WO2019191668A1 - Binding molecules specific for fcyriia and uses thereof - Google Patents

Binding molecules specific for fcyriia and uses thereof Download PDF

Info

Publication number
WO2019191668A1
WO2019191668A1 PCT/US2019/024977 US2019024977W WO2019191668A1 WO 2019191668 A1 WO2019191668 A1 WO 2019191668A1 US 2019024977 W US2019024977 W US 2019024977W WO 2019191668 A1 WO2019191668 A1 WO 2019191668A1
Authority
WO
WIPO (PCT)
Prior art keywords
fcgriia
binding molecule
seq
antibody
binding
Prior art date
Application number
PCT/US2019/024977
Other languages
French (fr)
Inventor
Katherine Ann Vousden
Bo Chen
Gary Patrick SIMS
Original Assignee
Katherine Ann Vousden
Bo Chen
Sims Gary Patrick
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 Katherine Ann Vousden, Bo Chen, Sims Gary Patrick filed Critical Katherine Ann Vousden
Publication of WO2019191668A1 publication Critical patent/WO2019191668A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • 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
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/33Crossreactivity, e.g. for species or epitope, or lack of said crossreactivity
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • 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
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • 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
    • C07K2317/77Internalization into the cell
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value

Definitions

  • FcyRs are a family of cell surface receptors that bind to the Fc portion of antibodies of the immunoglobulin G (IgG) subclasses.
  • Human Fc gamma receptors differ in function, binding affinity, and in their cellular distribution 1 .
  • FcyRs there are five FcyRs: the high-affinity receptor FcyRI (CD64), which can bind monomeric IgG; and the low-affinity receptors FcyRI I A (CD32A), FcyRI IB (CD32B), FcyRIIIA (CD16A), and FcyRIIIB (CD16B), which bind weakly to monomeric IgG, but avidly to immune complexes of IgG.
  • FcyRI, FcgRIIA, FcyRIIIA, and FcyRIIIB are considered to have activating properties, whereas FcyRI IB is predominately inhibitory.
  • FcgRIIA and FcyRI IB are the most closely related receptors. The extracellular regions of these receptors, which are responsible for interactions with IgG, share greater than 90% sequence identity 2 . Sequence differences in the intracellular signaling regions of FcgRIIA and FcyRI IB mediate the alternative cellular responses.
  • FcyRs mediate several cellular processes, including antigen or pathogen uptake, degranulation, antigen presentation, and antibody-dependent cellular cytotoxicity (ADCC).
  • ADCC antibody-dependent cellular cytotoxicity
  • FcyRs can interact with other receptors to influence the production of specific cytokines. Failure of the immune system to appropriately limit the reactivity of FcyRs can play a role in the development of inflammatory, immune-mediated, or autoimmune diseases or disorders 3 .
  • SLE Systemic lupus erythematosus
  • dsDNA nuclear antigens
  • ssDNA nuclear antigens
  • nucleic acid associated proteins e.g ., RNP, histones, Smith, Ro.
  • Disease manifestations in the skin, lung, and kidney are associated with deposition of immune complexes 4 .
  • Immune-complex -mediated activation of FcgRIIA has been implicated to play a role in the pathogenesis of SLE 5 .
  • Anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV) refers to a heterogeneous group of inflammatory diseases of blood vessels with multisystem
  • AAV comprises granulomatosis with polyangiitis (GPA), microscopic polyangiitis (MPA), necrotizing crescentic glomerulonephritis (NCGN), and eosinophilic granulomatosis (EGPA).
  • GPA polyangiitis
  • MPA microscopic polyangiitis
  • NCGN necrotizing crescentic glomerulonephritis
  • EGPA eosinophilic granulomatosis
  • MPO myeloperoxidase
  • PR3 proteinase 3
  • lactoferrin lactoferrin
  • ANCAs not only serve as a diagnostic marker, but they also play a direct pathogenic role in the disease.
  • ANCAs can induce the direct activation of neutrophils via FcgRIIA, which can drive vascular injury 9 .
  • ANCAs may also trigger FcgRIIA-dependent induction of neutrophil extracellular traps (NETs), cytokines, and chemokines, which may contribute to inflammation and the autoimmune response 10 . Therefore, FcgRIIA appears to play a central role in the development and pathology of AAV.
  • NETs neutrophil extracellular traps
  • cytokines cytokines
  • chemokines which may contribute to inflammation and the autoimmune response 10 . Therefore, FcgRIIA appears to play a central role in the development and pathology of AAV.
  • Immune thrombocytopenia is an autoimmune bleeding disorder characterized by the production of auto-reactive antibodies directed against platelet antigens. Autoantibodies coating the surface of platelets promote their clearance by phagocytic macrophages of the reticuloendothelial system.
  • the repertoire and cellular expression of FcyRs differ between mouse and human, and although FcyRII is the most broadly expressed FcyR in humans, it is absent in mice. Using mice, transgenic for human FcgRIIA and deficient for the murine activating FcyRs, it was demonstrated that passively administered anti-platelet antibodies triggered immune thrombocytopenia in an FcgRIIA-dependent manner 11 .
  • NETs neutrophil extracellular traps
  • NETs neutrophil extracellular traps
  • ANCA-associated vasculitis In addition to their pathogenic role in ANCA-associated vasculitis, there is evidence to indicate that NETs may contribute to sepsis, thrombosis, acute kidney injury, acute lung injury, chronic obstructive pulmonary disease, glomerulonephritis, toxic liver injury, stroke, atherogenesis and Type I diabetes 21,22 . Since FcgRIIA plays a crucial role in the formation of NETs 10 , blocking FcgRIIA may have a beneficial role in the treatment of NET-associated disorders.
  • AD As Anti-drug antibodies
  • FcgRIIA is the predominant activating FcyR that is responsible for immune complex-mediated effector functions, blocking FcgRIIA may be able to inhibit ADA-mediated adverse effects.
  • compositions that specifically bind to FcgRIIA, and methods for the use of such compositions, such as for the treatment or prevention of an inflammatory, immune-mediated, or autoimmune disease or disorder.
  • the disclosure provides FcgRIIA-binding molecules, for example, humanized monoclonal antibodies capable of inhibiting FcgRIIA activity, and methods of using the FcgRIIA binding molecules, for example, in treating or preventing inflammatory, immune-mediated, or autoimmune diseases or disorders.
  • the invention provides an isolated binding molecule that specifically binds to FcgRIIA, wherein the binding molecule comprises an immunoglobulin variable heavy chain complementarity determining region 2 (VH-CDR2) comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 19 and SEQ ID NO: 20.
  • the invention provides an isolated binding molecule that specifically binds to FcgRIIA, wherein the binding molecule comprises an immunoglobulin variable light chain complementarity determining region 1 (VL-CDR1) comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 22, SEQ ID NO: 23, SEQ ID NO: 24, SEQ ID NO: 25, and SEQ ID NO: 26.
  • VH-CDR2 immunoglobulin variable heavy chain complementarity determining region 2
  • VL-CDR1 immunoglobulin variable light chain complementarity determining region 1
  • a binding molecule of the invention comprises a VH-CDR2 and a VL-CDR1 comprising amino acid sequences: (i) SEQ ID NO: 19 and SEQ ID NO: 22; (ii) SEQ ID NO: 19 and SEQ ID NO: 23; (iii) SEQ ID NO: 19 and SEQ ID NO: 24; (iv) SEQ ID NO: 20 and SEQ ID NO: 25; (v) SEQ ID NO: 20 and SEQ ID NO: 23; or (vi) SEQ ID NO: 20 and SEQ ID NO: 26, respectively.
  • the invention provides an isolated binding molecule that specifically binds to FcgRIIA, wherein the binding molecule comprises: (a) an immunoglobulin variable heavy chain complementarity determining region 2 (VH-CDR2) and an immunoglobulin variable light chain complementarity determining region 1 (VL-CDR1) comprising amino acid sequences: (i) SEQ ID NO: 19 and SEQ ID NO: 22; (ii) SEQ ID NO: 19 and SEQ ID NO: 23; (iii) SEQ ID NO: 19 and SEQ ID NO: 24; (iv) SEQ ID NO: 20 and SEQ ID NO: 25; (v) SEQ ID NO: 20 and SEQ ID NO: 23; or (vi) SEQ ID NO: 20 and SEQ ID NO: 26, respectively; (b) an immunoglobulin variable heavy chain complementarity determining region 1 (VH-CDR1) comprising SEQ ID NO: 29; (c) an immunoglobulin variable heavy chain complementarity determining region 3 (VH-CDR3)
  • the binding molecule comprises a heavy chain variable (VH) region and a light chain variable (VL) region, comprising amino acid sequences selected from the group consisting of: (i) SEQ ID NO: 33 and SEQ ID NO: 34, (ii) SEQ ID NO: 35 and SEQ ID NO: 36, (iii) SEQ ID NO: 37 and SEQ ID NO: 38, (iv) SEQ ID NO: 39 and SEQ ID NO: 40, (v) SEQ ID NO: 41 and SEQ ID NO: 42, and (vi) SEQ ID NO: 43 and SEQ ID NO: 44, respectively.
  • the binding molecule comprises SEQ ID NO: 33 and SEQ ID NO: 34.
  • the invention provides an isolated binding molecule that competes or cross-competes with one or more of the binding molecules described above.
  • the binding molecule of the invention is selected from a murine antibody, a human antibody, a humanized antibody, a chimeric antibody, monoclonal antibody, a polyclonal antibody, a recombinant antibody, a bi-specific antibody, a multi-specific antibody, and an antigen-binding fragment thereof.
  • the binding molecule of the invention is selected from an Fv, an Fab, an F(ab’)2, an Fab’, a dsFv fragment, a single chain Fv (scFV), an sc(Fv)2, a disulfide- linked (dsFv), a diabody, a triabody, a tetrabody, a minibody, or a single chain antibody.
  • the binding molecule of the invention can comprise an immunoglobulin (Ig) heavy chain constant region.
  • the constant region is a human IgG constant region.
  • the constant region comprises amino acid substitutions at Kabat positions 234, 235, and 331, wherein: the amino acid at Kabat position 234 is substituted with Phenylalanine (F), the amino acid at Kabat position 235 is substituted with Glutamic acid (E), and the amino acid at Kabat position 331 is substituted with Serine (S).
  • the constant region comprises one or more substitutions of amino acid residues at positions 251-257, 285-290, 308-314, 385-389, and 428-436, relative to a wild-type human IgG constant region, wherein the amino acid position numbering is according to the EU index as set forth in Kabat.
  • the constant region comprises amino acid substitutions at Kabat positions 252, 254, and 256, wherein: the amino acid at Kabat position 252 is substituted with Tyrosine (Y), the amino acid at Kabat position 254 is substituted with Threonine (T), and the amino acid at Kabat position 256 is substituted with Glutamic acid (E).
  • the binding molecule of the invention can comprise an immunoglobulin light chain constant region.
  • the light chain constant region is a human kappa constant region.
  • the binding molecule of the invention specifically binds human FcgRIIA 131R with an affinity characterized by a dissociation constant (K D ) of about 0.16 nM, as measured by a BIAcore assay.
  • the binding molecule of the invention specifically binds human FcgRIIA 131H with an affinity characterized by a dissociation constant (K D ) of about 0.13 nM, as measured by a BIAcore assay.
  • the binding molecule does not specifically bind to FcyRI, FcyRIIB, or FcyRIII.
  • the binding molecule of the invention can be conjugated to an agent, for example, an agent selected from the group consisting of an antimicrobial agent, a therapeutic agent, a prodrug, a peptide, a protein, an enzyme, a lipid, a biological response modifier, a phannaceutical agent, a lymphokine, a heterologous antibody or fragment thereof, a detectable label, a polyethylene glycol (PEG), a toxin, and a combination of two or more of any said agents.
  • an agent selected from the group consisting of an antimicrobial agent, a therapeutic agent, a prodrug, a peptide, a protein, an enzyme, a lipid, a biological response modifier, a phannaceutical agent, a lymphokine, a heterologous antibody or fragment thereof, a detectable label, a polyethylene glycol (PEG), a toxin, and a combination of two or more of any said agents.
  • an agent selected from the group consisting of an antim
  • the invention further provides a composition comprising a binding molecule of the invention.
  • the composition is a diagnostic reagent.
  • the invention provides a method for inhibiting
  • RNP-IC ribonucleoprotein-immune complex
  • PBMC peripheral blood mononuclear cell
  • ANCA anti -neutrophil cytoplasmic antibody
  • the invention provides a method for inhibiting
  • ribonucleoprotein-immune complex RNP-IC-mediated pro-inflammatory molecule production in a peripheral blood mononuclear cell (PBMC).
  • the method comprising contacting the PBMC with a binding molecule of the invention.
  • the pro-inflammatory molecule may be interleukin-6 or may be tumomecrosis factor alpha.
  • the invention provides a method of treating or preventing an inflammatory, immune-mediated, or autoimmune disease or disorder in a subject, the method comprising administering to a subject in need of treatment or to a subject susceptible to the disease or disorder an effective amount of a binding molecule or composition of the invention.
  • the disease or disorder is preferably selected from ANCA-associated vasculitis (AAV), systemic lupus erythematosus (SLE), lupus nephritis, membranous nephritis, immune thrombocytopenia (ITP), rheumatoid arthritis, polymyositis, dermatomyositis, pemphigus, hemolytic anemia, mixed connective tissue disease, Sjogren’s syndrome, scleroderma, sepsis, thrombosis, acute kidney injury, acute lung injury, chronic obstructive pulmonary disease, glomerulonephritis, toxic liver injury, stroke, atherogenesis and Type I diabetes, an autoantibody disorder, and an immune- complex-mediated disorder.
  • the method comprises administering a second active agent.
  • the invention provides an isolated nucleic acid molecule comprising a nucleotide sequence encoding a binding molecule of the invention, optionally linked to a regulatory sequence; a host cell transformed with the nucleic acid molecule, preferably a mammalian host cell; and a vector comprising the nucleic acid molecule. Also provided is a composition comprising the nucleic acid molecule, host cell, or vector of the invention.
  • the invention provides a method of making a binding molecule that specifically binds FcgRIIA, the method comprising culturing the host cell of the invention under suitable conditions for producing the binding molecule. In some aspects, the method further comprises isolating the binding molecule.
  • the invention provides a kit comprising a binding molecule or a nucleic acid molecule of the invention.
  • FIGS. 1A-1C show consensus FcgRIIA cynomolgus sequences and alignments to human FcgRIIA
  • FIG. 1A shows the consensus amino acid sequence of cynomolgus FcgRIIA (SEQ ID NO: 1). Single nucleotide polymorphism (SNP) variants among cynomolgus populations are shown in bold type. Minor alleles corresponding to the major human allele are underlined. Shaded residues indicate minor allele frequency (MAF) of >20%.
  • SNP single nucleotide polymorphism
  • FIG. 1B shows alignment of human FcgRIIA (P12318; as 1-317; SEQ ID NO: 2) and cynomolgus FcgRIIA (SEQ ID NO: 3). Cyno amino acids that are non-homologous to human are shaded. Variations where the minor cyno allele corresponds to human are underlined.
  • FIG. 1C shows the consensus FcgRIIA full-length cynomolgus transcript (SEQ ID NO: 4).
  • FIG. 2 shows epitope competition assay data for humanization of IV.3.
  • FIG. 3 shows alignment of mouse IV.3 VH (SEQ ID NO: 5) and VL (SEQ ID NO: 8) with humanized IV.3 (CamIV3 VH (SEQ ID NO: 6) and VL (SEQ ID NO: 9)) and selected human germline sequences (SEQ ID NO: 7, SEQ ID NO: 10).
  • FIGS. 4A-4B show epitope competition assay data for a panel of humanized IV.3 antibodies.
  • FIG. 4A shows binding to human FcgRIIA 131H.
  • FIG. 4B shows binding to human FcgRIIA 131R.
  • FIGS. 5A-5G show that optimized IV.3 Abs are specific for human FcgRIIA binding, but not for other FcyRs; and that MED 19600 lacks CDC and ADCC effector functions.
  • FIG. 5A shows binding data for human FcgRIIA.
  • FIG. 5B shows binding data for FcyRIIB.
  • FIG. 5C shows binding data for human FcyRI.
  • FIG. 5A-5G show that optimized IV.3 Abs are specific for human FcgRIIA binding, but not for other FcyRs; and that MED 19600 lacks CDC and ADCC effector functions.
  • FIG. 5A shows binding data for human FcgRIIA.
  • FIG. 5B shows binding data
  • FIG. 5D shows binding data for human FcyRIIIA - 158F allotype.
  • FIG. 5E shows binding data for human FcyRIIIA - 158V allotype.
  • FIG. 5F shows CDC activity of MED 19600 compared with wild type control 9600 IgGl and isotype control IgG (R347-Tm) antibodies.
  • FIG. 5G shows ADCC activity of MED 19600 compared with wild type control 9600 IgGl and isotype control IgG (R347-Tm) antibodies.
  • the plots represent the mean ⁇ standard deviation. Representative plots of three independent experiments are shown.
  • FIG. 6 shows that MEDI9600 competes with intravenous immunoglobulin (IVIG) for binding to FcgRIIA.
  • IVIG intravenous immunoglobulin
  • FIGS. 7A-7G show that optimized IV.3 Abs internalize FcgRIIA from the surface of monocytes from human 131H/H donors (FIG. 7 A), human 131R/R donors (FIG. 7B), and cynomolgous monkeys (FIG. 7C).
  • FIG. 7D shows internalization of FcgRIIA by MED 19600 using confocal microscopy.
  • FIGS. 7E-G show that MEDI9600 internalizes FcgRIIA from the surface of neutrophils from human 131H/H donors (FIG. 7E), human 131R/R donors (FIG. 7F), and cynomolgous monkeys (FIG. 7G).
  • FIGS. 7E-G representative data from three independent human and monkey experiments are shown.
  • FIGS. 8A-8D show that optimized IV.3 Abs block RNP-IC-induced IFN-a expression from human and cyno PBMC, using cells from human 131 H/H donors (FIG. 8A), human 131R/R donors (FIG. 8B), and cynomologus monkeys (FIG. 8C).
  • FIG. 8D shows that MEDI9600 (30 pg/mL) inhibited Ig-IC induced TNF-a and IL-6 protein in whole blood.
  • FIGS. 9A-9G show that MED 19600 (clone 32LO0352) specifically blocks anti neutrophil cytoplasmic antibody (ANCA) induced neutrophil activation.
  • MED 19600 clone 32LO0352
  • ANCA neutrophil cytoplasmic antibody
  • FIGS. 10A-10G show that MEDI9600 protects mice from anti platelets antibody— induced thrombocytopenia.
  • FIG. 10D shows free FcgRIIA on platelets, measured by flow cytometry, at 24, 48, and 96 hrs after a single lmg/kg or 10 mg/kg i.p dose of MED 19600.
  • FIG. 10E shows free FcgRIIA on neutrophils, measured by flow cytometry, at 24, 48, and 96 hrs after a single lmg/kg or 10 mg/kg i.p dose of MED 19600.
  • FIG. 10D shows free FcgRIIA on platelets, measured by flow cytometry, at 24, 48, and 96 hrs after a single lmg/kg or 10 mg/kg i.p dose of MED 19600.
  • FIG. 10E shows free FcgRIIA on neutrophils, measured by flow cytometry, at 24, 48, and 96 hrs after a single l
  • FIG. 10F shows serum concentration of MED 19600 in the mice, measured by human IgG ELISA at 24, 48, and 96 hrs following delivery of a lmg/kg or 10 mg/kg i.p dose of MED 19600.
  • FIG. 10G shows platelet numbers in the mice after prophylactic treatment with MEDI9600.
  • MEDI9600 or control Ab R347-Tm was injected i.p. at lOmg/ml 24hrs before treatment, thrombocytopenia was induced with 2 pg rat anti-mouse CD41 Ab delivered i.p. at day 0.
  • FIGS. 11A-11C show that blockade of FcgRIIA by MEDI9600 has no adverse effects on neutrophil function.
  • FIG. 12A - 12B shows that ex vivo treatment of human whole blood with MED 19600 alone has no effect on the protein expression profile.
  • FIGS. 13A-13C show the results of a single-dose pharmacokinetic and exploratory pharmaco-dynamic study of MEDI9600.
  • FIG. 13A shows the serum concentration of
  • MED 19600 in cynomolgus monkeys at various time points after a single dose shows that MED 19600 induced a dose- response reduction of FcgRIIA fluorescence intensity on monocytes (FIG. 13B) and
  • FIG. 13C granulocytes
  • FIG. 14A - 14B show that, in a 13 week safety study, MED 19600 did not affect cynomologous monkey platelet counts (FIG. 14A) or body weights (FIG. 14B).
  • the platelet count and body weight measurements were performed on (5/sex/group) monkeys that had been treated with weekly injections of vehicle control (IV and SC), 7.5 mg/kg (IV), 30 mg/kg (IV),
  • the present invention provides molecules that bind to FcgRIIA
  • such molecules are antibodies or antigen-binding fragments thereof, which specifically bind to FcgRIIA.
  • Related polynucleotides, compositions comprising the anti-FcgRIIA binding molecules, and methods of making the anti-FcgRIIA binding molecules are also provided.
  • Methods of using the novel anti-FcgRIIA antibodies such as diagnostic methods and methods of treating inflammatory, immune-mediated, or autoimmune disease or disorders are further provided.
  • “and/or” is to be taken as specific disclosure of each of the two specified features or components with or without the other.
  • the term“and/or” as used in a phrase such as“A and/or B” is intended to include A and B, A or B, A (alone), and B (alone).
  • the term“and/or” as used in a phrase such as“A, B, and/or C” is intended to include A, B, and C; A, B, or C; A or B; A or C; B or C; A and B; A and C; B and C; A (alone); B (alone); and C (alone).
  • FcgRIIA refers to Fc receptor-gamma IIA.
  • the full-length amino acid and nucleotide sequences for human and cynomolgus monkey ( Macaca fasciculari ) FcgRIIA are known in the art. (See, e.g., FIG. 1.)
  • the terms“FcgRIIA” and“CD32A” are used
  • antibody refers to an immunoglobulin molecule that recognizes and specifically binds to a target, such as a protein, polypeptide, peptide, carbohydrate, polynucleotide, lipid, or combinations of the foregoing through at least one antigen recognition site within the variable region of the immunoglobulin molecule.
  • a target such as a protein, polypeptide, peptide, carbohydrate, polynucleotide, lipid, or combinations of the foregoing through at least one antigen recognition site within the variable region of the immunoglobulin molecule.
  • the term “antibody” encompasses polyclonal antibodies; monoclonal antibodies; multispecific antibodies, such as bispecific antibodies generated from at least two intact antibodies; humanized antibodies; human antibodies; chimeric antibodies; fusion proteins comprising an antigen-determination portion of an antibody; and any other modified immunoglobulin molecule comprising an antigen recognition site, so long as the antibodies exhibit the desired biological activity.
  • Antibodies can be of any the five major classes of immunoglobulins: IgA, IgD, IgE, IgG and IgM, or subclasses (isotypes) thereof (e.g. IgGl, IgG2, IgG3, IgG4, IgAl and IgA2), based on the identity of their heavy-chain constant domains referred to as alpha, delta, epsilon, gamma, and mu respectively.
  • the different classes of immunoglobulins have different and well-known subunit structures and three-dimensional configurations. There are two classes of mammalian light chains, lambda and kappa. Antibodies can be naked or conjugated to other molecules such as toxins, radioisotopes, etc.
  • antibody fragment refers to a portion of an intact antibody comprising the complementarity determining variable regions of the antibody. Fragments of a full-length antibody can be an antigen-binding fragment of an antibody. Examples of antibody fragments include, but are not limited to Fab, Fab’, F(ab’)2, and Fv fragments, linear antibodies, single chain antibodies (e.g., ScFvs), and multispecific antibodies formed from antibody fragments.
  • A“monoclonal antibody” refers to a homogeneous antibody population that is involved in the highly specific recognition and binding of a single antigenic determinant, or epitope. This is in contrast to polyclonal antibodies, which typically include different antibodies directed against different antigenic determinants.
  • the term“monoclonal” can apply to both intact and full-length monoclonal antibodies, as well as to antibody fragments (such as Fab, Fab’, F(ab’)2, Fv), single chain (scFv) mutants, fusion proteins comprising an antibody portion, and any other modified immunoglobulin molecule comprising an antigen recognition site.
  • “monoclonal antibody” refers to such antibodies made in any number of ways including, but not limited to, by hybridoma, phage selection, recombinant expression, and transgenic animals.
  • the term“humanized antibody” refers to an antibody derived from a non-human (e.g ., murine) immunoglobulin, which has been engineered to contain minimal non-human (e.g., murine) sequences.
  • humanized antibodies are human immunoglobulins in which residues from the complementary determining region (CDR) are replaced by residues from the CDR of a non-human species (e.g, mouse, rat, rabbit, or hamster) that have the desired specificity, affinity, and capability (Jones et al., 1986, Nature, 321 :522-525; Riechmann et al., 1988, Nature, 332:323-327; Verhoeyen et al, 1988, Science, 239: 1534-1536).
  • the Fv framework region (FW) residues of a human immunoglobulin are replaced with the corresponding residues in an antibody from a non-human species that has the desired specificity, affinity, and capability.
  • Humanized antibodies can be further modified by the substitution of additional residues either in the Fv framework region and/or within the replaced non-human residues to refine and optimize antibody specificity, affinity, and/or capability.
  • humanized antibodies will comprise substantially all of at least one, and typically two or three, variable domains containing all or substantially all of the CDR regions that correspond to the non-human immunoglobulin whereas all or substantially all of the FR regions are those of a human immunoglobulin consensus sequence.
  • Humanized antibody can also comprise at least a portion of an
  • immunoglobulin constant region or domain typically that of a human immunoglobulin.
  • Fc immunoglobulin constant region or domain
  • human antibody means an antibody produced by a human or an antibody having an amino acid sequence corresponding to an antibody produced by a human made using any technique known in the art.
  • the definition of a human antibody includes intact or full-length antibodies comprising at least one human heavy and/or light chain polypeptide such as, for example, an antibody comprising murine light chain and human heavy chain polypeptides.
  • chimeric antibodies refers to antibodies wherein the amino acid sequence of the immunoglobulin molecule is derived from two or more species.
  • the variable region of both light and heavy chains corresponds to the variable region of antibodies derived from one species of mammals (e.g, mouse, rat, rabbit, etc.) with the desired specificity, affinity, and capability while the constant regions are homologous to the sequences in antibodies derived from another (usually human) to avoid eliciting an immune response in that species.
  • A“blocking” antibody or an“antagonist” antibody is one that inhibits or reduces biological activity of the antigen it binds, such as FcgRIIA.
  • blocking antibodies or antagonist antibodies substantially or completely inhibit the biological activity of the antigen. Desirably, the biological activity is reduced by 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or even 100%.
  • germlining means that amino acids at specific positions in an antibody are mutated back to those in the germ line.
  • The“IgGl triple mutant” or“IgGl-TM” antibody format is a human IgGl isotype containing three single amino acid substitutions, L234F/L235E/P331S, within the lower hinge and CH2 domain (Oganesyan et al., Acta Crystallogr. D Biol. Crystallogr. 64:700-704, 2008).
  • the TM causes a profound decrease in binding to human FcyRI, FcyRII, FcyRIII, and Clq, resulting in a human isotype with very low effector function.
  • YTE or“YTE mutant” refer to a mutation in IgGl Fc that results in an increase in the binding to human FcRn and improves the serum half-life of the antibody having the mutation.
  • a YTE mutant comprises a combination of three mutations,
  • a typical antibody comprises at least two heavy (H) chains and two light (L) chains interconnected by disulfide bonds.
  • Each heavy chain is comprised of a heavy chain variable region (abbreviated herein as VH) and a heavy chain constant region.
  • the heavy chain constant region is comprised of three domains, CH1, CH2, and CH3.
  • Each light chain is comprised of a light chain variable region (abbreviated herein as VL) and a light chain constant region (CL).
  • the light chain constant region is comprised of one domain, Cl.
  • the variable regions of the heavy and light chains contain a binding domain that interacts with an antigen.
  • the constant regions of the antibodies can mediate the binding of the immunoglobulin to host tissues or factors, including various cells of the immune system (e.g. effector cells) and the first component (Clq) of the classical complement system.
  • VH and VL regions can be further subdivided into regions of hypervariability, termed complementarity-determining regions (CDRs), interspersed with regions that are more conserved, termed framework (FW) regions.
  • CDRs complementarity-determining regions
  • FW framework regions
  • the CDRs in each chain are held together in close proximity by the FW regions and, with the CDRs from the other chain, contribute to the formation of the antigen-binding site of antibodies.
  • Each VH and VL is composed of three CDRs and four FWs, arranged from amino-terminus to carboxy-terminus in the following order: FW1, CDR1, FW2, CDR2, FW3, CDR3, FW4.
  • CDRs There are at least two techniques for determining CDRs: (1) an approach based on cross-species sequence variability (Kabat et al, Sequences of Proteins of Immunological Interest , 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md. (1991)); and (2) an approach based on crystallographic studies of antigen-antibody complexes (Al-lazikani et al ., ./. Molec. Biol. 273:927-948 (1997)). In addition, combinations of these two approaches are sometimes used in the art to determine CDRs.
  • the amino acid position numbering as in Kabat refers to the numbering system used for heavy chain variable domains or light chain variable domains (approximately residues 1-107 of the light chain and residues 1-113 of the heavy chain). ETsing this numbering system, the actual linear amino acid sequence can contain fewer or additional amino acids corresponding to a shortening of, or insertion into, a FW or CDR of the variable domain.
  • a heavy chain variable domain can include a single amino acid insert (residue 52a, according to Kabat) after residue 52 of H2 and inserted residues (e.g, residues 82a, 82b, and 82c, etc., according to Kabat) after heavy chain FW residue 82.
  • the Kabat numbering of residues can be determined for a given antibody by alignment at regions of homology of the sequence of the antibody with a“standard” Kabat numbered sequence. Chothia refers instead to the location of the structural loops (Chothia and Lesk, J Mol. Biol. 196:901-917 (1987)).
  • the end of the Chothia CDR-H1 loop when numbered using the Kabat numbering convention varies between H32 and H34 depending on the length of the loop (this is because the Kabat numbering scheme places the insertions at H35A and H35B; if neither 35A nor 35B is present, the loop ends at 32; if only 35A is present, the loop ends at 33; if both 35 A and 35B are present, the loop ends at 34).
  • the AbM hypervariable regions represent a compromise between the Kabat CDRs and Chothia structural loops, and are used by Oxford Molecular’s AbM antibody modeling software. See Table 1.
  • IMGT immunoglobulin variable regions
  • CDR the numbering system for the immunoglobulin variable regions. See, e.g. , Lefranc, M.P. et al. , Dev. Comp. Immunol. 27: 55-77 (2003).
  • the IMGT numbering system was based on an alignment of more than 5,000 sequences, structural data, and characterization of hypervariable loops and allows for easy comparison of the variable and CDR regions for all species.
  • VH-CDR1 is at positions 26 to 35
  • VH-CDR2 is at positions 51 to 57
  • VH-CDR3 is at positions 93 to 102
  • VL-CDR1 is at positions 27 to 32
  • VL-CDR2 is at positions 50 to 52
  • VL-CDR3 is at positions 89 to 97.
  • VH CDRs sequences described correspond to the classical Kabat numbering locations, namely Kabat VH-CDR1 is at positions 31-35, VH- CDR2 is a positions 50-65, and VH-CDR3 is at positions 95-102.
  • VL-CDR1, VL-CDR2 and VL-CDR3 also correspond to classical Kabat numbering locations, namely positions 24-34, 50- 56 and 89-97, respectively.
  • Binding affinity generally refers to the strength of the sum total of non-covalent interactions between a single binding site of a molecule (e.g, an antibody) and its binding partner (e.g, an antigen). Unless indicated otherwise, as used herein,“binding affinity” refers to intrinsic binding affinity which reflects a 1 : 1 interaction between members of a binding pair (e.g, antibody and antigen).
  • the affinity of a molecule X for its partner Y can generally be represented by the dissociation constant (KD). Affinity can be measured by common methods known in the art, including those described herein. Low-affinity antibodies generally bind antigen slowly and tend to dissociate readily, whereas high-affinity antibodies generally bind antigen faster and tend to remain bound longer.
  • the affinity or avidity of an antibody for an antigen can be determined experimentally using any suitable method known in the art, e.g ., flow cytometry, enzyme-linked immunosorbent assay (ELISA), or radioimmunoassay (RIA), or kinetics (e.g, KINEXA® or BIACORETM analysis).
  • ELISA enzyme-linked immunosorbent assay
  • RIA radioimmunoassay
  • kinetics e.g, KINEXA® or BIACORETM analysis.
  • Direct binding assays as well as competitive binding assay formats can be readily employed. (See, e.g, Berzofsky et a/.,“Antibody -Antigen Interactions,” In Fundamental Immunology, Paul, W. E., ed., Raven Press: New York, N.Y. (1984); Kuby, Immunology, W. H. Freeman and Company: New York, N.Y.
  • the measured affinity of a particular antibody-antigen interaction can vary if measured under different conditions (e.g, salt concentration, pH, temperature).
  • affinity and other antigen-binding parameters e.g, K D or Kd, K on , K 0ff
  • measurements of affinity and other antigen-binding parameters are made with standardized solutions of antibody and antigen, and a standardized buffer, as known in the art.
  • IC50 is normally expressed as an IC50 value, in nM or pM, unless otherwise stated.
  • IC50 is the median inhibitory concentration of an antibody molecule. In functional assays, IC50 is the concentration that reduces a biological response by 50% of its maximum. In ligand-binding studies, IC50 is the concentration that reduces receptor binding by 50% of maximal specific binding level. IC50 can be calculated by any number of means known in the art.
  • the fold improvement in potency for the antibodies or polypeptides of the invention as compared to a reference antibody can be at least about 2-fold, at least about 4-fold, at least about 6-fold, at least about 8-fold, at least about lO-fold, at least about 20-fold, at least about 30-fold, at least about 40-fold, at least about 50-fold, at least about 60-fold, at least about 70-fold, at least about 80-fold, at least about 90-fold, at least about lOO-fold, at least about 1 lO-fold, at least about l20-fold, at least about l30-fold, at least about l40-fold, at least about l50-fold, at least about 160-fold, at least about 170-fold, or at least about 180-fold or more.
  • the terms“inhibit,”“block,” and“suppress” are used interchangeably and refer to any statistically significant decrease in biological activity, including full blocking of the activity.
  • “inhibition” can refer to a decrease of about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or 100% in biological activity.
  • the terms“inhibition” or “suppression” are applied to describe, e.g, an effect on the FcgRIIA signal transduction pathway, the terms refer to the ability of an FcgRIIA binding molecule to statistically significantly decrease FcgRIIA-induced cell activation or signal transduction relative to an untreated (control) cell.
  • the cell that expresses FcgRIIA can be a naturally occurring cell or cell line, (e.g. , macrophage, neutrophil, eosinophil, platelet) or can be recombinantly produced by introducing a nucleic acid encoding FcgRIIA into a host cell.
  • the FcgRIIA binding molecule can inhibit FcgRIIA-mediated cell activation or signal transduction in an FcgRIIA- expressing cell by at least 10%, or at least 20%, or at least 30%, or at least 40%, or at least 50%, or at least 60%, or at least 70%, or at least 80%, or at least 90% or about 100%, as determined, for example, by flow cytometry, Western blotting, ELISA, or other assays known to those of skill in the art.
  • An“isolated” polypeptide, antibody, binding molecule, polynucleotide, vector, or cell is in a form not found in nature.
  • Isolated polypeptides, antibodies, binding molecules, polynucleotides, vectors, or cells include those which have been purified to a degree that they are no longer in a form in which they are found in nature.
  • a polypeptide, antibody, binding molecule, polynucleotide, vector, or cell that is isolated is substantially pure.
  • the term“substantially pure” refers to purity of greater than 75%, preferably greater than 80% or 90%, and most preferably greater than 95%.
  • “subject” or“individual” or“animal” or“patient” or“mammal,” is meant any subject, particularly a mammalian subject, for whom diagnosis, prognosis, or therapy is desired.
  • Mammalian subjects include humans, domestic animals, farm animals, sports animals, and zoo animals including, e.g., humans, non-human primates, dogs, cats, guinea pigs, rabbits, rats, mice, horses, cattle, bears, and so on.
  • composition refers to a preparation that is in such form as to permit the biological activity of the active ingredient to be effective and which contains no additional components that are unacceptably toxic to a subject to which the composition would be administered.
  • Such composition can be sterile and can comprise a pharmaceutically acceptable carrier, such as physiological saline.
  • Suitable pharmaceutical compositions can comprise one or more of a buffer, a surfactant, a stabilizing agent, a preservative, an absorption promoter to enhance bioavailability and/or other conventional solubilizing or dispersing agents.
  • An“effective amount” of a binding molecule as disclosed herein is an amount sufficient to carry out a specifically stated purpose. An“effective amount” can be determined empirically and in a routine manner, in relation to the stated purpose.
  • label when used herein refers to a detectable compound or composition that is conjugated directly or indirectly to a binding molecule, so as to generate a“labeled” binding molecule.
  • the label can be detectable by itself (e.g ., radioisotope labels or fluorescent labels) or, as in the case of, e.g., an enzymatic label, can catalyze chemical alteration of a substrate compound or composition that is detectable.
  • Terms such as“treating” or“treatment” or“to treat” or“alleviating” or“to alleviate” refer to therapeutic measures that cure, slow down, lessen symptoms of, and/or halt progression of a diagnosed pathologic condition or disorder. Thus, those in need of treatment include those already with the disorder.
  • a subject is successfully“treated” for an inflammatory, immune-mediated, or autoimmune disease or disorder according to the methods provided herein if the patient shows, e.g, total, partial, or transient alleviation or elimination of symptoms associated with the disease or disorder.
  • Prevent refers to prophylactic or preventative measures that prevent and/or slow the development of a targeted pathologic condition or disorder.
  • those in need of prevention include those prone to have or susceptible to the disorder.
  • an inflammatory, immune-mediated, or autoimmune disease or disorder is successfully prevented according to the methods provided herein if the patient develops, transiently or permanently, e.g, fewer or less severe symptoms associated with the disease or disorder, or a later onset of symptoms associated with the disease or disorder, than a patient who has not been subject to the methods of the invention.
  • polypeptide “peptide,” and“protein” are used interchangeably herein to refer to polymers of amino acids of any length.
  • the polymer can be linear or branched, it can comprise modified amino acids and non-amino acids can interrupt it.
  • the terms also encompass an amino acid polymer that has been modified naturally or by intervention; for example, disulfide bond formation, glycosylation, lipidation, acetylation, phosphorylation or any other manipulation or modification such as conjugation with a labeling component.
  • polypeptides containing one or more analogs of an amino acid including, for example, unnatural amino acids, etc.
  • the polypeptides can occur as single chains or associated chains.
  • A“conservative amino acid substitution” is one in which one amino acid residue is replaced with another amino acid residue having a similar side chain.
  • Families of amino acid residues having similar side chains have been defined in the art, including basic side chains (e.g ., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g, asparagine, glutamine, serine, threonine, tyrosine, cysteine), nonpolar side chains (e.g, glycine, alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan), beta-branched side chains (e.g, threonine, valine, isoleucine) and aromatic side chains (e.g, tyrosine, phenylalanine, tryptophan, histidine).
  • basic side chains e.g
  • substitution of a phenylalanine for a tyrosine is a conservative substitution.
  • conservative substitutions in the amino acid sequences of the binding molecules of the invention do not abrogate the binding of the binding molecule to the antigen(s), i.e., FcgRIIA, to which the binding molecule binds.
  • Methods of identifying conservative nucleotide and amino acid substitutions which do not eliminate antigen-binding are well-known in the art (see, e.g, Brummell et al., Biochem. 32: 1180-1 187 (1993); Kobayashi et al. , Protein Eng. 12(10):879- 884 (1999); and Burks et al., Proc. Natl. Acad. Sci. U.S.A. 94:.412-417 (1997)).
  • A“polynucleotide,” as used herein can include one or more“nucleic acids,”“nucleic acid molecules,” or“nucleic acid sequences,” and refers to a polymer of nucleotides of any length, and includes DNA and RNA.
  • the polynucleotides can be deoxyribonucleotides, ribonucleotides, modified nucleotides or bases, and/or their analogs, or any substrate that can be incorporated into a polymer by DNA or RNA polymerase.
  • a polynucleotide can comprise modified nucleotides, such as methylated nucleotides and their analogs. The preceding description applies to all polynucleotides referred to herein, including RNA and DNA.
  • vector means a construct, which is capable of delivering and, in some embodiments expressing, one or more gene(s) or sequence(s) of interest in a host cell.
  • vectors include, but are not limited to, viral vectors, naked DNA or RNA expression vectors, plasmid, cosmid or phage vectors, DNA or RNA expression vectors associated with cationic condensing agents, DNA or RNA expression vectors encapsulated in liposomes, and certain eukaryotic cells, such as producer cells.
  • nucleic acids or polypeptides refer to two or more sequences or subsequences that are the same or have a specified percentage of nucleotides or amino acid residues that are the same, when compared and aligned (introducing gaps, if necessary) for maximum correspondence, not considering any conservative amino acid substitutions as part of the sequence identity.
  • the percent identity can be measured using sequence comparison software or algorithms or by visual inspection. Various algorithms and software are known in the art that can be used to obtain alignments of amino acid or nucleotide sequences.
  • sequence alignment algorithm is the algorithm described in Karlin et al. , Proc. Natl. Acad. Sci., 87:2264-2268 (1990), as modified in Karlin et al, Proc. Natl. Acad. Sci., 90:5873-5877 (1993), and incorporated into the NBLAST and
  • XBLAST programs Altschul et al, Nucleic Acids Res., 25:3389-3402 (1991)). In certain embodiments, Gapped BLAST can be used as described in Altschul et al, Nucleic Acids Res. 25:3389-3402 (1997).
  • BLAST-2, WU-BLAST-2 Altschul et al, Methods in Enzymology, 266:460-480 (1996)), ALIGN, ALIGN-2 (Genentech, South San Francisco, California) or Megalign (DNASTAR) are additional publicly available software programs that can be used to align sequences.
  • the percent identity between two nucleotide sequences is determined using the GAP program in the GCG software package ( e.g ., using a
  • the GAP program in the GCG software package which incorporates the algorithm of Needleman and Wunsch (J. Mol. Biol. (48):444-453 (1970)), can be used to determine the percent identity between two amino acid sequences (e.g., using either a BLOSUM 62 matrix or a PAM250 matrix, and a gap weight of 16, 14, 12, 10, 8, 6, or 4 and a length weight of 1, 2, 3, 4, 5).
  • the percent identity between nucleotide or amino acid sequences is determined using the algorithm of Myers and Miller ( CABIOS 4: 11-17 (1989)).
  • the percent identity can be determined using the ALIGN program (version 2.0) and using a PAM120 with residue table, a gap length penalty of 12 and a gap penalty of 4.
  • One skilled in the art can determine appropriate parameters for maximal alignment by particular alignment software. In certain embodiments, the default parameters of the alignment software are used.
  • the percentage identity“X” of a first amino acid sequence to a second sequence amino acid is calculated as 100 x (Y/Z), where Y is the number of amino acid residues scored as identical matches in the alignment of the first and second sequences (as aligned by visual inspection or a particular sequence alignment program) and Z is the total number of residues in the second sequence. If the length of a first sequence is longer than the second sequence, the percent identity of the first sequence to the second sequence will be higher than the percent identity of the second sequence to the first sequence.
  • the present invention provides FcgRIIA binding molecules, /. e. , anti -FcgRIIA antibodies and antigen-binding fragments thereof, which specifically bind FcgRIIA
  • FcgRIIA binding molecule or“binding molecule that binds to FcgRIIA” or“anti -FcgRIIA” refers to a binding molecule that is capable of binding FcgRIIA with sufficient affinity such that the binding molecule is useful as a therapeutic agent or diagnostic reagent in targeting FcgRIIA.
  • a binding molecule that“specifically binds to FcgRIIA” binds to an unrelated, non-FcgRIIA protein to an extent of less than about 10% of the binding of the binding molecule to FcgRIIA, as measured, e.g ., by a radioimmunoassay (RIA), BIACORE® (using recombinant FcgRIIA as the analyte and binding molecule as the ligand, or vice versa), KINEXA®, or other binding assays known in the art.
  • RIA radioimmunoassay
  • BIACORE® using recombinant FcgRIIA as the analyte and binding molecule as the ligand, or vice versa
  • KINEXA® or other binding assays known in the art.
  • binding molecule that binds to FcgRIIA has a dissociation constant (K D ) of ⁇ 1 mM, ⁇ 100 nM, ⁇ 10 nM, ⁇ 1 nM, ⁇ 0.1 nM, ⁇ 10 pM, ⁇ 1 pM, or ⁇ 0.1 pM.
  • Exemplary binding molecules of the present disclosure include variants of the mouse monoclonal antibody IV.3 (Looney RJ. et al, ./. Immunol. 136(5): 1641 (1986)), including humanized, optimized, germlined, and/or other versions of these antibodies, anti-FcgRIIA TM antibodies, and serum half-life-optimized anti-FcgRIIA YTE antibodies (e.g, K44VHa-N56Q, K44VHa6-N56Q, or K2Ha-N56Q).
  • Exemplary antibodies of the present disclosure include clones 32L00350, 32LO0351, 32LO0352, 32LO0354, 32LO0355, and 32LO0356.
  • “Clone 32LO0352” and“MEDI9600” refer to the same molecule and the terms are used interchangeably herein.
  • the invention also embraces variants and equivalents that are substantially homologous to the FcgRIIA-binding molecules set forth herein. These can contain, for example, conservative amino acid substitutions.
  • this disclosure provides an FcgRIIA binding molecule that can specifically bind to the same FcgRIIA epitope as a binding molecule comprising the heavy chain variable region (VH) and light chain variable region (VL) of any one of clones 32L00350, 32LO0351, 32LO0352, 32LO0354, 32LO0355, or 32LO0356.
  • epitope refers to a target protein determinant capable of binding to a binding molecule of the invention.
  • Epitopes usually consist of chemically active surface groupings of molecules such as amino acids or sugar side chains, and usually have specific three-dimensional structural characteristics, as well as specific charge characteristics. Conformational and non-conformational epitopes are
  • binding molecules can be identified based on their ability to cross- compete (e.g ., to competitively inhibit the binding of, in a statistically significant manner) with binding molecules such as clones 32L00350, 32LO0351, 32LO0352, 32LO0354, 32LO0355, or 32LO0356, in standard FcgRIIA binding or activity assays.
  • the invention provides FcgRIIA binding molecules that compete for binding to FcgRIIA with another FcgRIIA binding molecule of the invention, such as one of clones 32L00350, 32LO0351, 32LO0352, 32LO0354, 32LO0355, or 32LO0356.
  • Another FcgRIIA binding molecule of the invention such as one of clones 32L00350, 32LO0351, 32LO0352, 32LO0354, 32LO0355, or 32LO0356.
  • the ability of a binding molecule to inhibit the binding of, e.g., clones 32L00350, 32LO0351, 32LO0352, 32LO0354, 32LO0355, or 32LO0356 demonstrates that the test binding molecule can compete with clones 32L00350, 32LO0351, 32LO0352, 32LO0354, 32LO0355, or
  • binding molecule can, according to non-limiting theory, bind to the same or a related (e.g, a structurally similar or spatially proximal) epitope on FcgRIIA as the FcgRIIA binding molecule with which it competes.
  • FcgRIIA as any of clones 32L00350, 32LO0351, 32LO0352, 32LO0354, 32LO0355, or 32LO0356.
  • the term“competes” indicates that a binding molecule competes uni directionally for binding to FcgRIIA with any one of 32L00350, 32LO0351, 32LO0352, 32LO0354,
  • 32LO0355, or 32LO0356 The term“cross-competes” indicates that a binding molecule competes bidirectionally for binding to FcgRIIA with any one of 32L00350, 32LO0351, 32LO0352, 32LO0354, 32LO0355, or 32LO0356.
  • the FcgRIIA binding molecule is a murine antibody, a human antibody, a humanized antibody, a chimeric antibody, a monoclonal antibody, a polyclonal antibody, a recombinant antibody, a bi-specific antibody, a multi specific antibody, or any combination thereof.
  • FcgRIIA binding molecules comprise a Fab, a Fab’, a F(ab’) 2 , a Fd, a Fv, a scFv, a disulfide linked Fv, a V-NAR domain, an IgNar, an intrabody, an IgGACFLZ, a minibody, a F(ab’) 3 , a tetrabody, a triabody, a diabody, a single-domain antibody, DVD-Ig, Fcab, mAb 2 , a (scFv) 2 , or a scFv-Fc.
  • An FcgRIIA binding molecule provided herein can include, in addition to a VH and a VL, a heavy chain constant region or fragment thereof.
  • the heavy chain constant region is a human heavy chain constant region, e.g ., a human IgG constant region, e.g. , a human IgGl constant region.
  • binding molecules of the invention are produced to comprise an altered Fc region, in which one or more alterations have been made in the Fc region in order to change functional and/or pharmacokinetic properties of the binding molecule. Such alterations may result in altered effector function, reduced immunogenicity, and/or an increased serum half-life.
  • the Fc region interacts with a number of ligands, including Fc receptors, the complement protein Clq, and other molecules, such as proteins A and G. These interactions are essential for a variety of effector functions and downstream signaling events including antibody dependent cell-mediated cytotoxicity (ADCC) and complement dependent cytotoxicity (CDC).
  • ADCC antibody dependent cell-mediated cytotoxicity
  • CDC complement dependent cytotoxicity
  • the FcgRIIA binding molecules of the invention have reduced or ablated affinity for an Fc ligand responsible for facilitating effector function, compared to an FcgRIIA binding molecule not comprising the modification in the Fc region.
  • the FcgRIIA binding molecule has no ADCC activity and/or no CDC activity.
  • the FcgRIIA binding molecule does not bind to an Fc receptor and/or complement factors.
  • the FcgRIIA binding molecule has no effector function. Selecting particular constant domains to optimize desired effector functions is within the ordinary skill in the art.
  • the binding molecule is of the IgGl subtype, and optionally comprises the TM format (L234F/L235E/P331S), as disclosed supra in the
  • a heavy chain constant region or fragment thereof can include one or more amino acid substitutions relative to a wild-type IgG constant domain, wherein the modified IgG has an increased half-life compared to the half-life of an IgG having the wild-type IgG constant domain.
  • the IgG constant domain can contain one or more amino acid substitutions of amino acid residues at positions 251-257, 285-290, 308-314, 385-389, and 428- 436, wherein the amino acid position numbering is according to the EU index as set forth in Kabat.
  • the IgG constant domain can contain one or more of a substitution of the amino acid at Kabat position 252 with Tyrosine (Y), Phenylalanine (F), Tryptophan (W), or Threonine (T), a substitution of the amino acid at Kabat position 254 with Threonine (T), a substitution of the amino acid at Kabat position 256 with Serine (S), Arginine (R), Glutamine (Q), Glutamic acid (E), Aspartic acid (D), or Threonine (T), a substitution of the amino acid at Kabat position 257 with Leucine (L), a substitution of the amino acid at Kabat position 309 with Proline (P), a substitution of the amino acid at Kabat position 311 with Serine (S), a substitution of the amino acid at Kabat position 428 with Threonine (T), Leucine (L), Phenylalanine (F), or Serine (S), a substitution of the amino acid at Kabat position 433 with Arginine (R)
  • the IgG constant domain can contain amino acid substitutions relative to a wild-type human IgG constant domain including as substitution of the amino acid at Kabat position 252 with Tyrosine (Y), a substitution of the amino acid at Kabat position 254 with Threonine (T), and a substitution of the amino acid at Kabat position 256 with Glutamic acid (E).
  • the binding molecule is of the IgGl subtype, and optionally comprises the triple mutant YTE, as disclosed supra in the Definitions section.
  • An FcgRIIA binding molecule provided herein can include a light chain constant region or fragment thereof.
  • the light chain constant region is a kappa constant region or a lambda constant region, e.g ., a human kappa constant region or a human lambda constant region.
  • FcgRIIA binding molecules can have beneficial properties.
  • the binding molecule can inhibit, suppress, or block various FcgRIIA-mediated activities, e.g. , immune-complex -induced type I interferon expression in plasmacytoid dendritic cells (DCs), immune-complex-induced cytokines/chemokines expression in DCs, immune- complex-induced platelet activation, immune-complex -induced antigen presentation, immune- complex-induced neutrophil extracellular traps (NETs) formation, and degranulation in neutrophil activation.
  • DCs plasmacytoid dendritic cells
  • NETs immune-complex-induced cytokines/chemokines expression in DCs
  • NETs immune-complex induced neutrophil extracellular traps
  • the binding molecules provided herein can bind to FcgRIIA with a binding affinity characterized by a dissociation constant (K D ) of about 100 nM to about 0.1 nM as measured by a BiacoreTM assay or on a Kinetic Exclusion Assay (KinExA) 3000 platform.
  • K D dissociation constant
  • an anti-FcgRIIA antibody or antigen-binding fragment thereof can specifically bind to FcgRIIA, e.g., human FcgRIIA or cynomolgus monkey FcgRIIA, or an antigenic fragment thereof, with a dissociation constant or K D of less than 10 -6 M, of less than 10 -7 M, of less than 10 -8 M, of less than 10 -9 M, of less than 10 -10 M, of less than 10 -11 M, of less than 10 -12 M, of less than 10 -13 M, of less than 10 -14 M, or of less than 10 -15 M as measured, e.g. , by BiacoreTM or KinExA®.
  • the humanized anti-FcgRIIA antibody or antigen-binding fragment thereof can specifically bind to FcgRIIA, e.g., human FcgRIIA or cynomolgus monkey FcgRIIA, or an antigenic fragment thereof, with a dissoci
  • MED 19600 can bind to human FcgRIIA (131R) with a Ko of about 0.15 nM, to human FcgRIIA (131H) with a K D of about 0.13 nM, and to cynomolgus monkey FcgRIIA with a K D of about 31.3 nM, as measured by a BIAcore assay.
  • an FcgRIIA binding molecule of the invention binds to
  • an FcgRIIA binding molecule binds to FcgRIIA or an antigenic fragment thereof with a K 0ff of less than 10 -3 s -1 , less than 5x 10 -3 s -1 , less than 10 -4 s -1 , less than 5> ⁇ l0 _4 s -1 , less than 10 -5 s -1 , less than 5x l0 -5 s -1 , less than 10 -6 s -1 , less than 5x 1o -6 s -1 , less than less than 5x l0 -7 s -1 , less than 10 -8 s -1 , less than 5x l0 -8 s -1 , less than 10 -9 s -1 , less than 5x l0
  • the humanized anti-FcgRIIA antibody MED 19600 can bind to human FcgRIIA (131R) with a K 0ff of about 7.35 x 10 ⁇ s -1 , to human FcgRIIA (131H) with a K 0ff of about 3.37 x 10 -4 s -1 , and to cynomolgus monkey F cyRIIA with a K 0ff of about 9.04 x 10 -2 s -1 , as measured by a BIAcore assay.
  • an FcgRIIA binding molecule of the invention binds to
  • MED 19600 can bind to human FcgRIIA (131R) with a K on of about 4.98 x 10 6 M -1 s -1 , to human FcgRIIA (131H) with a K on of about 2.60 x 10 6 M -1 s -1 , and to cynomolgus monkey FcgRIIA with a K on of about 2.88 x 10 6 M -1 s -1 , as measured by a BIAcore assay.
  • a VH and/or VL amino acid sequence or portion thereof, including a CDR sequence can be, e.g.
  • an FcyRIIA-binding molecule having VH and VL regions with a certain percent similarity to a VH region or VL region, or having one or more substitutions, e.g, conservative substitutions, can be obtained by
  • mutagenesis e.g, site-directed or PCR-mediated mutagenesis
  • nucleic acid molecules encoding VH and/or VL regions described herein
  • testing of the encoded altered binding molecule for binding to FcgRIIA and optionally testing for retained function using the functional assays described herein.
  • the disclosure further provides an FcgRIIA binding molecule that is conjugated to a heterologous agent.
  • the agent can be an antimicrobial agent, a therapeutic agent, a prodrug, a peptide, a protein, an enzyme, a lipid, a biological response modifier, a pharmaceutical agent, a lymphokine, a heterologous antibody or fragment thereof, a detectable label, a polyethylene glycol (PEG), or a combination of two or more of any said agents.
  • PEG polyethylene glycol
  • Heteroconjugate anti -FcgRIIA antibodies are discussed in more detail elsewhere herein.
  • the term“binding molecule” includes antibodies and antigen-binding fragments thereof.
  • the FcgRIIA-binding molecule is a polypeptide that is not an antibody.
  • a variety of methods for identifying and producing non-antibody polypeptides that bind with high affinity to a protein target are known in the art. See, e.g, Skerra, Curr. Opin. Biotechnol. 18:295-304 (2007), Hosse et al, Protein Science 15:14-27 (2006), Gill et al, Curr. Opin. Biotechnol. 17:653-658 (2006), Nygren, FEBS J. 275:2668-76 (2008), and Skerra, FEBSJ.
  • phage display technology can been used to identify and/or produce an FcgRIIA-binding polypeptide.
  • the polypeptide comprises a protein scaffold of a type selected from the group consisting of protein A, a lipocalin, a fibronectin domain, an ankyrin consensus repeat domain, and thioredoxin.
  • Monoclonal anti-FcgRIIA antibodies can be prepared using hybridoma methods, such as those described by Kohler and Milstein Nature 256:495 (1975). Using the hybridoma method, a mouse, hamster, or other appropriate host animal, is immunized to elicit the production by lymphocytes of antibodies that will specifically bind to an immunizing antigen. Lymphocytes can also be immunized in vitro. Following immunization, the lymphocytes are isolated and fused with a suitable myeloma cell line using, for example, polyethylene glycol (PEG), to form hybridoma cells that can then be selected away from unfused lymphocytes and myeloma cells.
  • PEG polyethylene glycol
  • Hybridomas that produce monoclonal antibodies directed specifically against a chosen antigen as determined by immunoprecipitation, immunoblotting, or by an in vitro binding assay can then be propagated either in in vitro culture using standard methods (Goding, Monoclonal Antibodies: Principles and Practice , Academic Press, 1986) or in vivo as ascites tumors in an animal.
  • the monoclonal antibodies can then be purified from the culture medium or ascites fluid.
  • FcgRIIA-binding molecules can also be made using recombinant DNA methods, for example, as described in U.S. Patent No. 4,816,567.
  • the polynucleotides encoding a monoclonal antibody are isolated from mature B-cells or hybridoma cell, such as by RT-PCR using oligonucleotide primers that specifically amplify the genes encoding the heavy and light chains of the antibody, and their sequence is determined using conventional procedures.
  • the isolated polynucleotides encoding the heavy and light chains or antigen-binding fragments thereof are then cloned into suitable expression vectors, which when transfected into host cells such as E.
  • FcgRIIA-binding molecules can be isolated from phage display libraries expressing CDRs of the desired species, as described by McCafferty et al. (Nature, 348:552-554 (1990)); Clackson et al. (Nature, 352:624-628 (1991)); and Marks et al. (J. Mol. Biol., 222:581-597 (1991)). Production and expression of nucleic acids comprising nucleotide sequences encoding FcgRIIA-binding molecules are discussed in more detail in the next section.
  • the polynucleotide(s) encoding a binding molecule can further be modified in a number of different manners using recombinant DNA technology to generate alternative binding molecules.
  • the constant domains of the light and heavy chains of, for example, a mouse monoclonal antibody can be substituted (1) for those regions of, for example, a human antibody to generate a chimeric antibody or (2) for a non-immunoglobulin polypeptide to generate a fusion antibody.
  • the constant regions are truncated or removed to generate the desired antibody fragment of a monoclonal antibody. Site-directed or high-density mutagenesis of the variable region can be used to optimize specificity, affinity, etc. of a monoclonal antibody.
  • the FcgRIIA-binding molecule is a human antibody or antigen binding fragment thereof.
  • Human antibodies can be directly prepared using various techniques known in the art. Immortalized human B lymphocytes immunized in vitro or isolated from an immunized individual that produce an antibody directed against a target antigen can be generated ⁇ See, e.g. , Cole el al. , Monoclonal Antibodies and Cancer Therapy , Alan R. Liss, p. 77 (1985); Boemer et al., J Immunol. 147 (l):86-95 (1991); and U.S. Patent No. 5,750,373).
  • the FcgRIIA-binding molecule can be selected from a phage library, where the phage library expresses human antibodies, as described, for example, by Vaughan et al. ⁇ Nat.
  • Affinity maturation strategies and chain shuffling strategies are known in the art and can be employed to generate high affinity human antibodies or antigen-binding fragments thereof. ⁇ See Marks et al., Bio/Technology 10:779-783 (1992)).
  • the FcgRIIA-binding molecule can be a humanized antibody or antigen-binding fragment thereof.
  • Methods for engineering, humanizing, or resurfacing non human or human antibodies can also be used and are well known in the art.
  • a humanized, resurfaced, or similarly engineered antibody can have one or more amino acid residues from a source that is non-human, e.g, mouse, rat, rabbit, non-human primate, or other mammal. These non-human amino acid residues are replaced by residues that are often referred to as“import” residues, which are typically taken from an“import” variable, constant, or other domain of a known human sequence.
  • CDR residues are directly and most substantially involved in influencing FcgRIIA binding. Accordingly, part or all of the non-human or human CDR sequences are maintained while the non-human sequences of the variable and constant regions can be replaced with human or other amino acids.
  • Antibodies can also optionally be humanized, resurfaced, engineered, or human antibodies engineered with retention of high affinity for FcgRIIA and other favorable biological properties.
  • humanized (or human) or engineered anti-FcgRIIA antibodies and resurfaced antibodies can be optionally prepared by a process of analyzing the parental sequences and various conceptual humanized and engineered products, using three-dimensional models of the parental, engineered, and humanized sequences. Three-dimensional
  • immunoglobulin models are commonly available and are familiar to those skilled in the art.
  • Computer programs are available which illustrate and display probable three-dimensional conformational structures of selected candidate immunoglobulin sequences. Inspection of these displays permits analysis of the likely role of the residues in the functioning of the candidate binding molecule sequence, i.e., the analysis of residues that influence the ability of the candidate binding molecule to bind its target, such as FcgRIIA
  • framework (FW) residues can be selected and combined from the consensus and import sequences so that the desired binding molecule characteristic, such as increased affinity for the target, is achieved.
  • Anti-FcgRIIA humanized antibodies and antigen-binding fragments thereof can also be made in transgenic mice containing human immunoglobulin loci that are capable, upon immunization, of producing the full repertoire of human antibodies in the absence of endogenous immunoglobulin production. This approach is described in U.S. Patent Nos. 5,545,807;
  • an anti-FcgRIIA antibody fragment is provided.
  • Various techniques are known for the production of antibody fragments. Traditionally, these fragments are derived via proteolytic digestion of intact antibodies. See , e.g. , Morimoto et al ., ./. Biochem. Biophys. Meth. 24: 107-117 (1993); Brennan et al ., Science , 229:81-83 (1985).
  • anti-FcgRIIA antibody fragments are produced recombinantly. Fab, Fv, and scFv antibody fragments can all be expressed in and secreted from E. coli or other host cells, thus allowing the production of large amounts of these fragments.
  • Anti-FcgRIIA antibody fragments can also be isolated from the antibody phage libraries discussed above.
  • Anti-FcgRIIA antibody fragments can also be linear antibodies, as described in U.S. Patent No. 5,641,870. Other techniques for the production of antibody fragments will be apparent to the skilled practitioner.
  • Antibody fragments can also be produced by techniques in the art including, but not limited to: (a) a F(ab’)2 fragment produced by pepsin digestion of an antibody molecule; (b) a Fab fragment generated by reducing the disulfide bridges of an F(ab’)2 fragment, (c) a Fab fragment generated by the treatment of the antibody molecule with papain and a reducing agent, and (d) Fv fragments.
  • FcgRIIA-binding molecule can be modified in order to reduce or eliminate effector function. This can be achieved, for example, by the triple mutation (TM) L234F/L235E/P331S in the Fc domain of IgGl. Other mutations that reduce effector function are known in the art. See, e.g., Armour et al, Eur. J. Immunol. 29:2613-2624, 1999; Shields et al, J. Biol. Chem. 276:6591-6604, 2001.
  • an FcgRIIA-binding molecule can be modified in order to increase its serum half-life. This can be achieved, for example, by incorporation of a salvage receptor binding epitope into the binding molecule by mutation of the appropriate region, or by incorporating the epitope into a peptide tag that is then fused to the binding molecule at either end or in the middle ( e.g ., by DNA or peptide synthesis), or by YTE mutation.
  • Other methods to increase the serum half-life of an antibody or antigen-binding fragment thereof, e.g., conjugation to a heterologous molecule such as PEG, are known in the art.
  • Heteroconjugate FcgRIIA antibodies and antigen-binding fragments thereof are also within the scope of the present invention.
  • Heteroconjugate antibodies are composed of two covalently joined antibodies. Such antibodies have, for example, been proposed to target immune cells to unwanted cells (see, e.g, ET.S. Pat. No. 4,676,980).
  • heteroconjugate anti-FcgRIIA antibodies and antigen-binding fragments thereof can be prepared in vitro using known methods in synthetic protein chemistry, including those involving crosslinking agents.
  • immunotoxins can be constructed using a disulfide exchange reaction or by forming a thioether bond. Examples of suitable reagents for this purpose include iminothiolate and methyl-4-mercaptobutyrimidate.
  • An FcgRIIA-binding molecule can be modified to contain additional chemical moieties not normally part of the protein. Such moieties can improve the characteristics of the binding molecule, for example, solubility, biological half-life, or absorption. The moieties can also reduce or eliminate any undesirable side effects of the binding molecule. An overview of those moieties can be found in Remington’s Pharmaceutical Sciences, 20th ed., Mack Publishing Co., Easton, PA (2000).
  • the disclosure provides a composition, e.g, a pharmaceutical composition, comprising an FcgRIIA-binding molecule of the invention, optionally further comprising one or more carriers, diluents, excipients, or other additives.
  • a composition e.g, a pharmaceutical composition, comprising an FcgRIIA-binding molecule of the invention, optionally further comprising one or more carriers, diluents, excipients, or other additives.
  • polynucleotides comprising nucleic acid sequences that encode an FcgRIIA-binding molecule, e.g, a polypeptide that specifically binds FcgRIIA
  • the invention provides a polynucleotide comprising a nucleic acid sequence that encodes an anti-FcgRIIA antibody or encodes an antigen-binding fragment of such an antibody.
  • the polynucleotides of the invention can be in the form of RNA or in the form of DNA.
  • DNA includes cDNA, genomic DNA, and synthetic DNA; and can be double-stranded or single- stranded, and, if single stranded, can be the coding strand or non-coding (anti-sense) strand.
  • the polynucleotide can be isolated. In certain embodiments, the polynucleotide can be substantially pure. In certain embodiments, the polynucleotide can be cDNA or are derived from cDNA. In certain embodiments, the polynucleotide can be recombinantly produced.
  • the polynucleotide can comprise the coding sequence for a mature polypeptide, fused in the same reading frame to a polynucleotide which aids, for example, in expression and optionally, secretion, of a polypeptide from a host cell (e.g ., a promoter or other regulatory sequence, a leader sequence that functions as a secretory sequence for controlling transport of a polypeptide from the cell).
  • a host cell e.g ., a promoter or other regulatory sequence, a leader sequence that functions as a secretory sequence for controlling transport of a polypeptide from the cell.
  • the polypeptide having a leader sequence is a pre-protein and can have the leader sequence cleaved by the host cell to form the mature form of the polypeptide.
  • the polynucleotide can also encode an FcgRIIA-binding pro- protein which is the mature protein plus additional 5’ amino acid residues.
  • the disclosure provides an isolated polynucleotide comprising a nucleic acid encoding an FcgRIIA-binding molecule comprising an amino acid sequence from a VH and/or VL domain having 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% similarity to an amino acid sequence set forth herein, and/or comprising 0, 1, 2, 3, 4, 5 or more amino acid substitutions, e.g.,
  • the polynucleotide that comprises the coding sequence for the FcgRIIA-binding molecule is fused in the same reading frame as a marker sequence that allows, for example, for purification of the encoded polypeptide.
  • the marker sequence can be a hexa-histidine tag supplied by a pQE-9 vector to provide for purification of the mature polypeptide fused to the marker in the case of a bacterial host, or the marker sequence can be a hemagglutinin (HA) tag derived from the influenza hemagglutinin protein when a mammalian host (e.g, COS-7 cells) is used.
  • a mammalian host e.g, COS-7 cells
  • Polynucleotide variants are also provided. Polynucleotide variants can contain alterations in the coding regions, non-coding regions, or both. In some embodiments, polynucleotide variants contain alterations that produce silent substitutions, additions, or deletions, but do not alter the properties or activities of the encoded polypeptide. In some embodiments, polynucleotide variants are produced by silent substitutions due to the degeneracy of the genetic code. Polynucleotide variants can be produced for a variety of reasons, e.g, to optimize codon expression for a particular host (change codons in the human mRNA to those preferred by a bacterial host such as E. coli). [00131] The invention includes vectors comprising the polynucleotides described above.
  • Suitable vectors are described elsewhere herein, and are known to those of ordinary skill in the art.
  • a polynucleotide comprising a nucleic acid encoding a VH domain or portion thereof and the polynucleotide comprising a nucleic acid encoding a VL domain or portion thereof can reside in a single vector, or can be on separate vectors. Accordingly, the disclosure provides one or more vectors comprising the polynucleotides described above.
  • the disclosure provides a composition, e.g ., a pharmaceutical composition, comprising a polynucleotide or vector as described above, optionally further comprising one or more carriers, diluents, excipients, or other additives.
  • a composition e.g ., a pharmaceutical composition, comprising a polynucleotide or vector as described above, optionally further comprising one or more carriers, diluents, excipients, or other additives.
  • the disclosure further provides a host cell comprising a polynucleotide or vector of the invention, wherein the host cell can, in some instances, express a binding molecule that specifically binds to FcyRIIA.
  • a host cell can be utilized in a method of making an
  • FcgRIIA-binding molecule where the method includes (a) culturing the host cell and (b) isolating the binding molecule from the host cell or from the culture medium, if the binding molecule is secreted by the host cell.
  • nucleotide sequence encoding an FcgRIIA-binding molecule can be constructed by chemical synthesis using an oligonucleotide synthesizer.
  • oligonucleotides can be designed based on the amino acid sequence of the desired polypeptide and selecting those codons that are favored in the host cell in which the recombinant polypeptide of interest will be produced. Standard methods can be applied to synthesize an isolated polynucleotide sequence encoding an isolated polypeptide of interest. For example, a complete amino acid sequence can be used to construct a back-translated gene. Further, a nucleotide oligomer containing a nucleotide sequence coding for the particular isolated polypeptide can be synthesized. For example, several small oligonucleotides coding for portions of the desired polypeptide can be synthesized and then ligated. The individual oligonucleotides typically contain 5’ or 3’ overhangs for complementary assembly.
  • the polynucleotide sequences encoding the binding molecule can be inserted into an expression vector and operatively linked to an expression control sequence appropriate for expression of the binding molecule in a desired host. Proper assembly can be confirmed, e.g. , by nucleotide sequencing, restriction mapping, and/or expression of a biologically active polypeptide in a suitable host. In order to obtain high expression levels of a transfected gene in a host, the gene can be operatively linked to or associated with transcriptional and translational expression control sequences that are functional in the chosen expression host.
  • recombinant expression vectors are used to amplify and express DNA encoding FcgRIIA-binding molecules.
  • Recombinant expression vectors are replicable DNA constructs that have synthetic or cDNA-derived DNA fragments encoding a polypeptide chain of an FcgRIIA-binding molecule, operatively linked to suitable transcriptional or translational regulatory elements derived from mammalian, microbial, viral or insect genes.
  • a transcriptional unit generally comprises an assembly of (1) a genetic element or elements having a regulatory role in gene expression, for example, transcriptional promoters or enhancers, (2) a structural or coding sequence which is transcribed into mRNA and translated into protein, and (3) appropriate transcription and translation initiation and termination sequences, as described in detail below.
  • a genetic element or elements having a regulatory role in gene expression for example, transcriptional promoters or enhancers
  • a structural or coding sequence which is transcribed into mRNA and translated into protein
  • appropriate transcription and translation initiation and termination sequences as described in detail below.
  • Such regulatory elements can include an operator sequence to control
  • DNA regions are operatively linked when they are functionally related to each other.
  • DNA for a signal peptide secretory leader
  • a promoter is operatively linked to a coding sequence if it controls the transcription of the sequence
  • a ribosome binding site is operatively linked to a coding sequence if it is positioned so as to permit translation.
  • Structural elements intended for use in yeast expression systems include a leader sequence enabling extracellular secretion of translated protein by a host cell.
  • the protein can include an N-terminal methionine residue. This residue can optionally be subsequently cleaved from the expressed recombinant protein to provide a final product.
  • Useful expression vectors for eukaryotic hosts include, for example, vectors comprising expression control sequences from SV40, bovine papilloma virus, adenovirus, and cytomegalovirus.
  • Useful expression vectors for bacterial hosts include known bacterial plasmids, such as plasmids from E. coli , including pCR 1, pBR322, pMB9 and their derivatives, wider host range plasmids, such as Ml 3, and filamentous single-stranded DNA phages.
  • Suitable host cells for expression of an FcgRIIA-binding molecule include prokaryotes, yeast, insect, or higher eukaryotic cells under the control of appropriate promoters.
  • Prokaryotes include gram negative or gram positive organisms, for example E. coli or bacilli.
  • Higher eukaryotic cells include established cell lines of mammalian origin as described below. Cell-free translation systems could also be employed. Additional information regarding methods of protein production, including antibody production, can be found in, e.g ., U.S. Patent Publication No. 2008/0187954, U.S. Patent Nos. 6,413,746 and 6,660,501, and International Patent
  • Suitable mammalian host cell lines include HEK-293 and HEK-293T, the COS-7 lines of monkey kidney cells, described by Gluzman ( Cell 23: 175, (1981)), and other cell lines including, for example, L cells, C127, 3T3, Chinese hamster ovary (CHO), HeLa, and BHK cell lines.
  • Mammalian expression vectors can comprise non-transcribed elements, such as an origin of replication, a suitable promoter and enhancer linked to the gene to be expressed, and other 5’ or 3’ flanking non-transcribed sequences, and 5’ or 3’ non-translated sequences, such as necessary ribosome binding sites, a polyadenylation site, splice donor and acceptor sites, and transcriptional termination sequences.
  • non-transcribed elements such as an origin of replication, a suitable promoter and enhancer linked to the gene to be expressed, and other 5’ or 3’ flanking non-transcribed sequences, and 5’ or 3’ non-translated sequences, such as necessary ribosome binding sites, a polyadenylation site, splice donor and acceptor sites, and transcriptional termination sequences.
  • Baculovirus systems for production of heterologous proteins in insect cells are reviewed by Luckow and Summers ( BioTechnology 6:47 (1988)).
  • FcgRIIA-binding molecules produced by a transformed host can be purified according to any suitable method.
  • standard methods include chromatography (e.g, ion exchange, affinity, and sizing column chromatography), centrifugation, differential solubility, or by any other standard technique for protein purification.
  • Affinity tags such as hexahistidine, maltose binding domain, influenza coat sequence, and glutathione-S-transferase can be attached to the protein to allow easy purification by passage over an appropriate affinity column.
  • Isolated proteins can also be physically characterized using such techniques as proteolysis, nuclear magnetic resonance and x-ray crystallography.
  • supernatants from systems that secrete recombinant protein into culture media can be first concentrated using a commercially available protein concentration filter, for example, an Amicon or Millipore Pellicon ultrafiltration unit. Following the concentration step, the concentrate can be applied to a suitable purification matrix.
  • a suitable purification matrix for example, an anion exchange resin can be employed, for example, a matrix or substrate having pendant diethylaminoethyl (DEAE) groups.
  • the matrices can be acrylamide, agarose, dextran, cellulose, or other types commonly employed in protein purification.
  • a cation exchange step can be employed. Suitable cation exchangers include various insoluble matrices comprising sulfopropyl or carboxymethyl groups.
  • RP-HPLC reversed-phase high performance liquid chromatography
  • a recombinant FcgRIIA-binding molecule produced in bacterial culture can be isolated, for example, by initial extraction from cell pellets, followed by one or more concentration, salting-out, aqueous ion exchange, or size exclusion chromatography steps. High performance liquid chromatography (HPLC) can be employed for final purification steps.
  • Microbial cells employed in expression of a recombinant protein can be disrupted by any convenient method, including freeze-thaw cycling, sonication, mechanical disruption, or use of cell lysing agents.
  • Methods known in the art for purifying antibodies and other proteins also include, for example, those described in ET.S. Patent Publication Nos. 2008/0312425, 2008/0177048, and 2009/0187005.
  • Methods are provided for the use of FcgRIIA binding molecules to treat patients having a disease or disorder associated with inappropriate FcgRIIA activation, such as disease or disorders characterized by immune-complex deposition, immune-complex mediated NETosis, ANCA-induced FcgRIIA activation, and anti-platelet antibody-triggered FcgRIIA activation.
  • a disease or disorder associated with inappropriate FcgRIIA activation such as disease or disorders characterized by immune-complex deposition, immune-complex mediated NETosis, ANCA-induced FcgRIIA activation, and anti-platelet antibody-triggered FcgRIIA activation.
  • treatment or prevention includes the application or administration of an FcgRIIA binding molecule or a composition comprising FcgRIIA binding molecule to a subject or patient, or application or administration of the FcgRIIA binding molecule to an isolated tissue or cell line from a subject or patient, where the subject or patient has a disease, a symptom of a disease, or a predisposition toward a disease.
  • the composition is preferably a pharmaceutical composition.
  • FcgRIIA binding molecules are useful for the treatment or prevention of certain inflammatory, immune-mediated, or autoimmune diseases or disorders.
  • inflammatory, immune-mediated, or autoimmune disease or disorders include, but are not limited to vasculitis, e.g ., Anti-neutrophil cytoplasm antibodies (ANCA), ANCA-associated vasculitis (AAV) or giant cell arteritis (GCA) vasculitis, Sjogren’s syndrome, inflammatory bowel disease (IBD), pemphigus vulgaris, lupus nephritis, psoriasis, thyroiditis, Type I Diabetes, immune thrombocytopenia (FTP), ankylosing spondylitis, multiple sclerosis, systemic lupus
  • vasculitis e.g ., Anti-neutrophil cytoplasm antibodies (ANCA), ANCA-associated vasculitis (AAV) or giant cell arteritis (GCA) vasculitis, Sjogren’s syndrome,
  • erythematosus SLE
  • rheumatoid arthritis Crohn’s disease
  • Myasthenia Gravis neuromyelitis optica
  • NMO neuromyelitis optica
  • IDDM insulin-dependent diabetes mellitus
  • akylosing spondylitis atopic dermatitis
  • uveitis uveitis
  • Graft-versus-host disease GVHD
  • polymyositis dermatomyositis, membranous nephropathy, hemolytic anemia, mixed connective tissue disease, sclerodema, sepsis, thrombosis, acute kidney injury, acute lung injury, chronic obstructive pulmonary disease, glomerulonephritis, toxic liver injury, stroke, atherogenesis, IgG mediated hypersensitive reaction, anti-drug immune complex mediated adverse effects, and other autoantibody or immune-complex-mediated disorders.
  • Clinical response to administration of an FcgRIIA-binding molecule can be assessed using screening techniques such as magnetic resonance imaging (MRI), x-radiographic imaging, computed tomographic (CT) scan, flow cytometry or fluorescence-activated cell sorter (FACS) analysis, histology, gross pathology, and blood chemistry, including but not limited to changes detectable by ELISA, ELISPOT, RIA, chromatography, and the like. Further, the subject undergoing therapy with the FcgRIIA-binding molecule can experience improvement in the symptoms associated with the disease or disorder.
  • screening techniques such as magnetic resonance imaging (MRI), x-radiographic imaging, computed tomographic (CT) scan, flow cytometry or fluorescence-activated cell sorter (FACS) analysis, histology, gross pathology, and blood chemistry, including but not limited to changes detectable by ELISA, ELISPOT, RIA, chromatography, and the like.
  • Methods of preparing and administering FcgRIIA-binding molecules to a subject in need thereof are well-known to or can be readily determined by those skilled in the art.
  • the route of administration of the FcgRIIA binding molecule can be, for example, oral, parenteral, by inhalation, or topical.
  • parenteral as used herein includes, e.g, intravenous, intraarterial, intraperitoneal, intramuscular, subcutaneous, rectal, and vaginal administration.
  • Oral dosage forms include, e.g. , capsules, tablets, aqueous suspensions, and solutions.
  • Nasal aerosol or inhalation dosage forms can be prepared, for example, as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, and/or other conventional solubilizing or dispersing agents.
  • a suitable pharmaceutical composition can comprise a buffer, optionally a surfactant, optionally a stabilizer agent, etc.
  • the form and character of the pharmaceutically acceptable carrier or diluent can be dictated by the amount of active ingredient with which it is to be combined, the route of administration and other well-known variables.
  • a cocktail comprising one or more species of FcyRIIA binding molecules, e.g. , anti-FcgRIIA antibodies, or antigen binding fragments, variants, or derivatives thereof, of the invention can also be used.
  • FcgRIIA-binding molecules can be delivered directly to the site of the adverse cellular population, thereby increasing the exposure of the diseased tissue to the therapeutic agent.
  • the administration is directly to the airway, e.g. , by inhalation or intranasal administration.
  • FcgRIIA binding molecules can be administered in a
  • F cy R 11 A -m edi ated diseases such as inflammatory, immune-mediated, or autoimmune diseases or disorders.
  • F cy R 11 A -m edi ated diseases such as inflammatory, immune-mediated, or autoimmune diseases or disorders.
  • the disclosed binding molecules can be formulated so as to facilitate
  • compositions in accordance with the present invention can comprise a pharmaceutically acceptable, non-toxic, sterile carrier such as physiological saline, non-toxic buffers, preservatives and the like.
  • a“therapeutically effective amount” of an FcgRIIA-binding molecule means an amount sufficient to achieve a benefit, e.g. , to ameliorate symptoms of a disease or condition or to detect a substance or a cell.
  • Suitable formulations for use in the therapeutic methods disclosed herein are described in Remington’s Pharmaceutical Sciences , 20th ed., Mack Publishing Co., Easton, PA (2000).
  • the composition can be administered as a single dose, multiple doses, or over an established period of time in an infusion. Dosage regimens also can be adjusted to provide the optimum desired response (e.g, a therapeutic or prophylactic response).
  • the amount of an FcgRIIA-binding molecule that can be combined with carrier materials to produce a dosage form will vary depending upon many different factors, including means of administration, target site, physiological state of the patient (z.e., the severity of the disease, the history of the disease, and the age, height, weight, health, and physical condition of the individual undergoing therapy), whether treatment is prophylactic or therapeutic, other medications administered, and whether the patient is human or an animal.
  • the patient is a human, but non-human mammals, including transgenic mammals can also be treated.
  • the amount of FcgRIIA-binding molecule to be administered is can be determined by one of ordinary skill in the art. Treatment dosages can be titrated using routine methods known to those of skill in the art to optimize safety and efficacy.
  • This disclosure also provides for the use of an FcgRIIA-binding molecule as described herein to treat or prevent an inflammatory, immune-mediated, or autoimmune disease or disorder, e.g ., vasculitis, such as ANCA or GCA vasculitis, immune thrombocytopenia, systemic lupus erythematosus, lupus nephritis, Sjogren’s syndrome, rheumatoid arthritis, Crohn’s disease, Myasthenia Gravis, GVHD, ADA-mediated adverse effects, NETosis, and NETosis-associated disorders, including sepsis, thrombosis, acute kidney injury, acute lung injury, chronic obstructive pulmonary disease, glomerulonephritis, toxic liver injury, stroke, atherogenesis, Type I diabetes, and IgG mediated hypersensitive reaction.
  • vasculitis such as ANCA or GCA vasculitis
  • immune thrombocytopenia systemic
  • This disclosure also provides for the use of an FcgRIIA-binding molecule as described herein in the manufacture of a medicament for treating or preventing an inflammatory, immune- mediated, or autoimmune disease or disorder, e.g. , vasculitis, such as ANCA or GCA vasculitis, immune thrombocytopenia, systemic lupus erythematosus, lupus nephritis, Sjogren’s syndrome, rheumatoid arthritis, Crohn’s disease, Myasthenia Gravis, GVHD, ADA-mediated adverse effects, NETosis, and NETosis-associated disorders, including sepsis, thrombosis, acute kidney injury, acute lung injury, chronic obstructive pulmonary disease, glomerulonephritis, toxic liver injury, stroke, atherogenesis, Type I diabetes, and IgG mediated hypersensitive reaction.
  • vasculitis such as ANCA or GCA vasculitis
  • FcgRIIA-binding molecules of the invention can be used for diagnosis of FcgRIIA- mediated diseases such as certain inflammatory, immune-mediated, or autoimmune diseases or disorders, and/or for diagnostic monitoring of protein levels as part of a clinical testing procedure, e.g, to determine the efficacy of a given treatment regimen.
  • Such methods typically involve using the FcgRIIA-binding molecules described herein to assay the expression level FcgRIIA.
  • test the expression level of FcgRIIA is intended to mean qualitatively or quantitatively measuring or estimating the level of FcgRIIA in a first biological sample either directly (e.g, by determining or estimating absolute protein level) or relatively (e.g, by comparing to the disease associated polypeptide level in a second biological sample).
  • the FcgRIIA expression level in the first biological sample can be measured or estimated and compared to a standard FcgRIIA level, the standard being taken from a second biological sample obtained from an individual not having the disorder, or being determined by averaging levels from a population of individuals not having the disorder.
  • an increase in the protein level of the test sample compared to the standard sample is indicative of a disease or disorder treatable by an FcgRIIA-binding molecule of the invention.
  • an FcgRIIA-binding molecule of the invention As will be appreciated in the art, once the“standard” FcgRIIA level is known, it can be used repeatedly as a standard for comparison.
  • biological sample any biological sample obtained from an individual, cell line, tissue culture, or other source of cells potentially expressing FcgRIIA. Methods for obtaining tissue biopsies and body fluids from mammals are known in the art.
  • the FcgRIIA-binding molecules of the invention can be used to assay FcyRIIA protein levels in a biological sample using classical immunohistological methods known to those of skill in the art (e.g., see Jalkanen, el al., J. Cell. Biol. 707:976-985 (1985); Jalkanen el al., J. Cell Biol.
  • Immunoassays that can be used include but are not limited to competitive and non-competitive assay systems using techniques such as Western blots, radioimmunoassays, ELISA (enzyme linked immunosorbent assay), ELISPOT,“sandwich” immunoassays, immunoprecipitation assays, precipitin reactions, gel diffusion precipitin reactions, immunodiffusion assays, agglutination assays, complement-fixation assays, immunoradiometric assays, fluorescent immunoassays, protein A immunoassays, and immunoelectron microscopy, to name some examples.
  • Such assays are routine and well known in the art. Those skilled in the art will be able to determine operative and optimal assay conditions for each determination by employing routine experimentation.
  • Detection of FcgRIIA can be facilitated by coupling the binding molecule to a detectable substance or label.
  • detectable substances include various enzymes, prosthetic groups, fluorescent materials, luminescent materials, bioluminescent materials, and radioactive materials.
  • suitable enzymes include horseradish peroxidase, alkaline phosphatase, b-galactosidase, or acetylcholinesterase;
  • suitable prosthetic group complexes include streptavidin/biotin and avidin/biotin;
  • suitable fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine,
  • An example of a luminescent material is luminol.
  • bioluminescent materials include luciferase, luciferin, and aequorin.
  • suitable radioactive material include 125 I, 131 1, 35 S, or 3 H.
  • In situ detection can be accomplished by removing a histological specimen, for example a blood sample, from a patient, and applying thereto a labeled FcgRIIA-binding molecule, applied by overlaying the labeled FcgRIIA-binding molecule onto a biological sample.
  • a histological specimen for example a blood sample
  • FcgRIIA-binding molecule applied by overlaying the labeled FcgRIIA-binding molecule onto a biological sample.
  • FcgRIIA but also its distribution in the examined tissue. Rising the present invention, those of ordinary skill will readily perceive that any of a wide variety of histological methods (such as staining procedures) can be modified in order to achieve such in situ detection.
  • Kits comprising FcgRIIA-binding Molecules
  • kits that comprise an FcgRIIA-binding molecule, which can be used to perform the methods described herein.
  • a kit comprises at least one purified FcgRIIA-binding molecule in one or more containers.
  • the kits contain all of the components necessary and/or sufficient to perform a detection assay, including all controls, directions for performing assays, and any necessary software for analysis and presentation of results.
  • the disclosed FcgRIIA-binding molecules can be readily incorporated into one of the established kit formats which are well known in the art. [00161] All of the references cited in this disclosure are hereby incorporated by reference in their entireties. In addition, any manufacturers’ instructions or catalogues for any products cited or mentioned herein are incorporated by reference. Documents incorporated by reference into this text, or any teachings therein, can be used in the practice of the present invention.
  • Embodiments of the present disclosure can be further defined by reference to the following non-limiting examples, which describe in detail preparation of certain antibodies of the present disclosure and methods for using antibodies of the present disclosure. It will be apparent to those skilled in the art that many modifications, both to materials and methods, can be practiced without departing from the scope of the present disclosure.
  • cDNA molecules encoding the extracellular domains of CD32A, CD32B from human were synthesized with primer extension PCR cloning using database sequences as references (see Table 2). Sequences were cloned into the pEBNA mammalian expression vector (Thermo Fisher, Cat. No. A10898) fused to a 6xHis_Flag tag at the C-terminus (HHHHHHDYKDDDDK) (SEQ ID NO: 18). Proteins were expressed in FreeStyle 293F cells (Thermo Fisher Cat. No. R790-07). Expressed protein in supernatant was purified using Ni-NTA affinity chromatography (Histrap HP column (GE Healthcare, Cat. No. 17-5248-02)) followed by Size Exclusion chromatography (Superdex 200 column (GE Healthcare, Cat. No. 17-1069-01)).
  • CD32A Three versions of cynomologus CD32A were made based on polymorphisms around the epitope of IV3 (Ramsland et. al. , ./. Immunol. 187:3208-3217 (2011)). These proteins were expressed in HEK cells by GeneArt and purified by Ni-NTA affinity chromatography utilizing a c-terminal 6xhis tag.
  • IgGs and modified proteins used herein were biotinylated via free amines using EZ link Sulfo-NHS-LC -Biotin (Thermo/Pierce, Cat. No. 21335).
  • the biotin reagent was dissolved in anhydrous dimethylformamide and PBS based protein solutions were adjusted to pH ⁇ 8 with 1 M NaHCCb in D-PBS (Dulbecco’s phosphate buffered saline).
  • CD32 proteins used herein were biotinylated via free cysteines using EZ link Biotin-BMCC (Perbio/Pierce, Product No. 21900).
  • the biotin reagent was dissolved in anhydrous dimethylformamide and mixed 3 : 1 with D-PBS protein solutions. Label incorporations were assessed by MALDI-TOF mass
  • concentrations were determined by 280 nm absorbance using extinction coefficients calculated from amino acid sequences.
  • Variable domains were converted to whole immunoglobulin Gl (IgGi) antibody format essentially as described by Persic et al. ⁇ Gene 187(1):9-18 (1997)) with the following
  • VH variable heavy domain
  • the heavy chain constant domains contained three mutations shown to significantly reduce Fc effector function (Oganesyan et al ., Acta Crystallographica Section D: Biological Crystallography 64(6): 700 (2008)), to avoid engagement of CD32A via the Fc portion of full-length antibody.
  • variable light (VL) domain was cloned into a vector for the expression of the human light chain (kappa) constant domains and regulatory elements to express whole IgG light chain in mammalian cells.
  • the heavy and light chain IgG-expressing vectors were transfected into CHO- transient mammalian cells (Daramola et al. , Biotechnol Prog. 30(1): 132-41 (2014)). IgGs were expressed and secreted into the medium. Harvests were filtered prior to purification, then IgG was purified using Protein A chromatography.
  • IgGi immunoglobulin Gl antibodies
  • Antibodies were characterized in an epitope competition assay as described in Example 11. Briefly, homogenous time-resolved fluorescence (HTRF®, Cisbio International) was used to detect binding of IV.3 IgG (Stemcell Technologies, Cat. No. 01470) to biotinylated human CD32A 131H. Fluorescence resonance energy transfer (FRET) between IV.3 and CD32A was detected using DyLight 649 conjugated anti-mouse antibody and streptavidin Terbium cryptate (Cisbio International, Product No. 610SATLB), respectively.
  • HTRF® homogenous time-resolved fluorescence
  • FRET Fluorescence resonance energy transfer
  • amino acid sequences of the VH and VL domains of the clones 32L00350, 32LO0351, 32LO0352, 32LO0354, 32LO0355, and 32LO0356 are shown in Table 7.
  • ADCC and CDC assays were performed.
  • the effects of MED 19600 were compared with wild type control 9600 IgGl (identical Fab as MED 19600, but with wild-type IgG Fc) and isotype control IgG (R347-Tm) antibodies.
  • ADCC assay primary NK cells (effectors) were incubated with adherent FcgRIIA-expressing HEK-293 cells (targets) for 5 h, and % cytotoxicity was determined.
  • baby rabbit complement was incubated with adherent
  • FcgRIIA-expressing HEK-293 cells targets and % cytotoxicity was determined after 1 h. No notable ADCC (FIG. 5F) or CDC (FIG. 5G) activity was detected with MEDI9600, whereas the 9600 IgGl control antibody induced cytotoxicity of FcgRIIA-expressing HEK cells. Therefore the TM incorporated in MED 19600’s heavy chain constant region renders MEDI9600 effector null.
  • IVIG immunoglobulin
  • MED 19600 bound to human FcgRIIA-expressing neutrophils in a dose-dependent manner.
  • the EC50 value of MEDI9600 binding to human FcgRIIA at the neutrophil surface was 0.03 nM
  • FIG. 6 In the presence of 10 mg/mL IVIG, the EC50 value of MED 19600 binding to human FcgRIIA at the neutrophil surface was increased to 3.34 nM (FIG. 6), suggesting that MEDI9600 competes with immunoglobulin for binding to FcgRIIA. No binding was observed with the human IgGl isotype R347-TM control (FIG. 6). These data indicate that MEDI9600 is a ligand- blocking antibody.
  • the murine IV.3 Ab has been shown to internalize FcgRIIA and subsequently degrade the receptor in the lysosome 13 . Consequently, removal of FcgRIIA from the surface of the cell upon antibody binding can be used as a measure of receptor internalization.
  • This mode of action was assessed the potency of the anti-FcgRIIA antibody variants in a whole blood internalization assay. This assay format was used to mimic the cellular composition and the levels of competing immunoglobulin found under physiological conditions. It has previously been shown that two common human polymorphic variants of FcgRIIA, 131H and 131R, differentially bind IgG subclasses 14 .
  • the ECso for the optimized antibodies ranged from 0.04 nM to 0.09 nM compared to 0.19 nM for the chimeric IV.3 antibody for the 131H/H donor, and 0.04 nM to 0.06 nM compared to 0.20 nM for the chimeric IV.3 antibody for the 131 R/R donors (FIG. 7A, 7B).
  • the optimized antibodies also internalized FcgRIIA on the surface of monocytes from cynomolgous monkeys (FIG. 7C). The potency of the optimized antibodies was up to seven-fold greater than the chimeric IV.3 Ab.
  • MED 19600 mediated internalization of FcgRIIA from the surface of neutrophils from human donors having the 131H/H (Fig. 7E) or 131R/R genotype (Fig. 7F), and from the suface of neutrophils from cynomolgus monkey whole blood (Fig.7G).
  • the neutrophils from human (131R/R or 131H/H phenotypes) and cyomolgus monkey whole blood were examined following a 12 hour incubation with MED 19600 or control Ab (R347-TM) by flow cytometry (M.F.I.). Together these data demonstrate that MED 19600 reduces monocyte and neutrophil cell surface expression of FcgRIIA available for ligand engagement.
  • the potency of the optimized antibodies were ⁇ 3-8 fold greater than the humanized CamIV.3 antibody with the 131 H/H donor (FIG. 8A), and ⁇ 5-12 fold greater than CamIV.3 for the 131 R/R donor (FIG. 8B). Three of the four optimized antibodies also exhibited greater potency than the parental and humanized versions of IV.3 in the RNP-IC-induced IFNa assay using cynomolgus monkey cells (FIG. 8C).
  • immune complexes can also trigger monocytes to produce TNFa and IL- 6, key cytokines involved in the pathogenesis of RA [21] Compared to control antibody,
  • MED 19600 (30 pg/mL) inhibited the immune complex induction of IL-6 and TNFa in whole blood by approximately 60% and 80% respectively (Fig. 8D; mean ⁇ standard deviation percentage inhibition relative to no antibody presented).
  • Fig. 8D mean ⁇ standard deviation percentage inhibition relative to no antibody presented.
  • MEDI9600 can inhibit IC-mediated induction of TNFa and IL-6 among other proinflammatory molecules would suggest that MED 19600 may provide an alternative option for the treatment of RA.
  • Treatments targeting the TNFa or IL-6 pathways have been approved for RA highlighting the importance of these cytokines in the pathology of the disease.
  • a marker of neutrophil activation is the release of reactive oxygen species, which can measured in a ferri-cytochrome C reduction assay, or by flow cytometry based DH123 assay 15 .
  • the ability of optimized IV.3 Ab clone 32LO0352 (MEDI9600) to inhibit ANCA induced neutrophil activation was first examined in ferri-cytochrome C reduction assay.
  • MED 19600 specially blocked ANCA- (both Anti-PR3 Ab and anti-MPO Ab) induced neutrophil production of reactive oxygen species (FIG. 9A, 9B).
  • neutrophils were treated with PMA in the presence and absence of the FcgRIIA antibody.
  • IgG was purified from AAV patients’ sera with autoantibodies against either PR3 or MPO, and used as stimuli for neutrophils.
  • MED 19600 significantly blocked neutrophil activation triggered by AAV patient IgG (FIG. 9F, 9G).
  • MEDI9600 anti-FcgRIIA Ab is able to specifically block anti-neutrophil cytoplasmic antibody (ANCA) induced neutrophil activation, and supports the use of the humanized, optimized MEDI9600 anti-FcgRIIA antibody for the treatment of ANCA-associated vasculitis.
  • ANCA anti-neutrophil cytoplasmic antibody
  • FIG. 10G A summary of MEDI9600 inhibition of anti -platelet (anti-CD4l Ab) induced thrombocytopenia in FcgRIIA transgenic mice is provided at FIG. 10G. Bars in the graph show platelet numbers in the transgenic mice prophylactically treated with MEDI9600 or control antibody prior to induction of thrombocytopenia.
  • the MED 19600 or control antibody R347- Tm
  • thrombocytopenia was induced with 2 pg rat anti-mouse CD4lAb delivered i.p. at day 0.
  • Preincubation with 10 mg/kg of MED 19600 prevented anti-platelet mediated thrombocytopenia compared to the control antibody where there was -80% reduction in platelet numbers (FIG. 10G).
  • Neutrophils play a critical role in host defense by sensing infection and tissue injury, and initiating an acute inflammatory response, which serves to recruit leukocytes, clear infections, engage the adaptive immune system, and promote repair.
  • neutrophils Upon exposure to an insult, neutrophils rapidly migrate through the blood vessels to the site of the tissue injury, following chemical signals derived from pathogens and damaged host cells.
  • Neutrophils directly engage pathogens through interactions with pattern recognition receptors, complement receptors, and immunoglobulin receptors, which results in the release of toxic substances that kill pathogens and/or the clearance of the pathogens by phagocytosis. Since neutrophil functions are important in host defense, we assessed the impact of FcgRIIA blockade with MED 19600 on different neutrophil functions.
  • MEDI9600 affects anti-Psl mAb-mediated opsonophagocytic killing (OPK) of Pseudomonas aeruginosa.
  • OPK opsonophagocytic killing
  • Anti-Psl mAbs were previously shown to mediate potent complement dependent killing of P. aeruginosa in the presence of neutrophils 16 .
  • a luminescent/ 1 aeruginosa strain was used in this assay to assess bacterial killing. The level of luminescence correlates with the frequency of live bacteria. Neutrophils were pre-incubated with MED 19600, anti-FcyRIIB Ab, or anti-FcyRIII Ab, and then they were incubated with luminescent bacteria for 2 hours.
  • MEDI9600 had a minimal effect on the ability of neutrophils to kill P. aeruginosa in the presence of anti-Psl mAb Psl0096, whereas blockade of FcyRIII clearly inhibited OPK activity (FIG. 11C).
  • MEDI9600 A Single-Dose Pharmacokinetic and Exploratory Pharmacodynamic Study in Cynomolgus Monkeys
  • a single dose of MED 19600 administered at 1, 10, or 100 mg/kg induced a dose- response reduction of FcgRIIA fluorescence intensity on monocytes and granulocytes (FIG.
  • MED 19600 resulted in dose-dependent suppression of FcgRIIA expression on monocytes and granulocytes, followed by a slow recovery to baseline levels consistent with the duration of the MED 19600 pharmacokinetic exposure, indicating a strong relationship between pharmacokinetic exposure and pharmacodynamic response.
  • Primers were designed using the available cynomolgus monkey draft genome sequence derived from a single animal of Vietnamese origin (Beijing Genome Institute, 2011).
  • the Protein Science cynomolgus genomic DNA bank comprised of 60 individuals (20 Chinese, 20 Vietnamese, 20 Mauritian), was utilized to sequence FcylLA PCR amplification was performed using Qiagen HotStar Taq Master Mix followed by in-house sequencing of the PCR products.
  • Raw sequence alignments, consensus transcript builds, SNP variant identification, and comparison to human FcyllA (CCDS 44264 / ETniProt P12318) were completed using SeqMan (Lasergene) and CloneManager software.
  • FcyRI Recombinant FcyRI, FcgRIIA- 13 1 H/H allotype, FcyRI IB, FcyRIII - 158F allotype, and FcyRIII - 158V allotype, were generated at Medlmmune.
  • Anti-FcyRIII antibody (3G8) was purchased from Abeam (Cambridge, MA).
  • Anti- FcyRI Ab (16-115) was purchased from Antibodies-online (Atlanta, GA).
  • Anti- FcyRI IB Ab and isotype control R347-Tm were generated at Medlmmune.
  • the color reaction was stopped with 50m1 0.2M FhsSCL and the absorbance was measured using a spectrophotometer (Molecular Devices, CA) at A 405nm.
  • the assay was used to identify improvements in crude IgG samples by measuring inhibition of the interaction between IV.3 IgG and biotinylated human CD32a 131H.
  • a pre mixed solution containing 4 nM biotinylated human CD32a 131H and 2.67 nM streptavidin Terbium cryptate is prepared as a 4x stock. 5m1 is added to the assay plate to give a final concentration of 1 nM biotinylated human CD32a 131H and 0.67 nM streptavidin Terbium cryptate. Non-specific binding wells (negative controls) were defined for each plate by omitting biotinylated human CD32a 131H. Assay plates were incubated for 3 hours at room temperature prior to reading time resolved fluorescence at 620 nm and 665 nm emission wavelengths using an EnVision plate reader (Perkin Elmer).
  • IC50 values were determined using GraphPad Prism software by curve fitting using a four-parameter logistic equation (Equation 4).
  • X is the logarithm of concentration.
  • Y is specific binding
  • Y starts at Bottom and goes to Top with a sigmoid shape.
  • the selectivity of lead antibodies was assessed using a homogenous time-resolved fluorescence assay (HTRF®, Cisbio International) in which the binding of the IgG to biotinylated human CD32b was measured.
  • Cross-reactivity was assessed using a homogenous time-resolved fluorescence assay in which the binding of the IgG to cyno CD32a and CD32b was measured.
  • Assay buffer consisting of PBS (Invitrogen 14190-185), 0.2 % BSA (Sigma A9576) and 0.4 M KF (BDH103444T) is prepared and 5m1 added to the assay plate (384 black, shallow well, Costar, 3676).
  • CD32 is diluted as a 4x working stock solution in assay buffer.
  • Human CD32b, cyno CD32a and cyno CD32b are prepared at 20 nM and 5m1 added to the assay plate to give a final concentration of 5 nM.
  • Serial dilutions of IgGs are prepared in duplicate in assay buffer using a Greiner 384 well V bottom plate (Greiner 781280) and 5m1 of each dilution of peptide transferred to the assay plate using a BravoTM (Agilent). Streptavidin terbium cryptate and anti-human Fc IgG XL665 were diluted to 0.67 nM and 15 nM respectively (final concentrations) in assay buffer and 5m1 of this premixed solution was added to the assay plate. A negative control was defined for each plate by replacing sample IgG with 5m1 assay buffer.
  • IgG samples were quantified using Protein A Biosensors (ForteBio) in conjunction with the OctetRED384 system (ForteBio).
  • 25m1 of sample is mixed with 25m1 assay buffer consisting of PBS, 0.02 % Tween20, 1 mg/ml BSA (0.1%).
  • the rate of binding of IgG in the sample to the protein A biosensor is measured by Bio-Layer Interferometry (BLI).
  • An IgG standard curve is generated using a control IgG of known concentration and preparing a 12-point 1 in 2 dilution series from a top final concentration of 500 pg/ml.
  • the Octet system software calculates the binding rates from standards with known concentrations to generate a standard curve. The binding rate is proportional to the standard concentration.
  • Data is analysed using Data Analysis software package provided with the OctetRED384 system. Data is uploaded into the software and analysis performed using initial slope and the Dose Response - 4PL (Default; Unweighted) equations. Unknown concentrations are calculated from comparison with the standard curve.
  • MFI mean fluorescence intensity
  • PBMCs peripheral blood mononuclear cells
  • the PBMCs were first stained with CDl4-Alexa 488, and CDl4-positive monocytes were sorted. The cells were then stained with MED 19600- Alexa 647 on ice for 30 min, washed once and incubated at room temperature for confocal microscopy.
  • Cell imaging was performed using a Leica TCS SP5 confocal system consisting of a Leica DMI6000 B inverted microscope (Leica Microsystems). Images were acquired at time- points noted in the figure legends and were analyzed using the LAS AF version 2.2.1 Leica Application Suite software (Leica Microsystems).
  • Anti-FcgRIIA-Tm Ab-mediated receptor internalization was examined by a FACS- based assay. Human or cynomolgus monkey blood was collected into heparin tubes. A three fold dilution series of unlabeled anti-FcgRIIA antibody or control antibody was added to 50 m ⁇ whole blood for 2 hours at 37°C. The whole blood was washed with FACS buffer (5% fetal bovine serum in PBS), and stained with Ab cocktail (CD14-PE, CD20-Pacific Blue, and IV.3- Alexa 647) on ice for 1 hour.
  • FACS buffer 5% fetal bovine serum in PBS
  • the cells were incubated with 1 ml RBC lysis buffer (Biolegend, CA) at room temperature for 3 minutes, washed and re-suspended in 150 m ⁇ FACS buffer.
  • FcgRIIA surface expression on human or cynomolgus monkey monocytes was measured on a FACS LSRII flow cytometer (BD Bioscience, CA) and analysis was performed using Flowjo software. Monocytes were gated on the CDl4-positive cells, and neutrophils were gated on the basis of the forward/side scatter characteristics.
  • FcgRIIA blockade on RNP-IC -induced IFNa expression was examined by ELISA. Briefly, 100 m ⁇ human or cynomolgous monkey PBMC (5xl0 6 cell/ml) were incubated in a tissue culture flask for 1 hour. Non-adherent cells were collected, and this monocyte-depleted fraction was incubated with anti- FcgRIIA Abs or its isotype control (R347- Tm Ab) from 300 pg/ml to 0.001 pg/ml with 3-fold dilution at 37°C incubator for 2 hours.
  • Peripheral blood from normal volunteers was drawn into vacutainer tubes containing heparin, and the blood was diluted 1 : 1 in HBSS. Neutrophils were separated by centrifugation on a lymphoprep density gradient. Erythrocytes were removed by RBC lysis buffer (Biolegend, CA). Neutrophils were further enriched using the Human Neutrophil Enrichment Kit (StemCell Technologies, CA).
  • Neutrophil superoxide production induced by anti-neutrophil cytoplasmic antibody (ANCA) or phorbol myri state acetate (PMA) was measured by the ferri-cytochrome C reduction assay. Briefly, isolated human neutrophils were suspended at 4 x 10 6 cells/ml in HBSS/20mM Hepes/Ca 2+ , Mg 2+ free buffer, and primed with recombinant 2 ng/ml TNF-a (R&D Systems,
  • neutrophils were pre-incubated with 10 pg/ml MED 19600 for 30 min at room temperature before they were added to the 96 well tissue culture plates.
  • the 96-well tissue culture plates were pre-treated with 100 pl of 0.5% milk in HBSS/20 mM Hepes for 1 hour at room temperature. Plates were washed three times with wash buffer (HBSS/20mM Hepes/Ca 2+ , Mg 2+ free buffer).
  • Neutrophil superoxide production induced by ANCA was measured by flow cytometry.
  • Neutrophils (lx l0 5 /ml) were resuspended in RPMI 1640 buffer with 1 pg/ml dihydrohodamine 123 (DHR) (Life Technologies, CA). Some cells were pre-incubated with 10 pg/ml MED19600, anti-FcyRIII Ab (Biolegend, CA), or isotype control Ab at room temperature for 30 minutes.
  • MA 10 pg/ml anti-PR3 Ab
  • MA 80 pg/ml IgG purified from sera of healthy volunteers or AAV patients (Tissue Solutions, UK), pelleted at 200g, and resuspended in ice- cold HBSS at a density of 5xl0 6 cells/ml.
  • Mean fluorescence intensity M.F.I was measured by LSRII flow cytometer (BD Bioscience, CA) and analysis was performed using Flowjo software.
  • Neutrophil migration was assessed using a 96-well Chemo TX system with 5 pm filter (Neuro Probe, MD).
  • Human IL-8 (R&D Systems, MN) was diluted in RPMI 1640 containing 1% BSA to a final concentration of 20 nM and placed in the lower chamber. Cells were washed and suspended in the same medium. Human neutrophils were incubated with 10 pg/ml
  • the migration index is the ratio of the number of cells that migrate in response to the chemotactic agent versus the number of cells that migrate in its absence.
  • ADCC assays were performed using a Celigo Imaging Cytometer (Nexelcom,
  • HEK-293 cells stably expressing CD32A were stained with Calcein AM dye (Thermo Fisher, Waltham, MA) following manufacturer’s protocol. Cells were then washed 3x in PBS and resuspended in RPMI 1640 media (lacking phenol red) with 3% FBS. Cells were then plated at ⁇ l0,000 cells/well a black-walled 96-well clear bottom plates
  • NK cells served as effector cells were isolated from healthy donors via the EasySep Human NK Cell Enrichment Kit (StemCell) following manufacturer’s directions. NK cells were added at a ratio of 3: 1 effector: target cells. Cells were then incubated at 37°C in 5% C0 2. After 5 h, Calcein AM positive cells were quantified in each well. Wells lacking antibody served as the“no specific lysis” control wells. Specific lysis (% cytotoxicity) was calculated as 100 - ((# of Calcein AM positive cells in test well)/ (mean # of Calcein AM cells in no effector cell control wells) c 100). Results shown are representative of 3 independent experiments. Data were graphed and analyzed using
  • CDC Complement-dependent cytotoxicity assays were performed using a Celigo Imaging Cytometer (Nexelcom). Adherent HEK-293 cells stably expressing CD32A were stained with Calcein AM dye (Thermo Fisher) following manufacturers protocol. Cells were then washed 3x in PBS and resuspended in RPMI 1640 media (lacking phenol red). Cells were then plated at ⁇ 10,000 cells/well in black-walled 96-well clear bottom plates (Greiner). Complement (baby rabbit complement; Cedarlane, Burlington, ON) was then added to a final concentration of 8% and cells were incubated at 37°C in 5% C0 2.
  • Luminescent P. aeruginosa were constructed using vector mini-CTX-lac-lux as previously described (DiGiandomenico et al. , J Exp. Med. 209:1273-1287 (2012)). P.
  • aeruginosa strains were grown to single colony on overnight plates of trypticase soy agar (TSA) at 37°C, followed by inoculation of a single colony into 10 mL Luria Bertani (LB) broth, and grown to an optical density at 650 nm of 0.4 (approximately 5xl0 8 colony forming units
  • Baby rabbit serum (BRS) (CedarLane, Hornby, Ontario, Canada) was used as the complement source for OPK assays.
  • BRS Baby rabbit serum
  • One milliliter of lyophilized BRS was reconstituted with ice-cold distilled water, followed by preparation of a 1 : 10 dilution in OPK assay buffer. Diluted BRS was kept on ice until preparation of all OPK components was completed.
  • MEDI9600 anti- FcgRIIA
  • H2B anti- FcyRIIB
  • 3G8 anti- FcyRIII
  • the final concentration of FcyR mAbs and primary neutrophils was 2 pg/ml and 2xl0 7 cells/ml, respectively.
  • dilutions of the anti-Psl antibody, diluted complement, and bacteria were added to each well, and the plate was covered with a breathable sealing film.
  • Wells lacking anti-Psl mAb antibody served as assay controls.
  • FIG. 12A Human blood was collected in heparin tubes. Five milliliters of blood was incubated with anti- FcgRIIA Ab (30 pg/ml), RNP-IC (1 mg/ml RNP /2% anti-RNP antibody seropositive SLE serum), or Ig-IC (100 pg/ml biotinylated-NMGC Ab-Streptavidin, 2: 1 mol: mol ratio) for 16 hours at 37°C. The plasma fraction was collected after centrifugation for 10 minutes at l300g. Over 200 protein analytes were measured using Rules-Based-Medicine Discovery MAP (Austin, TX). Data was generated from five individual donors: two 131H/H donors, two 131 R/R donors, and one 131 H/R donor, which covered all the FcgRIIA
  • FIG. 12B Human blood was collected in heparin tubes. Five milliliters of blood (5 mL) was incubated with the MED 19600 antibody (30 pg/mL), RNP-IC (1 pg/mL RNP /2% anti-RNP antibody seropositive SLE serum; RNP and SLE serum was pre-incubated at room temperature for 1 h to form immune complexes) or IgG-IC (final concentration: 100 pg/mL biotinylated-IgGl [Clone NMGC] antibody-streptavidin, 2: 1 molar ratio; biotinylated-IgGl antibody was pre-incubated with streptavidin at room temperature for 1 h to form immune complexes) for 16 h at 37°C.
  • MED 19600 antibody 30 pg/mL
  • RNP-IC 1 pg/mL RNP /2% anti-RNP antibody seropositive SLE serum
  • RNP and SLE serum was pre-in
  • FIG. 12B lists the panel of analytes examined.
  • the data were generated from five individual donors homozygous or heterozygous for the FcgRIIA 131 polymorphism (two 131H/H donors, two 131 R/R donors and one 131 H/R donor).
  • the data were z-score transformed, and heat-maps were generated in R using the heatmap.2 function of the gplots package.
  • the samples were clustered by type, although the protein clustering structure was unsupervised. The average fold change of treated blood compared to the untreated whole blood is listed in FIG. 12B.
  • IgGl Ab and control rat IgGl Ab were purchased from BD Bioscience.
  • FIG. 10A-C Wild- type and transgenic mice were injected intraperitoneally with 2pg of anti-CD4l at day 0. Some mice were pretreated 24 hours prior with MEDI9600 at 10 mg/kg. Blood was collected at day 1, day 5, day 8, and day 12 by retro-orbital bleeding, and platelets were counted using the Sysmex hematology analyzer. Platelet FcyRII A expression was determined by flow cytometry.
  • FIG. 10D-G MEDI9600 or control Ab (R347-Tm) was injected i.p. 24hrs before treatment.
  • Platelet numbers were assessed using a sysmex counter before the treatment and dayl, day 2 post treatment.
  • MEDI9600 was given by i.p at day 0, blood was collected at dayl, day2 and day 4 post injection, FcgRIIA level at platelets and neutrophils were determined by FACS and serum concentration of MEDI9600 was determined by human IgG ELISA.
  • IV Intravenous
  • This method utilized an indirect ELISA format to measure the concentration of
  • MEDI9600 in cynomolgus monkey serum Standards, controls, and test samples were incubated with sheep anti-human IgG (H+L) that had been immobilized on a microtiter plate. After incubation, unbound material was washed away and MEDI9600 was detected using goat anti- human IgG-(H+L)-HRP conjugate, and visualized with the addition of a tetramethylbenzidine (TMB) substrate solution. The color development was stopped, and the intensity of the color was measured at 450/650 nm.
  • TMB tetramethylbenzidine
  • Blood (approximately 1 mL) was collected for flow cytometry via a femoral vein from all animals on Days 12 and 20 of the pre-dose phase; at approximately 0.5 hours, 4 hours, 8 hours, and 24 hours post-dose; once on Days 3, 4, 5, and 8 of the dosing phase; and weekly thereafter (based on Day 1 of the dosing phase). Animals were not fasted unless samples were collected in conjunction with clinical pathology sampling. The anticoagulant was acid-citrate- dextrose. Parameters were expressed as percentages (of total CD45+ white blood cells) and absolute values (cells/pL).
  • FcgRIIA expression levels were described in fluorescence intensity and in calculated molecules of equivalent soluble fluorescence (MESF) values using median and/or geometric mean fluorescence intensity values.
  • the lymphocyte subsets shown in Table 10 were quantitated using flow cytometry.
  • Results were enumerated as percent relative (% of total CD45+ white blood cells) and absolute (cells/pL) values for each phenotype.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Molecular Biology (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Genetics & Genomics (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

The disclosure provides FcyRIIA-binding molecules, for example, humanized monoclonal antibodies capable of inhibiting FcyRIIA activity, and methods of using the FcyRIIA binding molecules, for example, in treating or preventing inflammatory, immune-mediated, or autoimmune diseases or disorders.

Description

BINDING MOLECULES SPECIFIC FOR FcgRIIA AND USES THEREOF
BACKGROUND
[0001] FcyRs are a family of cell surface receptors that bind to the Fc portion of antibodies of the immunoglobulin G (IgG) subclasses. Human Fc gamma receptors (FcyRs) differ in function, binding affinity, and in their cellular distribution1.
[0002] In humans, there are five FcyRs: the high-affinity receptor FcyRI (CD64), which can bind monomeric IgG; and the low-affinity receptors FcyRI I A (CD32A), FcyRI IB (CD32B), FcyRIIIA (CD16A), and FcyRIIIB (CD16B), which bind weakly to monomeric IgG, but avidly to immune complexes of IgG. FcyRI, FcgRIIA, FcyRIIIA, and FcyRIIIB are considered to have activating properties, whereas FcyRI IB is predominately inhibitory. FcgRIIA and FcyRI IB are the most closely related receptors. The extracellular regions of these receptors, which are responsible for interactions with IgG, share greater than 90% sequence identity2. Sequence differences in the intracellular signaling regions of FcgRIIA and FcyRI IB mediate the alternative cellular responses.
[0003] FcyRs mediate several cellular processes, including antigen or pathogen uptake, degranulation, antigen presentation, and antibody-dependent cellular cytotoxicity (ADCC). In addition, FcyRs can interact with other receptors to influence the production of specific cytokines. Failure of the immune system to appropriately limit the reactivity of FcyRs can play a role in the development of inflammatory, immune-mediated, or autoimmune diseases or disorders3.
[0004] Systemic lupus erythematosus (SLE) is a heterogeneous autoantibody driven immune- complex mediated autoimmune disease. A hallmark of SLE patients is the presence of autoantibodies directed against nuclear antigens, including dsDNA, ssDNA, and nucleic acid associated proteins ( e.g ., RNP, histones, Smith, Ro). Disease manifestations in the skin, lung, and kidney are associated with deposition of immune complexes4. Immune-complex -mediated activation of FcgRIIA has been implicated to play a role in the pathogenesis of SLE5.
Approximately 60% of SLE patients have a type I interfereon (IFN) gene signature which is most prevalent in patients with severe disease activity. Importantly, immune complexes containing DNA and RNA induce plasmacytoid dendritic cells to produce type I IFNa in an FcgRIIA-dependent manner5,67. [0005] Anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV) refers to a heterogeneous group of inflammatory diseases of blood vessels with multisystem
manifestations8. AAV comprises granulomatosis with polyangiitis (GPA), microscopic polyangiitis (MPA), necrotizing crescentic glomerulonephritis (NCGN), and eosinophilic granulomatosis (EGPA). A hallmark of AAV patients is the presence of autoantibodies directly against neutrophil cytoplasmic antigens. The target antigens of ANCA include myeloperoxidase (MPO), proteinase 3 (PR3), lactoferrin, and others. GPA is primarily associated with antibodies to PR3, whereas MPA and EGPA are both associated with antibodies to MPO. ANCAs not only serve as a diagnostic marker, but they also play a direct pathogenic role in the disease. ANCAs can induce the direct activation of neutrophils via FcgRIIA, which can drive vascular injury9. Moreover, ANCAs may also trigger FcgRIIA-dependent induction of neutrophil extracellular traps (NETs), cytokines, and chemokines, which may contribute to inflammation and the autoimmune response10. Therefore, FcgRIIA appears to play a central role in the development and pathology of AAV.
[0006] Immune thrombocytopenia (ITP) is an autoimmune bleeding disorder characterized by the production of auto-reactive antibodies directed against platelet antigens. Autoantibodies coating the surface of platelets promote their clearance by phagocytic macrophages of the reticuloendothelial system. The repertoire and cellular expression of FcyRs differ between mouse and human, and although FcyRII is the most broadly expressed FcyR in humans, it is absent in mice. Using mice, transgenic for human FcgRIIA and deficient for the murine activating FcyRs, it was demonstrated that passively administered anti-platelet antibodies triggered immune thrombocytopenia in an FcgRIIA-dependent manner11. In addition, significantly higher FcgRIIA/B ratio was observed on monocytes in patients with primary ITP; high-dose dexamethasone treatment, which is used as a first-line therapy for ITP patients, decreased the FcgRIIA/B ratio12. These in vitro and in vivo data indicate that human FcgRIIA plays a significant role in the pathogenesis of ITP.
[0007] The formation of neutrophil extracellular traps (NETs) are believed to be important in host defense against bacterial infections17. Conversely, formation of NETs is also associated with detrimental effects such as thrombosis, inflammation and endothelial dysfunction18 19,20. In addition to their pathogenic role in ANCA-associated vasculitis, there is evidence to indicate that NETs may contribute to sepsis, thrombosis, acute kidney injury, acute lung injury, chronic obstructive pulmonary disease, glomerulonephritis, toxic liver injury, stroke, atherogenesis and Type I diabetes21,22. Since FcgRIIA plays a crucial role in the formation of NETs10, blocking FcgRIIA may have a beneficial role in the treatment of NET-associated disorders.
[0008] Anti-drug antibodies (AD As) can be elicited in vivo in response to a therapeutic antibody. In addition to the impact of AD As on therapeutic exposure, the formation of immune complexes between the drug and AD As can elicit potential harmful FcR-mediated
hypersensitivity reactions23. Since FcgRIIA is the predominant activating FcyR that is responsible for immune complex-mediated effector functions, blocking FcgRIIA may be able to inhibit ADA-mediated adverse effects.
[0009] This disclosure provides compositions that specifically bind to FcgRIIA, and methods for the use of such compositions, such as for the treatment or prevention of an inflammatory, immune-mediated, or autoimmune disease or disorder.
SUMMARY OF THE INVENTION
[0010] Some of the main aspects of the present invention are summarized below. Additional aspects are described in the Detailed Description of the Invention, Examples, Drawings, and Claims sections of this disclosure. The description in each section of this disclosure is intended to be read in conjunction with the other sections. Furthermore, the various embodiments described in each section of this disclosure can be combined in various different ways, and all such combinations are intended to fall within the scope of the present invention.
[0011] The disclosure provides FcgRIIA-binding molecules, for example, humanized monoclonal antibodies capable of inhibiting FcgRIIA activity, and methods of using the FcgRIIA binding molecules, for example, in treating or preventing inflammatory, immune-mediated, or autoimmune diseases or disorders.
[0012] In one aspect, the invention provides an isolated binding molecule that specifically binds to FcgRIIA, wherein the binding molecule comprises an immunoglobulin variable heavy chain complementarity determining region 2 (VH-CDR2) comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 19 and SEQ ID NO: 20. In one aspect, the invention provides an isolated binding molecule that specifically binds to FcgRIIA, wherein the binding molecule comprises an immunoglobulin variable light chain complementarity determining region 1 (VL-CDR1) comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 22, SEQ ID NO: 23, SEQ ID NO: 24, SEQ ID NO: 25, and SEQ ID NO: 26. In a particular aspect, a binding molecule of the invention comprises a VH-CDR2 and a VL-CDR1 comprising amino acid sequences: (i) SEQ ID NO: 19 and SEQ ID NO: 22; (ii) SEQ ID NO: 19 and SEQ ID NO: 23; (iii) SEQ ID NO: 19 and SEQ ID NO: 24; (iv) SEQ ID NO: 20 and SEQ ID NO: 25; (v) SEQ ID NO: 20 and SEQ ID NO: 23; or (vi) SEQ ID NO: 20 and SEQ ID NO: 26, respectively.
[0013] The invention provides an isolated binding molecule that specifically binds to FcgRIIA, wherein the binding molecule comprises: (a) an immunoglobulin variable heavy chain complementarity determining region 2 (VH-CDR2) and an immunoglobulin variable light chain complementarity determining region 1 (VL-CDR1) comprising amino acid sequences: (i) SEQ ID NO: 19 and SEQ ID NO: 22; (ii) SEQ ID NO: 19 and SEQ ID NO: 23; (iii) SEQ ID NO: 19 and SEQ ID NO: 24; (iv) SEQ ID NO: 20 and SEQ ID NO: 25; (v) SEQ ID NO: 20 and SEQ ID NO: 23; or (vi) SEQ ID NO: 20 and SEQ ID NO: 26, respectively; (b) an immunoglobulin variable heavy chain complementarity determining region 1 (VH-CDR1) comprising SEQ ID NO: 29; (c) an immunoglobulin variable heavy chain complementarity determining region 3 (VH-CDR3) comprising SEQ ID NO: 30 or SEQ ID NO: 45; (d) an immunoglobulin variable light chain complementarity determining region 2 (VL-CDR2) comprising SEQ ID NO: 31; and (e) an immunoglobulin variable light chain complementarity determining region 3 (VL-CDR3) comprising SEQ ID NO: 32. In one embodiment, the binding molecule comprises SEQ ID NO: 19 and SEQ ID NO: 22.
[0014] In one embodiment, the binding molecule comprises a heavy chain variable (VH) region and a light chain variable (VL) region, comprising amino acid sequences selected from the group consisting of: (i) SEQ ID NO: 33 and SEQ ID NO: 34, (ii) SEQ ID NO: 35 and SEQ ID NO: 36, (iii) SEQ ID NO: 37 and SEQ ID NO: 38, (iv) SEQ ID NO: 39 and SEQ ID NO: 40, (v) SEQ ID NO: 41 and SEQ ID NO: 42, and (vi) SEQ ID NO: 43 and SEQ ID NO: 44, respectively. In one embodiment, the binding molecule comprises SEQ ID NO: 33 and SEQ ID NO: 34.
[0015] In another aspect, the invention provides an isolated binding molecule that competes or cross-competes with one or more of the binding molecules described above.
[0016] In some embodiments, the binding molecule of the invention is selected from a murine antibody, a human antibody, a humanized antibody, a chimeric antibody, monoclonal antibody, a polyclonal antibody, a recombinant antibody, a bi-specific antibody, a multi-specific antibody, and an antigen-binding fragment thereof.
[0017] In some embodiments, the binding molecule of the invention is selected from an Fv, an Fab, an F(ab’)2, an Fab’, a dsFv fragment, a single chain Fv (scFV), an sc(Fv)2, a disulfide- linked (dsFv), a diabody, a triabody, a tetrabody, a minibody, or a single chain antibody.
[0018] The binding molecule of the invention can comprise an immunoglobulin (Ig) heavy chain constant region. In one aspect, the constant region is a human IgG constant region.
[0019] In a particular embodiment, the constant region comprises amino acid substitutions at Kabat positions 234, 235, and 331, wherein: the amino acid at Kabat position 234 is substituted with Phenylalanine (F), the amino acid at Kabat position 235 is substituted with Glutamic acid (E), and the amino acid at Kabat position 331 is substituted with Serine (S).
[0020] In some embodiments, the constant region comprises one or more substitutions of amino acid residues at positions 251-257, 285-290, 308-314, 385-389, and 428-436, relative to a wild-type human IgG constant region, wherein the amino acid position numbering is according to the EU index as set forth in Kabat. In a particular embodiment, the constant region comprises amino acid substitutions at Kabat positions 252, 254, and 256, wherein: the amino acid at Kabat position 252 is substituted with Tyrosine (Y), the amino acid at Kabat position 254 is substituted with Threonine (T), and the amino acid at Kabat position 256 is substituted with Glutamic acid (E).
[0021] The binding molecule of the invention can comprise an immunoglobulin light chain constant region. In some embodiments, the light chain constant region is a human kappa constant region.
[0022] In one aspect, the binding molecule of the invention specifically binds human FcgRIIA 131R with an affinity characterized by a dissociation constant (KD) of about 0.16 nM, as measured by a BIAcore assay. In another aspect, the binding molecule of the invention specifically binds human FcgRIIA 131H with an affinity characterized by a dissociation constant (KD) of about 0.13 nM, as measured by a BIAcore assay. Preferably, the binding molecule does not specifically bind to FcyRI, FcyRIIB, or FcyRIII.
[0023] The binding molecule of the invention can be conjugated to an agent, for example, an agent selected from the group consisting of an antimicrobial agent, a therapeutic agent, a prodrug, a peptide, a protein, an enzyme, a lipid, a biological response modifier, a phannaceutical agent, a lymphokine, a heterologous antibody or fragment thereof, a detectable label, a polyethylene glycol (PEG), a toxin, and a combination of two or more of any said agents.
[0024] The invention further provides a composition comprising a binding molecule of the invention. In one embodiment, the composition is a diagnostic reagent.
[0025] In further embodiments, the invention provides a method for inhibiting
ribonucleoprotein-immune complex (RNP-IC)-mediated type I IFNa in a peripheral blood mononuclear cell (PBMC), the method comprising contacting the PBMC with a binding molecule of the invention. Also provided is a method for inhibiting anti -neutrophil cytoplasmic antibody (ANCA)-induced neutrophil activation, the method comprising contacting a neutrophil with a binding molecule of the invention.
[0026] In another embodiment, the invention provides a method for inhibiting
ribonucleoprotein-immune complex (RNP-IC)-mediated pro-inflammatory molecule production in a peripheral blood mononuclear cell (PBMC). The method comprising contacting the PBMC with a binding molecule of the invention. The pro-inflammatory molecule may be interleukin-6 or may be tumomecrosis factor alpha.
[0027] In some aspects, the invention provides a method of treating or preventing an inflammatory, immune-mediated, or autoimmune disease or disorder in a subject, the method comprising administering to a subject in need of treatment or to a subject susceptible to the disease or disorder an effective amount of a binding molecule or composition of the invention. The disease or disorder is preferably selected from ANCA-associated vasculitis (AAV), systemic lupus erythematosus (SLE), lupus nephritis, membranous nephritis, immune thrombocytopenia (ITP), rheumatoid arthritis, polymyositis, dermatomyositis, pemphigus, hemolytic anemia, mixed connective tissue disease, Sjogren’s syndrome, scleroderma, sepsis, thrombosis, acute kidney injury, acute lung injury, chronic obstructive pulmonary disease, glomerulonephritis, toxic liver injury, stroke, atherogenesis and Type I diabetes, an autoantibody disorder, and an immune- complex-mediated disorder. In some embodiments, the method comprises administering a second active agent.
[0028] Further provided is a method for detecting FcgRIIA in a sample, the method comprising (a) contacting the sample with a binding molecule of the invention, and (b) detecting binding of the binding molecule to FcgRIIA, thereby detecting FcgRIIA in the sample. In some instances, the method is a diagnostic method. [0029] In additional embodiments, the invention provides an isolated nucleic acid molecule comprising a nucleotide sequence encoding a binding molecule of the invention, optionally linked to a regulatory sequence; a host cell transformed with the nucleic acid molecule, preferably a mammalian host cell; and a vector comprising the nucleic acid molecule. Also provided is a composition comprising the nucleic acid molecule, host cell, or vector of the invention.
[0030] The invention provides a method of making a binding molecule that specifically binds FcgRIIA, the method comprising culturing the host cell of the invention under suitable conditions for producing the binding molecule. In some aspects, the method further comprises isolating the binding molecule.
[0031] In one embodiment, the invention provides a kit comprising a binding molecule or a nucleic acid molecule of the invention.
BRIEF DESCRIPTION OF THE DRAWINGS
[0032] FIGS. 1A-1C show consensus FcgRIIA cynomolgus sequences and alignments to human FcgRIIA FIG. 1A shows the consensus amino acid sequence of cynomolgus FcgRIIA (SEQ ID NO: 1). Single nucleotide polymorphism (SNP) variants among cynomolgus populations are shown in bold type. Minor alleles corresponding to the major human allele are underlined. Shaded residues indicate minor allele frequency (MAF) of >20%. FIG. IB shows alignment of human FcgRIIA (P12318; as 1-317; SEQ ID NO: 2) and cynomolgus FcgRIIA (SEQ ID NO: 3). Cyno amino acids that are non-homologous to human are shaded. Variations where the minor cyno allele corresponds to human are underlined. FIG. 1C shows the consensus FcgRIIA full-length cynomolgus transcript (SEQ ID NO: 4).
[0033] FIG. 2 shows epitope competition assay data for humanization of IV.3.
[0034] FIG. 3 shows alignment of mouse IV.3 VH (SEQ ID NO: 5) and VL (SEQ ID NO: 8) with humanized IV.3 (CamIV3 VH (SEQ ID NO: 6) and VL (SEQ ID NO: 9)) and selected human germline sequences (SEQ ID NO: 7, SEQ ID NO: 10).
[0035] FIGS. 4A-4B show epitope competition assay data for a panel of humanized IV.3 antibodies. FIG. 4A shows binding to human FcgRIIA 131H. FIG. 4B shows binding to human FcgRIIA 131R. [0036] FIGS. 5A-5G show that optimized IV.3 Abs are specific for human FcgRIIA binding, but not for other FcyRs; and that MED 19600 lacks CDC and ADCC effector functions. FIG. 5A shows binding data for human FcgRIIA. FIG. 5B shows binding data for FcyRIIB. FIG. 5C shows binding data for human FcyRI. FIG. 5D shows binding data for human FcyRIIIA - 158F allotype. FIG. 5E shows binding data for human FcyRIIIA - 158V allotype. FIG. 5F shows CDC activity of MED 19600 compared with wild type control 9600 IgGl and isotype control IgG (R347-Tm) antibodies. FIG. 5G shows ADCC activity of MED 19600 compared with wild type control 9600 IgGl and isotype control IgG (R347-Tm) antibodies. For both FIG. 5F and FIG. 5G, the plots represent the mean ± standard deviation. Representative plots of three independent experiments are shown.
[0037] FIG. 6 shows that MEDI9600 competes with intravenous immunoglobulin (IVIG) for binding to FcgRIIA.
[0038] FIGS. 7A-7G. FIGS. 7A-7C show that optimized IV.3 Abs internalize FcgRIIA from the surface of monocytes from human 131H/H donors (FIG. 7 A), human 131R/R donors (FIG. 7B), and cynomolgous monkeys (FIG. 7C). FIG. 7D shows internalization of FcgRIIA by MED 19600 using confocal microscopy. FIGS. 7E-G show that MEDI9600 internalizes FcgRIIA from the surface of neutrophils from human 131H/H donors (FIG. 7E), human 131R/R donors (FIG. 7F), and cynomolgous monkeys (FIG. 7G). For FIGS. 7E-G, representative data from three independent human and monkey experiments are shown.
[0039] FIGS. 8A-8D. FIGS. 8A-8C show that optimized IV.3 Abs block RNP-IC-induced IFN-a expression from human and cyno PBMC, using cells from human 131 H/H donors (FIG. 8A), human 131R/R donors (FIG. 8B), and cynomologus monkeys (FIG. 8C). FIG. 8D shows that MEDI9600 (30 pg/mL) inhibited Ig-IC induced TNF-a and IL-6 protein in whole blood.
[0040] FIGS. 9A-9G show that MED 19600 (clone 32LO0352) specifically blocks anti neutrophil cytoplasmic antibody (ANCA) induced neutrophil activation.
[0041] FIGS. 10A-10G. FIGS. 10A-10C show that MEDI9600 protects mice from anti platelets antibody— induced thrombocytopenia. FIG. 10D shows free FcgRIIA on platelets, measured by flow cytometry, at 24, 48, and 96 hrs after a single lmg/kg or 10 mg/kg i.p dose of MED 19600. FIG. 10E shows free FcgRIIA on neutrophils, measured by flow cytometry, at 24, 48, and 96 hrs after a single lmg/kg or 10 mg/kg i.p dose of MED 19600. FIG. 10F shows serum concentration of MED 19600 in the mice, measured by human IgG ELISA at 24, 48, and 96 hrs following delivery of a lmg/kg or 10 mg/kg i.p dose of MED 19600. FIG. 10G shows platelet numbers in the mice after prophylactic treatment with MEDI9600. MEDI9600 or control Ab (R347-Tm) was injected i.p. at lOmg/ml 24hrs before treatment, thrombocytopenia was induced with 2 pg rat anti-mouse CD41 Ab delivered i.p. at day 0. A representative plot of two independent experiments is shown. Error bars represent the mean ± standard deviation from one experiment. (n=3 replicates).
[0042] FIGS. 11A-11C show that blockade of FcgRIIA by MEDI9600 has no adverse effects on neutrophil function.
[0043] FIG. 12A - 12B. FIG. 12A shows that ex vivo treatment of human whole blood with MED 19600 alone has no effect on the protein expression profile. FIG. 12B provides the panel of analytes examined to determine lack of effect of MED 19600 on protein expression profile. Fold changes provided are changes for the treatment group versus untreated (n=5); P value was generated by paired Student’s t-test; statistical significance was ascribed to the data when P<0.05.
[0044] FIGS. 13A-13C show the results of a single-dose pharmacokinetic and exploratory pharmaco-dynamic study of MEDI9600. FIG. 13A shows the serum concentration of
MED 19600 in cynomolgus monkeys at various time points after a single dose. Flow cytometry analysis of blood cells from cynomolgus monkeys shows that MED 19600 induced a dose- response reduction of FcgRIIA fluorescence intensity on monocytes (FIG. 13B) and
granulocytes (FIG. 13C).
[0045] FIG. 14A - 14B show that, in a 13 week safety study, MED 19600 did not affect cynomologous monkey platelet counts (FIG. 14A) or body weights (FIG. 14B). The platelet count and body weight measurements were performed on (5/sex/group) monkeys that had been treated with weekly injections of vehicle control (IV and SC), 7.5 mg/kg (IV), 30 mg/kg (IV),
100 mg/kg (IV), or 100 mg/kg (SC) MED 19600 for a total of 13 weeks followed by an 8-week treatment-free period. Data are presented as mean ± SEM.
DETAILED DESCRIPTION OF THE INVENTION
[0046] The present invention provides molecules that bind to FcgRIIA In some embodiments, such molecules are antibodies or antigen-binding fragments thereof, which specifically bind to FcgRIIA. Related polynucleotides, compositions comprising the anti-FcgRIIA binding molecules, and methods of making the anti-FcgRIIA binding molecules are also provided.
Methods of using the novel anti-FcgRIIA antibodies, such as diagnostic methods and methods of treating inflammatory, immune-mediated, or autoimmune disease or disorders are further provided.
[0047] The practice of the present invention will employ, unless otherwise indicated, conventional techniques of cell biology, cell culture, molecular biology, transgenic biology, microbiology, recombinant DNA, and immunology, which are within the skill of the art. Such techniques are explained fully in the literature. See, e.g., Ausubel el al. eds. (2015) Current Protocols in Molecular Biology (John Wiley and Sons); Greenfield, ed. (2013) Antibodies: A Laboratory Manual (2nd ed., Cold Spring Harbor Press); Green and Sambrook, eds. (2012), Molecular Cloning: A Laboratory Manual (4th ed., Cold Spring Harbor Laboratory Press);
Krebs et al., eds. (2012) Lewin’s Genes XI (1 lth ed., Jones & Bartlett Learning); Freshney (2010) Culture Of Animal Cells (6th ed., Wiley); Weir and Blackwell, eds., (1996) Handbook Of Experimental Immunology, Volumes I-IV (5th ed., Wiley-Blackwell); Borrebaeck, ed. (1995) Antibody Engineering (2nd ed., Oxford Univ. Press); Glover and Hames, eds., (1995) DNA Cloning: A Practical Approach, Volumes I and II (2nd ed., IRL Press); Rees et al. , eds. (1993) Protein Engineering: A Practical Approach (lst ed., IRL Press); Mayer and Walker, eds. (1987) Immunochemical Methods In Cell And Molecular Biology (Academic Press, London); Nisonoff (1984) Introduction to Molecular Immunology (2nd ed., Sinauer Associates, Inc.); and Steward (1984) Antibodies: Their Structure and Function (lst ed., Springer Netherlands).
[0048] In order that the present invention can be more readily understood, certain terms are first defined. Additional definitions are set forth throughout the disclosure. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention is related. For example, The Dictionary of Cell and Molecular Biology (5th ed. J.M. Lackie ed., 2013), the Oxford Dictionary of Biochemistry and Molecular Biology (2d ed. R. Cammack et al. eds., 2008), and The Concise Dictionary of Biomedicine and Molecular Biology (2d ed. P-S. Juo, 2002) can provide one of skill with general definitions of some terms used herein.
I. Definitions [0049] As used in this specification and the appended claims, the singular forms“a,”“an,” and “the” include plural referents, unless the context clearly dictates otherwise. The terms“a” (or “an”) as well as the terms“one or more” and“at least one” can be used interchangeably.
[0050] Furthermore,“and/or” is to be taken as specific disclosure of each of the two specified features or components with or without the other. Thus, the term“and/or” as used in a phrase such as“A and/or B” is intended to include A and B, A or B, A (alone), and B (alone).
Likewise, the term“and/or” as used in a phrase such as“A, B, and/or C” is intended to include A, B, and C; A, B, or C; A or B; A or C; B or C; A and B; A and C; B and C; A (alone); B (alone); and C (alone).
[0051] Units, prefixes, and symbols are denoted in their Systeme International de Unites (SI) accepted form. Numeric ranges are inclusive of the numbers defining the range. Unless otherwise indicated, amino acid sequences are written left to right in amino to carboxy orientation, and nucleic acid sequences are written left to right in 5’ to 3’ orientation. The headings provided herein are not limitations of the various aspects or embodiments of the invention, which can be had by reference to the specification as a whole. Accordingly, the terms defined immediately below are more fully defined by reference to the specification in its entirety.
[0052] Wherever embodiments are described with the language“comprising,” otherwise analogous embodiments described in terms of“consisting of’ and/or“consisting essentially of’ are included.
[0053] Amino acids are referred to herein by their commonly known three-letter symbols or by the one-letter symbols recommended by the IUPAC-IUB Biochemical Nomenclature
Commission. Nucleotides, likewise, are referred to by their commonly accepted single-letter codes.
[0054] “FcgRIIA” refers to Fc receptor-gamma IIA. The full-length amino acid and nucleotide sequences for human and cynomolgus monkey ( Macaca fasciculari ) FcgRIIA are known in the art. (See, e.g., FIG. 1.) The terms“FcgRIIA” and“CD32A” are used
interchangeably throughout this disclosure. The terms“FcyRIIB” and“CD32B” are also used interchangeably throughout this disclosure, as are the terms“FcyRIIIA” and“CD16A,” and “FcyRIIIB” and“CD16B,” and“FcyRI” and“CD64,” respectively.
[0055] The term“antibody” refers to an immunoglobulin molecule that recognizes and specifically binds to a target, such as a protein, polypeptide, peptide, carbohydrate, polynucleotide, lipid, or combinations of the foregoing through at least one antigen recognition site within the variable region of the immunoglobulin molecule. As used herein, the term “antibody” encompasses polyclonal antibodies; monoclonal antibodies; multispecific antibodies, such as bispecific antibodies generated from at least two intact antibodies; humanized antibodies; human antibodies; chimeric antibodies; fusion proteins comprising an antigen-determination portion of an antibody; and any other modified immunoglobulin molecule comprising an antigen recognition site, so long as the antibodies exhibit the desired biological activity. Antibodies can be of any the five major classes of immunoglobulins: IgA, IgD, IgE, IgG and IgM, or subclasses (isotypes) thereof (e.g. IgGl, IgG2, IgG3, IgG4, IgAl and IgA2), based on the identity of their heavy-chain constant domains referred to as alpha, delta, epsilon, gamma, and mu respectively. The different classes of immunoglobulins have different and well-known subunit structures and three-dimensional configurations. There are two classes of mammalian light chains, lambda and kappa. Antibodies can be naked or conjugated to other molecules such as toxins, radioisotopes, etc.
[0056] The term“anti gen -binding fragment” refers to a portion of an intact antibody comprising the complementarity determining variable regions of the antibody. Fragments of a full-length antibody can be an antigen-binding fragment of an antibody. Examples of antibody fragments include, but are not limited to Fab, Fab’, F(ab’)2, and Fv fragments, linear antibodies, single chain antibodies (e.g., ScFvs), and multispecific antibodies formed from antibody fragments.
[0057] A“monoclonal antibody” (mAb) refers to a homogeneous antibody population that is involved in the highly specific recognition and binding of a single antigenic determinant, or epitope. This is in contrast to polyclonal antibodies, which typically include different antibodies directed against different antigenic determinants The term“monoclonal” can apply to both intact and full-length monoclonal antibodies, as well as to antibody fragments (such as Fab, Fab’, F(ab’)2, Fv), single chain (scFv) mutants, fusion proteins comprising an antibody portion, and any other modified immunoglobulin molecule comprising an antigen recognition site.
Furthermore,“monoclonal antibody” refers to such antibodies made in any number of ways including, but not limited to, by hybridoma, phage selection, recombinant expression, and transgenic animals. [0058] The term“humanized antibody” refers to an antibody derived from a non-human ( e.g ., murine) immunoglobulin, which has been engineered to contain minimal non-human (e.g., murine) sequences. Typically, humanized antibodies are human immunoglobulins in which residues from the complementary determining region (CDR) are replaced by residues from the CDR of a non-human species (e.g, mouse, rat, rabbit, or hamster) that have the desired specificity, affinity, and capability (Jones et al., 1986, Nature, 321 :522-525; Riechmann et al., 1988, Nature, 332:323-327; Verhoeyen et al, 1988, Science, 239: 1534-1536). In some instances, the Fv framework region (FW) residues of a human immunoglobulin are replaced with the corresponding residues in an antibody from a non-human species that has the desired specificity, affinity, and capability.
[0059] Humanized antibodies can be further modified by the substitution of additional residues either in the Fv framework region and/or within the replaced non-human residues to refine and optimize antibody specificity, affinity, and/or capability. In general, humanized antibodies will comprise substantially all of at least one, and typically two or three, variable domains containing all or substantially all of the CDR regions that correspond to the non-human immunoglobulin whereas all or substantially all of the FR regions are those of a human immunoglobulin consensus sequence. Humanized antibody can also comprise at least a portion of an
immunoglobulin constant region or domain (Fc), typically that of a human immunoglobulin. Examples of methods used to generate humanized antibodies are described in U.S. Pat. Nos. 5,225,539 and 5,639,641.
[0060] The term“human antibody” means an antibody produced by a human or an antibody having an amino acid sequence corresponding to an antibody produced by a human made using any technique known in the art. The definition of a human antibody includes intact or full-length antibodies comprising at least one human heavy and/or light chain polypeptide such as, for example, an antibody comprising murine light chain and human heavy chain polypeptides.
[0061] The term“chimeric antibodies” refers to antibodies wherein the amino acid sequence of the immunoglobulin molecule is derived from two or more species. Typically, the variable region of both light and heavy chains corresponds to the variable region of antibodies derived from one species of mammals (e.g, mouse, rat, rabbit, etc.) with the desired specificity, affinity, and capability while the constant regions are homologous to the sequences in antibodies derived from another (usually human) to avoid eliciting an immune response in that species. [0062] A“blocking” antibody or an“antagonist” antibody is one that inhibits or reduces biological activity of the antigen it binds, such as FcgRIIA. In certain aspects, blocking antibodies or antagonist antibodies substantially or completely inhibit the biological activity of the antigen. Desirably, the biological activity is reduced by 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or even 100%.
[0063] The term“germlining” means that amino acids at specific positions in an antibody are mutated back to those in the germ line.
[0064] The“IgGl triple mutant” or“IgGl-TM” antibody format is a human IgGl isotype containing three single amino acid substitutions, L234F/L235E/P331S, within the lower hinge and CH2 domain (Oganesyan et al., Acta Crystallogr. D Biol. Crystallogr. 64:700-704, 2008). The TM causes a profound decrease in binding to human FcyRI, FcyRII, FcyRIII, and Clq, resulting in a human isotype with very low effector function.
[0065] The terms“YTE” or“YTE mutant” refer to a mutation in IgGl Fc that results in an increase in the binding to human FcRn and improves the serum half-life of the antibody having the mutation. A YTE mutant comprises a combination of three mutations,
M252Y/S254T/T256E (EEG numbering Rabat et al. (1991) Sequences of Proteins of
Immunological Interest, ET.S. Public Health Service, National Institutes of Health, Washington, D.C.), introduced into the heavy chain of an IgGl. See ET.S. Patent No. 7,658,921, which is incorporated by reference herein. The YTE mutant has been shown to increase the serum half- life of antibodies approximately four-times as compared to wild-type versions of the same antibody (DalF Acqua et al, ./. Biol. Chem. 281 :23514-24 (2006); Robbie et al, Antimicrob. Agents Chemother. 57, 6147-6153 (2013)). See also U.S. Patent No. 7,083,784, which is hereby incorporated by reference in its entirety.
[0066] The terms“antibody” or“immunoglobulin” are used interchangeably herein. A typical antibody comprises at least two heavy (H) chains and two light (L) chains interconnected by disulfide bonds. Each heavy chain is comprised of a heavy chain variable region (abbreviated herein as VH) and a heavy chain constant region. The heavy chain constant region is comprised of three domains, CH1, CH2, and CH3. Each light chain is comprised of a light chain variable region (abbreviated herein as VL) and a light chain constant region (CL). The light chain constant region is comprised of one domain, Cl. The variable regions of the heavy and light chains contain a binding domain that interacts with an antigen. The constant regions of the antibodies can mediate the binding of the immunoglobulin to host tissues or factors, including various cells of the immune system (e.g. effector cells) and the first component (Clq) of the classical complement system.
[0067] The VH and VL regions can be further subdivided into regions of hypervariability, termed complementarity-determining regions (CDRs), interspersed with regions that are more conserved, termed framework (FW) regions. The CDRs in each chain are held together in close proximity by the FW regions and, with the CDRs from the other chain, contribute to the formation of the antigen-binding site of antibodies. Each VH and VL is composed of three CDRs and four FWs, arranged from amino-terminus to carboxy-terminus in the following order: FW1, CDR1, FW2, CDR2, FW3, CDR3, FW4.
[0068] There are at least two techniques for determining CDRs: (1) an approach based on cross-species sequence variability (Kabat et al, Sequences of Proteins of Immunological Interest , 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md. (1991)); and (2) an approach based on crystallographic studies of antigen-antibody complexes (Al-lazikani et al ., ./. Molec. Biol. 273:927-948 (1997)). In addition, combinations of these two approaches are sometimes used in the art to determine CDRs.
[0069] The amino acid position numbering as in Kabat, refers to the numbering system used for heavy chain variable domains or light chain variable domains (approximately residues 1-107 of the light chain and residues 1-113 of the heavy chain). ETsing this numbering system, the actual linear amino acid sequence can contain fewer or additional amino acids corresponding to a shortening of, or insertion into, a FW or CDR of the variable domain. For example, a heavy chain variable domain can include a single amino acid insert (residue 52a, according to Kabat) after residue 52 of H2 and inserted residues (e.g, residues 82a, 82b, and 82c, etc., according to Kabat) after heavy chain FW residue 82.
Figure imgf000016_0001
Figure imgf000017_0001
[0070] The Kabat numbering of residues can be determined for a given antibody by alignment at regions of homology of the sequence of the antibody with a“standard” Kabat numbered sequence. Chothia refers instead to the location of the structural loops (Chothia and Lesk, J Mol. Biol. 196:901-917 (1987)). The end of the Chothia CDR-H1 loop when numbered using the Kabat numbering convention varies between H32 and H34 depending on the length of the loop (this is because the Kabat numbering scheme places the insertions at H35A and H35B; if neither 35A nor 35B is present, the loop ends at 32; if only 35A is present, the loop ends at 33; if both 35 A and 35B are present, the loop ends at 34). The AbM hypervariable regions represent a compromise between the Kabat CDRs and Chothia structural loops, and are used by Oxford Molecular’s AbM antibody modeling software. See Table 1.
[0071] IMGT (ImMunoGeneTics) also provides a numbering system for the immunoglobulin variable regions, including the CDRs. See, e.g. , Lefranc, M.P. et al. , Dev. Comp. Immunol. 27: 55-77 (2003). The IMGT numbering system was based on an alignment of more than 5,000 sequences, structural data, and characterization of hypervariable loops and allows for easy comparison of the variable and CDR regions for all species. According to the IMGT numbering schema VH-CDR1 is at positions 26 to 35, VH-CDR2 is at positions 51 to 57, VH-CDR3 is at positions 93 to 102, VL-CDR1 is at positions 27 to 32, VL-CDR2 is at positions 50 to 52, and VL-CDR3 is at positions 89 to 97.
[0072] As used throughout the specification the VH CDRs sequences described correspond to the classical Kabat numbering locations, namely Kabat VH-CDR1 is at positions 31-35, VH- CDR2 is a positions 50-65, and VH-CDR3 is at positions 95-102. VL-CDR1, VL-CDR2 and VL-CDR3 also correspond to classical Kabat numbering locations, namely positions 24-34, 50- 56 and 89-97, respectively.
[0073] “Binding affinity” generally refers to the strength of the sum total of non-covalent interactions between a single binding site of a molecule (e.g, an antibody) and its binding partner (e.g, an antigen). Unless indicated otherwise, as used herein,“binding affinity” refers to intrinsic binding affinity which reflects a 1 : 1 interaction between members of a binding pair (e.g, antibody and antigen). The affinity of a molecule X for its partner Y can generally be represented by the dissociation constant (KD). Affinity can be measured by common methods known in the art, including those described herein. Low-affinity antibodies generally bind antigen slowly and tend to dissociate readily, whereas high-affinity antibodies generally bind antigen faster and tend to remain bound longer.
[0074] The affinity or avidity of an antibody for an antigen can be determined experimentally using any suitable method known in the art, e.g ., flow cytometry, enzyme-linked immunosorbent assay (ELISA), or radioimmunoassay (RIA), or kinetics (e.g, KINEXA® or BIACORE™ analysis). Direct binding assays as well as competitive binding assay formats can be readily employed. (See, e.g, Berzofsky et a/.,“Antibody -Antigen Interactions,” In Fundamental Immunology, Paul, W. E., ed., Raven Press: New York, N.Y. (1984); Kuby, Immunology, W. H. Freeman and Company: New York, N.Y. (1992)) The measured affinity of a particular antibody-antigen interaction can vary if measured under different conditions (e.g, salt concentration, pH, temperature). Thus, measurements of affinity and other antigen-binding parameters (e.g, KD or Kd, Kon, K0ff) are made with standardized solutions of antibody and antigen, and a standardized buffer, as known in the art.
[0075] “Potency” is normally expressed as an IC50 value, in nM or pM, unless otherwise stated. IC50 is the median inhibitory concentration of an antibody molecule. In functional assays, IC50 is the concentration that reduces a biological response by 50% of its maximum. In ligand-binding studies, IC50 is the concentration that reduces receptor binding by 50% of maximal specific binding level. IC50 can be calculated by any number of means known in the art.
[0076] The fold improvement in potency for the antibodies or polypeptides of the invention as compared to a reference antibody can be at least about 2-fold, at least about 4-fold, at least about 6-fold, at least about 8-fold, at least about lO-fold, at least about 20-fold, at least about 30-fold, at least about 40-fold, at least about 50-fold, at least about 60-fold, at least about 70-fold, at least about 80-fold, at least about 90-fold, at least about lOO-fold, at least about 1 lO-fold, at least about l20-fold, at least about l30-fold, at least about l40-fold, at least about l50-fold, at least about 160-fold, at least about 170-fold, or at least about 180-fold or more.
[0077] The terms“inhibit,”“block,” and“suppress” are used interchangeably and refer to any statistically significant decrease in biological activity, including full blocking of the activity. For example,“inhibition” can refer to a decrease of about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or 100% in biological activity. Accordingly, when the terms“inhibition” or “suppression” are applied to describe, e.g, an effect on the FcgRIIA signal transduction pathway, the terms refer to the ability of an FcgRIIA binding molecule to statistically significantly decrease FcgRIIA-induced cell activation or signal transduction relative to an untreated (control) cell. The cell that expresses FcgRIIA can be a naturally occurring cell or cell line, (e.g. , macrophage, neutrophil, eosinophil, platelet) or can be recombinantly produced by introducing a nucleic acid encoding FcgRIIA into a host cell. In one embodiment, the FcgRIIA binding molecule can inhibit FcgRIIA-mediated cell activation or signal transduction in an FcgRIIA- expressing cell by at least 10%, or at least 20%, or at least 30%, or at least 40%, or at least 50%, or at least 60%, or at least 70%, or at least 80%, or at least 90% or about 100%, as determined, for example, by flow cytometry, Western blotting, ELISA, or other assays known to those of skill in the art.
[0078] An“isolated” polypeptide, antibody, binding molecule, polynucleotide, vector, or cell is in a form not found in nature. Isolated polypeptides, antibodies, binding molecules, polynucleotides, vectors, or cells include those which have been purified to a degree that they are no longer in a form in which they are found in nature. In some embodiments, a polypeptide, antibody, binding molecule, polynucleotide, vector, or cell that is isolated is substantially pure. When used herein, the term“substantially pure” refers to purity of greater than 75%, preferably greater than 80% or 90%, and most preferably greater than 95%.
[0079] By“subject” or“individual” or“animal” or“patient” or“mammal,” is meant any subject, particularly a mammalian subject, for whom diagnosis, prognosis, or therapy is desired. Mammalian subjects include humans, domestic animals, farm animals, sports animals, and zoo animals including, e.g., humans, non-human primates, dogs, cats, guinea pigs, rabbits, rats, mice, horses, cattle, bears, and so on.
[0080] The term“pharmaceutical composition” refers to a preparation that is in such form as to permit the biological activity of the active ingredient to be effective and which contains no additional components that are unacceptably toxic to a subject to which the composition would be administered. Such composition can be sterile and can comprise a pharmaceutically acceptable carrier, such as physiological saline. Suitable pharmaceutical compositions can comprise one or more of a buffer, a surfactant, a stabilizing agent, a preservative, an absorption promoter to enhance bioavailability and/or other conventional solubilizing or dispersing agents. [0081] An“effective amount” of a binding molecule as disclosed herein is an amount sufficient to carry out a specifically stated purpose. An“effective amount” can be determined empirically and in a routine manner, in relation to the stated purpose.
[0082] The term“label” when used herein refers to a detectable compound or composition that is conjugated directly or indirectly to a binding molecule, so as to generate a“labeled” binding molecule. The label can be detectable by itself ( e.g ., radioisotope labels or fluorescent labels) or, as in the case of, e.g., an enzymatic label, can catalyze chemical alteration of a substrate compound or composition that is detectable.
[0083] Terms such as“treating” or“treatment” or“to treat” or“alleviating” or“to alleviate” refer to therapeutic measures that cure, slow down, lessen symptoms of, and/or halt progression of a diagnosed pathologic condition or disorder. Thus, those in need of treatment include those already with the disorder. In certain embodiments, a subject is successfully“treated” for an inflammatory, immune-mediated, or autoimmune disease or disorder according to the methods provided herein if the patient shows, e.g, total, partial, or transient alleviation or elimination of symptoms associated with the disease or disorder.
[0084] “Prevent” or“prevention” refer to prophylactic or preventative measures that prevent and/or slow the development of a targeted pathologic condition or disorder. Thus, those in need of prevention include those prone to have or susceptible to the disorder. In certain embodiments, an inflammatory, immune-mediated, or autoimmune disease or disorder is successfully prevented according to the methods provided herein if the patient develops, transiently or permanently, e.g, fewer or less severe symptoms associated with the disease or disorder, or a later onset of symptoms associated with the disease or disorder, than a patient who has not been subject to the methods of the invention.
[0085] The terms“polypeptide,”“peptide,” and“protein” are used interchangeably herein to refer to polymers of amino acids of any length. The polymer can be linear or branched, it can comprise modified amino acids and non-amino acids can interrupt it. The terms also encompass an amino acid polymer that has been modified naturally or by intervention; for example, disulfide bond formation, glycosylation, lipidation, acetylation, phosphorylation or any other manipulation or modification such as conjugation with a labeling component. Also included within the definition are, for example, polypeptides containing one or more analogs of an amino acid (including, for example, unnatural amino acids, etc.), as well as other modifications known in the art. In certain embodiments, the polypeptides can occur as single chains or associated chains.
[0086] A“conservative amino acid substitution” is one in which one amino acid residue is replaced with another amino acid residue having a similar side chain. Families of amino acid residues having similar side chains have been defined in the art, including basic side chains ( e.g ., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g, asparagine, glutamine, serine, threonine, tyrosine, cysteine), nonpolar side chains (e.g, glycine, alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan), beta-branched side chains (e.g, threonine, valine, isoleucine) and aromatic side chains (e.g, tyrosine, phenylalanine, tryptophan, histidine). For example, substitution of a phenylalanine for a tyrosine is a conservative substitution. In certain embodiments, conservative substitutions in the amino acid sequences of the binding molecules of the invention do not abrogate the binding of the binding molecule to the antigen(s), i.e., FcgRIIA, to which the binding molecule binds. Methods of identifying conservative nucleotide and amino acid substitutions which do not eliminate antigen-binding are well-known in the art (see, e.g, Brummell et al., Biochem. 32: 1180-1 187 (1993); Kobayashi et al. , Protein Eng. 12(10):879- 884 (1999); and Burks et al., Proc. Natl. Acad. Sci. U.S.A. 94:.412-417 (1997)).
[0087] A“polynucleotide,” as used herein can include one or more“nucleic acids,”“nucleic acid molecules,” or“nucleic acid sequences,” and refers to a polymer of nucleotides of any length, and includes DNA and RNA. The polynucleotides can be deoxyribonucleotides, ribonucleotides, modified nucleotides or bases, and/or their analogs, or any substrate that can be incorporated into a polymer by DNA or RNA polymerase. A polynucleotide can comprise modified nucleotides, such as methylated nucleotides and their analogs. The preceding description applies to all polynucleotides referred to herein, including RNA and DNA.
[0088] The term“vector” means a construct, which is capable of delivering and, in some embodiments expressing, one or more gene(s) or sequence(s) of interest in a host cell. Examples of vectors include, but are not limited to, viral vectors, naked DNA or RNA expression vectors, plasmid, cosmid or phage vectors, DNA or RNA expression vectors associated with cationic condensing agents, DNA or RNA expression vectors encapsulated in liposomes, and certain eukaryotic cells, such as producer cells. [0089] The terms“identical” or percent“identity” in the context of two or more nucleic acids or polypeptides, refer to two or more sequences or subsequences that are the same or have a specified percentage of nucleotides or amino acid residues that are the same, when compared and aligned (introducing gaps, if necessary) for maximum correspondence, not considering any conservative amino acid substitutions as part of the sequence identity. The percent identity can be measured using sequence comparison software or algorithms or by visual inspection. Various algorithms and software are known in the art that can be used to obtain alignments of amino acid or nucleotide sequences.
[0090] One such non-limiting example of a sequence alignment algorithm is the algorithm described in Karlin et al. , Proc. Natl. Acad. Sci., 87:2264-2268 (1990), as modified in Karlin et al, Proc. Natl. Acad. Sci., 90:5873-5877 (1993), and incorporated into the NBLAST and
XBLAST programs (Altschul et al, Nucleic Acids Res., 25:3389-3402 (1991)). In certain embodiments, Gapped BLAST can be used as described in Altschul et al, Nucleic Acids Res. 25:3389-3402 (1997). BLAST-2, WU-BLAST-2 (Altschul et al, Methods in Enzymology, 266:460-480 (1996)), ALIGN, ALIGN-2 (Genentech, South San Francisco, California) or Megalign (DNASTAR) are additional publicly available software programs that can be used to align sequences. In certain embodiments, the percent identity between two nucleotide sequences is determined using the GAP program in the GCG software package ( e.g ., using a
NWSgapdna.CMP matrix and a gap weight of 40, 50, 60, 70, or 90 and a length weight of 1, 2, 3, 4, 5, or 6). In certain alternative embodiments, the GAP program in the GCG software package, which incorporates the algorithm of Needleman and Wunsch (J. Mol. Biol. (48):444-453 (1970)), can be used to determine the percent identity between two amino acid sequences (e.g., using either a BLOSUM 62 matrix or a PAM250 matrix, and a gap weight of 16, 14, 12, 10, 8, 6, or 4 and a length weight of 1, 2, 3, 4, 5). Alternatively, in certain embodiments, the percent identity between nucleotide or amino acid sequences is determined using the algorithm of Myers and Miller ( CABIOS 4: 11-17 (1989)). For example, the percent identity can be determined using the ALIGN program (version 2.0) and using a PAM120 with residue table, a gap length penalty of 12 and a gap penalty of 4. One skilled in the art can determine appropriate parameters for maximal alignment by particular alignment software. In certain embodiments, the default parameters of the alignment software are used. [0091] In certain embodiments, the percentage identity“X” of a first amino acid sequence to a second sequence amino acid is calculated as 100 x (Y/Z), where Y is the number of amino acid residues scored as identical matches in the alignment of the first and second sequences (as aligned by visual inspection or a particular sequence alignment program) and Z is the total number of residues in the second sequence. If the length of a first sequence is longer than the second sequence, the percent identity of the first sequence to the second sequence will be higher than the percent identity of the second sequence to the first sequence.
II. FcgRIIA-Binding Molecules
[0092] The present invention provides FcgRIIA binding molecules, /. e. , anti -FcgRIIA antibodies and antigen-binding fragments thereof, which specifically bind FcgRIIA The term “FcgRIIA binding molecule” or“binding molecule that binds to FcgRIIA” or“anti -FcgRIIA” refers to a binding molecule that is capable of binding FcgRIIA with sufficient affinity such that the binding molecule is useful as a therapeutic agent or diagnostic reagent in targeting FcgRIIA. A binding molecule that“specifically binds to FcgRIIA” binds to an unrelated, non-FcgRIIA protein to an extent of less than about 10% of the binding of the binding molecule to FcgRIIA, as measured, e.g ., by a radioimmunoassay (RIA), BIACORE® (using recombinant FcgRIIA as the analyte and binding molecule as the ligand, or vice versa), KINEXA®, or other binding assays known in the art. In certain embodiments, binding molecule that binds to FcgRIIA has a dissociation constant (KD) of <1 mM, <100 nM, <10 nM, <1 nM, <0.1 nM, <10 pM, <1 pM, or <0.1 pM.
[0093] Exemplary binding molecules of the present disclosure include variants of the mouse monoclonal antibody IV.3 (Looney RJ. et al, ./. Immunol. 136(5): 1641 (1986)), including humanized, optimized, germlined, and/or other versions of these antibodies, anti-FcgRIIA TM antibodies, and serum half-life-optimized anti-FcgRIIA YTE antibodies (e.g, K44VHa-N56Q, K44VHa6-N56Q, or K2Ha-N56Q). Exemplary antibodies of the present disclosure include clones 32L00350, 32LO0351, 32LO0352, 32LO0354, 32LO0355, and 32LO0356. “Clone 32LO0352” and“MEDI9600” refer to the same molecule and the terms are used interchangeably herein. The invention also embraces variants and equivalents that are substantially homologous to the FcgRIIA-binding molecules set forth herein. These can contain, for example, conservative amino acid substitutions. [0094] In certain aspects, this disclosure provides an FcgRIIA binding molecule that can specifically bind to the same FcgRIIA epitope as a binding molecule comprising the heavy chain variable region (VH) and light chain variable region (VL) of any one of clones 32L00350, 32LO0351, 32LO0352, 32LO0354, 32LO0355, or 32LO0356. The term“epitope” refers to a target protein determinant capable of binding to a binding molecule of the invention. Epitopes usually consist of chemically active surface groupings of molecules such as amino acids or sugar side chains, and usually have specific three-dimensional structural characteristics, as well as specific charge characteristics. Conformational and non-conformational epitopes are
distinguished in that the binding to the former but not the latter is lost in the presence of denaturing solvents. Such binding molecules can be identified based on their ability to cross- compete ( e.g ., to competitively inhibit the binding of, in a statistically significant manner) with binding molecules such as clones 32L00350, 32LO0351, 32LO0352, 32LO0354, 32LO0355, or 32LO0356, in standard FcgRIIA binding or activity assays.
[0095] Accordingly, in one embodiment, the invention provides FcgRIIA binding molecules that compete for binding to FcgRIIA with another FcgRIIA binding molecule of the invention, such as one of clones 32L00350, 32LO0351, 32LO0352, 32LO0354, 32LO0355, or 32LO0356. The ability of a binding molecule to inhibit the binding of, e.g., clones 32L00350, 32LO0351, 32LO0352, 32LO0354, 32LO0355, or 32LO0356, demonstrates that the test binding molecule can compete with clones 32L00350, 32LO0351, 32LO0352, 32LO0354, 32LO0355, or
32LO0356 for binding to FcgRIIA; such a binding molecule can, according to non-limiting theory, bind to the same or a related (e.g, a structurally similar or spatially proximal) epitope on FcgRIIA as the FcgRIIA binding molecule with which it competes. In one embodiment, the anti- FcgRIIA antibody or antigen-binding fragment thereof that binds to the same epitope on
FcgRIIA as any of clones 32L00350, 32LO0351, 32LO0352, 32LO0354, 32LO0355, or 32LO0356. The term“competes” indicates that a binding molecule competes uni directionally for binding to FcgRIIA with any one of 32L00350, 32LO0351, 32LO0352, 32LO0354,
32LO0355, or 32LO0356. The term“cross-competes” indicates that a binding molecule competes bidirectionally for binding to FcgRIIA with any one of 32L00350, 32LO0351, 32LO0352, 32LO0354, 32LO0355, or 32LO0356.
[0096] In some embodiments, the FcgRIIA binding molecule is a murine antibody, a human antibody, a humanized antibody, a chimeric antibody, a monoclonal antibody, a polyclonal antibody, a recombinant antibody, a bi-specific antibody, a multi specific antibody, or any combination thereof. In some embodiments, FcgRIIA binding molecules comprise a Fab, a Fab’, a F(ab’)2, a Fd, a Fv, a scFv, a disulfide linked Fv, a V-NAR domain, an IgNar, an intrabody, an IgGACFLZ, a minibody, a F(ab’)3, a tetrabody, a triabody, a diabody, a single-domain antibody, DVD-Ig, Fcab, mAb2, a (scFv)2, or a scFv-Fc.
[0097] An FcgRIIA binding molecule provided herein can include, in addition to a VH and a VL, a heavy chain constant region or fragment thereof. In certain aspects the heavy chain constant region is a human heavy chain constant region, e.g ., a human IgG constant region, e.g. , a human IgGl constant region.
[0098] In certain embodiments, binding molecules of the invention are produced to comprise an altered Fc region, in which one or more alterations have been made in the Fc region in order to change functional and/or pharmacokinetic properties of the binding molecule. Such alterations may result in altered effector function, reduced immunogenicity, and/or an increased serum half-life. The Fc region interacts with a number of ligands, including Fc receptors, the complement protein Clq, and other molecules, such as proteins A and G. These interactions are essential for a variety of effector functions and downstream signaling events including antibody dependent cell-mediated cytotoxicity (ADCC) and complement dependent cytotoxicity (CDC). Accordingly, in certain embodiments the FcgRIIA binding molecules of the invention have reduced or ablated affinity for an Fc ligand responsible for facilitating effector function, compared to an FcgRIIA binding molecule not comprising the modification in the Fc region. In particular embodiments, the FcgRIIA binding molecule has no ADCC activity and/or no CDC activity. In certain aspects, the FcgRIIA binding molecule does not bind to an Fc receptor and/or complement factors. In certain aspects, the FcgRIIA binding molecule has no effector function. Selecting particular constant domains to optimize desired effector functions is within the ordinary skill in the art. In some embodiments, the binding molecule is of the IgGl subtype, and optionally comprises the TM format (L234F/L235E/P331S), as disclosed supra in the
Definitions section.
[0099] In certain aspects, a heavy chain constant region or fragment thereof can include one or more amino acid substitutions relative to a wild-type IgG constant domain, wherein the modified IgG has an increased half-life compared to the half-life of an IgG having the wild-type IgG constant domain. For example, the IgG constant domain can contain one or more amino acid substitutions of amino acid residues at positions 251-257, 285-290, 308-314, 385-389, and 428- 436, wherein the amino acid position numbering is according to the EU index as set forth in Kabat. In certain aspects the IgG constant domain can contain one or more of a substitution of the amino acid at Kabat position 252 with Tyrosine (Y), Phenylalanine (F), Tryptophan (W), or Threonine (T), a substitution of the amino acid at Kabat position 254 with Threonine (T), a substitution of the amino acid at Kabat position 256 with Serine (S), Arginine (R), Glutamine (Q), Glutamic acid (E), Aspartic acid (D), or Threonine (T), a substitution of the amino acid at Kabat position 257 with Leucine (L), a substitution of the amino acid at Kabat position 309 with Proline (P), a substitution of the amino acid at Kabat position 311 with Serine (S), a substitution of the amino acid at Kabat position 428 with Threonine (T), Leucine (L), Phenylalanine (F), or Serine (S), a substitution of the amino acid at Kabat position 433 with Arginine (R), Serine (S), Isoleucine (I), Proline (P), or Glutamine (Q), or a substitution of the amino acid at Kabat position 434 with Tryptophan (W), Methionine (M), Serine (S), Histidine (H), Phenylalanine (F), or Tyrosine. More specifically, the IgG constant domain can contain amino acid substitutions relative to a wild-type human IgG constant domain including as substitution of the amino acid at Kabat position 252 with Tyrosine (Y), a substitution of the amino acid at Kabat position 254 with Threonine (T), and a substitution of the amino acid at Kabat position 256 with Glutamic acid (E). In some embodiments, the binding molecule is of the IgGl subtype, and optionally comprises the triple mutant YTE, as disclosed supra in the Definitions section.
[00100] An FcgRIIA binding molecule provided herein can include a light chain constant region or fragment thereof. In certain aspects the light chain constant region is a kappa constant region or a lambda constant region, e.g ., a human kappa constant region or a human lambda constant region.
[00101] FcgRIIA binding molecules provided herein can have beneficial properties. For example, the binding molecule can inhibit, suppress, or block various FcgRIIA-mediated activities, e.g. , immune-complex -induced type I interferon expression in plasmacytoid dendritic cells (DCs), immune-complex-induced cytokines/chemokines expression in DCs, immune- complex-induced platelet activation, immune-complex -induced antigen presentation, immune- complex-induced neutrophil extracellular traps (NETs) formation, and degranulation in neutrophil activation. [00102] In certain aspects, the binding molecules provided herein can bind to FcgRIIA with a binding affinity characterized by a dissociation constant (KD) of about 100 nM to about 0.1 nM as measured by a Biacore™ assay or on a Kinetic Exclusion Assay (KinExA) 3000 platform.
[00103] In certain aspects, an anti-FcgRIIA antibody or antigen-binding fragment thereof can specifically bind to FcgRIIA, e.g., human FcgRIIA or cynomolgus monkey FcgRIIA, or an antigenic fragment thereof, with a dissociation constant or KD of less than 10-6 M, of less than 10-7 M, of less than 10-8 M, of less than 10-9 M, of less than 10-10 M, of less than 10-11 M, of less than 10-12 M, of less than 10-13 M, of less than 10-14 M, or of less than 10-15 M as measured, e.g. , by Biacore™ or KinExA®. In a particular aspect, the humanized anti-FcgRIIA antibody
MED 19600 can bind to human FcgRIIA (131R) with a Ko of about 0.15 nM, to human FcgRIIA (131H) with a KD of about 0.13 nM, and to cynomolgus monkey FcgRIIA with a KD of about 31.3 nM, as measured by a BIAcore assay.
[00104] In another embodiment, an FcgRIIA binding molecule of the invention binds to
F cyRIIA or an antigenic fragment thereof with a K0ff of less than 1 x 10-3 s-1 , or less than 2 x 10-3 s-1. In other embodiments, an FcgRIIA binding molecule binds to FcgRIIA or an antigenic fragment thereof with a K0ff of less than 10-3 s-1, less than 5x 10-3 s-1, less than 10-4 s-1, less than 5>< l0_4 s-1, less than 10-5 s-1, less than 5x l0-5 s-1, less than 10-6 s-1, less than 5x 1o-6 s-1, less than less than 5x l0-7 s-1, less than 10-8 s-1, less than 5x l0-8 s-1, less than 10-9 s-1, less than 5x l0-9 s-1, or less than 10-10 s-1 as measured, e.g. , by Biacore™ or KinExA®. In a particular aspect, the humanized anti-FcgRIIA antibody MED 19600 can bind to human FcgRIIA (131R) with a K0ff of about 7.35 x 10^ s-1, to human FcgRIIA (131H) with a K0ff of about 3.37 x 10-4 s-1, and to cynomolgus monkey F cyRIIA with a K0ff of about 9.04 x 10-2 s-1, as measured by a BIAcore assay.
[00105] In another embodiment, an FcgRIIA binding molecule of the invention binds to
FcgRIIA or an antigenic fragment thereof with an association rate constant or Kon rate of at least 105 M-1 s-1, at least 5x l0-5 M-1 s-1, at least 10-6 M-1 s-1, at least 5x 10-6 M-1 s-1, at least 10-7 M-1 s-1, at least 5x lO-7 M-1 s-1, at least 10-8 M-1 s-1, or at least 10-9 M-1 s-1 as measured, e.g. , by Biacore™ or KinExA®. In a particular aspect, the humanized anti-FcgRIIA antibody
MED 19600 can bind to human FcgRIIA (131R) with a Kon of about 4.98 x 106 M-1 s-1, to human FcgRIIA (131H) with a Kon of about 2.60 x 106 M-1 s-1, and to cynomolgus monkey FcgRIIA with a Kon of about 2.88 x 106 M-1 s-1, as measured by a BIAcore assay. [00106] A VH and/or VL amino acid sequence or portion thereof, including a CDR sequence, can be, e.g. , 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% similar to a sequence set forth herein, and/or comprise 0, 1, 2, 3, 4, 5 or more substitutions, e.g., conservative substitutions, relative to a sequence set forth herein, such as a sequence from any of clones 32L00350, 32LO0351, 32LO0352, 32LO0354, 32LO0355, or 32LO0356. An FcyRIIA-binding molecule having VH and VL regions with a certain percent similarity to a VH region or VL region, or having one or more substitutions, e.g, conservative substitutions, can be obtained by
mutagenesis (e.g, site-directed or PCR-mediated mutagenesis) of nucleic acid molecules encoding VH and/or VL regions described herein, followed by testing of the encoded altered binding molecule for binding to FcgRIIA, and optionally testing for retained function using the functional assays described herein.
[00107] The disclosure further provides an FcgRIIA binding molecule that is conjugated to a heterologous agent. In certain aspects, the agent can be an antimicrobial agent, a therapeutic agent, a prodrug, a peptide, a protein, an enzyme, a lipid, a biological response modifier, a pharmaceutical agent, a lymphokine, a heterologous antibody or fragment thereof, a detectable label, a polyethylene glycol (PEG), or a combination of two or more of any said agents.
Heteroconjugate anti -FcgRIIA antibodies are discussed in more detail elsewhere herein.
[00108] The term“binding molecule” includes antibodies and antigen-binding fragments thereof. In certain embodiments, the FcgRIIA-binding molecule is a polypeptide that is not an antibody. A variety of methods for identifying and producing non-antibody polypeptides that bind with high affinity to a protein target are known in the art. See, e.g, Skerra, Curr. Opin. Biotechnol. 18:295-304 (2007), Hosse et al, Protein Science 15:14-27 (2006), Gill et al, Curr. Opin. Biotechnol. 17:653-658 (2006), Nygren, FEBS J. 275:2668-76 (2008), and Skerra, FEBSJ. 275:2677-83 (2008). In certain embodiments, phage display technology can been used to identify and/or produce an FcgRIIA-binding polypeptide. In certain embodiments, the polypeptide comprises a protein scaffold of a type selected from the group consisting of protein A, a lipocalin, a fibronectin domain, an ankyrin consensus repeat domain, and thioredoxin.
III. Preparation of FcgRIIA-Binding Molecules
[00109] Monoclonal anti-FcgRIIA antibodies can be prepared using hybridoma methods, such as those described by Kohler and Milstein Nature 256:495 (1975). Using the hybridoma method, a mouse, hamster, or other appropriate host animal, is immunized to elicit the production by lymphocytes of antibodies that will specifically bind to an immunizing antigen. Lymphocytes can also be immunized in vitro. Following immunization, the lymphocytes are isolated and fused with a suitable myeloma cell line using, for example, polyethylene glycol (PEG), to form hybridoma cells that can then be selected away from unfused lymphocytes and myeloma cells. Hybridomas that produce monoclonal antibodies directed specifically against a chosen antigen as determined by immunoprecipitation, immunoblotting, or by an in vitro binding assay (e.g. RIA or ELISA) can then be propagated either in in vitro culture using standard methods (Goding, Monoclonal Antibodies: Principles and Practice , Academic Press, 1986) or in vivo as ascites tumors in an animal. The monoclonal antibodies can then be purified from the culture medium or ascites fluid.
[00110] FcgRIIA-binding molecules can also be made using recombinant DNA methods, for example, as described in U.S. Patent No. 4,816,567. In some instances, the polynucleotides encoding a monoclonal antibody are isolated from mature B-cells or hybridoma cell, such as by RT-PCR using oligonucleotide primers that specifically amplify the genes encoding the heavy and light chains of the antibody, and their sequence is determined using conventional procedures. The isolated polynucleotides encoding the heavy and light chains or antigen-binding fragments thereof are then cloned into suitable expression vectors, which when transfected into host cells such as E. coli cells, simian COS cells, Chinese hamster ovary (CHO) cells, or myeloma cells that do not otherwise produce immunoglobulin protein, binding molecules are generated by the host cells. Also, recombinant FcgRIIA-binding molecules can be isolated from phage display libraries expressing CDRs of the desired species, as described by McCafferty et al. (Nature, 348:552-554 (1990)); Clackson et al. (Nature, 352:624-628 (1991)); and Marks et al. (J. Mol. Biol., 222:581-597 (1991)). Production and expression of nucleic acids comprising nucleotide sequences encoding FcgRIIA-binding molecules are discussed in more detail in the next section.
[00111] The polynucleotide(s) encoding a binding molecule can further be modified in a number of different manners using recombinant DNA technology to generate alternative binding molecules. In some embodiments, the constant domains of the light and heavy chains of, for example, a mouse monoclonal antibody can be substituted (1) for those regions of, for example, a human antibody to generate a chimeric antibody or (2) for a non-immunoglobulin polypeptide to generate a fusion antibody. In some embodiments, the constant regions are truncated or removed to generate the desired antibody fragment of a monoclonal antibody. Site-directed or high-density mutagenesis of the variable region can be used to optimize specificity, affinity, etc. of a monoclonal antibody.
[00112] In certain embodiments, the FcgRIIA-binding molecule is a human antibody or antigen binding fragment thereof. Human antibodies can be directly prepared using various techniques known in the art. Immortalized human B lymphocytes immunized in vitro or isolated from an immunized individual that produce an antibody directed against a target antigen can be generated {See, e.g. , Cole el al. , Monoclonal Antibodies and Cancer Therapy , Alan R. Liss, p. 77 (1985); Boemer et al., J Immunol. 147 (l):86-95 (1991); and U.S. Patent No. 5,750,373).
[00113] The FcgRIIA-binding molecule can be selected from a phage library, where the phage library expresses human antibodies, as described, for example, by Vaughan et al. {Nat.
Biotechnol. , 14:309-314 (1996)), Sheets et al. {Proc. Nat’l. Acad. Sci. U.S. A. 95:6157-6162 (1998)), Hoogenboom et al. {J. Mol. Biol. 227:381 (1991)), and Marks et al. {J. Mol. Biol.
222:581 (1991)). Techniques for the generation and use of antibody phage libraries are also described in U.S. Patent Nos. 5,969,108, 6,172,197, 5,885,793, 6,521,404; 6,544,731; 6,555,313; 6,582,915; 6,593,081; 6,300,064; 6,653,068; 6,706,484; and 7,264,963; and in Rothe et al., j. Mol. Biol. 375: 1182-1200 (2007).
[00114] Affinity maturation strategies and chain shuffling strategies are known in the art and can be employed to generate high affinity human antibodies or antigen-binding fragments thereof. {See Marks et al., Bio/Technology 10:779-783 (1992)).
[00115] In some embodiments, the FcgRIIA-binding molecule can be a humanized antibody or antigen-binding fragment thereof. Methods for engineering, humanizing, or resurfacing non human or human antibodies can also be used and are well known in the art. A humanized, resurfaced, or similarly engineered antibody can have one or more amino acid residues from a source that is non-human, e.g, mouse, rat, rabbit, non-human primate, or other mammal. These non-human amino acid residues are replaced by residues that are often referred to as“import” residues, which are typically taken from an“import” variable, constant, or other domain of a known human sequence. Such imported sequences can be used to reduce immunogenicity or reduce, enhance, or modify binding, affinity, on-rate, off-rate, avidity, specificity, half-life, or any other suitable characteristic, as known in the art. In general, the CDR residues are directly and most substantially involved in influencing FcgRIIA binding. Accordingly, part or all of the non-human or human CDR sequences are maintained while the non-human sequences of the variable and constant regions can be replaced with human or other amino acids.
[00116] Antibodies can also optionally be humanized, resurfaced, engineered, or human antibodies engineered with retention of high affinity for FcgRIIA and other favorable biological properties. To achieve this goal, humanized (or human) or engineered anti-FcgRIIA antibodies and resurfaced antibodies can be optionally prepared by a process of analyzing the parental sequences and various conceptual humanized and engineered products, using three-dimensional models of the parental, engineered, and humanized sequences. Three-dimensional
immunoglobulin models are commonly available and are familiar to those skilled in the art. Computer programs are available which illustrate and display probable three-dimensional conformational structures of selected candidate immunoglobulin sequences. Inspection of these displays permits analysis of the likely role of the residues in the functioning of the candidate binding molecule sequence, i.e., the analysis of residues that influence the ability of the candidate binding molecule to bind its target, such as FcgRIIA In this way, framework (FW) residues can be selected and combined from the consensus and import sequences so that the desired binding molecule characteristic, such as increased affinity for the target, is achieved.
[00117] Humanization, resurfacing, or engineering of anti-FcgRIIA antibodies or antigen binding fragments thereof can be performed using any known method, such as, but not limited to, those described in, Jones et al. , Nature 321 :522 (1986); Riechmann et al. , Nature 332:323 (1988); Verhoeyen et al, Science 239: 1534 (1988)), Sims et al, J Immunol. 151 : 2296 (1993); Chothia and Lesk, J. Mol. Biol. 196:901 (1987), Carter et al ., Proc. Natl. Acad. Sci. U.S.A.
89:4285 (1992); Presta et al., J. Immunol. 151 :2623 (1993), U.S. Pat. Nos. 5,639,641, 5,723,323; 5,976,862; 5,824,514; 5,817,483; 5,814,476; 5,763,192; 5,723,323; 5,766,886; 5,714,352;
6,204,023; 6,180,370; 5,693,762; 5,530,101; 5,585,089; 5,225,539; 4,816,567, 7,557,189;
7,538,195; and 7,342,110; International Application Nos. PCT/US98/16280; PCT/US96/18978; PCT/US91/09630; PCT/US91/05939; PCT/US94/01234; PCT/GB89/01334; PCT/GB91/01134;
PCT/GB92/01755; International Patent Application Publication Nos. W090/14443;
W090/14424; W090/14430; and European Patent Publication No. EP 229246.
[00118] Anti-FcgRIIA humanized antibodies and antigen-binding fragments thereof can also be made in transgenic mice containing human immunoglobulin loci that are capable, upon immunization, of producing the full repertoire of human antibodies in the absence of endogenous immunoglobulin production. This approach is described in U.S. Patent Nos. 5,545,807;
5,545,806; 5,569,825; 5,625,126; 5,633,425; and 5,661,016.
[00119] In certain embodiments an anti-FcgRIIA antibody fragment is provided. Various techniques are known for the production of antibody fragments. Traditionally, these fragments are derived via proteolytic digestion of intact antibodies. See , e.g. , Morimoto et al ., ./. Biochem. Biophys. Meth. 24: 107-117 (1993); Brennan et al ., Science , 229:81-83 (1985). In certain embodiments, anti-FcgRIIA antibody fragments are produced recombinantly. Fab, Fv, and scFv antibody fragments can all be expressed in and secreted from E. coli or other host cells, thus allowing the production of large amounts of these fragments. Such anti-FcgRIIA antibody fragments can also be isolated from the antibody phage libraries discussed above. Anti-FcgRIIA antibody fragments can also be linear antibodies, as described in U.S. Patent No. 5,641,870. Other techniques for the production of antibody fragments will be apparent to the skilled practitioner.
[00120] According to the present invention, techniques can be adapted for the production of single-chain antibodies specific to FcgRIIA (see, e.g., U.S. Pat. No. 4,946,778). In addition, methods can be adapted for the construction of Fab expression libraries (see, e.g. , Huse et al, Science 246:1275-1281 (1989)) to allow rapid and effective identification of monoclonal Fab fragments with the desired specificity for FcgRIIA Antibody fragments can also be produced by techniques in the art including, but not limited to: (a) a F(ab’)2 fragment produced by pepsin digestion of an antibody molecule; (b) a Fab fragment generated by reducing the disulfide bridges of an F(ab’)2 fragment, (c) a Fab fragment generated by the treatment of the antibody molecule with papain and a reducing agent, and (d) Fv fragments.
[00121] In some aspects, FcgRIIA-binding molecule can be modified in order to reduce or eliminate effector function. This can be achieved, for example, by the triple mutation (TM) L234F/L235E/P331S in the Fc domain of IgGl. Other mutations that reduce effector function are known in the art. See, e.g., Armour et al, Eur. J. Immunol. 29:2613-2624, 1999; Shields et al, J. Biol. Chem. 276:6591-6604, 2001.
[00122] In certain aspects, an FcgRIIA-binding molecule can be modified in order to increase its serum half-life. This can be achieved, for example, by incorporation of a salvage receptor binding epitope into the binding molecule by mutation of the appropriate region, or by incorporating the epitope into a peptide tag that is then fused to the binding molecule at either end or in the middle ( e.g ., by DNA or peptide synthesis), or by YTE mutation. Other methods to increase the serum half-life of an antibody or antigen-binding fragment thereof, e.g., conjugation to a heterologous molecule such as PEG, are known in the art.
[00123] Heteroconjugate FcgRIIA antibodies and antigen-binding fragments thereof are also within the scope of the present invention. Heteroconjugate antibodies are composed of two covalently joined antibodies. Such antibodies have, for example, been proposed to target immune cells to unwanted cells (see, e.g, ET.S. Pat. No. 4,676,980). It is contemplated that heteroconjugate anti-FcgRIIA antibodies and antigen-binding fragments thereof can be prepared in vitro using known methods in synthetic protein chemistry, including those involving crosslinking agents. For example, immunotoxins can be constructed using a disulfide exchange reaction or by forming a thioether bond. Examples of suitable reagents for this purpose include iminothiolate and methyl-4-mercaptobutyrimidate.
[00124] An FcgRIIA-binding molecule can be modified to contain additional chemical moieties not normally part of the protein. Such moieties can improve the characteristics of the binding molecule, for example, solubility, biological half-life, or absorption. The moieties can also reduce or eliminate any undesirable side effects of the binding molecule. An overview of those moieties can be found in Remington’s Pharmaceutical Sciences, 20th ed., Mack Publishing Co., Easton, PA (2000).
[00125] In certain aspects, the disclosure provides a composition, e.g, a pharmaceutical composition, comprising an FcgRIIA-binding molecule of the invention, optionally further comprising one or more carriers, diluents, excipients, or other additives.
IV. Polynucleotides Encoding FcgRIIA-Binding Molecules, Preparation and Expression Thereof
[00126] This disclosure provides polynucleotides comprising nucleic acid sequences that encode an FcgRIIA-binding molecule, e.g, a polypeptide that specifically binds FcgRIIA For example, the invention provides a polynucleotide comprising a nucleic acid sequence that encodes an anti-FcgRIIA antibody or encodes an antigen-binding fragment of such an antibody. The polynucleotides of the invention can be in the form of RNA or in the form of DNA. DNA includes cDNA, genomic DNA, and synthetic DNA; and can be double-stranded or single- stranded, and, if single stranded, can be the coding strand or non-coding (anti-sense) strand.
[00127] In certain embodiments, the polynucleotide can be isolated. In certain embodiments, the polynucleotide can be substantially pure. In certain embodiments, the polynucleotide can be cDNA or are derived from cDNA. In certain embodiments, the polynucleotide can be recombinantly produced. In certain embodiments, the polynucleotide can comprise the coding sequence for a mature polypeptide, fused in the same reading frame to a polynucleotide which aids, for example, in expression and optionally, secretion, of a polypeptide from a host cell ( e.g ., a promoter or other regulatory sequence, a leader sequence that functions as a secretory sequence for controlling transport of a polypeptide from the cell). The polypeptide having a leader sequence is a pre-protein and can have the leader sequence cleaved by the host cell to form the mature form of the polypeptide. The polynucleotide can also encode an FcgRIIA-binding pro- protein which is the mature protein plus additional 5’ amino acid residues.
[00128] The disclosure provides an isolated polynucleotide comprising a nucleic acid encoding an FcgRIIA-binding molecule comprising an amino acid sequence from a VH and/or VL domain having 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% similarity to an amino acid sequence set forth herein, and/or comprising 0, 1, 2, 3, 4, 5 or more amino acid substitutions, e.g.,
conservative substitutions, relative to an amino acid sequence set forth herein, such as a sequence from any of clones 32L00350, 32LO0351, 32LO0352, 32LO0354, 32LO0355, or 32LO0356.
[00129] In certain embodiments the polynucleotide that comprises the coding sequence for the FcgRIIA-binding molecule is fused in the same reading frame as a marker sequence that allows, for example, for purification of the encoded polypeptide. For example, the marker sequence can be a hexa-histidine tag supplied by a pQE-9 vector to provide for purification of the mature polypeptide fused to the marker in the case of a bacterial host, or the marker sequence can be a hemagglutinin (HA) tag derived from the influenza hemagglutinin protein when a mammalian host (e.g, COS-7 cells) is used.
[00130] Polynucleotide variants are also provided. Polynucleotide variants can contain alterations in the coding regions, non-coding regions, or both. In some embodiments, polynucleotide variants contain alterations that produce silent substitutions, additions, or deletions, but do not alter the properties or activities of the encoded polypeptide. In some embodiments, polynucleotide variants are produced by silent substitutions due to the degeneracy of the genetic code. Polynucleotide variants can be produced for a variety of reasons, e.g, to optimize codon expression for a particular host (change codons in the human mRNA to those preferred by a bacterial host such as E. coli). [00131] The invention includes vectors comprising the polynucleotides described above.
Suitable vectors are described elsewhere herein, and are known to those of ordinary skill in the art. In some embodiments, a polynucleotide comprising a nucleic acid encoding a VH domain or portion thereof and the polynucleotide comprising a nucleic acid encoding a VL domain or portion thereof can reside in a single vector, or can be on separate vectors. Accordingly, the disclosure provides one or more vectors comprising the polynucleotides described above.
[00132] In certain aspects, the disclosure provides a composition, e.g ., a pharmaceutical composition, comprising a polynucleotide or vector as described above, optionally further comprising one or more carriers, diluents, excipients, or other additives.
[00133] The disclosure further provides a host cell comprising a polynucleotide or vector of the invention, wherein the host cell can, in some instances, express a binding molecule that specifically binds to FcyRIIA. Such a host cell can be utilized in a method of making an
FcgRIIA-binding molecule, where the method includes (a) culturing the host cell and (b) isolating the binding molecule from the host cell or from the culture medium, if the binding molecule is secreted by the host cell.
[00134] In some embodiments a nucleotide sequence encoding an FcgRIIA-binding molecule, can be constructed by chemical synthesis using an oligonucleotide synthesizer. Such
oligonucleotides can be designed based on the amino acid sequence of the desired polypeptide and selecting those codons that are favored in the host cell in which the recombinant polypeptide of interest will be produced. Standard methods can be applied to synthesize an isolated polynucleotide sequence encoding an isolated polypeptide of interest. For example, a complete amino acid sequence can be used to construct a back-translated gene. Further, a nucleotide oligomer containing a nucleotide sequence coding for the particular isolated polypeptide can be synthesized. For example, several small oligonucleotides coding for portions of the desired polypeptide can be synthesized and then ligated. The individual oligonucleotides typically contain 5’ or 3’ overhangs for complementary assembly.
[00135] Once assembled (by synthesis, site-directed mutagenesis, or another method), the polynucleotide sequences encoding the binding molecule can be inserted into an expression vector and operatively linked to an expression control sequence appropriate for expression of the binding molecule in a desired host. Proper assembly can be confirmed, e.g. , by nucleotide sequencing, restriction mapping, and/or expression of a biologically active polypeptide in a suitable host. In order to obtain high expression levels of a transfected gene in a host, the gene can be operatively linked to or associated with transcriptional and translational expression control sequences that are functional in the chosen expression host.
[00136] In certain embodiments, recombinant expression vectors are used to amplify and express DNA encoding FcgRIIA-binding molecules. Recombinant expression vectors are replicable DNA constructs that have synthetic or cDNA-derived DNA fragments encoding a polypeptide chain of an FcgRIIA-binding molecule, operatively linked to suitable transcriptional or translational regulatory elements derived from mammalian, microbial, viral or insect genes. A transcriptional unit generally comprises an assembly of (1) a genetic element or elements having a regulatory role in gene expression, for example, transcriptional promoters or enhancers, (2) a structural or coding sequence which is transcribed into mRNA and translated into protein, and (3) appropriate transcription and translation initiation and termination sequences, as described in detail below. Such regulatory elements can include an operator sequence to control
transcription. The ability to replicate in a host, usually conferred by an origin of replication, and a selection gene to facilitate recognition of transformants can additionally be incorporated. DNA regions are operatively linked when they are functionally related to each other. For example, DNA for a signal peptide (secretory leader) is operatively linked to DNA for a polypeptide if it is expressed as a precursor which participates in the secretion of the polypeptide; a promoter is operatively linked to a coding sequence if it controls the transcription of the sequence; or a ribosome binding site is operatively linked to a coding sequence if it is positioned so as to permit translation. Structural elements intended for use in yeast expression systems include a leader sequence enabling extracellular secretion of translated protein by a host cell. Alternatively, where a recombinant protein is expressed without a leader or transport sequence, the protein can include an N-terminal methionine residue. This residue can optionally be subsequently cleaved from the expressed recombinant protein to provide a final product.
[00137] The choice of expression control sequence and expression vector will depend upon the choice of host. A wide variety of expression host/vector combinations can be employed. Useful expression vectors for eukaryotic hosts, include, for example, vectors comprising expression control sequences from SV40, bovine papilloma virus, adenovirus, and cytomegalovirus. Useful expression vectors for bacterial hosts include known bacterial plasmids, such as plasmids from E. coli , including pCR 1, pBR322, pMB9 and their derivatives, wider host range plasmids, such as Ml 3, and filamentous single-stranded DNA phages.
[00138] Suitable host cells for expression of an FcgRIIA-binding molecule include prokaryotes, yeast, insect, or higher eukaryotic cells under the control of appropriate promoters. Prokaryotes include gram negative or gram positive organisms, for example E. coli or bacilli. Higher eukaryotic cells include established cell lines of mammalian origin as described below. Cell-free translation systems could also be employed. Additional information regarding methods of protein production, including antibody production, can be found in, e.g ., U.S. Patent Publication No. 2008/0187954, U.S. Patent Nos. 6,413,746 and 6,660,501, and International Patent
Publication No. WO 04009823.
[00139] Various mammalian or insect cell culture systems can be advantageously employed to express recombinant FcgRIIA-binding molecules. Expression of recombinant proteins in mammalian cells can be performed because such proteins are generally correctly folded, appropriately modified, and completely functional. Examples of suitable mammalian host cell lines include HEK-293 and HEK-293T, the COS-7 lines of monkey kidney cells, described by Gluzman ( Cell 23: 175, (1981)), and other cell lines including, for example, L cells, C127, 3T3, Chinese hamster ovary (CHO), HeLa, and BHK cell lines. Mammalian expression vectors can comprise non-transcribed elements, such as an origin of replication, a suitable promoter and enhancer linked to the gene to be expressed, and other 5’ or 3’ flanking non-transcribed sequences, and 5’ or 3’ non-translated sequences, such as necessary ribosome binding sites, a polyadenylation site, splice donor and acceptor sites, and transcriptional termination sequences. Baculovirus systems for production of heterologous proteins in insect cells are reviewed by Luckow and Summers ( BioTechnology 6:47 (1988)).
[00140] FcgRIIA-binding molecules produced by a transformed host can be purified according to any suitable method. Such standard methods include chromatography (e.g, ion exchange, affinity, and sizing column chromatography), centrifugation, differential solubility, or by any other standard technique for protein purification. Affinity tags such as hexahistidine, maltose binding domain, influenza coat sequence, and glutathione-S-transferase can be attached to the protein to allow easy purification by passage over an appropriate affinity column. Isolated proteins can also be physically characterized using such techniques as proteolysis, nuclear magnetic resonance and x-ray crystallography. [00141] For example, supernatants from systems that secrete recombinant protein into culture media can be first concentrated using a commercially available protein concentration filter, for example, an Amicon or Millipore Pellicon ultrafiltration unit. Following the concentration step, the concentrate can be applied to a suitable purification matrix. Alternatively, an anion exchange resin can be employed, for example, a matrix or substrate having pendant diethylaminoethyl (DEAE) groups. The matrices can be acrylamide, agarose, dextran, cellulose, or other types commonly employed in protein purification. Alternatively, a cation exchange step can be employed. Suitable cation exchangers include various insoluble matrices comprising sulfopropyl or carboxymethyl groups. Finally, one or more reversed-phase high performance liquid chromatography (RP-HPLC) steps employing hydrophobic RP-HPLC media, e.g, silica gel having pendant methyl or other aliphatic groups, can be employed to further purify an FcgRIIA- binding molecule. Some or all of the foregoing purification steps, in various combinations, can also be employed to provide a homogeneous recombinant protein.
[00142] A recombinant FcgRIIA-binding molecule produced in bacterial culture can be isolated, for example, by initial extraction from cell pellets, followed by one or more concentration, salting-out, aqueous ion exchange, or size exclusion chromatography steps. High performance liquid chromatography (HPLC) can be employed for final purification steps. Microbial cells employed in expression of a recombinant protein can be disrupted by any convenient method, including freeze-thaw cycling, sonication, mechanical disruption, or use of cell lysing agents.
[00143] Methods known in the art for purifying antibodies and other proteins also include, for example, those described in ET.S. Patent Publication Nos. 2008/0312425, 2008/0177048, and 2009/0187005.
V. Treatment Methods Rising FcgRIIA-Binding Molecules
[00144] Methods are provided for the use of FcgRIIA binding molecules to treat patients having a disease or disorder associated with inappropriate FcgRIIA activation, such as disease or disorders characterized by immune-complex deposition, immune-complex mediated NETosis, ANCA-induced FcgRIIA activation, and anti-platelet antibody-triggered FcgRIIA activation.
The following discussion refers to diagnostic methods and methods of treatment of various diseases and disorders with an FcgRIIA-binding molecule that is capable of specifically binding FcgRIIA and antagonizing FcgRIIA activity. [00145] In one embodiment, treatment or prevention includes the application or administration of an FcgRIIA binding molecule or a composition comprising FcgRIIA binding molecule to a subject or patient, or application or administration of the FcgRIIA binding molecule to an isolated tissue or cell line from a subject or patient, where the subject or patient has a disease, a symptom of a disease, or a predisposition toward a disease. The composition is preferably a pharmaceutical composition.
[00146] FcgRIIA binding molecules provided herein are useful for the treatment or prevention of certain inflammatory, immune-mediated, or autoimmune diseases or disorders. Examples of inflammatory, immune-mediated, or autoimmune disease or disorders include, but are not limited to vasculitis, e.g ., Anti-neutrophil cytoplasm antibodies (ANCA), ANCA-associated vasculitis (AAV) or giant cell arteritis (GCA) vasculitis, Sjogren’s syndrome, inflammatory bowel disease (IBD), pemphigus vulgaris, lupus nephritis, psoriasis, thyroiditis, Type I Diabetes, immune thrombocytopenia (FTP), ankylosing spondylitis, multiple sclerosis, systemic lupus
erythematosus (SLE), rheumatoid arthritis, Crohn’s disease, Myasthenia Gravis, neuromyelitis optica (NMO), systemic sclerosis, insulin-dependent diabetes mellitus (IDDM), akylosing spondylitis, atopic dermatitis, uveitis, Graft-versus-host disease (GVHD), polymyositis, dermatomyositis, membranous nephropathy, hemolytic anemia, mixed connective tissue disease, sclerodema, sepsis, thrombosis, acute kidney injury, acute lung injury, chronic obstructive pulmonary disease, glomerulonephritis, toxic liver injury, stroke, atherogenesis, IgG mediated hypersensitive reaction, anti-drug immune complex mediated adverse effects, and other autoantibody or immune-complex-mediated disorders.
[00147] Clinical response to administration of an FcgRIIA-binding molecule can be assessed using screening techniques such as magnetic resonance imaging (MRI), x-radiographic imaging, computed tomographic (CT) scan, flow cytometry or fluorescence-activated cell sorter (FACS) analysis, histology, gross pathology, and blood chemistry, including but not limited to changes detectable by ELISA, ELISPOT, RIA, chromatography, and the like. Further, the subject undergoing therapy with the FcgRIIA-binding molecule can experience improvement in the symptoms associated with the disease or disorder.
[00148] Methods of preparing and administering FcgRIIA-binding molecules to a subject in need thereof are well-known to or can be readily determined by those skilled in the art. The route of administration of the FcgRIIA binding molecule can be, for example, oral, parenteral, by inhalation, or topical. The term“parenteral” as used herein includes, e.g, intravenous, intraarterial, intraperitoneal, intramuscular, subcutaneous, rectal, and vaginal administration.
Oral dosage forms include, e.g. , capsules, tablets, aqueous suspensions, and solutions. Nasal aerosol or inhalation dosage forms can be prepared, for example, as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, and/or other conventional solubilizing or dispersing agents.
[00149] Usually, a suitable pharmaceutical composition can comprise a buffer, optionally a surfactant, optionally a stabilizer agent, etc. The form and character of the pharmaceutically acceptable carrier or diluent can be dictated by the amount of active ingredient with which it is to be combined, the route of administration and other well-known variables. A cocktail comprising one or more species of FcyRIIA binding molecules, e.g. , anti-FcgRIIA antibodies, or antigen binding fragments, variants, or derivatives thereof, of the invention can also be used. In other methods, FcgRIIA-binding molecules can be delivered directly to the site of the adverse cellular population, thereby increasing the exposure of the diseased tissue to the therapeutic agent. In one embodiment, the administration is directly to the airway, e.g. , by inhalation or intranasal administration.
[00150] As discussed herein, FcgRIIA binding molecules can be administered in a
therapeutically effective amount for the in vivo treatment of F cy R 11 A -m edi ated diseases such as inflammatory, immune-mediated, or autoimmune diseases or disorders. In this regard, it will be appreciated that the disclosed binding molecules can be formulated so as to facilitate
administration and promote stability of the active agent. Pharmaceutical compositions in accordance with the present invention can comprise a pharmaceutically acceptable, non-toxic, sterile carrier such as physiological saline, non-toxic buffers, preservatives and the like. For the purposes of the instant application, a“therapeutically effective amount” of an FcgRIIA-binding molecule means an amount sufficient to achieve a benefit, e.g. , to ameliorate symptoms of a disease or condition or to detect a substance or a cell. Suitable formulations for use in the therapeutic methods disclosed herein are described in Remington’s Pharmaceutical Sciences , 20th ed., Mack Publishing Co., Easton, PA (2000).
[00151] The composition can be administered as a single dose, multiple doses, or over an established period of time in an infusion. Dosage regimens also can be adjusted to provide the optimum desired response (e.g, a therapeutic or prophylactic response). The amount of an FcgRIIA-binding molecule that can be combined with carrier materials to produce a dosage form will vary depending upon many different factors, including means of administration, target site, physiological state of the patient (z.e., the severity of the disease, the history of the disease, and the age, height, weight, health, and physical condition of the individual undergoing therapy), whether treatment is prophylactic or therapeutic, other medications administered, and whether the patient is human or an animal. Usually, the patient is a human, but non-human mammals, including transgenic mammals can also be treated. The amount of FcgRIIA-binding molecule to be administered is can be determined by one of ordinary skill in the art. Treatment dosages can be titrated using routine methods known to those of skill in the art to optimize safety and efficacy.
[00152] This disclosure also provides for the use of an FcgRIIA-binding molecule as described herein to treat or prevent an inflammatory, immune-mediated, or autoimmune disease or disorder, e.g ., vasculitis, such as ANCA or GCA vasculitis, immune thrombocytopenia, systemic lupus erythematosus, lupus nephritis, Sjogren’s syndrome, rheumatoid arthritis, Crohn’s disease, Myasthenia Gravis, GVHD, ADA-mediated adverse effects, NETosis, and NETosis-associated disorders, including sepsis, thrombosis, acute kidney injury, acute lung injury, chronic obstructive pulmonary disease, glomerulonephritis, toxic liver injury, stroke, atherogenesis, Type I diabetes, and IgG mediated hypersensitive reaction.
[00153] This disclosure also provides for the use of an FcgRIIA-binding molecule as described herein in the manufacture of a medicament for treating or preventing an inflammatory, immune- mediated, or autoimmune disease or disorder, e.g. , vasculitis, such as ANCA or GCA vasculitis, immune thrombocytopenia, systemic lupus erythematosus, lupus nephritis, Sjogren’s syndrome, rheumatoid arthritis, Crohn’s disease, Myasthenia Gravis, GVHD, ADA-mediated adverse effects, NETosis, and NETosis-associated disorders, including sepsis, thrombosis, acute kidney injury, acute lung injury, chronic obstructive pulmonary disease, glomerulonephritis, toxic liver injury, stroke, atherogenesis, Type I diabetes, and IgG mediated hypersensitive reaction.
VI. Assays and Diagnostics
[00154] FcgRIIA-binding molecules of the invention can be used for diagnosis of FcgRIIA- mediated diseases such as certain inflammatory, immune-mediated, or autoimmune diseases or disorders, and/or for diagnostic monitoring of protein levels as part of a clinical testing procedure, e.g, to determine the efficacy of a given treatment regimen. Such methods typically involve using the FcgRIIA-binding molecules described herein to assay the expression level FcgRIIA. By“assay the expression level of FcgRIIA” is intended to mean qualitatively or quantitatively measuring or estimating the level of FcgRIIA in a first biological sample either directly (e.g, by determining or estimating absolute protein level) or relatively (e.g, by comparing to the disease associated polypeptide level in a second biological sample). The FcgRIIA expression level in the first biological sample can be measured or estimated and compared to a standard FcgRIIA level, the standard being taken from a second biological sample obtained from an individual not having the disorder, or being determined by averaging levels from a population of individuals not having the disorder. In some aspects, an increase in the protein level of the test sample compared to the standard sample is indicative of a disease or disorder treatable by an FcgRIIA-binding molecule of the invention. As will be appreciated in the art, once the“standard” FcgRIIA level is known, it can be used repeatedly as a standard for comparison.
[00155] By“biological sample” is intended any biological sample obtained from an individual, cell line, tissue culture, or other source of cells potentially expressing FcgRIIA. Methods for obtaining tissue biopsies and body fluids from mammals are known in the art.
[00156] The FcgRIIA-binding molecules of the invention can be used to assay FcyRIIA protein levels in a biological sample using classical immunohistological methods known to those of skill in the art (e.g., see Jalkanen, el al., J. Cell. Biol. 707:976-985 (1985); Jalkanen el al., J. Cell Biol. 105:3087-3096 (1987)) Immunoassays that can be used include but are not limited to competitive and non-competitive assay systems using techniques such as Western blots, radioimmunoassays, ELISA (enzyme linked immunosorbent assay), ELISPOT,“sandwich” immunoassays, immunoprecipitation assays, precipitin reactions, gel diffusion precipitin reactions, immunodiffusion assays, agglutination assays, complement-fixation assays, immunoradiometric assays, fluorescent immunoassays, protein A immunoassays, and immunoelectron microscopy, to name some examples. Such assays are routine and well known in the art. Those skilled in the art will be able to determine operative and optimal assay conditions for each determination by employing routine experimentation.
[00157] Detection of FcgRIIA can be facilitated by coupling the binding molecule to a detectable substance or label. Examples of detectable substances include various enzymes, prosthetic groups, fluorescent materials, luminescent materials, bioluminescent materials, and radioactive materials. Examples of suitable enzymes include horseradish peroxidase, alkaline phosphatase, b-galactosidase, or acetylcholinesterase; examples of suitable prosthetic group complexes include streptavidin/biotin and avidin/biotin; examples of suitable fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine,
dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin. An example of a luminescent material is luminol. Examples of bioluminescent materials include luciferase, luciferin, and aequorin. Examples of suitable radioactive material include 125I, 1311, 35S, or 3H.
[00158] In situ detection can be accomplished by removing a histological specimen, for example a blood sample, from a patient, and applying thereto a labeled FcgRIIA-binding molecule, applied by overlaying the labeled FcgRIIA-binding molecule onto a biological sample. Through the use of such a procedure, it is possible to determine not only the presence of
FcgRIIA, but also its distribution in the examined tissue. Rising the present invention, those of ordinary skill will readily perceive that any of a wide variety of histological methods (such as staining procedures) can be modified in order to achieve such in situ detection.
[00159] Methods and reagents suitable for determination of binding characteristics of an isolated FcgRIIA-binding molecule are known in the art and/or are commercially available. Equipment and software designed for such kinetic analyses are commercially available ( e.g ., BIAcore®, BIAevaluation® software, GE Healthcare; KINEXA® Software, Sapidyne
Instruments).
VII. Kits comprising FcgRIIA-binding Molecules
[00160] This disclosure further provides kits that comprise an FcgRIIA-binding molecule, which can be used to perform the methods described herein. In certain embodiments, a kit comprises at least one purified FcgRIIA-binding molecule in one or more containers. In some embodiments, the kits contain all of the components necessary and/or sufficient to perform a detection assay, including all controls, directions for performing assays, and any necessary software for analysis and presentation of results. One skilled in the art will readily recognize that the disclosed FcgRIIA-binding molecules can be readily incorporated into one of the established kit formats which are well known in the art. [00161] All of the references cited in this disclosure are hereby incorporated by reference in their entireties. In addition, any manufacturers’ instructions or catalogues for any products cited or mentioned herein are incorporated by reference. Documents incorporated by reference into this text, or any teachings therein, can be used in the practice of the present invention.
Documents incorporated by reference into this text are not admitted to be prior art.
[00162] The invention is further described in the following non-limiting Examples.
EXAMPLES
[00163] Embodiments of the present disclosure can be further defined by reference to the following non-limiting examples, which describe in detail preparation of certain antibodies of the present disclosure and methods for using antibodies of the present disclosure. It will be apparent to those skilled in the art that many modifications, both to materials and methods, can be practiced without departing from the scope of the present disclosure.
Example 1. Humanization and Optimization of Anti-FcyRIIA Murine Monoclonal
Antibody
Cloning, Expression, Purification of FcgRIIA and FcyRIIB from Human and Cynomolgus Monkey
[00164] cDNA molecules encoding the extracellular domains of CD32A, CD32B from human were synthesized with primer extension PCR cloning using database sequences as references (see Table 2). Sequences were cloned into the pEBNA mammalian expression vector (Thermo Fisher, Cat. No. A10898) fused to a 6xHis_Flag tag at the C-terminus (HHHHHHDYKDDDDK) (SEQ ID NO: 18). Proteins were expressed in FreeStyle 293F cells (Thermo Fisher Cat. No. R790-07). Expressed protein in supernatant was purified using Ni-NTA affinity chromatography (Histrap HP column (GE Healthcare, Cat. No. 17-5248-02)) followed by Size Exclusion chromatography (Superdex 200 column (GE Healthcare, Cat. No. 17-1069-01)).
TABLE 2
Figure imgf000044_0001
FcgRIIA Consensus Sequence in Cynomolgus Monkey ( Macaca fascicularis)
[00165] There are 36 amino acids in cynomolgus FcgRIIA that are non-homologous to human FcgRIIA (UniProt P12318) resulting in 88% sequence identity (aa 1-317). Table 3 shows non- homologous amino acids.
TABLE 3
Figure imgf000045_0001
Figure imgf000046_0001
Variations where the minor cynomolgus allele corresponds to human are shown in bold type.
[00166] An additional 7 coding polymorphisms were identified among cynomolgus populations with a minor allele frequency (MAF) of >20%. Table 4 shows single nucleotide polymorphism variation.
TABLE 4
Figure imgf000046_0002
Figure imgf000047_0001
MAF > 20% shown in bold type; minor alleles corresponding to the major human allele shown in italics.
[00167] Consensus cynomolgus sequences and alignments to human FcyllA are shown in
FIGS. 1A-1C.
Cynomolgus CD32A Proteins
[00168] Three versions of cynomologus CD32A were made based on polymorphisms around the epitope of IV3 (Ramsland et. al. , ./. Immunol. 187:3208-3217 (2011)). These proteins were expressed in HEK cells by GeneArt and purified by Ni-NTA affinity chromatography utilizing a c-terminal 6xhis tag.
Figure imgf000048_0001
Protein Modifications
[00169] IgGs and modified proteins used herein were biotinylated via free amines using EZ link Sulfo-NHS-LC -Biotin (Thermo/Pierce, Cat. No. 21335). The biotin reagent was dissolved in anhydrous dimethylformamide and PBS based protein solutions were adjusted to pH ~8 with 1 M NaHCCb in D-PBS (Dulbecco’s phosphate buffered saline). CD32 proteins used herein were biotinylated via free cysteines using EZ link Biotin-BMCC (Perbio/Pierce, Product No. 21900). The biotin reagent was dissolved in anhydrous dimethylformamide and mixed 3 : 1 with D-PBS protein solutions. Label incorporations were assessed by MALDI-TOF mass
spectrometry in all cases and unreacted reagents were cleared by buffer exchange using D-PBS equilibrated disposable Sephadex G25 columns. For biotinylation, the final protein
concentrations were determined by 280 nm absorbance using extinction coefficients calculated from amino acid sequences.
Cloning and Expression of IgG Molecules
[00170] Variable domains were converted to whole immunoglobulin Gl (IgGi) antibody format essentially as described by Persic et al. {Gene 187(1):9-18 (1997)) with the following
modifications. An OriP fragment was included in the expression vectors to facilitate use with CHO-transient cells and to allow episomal replication. The variable heavy (VH) domain was cloned into a vector containing the human heavy chain constant domains and regulatory elements to express whole IgGi heavy chain in mammalian cells. The heavy chain constant domains contained three mutations shown to significantly reduce Fc effector function (Oganesyan et al ., Acta Crystallographica Section D: Biological Crystallography 64(6): 700 (2008)), to avoid engagement of CD32A via the Fc portion of full-length antibody. Similarly, the variable light (VL) domain was cloned into a vector for the expression of the human light chain (kappa) constant domains and regulatory elements to express whole IgG light chain in mammalian cells. To obtain IgGs, the heavy and light chain IgG-expressing vectors were transfected into CHO- transient mammalian cells (Daramola et al. , Biotechnol Prog. 30(1): 132-41 (2014)). IgGs were expressed and secreted into the medium. Harvests were filtered prior to purification, then IgG was purified using Protein A chromatography. Culture supernatants were loaded on a column of appropriate size of Ceramic Protein A (BioSepra) and washed with 50 mM Tris-HCl pH 8.0, 250 mM NaCl. Bound IgG was eluted from the column using 0.1 M Sodium Citrate (pH 3.0) and neutralized by the addition of Tris-HCl (pH 9.0). The eluted material was buffer exchanged into PBS using NaplO columns (Amersham Cat. No. 17-0854-02) and the concentration of IgG was determined spectrophotometrically using an extinction coefficient based on the amino acid sequence of the IgG (Mach et al. , Anal. Biochem. 200(l):74-80 (1992)). The purified IgGs were analyzed for aggregation and degradation purity using SEC-HPLC and by SDS-PAGE.
Humanization of IV.3 Ab
[00171] Mouse monoclonal antibody IV.3 was first described in 1986 (Looney RJ. et al., J. Immunol. 136(5): 1641). The hybridoma cell line was obtained from ATCC and the heavy chain variable (VH) and light chain variable (VL) segments of IV.3 were sequenced (SEQ ID NO: 5 and SEQ ID NO: 8).
[00172] Humanization of the variable domains of mouse mAb IV.3 was performed by grafting the heavy and light chain complementarity determining regions (CDRs) of IV.3 onto selected human germline frameworks (FWs). The amino acid sequences of the VH and VL domains of IV.3 were aligned to the known human germline sequences in the IMGT database (Lefranc, M.
P. et al, Nucl. Acids Res. 37(Database issue): D1006-D1012 (2009)), and appropriate human germlines were identified by sequence similarity, including matching of critical residues (Vernier zone, canonical class residues, and VH/VL interface residues), immunogenicity (germline frequency), stability, and expression. For the VH domain of the IV.3 antibody, the chosen human germline was IGHVl-3*0l/IGHJ4. For the VL domain, it was IGKV2-28*0l/IGKJ2.
[00173] A series of chimeric variable heavy and light chains were designed with one or more mouse FW regions replaced with the equivalent selected human FW. All CDR and FW regions were as defined by Rabat. Fully mouse and chimeric IV.3 variable regions were codon optimized for CHO expression and synthesized by GeneArt® Gene Synthesis (Life
Technologies). These were subsequently expressed as whole immunoglobulin Gl (IgGi) antibodies as described above to generate a panel of partially and wholly humanized IV.3 variants. Antibodies were characterized in an epitope competition assay as described in Example 11. Briefly, homogenous time-resolved fluorescence (HTRF®, Cisbio International) was used to detect binding of IV.3 IgG (Stemcell Technologies, Cat. No. 01470) to biotinylated human CD32A 131H. Fluorescence resonance energy transfer (FRET) between IV.3 and CD32A was detected using DyLight 649 conjugated anti-mouse antibody and streptavidin Terbium cryptate (Cisbio International, Product No. 610SATLB), respectively. The signal is disrupted by the addition of sample IgGs that compete with IV.3 for CD32A binding. Full humanization of both VH and VL chains of IV.3 was associated with a lO-fold loss of potency in this assay. A mouse residue was introduced back into the human light chain germline framework, using standard molecular biology techniques, which restored potency to within 5-fold of the mouse IgG (mutation Y36F). Data obtained is exemplified in FIG. 2. Subsequently, a potential deamination site was removed in VL CDR1 (N28L) to reduce the risk of chemical modification during manufacture. An alignment of the parental VH and VL chains (IV.3), the final humanized VH and VL chains (CamIV3) and the selected human germline sequences is shown in FIG. 3.
Optimization of CamIV3 Ab
[00174] Single amino acid substitutions were made at select positions of VH CDR2 and VL CDR1 of CamIV.3 using standard molecular biology techniques. Variants were expressed directly as IgG and supernatants quantified and screened in an IV.3 epitope competition assay as described in Example 11. Beneficial individual mutations in VH CDR2 and VL CDR1 were combined and IgGs expressed and screened. Six optimized CamIV.3 variants were chosen for further analysis, 32L00350, 32LO0351, 32LO0352, 32LO0354, 32LO0355 and 32LO0356. NIP228 IgGlTM was used as a negative control. The activity of these variants in the IV.3 epitope competition assay is shown in FIG. 4. The optimized variants differed from CamIV3 only in HCDR2 and LCDR2 sequence, as shown in Table 5.
TABLE 5
Figure imgf000051_0001
Figure imgf000052_0001
Amino acid differences from IV.3 are in bold type.
The CDR sequences common to IV.3, CamIV3, 32L00350, 32LO0351, 32LO0352
(MED 19600), 32LO0354, 32LO0355, and 32LO0356 are shown in Table 6. (See also FIG. 3.)
TABLE 6
Figure imgf000052_0002
The amino acid sequences of the VH and VL domains of the clones 32L00350, 32LO0351, 32LO0352, 32LO0354, 32LO0355, and 32LO0356 are shown in Table 7.
TABLE 7
Figure imgf000052_0003
Figure imgf000053_0001
Affinity of IV.3 and 32LO0352 for Human and Cynomolgus CD32A
[00175] The affinity for IV.3 and 32LO0352 was measured by Biacore™ analysis. In brief, human IgGl-TM (chimeric IV.3 or 32LO0352) was captured on a Cl-Protein G chip. Serial dilutions (25 nM to 0.3906 nM in log2 steps) of human CD32A_l3 l Arg, human CD32A_l3 lH or cyno CD32A (version 3) were used as the analyte. The data were fitted to a 1 : 1 Langmuir dissociation model and shown in Table 8 (average data from two independent experiments).
TABLE 8
Figure imgf000054_0001
Example 2. Optimized IV.3 Abs Specific for Human FcyRIIA (CD32A) Binding and
Lacking in Effector Function
[00176] To examine the specificity of the humanized, optimized IV.3 Abs, the binding of antibodies to recombinant FcyRI, FcgRIIA- 13 1 H/H allotype, FcyRIIB, FcyRIII - 158F allotype, or FcyRIII - 158V allotype was assessed by ELISA. R347 Tm Ab was used as human IgGl isotype control, and the antibodies H2B6-Tm, 3G8, 16-115 were used as positive controls for FcyRIIB, FcyRIII, FcgRIIA, and FcyRI, respectively. These four humanized, optimized IV.3 clones, 32L00350, 32LO0352, 32LO0354, and 32LO0355, exhibited high binding specificity for human FcgRIIA (FIG. 5A) but not for other FcyRs (FIG. 5B-5E). These data demonstrate that the humanized, optimized antibody variants retain their specificity for FcgRIIA Importantly, despite the similarity in the extracellular domains of FcgRIIA and FcyRIIB, these antibodies bind FcgRIIA but not FcyRIIB in this assay.
[00177] To determine whether the triple mutation (TM) (L234F/L235E/P331 S) incorporated in MED 19600’s heavy chain constant region reduced MED 19600’ s effector functions, ADCC and CDC assays were performed. In the assays, the effects of MED 19600 were compared with wild type control 9600 IgGl (identical Fab as MED 19600, but with wild-type IgG Fc) and isotype control IgG (R347-Tm) antibodies. In the ADCC assay, primary NK cells (effectors) were incubated with adherent FcgRIIA-expressing HEK-293 cells (targets) for 5 h, and % cytotoxicity was determined. For CDC assays, baby rabbit complement was incubated with adherent
FcgRIIA-expressing HEK-293 cells (targets) and % cytotoxicity was determined after 1 h. No notable ADCC (FIG. 5F) or CDC (FIG. 5G) activity was detected with MEDI9600, whereas the 9600 IgGl control antibody induced cytotoxicity of FcgRIIA-expressing HEK cells. Therefore the TM incorporated in MED 19600’s heavy chain constant region renders MEDI9600 effector null.
Example 3. MEDI9600 (32LO0352) binding competition with intravenous
immunoglobulin
[00178] To determine whether MED 19600 has a competitive or non-competitive mode of action, a binding competition assay was conducted with IgG. The binding of MEDI9600 to human neutrophils expressing FcgRIIA in the presence and absence of intravenous
immunoglobulin (IVIG), which mimics the physiological concentration of circulating
immunoglobulin in whole blood, was assessed by flow cytometry. As shown in FIG.6,
MED 19600 bound to human FcgRIIA-expressing neutrophils in a dose-dependent manner. The EC50 value of MEDI9600 binding to human FcgRIIA at the neutrophil surface was 0.03 nM
(FIG. 6). In the presence of 10 mg/mL IVIG, the EC50 value of MED 19600 binding to human FcgRIIA at the neutrophil surface was increased to 3.34 nM (FIG. 6), suggesting that MEDI9600 competes with immunoglobulin for binding to FcgRIIA. No binding was observed with the human IgGl isotype R347-TM control (FIG. 6). These data indicate that MEDI9600 is a ligand- blocking antibody.
Example 4. Optimized IV.3 Abs Internalize Human and Cynomolgus Monkey FcyRIIA in Whole Blood Assay
[00179] The murine IV.3 Ab has been shown to internalize FcgRIIA and subsequently degrade the receptor in the lysosome13. Consequently, removal of FcgRIIA from the surface of the cell upon antibody binding can be used as a measure of receptor internalization. We verified this mode of action and assessed the potency of the anti-FcgRIIA antibody variants in a whole blood internalization assay. This assay format was used to mimic the cellular composition and the levels of competing immunoglobulin found under physiological conditions. It has previously been shown that two common human polymorphic variants of FcgRIIA, 131H and 131R, differentially bind IgG subclasses14. Consequently, the capacity of the antibodies to internalize FcgRIIA from donors homozygous for 131H and 131R was also assessed. To facilitate pharmacology and toxicology assessment, the capacity of the antibodies to internalize FcyRII from monocytes and neutrophils was also assessed in whole blood from cynomolgus monkey. [00180] The chimeric and optimized antibodies internalized FcgRIIA from the surface of monocytes from human 131H/H donors (FIG. 7A) and human 131R/R donors (FIG. 7B). The optimized IV.3 Abs exhibited improved potency from two- to five-fold compared to the parent IV.3 chimeric Ab for both the 131H/H and 131R/R donors. The ECso for the optimized antibodies ranged from 0.04 nM to 0.09 nM compared to 0.19 nM for the chimeric IV.3 antibody for the 131H/H donor, and 0.04 nM to 0.06 nM compared to 0.20 nM for the chimeric IV.3 antibody for the 131 R/R donors (FIG. 7A, 7B). The optimized antibodies also internalized FcgRIIA on the surface of monocytes from cynomolgous monkeys (FIG. 7C). The potency of the optimized antibodies was up to seven-fold greater than the chimeric IV.3 Ab. Taken together, these data demonstrate that the changes introduced into the Fab regions of the heavy and light chains in the optimized variants of the IV.3 antibody are beneficial. The FcgRIIA binding specificity has been retained, murine residues have been removed, and key changes have enhanced the potency of the antibody above and beyond the parental version of the antibody in a physiologically relevant assay.
[00181] In an additional experiment, MEDI9600-mediated receptor internalization was confirmed by confocal microscopy. At time 0, both CD14 (as a negative control) and FcgRIIA showed cell surface staining. However, after a 1 hour incubation at 37°C, almost complete FcgRIIA internalization was stimulated by binding of MED 19600, while CD 14 remained on the cell surface (FIG. 7D).
[00182] Similar to the results with the human and cynomologous monkey monocytes,
MED 19600 mediated internalization of FcgRIIA from the surface of neutrophils from human donors having the 131H/H (Fig. 7E) or 131R/R genotype (Fig. 7F), and from the suface of neutrophils from cynomolgus monkey whole blood (Fig.7G). The neutrophils from human (131R/R or 131H/H phenotypes) and cyomolgus monkey whole blood were examined following a 12 hour incubation with MED 19600 or control Ab (R347-TM) by flow cytometry (M.F.I.). Together these data demonstrate that MED 19600 reduces monocyte and neutrophil cell surface expression of FcgRIIA available for ligand engagement.
Example 5. Optimized IV.3 Abs Block RNP-IC-Induced Inflammatory Responses from
Human and Cynomolgus Monkey PBMC [00183] We assessed the capacity of the anti- FcgRIIA antibodies to inhibit Ribonucleoprotein- Immune complex (RNP-IC)-mediated inflammatory responses. The activity of the antibodies was examined with cells from healthy donors with either a 131H/H or 131R/R haplotype, and with cells from cynomolgus monkeys. The optimized IV.3 Abs exhibited up to a ~ 2 fold increase in potency in the RNP-IC -induced type I IFNa assay compared to the parental IV.3 antibody using cells from 131 H/H and 131R/R donors (FIG. 8A, 8B). The potency of the optimized antibodies were ~ 3-8 fold greater than the humanized CamIV.3 antibody with the 131 H/H donor (FIG. 8A), and ~ 5-12 fold greater than CamIV.3 for the 131 R/R donor (FIG. 8B). Three of the four optimized antibodies also exhibited greater potency than the parental and humanized versions of IV.3 in the RNP-IC-induced IFNa assay using cynomolgus monkey cells (FIG. 8C).
[00184] The increased potency of these optimized human anti- FcgRIIA antibodies to block immune-complex mediated induction of type I IFN is consistent with improvements in the antibody-mediated FcgRIIA uptake in the whole blood internalization assay. Since IFNa appears to play an important role in the pathogenesis of SLE, these data support the use of these humanized, optimized anti-FcgRIIA antibodies for the treatment of SLE.
[00185] Furthermore, immune complexes can also trigger monocytes to produce TNFa and IL- 6, key cytokines involved in the pathogenesis of RA [21] Compared to control antibody,
MED 19600 (30 pg/mL) inhibited the immune complex induction of IL-6 and TNFa in whole blood by approximately 60% and 80% respectively (Fig. 8D; mean ± standard deviation percentage inhibition relative to no antibody presented). The demonstration that MEDI9600 can inhibit IC-mediated induction of TNFa and IL-6 among other proinflammatory molecules would suggest that MED 19600 may provide an alternative option for the treatment of RA. Treatments targeting the TNFa or IL-6 pathways have been approved for RA highlighting the importance of these cytokines in the pathology of the disease.
Example 6. Optimized IV.3 Ab, Clone 32LO0352 (MEDI9600), Specifically Blocks Anti- Neutrophil Cytoplasmic Antibody (ANCA) Induced Neutrophil Activation
[00186] A marker of neutrophil activation is the release of reactive oxygen species, which can measured in a ferri-cytochrome C reduction assay, or by flow cytometry based DH123 assay15. The ability of optimized IV.3 Ab clone 32LO0352 (MEDI9600) to inhibit ANCA induced neutrophil activation was first examined in ferri-cytochrome C reduction assay. MED 19600 specially blocked ANCA- (both Anti-PR3 Ab and anti-MPO Ab) induced neutrophil production of reactive oxygen species (FIG. 9A, 9B). To determine if MEDI9600 could non-specifically impact neutrophil activation, neutrophils were treated with PMA in the presence and absence of the FcgRIIA antibody. PMA induced reactive oxygen species, but inhibition of FcgRIIA with MED 19600 had no effect on PMA-induced neutrophil activation (FIG. 9C). These data demonstrate that MED 19600 specifically blocks antibody-mediated induction of neutrophil activation, whereas other mechanisms of neutrophil activation may not be impeded by this treatment.
[00187] The ability of MEDI9600 to block ANCA-induced neutrophil activation was also assessed by a more sensitive flow cytometry based DHR123 assay. Rising this assay format, MED 19600 again inhibited anti-MPO and anti-PR3 Ab-induced neutrophil activation in a dose dependent manner, whereas the isotype control antibody had no effect (FIG. 9D, 9E). Both of the previous experiments used commercially available anti-MPO and anti-PR3 Abs as stimuli.
To verify the ability of optimized IV.3 Ab to inhibit ANCA-induced neutrophil activation, IgG was purified from AAV patients’ sera with autoantibodies against either PR3 or MPO, and used as stimuli for neutrophils. Using the flow cytometry-based DHR123 assay, MED 19600 significantly blocked neutrophil activation triggered by AAV patient IgG (FIG. 9F, 9G).
[00188] Taken together, these data indicate that MEDI9600 anti-FcgRIIA Ab is able to specifically block anti-neutrophil cytoplasmic antibody (ANCA) induced neutrophil activation, and supports the use of the humanized, optimized MEDI9600 anti-FcgRIIA antibody for the treatment of ANCA-associated vasculitis.
Example 7. Optimized IV.3 Ab (MEDI9600) Protects Mice from Anti-Platelet Antibody- Induced Thrombocytopenia
[00189] Using the transgenic human FcgRIIA murine-activating FcyR-deficient mice, the efficacy of the optimized IV.3 Ab (MED 19600) to block anti -platelet antibody induced thrombocytopenia was examined. Treatment of the FcgRIIA transgenic mice with anti-platelet antibody caused a rapid depletion of platelets (FIG. 10A). Prophylactic dosing with the optimized IV.3 Ab (MEDI9600) significantly blocked platelet clearance in this model (FIG.
10B, 10C). Importantly, the inhibition of platelet depletion with MEDI9600 was associated with the internalization of FcgRIIA on the platelets (FIG. 10D). This internalization of FcgRIIA was not only observed on the platelets, but also on neutrophils (FIG. 10E), and it was dose- dependent over a 4-day period. With 10 mg/kg of MEDI9600, no free FcgRIIA was detected on the platelets (FIG. 10D) or neutrophils (FIG. 10E) through 48 h, and this observation was consistent with the presence of circulating MED 19600 at that time (FIG. 10F).
[00190] A summary of MEDI9600 inhibition of anti -platelet (anti-CD4l Ab) induced thrombocytopenia in FcgRIIA transgenic mice is provided at FIG. 10G. Bars in the graph show platelet numbers in the transgenic mice prophylactically treated with MEDI9600 or control antibody prior to induction of thrombocytopenia. The MED 19600 or control antibody (R347- Tm) was injected i.p. at lOmg/ml 24hrs before treatment, thrombocytopenia was induced with 2 pg rat anti-mouse CD4lAb delivered i.p. at day 0. Preincubation with 10 mg/kg of MED 19600 prevented anti-platelet mediated thrombocytopenia compared to the control antibody where there was -80% reduction in platelet numbers (FIG. 10G).
[00191] Taken together, these data indicate that humanized anti -FcgRIIA antibody, MED 19600, inhibits anti-platelet antibody-induced thrombocytopenia in vivo , and support the use of this antibody for the treatment of immune thrombocytopenia.
Example 8. Blockade of FcyRIIA by MEDI9600 Has No Adverse Effects on Neutrophil
Function
[00192] Neutrophils play a critical role in host defense by sensing infection and tissue injury, and initiating an acute inflammatory response, which serves to recruit leukocytes, clear infections, engage the adaptive immune system, and promote repair. Upon exposure to an insult, neutrophils rapidly migrate through the blood vessels to the site of the tissue injury, following chemical signals derived from pathogens and damaged host cells. Neutrophils directly engage pathogens through interactions with pattern recognition receptors, complement receptors, and immunoglobulin receptors, which results in the release of toxic substances that kill pathogens and/or the clearance of the pathogens by phagocytosis. Since neutrophil functions are important in host defense, we assessed the impact of FcgRIIA blockade with MED 19600 on different neutrophil functions.
[00193] First, we examined if blockade of FcgRIIA would affect neutrophil activation in the presence of the synthetic triacylated lipoprotein Pam3Sk4, which is a toll-like receptor 2 (TLR2) agonist and mimic of the acylated amino terminus of bacterial lipoproteins. An anti-TLR2 Ab inhibited the Pam3Sk4-induced neutrophil activation, whereas the MEDI9600 anti-FcgRIIA antibody and the isotype control IgG antibody had no effect on TLR2 -mediated neutrophil activation (FIG. 11 A).
[00194] Next, we examined if blockade of FcgRIIA would affect IL-8-induced neutrophil migration. IL-8-induced neutrophil migration was not impacted by MEDI9600 or the control antibody (FIG. 11B).
[00195] Finally, we examined if MEDI9600 affects anti-Psl mAb-mediated opsonophagocytic killing (OPK) of Pseudomonas aeruginosa. Anti-Psl mAbs were previously shown to mediate potent complement dependent killing of P. aeruginosa in the presence of neutrophils16. A luminescent/1 aeruginosa strain was used in this assay to assess bacterial killing. The level of luminescence correlates with the frequency of live bacteria. Neutrophils were pre-incubated with MED 19600, anti-FcyRIIB Ab, or anti-FcyRIII Ab, and then they were incubated with luminescent bacteria for 2 hours. MEDI9600 had a minimal effect on the ability of neutrophils to kill P. aeruginosa in the presence of anti-Psl mAb Psl0096, whereas blockade of FcyRIII clearly inhibited OPK activity (FIG. 11C). These data indicate that antibody-mediated phagocytosis of a clinically resistant strain of bacteria is largely independent of FcyllA, and blockade of this receptor is not predicted to adversely impact phagocytosis of bacteria by neutrophils.
[00196] Taken together, these data indicate that FcgRIIA blockade will not adversely impact normal neutrophil functions such as chemotaxis, activation induced by pathogen associated molecular patterns, and phagocytosis.
Example 9. MEDI9600 Has No Impact on Protein Induction from Cells in Whole Blood
[00197] Adverse effects of antibody treatments, such as cytokine storms, can be detected by examining the protein profile of whole blood. To assess the impact of MEDI9600, the gene expression profiles of unstimulated whole blood, and whole blood stimulated by MEDI9600, Ig- IC, or RNP-IC were assessed in five normal health donors. The Ig-IC or RNP-IC had a profound impact on the protein expression profile, and as expected, there were no differences in the expression profile between the untreated and MEDI9600-stimulated conditions (FIG. 12A and 12B). These data indicate that MED 19600 does not activate cells in whole blood, and are indicative of a good safety profile. Example 10. MEDI9600: A Single-Dose Pharmacokinetic and Exploratory Pharmacodynamic Study in Cynomolgus Monkeys
[00198] The purpose of this study was to characterize the pharmacokinetic/pharmacodynamic relationship of MED 19600 in cynomolgus monkeys when given as a single dose by intravenous injection. Results of this study were used to inform pharmacokinetic/ pharmacodynamic modeling, which supported administration of MEDI9600 to humans.
[00199] The pharmacokinetics were nonlinear; an inspection of Cmax /Dose, AUC (0- )/Dose, and half-life show a clear trend for these to increase with dose (FIG. 13A). The terminal half- lives obtained from the non-compartmental analysis were shorter than expected for a human IgGl antibody in the cynomolgus monkey, with those for the group given 1 mg/kg only 1.08-1.2 days, increasing to 2.84-4.3 days for the group given 100 mg/kg. However, the tendency for the half-life to increase with dose, along with an inspection of the pharmacokinetic curves, reveals a pharmacokinetic profile consistent with target-mediated elimination of the antibody. For the highest dose of 100 mg/kg, which had less interference from target-mediated elimination, initial volumes of distribution of 0.024-0.052 L/kg were similar to plasma volume, as would be expected for a human IgGl antibody. Pharmacokinetic variability appears low, with coefficient of variation values around 20% or less for the earlier samples. Pharmacokinetics became markedly more variable approaching the terminal phase.
[00200] A single dose of MED 19600 administered at 1, 10, or 100 mg/kg induced a dose- response reduction of FcgRIIA fluorescence intensity on monocytes and granulocytes (FIG.
13B, 13C). This reduction was completely reversible, with a full recovery achieved first in animals given 1 mg/kg, followed by animals given 10 mg/kg, and animals given 100 mg/kg. FcgRIIA expression on monocytes and granulocytes showed a dose-dependent suppression on dosing with MEDI9600, and was similar for monocytes and granulocytes. Suppression was observed by the 30-minute time point, indicating a rapid suppression in dosing. This suppression was only partial for Group 1 (1 mg/kg) but appeared to cause maximum suppression for Groups 2 and 3, judging by the similar suppression achieved by these dose levels at their nadir. FcgRIIA expression levels returned to baseline over a dose-dependent period of days to weeks. It was notable that this rapid suppression and slower recovery mirrored the pharmacokinetic profile of MED 19600. In particular, the recovery to baseline of FcgRIIA expression matched the reduction of pharmacokinetic exposure to concentrations of below 1,000 ng/ml; this occurred at around 7 days post-dose for Group 1, around 14 days post-dose for Group 2, and around 49 days post-dose for Group 3, indicating a strong and direct relationship between pharmacokinetic exposure and pharmacodynamic response in the form of FcgRIIA expression.
[00201] To summarize, male monkeys were given MED 19600 once at a dose level of 1, 10 or 100 mg/kg with a dose volume of 2 mL/kg via intravenous injection. Administration of
MED 19600 resulted in dose-dependent suppression of FcgRIIA expression on monocytes and granulocytes, followed by a slow recovery to baseline levels consistent with the duration of the MED 19600 pharmacokinetic exposure, indicating a strong relationship between pharmacokinetic exposure and pharmacodynamic response.
[00202] In addition, safety end points were evaluated during the study; administration of MED 19600 had no effect on D-dimer concentrations or on clinical observations. A l3-week subcutaneous and intravenous GLP toxicity study indicated that a single weekly IV dose (up to 100 mg/kg) did not result in any changes in clinical chemistry. Furthermore, MEDI9600 treatment did not result in body weight changes (FIG. 14A) or changes in platelet counts (FIG. 14B) after 13 weeks treatment. In addition, no adverse effects on other hematology, clinical chemistry, organ weight, histopathology, qualitative food consumption, or coagulation were noted. MEDI9600-r elated transient erythema and edema at the injection site was considered non-adverse.
Example 11. Materials and Methods
Cynomolgus Monkey FcgRIIA Sequencing
[00203] Primers were designed using the available cynomolgus monkey draft genome sequence derived from a single animal of Vietnamese origin (Beijing Genome Institute, 2011). The Protein Science cynomolgus genomic DNA bank, comprised of 60 individuals (20 Chinese, 20 Vietnamese, 20 Mauritian), was utilized to sequence FcylLA PCR amplification was performed using Qiagen HotStar Taq Master Mix followed by in-house sequencing of the PCR products. Raw sequence alignments, consensus transcript builds, SNP variant identification, and comparison to human FcyllA (CCDS 44264 / ETniProt P12318) were completed using SeqMan (Lasergene) and CloneManager software.
[00204] Sequence variation was confirmed using an internal cynomolgus genome browser, which contains whole exome sequence data from 48 individuals (16 Chinese, 16 Vietnamese, 16 Mauritian). Minor allele frequencies derived from this internal database are shown in Table 4. FcyllA exon arrangement was confirmed using blood-derived cDNA from 6 individuals.
Fey Receptor Binding Assay
[00205] Recombinant FcyRI, FcgRIIA- 13 1 H/H allotype, FcyRI IB, FcyRIII - 158F allotype, and FcyRIII - 158V allotype, were generated at Medlmmune. Anti-FcyRIII antibody (3G8) was purchased from Abeam (Cambridge, MA). Anti- FcyRI Ab (16-115) was purchased from Antibodies-online (Atlanta, GA). Anti- FcyRI IB Ab and isotype control R347-Tm were generated at Medlmmune.
[00206] Costar 96-well microplates (Fisher Scientific, PA) were coated with 2pg/ml of Fey Receptor (FcyRI, FcgRIIA-l3 lH/H allotype, FcyRIIB, FcyRIII - 158F allotype, or FcyRIII - 158V allotype) overnight at 4°C. Plates were washed five times with 200 mΐ of wash buffer (PBS containing 0.1% Tween 20), blocked with blocking buffer containing 5% milk in PBS for 1 hour at room temperature, and washed five additional times with wash buffer. Three-fold serial dilutions of the test antibody or control antibody (R347-Tm) were added to wells in duplicate. Plates were incubated for 2 hours at room temperature, and washed five times with wash buffer. Binding of the antibodies to the FcyRs was detected by adding 50 mΐ of goat anti-human Fc-HRP (Jackson ImmunoResearch Laboratories, PA) to the wells and incubating for 1 hour at room temperature, washing 10 times, and adding 50m1 TMB substrate (Pierce Biotechnology, MA).
The color reaction was stopped with 50m1 0.2M FhsSCL and the absorbance was measured using a spectrophotometer (Molecular Devices, CA) at A405nm.
Epitope competition assay
[00207] To identify IgG improvements during Lead Optimisation, antibody samples were assessed using an epitope competition assay using homogenous time-resolved fluorescence (HTRF®, Cisbio International) in which the binding of IV.3 IgG (Stemcell Technologies, 01470) to biotinylated human CD32a 131H was measured.
[00208] The binding of IV.3 IgG to biotinylated human CD32a 131H was assessed by measuring the FRET signal between a IV.3 and biotinylated human CD32a 131H using DyLight 649 conjugated anti-mouse detection antibody streptavidin Terbium cryptate (Cisbio
International, 610SATLB) detection. The assay was used to identify improvements in crude IgG samples by measuring inhibition of the interaction between IV.3 IgG and biotinylated human CD32a 131H.
[00209] An 8.0 nM solution of IV.3 IgG is prepared in assay buffer consisting of PBS
(Invitrogen 14190-185), 0.2 % BSA (Sigma A9576) and 0.4 M KF (BDH 103444T). 5 mΐ is added to the assay plate (384 black, shallow well, Costar, 3676), to give a final concentration of 2.0 nM. A 40nM solution of DyLight 649 conjugated anti-mouse detection antibody is prepared in assay buffer and 5 mΐ is added to the assay plate to give a final concentration of 10 nM. 5 mΐ of each IgG sample is transferred to the assay plate using a MiniTrak™ (Perkin Elmer). A pre mixed solution containing 4 nM biotinylated human CD32a 131H and 2.67 nM streptavidin Terbium cryptate is prepared as a 4x stock. 5m1 is added to the assay plate to give a final concentration of 1 nM biotinylated human CD32a 131H and 0.67 nM streptavidin Terbium cryptate. Non-specific binding wells (negative controls) were defined for each plate by omitting biotinylated human CD32a 131H. Assay plates were incubated for 3 hours at room temperature prior to reading time resolved fluorescence at 620 nm and 665 nm emission wavelengths using an EnVision plate reader (Perkin Elmer).
[00210] Data was analysed by calculating the 665/620 nm ratio followed by the % Delta F values for each sample.
[00211] The 665/620 nm ratio was used to correct for sample interference using Equation 1.
Equation 1 :
Figure imgf000064_0001
Figure imgf000065_0001
[00212] To confirm improvements, crude or purified IgG samples were assessed in the same assay by serially diluting samples in assay buffer using a Greiner 384 well V bottom plate (Greiner 781280). 5 mΐ of each dilution of scFv was transferred in duplicate to the assay plate (384 black, shallow well, Costar, 3676) using a Bravo™ (Agilent). Assay reagents were then added as described above.
[00213] Data was analysed by calculating the 665/620 nm ratio followed by the % Delta F values as described previously. The 665/620 nm ratio was used to correct for sample interference as described in Equation 1. % Delta F was calculated using Equation 2. %Specific binding was calculated for each condition using Equation 3.
[00214] IC50 values were determined using GraphPad Prism software by curve fitting using a four-parameter logistic equation (Equation 4).
Equation 4:
Y=Bottom + (Top-Bottom)/(l+l0A((LogEC50-X)*HillSlope))
X is the logarithm of concentration. Y is specific binding
Y starts at Bottom and goes to Top with a sigmoid shape.
HTRF binding assay for selectivity and cross-reactivity
[00215] The selectivity of lead antibodies was assessed using a homogenous time-resolved fluorescence assay (HTRF®, Cisbio International) in which the binding of the IgG to biotinylated human CD32b was measured. Cross-reactivity was assessed using a homogenous time-resolved fluorescence assay in which the binding of the IgG to cyno CD32a and CD32b was measured.
[00216] The binding of purified IgG to biotinylated CD32 was assessed by measuring the FRET signal between the IgG and the biotinylated CD32 using streptavidin terbium cryptate (Cisbio International, 610SATLB) and anti-human Fc antibody conjugated with XL665 (Cisbio
International, 61HFCXLB) detection reagents.
[00217] Assay buffer consisting of PBS (Invitrogen 14190-185), 0.2 % BSA (Sigma A9576) and 0.4 M KF (BDH103444T) is prepared and 5m1 added to the assay plate (384 black, shallow well, Costar, 3676). CD32 is diluted as a 4x working stock solution in assay buffer. Human CD32b, cyno CD32a and cyno CD32b are prepared at 20 nM and 5m1 added to the assay plate to give a final concentration of 5 nM. Serial dilutions of IgGs are prepared in duplicate in assay buffer using a Greiner 384 well V bottom plate (Greiner 781280) and 5m1 of each dilution of peptide transferred to the assay plate using a Bravo™ (Agilent). Streptavidin terbium cryptate and anti-human Fc IgG XL665 were diluted to 0.67 nM and 15 nM respectively (final concentrations) in assay buffer and 5m1 of this premixed solution was added to the assay plate. A negative control was defined for each plate by replacing sample IgG with 5m1 assay buffer.
Assay plates were incubated for 3 hours at room temperature prior to reading time resolved fluorescence at 620 nm and 665 nm emission wavelengths using an EnVision plate reader (Perkin Elmer).
[00218] Data was analysed by calculating the 665/620 nm ratio followed by the % Delta F values as described previously (see epitope competition protocol for equations). The 665/620 nm ratio was used to correct for sample interference as described in Equation 1. % Delta F was calculated using Equation 2. %Specific binding was calculated for each condition using
Equation 3.
Octet IgG quantification
[00219] IgG samples were quantified using Protein A Biosensors (ForteBio) in conjunction with the OctetRED384 system (ForteBio).
[00220] 25m1 of sample is mixed with 25m1 assay buffer consisting of PBS, 0.02 % Tween20, 1 mg/ml BSA (0.1%). The rate of binding of IgG in the sample to the protein A biosensor is measured by Bio-Layer Interferometry (BLI). An IgG standard curve is generated using a control IgG of known concentration and preparing a 12-point 1 in 2 dilution series from a top final concentration of 500 pg/ml. The Octet system software calculates the binding rates from standards with known concentrations to generate a standard curve. The binding rate is proportional to the standard concentration. [00221] Data is analysed using Data Analysis software package provided with the OctetRED384 system. Data is uploaded into the software and analysis performed using initial slope and the Dose Response - 4PL (Default; Unweighted) equations. Unknown concentrations are calculated from comparison with the standard curve.
Binding competition assay with IVIG
[00222] Human neutrophils were re-suspended in blocking buffer [phosphate-buffered saline (PBS) supplemented with 10% fetal bovine serum] and transferred to round-bottom 96-well culture plates (0.5 xl06 cells/well). Cells were incubated with increasing concentrations of fluorescently labeled MEDI9600 or the R347-Tm control antibody (ranging from 0.0003 to 66.67 nM) for 30 minutes at 4°C in the presence or absence of 10 mg/mL of Intravenous Immunoglobulin (IVIG). After incubation, cells were washed with cold PBS and the mean fluorescence intensity (MFI) of cell-bound antibodies was assessed by flow cytometry using an LSRII (BD;Franklin Lakes, NJ). The ECso for binding was calculated using a non-linear fit equation in GraphPad Prism 6 software.
Whole Blood Internalization Assay
[00223] To demonstrate MEDI9600-mediated FcgRIIA internalization in human monocytes by confocal microscopy, human peripheral blood mononuclear cells (PBMCs) were isolated from whole blood that was collected into CPT tubes. The PBMCs were first stained with CDl4-Alexa 488, and CDl4-positive monocytes were sorted. The cells were then stained with MED 19600- Alexa 647 on ice for 30 min, washed once and incubated at room temperature for confocal microscopy. Cell imaging was performed using a Leica TCS SP5 confocal system consisting of a Leica DMI6000 B inverted microscope (Leica Microsystems). Images were acquired at time- points noted in the figure legends and were analyzed using the LAS AF version 2.2.1 Leica Application Suite software (Leica Microsystems).
[00224] Anti-FcgRIIA-Tm Ab-mediated receptor internalization was examined by a FACS- based assay. Human or cynomolgus monkey blood was collected into heparin tubes. A three fold dilution series of unlabeled anti-FcgRIIA antibody or control antibody was added to 50 mΐ whole blood for 2 hours at 37°C. The whole blood was washed with FACS buffer (5% fetal bovine serum in PBS), and stained with Ab cocktail (CD14-PE, CD20-Pacific Blue, and IV.3- Alexa 647) on ice for 1 hour. After washing, the cells were incubated with 1 ml RBC lysis buffer (Biolegend, CA) at room temperature for 3 minutes, washed and re-suspended in 150 mΐ FACS buffer. FcgRIIA surface expression on human or cynomolgus monkey monocytes was measured on a FACS LSRII flow cytometer (BD Bioscience, CA) and analysis was performed using Flowjo software. Monocytes were gated on the CDl4-positive cells, and neutrophils were gated on the basis of the forward/side scatter characteristics.
Ribonucleoprotein-Immune Complex (RNP-IC)-Induced Type I IFNa Assay
[00225] The impact of FcgRIIA blockade on RNP-IC -induced IFNa expression was examined by ELISA. Briefly, 100 mΐ human or cynomolgous monkey PBMC (5xl06 cell/ml) were incubated in a tissue culture flask for 1 hour. Non-adherent cells were collected, and this monocyte-depleted fraction was incubated with anti- FcgRIIA Abs or its isotype control (R347- Tm Ab) from 300 pg/ml to 0.001 pg/ml with 3-fold dilution at 37°C incubator for 2 hours. Cells were then treated with 100 pl RNP-IC (1 pg/ml RNP (Biomeda)/2% anti-RNP antibody seropositive SLE serum) for 16 hours. Concentrations of IFNa in the cell culture supernatants were determined using a human IFNa ELISA kit (PBL Assay Science, NJ).
Isolation of Human Neutrophils
[00226] Peripheral blood from normal volunteers was drawn into vacutainer tubes containing heparin, and the blood was diluted 1 : 1 in HBSS. Neutrophils were separated by centrifugation on a lymphoprep density gradient. Erythrocytes were removed by RBC lysis buffer (Biolegend, CA). Neutrophils were further enriched using the Human Neutrophil Enrichment Kit (StemCell Technologies, CA).
Detection of Superoxide Production Using the Ferri-Cytochrome C Reduction Assay
[00227] Neutrophil superoxide production induced by anti-neutrophil cytoplasmic antibody (ANCA) or phorbol myri state acetate (PMA) was measured by the ferri-cytochrome C reduction assay. Briefly, isolated human neutrophils were suspended at 4 x 106 cells/ml in HBSS/20mM Hepes/Ca2+ , Mg2+ free buffer, and primed with recombinant 2 ng/ml TNF-a (R&D Systems,
MN) for 15 minutes. To assess the effects of FcyRIIA blockade, neutrophils were pre-incubated with 10 pg/ml MED 19600 for 30 min at room temperature before they were added to the 96 well tissue culture plates. The 96-well tissue culture plates were pre-treated with 100 pl of 0.5% milk in HBSS/20 mM Hepes for 1 hour at room temperature. Plates were washed three times with wash buffer (HBSS/20mM Hepes/Ca2+, Mg2+ free buffer). Fifty microliters of 2x Cytochrome C buffer (0.2 mM Cytochrome C, 1 mM MgCh, 2 mM CaCl2) were added to each well, followed by 10 mI of stimulus (10 pg/ml of anti-PR3 Ab, 30 pg/ml of anti-MPO Ab, at or 20 nM PMA) and 50 mΐ of neutrophils. The plate was immediately put into a pre-warmed plate reader (37 °C), and OD (550nm and 490nm) was measured continuously for 2 hours at 37 °C.
Detection of Superoxide Production Using the Flow Cytometry (DHR 123 Assay)
[00228] Neutrophil superoxide production induced by ANCA was measured by flow cytometry. Neutrophils (lx l05/ml) were resuspended in RPMI 1640 buffer with 1 pg/ml dihydrohodamine 123 (DHR) (Life Technologies, CA). Some cells were pre-incubated with 10 pg/ml MED19600, anti-FcyRIII Ab (Biolegend, CA), or isotype control Ab at room temperature for 30 minutes.
The cells were then incubated for 30 minutes at 37°C with 30 pg/ml anti-MPO Ab (Abeam,
MA), 10 pg/ml anti-PR3 Ab (Abeam, MA), or 80 pg/ml IgG purified from sera of healthy volunteers or AAV patients (Tissue Solutions, UK), pelleted at 200g, and resuspended in ice- cold HBSS at a density of 5xl06 cells/ml. Mean fluorescence intensity (M.F.I) was measured by LSRII flow cytometer (BD Bioscience, CA) and analysis was performed using Flowjo software.
Neutrophil Migration Assay
[00229] Neutrophil migration was assessed using a 96-well Chemo TX system with 5 pm filter (Neuro Probe, MD). Human IL-8 (R&D Systems, MN) was diluted in RPMI 1640 containing 1% BSA to a final concentration of 20 nM and placed in the lower chamber. Cells were washed and suspended in the same medium. Human neutrophils were incubated with 10 pg/ml
MED19600 or isotype control antibody for 30 minutes at 37°C before the cells were added to the upper chamber. Neutrophils were allowed to migrate for 1 hour. Cells that migrated to the lower chamber were enumerated by flow cytometry. The migration index is the ratio of the number of cells that migrate in response to the chemotactic agent versus the number of cells that migrate in its absence.
Neutrophil Activation Assay
[00230] Human blood was collected in EDTA tubes, and 50 pl aliquots of blood were incubated with Anti-TLR2 Ab, MEDI9600, or isotype control Ab at 10 pg/ml for 2 hours at 37°C. Fifty microliters of Pam3CSK4 (Invivogen, CA) were added to the whole blood to a final concentration of 100 ng/ml and incubated for 45 minutes. The cells were washed two times in FACS buffer (3% FBS in PBS), stained with CD1 lb-PE (Biolegend, CA) on ice for 30 minutes, and washed once with FACS buffer. Red blood cells were lysed with lysis/Fix buffer (BD Bioscience, CA). CDllb expression in the neutrophils was measured by flow cytometry.
Antibody-Dependent Cell-Mediated Cytotoxicity Assay
[00231] ADCC assays were performed using a Celigo Imaging Cytometer (Nexelcom,
Lawrence, MA). Adherent HEK-293 cells stably expressing CD32A were stained with Calcein AM dye (Thermo Fisher, Waltham, MA) following manufacturer’s protocol. Cells were then washed 3x in PBS and resuspended in RPMI 1640 media (lacking phenol red) with 3% FBS. Cells were then plated at ~l0,000 cells/well a black-walled 96-well clear bottom plates
(Greiner). Antibodies at various concentrations were then added. Unlabeled NK cells served as effector cells were isolated from healthy donors via the EasySep Human NK Cell Enrichment Kit (StemCell) following manufacturer’s directions. NK cells were added at a ratio of 3: 1 effector: target cells. Cells were then incubated at 37°C in 5% C02. After 5 h, Calcein AM positive cells were quantified in each well. Wells lacking antibody served as the“no specific lysis” control wells. Specific lysis (% cytotoxicity) was calculated as 100 - ((# of Calcein AM positive cells in test well)/ (mean # of Calcein AM cells in no effector cell control wells) c 100). Results shown are representative of 3 independent experiments. Data were graphed and analyzed using
GraphPad Prism 7.0.
Complement-Dependent Cytotoxicity Assays
[00232] Complement-dependent cytotoxicity (CDC) assays were performed using a Celigo Imaging Cytometer (Nexelcom). Adherent HEK-293 cells stably expressing CD32A were stained with Calcein AM dye (Thermo Fisher) following manufacturers protocol. Cells were then washed 3x in PBS and resuspended in RPMI 1640 media (lacking phenol red). Cells were then plated at ~ 10,000 cells/well in black-walled 96-well clear bottom plates (Greiner). Complement (baby rabbit complement; Cedarlane, Burlington, ON) was then added to a final concentration of 8% and cells were incubated at 37°C in 5% C02. After 1 h, Calcein AM positive cells were quantified in each well. Six wells per plate without added antibodies (but with complement) served as the no specific lysis control wells. Specific lysis (% cytotoxicity) was calculated as 100 - ((# of Calcein AM positive cells in test well)/(mean # of Calcein AM cells in no antibody control wells) c 100). Results shown are representative of 3 independent experiments. Data were graphed and analyzed using GraphPad Prism 7.0.
Opsonophagocytic Killing (OPK) Assay
[00233] Luminescent P. aeruginosa were constructed using vector mini-CTX-lac-lux as previously described (DiGiandomenico et al. , J Exp. Med. 209:1273-1287 (2012)). P.
aeruginosa strains were grown to single colony on overnight plates of trypticase soy agar (TSA) at 37°C, followed by inoculation of a single colony into 10 mL Luria Bertani (LB) broth, and grown to an optical density at 650 nm of 0.4 (approximately 5xl08 colony forming units
[CFU]/mL). One milliliter of cells was pelleted by centrifugation at 14,000 x g, followed by suspension in OPK assay buffer, and further diluted 1 :200 (approximately 2.5xl06CFU/mL). Bacteria were kept on ice until preparation of all OPK components was completed.
[00234] Baby rabbit serum (BRS) (CedarLane, Hornby, Ontario, Canada) was used as the complement source for OPK assays. One milliliter of lyophilized BRS was reconstituted with ice-cold distilled water, followed by preparation of a 1 : 10 dilution in OPK assay buffer. Diluted BRS was kept on ice until preparation of all OPK components was completed.
[00235] MEDI9600 (anti- FcgRIIA), H2B (anti- FcyRIIB), or 3G8 (anti- FcyRIII) Ab was added to white flat-bottom 96-well plates (12.5 mΐ per well), followed by the addition of 12.5 mΐ of the primary neutrophil preparation. The final concentration of FcyR mAbs and primary neutrophils was 2 pg/ml and 2xl07 cells/ml, respectively. After 20-minute incubation at room temperature, dilutions of the anti-Psl antibody, diluted complement, and bacteria were added to each well, and the plate was covered with a breathable sealing film. Wells lacking anti-Psl mAb antibody served as assay controls. Plates were incubated for 120 minutes at 37°C with shaking at 250 revolutions per minute (rpm). Following the incubation, relative luciferase units (RLU) were measured using the Perkin Elmer Envision Multi -Label Reader. The amount of luminescence correlates directly with the frequency of viable bacteria remaining in culture. All samples were run in duplicate. Percent killing of P. aeruginosa was calculated using the following formula:
% Killing = l00-([RLU experimental wells/RLU control wells] x 100)
EC50 values were calculated with GraphPad Prism (version 5) using a 4-parameter logistic, nonlinear regression model for curve fitting analysis. Whole Blood Proteomic Assessment
[00236] For Figure 12A: Human blood was collected in heparin tubes. Five milliliters of blood was incubated with anti- FcgRIIA Ab (30 pg/ml), RNP-IC (1 mg/ml RNP /2% anti-RNP antibody seropositive SLE serum), or Ig-IC (100 pg/ml biotinylated-NMGC Ab-Streptavidin, 2: 1 mol: mol ratio) for 16 hours at 37°C. The plasma fraction was collected after centrifugation for 10 minutes at l300g. Over 200 protein analytes were measured using Rules-Based-Medicine Discovery MAP (Austin, TX). Data was generated from five individual donors: two 131H/H donors, two 131 R/R donors, and one 131 H/R donor, which covered all the FcgRIIA
polymorphic variants.
[00237] For Figure 12B: Human blood was collected in heparin tubes. Five milliliters of blood (5 mL) was incubated with the MED 19600 antibody (30 pg/mL), RNP-IC (1 pg/mL RNP /2% anti-RNP antibody seropositive SLE serum; RNP and SLE serum was pre-incubated at room temperature for 1 h to form immune complexes) or IgG-IC (final concentration: 100 pg/mL biotinylated-IgGl [Clone NMGC] antibody-streptavidin, 2: 1 molar ratio; biotinylated-IgGl antibody was pre-incubated with streptavidin at room temperature for 1 h to form immune complexes) for 16 h at 37°C. Both RNP-IC and IgG-IC were used as positive control Fey receptor agonists. The plasma fraction was collected after centrifugation for 10 min at 1300 xg, and over 200 protein analytes were measured using a Multi-Analyte Profiling (MAP) technology platform (Myriad RBM Discovery, Austin, TX). FIG. 12B lists the panel of analytes examined. The data were generated from five individual donors homozygous or heterozygous for the FcgRIIA 131 polymorphism (two 131H/H donors, two 131 R/R donors and one 131 H/R donor). The data were z-score transformed, and heat-maps were generated in R using the heatmap.2 function of the gplots package. The samples were clustered by type, although the protein clustering structure was unsupervised. The average fold change of treated blood compared to the untreated whole blood is listed in FIG. 12B.
Passive Immune Thrombocytopenia Animal Model
[00238] The effect of anti- FcgRIIA Ab on anti-platelet antibody-induced thrombocytopenia was measured in a passive immune thrombocytopenia animal model. Wild-type and FcgRIIA transgenic mice were obtained from The Jackson Laboratory. Rat anti-mouse CD41 (GpIIa)
IgGl Ab and control rat IgGl Ab were purchased from BD Bioscience. For FIG. 10A-C: Wild- type and transgenic mice were injected intraperitoneally with 2pg of anti-CD4l at day 0. Some mice were pretreated 24 hours prior with MEDI9600 at 10 mg/kg. Blood was collected at day 1, day 5, day 8, and day 12 by retro-orbital bleeding, and platelets were counted using the Sysmex hematology analyzer. Platelet FcyRII A expression was determined by flow cytometry. For FIG. 10D-G: MEDI9600 or control Ab (R347-Tm) was injected i.p. 24hrs before treatment. Platelet numbers were assessed using a sysmex counter before the treatment and dayl, day 2 post treatment. For initial dose range finding studies, MEDI9600 was given by i.p at day 0, blood was collected at dayl, day2 and day 4 post injection, FcgRIIA level at platelets and neutrophils were determined by FACS and serum concentration of MEDI9600 was determined by human IgG ELISA.
MED 19600: A Single-Dose Pharmacokinetic and Exploratory Pharmacodynamics Study in the Cynomolgus Monkey
[00239] Male cynomolgus monkeys were assigned to groups and administered MED 19600, as indicated in Table 9. Animals were dosed via intravenous injection.
TABLE 9
Figure imgf000073_0001
IV = Intravenous
a Animals in Group 3 received the test article as supplied by the sponsor (nominal
concentration of 50 mg/mL) with no further adjustment of dose volume.
Quantitative Determination of MEDI9600 in Cynomolgus Monkey Serum ETsing an Enzyme- Linked Immunosorbent Assay (ELISA) (Non-GLP)
[00240] This method utilized an indirect ELISA format to measure the concentration of
MEDI9600 in cynomolgus monkey serum. Standards, controls, and test samples were incubated with sheep anti-human IgG (H+L) that had been immobilized on a microtiter plate. After incubation, unbound material was washed away and MEDI9600 was detected using goat anti- human IgG-(H+L)-HRP conjugate, and visualized with the addition of a tetramethylbenzidine (TMB) substrate solution. The color development was stopped, and the intensity of the color was measured at 450/650 nm.
Pharmacodynamics - Flow Cytometry
[00241] Blood (approximately 1 mL) was collected for flow cytometry via a femoral vein from all animals on Days 12 and 20 of the pre-dose phase; at approximately 0.5 hours, 4 hours, 8 hours, and 24 hours post-dose; once on Days 3, 4, 5, and 8 of the dosing phase; and weekly thereafter (based on Day 1 of the dosing phase). Animals were not fasted unless samples were collected in conjunction with clinical pathology sampling. The anticoagulant was acid-citrate- dextrose. Parameters were expressed as percentages (of total CD45+ white blood cells) and absolute values (cells/pL). FcgRIIA expression levels were described in fluorescence intensity and in calculated molecules of equivalent soluble fluorescence (MESF) values using median and/or geometric mean fluorescence intensity values. The lymphocyte subsets shown in Table 10 were quantitated using flow cytometry.
TABLE 10
Figure imgf000074_0001
Note: All populations were derived from FSC/SSC and/or SSC/CD45 gates.
Results were enumerated as percent relative (% of total CD45+ white blood cells) and absolute (cells/pL) values for each phenotype.
aMedian and/or geometric fluorescence intensity values for C FcgRIIA were also used
Statistical Analysis
[00242] The statistical significance of the difference between two groups was analyzed using unpaired Student’s t-test or non-parametric Mann Whitney test. Statistical significance was ascribed to the data when p < 0.05. REFERENCES Nimmeijahn et al. Fc-gamma receptors as regulators of immune responses. Nat Rev Immunol. 2008 Jan; 8(l):34-47.
Hogarth et al. Fc receptor-targeted therapies for the treatment of inflammation, cancer and beyond. Nat Rev Drug Discov. 2012 Mar 30; 11(4):311-31.
Bruhns et al. Properties of mouse and human IgG receptors and their contribution to disease models. Blood. 2012 Jun 14; H9(24):5640-9.
Schiffenbauer et al. Biomarkers, surrogate markers, and design of clinical trials of new therapies for systemic lupus erythematosus. Arthritis Rheum. 2004 Aug; 50(8):2415-22. Means et al. Human lupus autoantibody-DNA complexes activate DCs through cooperation of CD32 and TLR9. J Clin Invest. 2005 Feb; 115(2):407-17.
Bennett et al. Interferon and granulopoiesis signatures in systemic lupus erythematosus blood. J Exp Med. 2003 Mar 17; 197(6):711-23.
Bave et al. Fc gamma Rlla is expressed on natural IFN-alpha-producing cells
(plasmacytoid dendritic cells) and is required for the IFN-alpha production induced by apoptotic cells combined with lupus IgG. J Immunol. 2003 Sep 15; 171(6):3296-302. Kallenberg et al. Pathogenesis of ANCA-associated vasculitis: new possibilities for intervention. Am J Kidney Dis. 2013 Dec; 62(6): 1176-87.
Porges et al. Anti-neutrophil cytoplasmic antibodies engage and activate human neutrophils via Fc gamma Rlla. J Immunol. 1994 Aug 1; 153(3): 1271-80.
Chen et al. Endocytosis of soluble immune complexes leads to their clearance by FcyRIIIB but induces neutrophil extracellular traps via FcgRIIA in vivo. Blood. 2012 Nov 22; l20(22):442l-3.
Reilly et al. Heparin-induced thrombocytopenia/thrombosis in a transgenic mouse model requires human platelet factor 4 and platelet activation through FcgammaRIIA. Blood. 2001 Oct 15; 98(8):2442-7.
Liu et al. High-dose dexamethasone shifts the balance of stimulatory and inhibitory Fcgamma receptors on monocytes in patients with primary immune thrombocytopenia. Blood. 2011 Feb 10; 1 l7(6):206l-9. Zhang et al. Divergent intracellular sorting of Fc-gamma-RIIA and Fc-gamma-RIIB2. J Biol Chem. 2010 Oct 29; 285(44):34250-8.
Warmerdam et al. A single amino acid in the second Ig-like domain of the human Fey receptor II is critical for human IgG2 binding. J. Immunol. 1991; 27: 1338-1343
Mulder et al. Activation of granulocytes by anti -neutrophil cytoplasmic antibodies (ANCA): a Fc gamma RH-dependent process. Clin Exp Immunol. 1994 Nov; 98(2):270- 8
DiGiandomenico et al. Identification of broadly protective human antibodies to
Pseudomonas aeruginosa exopolysaccharide Psl by phenotypic screening. J Exp Med. 2012 Jul 2; 209(7): 1273-87.
Brinkmann et al. A Neutrophil extracellular traps kill bacteria. Science 2004 Mar 5; 303(5663): l532-5.
Fuchs et al. Neutrophil extracellular trap (NET) impact on deep vein thrombosis.
Arterioscler. Thromb. Vase. Biol. 2012; 201232(8): 1777-1783.
Villanueva et al. Netting neutrophils induce endothelial damage, infiltrate tissues, and expose immune stimulatory molecules in systemic lupus erythematosus. J. Immunol. 2011; l87(l):538-552.
Carmona-Rivera et al. Neutrophil extracellular traps induce endothelial dysfunction in systemic lupus erythematosus through the activation of matrixmetalloproteinase-2. Ann. Rheum. Dis. 2015 Jul; 74(7): 1417-24.
Allam et al. Extracellular histones in tissue injury and inflammation. J. Mol. Med. (Berk) 2014 May; 92(5):465-72.
Wang et al. Increased neutrophil elastase and proteinase 3 and augmented NETosis are closely associated with (3-cell autoimmunity in patients with type 1 diabetes. Diabetes 2014 Dec; 63(l2):4239-48.
Krishna et al. Immunogenicity to Biotherapeutics - The Role of Anti-drug Immune Complexes. Front. Immunol. 2016 Feb 2; 7:21. [00243] The foregoing description of the specific embodiments will so fully reveal the general nature of the invention that others can, by applying knowledge within the skill of the art, readily modify and/or adapt for various applications such specific embodiments, without undue experimentation, without departing from the general concept of the present invention. Therefore, such adaptations and modifications are intended to be within the meaning and range of equivalents of the disclosed embodiments, based on the teaching and guidance presented herein. It is to be understood that the phraseology or terminology herein is for the purpose of description and not of limitation, such that the terminology or phraseology of the present specification is to be interpreted by the skilled artisan in light of the teachings and guidance. The present invention is further described by the following claims.

Claims

1. An isolated binding molecule that specifically binds to FcgRIIA, wherein the binding molecule comprises:
a. an immunoglobulin variable heavy chain complementarity determining region 2 (VH-CDR2) and an immunoglobulin variable light chain complementarity determining region 1 (VL-CDR1) comprising amino acid sequences selected from the group consisting of: (i) SEQ ID NO: 19 and SEQ ID NO: 22; (ii) SEQ ID NO: 19 and SEQ ID NO: 23; (iii) SEQ ID NO: 19 and SEQ ID NO: 24; (iv) SEQ ID NO: 20 and SEQ ID NO: 25; (v) SEQ ID NO: 20 and SEQ ID NO: 23; and (vi) SEQ ID NO: 20 and SEQ ID NO: 26, respectively;
b. an immunoglobulin variable heavy chain complementarity determining region 1 (VH-CDR1) comprising SEQ ID NO: 29;
c. an immunoglobulin variable heavy chain complementarity determining region 3 (VH-CDR3) comprising SEQ ID NO: 30 or SEQ ID NO: 45;
d. an immunoglobulin variable light chain complementarity determining region 2 (VL-CDR2) comprising SEQ ID NO: 31; and
e. an immunoglobulin variable light chain complementarity determining region 3 (VL-CDR3) comprising SEQ ID NO: 32.
2. The binding molecule of claim 1, comprising SEQ ID NO: 19 and SEQ ID NO: 22.
3. The binding molecule of claim 1, comprising a heavy chain variable (VH) region and a light chain variable (VL) region, comprising amino acid sequences selected from the group consisting of: (i) SEQ ID NO: 33 and SEQ ID NO: 34, (ii) SEQ ID NO: 35 and SEQ ID NO: 36, (iii) SEQ ID NO: 37 and SEQ ID NO: 38, (iv) SEQ ID NO: 39 and SEQ ID NO: 40, (v) SEQ ID NO: 41 and SEQ ID NO: 42, and (vi) SEQ ID NO: 43 and SEQ ID NO: 44, respectively.
4. The binding molecule of claim 3, comprising a VH region and a VL region, comprising the amino acid sequences of SEQ ID NO: 33 and SEQ ID NO: 34, respectively.
5. An isolated binding molecule that competes or cross-competes with the binding molecule of any preceding claim.
6. The binding molecule of any preceding claim, which is selected from a murine antibody, a human antibody, a humanized antibody, a chimeric antibody, a monoclonal antibody, a polyclonal antibody, a recombinant antibody, a bi-specific antibody, a multi-specific antibody, and an antigen-binding fragment thereof.
7. The binding molecule of any preceding claim, which is selected from an Fv, an Fab, an F(ab’)2, an Fab’, a dsFv fragment, a single chain Fv (scFV), an sc(Fv)2, a disulfide-linked (dsFv), a diabody, a triabody, a tetrabody, a minibody, or a single chain antibody.
8. The binding molecule of any preceding claim, comprising an immunoglobulin (Ig) heavy chain constant region.
9. The binding molecule of claim 8, wherein the constant region is a human IgG constant region.
10. The binding molecule of claim 9, wherein the constant region comprises amino acid substitutions at Kabat positions 234, 235, and 331, wherein:
a. the amino acid at Kabat position 234 is substituted with Phenylalanine (F), b. the amino acid at Kabat position 235 is substituted with Glutamic acid (E), and c. the amino acid at Kabat position 331 is substituted with Serine (S).
11. The binding molecule of claim 9 or 10, wherein the constant region comprises one or more substitutions of amino acid residues at positions 251-257, 285-290, 308-314, 385-389, and 428-436, relative to a wild-type human IgG constant region, wherein the amino acid position numbering is according to the EU index as set forth in Kabat.
12. The binding molecule of claim 11, wherein the constant region comprises amino acid substitutions at Kabat positions 252, 254, and 256, wherein:
a. the amino acid at Kabat position 252 is substituted with Tyrosine (Y), b. the amino acid at Kabat position 254 is substituted with Threonine (T), and c. the amino acid at Kabat position 256 is substituted with Glutamic acid (E).
13. The binding molecule of any preceding claim, comprising an immunoglobulin light chain constant region.
14. The binding molecule of claim 13, wherein the light chain constant region is a human kappa constant region.
15. The binding molecule of any preceding claim, which specifically binds human FcgRIIA 131R with an affinity characterized by a dissociation constant (KD) of about 0.15 nM, as measured by a BIAcore assay.
16. The binding molecule of any preceding claim, which specifically binds human FcgRIIA 131H with an affinity characterized by a dissociation constant (KD) of about 0.13 nM, as measured by a BIAcore assay.
17. The binding molecule of any preceding claim, which does not specifically bind to FcyRI, FcyRIIB, or FcyRIII.
18. The binding molecule of any preceding claim, which is conjugated to an agent selected from the group consisting of an antimicrobial agent, a therapeutic agent, a prodrug, a peptide, a protein, an enzyme, a lipid, a biological response modifier, a pharmaceutical agent, a
lymphokine, a heterologous antibody or fragment thereof, a detectable label, a polyethylene glycol (PEG), a toxin, and a combination of two or more of any said agents.
19. A composition comprising the binding molecule of any preceding claim and a carrier.
20. The composition of claim 19, which is a diagnostic reagent.
21. A method for inhibiting ribonucleoprotein-immune complex (RNP-IC)-mediated type I IFNa in a peripheral blood mononuclear cell (PBMC), the method comprising contacting the PBMC with the binding molecule of any one of claims 1 to 18.
22. A method for inhibiting anti-neutrophil cytoplasmic antibody (ANCA)-induced neutrophil activation, the method comprising contacting a neutrophil with the binding molecule of any one of claims 1 to 18.
23. A method of treating an inflammatory, immune-mediated, or autoimmune disease or disorder in a subject, the method comprising administering to a subject in need of treatment an effective amount of the binding molecule of any one of claims 1 to 18 or the composition of claim 19.
24. A method of preventing an inflammatory, immune-mediated, or autoimmune disease or disorder in a subject, the method comprising administering to a subject susceptible to the disease or disorder an effective amount of the binding molecule of any one of claims 1 to 18 or the composition of claim 19.
25. The method of claim 23 or claim 24, wherein the method comprises administering a second active agent.
26. The method of any one of claims 23 to 25, wherein the disease or disorder is ANCA- associated vasculitis (AAV), systemic lupus erythematosus (SLE), lupus nephritis, membranous nephritis, giant cell arteritis (GCA) vasculitis, immune thrombocytopenia (ITP), rheumatoid arthritis, polymyositis, dermatomyositis, pemphigus, hemolytic anemia, mixed connective tissue disease, Sjogren’s syndrome, scleroderma, an autoantibody disorder, an immune-complex- mediated disorder, ADA-mediated adverse effects, NETosis, and NETosis-associated disorders, including sepsis, thrombosis, acute kidney injury, acute lung injury, chronic obstructive pulmonary disease, glomerulonephritis, toxic liver injury, stroke, atherogenesis, Type I diabetes, and IgG mediated hypersensitive reaction.
27. A method for detecting FcgRIIA in a sample, the method comprising (a) contacting the sample with the binding molecule of any one of claims 1 to 18, and (b) detecting binding of the binding molecule to FcgRIIA, thereby detecting FcgRIIA in the sample.
28. The method of claim 27, which is a diagnostic method.
29. An isolated nucleic acid molecule comprising a nucleotide sequence encoding the binding molecule of any one of claims 1 to 18.
30. The nucleic acid molecule of claim 29 operably linked to a regulatory sequence.
31. A vector comprising the nucleic acid molecule of claim 29 or claim 30.
32. A host cell transformed with a nucleic acid molecule of claim 29 or claim 30, or the vector of claim 31.
33. The host cell of claim 32, which is a mammalian host cell.
34. A composition comprising the nucleic acid molecule of claim 29 or claim 30, the vector of claim 31, or the host cell of claim 32 or claim 33.
35. A method of making a binding molecule that specifically binds FcgRIIA, the method comprising culturing the host cell of claim 32 or claim 33 under suitable conditions for producing the binding molecule.
36. The method of claim 35, further comprising isolating the binding molecule.
37. A kit comprising the binding molecule of any one of claims 1 to 18 or the nucleic acid molecule of claim 29 or claim 30.
38. A method for inhibiting ribonucleoprotein-immune complex (RNP-IC)-mediated pro- inflammatory molecule production in a peripheral blood mononuclear cell (PBMC), the method comprising contacting the PBMC with the binding molecule of any one of claims 1 to 18.
39. The method of claim 38, wherein the pro-inflammatory molecule is interleukin-6.
40. The method of claim 39, wherein the pro-inflammatory molecule is tumor necrosis factor alpha.
41. The method of claim 26, wherein the disease or disorder is rheumatoid arthritis.
PCT/US2019/024977 2018-03-30 2019-03-29 Binding molecules specific for fcyriia and uses thereof WO2019191668A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201862650644P 2018-03-30 2018-03-30
US62/650,644 2018-03-30

Publications (1)

Publication Number Publication Date
WO2019191668A1 true WO2019191668A1 (en) 2019-10-03

Family

ID=68060772

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2019/024977 WO2019191668A1 (en) 2018-03-30 2019-03-29 Binding molecules specific for fcyriia and uses thereof

Country Status (1)

Country Link
WO (1) WO2019191668A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020191441A1 (en) * 2019-03-25 2020-10-01 Newsouth Innovations Pty Limited Treating immune platelet disorders using antigen-binding fragments

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20150344575A1 (en) * 2003-08-14 2015-12-03 Macrogenics, Inc. FcGammaRIIB Specific Antibodies and Methods of Use Thereof
WO2017186908A1 (en) * 2016-04-29 2017-11-02 Medimmune Limited BINDING MOLECULES SPECIFIC FOR FCγGAMMA RIIA AND USES THEREOF

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20150344575A1 (en) * 2003-08-14 2015-12-03 Macrogenics, Inc. FcGammaRIIB Specific Antibodies and Methods of Use Thereof
WO2017186908A1 (en) * 2016-04-29 2017-11-02 Medimmune Limited BINDING MOLECULES SPECIFIC FOR FCγGAMMA RIIA AND USES THEREOF

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020191441A1 (en) * 2019-03-25 2020-10-01 Newsouth Innovations Pty Limited Treating immune platelet disorders using antigen-binding fragments
CN114430683A (en) * 2019-03-25 2022-05-03 新南创新私人有限公司 Treatment of immune platelet disorders using antigen binding fragments

Similar Documents

Publication Publication Date Title
US11746153B2 (en) Binding molecules specific for FcγRIIA and uses thereof
CN109608544B (en) PD-L1 antibody, antigen binding fragment thereof and medical application thereof
AU2015322543B2 (en) Cytotoxicity-inducing therapeutic agent
CN107849136B (en) anti-TfR antibodies and their use in the treatment of proliferative and inflammatory diseases
EP3702373B1 (en) Anti-pd1 antibodies and their use as therapeutics and diagnostics
CA2838497C (en) Therapeutic antibodies
CA2977621C (en) Antibody binding to tfpi and composition comprising the same
US20210171625A1 (en) Anti-interferon gamma antibodies and uses thereof
KR20240093751A (en) Antibody drug conjugates for ablating hematopoietic stem cells
CN112409483A (en) anti-PD-L1 nano antibody
JP2024518724A (en) Anti-MASP2 antibodies, antigen-binding fragments thereof, and medical uses
AU2022271678A1 (en) Antibodies for treating alpha-synucleinopathies
US20230272067A1 (en) Human monoclonal antibodies against tigit for immune related diseases
WO2022247826A1 (en) Specific binding protein targeting pd-l1 and cd73
WO2019191668A1 (en) Binding molecules specific for fcyriia and uses thereof
EP4108683A1 (en) Anti-il-2 antibody, and antigen-binding fragment thereof and medical use thereof
WO2018039107A1 (en) Binding molecules specific for notch4 and uses thereof
TWI833227B (en) Specific binding protein targeting pd-l1 and cd73 and application thereof
TW202313693A (en) CDC platform antibody
KR20230087552A (en) Binding molecule that multimerizes CD45
EA045935B1 (en) ANTIBODIES TO CD3 AND THEIR APPLICATION
NZ730615B2 (en) Cytotoxicity-inducing therapeutic agent

Legal Events

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

Ref document number: 19776818

Country of ref document: EP

Kind code of ref document: A1

122 Ep: pct application non-entry in european phase

Ref document number: 19776818

Country of ref document: EP

Kind code of ref document: A1