US20220119533A1 - Heterodimeric fc-fused proteins - Google Patents

Heterodimeric fc-fused proteins Download PDF

Info

Publication number
US20220119533A1
US20220119533A1 US17/287,849 US201917287849A US2022119533A1 US 20220119533 A1 US20220119533 A1 US 20220119533A1 US 201917287849 A US201917287849 A US 201917287849A US 2022119533 A1 US2022119533 A1 US 2022119533A1
Authority
US
United States
Prior art keywords
antibody
domain polypeptide
seq
domain
subunit
Prior art date
Legal status (The legal status 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 status listed.)
Pending
Application number
US17/287,849
Other languages
English (en)
Inventor
Ann F. Cheung
Jean-Marie CULLEROT
Asya Grinberg
Eva Gutierrez
William Haney
Nicolai Wagtmann
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Dragonfly Therapeutics Inc
Original Assignee
Dragonfly Therapeutics Inc
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
Application filed by Dragonfly Therapeutics Inc filed Critical Dragonfly Therapeutics Inc
Priority to US17/287,849 priority Critical patent/US20220119533A1/en
Publication of US20220119533A1 publication Critical patent/US20220119533A1/en
Assigned to DRAGONFLY THERAPEUTICS, INC. reassignment DRAGONFLY THERAPEUTICS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CHEUNG, Ann F., CUILLEROT, Jean-Marie, GRINBERG, ASYA, GUTIERREZ, Eva, HANEY, WILLIAM, Wagtmann, Nicolai
Pending legal-status Critical Current

Links

Images

Classifications

    • 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
    • C07K16/2851Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the lectin superfamily, e.g. CD23, CD72
    • 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
    • C07K14/54Interleukins [IL]
    • C07K14/5434IL-12
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • 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
    • C07K16/283Immunoglobulins [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 Fc-receptors, e.g. CD16, CD32, CD64
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype
    • C07K2317/526CH3 domain
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/71Decreased effector function due to an Fc-modification
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/30Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/70Fusion polypeptide containing domain for protein-protein interaction

Definitions

  • the invention generally relates to heterodimeric Fc-fused proteins and pharmaceutical compositions comprising such proteins, and methods of use in treating a disease or disorder in a human patient.
  • Physiologically active proteins mostly have the disadvantage of having a short in vivo half-life.
  • PEG polyethylene glycol
  • Fc crystallizable fragment
  • Proteins composed of two or more different subunits, in which the two or more different subunits form a protein complex to exhibit physiological activity can also be fused to heterodimeric Fc regions derived not only from IgG1, but also from other isotype antibodies such as IgG2, IgG3 and IgG4, to form a heterodimeric Fc-fused protein.
  • one or more subunit(s) of the protein which is composed of two or more different subunits and in which two or more subunits exhibit physiological activity by forming a protein complex, can be fused to the terminus of heterodimeric Fc variant regions to form improved Fc-fused protein forms.
  • Fc heterodimerization is a technology that induces mutations in two different CH3 domains of Fc by genetic engineering, such that the two Fc fragments form a heterodimer with minimal sequence variations while they have tertiary structures very similar to those of naturally occurring antibodies (see, e.g., U.S. Pat. No. 7,695,936).
  • inventions described in the present disclosure provide designs for improving the Fc-fused protein forms, in which the two subunits of a heterodimeric protein are connected to two Fc domains having different heterodimerization domains, by introducing linkers of varying lengths, or mutations in the CH2 and the CH3 domains of the Fc.
  • the invention generally relates to heterodimeric Fc-fused proteins and pharmaceutical compositions comprising such proteins.
  • the present invention provides a heterodimeric Fc-fused protein comprising a first polypeptide comprising a first antibody Fc domain polypeptide and a first subunit of a multisubunit protein; and a second polypeptide comprising a second antibody Fc domain polypeptide and a second, different subunit of the multisubunit protein, wherein the first and second antibody Fc domain polypeptides each comprise different mutations promoting heterodimerization, wherein the first and/or second antibody Fc domain polypeptides comprise one or more mutation(s) that reduce(s) an effector function of an Fc, and wherein the first subunit and second, different subunit of the multisubunit protein are bound to each other.
  • the effector function comprises the ability of an Fc domain polypeptide to induce antibody-dependent cellular cytotoxicity (ADCC), antibody-dependent cellular phagocytosis (ADCP), and/or complement dependent cytotoxicity (CDC).
  • the first and second antibody Fc domain polypeptides are human antibody Fc domain polypeptides.
  • the first and second antibody Fc domain polypeptides are IgG1, IgG2, IgG3, IgG4, IgM, IgA, IgD, or IgE Fc domain polypeptides (e.g., human IgG1, IgG2, IgG3, IgG4, IgM, IgA, IgD, or IgE Fc domain polypeptides).
  • the first and/or second antibody Fc domain polypeptides comprise one or more mutation(s) at position(s) 233, 234, 235, 236, 237, 297, 318, 320, 322, 329, 330, and/or 331 under EU numbering.
  • the heterodimeric Fc-fused protein is fucosylated.
  • the first and second antibody Fc domain polypeptides are human IgG1 Fc domain polypeptides. In some embodiments, the first and/or second antibody Fc domain polypeptides comprise one or more mutation(s) at position(s) 234, 235, 237, 329, 330, and/or 331 under EU numbering. In some embodiments, the first and/or second antibody Fc domain polypeptides comprise one or more mutation(s) selected from L234A, L235A, L235E, G237A, P329A, A330S, and P331S. In some embodiments, the first and second antibody Fc domain polypeptides each comprise mutations L234A and L235A.
  • the first and second antibody Fc domain polypeptides each comprise mutations L234A, L235A, and P329A. In some embodiments, the first and second antibody Fc domain polypeptides each comprise mutations L234A, L235E, G237A, A330S, and P331S. In some embodiments, the first and second antibody Fc domain polypeptides each comprise mutation C220S.
  • the first and second antibody Fc domain polypeptides are human IgG4 Fc domain polypeptides. In some embodiments, the first and/or second antibody Fc domain polypeptides comprise one or more mutation(s) at position(s) 235 and/or 329 under EU numbering. In some embodiments, the first and/or second antibody Fc domain polypeptides comprise one or more mutation(s) selected from L235E and P329A. In some embodiments, the first and second antibody Fc domain polypeptides each comprise mutation L235E. In some embodiments, the first and second antibody Fc domain polypeptides each comprise mutations L235E and P329A. In some embodiments, the first and second antibody Fc domain polypeptides each comprise mutation S228P.
  • the present invention provides a heterodimeric Fc-fused protein comprising a first polypeptide comprising the amino acid sequence of SEQ ID NO:290 and a second polypeptide comprising the amino acid sequence of SEQ ID NO:291.
  • the present invention provides a polypeptide comprising a subunit of a multisubunit cytokine and an immunoglobulin Fc domain polypeptide; the Fc domain polypeptide comprises mutations for promoting heterodimerization with a different immunoglobulin Fc domain polypeptide, and one or more mutation(s) that reduce(s) an effector function of an Fc.
  • the Fc domain polypeptide comprising the mutations can be a human IgG1 antibody Fc domain polypeptide.
  • the one or more mutation(s) that reduce(s) an effector function of an Fc is selected from L234A, L235A or L235E, G237A, P329A, A330S, and P331S, numbered according to the EU numbering system.
  • the mutations that reduce an effector function of an Fc are L234A, L235A, and P329A, numbered according to the EU numbering system.
  • the mutations for promoting heterodimerization are K360E and K409W, numbered according to the EU numbering system. In some other embodiments, the mutations for promoting heterodimerization are Q347R, D399V, and F405T, numbered according to the EU numbering system.
  • the Fc domain polypeptide further comprises a mutation for promoting disulfide bond formation with a different immunoglobulin Fc domain polypeptide.
  • the mutation for promoting disulfide bond formation is Y349C, numbered according to the EU numbering system.
  • the mutation for promoting disulfide bond formation is S354C, numbered according to the EU numbering system.
  • a polypeptide comprising a subunit of a multisubunit cytokine and an immunoglobulin Fc domain polypeptide comprises an amino acid sequence of SEQ ID NO:290 or an amino acid sequence of SEQ ID NO:291.
  • the present invention provides a nucleic acid encoding a polypeptide comprising an amino acid sequence of SEQ ID NO:290 or SEQ ID NO:291.
  • the present invention provides an expression vector comprising a nucleic acid comprising a sequence encoding a polypeptide comprising an amino acid sequence of SEQ ID NO:290 or SEQ ID NO:291.
  • the present invention provides a cell comprising a nucleic acid encoding a polypeptide comprising an amino acid sequence of SEQ ID NO:290 or SEQ ID NO:291, or an expression vector comprising a nucleic acid comprising a sequence encoding a polypeptide comprising an amino acid sequence of SEQ ID NO:290 or SEQ ID NO:291.
  • the present invention provides a heterodimeric Fc-fused protein comprising a first Fc domain polypeptide and a second, different Fc domain polypeptide of an immunoglobulin Fc and a first subunit and a second, different subunit of a multisubunit protein wherein the first subunit and the second, different subunit are bound to each other and linked to one or more end(s) of the N-terminus or C-terminus of the first Fc domain polypeptide and/or the second, different Fc domain polypeptide; wherein the first Fc domain polypeptide and the second Fc domain polypeptide are mutated so as to promote heterodimeric Fc formation and to reduce an effector function of an Fc.
  • the multisubunit protein is IL-12 (e.g., human IL-12), such that in a heterodimeric Fc-fused protein the p35 and p40 subunits of IL-12 are bound to each other and linked to one or more end(s) of the N-terminus or C-terminus of the first Fc domain polypeptide and/or the second Fc domain polypeptide; wherein the first Fc domain polypeptide and the second Fc domain polypeptide are mutated so as to promote heterodimeric Fc formation and to reduce an effector function of an Fc.
  • IL-12 e.g., human IL-12
  • the present invention provides a heterodimeric Fc-fused protein comprising: a first polypeptide comprising a first antibody Fc domain polypeptide and a second polypeptide comprising a second antibody Fc domain polypeptide bound to the first antibody Fc domain polypeptide, in which the first polypeptide further comprises a first subunit of a multisubunit protein fused by a linker comprising amino acid sequence PKSSDKTHTCPPCPAPEX 1 X 2 GX 3 (SEQ ID NO:237) or EPKSSDKTHTCPPCPAPEX 1 X 2 GX 3 (SEQ ID NO:6) to the first antibody Fc domain polypeptide, wherein X 1 represents L or A, X 2 represents L, E, or A, and X 3 represents A or G; a second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide and the subunits of the multisubunit protein are bound to each other; when X 1 represents L and/or X 2 represents L, at least
  • the linker connecting the first subunit of the multisubunit protein to the first antibody Fc domain polypeptide comprises amino acid sequence PKSSDKTHTCPPCPAPEAAGG (SEQ ID NO:239) or EPKSSDKTHTCPPCPAPEAAGG (SEQ ID NO:9). In some embodiments, within the heterodimeric Fc-fused protein, the linker connecting the first subunit of the multisubunit protein to the first antibody Fc domain polypeptide consists of amino acid sequence PKSSDKTHTCPPCPAPEAAGG (SEQ ID NO:239) or EPKSSDKTHTCPPCPAPEAAGG (SEQ ID NO:9).
  • the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker comprising amino acid sequence GGGGSGGGGSGGGGSEPKSSDKTHTCPPCPAPEAAGG (SEQ ID NO:10) or GGGGSGGGGSGGGGSPKSSDKTHTCPPCPAPEAAGG (SEQ ID NO:244).
  • the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker consisting of amino acid sequence GGGGSGGGGSGGGGSEPKSSDKTHTCPPCPAPEAAGG (SEQ ID NO:10) or GGGGSGGGGSGGGGSPKSSDKTHTCPPCPAPEAAGG (SEQ ID NO:244).
  • the linker connecting the first subunit of the multisubunit protein to the first antibody Fc domain polypeptide comprises amino acid sequence PKSSDKTHTCPPCPAPELLGG (SEQ ID NO:238) or EPKSSDKTHTCPPCPAPELLGG (SEQ ID NO:7).
  • the linker fusing the first subunit of the multisubunit protein to the first antibody Fc domain polypeptide consists of amino acid sequence PKSSDKTHTCPPCPAPELLGG (SEQ ID NO:238) or EPKSSDKTHTCPPCPAPELLGG (SEQ ID NO:7).
  • the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker comprising amino acid sequence GGGGSGGGGSGGGGSEPKSSDKTHTCPPCPAPELLGG (SEQ ID NO:8) or GGGGSGGGGSGGGGSPKSSDKTHTCPPCPAPELLGG (SEQ ID NO:241).
  • the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker consisting of amino acid sequence GGGGSGGGGSGGGGSEPKSSDKTHTCPPCPAPELLGG (SEQ ID NO:8) or GGGGSGGGGSGGGGSPKSSDKTHTCPPCPAPELLGG (SEQ ID NO:241).
  • the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker comprising amino acid sequence GGGGSEPKSSDKTHTCPPCPAPELLGG (SEQ ID NO:15) or GGGGSPKSSDKTHTCPPCPAPELLGG (SEQ ID NO:242).
  • the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker consisting of amino acid sequence GGGGSEPKSSDKTHTCPPCPAPELLGG (SEQ ID NO:15) or GGGGSPKSSDKTHTCPPCPAPELLGG (SEQ ID NO:242).
  • the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker comprising amino acid sequence GGGGSGGGGSEPKSSDKTHTCPPCPAPELLGG (SEQ ID NO:16) or GGGGSGGGGSPKSSDKTHTCPPCPAPELLGG (SEQ ID NO:243).
  • the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker consisting of amino acid sequence GGGGSGGGGSEPKSSDKTHTCPPCPAPELLGG (SEQ ID NO:16) or GGGGSGGGGSPKSSDKTHTCPPCPAPELLGG (SEQ ID NO:243).
  • the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker comprising amino acid sequence GGGGSEPKSSDKTHTCPPCPAPEAAGG (SEQ ID NO:65) or GGGGSPKSSDKTHTCPPCPAPEAAGG (SEQ ID NO:245).
  • the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker consisting of amino acid sequence GGGGSEPKSSDKTHTCPPCPAPEAAGG (SEQ ID NO:65) or GGGGSPKSSDKTHTCPPCPAPEAAGG (SEQ ID NO:245).
  • the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker comprising amino acid sequence GGGGSGGGGSEPKSSDKTHTCPPCPAPEAAGG (SEQ ID NO:66) or GGGGSGGGGSPKSSDKTHTCPPCPAPEAAGG (SEQ ID NO:246).
  • the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker consisting of amino acid sequence GGGGSGGGGSEPKSSDKTHTCPPCPAPEAAGG (SEQ ID NO:66) or GGGGSGGGGSPKSSDKTHTCPPCPAPEAAGG (SEQ ID NO:246).
  • the linker connecting the first subunit of the multisubunit protein to the first antibody Fc domain polypeptide comprises amino acid sequence EPKSSDKTHTCPPCPAPEAEGA (SEQ ID NO:11) or PKSSDKTHTCPPCPAPEAEGA (SEQ ID NO:240).
  • the linker fusing the first subunit of the multisubunit protein to the first antibody Fc domain polypeptide consists of amino acid sequence EPKSSDKTHTCPPCPAPEAEGA (SEQ ID NO:11) or PKSSDKTHTCPPCPAPEAEGA (SEQ ID NO:240).
  • the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker comprising amino acid sequence GGGGSGGGGSGGGGSEPKSSDKTHTCPPCPAPEAEGA (SEQ ID NO:12) or GGGGSGGGGSGGGGSPKSSDKTHTCPPCPAPEAEGA (SEQ ID NO:247).
  • the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker consisting of amino acid sequence GGGGSGGGGSGGGGSEPKSSDKTHTCPPCPAPEAEGA (SEQ ID NO:12) or GGGGSGGGGSGGGGSPKSSDKTHTCPPCPAPEAEGA (SEQ ID NO:247).
  • the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker comprising amino acid sequence GGGGSEPKSSDKTHTCPPCPAPEAEGA (SEQ ID NO:67) or GGGGSPKSSDKTHTCPPCPAPEAEGA (SEQ ID NO:248).
  • the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker consisting of amino acid sequence GGGGSEPKSSDKTHTCPPCPAPEAEGA (SEQ ID NO:67) or GGGGSPKSSDKTHTCPPCPAPEAEGA (SEQ ID NO:248).
  • the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker comprising amino acid sequence GGGGSGGGGSEPKSSDKTHTCPPCPAPEAEGA (SEQ ID NO:68) or GGGGSGGGGSPKSSDKTHTCPPCPAPEAEGA (SEQ ID NO:249).
  • the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker consisting of amino acid sequence GGGGSGGGGSEPKSSDKTHTCPPCPAPEAEGA (SEQ ID NO:68) or GGGGSGGGGSPKSSDKTHTCPPCPAPEAEGA (SEQ ID NO:249).
  • the present invention provides a heterodimeric Fc-fused protein comprising a subunit of a multisubunit cytokine connected to an immunoglobulin Fc domain polypeptide by a linker comprising an amino acid sequence PKSSDKTHTCPPCPAPEAAGG (SEQ ID NO:239); the Fc domain polypeptide comprises mutations for promoting heterodimerization with a different immunoglobulin Fc domain polypeptide, and L234A, L235A, and P329A substitutions for reducing an effector function of an Fc.
  • the present invention provides a heterodimeric Fc-fused protein comprising a p40 subunit of human IL-12 connected to a first human IgG1 (hIgG1) Fc domain polypeptide by a linker comprising or consisting of an amino acid sequence PKSSDKTHTCPPCPAPEAAGG (SEQ ID NO:239), and a p35 subunit of human IL-12 connected to a second hIgG1 Fc domain polypeptide by a linker comprising or consisting of an amino acid sequence GGGGSGGGGSGGGGSEPKSSDKTHTCPPCPAPEAAGG (SEQ ID NO:10); the first and the second Fc domain polypeptides comprise mutations promoting heterodimerization, and L234A, L235A, and P329A substitutions for reducing an effector function of a hIgG1 Fc.
  • the present invention provides a heterodimeric Fc-fused protein comprising: a first polypeptide comprising a first antibody Fc domain polypeptide and a second polypeptide comprising a second antibody Fc domain polypeptide, in which the first polypeptide further comprises a first subunit of a multisubunit protein, in which the protein sequence is fused by a linker comprising amino acid sequence RVESKYGPPCPPCPAPEFXGG (SEQ ID NO:1) to the first antibody Fc domain polypeptide, in which X represents L or E; a second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide, and the subunits of the multisubunit protein are bound to each other, the first antibody Fc domain polypeptide and the second antibody Fc domain polypeptide each contain different mutations promoting heterodimerization, and the first antibody Fc domain polypeptide and the second antibody Fc domain polypeptide are bound to each other.
  • a heterodimeric Fc-fused protein of the present invention comprises a first polypeptide comprising a first antibody Fc domain polypeptide and a second polypeptide comprising a second antibody Fc domain polypeptide, in which the first polypeptide further comprises a first subunit of a multisubunit protein, in which the protein sequence is fused by a linker to the first antibody Fc domain polypeptide; and the second polypeptide further comprises a second, different subunit of a multisubunit protein, in which the protein sequence is fused by a linker to the second antibody Fc domain polypeptide.
  • the linker connecting the protein sequence of the second, different subunit of a multisubunit protein to the second antibody Fc domain polypeptide may include G45 (SEQ ID NO:110), (G45) 2 (SEQ ID NO:109), or (G4S) 3 (SEQ ID NO:108).
  • the linker connecting the subunit of the multisubunit protein to the first antibody Fc domain polypeptide comprises amino acid sequence RVESKYGPPCPPCPAPEFLGG (SEQ ID NO:2).
  • the linker fusing the first subunit of the multisubunit protein to the first antibody Fc domain polypeptide consists of amino acid sequence RVESKYGPPCPPCPAPEFLGG (SEQ ID NO:2).
  • the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker comprising amino acid sequence GGGGSGGGGSGGGGSRVESKYGPPCPPCPAPEFLGG (SEQ ID NO:3). In some embodiments, the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker consisting of amino acid sequence GGGGSGGGGSGGGGSRVESKYGPPCPPCPAPEFLGG (SEQ ID NO:3).
  • the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker comprising amino acid sequence GGGGSRVESKYGPPCPPCPAPEFLGG (SEQ ID NO:13). In some embodiments, the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker consisting of amino acid sequence GGGGSRVESKYGPPCPPCPAPEFLGG (SEQ ID NO:13).
  • the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker comprising amino acid sequence GGGGSGGGGSRVESKYGPPCPPCPAPEFLGG (SEQ ID NO:14). In some embodiments, the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker consisting of amino acid sequence GGGGSGGGGSRVESKYGPPCPPCPAPEFLGG (SEQ ID NO:14).
  • the linker connecting the subunit of the multisubunit protein to the first antibody Fc domain polypeptide comprises amino acid sequence RVESKYGPPCPPCPAPEFEGG (SEQ ID NO:4).
  • the linker fusing the first subunit of the multisubunit protein to the first antibody Fc domain polypeptide consists of amino acid sequence RVESKYGPPCPPCPAPEFEGG (SEQ ID NO:4).
  • the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker comprising amino acid sequence GGGGSGGGGSGGGGSRVESKYGPPCPPCPAPEFEGG (SEQ ID NO:5). In some embodiments, the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker consisting of amino acid sequence GGGGSGGGGSGGGGSRVESKYGPPCPPCPAPEFEGG (SEQ ID NO:5).
  • the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker comprising amino acid sequence GGGGSRVESKYGPPCPPCPAPEFEGG (SEQ ID NO:63). In some embodiments, the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker consisting of amino acid sequence GGGGSRVESKYGPPCPPCPAPEFEGG (SEQ ID NO:63).
  • the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker comprising amino acid sequence GGGGSGGGGSRVESKYGPPCPPCPAPEFEGG (SEQ ID NO:64). In some embodiments, the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker consisting of amino acid sequence GGGGSGGGGSRVESKYGPPCPPCPAPEFEGG (SEQ ID NO:64).
  • Some heterodimeric Fc-fused proteins described herein include a first IgG4 antibody Fc domain polypeptide and a second, different IgG4 antibody Fc domain polypeptide, each mutated to promote heterodimerization with each other.
  • the first IgG4 antibody Fc domain polypeptide includes one or more mutation(s) selected from K370E and R409W
  • the second, different IgG4 antibody Fc domain polypeptide includes one or more mutation(s) selected from E357N, D399V, and F405T.
  • the first IgG4 antibody Fc domain polypeptide includes one or more mutation(s) selected from E357N, D399V, and F405T
  • the second, different IgG4 antibody Fc domain polypeptide includes one or more mutation(s) selected from K370E and R409W
  • the first IgG4 antibody Fc domain polypeptide includes mutations K370E and R409W
  • the second, different IgG4 antibody Fc domain polypeptide includes mutations E357N, D399V, and F405T.
  • the first IgG4 antibody Fc domain polypeptide includes mutations E357N, D399V, and F405T, and the second, different IgG4 antibody Fc domain polypeptide includes mutations K370E and R409W. In some embodiments, the first IgG4 Fc domain polypeptide includes one or more mutation(s) selected from K360E and R409W, and the second, different IgG4 Fc domain polypeptide includes one or more mutation(s) selected from Q347R, D399V, and F405T.
  • the first IgG4 Fc domain polypeptide includes one or more mutation(s) selected from Q347R, D399V, and F405T
  • the second, different IgG4 Fc domain polypeptide includes one or more mutation(s) selected from K360E and R409W
  • the first IgG4 Fc domain polypeptide includes mutations K360E and R409W
  • the second, different IgG4 Fc domain polypeptide includes mutations Q347R, D399V, and F405T.
  • the first IgG4 Fc domain polypeptide includes mutations Q347R, D399V, and F405T
  • the second, different IgG4 Fc domain polypeptide includes mutations K360E and R409W.
  • Some heterodimeric Fc-fused proteins disclosed herein include a first IgG1 antibody Fc domain polypeptide and a second, different IgG1 antibody Fc domain polypeptide, each mutated to promote heterodimerization with each other.
  • the first IgG1 Fc domain polypeptide includes one or more mutation(s) selected from K360E and K409W
  • the second, different IgG1 Fc domain polypeptide includes one or more mutation(s) selected from Q347R, D399V, and F405T.
  • the first IgG1 Fc domain polypeptide includes one or more mutation(s) selected from Q347R, D399V, and F405T
  • the second, different IgG1 Fc domain polypeptide includes one or more mutation(s) selected from K360E and K409W
  • the first IgG1 antibody Fc domain polypeptide includes mutations K360E and K409W
  • the second, different IgG1 antibody Fc domain polypeptide includes mutations Q347R, D399V, and F405T.
  • the first IgG1 antibody Fc domain polypeptide includes mutations Q347R, D399V, and F405T, and the second, different IgG1 antibody Fc domain polypeptide includes mutations K360E and K409W.
  • a heterodimeric Fc-fused protein described herein comprises IgG4 or IgG1 Fc domain polypeptides further mutated to reduce effector functions.
  • the first antibody Fc domain polypeptide and the second antibody Fc domain polypeptide each contain the mutation P329G or P329A.
  • the first IgG4 antibody Fc domain polypeptide and the second, different IgG4 antibody Fc domain polypeptide each contain the mutation P329G or P329A.
  • the first IgG1 antibody Fc domain polypeptide and the second, different IgG1 antibody Fc domain polypeptide each contain the mutation P329G or P329A.
  • the first IgG4 antibody Fc domain polypeptide and the second, different IgG4 antibody Fc domain polypeptide each contain the mutation P329A. In some embodiments, the first IgG1 antibody Fc domain polypeptide and the second, different IgG1 antibody Fc domain polypeptide each contain the mutation P329A.
  • a heterodimeric Fc-based fusion described herein incorporates a first IgG1 antibody Fc domain polypeptide and a second, different IgG1 antibody Fc domain polypeptide each containing a mutation selected from A330S and P331S. In some embodiments, a heterodimeric Fc-based fusion described herein incorporates a first IgG1 antibody Fc domain polypeptide and a second, different IgG1 antibody Fc domain polypeptide each containing the mutations A330S and P331S.
  • a heterodimeric Fc-based protein described herein incorporates IgG4 or IgG1 Fc domain polypeptides that are further mutated to introduce an inter-chain disulfide bond.
  • the first IgG4 or IgG1 Fc domain polypeptide includes mutation Y349C
  • the second, different IgG4 or IgG1 Fc domain polypeptide includes mutation S354C
  • the first IgG4 or IgG1 Fc domain polypeptide includes mutation S354C
  • the second, different IgG4 or IgG1 Fc domain polypeptide includes mutation Y349C.
  • heterodimeric Fc-fused proteins of the present invention include a native disulfide bond between the first subunit of a multisubunit protein and the second, different subunit of the multisubunit protein.
  • a heterodimeric Fc-fused protein according to the invention includes a native heterodimer disulfide bond between p35 and p40 subunits of IL-12.
  • Such a protein includes the native disulfide bond between C74 of p35 and C177 of p40.
  • heterodimeric Fc-fused proteins of the present invention include an artificial or engineered heterodimer disulfide bond between the first subunit of a multisubunit protein and the second, different subunit of the multisubunit protein.
  • a heterodimeric Fc-fused protein according to the invention includes an artificial or engineered heterodimer disulfide bond between p35 and p40 subunits of IL-12.
  • Such a protein includes an artificial or engineered disulfide bond between V185C of p35 and Y292C of p40.
  • Some heterodimeric Fc-fused proteins of the present invention include a native disulfide bond between the first subunit of a multisubunit protein and the second, different subunit of the multisubunit protein, and an artificial or engineered heterodimer disulfide bond between the first subunit of a multisubunit protein and the second, different subunit of the multisubunit protein.
  • a native heterodimer disulfide bond between p35 and p40 subunits of IL-12 and includes an artificial or engineered heterodimer disulfide bond between p35 and p40 subunits of IL-12.
  • Such a protein includes the native disulfide bond between C74 of p35 and C177 of p40, and an artificial or engineered disulfide bond between V185C of p35 and Y292C of p40.
  • a heterodimeric Fc-fused protein according to the invention includes p35 of IL-12 in which the native C74 is mutated to serine, and a p40 of 11-12 in which the native C177 is mutated to serine, thereby removing the native disulfide bond between p35 and p40 subunits of IL-12.
  • p35 of IL-12 in which the native C74 is mutated to serine
  • a p40 of 11-12 in which the native C177 is mutated to serine
  • a first polypeptide and a second, different polypeptide comprise a first subunit and a second, different subunit of a multisubunit cytokine, respectively.
  • a first polypeptide and a second, different polypeptide comprise a second, different subunit and a first subunit of a multisubunit cytokine, respectively.
  • the cytokine is IL-12 (e.g., human IL-12).
  • Formulations containing any one of the heterodimeric Fc-fused proteins described herein, cells containing one or more nucleic acid(s) expressing the heterodimeric Fc-fused proteins or vector(s) expressing the heterodimeric Fc-fused protein, and methods of enhancing tumor cell death using the heterodimeric Fc-fused proteins are also provided.
  • the invention provides a formulation that includes a heterodimeric Fc-fused protein described herein and a pharmaceutically acceptable carrier.
  • a heterodimeric Fc-fused protein of the present invention further comprises at least one antibody variable domain (e.g., an antibody heavy chain variable domain).
  • the at least one antibody heavy chain variable domain binds to an antibody light chain variable region to form an Fab, and the heavy chain variable domain or the light chain variable domain of the Fab is fused at the N-terminus of the first antibody Fc domain polypeptide and/or the second antibody Fc domain polypeptide.
  • the heterodimeric Fc-fused protein comprising at least one antibody variable domain of the present invention has a first subunit of a multisubunit protein and a second, different subunit of the multisubunit protein connected to the C-terminus of the first antibody Fc domain polypeptide and the second antibody Fc domain polypeptide, respectively.
  • the heterodimeric Fc-fused protein comprising at least one antibody variable domain of the present invention has a first subunit of a multisubunit protein and a second, different subunit of the multisubunit protein connected to the C-terminus of the second antibody Fc domain polypeptide and the first antibody Fc domain polypeptide, respectively.
  • a heterodimeric Fc-fused protein of the present invention comprises an antibody heavy chain variable domain positioned at the C-terminus to the first antibody Fc domain polypeptide of the first polypeptide. In certain embodiments, a heterodimeric Fc-fused protein of the present invention comprises an antibody heavy chain variable domain positioned C-terminally to the second antibody Fc domain polypeptide of the second polypeptide. In certain embodiments, the antibody heavy chain variable region binds to an antibody light chain variable region to form an scFv.
  • a first subunit of a multisubunit protein and the second, different subunit of the multisubunit protein are connected to the N-terminus of the first antibody Fc domain polypeptide and the second antibody Fc domain polypeptide, respectively. In certain embodiments, a first subunit of a multisubunit protein and the second, different subunit of the multisubunit protein are connected to the N-terminus of the second antibody Fc domain polypeptide and the first antibody Fc domain polypeptide, respectively.
  • a heterodimeric Fc-fused protein of the present invention does not comprise an antibody variable domain.
  • the heterodimeric Fc-fused protein consists of or consists essentially of a first subunit of a multisubunit protein (e.g., an IL-12 protein), a linker optionally comprising a spacer peptide, and a first antibody Fc domain polypeptide; and a second, different subunit of the multisubunit protein (e.g., an IL-12 protein), a linker optionally comprising a spacer peptide, and a second antibody Fc domain polypeptide.
  • a multisubunit protein e.g., an IL-12 protein
  • a heterodimeric Fc-fused protein of the present invention further comprises a proteoglycan-binding domain.
  • the proteoglycan-binding domain binds one or more proteoglycans that are specifically expressed in a tumor.
  • the proteoglycan-binding domain binds one or more proteoglycans selected from syndecan, serglycin, CSPG4, betaglycan, glypican, perlecan, versican, brevican, and small leucine-rich proteoglycans (SLRPs).
  • the SLRPs are selected from decorin, biglycan, asporin, fibrodulin, and lumican.
  • the proteoglycan-binding domain is linked to the C-terminus of the first antibody Fc domain polypeptide. In some embodiments, the proteoglycan-binding domain is linked to the C-terminus of the second antibody Fc domain polypeptide.
  • a heterodimeric Fc-fused protein of the present invention further comprises a collagen-binding domain.
  • the collagen-binding domain binds one or more collagens that are specifically expressed in a tumor.
  • the collagen-binding domain is linked to the C-terminus of the first antibody Fc domain polypeptide.
  • the collagen-binding domain is linked to the C-terminus of the second antibody Fc domain polypeptide.
  • a heterodimeric Fc-fused protein of the present invention further comprises a hyaluronic acid-binding domain.
  • the hyaluronic acid-binding domain is linked to the C-terminus of the first antibody Fc domain polypeptide.
  • the hyaluronic acid-binding domain is linked to the C-terminus of the second antibody Fc domain polypeptide.
  • Another aspect of the invention provides a method of treating cancer in a patient.
  • the method comprises administering to a patient, for example, a patient in need thereof, an effective amount of a heterodimeric Fc-fused protein described herein or a formulation that includes an effective amount of a multi-specific binding protein described herein.
  • the method of treating cancer includes administering to a patient, for example, a patient in need of treatment, a formulation that includes an effective amount of a heterodimeric Fc-fused protein described herein and a pharmaceutically acceptable carrier.
  • the present disclosure provides a method of treating cancer, the method comprising administering to a patient only a single dose of a heterodimeric IL-12-Fc-fused protein.
  • the single dose is in an amount sufficient to induce a complete response to the cancer.
  • the single dose is in an amount sufficient to delay or prevent recurrence of the cancer.
  • the present disclosure provides a method of treating cancer, the method comprising administering to a patient only a single dose of a heterodimeric IL-12-Fc-fused protein comprising a first polypeptide comprising the amino acid sequence of SEQ ID NO:290 and a second polypeptide comprising the amino acid sequence of SEQ ID NO:291, or a formulation comprising the heterodimeric IL-12-Fc-fused protein and a pharmaceutically acceptable carrier.
  • a single dose of a heterodimeric IL-12-Fc-fused protein comprising a first polypeptide comprising the amino acid sequence of SEQ ID NO:290 and a second polypeptide comprising the amino acid sequence of SEQ ID NO:291, or a formulation comprising the heterodimeric IL-12-Fc-fused protein and a pharmaceutically acceptable carrier is in an amount sufficient to induce a complete response to the cancer.
  • the single dose is in an amount sufficient to delay or prevent recurrence of the cancer.
  • the acute radiation syndrome comprises one or more syndrome(s) selected from hematopoietic radiation syndrome, gastrointestinal radiation syndrome, neurovascular radiation syndrome, and cutaneous radiation syndrome.
  • the acute radiation syndrome comprises a syndrome selected from the group consisting of hematopoietic radiation syndrome, gastrointestinal radiation syndrome, neurovascular radiation syndrome, and cutaneous radiation syndrome.
  • the present invention provides heterodimeric Fc-fused protein constructs of multisubunit proteins. These fusion protein constructs can exhibit a higher serum half-life compared to a native/natural multisubunit protein, improved yield during production, enhanced stability during storage, and/or improved efficacy when used as a therapeutic.
  • FIG. 1A illustrates an exemplary heterodimeric Fc-fused protein comprising a first subunit of a multisubunit protein connected by a linker to a first antibody Fc domain polypeptide, and a second, different subunit of a multisubunit protein connected by a linker to a second antibody Fc domain polypeptide.
  • the first antibody Fc domain polypeptide and the second antibody Fc domain polypeptide each contain different mutations promoting heterodimerization, and a disulfide bond between the Fc domain polypeptides stabilizes the heterodimer.
  • the linker connecting the additional subunit to the second antibody Fc domain polypeptide may include (G4S) 3 sequence (SEQ ID NO:108).
  • FIG. 1B illustrates an exemplary heterodimeric Fc-fused protein similar to the protein illustrated in FIG. 1A , but also including mutations in the Fc domain polypeptide to reduce Fc ⁇ R binding.
  • FIG. 1C illustrates an exemplary protein in which the second, different subunit of the multisubunit protein is connected by a linker to the first antibody Fc domain polypeptide, and the other subunit of the multisubunit protein is connected by another linker to the second antibody Fc domain polypeptide.
  • FIG. 1D illustrates an exemplary heterodimeric Fc-fused protein similar to the protein illustrated in FIG. 1C , but also including mutations in the Fc domain polypeptide to reduce Fc ⁇ R binding.
  • FIG. 1E illustrates an exemplary heterodimeric Fc-fused protein similar to the protein illustrated in FIG. 1B , except that the second, different subunit of a multisubunit protein is positioned at the C-terminus of the second antibody Fc domain polypeptide in the second polypeptide.
  • FIG. 1F illustrates an exemplary heterodimeric Fc-fused protein similar to the protein illustrated in FIG. 1E , except that the second, different subunit of a multisubunit protein is positioned at the C-terminus of the first antibody Fc domain polypeptide in the first polypeptide.
  • FIG. 2A illustrates an exemplary heterodimeric Fc-fused protein comprising a first subunit of a multisubunit protein connected by a linker to a first antibody Fc domain polypeptide, and a second, different subunit of a multisubunit protein connected by another linker to a second antibody Fc domain polypeptide, in which the subunits are connected by two disulfide bonds.
  • the first antibody Fc domain polypeptide and the second antibody Fc domain polypeptide each contain different mutations promoting heterodimerization, and a disulfide bond between the Fc domain polypeptides stabilizes the heterodimer.
  • the illustrated protein also includes mutations in the Fc domain polypeptide to reduce Fc ⁇ R binding.
  • FIG. 2B illustrates an exemplary heterodimeric Fc-fused protein comprising a first subunit of a multisubunit protein connected by a linker to a first antibody Fc domain polypeptide, and a second, different subunit of a multisubunit protein connected by another linker to a second antibody Fc domain polypeptide, in which the subunits are connected by one non-native disulfide bond.
  • the native disulfide bond has been removed and replaced with an artificial disulfide bond.
  • an IL-12 construct can incorporate mutations p35-V185C/C74S and p40-Y292C/C177S.
  • the first antibody Fc domain polypeptide and the second antibody Fc domain polypeptide each contain different mutations promoting heterodimerization, and a disulfide bond between the Fc domain polypeptides stabilizes the heterodimer.
  • the protein also includes mutations in the Fc domain polypeptide to reduce Fc ⁇ R binding.
  • FIG. 3A illustrates an exemplary heterodimeric Fc-fused protein comprising a first subunit of a multisubunit protein connected by a linker to a first antibody Fc domain polypeptide, and a second, different subunit of a multisubunit protein connected by another linker having the same amino acid sequence to a second antibody Fc domain polypeptide.
  • the first antibody Fc domain polypeptide and the second antibody Fc domain polypeptide each contain different mutations promoting heterodimerization, and a disulfide bond between the Fc domain polypeptides stabilizes the heterodimer.
  • FIG. 3B illustrates an exemplary heterodimeric Fc-fused protein comprising a first subunit of a multisubunit protein connected by a linker to a first antibody Fc domain polypeptide, and a second, different subunit of a multisubunit protein connected by another linker to a second antibody Fc domain polypeptide.
  • the first antibody Fc domain polypeptide and the second antibody Fc domain polypeptide each contain different mutations promoting heterodimerization, and a disulfide bond between the Fc domain polypeptides stabilizes the heterodimer.
  • the linker connecting the second, different subunit of the multisubunit protein to the second antibody Fc domain polypeptide may include a (G4S) 2 (SEQ ID NO:109) or G45 (SEQ ID NO:110) sequence.
  • FIGS. 4A-4L illustrate exemplary scFv fusion heterodimeric Fc-fused protein constructs ( FIGS. 4A-4H ), and mAb fusion heterodimeric Fc-fused protein constructs ( FIGS. 4I-4L ) in which a first subunit of a multisubunit protein is connected by a linker to a first antibody Fc domain polypeptide, and a second, different subunit of a multisubunit protein is connected by another linker to a second antibody Fc domain polypeptide.
  • FIGS. 4A-4D illustrate exemplary Fc-fused proteins including a first scFv linked to the C-terminus of the first antibody Fc domain polypeptide and a second scFv linked to the C-terminus of the second antibody Fc domain polypeptide.
  • the first scFv and the second scFv can be the same or different (e.g., bind to different antigens or different epitopes on a single antigen).
  • the exemplary Fc-fused proteins illustrated in FIG. 4A and FIG. 4C comprise different pairs of heterodimerization Fc variants.
  • the exemplary Fc-fused proteins illustrated in FIG. 4B and FIG. 4D comprise different pairs of heterodimerization Fc variants.
  • FIG. 4A and FIG. 4D illustrate exemplary Fc-fused proteins in which the first polypeptide comprises “hole” mutation(s) and the second polypeptide comprises “knob” mutation(s);
  • FIG. 4B and FIG. 4C illustrate exemplary Fc-fused proteins in which the first polypeptide comprises “knob” mutation(s) and the second polypeptide comprises “hole” mutation(s).
  • FIG. 4A and FIG. 4B illustrate exemplary Fc-fused proteins in which the protein sequence of a first subunit of a multisubunit protein is connected to the first polypeptide;
  • FIG. 4C and FIG. 4D illustrate exemplary Fc-fused proteins in which the second, different subunit of the multisubunit protein is connected to the first polypeptide.
  • Heterodimeric Fc-fused proteins with other types of heterodimerization mutations are similarly contemplated.
  • FIGS. 4E and 4G illustrate exemplary Fc-fused proteins including an scFv linked to the C-terminus of the first antibody Fc domain polypeptide.
  • FIGS. 4F and 4H illustrate exemplary Fc-fused proteins including an scFv linked to the C-terminus of the second antibody Fc domain polypeptide.
  • FIGS. 4I-4L illustrate exemplary Fc-fused proteins including a first Fab linked to the N-terminus of the first antibody Fc domain polypeptide and a second Fab linked to the N-terminus of the second antibody Fc domain polypeptide.
  • FIG. 4I and FIG. 4K illustrate exemplary Fc-fused proteins in which a first subunit of a multisubunit protein is connected to the first polypeptide;
  • FIG. 4J and FIG. 4L illustrate exemplary Fc-fused proteins in which the second, different subunit of the multisubunit protein is connected to the first polypeptide.
  • FIG. 4K differs from FIG.
  • FIG. 4I in having a longer amino acid sequence (e.g., spacer peptide disclosed herein) that connects the antibody Fc domain polypeptide and the a first subunit of a multisubunit protein;
  • FIG. 4J differs from FIG. 4L in having a longer amino acid sequence (e.g., spacer peptide disclosed herein) that connects the antibody Fc domain polypeptide and the first subunit of a multisubunit protein.
  • FIGS. 5A-5C are graphs showing tumor growth curves of individual mice inoculated with CT26 tumor cells and treated with recombinant mouse IL-12 (rmIL-12)( FIG. 5A ), DF-mIL-12-Fc wt ( FIG. 5B ), DF-mIL-12-Fc si ( FIG. 5C ), or mIgG2a isotype control once a week.
  • FIG. 6 is a graph showing Kaplan-Meier survival curves of mice inoculated with CT26 tumor cells and treated with rmIL-12, DF-mIL-12-Fc wt, DF-mIL-12-Fc si, or mIgG2a isotype control once a week.
  • FIGS. 7A-7D are graphs showing tumor growth curves of individual mice inoculated with CT26 tumor cells and treated with DF-mIL-12-Fc wt at a molar equivalent of 1 ⁇ g rmIL-12 ( FIG. 7A ), DF-mIL-12-Fc si at a molar equivalent of 1 ⁇ g rmIL-12 ( FIG. 7B ), DF-mIL-12-Fc wt at a molar equivalent of 0.1 ⁇ g rmIL-12 ( FIG. 7C ), DF-mIL-12-Fc si at a molar equivalent of 0.1 ⁇ g rmIL-12 ( FIG. 7D ), or mIgG2a isotype control once a week.
  • FIG. 8 is a graph showing Kaplan-Meier survival curves of mice inoculated with CT26 tumor cells and treated with DF-mIL-12-Fc wt at a molar equivalent of 1 ⁇ g rmIL-12, DF-mIL-12-Fc si at a molar equivalent of 1 ⁇ g rmIL-12, DF-mIL-12-Fc wt at a molar equivalent of 0.1 ⁇ g rmIL-12, DF-mIL-12-Fc si at a molar equivalent of 0.1 ⁇ g rmIL-12, or mIgG2a isotype control once a week.
  • FIGS. 9A-9C are graphs showing tumor growth curves of individual mice inoculated with B16F10 melanoma cells and treated with rmIL-12 ( FIG. 9A ), DF-mIL-12-Fc wt ( FIG. 9B ), DF-mIL-12-Fc si ( FIG. 9C ), or mIgG2a isotype control once a week.
  • FIG. 10 is a graph showing Kaplan-Meier survival curves of mice inoculated with B16F10 melanoma cells and treated with rmIL-12, DF-mIL-12-Fc wt, DF-mIL-12-Fc si, or mIgG2a isotype control once a week.
  • FIGS. 11A-11D are graphs showing tumor growth curves of individual mice inoculated with B16F10 melanoma cells and treated with DF-mIL-12-Fc wt at a molar equivalent of 0.5 ⁇ g rmIL-12 ( FIG. 11A ), DF-mIL-12-Fc si at a molar equivalent of 0.5 ⁇ g rmIL-12 ( FIG. 11B ), DF-mIL-12-Fc wt at a molar equivalent of 0.1 ⁇ g rmIL-12 ( FIG. 11C ), DF-mIL-12-Fc si at a molar equivalent of 0.1 ⁇ g rmIL-12 ( FIG. 11D ), or mIgG2a isotype control once a week.
  • FIG. 12 is a graph showing Kaplan-Meier survival curves of mice inoculated with B16F10 melanoma cells and treated with DF-mIL-12-Fc wt at a molar equivalent of 0.5 ⁇ g rmIL-12, DF-mIL-12-Fc si at a molar equivalent of 0.5 ⁇ g rmIL-12, DF-mIL-12-Fc wt at a molar equivalent of 0.1 ⁇ g rmIL-12, DF-mIL-12-Fc si at a molar equivalent of 0.1 ⁇ g rmIL-12, or mIgG2a isotype control once a week.
  • FIG. 13A is a graph showing IL-12 response to treatment with DF-hIL-12-Fc si (DF IL-12-Fc) or recombinant human IL-12 (rhIL-12) using a HEK-Blue IL-12 reporter assay.
  • FIG. 13B is a graph showing IFN ⁇ production by peripheral blood mononuclear cells (PBMCs) in response to treatment with DF-hIL-12-Fc si (DF IL-12-Fc) and rhIL-12.
  • PBMCs peripheral blood mononuclear cells
  • FIG. 14 is a graph showing the relative plasma concentrations of DF-hIL-12-Fc si, rhIL-12, and IFN ⁇ in cynomolgus monkey K2 EDTA plasma following a single intravenous dose of equimolar amounts of DF-hIL-12-Fc si or wild type rhIL-12 at 10 ⁇ g/kg.
  • FIGS. 15A-15B are graphs showing the PK/PD profile of rmIL-12 ( FIG. 15A ) and DF-mIL-12-Fc si ( FIG. 15B ) in na ⁇ ve Balb/c mice.
  • FIG. 15A shows the PK/PD profile of rmIL-12 in na ⁇ ve Balb/c mice and
  • FIG. 15B shows the PK/PD profile of DF-mIL-12-Fc si in na ⁇ ve Balb/c micelL-12.
  • IL-12 and IFN ⁇ levels in serum were analyzed by ELISA.
  • FIG. 15C is a graph showing the PK/PD profile of DF-mIL-12-Fc si administered intravenously in na ⁇ ve Balb/c mice.
  • FIG. 15D is a graph showing the PK/PD profile of DF-mIL-12-Fc si administered intraperitoneally in na ⁇ ve Balb/c mice.
  • FIG. 15E is a graph showing the PK/PD profile of DF-mIL-12-Fc si administered subcutaneously in na ⁇ ve Balb/c mice. Average serum levels represent the mean ⁇ SEM.
  • FIGS. 16A-16C are graphs showing tumor growth curves of B16F10 tumor-bearing mice treated with DF-mIL-12-Fc si, anti-PD-1, or a combination thereof.
  • Mice were treated intraperitoneally with 0.5 ⁇ g isotype control or 0.5 ⁇ g DF-mIL-12-Fc si ( FIG. 16A ), isotype control or anti-PD-1 ( FIG. 16B ), and isotype control or DF-mIL-12-Fc si/anti-PD-1 ( FIG. 16C ).
  • Animals were injected once a week with DF-mIL-12-Fc si and twice weekly with anti-PD-1 as indicated above. Tumor growth was assessed for 60 days. Graphs show tumor growth curves of individual mice.
  • FIGS. 17A-17B are graphs showing survival and body weights of B16F10 tumor-bearing mice treated DF-mIL-12-Fc si, anti-PD-1, or a combination thereof. Mice were treated with isotype, DF-mIL-12-Fc si, anti-PD-1 or in combination of DF-mIL-12-Fc si and anti-PD-1. Animals were injected once a week with 0.5 ⁇ g DF-mIL-12-Fc si and twice weekly with 200 ⁇ g anti-PD-1 or isotype.
  • FIG. 17A shows Kaplan-Meier survival curves.
  • FIG. 17B shows body weights of mice as averages ⁇ standard deviation.
  • FIGS. 18A-18C are graphs showing tumor growth curves of B16F10 tumor-bearing mice treated DF-mIL-12-Fc si, mcFAE-C26.99 TriNKETs, or a combination thereof.
  • Mice were treated intraperitoneally with 150 ⁇ g isotype control or 0.5 ⁇ g DF-mIL-12-Fc si ( FIG. 18A ), isotype control or 150 ⁇ g TriNKET ( FIG. 18B ), and isotype control or DF-mIL-12-Fc si/TriNKET ( FIG. 18C ).
  • Animals were injected once a week with DF-mIL-12-Fc si and thrice weekly with TriNKET as indicated above. Tumor growth was assessed for 72 days. Graphs show tumor growth curves of individual mice.
  • FIGS. 19A-19B are graphs showing survival and body weights of B16F10 tumor-bearing mice treated with DF-mIL-12-Fc si, mcFAE-C26.99 TriNKETs, or a combination thereof. Mice were treated with isotype, DF-mIL-12-Fc si, TriNKET, or a combination of DF-mIL-12-Fc si and TriNKET. Animals were injected once a week with 0.5 ⁇ g DF-mIL-12-Fc si and thrice weekly with 150 ⁇ g TriNKET or isotype.
  • FIG. 19A shows Kaplan-Meier survival curves.
  • FIG. 19B shows body weights of mice as averages+standard deviation.
  • FIG. 21A is a graph showing tumor growth curves of individual mice inoculated with CT26 tumor cells and administered a single dose of DF-mIL-12-Fc si or mIgG2a isotype.
  • FIG. 21B is a graph showing body weights ⁇ standard deviation of mice inoculated with CT26 tumor cells and administered a weekly dose of DF-mIL-12-Fc si, mIgG2a isotype, or rmIL-12.
  • FIG. 21C is a graph showing tumor growth curves of re-challenged individual mice that were either na ⁇ ve or complete responders (CR) when previously administered a single dose of DF-mIL-12-Fc si in a CT26 tumor model.
  • FIGS. 22A-22B are graphs showing tumor growth curves of individual mice inoculated with CT26 tumor cells and administered a weekly dose of DF-mIL-12-Fc si or mIgG2a isotype either intraperitoneally (IP)( FIG. 22A ) or subcutaneously (SC) ( FIG. 22B ).
  • IP intraperitoneally
  • SC subcutaneously
  • FIG. 23 is a graph showing tumor growth curves of individual mice inoculated with B16F10 melanoma cells and administered a single dose of DF-mIL-12-Fc si or mIgG2a isotype.
  • FIGS. 24A-24B are graphs showing tumor growth curves of individual mice inoculated with B16F10 melanoma cells and administered a weekly dose of DF-mIL-12-Fc si or mIgG2a isotype either intraperitoneally (IP) ( FIG. 24A ) or subcutaneously (SC) ( FIG. 24B ).
  • IP intraperitoneally
  • SC subcutaneously
  • FIGS. 25A-25B are graphs showing tumor growth curves of individual mice inoculated with CT26 tumor cells and administered a single dose ( FIG. 25A ) or once weekly dose ( FIG. 25B ) of DF-mIL-12-Fc si or mIgG2A isotype intraperitoneally at a molar equivalent of 1 ⁇ g of rmIL-12.
  • FIGS. 26A-26B are graphs showing tumor growth curves of individual mice inoculated with CT26 tumor cells and administered a once weekly dose of DF-mIL-12-Fc si subcutaneously.
  • FIG. 26A is a graph showing tumor growth curves of individual mice inoculated with CT26-Tyrpl tumor cells and treated once (weekly) with either 2 ⁇ g mIgG2a isotype control or 1 ⁇ g DF-mIL-12-Fc si.
  • FIG. 26B is a graph showing tumor growth curves of individual mice inoculated with CT26-Tyrpl tumor cells and treated once (weekly) with either 2 ⁇ g mIgG2a isotype control or 2 ⁇ g DF-mIL-12-Fc si.
  • FIGS. 27A-27C are graphs showing IFN ⁇ ( FIG. 27A ), CXCL9 ( FIG. 27B ), and CXCL10 ( FIG. 27C ) levels in blood (left) and tumor (right) samples at 72 hours following a single dose of DF-mIL-12-Fc si in C57BL/6 mice bearing B16F10 tumors.
  • FIGS. 28A-28C are line graphs showing pharmacokinetics of DF-hIL-12-Fc si in cynomolgus monkeys treated with a single subcutaneous dose of 1 ⁇ g/kg ( FIG. 28A ), 2 ⁇ g/kg ( FIG. 28B ), or 4 ⁇ g/kg ( FIG. 28C ) of DF-hIL-12-Fc si. 2240, 2241, 2740, 2741 denote individual cynomolgus monkey subjects.
  • FIGS. 29A-29F are line graphs showing concentrations of IFN ⁇ and IP10/CXCL10 in cynomolgus monkeys treated with a single subcutaneous dose of DF-hIL-12-Fc si.
  • FIGS. 29A, 29C and 29E show IFN ⁇ concentrations/levels of expression in cynomolgus monkeys treated with 1 ⁇ g/kg, 2 ⁇ g/kg, and 4 ⁇ g/kg of DF-hIL-12-Fc si, respectively.
  • FIGS. 29A, 29C and 29E show IFN ⁇ concentrations/levels of expression in cynomolgus monkeys treated with 1 ⁇ g/kg, 2 ⁇ g/kg, and 4 ⁇ g/kg of DF-hIL-12-Fc si, respectively.
  • 29B, 29D and 29F show IP10/CXCL10 concentrations/levels of expression in cynomolgus monkeys treated with 1 ⁇ g/kg, 2 ⁇ g/kg, and 4 ⁇ g/kg of DF-hIL-12-Fc si, respectively.
  • 3240, 3241, 3740, 3741 denote individual cynomolgus monkey subjects.
  • FIG. 30 is a graph showing tumor growth curves of individual mice inoculated with breast cancer cells and administered a weekly dose of a monotherapy (isotype control, DF-mIL-12-Fc si, Doxil® (chemotherapy), or irradiated with 10 Gy) or combination therapy (DF-mIL-12-Fc si in combination with Doxil® or radiation).
  • a monotherapy isotype control, DF-mIL-12-Fc si, Doxil® (chemotherapy), or irradiated with 10 Gy
  • combination therapy DF-mIL-12-Fc si in combination with Doxil® or radiation
  • FIG. 31A is a graph showing tumor growth curves of individual mice inoculated with CT26-Tyrpl tumor cells and treated (bi-weekly) either with isotype control or anti-PD-1 antibody.
  • FIG. 31B is a graph showing tumor growth curve of Balb/c mice inoculated with CT26-Tyrpl tumor cells and treated (bi-weekly) either with isotype control or anti-PD-1 antibody.
  • FIG. 31B also shows tumor growth curves of individual mice previously treated with anti-PD-1 antibody that were administered anti-PD-1 antibody (bi-weekly) along with weekly treatment of 1 ⁇ g of DF-mIL-12-Fc si.
  • FIG. 32A is a graph showing tumor growth curves of treated (Tr) tumors inindividual mice inoculated with CT26-Tyrpl tumor cells and intratumorally treated once (weekly) with either isotype control or DF-mIL-12-Fc si.
  • FIG. 32B is a graph showing tumor growth curves of non-treated (NT) CT26-Tyrpl tumors in the individual mice described in FIG. 32A .
  • FIG. 33A is a graph showing tumor growth curves of individual mice inoculated with CT26-Tyrpl tumor cells and treated once with either 2 ⁇ g mIgG2a isotype control or 2 ⁇ g DF-mIL-12-Fc si.
  • FIG. 33B is a graph showing average tumor growth curves of individual mice inoculated with CT26-Tyrpl tumor cells and treated with 2 ⁇ g mIgG2a isotype control, 1 ⁇ g DF-mIL-12-Fc si (weekly administration), 2 ⁇ g DF-mIL-12-Fc si (weekly administration), or 2 ⁇ g DF-mIL-12-Fc si (once)
  • FIG. 34A is a graph showing IFN ⁇ production of PHA-stimulated PBMCs treated with DF hIL-12-Fc-si having L234A, L235A, and P329A mutations (LALAPA), or L234A, L235A, P329G mutations (LALAPG).
  • FIG. 34B shows flow cytometry histograms of fluorophore-conjugated hIgG1 binding to THP-1 cells in the presence or absence of DF hIL-12-Fc-si having LALAPA mutations, or LALAPG mutations.
  • the invention provides improvements on heterodimeric Fc-fused proteins, pharmaceutical compositions comprising such proteins, and therapeutic methods using such proteins and pharmaceutical compositions, including for the treatment of cancer.
  • the terms “subject” and “patient” refer to an organism to be treated by the methods and compositions described herein. Such organisms preferably include, but are not limited to, mammals (e.g., murines, simians, equines, bovines, porcines, canines, felines, and the like), and more preferably include humans.
  • the term “effective amount” refers to the amount of a compound (e.g., a compound of the present invention) sufficient to effect beneficial or desired results (e.g., a desired prophylactic or therapeutic effect).
  • An effective amount can be administered in one or more administration(s), application(s) or dosage(s) and is not intended to be limited to a particular formulation or administration route.
  • the term “treating” includes any effect, e.g., lessening, reducing, modulating, ameliorating or eliminating, that results in the improvement of the condition, disease, disorder, and the like, or ameliorating a symptom thereof.
  • composition refers to the combination of an active agent with a carrier, inert or active, making the composition especially suitable for diagnostic or therapeutic use in vivo or ex vivo.
  • the term “pharmaceutically acceptable carrier” refers to any of the standard pharmaceutical carriers, such as a phosphate buffered saline solution, water, emulsions (e.g., such as an oil/water or water/oil emulsions), and various types of wetting agents.
  • the compositions also can include stabilizers and preservatives.
  • stabilizers and adjuvants see e.g., Martin, Remington's Pharmaceutical Sciences, 15th Ed., Mack Publ. Co., Easton, Pa. [1975].
  • the term “pharmaceutically acceptable salt” refers to any pharmaceutically acceptable salt (e.g., acid or base) of a compound of the present invention which, upon administration to a subject, is capable of providing a compound of this invention or an active metabolite or residue thereof.
  • salts of the compounds of the present invention may be derived from inorganic or organic acids and bases.
  • Exemplary acids include, but are not limited to, hydrochloric, hydrobromic, sulfuric, nitric, perchloric, fumaric, maleic, phosphoric, glycolic, lactic, salicylic, succinic, toluene-p-sulfonic, tartaric, acetic, citric, methanesulfonic, ethanesulfonic, formic, benzoic, malonic, naphthalene-2-sulfonic, benzenesulfonic acid, and the like.
  • Other acids such as oxalic, while not in themselves pharmaceutically acceptable, may be employed in the preparation of salts useful as intermediates in obtaining the compounds of the invention and their pharmaceutically acceptable acid addition salts.
  • Exemplary bases include, but are not limited to, alkali metal (e.g., sodium) hydroxides, alkaline earth metal (e.g., magnesium) hydroxides, ammonia, and compounds of formula NW4 + , wherein W is C 1-4 alkyl, and the like.
  • alkali metal e.g., sodium
  • alkaline earth metal e.g., magnesium
  • W is C 1-4 alkyl
  • Exemplary salts include, but are not limited to: acetate, adipate, alginate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, citrate, camphorate, camphorsulfonate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, flucoheptanoate, glycerophosphate, hemisulfate, heptanoate, hexanoate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxyethanesulfonate, lactate, maleate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, oxalate, palmoate, pectinate, persulfate, phenylpropionate, picrate, pivalate, propionate, succinate, tartrate, thiocyanate
  • salts of the compounds of the present invention are contemplated as being pharmaceutically acceptable.
  • salts of acids and bases that are non-pharmaceutically acceptable may also find use, for example, in the preparation or purification of a pharmaceutically acceptable compound.
  • compositions are described as having, including, or comprising specific components, or where processes and methods are described as having, including, or comprising specific steps, it is contemplated that, additionally, there are compositions of the present invention that consist essentially of, or consist of, the recited components, and that there are processes and methods according to the present invention that consist essentially of, or consist of, the recited processing steps.
  • compositions specifying a percentage are by weight unless otherwise specified. Further, if a variable is not accompanied by a definition, then the previous definition of the variable controls.
  • the present invention provides Fc-fused protein constructs comprising the amino acid sequences of a multisubunit protein. These fusion protein constructs can exhibit a higher serum half-life compared to a native/natural multisubunit protein, improved yield during production, enhanced stability during storage, and/or improved efficacy when used as a therapeutic.
  • the present invention provides a heterodimeric IgG1 Fc-fused protein comprising: a first polypeptide comprising a first antibody IgG1 Fc domain polypeptide and a second polypeptide comprising a second antibody IgG1 Fc domain polypeptide bound to the first antibody Fc domain, in which the first polypeptide further comprises a first subunit of a multisubunit protein fused by a linker comprising amino acid sequence PKSSDKTHTCPPCPAPEX 1 X 2 GX 3 (SEQ ID NO:237) or EPKSSDKTHTCPPCPAPEX 1 X 2 GX 3 (SEQ ID NO:6) to the first antibody Fc domain polypeptide, wherein X 1 represents L or A, X 2 represents L, E, or A, and X 3 represents A or G; a second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide and the subunits of the multisubunit protein are bound to each other; when X 1 represents L and
  • the linker connecting the a first subunit of a multisubunit protein to the first antibody Fc domain polypeptide consists of amino acid sequence PKSSDKTHTCPPCPAPEX 1 X 2 GX 3 (SEQ ID NO:237) or EPKSSDKTHTCPPCPAPEX 1 X 2 GX 3 (SEQ ID NO:6), wherein X 1 represents L or A, X 2 represents L, E, or A, and X 3 represents A or G.
  • the linker connecting the first subunit of a multisubunit protein to the first antibody Fc domain polypeptide further comprises a spacer peptide.
  • the linker comprises a sequence of SEQ ID NO:237 or SEQ ID NO:6, and a spacer peptide.
  • the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker that comprises a sequence PKSSDKTHTCPPCPAPEX 1 X 2 GX 3 (SEQ ID NO:237) or EPKSSDKTHTCPPCPAPEX 1 X 2 GX 3 (SEQ ID NO:6), wherein X 1 represents L or A, X 2 represents L, E, or A, and X 3 represents A or G, and a spacer peptide.
  • a linker that comprises a sequence PKSSDKTHTCPPCPAPEX 1 X 2 GX 3 (SEQ ID NO:237) or EPKSSDKTHTCPPCPAPEX 1 X 2 GX 3 (SEQ ID NO:6), wherein X 1 represents L or A, X 2 represents L, E, or A, and X 3 represents A or G, and a spacer peptide.
  • the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker that consists of the amino acid sequence PKSSDKTHTCPPCPAPEX 1 X 2 GX 3 (SEQ ID NO:237) or EPKSSDKTHTCPPCPAPEX 1 X 2 GX 3 (SEQ ID NO:6), wherein X 1 represents L or A, X 2 represents L, E, or A, and X 3 represents A or G.
  • the amino acid sequence of the linker connecting the second, different subunit of the multisubunit protein to the second antibody Fc domain polypeptide is identical to the amino acid sequence of the linker connecting the subunit of the multisubunit protein to the first antibody Fc domain polypeptide.
  • the spacer peptide comprises an amino acid sequence set forth in any one of SEQ ID NOs:107-120.
  • the spacer peptide consists of an amino acid sequence set forth in any one of SEQ ID NOs:107-120.
  • the linker connecting the subunit of a multisubunit protein to the first antibody Fc domain polypeptide consists of, or consists essentially of, a spacer peptide disclosed herein and a peptide having the sequence of SEQ ID NO:237 or SEQ ID NO:6.
  • the linker connecting the second, different subunit of the multisubunit protein to the second antibody Fc domain polypeptide consists of, or consists essentially of, a spacer peptide disclosed herein and a peptide having the sequence of SEQ ID NO:237 or SEQ ID NO:6.
  • the spacer peptide is N-terminal to the either or both of the linkers.
  • the linker connecting the first subunit of the multisubunit protein to the first antibody Fc domain polypeptide comprises amino acid sequence PKSSDKTHTCPPCPAPEAAGG (SEQ ID NO:239) or EPKSSDKTHTCPPCPAPEAAGG (SEQ ID NO:9). In some embodiments, within the heterodimeric Fc-fused protein, the linker connecting the first subunit of the multisubunit protein to the first antibody Fc domain polypeptide consists of amino acid sequence PKSSDKTHTCPPCPAPEAAGG (SEQ ID NO:239) or EPKSSDKTHTCPPCPAPEAAGG (SEQ ID NO:9).
  • the linker connecting the first subunit of the multisubunit protein to the first antibody Fc domain polypeptide comprises amino acid sequence PKSSDKTHTCPPCPAPEAAGG (SEQ ID NO:239). In some embodiments, within the heterodimeric Fc-fused protein, the linker connecting the first subunit of the multisubunit protein to the first antibody Fc domain polypeptide consists of amino acid sequence PKSSDKTHTCPPCPAPEAAGG (SEQ ID NO:239).
  • the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker comprising amino acid sequence GGGGSGGGGSGGGGSEPKSSDKTHTCPPCPAPEAAGG (SEQ ID NO:10) or GGGGSGGGGSGGGGSPKSSDKTHTCPPCPAPEAAGG (SEQ ID NO:244).
  • the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker consisting of amino acid sequence GGGGSGGGGSGGGGSEPKSSDKTHTCPPCPAPEAAGG (SEQ ID NO:10) or GGGGSGGGGSGGGGSPKSSDKTHTCPPCPAPEAAGG (SEQ ID NO:244).
  • the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker comprising amino acid sequence GGGGSGGGGSGGGGSEPKSSDKTHTCPPCPAPEAAGG (SEQ ID NO:10).
  • the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker consisting of amino acid sequence GGGGSGGGGSGGGGSEPKSSDKTHTCPPCPAPEAAGG (SEQ ID NO:10).
  • the linker connecting the subunit of the multisubunit protein to the first antibody Fc domain polypeptide comprises amino acid sequence PKSSDKTHTCPPCPAPELLGG (SEQ ID NO:238) or EPKSSDKTHTCPPCPAPELLGG (SEQ ID NO:7).
  • the linker fusing the first subunit of the multisubunit protein to the first antibody Fc domain polypeptide consists of amino acid sequence PKSSDKTHTCPPCPAPELLGG (SEQ ID NO:238) or EPKSSDKTHTCPPCPAPELLGG (SEQ ID NO:7).
  • the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker comprising amino acid sequence GGGGSGGGGSGGGGSEPKSSDKTHTCPPCPAPELLGG (SEQ ID NO:8) or GGGGSGGGGSGGGGSPKSSDKTHTCPPCPAPELLGG (SEQ ID NO:241).
  • the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker consisting of amino acid sequence GGGGSGGGGSGGGGSEPKSSDKTHTCPPCPAPELLGG (SEQ ID NO:8) or GGGGSGGGGSGGGGSPKSSDKTHTCPPCPAPELLGG (SEQ ID NO:241).
  • the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker comprising amino acid sequence GGGGSEPKSSDKTHTCPPCPAPELLGG (SEQ ID NO:15) or GGGGSPKSSDKTHTCPPCPAPELLGG (SEQ ID NO:242).
  • the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker consisting of amino acid sequence GGGGSEPKSSDKTHTCPPCPAPELLGG (SEQ ID NO:15) or GGGGSPKSSDKTHTCPPCPAPELLGG (SEQ ID NO:242).
  • the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker comprising amino acid sequence GGGGSGGGGSEPKSSDKTHTCPPCPAPELLGG (SEQ ID NO:16) or GGGGSGGGGSPKSSDKTHTCPPCPAPELLGG (SEQ ID NO:243).
  • the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker consisting of amino acid sequence GGGGSGGGGSEPKSSDKTHTCPPCPAPELLGG (SEQ ID NO:16) or GGGGSGGGGSPKSSDKTHTCPPCPAPELLGG (SEQ ID NO:243).
  • the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker comprising amino acid sequence GGGGSEPKSSDKTHTCPPCPAPEAAGG (SEQ ID NO:65) or GGGGSPKSSDKTHTCPPCPAPEAAGG (SEQ ID NO:245).
  • the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker consisting of amino acid sequence GGGGSEPKSSDKTHTCPPCPAPEAAGG (SEQ ID NO:65) or GGGGSPKSSDKTHTCPPCPAPEAAGG (SEQ ID NO:245).
  • the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker comprising amino acid sequence GGGGSGGGGSEPKSSDKTHTCPPCPAPEAAGG (SEQ ID NO:66) or GGGGSGGGGSPKSSDKTHTCPPCPAPEAAGG (SEQ ID NO:246).
  • the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker consisting of amino acid sequence GGGGSGGGGSEPKSSDKTHTCPPCPAPEAAGG (SEQ ID NO:66) or GGGGSGGGGSPKSSDKTHTCPPCPAPEAAGG (SEQ ID NO:246).
  • the linker connecting the first subunit of the multisubunit protein to the first antibody Fc domain polypeptide comprises amino acid sequence EPKSSDKTHTCPPCPAPEAEGA (SEQ ID NO:11) or PKSSDKTHTCPPCPAPEAEGA (SEQ ID NO:240).
  • the linker fusing the first subunit of the multisubunit protein to the first antibody Fc domain polypeptide consists of amino acid sequence EPKSSDKTHTCPPCPAPEAEGA (SEQ ID NO:11) or PKSSDKTHTCPPCPAPEAEGA (SEQ ID NO:240).
  • the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker comprising amino acid sequence GGGGSGGGGSGGGGSEPKSSDKTHTCPPCPAPEAEGA (SEQ ID NO:12) or GGGGSGGGGSGGGGSPKSSDKTHTCPPCPAPEAEGA (SEQ ID NO:247).
  • the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker consisting of amino acid sequence GGGGSGGGGSGGGGSEPKSSDKTHTCPPCPAPEAEGA (SEQ ID NO:12) or GGGGSGGGGSGGGGSPKSSDKTHTCPPCPAPEAEGA (SEQ ID NO:247).
  • the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker comprising amino acid sequence GGGGSEPKSSDKTHTCPPCPAPEAEGA (SEQ ID NO:67) or GGGGSPKSSDKTHTCPPCPAPEAEGA (SEQ ID NO:248).
  • the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker consisting of amino acid sequence GGGGSEPKSSDKTHTCPPCPAPEAEGA (SEQ ID NO:67) or GGGGSPKSSDKTHTCPPCPAPEAEGA (SEQ ID NO:248).
  • the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker comprising amino acid sequence GGGGSGGGGSEPKSSDKTHTCPPCPAPEAEGA (SEQ ID NO:68) or GGGGSGGGGSPKSSDKTHTCPPCPAPEAEGA (SEQ ID NO:249).
  • the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker consisting of amino acid sequence GGGGSGGGGSEPKSSDKTHTCPPCPAPEAEGA (SEQ ID NO:68) or GGGGSGGGGSPKSSDKTHTCPPCPAPEAEGA (SEQ ID NO:249).
  • the Fc domain polypeptide is that of an IgG1 Fc.
  • a protein of the current invention includes, a first antibody Fc domain polypeptide and a second antibody Fc domain polypeptide, which are both mutated IgG1 Fc domain polypeptides that promote heterodimerization with each other.
  • the Fc domain can comprise an amino acid sequence at least 90% identical to amino acids 234-332 of a human IgG1 antibody, and differ at one or more position(s) selected from the group consisting of Q347, Y349, L351, 5354, E356, E357, K360, Q362, 5364, T366, L368, K370, N390, K392, T394, D399, 5400, D401, F405, Y407, K409, T411, and K439.
  • the antibody constant domain can comprise an amino acid sequence at least 90% identical to amino acids 234-332 of a human IgG1 antibody, and differ by one or more substitution(s) selected from the group consisting of Q347E, Q347R, Y349S, Y349K, Y349T, Y349D, Y349E, Y349C, L351K, L351D, L351Y, S354C, E356K, E357Q, E357L, E357W, K360E, K360W, Q362E, S364K, S364E, S364H, S364D, T366V, T366I, T366L, T366M, T366K, T366W, T366S, L368E, L368A, L368D, K370S, N390D, N390E, K392L, K392M, K392V, K392F
  • the first antibody IgG1 Fc domain polypeptide includes one or more mutation(s) selected from K360E and K409W
  • the second antibody IgG1 Fc domain polypeptide includes one or more mutation(s) selected from Q347R, D399V, and F405T
  • the first antibody IgG1 Fc domain polypeptide includes one or more mutation(s) selected from Q347R, D399V, and F405T
  • the second antibody IgG1 Fc domain polypeptide includes one or more mutation(s) selected from K360E and K409W.
  • the first antibody IgG1 Fc domain polypeptide includes mutations K360E and K409W
  • the second antibody IgG1 Fc domain polypeptide includes mutations Q347R, D399V, and F405T
  • the first antibody IgG1 Fc domain polypeptide includes mutations Q347R, D399V, and F405T
  • the second antibody IgG1 Fc domain polypeptide includes mutations K360E and K409W.
  • a heterodimeric Fc-fused protein of the present invention with an IgG1 Fc includes one or more mutation(s) to reduce binding to an Fc ⁇ R (e.g., Fc ⁇ RI, Fc ⁇ RIIA, Fc ⁇ RIIB, Fc ⁇ RIIIA, or Fc ⁇ RIIIB) or a complement component (e.g., C1q) in the first and/or second polypeptides.
  • Fc ⁇ R e.g., Fc ⁇ RI, Fc ⁇ RIIA, Fc ⁇ RIIB, Fc ⁇ RIIIA, or Fc ⁇ RIIIB
  • complement component e.g., C1q
  • a protein of the present disclosure includes LALA (L234A and L235A) mutations, LALAPA (L234A, L235A, and P329A) mutations, LALAPG (L234A, L235A, and P329G) mutations, or LALEGAASPS (L234A, L235E, G237A, A330S, and P331S) mutations.
  • a heterodimeric Fc-fused protein according to the invention includes a first antibody IgG4 or IgG1 Fc domain polypeptide and the second antibody IgG4 or IgG1 Fc domain polypeptide each containing the mutation P329G or P329A.
  • a heterodimeric Fc-fused protein according to the invention comprises a first antibody IgG4 or IgG1 Fc domain polypeptide and a second antibody IgG4 or IgG1 Fc domain polypeptide each comprising the mutation P329A.
  • the first IgG1 antibody Fc domain polypeptide and the second, different IgG1 antibody Fc domain polypeptide each contain a mutation selected from A330S and P331S. In some embodiments, the first IgG1 antibody Fc domain polypeptide and the second, different IgG1 antibody Fc domain polypeptide each contain the mutations A330S and P331S.
  • an additional disulfide bond between IgG1 Fc monomers is introduced, which improves the stability of the heterodimer.
  • the first antibody Fc domain polypeptide fused to the first subunit of a multisubunit protein includes a Y349C substitution in the CH3 domain, which forms a disulfide bond with an S354C substitution on the second antibody Fc domain polypeptide fused to the second, different subunit of a multisubunit protein.
  • the first antibody Fc domain polypeptide fused to the first subunit of a multisubunit protein includes an S354C substitution in the CH3 domain, which forms a disulfide bond with a Y349C substitution on the second antibody Fc domain polypeptide fused to the second, different subunit of a multisubunit protein.
  • IgG1 antibody Fc domain polypeptides provided in Table 2 below can be employed in combination with any of the IgG1 hinge sequences (which, in the current invention, is part or the entirety of a linker connecting the protein sequence of the first subunit of the multisubunit protein to the first IgG1 antibody Fc domain polypeptide, or a linker connecting the additional subunit to the second, different IgG1 antibody Fc domain polypeptide) provided in Table 1 below.
  • Exemplary IgG1 hinge-Fc domain polypeptides are provided in Table 3 below.
  • the first and second polypeptides of the Fc-fused protein comprise the amino acid sequences of SEQ ID NOs: 212 and 212; 213 and 214; 215 and 216; 217 and 218; 214 and 213; 216 and 215; or 218 and 217, respectively.
  • the first and second polypeptides of the Fc-fused protein comprise the amino acid sequences of SEQ ID NOs:228 and 228; 229 and 230; 231 and 232; 233 and 234; 235 and 236; 230 and 229; 232 and 231; 234 and 233; 236 and 235; 228 and 250; 250 and 228; 250 and 250; 229 and 252; 252 and 229; 251 and 230; 230 and 251; 253 and 232; 232 and 253; 231 and 254; 254 and 231; 255 and 234; 234 and 255; 233 and 256; 256 and 233; 257 and 236; 236 and 257; 258 and 235; or 235 and 258, respectively.
  • the current invention provides an improvement on a multisubunit protein.
  • the present invention provides a heterodimeric IgG4 Fc-fused protein comprising: a first polypeptide comprising a first antibody IgG4 Fc domain polypeptide and a second polypeptide comprising a second, different antibody IgG4 Fc domain polypeptide bound to the first antibody Fc domain polypeptide, in which the first polypeptide further comprises a first subunit of a multisubunit protein fused by a linker comprising amino acid sequence RVESKYGPPCPPCPAPEFXGG (SEQ ID NO:1) to the first antibody IgG4 Fc domain polypeptide, in which X represents L or E; a second, different subunit of the multisubunit protein is fused to the second antibody IgG4 Fc domain polypeptide and the subunits of the multisubunit protein are bound to each other; the first antibody Fc domain polypeptide and the second antibody IgG4 Fc domain polypeptide each contain different mutations
  • the linker connecting the a first subunit of a multisubunit protein to the first antibody Fc domain polypeptide consists of amino acid sequence RVESKYGPPCPPCPAPEFXGG, wherein X represents L or E (SEQ ID NO:1).
  • the linker connecting the protein sequence of a first subunit of a multisubunit protein to the first antibody Fc domain polypeptide further comprises a spacer peptide.
  • the linker comprises a sequence of SEQ ID NO:1 and a spacer peptide.
  • the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker that comprises a sequence RVESKYGPPCPPCPAPEFXGG, wherein X represents L or E (SEQ ID NO:1), and a spacer peptide.
  • the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker that consists of the amino acid sequence RVESKYGPPCPPCPAPEFXGG, wherein X represents L or E (SEQ ID NO:1).
  • amino acid sequence of the linker connecting the second, different subunit of the multisubunit protein to the second antibody Fc domain polypeptide is identical to the amino acid sequence of the linker connecting the subunit of the multisubunit protein to the first antibody Fc domain polypeptide.
  • the spacer peptide comprises an amino acid sequence set forth in any one of SEQ ID NOs: 107-120.
  • the spacer peptide consists of an amino acid sequence set forth in any one of SEQ ID NOs: 107-120.
  • the linker connecting the subunit of the multisubunit protein to the first antibody Fc domain polypeptide consists of, or consists essentially of, a spacer peptide disclosed herein and SEQ ID NO:1.
  • the linker consists of, or consists essentially of, a spacer peptide disclosed herein and SEQ ID NO:1.
  • the spacer peptide is N-terminal to the first linker and/or the second linker.
  • the linker connecting the subunit of the multisubunit protein to the first antibody Fc domain polypeptide comprises amino acid sequence RVESKYGPPCPPCPAPEFLGG (SEQ ID NO:2).
  • the linker fusing the first subunit of the multisubunit protein to the first antibody Fc domain polypeptide consists of amino acid sequence RVESKYGPPCPPCPAPEFLGG (SEQ ID NO:2).
  • the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker comprising amino acid sequence GGGGSGGGGSGGGGSRVESKYGPPCPPCPAPEFLGG (SEQ ID NO:3). In some embodiments, the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker consisting of amino acid sequence GGGGSGGGGSGGGGSRVESKYGPPCPPCPAPEFLGG (SEQ ID NO:3).
  • the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker comprising amino acid sequence GGGGSRVESKYGPPCPPCPAPEFLGG (SEQ ID NO:13). In some embodiments, the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker consisting of amino acid sequence GGGGSRVESKYGPPCPPCPAPEFLGG (SEQ ID NO:13).
  • the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker comprising amino acid sequence GGGGSGGGGSRVESKYGPPCPPCPAPEFLGG (SEQ ID NO:14). In some embodiments, the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker consisting of amino acid sequence GGGGSGGGGSRVESKYGPPCPPCPAPEFLGG (SEQ ID NO:14).
  • the linker connecting the first subunit of the multisubunit protein to the first antibody Fc domain polypeptide comprises amino acid sequence RVESKYGPPCPPCPAPEFEGG (SEQ ID NO:4).
  • the linker fusing the first subunit of the multisubunit protein to the first antibody Fc domain polypeptide consists of amino acid sequence RVESKYGPPCPPCPAPEFEGG (SEQ ID NO:4).
  • the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker comprising amino acid sequence GGGGSGGGGSGGGGSRVESKYGPPCPPCPAPEFEGG (SEQ ID NO:5). In some embodiments, the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker consisting of amino acid sequence GGGGSGGGGSGGGGSRVESKYGPPCPPCPAPEFEGG (SEQ ID NO:5).
  • the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker comprising amino acid sequence GGGGSRVESKYGPPCPPCPAPEFEGG (SEQ ID NO:63). In some embodiments, the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker consisting of amino acid sequence GGGGSRVESKYGPPCPPCPAPEFEGG (SEQ ID NO:63).
  • the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker comprising amino acid sequence GGGGSGGGGSRVESKYGPPCPPCPAPEFEGG (SEQ ID NO:64). In some embodiments, the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a linker consisting of amino acid sequence GGGGSGGGGSRVESKYGPPCPPCPAPEFEGG (SEQ ID NO:64).
  • the Fc domain polypeptide is that of an IgG4 Fc.
  • IgG4 is an unstable dimer that can undergo a Fab-arm exchange and pair with other IgG4 antibodies in the body.
  • a S228P mutation is introduced within the hinge (which, in the current invention, is part or the entirety of a linker connecting the first subunit of the multisubunit protein to the first IgG4 antibody Fc domain polypeptide, or a linker connecting the additional subunit to the second, different IgG4 antibody Fc domain polypeptide), which increases the stability of the hinge region and reduces the chance for Fab-arm exchange.
  • an additional disulfide bond between Fc domain polypeptide monomers is introduced, which improves the stability of the heterodimer.
  • the first antibody Fc domain polypeptide linked to the first subunit of the multisubunit protein includes a Y349C substitution in the CH3 domain, which forms a disulfide bond with an S354C substitution on the second antibody Fc domain polypeptide linked to the second, different subunit of the multisubunit proteinsecond antibody Fc domain polypeptide.
  • the first antibody Fc domain polypeptide linked to the first subunit of the multisubunit protein includes an S354C substitution in the CH3 domain, which forms a disulfide bond with a Y349C substitution on the second antibody Fc domain polypeptide linked to the second, different subunit of the multisubunit protein.
  • a protein of the current invention includes, a first antibody Fc domain polypeptide and a second antibody Fc domain polypeptide, which are both mutated IgG4 Fc domain polypeptides that promote heterodimerization with each other.
  • the first antibody IgG4 Fc domain polypeptide includes one or more mutation(s) selected from K360E, K370E, and R409W
  • the second antibody IgG4 Fc domain polypeptide includes one or more mutation(s) selected from E357N, Q347R, D399V, and F405T.
  • the first antibody IgG4 Fc domain polypeptide includes mutations K370E and R409W
  • the second antibody IgG4 Fc domain polypeptide includes mutations E357N, D399V, and F405T.
  • the first antibody IgG4 Fc domain polypeptide includes mutations E357N, D399V, and F405T
  • the second antibody IgG4 Fc domain polypeptide includes mutations K370E and R409W
  • the first antibody IgG4 Fc domain polypeptide includes mutations K360E and R409W
  • the second antibody IgG4 Fc domain polypeptide includes mutations Q347R, D399V, and F405T.
  • the first antibody IgG4 Fc domain polypeptide includes mutations Q347R, D399V, and F405T
  • the second antibody IgG4 Fc domain polypeptide includes mutations K360E and R409W.
  • a heterodimeric Fc-fused protein of the present invention with an IgG4 Fc includes one or more mutation(s) to reduce binding to an Fc ⁇ R (e.g., Fc ⁇ RI, Fc ⁇ RIIA, Fc ⁇ RIIB, Fc ⁇ RIIIA, or Fc ⁇ RIIIB) or a complement component (e.g., C1q) in the first and/or second polypeptide(s).
  • Fc ⁇ R e.g., Fc ⁇ RI, Fc ⁇ RIIA, Fc ⁇ RIIB, Fc ⁇ RIIIA, or Fc ⁇ RIIIB
  • complement component e.g., C1q
  • a protein of the present disclosure includes SPLE (S228P and L235E) mutations, SPLEPA (S228P, L235E, and P329A) mutations, or SPLEPG (S228P, L235E, and P329G) mutations.
  • any of the IgG4 antibody Fc domain polypeptides provided in Table 2 can be employed in combination with any of the IgG4 hinge sequences (which, in the current invention, is part or the entirety of a linker connecting the first subunit of the multisubunit protein to the first IgG4 antibody Fc domain polypeptide, or a linker connecting the second, different subunit of the multisubunit protein to the second, different IgG4 antibody Fc domain polypeptide) provided in Table 1.
  • Exemplary IgG4 hinge-Fc domain polypeptides are provided in Table 3.
  • the first and second polypeptides of the Fc-fused protein comprise the amino acid sequences of SEQ ID NOs:205 and 205; 206 and 207; 208 and 209; 210 and 211; 207 and 206; 209 and 208; or 211 and 210, respectively.
  • the first and second polypeptides of the Fc-fused protein comprise the amino acid sequences of SEQ ID NOs:219 and 219; 220 and 221; 222 and 223; 224 and 225; 226 and 227; 221 and 220; 223 and 222; 225 and 224; or 227 and 226, respectively.
  • heterodimeric Fc-fused proteins of the present invention include the native heterodimer disulfide bond between the first subunit of a multisubunit protein and the second, different subunit of the multisubunit protein.
  • a heterodimeric Fc-fused protein according to the invention includes a native heterodimer disulfide bond between p35 and p40 subunits of IL-12.
  • Such a protein includes the native disulfide bond between C74 of p35 and C177 of p40.
  • heterodimeric Fc-fused proteins of the present invention include an artificial or engineered heterodimer disulfide bond between the first subunit of a multisubunit protein and the second, different subunit of the multisubunit protein.
  • a heterodimeric Fc-fused protein according to the invention includes an artificial or engineered heterodimer disulfide bond between p35 and p40 subunits of IL-12.
  • Such a protein includes an artificial or engineered disulfide bond between V185C of p35 and Y292C of p40.
  • heterodimeric Fc-fused proteins of the present invention include the native heterodimer disulfide bond between the first subunit of a multisubunit protein and the second, different subunit of the multisubunit protein, and an artificial or engineered heterodimer disulfide bond between the first subunit of a multisubunit protein and the second, different subunit of the multisubunit protein.
  • a native heterodimer disulfide bond between p35 and p40 subunits of IL-12 and includes an artificial or engineered heterodimer disulfide bond between p35 and p40 subunits of IL-12.
  • Such a protein includes the native disulfide bond between C74 of p35 and C177 of p40, and an artificial or engineered disulfide bond between V185C of p35 and Y292C of p40.
  • a heterodimeric Fc-fused protein according to the invention includes p35 of IL-12 in which the native C74 is mutated to serine, and a p40 of IL-12 in which the native C177 is mutated to serine, thereby removing the native disulfide bond between p35 and p40 subunits of IL-12.
  • p35 of IL-12 in which the native C74 is mutated to serine
  • a p40 of IL-12 in which the native C177 is mutated to serine
  • Exemplary heterodimeric Fc-fused proteins of the present invention are constructed with any one of the IgG1 or IgG4 Fc variant sequences and any one of the corresponding linker sequences described in the Tables 1-2 below.
  • the fusion protein constructs of the present invention can confer a higher serum half-life compared to a native/natural multisubunit protein, improve yield of the proteins during production, enhance stability during storage, and/or improve efficacy when used as a therapeutic.
  • Tables 4 and 5 list amino acid sequences of exemplary protein constructs of the present invention. All mutations/substitutions in the Fc domain polypeptides are numbered according to the EU numbering system. Mutations in p35 and p40 subunits are numbered starting from the respective N-terminal amino acids of these subunits.
  • IgG4 antibody Fc variant domain polypeptides provided in Table 2 below can be employed in combination with any of the IgG4 hinge sequences provided in Table 1 below.
  • any of the IgG1 antibody Fc variant domain polypeptides provided in Table 2 below can be employed in combination with any of the IgG1 hinge sequences provided in Table 1 below.
  • Exemplary IgG1 hinge-Fc domain polypeptides are provided in Table 3 below.
  • IL-12 is a multisubunit protein including a p40 subunit and a p35 subunit.
  • the amino acid sequence of mature wild-type IL-12 p40 is amino acids 23-328 of the GenBank Accession No. NP_002178.2, set forth in SEQ ID NO:127 below.
  • the amino acid sequence of mature wild-type IL-12 p35 is amino acids 57-253 of GenBank Accession No. NP_000873.2, set forth in SEQ ID NO:128 below.
  • the numbering of amino acid residues of p40 and p35 used herein corresponds to the mature wild-type protein sequences.
  • an IL-12 p40 subunit comprises an amino acid sequence at least 90% (e.g., at least 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%) identical to SEQ ID NO:127.
  • an IL-12 p35 subunit comprises an amino acid sequence at least 90% (e.g., at least 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%) identical to SEQ ID NO:128.
  • the p40 and p35 subunits of IL-12 comprise the amino acid sequences of SEQ ID NOs: 121 and 122; 127 and 128; 201 and 202; 203 and 204; 123 and 124; or 125 and 126, respectively.
  • the first polypeptide comprises the amino acid sequence of a p40 subunit of IL-12
  • the second polypeptide comprises the amino acid sequence of a p35 subunit of IL-12.
  • the first polypeptide comprises the amino acid sequence of a p35 subunit of IL-12
  • the second polypeptide comprises the amino acid sequence of a p40 subunit of IL-12.
  • the present disclosure includes a heterodimeric Fc-fused protein comprising: a first polypeptide comprising a first antibody Fc domain polypeptide and a second polypeptide comprising a second antibody Fc domain polypeptide, wherein the first polypeptide further comprises a first subunit of IL-12 fused to the first antibody Fc domain polypeptide by a linker; and a second, different subunit of IL-12 is fused to the second antibody Fc domain polypeptide, wherein the first and second, different subunits of IL-12 are bound to each other, wherein the first antibody Fc domain polypeptide and the second antibody Fc domain polypeptide each contain different mutations promoting heterodimerization, wherein the first antibody Fc domain polypeptide and the second antibody Fc domain polypeptide are bound to each other, and wherein the first subunit of IL-12 is a p40 subunit with a Y292C substitution, and the second, different subunit of IL-12 is a p35 subunit with
  • the first subunit and second, different subunit of IL-12 can be fused to any of the antibody Fc domain polypeptides via any linkers disclosed herein to form Fc-fused proteins having sequences including but not limited to Constructs 120, 120-1, 120-2, 120-3, 120-4, 120-5, 120-6, and 120-7 as described in Table 4 and Constructs 20, 20-1, 20-2, 20-3, 20-4, 20-5, 20-6, 20-7, 20-8, and 20-9 as described in Table 5.
  • the p40 subunit of IL-12 further comprises a replacement of C177
  • the p35 subunit of IL-12 further comprises a replacement of C74.
  • C177 in the p40 subunit of IL-12 is replaced by S
  • C74 in the p35 subunit of IL-12 is replaced by S.
  • the p40 and p35 subunits of IL-12 comprise the amino acid sequences of SEQ ID NOs: 123 and 124, respectively.
  • the first subunit and second, different subunit of IL-12 can be fused to any of the antibody Fc domain polypeptides via any linkers disclosed herein to form Fc-fused proteins having sequences including but not limited to Constructs 119, 119-1, 119-2, 119-3, 119-4, 119-5, 119-6, 119-7, and 119-8 as described in Table 4, and Constructs 19, 19-1, 19-2, 19-3, 19-4, 19-5, 19-6, 19-7, 19-8, 19-9, and 19-10 as described in Table 5.
  • a heterodimeric Fc-fused protein of the present invention comprises a first polypeptide comprising the amino acid sequence of SEQ ID NO:290 and a second polypeptide comprising the amino acid sequence of SEQ ID NO:291.
  • the heterodimeric Fc-fused protein of the present invention comprising SEQ ID NO:290 and SEQ ID NO:291 comprises a Y349C mutation in the CH3 domain of the first antibody Fc domain polypeptide and a S354C mutation in the CH3 domain of the second antibody Fc domain polypeptide.
  • the heterodimeric Fc-fused protein of the present invention comprising SEQ ID NO:290 and SEQ ID NO:291 comprise different mutations in the respective Fc domain polypeptide sequences for promoting heterodimerization between the Fc domains.
  • the first polypeptide sequence comprises a first antibody Fc domain polypeptide (human IgG1) sequence comprising K360E and K409W substitutions.
  • the second polypeptide sequence comprises a second antibody Fc domain polypeptide (human IgG1) sequence comprising Q347R, D399V, and F405T substitutions.
  • the first polypeptide and second polypeptide amino acid sequences comprise one or more mutations for reducing effector functions.
  • the heterodimeric Fc-fused protein of the present invention comprises LALAPA (L234A, L235A, and P329A) mutations.
  • the p40 subunit of human IL-12 is fused to the first antibody Fc domain polypeptide by a first linker comprising a first amino acid sequence
  • the p35 subunit of human IL-12 is fused to the second antibody Fc domain polypeptide by a second linker comprising a second amino acid sequence
  • SEQ ID NO:290 is a sequence of p40 subunit of human IL-12 (underlined amino acids) fused to human IgG1 Fc domain polypeptide. Mutations are shown in bold.
  • SEQ ID NO:291 is a sequence of p35 subunit of human IL-12 (underlined amino acids) fused to human IgG1 Fc domain polypeptide. Mutations are shown in bold.
  • the first antibody Fc domain polypeptide sequence (human IgG1) in SEQ ID NO:290 includes K360E and K409W substitutions in the CH3 domain
  • the second, different Fc domain polypeptide sequence (human IgG1) in SEQ ID NO:291 includes Q347R, D399V, and F405T substitutions in the CH3 domain (Fc numbering according to the EU system).
  • the first antibody Fc domain polypeptide sequence and the second, different Fc domain polypeptide sequence (human IgG1) in SEQ ID NO:290 and SEQ ID NO:291 also include L234A, L235A, and P329A (LALAPA) mutations for reducing effector functions.
  • Exemplary spacer peptide sequences are provided in Table 7, and exemplary full length linker sequences are provided in Tables 4 and 5.
  • a first subunit of a multisubunit protein is fused via a linker to a first antibody Fc domain polypeptide (e.g., an IgG4 antibody Fc variant sequence or an IgG1 antibody Fc variant sequence, as disclosed in Table 2), in an amino-to-carboxyl direction.
  • a second, different subunit of a multisubunit protein is fused via a linker to a second antibody Fc domain polypeptide (e.g., an IgG4 antibody Fc variant sequence or an IgG1 antibody Fc variant sequence, as disclosed in Table 2), in an amino-to-carboxyl direction.
  • the first subunit of a multisubunit protein of the present invention is fused via a linker to a first antibody Fc domain sequence, wherein the linker comprises or consists of a spacer peptide L 1 and the amino acid sequence of SEQ ID NO:1, 2, 4, 6, 7, 9, 11, 237, 238, 239, or 240.
  • the second, different subunit of the multisubunit protein is fused to a second antibody Fc domain polypeptide via a linker, wherein the linker comprises or consists of a spacer peptide L 2 and the amino acid sequence of SEQ ID NO:1, 2, 4, 6, 7, 9, 11, 237, 238, 239, or 240.
  • L 1 and L 2 are peptide linkers, for example, L 1 and/or L 2 include(s) 4-50 amino acid residues. In certain embodiments, L 1 consists of 4-50 amino acid residues. In certain embodiments, L 1 consists of 4-20 amino acid residues. In certain embodiments, L 2 consists of 4-50 amino acid residues. In certain embodiments, L 2 consists of about 4-20 amino acid residues. In certain embodiments, L 1 and L 2 each independently consist of about 4-50 amino acid residues. In certain embodiments, L 1 and L 2 each independently consist of 4-20 amino acid residues.
  • L 1 and L 2 have an optimized length and/or amino acid composition. In some embodiments, L 1 and L 2 are of the same length and have the same amino acid composition. In other embodiments, L 1 and L 2 are different.
  • L 1 is of equal number of amino acids to Lz; in certain embodiments L 1 is longer (i.e., more in the number of amino acids) than Lz; in certain embodiments L 1 is shorter (i.e., fewer number of amino acids) than Lz.
  • L 1 and/or L 2 are “short,” e.g., consist of 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12 amino acid residues.
  • the spacer peptides consist of about 12 or fewer amino acid residues. In the case of 0 amino acid residues, the spacer peptide is a peptide bond.
  • L 1 and/or L 2 are “long,” e.g., consist of 15, 20 or 25 amino acid residues. In some embodiments, the spacer peptides consist of about 3 to about 15, for example 8, 9 or 10 contiguous amino acid residues.
  • peptides are selected with properties that confer flexibility to first and the second polypeptides of the proteins of the present invention, do not interfere with the binding of the first and the second, different subunits to each other, as well as resist cleavage from proteases.
  • glycine and serine residues generally provide protease resistance.
  • the spacer peptides suitable for linking the first subunit of the multisubunit protein to the amino acid sequence of SEQ ID NO:1, 2, 4, 6, 7, 9, 11, 237, 238, 239, or 240, and/or suitable for linking the second, different subunit of the multisubunit protein to the amino acid sequence of SEQ ID NO:1, 2, 4, 6, 7, 9, 11, 237, 238, 239, or 240 may include, as part of a linker, a (GS) n , (GGS) n , (GGGS) n , (GGSG) n , (GGSGG) n , and (GGGGS) n sequence, wherein n is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20.
  • L 1 and/or L 2 independently include a (GGGGS) 4 (SEQ ID NO:107) or (GGGGS) 3 (SEQ ID NO:108) sequence as part of a linker.
  • L 1 and/or L 2 independently include a peptide sequence, as part of a linker, as set forth in the sequences selected from: SEQ ID NO:111, SEQ ID NO:112, SEQ ID NO: 113, SEQ ID NO:114, SEQ ID NO:115, SEQ ID NO:116, SEQ ID NO:117, SEQ ID NO:118, SEQ ID NO:119, and SEQ ID NO:120, as listed in Table 7.
  • L 1 as part of a linker, does not include a sequence as set forth in SEQ ID NO:107, SEQ ID NO:108, SEQ ID NO:109, SEQ ID NO:110, SEQ ID NO:111, SEQ ID NO:112, SEQ ID NO:113, SEQ ID NO:114, SEQ ID NO:115, SEQ ID NO:116, SEQ ID NO:117, SEQ ID NO:118, SEQ ID NO:119, or SEQ ID NO:120.
  • only L 2 includes a sequence as set forth in SEQ ID NO:107, SEQ ID NO:108, SEQ ID NO:109, SEQ ID NO:110, SEQ ID NO:111, SEQ ID NO:112, SEQ ID NO:113, SEQ ID NO:114, SEQ ID NO:115, SEQ ID NO:116, SEQ ID NO:117, SEQ ID NO:118, SEQ ID NO:119, or SEQ ID NO:120.
  • neither L 1 nor L 2 includes a sequence as set forth in SEQ ID NO:107, SEQ ID NO:108, SEQ ID NO:109, SEQ ID NO:110, SEQ ID NO:111, SEQ ID NO:112, SEQ ID NO:113, SEQ ID NO:114, SEQ ID NO:115, SEQ ID NO:116, SEQ ID NO:117, SEQ ID NO:118, SEQ ID NO:119, or SEQ ID NO:120.
  • Some heterodimeric Fc-fused proteins of the present invention comprise a first polypeptide comprising a first subunit of a multisubunit protein and a first antibody Fc domain polypeptide, in which a linker comprising GGGGSGGGGSGGGGS (SEQ ID NO:118; (G4S) 3 ) connects the first subunit of a multisubunit protein to the first antibody Fc domain polypeptide, for example an Fc domain polypeptide of an IgG4 antibody or an Fc domain polypeptide of an IgG1 antibody.
  • Some heterodimeric Fc-fused proteins of the present invention comprise a second polypeptide comprising a second, different subunit of a multisubunit protein and a second antibody Fc domain polypeptide, in which a linker comprising GGGGSGGGGSGGGGS (SEQ ID NO:118; (G4S) 3 ) connects the second, different subunit of a multisubunit protein to the second antibody Fc domain polypeptide, for example an Fc domain polypeptide of an IgG4 antibody or an Fc domain polypeptide of an IgG1 antibody.
  • some heterodimeric Fc-fused proteins of the present invention comprise a first polypeptide comprising a first subunit of a multisubunit protein and a first antibody Fc domain polypeptide, in which a linker comprising GGGGSGGGGSGGGGS (SEQ ID NO:118; (G4S) 3 ) connects the first subunit of a multisubunit protein to the Fc domain polypeptide, and a second polypeptide comprising a second, different subunit of a multisubunit protein and a second antibody Fc domain polypeptide, in which the additional subunit is connected to the second antibody Fc domain polypeptide with a linker that does not comprise GGGGSGGGGSGGGGS (SEQ ID NO:118; (G4S) 3 ).
  • some heterodimeric Fc-fused proteins of the present invention comprise a first polypeptide comprising a first subunit of a multisubunit protein and a first antibody Fc domain polypeptide, in which a linker that does not comprise GGGGSGGGGSGGGGS (SEQ ID NO:118; (G4S) 3 ) connects the first subunit of a multisubunit protein to the Fc domain polypeptide, and a second polypeptide comprising a second, different subunit of a multisubunit protein and a second antibody Fc domain polypeptide, in which the second, different subunit of a multisubunit protein is connected to the second antibody Fc domain polypeptide with a linker comprising GGGGSGGGGSGGGGS (SEQ ID NO:118; (G4S) 3 ).
  • the assembly of proteins of the present invention can be accomplished by expressing a first polypeptide comprising a first subunit of a multisubunit protein sequence fused to a first antibody Fc domain polypeptide (e.g., an IgG4 antibody Fc variant sequence or an IgG1 antibody Fc variant sequence, as disclosed in Table 2), and a second polypeptide comprising a second, different subunit of a multisubunit protein sequence fused to a second antibody Fc domain polypeptide (e.g., an IgG4 antibody Fc variant sequence or an IgG1 antibody Fc variant sequence, as disclosed in Table 2) in the same cell, which leads to the assembly of a heterodimeric Fc-fused protein according to the invention.
  • a first antibody Fc domain polypeptide e.g., an IgG4 antibody Fc variant sequence or an IgG1 antibody Fc variant sequence, as disclosed in Table 2
  • a second antibody Fc domain polypeptide e.g., an IgG4 antibody Fc variant sequence or
  • the assembled proteins have heterodimeric Fc domain polypeptides with the first antibody Fc domain polypeptide and the second antibody Fc domain polypeptide bound to each other. Promoting the preferential assembly of heterodimers of the Fc can be accomplished by incorporating different mutations in the CH3 domain of each antibody heavy chain constant region as shown in U.S. Ser. Nos. 13/494,870, 16/028,850, 11/533,709, 12/875,015, 13/289,934, 14/773,418, 12/811,207, 13/866,756, 14/647,480, and 14/830,336.
  • mutations can be made in the CH3 domain based on human IgG1 and incorporating distinct pairs of amino acid substitutions within a first antibody Fc domain polypeptide and a second antibody Fc domain polypeptide that allow these two chains to selectively heterodimerize with each other.
  • the positions of amino acid substitutions illustrated below are all numbered according to the EU index as in Kabat.
  • an amino acid substitution in the first antibody Fc domain polypeptide replaces the original amino acid with a larger amino acid, selected from arginine (R), phenylalanine (F), tyrosine (Y) or tryptophan (W), and at least one amino acid substitution in the second antibody Fc domain polypeptide replaces the original amino acid(s) with a smaller amino acid(s), chosen from alanine (A), serine (S), threonine (T), or valine (V), such that the larger amino acid substitution (a protuberance) fits into the surface of the smaller amino acid substitutions (a cavity).
  • one antibody Fc domain polypeptide can incorporate a T366W substitution, and the other can incorporate three substitutions including T366S, L368A, and Y407V.
  • a first polypeptide comprising a first subunit of a multisubunit protein sequence or a second polypeptide comprising a second, different subunit of a multisubunit protein sequence of the invention can optionally be coupled to an amino acid sequence at least 90% (e.g., at least 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%) identical to an antibody constant region, such as an IgG constant region including hinge, CH2 and CH3 domains with or without a CH1 domain.
  • an antibody constant region such as an IgG constant region including hinge, CH2 and CH3 domains with or without a CH1 domain.
  • the amino acid sequence of the constant region is at least 90% (e.g., at least 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%) identical to a human antibody constant region, such as a human IgG1 constant region, an IgG2 constant region, an IgG3 constant region, or an IgG4 constant region.
  • the amino acid sequence of the constant region is at least 90% (e.g., at least 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%) identical to an antibody constant region from another mammal, such as rabbit, dog, cat, mouse, or horse.
  • One or more mutation(s) can be incorporated into the constant region as compared to the human IgG1 constant region, for example at Q347, Y349, L351, 5354, E356, E357, K360, Q362, 5364, T366, L368, K370, N390, K392, T394, D399, 5400, D401, F405, Y407, K409, T411 and/or K439.
  • substitutions include, for example, Q347E, Q347R, Y349S, Y349K, Y349T, Y349D, Y349E, Y349C, T350V, L351K, L351D, L351Y, S354C, E356K, E357Q, E357L, E357W, K360E, K360W, Q362E, S364K, S364E, S364H, S364D, T366V, T366I, T366L, T366M, T366K, T366W, T366S, L368E, L368A, L368D, K370S, N390D, N390E, K392L, K392M, K392V, K392F, K392D, K392E, T394F, T394W, D399R, D399K, D399V, S400K,
  • mutations that can be incorporated into the CH1 of a human IgG1 constant region may be at amino acids V125, F126, P127, T135, T139, A140, F170, P171, and/or V173.
  • mutations that can be incorporated into the C ⁇ of a human IgG1 constant region may be at amino acids E123, F116, S176, V163, 5174, and/or T164.
  • Amino acid substitutions could be selected from the following sets of substitutions shown in Table 8.
  • amino acid substitutions could be selected from the following sets of substitutions shown in Table 9.
  • amino acid substitutions could be selected from the following set of substitutions shown in Table 10.
  • At least one amino acid substitution in each polypeptide chain could be selected from Table 11.
  • At least one amino acid substitutions could be selected from the following set of substitutions in Table 12, where the position(s) indicated in the First Polypeptide column is replaced by any known negatively-charged amino acid, and the position(s) indicated in the Second Polypeptide Column is replaced by any known positively-charged amino acid.
  • At least one amino acid substitutions could be selected from the following set of in Table 13, where the position(s) indicated in the First Polypeptide column is replaced by any known positively-charged amino acid, and the position(s) indicated in the Second Polypeptide Column is replaced by any known negatively-charged amino acid.
  • amino acid substitutions could be selected from the following set in Table 14.
  • the structural stability of a heterodimeric Fc-fused protein according to the invention may be increased by introducing S354C on either of the first or second polypeptide chain, and Y349C on the opposing polypeptide chain, which forms an artificial disulfide bond within the interface of the two polypeptides.
  • the amino acid sequence of one polypeptide chain of the antibody constant region differs from the amino acid sequence of an IgG1 constant region at position T366, and the amino acid sequence of the other polypeptide chain of the antibody constant region differs from the amino acid sequence of an IgG1 constant region at one or more position(s) selected from T366, L368 and Y407.
  • the amino acid sequence of one polypeptide chain of the antibody constant region differs from the amino acid sequence of an IgG1 constant region at one or more position(s) selected from T366, L368 and Y407, and the amino acid sequence of the other polypeptide chain of the antibody constant region differs from the amino acid sequence of an IgG1 constant region at position T366.
  • the amino acid sequence of one polypeptide chain of the antibody constant region differs from the amino acid sequence of an IgG1 constant region at one or more position(s) selected from E357, K360, Q362, 5364, L368, K370, T394, D401, F405, and T411 and the amino acid sequence of the other polypeptide chain of the antibody constant region differs from the amino acid sequence of an IgG1 constant region at one or more position(s) selected from Y349, E357, S364, L368, K370, T394, D401, F405 and T411.
  • the amino acid sequence of one polypeptide chain of the antibody constant region differs from the amino acid sequence of an IgG1 constant region at one or more position(s) selected from Y349, E357, S364, L368, K370, T394, D401, F405 and T411 and the amino acid sequence of the other polypeptide chain of the antibody constant region differs from the amino acid sequence of an IgG1 constant region at one or more position(s) selected from E357, K360, Q362, S364, L368, K370, T394, D401, F405, and T411.
  • the amino acid sequence of one polypeptide chain of the antibody constant region differs from the amino acid sequence of an IgG1 constant region at one or more position(s) selected from L351, D399, 5400 and Y407 and the amino acid sequence of the other polypeptide chain of the antibody constant region differs from the amino acid sequence of an IgG1 constant region at one or more position(s) selected from T366, N390, K392, K409 and T411.
  • the amino acid sequence of one polypeptide chain of the antibody constant region differs from the amino acid sequence of an IgG1 constant region at one or more position(s) selected from T366, N390, K392, K409 and T411 and the amino acid sequence of the other polypeptide chain of the antibody constant region differs from the amino acid sequence of an IgG1 constant region at one or more position(s) selected from L351, D399, 5400 and Y407.
  • the amino acid sequence of one polypeptide chain of the antibody constant region differs from the amino acid sequence of an IgG1 constant region at one or more position(s) selected from Q347, Y349, K360, and K409, and the amino acid sequence of the other polypeptide chain of the antibody constant region differs from the amino acid sequence of an IgG1 constant region at one or more position(s) selected from Q347, E357, D399 and F405.
  • the amino acid sequence of one polypeptide chain of the antibody constant region differs from the amino acid sequence of an IgG1 constant region at one or more position(s) selected from Q347, E357, D399 and F405, and the amino acid sequence of the other polypeptide chain of the antibody constant region differs from the amino acid sequence of an IgG1 constant region at one or more position(s) selected from Y349, K360, Q347 and K409.
  • the amino acid sequence of one polypeptide chain of the antibody constant region differs from the amino acid sequence of an IgG1 constant region at one or more position(s) selected from K370, K392, K409 and K439, and the amino acid sequence of the other polypeptide chain of the antibody constant region differs from the amino acid sequence of an IgG1 constant region at one or more position(s) selected from D356, E357 and D399.
  • the amino acid sequence of one polypeptide chain of the antibody constant region differs from the amino acid sequence of an IgG1 constant region at one or more position(s) selected from D356, E357 and D399, and the amino acid sequence of the other polypeptide chain of the antibody constant region differs from the amino acid sequence of an IgG1 constant region at one or more position(s) selected from K370, K392, K409 and K439.
  • the amino acid sequence of one polypeptide chain of the antibody constant region differs from the amino acid sequence of an IgG1 constant region at one or more position(s) selected from L351, E356, T366 and D399, and the amino acid sequence of the other polypeptide chain of the antibody constant region differs from the amino acid sequence of an IgG1 constant region at one or more position(s) selected from Y349, L351, L368, K392 and K409.
  • the amino acid sequence of one polypeptide chain of the antibody constant region differs from the amino acid sequence of an IgG1 constant region at one or more position(s) selected from Y349, L351, L368, K392 and K409, and the amino acid sequence of the other polypeptide chain of the antibody constant region differs from the amino acid sequence of an IgG1 constant region at one or more position(s) selected from L351, E356, T366 and D399.
  • the amino acid sequence of one polypeptide chain of the antibody constant region differs from the amino acid sequence of an IgG1 constant region by an S354C substitution and the amino acid sequence of the other polypeptide chain of the antibody constant region differs from the amino acid sequence of an IgG1 constant region by a Y349C substitution.
  • the amino acid sequence of one polypeptide chain of the antibody constant region differs from the amino acid sequence of an IgG1 constant region by a Y349C substitution and the amino acid sequence of the other polypeptide chain of the antibody constant region differs from the amino acid sequence of an IgG1 constant region by an S354C substitution.
  • the amino acid sequence of one polypeptide chain of the antibody constant region differs from the amino acid sequence of an IgG1 constant region by K360E and K409W substitutions and the amino acid sequence of the other polypeptide chain of the antibody constant region differs from the amino acid sequence of an IgG1 constant region by O347R, D399V and F405T substitutions.
  • the amino acid sequence of one polypeptide chain of the antibody constant region differs from the amino acid sequence of an IgG1 constant region by O347R, D399V and F405T substitutions and the amino acid sequence of the other polypeptide chain of the antibody constant region differs from the amino acid sequence of an IgG1 constant region by K360E and K409W substitutions.
  • the amino acid sequence of one polypeptide chain of the antibody constant region differs from the amino acid sequence of an IgG1 constant region by a T366W substitutions and the amino acid sequence of the other polypeptide chain of the antibody constant region differs from the amino acid sequence of an IgG1 constant region by T366S, T368A, and Y407V substitutions.
  • the amino acid sequence of one polypeptide chain of the antibody constant region differs from the amino acid sequence of an IgG1 constant region by T366S, T368A, and Y407V substitutions and the amino acid sequence of the other polypeptide chain of the antibody constant region differs from the amino acid sequence of an IgG1 constant region by a T366W substitution.
  • the amino acid sequence of one polypeptide chain of the antibody constant region differs from the amino acid sequence of an IgG1 constant region by T350V, L351Y, F405A, and Y407V substitutions and the amino acid sequence of the other polypeptide chain of the antibody constant region differs from the amino acid sequence of an IgG1 constant region by T350V, T366L, K392L, and T394W substitutions.
  • the amino acid sequence of one polypeptide chain of the antibody constant region differs from the amino acid sequence of an IgG1 constant region by T350V, T366L, K392L, and T394W substitutions and the amino acid sequence of the other polypeptide chain of the antibody constant region differs from the amino acid sequence of an IgG1 constant region by T350V, L351Y, F405A, and Y407V substitutions.
  • N-terminal glutamate (E) or glutamine (Q) can be cyclized to form a lactam (e.g., spontaneously or catalyzed by an enzyme present during production and/or storage). Accordingly, in some embodiments where the N-terminal residue of an amino acid sequence of a polypeptide is E or Q, a corresponding amino acid sequence with the E or Q replaced with pyroglutamate is also contemplated herein.
  • the C-terminal lysine (K) of a protein can be removed (e.g., spontaneously or catalyzed by an enzyme present during production and/or storage). Such removal of K is often observed with proteins that comprise a Fc domain at its C-terminus. Accordingly, in some embodiments where the C-terminal residue of an amino acid sequence of a polypeptide (e.g., a Fc domain sequence) is K, a corresponding amino acid sequence with the K removed is also contemplated herein.
  • the present invention provides a heterodimeric Fc-fused protein comprising (a) a first polypeptide comprising a first antibody Fc domain polypeptide and a first subunit of a multisubunit protein; and (b) a second polypeptide comprising a second antibody Fc domain polypeptide and a second, different subunit of the multisubunit protein, wherein the first and second antibody Fc domain polypeptides each comprise different mutations promoting heterodimerization, wherein the first and/or second antibody Fc domain polypeptides comprise one or more mutation(s) that reduce(s) an effector function of an Fc, and wherein the first subunit and second, different subunit of the multisubunit protein are bound to each other.
  • a heterodimeric Fc-fused protein disclosed herein comprising one or more mutation(s) that reduce(s) an effector function of an Fc has an increased activity to inhibit tumor growth than its counterpart without the Fc mutation(s) that reduce(s) the effector function.
  • the mutations contemplated herein include substitution, insertion, and deletion of amino acid residues. All the amino acid positions in an Fc domain or hinge region disclosed herein are numbered according to EU numbering.
  • the first and/or second antibody Fc domain polypeptides comprise one or more mutation(s) that reduce(s) the ability of the Fc domain polypeptide to induce antibody-dependent cellular cytotoxicity (ADCC) and/or antibody-dependent cellular phagocytosis (ADCP).
  • ADCC and ADCP are typically mediated by an Fc receptor.
  • the first and second antibody Fc domain polypeptides are human IgG (e.g., human IgG1, human IgG2, human IgG3, or human IgG4) antibody sequences.
  • Fc receptors of human IgG also called Fc gamma receptors (Fc ⁇ Rs)
  • Fc ⁇ Rs include but are not limited to activating Fc gamma receptors Fc ⁇ RI (CD64), Fc ⁇ RIIA (CD32A), Fc ⁇ RIIIA (CD16 or CD16A), and Fc ⁇ RIIIB (CD16B), and inhibitor Fc gamma receptor Fc ⁇ RIIB (CD32B).
  • a heterodimeric Fc-fused protein of the present invention includes one or more mutation(s) to reduce binding to an activating Fc ⁇ R (e.g., Fc ⁇ RI, Fc ⁇ RIIA, Fc ⁇ RIIIA, or Fc ⁇ RIIIB) in the first and/or second polypeptides.
  • a heterodimeric Fc-fused protein of the present invention includes one or more mutation(s) to increase binding to an inhibitory Fc ⁇ R (e.g., Fc ⁇ RIIB) in the first and/or second polypeptides.
  • Fc mutations that reduce binding to an activating Fc ⁇ R and/or increase binding to an inhibitory Fc ⁇ R are known in the art.
  • CD16 binding is mediated by the hinge region and the CH2 domain.
  • the interaction with CD16 is primarily focused on amino acid residues Asp 265-Glu 269, Asn 297-Thr 299, Ala 327-Ile 332, Leu 234-Ser 239, and carbohydrate residue N-acetyl-D-glucosamine in the CH2 domain (see, Sondermann et al, Nature, 406 (6793):267-273).
  • mutations can be selected to enhance or reduce the binding affinity to CD16, such as by using phage-displayed libraries or yeast surface-displayed cDNA libraries, or can be designed based on the known three-dimensional structure of the interaction.
  • mutation at position 329 also reduces activating Fc ⁇ R binding.
  • Additional amino acid positions and mutations e.g., E233P mutation
  • Additional amino acid positions and mutations implicated in activating Fc ⁇ R binding are disclosed in U.S. Pat. No. 7,943,743 and Isaacs et al., J. Immunol. (1998) 161:3862-69.
  • the first and second antibody Fc domain polypeptides comprise a mutation (e.g., substitution relative to wild-type human IgG1) at one or more of positions selected from 233, 234, 235, 297, and 329.
  • the first and second antibody Fc domain polypeptides are human IgG1 antibody Fc domain polypeptides comprising mutation(s) E233P; L234A (human IgG1) or F234A (human IgG4); L235A or L235E; N297A, N297Q, N297G, or N297D; and/or P329A, P329G, or P329R.
  • the first and second antibody Fc domain polypeptides are human IgG1 antibody Fc domain polypeptides comprising mutations L234A and L235A. In certain embodiments, the first and second antibody Fc domain polypeptides are human IgG1 antibody Fc domain polypeptides comprising mutations L234A, L235A, and P329A. In certain embodiments, the first and second antibody Fc domain polypeptides are human IgG4 antibody Fc domain polypeptides comprising mutation L235E. In certain embodiments, the first and second antibody Fc domain polypeptides are human IgG1 antibody Fc domain polypeptides comprising mutations L235E and P329A.
  • the first and/or second antibody Fc domain polypeptides comprise one or more mutation(s) that reduce(s) the ability of the Fc domain polypeptide to induce complement dependent cytotoxicity (CDC).
  • CDC is typically mediated by a complement component (e.g., C1q).
  • a heterodimeric Fc-fused protein of the present invention includes one or more mutation(s) to reduce binding to a complement component (e.g., C1q) in the first and/or second polypeptides.
  • Fc mutations that reduce binding to C1q are known in the art.
  • the amino acid residues of Fc at positions 234, 235, 236, 237, 297, 318, 320, and 322 are implicated in C1q binding.
  • residue Pro at position 331 is implicated in C1q binding.
  • the first and second antibody Fc domain polypeptides comprise a mutation (e.g., substitution relative to wild-type human IgG1) at one or more of positions selected from 234, 235, 236, 237, 270, 297, 318, 320, 322, 329, and 331.
  • the first and second antibody Fc domain polypeptides are human IgG1 antibody Fc domain polypeptides comprising mutation(s) G237A, A330S, P331S, and/or P329A.
  • the first and second antibody Fc domain polypeptides are human IgG1 antibody Fc domain polypeptides comprising mutations G237A, A330S, and P331S.
  • the first and second antibody Fc domain polypeptides are human IgG1 antibody Fc domain polypeptides comprising mutation P329A.
  • the mutations that reduce ADCC and/or ADCP and the mutations that reduce CDC can be combined.
  • the first and/or second antibody Fc domain polypeptides comprise one or more mutation(s) that reduce(s) the ability of the Fc domain polypeptide to induce ADCC and/or ADCP and further comprise one or more mutation(s) that reduce(s) the ability of the Fc domain polypeptide to induce CDC.
  • the first and second antibody Fc domain polypeptides each comprise one or more mutation(s) that reduce(s) the ability of the Fc domain polypeptide to induce ADCC and/or ADCP and further comprise one or more mutation(s) that reduce(s) the ability of the Fc domain polypeptide to induce CDC.
  • a heterodimeric Fc-fused protein of the present invention with an IgG4 Fc includes one or more mutation(s) to reduce binding to an Fc ⁇ R (e.g., Fc ⁇ RI, Fc ⁇ RIIA, Fc ⁇ RIIB, Fc ⁇ RIIIA, or Fc ⁇ RIIIB) or a complement component (e.g., C1q) in the first and/or second polypeptides.
  • Fc ⁇ R e.g., Fc ⁇ RI, Fc ⁇ RIIA, Fc ⁇ RIIB, Fc ⁇ RIIIA, or Fc ⁇ RIIIB
  • complement component e.g., C1q
  • a protein of the present disclosure can include SPLE (S228P and L235E) mutations, SPLEPA (S228P, L235E, and P329A) mutations, or SPLEPG (S228P, L235E, and P329G) mutations.
  • SPLE S228P and L235E
  • SPLEPA S228P, L235E, and P329A
  • SPLEPG S228P, L235E, and P329G mutations.
  • a heterodimeric Fc-fused protein of the present invention with an IgG1 Fc includes one or more mutation(s) to reduce binding to an Fc ⁇ R (e.g., Fc ⁇ RI, Fc ⁇ RIIA, Fc ⁇ RIIB, Fc ⁇ RIIIA, or Fc ⁇ RIIIB) or a complement component (e.g., C1q) in the first and/or second polypeptides.
  • Fc ⁇ R e.g., Fc ⁇ RI, Fc ⁇ RIIA, Fc ⁇ RIIB, Fc ⁇ RIIIA, or Fc ⁇ RIIIB
  • complement component e.g., C1q
  • a protein of the present disclosure can include LALA (L234A and L235A) mutations, LALAPA (L234A, L235A, and P329A) mutations, LALAPG (L234A, L235A, and P329G) mutations, or LALEGAASPS (L234A, L235E, G237A, A330S, and P331S) mutations.
  • LALA L234A and L235A
  • LALAPA L234A, L235A, and P329A
  • LALAPG L234A, L235A, and P329G mutations
  • LALEGAASPS L234A, L235E, G237A, A330S, and P331S
  • a heterodimeric Fc-fused protein according to the invention includes a first antibody IgG4 or IgG1 Fc domain polypeptide and a second antibody IgG4 or IgG1 Fc domain polypeptide each containing the mutation P329G or P329A.
  • the first antibody Fc domain polypeptide and the second antibody Fc domain polypeptide each contain a mutation selected from A330S and P331S.
  • the first antibody Fc domain polypeptide and the second antibody Fc domain polypeptide each contain the mutations A330S and P331S.
  • the first subunit of the multisubunit protein is fused to the first antibody Fc domain polypeptide by a first linker.
  • the second, different subunit of the multisubunit protein is fused to the second antibody Fc domain polypeptide by a second linker.
  • linkers suitable for such use are described under the headings “IgG4 constructs” and “IgG1 constructs.” Additional linker sequences suitable for use in the first and/or second polypeptides include but are not limited to wild-type IgG (e.g., human IgG1, human IgG2, human IgG3, or human IgG4) hinge sequences and mutant forms thereof.
  • the first and second linkers each comprise amino acid sequence ESKYGPPCPPCPAPEFXGG, wherein X is L or E (SEQ ID NO:280) or SKYGPPCPPCPAPEFXGG, wherein X is L or E (SEQ ID NO:281).
  • the first and second linkers each comprise amino acid sequence ESKYGPPCPPCPAPEFLGG (SEQ ID NO:282) or SKYGPPCPPCPAPEFLGG (SEQ ID NO:283). In certain embodiments, the first and second linkers each comprise amino acid sequence ESKYGPPCPPCPAPEFEGG (SEQ ID NO:284) or SKYGPPCPPCPAPEFEGG (SEQ ID NO:285).
  • Heterodimeric Fc-fused proteins according to the invention have pharmacokinetic properties suitable for therapeutic use.
  • a heterodimeric Fc-fused protein according to the invention has a serum half-life of at least about 50 hours.
  • a heterodimeric Fc-fused protein according to the invention has a serum half-life of at least about 100 hours.
  • the serum concentration of the heterodimeric Fc-fused protein according to the invention is at least 10% of the serum concentration of the protein of the present invention 1 hour after the administration in said subject.
  • a heterodimeric Fc-fused protein according to the invention has a serum half-life that is at least 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90% longer than the multisubunit protein not fused to Fc domain polypeptides.
  • a heterodimeric Fc-fused protein comprising a protein sequence of a multisubunit protein according to the present invention has a serum half-life that is at least 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold, 15-fold, or 20-fold longer than the multisubunit protein not fused to Fc domain polypeptides.
  • heterodimeric Fc-fused proteins of the invention can optionally incorporate additional features to enhance retention of the proteins at the tumor site.
  • the heterodimeric Fc-fused protein further comprises a proteoglycan-binding domain, a collagen-binding domain, and/or a hyaluronic acid-binding domain.
  • the heterodimeric Fc-fused protein further comprises a proteoglycan-binding domain that binds one or more proteoglycans (e.g., proteoglycans known in the art, e.g., as disclosed in Lozzo et al., Matrix Bio (2015) 42:11-55; and Nikitovic et al., Frontiers in Endocrinology (2016) 9:69) that are present in a tumor (e.g., on the surface of a tumor cell, in a pericellular matrix in a tumor, or in a extracellular matrix in a tumor).
  • proteoglycans e.g., proteoglycans known in the art, e.g., as disclosed in Lozzo et al., Matrix Bio (2015) 42:11-55; and Nikitovic et al., Frontiers in Endocrinology (2016) 9:69
  • the collagen-binding domain binds one or more collagens that are present in a tumor (e.g., on the surface of a tumor cell, in a pericellular matrix in a tumor, or in a extracellular matrix in a tumor).
  • the heterodimeric Fc-fused protein further comprises a h acid-binding domain that binds to one ore more hyaluronic acid that are present in a tumor.
  • Such heterodimeric Fc-fused proteins have enhanced retention in tumors and may be administered to a subject intratumorally at a lower dose and/or frequency.
  • the proteoglycan-binding domain comprised in the heterodimeric Fc-fused protein binds one or more proteoglycans that are specifically expressed in a tumor (e.g., on the surface of a tumor cell, in a pericellular matrix in a tumor, or in a extracellular matrix in a tumor).
  • the collagen-binding domain comprised in the heterodimeric Fc-fused protein binds one or more collagens that are specifically expressed in a tumor (e.g., on the surface of a tumor cell, in a pericellular matrix in a tumor, or in a extracellular matrix in a tumor).
  • Such heterodimeric Fc-fused proteins may be enriched in tumors after administration (e.g., intravenous, subcutaneous, or pulmonary administration) and have enhanced tumor retention, thereby allowing administration at a lower dose and/or frequency.
  • the heterodimeric Fc-fused protein of the present invention further comprises a proteoglycan-binding domain that binds one or more proteoglycans selected from syndecan, chondroitin sulfate proteoglycan 4 (CSPG4), betaglycan, phosphacan, glypican, perlecan, agrin, collagen (e.g., collagen IX, XII, XV, or XVIII), hyalectan, aggrecan, versican, neurocan, brevican, and a small leucine-rich proteoglycan (SLRP).
  • proteoglycan-binding domain that binds one or more proteoglycans selected from syndecan, chondroitin sulfate proteoglycan 4 (CSPG4), betaglycan, phosphacan, glypican, perlecan, agrin, collagen (e.g., collagen IX, XII, XV,
  • Proteoglycans implicated in cancer include but are not limited to collagen, syndecan (e.g., syndecan-1 or syndecan-2), serglycin, CSPG4, betaglycan, glypican (e.g., glypican-1 or glypican-3), perlecan, versican, brevican, and SLPR (e.g., decorin, biglycan, asporin, fibrodulin, and lumican).
  • syndecan e.g., syndecan-1 or syndecan-2
  • serglycin e.g., serglycin
  • CSPG4 betaglycan
  • glypican e.g., glypican-1 or glypican-3
  • perlecan e.g., versican, brevican
  • SLPR e.g., decorin, biglycan, asporin, fibrodulin, and lumican
  • the proteoglycan-binding domain comprised in the heterodimeric Fc-fused protein binds one or more proteoglycans selected from syndecan (e.g., syndecan-1 or syndecan-2), serglycin, CSPG4, betaglycan, glypican (e.g., glypican-1 or glypican-3), perlecan, versican, brevican, and a SLPR.
  • the proteoglycan-binding domain comprised in the heterodimeric Fc-fused protein binds one or more SLPRs selected from decorin, biglycan, asporin, fibrodulin, and lumican.
  • the proteoglycan-binding domain comprised in the heterodimeric Fc-fused protein can be a protein (e.g., an antibody or an antigen-binding fragment thereof), a peptide (e.g., a portion of a proteoglycan-binding protein or a variant thereof), an aptamer, a small molecule, or a combination thereof.
  • Proteoglycan-binding domains are also known in the art. For example, syndecan-binding domains are disclosed in U.S. Pat. Nos. 6,566,489, 8,647,828, and 10,124,038; U.S. Patent Application Publication No. 2009/0297479; and PCT Patent Application Publication No. WO2018199176A1.
  • CSPG4-binding domains are disclosed in U.S. Pat. Nos. 9,801,928 and 10,093,745; and U.S. Patent Application Publication Nos. 2016/0032007, 2017/0342151, and 2018/0072811.
  • ⁇ -glycan-binding domains are disclosed in U.S. Pat. No. 7,455,839.
  • Glypican-binding domains are disclosed in U.S. Pat. Nos. 7,919,086, 7,776,329, 8,680,247, 8,388,937, 9,260,492, 9,394,364, 9,790,267, 9,522,940, and 9,409,994; U.S. Patent Application Publication Nos.
  • the heterodimeric Fc-fused protein of the present invention further comprises a collagen-binding domain.
  • Collagen is a class of proteins having at least 28 different types identified in vertebrates. Each type of collagen has its unique structural characteristics and distribution pattern, as disclosed in Fang et al., Tumor Biol. (2014) 35:2871-82 and Xiong et al., J. Cancer Metasta. Treat. (2016) 2:357-64.
  • Various types of collagens are implicated in cancer, including but not limited to Col3A1, Col5A2, Col6, Col7A1, Col15A1 Col19A1, and Col22A1.
  • the collagen-binding domain can be a protein (e.g., an antibody or an antigen-binding fragment thereof), a peptide (e.g., a portion of a collagen-binding protein or a variant thereof), an aptamer, a small molecule, or a combination thereof.
  • Collagen-binding domains are known in the art, and are disclosed in, for example, U.S. Pat. Nos. 5,788,966, 5,587,360, 5,851,794, 5,741,670, 5,849,701, 6,288,214, 6,387,663, 6,908,994, 7,169,902, 7,488,792, 7,820,401, 8,956,612, 8,642,728, and 8,906,649, and U.S. Patent Application Publication Nos. 2007/0161062, 2009/0142345, and 2012/0100106.
  • the heterodimeric Fc-fused protein of the present invention further comprises a hyaluronic acid-binding domain.
  • the hyaluronic acid-binding domain can be a protein (e.g., an antibody or an antigen-binding fragment thereof), a peptide (e.g., a portion of a hyaluronic acid-binding protein or a variant thereof), an aptamer, a small molecule, or a combination thereof.
  • Hyaluronic acid-binding domains are known in the art, and are disclosed in, for example, U.S. Pat. Nos.
  • a proteoglycan-binding domain, collagen-binding domain, and/or hyaluronic acid-binding domain can be at any position of the heterodimeric Fc-fused protein.
  • a proteoglycan-binding domain, a collagen-binding domain, and/or a hyaluronic acid-binding domain as disclosed herein can be fused to the C-terminus of the first antibody Fc domain polypeptide and/or to the C-terminus of the second antibody Fc domain polypeptide.
  • a proteoglycan-binding domain, a collagen-binding domain, and/or a hyaluronic acid-binding domain as disclosed herein can be fused to the N-terminus of the first antibody Fc domain polypeptide and/or to the N-terminus of the second antibody Fc domain polypeptide.
  • a proteoglycan-binding domain, collagen-binding domain, and/or hyaluronic acid-binding domain, if present, can be fused to the rest of the heterodimeric Fc-fused protein through a linker.
  • the proteoglycan-binding domain is fused to the rest of the heterodimeric Fc-fused protein through a peptide linker.
  • the peptide linker includes a spacer peptide disclosed herein.
  • the proteins of the present invention can be made using recombinant DNA technology well known to a skilled person in the art.
  • a first nucleic acid sequence encoding a first polypeptide comprising a first subunit of a multisubunit protein sequence fused to a first antibody Fc domain polypeptide can be cloned into a first expression vector;
  • a second nucleic acid sequence encoding a second polypeptide comprising a second, different subunit of a multisubunit fused to a second antibody Fc domain polypeptide can be cloned into a second expression vector; and the first and the second expression vectors can be stably transfected together into host cells to produce the multimeric proteins.
  • Clones can be cultured under conditions suitable for bio-reactor scale-up and maintained expression of the proteins of the present invention.
  • the proteins can be isolated and purified using methods known in the art including centrifugation, depth filtration, cell lysis, homogenization, freeze-thawing, affinity purification, gel filtration, ion exchange chromatography, hydrophobic interaction exchange chromatography, and mixed-mode chromatography.
  • compositions that contain an effective amount of a protein described herein.
  • the composition can be formulated for use in a variety of drug delivery systems.
  • One or more physiologically acceptable excipient(s) or carrier(s) can also be included in the composition for proper formulation.
  • suitable formulations for use in the present disclosure are found in Remington's Pharmaceutical Sciences, Mack Publishing Company, Philadelphia, Pa., 17th ed., 1985.
  • Langer Science 249:1527-1533, 1990).
  • the intravenous drug delivery formulation of the present disclosure may be contained in a bag, a pen, or a syringe.
  • the bag may be connected to a channel comprising a tube and/or a needle.
  • the formulation may be a lyophilized formulation or a liquid formulation.
  • the formulation may freeze-dried (lyophilized) and contained in about 12-60 vials.
  • the formulation may be freeze-dried and 45 mg of the freeze-dried formulation may be contained in one vial.
  • the about 40 mg-about 100 mg of freeze-dried formulation may be contained in one vial.
  • freeze dried formulation from 12, 27, or 45 vials are combined to obtained a therapeutic dose of the protein in the intravenous drug formulation.
  • the formulation may be a liquid formulation and stored as about 250 mg/vial to about 1000 mg/vial. In certain embodiments, the formulation may be a liquid formulation and stored as about 600 mg/vial. In certain embodiments, the formulation may be a liquid formulation and stored as about 250 mg/vial.
  • a heterodimeric Fc-fused protein of the present invention could exist in a liquid aqueous pharmaceutical formulation including an effective amount of the protein in a buffered solution forming a formulation.
  • compositions may be sterilized by conventional sterilization techniques, or may be sterile filtered.
  • the resulting aqueous solutions may be packaged for use as-is, or lyophilized, the lyophilized preparation being combined with a sterile aqueous carrier prior to administration.
  • the pH of the preparations typically will be between 3 and 11, more preferably between 5 and 9 or between 6 and 8, and most preferably between 7 and 8, such as 7 to 7.5.
  • the resulting compositions in solid form may be packaged in multiple single dose units, each containing a fixed amount of the above-mentioned agent or agents.
  • the composition in solid form can also be packaged in a container for a flexible quantity.
  • the present disclosure provides a formulation with an extended shelf life including the heterodimeric Fc-fused protein of the present disclosure, in combination with mannitol, citric acid monohydrate, sodium citrate, disodium phosphate dihydrate, sodium dihydrogen phosphate dihydrate, sodium chloride, polysorbate 80, water, and sodium hydroxide.
  • an aqueous formulation is prepared including the heterodimeric Fc-fused protein of the present disclosure in a pH-buffered solution.
  • the buffer of this invention may have a pH ranging from about 4 to about 8, e.g., from about 4.5 to about 6.0, or from about 4.8 to about 5.5, or may have a pH of about 5.0 to about 5.2. Ranges intermediate to the above recited pH's are also intended to be part of this disclosure. For example, ranges of values using a combination of any of the above recited values as upper and/or lower limits are intended to be included. Examples of buffers that will control the pH within this range include acetate (e.g., sodium acetate), succinate (such as sodium succinate), gluconate, histidine, citrate and other organic acid buffers.
  • the formulation includes a buffer system which contains citrate and phosphate to maintain the pH in a range of about 4 to about 8.
  • the pH range may be from about 4.5 to about 6.0, or from about pH 4.8 to about 5.5, or in a pH range of about 5.0 to about 5.2.
  • the buffer system includes citric acid monohydrate, sodium citrate, disodium phosphate dihydrate, and/or sodium dihydrogen phosphate dihydrate.
  • the buffer system includes about 1.3 mg/mL of citric acid (e.g., 1.305 mg/mL), about 0.3 mg/mL of sodium citrate (e.g., 0.305 mg/mL), about 1.5 mg/mL of disodium phosphate dihydrate (e.g., 1.53 mg/mL), about 0.9 mg/mL of sodium dihydrogen phosphate dihydrate (e.g., 0.86), and about 6.2 mg/mL of sodium chloride (e.g., 6.165 mg/mL).
  • citric acid e.g., 1.305 mg/mL
  • sodium citrate e.g. 0.305 mg/mL
  • 1.5 mg/mL of disodium phosphate dihydrate e.g., 1.53 mg/mL
  • about 0.9 mg/mL of sodium dihydrogen phosphate dihydrate e.g., 0.86
  • sodium chloride e.g., 6.165 mg/mL
  • the buffer system includes 1-1.5 mg/mL of citric acid, 0.25 to 0.5 mg/mL of sodium citrate, 1.25 to 1.75 mg/mL of disodium phosphate dihydrate, 0.7 to 1.1 mg/mL of sodium dihydrogen phosphate dihydrate, and 6.0 to 6.4 mg/mL of sodium chloride.
  • the pH of the formulation is adjusted with sodium hydroxide.
  • a polyol which acts as a tonicifier and may stabilize the antibody, may also be included in the formulation.
  • the polyol is added to the formulation in an amount which may vary with respect to the desired isotonicity of the formulation.
  • the aqueous formulation may be isotonic.
  • the amount of polyol added may also be altered with respect to the molecular weight of the polyol. For example, a lower amount of a monosaccharide (e.g., mannitol) may be added, compared to a disaccharide (such as trehalose).
  • the polyol which may be used in the formulation as a tonicity agent is mannitol.
  • the mannitol concentration may be about 5 to about 20 mg/mL. In certain embodiments, the concentration of mannitol may be about 7.5 to 15 mg/mL. In certain embodiments, the concentration of mannitol may be about 10-14 mg/mL. In certain embodiments, the concentration of mannitol may be about 12 mg/mL. In certain embodiments, the polyol sorbitol may be included in the formulation.
  • a detergent or surfactant may also be added to the formulation.
  • exemplary detergents include nonionic detergents such as polysorbates (e.g., polysorbates 20, 80 etc.) or poloxamers (e.g., poloxamer 188).
  • the amount of detergent added is such that it reduces aggregation of the formulated antibody and/or minimizes the formation of particulates in the formulation and/or reduces adsorption.
  • the formulation may include a surfactant which is a polysorbate.
  • the formulation may contain the detergent polysorbate 80 or Tween 80.
  • Tween 80 is a term used to describe polyoxyethylene (20) sorbitanmonooleate (see Fiedler, Lexikon der Hifsstoffe, Editio Cantor Verlag Aulendorf, 4th ed., 1996).
  • the formulation may contain between about 0.1 mg/mL and about 10 mg/mL of polysorbate 80, or between about 0.5 mg/mL and about 5 mg/mL. In certain embodiments, about 0.1% polysorbate 80 may be added in the formulation.
  • the protein product of the present disclosure is formulated as a liquid formulation.
  • the liquid formulation may be presented at a 10 mg/mL concentration in either a USP/Ph Eur type I 50R vial closed with a rubber stopper and sealed with an aluminum crimp seal closure.
  • the stopper may be made of elastomer complying with USP and Ph Eur.
  • vials may be filled with 61.2 mL of the protein product solution in order to allow an extractable volume of 60 mL.
  • the liquid formulation may be diluted with 0.9% saline solution.
  • the liquid formulation of the disclosure may be prepared as a 10 mg/mL concentration solution in combination with a sugar at stabilizing levels.
  • the liquid formulation may be prepared in an aqueous carrier.
  • a stabilizer may be added in an amount no greater than that which may result in a viscosity undesirable or unsuitable for intravenous administration.
  • the sugar may be disaccharides, e.g., sucrose.
  • the liquid formulation may also include one or more of a buffering agent, a surfactant, and a preservative.
  • the pH of the liquid formulation may be set by addition of a pharmaceutically acceptable acid and/or base.
  • the pharmaceutically acceptable acid may be hydrochloric acid.
  • the base may be sodium hydroxide.
  • deamidation is a common product variant of peptides and proteins that may occur during fermentation, harvest/cell clarification, purification, drug substance/drug product storage and during sample analysis.
  • Deamidation is the loss of NH 3 from a protein forming a succinimide intermediate that can undergo hydrolysis.
  • the succinimide intermediate results in a 17 dalton mass decrease of the parent peptide.
  • the subsequent hydrolysis results in an 18 dalton mass increase.
  • Isolation of the succinimide intermediate is difficult due to instability under aqueous conditions. As such, deamidation is typically detectable as a 1 dalton mass increase.
  • Deamidation of an asparagine results in either aspartic or isoaspartic acid.
  • the parameters affecting the rate of deamidation include pH, temperature, solvent dielectric constant, ionic strength, primary sequence, local polypeptide conformation and tertiary structure.
  • the amino acid residues adjacent to Asn in the peptide chain affect deamidation rates. Gly and Ser following an Asn in protein sequences results in a higher susceptibility to deamidation.
  • the liquid formulation of the present disclosure may be preserved under conditions of pH and humidity to prevent deamination of the protein product.
  • the aqueous carrier of interest herein is one which is pharmaceutically acceptable (safe and non-toxic for administration to a human) and is useful for the preparation of a liquid formulation.
  • Illustrative carriers include sterile water for injection (SWFI), bacteriostatic water for injection (BWFI), a pH buffered solution (e.g., phosphate-buffered saline), sterile saline solution, Ringer's solution or dextrose solution.
  • a preservative may be optionally added to the formulations herein to reduce bacterial action.
  • the addition of a preservative may, for example, facilitate the production of a multi-use (multiple-dose) formulation.
  • Intravenous (IV) formulations may be the preferred administration route in particular instances, such as when a patient is in the hospital after transplantation receiving all drugs via the IV route.
  • the liquid formulation is diluted with 0.9% sodium chloride solution before administration.
  • the diluted drug product for injection is isotonic and suitable for administration by intravenous infusion.
  • a salt or buffer components may be added in an amount of 10 mM-200 mM.
  • the salts and/or buffers are pharmaceutically acceptable and are derived from various known acids (inorganic and organic) with “base forming” metals or amines.
  • the buffer may be phosphate buffer.
  • the buffer may be glycinate, carbonate, citrate buffers, in which case, sodium, potassium or ammonium ions can serve as counterion.
  • a preservative may be optionally added to the formulations herein to reduce bacterial action.
  • the addition of a preservative may, for example, facilitate the production of a multi-use (multiple-dose) formulation.
  • the aqueous carrier of interest herein is one which is pharmaceutically acceptable (safe and non-toxic for administration to a human) and is useful for the preparation of a liquid formulation.
  • Illustrative carriers include sterile water for injection (SWFI), bacteriostatic water for injection (BWFI), a pH buffered solution (e.g., phosphate-buffered saline), sterile saline solution, Ringer's solution or dextrose solution.
  • the protein of the present disclosure could exist in a lyophilized formulation including the proteins and a lyoprotectant.
  • the lyoprotectant may be sugar, e.g., disaccharides.
  • the lyoprotectant may be sucrose or maltose.
  • the lyophilized formulation may also include one or more of a buffering agent, a surfactant, a bulking agent, and/or a preservative.
  • the amount of sucrose or maltose useful for stabilization of the lyophilized drug product may be in a weight ratio of at least 1:2 protein to sucrose or maltose.
  • the protein to sucrose or maltose weight ratio may be of from 1:2 to 1:5.
  • the pH of the formulation, prior to lyophilization may be set by addition of a pharmaceutically acceptable acid and/or base.
  • the pharmaceutically acceptable acid may be hydrochloric acid.
  • the pharmaceutically acceptable base may be sodium hydroxide.
  • the pH of the solution containing the protein of the present disclosure may be adjusted between 6 to 8.
  • the pH range for the lyophilized drug product may be from 7 to 8.
  • a salt or buffer components may be added in an amount of 10 mM-200 mM.
  • the salts and/or buffers are pharmaceutically acceptable and are derived from various known acids (inorganic and organic) with “base forming” metals or amines.
  • the buffer may be phosphate buffer.
  • the buffer may be glycinate, carbonate, citrate buffers, in which case, sodium, potassium or ammonium ions can serve as counterion.
  • a “bulking agent” may be added.
  • a “bulking agent” is a compound which adds mass to a lyophilized mixture and contributes to the physical structure of the lyophilized cake (e.g., facilitates the production of an essentially uniform lyophilized cake which maintains an open pore structure).
  • Illustrative bulking agents include mannitol, glycine, polyethylene glycol and sorbitol. The lyophilized formulations of the present invention may contain such bulking agents.
  • a preservative may be optionally added to the formulations herein to reduce bacterial action.
  • the addition of a preservative may, for example, facilitate the production of a multi-use (multiple-dose) formulation.
  • the lyophilized drug product may be constituted with an aqueous carrier.
  • the aqueous carrier of interest herein is one which is pharmaceutically acceptable (e.g., safe and non-toxic for administration to a human) and is useful for the preparation of a liquid formulation, after lyophilization.
  • diluents include SWFI, BWFI, a pH buffered solution (e.g., phosphate-buffered saline), sterile saline solution, Ringer's solution or dextrose solution.
  • the lyophilized drug product of the current disclosure is reconstituted with either SWFI, USP or 0.9% sodium chloride Injection, USP. During reconstitution, the lyophilized powder dissolves into a solution.
  • the lyophilized protein product of the instant disclosure is constituted to about 4.5 mL water for injection and diluted with 0.9% saline solution (sodium chloride solution).
  • Actual dosage levels of the active ingredients in the pharmaceutical compositions of this invention may be varied so as to obtain an amount of the active ingredient which is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient.
  • the specific dose can be a uniform dose for each patient, for example, 50-5000 mg of protein.
  • a patient's dose can be tailored to the approximate body weight or surface area of the patient.
  • Other factors in determining the appropriate dosage can include the disease or condition to be treated or prevented, the severity of the disease, the route of administration, and the age, sex and medical condition of the patient. Further refinement of the calculations necessary to determine the appropriate dosage for treatment is routinely made by those skilled in the art, especially in light of the dosage information and assays disclosed herein.
  • the dosage can also be determined through the use of known assays for determining dosages used in conjunction with appropriate dose-response data. An individual patient's dosage can be adjusted as the progress of the disease is monitored.
  • Blood levels of the targetable construct or complex in a patient can be measured to see if the dosage needs to be adjusted to reach or maintain an effective concentration.
  • Pharmacogenomics may be used to determine which targetable constructs and/or complexes, and dosages thereof, are most likely to be effective for a given individual (Schmitz et al., Clinica Chimica Acta 308: 43-53, 2001; Steimer et al., Clinica Chimica Acta 308: 33-41, 2001).
  • dosages based on body weight are from about 0.01 ⁇ g to about 100 mg per kg of body weight, such as about 0.01 ⁇ g to about 100 mg/kg of body weight, about 0.01 ⁇ g to about 50 mg/kg of body weight, about 0.01 ⁇ g to about 10 mg/kg of body weight, about 0.01 ⁇ g to about 1 mg/kg of body weight, about 0.01 ⁇ g to about 100 ⁇ g/kg of body weight, about 0.01 ⁇ g to about 50 ⁇ g/kg of body weight, about 0.01 ⁇ g to about 10 ⁇ g/kg of body weight, about 0.01 ⁇ g to about 1 ⁇ g/kg of body weight, about 0.01 ⁇ g to about 0.1 ⁇ g/kg of body weight, about 0.1 ⁇ g to about 100 mg/kg of body weight, about 0.1 ⁇ g to about 50 mg/kg of body weight, about 0.1 ⁇ g to about 10 mg/kg of body weight, about 0.1 ⁇ g to about 1 mg/kg of body weight, about 0.1 ⁇ g to about
  • Doses may be given once or more times daily, weekly, monthly or yearly, or even once every 2 to 20 years. Persons of ordinary skill in the art can easily estimate repetition rates for dosing based on measured residence times and concentrations of the targetable construct or complex in bodily fluids or tissues.
  • Administration of the present invention could be intravenous, intraarterial, intraperitoneal, intramuscular, subcutaneous, intrapleural, intrathecal, intracavitary, by perfusion through a catheter or by direct intralesional injection. This may be administered once or more times daily, once or more times weekly, once or more times monthly, and once or more times annually.
  • the heterodimeric Fc-fused protein (e.g., a heterodimeric Fc-fused protein comprising IL-12 subunits) is administered intratumorally. In some embodiments, the heterodimeric Fc-fused protein is administered intratumorally at a lower dose or frequency than when administered systemically. In some embodiments, the heterodimeric Fc-fused protein is administered intratumorally following administration of a local anesthetic. In some embodiments, the heterodimeric Fc-fused protein is administered intratumorally under direct palpation of a tumor mass.
  • the invention provides methods for treating cancer using a heterodimeric Fc-fused binding protein described herein and/or a pharmaceutical composition described herein.
  • the methods may be used to treat a variety of cancers by administering to a patient in need thereof an effective amount of a heterodimeric Fc-fused protein described herein.
  • the cancer that is treated by a method disclosed herein is a locally advanced malignancy.
  • the locally advanced malignancy can be fully resected.
  • the locally advanced malignancy has been fully resected, and the treatment is provided subsequent to the resection.
  • a heterodimeric Fc-fused protein of the present invention (e.g., a heterodimeric Fc-fused protein comprising IL-12 subunits) is used for treating an advanced malignancy as a monotherapy.
  • a heterodimeric Fc-fused protein of the present invention (e.g., a heterodimeric Fc-fused protein comprising IL-12 subunits) is used as an adjuvant for active immunotherapy of severe infectious diseases.
  • a heterodimeric Fc-fused protein of the present invention (e.g., a heterodimeric Fc-fused protein comprising IL-12 subunits) is used as an adjuvant for prophylactic vaccination.
  • a heterodimeric Fc-fused protein of the present invention is used for treating myelosuppression following radiotherapy.
  • the present invention also provides a method of reducing hematopoietic toxicity.
  • Hematopoietic toxicity can result from genetic, infective, or environmental causes, including but not limited to irradiation and chemotherapy.
  • the present invention provides a method of treating a disease or disorder associated with radiation (e.g., ionizing radiation, alpha radiation, beta radiation, gamma radiation, X radiation, or neutron radiation).
  • a disease or disorder associated with radiation e.g., ionizing radiation, alpha radiation, beta radiation, gamma radiation, X radiation, or neutron radiation.
  • the present invention provides a method of treating myelosuppression following a radiation therapy, the method comprising administering to a patient in need thereof an effective amount of a heterodimeric Fc-fused protein or a formulation described herein.
  • the dose of the radiation therapy is at least 1, 5, 10, 15, or 20 Gy.
  • the radiation therapy causes damage in a system, organ, or tissue selected from the group consisting of bone marrow, lymphatic system, immune system, mucosal tissue, mucosal immune system, gastrointestinal system, cardiovascular system, nervous system, reproductive organs, prostate, ovaries, lung, kidney, skin and brain.
  • the method of the present invention reduces the damage.
  • the present invention provides a method of treating myelosuppression occurring in the context of an accidental exposure to radiation, the method comprising administering to a patient in need thereof a therapeutically effective amount of a heterodimeric Fc-fused protein or a formulation described herein.
  • the present invention provides a method of treating acute radiation syndrome (ARS), the method comprising administering to a patient in need thereof an effective amount of a heterodimeric Fc-fused protein or a formulation described herein.
  • ARS includes but is not limited to hematopoietic radiation syndrome, gastrointestinal radiation syndrome, neurovascular radiation syndrome, and cutaneous radiation syndrome.
  • hematopoietic radiation syndrome results from, at least in part, depletion of the hematopoietic stem cell pool and shows signs of lymphopenia and granulocytopenia.
  • Gastrointestinal syndrome results from, at least in part, damage of stem cells and progenitor cells located in the crypts and failure to replace the cells in the surface of the villi and shows signs of watery diarrhea, dehydration, electrolyte loss, gastrointestinal bleeding, and perforation.
  • the method of treatment provided herein is conducted at the prodromal phase of the ARS.
  • the prodromal phase is the initial phase of acute illness, characterized by the symptoms of nausea, vomiting, anorexia, fever, headache, and/or early skin erythema, typically within 1-3 days after the exposure to radiation.
  • the method of treatment provided herein is conducted at the latent phase of the ARS.
  • the latent phase is a phase characterized by improvement of symptoms but exhibition of lymphopenia and granulocytopenia in lab tests, and may last hours to weeks depending on the dose of exposure.
  • Treatment in the prodromal phase or latent phase may mitigate the development of the syndromes in the affected systems, organs, and/or tissues.
  • the method of treatment provided herein is conducted at the manifest illness phase of the ARS. Treatment in this phase may still promote recovery from the ARS.
  • the present invention also provides a method of increasing the survival, proliferation, differentiation, and/or activity of an immune cell, the method comprising contacting the immune cell with a heterodimeric Fc-fused protein or a formulation disclosed herein.
  • the immune cell is a T cell (e.g., CD4 + T cells).
  • the immune cell is an NK cell.
  • the heterodimeric Fc-fused protein of the present invention (e.g., a heterodimeric Fc-fused protein comprising IL-12 subunits) is used in treating a subject diagnosed with cancer.
  • the cancer is bladder cancer, breast cancer, cervical cancer, colorectal cancer, esophageal cancer, gastric cancer, head and neck cancer, hepatocellular cancer, leukemia, lung cancer, lymphoma, mesothelioma, melanoma, myeloma, ovarian cancer, endometrial cancer, prostate cancer, pancreatic cancer, renal cell cancer, non-small cell lung cancer, small cell lung cancer, brain cancer, sarcoma, neuroblastoma, or squamous cell carcinoma of the head and neck cancerous cells.
  • the cancer is colon cancer. In certain embodiments, the heterodimeric Fc-fused protein is administered as a monotherapy to a subject diagnosed with colon cancer. In certain embodiments, the cancer is melanoma. In certain embodiments, the heterodimeric Fc-fused protein is administered as a monotherapy to a subject diagnosed with melanoma. In certain embodiments, the cancer is breast cancer. In certain embodiments, the heterodimeric Fc-fused protein is administered as a monotherapy to a subject diagnosed with breast cancer.
  • the present disclosure provides a method of treating cancer, the method comprising administering to a patient only a single dose of a heterodimeric IL-12-Fc-fused protein.
  • the single dose is in an amount sufficient to induce a complete response to the cancer.
  • the single dose is in an amount sufficient to delay or prevent recurrence of the cancer.
  • the present disclosure provides a method of treating cancer, the method comprising administering to a patient only a single dose of a heterodimeric IL-12-Fc-fused protein comprising a first polypeptide comprising the amino acid sequence of SEQ ID NO:290 and a second polypeptide comprising the amino acid sequence of SEQ ID NO:291, or a formulation comprising the heterodimeric IL-12-Fc-fused protein and a pharmaceutically acceptable carrier.
  • a single dose of a heterodimeric IL-12-Fc-fused protein comprising a first polypeptide comprising the amino acid sequence of SEQ ID NO:290 and a second polypeptide comprising the amino acid sequence of SEQ ID NO:291, or a formulation comprising the heterodimeric IL-12-Fc-fused protein (e.g., comprising SEQ ID NO:290 and SEQ ID NO:291) and a pharmaceutically acceptable carrier is in an amount sufficient to induce a complete response to the cancer.
  • the single dose is in an amount sufficient to delay or prevent recurrence of the cancer.
  • a recurrence of a completely treated cancer is delayed or prevented by 6 months to 72 months or more (e.g., 6 months, 12 months, 24 months, 36 months, 48 months, 60 months, 72 months, 84 months, or 96 months).
  • the cancer treated with a single dose or more of a heterodimeric IL-12-Fc-fused protein is a metastatic cancer.
  • the metastatic cancer is a local, regional, or distant metastatic cancer.
  • a single or multiple dose of a heterodimeric IL-12-Fc-fused protein treats a distant cancer, which is not the primary cancer of the source organ or tissue and/or the direct target of a treatment regimen, by an abscopal effect.
  • a heterodimeric IL-12-Fc-fused protein treats a distant cancer, which is not the primary cancer of the source organ or tissue and/or the direct target of a treatment regimen, by an abscopal effect.
  • the abscopal effect of a heterodimeric IL-12-Fc-fused protein is enhanced during and/or after a treatment plan including radiation and/or chemotherapy.
  • a single or multiple dose of a heterodimeric IL-12-Fc-fused protein treats cancer in a patient by inducing a systemic anti-tumor response, determined, for example, by increased expression of IFN ⁇ , CXCL9, and/or CXCL10 in the serum and/or the tumor of the patient.
  • a multi-specific binding protein described herein can be used in combination with additional therapeutic agents to treat the cancer.
  • the heterodimeric Fc-fused protein of the present invention is administered as a combination therapy to treat a subject diagnosed with cancer.
  • the cancer is bladder cancer, breast cancer, cervical cancer, colorectal cancer, esophageal cancer, gastric cancer, head and neck cancer, hepatocellular cancer, leukemia, lung cancer, lymphoma, mesothelioma, melanoma, myeloma, ovarian cancer, endometrial cancer, prostate cancer, pancreatic cancer, renal cell cancer, non-small cell lung cancer, small cell lung cancer, brain cancer, sarcoma, neuroblastoma, or squamous cell carcinoma of the head and neck cancerous cells.
  • the cancer is colon cancer. In certain embodiments, the heterodimeric Fc-fused protein is administered as a combination therapy to a subject diagnosed with colon cancer. In certain embodiments, the cancer is melanoma. In certain embodiments, the heterodimeric Fc-fused protein is administered as a combination therapy to a subject diagnosed with melanoma. In certain embodiments, the cancer is breast cancer. In certain embodiments, the heterodimeric Fc-fused protein is administered as a combination therapy to a subject diagnosed with breast cancer.
  • a heterodimeric Fc-fused protein of the present invention (e.g., a heterodimeric Fc-fused protein comprising IL-12 subunits) is used in treating an advanced malignancy in combination with another therapeutic agent selected from: cytotoxic chemotherapy; radiotherapy; an antibody that targets a molecule involved in an anti-tumor immune response, such as CTLA-4, PD-1, PD-L 1 , or TGF- ⁇ ; an antibody that acts by ADCC on a tumor-associated antigen; a multispecific antibody binding NKG2D, CD16, and a tumor-associated antigen, optionally administered in combination with an antibody that targets PD-1 or PD-L1; a personalized cancer vaccine; an oncolytic cancer vaccine; and a personalized vaccine administered in combination with an antibody that targets PD-1 or PD-L1.
  • another therapeutic agent selected from: cytotoxic chemotherapy; radiotherapy; an antibody that targets a molecule involved in an anti-tumor immune response, such as CTLA-4, PD-1, PD-L
  • a heterodimeric Fc-fused protein of the present invention is used in treating malignancy (e.g., an advanced malignancy) in combination with another therapy including, but not limited to, an NK-targeting therapy (e.g., CAR-NK therapy), an antibody therapy, a checkpoint inhibitor therapy, an additional cytokine therapy, an innate immune system agonist therapy, a chemotherapy, a target agent therapy, a radiotherapy, an adoptive NK therapy, a stem cell transplant (SCT) therapy, an agonistic antibody, a chimeric antigen receptor (CAR) T cell therapy, a T-cell receptor (TCR) engineered therapy, a multi-specific binding protein (TriNKET), an agent that induces cellular senescence, and a vaccine and/or oncolytic virus therapy.
  • an NK-targeting therapy e.g., CAR-NK therapy
  • an antibody therapy e.g., a checkpoint inhibitor therapy
  • an additional cytokine therapy e.g., an
  • a heterodimeric Fc-fused protein of the present invention is used in treating malignancy (e.g., an advanced malignancy) in combination with two or more additional therapies selected from an NK-targeting therapy (e.g., CAR-NK therapy), an antibody therapy, a checkpoint inhibitor therapy, an additional cytokine therapy, an innate immune system agonist therapy, a chemotherapy, a target agent therapy, a radiotherapy, an adoptive NK therapy, a stem cell transplant (SCT) therapy, an agonistic antibody, a chimeric antigen receptor (CAR) T cell therapy, a T-cell receptor (TCR) engineered therapy, a multi-specific binding protein (TriNKET), an agent that induces cellular senescence, and a vaccine and/or oncolytic virus therapy.
  • an NK-targeting therapy e.g., CAR-NK therapy
  • an antibody therapy e.g., a checkpoint inhibitor therapy
  • an additional cytokine therapy e.g., an cytokin
  • a heterodimeric Fc-fused protein of the present invention e.g., a heterodimeric Fc-fused protein comprising IL-12
  • a heterodimeric Fc-fused protein comprising IL-12 is used in treating locally advanced malignancy that can be fully resected, in combination with a cancer vaccine or an antibody that targets PD-1 or PD-L1.
  • Proteins of the invention can also be used as an adjunct to surgical removal of the primary lesion.
  • heterodimeric Fc-fused protein of the present invention e.g., a heterodimeric Fc-fused protein comprising IL-12
  • additional therapeutic agent and the relative timing of administration may be selected in order to achieve a desired combined therapeutic effect.
  • the therapeutic agents in the combination, or a pharmaceutical composition or compositions comprising the therapeutic agents may be administered in any order such as, for example, sequentially, concurrently, together, simultaneously and the like.
  • a heterodimeric Fc-fused protein may be administered during a time when the additional therapeutic agent(s) exerts its prophylactic or therapeutic effect, or vice versa.
  • the methods of the invention include coadministration of the combination of a heterodimeric Fc-fused protein (e.g., a heterodimeric Fc-fused protein comprising IL-12 subunits) and an additional therapeutic agent.
  • a heterodimeric Fc-fused protein e.g., a heterodimeric Fc-fused protein comprising IL-12 subunits
  • the methods of the invention include coadministration of the combination of a heterodimeric Fc-fused protein comprising IL-12 subunits and an additional therapeutic agent.
  • Coadministered encompasses methods where a heterodimeric Fc-fused protein (e.g., a heterodimeric Fc-fused protein comprising IL-12 subunits) and an additional therapeutic agent are given simultaneously, where a heterodimeric Fc-fused protein and an additional therapeutic agent are given sequentially, and where either one of, or both of, a heterodimeric Fc-fused protein and an additional therapeutic agent are given intermittently or continuously, or any combination of: simultaneously, sequentially, intermittently and/or continuously.
  • intermittent administration is not necessarily the same as sequential because intermittent also includes a first administration of an agent and then another administration later in time of that very same agent.
  • intermittent administration also encompasses sequential administration in some embodiments because intermittent administration does include interruption of the first administration of an agent with an administration of a different agent before the first agent is administered again. Further, the skilled artisan will also know that continuous administration can be accomplished by a number of routes including intravenous drip (IV infusion) or feeding tubes, etc.
  • IV infusion intravenous drip
  • feeding tubes etc.
  • the term “coadministered” encompasses any and all methods where the individual administration of a heterodimeric Fc-fused protein and the individual administration of an additional therapeutic agent to a subject overlap during any timeframe.
  • the frequency of administration of a heterodimeric Fc-fused protein or an additional therapeutic agent to a subject is known in the art as Qnd or qnd where n is the frequency in days for successive administration of that agent.
  • Q3d would be an administration of an agent once every three (3) days.
  • the method comprises administering either one of, or both of, or any combinations thereof, a heterodimeric Fc-fused protein and/or an additional therapeutic agent to a subject for Q1d, Q2d, Q3d, Q4d, Q5d, Q6d, Q7d, Q8d, Q9d, Q10d, Q14d, Q21d, Q28d, Q30d, Q90d, Q120d, Q240d, or Q365d.
  • either one of or both of a heterodimeric Fc-fused protein and/or an additional therapeutic agent are administered intermittently.
  • the method includes administering either one of, or both of a heterodimeric Fc-fused protein or an additional therapeutic agent to a subject with a delay of at least ten (10) minutes, fifteen (15) minutes, twenty (20) minutes, thirty (30) minutes, forty (40) minutes, sixty (60) minutes, two (2) hours, three (3) hour, four (4) hours, six (6) hours, eight (8) hours, ten (10) hours, twelve (12) hours, fourteen (14) hours, eighteen (18) hours, twenty-four (24) hours, thirty-six (36) hours, forty-eight (48) hours, three (3) days, four (4) days, five (5) days, six (6) days, seven (7) days, eight (8) days, nine (9) days, ten (10) days, eleven (11) days, twelve (12) days, thirteen (13) days, fourteen (14) days, three (3) weeks, or four (4) weeks between administrations
  • the administration with a delay follows a pattern where one of, or both of, or any combination thereof, of a heterodimeric Fc-fused protein and/or an additional therapeutic agent are administered continuously for a given period of time from about ten (10) minutes to about three hundred and sixty five (365) days and then is not administered for a given period of time from about ten (10) minutes to about thirty (30) days.
  • either one of, or any combination of, a heterodimeric Fc-fused protein and/or an additional therapeutic agent are administered intermittently while the other is given continuously.
  • the combination of the first effective amount of a heterodimeric Fc-fused protein is administered sequentially with the second effective amount of an additional therapeutic agent.
  • a heterodimeric Fc-fused protein and an additional therapeutic agent are administered simultaneously.
  • the combination of the first effective amount of a heterodimeric Fc-fused protein is administered sequentially with the second effective amount of an additional therapeutic agent.
  • the combination is also said to be “coadministered” since the term includes any and all methods where the subject is exposed to both components in the combination.
  • such embodiments are not limited to the combination being given just in one formulation or composition. It may be that certain concentrations of a heterodimeric Fc-fused protein and the additional therapeutic agent are more advantageous to deliver at certain intervals and as such, the first effective amount and second effective amount may change according to the formulation being administered.
  • a heterodimeric Fc-fused protein and the additional therapeutic agent are administered simultaneously or sequentially.
  • the first effective amount of a heterodimeric Fc-fused protein is administered sequentially after the second effective amount of an additional therapeutic agent.
  • the second effective amount of an additional therapeutic agent is administered sequentially after the first effective amount of a heterodimeric Fc-fused protein.
  • the combination of a heterodimeric Fc-fused protein e.g., a heterodimeric Fc-fused protein comprising IL-12 subunits
  • an additional therapeutic agent is administered in one formulation.
  • the combination is administered in two (2) compositions where the first effective amount of a heterodimeric Fc-fused protein is administered in a separate formulation from the formulation of the second effective amount of an additional therapeutic agent.
  • the combination is administered in two (2) compositions where the first effective amount of the heterodimeric Fc-fused protein is administered in a separate formulation from the formulation of the second effective amount of an additional therapeutic agent.
  • the first effective amount of a heterodimeric Fc-fused protein is administered sequentially after the second effective amount of an additional therapeutic agent.
  • the second effective amount of an additional therapeutic agent is administered sequentially after the first effective amount of a heterodimeric Fc-fused protein.
  • a heterodimeric Fc-fused protein and the additional therapeutic agent are administered; and subsequently both the heterodimeric Fc-fused protein and the additional therapeutic agent are administered intermittently for at least twenty-four (24) hours.
  • the heterodimeric Fc-fused protein and the additional therapeutic agent are administered on a non-overlapping every other day schedule.
  • the first effective amount of a heterodimeric Fc-fused protein is administered no less than four (4) hours after the second effective amount of an additional therapeutic agent.
  • the first effective amount of a heterodimeric Fc-fused protein is administered no less than ten (10) minutes, no less than fifteen (15) minutes, no less than twenty (20) minutes, no less than thirty (30) minutes, no less than forty (40) minutes, no less than sixty (60) minutes, no less than one (1) hour, no less than two (2) hours, no less than four (4) hours, no less than six (6) hours, no less than eight (8) hours, no less than ten (10) hours, no less than twelve (12) hours, no less than twenty four (24) hours, no less than two (2) days, no less than four (4) days, no less than six (6) days, no less than eight (8) days, no less than ten (10) days, no less than twelve (12) days, no less than fourteen (14) days, no less than twenty one (21) days, or no less than thirty (30) days after
  • the second effective amount of an additional therapeutic agent is administered no less than ten (10) minutes, no less than fifteen (15) minutes, no less than twenty (20) minutes, no less than thirty (30) minutes, no less than forty (40) minutes, no less than sixty (60) minutes, no less than one (1) hour, no less than two (2) hours, no less than four (4) hours, no less than six (6) hours, no less than eight (8) hours, no less than ten (10) hours, no less than twelve (12) hours, no less than twenty four (24) hours, no less than two (2) days, no less than four (4) days, no less than six (6) days, no less than eight (8) days, no less than ten (10) days, no less than twelve (12) days, no less than fourteen (14) days, no less than twenty one (21) days, or no less than thirty (30) days after the first effective amount of a heterodimeric Fc-fused protein.
  • either one of, or both of a heterodimeric Fc-fused protein and/or additional therapeutic agent are administered by a route selected from the group consisting of: intravenous, subcutaneous, cutaneous, oral, intramuscular, and intraperitoneal.
  • a route selected from the group consisting of: intravenous, subcutaneous, cutaneous, oral, intramuscular, and intraperitoneal are administered by intravenously.
  • a heterodimeric Fc-fused protein and/or additional therapeutic agent are administered orally.
  • the unit dose forms of the present disclosure may be administered in the same or different physical forms, i.e. orally via capsules or tablets and/or by liquid via IV infusion, and so on.
  • the unit dose forms for each administration may differ by the particular route of administration.
  • Several various dosage forms may exist for either one of, or both of, the combination of a heterodimeric Fc-fused protein and additional therapeutic agents. Because different medical conditions can warrant different routes of administration, the same components of the combination described herein may be exactly alike in composition and physical form and yet may need to be given in differing ways and perhaps at differing times to alleviate the condition.
  • a condition such as persistent nausea, especially with vomiting, can make it difficult to use an oral dosage form, and in such a case, it may be necessary to administer another unit dose form, perhaps even one identical to other dosage forms used previously or afterward, with an inhalation, buccal, sublingual, or suppository route instead or as well.
  • the specific dosage form may be a requirement for certain combinations of a heterodimeric Fc-fused protein and additional therapeutic agents, as there may be issues with various factors like chemical stability or pharmacokinetics.
  • the heterodimeric Fc-fused protein therapy is combined with NK targeting therapies.
  • the heterodimeric Fc-fused protein is coadministered with a therapeutic agent that targets NKp46.
  • the therapeutic agent that targets NKp46 also binds CD16, one or more tumor-associated antigens, or a combination thereof. Exemplary therapeutic agents that target NKp46 are described in more detail in U.S. Application No. US20170198038A1, herein incorporated by reference for all purposes.
  • the heterodimeric Fc-fused protein therapy is combined with bi- and tri-specific killer engagers (BiKEs and TriKEs) therapies, including BiKE and TriKE therapies targeting NK cells.
  • BiKEs and TriKEs are constructed from a single heavy (VH) and light (VL) chain of the variable region of each antibody of interest. VH and VL domains are joined by a short flexible polypeptide linker to prevent dissociation.
  • BiKEs and TriKEs are described in more detail in U.S. Application Nos. US20180282386A1 and US20180258396A1, herein incorporated by reference for all purposes.
  • BiKEs and TriKEs can contain a binding domain specific for an NK cell.
  • BiKE and TriKE therapies are used in combination with the heterodimeric Fc-fused protein therapy to treat subjects known or suspected of having High-risk Myelodysplastic Syndrome, Acute Myelogenous Leukemia, Systemic Mastocytosis, or Mast Cell Leukemia.
  • BiKE and TriKE therapies are administered as a single course of 3 weekly treatment blocks.
  • a treatment block comprises 4 consecutive 24-hour continuous infusions (approximately 96 hours) followed by a 72 hour break.
  • BiKE and TriKE therapies are administered at a dose of 5 ⁇ g/kg/day, 10 ⁇ g/kg/day, 25 ⁇ g/kg/day, 50 ⁇ g/kg/day, 100 ⁇ g/kg/day, or 200 ⁇ g/kg/day. In certain embodiments, BiKE and TriKE therapies are administered at a dose of at least 5 ⁇ g/kg/day, at least 10 ⁇ g/kg/day, at least 25 ⁇ g/kg/day, at least 50 ⁇ g/kg/day, at least 100 ⁇ g/kg/day, or at least 200 ⁇ g/kg/day. In certain embodiments, BiKE and TriKE therapies are administered at a dose of at least 1 ⁇ g/kg/day.
  • BiKE and TriKE therapies are administered at a dose of at least 5 ⁇ g/kg/day. In certain embodiments, BiKE and TriKE therapies are administered at a dose of at least 200 ⁇ g/kg/day. In certain embodiments, BiKE and TriKE therapies are administered at a dose of at least 500 ⁇ g/kg/day. In certain embodiments, BiKE and TriKE therapies are administered at a dose of at least 1000 ⁇ g/kg/day. In certain embodiments, BiKE and TriKE therapies are administered at a dose of 200 ⁇ g/kg/day or less. In certain embodiments, BiKE and TriKE therapies are administered at a dose of 500 ⁇ g/kg/day or less.
  • BiKE and TriKE therapies are administered at a dose of 1000 ⁇ g/kg/day or less. In certain embodiments, BiKE and TriKE therapies are administered at a dose of 1-200 ⁇ g/kg/day. In certain embodiments, BiKE and TriKE therapies are administered at a dose of 5-200 ⁇ g/kg/day. In certain embodiments, BiKE and TriKE therapies are administered at a dose of 1-500 ⁇ g/kg/day. In certain embodiments, BiKE and TriKE therapies are administered at a dose of 1-1000 ⁇ g/kg/day. In certain embodiments, BiKE and TriKE therapies are administered at a dose of 5-500 ⁇ g/kg/day.
  • BiKE and TriKE therapies are administered at a dose of 5-1000 ⁇ g/kg/day. In certain embodiments, BiKE and TriKE therapies are administered at a maximum-tolerated dose. In certain embodiments, BiKE and TriKE therapies are administered at less than maximum-tolerated dose.
  • TriNKET Multi-Specific Binding Protein
  • the heterodimeric Fc-fused protein therapy is combined with a therapy comprising a multi-specific binding protein, which comprises: (a) a first antigen-binding site that binds NKG2D; (b) a second antigen-binding site that binds a tumor-associated antigen; and (c) an antibody Fc domain or a portion thereof sufficient to bind CD16, or a third antigen-binding site that binds CD16 (“TriNKET”) (for example, multi-specific binding proteins comprising various NKG2D-binders and tumor-associated antigen-binding sites described in international publication no.
  • a therapy comprising a multi-specific binding protein, which comprises: (a) a first antigen-binding site that binds NKG2D; (b) a second antigen-binding site that binds a tumor-associated antigen; and (c) an antibody Fc domain or a portion thereof sufficient to bind CD16, or a third antigen-binding site that binds CD16 (“
  • tumor-associated antigens include, but are not limited to, HER2, CD20, CD33, B-cell maturation antigen (BCMA), EpCAM, CD2, CD19, CD25, CD30, CD38, CD40, CD52, CD70, CLL1/CLEC12A, FLT3, EGFR/ERBB1, IGF1R, HER3/ERBB3, HER4/ERBB4, MUC1, cMET, SLAMF7, PSCA, MICA, MICB, TRAILR1, TRAILR2, MAGE-A3, B7.1, B7.2, CTLA4, HLA-E, and PD-L1.
  • BCMA B-cell maturation antigen
  • EpCAM EpCAM
  • CD2 CD19, CD25, CD30, CD38
  • CD40 CD52
  • CD70 CD70
  • CLL1/CLEC12A FLT3, EGFR/ERBB1, IGF1R, HER3/ERBB3, HER4/ERBB4, MUC1, cMET
  • SLAMF7 PSCA
  • MICA MICA
  • the heterodimeric Fc-fused protein therapy is combined with a therapy comprising a dose of a multi-specific binding protein based on body weight.
  • doses of a multi-specific binding protein based on body weight are from about 0.01 ⁇ g to about 100 mg per kg of body weight, such as about 0.01 ⁇ g to about 100 mg/kg of body weight, about 0.01 ⁇ g to about 50 mg/kg of body weight, about 0.01 ⁇ g to about 10 mg/kg of body weight, about 0.01 ⁇ g to about 1 mg/kg of body weight, about 0.01 ⁇ g to about 100 ⁇ g/kg of body weight, about 0.01 ⁇ g to about 50 ⁇ g/kg of body weight, about 0.01 ⁇ g to about 10 ⁇ g/kg of body weight, about 0.01 ⁇ g to about 1 ⁇ g/kg of body weight, about 0.01 ⁇ g to about 0.1 ⁇ g/kg of body weight, about 0.1 ⁇ g to about 100 mg/kg of body weight, about 0.1 ⁇ g/kg of
  • the heterodimeric Fc-fused protein therapy is combined with a therapy comprising doses of a multi-specific binding protein given once or more times daily, weekly, monthly or yearly, or even once every 2 to 20 years.
  • a therapy comprising doses of a multi-specific binding protein given once or more times daily, weekly, monthly or yearly, or even once every 2 to 20 years.
  • Persons of ordinary skill in the art can easily estimate repetition rates for dosing based on measured residence times and concentrations of the targetable construct or complex in bodily fluids or tissues.
  • Administration of a multi-specific binding protein could be intravenous, intraarterial, intraperitoneal, intramuscular, subcutaneous, intrapleural, intrathecal, intracavitary, by perfusion through a catheter or by direct intralesional injection. This may be administered once or more times daily, once or more times weekly, once or more times monthly, and once or more times annually.
  • the heterodimeric Fc-fused protein therapy is combined with a CAR therapy.
  • chimeric antigen receptor or alternatively a “CAR” refers to a recombinant polypeptide construct comprising at least an extracellular antigen binding domain, a transmembrane domain and an intracellular signaling domain comprising a functional signaling domain derived from a stimulatory molecule (also referred to herein as a “primary signaling domain”).
  • the CAR comprises an extracellular antigen-binding site that binds tumor-associated antigen, a transmembrane domain, and an intracellular signaling domain comprising a primary signaling domain.
  • the CAR further comprises one or more functional signaling domains derived from at least one costimulatory molecule (also referred to as a “costimulatory signaling domain”).
  • the CAR comprises a chimeric fusion protein comprising a tumor-associated antigen-binding domain (e.g., tumor-associated antigen-binding scFv domain) comprising a heavy chain variable domain and a light chain variable domain as an extracellular antigen binding domain, a transmembrane domain, and an intracellular signaling domain comprising a primary signaling domain.
  • a tumor-associated antigen-binding domain e.g., tumor-associated antigen-binding scFv domain
  • a tumor-associated antigen-binding domain comprising a heavy chain variable domain and a light chain variable domain as an extracellular antigen binding domain, a transmembrane domain, and an intracellular signaling domain comprising a primary signaling domain.
  • the CAR comprises a chimeric fusion protein comprising a tumor-associated antigen-binding domain (e.g., tumor-associated antigen-binding scFv domain) comprising a heavy chain variable domain and a light chain variable domain as an extracellular antigen binding domain, a transmembrane domain and an intracellular signaling domain comprising a costimulatory signaling domain and a primary signaling domain.
  • a tumor-associated antigen-binding domain e.g., tumor-associated antigen-binding scFv domain
  • the CAR comprises a chimeric fusion protein comprising a tumor-associated antigen-binding domain (e.g., tumor-associated antigen-binding scFv domain) comprising a heavy chain variable domain and a light chain variable domain as an extracellular antigen binding domain, a transmembrane domain, and an intracellular signaling domain comprising two costimulatory signaling domains and a primary signaling domain.
  • a tumor-associated antigen-binding domain e.g., tumor-associated antigen-binding scFv domain
  • a tumor-associated antigen-binding domain comprising a heavy chain variable domain and a light chain variable domain as an extracellular antigen binding domain, a transmembrane domain, and an intracellular signaling domain comprising two costimulatory signaling domains and a primary signaling domain.
  • the CAR comprises a chimeric fusion protein comprising a tumor-associated antigen-binding domain comprising a heavy chain variable domain and a light chain variable domain as an extracellular antigen binding domain, a transmembrane domain, and an intracellular signaling domain comprising at least two costimulatory signaling domains and a primary signaling domain.
  • the CAR is designed to comprise a transmembrane domain that is fused to the extracellular domain of the CAR.
  • the transmembrane domain is one that naturally is associated with one of the domains in the CAR.
  • the transmembrane domain can be selected or modified by amino acid substitution to avoid binding of such domains to the transmembrane domains of the same or different surface membrane proteins to minimize interactions with other members of the receptor complex.
  • the transmembrane domain is capable of homodimerization with another CAR on the CAR T cell surface.
  • the amino acid sequence of the transmembrane domain may be modified or substituted so as to minimize interactions with the binding domains of the native binding partner present in the same CAR T cell.
  • the transmembrane domain may be derived from any naturally occurring membrane-bound or transmembrane protein.
  • the transmembrane region is capable of signaling to the intracellular domain(s) whenever the CAR has bound to a target.
  • the transmembrane domain comprises the transmembrane region(s) of one or more proteins selected from the group consisting of TCR ⁇ chain, TCR ⁇ chain, TCR chain, CD28, CD3E, CD45, CD4, CD5, CD8, CD9, CD16, CD22, CD33, CD37, CD64, CD80, CD86, CD134, CD137, and CD154.
  • the transmembrane domain comprises the transmembrane region(s) of one or more protein(s) selected from the group consisting of KIRDS2, OX40, CD2, CD27, LFA-1 (CD11a, CD18), ICOS (CD278), 4-1BB (CD137), GITR, CD40, BAFFR, HVEM (LIGHTR), SLAMF7, NKp80 (KLRF1), NKp44, NKp30, NKp46, CD160, CD19, IL2R ⁇ , IL2R ⁇ , IL7R ⁇ , ITGA1, VLA1, CD49a, ITGA4, IA4, CD49D, ITGA6, VLA-6, CD49f, ITGAD, CD11d, ITGAE, CD103, ITGAL, CD11a, LFA-1, ITGAM, CD11b, ITGAX, CD11c, ITGB1, CD29, ITGB2, CD18, LFA-1, ITGB7, TNFR2, DNAM1 (CD226)
  • the extracellular tumor-associated antigen-binding domain (e.g., tumor-associated antigen-binding scFv domain) can be connected to the transmembrane domain by a hinge region.
  • a hinge region can be employed, including but not limited to the human Ig hinge (e.g., an IgG4 hinge, an IgD hinge), a Gly-Ser linker, a (G45) 4 linker, a KIR2DS2 hinge, and a CD8a hinge.
  • the intracellular signaling domain of the CAR is responsible for activation of at least one of the specialized functions of the immune cell (e.g., cytolytic activity or helper activity, including the secretion of cytokines, of a T cell) in which the CAR has been placed in.
  • the term “intracellular signaling domain” refers to the portion of a protein which transduces an effector function signal and directs the cell to perform a specialized function. While usually the entire intracellular signaling domain can be employed, in many cases it is not necessary to use the entire chain.
  • intracellular signaling domain is thus meant to include any truncated portion of the intracellular signaling domain sufficient to transduce the effector function signal.
  • the intracellular signaling domain of the CAR comprises a primary signaling domain (i.e. a functional signaling domain derived from a stimulatory molecule) and one or more costimulatory signaling domains (i.e. functional signaling domains derived from at least one costimulatory molecule).
  • a primary signaling domain i.e. a functional signaling domain derived from a stimulatory molecule
  • costimulatory signaling domains i.e. functional signaling domains derived from at least one costimulatory molecule
  • the term “stimulatory molecule” refers to a molecule expressed by an immune cell, e.g., a T cell, an NK cell, or a B cell, that provide the cytoplasmic signaling sequence(s) that regulate activation of the immune cell in a stimulatory way for at least some aspect of the immune cell signaling pathway.
  • the signal is a primary signal that is initiated by, for instance, binding of a TCR/CD3 complex with an MHC molecule loaded with a peptide, and which leads to mediation of a T cell response, including, but not limited to, proliferation, activation, differentiation, and the like.
  • Primary signaling domains that act in a stimulatory manner may contain signaling motifs which are known as immunoreceptor tyrosine-based activation motifs or ITAMs.
  • ITAM containing cytoplasmic signaling sequences that are of particular use in the present disclosure include those derived from CD3 zeta, common FcR gamma (FCER1G), Fc gamma RIIa, FcR beta (Fc Epsilon Rib), CD3 gamma, CD3 delta, CD3 epsilon, CD79a, CD79b, DAP10, and DAP12.
  • the primary signaling domain in any one or more CARs comprises a cytoplasmic signaling sequence derived from CD3-zeta.
  • the primary signaling domain is a functional signaling domain of TCR zeta, FcR gamma, FcR beta, CD3 gamma, CD3 delta, CD3 epsilon, CD5, CD22, CD79a, CD79b, CD66d, 4-1BB, and/or CD3-zeta.
  • the intracellular signaling domain comprises a functional signaling domain of CD3 zeta, common FcR gamma (FCER1G), Fc gamma RIIa, FcR beta (Fc Epsilon Rib), CD3 gamma, CD3 delta, CD3 epsilon, CD79a, CD79b, DAP10, and/or DAP12.
  • the primary signaling domain is a functional signaling domain of the zeta chain associated with the T cell receptor complex.
  • costimulatory molecule refers to a cognate binding partner on a T cell that specifically binds with a costimulatory ligand, thereby mediating a costimulatory response by the T cell, such as, but not limited to, proliferation.
  • a costimulatory molecule is a cell surface molecule other than an antigen receptor or its ligands that is required for an efficient response of lymphocytes to an antigen.
  • Examples of such molecules include CD27, CD28, 4-1BB (CD137), OX40, CD30, CD40, PD-1, ICOS, lymphocyte function-associated antigen-1 (LFA-1, CD11a/CD18), CD2, CD7, CD258 (LIGHT), NKG2C, B7-H3, and a ligand that specifically binds with CD83, and the like.
  • costimulatory molecules include CD5, ICAM-1, GITR, BAFFR, HVEM (LIGHTR), SLAMF7, NKp80 (KLRF1), NKp44, NKp30, NKp46, CD160, CD19, CD4, CD8alpha, CD8beta, IL2R beta, IL2R gamma, IL7R alpha, ITGA4, VLA1, CD49a, ITGA4, IA4, CD49D, ITGA6, VLA-6, CD49f, ITGAD, CD11d, ITGAE, CD103, ITGAL, CD11a, LFA-1, ITGAM, CD11b, ITGAX, CD11c, ITGB1, CD29, ITGB2, CD18, LFA-1, ITGB7, NKG2D, NKG2C, TNFR2, TRANCE/RANKL, DNAM1 (CD226), SLAMF4 (CD244, 2B4), CD84, CD96 (Tactile), CEACAM1, CRTAM
  • the costimulatory signaling domain of the CAR is a functional signaling domain of a costimulatory molecule described herein, e.g., OX40, CD27, CD28, CD30, CD40, PD-1, CD2, CD7, CD258, NKG2C, B7-H3, a ligand that binds to CD83, ICAM-1, LFA-1 (CD11a/CD18), ICOS and 4-1BB (CD137), or any combination thereof.
  • a costimulatory molecule described herein e.g., OX40, CD27, CD28, CD30, CD40, PD-1, CD2, CD7, CD258, NKG2C, B7-H3, a ligand that binds to CD83, ICAM-1, LFA-1 (CD11a/CD18), ICOS and 4-1BB (CD137), or any combination thereof.
  • signaling domain refers to the functional portion of a protein which acts by transmitting information within the cell to regulate cellular activity via defined signaling pathways by generating second messengers or functioning as effectors by responding to such messengers.
  • cytoplasmic signaling sequences within the cytoplasmic signaling portion of the CAR may be linked to each other in a random or specified order.
  • a short oligo- or polypeptide linker for example, between 2 and 10 amino acids in length may form the linkage.
  • the heterodimeric Fc-fused protein therapy is combined with an antibody-therapy to treat subjects known or suspected of having cancer.
  • the heterodimeric Fc-fused protein is combined with a therapy comprising an anti-HER2 binding domain, such as an anti-HER2 antibody or anti-HER2 antibody platforms (e.g., a bi-specific or tri-specific antibody comprising an anti-HER2 binding domain, anti-HER2 antibody-drug conjugates, or anti-HER2 CAR).
  • Anti-HER2 antibodies include, but are not limited to, trastuzumab (HERCEPTIN®—Roche/Genentech; Kanjinti—Amgen), pertuzumab (PERJETA®—Roche/Genentech), and MGAH22 (described in detail in U.S. Pat. No. 8,802,093, herein incorporated by reference for all purposes).
  • Anti-HER2 antibody platforms include, but are not limited to, ertumaxomab (REXOMUN®—Creative Biolabs) and trastuzumab emtansine (ado-trastuzumab emtansine/T-DM1; KADCYLA®—Roche/Genentech).
  • the anti-HER2 binding domain therapy is used in combination with the heterodimeric Fc-fused protein therapy to treat subjects known or suspected of having cancer.
  • the anti-HER2 binding domain therapy is administered by IV infusion.
  • the anti-HER2 binding domain therapy is administered at a dose of 1 mg/kg/day, 2 mg/kg/day, 3 mg/kg/day, 4 mg/kg/day, 5 mg/kg/day, 6 mg/kg/day, 7 mg/kg/day, 8 mg/kg/day, 9 mg/kg/day, 10 mg/kg/day.
  • the anti-HER2 binding domain therapy is administered at a dose of at least 1 mg/kg/day, at least 2 mg/kg/day, at least 3 mg/kg/day, at least 4 mg/kg/day, at least 5 mg/kg/day, at least 6 mg/kg/day, at least 7 mg/kg/day, at least 8 mg/kg/day, at least 9 mg/kg/day, at least 10 mg/kg/day. In certain embodiments, the anti-HER2 binding domain therapy is administered at a dose of less than 1 mg/kg/day.
  • the anti-HER2 binding domain therapy is used in combination with the heterodimeric Fc-fused protein therapy to treat subjects known or suspected of having breast cancer, e.g., a subject diagnosed with metastatic HER2-overexpressing breast cancer.
  • the anti-HER2 binding domain therapy is administered at 4 mg/kg/day.
  • the anti-HER2 binding domain therapy is administered at 4 mg/kg/day by IV infusion over 90 minutes.
  • the anti-HER2 binding domain therapy is administered at 2 mg/kg/day.
  • the anti-HER2 binding domain therapy is administered at 2 mg/kg/day by IV infusion over 30 minutes.
  • the anti-HER2 binding domain therapy is administered at an initial dose of 4 mg/kg/day, then subsequently administered weekly at 2 mg/kg/day. In certain embodiments, the anti-HER2 binding domain therapy is administered at an initial dose of 4 mg/kg/day, then subsequently administered weekly at 2 mg/kg/day for 52 weeks.
  • the anti-HER2 binding domain therapy is used in combination with the heterodimeric Fc-fused protein therapy to treat subjects known or suspected of having gastric cancer, e.g., a subject diagnosed with metastatic HER2-overexpressing gastric cancer.
  • the anti-HER2 binding domain therapy is administered at 8 mg/kg/day.
  • the anti-HER2 binding domain therapy is administered at 8 mg/kg/day by IV infusion over 90 minutes.
  • the anti-HER2 binding domain therapy is administered at 6 mg/kg/day.
  • the anti-HER2 binding domain therapy is administered at 6 mg/kg/day by IV infusion over 30-90 minutes.
  • the anti-HER2 binding domain therapy is administered at an initial dose of 8 mg/kg/day, then subsequently administered weekly at 6 mg/kg/day. In certain embodiments, the anti-HER2 binding domain therapy is administered at an initial dose of 8 mg/kg/day, then subsequently administered weekly at 6 mg/kg/day for 52 weeks.
  • the heterodimeric Fc-fused protein therapy is combined with a therapy comprising an anti-CD20 binding domain, such as an anti-CD20 antibody or anti-CD20 antibody platforms (e.g., a bi-specific or tri-specific antibody comprising an anti-CD20 binding domain, anti-CD20 antibody-drug conjugates, or anti-CD20 CAR).
  • a therapy comprising an anti-CD20 binding domain, such as an anti-CD20 antibody or anti-CD20 antibody platforms (e.g., a bi-specific or tri-specific antibody comprising an anti-CD20 binding domain, anti-CD20 antibody-drug conjugates, or anti-CD20 CAR).
  • Anti-CD20 antibodies include, but are not limited to, rituximab (RITUXAN®—Roche/Genentech), ocrelizumab (OCREVUS®—Roche/Genentech), obinutuzumab (GAZYVA®—Roche/Genentech), ofatumumab (ARZERRA®—Novartis), and veltuzumab.
  • the anti-CD20 binding domain therapy is used in combination with the heterodimeric Fc-fused protein therapy to treat subjects known or suspected of having cancer.
  • the anti-CD20 binding domain therapy is administered by IV infusion. In certain embodiments, the anti-CD20 binding domain therapy is administered at a dose of 100 mg/m 2 , 200 mg/m 2 , 300 mg/m 2 , 400 mg/m 2 , 500 mg/m 2 , 600 mg/m 2 , 700 mg/m 2 , 800 mg/m 2 , 900 mg/m 2 , or 1000 mg/m 2 . In certain embodiments, the anti-CD20 binding domain therapy is administered at a dose of 375 mg/m 2 .
  • the anti-CD20 binding domain therapy is administered at a dose of at least 100 mg/m 2 , at least 200 mg/m 2 , at least 300 mg/m 2 , at least 400 mg/m 2 , at least 500 mg/m 2 , at least 600 mg/m 2 , at least 700 mg/m 2 , at least 800 mg/m 2 , at least 900 mg/m 2 , or at least 1000 mg/m 2 .
  • the anti-CD20 binding domain therapy is administered at a dose of less than 400 mg/m 2 .
  • the anti-CD20 binding domain therapy is administered at a dose of less than 375 mg/m 2 .
  • the anti-CD20 binding domain therapy is used in combination with the heterodimeric Fc-fused protein therapy to treat subjects known or suspected of having Non-Hodgkin's Lymphoma (NHL).
  • the anti-CD20 binding domain therapy is administered at a dose of 375 mg/m 2 by IV-infusion. In certain embodiments, the anti-CD20 binding domain therapy is administered at a dose less than 375 mg/m 2 by IV-infusion.
  • the anti-CD20 binding domain therapy is used in combination with the heterodimeric Fc-fused protein therapy to treat subjects known or suspected of having Chronic Lymphocytic Leukemia (CLL).
  • CLL Chronic Lymphocytic Leukemia
  • the anti-CD20 binding domain therapy is administered at a dose of 375 mg/m 2 by IV-infusion in a first cycle, and at a dose of 500 mg/m 2 by IV-infusion per cycle in an additional 2-6 cycles.
  • the anti-CD20 binding domain therapy is administered at a dose less than 375 mg/m 2 by IV-infusion.
  • the combined anti-CD20 binding domain and heterodimeric Fc-fused protein therapy can be used in combination with fludarabine and cyclophosphamide (FC).
  • the anti-CD20 binding domain therapy is used in combination with the heterodimeric Fc-fused protein therapy to treat subjects known or suspected of having Rheumatoid Arthritis (RA).
  • the anti-CD20 binding domain therapy is administered as two doses of 1000 mg, doses separated 2 weeks, by IV-infusion.
  • the anti-CD20 binding domain therapy is administered as two doses of 1000 mg, doses separated 2 weeks, by IV-infusion up to 24 weeks.
  • the combined anti-CD20 binding domain and heterodimeric Fc-fused protein therapy is coadministered with methotrexate.
  • the heterodimeric Fc-fused protein therapy is combined with a therapy comprising an antibody therapy comprising an agonist antibody.
  • the agonist antibody is an anti-4-1BB antibody, an anti-CD137 antibody, an anti-FAP antibody, an anti-OX40 antibody, an anti-CD40 antibody, an anti-GITR antibody, or an anti-CD27 antibody.
  • the agonist antibody is a bispecific antibody.
  • the agonist antibody is a multispecific antibody, e.g., abispecific antibody, comprising two or more antigen binding domains selected from an anti-4-1BB antibody, an anti-CD137 antibody, an anti-FAP antibody, an anti-OX40 antibody, an anti-CD40 antibody, an anti-GITR antibody, or an anti-CD27 antibody.
  • An illustrative example is a bispecific agonist antibody targeting 4-1BB and CD137, such as utomilumab (Pfizer).
  • the heterodimeric Fc-fused protein therapy can be combined with a checkpoint inhibitor therapy.
  • Illustrative immune checkpoint molecules that can be targeted for blocking or inhibition include, but are not limited to, CTLA-4, 4-1BB (CD137), 4-1BBL (CD137L), PDL1, PDL2, PD1, B7-H3, B7-H4, BTLA, HVEM, TIM3, GAL9, LAG3, TIM3, B7H3, B7H4, VISTA, KIR, 2B4 (belongs to the CD2 family of molecules and is expressed on all NK, ⁇ , and memory CD8+ ⁇ ( ⁇ ) T cells), CD160 (also referred to as BY55), and CGEN-15049.
  • Immune checkpoint inhibitors include antibodies, or antigen binding fragments thereof, or other binding proteins, that bind to and block or inhibit the activity of one or more of CTLA-4, PDL1, PDL2, PD1, B7-H3, B7-H4, BTLA, HVEM, TIM3, GAL9, LAG3, TIM3, B7H3, B7H4, VISTA, KIR, 2B4, CD160, and CGEN-15049.
  • Illustrative immune checkpoint inhibitors include Illustrative immune checkpoint inhibitors include nivolumamb (anti-PD-1; OPDIVO®—BMS), AMP224 (anti-PD-1; NCI), pembrolizumab (anti-PD-1; MK-3475/KEYTRUDA®—Merck), pidilizumab (anti-PD-1 antibody; CT-011—Teva/CureTech), atezolizumab (anti-PD-L1; TECENTRIQ®—Roche/Genentech), durvalumab (anti-PD-L1; MEDI4736/IMFINZI®—Medimmune/AstraZeneca), avelumab (anti-PD-L1; BAVENCIO®—Pfizer), BMS-936559 (anti-PD-L1—BMS), ipilimumab (anti-CTLA-4; YERVOY®—BMS), tremelimumab (anti-CTLA-4; Medimmune
  • the checkpoint inhibitor therapy is used in combination with the heterodimeric Fc-fused protein therapy to treat subjects known or suspected of having cancer.
  • the checkpoint inhibitor therapy is administered by IV infusion.
  • the checkpoint inhibitor therapy is administered by IV infusion over 30 minutes.
  • the checkpoint inhibitor therapy is administered every 3 weeks.
  • the checkpoint inhibitor therapy is administered at a dose of 100 mg, 200 mg, 300 mg, 400 mg, 500 mg, 600 mg, 700 mg, 800 mg, 900 mg, or 1000 mg.
  • the checkpoint inhibitor therapy is administered at a dose of 200 mg.
  • the checkpoint inhibitor therapy is administered at a dose of at least 100 mg, at least 200 mg, at least 300 mg, at least 400 mg, at least 500 mg, at least 600 mg, at least 700 mg, at least 800 mg, at least 900 mg, or at least 1000 mg. In certain embodiments, the checkpoint inhibitor therapy is administered at a dose of less than 200 mg.
  • the checkpoint inhibitor therapy is used in combination with the heterodimeric Fc-fused protein therapy to treat subjects known or suspected of having Melanoma, Non-Small Cell Lung Cancer (NSCLC), Head and Neck Squamous Cell Cancer (HNSCC), Classical Hodgkin Lymphoma (cHL), Primary Mediastinal Large B-Cell Lymphoma (PMBCL), Urothelial Carcinoma, Microsatellite Instability-High Cancer, Gastric Cancer, Cervical Cancer, Hepatocellular Carcinoma (HCC), Merkel Cell Carcinoma (MCC), Renal Cell Carcinoma (RCC).
  • NSCLC Non-Small Cell Lung Cancer
  • HNSCC Head and Neck Squamous Cell Cancer
  • cHL Classical Hodgkin Lymphoma
  • PMBCL Primary Mediastinal Large B-Cell Lymphoma
  • Urothelial Carcinoma Microsatellite Instability-High Cancer
  • Gastric Cancer Cervical Cancer
  • the heterodimeric Fc-fused protein therapy is combined with one or more additional cytokine therapies, one or more chemokine therapies, or combinations thereof. In some embodiments, the heterodimeric Fc-fused protein therapy is combined with one or more additional cytokine therapies. In some embodiments, the heterodimeric Fc-fused protein therapy is combined with one or more chemokine therapies.
  • the cytokine therapy comprises a pro-inflammatory cytokine, a Th1 cytokine, or a Th2 cytokine. In some embodiments, the cytokine therapy comprises a recombinant human cytokine or chemokine.
  • the cytokine therapy includes a cytokine that is an interleukin (e.g., IL-1, IL-2, IL-6, IL-7, IL-8, IL-12, IL-13, IL-15, IL-16, IL-18, IL-21 and IL-22).
  • the cytokine therapy includes a cytokine that is growth factor (e.g., tumor necrosis factor (TNF), LT, EMAP-II, GM-CSF, FGF and PDGF)
  • TNF tumor necrosis factor
  • LT LT
  • EMAP-II EMAP-II
  • GM-CSF GM-CSF
  • FGF fibroblast growth factor
  • PDGF e.g., PDGF
  • the cytokine therapy comprises an anti-inflammatory cytokine (e.g., IL-4, IL-10, IL-11, IL-13 and TGF).
  • the chemokine therapy includes a pro-inflammatory chemokine (e.g., GRO- ⁇ , GRO-b, LIX, GCP-2, MIG, IP10, I-TAC, and MCP-1, RANTES, Eotaxin, SDF-1, and MIP3a).
  • a pro-inflammatory chemokine e.g., GRO- ⁇ , GRO-b, LIX, GCP-2, MIG, IP10, I-TAC, and MCP-1, RANTES, Eotaxin, SDF-1, and MIP3a.
  • the chemokine therapy includes a chemokine receptor.
  • the chemokine therapy includes a CXC chemokine receptor (e.g., CXCR1, CXCR2, CXCR3, CXCR4, CXCR5, CXCR6, and CXCR7), a CC chemokine receptor (e.g., CCR1, CCR2, CCR3, CCR4, CCR5, CCR6, CCR7, CCR8, CCR9, CCR10, and CCR11), a CX3C chemokine receptor (e.g., CX3C11), or a XC chemokine receptor (e.g., XCR1).
  • the chemokine therapy comprises a G protein-linked transmembrane receptor.
  • the cytokine therapy comprises a cytokine therapy that synergizes with the IL-12 signaling.
  • the cytokine therapy comprises an IL-2 cytokine or a derivative thereof.
  • the IL-2 therapy is aldesleukin (Proleukin—Prometheus Therapeutics).
  • the IL-2 therapy and/or aldesleukin is administered intravenously.
  • the cytokine therapy comprises an IL-15 cytokine or a derivative thereof.
  • the IL-15 therapy is ALT-803 (Altor Bioscience) or NKTR-255 (Nektar).
  • the IL-15 therapy, NKTR-255, and/or ALT-803 is administered subcutaneously.
  • the chemokine therapy comprises a CXCL9 chemokine, a CXCL10 chemokine, or derivatives thereof.
  • the cytokine or chemokine therapy includes administering a cytokine or chemokine to a subject. In some embodiments, the cytokine or chemokine therapy includes administering a recombinant cytokine or chemokine to a subject. In some embodiments, the cytokine or chemokine therapy includes engineering a cell to produce the cytokine or chemokine. In some embodiments, the cytokine or chemokine therapy includes engineering a cell ex vivo, in vitro, or in vivo to produce the cytokine or chemokine.
  • the cytokine or chemokine therapy includes engineering a cell to produce the cytokine or chemokine using a viral vector-based delivery platform such as vaccinia, fowlpox, self-replicating alphavirus, marabavirus, adenovirus (See, e.g., Tatsis et al., Adenoviruses, Molecular Therapy (2004) 10, 616-629), a lentivirus, including but not limited to second, third or hybrid second/third generation lentivirus and recombinant lentivirus of any generation designed to target specific cell types or receptors (See, e.g., Hu et al., Immunization Delivered by Lentiviral Vectors for Cancer and Infectious Diseases, Immunol Rev.
  • a viral vector-based delivery platform such as vaccinia, fowlpox, self-replicating alphavirus, marabavirus, adenovirus (See, e.g., Tatsis et al., A
  • the cytokine or chemokine therapy includes engineering a cell to produce the cytokine or chemokine using a LNP, liposome, or an exosome.
  • the cytokine or chemokine therapy includes engineering a cell to produce the cytokine or chemokine using genome editing, such as using a nuclease-based genome editing systems (e.g., a Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) family, a Transcription activator-like effector nuclease (TALEN), a zinc-finger nuclease (ZFN), and a homing endonuclease (HE) based genome editing system or a derivative thereof).
  • CRISPR Clustered Regularly Interspaced Short Palindromic Repeats
  • TALEN Transcription activator-like effector nuclease
  • ZFN zinc-finger nuclease
  • HE homing endonuclease
  • the heterodimeric Fc-fused protein therapy is combined with one or more innate immune system agonists.
  • the innate immune system agonist comprises a toll-like receptor (TLR) agonist.
  • the TLR agonist comprises a TLR1/2, TLR2/6, TLR3, TLR4, TLR7, TLR8, TLR7/8, or TLR9 agonist.
  • a TLR2/6 agonist comprises lipoproteins, such as bacterial lipoproteins or derivatives, such as Pam2CSK4.
  • a TLR1/2 agonist comprises lipoproteins.
  • a TLR3 agonist comprises a dsRNA analog, such as rintatolimod (AMPLIGEN®—Hemispherx Biopharma) or poly IC-LC (e.g., HILTONOL®).
  • a TLR4 agonist comprises lipopolysaccharide (LPS, also referred to as endotoxin) or derivatives, such as lipid A.
  • a TLR7 agonist comprises a ssRNA or derivatives or imidazoquinoline derivatives including, but not limited to, resiquimod (also referred to as R848), imiquimod (ZYCLARA®, Aldara—Medicis), and gardiquimod.
  • a TLR7 agonist is also a TLR8 agonist, such as imiquimod or Medi-9197 (AstraZeneca/MedImmune).
  • a TLR9 agonist comprises a CpG-containing oligodeoxynucleotide (CpG-ODN) or SD-101 (Dynavax).
  • the innate immune system agonist comprises a Stimulator of interferon genes (STING) agonist.
  • the STING agonist comprises a cyclic-di-nucleotide (CDN).
  • the CDN comprises a cyclic-di-AMP, a cyclic-di-GMP, or a cyclic-GMP-AMP (cGAMP).
  • the STING agonist comprises a nucleic acid (e.g., DNA or RNA) that stimulates cGAS.
  • the STING agonist is ADU-S100 (also referred to as MIW815—Aduro/Novartis).
  • the heterodimeric Fc-fused protein therapy is combined with one or more chemotherapies. In certain embodiments, the heterodimeric Fc-fused protein therapy is combined with one or more chemotherapies to treat a subject diagnosed with cancer.
  • chemotherapy agents include aldesleukin, alvocidib, antineoplaston AS2-1, antineoplaston A10, anti-thymocyte globulin, amifostine trihydrate, aminocamptothecin, arsenic trioxide, beta alethine, Bcl-2 family protein inhibitor ABT-263, ABT-199, BMS-345541, bortezomib (VELCADE®), bryostatin 1, busulfan, carboplatin, campath-1H, CC-5103, carmustine, caspofungin acetate, clofarabine, cisplatin, Cladribine (LEUSTARIN®), Chlorambucil (LEUKERAN®), Curcumin, cyclosporine, Cyclophosphamide (Cyloxan, Endoxan, Endoxana, Cyclostin), cytarabine, denileukin diftitox, dexamethasone, DT PACE, docetaxe
  • the heterodimeric Fc-fused protein therapy is combined with one or more chemotherapies to treat a subject diagnosed with colon cancer, rectal cancer, or colorectal cancer (CRC).
  • the chemotherapy comprises FOLFOX (5-FU, leucovorin, and oxaliplatin/Eloxatin), FOLFIRI (leucovorin, 5-FU, and irinotecan/Camptosar), FOLFOXIRI (leucovorin, 5-FU, oxaliplatin, and irinotecan), CapeOx (capecitabine and oxaliplatin), 5-FU coadministered with leucovorin, capecitabine (XELODA®) alone, or Trifluridine and tipiracil (LONSURF®).
  • FOLFOX 5-FU, leucovorin, and oxaliplatin/Eloxatin
  • FOLFIRI leucovorin, 5-FU, and irinotecan/Camptosar
  • the chemotherapy comprises a VEGF targeting agent, such as bevacizumab (AVASTIN®), ziv-aflibercept (ZALTRAP®), ramucirumab (CYRAMZA®), or Regorafenib (STIVARGA®), or an EGFR targeting agent such as cetuximab (ERBITUX) or panitumumab (VECTIBIX®).
  • a VEGF targeting agent such as bevacizumab (AVASTIN®), ziv-aflibercept (ZALTRAP®), ramucirumab (CYRAMZA®), or Regorafenib (STIVARGA®
  • an EGFR targeting agent such as cetuximab (ERBITUX) or panitumumab (VECTIBIX®).
  • the chemotherapy coadministers a chemotherapy selected from FOLFOX, FOLFIRI, FOLFOXIRI, CapeOx, 5-FU coadministered with leucovorin, capecitabine
  • the heterodimeric Fc-fused protein therapy is combined with one or more chemotherapies to treat a subject diagnosed with breast cancer.
  • the chemotherapy comprises doxorubicin (ADRIAMYCIN®), pegylated liposomal doxorubicin, epirubicin (ELLENCE®), paclitaxel (Taxol), docetaxel (TAXOTERE®), albumin-bound paclitaxel (ABRAXANE®), 5-fluorouracil (5-FU), cyclophosphamide (CYTOXAN®), carboplatin (PARAPLATIN®), cisplatin, vinorelbine (NAVELBINE®), capecitabine (XELODA), gemcitabine (GEMZAR®), ixabepilone (IXEMPRA®), or eribulin (HALAVEN).
  • ADRIAMYCIN® doxorubicin
  • pegylated liposomal doxorubicin epi
  • the chemotherapy comprises a combination of two or more chemotherapies selected from doxorubicin (ADRIAMYCIN®), pegylated liposomal doxorubicin, epirubicin (ELLENCE®), paclitaxel (Taxol), docetaxel (TAXOTERE®), albumin-bound paclitaxel (ABRAXANE®), 5-fluorouracil (5-FU), cyclophosphamide (CYTOXAN®), carboplatin (PARAPLATIN®), cisplatin, vinorelbine (NAVELBINE®), capecitabine (XELODA®), gemcitabine (GEMZAR®), ixabepilone (IXEMPRA®), and eribulin (HALAVEN®).
  • doxorubicin ADRIAMYCIN®
  • pegylated liposomal doxorubicin epirubicin
  • paclitaxel Taxol
  • the heterodimeric Fc-fused protein therapy is combined with one or more chemotherapies to treat a subject diagnosed with melanoma/skin-cancer.
  • the chemotherapy comprises dacarbazine (also called DTIC), temozolomide, nab-paclitaxel, paclitaxel, cisplatin, carboplatin, or vinblastine.
  • the heterodimeric Fc-fused protein therapy is combined with one or more targeted agents.
  • targeted agents act on specific molecular targets, such as targets associated with cancer.
  • Targeted agents are differentiated from standard chemotherapies in that standard chemotherapies act on all rapidly dividing normal and cancerous cells.
  • Targeted agents include, but are not limited to, a hormone therapy, a signal transduction inhibitor, a gene expression modulator, an apoptosis inducer, an angiogenesis inhibitor, an immunotherapy, a toxin delivery molecule (e.g., an antibody drug-conjugate), and a kinase inhibitor.
  • a targeted agent comprises a receptor agonist or ligand.
  • the heterodimeric Fc-fused protein therapy is combined with one or more targeted agents to treat a subject diagnosed with colon cancer, rectal cancer, or colorectal cancer (CRC).
  • the targeted agent comprises cetuximab (ERBITUX®), panitumumab (VECTIBIX®), bevacizumab (AVASTIN®), ziv-aflibercept (ZALTRAP®), regorafenib (STIVARGA®), ramucirumab (CYRAMZA®), nivolumab (OPDIVO®), or ipilimumab (YERVOY®).
  • the heterodimeric Fc-fused protein therapy is combined with one or more targeted agents to treat a subject diagnosed with breast cancer.
  • the targeted agent comprises everolimus (AFINITOR®), tamoxifen (NOLVADEX®), toremifene (FARESTON®), trastuzumab (HERCEPTIN®), fulvestrant (FASLODEX®), anastrozole (ARIMIDEX®), exemestane (AROMASIN®), lapatinib (TYKERB®), letrozole (FEMARA®), pertuzumab (PERJETA®), ado-trastuzumab emtansine (KADCYLA®), palbociclib (IBRANCE®), ribociclib (KISQALI®), neratinib maleate (NERLYNXTM), abemaciclib (VERZENIOTM), olaparib (LYNPARZATM) atezolizumab (TECENTRIQ
  • the heterodimeric Fc-fused protein therapy is combined with one or more targeted agents to treat a subject diagnosed with melanoma/skin-cancer.
  • the targeted agent comprises Vismodegib (ERIVEDGE®), sonidegib (ODOMZO®), ipilimumab (YERVOY®), vemurafenib (ZELBORAF®), trametinib (MEKINIST®), dabrafenib (TAFINLAR®), pembrolizumab (KEYTRUDA®), nivolumab (OPDIVO®), cobimetinib (COTELLICTM), alitretinoin (PANRETIN®), avelumab (BAVENCIO®), encorafenib (BRAFTOVITM), binimetinib (MEKTOVI®), or cemiplimab-rwlc (LIBTAYO®).
  • the heterodimeric Fc-fused protein therapy is combined with a receptor agonist or ligand therapy.
  • the receptor agonist or ligand therapy comprises an agonist antibody.
  • the receptor agonist or ligand therapy comprises a receptor ligand, such as 4-1BBL or CD40L.
  • the heterodimeric Fc-fused protein therapy is combined with radiotherapy.
  • the heterodimeric Fc-fused protein therapy is combined with a radioisotope particle, such as indium In-111, yttrium Y-90, or iodine 1-131.
  • a radioisotope particle such as indium In-111, yttrium Y-90, or iodine 1-131.
  • combination therapies include, but are not limited to, Iodine-131 tositumomab (BEXXAR®), Yttrium-90 ibritumomab tiuxetan (ZEVALIN®), and BEXXAR® with CHOP.
  • the radiotherapy comprises external-beam radiation therapy (EBRT), internal radiation therapy (brachytherapy), endocavitary radiation therapy, interstitial brachytherapy, radioembolization, hypofractionated radiation therapy, intraoperative radiation therapy (IORT), 3D-conformal radiotherapy, stereotactic radiosurgery (SRS), or stereotactic body radiation therapy (SBRT).
  • EBRT external-beam radiation therapy
  • brachytherapy internal radiation therapy
  • endocavitary radiation therapy endocavitary radiation therapy
  • interstitial brachytherapy radioembolization
  • hypofractionated radiation therapy IORT
  • 3D-conformal radiotherapy 3D-conformal radiotherapy
  • SRS stereotactic radiosurgery
  • SBRT stereotactic body radiation therapy
  • the heterodimeric Fc-fused protein therapy is combined with one or more radiotherapies to treat a subject diagnosed with colon cancer, rectal cancer, or colorectal cancer (CRC).
  • the radiotherapy comprises external-beam radiation therapy (EBRT), internal radiation therapy (brachytherapy), endocavitary radiation therapy, interstitial brachytherapy, or radioembolization.
  • the heterodimeric Fc-fused protein therapy is combined with one or more radiotherapies to treat a subject diagnosed with breast cancer.
  • the radiotherapy comprises external-beam radiation therapy, hypofractionated radiation therapy, intraoperative radiation therapy (IORT), or 3D-conformal radiotherapy.
  • the heterodimeric Fc-fused protein therapy is combined with one or more radiotherapies to treat a subject diagnosed with melanoma/skin-cancer.
  • the radiotherapy comprises stereotactic radiosurgery (SRS; e.g., using a Gamma Knife or linear accelerator) or stereotactic body radiation therapy (SBRT).
  • the heterodimeric Fc-fused protein therapy is combined with one or more immunogenic compositions, e.g., a vaccine composition or an oncolytic virus, capable of raising a specific immune response, e.g., a tumor-specific immune response.
  • immunogenic compositions e.g., a vaccine composition or an oncolytic virus, capable of raising a specific immune response, e.g., a tumor-specific immune response.
  • the heterodimeric Fc-fused protein therapy is combined with a vaccine composition.
  • Vaccine compositions typically comprise a plurality of antigens and or neoantigens specific for the tumor to be targeted. Vaccine compositions can also be referred to as vaccines.
  • a vaccine composition further comprises an adjuvant and/or a carrier.
  • a vaccine composition associates with a carrier such as a protein or an antigen-presenting cell such as a dendritic cell (DC) capable of presenting the peptide to a T-cell.
  • carriers are scaffold structures, for example a polypeptide or a polysaccharide, to which an antigen or neoantigen, is capable of being associated.
  • adjuvants are any substance whose admixture into a vaccine composition increases or otherwise modifies the immune response to an antigen or neoantigen.
  • adjuvants are conjugated covalently or non-covalently.
  • the ability of an adjuvant to increase an immune response to an antigen is typically manifested by a significant or substantial increase in an immune-mediated reaction, or reduction in disease symptoms.
  • an increase in humoral immunity is typically manifested by a significant increase in the titer of antibodies raised to the antigen
  • an increase in T-cell activity is typically manifested in increased cell proliferation, or cellular cytotoxicity, or cytokine secretion.
  • An adjuvant may also alter an immune response, for example, by changing a primarily humoral or Th response into a primarily cellular, or Th response.
  • Suitable adjuvants include, but are not limited to 1018 ISS, alum, aluminium salts, Amplivax, AS15, BCG, CP-870,893, CpG7909, CyaA, dSLIM, GM-CSF, IC30, IC31, Imiquimod, ImuFact IMP321, IS Patch, ISS, ISCOMATRIX, JuvImmune, LipoVac, MF59, monophosphoryl lipid A, Montanide IMS 1312, Montanide ISA 206, Montanide ISA 50V, Montanide ISA-51, OK-432, OM-174, OM-197-MP-EC, ONTAK, PepTel vector system, PLG microparticles, resiquimod, SRL172, Virosomes and other Virus-like particles, YF-17
  • Adjuvants such as incomplete Freund's or GM-CSF are useful.
  • GM-CSF Several immunological adjuvants (e.g., MF59) specific for dendritic cells and their preparation have been described previously (Dupuis M, et al., Cell Immunol. 1998; 186(1):18-27; Allison A C; Dev Biol Stand. 1998; 92:3-11). Cytokines can also be used.
  • an adjuvant comprises a CpG immunostimulatory oligonucleotide.
  • an adjuvant comprises a TLR agonist.
  • useful adjuvants include, but are not limited to, chemically modified CpGs (e.g. CpR, Idera), Poly(I:C)(e.g. polyi:CI2U), non-CpG bacterial DNA or RNA as well as immunoactive small molecules and antibodies such as cyclophosphamide, sunitinib, bevacizumab, celebrex, NCX-4016, sildenafil, tadalafil, vardenafil, sorafinib, XL-999, CP-547632, pazopanib, ZD2171, AZD2171, ipilimumab, tremelimumab, and SC58175, which may act therapeutically and/or as an adjuvant.
  • CpGs e.g. CpR, Idera
  • non-CpG bacterial DNA or RNA as well as immunoactive small molecules and
  • adjuvants and additives can readily be determined by the skilled artisan without undue experimentation.
  • Additional adjuvants include colony-stimulating factors, such as Granulocyte Macrophage Colony Stimulating Factor (GM-CSF, sargramostim).
  • GM-CSF Granulocyte Macrophage Colony Stimulating Factor
  • a vaccine composition comprises more than one different adjuvant.
  • a vaccine composition comprises any adjuvant substance including any of the above or combinations thereof. It is also contemplated that a vaccine and an adjuvant can be administered together or separately in any appropriate sequence.
  • a carrier (or excipient) is present independently of an adjuvant.
  • the function of a carrier is to increase the molecular weight, increase activity or immunogenicity, to confer stability, to increase the biological activity, or to increase serum half-life.
  • a carrier aids presenting peptides to T-cells.
  • a carrier comprises any suitable carrier known to the person skilled in the art, for example a protein or an antigen presenting cell.
  • carrier proteins include, but are not limited to, keyhole limpet hemocyanin, serum proteins such as transferrin, bovine serum albumin, human serum albumin, thyroglobulin or ovalbumin, immunoglobulins, or hormones, such as insulin or palmitic acid.
  • the carrier is generally a physiologically acceptable carrier acceptable to humans and safe.
  • tetanus toxoid and/or diphtheria toxoid are suitable carriers.
  • the carrier can be dextrans, for example sepharose.
  • a vaccine comprises a viral vector-based vaccine platform, such as vaccinia, fowlpox, self-replicating alphavirus, marabavirus, adenovirus (See, e.g., Tatsis et al., Adenoviruses, Molecular Therapy (2004) 10, 616-629), or lentivirus, including but not limited to second, third or hybrid second/third generation lentivirus and recombinant lentivirus of any generation designed to target specific cell types or receptors (See, e.g., Hu et al., Immunization Delivered by Lentiviral Vectors for Cancer and Infectious Diseases, Immunol Rev .
  • vaccinia such as vaccinia, fowlpox, self-replicating alphavirus, marabavirus, adenovirus (See, e.g., Tatsis et al., Adenoviruses, Molecular Therapy (2004) 10, 616-629), or lentivirus, including but not limited to second
  • the vaccine composition comprises one or more viral-vectors.
  • viral-vectors comprise sequences flanked by non-mutated sequences, separated by linkers, or preceded with one or more sequences targeting a subcellular compartment (See, e.g., Gros et al., Prospective identification of neoantigen-specific lymphocytes in the peripheral blood of melanoma patients, Nat Med (2016) 22 (4):433-8, Stronen et al., Targeting of cancer neoantigens with donor-derived T cell receptor repertoires, Science.
  • Vaccinia vectors and methods useful in immunization protocols are described in, e.g., U.S. Pat. No. 4,722,848.
  • Another vector is BCG (Bacille Calmette Guerin).
  • BCG vectors are described in Stover et al. (Nature 351:456-460 (1991)).
  • a wide variety of other vaccine vectors useful for therapeutic administration or immunization of neoantigens e.g., Salmonella typhi vectors, and the like will be apparent to those skilled in the art from the description herein.
  • the heterodimeric Fc-fused protein therapy is combined with an oncolytic virus therapy.
  • an oncolytic virus is a virus engineered to infect and kill mainly cancer cells.
  • the oncolytic virus in addition to an oncolytic virus killing a cancer cell, induces an immune response to the cancer cell.
  • the heterodimeric Fc-fused protein therapy is combined with oncolytic virus therapy to treat a subject diagnosed with melanoma/skin-cancer.
  • the oncolytic virus comprises talimogene laherparepvec (IMLYGIC®), also referred to as T-VEC.
  • IMLYGIC® talimogene laherparepvec
  • T-VEC T-VEC
  • a heterodimeric Fc-fused protein comprising subunits of IL-12 is used for treating cancer (e.g., an advanced malignancy) in combination with an oncolytic virus (for example, Talimogene Laherparepvec (IMLYGIC®) or T-VEC).
  • the proteins of the present invention are typically made using recombinant DNA technology.
  • a first nucleic acid sequence encoding the first polypeptide comprising a first subunit of a multisubunit protein (p40 subunit of human IL-12) fused to a first antibody Fc domain polypeptide was cloned into a first expression vector (pET-pSURE-Puro);
  • a second nucleic acid sequence encoding a second polypeptide comprising a second, different subunit of a multisubunit protein (p35 subunit of human IL-12) fused to a second antibody Fc domain polypeptide was cloned into a second expression vector (pET-pSURE-Puro); and the first and the second expression vectors were stably transfected together into host cells to produce the heterodimeric Fc-fused proteins.
  • Exemplary amino acid sequence encoded by the first expression vector is shown in SEQ ID NO:292.
  • the first expression vector encoded a first polypeptide comprising a p40 subunit of human IL-12 fused to a human IgG1 Fc sequence comprising a Y349C mutation.
  • the first polypeptide also included K360E and K409W mutations that promote heterodimerization, and LALAPA (L234A, L235A, and P329A) mutations that reduce effector functions.
  • leader sequence is shown in italics, the p40 subunit sequence of human IL-12 is underlined, and the mutations are shown in bold.
  • Exemplary amino acid sequence encoded from the second expression vector is shown in SEQ ID NO:293.
  • the second expression vector encoded a second polypeptide comprising a p35 subunit of human IL-12 fused to a human IgG1 Fc sequence comprising a S354C mutation.
  • the second polypeptide also included Q347R, D399V, and F405T mutations that promote heterodimerization, and LALAPA (L234A, L235A, and P329A) mutations that reduce effector functions.
  • leader sequence is shown in italics
  • the p35 subunit sequence of human IL-12 is underlined
  • mutations are shown in bold.
  • Clones are cultured under conditions suitable for bio-reactor scale-up and maintained expression of the proteins.
  • the proteins are isolated and purified using methods known in the art including centrifugation, depth filtration, cell lysis, homogenization, freeze-thawing, affinity purification, gel filtration, ion exchange chromatography, hydrophobic interaction exchange chromatography, and mixed-mode chromatography.
  • This example describes relative abilities of two IL-12-Fc fusion constructs of recombinant murine IL-12 (rmIL-12) to control tumor progression in a mouse colon cancer model.
  • the two IL-12-Fc fusion variants used in this example were mIL-12-Fc wildtype (DF-mIL-12-Fc wt), which includes wild-type murine IL-12 p40 and p35 subunits fused to the N-termini of wild-type murine IgG2a Fc domain polypeptides, and mIL-12-Fc silent (DF-mIL-12-Fc si), which includes wild-type murine IL-12 p40 and p35 subunits fused to the N-termini of murine IgG2a Fc domain polypeptides with mutations L234A, L235A, and P329G.
  • the amino acid sequences of the proteins are shown below:
  • mIL-12-p40-mIgG2A-EW (first chain of DF-mIL-12-Fc wt) (SEQ ID NO: 286) MWELEKDVYVVEVDWTPDAPGETVNLTCDTPEEDD ITWTSDQRHGVIGSGKTLTITVKEFLDAGQYTCHK GGETLSHSHLLLHKKENGIWSTEILKNFKNKTFLK CEAPNYSGRFTCSWLVQRNMDLKFNIKSSSSSPDS RAVTCGMASLSAEKVTLDQRDYEKYSVSCQEDVTC PTAEETLPIELALEARQQNKYENYSTSFFIRDIIK PDPPKNLQMKPLKNSQVEVSWEYPDSWSTPHSYFS LKFFVRIQRKKEKMKETEEGCNQKGAFLVEKTSTE VQCKGGNVCVQAQDRYYNSSCSKWACVPCRVRSPR GPTIKPCPPCKCPAPNLLGGPSVFIFPPKIKDVLM ISLSPIVTCVV
  • FIGS. 5A-5C Although IL-12 ( FIG. 5A ) and DF-mIL-12-Fc wt ( FIG. 5B ) were efficient in controlling tumor progression in some mice, only DF-mIL-12-Fc si induced robust tumor regression and yielded 100% complete tumor regression ( FIG. 5C ). Moreover, overall survival was significantly extended by the treatment of DF-mIL-12-Fc si therapy—100% of treated mice were still alive at day 60, whereas median survival times of the mice treated with isotype control, DF-mIL-12-Fc wt, and IL-12 were 27 days, 33 days, and 46 days, respectively ( FIG. 6 ).
  • DF-mIL-12-Fc wt and DF-mIL-12-Fc si were compared.
  • 10 6 CT26-Tyrpl colon carcinoma cells were injected subcutaneously into the flank of Balb/c mice.
  • tumor volume reached 300 mm 3
  • Tumor growth was assessed for 55 days.
  • FIGS. 7A-7D treatment with DF-mIL-12-Fc wt led to reduced tumor progression in some mice and complete regression in two mice at the 1 ⁇ g rmIL-12 molar equivalents dose ( FIG. 7A ), but no tumor suppression was observed at the 0.1 ⁇ g IL-12 molar equivalents dose ( FIG. 7C ).
  • the DF-mIL-12-Fc si treatment at the 1 ⁇ g IL-12 molar equivalents dose yielded 100% complete tumor regression ( FIG. 7B ) and induced a robust delay in tumor growth at the lower dose of 0.1 ⁇ g IL-12 molar equivalents ( FIG. 7D ).
  • mice treated with 1 ⁇ g IL-12 molar equivalents of DF-mIL-12-Fc wt was 32 days, similar to the 34 days of median survival of the mice treated with 0.1 ⁇ g IL-12 molar equivalents of DF-mIL-12-Fc si, suggesting that DF-mIL-12-Fc si was 10-fold more potent than its wildtype variant ( FIG. 8 ).
  • DF-mIL-12-Fc wt was not efficient at the dose of 0.1 ⁇ g IL-12 molar equivalents, and showed the same median survival of 24 days as the isotype treated group.
  • FIGS. 22A-22B both intraperitoneal ( FIG. 22A ) and subcutaneous ( FIG. 22B ) administration of DF-mIL-12-Fc si induced robust tumor regression and yielded 100% complete tumor regression.
  • DF-mIL-12-Fc si treatment demonstrated efficacy using various routes of administration.
  • Example 3 Tumor Suppression by IL-12 Fused with a Silent Fc Domain Polypeptide in a B16F10 Tumor Model
  • This example describes relative abilities of DF-mIL-12-Fc wt and DF-mIL-12-Fc si in controlling tumor progression in a mouse melanoma model. Briefly, 10 6 B16F10 melanoma cells were injected subcutaneously into C57BL/6 mice.
  • DF-mIL-12-Fc si was the most efficient in controlling tumor growth.
  • Median survival time of the mice treated with DF-mIL-12-Fc si was 29 days, which was longer than the median survival times of the mice treated with isotype control, DF-mIL-12-Fc wt, and IL-12, which were 16 days, 26 days, and 22 days, respectively ( FIG. 10 ).
  • DF-mIL-12-Fc si was superior to DF-mIL-12-Fc wt in suppression of tumor growth at both doses.
  • the median survival of the mice treated with DF-mIL-12-Fc wt was 20 days.
  • the median survival of the mice treated with 0.1 ⁇ g IL-12 molar equivalents of DF-mIL-12-Fc si was 21 days, and the median survival of the mice treated with 0.5 ⁇ g IL-12 molar equivalents of DF-mIL-12-Fc si was 28 days ( FIG. 12 ).
  • a single administration of DF-mIL-12-Fc si resulted in reduced tumor outgrowth in 100% of mice, although tumor outgrowth occurred sooner when compared to weekly administrations ( FIG. 9C ). Additionally, mice demonstrated transient weight loss, but after the first dose only (data not shown). Accordingly, a single administration of DF-mIL-12-Fc si demonstrated initial efficacy in a hard-to-treat tumor model, although subsequent weekly administrations are better at delaying tumor outgrowth in this model.
  • FIGS. 24A-24B both intraperitoneal ( FIG. 24A ) and subcutaneous ( FIG. 24B ) administration of DF-mIL-12-Fc si induced tumor regression in 100% of mice.
  • DF-mIL-12-Fc si treatment demonstrated efficacy using various routes of administration.
  • the potency of DF-hIL-12-Fc si in comparison to rhIL-12 was assessed using in vitro bioassays.
  • IL-12 potency was assessed using a HEK-Blue IL-12 reporter assay.
  • IL-12R+ HEK-Blue reporter cells (InvivoGen) were harvested from culture and adjusted to 1 ⁇ 10 6 cells/mL in culture media.
  • DF-hIL-12-Fc si (DF IL-12-Fc) and recombinant human IL-12 (rhIL-12; PeproTech) were diluted in media.
  • 100 ⁇ L of PBMC suspension was mixed with 100 ⁇ L of diluted test article and incubated for 48 hours. The supernatant was harvested and engagement of IL-12 receptor and signaling components stably expressed by the reporter cells was detected by measurement of secreted embryonic alkaline phosphatase from the cells following manufacturer instructions.
  • sample supernatant was mixed with 200 ⁇ L of QUANTI-Blue reagent and incubated in the dark at RT for 10 minutes.
  • the plate was then read with a SpectraMax i3x plate-reader at 620 nM and optical density reported to represent relative IL-12 activity.
  • production of SEAP by IL-12R+ HEK reporter cells increased with increasing concentrations of DF-hIL-12-Fc si or rhIL-12.
  • the measured IL-12 responses in the HEK-Blue reporter assay were comparable between DF-hIL-12-Fc si and rhIL-12 at the concentrations examined.
  • IL-12 potency was assessed by quantifying IFN ⁇ production from human PBMCs.
  • PBMCs were isolated from human peripheral blood buffy coats using density gradient centrifugation and adjusted to 1 ⁇ 10 6 cells/mL in culture media.
  • DF-hIL-12-Fc si and recombinant human IL-12 (rhIL-12) were diluted in media.
  • 100 ⁇ L of PBMC suspension was mixed with 100 ⁇ L of diluted test article and incubated for 48 hrs. The supernatant was harvested and IFN ⁇ was quantified using a Human IFN- ⁇ ELISA MAX kit (BioLegend).
  • IFN ⁇ ELISA plates After development of the IFN ⁇ ELISA plates, they were read using a SpectraMax i3x instrument at 450 nm with a background subtraction at 540 nm. IFN ⁇ content in sample wells was approximated by interpolating sample readings from the assay standard curve.
  • IFN ⁇ production increased when human PBMCs were cultured with DF-hIL-12-Fc si or rhIL-12, with concurrent treatment with 5 ⁇ g/ml of PHA to amplify the magnitude of IFN ⁇ responses.
  • IFN- ⁇ production following IL-12 stimulation was comparable between DF-hIL-12-Fc si and rhIL-12 at the concentrations examined.
  • Example 5 IL-12, DF-hIL-12-Fc Si and IFN ⁇ Concentrations in Monkey Plasma Following IV Infusion of DF-hIL-12-Fc Si or rhlL-12
  • PD pharmacodynamics
  • PK pharmacokinetics
  • Cynomolgus monkeys were administered DF-hIL-12-Fc si and recombinant human IL-12 at 10 ⁇ g/kg by IV-infusion.
  • An immunoassay was used to detect DF-hIL-12-Fc si and Human IL-12 based on a Quantikine ELISA Human IL-12 p70 Immunoassay kit: This assay employed the quantitative sandwich enzyme immunoassay technique. A monoclonal antibody specific for human IL-12 p70 was used as a solid phase capture and detection was accomplished using an antibody HRP-tagged reporter. Standards and QCs spiked with rhIL-12 or DF-hIL-12-Fc si reference standard, along with test samples were pipetted into the wells of microtiter plate and any IL-12 p70 present in the samples were bound by the immobilized antibody, on the solid phase.
  • An immunoassay (meso scale discovery (MSD)—an ELISA like immunoassay) was also used to detect DF-hIL-12-Fc si that involved coating an untreated MSD microtiter plate with monkey-adsorbed goat anti-human IgG and incubating at room temperature. The plate was washed, blocked, washed, and incubated with standard curve and quality control samples spiked with DF-hIL-12-Fc si reference standard, along with test samples. After this incubation, the plate was washed and biotin anti-human IL-12/IL-23 p40 was added to the plate as the primary detection antibody. After another wash step, streptavidin-conjugated Sulfo-Tag was added as the secondary detection antibody.
  • MSD Read Buffer T was added to the plate, and the plate was read using a MSD Sector Imager 5600.
  • Raw MSD data was exported into a text file, which was then converted into a Watson LIMS compatible file using a programmed Excel spreadsheet, which was custom designed at Envigo. Data was imported and regressed in Watson LIMS Software v.7.2.0.02.
  • a meso scale discovery method was performed for the relative quantitative measurement of NHP proinflammatory biomarkers in cynomolgus monkey plasma.
  • the method used a sandwich immunoassay procedure for the relative quantitative measurement of Pro-inflammatory Panel 1 Biomarkers: IFN ⁇ , IL-1 ⁇ , IL-2, IL-6 IL-8, and IL-10 in cynomolgus monkey K2 EDTA plasma (referred to as monkey plasma).
  • the method is based on MSD non-human primate (NHP) kits for V-PLEX and V-PLEX Plus, Catalog No. K15056D-1, K15056D-2, K15056D-4, K15056D-6, K15056G-1, K15056G-2, K15056G-4, K15056G-6.
  • NHS MSD non-human primate
  • the method employs human capture and detection antibodies that react with cynomolgus monkeys.
  • the kit provides plates pre-coated with capture antibodies on independent well-defined spots in each well of a 96-well multi-spot plate. The plate was incubated with monkey plasma samples, washed and then incubated with detection antibodies (specific for each analyte) that are conjugated with electrochemiluminescent (ECL) labels (MSD SULFO-TAG). Analytes in the sample bind to capture antibodies immobilized on the working electrode surface; recruitment of the detection antibodies by the bound analytes completes the sandwich. The plate was washed and an MSD Read Buffer was added to create the appropriate chemical environment for electrochemiluminiscence (ECL).
  • ECL electrochemiluminescent
  • the plate was loaded into an MSD Sector Imager 600 (SI600) instrument where a voltage was applied to the plate electrodes causing the captured labels to emit light.
  • SI600 MSD Sector Imager 600
  • the instrument measures the intensity of emitted light in terms of Relative Light Units (RLU) to provide a relative quantitative measure of analytes in the sample.
  • Raw RLU data was exported into a text file, which then was converted into a Watson LIMS compatible file using a programmed Excel spread sheet, which was custom designed at Envigo. Data was subsequently imported and regressed in Watson LIMS Software v.7.2.0.02.
  • FIG. 14 shows the relative plasma concentrations of DF-hIL-12-Fc si and recombinant human IL-12 over time following IV-administration.
  • concentrations of DF-hIL-12-Fc si and rhIL-12 decreased over time, as expected.
  • DF-hIL-12-Fc si demonstrated a prolonged half-life and overall greater exposure compared to rhIL-12 over the time course.
  • FIG. 14 also shows the relative concentrations of IFN ⁇ (PD) in monkey plasma following IV-administration.
  • the data indicate that the pharmacodynamics of DF-hIL-12-Fc si and rhIL-12, as assessed by IFN ⁇ production, both demonstrated activity following IV-administration.
  • DF-hIL-12-Fc si demonstrated a higher peak activity and a longer duration compared to rhIL-12.
  • DF-mIL-12-Fc si The serum half-life and in vivo pharmacodynamics of a half-life prolonged murine IL-12 variant, designated DF-mIL-12-Fc si, was examined.
  • DF-mIL-12-Fc si An equivalent molar amount of DF-mIL-12-Fc si, corresponding to 1 ⁇ g IL-12, was intravenously injected in non-tumor bearing Balb/c mice and PK/PD characteristics were compared to IL-12.
  • Blood was sampled at 0.017, 0.5, 3, 6, 24, 48, 72, 96, 144 and 219 hours post-injection.
  • IL-12 and IFN ⁇ levels in serum were analyzed by ELISA, as previously described.
  • PK/PD properties for different routes of administering DF-mIL-12-Fc si were compared.
  • intravenous ( FIG. 15C ), intraperitoneal ( FIG. 15D ), or subcutaneous ( FIG. 15E ) administration all resulted in DF-mIL-12-Fc si-mediated IFN ⁇ production comparable across the different routes of administration.
  • subcutaneous administration resulted in a lower IL-12 Cmax.
  • the pharmacokinetic properties (e.g., IL-12 concentration) of DF-mIL-12-Fc si administration varied depending on the route of administration, while the pharmacodynamic properties (IFN ⁇ production) remained protracted and relatively comparable across the different routes.
  • Combination therapy of DF-mIL-12-Fc si and PD-1 blockade was performed to analyze whether anti-tumor immune response can be amplified in established B16F10 tumors.
  • FIGS. 16A-16C While administration of DF-mIL-12-Fc si alone delayed tumor regression ( FIG. 16A ) and PD-1 alone had a minimal effect on tumor growth ( FIG. 16B ), the combination of DF-mIL-12-Fc si with PD-1 blockade further delayed tumor growth ( FIG. 16C ), suggesting anti-PD-1 treatment further amplified anti-tumor responses to DF-mIL-12-Fc si treatment.
  • a combination therapy of DF-mIL-12-Fc si and PD-1 blockade demonstrated improved efficacy compared to either treatment alone.
  • Combination therapy of DF-mIL-12-Fc si and mcFAE-C26.99 TriNKETs was performed to analyze whether anti-tumor immune response can be amplified in established B16F10 tumors.
  • mice C57BL/6 mice were injected with 10 6 B16F10 melanoma cells subcutaneously into the flank of the mice.
  • the three complete responders (the CRs from the experiment described above and the data presented in FIG. 18C ) were re-challenged with 2 ⁇ 10 6 B16F10 melanoma cells 72 days after first tumor inoculation.
  • Age-matched na ⁇ ve C57BL/6 mice were used as control group.
  • 1 out of 3 mice from the initial DF-mIL-12-Fc si/TriNKET combo treated group remained tumor-free, another mouse showed tumor formation starting at day 95 and the tumor progression of the third mouse was similar to the age-matched control group ( FIG. 20 ), suggesting the formation of immunological memory with combination therapy.
  • a combination therapy of DF-mIL-12-Fc si and TriNKETs demonstrated improved efficacy compared to either treatment alone, including demonstrating a complete, durable response in a population of mice.
  • This example shows that treatment with DF-mIL-12-Fc si promotes recovery in mice bearing CT26 tumors.
  • FIG. 21A is a graph showing tumor growth curves of individual mice inoculated with CT26 tumor cells and administered a single dose of 1 ⁇ g of DF-mIL-12-Fc si or mIgG2a isotype.
  • FIG. 21B is a graph showing body weights of individual mice inoculated with CT26 tumor cells and administered a weekly dose of 1 ⁇ g of DF-mIL-12-Fc si or mIgG2a isotype.
  • FIG. 21C is a graph showing tumor growth curves of individual mice re-challenged with inoculation of CT26 tumor cells.
  • FIGS. 21A-B administration of DF-mIL-12-Fc si resulted in robust tumor regression in comparison to mIgG2a isotype with no observable toxicity affecting the body weight of treatment animals.
  • FIG. 21C the initial DF-mIL-12-Fc si treated mice remained tumor-free suggesting the formation of immunological memory with DF-mIL-12-Fc si treatment.
  • FIG. 22A is a graph showing tumor growth curve of individual mice inoculated with CT26 tumor cells and administered a weekly dose of 1 ⁇ g of DF-mIL-12-Fc si or mIgG2a isotype delivered intraperitoneally.
  • FIG. 22B is a graph showing tumor growth curve of individual mice inoculated with CT26 tumor cells and administered a weekly dose of 1 ⁇ g of DF-mIL-12-Fc si or mIgG2a isotype delivered subcutaneously.
  • This example shows that a single dose of DF-mI12-Fc si is effective at reducing tumor volume in mice bearing B16F10 melanoma tumors.
  • mice were injected with 10 6 B16F10 melanoma cells subcutaneously into the flank of the mice.
  • mice were randomized. When tumor average reached 200 mm 3 , mice were treated intraperitoneally with a single dose of isotype control, or 1 ⁇ g of DF-mIL-12-Fc si. Tumor growth was assessed for 50 days.
  • a single administration of ⁇ g of DF-mIL-12-Fc si is effective to reduce tumor volume in B16F10 tumor-bearing mice.
  • mice were randomized. When tumor average reached 200 mm 3 , mice were intraperitoneally injected with 1 ⁇ g of DF-mIL-12-Fc si at a molar dose equivalent to 1 ⁇ g IL-12 or 1 ⁇ g of mIgG2a isotype control once a week, or subcutaneously injected with 1 ⁇ g of DF-mIL-12-Fc si at a molar dose equivalent to 1 ⁇ g IL-12 or 1 ⁇ g of mIgG2a isotype control once a week. Tumor growth was assessed for 40 days.
  • Example 13 DF-mIL-12-Fc Si is Efficacious as a Single Dose
  • CT26-Tyrpl colon carcinoma cells were injected subcutaneously into the flank of Balb/c mice.
  • tumor volume reached 270 mm 3
  • mice were intraperitoneally injected with 1 ⁇ g of DF-mIL-12-Fc si at a molar dose equivalent to 1 ⁇ g IL-12 or 1 ⁇ g of mIgG2a isotype control once a week. Tumor growth was assessed for more than 60 days.
  • FIG. 25A is a graph showing tumor growth curve of individual mice inoculated with CT26 tumor cells and administered a single dose of 1 ⁇ g of DF-mIL-12-Fc si or mIgG2a isotype.
  • FIG. 25B is a graph showing tumor growth curve of individual mice inoculated with CT26 tumor cells and administered a weekly dose of 1 ⁇ g of DF-mIL-12-Fc si or mIgG2a isotype.
  • a single administration of 1 ⁇ g of DF-mIL-12-Fc si resulted in robust 70% complete recovery of tumor-bearing mice as compared to mIgG2a isotype.
  • FIG. 5C and FIG. 25B repeat weekly dosing of 1 ⁇ g of DF-mIL-12-Fc si ensured 100% complete recovery of tumor-bearing mice as compared to mIgG2a isotype.
  • FIG. 21B even repeat administration of DF-mIL-12-Fc si was well-tolerated with no toxicities observed, as assessed by body weight.
  • Pharmacokinetics were determined following a subcutaneous injection of DF-hIL-12-Fc si at 1 ⁇ g/kg ( FIG. 28A ), 2 ⁇ g/kg ( FIG. 28B ), or 4 ⁇ g/kg ( FIG. 28C ) in cynomolgus monkeys utilizing an ELISA like immunoassay-Meso Scale Discovery (MSD) immunoassay method. Briefly, an untreated MSD microtiter plate was coated with monkey-adsorbed goat anti-human IgG and incubated at room temperature.
  • MSD Immunassay-Meso Scale Discovery
  • the plate was washed, blocked, washed, and incubated with standard curve and quality control samples spiked with a DF-hIL-12-Fc si reference standard, along with test samples. Following incubation, the plate was washed and biotin anti-human IL-12/IL-23 p40 was added to the plate as the primary detection antibody. Following another wash step, streptavidin-conjugated Sulfo-Tag was added as the secondary detection antibody. The plate was washed a final time before adding MSD read buffer T to the plate. The plate was read using an MSD Sector Imager 56000.
  • FIGS. 28A-28C are line graphs showing pharmacokinetics in cynomolgus monkeys treated with a single subcutaneous dose of 1 ⁇ g/kg ( FIG. 28A ), 2 ⁇ g/kg ( FIG. 28B ), or 4 ⁇ g/kg ( FIG. 28C ) of DF-hIL-12-Fc si.
  • FIGS. 29A and 29B Quantitative measurements of cytokines following a subcutaneous injection of DF-hIL-12-Fc si at 1 ⁇ g/kg ( FIGS. 29A and 29B ), 2 ⁇ g/kg ( FIGS. 29C and 29D ), or 4 ⁇ g/kg ( FIGS. 29E and 29F ) in cynomolgus monkeys were determined using MSD immunoassay kits.
  • the method used sandwich immunoassay kits (Pro-inflammatory Panel 1 Biomarkers and V-PLEX Plus Chemokine Panel 1 NHP Kit) for the relative quantitative measurement of Pro-inflammatory Panel 1 Biomarkers: IFN ⁇ , IL-1 ⁇ , IL-2, IL-6 IL-8, and IL-10 in cynomolgus monkey K2 EDTA plasma (referred to as monkey plasma).
  • the method is based on MSD non-human primate (NHP) kits for V-PLEX and V-PLEX Plus, Catalog No. K15056D-1, K15056D-2, K15056D-4, K15056D-6, K15056G-1, K15056G-2, K15056G-4, K15056G-6.
  • NHS MSD non-human primate
  • the method employs human capture and detection antibodies that react with cynomolgus monkeys.
  • the kit provides plates pre-coated with capture antibodies on independent, well-defined spots within each well of a 96-well multi-spot plate.
  • the plate was incubated with monkey plasma samples, washed and then incubated with detection antibodies (specific for each analyte) that are conjugated with electrochemiluminescent (ECL) labels (MSD SULFO-TAG).
  • ECL electrochemiluminescent
  • Analytes in the sample bind to capture antibodies immobilized on the working electrode surface; recruitment of the detection antibodies by the bound analytes completes the sandwich.
  • the plate was washed and an MSD Read Buffer was added to create the appropriate chemical environment for electrochemiluminiscence (ECL).
  • the plate was loaded into an MSD Sector Imager 600 (SI600) instrument where a voltage was applied to the plate electrodes causing the captured labels to emit light.
  • SI600 MSD Sector Imager 600
  • the instrument measures the intensity of emitted light in terms of Relative Light Units (RLU) to provide a relative quantitative measure of analytes in the sample.
  • Raw RLU data was exported into a text file, which then was converted into a Watson LIMS compatible file using a programmed Excel spread sheet, which was custom designed at Envigo. Data was subsequently imported and regressed in Watson LIMS Software v.7.2.0.02.
  • FIGS. 29A-29F are line graphs showing concentrations of IFN ⁇ ( FIGS. 29A, 29C, and 29E ) and IP10/CXCL10 ( FIGS. 29B, 29D, and 29F ) in cynomolgus monkeys treated with a single subcutaneous dose of 1 ⁇ g/kg ( FIGS. 29A and 29B ), 2 ⁇ g/kg ( FIGS. 29C and 29D ), or 4 ⁇ g/kg ( FIGS. 29E and 29F ) of DF-hIL-12-Fc si.
  • a single subcutaneous dose of DF-hIL-12-Fc si at 1 ⁇ g/kg did not result in detectable levels of IFN ⁇ .
  • Subcutaneous doses of DF-hIL-12-Fc si at 2 ⁇ g/kg and 4 ⁇ g/kg resulted in an increase in IFN ⁇ levels in some animals that peaked at day 4 post-dosing ( FIGS. 29C and 29E ).
  • Subcutaneous doses of DF-hIL-12-Fc si at 1 ⁇ g/kg, 2 ⁇ g/kg, and 4 ⁇ g/kg all resulted in elevated IP10/CXCL10 levels that peaked at day 4 post-dosing ( FIGS. 29B, 29D, and 29F ).
  • FIG. 30 is a graph showing tumor growth curves of individual mice inoculated with breast cancer cells and administered a weekly dose of isotype control, DF-mIL-12-Fc si, Doxil (chemotherapy), or irradiated with 10 Gy as monotherapy or DF-mIL-12-Fc si in combination with Doxil® or radiation.
  • Graph shows group averages of tumor growth ⁇ standard error mean.
  • Example 17 DF-mIL-12-Fc Si Mediated Anti-Tumor Efficacy Against Large, PD-1 Blockade-Resistant CT26 Colon Carcinoma Tumors
  • FIG. 31A is a graph showing tumor growth curve of Balb/c mice inoculated with CT26-Tyrpl tumor cells and treated (bi-weekly) either with isotype control or anti-PD-1 antibody.
  • FIG. 31B is a graph showing tumor growth curve of the previously anti-PD-1 antibody-treated Balb/c mice treated with anti-PD-1 antibody (bi-weekly) along with weekly treatment with 1 ⁇ g of DF-mIL-12-Fc si.
  • Example 18 Local Treatment of DF-mIL-12-Fc Si against Large CT26 Colon Carcinoma Tumors Induces Abscopal Anti-Tumor Responses
  • FIG. 32A is a graph showing tumor growth curve of the treated (Tr) tumor in Balb/c mice inoculated with CT26-Tyrpl tumor cells and treated once (weekly) with either isotype control or DF-mIL-12-Fc si.
  • Right tumors were left untreated (NT).
  • FIG. 32B is a graph showing tumor growth curves of the untreated (NT) tumors in Balb/c mice inoculated with CT26-Tyrpl tumor cells.
  • control isotype-treated tumors grew progressively at both right and left sites.
  • DF-mIL-12-Fc si caused effective anti-tumor responses at the local injected site ( FIG. 32A ) and the distant non-treated tumor ( FIG. 32B ) indicating abscopal therapeutic effects.
  • DF-mIL-12-Fc si which includes wild-type murine IL-12 p40 and p35 subunits fused to the N-termini of murine IgG2a Fc domain polypeptides with mutations L234A, L235A, and P329G (discussed in Example 2) is efficacious against larger tumor volumes.
  • FIG. 26A is a graph showing tumor growth curves of Balb/c mice inoculated with CT26-Tyrpl tumor cells and treated once (weekly) with either 2 ⁇ g mIgG2a isotype control or 1 ⁇ g DF-mIL-12-Fc si.
  • FIG. 26B is a graph showing tumor growth curves of Balb/c mice inoculated with CT26-Tyrpl tumor cells and treated once (weekly) with either 2 ⁇ g mIgG2a isotype control or 2 ⁇ g DF-mIL-12-Fc si.
  • FIG. 26A is a graph showing tumor growth curves of Balb/c mice inoculated with CT26-Tyrpl tumor cells and treated once (weekly) with either 2 ⁇ g mIgG2a isotype control or 2 ⁇ g DF-mIL-12-Fc si.
  • FIG. 33A is a graph showing tumor growth curves of Balb/c mice inoculated with CT26-Tyrpl tumor cells and treated once with either 2 ⁇ g mIgG2a isotype control or 2 ⁇ g DF-mIL-12-Fc si.
  • FIG. 33B is a graph showing average tumor growth curves of Balb/c mice inoculated with CT26-Tyrpl tumor cells and treated with 2 ⁇ g mIgG2a isotype control, 1 ⁇ g DF-mIL-12-Fc si (weekly administration), 2 ⁇ g DF-mIL-12-Fc si (weekly administration), or 2 ⁇ g DF-mIL-12-Fc si (once).
  • FIGS. 26A, 26B, and 33A show tumor growth curves of individual mice.
  • FIG. 33B shows tumor average ⁇ standard error mean.
  • Example 20 DF-mIL-12-Fc Si Treatment against B16F10 Melanomas Induces Production of Cytokines and Chemokines in Serum and in Tumors
  • DF-mIL-12-Fc si treatment results in elevated levels of IFN ⁇ , CXCL9, and CXCL10 in blood and tumors of C57BL/6 mice bearing B16F10 tumors.
  • C57BL/6 mice were injected subcutaneously with 10 6 B16F10 melanoma cells.
  • Mice were treated intraperitoneally with isotype control, IL-12, or DF-mIL-12-Fc equimolar to 1 ⁇ g IL-12.
  • FIGS. 34A-C show average cytokine/chemokine levels in mice.
  • FIGS. 27A-27C a single administration of 0.5 ⁇ g of DF-mIL-12-Fc si resulted in increased expression of IFN ⁇ ( FIG. 27A ), CXCL9 ( FIG. 27B ), and CXCL10 ( FIG. 27C ) in serum (left panel) and within tumors (right panel), whereas IL-12 treatment had little or no effect.
  • This example shows the IFN ⁇ -stimulating and Fc ⁇ R-binding activities of DF hIL-12-Fc si with IgG1 Fc having LALAPA (L234A, L235A, and P329A) mutations, or LALAPG (L234A, L235A, and P329G) mutations.
  • human PBMCs were cultured for 2 days with both 5 ⁇ g/ml phytohaemagglutinin (PHA) and a dose-titration of DF hIL-12-Fc-si, having LALAPA or LALAPG mutations. After 2-day stimulation, supernatants were harvested and IFN ⁇ content measured by ELISA.
  • PHA phytohaemagglutinin
  • fluorophore-conjugated hIgG1 isotype antibody (83 nM) bound to THP-1 cells that express high affinity Fc ⁇ Rs CD32 and CD64 was detected by flow cytometry.
  • hIL-12-Fc-LALAPA and hIL-12-Fc-LALAPG have similar abilities to stimulate IFN ⁇ production from PBMCs concurrently with PHA, well above the amount produced with PHA alone.
  • Embodiments disclosed herein include embodiments P1 to P121 as provided in the numbered embodiments of the disclosure.
  • a heterodimeric Fc-fused protein comprising:
  • first polypeptide comprising a first antibody Fc sequence and a second polypeptide comprising a second, different antibody Fc sequence
  • the first polypeptide further comprises a protein sequence of a subunit of a multisubunit protein
  • protein sequence is fused by a linker comprising amino acid sequence RVESKYGPPCPPCPAPEFXGG (SEQ ID NO:1) to the first antibody Fc sequence, wherein X represents L or E; and
  • an additional subunit of the multisubunit protein is fused to the second, different antibody Fc sequence, and the subunits of the multisubunit protein are bound to each other,
  • first antibody Fc sequence and the second, different antibody Fc sequence each comprise different mutations promoting heterodimerization
  • first antibody Fc sequence and the second, different antibody Fc sequence are bound to each other.
  • heterodimeric Fc-fused protein of embodiment P2 wherein the additional subunit of the multisubunit protein is fused to the second, different antibody Fc sequence by a linker comprising amino acid sequence GGGGSGGGGSGGGGSRVESKYGPPCPPCPAPEFLGG (SEQ ID NO:3).
  • heterodimeric Fc-fused protein of embodiment P2 wherein the additional subunit of the multisubunit protein is fused to the second, different antibody Fc sequence by a linker comprising amino acid sequence GGGGSRVESKYGPPCPPCPAPEFLGG (SEQ ID NO:13).
  • heterodimeric Fc-fused protein of embodiment P5 wherein the additional subunit of the multisubunit protein is fused to the second, different antibody Fc sequence by a linker consisting of amino acid sequence GGGGSRVESKYGPPCPPCPAPEFLGG (SEQ ID NO:13).
  • heterodimeric Fc-fused protein of embodiment P2 wherein the additional subunit of the multisubunit protein is fused to the second, different antibody Fc sequence by a linker comprising amino acid sequence GGGGSGGGGSRVESKYGPPCPPCPAPEFLGG (SEQ ID NO:14).
  • heterodimeric Fc-fused protein of embodiment P7 wherein the additional subunit of the multisubunit protein is fused to the second, different antibody Fc sequence by a linker consisting of amino acid sequence GGGGSGGGGSRVESKYGPPCPPCPAPEFLGG (SEQ ID NO:14).
  • a heterodimeric Fc-fused protein according to any one of embodiments P2 to P8, wherein the linker fusing the protein sequence to the first antibody Fc sequence consists of amino acid sequence RVESKYGPPCPPCPAPEFLGG (SEQ ID NO:2).
  • heterodimeric Fc-fused protein of embodiment P10 wherein the additional subunit of the multisubunit protein is fused to the second, different antibody Fc sequence by a linker comprising amino acid sequence GGGGSGGGGSGGGGSRVESKYGPPCPPCPAPEFEGG (SEQ ID NO:5).
  • heterodimeric Fc-fused protein of embodiment P11 wherein the additional subunit of the multisubunit protein is fused to the second, different antibody Fc sequence by a linker consisting of amino acid sequence GGGGSGGGGSGGGGSRVESKYGPPCPPCPAPEFEGG (SEQ ID NO:5).
  • heterodimeric Fc-fused protein of embodiment P10 wherein the additional subunit of the multisubunit protein is fused to the second, different antibody Fc sequence by a linker comprising amino acid sequence GGGGSRVESKYGPPCPPCPAPEFEGG (SEQ ID NO:63).
  • heterodimeric Fc-fused protein of embodiment P13 wherein the additional subunit of the multisubunit protein is fused to the second, different antibody Fc sequence by a linker consisting of amino acid sequence GGGGSRVESKYGPPCPPCPAPEFEGG (SEQ ID NO:63).
  • heterodimeric Fc-fused protein of embodiment P10 wherein the additional subunit of the multisubunit protein is fused to the second, different antibody Fc sequence by a linker comprising amino acid sequence GGGGSGGGGSRVESKYGPPCPPCPAPEFEGG (SEQ ID NO:64).
  • heterodimeric Fc-fused protein of embodiment P15 wherein the additional subunit of the multisubunit protein is fused to the second, different antibody Fc sequence by a linker consisting of amino acid sequence GGGGSGGGGSRVESKYGPPCPPCPAPEFEGG (SEQ ID NO:64).
  • a heterodimeric Fc-fused protein according to any one of embodiments P10 to P16, wherein the linker fusing the protein sequence to the first antibody
  • Fc sequence consists of amino acid sequence RVESKYGPPCPPCPAPEFEGG (SEQ ID NO:4).
  • a heterodimeric Fc-fused protein comprising:
  • first polypeptide comprising a first antibody Fc sequence and a second polypeptide comprising a second, different antibody Fc sequence
  • the first polypeptide further comprises a protein sequence of a subunit of a multisubunit protein
  • protein sequence is fused by a linker comprising amino acid sequence EPKSSDKTHTCPPCPAPEX 1 X 2 GX 3 (SEQ ID NO:6) or PKSSDKTHTCPPCPAPEX 1 X 2 GX 3 (SEQ ID NO:237) to the first antibody Fc sequence, wherein X 1 represents L or A, X 2 represents L, E, or A, and X 3 represents A or G; and
  • an additional subunit of the multisubunit protein is fused to the second, different antibody Fc sequence, and the subunits of the multisubunit protein are bound to each other, and
  • X 1 represents L and/or X 2 represents L
  • at least one of the first antibody Fc sequence, and the second, different antibody Fc sequence comprises a Q347R mutation for promoting heterodimerization
  • first antibody Fc sequence and the second, different antibody Fc sequence are bound to each other.
  • heterodimeric Fc-fused protein of embodiment P19 wherein the additional subunit of the multisubunit protein is fused to the second, different antibody Fc sequence by a linker comprising amino acid sequence GGGGSGGGGSGGGGSEPKSSDKTHTCPPCPAPELLGG (SEQ ID NO:8) or GGGGSGGGGSGGGGSPKSSDKTHTCPPCPAPELLGG (SEQ ID NO:241).
  • heterodimeric Fc-fused protein of embodiment P20 wherein the additional subunit of the multisubunit protein is fused to the second, different antibody Fc sequence by a linker consisting of amino acid sequence GGGGSGGGGSGGGGSEPKSSDKTHTCPPCPAPELLGG (SEQ ID NO:8) or GGGGSGGGGSGGGGSPKSSDKTHTCPPCPAPELLGG (SEQ ID NO:241).
  • heterodimeric Fc-fused protein of embodiment P19 wherein the additional subunit of the multisubunit protein is fused to the second, different antibody Fc sequence by a linker comprising amino acid sequence GGGGSEPKSSDKTHTCPPCPAPELLGG (SEQ ID NO:15) or GGGGSPKSSDKTHTCPPCPAPELLGG (SEQ ID NO:242).
  • heterodimeric Fc-fused protein of embodiment P22 wherein the additional subunit of the multisubunit protein is fused to the second, different antibody Fc sequence by a linker consisting of amino acid sequence GGGGSEPKSSDKTHTCPPCPAPELLGG (SEQ ID NO:15) or GGGGSPKSSDKTHTCPPCPAPELLGG (SEQ ID NO:242).
  • heterodimeric Fc-fused protein of embodiment P19 wherein the additional subunit of the multisubunit protein is fused to the second, different antibody Fc sequence by a linker comprising amino acid sequence GGGGSGGGGSEPKSSDKTHTCPPCPAPELLGG (SEQ ID NO:16) or GGGGSGGGGSPKSSDKTHTCPPCPAPELLGG (SEQ ID NO:243).
  • heterodimeric Fc-fused protein of embodiment P24 wherein the additional subunit of the multisubunit protein is fused to the second, different antibody Fc sequence by a linker consisting of amino acid sequence GGGGSGGGGSEPKSSDKTHTCPPCPAPELLGG (SEQ ID NO:16) or GGGGSGGGGSPKSSDKTHTCPPCPAPELLGG (SEQ ID NO:243).
  • a heterodimeric Fc-fused protein according to any one of embodiments P19 to P25, wherein the linker fusing the protein sequence to the first antibody Fc sequence consists of amino acid sequence EPKSSDKTHTCPPCPAPELLGG (SEQ ID NO:7) or PKSSDKTHTCPPCPAPELLGG (SEQ ID NO:238).
  • heterodimeric Fc-fused protein of embodiment P27 wherein the additional subunit of the multisubunit protein is fused to the second, different antibody Fc sequence by a linker comprising amino acid sequence GGGGSGGGGSGGGGSEPKSSDKTHTCPPCPAPEAAGG (SEQ ID NO:10) or GGGGSGGGGSGGGGSPKSSDKTHTCPPCPAPEAAGG (SEQ ID NO:244).
  • heterodimeric Fc-fused protein of embodiment P28 wherein the additional subunit of the multisubunit protein is fused to the second, different antibody Fc sequence by a linker consisting of amino acid sequence GGGGSGGGGSGGGGSEPKSSDKTHTCPPCPAPEAAGG (SEQ ID NO:10) or GGGGSGGGGSGGGGSPKSSDKTHTCPPCPAPEAAGG (SEQ ID NO:244).
  • heterodimeric Fc-fused protein of embodiment P27 wherein the additional subunit of the multisubunit protein is fused to the second, different antibody Fc sequence by a linker comprising amino acid sequence GGGGSEPKSSDKTHTCPPCPAPEAAGG (SEQ ID NO:65) or GGGGSPKSSDKTHTCPPCPAPEAAGG (SEQ ID NO:245).
  • heterodimeric Fc-fused protein of embodiment P30 wherein the additional subunit of the multisubunit protein is fused to the second, different antibody Fc sequence by a linker consisting of amino acid sequence GGGGSEPKSSDKTHTCPPCPAPEAAGG (SEQ ID NO:65) or GGGGSPKSSDKTHTCPPCPAPEAAGG (SEQ ID NO:245).
  • heterodimeric Fc-fused protein of embodiment P27 wherein the additional subunit of the multisubunit protein is fused to the second, different antibody Fc sequence by a linker comprising amino acid sequence GGGGSGGGGSEPKSSDKTHTCPPCPAPEAAGG (SEQ ID NO:66) or GGGGSGGGGSPKSSDKTHTCPPCPAPEAAGG (SEQ ID NO:246).
  • heterodimeric Fc-fused protein of embodiment P32 wherein the additional subunit of the multisubunit protein is fused to the second, different antibody Fc sequence by a linker consisting of amino acid sequence GGGGSGGGGSEPKSSDKTHTCPPCPAPEAAGG (SEQ ID NO:66) or GGGGSGGGGSPKSSDKTHTCPPCPAPEAAGG (SEQ ID NO:246).
  • a heterodimeric Fc-fused protein according to any one of embodiments P27 to P33, wherein the linker fusing the protein sequence to the first antibody Fc sequence consists of amino acid sequence EPKSSDKTHTCPPCPAPEAAGG (SEQ ID NO:9) or PKSSDKTHTCPPCPAPEAAGG (SEQ ID NO:239).
  • heterodimeric Fc-fused protein of embodiment P35 wherein the additional subunit of the multisubunit protein is fused to the second, different antibody Fc sequence by a linker comprising amino acid sequence GGGGSGGGGSGGGGSEPKSSDKTHTCPPCPAPEAEGA (SEQ ID NO:12) or GGGGSGGGGSGGGGSPKSSDKTHTCPPCPAPEAEGA (SEQ ID NO:247).
  • heterodimeric Fc-fused protein of embodiment P36 wherein the additional subunit of the multisubunit protein is fused to the second, different antibody Fc sequence by a linker consisting of amino acid sequence GGGGSGGGGSGGGGSEPKSSDKTHTCPPCPAPEAEGA (SEQ ID NO:12) or GGGGSGGGGSGGGGSPKSSDKTHTCPPCPAPEAEGA (SEQ ID NO:247).
  • heterodimeric Fc-fused protein of embodiment P35 wherein the additional subunit of the multisubunit protein is fused to the second, different antibody Fc sequence by a linker comprising amino acid sequence GGGGSEPKSSDKTHTCPPCPAPEAEGA (SEQ ID NO:67) or GGGGSPKSSDKTHTCPPCPAPEAEGA (SEQ ID NO:248).
  • heterodimeric Fc-fused protein of embodiment P38 wherein the additional subunit of the multisubunit protein is fused to the second, different antibody Fc sequence by a linker consisting of amino acid sequence GGGGSEPKSSDKTHTCPPCPAPEAEGA (SEQ ID NO:67) or GGGGSPKSSDKTHTCPPCPAPEAEGA (SEQ ID NO:248).
  • heterodimeric Fc-fused protein of embodiment P35 wherein the additional subunit of the multisubunit protein is fused to the second, different antibody Fc sequence by a linker comprising amino acid sequence GGGGSGGGGSEPKSSDKTHTCPPCPAPEAEGA (SEQ ID NO:68) or GGGGSGGGGSPKSSDKTHTCPPCPAPEAEGA (SEQ ID NO:249).
  • heterodimeric Fc-fused protein of embodiment P40 wherein the additional subunit of the multisubunit protein is fused to the second, different antibody Fc sequence by a linker consisting of amino acid sequence GGGGSGGGGSEPKSSDKTHTCPPCPAPEAEGA (SEQ ID NO:68) or GGGGSGGGGSPKSSDKTHTCPPCPAPEAEGA (SEQ ID NO:249).
  • a heterodimeric Fc-fused protein according to any one of embodiments P35 to P41, wherein the linker fusing the protein sequence to the first antibody Fc sequence consists of amino acid sequence EPKSSDKTHTCPPCPAPEAEGA (SEQ ID NO:11) or PKSSDKTHTCPPCPAPEAEGA (SEQ ID NO:240).
  • a heterodimeric Fc-fused protein according to any one of embodiments P1 to P17, wherein the first antibody Fc sequence and the second, different antibody Fc sequence are IgG4 Fc sequences mutated to promote heterodimerization with each other.
  • the heterodimeric Fc-fused protein of embodiment P43 wherein the first antibody Fc sequence comprises one or more mutation(s) selected from the group consisting of K370E, R409W, and a combination thereof, and the second, different antibody Fc sequence comprises one or more mutation(s) selected from the group consisting of E357N, D399V, F405T, and any combination(s) thereof.
  • the heterodimeric Fc-fused protein of embodiment P43 wherein the first antibody Fc sequence comprises one or more mutation(s) selected from the group consisting of E357N, D399V, F405T, and any combination(s) thereof, and the second, different antibody Fc sequence comprises one or more mutation(s) selected from the group consisting of K370E, R409W, and a combination thereof.
  • the heterodimeric Fc-fused protein of embodiment P43 wherein the first antibody Fc sequence comprises mutations K370E and R409W, and the second, different antibody Fc sequence comprises mutations E357N, D399V, and F405T.
  • the heterodimeric Fc-fused protein of embodiment P43 wherein the first antibody Fc sequence comprises mutations E357N, D399V, and F405T, and the second, different antibody Fc sequence comprises mutations K370E and R409W.
  • the heterodimeric Fc-fused protein of embodiment P43 wherein the first antibody Fc sequence comprises one or more mutation(s) selected from the group consisting of K360E, R409W, and a combination thereof, and the second, different antibody Fc sequence comprises one or more mutation(s) selected from the group consisting of Q347R, D399V, F405T, and any combination(s) thereof.
  • Embodiment P49 The heterodimeric Fc-fused protein of embodiment P43, wherein the first antibody Fc sequence comprises one or more mutation(s) selected from the group consisting of Q347R, D399V, F405T, and any combination(s) thereof, and the second, different antibody Fc sequence comprises one or more mutation(s) selected from the group consisting of K360E, R409W, and a combination thereof.
  • the heterodimeric Fc-fused protein of embodiment P43 wherein the first antibody Fc sequence comprises mutations K360E and R409W, and the second, different antibody Fc sequence comprises mutations Q347R, D399V, and F405T.
  • the heterodimeric Fc-fused protein of embodiment P43 wherein the first antibody Fc sequence comprises mutations Q347R, D399V, and F405T, and the second, different antibody Fc sequence comprises mutations K360E and R409W.
  • a heterodimeric Fc-fused protein according to any one of embodiments P18 to P42, wherein the first antibody Fc sequence and the second, different antibody Fc sequence are IgG1 Fc sequence are mutated to promote heterodimerization with each other.
  • the heterodimeric Fc-fused protein of embodiment P52 wherein the first antibody Fc sequence comprises one or more mutation(s) selected from the group consisting of K360E, K409W, and a combination thereof, and the second, different antibody Fc sequence comprises one or more mutation(s) selected from the group consisting of Q347R, D399V, F405T, and any combination(s) thereof.
  • the heterodimeric Fc-fused protein of embodiment P52 wherein the first antibody Fc sequence comprises one or more mutation(s) selected from the group consisting of Q347R, D399V, F405T, and any combination(s) thereof, and the second, different antibody Fc sequence comprises one or more mutation(s) selected from the group consisting of K360E, K409W, and a combination thereof.
  • the heterodimeric Fc-fused protein of embodiment P52 wherein the first antibody Fc sequence comprises mutations K360E and K409W, and the second, different antibody Fc sequence comprises mutations Q347R, D399V, and F405T.
  • the heterodimeric Fc-fused protein of embodiment P52 wherein the first antibody Fc sequence comprises mutations Q347R, D399V, and F405T, and the second, different antibody Fc sequence comprises mutations K360E and K409W.
  • a heterodimeric Fc-fused protein according to any one of embodiments P43 to P56, wherein the IgG4 or IgG1 Fc sequences are further mutated to reduce effector functions.
  • the heterodimeric Fc-fused protein of embodiment P57 wherein the first antibody Fc sequence and the second, different antibody Fc sequence each comprise the mutation L234A, L235A or L235E, and/or P329A.
  • a heterodimeric Fc-fused protein according to any one of embodiments P43 to P60, wherein the IgG4 or IgG1 Fc sequences are further mutated to introduce an inter-chain disulfide bridge.
  • the heterodimeric Fc-fused protein of embodiment P61 wherein the first antibody Fc sequence comprises mutation Y349C, and the second, different antibody Fc sequence comprises mutation S354C.
  • the heterodimeric Fc-fused protein of embodiment P61 wherein the first antibody Fc sequence comprises mutation S354C, and the second, different antibody Fc sequence comprises mutation Y349C.
  • Embodiment P65 The heterodimeric Fc-fused protein of embodiment P64, wherein the cytokine is IL-12.
  • the heterodimeric Fc-fusion protein according to embodiment P66 wherein the at least one antibody heavy chain variable region is connected to an antibody light chain variable region to form an Fab, wherein the Fab is connected at the N-terminus of the first antibody Fc sequence and/or the second, different antibody Fc sequence.
  • heterodimeric Fc-fusion protein according to embodiment P67, wherein the protein sequence of a subunit of a multisubunit protein and the additional subunit of the multisubunit protein are connected to the C-terminus of the first antibody Fc sequence and the second, different antibody Fc sequence, respectively.
  • a heterodimeric Fc-fusion protein according to embodiment P66 or embodiment P70, wherein the second polypeptide comprises an antibody heavy chain variable domain positioned C-terminal to the second, different antibody Fc sequence.
  • heterodimeric Fc-fusion protein according to embodiment P70 or embodiment P71, wherein the antibody heavy chain variable region is connected to an antibody light chain variable region to form an scFv.
  • heterodimeric Fc-fusion protein according to any one of embodiments P70 to 72, wherein the protein sequence of a subunit of a multisubunit protein and the additional subunit of the multisubunit protein are connected to the N-terminus of the first antibody Fc sequence and the second, different antibody Fc sequence, respectively.
  • heterodimeric Fc-fusion protein according to any one of embodiments P70 to P72, wherein the protein sequence of a subunit of a multisubunit protein and the additional subunit of the multisubunit protein are connected to the N-terminus of the second, different antibody Fc sequence and the first antibody Fc sequence, respectively.
  • heterodimeric Fc-fusion protein according to any one of embodiments P1 to P65 or embodiment P75, further comprising a proteoglycan-binding domain.
  • the heterodimeric Fc-fusion protein according to embodiment P76 wherein the proteoglycan-binding domain binds one or more proteoglycans that are specifically expressed in a tumor.
  • the heterodimeric Fc-fusion protein according to embodiment P77 wherein the proteoglycan-binding domain binds one or more proteoglycans selected from syndecan, serglycin, CSPG4, betaglycan, glypican, perlecan, versican, brevican, and small leucine-rich proteoglycans (SLRPs).
  • proteoglycans selected from syndecan, serglycin, CSPG4, betaglycan, glypican, perlecan, versican, brevican, and small leucine-rich proteoglycans (SLRPs).
  • heterodimeric Fc-fusion protein according to embodiment P78, wherein the SLRPs are selected from decorin, biglycan, asporin, fibrodulin, and lumican.
  • heterodimeric Fc-fusion protein according to any one of embodiments P1 to P65 or embodiments P75 to P79, further comprising a collagen-binding domain.
  • collagen-binding domain binds one or more collagens that are specifically expressed in a tumor.
  • heterodimeric Fc-fusion protein according to any one of embodiments P1 to P65 or embodiments P75 to P81, further comprising a hyaluronic acid-binding domain.
  • heterodimeric Fc-fusion protein according to any one of embodiments P76 to P82, wherein the proteoglycan-binding domain, collagen-binding domain, or hyaluronic acid-binding domain is fused to the C-terminus of the first antibody Fc domain polypeptide or the second antibody Fc domain polypeptide.
  • a formulation comprising a heterodimeric Fc-fused protein according to any one of the preceding embodiments and a pharmaceutically acceptable carrier.
  • a cell comprising one or more nucleic acid(s) encoding a heterodimeric Fc-fused protein according to any one of embodiments P1 to P83.
  • a method of treating cancer comprising administering a heterodimeric Fc-fused protein according to any one of embodiments P1 to P83 or a formulation according to embodiment P84 to a patient.
  • a method of treating acute radiation syndrome comprising administering a heterodimeric Fc-fused protein according to any one of embodiments P1 to P83 or a formulation according to embodiment P84 to a patient.
  • the acute radiation syndrome comprises one or more syndrome(s) selected from the group consisting of hematopoietic radiation syndrome, gastrointestinal radiation syndrome, neurovascular radiation syndrome, cutaneous radiation syndrome, and any combination(s) thereof.
  • a heterodimeric Fc-fused protein comprising a first polypeptide comprising the amino acid sequence of SEQ ID NO:290 and a second polypeptide comprising the amino acid sequence of SEQ ID NO:291.
  • a polypeptide comprising a subunit of a multisubunit cytokine and an immunoglobulin Fc domain polypeptide, wherein the Fc domain polypeptide comprises mutations for promoting heterodimerization with a different immunoglobulin Fc domain polypeptide, and one or more mutation(s) that reduce(s) an effector function of an Fc.
  • polypeptide of embodiment P90 wherein the Fc domain is a human IgG1 antibody Fc domain.
  • polypeptide of embodiment P91 wherein the one or more mutation(s) that reduce(s) an effector function of an Fc is selected from L234A, L235A or L235E, G237A, P329A, A330S, and P331S, numbered according to the EU numbering system.
  • polypeptide of embodiment P91 or embodiment P92, wherein the mutations that reduce an effector function of an Fc are L234A, L235A, and P329A, numbered according to the EU numbering system.
  • polypeptide of any one of embodiments P91-P93, wherein the mutations for promoting heterodimerization are K360E and K409W, numbered according to the EU numbering system.
  • polypeptide of any one of embodiments P91-P93, wherein the mutations for promoting heterodimerization are Q347R, D399V, and F405T, numbered according to the EU numbering system.
  • polypeptide of embodiment P94 or P95 wherein the Fc domain further comprises a mutation for promoting disulfide bond formation with a different immunoglobulin Fc domain polypeptide.
  • polypeptide of embodiment P96 wherein when the heterodimerization mutations are K360E and K409W, the mutation for promoting disulfide bond formation is Y349C, numbered according to the EU numbering system.
  • polypeptide of embodiment P96 wherein when the heterodimerization mutations are Q347R, D399V, and F405T, the mutation for promoting disulfide bond formation is S354C, numbered according to the EU numbering system.
  • polypeptide of embodiment P97 comprising an amino acid sequence of SEQ ID NO:290.
  • polypeptide of embodiment P98 comprising an amino acid sequence of SEQ ID NO:291.
  • polypeptide of any one of embodiments P90-P100 further comprising an antibody variable domain.
  • the polypeptide of embodiment P101, wherein the antibody variable domain comprises an antibody heavy chain variable domain.
  • polypeptide of embodiment P102 wherein the antibody heavy chain variable domain is fused to the N-terminus of the polypeptide.
  • polypeptide of any one of embodiments P101-P105 wherein the subunit of the multisubunit cytokine is fused to the C-terminus of the immunoglobulin Fc domain.
  • polypeptide of embodiment P101 wherein the antibody heavy chain variable domain is fused to the C-terminus of the polypeptide.
  • polypeptide of embodiment P107 or P108 wherein the subunit of the multi-subunit cytokine is fused to the N-terminus of the immunoglobulin Fc domain.
  • proteoglycan-binding domain binds one or more proteoglycan(s) specifically expressed in a tumor.
  • proteoglycan-binding domain binds one or more proteoglycan(s) selected from syndecan, serglycin, CSPG4, betaglycan, glypican, perlecan, versican, brevican, and small leucine-rich proteoglycans (SLRPs).
  • proteoglycan(s) selected from syndecan, serglycin, CSPG4, betaglycan, glypican, perlecan, versican, brevican, and small leucine-rich proteoglycans (SLRPs).
  • polypeptide of embodiment P113 wherein the SLRPs are selected from decorin, biglycan, asporin, fibrodulin, and lumican.
  • polypeptide of any one of embodiments P90-P100, or 110-114 further comprising a collagen-binding domain.
  • polypeptide of any one of embodiments P90-P100, or 110-116, further comprising a hyaluronic acid-binding domain is provided.
  • An expression vector comprising a nucleic acid comprising a sequence encoding a polypeptide comprising the amino acid sequence of SEQ ID NO:290 or SEQ ID NO:291, according to any one of embodiments P89-P118.
  • a cell comprising a nucleic acid of embodiment P119 or an expression vector of embodiment P120.
US17/287,849 2018-10-23 2019-10-23 Heterodimeric fc-fused proteins Pending US20220119533A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US17/287,849 US20220119533A1 (en) 2018-10-23 2019-10-23 Heterodimeric fc-fused proteins

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
US201862749489P 2018-10-23 2018-10-23
US201862781898P 2018-12-19 2018-12-19
US201962788499P 2019-01-04 2019-01-04
US201962827347P 2019-04-01 2019-04-01
US201962895889P 2019-09-04 2019-09-04
US17/287,849 US20220119533A1 (en) 2018-10-23 2019-10-23 Heterodimeric fc-fused proteins
PCT/US2019/057721 WO2020086758A1 (en) 2018-10-23 2019-10-23 Heterodimeric fc-fused proteins

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2019/057721 A-371-Of-International WO2020086758A1 (en) 2018-10-23 2019-10-23 Heterodimeric fc-fused proteins

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US17/929,282 Continuation US11787864B2 (en) 2018-10-23 2022-09-01 Heterodimeric Fc-fused proteins

Publications (1)

Publication Number Publication Date
US20220119533A1 true US20220119533A1 (en) 2022-04-21

Family

ID=68583501

Family Applications (2)

Application Number Title Priority Date Filing Date
US17/287,849 Pending US20220119533A1 (en) 2018-10-23 2019-10-23 Heterodimeric fc-fused proteins
US17/929,282 Active US11787864B2 (en) 2018-10-23 2022-09-01 Heterodimeric Fc-fused proteins

Family Applications After (1)

Application Number Title Priority Date Filing Date
US17/929,282 Active US11787864B2 (en) 2018-10-23 2022-09-01 Heterodimeric Fc-fused proteins

Country Status (17)

Country Link
US (2) US20220119533A1 (zh)
EP (1) EP3870598A1 (zh)
JP (1) JP2022505871A (zh)
KR (1) KR20210080460A (zh)
CN (1) CN113286808A (zh)
AU (1) AU2019366956A1 (zh)
BR (1) BR112021007175A2 (zh)
CA (1) CA3117212A1 (zh)
CL (2) CL2021001058A1 (zh)
CO (1) CO2021006474A2 (zh)
IL (1) IL281954A (zh)
MA (1) MA53982A (zh)
MX (1) MX2021004711A (zh)
PE (1) PE20211279A1 (zh)
SG (1) SG11202104136YA (zh)
TW (1) TW202033547A (zh)
WO (1) WO2020086758A1 (zh)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11787864B2 (en) 2018-10-23 2023-10-17 Dragonfly Therapeutics, Inc. Heterodimeric Fc-fused proteins

Families Citing this family (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016104657A1 (ja) * 2014-12-26 2016-06-30 橋本 正 癌治療剤
CA3033475A1 (en) 2016-08-10 2018-02-15 Ajou University Industry-Academic Cooperation Foundation Heterodimeric fc-fused cytokine and pharmaceutical composition comprising the same
CN110944651A (zh) 2017-02-08 2020-03-31 蜻蜓疗法股份有限公司 用于自然杀伤细胞激活的多特异性结合蛋白及其治疗癌症的治疗性用途
PE20220278A1 (es) 2018-02-08 2022-02-25 Dragonfly Therapeutics Inc Dominios variables de anticuerpos que se dirigen al receptor nkg2d
AU2019218125A1 (en) * 2018-02-08 2020-08-20 Dragonfly Therapeutics, Inc. Combination therapy of cancer involving multi-specific binding proteins that activate natural killer cells
MA52366A (fr) 2018-04-25 2021-03-03 Prometheus Biosciences Inc Anticorps anti-tl1a optimisés
EP3818083A2 (en) 2018-07-03 2021-05-12 Elstar Therapeutics, Inc. Anti-tcr antibody molecules and uses thereof
MX2021003543A (es) 2018-09-27 2021-06-23 Xilio Dev Inc Polipeptidos de citocinas enmascaradas.
US11358999B2 (en) 2018-10-03 2022-06-14 Xencor, Inc. IL-12 heterodimeric Fc-fusion proteins
US20210130495A1 (en) * 2019-09-27 2021-05-06 Agenus Inc. Heterodimeric proteins
CA3157024A1 (en) 2019-10-03 2021-04-08 Xencor, Inc. Targeted il-12 heterodimeric fc-fusion proteins
KR20220103721A (ko) 2019-10-24 2022-07-22 프로메테우스 바이오사이언시즈, 인크. Tnf 유사 리간드 1a(tl1a)에 대한 인간화 항체 및 그의 용도
IL297225A (en) 2020-04-10 2022-12-01 Cytomx Therapeutics Inc Activatable cytokine constructs and related compositions and methods
EP4138778A1 (en) * 2020-04-22 2023-03-01 Dragonfly Therapeutics, Inc. Formulation, dosage regimen, and manufacturing process for heterodimeric fc-fused proteins
CN115803091A (zh) * 2020-05-22 2023-03-14 福迈康股份公司 Ace2-fc融合蛋白及其用途
WO2022090469A2 (en) * 2020-10-29 2022-05-05 Formycon Ag Ace2 fusion proteins and uses thereof
TW202304958A (zh) 2021-03-16 2023-02-01 美商Cytomx生物製藥公司 經遮蔽之可活化之細胞介素構築體及相關之組成物及方法
KR20240036570A (ko) * 2021-07-22 2024-03-20 에프. 호프만-라 로슈 아게 이종이량체 Fc 도메인 항체
CN115925984A (zh) * 2021-08-24 2023-04-07 广东东阳光药业有限公司 Gdf15融合蛋白及其用途
WO2023070056A2 (en) * 2021-10-20 2023-04-27 Synthekine, Inc. Heterodimeric fc cytokines and uses thereof
WO2023086772A1 (en) 2021-11-12 2023-05-19 Xencor, Inc. Bispecific antibodies that bind to b7h3 and nkg2d
WO2024086739A1 (en) 2022-10-20 2024-04-25 Synthekine, Inc. Methods and compositions of il12 muteins and il2 muteins

Family Cites Families (125)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4722848A (en) 1982-12-08 1988-02-02 Health Research, Incorporated Method for immunizing animals with synthetically modified vaccinia virus
WO1988007089A1 (en) 1987-03-18 1988-09-22 Medical Research Council Altered antibodies
US5391481A (en) 1990-08-31 1995-02-21 The Trustees Of Columbia University Antibody which is directed against and inhibits collagen binding to a VLA-1 epitope and uses thereof
GB9022040D0 (en) 1990-10-10 1990-11-21 Biopharm Ltd Platelet adhesion inhibitor
US5851794A (en) 1990-10-22 1998-12-22 Alfa Laval Ab Collagen binding protein as well as its preparation
US5173414A (en) 1990-10-30 1992-12-22 Applied Immune Sciences, Inc. Production of recombinant adeno-associated virus vectors
CA2117780A1 (en) 1992-04-10 1993-10-28 Paul F. Goetinck Cartillage matrix protein and methods for use
US5491130A (en) 1992-11-10 1996-02-13 The United States Of America As Represented By The Department Of Health And Human Services Peptide inhibitors of fibronectin and related collagen-binding proteins
US5731168A (en) 1995-03-01 1998-03-24 Genentech, Inc. Method for making heteromultimeric polypeptides
US5849589A (en) 1996-03-11 1998-12-15 Duke University Culturing monocytes with IL-4, TNF-α and GM-CSF TO induce differentiation to dendric cells
EP0950068B1 (en) 1996-05-16 2005-11-09 THE TEXAS A&M UNIVERSITY SYSTEM Collagen binding protein compositions and methods of use
US7951917B1 (en) 1997-05-02 2011-05-31 Genentech, Inc. Method for making multispecific antibodies having heteromultimeric and common components
WO1999029732A2 (en) 1997-12-08 1999-06-17 Lexigen Pharmaceuticals Corporation Heterodimeric fusion proteins useful for targeted immune therapy and general immune stimulation
US6194551B1 (en) 1998-04-02 2001-02-27 Genentech, Inc. Polypeptide variants
US6387663B1 (en) 1998-07-31 2002-05-14 University Of Southern California Targeting pharmaceutical agents to injured tissues
WO2000021989A1 (en) 1998-10-09 2000-04-20 Medimmune, Inc. Decorin binding proteins dbp a and b and genes encoding them
US6737056B1 (en) 1999-01-15 2004-05-18 Genentech, Inc. Polypeptide variants with altered effector function
US7183387B1 (en) 1999-01-15 2007-02-27 Genentech, Inc. Polypeptide variants with altered effector function
US6566489B1 (en) 1999-03-15 2003-05-20 The General Hospital Corporation Syndecan-4 binding protein (S4BP) and uses thereof
US6864235B1 (en) 1999-04-01 2005-03-08 Eva A. Turley Compositions and methods for treating cellular response to injury and other proliferating cell disorders regulated by hyaladherin and hyaluronans
US6908994B1 (en) 1999-05-10 2005-06-21 The Texas A&M University System Collagen-binding proteins from enterococcal bacteria
AU5495500A (en) 1999-06-16 2001-01-02 The Texas A & M University System Decorin binding protein essential peptides and methods of use
AU5622800A (en) 1999-06-18 2001-01-09 Med Immune, Inc. Combined decorin binding protein and outer surface protein compositions and methods of use
CA2394576A1 (en) 1999-12-15 2001-06-21 Research Development Foundation Betaglycan as an inhibin receptor and uses thereof
US6777547B1 (en) 2000-01-31 2004-08-17 Andreas Podbielski Collagen-binding proteins from streptococcus pyogenes
US6517838B1 (en) 2000-06-16 2003-02-11 The Texas A&M University System Decorin binding protein essential peptides and methods of use
WO2002024223A2 (en) 2000-09-21 2002-03-28 The Brigham And Women's Hospital, Inc. Prevention and treatment of streptococcal and staphylococcal infection
JP4281869B2 (ja) 2001-06-22 2009-06-17 中外製薬株式会社 抗グリピカン3抗体を含む細胞増殖抑制剤
US8192744B2 (en) 2002-08-26 2012-06-05 Ibcc Holding As Drug for treating states related to the inhibition of angiogenesis and/or endothelial cell proliferation
US7488792B2 (en) 2002-08-28 2009-02-10 Burnham Institute For Medical Research Collagen-binding molecules that selectively home to tumor vasculature and methods of using same
GB0406415D0 (en) 2004-03-22 2004-04-21 Inst Of Cancer Res The Materials and methods for treatment of cancer
WO2005114186A1 (ja) 2004-05-20 2005-12-01 Wako Pure Chemical Industries, Ltd. ヒアルロン酸バインディングプロテインを用いたヒアルロン酸の測定方法
WO2005124356A2 (en) 2004-06-18 2005-12-29 Roche Diagnostics Gmbh Use of protein cbp2 as a marker for colorectal cancer
NZ579543A (en) 2004-07-09 2011-07-29 Chugai Pharmaceutical Co Ltd Anti-glypican 3 antibody
US7820401B2 (en) 2004-08-23 2010-10-26 Albert Einstein College Of Medicine Of Yeshiva University Collagen VI and cancer
US20090142345A1 (en) 2005-03-15 2009-06-04 Takeda Pharmaceutical Company Limited Prophylactic/therapeutic agent for cancer
JP5140579B2 (ja) 2005-06-08 2013-02-06 カンジェーン コーポレイション 病原菌に対する生体防御を増強するヒアルロン酸結合性ペプチド
CN104356236B (zh) 2005-07-01 2020-07-03 E.R.施贵宝&圣斯有限责任公司 抗程序性死亡配体1(pd-l1)的人单克隆抗体
EP1999154B1 (en) 2006-03-24 2012-10-24 Merck Patent GmbH Engineered heterodimeric protein domains
EP1864996A1 (en) 2006-06-06 2007-12-12 Helmholtz-Zentrum für Infektionsforschung GmbH Peptide associated with rheumatic fever (PARF) and its use as a diagnostic marker.
HU0600578D0 (en) 2006-07-13 2006-09-28 Szilak Labor Bioinformatikai E Nuclear protein transport
CA2660455A1 (en) 2006-08-08 2008-02-14 Seikagaku Corporation Method for determination of molecular weight of hyaluronic acid
WO2008089448A2 (en) 2007-01-19 2008-07-24 Cornell Presearch Foundation, Inc. Methods and compositions for promoting survival & proliferation of endothelial cells & stimulating angiogenesis
US8680247B2 (en) 2007-07-17 2014-03-25 Medarex, L.L.C. Monoclonal antibodies against glypican-3
WO2009089004A1 (en) 2008-01-07 2009-07-16 Amgen Inc. Method for making antibody fc-heterodimeric molecules using electrostatic steering effects
US8663929B2 (en) 2008-03-17 2014-03-04 University Of Miyazaki Method for detection of liver cancer cell using anti-glypican-3 antibody
US20090297479A1 (en) 2008-03-28 2009-12-03 Kiyoshi Ariizumi Dc-hil conjugates for treatment of t-cell disorders
CA2720368C (en) 2008-04-02 2017-08-22 Macrogenics, Inc. Her2/neu-specific antibodies and methods of using same
JP5477287B2 (ja) 2008-04-15 2014-04-23 和光純薬工業株式会社 新規なヒアルロン酸結合能を有するタンパク質及びこれを用いたヒアルロン酸の測定方法
WO2010033866A2 (en) 2008-09-19 2010-03-25 University Of Pittsburgh-Of The Commonwealth System Of Higher Education Monoclonal antibodies for cspg4 for the diagnosis and treatment of basal breast carcinoma
GB0818273D0 (en) 2008-10-06 2008-11-12 Cambridge Entpr Ltd Modulation of cellular activity and differentiation
KR101105428B1 (ko) 2009-02-12 2012-01-17 경북대학교 산학협력단 글리피칸-3 단백질과 특이적으로 결합하는 펩타이드
EP2417159A1 (en) 2009-04-07 2012-02-15 Roche Glycart AG Bispecific anti-erbb-3/anti-c-met antibodies
US20120100106A1 (en) 2009-05-04 2012-04-26 Purdue Research Foundation Collagen-binding synthetic peptidoglycans for wound healing
JP5764127B2 (ja) 2009-08-17 2015-08-12 ロシュ グリクアート アーゲー 標的化イムノコンジュゲート
US9493578B2 (en) 2009-09-02 2016-11-15 Xencor, Inc. Compositions and methods for simultaneous bivalent and monovalent co-engagement of antigens
SG184427A1 (en) * 2010-04-20 2012-11-29 Genmab As Heterodimeric antibody fc-containing proteins and methods for production thereof
RU2624027C2 (ru) 2010-04-23 2017-06-30 Дженентек, Инк. Получение гетеромультимерных белков
CN103068846B9 (zh) 2010-08-24 2016-09-28 弗·哈夫曼-拉罗切有限公司 包含二硫键稳定性Fv片段的双特异性抗体
WO2012032080A1 (en) 2010-09-07 2012-03-15 F-Star Biotechnologische Forschungs- Und Entwicklungsges.M.B.H Stabilised human fc
US8906649B2 (en) 2010-09-27 2014-12-09 Janssen Biotech, Inc. Antibodies binding human collagen II
JP6167040B2 (ja) 2010-11-05 2017-07-19 ザイムワークス,インコーポレイテッド Fcドメイン中に突然変異を有する、安定したヘテロ二量体抗体の設計
JP6100694B2 (ja) 2010-11-15 2017-03-22 ピエリス ファーマシューティカルズ ゲーエムベーハー グリピカン−3(gpc3)に対して親和性を有するヒトリポカリン2の突然変異タンパク質
BR112013013311A2 (pt) * 2010-11-30 2017-09-19 Chugai Pharmaceutical Co Ltd agente terapêutico de indução de citotoxicidade
US10689447B2 (en) 2011-02-04 2020-06-23 Genentech, Inc. Fc variants and methods for their production
DK3489255T3 (da) * 2011-02-10 2021-08-23 Roche Glycart Ag Muterede interleukin-2-polypeptider
EP3590965A1 (en) 2011-03-29 2020-01-08 Roche Glycart AG Antibody fc variants
EP3070104B1 (en) 2011-04-19 2017-12-27 The United States of America, as represented by The Secretary, Department of Health and Human Services Human monoclonal antibodies specific for glypican-3 and use thereof
EA201892619A1 (ru) 2011-04-29 2019-04-30 Роше Гликарт Аг Иммуноконъюгаты, содержащие мутантные полипептиды интерлейкина-2
ES2708076T3 (es) 2011-05-24 2019-04-08 Symic Ip Llc Peptidoglicanos sintéticos que se unen al ácido hialurónico, preparación y métodos de uso
PL3351261T3 (pl) * 2011-10-11 2021-12-06 Universität Zürich Lek złożony zawierający IL-12 i środek do blokowania cząsteczek hamujących komórki T w terapii nowotworów
US20140322216A1 (en) 2011-11-08 2014-10-30 The Trustees Of The University Of Pennsylvania Glypican-3-specific antibody and uses thereof
EP2804586A4 (en) 2012-01-19 2016-03-16 Univ Johns Hopkins BIOMATERIALS COMPRISING HYALURONIC ACID-BINDING PEPTIDES AND BIFUNCTIONAL BIOPOLYMER MOLECULES FOR HYALURONIC ACID RETENTION AND TISSUE ENGINEERING APPLICATIONS
NZ630551A (en) 2012-04-20 2017-11-24 Merus Nv Methods and means for the production of ig-like molecules
WO2013163766A1 (en) 2012-05-04 2013-11-07 Cangene Corporation ANTIMICROBIAL COMPOSITIONS COMPRISING A HYALURONIC ACID BINDING PEPTIDE AND A β-LACTAM ANTIBIOTIC
WO2013174783A1 (en) 2012-05-23 2013-11-28 Pieris Ag Lipocalin muteins with binding-affinity for glypican-3 (gpc-3) and use of lipocalin muteins for target-specific delivery to cells expressing gpc-3
JP6494507B2 (ja) 2012-06-01 2019-04-03 ザ ユナイテッド ステイツ オブ アメリカ, アズ リプレゼンテッド バイ ザ セクレタリー, デパートメント オブ ヘルス アンド ヒューマン サービシーズ グリピカン−3に対する高親和性モノクローナル抗体およびその使用
DK2880170T3 (en) 2012-08-02 2016-10-24 Hoffmann La Roche PROCEDURE FOR PREPARING SOLUBLE FcR AS Fc FUSION WITH INERT IMMUNOGLOBULIN Fc REGION AND APPLICATIONS THEREOF
SG11201408530YA (en) 2012-08-02 2015-03-30 Hoffmann La Roche Method for producing monomeric and multimeric molecules and uses thereof
MX365382B (es) 2012-08-07 2019-05-31 Roche Glycart Ag Una combinación de inmunoconjugado y anticuerpo para usarse en el tratamiento de cáncer.
US9951145B2 (en) 2012-11-27 2018-04-24 Ajou University Industry—Academic Cooperation Foundation CH3 domain variant pair inducing formation of heterodimer of heavy chain constant region of antibody at high efficiency, method for preparing same, and use thereof
US20140302037A1 (en) * 2013-03-15 2014-10-09 Amgen Inc. BISPECIFIC-Fc MOLECULES
SG11201507429TA (en) * 2013-03-15 2015-10-29 Genentech Inc Il-22 polypeptides and il-22 fc fusion proteins and methods of use
AU2014232416B2 (en) 2013-03-15 2017-09-28 Xencor, Inc. Modulation of T Cells with Bispecific Antibodies and FC Fusions
US10047167B2 (en) 2013-03-15 2018-08-14 Eli Lilly And Company Methods for producing fabs and bi-specific antibodies
EP2992010B1 (en) 2013-04-29 2021-03-24 F.Hoffmann-La Roche Ag Fc-receptor binding modified asymmetric antibodies and methods of use
US10093745B2 (en) 2013-05-29 2018-10-09 The Regents Of The University Of California Anti-CSPG4 fusions with interferon for the treatment of malignancy
AR096891A1 (es) 2013-07-12 2016-02-03 Hanmi Pharm Ind Co Ltd Conjugado de monómero polipéptido biológicamente activo y conjugado de fragmento fc de inmunoglobulina, que muestra aclaramiento mediado por receptor reducido, y el método para la preparación del mismo
UA117289C2 (uk) 2014-04-02 2018-07-10 Ф. Хоффманн-Ля Рош Аг Мультиспецифічне антитіло
US20160032007A1 (en) 2014-04-28 2016-02-04 Duke University Human Antibody Fragments Against Chondroitin Sulfate Proteoglycan 4 (CSPG4)
BR122021009041B1 (pt) 2014-05-06 2022-11-29 Genentech, Inc Métodos para a preparação de uma proteína heteromultimérica
CA2952727A1 (en) 2014-06-27 2015-12-30 Innate Pharma Multispecific nkp46 binding proteins
WO2016007919A2 (en) 2014-07-11 2016-01-14 Regents Of The University Of Minnesota Antibody fragments for detecting cancer and methods of use
MA40764A (fr) 2014-09-26 2017-08-01 Chugai Pharmaceutical Co Ltd Agent thérapeutique induisant une cytotoxicité
RU2713131C1 (ru) 2014-11-06 2020-02-03 Ф. Хоффманн-Ля Рош Аг ВАРИАНТЫ Fc-ОБЛАСТИ С МОДИФИЦИРОВАННЫМИ СВОЙСТВАМИ СВЯЗЫВАНИЯ FcRn И БЕЛКА А
SI3215528T1 (sl) 2014-11-06 2019-11-29 Hoffmann La Roche Variante regije Fc s spremenjeno vezavo FcRn in postopki uporabe
CA2967350C (en) 2014-11-12 2021-11-23 The General Hospital Corporation Anti-chondroitin sulfate proteoglycan 4 antibodies and uses thereof
WO2016154019A1 (en) 2015-03-20 2016-09-29 Orbsen Therapeutics Limited Modulators of syndecan-2 and uses thereof
US10881711B2 (en) 2015-04-06 2021-01-05 The General Hospital Corporation Anti-CSPG4 reagents and methods of treating cancer
WO2016208754A1 (ja) 2015-06-24 2016-12-29 学校法人慶應義塾 抗グリピカン-1-免疫抗原受容体
RU2740672C2 (ru) * 2015-08-07 2021-01-19 ЭйЭлЭкс Онколоджи Инк. Конструкции, имеющие sirp-альфа домен или его вариант
AU2016322919B2 (en) 2015-09-14 2022-12-22 Regents Of The University Of Minnesota NK cells exhibiting an adaptive phenotype and methods for preparing and for using
ES2809125T3 (es) 2015-09-23 2021-03-03 Bristol Myers Squibb Co Moléculas de armazón a base de fibronectina de unión a glipicano-3
WO2017062604A1 (en) 2015-10-06 2017-04-13 Regents Of The University Of Minnesota Therapeutic compounds and methods
KR101851380B1 (ko) 2015-10-12 2018-04-23 아주대학교산학협력단 효모접합을 이용한 항체 ch3 도메인 이종이중체 돌연변이쌍 제조 방법 및 이에 의하여 제조된 ch3 돌연변이체 쌍
KR101796688B1 (ko) 2015-10-29 2017-12-01 재단법인 목암생명과학연구소 신규 항-글리피칸 3 항체 및 이를 포함하는 약학적 조성물
JP6983824B2 (ja) 2016-07-04 2021-12-17 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft 新規抗体フォーマット
AU2017305170A1 (en) 2016-08-02 2019-02-14 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Monoclonal antibodies targeting glypican-2 (GPC2) and use thereof
WO2018030806A1 (ko) * 2016-08-10 2018-02-15 아주대학교산학협력단 항체 중쇄불변부위 이종이중체에 융합된 사이토카인 및 이를 포함하는 약제학적 조성물
CA3033475A1 (en) 2016-08-10 2018-02-15 Ajou University Industry-Academic Cooperation Foundation Heterodimeric fc-fused cytokine and pharmaceutical composition comprising the same
CN110214148A (zh) * 2016-10-14 2019-09-06 Xencor股份有限公司 含有IL-15/IL-15Rα Fc融合蛋白和PD-1抗体片段的双特异性异源二聚体融合蛋白
CA3054079A1 (en) 2017-02-20 2018-08-23 Dragonfly Therapeutics, Inc. Proteins binding her2, nkg2d and cd16
CN110831634A (zh) 2017-03-08 2020-02-21 密歇根大学董事会 磷脂酰肌醇蛋白聚糖-3肽试剂和方法
RU2019133202A (ru) 2017-03-27 2021-04-28 Ф. Хоффманн-Ля Рош Аг Улучшенные антигенсвязывающие рецепторы
US11242403B2 (en) 2017-04-26 2022-02-08 Mitsubishi Tanabe Pharma Corporation Syndecan-1 (CD138) binding agents and uses thereof
WO2018199318A1 (ja) 2017-04-28 2018-11-01 国立大学法人高知大学 抗gpc-1抗体
JP2019014449A (ja) 2017-07-10 2019-01-31 トヨタ自動車株式会社 車両用動力伝達装置
CA3112984A1 (en) 2017-09-07 2019-03-14 Dragonfly Therapeutics, Inc. Proteins binding nkg2d, cd16 and a tumor-associated antigen
EP3684791A1 (en) * 2017-09-21 2020-07-29 Merck Patent GmbH Fusion protein comprising an fgf-18 moiety
BR112020005676A2 (pt) * 2017-09-22 2020-10-20 WuXi Biologics Ireland Limited novos complexos polipeptídicos biespecíficos
CN111246885A (zh) 2017-10-20 2020-06-05 豪夫迈·罗氏有限公司 从单特异性抗体生成多特异性抗体的方法
AU2019218125A1 (en) 2018-02-08 2020-08-20 Dragonfly Therapeutics, Inc. Combination therapy of cancer involving multi-specific binding proteins that activate natural killer cells
DE102018208278A1 (de) 2018-05-25 2019-11-28 Robert Bosch Gmbh Betriebsassistenzverfahren, Steuereinheit, Betriebsassistenzsystem und Arbeitsvorrichtung
MX2021000287A (es) * 2018-07-11 2021-09-08 Momenta Pharmaceuticals Inc Composiciones y métodos relacionados con constructos de dominio de unión a antígeno-fc modificados genéticamente.
PE20211279A1 (es) 2018-10-23 2021-07-19 Dragonfly Therapeutics Inc Proteinas heterodimericas fusionadas con fc

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11787864B2 (en) 2018-10-23 2023-10-17 Dragonfly Therapeutics, Inc. Heterodimeric Fc-fused proteins

Also Published As

Publication number Publication date
WO2020086758A8 (en) 2021-05-06
AU2019366956A1 (en) 2021-05-20
PE20211279A1 (es) 2021-07-19
IL281954A (en) 2021-05-31
US20230033425A1 (en) 2023-02-02
CA3117212A1 (en) 2020-04-30
KR20210080460A (ko) 2021-06-30
MX2021004711A (es) 2021-07-02
SG11202104136YA (en) 2021-05-28
EP3870598A1 (en) 2021-09-01
US11787864B2 (en) 2023-10-17
MA53982A (fr) 2022-01-26
BR112021007175A2 (pt) 2021-08-10
CL2022000900A1 (es) 2023-01-20
JP2022505871A (ja) 2022-01-14
CL2021001058A1 (es) 2021-11-12
TW202033547A (zh) 2020-09-16
WO2020086758A1 (en) 2020-04-30
CO2021006474A2 (es) 2021-06-10
CN113286808A (zh) 2021-08-20

Similar Documents

Publication Publication Date Title
US11787864B2 (en) Heterodimeric Fc-fused proteins
KR20210003170A (ko) IL-15/IL-15Rα 이종이량체 Fc 융합 단백질 및 이의 용도
AU2021201451A1 (en) Proteins binding NKG2D, CD16, and EGFR, CCR4, or PD-L1
JP7397055B2 (ja) Cd137及びox40に結合する抗体分子
JP2021512630A (ja) Nkg2d受容体を標的とする抗体可変ドメイン
KR20210013160A (ko) 다중-특이적 결합 단백질 및 그에 대한 개선
KR102645411B1 (ko) Her2, nkg2d 및 cd16에 결합하는 다중-특이적 결합 단백질 및 사용 방법
WO2020011968A1 (en) Anti-cd137 antibodies
BR112020016939A2 (pt) Proteínas de ligação multiespecíficas que se ligam a cd33, nkg2d, e cd16 e métodos de uso
KR20220035122A (ko) 신규한 인터루킨-2 변이체 및 이의 이작용성 융합 분자
CN114901679A (zh) 全新被掩蔽的细胞因子及其应用
US20230272041A1 (en) Formulation, Dosage Regimen, and Manufacturing Process for Heterodimeric FC-Fused Proteins
JP2022105121A (ja) Psma、nkg2dおよびcd16に結合するタンパク質
KR20220083770A (ko) Nkg2d, cd16 및 flt3에 결합하는 단백질
KR20230162013A (ko) Nk 세포 관여를 위해 사이토카인에 융합된 nkp46-결합 부위, 암 항원 결합 부위를 포함하는 다중특이적 단백질
US20230257467A1 (en) Proteins binding nkg2d, cd16 and egfr

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

AS Assignment

Owner name: DRAGONFLY THERAPEUTICS, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:CHEUNG, ANN F.;CUILLEROT, JEAN-MARIE;GRINBERG, ASYA;AND OTHERS;REEL/FRAME:063970/0497

Effective date: 20200304