US20140112926A1 - Fc VARIANTS - Google Patents

Fc VARIANTS Download PDF

Info

Publication number
US20140112926A1
US20140112926A1 US14/005,517 US201214005517A US2014112926A1 US 20140112926 A1 US20140112926 A1 US 20140112926A1 US 201214005517 A US201214005517 A US 201214005517A US 2014112926 A1 US2014112926 A1 US 2014112926A1
Authority
US
United States
Prior art keywords
substitutions
chain comprises
seq
chain
containing protein
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US14/005,517
Other languages
English (en)
Inventor
Zhi Liu
Gunasekaran Kannan
Wei Yan
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Amgen Inc
Original Assignee
Amgen Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Amgen Inc filed Critical Amgen Inc
Priority to US14/005,517 priority Critical patent/US20140112926A1/en
Assigned to AMGEN INC. reassignment AMGEN INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: YAN, WEI, LIU, ZHI, KANNAN, GUNASEKARAN
Publication of US20140112926A1 publication Critical patent/US20140112926A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [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
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/39558Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against tumor tissues, cells, antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/31Immunoglobulins specific features characterized by aspects of specificity or valency multispecific
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/40Immunoglobulins specific features characterized by post-translational modification
    • C07K2317/41Glycosylation, sialylation, or fucosylation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype
    • C07K2317/524CH2 domain
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype
    • C07K2317/526CH3 domain
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/72Increased 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
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • C07K2317/732Antibody-dependent cellular cytotoxicity [ADCC]

Definitions

  • the invention relates to polypeptides comprising variant Fc regions that can be heterodimeric and contain amino acid substitutions.
  • the invention further relates to methods of making and using such polypeptides.
  • Therapeutic monoclonal antibodies have been successfully used in various oncologic indications. See, e.g., Reichert et al. (2007), Nature Rev. Drug Discovery 6: 349-356. Efficacy can be dependent upon effector functions of the antibody, such as complement dependent cytotoxicity (CDC), antibody dependent cellular cytotoxicity (ADCC), and/or antibody dependent cell-mediated phagocytosis (ADCP) or upon antibody-induced formation of complexes of the antigen on the tumor cell surface, which can, in some cases, induce apoptosis. See, e.g., Deans et al. (2002), Immunology 107: 176-182.
  • CDC complement dependent cytotoxicity
  • ADCC antibody dependent cellular cytotoxicity
  • ADCP antibody dependent cell-mediated phagocytosis
  • Fc ⁇ Rs Fc gamma receptors
  • the invention includes an Fc-containing protein comprising a heterodimeric human IgG Fc region, which comprises an A chain and a B chain, which each comprise from 1 to 10 amino acid substitutions relative to a wild type human Fc polypeptide chain, wherein the Fc-containing protein binds to a human Fc ⁇ RIIIA-158V and/or Fc ⁇ RIIIA-158F with a K D of less than or equal to one fifth of the K D with which a second protein binds to human Fc ⁇ RIIIA-158V and/or Fc ⁇ RIIIA-158F, wherein the second protein is the same as the Fc-containing protein except that it contains a wild type human IgG Fc region without substitutions.
  • the human IgG Fc region can be a human IgG1 or IgG3 Fc region.
  • the Fc-containing protein can bind to human Fc ⁇ RIIIA-158V and/or Fc ⁇ RIIIA-158F with a K D of less than or equal to one tenth or one twentieth of the K D with which the second protein binds to human Fc ⁇ RIIIA-158V and/or Fc ⁇ RIIIA-158F.
  • the IgG Fc region of the Fc-containing protein can be an IgG1 Fc region, and the Fc region can be defucosylated.
  • the A chain and the B chain of the Fc-containing protein each comprise from 1 to 6 amino acid substitutions relative to a wild type human Fc polypeptide chain. At least one of these substitutions can be a heterodimerizing alteration.
  • the A chain and the B chain can each contain at least two amino acid substitutions that are heterodimerizing alterations and can, for example, contain two or three substitutions that are heterodimerizing alterations.
  • the heterodimerizing alterations can be charge pair mutations, such as the substitutions K392D and K409D in the A chain and the substitutions E356K and D399K in the B chain, or vice versa.
  • the heterodimerizing alterations can be pairs of knobs and holes substitutions.
  • the Fc-containing protein can comprise an Fc region in which the following substitutions are present: (a) the A chain comprises Q311M and K334V substitutions and the B chain comprises L234Y, E294L, and Y296W substitutions or vice versa; (b) the A chain comprises E233L, Q311M, and K334V substitutions and the B chain comprises L234Y, E294L, and Y296W substitutions or vice versa; (c) the A chain comprises L2341, Q311M, and K334V substitutions and the B chain comprises L234Y, E294L, and Y296W substitutions or vice versa; (d) the A chain comprises S298T and K334V substitutions and the B chain comprises L234Y, K290Y, and Y296W substitutions or vice versa; (e) the A chain comprises A330M and K334V substitutions and the B chain comprises L234Y, K290Y, and Y296W
  • the A chain can comprise the amino acid sequence of SEQ ID NO:8, 12, 14, 16, 20, 22, 24, 26, 28, 30, 32, 34, or 37 and the B chain can comprise the amino acid sequence of SEQ ID NO: 10, 18, 39, or 41.
  • the B chain can comprise the amino acid sequence of SEQ ID NO:8, 12, 14, 16, 20, 22, 24, 26, 28, 30, 32, or 34, and the A chain can comprise the amino acid sequence of SEQ ID NO:8, 10, or 18.
  • the Fc-containing protein can comprise one of the following combinations of amino acid sequences: SEQ ID NO: 8 and SEQ ID NO:10; SEQ ID NO: 16 and SEQ ID NO:18; SEQ ID NO: 12 and SEQ ID NO:10; SEQ ID NO: 14 and SEQ ID NO:10; SEQ ID NO: 20 and SEQ ID NO:18; SEQ ID NO: 22 and SEQ ID NO:18; SEQ ID NO: 24 and SEQ ID NO:10; SEQ ID NO: 26 and SEQ ID NO:10; SEQ ID NO: 28 and SEQ ID NO:18; SEQ ID NO: 30 and SEQ ID NO:18; SEQ ID NO: 32 and SEQ ID NO:18; SEQ ID NO: 34 and SEQ ID NO:18; SEQ ID NO:37 and SEQ ID NO:39; or SEQ ID NO:37 and SEQ ID NO:41.
  • any of the Fc-containing proteins described herein can be an antibody or an Fc fusion protein and can be made in a CHO cell, a HEK 293 cell, or NS0 cell.
  • Such an antibody can be a full length human IgG1 antibody, which can be monospecific, bispecific, trispecific or multispecific and/or can be monovalent or multivalent, including bivalent or tetravalent.
  • the Fc-containing protein can bind to one or more target molecules selected from the group consisting of WT1, MUC1, LMP2, EGFRvIII, HER-2/neu, MAGE-A3, NY-ESO-1, PSMA, GM2/GD2 synthase, CEA, MLANA/MART1, gp100, survivin, prostate-specific antigen (PSA), telomerase reverse transcriptase (hTERT), sarcoma translocation breakpoints, EPHA2, prostatic acid phosphatase (PAP), melanoma inhibitor of apoptosis (ML-IAP), ⁇ -fetoprotein (AFP), epithelial cell adhesion molecule (EpCAM), ERG, NA17.A2 peptide (VLPDVFIRC), paired box 3 (PAX3), anaplastic lymphoma kinase (ALK), androgen receptor, claudin 3, claudin 4, claudin 6, claudin 9, cyclin B1, polys
  • the Fc-containing proteins described herein can bind to HER-2/neu or mesothelin or can bind to both CD38 and CD138.
  • the invention includes a pharmaceutical composition comprising a therapeutically effective amount of any of the Fc-containing proteins described above and below plus a pharmaceutically acceptable carrier.
  • nucleic acids encoding any of the Fc-containing proteins described above and below plus a host cell containing such nucleic acids.
  • an A chain and a B chain are encoded by separate nucleic acid molecules, whereas in other embodiments an A chain and a B chain can be encoded on the same nucleic acid molecule.
  • the host cell can be a CHO cell, a HEK 293 cell, or an NS0 cell.
  • described herein is a method of making an Fc-containing protein comprising a heterodimeric Fc region comprising culturing the host cell under conditions such that the Fc-containing protein will be expressed and, in some embodiments, recovering the polypeptide from the cell mass or the culture medium.
  • Also described herein is a method of making a pharmaceutical composition comprising an Fc-containing protein containing a heterodimeric Fc region comprising the following steps: (a) culturing a host cell containing one or more nucleic acids encoding a heterodimeric Fc-containing protein as described herein under conditions such that the Fc-containing protein will be expressed; (b) recovering the Fc-containing protein from the cell mass or the culture medium; and (c) formulating the Fc-containing protein with a pharmaceutically acceptable carrier.
  • Also described herein is a method of making an Fc-containing protein containing a heterodimeric Fc region comprising the following steps: (a) providing a host cell containing one or more nucleic acids encoding an Fc-containing protein comprising a heterodimeric human IgG Fc region and a binding region, wherein the Fc region comprises an A chain and a B chain, which each comprise from 1 to 10 amino acid substitutions relative to a wild type human Fc polypeptide chain, wherein the Fc-containing protein binds to human Fc ⁇ RIIIA-158F and/or Fc ⁇ RIIIA-158V with a K D of less than or equal to one fifth of the K D with which a second protein binds to human Fc ⁇ RIIIA-158F or Fc ⁇ RIIIA-158V, wherein the second protein is the same as the Fc-containing protein except that it contains a wild type human IgG Fc region without substitutions; (b) culturing the host cell containing one or more nucle
  • a method for treating cancer comprising administering to a patient in need thereof a therapeutically effective amount of the Fc-containing protein or pharmaceutical composition described above or below, wherein the Fc-containing protein binds to a molecule that is displayed on the cancer cells.
  • a chemotherapeutic agent or a non-chemotherapeutic anti-neoplastic agent can be administered to the patient before, after, or concurrently with administration of the Fc-containing protein.
  • the cancer can be selected from the group consisting of mesothelioma, squamous cell carcinoma, myeloma, osteosarcoma, glioblastoma, glioma, carcinoma, adenocarcinoma, melanoma, sarcoma, acute and chronic leukemia, lymphoma, meningioma, Hodgkin's disease, Sézary syndrome, multiple myeloma, and lung, non-small cell lung, small cell lung, laryngeal, breast, head and neck, bladder, ovarian, skin, prostate, cervical, vaginal, gastric, renal cell, kidney, pancreatic, colorectal, endometrial, esophageal, hepatobiliary, bone, skin, and hematologic cancers, as well as cancers of the nasal cavity and paranasal sinuses, the nasopharynx, the oral cavity, the oropharynx, the larynx
  • described herein are uses of the Fc-containing protein or pharmaceutical composition described above or below in the treatment of a human disease, for example autoimmune diseases, asthma, systemic lupus erythematosus, infectious diseases, or cell proliferative diseases such as cancer, or in the manufacture of a medicament, wherein the medicament can be for treating cancer, asthma, systemic lupus erythematosus, or an infectious disease.
  • a human disease for example autoimmune diseases, asthma, systemic lupus erythematosus, infectious diseases, or cell proliferative diseases such as cancer, or in the manufacture of a medicament, wherein the medicament can be for treating cancer, asthma, systemic lupus erythematosus, or an infectious disease.
  • FIG. 1 Diagram of the tertiary structure of Fc ⁇ RIIIB bound to an Fc region.
  • This figure is a representation of the X-ray crystal structure of the Fc-Fc ⁇ RIIIB (Protein Data Bank code: 1T83) complex, which includes the extracellular region of Fc ⁇ RIIIB and a dimeric Fc region.
  • the Fc ⁇ RIIIB structure is shown in a wire model above.
  • Fc Chain A and Fc Chain B are shown below in ribbon models.
  • the tertiary structures of the extracellular regions of Fc ⁇ RIIIA and Fc ⁇ RIIIB are expected to be similar since only five of the 176 amino acids in these two extracellular regions differ.
  • a later-determined structure of an Fc-Fc ⁇ RIIIA complex (Protein Data Bank code: 3SGK) is very similar to this Fc-Fc ⁇ RIIIB complex structure.
  • FIG. 2 The amino acid sequence of a human IgG1 Fc polypeptide.
  • the amino acid sequence of a human IgG1 Fc region, starting with the hinge region and ending with the carboxyl terminus of the C H 3 region, is shown in single letter notation and is numbered according to the EU system of Edelman et al. (1969), Proc. Natl. Acad. Sci. 63: 78-85.
  • the amino acids underlined and in boldface type were randomized in constructing the libraries as described in Example 1. Beneath each of these amino acids is a “1,” a “2,” or a “3,” which indicates that DNAs encoding variants at the corresponding site were included in a Tier 1, 2, or 3 library as described in Example 1.
  • FIG. 3 Diagram showing the primary screening and initial combinatorial screening for substitutions that enhance binding to Fc ⁇ RIIIA.
  • SIG represents a polynucleotide encoding a signal sequence, which facilitates protein secretion from mammalian cells.
  • a region encoding a hinge region is represented by a horizontal line labeled “hinge.”
  • a rectangle labeled “Fc polypeptide” represents a polynucleotide encoding an Fc polypeptide chain.
  • the five-pointed and four-pointed stars mean that the polynucleotides encoding the Fc polypeptide chains contain one randomized codon in each molecule at selected positions as explained in Example 1.
  • the circles labeled “VH” and “VL” represent regions encoding a heavy chain variable region and a light chain variable region, respectively.
  • the “++” and “ ⁇ ” signs in the rectangles labeled “Fc polypeptide” mean that these regions include mutations such that the encoded Fc polypeptide chain will have the substitutions E356K, D399K and K392D, K409D, respectively.
  • FIG. 4 Percent inhibition of AlphaLISA® signal by full length IgG1 antibodies containing variant Fc regions.
  • the graph shows the percent inhibition of an AlphaLISA® signal as a function of concentration of competitor.
  • the various competitors, which are human IgG1 antibodies, are indicated by alias in the graph, and the substitutions contained in each competitor are indicated in Table 3.
  • FIG. 5 Percent inhibition of AlphaLISA® signal by full length IgG1 antibodies containing variant Fc regions.
  • the graph shows the percent inhibition of an AlphaLISA® signal as a function of concentration of competitor.
  • the various competitors, which are human IgG1 antibodies, are indicated by alias in the graph, and the substitutions contained in each competitor are indicated in Table 3.
  • FIG. 6 Percent inhibition of AlphaLISA® signal by full length IgG1 antibodies containing variant Fc regions.
  • the graph shows the percent inhibition of an AlphaLISA® signal as a function of concentration of competitor.
  • the various competitors, which are human IgG1 antibodies, are indicated by alias in the graph, and the substitutions contained in each competitor are indicated in Table 3.
  • FIG. 7 Percent cell killing by full length IgG1 antibodies containing variant Fc regions.
  • the graph shows the percentage of cells killed in an assay for antibody-dependent cellular cytotoxity (% ADCC) versus antibody concentration.
  • the various human IgG1 antibodies used in these assays are indicated by alias in the graph, and the substitutions contained in each antibody are indicated in Table 3.
  • FIG. 8 Percent cell killing by full length IgG1 antibodies containing variant Fc regions.
  • the graph shows the percentage of cells killed in an assay for antibody-dependent cellular cytotoxity (% ADCC) versus antibody concentration.
  • the various human IgG1 antibodies used in these assays are indicated by alias in the graph, and the substitutions contained in each antibody are indicated in Table 3.
  • FIG. 9 Percent cell killing by full length IgG1 antibodies containing variant Fc regions.
  • the graph shows the percentage of cells killed in an assay for antibody-dependent cellular cytotoxity (% ADCC) versus antibody concentration.
  • the various human IgG1 antibodies used in these assays are indicated by alias in the graph, and the substitutions contained in each antibody are indicated in Table 3.
  • FIG. 10 Percent inhibition of AlphaLISA® signal for binding to human Fc ⁇ R IIIA (158F) allelic variant by full length IgG1 antibodies containing variant Fc regions.
  • the graph shows the percent inhibition of an AlphaLISA® signal as a function of the log of the competitor concentration.
  • the various competitors, which are human IgG1 antibodies, are indicated by alias in the graph, and the substitutions contained in each competitor are indicated in Tables 3 and 4.
  • the designation “AFUCO” preceding an alias means that the antibody lacks fucose.
  • FIG. 11 Percent inhibition of AlphaLISA® signal for binding to human Fc ⁇ R IIIA (158V) allelic variant by full length IgG1 antibodies containing variant Fc regions.
  • the graph shows the percent inhibition of an AlphaLISA® signal as a function of the log of the competitor concentration.
  • the various competitors, which are human IgG1 antibodies, are indicated by alias in the graph, and the substitutions contained in each competitor are indicated in Tables 3 and 4.
  • the designation “AFUCO” preceding an alias means that the antibody lacks fucose.
  • FIG. 12 Percent cell lysis of cells expressing high levels of antigen by full length IgG1 antibodies containing variant Fc regions.
  • the graph shows the percentage of cells killed in an assay for antibody-dependent cellular cytotoxicity (% Specific Lysis) versus the log of the antibody concentration (pM).
  • the various human IgG1 antibodies used in these assays are indicated by alias in the graph, and the substitutions contained in each competitor are indicated in Tables 3 and 4.
  • FIG. 13 Percent cell lysis of cells expressing moderate levels of antigen by full length IgG1 antibodies containing variant Fc regions.
  • the graph shows the percentage of cells killed in an assay for antibody-dependent cellular cytotoxicity (% Specific Lysis) versus the log of the antibody concentration (pM).
  • the various human IgG1 antibodies used in these assays are indicated by alias in the graph, and the substitutions contained in each competitor are indicated in Tables 3 and 4.
  • FIG. 14 Comparisons of ADCC activity of fucosylated and defucosylated preparations of IgG1 antibodies containing a wild type or a variant Fc region.
  • the graphs show the percentage of cells killed in an assay for antibody-dependent cellular cytotoxicity (% Specific Lysis) versus the log of the antibody concentration [pM].
  • a cell line that expresses high levels of the antigen that the antibody binds to was used as a target cell (SKBR3).
  • SKBR3 target cell
  • JIMT1 a cell line that expresses moderate levels of the antigen was used as a target cell
  • the various human IgG1 antibodies used in these assays are indicated in the graph as follows: “M01” indicates an IgG1 antibody containing a wild type Fc region that binds to the antigen; “AFUCO-M01” indicates a preparation of the same IgG1 antibody that contains no fucose; “W117” indicates an IgG1 antibody that binds to the antigen and contains a W117 variant Fc region; “AFUCO-W117” indicates a preparation of W117 that contains no fucose; “W125” indicates an IgG1 antibody that binds to the antigen and contains a W125 variant Fc region; and “AFUCO-W125” indicates a preparation of “W125” that contains no fucose.
  • ADCC Fc ⁇ receptors
  • asymmetric alterations in the Fc region may be needed to maximally enhance the interaction of Fc ⁇ RIIIA with the Fc region of an Fc-containing protein and, thus, enhance ADCC.
  • such heterodimeric Fc regions can also have different binding regions attached to each Fc polypeptide chain, thus creating a molecule that can have different binding specificities on each of its two binding arms.
  • the instant invention provides Fc-containing proteins comprising such asymmetric substitutions in their Fc regions and having increased binding to Fc ⁇ RIIIA and enhanced ADCC activity.
  • polypeptides can also be bispecific, or multispecific, that is, they may bind to two or more different target molecules.
  • FIG. 2 shows the sequence of the Fc region of a human IgG1 antibody numbered according to the EU system. Particular amino acid residues in an IgG1 constant region of an antibody are notated using the one letter code for amino acids and the EU numbering system.
  • D399 refers to an aspartic acid that is present in wild type IgG at position 399. Mutations at a particular residue are notated similarly.
  • D399K means that the aspartic acid that is present in a wild type IgG1 at position 399 has been changed to a lysine.
  • ADCC refers to a process called antibody-dependent cellular cytotoxicity, which is an immune response mediated primarily by natural killer (NK) cells in humans.
  • NK natural killer
  • Fc ⁇ RIII on the surface of an NK cell recognizes the Fc region of antibody that is bound to antigen displayed on the surface of a target cell. This activates the NK cell, which releases perforins and granzymes, leading to lysis and apoptosis of the target cells.
  • CDC refers to a complex process called complement-dependent cytotoxicity that can lead to cell killing through the action of a cascade of proteins that can act through either of two major pathways. See, e.g., Liszewski and Atkinson, Ch. 26 in F UNDAMENTAL I MMUNOLOGY, 3 rd ed., Paul, ed., Raven Press, New York, 1993, pp. 917-940, the portions of which describe CDC are incorporated herein by reference.
  • ADCP refers to a process called antibody dependent cell-mediated phagocytosis.
  • target cells to which antibodies are bound are engulfed by phagocytic cells, such as macrophage, monocytes, neutrophils, and dendritic cells. Multiple Fc receptors are involved in this process.
  • phagocytic cells such as macrophage, monocytes, neutrophils, and dendritic cells.
  • Multiple Fc receptors are involved in this process.
  • an “antibody,” as meant herein, is a protein containing at least one heavy or light chain immunoglobulin variable region, in many cases a heavy and a light chain variable region.
  • the term “antibody” encompasses single chain Fv antibodies (scFv, which contain heavy and light chain variable regions joined by a linker), Fab, F(ab) 2 ′, Fab′, scFv:Fc antibodies (as described in Carayannopoulos and Capra, Ch. 9 in F UNDAMENTAL I MMUNOLOGY, 3 rd ed., Paul, ed., Raven Press, New York, 1993, pp.
  • IgG antibodies can be of the IgG1, IgG2, IgG3, or IgG4 isotype and can be human antibodies.
  • the portions of Carayannopoulos and Capra that described the structure of antibodies are incorporated herein by reference.
  • antibody includes dimeric antibodies containing two heavy chains and no light chains such as the naturally-occurring antibodies found in camels and other dromedary species and sharks. See, e.g., Muldermans et al., 2001, J. Biotechnol.
  • An antibody can be monospecific (that is, binding to only one kind of antigen) or multispecific (that is, binding to more than one kind of antigen). In some embodiments, an antibody can be bispecific (that is, binding to two different kinds of antigen). Further, an antibody can be monovalent, bivalent, or multivalent, meaning that it can bind to one or two or more antigen molecules at once.
  • Some of the possible formats for such antibodies include monospecific or bispecific full length antibodies, monospecific monovalent antibodies (as described in International Application WO 2009/089004 and US Publication 2007/0105199, the relevant portions of which are incorporated herein by reference) that may inhibit or activate the molecule to which they bind, bivalent monospecific or bispecific dimeric Fv-Fc, scFv-Fc, or diabody Fc, monospecific monovalent scFv-Fc/Fc's, and the multispecific binding proteins and dual variable domain immunoglobulins described in US Publication 2009/0311253 (the relevant portions of which are incorporated herein by reference), among many other possible antibody formats.
  • an “Fc fusion protein,” as meant herein, is a protein containing an Fc polypeptide chain fused to another polypeptide, which comprises a binding region that binds to a target molecule, and which does not comprise a heavy or light chain variable region of an antibody.
  • the binding region of an Fc fusion protein can comprise a non-immunoglobulin polypeptide such as a soluble portion of a receptor or one or more peptides that bind to a target molecule (such as, for example, a “monomer domain” as defined in U.S. Pat. No. 7,820,790 that binds to a target protein, which can be selected as discussed in U.S. Pat. No.
  • scaffold domains include, for example, T-lymphocyte associated protein-4 (CTLA-4; Nuttall et al. (1999), Proteins 36: 217-227), the Z domain of Staphylococcal protein 1 (Nord et al. (1995), Protein Eng.
  • Fc fusion proteins like other proteins containing Fc polypeptide chains generally form multimers, which can be dimers. Since the Fc regions described herein are generally heterodimeric, such Fc fusion proteins can form heterodimers.
  • the polypeptide fused to the Fc polypeptide chain can be different in each of polypeptide chains that, together form the heterodimer.
  • an Fc fusion protein can be heterodimeric and bispecific or monospecific or multispecific.
  • a “binding region,” as meant herein, is a region of an Fc-containing protein as described herein that binds to a target molecule, such as, for example, a protein that is expressed at high levels on a cancer cell, on a cell mediating an autoimmune or inflammatory condition, on an infected cell, on an infectious agent, or on a cell mediating an immune effector function, for example, an NK cell.
  • a binding region can contain a heavy or light chain immunoglobulin variable region or a non-immunoglobulin polypeptide.
  • an “scFv-Fc,” as meant herein, is a polypeptide that consists of a heavy and a light chain variable region of an antibody joined by a linker, which is followed by an Fc polypeptide chain of an antibody, optionally the Fc region of a human IgG antibody, such as an IgG1, IgG2, IgG3, or IgG4 antibody.
  • a full length “heavy chain,” as meant herein, comprises a heavy chain variable region (V H ), a first heavy chain constant domain (C H 1), a hinge domain, a second heavy chain constant domain (C H 2), and a third heavy chain constant domain (C H 3).
  • a full length “light chain,” as meant herein, comprises a light chain variable region (VD and a light chain constant domain (C L ).
  • an “Fc region” is a dimer consisting of two polypeptide chains joined by one or more disulfide bonds, each chain comprising part or all of a hinge domain plus a C H 2 and a C H 3 domain.
  • Each of the polypeptide chains is referred to as an “Fc polypeptide chain.”
  • a chain an “A chain” and the other is referred to as a “B chain.”
  • the Fc regions contemplated for use with the present invention are IgG Fc regions, which can be mammalian or human IgG1, IgG2, IgG3, or IgG4 Fc regions.
  • allelic types are known.
  • One allelic type has the sequence as shown in FIG. 2 (SEQ ID NO:2).
  • Another has two substitutions relative to the sequence in FIG. 2 namely E356D and M358L.
  • the alanine at position 431 is a glycine.
  • a human IgG1 Fc region as meant herein can contain any of these amino acid sequence variations.
  • Fc-containing protein is a protein comprising an Fc region as described herein and a binding region that binds to a target molecule.
  • the term “Fc-containing protein” encompasses an antibody or an Fc fusion protein that contains an Fc region.
  • Fc ⁇ RIIIA-158V refers to the allelic variant of human Fc ⁇ RIIIA that has a valine at position 158 in the amino acid sequence of Fc ⁇ RIIIA as shown in SEQ ID NO:35.
  • Fc ⁇ RIIIA-158F refers to the allelic variant of human Fc ⁇ RIIIA that has a phenylalanine at position 158 in the amino acid sequence of Fc ⁇ RIIIA.
  • the sequence of human Fc ⁇ RIIIA-158F, including the 17 amino acid signal peptide is reported in NCBI Accession Number NP — 001121065, which is incorporated herein by reference. Since this sequence includes the signal peptide, which is absent in the mature protein, position 158 is equivalent to amino acid 176.
  • SEQ ID NO:35 contains the amino acid sequence of the mature form of Fc ⁇ RIIIA-158V.
  • a “heterodimeric” Fc region is one in which the A chain and the B chain of the Fc region have different amino acid sequences rather than identical amino acid sequences.
  • an “scFv-Fc/Fc” is a dimeric protein consisting essentially of an scFv-Fc plus an Fc polypeptide chain (referred to herein as a “dummy Fc”).
  • the scFv-Fc can be linked to the dummy Fc via one or more disulfide bridges.
  • the Fc region can contain “heterodimerizing alterations” in the C H 3 domains, such as one, two, three, or more pairs of charge pair substitutions, as described below.
  • the “C H 3-C H 3 interface” consists of those amino acids in the C H 3 region that come into close contact with residues of the other C H 3 region in the context of an Fc region and/or a full length antibody. More specifically these residues are within 4.5 ⁇ of an amino acid residue on the other C H 3 region in the context of an Fc region.
  • “Chemotherapy,” as used herein, means the treatment of a cancer patient with a “chemotherapeutic agent” that has cytotoxic or cytostatic effects on cancer cells.
  • a “chemotherapeutic agent” specifically targets cells engaged in cell division and not cells that are not engaged in cell division. Chemotherapeutic agents directly interfere with processes that are intimately tied to cell division such as, for example, DNA replication, RNA synthesis, protein synthesis, the assembly, disassembly, or function of the mitotic spindle, and/or the synthesis or stability of molecules that play a role in these processes, such as nucleotides or amino acids. A chemotherapeutic agent therefore has cytotoxic or cytostatic effects on both cancer cells and other cells that are engaged in cell division.
  • Chemotherapeutic agents are well-known in the art and include, for example: alkylating agents (e.g. busulfan, temozolomide, cyclophosphamide, lomustine (CCNU), methyllomustine, streptozotocin, cis-diamminedi-chloroplatinum, aziridinylbenzo-quinone, and thiotepa); inorganic ions (e.g. cisplatin and carboplatin); nitrogen mustards (e.g. melphalan hydrochloride, ifosfamide, chlorambucil, and mechlorethamine HCl); nitrosoureas (e.g.
  • alkylating agents e.g. busulfan, temozolomide, cyclophosphamide, lomustine (CCNU), methyllomustine, streptozotocin, cis-diamminedi-chloroplatinum, azirid
  • BCNU carmustine
  • anti-neoplastic antibiotics e.g. adriamycin (doxorubicin), daunomycin, mitomycin C, daunorubicin, idarubicin, mithramycin, and bleomycin
  • plant derivatives e.g. vincristine, vinblastine, vinorelbine, paclitaxel, docetaxel, vindesine, VP-16, and VM-26
  • antimetabolites e.g.
  • Alkylating agents and nitrogen mustard act by alkylating DNA, which restricts uncoiling and replication of strands.
  • Methotrexate, cytarabine, 6-mercaptopurine, 5-fluorouracil, and gemcitabine interfere with nucleotide synthesis.
  • Plant derivatives such a paclitaxel and vinblastine are mitotic spindle poisons. The podophyllotoxins inhibit topoisomerases, thus interfering with DNA replication.
  • Other mechanisms of action include carbamoylation of amino acids (lomustine, carmustine), and depletion of asparagine pools (asparaginase).
  • chemotherapeutic agents are those that directly affect the same cellular processes that are directly affected by the chemotherapeutic agents listed above.
  • Non-chemotherapeutic anti-neoplastic agents are chemical agents, compounds, or molecules having cytotoxic or cytostatic effects on cancer cells other than chemotherapeutic agents.
  • Non-chemotherapeutic antineoplastic agents may, however, be targeted to interact directly with molecules that indirectly affect cell division such as cell surface receptors, including receptors for hormones or growth factors.
  • non-chemotherapeutic antineoplastic agents do not interfere directly with processes that are intimately linked to cell division such as, for example, DNA replication, RNA synthesis, protein synthesis, or mitotic spindle function, assembly, or disassembly.
  • non-chemotherapeutic anti-neoplastic agents include inhibitors of Bcl2, inhibitors of farnesyltransferase, anti-estrogenic agents such as tamoxifen, anti-androgenic compounds, interferon, arsenic, retinoic acid, retinoic acid derivatives, antibodies targeted to tumor-specific antigens, and inhibitors of the Bcr-Abl tyrosine kinase (e.g. the small molecule STI-571 marketed under the trade name GLEEVECTM by Novartis, New York and New Jersey, USA and Basel, Switzerland), among many possible non-chemotherapeutic anti-neoplastic agents.
  • Bcr-Abl tyrosine kinase e.g. the small molecule STI-571 marketed under the trade name GLEEVECTM by Novartis, New York and New Jersey, USA and Basel, Switzerland
  • Heterodimerizing alterations generally refer to alterations in the A and B chains of an Fc region that facilitate the formation of heterodimeric Fc regions, that is, Fc regions in which the A chain and the B chain of the Fc region do not have identical amino acid sequences. Heterodimerizing alterations can be asymmetric, that is, a A chain having a certain alteration can pair with a B chain having a different alteration. These alterations facilitate heterodimerization and disfavor homodimerization. Whether hetero- or homo-dimers have formed can be assessed by size differences as determined by polyacrylamide gel electrophoresis in situations where one polypeptide chain is a dummy Fc and the other is an scFv-Fc.
  • knobs and holes substitutions are so-called “knobs and holes” substitutions. See, e.g., U.S. Pat. No. 7,695,936 and US Patent Application Publication 2003/0078385, the portions of which describe such mutations are incorporated herein by reference. As meant herein, an Fc region that contains one pair of knobs and holes substitutions, contains one substitution in the A chain and another in the B chain.
  • knobs and holes substitutions in the A and B chains of an IgG1 Fc region have been found to increase heterodimer formation as compared with that found with unmodified A and B chains: 1) Y407T in one chain and T366Y in the other; 2) Y407A in one chain and T366W in the other; 3) F405A in one chain and T394W in the other; 4) F405W in one chain and T394S in the other; 5) Y407T in one chain and T366Y in the other; 6) T366Y and F405A in one chain and T394W and Y407T in the other; 7) T366W and F405W in one chain and T394S and Y407A in the other; 8) F405W and Y407A in one chain and T366W and T394S in the other; and 9) T366W in one polypeptide of the Fc and T366S, L368A
  • substitutions creating new disulfide bridges can facilitate heterodimer formation. See, e.g., US Patent Application Publication 2003/0078385, the portions of which describe such mutations are incorporated herein by reference.
  • Such alterations in an IgG1 Fc region include, for example, the following substitutions: Y349C in one Fc polypeptide chain and S354C in the other; Y349C in one Fc polypeptide chain and E356C in the other; Y349C in one Fc polypeptide chain and E357C in the other; L351C in one Fc polypeptide chain and S354C in the other; T394C in one Fc polypeptide chain and E397C in the other; or D399C in one Fc polypeptide chain and K392C in the other.
  • substitutions changing the charge of a one or more residue can enhance heterodimer formation as explained in WO 2009/089004, the portions of which describe such substitutions are incorporated herein by reference.
  • Such substitutions are referred to herein as “charge pair substitutions,” and an Fc region containing one pair of charge pair substitutions contains one substitution in the A chain and a different substitution in the B chain.
  • charge pair substitutions include the following: 1) K409D or K409E in one chain plus D399K or D399R in the other; 2) K392D or K392E in one chain plus D399K or D399R in the other; 3) K439D or K439E in one chain plus E356K or E356R in the other; and 4) K370D or K370E in one chain plus E357K or E357R in the other.
  • the substitutions R355D, R355E, K360D, or K360R in both chains can stabilize heterodimers when used with other heterodimerizing alterations. Specific charge pair substitutions can be used either alone or with other charge pair substitutions.
  • single pairs of charge pair substitutions and combinations thereof include the following: 1) K409E in one chain plus D399K in the other; 2) K409E in one chain plus D399R in the other; 3) K409D in one chain plus D399K in the other; 4) K409D in one chain plus D399R in the other; 5) K392E in one chain plus D399R in the other; 6) K392E in one chain plus D399K in the other; 7) K392D in one chain plus D399R in the other; 8) K392D in one chain plus D399K in the other; 9) K409D and K360D in one chain plus D399K and E356K in the other; 10) K409D and K370D in one chain plus D399K and E357K in the other; 11) K409D and K392D in one chain plus D399K, E356K, and E357K in the other; 12) K409D and K370D
  • a “target molecule,” as meant herein, is a molecule to which the binding region of an Fc-containing protein described herein binds.
  • a target molecule is a protein that is expressed at high levels, for example, on a cancer cell, on a cell mediating an autoimmune or inflammatory condition, on an infected cell, on an infectious agent, or on a cell mediating an immune effector function, for example, an NK cell.
  • Tumor burden refers to the number of viable cancer cells, the number of tumor sites, and/or the size of the tumor(s) in a patient suffering from a cancer.
  • a reduction in tumor burden can be observed, for example, as a reduction in the amount of a tumor-associated antigen or protein in a patient's blood or urine, a reduction in the number of tumor cells or tumor sites, and/or a reduction in the size of one or more tumors.
  • a “therapeutically effective amount” of a protein comprising a variant Fc region as described herein is an amount that has the effect of, for example, reducing or eliminating the tumor burden of a cancer patient or reducing or eliminating the symptoms of any disease condition that the protein is used to treat.
  • a therapeutically effective amount need not completely eliminate all symptoms of the condition, but may reduce severity of one or more symptoms or delay the onset of more serious symptoms or a more serious disease that can occur with some frequency following the treated condition.
  • Treatment of any disease mentioned herein encompasses an alleviation of at least one symptom of the disease, a reduction in the severity of the disease, or the delay or prevention of disease progression to more serious symptoms that may, in some cases, accompany the disease or lead to at least one other disease. Treatment need not mean that the disease is totally cured. A useful therapeutic agent needs only to reduce the severity of a disease, reduce the severity of one or more symptoms associated with the disease or its treatment, or delay the onset of more serious symptoms or a more serious disease that can occur with some frequency following the treated condition.
  • the present invention encompasses Fc-containing proteins that comprise a binding region that binds to a target molecule and a variant Fc region.
  • Fc-containing proteins that comprise a binding region that binds to a target molecule and a variant Fc region.
  • These include antibodies and Fc fusion proteins, containing human or non-human IgG Fc regions, which could be IgG1, IgG2, IgG3, or IgG4 Fc regions, that are altered at selected amino acid residues as compared to an unchanged human or non-human Fc region and that bind to Fc ⁇ RIIIA with enhanced affinity as compared to the unchanged human or non-human Fc region.
  • Fc ⁇ RIIIA interacts with an Fc region in an asymmetric fashion, i.e., contacting different amino acid residues in the two Fc polypeptide chains that make up the Fc region
  • the asymmetrically altered Fc regions described herein can be particularly effective in enhancing affinity to Fc ⁇ RIIIA and, thus, ADCC.
  • the altered human Fc regions described herein can be altered such that the sequences of the two Fc polypeptide chains that make up an Fc region, that is, the A chain and the B chain, differ.
  • these Fc regions can also contain heterodimerizing alterations, which are different in the A and B chains, that discourage the formation of homodimeric Fc-containing proteins and encourage the formation of heterodimeric Fc-containing proteins.
  • Proteins containing the altered Fc regions can be more effective at binding to Fc ⁇ RIIIA and at eliciting ADCC as compared to proteins comprising an unaltered Fc region, or to an Fc region containing only heterodimerizing alterations, and can have increased efficacy as therapeutics in vivo, for example in oncologic or neoplastic indications and/or in treating autoimmune or infectious conditions.
  • Fc fusion proteins include heterodimers in which each Fc polypeptide chain is fused to a different protein. Such Fc fusion proteins are bivalent and bispecific. Also included are bivalent and monospecific Fc fusion protein. Similarly, monospecific or bispecific full length antibodies, monovalent antibodies, and bispecific or monospecific scFv-Fc's are among the many kinds of proteins that could contain the altered Fc regions described herein.
  • the invention also encompasses nucleic acids encoding the Fc polypeptide chains in the altered Fc regions and proteins containing these Fc polypeptide chains. Also provided are methods of making these proteins and methods of using these proteins to treat various human conditions.
  • Fc ⁇ Rs human Fc gamma receptors
  • Id immunoreceptor tyrosine-based activation motif
  • ITAM immunoreceptor tyrosine-based activation motif
  • Fc ⁇ RIIB receptors are the only known human inhibitory Fc ⁇ R and contain an immunoreceptor tyrosine-based inhibitory motif (ITIM) in the intracellular domain that mediates inhibition of cell activation. Id.
  • Enhanced affinity of an antibody for Fc ⁇ RIIIA can be indicative of enhanced clinical efficacy in oncologic indications.
  • Allelic variants of Fc ⁇ RIIIA having either a valine or a phenylalanine at amino acid 158 have been associated with higher or lower affinity binding to IgG, respectively. Koene et al. (1997), Blood 90(3): 1109-1114.
  • allelic differences also significantly correlate with clinical efficacy observed in patients with follicular lymphoma treated with rituximab (an IgG1 anti-CD20 monoclonal antibody) and in patients with solid tumors treated with either cetuximab (a chimeric IgG1 anti-epidermal growth factor receptor monoclonal antibody) or trastuzumab (an IgG1 anti-epidermal growth factor receptor 2 monoclonal antibody).
  • cetuximab a chimeric IgG1 anti-epidermal growth factor receptor monoclonal antibody
  • trastuzumab an IgG1 anti-epidermal growth factor receptor 2 monoclonal antibody
  • Each of the Fc polypeptide chains can have amino acid sequences that differ because of amino acid substitutions relative to the sequence of a human IgG Fc polypeptide chain.
  • An Fc polypeptide chain can be of a human IgG1 or IgG3 Fc polypeptide.
  • each Fc polypeptide chain comprises from one to twenty, one to ten, or one to five amino acid substitutions relative to a naturally-occurring human Fc sequence.
  • an Fc polypeptide chain can comprise zero, one, two, three, four, five, six, seven, eight, nine, or ten amino acid substitutions relative to a naturally-occurring human Fc polypeptide chain. In some embodiments, an Fc polypeptide chain can comprise no more than 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 amino acid substitutions.
  • substitutions can occur, for example, at one or more of the following sites in an Fc polypeptide chain: E233, L234, L235, S239, F241, F243, K246, K248, D249, L251, M252, I253, S254, R255, T256, E258, T260, V264, D265, S267, H268, E269, D270, E272, K274, F275, N276, Y278, V279, D280, V282, E283, V284, H285, N286, A287, K288, T289, K290, R292, E293, E294, Q295, Y296, S298, Y300, R301, V302, V303, V305, T307, L309, H310, Q311, D312, W313, L314, N315, K317, E318, K320, K322, S324, K326, A327, L328, A330, I332, E333, K334, T335, I33
  • the antibodies or Fc fusion proteins described herein, which comprise an Fc region can contain one or more of the following particular amino acid substitutions in one or both of the A chain and the B chain that make up the Fc region: E233L, L234I, L234Y, L235S, G236Y, S239D, S239E, S239N, S239T, F243M, F243L, F243V, F2431, K246W, K246E, K246S, K246V, K248Y, K248L, M252D, I253V, I253K, R255S, R255N, T256V, T256Q, E258S, E258V, H268E, H268K, A287F, K288T, K2881, K290G, K290F, K290S, K290W, K290Q, K290Y, E294L, Y296W, Y296L, S298A, S
  • the proteins of the invention can comprise an Fc region in which the A and B chains comprise the following substitutions: (1) K334V in one Fc polypeptide chain and Y296W plus S298C in the other; (2) K334V in one Fc polypeptide chain and L234Y, Y296W, and S298C in the other; (3) L235S, S239D, and K334V in one Fc polypeptide chain and L234Y, K290Y, and Y296W in the other; (4) L235S, S239D, and K334V in one Fc polypeptide chain and L234Y, Y296W, and S298C in the other; (5) Q311M and K334V in one Fc polypeptide chain and L234Y, F243V, and Y296W in the other; (6) Q311M and K334V in one Fc polypeptide chain and L234Y, E294L, and Y296W in the other
  • amino acid sequences of Fc polypeptide chains include SEQ ID NOs:8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 37, 39, and 41. These sequences contain heterodimerizing alterations and substitutions that enhance binding to Fc ⁇ RIIIA.
  • the Fc-containing proteins of the invention can contain heterodimeric human IgG1 Fc regions. That is, the two Fc polypeptide chains that, together, make up the Fc region each have a different amino acid sequence.
  • a heterodimeric Fc region of the invention contains heterodimerizing alterations (as defined above), thus greatly facilitating production of proteins containing the heterodimeric Fc.
  • An IgG Fc region is generally glycosylated at N297 when it is produced by a mammalian cell, and the absence of fucose in this carbohydrate can increase binding to Fc ⁇ RIII and the ability of an IgG antibody to elicit ADCC. Malphettes et al. (2010), Biotechnol. Bioeng. 106(5): 774-783.
  • the Fc-containing proteins, including antibodies and Fc fusion proteins, described herein can be “defucosylated,” that is, essentially free of fucose or containing only minor amounts of fucose. As meant herein, at least about 85%, 90%, or 95% of the glycans released from a defucosylated protein preparation do not contain fucose.
  • defucosylated and “afucosylated” are used interchangeably herein.
  • Such proteins can be produced as described above, for example, in FUT8 ⁇ / ⁇ or GFT / ⁇ CHO cells.
  • the fucose contents of a protein can be determined by as described by Ishiguro et al. (2010), Cancer Sci 101: 2227-2233, at 2228-2229, the relevant portion of which is incorporated herein by reference.
  • proteins are known to be expressed at high levels on cancer cells, on cells that mediate an autoimmune or inflammatory condition, or on infectious agents or infected cells. Such proteins are potential target molecules for therapeutic Fc-containing proteins described herein. Antibodies or Fc fusion proteins that bind to such potential target proteins are particularly appropriate for use with the present invention.
  • Potential target proteins known to be expressed on human cancer cells include the following human proteins: WT1, MUC1, LMP2, EGFRvIII, HER-2/neu, MAGE-A3, NY-ESO-1, PSMA, GM2/GD2 synthase, CEA, MLANA/MART1, gp100, survivin, prostate-specific antigen (PSA), telomerase reverse transcriptase (hTERT), sarcoma translocation breakpoints, EPHA2, prostatic acid phosphatase (PAP), melanoma inhibitor of apoptosis (ML-IAP), ⁇ -fetoprotein (AFP), epithelial cell adhesion molecule (EpCAM), ERG, NA17.A2 peptide (VLPDVFIRC), paired box 3 (PAX3), anaplastic lymphoma kinase (ALK), androgen receptor, cyclin B1, polysialic acid, rho-related GTP-binding protein RhoC, v-my
  • Cancer antigens also include the human herpes virus 4 protein LMP2, the human papillomavirus proteins E6 and E7, and the glycoceramide globo H (as described in Gilewski et al. (2001), Proc. Natl. Acad. Sci.
  • the ⁇ 4 subunit of the ⁇ 4 ⁇ 1 and ⁇ 4 ⁇ 7 integrins the ⁇ 4 ⁇ 7 integrin, BAFF, APRIL, CD2, CD3, CD20, CD52, CD73, CD80, CD86, the C 5 complement protein, IgE, IL-1 ⁇ , IL-5, IL-6R, IL-12, IL-23, and tumor necrosis factor ⁇ (TNF ⁇ ).
  • targets include proteins or other molecules displayed on the surface of pathogenic organisms including viruses, bacteria (including the species Borrelia, Staphylococcus, Escherichia , among many other species), fungi (including yeast), giardia , amoeba, eukarytic protists of the genus Plasmodium , ciliates, trypanosomes, nematodes, and other eukaryotic parasites.
  • bacteria including the species Borrelia, Staphylococcus, Escherichia , among many other species
  • fungi including yeast
  • giardia amoeba
  • amoeba eukarytic protists of the genus Plasmodium
  • ciliates trypanosomes
  • nematodes eukaryotic parasites.
  • the Fc-containing protein can bind one or two or multiple target molecules, which can be identical or different target molecules and can be monomers or multimers, on the same cells or different types of cells, to antagonize or agonize the signaling pathway; or to increase the avidity or specificity of an interaction between a target molecule and another molecule (which may or may not be a target molecule).
  • a bispecific or multispecific Fc-containing protein can bind to a target molecule, such as those mentioned in the paragraphs above, and another molecule, which can also be a target molecule, expressed at high levels on a cell involved in mediating a cytotoxic response by the immune system, such as, for example, NKG2D on NK cells or CD3 or T cell receptor on T cells.
  • the target molecule could be, for example, one of the following: (1) a human protein that is selectively expressed on cancer cells; (2) a protein of a virus or other pathogen that is highly expressed on the surface of the pathogen or on the surface of a pathogen-infected host cell; or (3) a human protein that is selectively expressed on the surface of a human cell type that mediates a condition such as an autoimmune or inflammatory disease.
  • nucleic acids encoding the Fc polypeptide chains of the Fc-containing proteins described herein are also provided.
  • nucleic acids are provided that encode Fc polypeptides, and/or Fc-containing proteins comprising them, comprising one or more of the following amino acid sequences: SEQ ID NOs:8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 37, 39, or 41.
  • sequences encoding such Fc polypeptide chains include SEQ ID NOs:7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 33, 36, 38, and 40.
  • These nucleic acids encode the amino acid sequences of SEQ ID NOs:8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 37, 39, and 41, respectively.
  • nucleic acid sequences encoding the many variant Fc polypeptide chains described herein, are also encompassed by the instant invention. These nucleic acids are useful for, inter alia, producing recombinant proteins containing altered Fc polypeptide chains, as described herein. These altered Fc polypeptide chains can be part of a heterodimeric Fc region that binds to Fc ⁇ RIIIA with enhanced affinity, as shown by binding with a lower K D than a wild type Fc region. Such nucleic acids can also encode a signal sequence that facilitates the secretion of a protein in mammalian cells and/or a binding region that binds to a target molecule.
  • signal sequences are cleaved from the remainder of a protein during maturation and are not part of a mature protein, even though they are encoded in a nucleic acid encoding the protein.
  • Signal sequences can be easily identified, e.g., as described by Kertein et al. (2000), Bioinformatics 16(8): 741-742, Nielsen and Krogh (1998), Proc. Sixth Int. Conf. on Intelligent Systems for Molecular Biol (AAAI Press): 122-130, Nielsen et al. (1997), Protein Eng. 10(1): 1-6, and Nielsen et al. (1997), Int. J. Neural Systems 8(5&6): 581-599.
  • the relevant portions of the references are incorporated herein by reference.
  • the nucleic acids of the invention include DNA and RNA in single- and double-stranded forms.
  • both polypeptide chains of a heterodimeric Fc-containing protein are encoded on a single nucleic acid molecule.
  • a heterodimeric Fc-containing protein can be encoded on two, three, or more nucleic acid molecules.
  • isolated nucleic acid is a nucleic acid that has been separated from adjacent sequences present in the genome of the organism from which the nucleic acid was initially isolated. For example, if the nucleic acid encodes an altered human IgG1 Fc region, the adjacent sequences would be the sequences adjacent to the sequences encoding an IgG1 Fc in the human genome. It is to be understood that nucleic acids synthesized chemically or produced enzymatically by PCR are “isolated nucleic acids,” as meant herein.
  • An isolated nucleic acid molecule refers to a nucleic acid molecule in the form of a separate fragment or as a component of a larger nucleic acid construct.
  • Fc-containing proteins such as antibodies and fusion proteins, encompassed by the invention can be made by methods known in the art. More specifically, a nucleic acid that encodes an Fc-containing protein including an altered Fc polypeptide chain, as described herein, can be introduced into a vector, which can be introduced into a host cell. Since the heterodimeric, Fc-containing proteins described herein necessarily contain at least two polypeptide chains, nucleic acids encoding these chains may be present on either a single vector or two or more vectors. If more than one vector is used, these vectors can be introduced together into a host cell. Vectors and host cells comprising nucleic acids encoding such a protein are encompassed by the invention.
  • the host cell containing the nucleic acids encoding the Fc-containing protein can be cultured under conditions such that the protein can be expressed.
  • the expressed protein can then be obtained from the medium in which the cells are cultured or from the cells themselves and purified by any of the many appropriate means known in the art.
  • genetic engineering methods for the production of proteins include the expression of the polynucleotide molecules in cell free expression systems, in cellular hosts, in tissues, and in animal models, according to known methods.
  • the vector can include a selectable marker and an origin of replication, for propagation in a host.
  • the vector can further include suitable transcriptional or translational regulatory sequences, such as those derived from mammalian, avian, microbial, viral, plant, or insect genes, operably linked to the nucleic acid encoding the protein. Examples of such regulatory sequences include transcriptional promoters, operators, or enhancers, mRNA ribosomal binding sites, and appropriate sequences that control transcription and translation. Nucleotide sequences are operably linked when the regulatory sequence functionally relates to the DNA encoding the target protein. Thus, a promoter nucleotide sequence is operably linked to a nucleic acid sequence if the promoter nucleotide sequence directs the transcription of the nucleic acid sequence.
  • Suitable host cells for expression of the antibodies or Fc fusion proteins described herein include prokaryotic cells, yeast cells, plant cells, insect cells, and higher eukaryotic cells, including mammalian or avian cells.
  • the regulatory sequences in the vector will be chosen such that they are operable in the host cell.
  • Suitable prokaryotic host cells include bacteria of the genera Escherichia, Bacillus , and Salmonella , as well as members of the genera Pseudomonas, Streptomyces , and Staphylococcus .
  • prokaryotic cells for example, in E.
  • the polynucleotide molecule encoding the protein preferably includes an N-terminal methionine residue to facilitate expression of the recombinant polypeptide.
  • the N-terminal methionine may optionally be cleaved from the expressed polypeptide.
  • Suitable yeast host cells include cells from genera including Saccharomyces, Pichia , and Kluveromyces .
  • Preferred yeast hosts are S. cerevisiae and P. pastoris .
  • a suitable system for expression in an insect host cell is described, for example, in the review by Luckow and Summers ((1988), BioTechnology 6:47), the relevant portions of which are incorporated herein by reference.
  • Suitable mammalian host cells include the COS-7 line of monkey kidney cells (Gluzman et al. (1981), Cell 23: 175-182), baby hamster kidney (BHK) cells, Chinese hamster ovary (CHO) cells (Puck et al. (1958), PNAS USA 60: 1275-1281), CV-1 (Fischer et al. (1970), Int. J. Cancer 5: 21-27), HEK 293 cells from human embryonic kidney (American Type Culture Collection (ATCC®) catalog no. CRL-1573), and human cervical carcinoma cells (HELA) (ATCC® CCL 2).
  • the relevant portions of the references referred to in this paragraph are incorporated herein by reference. Many other host cells are known in the art.
  • Expression vectors for use in cellular hosts generally comprise one or more phenotypic selectable marker genes. Such genes encode, for example, a protein that confers antibiotic resistance or that supplies an auxotrophic requirement.
  • phenotypic selectable marker genes encode, for example, a protein that confers antibiotic resistance or that supplies an auxotrophic requirement.
  • a wide variety of such vectors are readily available from commercial sources. Examples include pGEM® vectors (Promega), pSPORT vectors, and pPROEXTM vectors (InVitrogen, Life Technologies, Carlsbad, Calif.), Bluescript vectors (Stratagene), and pQE vectors (Qiagen).
  • Yeast vectors will often contain an origin of replication sequence from a 2 ⁇ yeast plasmid, an autonomously replicating sequence (ARS), a promoter region, sequences for polyadenylation, sequences for transcription termination, and a selectable marker gene.
  • Vectors replicable in both yeast and E. coli may also be used.
  • a shuttle vector will also include sequences for replication and selection in E. coli .
  • Direct secretion of the target polypeptides expressed in yeast hosts may be accomplished by the inclusion of nucleotide sequence encoding the yeast ⁇ -factor leader sequence at the 5′ end of the Fc-containing protein. Brake (1989), Biotechnology 13: 269-280.
  • suitable expression vectors for use in mammalian host cells include pcDNA3.1/Hygro + (Invitrogen), pDC409 (McMahan et al. (1991), EMBO J. 10: 2821-2832), and pSVL (Pharmacia Biotech).
  • Expression vectors for use in mammalian host cells can include transcriptional and translational control sequences derived from viral genomes. Commonly used promoter sequences and enhancer sequences that can be used to promote transcription of RNA encoding the proteins described herein include, but are not limited to, those derived from human cytomegalovirus (CMV), Adenovirus 2, Polyoma virus, and Simian virus 40 (SV40).
  • CMV cytomegalovirus
  • SV40 Simian virus 40
  • Fc-containing proteins of the invention can be used as therapeutics, particularly in disease contexts in which the selective killing of cells on which a particular target molecule is displayed is desirable.
  • the Fc-containing proteins of the invention can also be useful for eliminating soluble ligands, viruses, or foreign pathogenic cells. For example, in cancer patients, it is desirable to kill cancer cells, which may selectively express certain proteins that can be targeted by the Fc-containing proteins described herein.
  • antibodies or Fc fusion proteins that bind to such cancer target proteins and have enhanced cell killing properties can be desirable therapeutics in cancer indications.
  • cancer target protein that is, a protein expressed on a cancer cell
  • cytotoxic cell a protein expressed on a cytotoxic cell
  • CD16 which is expressed on NK cells
  • NKG2D which is expressed on cytotoxic T cells and NK cells
  • cytotoxic T cells and NK cells are proteins expressed on cytotoxic cells that can be target proteins.
  • asthma an inflammatory condition, it can be useful to kill eosinophils, which mediate damage of cells in the airway and induce hyperresponsiveness and mucus hypersecretion. Kolbeck et al. (2010), J. Allergy Clin. Immunol.
  • Fc-containing proteins with enhanced cell killing properties against antigens preferentially expressed on eosinophils can be useful in asthma.
  • viruses, foreign pathogenic cells, or infected host cells can also be targeted by the antibodies or Fc fusion proteins described herein.
  • the invention contemplates methods for treating patients suffering from a cell proliferative disease, including various forms of cancer, with the Fc-containing proteins described herein or with combinations including Fc-containing proteins comprising an altered Fc region plus other therapeutic agents.
  • the patient can be a human, but the methods may be applied to any mammal, including domestic animals such as pets and farm animals.
  • compositions for use in such methods that include a therapeutically effective amount of a protein containing an altered Fc region and, in some cases, an effective amount of another therapeutic agent, plus a suitable diluent, excipient, or carrier.
  • the Fc-containing proteins described herein can be administered with a variety of drugs and treatments have been widely employed in cancer treatment such as, for example, chemotherapeutic agents, non-chemotherpeutic, anti-neoplastic agents, and/or radiation.
  • chemotherapy and/or radiation can occur before, during, and/or after any of the treatments described herein.
  • chemotherapeutic agents include, but are not limited to, cisplatin, taxol, etoposide, mitoxantrone (Novantrone), actinomycin D, cycloheximide, camptothecin (or water soluble derivatives thereof), methotrexate, mitomycin (e.g., mitomycin C), dacarbazine (DTIC), anti-neoplastic antibiotics such as adriamycin (doxorubicin) and daunomycin, and all the chemotherapeutic agents mentioned above.
  • mitomycin e.g., mitomycin C
  • DTIC dacarbazine
  • anti-neoplastic antibiotics such as adriamycin (doxorubicin) and daunomycin
  • the Fc-containing proteins described herein can be used to treat cell proliferative diseases, including cancer, which involve the unregulated and/or inappropriate proliferation of cells, sometimes accompanied by destruction of adjacent tissue and growth of new blood vessels, which can allow invasion of cancer cells into new areas, i.e. metastasis.
  • cell proliferative diseases including cancer
  • cancer which involve the unregulated and/or inappropriate proliferation of cells, sometimes accompanied by destruction of adjacent tissue and growth of new blood vessels, which can allow invasion of cancer cells into new areas, i.e. metastasis.
  • non-malignant conditions that involve inappropriate cell growth, including colorectal polyps, cerebral ischemia, gross cystic disease, polycystic kidney disease, benign prostatic hyperplasia, and endometriosis.
  • cancers including mesotheliomas, squamous cell carcinomas, myelomas, osteosarcomas, glioblastomas, gliomas, carcinomas, adenocarcinomas, melanomas, sarcomas, acute and chronic leukemias, lymphomas, and meningiomas, Hodgkin's disease, Sézary syndrome, multiple myeloma, and lung, non-small cell lung, small cell lung, laryngeal, breast, head and neck, bladder, ovarian, skin, prostate, cervical, vaginal, gastric, renal cell, kidney, pancreatic, colorectal, endometrial, and esophageal, hepatobiliary, bone, skin, and hematologic cancers, as well as cancers of the nasal cavity and paranasal sinuses, the nasopharynx, the oral cavity
  • the Fc-containing proteins described herein can find further use in other kinds of conditions where it is beneficial to deplete certain cell types. For example, depletion of human eosinophils in asthma, excess human B cells in systemic lupus erythematosus, excess human Th2 T cells in autoimmune conditions, or pathogen-infected cells in infectious diseases can be beneficial.
  • compositions comprising the Fc-containing proteins described herein, such as antibodies or Fc fusion proteins.
  • Such compositions comprise a therapeutically effective amount of an Fc-containing protein having an altered Fc region with one or more additional components such as a physiologically acceptable carrier, excipient, or diluent.
  • additional components can include buffers, carbohydrates, polyols, amino acids, chelating agents, stabilizers, and/or preservatives, among many possibilities.
  • compositions comprising Fc-containing proteins comprising an altered Fc region described above can be administered by any appropriate means including, but not limited to, parenteral, topical, oral, nasal, vaginal, rectal, or pulmonary (by inhalation) administration. If injected, the composition(s) can be administered intra-articularly, intravenously, intraarterially, intramuscularly, intraarticularly, intraperitoneally, subcutaneously by bolus injection or continuous infusion. Localized administration, that is, at the site of disease, such as direct injection into a tumor, is contemplated, as are transdermal delivery and sustained release from implants or skin patches.
  • Delivery by inhalation includes, for example, nasal or oral inhalation, use of a nebulizer, inhalation in aerosol form, and the like.
  • Administration via a suppository inserted into a body cavity can be accomplished, for example, by inserting a solid form of the composition in a chosen body cavity and allowing it to dissolve.
  • Other alternatives include eye drops, oral preparations such as pills, lozenges, syrups, and chewing gum, and topical preparations such as lotions, gels, sprays, and ointments.
  • therapeutic molecules that are polypeptides such as those described herein can be administered topically or by injection or inhalation.
  • the Fc-containing proteins described herein can be administered at any dosage, frequency, and duration that can be effective to treat the condition being treated.
  • the therapeutically effective dosage depends on the molecular nature of the Fc-containing protein and the nature of the disorder being treated. Treatment may be continued as long as necessary to achieve the desired results.
  • the Fc-containing protein can be administered as a single dosage or as a series of dosages given periodically, including multiple times per day, daily, every other day, twice a week, three times per week, weekly, every other week, monthly, every six weeks, every two months, every three, four, five or six months, among other possible dosage regimens.
  • the periodicity of treatment may or may not be constant throughout the duration of the treatment.
  • treatment may initially occur at weekly intervals and later occur every other week or at longer intervals as mentioned above. Treatments having durations of days, weeks, months, or years are encompassed by the invention. Treatment may be discontinued and then restarted. Maintenance doses may be administered after an initial treatment.
  • Dosage may be measured as milligrams per kilogram of body weight (mg/kg) or as milligrams per square meter of skin surface (mg/m 2 ) or as a fixed dose, irrespective of height or weight. All of these are standard dosage units in the art. A person's skin surface area is calculated from her height and weight using a standard formula. With respect to the proteins containing an altered Fc region described herein, dosages can range from about 0.01 mg/kg to about 70 mg/kg, optionally from about 0.1 mg/kg to about 20 mg/kg, from about 0.1 mg/kg to about 5 mg/kg, from about 0.3 mg/kg to about 3 mg/kg, or about 2.5 mg/kg.
  • patients of all sizes can receive the same dosage, ranging from about 1 mg to about 500 mg, optionally from about 10 mg to about 100 mg, from about 25 mg to about 50 mg, from about 100 mg to about 300 mg, or from about 100 mg to about 200 mg.
  • the dosage may be from about 5 mg/m 2 to about 800 mg/m 2 , from about 10 mg/m 2 to about 600 mg/m 2 , or from about 25 mg/m 2 to about 500 mg/m 2 .
  • Dosages may or may not be constant throughout the duration of the treatment. For example, dosage may steadily escalate throughout the duration of the treatment.
  • a first dose may be higher than subsequent doses.
  • dosage may be reduced at later stages of the treatment.
  • nucleic acids encoding either an scFv-Fc containing the charge pair substitutions E356K and D399K or an Fc polypeptide chain (“dummy Fc”) containing the charge pair substitutions K392D and K409D, with additional alterations at selected sites within the Fc-encoding regions, were created using PCR. For each site within the Fc selected for substitution, the nucleic acid was changed such that Fc regions with all twenty different amino acids at the selected site would be generated. Each codon in the nucleic acid was randomized independently so that the nucleic acid molecules in the resulting library were each potentially modified within only one codon.
  • the library containing nucleic acids with mutations at sites encoding these residues was referred to as the “Tier 1” library.
  • Another group of sites were within the C H 2 region and were either close to or part of the area that contacts Fc ⁇ RIIIA (239, 241, 255, 256, 258, 264, 265, 267, 268, 269, 270, 272, 276, 280, 285, 286, 290, 294, 295, 296, 298, 300, 307, 309, 315, 326, 327, 328, 330, 332, 333, 334, 337, and 339; see FIG. 2 ).
  • the library containing nucleic acids with mutations at sites encoding these residues was referred to as the “Tier 2” library.
  • a third group included sites within the C H 2 region that were solvent-exposed, but were not close to or part of the area that contacts Fc ⁇ RIIIA (243, 246, 248, 249, 251, 252, 253, 254, 260, 274, 275, 278, 279, 282, 283, 284, 287, 288, 289, 292, 293, 301, 302, 303, 305, 310, 311, 312, 313, 314, 317, 318, 320, 322, 324, 335, 336, 338, and 340; see FIG. 2 ).
  • the library containing nucleic acids with mutations at sites encoding these residues was referred to as the “Tier 3” library.
  • FIG. 2 shows the positions of these sites within a human IgG1 Fc region.
  • a DNA fragment encoding the scFv of the M315 antibody (a rat-anti-mouse NKG2D antibody) fused to a human IgG1 Fc polypeptide with E356K and D399K charge pair mutations in C H 3 domain was subcloned into the mammalian expression vector pTTS. Zhang et al. (2009), Protein Expression and Purification 65(1): 77-82. A DNA fragment encoding a huIgG1 Fc polypeptide with K392D and K409D charge pair mutations in the C H 3 domain was also subcloned into pTTS.
  • the libraries were made as follows. For each of the 82 selected codons within each Fc-encoding region, an oligonucleotide randomized at the first two positions of the codon and having either a G or a C at third position (an “NNG/C codon”) was made (an “NNG/C oligonucleotide”). This NNG/C codon was placed in the middle of the NNG/C oligonucleotide with about 21 bases extending upstream and downstream. The NNG/C oligonucleotide was oriented such that its 5′ end was upstream of its 3′ end in the Fc-encoding region.
  • reverse oligonucleotides that match the upstream 21 bases of the NNG/C oligonucleotides were synthesized individually.
  • a universal downstream primer was combined with each of the NNG/C oligonucleotides and subjected to polymerase chain reaction (PCR) to produce downstream fragments.
  • PCR polymerase chain reaction
  • a universal upstream oligonucleotide and each of the reverse oligonucleotides were combined and subjected to PCR reactions to make upstream DNA fragments.
  • the NNG/C oligonucleotide may point upstream, and the reverse primer may point downstream.
  • the initial PCR reactions described above would include the NNG/C oligonucleotide plus the upstream oligonucleotide in one PCR reaction to produce an upstream fragment and the reverse oligonucleotide and the downstream oligonucleotide in another PCR reaction to produce a downstream fragment.
  • the upstream and downstream PCR fragments were purified using agarose gels, and the amounts of these PCR products were quantified.
  • the same molar amounts of individual upstream and downstream DNA fragments were combined with the universal upstream and downstream primers for a second round PCR reaction to assemble the full length PCR product.
  • Full length PCR fragments were then purified from agarose gels, and equal amounts of individual full length fragments from a tier were combined, digested with restriction enzymes Sal I and BamH I, and inserted into an expression vector.
  • a total of six libraries were made. Three libraries, a Tier 1, a Tier 2, and a Tier 3 library, having mutations in a nucleic acid encoding an scFv-Fc were made. Similarly, a Tier 1, a Tier 2, and a Tier 3 library having mutations at the same positions within the Fc-encoding region in a nucleic acid encoding a dummy Fc were made. As illustrated diagrammatically in FIG. 3 , initial screening was performed as follows. The libraries were introduced into Escherichia coli , and enough individual colonies were picked such that at least three times as many colonies were picked as there were different variants in the library. For example, each Tier 1 library contained twenty different amino acids at each of nine sites, for a total of 180 different variants.
  • ten microtiter plates of colonies (96 wells/plate for a total of about 960) were picked and grown. Plasmid DNA was isolated.
  • Each Tier 2 and Tier 3 library contained twenty different amino acids at each of 34 and 39 sites for a total of 680 and 780 different variants, respectively. Accordingly, 45 plates of colonies (for a total of 4320) were picked for each Tier 2 and Tier 3 library, and plasmid DNA was isolated. These mutated plasmid DNAs were combined with unaltered DNAs (if the altered DNA was an scFv-Fc, the unaltered DNA was a dummy Fc, and vice versa, as shown in FIG.
  • HEK 293 cells a transformed human embryonic kidney cell line
  • Transfectants were cultured, and the culture medium was assayed using an AlphaLISA® assay using reagents purchased from Perkin Elmer (catalog numbers 6760002 and AL109M).
  • the AlphaLISA® assay was performed as follows.
  • Cell culture medium from the transfected HEK 293 cells was added to wells containing streptavidin-coated donor beads (Perkin Elmer catalog number 6760002), a biotinylated human IgG antibody (which binds to the donor beads via the streptavidin-biotin interaction), acceptor beads (Perkin Elmer catalog number AL109M) conjugated to an anti-glutathione S-transferase (GST) antibody, a GST-tagged version of human Fc ⁇ RIIIA (which binds to the acceptor beads via the GST-anti-GST antibody interaction and which binds to the donor beads via the interaction of Fc ⁇ RIIIA with the biotinylated human IgG antibody).
  • streptavidin-coated donor beads Perkin Elmer catalog number 6760002
  • a biotinylated human IgG antibody which binds to the donor beads via the streptavidin-biotin interaction
  • acceptor beads
  • the donor beads are activated. If the acceptor beads are in close physical proximity to the activated donor cells, they will be activated by the donor beads to emit fluorescence at about 615 nm.
  • the cell culture supernatants that inhibited the signal to a greater extent than did supernatants from cells transfected with unmutated (except for the charge pair mutations which were also included in the libraries) versions of the scFv-Fc and dummy Fc were retested twice more to confirm that they were positive. In the third round of testing, tests were performed in duplicate.
  • the Fc-encoding regions of the plasmids encoding these scFv-Fc's or dummy Fc's that yielded a positive signal were sequenced.
  • Tables 1 and 2 below show the data only from these positive transfectants resulting from the Tier 1, 2, and 3 libraries that were mutated in the scFv-Fc- and dummy Fc-encoding nucleic acids, respectively.
  • N-glycosylation site N297-S298-T299
  • P329 site P329 site in either Fc chain
  • substitutions within both of these areas i.e., S298C, S298A, A330M, and A330V
  • S298C, S298A, A330M, and A330V had been found in the primary screen.
  • Combinations that appeared to be favorable based on molecular modeling as discussed below were constructed and tested for binding to Fc ⁇ RIIIA and, in some cases, for activity in an ADCC assay.
  • substitutions at positions S298, A327, and A330 were identified that might improve Fc binding to Fc ⁇ RIII using EGAD. Each of the three positions was changed to all other 19 amino acids in silico, and the change in the stability of the Fc-Fc ⁇ RIII interaction was predicted. EGAD was also used to analyze some of the combinations of mutations that bound well to Fc ⁇ Rs according to the data reported above. Examples of substitutions that EGAD predicted might enhance binding to Fc ⁇ RIIIA include S298C, S298I, S298V, S298T, A327Y, A327W, A327F, A327H, A330H, A330F, and A330M.
  • AlphaLISA® assay confirmed that some of the predicted mutations at positions S298 and A330 showed improved binding to the Fc ⁇ Rs.
  • the combination designated “W23,” which has L234Y, K290Y, and Y296W mutations in one chain and S298T and K334V mutations in the other chain resulted from this approach.
  • Beneficial combinations were selected and incorporated into DNA encoding an anti-human Protein X huIgG1 heavy chain using the SOE by PCR technique described above.
  • Heterodimeric huIgG1s were made by transiently transfecting HEK 293 cells at small scale. The crude supernatants were concentrated, and the buffer was exchanged. In such a way, a panel of heterodimeric huIgG1 antibodies containing novel Fc variants having multiple substitutions was created.
  • FIGS. 4-6 show the percent inhibition of AlphaLISA® signal as a function of the concentration of the competitor antibody. In the Tables 3 and 4 below, such data is presented as an “EC 50 ”, which is the concentration of antibody at which half of the maximal inhibition of the AlphaLISA® signal is achieved.
  • ADCC assays were performed as follows. Cell lines having high (tumor cell line SKBR3), medium (tumor cell line JIMT1), and low (tumor cell line MCF7) Protein X expression were used. These Protein X-expressing target cells were labeled with carboxyfluorescein succinimidyl ester (CFSE) and then washed once with phosphate buffered saline (PBS) before being deposited into 96-well microtiter plates with V-shaped wells. Purified NK cells from an Fc ⁇ RIIIA 158F/158F donor were added to each well.
  • CFSE carboxyfluorescein succinimidyl ester
  • PBS phosphate buffered saline
  • the heterodimeric human anti-Protein X IgG1 antibodies and an isotype-matched control antibody were diluted in a 1:3 series and added to each well.
  • the cells were incubated at 37° C. with 5% CO 2 for 3.5 hrs.
  • the cells were spun down and re-suspended in 1 ⁇ FACS buffer (1 ⁇ phosphate buffered saline (PBS) containing 0.5% fetal bovine serum (FBS)) with the dye TO-PRO®-3 iodide (Molecular Probes, Inc. Corporation, Oregon, USA), which stains dead cells, before analysis by fluorescence activated cell sorting (FACS).
  • the percentage of cell killing was calculated by dividing the number of dead cells (stained TO-PRO®-3 iodide) by number of total cells (stained with CFSE).
  • FIGS. 7-9 show the percentage of cells killed as a function of antibody concentration.
  • EC 50 's determined from such data are shown in Table 3. These data indicated that all fourteen of the antibodies containing variant Fc regions were very potent in killing tumor cells, each having an EC 50 of about 1 pM, which was much lower than the EC 50 of an unaltered antibody or an antibody containing only charge pair mutations. Table 3. Further, lower EC 50 's for ADCC generally correlated with lower EC 50 's for Fc ⁇ RIIIA binding, which would be expected since binding to Fc ⁇ RIIIA is a prerequisite for activity in this ADCC assay.
  • Binding of some of the Fc variants to recombinant human and murine Fc ⁇ Rs was tested using BiacoreTM technology by capturing His-tagged Fc ⁇ Rs using a murine anti-His antibody attached to a Sensor Chip CM5 (Biacore).
  • BiacoreTM technology by capturing His-tagged Fc ⁇ Rs using a murine anti-His antibody attached to a Sensor Chip CM5 (Biacore).
  • Fc ⁇ RIIA with a histidine at position 131 Fc ⁇ RIIIA with a valine at position 158
  • Fc ⁇ RIIIA with a phenylalanine at position 158 were tested.
  • Human IgG1 antibodies containing variant Fc regions were injected over the surface of the Sensor Chip CM5 to which the Fc ⁇ receptor was tethered and allowed to associate and disassociate from the Fc ⁇ receptor for defined times.
  • FIG. 10 shows that a human wild type IgG1 Fc (M01) inhibits IgG1 binding to Fc ⁇ RIIIA (158F) by only about 25% at the highest concentration tested (360 nM).
  • M01 a human wild type IgG1 Fc
  • M04 an IgG1 antibody that contains heterodimerizing alterations (K392D+K409D in one Fc polypeptide chain and E356K+ D399K in other), was only slightly more effective than M01.
  • W117, W125, and an afucosylated wild type human IgG1 competed much more strongly, as evidenced by a shift to the left in the curves. See W117, W125, and AFUCO-M01 in FIG. 10 .
  • Afucosylated preparations of W117 and W125 (AFUCO-W117 and AFUCO-W125) showed a very high affinity for the human Fc ⁇ RIIIA 158F, since these two preparations produced the leftmost curves in FIG. 10 .
  • Fc variants W157 and W165 also exhibited strong competition.
  • FIG. 11 Similar results for binding to Fc ⁇ RIIIA (158V) are shown in FIG. 11 . M01 and M04 exhibited weak competition for binding to Fc ⁇ RIIIA (158V). As in FIG. 10 , AFUCO-W117 and AFUCO-W125 were the most effective competitors, followed by W157 and W165. W117 and AFUCO-IgG1, followed by W125, were less effective, but still far more effective than M01 and M04. These data show that a synergistic enhancement of binding to Fc ⁇ RIIIA can be achieved using defucosylated preparations of IgG1 heterodimeric variants.
  • FIGS. 12 and 13 show the results obtained using SKBR3 cells.
  • Control antibodies M01 having a wild type Fc region
  • M04 having an Fc region containing only heterodimerizing alterations
  • Cell killing dropped off steeply at lower antibody concentrations of M01 and M04.
  • antibodies containing variant Fc regions including W23, W117, W125, W141, W144, W165, W168, and W187, exhibited higher levels of cell killing than either M01 or M04 at most antibody concentrations.
  • Variant W187 exhibited the highest activity in this assay, correlating with the fact that it also exhibited the highest affinity to human Fc ⁇ RIIIA. Table 4.
  • Variants W117, W125, W165, and W168 also elicited potent ADCC activity in this assay.
  • FIG. 14 shows the results (percent specific cell lysis) of an ADCC assay done using full length human IgG1 anti-Protein X antibodies and JIMT1 cells, which express moderate levels of Protein X.
  • M01 antibodies which contain a wild type Fc region
  • a defucosylated preparation of an M01 antibody achieved 86% specific cell lysis at the highest concentration tested and had an EC 50 of 0.274 pM in this assay.
  • FIG. 14 bottom panel. Since the defucoyslated versions of IgG1 antibodies containing either a W117 or a W125 Fc region both had much higher activity than the fucosylated versions of these antibodies, these data indicate a synergistic improvement in ADCC activity when the Fc region of an IgG1 antibody is defucosylated and also contains amino acid changes that increase its affinity to Fc ⁇ RIIIA.
  • Antibodies containing variant Fc regions had K D values for binding to human Fc ⁇ RIIIA, including the 158F and 158V allelic variants, ranging from 6.4 nM to 143 nM.
  • a defucosylated preparation had a K D for binding to human Fc ⁇ RIIIA (158F) about eleven fold lower than that of a fucosylated preparation (compare 9.0 nM to 105 nM) and had an EC 50 in the ADCC assay described in Example 4 that was about 100 fold lower (compare 0.015 pM to 1.15 pM).
  • the increases in activity in the ADCC assay of the defucosylated versus fucosylated preparations of the W117 and W125 antibodies were synergistic since they exceeded expectations based on the increases in binding affinity to Fc ⁇ RIIIA (158F).
US14/005,517 2011-03-16 2012-03-15 Fc VARIANTS Abandoned US20140112926A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US14/005,517 US20140112926A1 (en) 2011-03-16 2012-03-15 Fc VARIANTS

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201161453433P 2011-03-16 2011-03-16
US14/005,517 US20140112926A1 (en) 2011-03-16 2012-03-15 Fc VARIANTS
PCT/US2012/029271 WO2012125850A1 (fr) 2011-03-16 2012-03-15 Variants de fc

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2012/029271 A-371-Of-International WO2012125850A1 (fr) 2011-03-16 2012-03-15 Variants de fc

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/413,810 Continuation US11198732B2 (en) 2011-03-16 2019-05-16 Fc variants

Publications (1)

Publication Number Publication Date
US20140112926A1 true US20140112926A1 (en) 2014-04-24

Family

ID=45922822

Family Applications (2)

Application Number Title Priority Date Filing Date
US14/005,517 Abandoned US20140112926A1 (en) 2011-03-16 2012-03-15 Fc VARIANTS
US16/413,810 Active US11198732B2 (en) 2011-03-16 2019-05-16 Fc variants

Family Applications After (1)

Application Number Title Priority Date Filing Date
US16/413,810 Active US11198732B2 (en) 2011-03-16 2019-05-16 Fc variants

Country Status (8)

Country Link
US (2) US20140112926A1 (fr)
EP (1) EP2686345B1 (fr)
JP (2) JP5972915B2 (fr)
AU (1) AU2012229048B2 (fr)
CA (1) CA2830254C (fr)
ES (1) ES2668895T3 (fr)
MX (1) MX348071B (fr)
WO (1) WO2012125850A1 (fr)

Cited By (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20150056182A1 (en) * 2011-11-30 2015-02-26 Chugai Seiyaku Kabushiki Kaisha Drug containing carrier into cell for forming immune complex
US20150166636A1 (en) * 2012-06-14 2015-06-18 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule containing modified fc region
US9884921B2 (en) 2014-07-01 2018-02-06 Pfizer Inc. Bispecific heterodimeric diabodies and uses thereof
US9890218B2 (en) 2011-06-30 2018-02-13 Chugai Seiyaku Kabushiki Kaisha Heterodimerized polypeptide
WO2018217945A1 (fr) * 2017-05-23 2018-11-29 Dragonfly Therapeutics, Inc. Protéine de liaison nkg2d, cd16 et antigène associé à une tumeur
US10253100B2 (en) 2011-09-30 2019-04-09 Chugai Seiyaku Kabushiki Kaisha Therapeutic antigen-binding molecule with a FcRn-binding domain that promotes antigen clearance
US10618965B2 (en) 2011-02-25 2020-04-14 Chugai Seiyaku Kabushiki Kaisha Method for altering plasma retention and immunogenicity of antigen-binding molecule
US10766960B2 (en) 2012-12-27 2020-09-08 Chugai Seiyaku Kabushiki Kaisha Heterodimerized polypeptide
US10919953B2 (en) 2012-08-24 2021-02-16 Chugai Seiyaku Kabushiki Kaisha FcgammaRIIB-specific Fc region variant
US11130808B2 (en) 2016-05-26 2021-09-28 Qilu Puget Sound Biotherapeutics Corporation Mixtures of antibodies
US11154615B2 (en) 2014-11-11 2021-10-26 Chugai Seiyaku Kabushiki Kaisha Library of antigen-binding molecules including modified antibody variable region
WO2021232163A1 (fr) * 2020-05-20 2021-11-25 Zymeworks Inc. Variants de région fc d'immunoglobuline comprenant des mutations améliorant la stabilité
US11236168B2 (en) 2012-08-24 2022-02-01 Chugai Seiyaku Kabushiki Kaisha Mouse FcγammaRII-specific Fc antibody
US11267868B2 (en) 2013-04-02 2022-03-08 Chugai Seiyaku Kabushiki Kaisha Fc region variant
US11359194B2 (en) 2008-04-11 2022-06-14 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding two or more antigen molecules repeatedly
RU2788531C2 (ru) * 2017-05-23 2023-01-23 Драгонфлай Терапьютикс, Инк. Белок, связывающийся с nkg2d, cd16 и с опухолеспецифическим антигеном
US11739149B2 (en) 2013-11-11 2023-08-29 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule containing modified antibody variable region
US11814438B2 (en) * 2016-07-06 2023-11-14 Laboratoire Français Du Fractionnement Et Des Biotechnologies Fc mutants with improved functional activity
US11827699B2 (en) 2011-09-30 2023-11-28 Chugai Seiyaku Kabushiki Kaisha Methods for producing antibodies promoting disappearance of antigens having plurality of biological activities
US11834506B2 (en) 2017-02-08 2023-12-05 Dragonfly Therapeutics, Inc. Multi-specific binding proteins that bind NKG2D, CD16, and a tumor-associated antigen for activation of natural killer cells and therapeutic uses thereof to treat cancer
US11884732B2 (en) 2017-02-20 2024-01-30 Dragonfly Therapeutics, Inc. Proteins binding HER2, NKG2D and CD16
US11884733B2 (en) 2018-02-08 2024-01-30 Dragonfly Therapeutics, Inc. Antibody variable domains targeting the NKG2D receptor
US11952422B2 (en) 2017-12-05 2024-04-09 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule comprising altered antibody variable region binding CD3 and CD137

Families Citing this family (109)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2749539C (fr) 2009-01-21 2022-07-19 Amgen Inc. Compositions et methodes comprenant des mutants d'interleukine-2 destinees au traitement de maladies inflammatoires et auto-immunes
US9493578B2 (en) 2009-09-02 2016-11-15 Xencor, Inc. Compositions and methods for simultaneous bivalent and monovalent co-engagement of antigens
CA2806252C (fr) 2010-07-29 2019-05-14 Xencor, Inc. Anticorps dont les points isoelectriques sont modifies
WO2012138919A2 (fr) 2011-04-08 2012-10-11 Amgen Inc. Méthode de traitement ou d'amélioration de troubles métaboliques à l'aide du facteur 15 de différenciation de croissance (gdf-15)
CA2833019A1 (fr) 2011-04-22 2012-10-26 Emergent Product Development Seattle, Llc Proteines de liaison a un antigene membranaire specifique de la prostate et compositions et procedes associes
UA117901C2 (uk) 2011-07-06 2018-10-25 Ґенмаб Б.В. Спосіб посилення ефекторної функції вихідного поліпептиду, його варіанти та їх застосування
US10851178B2 (en) 2011-10-10 2020-12-01 Xencor, Inc. Heterodimeric human IgG1 polypeptides with isoelectric point modifications
EP3683228A3 (fr) * 2012-01-26 2020-07-29 Amgen Inc. Polypeptides du facteur de croissance et de différenciation 15 (gdf-15)
AU2013285355A1 (en) 2012-07-06 2015-01-29 Genmab B.V. Dimeric protein with triple mutations
US9714291B2 (en) 2012-10-05 2017-07-25 Kyowa Hakko Kirin Co., Ltd Heterodimer protein composition
EA201500741A1 (ru) * 2013-01-10 2016-01-29 Генмаб Б.В. ВАРИАНТЫ Fc-ОБЛАСТИ IGG1 ЧЕЛОВЕКА И ИХ ПРИМЕНЕНИЕ
US10131710B2 (en) 2013-01-14 2018-11-20 Xencor, Inc. Optimized antibody variable regions
CN110981964B (zh) 2013-01-14 2023-09-15 Xencor股份有限公司 新型异二聚体蛋白
US11053316B2 (en) 2013-01-14 2021-07-06 Xencor, Inc. Optimized antibody variable regions
US9605084B2 (en) 2013-03-15 2017-03-28 Xencor, Inc. Heterodimeric proteins
US9701759B2 (en) 2013-01-14 2017-07-11 Xencor, Inc. Heterodimeric proteins
US10487155B2 (en) 2013-01-14 2019-11-26 Xencor, Inc. Heterodimeric proteins
US10968276B2 (en) 2013-03-12 2021-04-06 Xencor, Inc. Optimized anti-CD3 variable regions
AU2014207549B2 (en) 2013-01-15 2018-12-06 Xencor, Inc. Rapid clearance of antigen complexes using novel antibodies
US9546203B2 (en) * 2013-03-14 2017-01-17 Amgen Inc. Aglycosylated Fc-containing polypeptides with cysteine substitutions
US10858417B2 (en) 2013-03-15 2020-12-08 Xencor, Inc. Heterodimeric proteins
US10519242B2 (en) 2013-03-15 2019-12-31 Xencor, Inc. Targeting regulatory T cells with heterodimeric proteins
AU2014232416B2 (en) 2013-03-15 2017-09-28 Xencor, Inc. Modulation of T Cells with Bispecific Antibodies and FC Fusions
US10106624B2 (en) 2013-03-15 2018-10-23 Xencor, Inc. Heterodimeric proteins
US20140302037A1 (en) 2013-03-15 2014-10-09 Amgen Inc. BISPECIFIC-Fc MOLECULES
WO2014144632A2 (fr) 2013-03-15 2014-09-18 Amgen Inc. Anticorps anti-pac1 humains
AU2014236769B2 (en) 2013-03-15 2018-09-27 Amgen Inc. Heterodimeric bispecific antibodies
EP2992010B1 (fr) * 2013-04-29 2021-03-24 F.Hoffmann-La Roche Ag Anticorps asymétrique modifié de liaison fc-receptor et procédés d'utilisation
PE20190920A1 (es) 2013-04-29 2019-06-26 Hoffmann La Roche Anticuerpos modificados de union a fcrn humano y metodos de utilizacion
AU2014273817B2 (en) * 2013-05-31 2019-03-14 Zymeworks Bc Inc. Heteromultimers with reduced or silenced effector function
LT3027642T (lt) 2013-07-31 2020-10-12 Amgen Inc. Augimo ir diferenciacijos faktoriaus 15(gdf-15) konstruktai
WO2015048272A1 (fr) 2013-09-25 2015-04-02 Amgen Inc. Anticorps v-c-fc-v-c
KR20160104636A (ko) 2014-01-15 2016-09-05 에프. 호프만-라 로슈 아게 단백질 A-결합이 개선된 Fc-영역 변이체
KR102497443B1 (ko) 2014-03-28 2023-02-08 젠코어 인코포레이티드 Cd38 및 cd3에 결합하는 이중특이적 항체
CA2947605A1 (fr) 2014-05-13 2015-11-19 Bioatla, Llc Proteines biologiques conditionnellement actives
SI3186283T1 (sl) * 2014-08-29 2020-04-30 F. Hoffmann-La Roche Ag Kombinirana terapija z imunocitokini različice IL-2, usmerjenimi proti tumorju in protitelesi proti humanemu PD-L1
EP3194435A1 (fr) 2014-09-15 2017-07-26 Amgen Inc. Protéines de liaison antigéniques bispécifiques anti-récepteur cgrp/récepteur pac1 et leurs utilisations
AU2015323313B2 (en) 2014-09-25 2021-04-01 Amgen Inc. Protease-activatable bispecific proteins
SI3215528T1 (sl) * 2014-11-06 2019-11-29 Hoffmann La Roche Variante regije Fc s spremenjeno vezavo FcRn in postopki uporabe
KR20170082594A (ko) * 2014-11-10 2017-07-14 에프. 호프만-라 로슈 아게 항-ang2 항체 및 사용 방법
BR112017009764A2 (pt) 2014-11-10 2018-02-20 Hoffmann La Roche anticorpos biespecíficos e métodos de uso em oftalmologia
JP6993228B2 (ja) * 2014-11-19 2022-03-03 ジェネンテック, インコーポレイテッド 抗トランスフェリン受容体/抗bace1多重特異性抗体および使用方法
US10259887B2 (en) 2014-11-26 2019-04-16 Xencor, Inc. Heterodimeric antibodies that bind CD3 and tumor antigens
PE20171103A1 (es) 2014-11-26 2017-08-07 Xencor Inc Anticuerpos heterodimericos que se unen a cd3 y cd38
CA2967426A1 (fr) 2014-11-26 2016-06-02 Xencor, Inc. Anticorps heterodimeriques se liant a l'antigene cd3 et a un antigene tumoral
US10428155B2 (en) 2014-12-22 2019-10-01 Xencor, Inc. Trispecific antibodies
US10227411B2 (en) 2015-03-05 2019-03-12 Xencor, Inc. Modulation of T cells with bispecific antibodies and FC fusions
UA126270C2 (uk) 2015-04-10 2022-09-14 Емджен Інк. Мутеїни інтерлейкіну-2 для росту регуляторних t-клітин
SG10201912593VA (en) * 2015-07-23 2020-02-27 Boehringer Ingelheim Int Compound targeting il-23a and b-cell activating factor (baff) and uses thereof
WO2017053469A2 (fr) 2015-09-21 2017-03-30 Aptevo Research And Development Llc Polypeptides de liaison à cd3
CN108699136B (zh) 2015-12-07 2022-03-18 Xencor股份有限公司 结合cd3和psma的异二聚抗体
EP3390447A1 (fr) 2015-12-15 2018-10-24 Amgen Inc. Anticorps pacap et leurs utilisations
MX2018012472A (es) 2016-04-15 2019-08-12 Alpine Immune Sciences Inc Proteinas inmunomoduladoras variantes de ligando icos y sus usos.
MA43552A (fr) 2016-04-15 2018-11-07 Alpine Immune Sciences Inc Protéines immunomodulatrices à variants de cd80 et leurs utilisations
RU2022104399A (ru) 2016-06-14 2022-05-05 Ксенкор, Инк. Биспецифические антитела-ингибиторы контрольных точек
AU2017290086A1 (en) 2016-06-28 2019-01-24 Xencor, Inc. Heterodimeric antibodies that bind somatostatin receptor 2
CA3032120A1 (fr) 2016-07-28 2018-02-01 Alpine Immune Sciences, Inc. Proteines immunomodulatrices a variants de cd155 et leurs utilisations
WO2018022945A1 (fr) 2016-07-28 2018-02-01 Alpine Immune Sciences, Inc. Protéines immunomodulatrices à variants de cd112 et utilisations associées
US11471488B2 (en) 2016-07-28 2022-10-18 Alpine Immune Sciences, Inc. CD155 variant immunomodulatory proteins and uses thereof
SG11201900746RA (en) 2016-08-12 2019-02-27 Janssen Biotech Inc Engineered antibodies and other fc-domain containing molecules with enhanced agonism and effector functions
US10793632B2 (en) 2016-08-30 2020-10-06 Xencor, Inc. Bispecific immunomodulatory antibodies that bind costimulatory and checkpoint receptors
CN110214148A (zh) 2016-10-14 2019-09-06 Xencor股份有限公司 含有IL-15/IL-15Rα Fc融合蛋白和PD-1抗体片段的双特异性异源二聚体融合蛋白
AU2018221731C1 (en) 2017-02-17 2021-11-18 Denali Therapeutics Inc. Engineered transferrin receptor binding polypeptides
SG11201907288RA (en) 2017-02-28 2019-09-27 Seattle Genetics Inc Cysteine mutated antibodies for conjugation
FI3596116T3 (fi) 2017-03-16 2023-11-09 Alpine Immune Sciences Inc Immunomodulatorisia pd-l1-varianttiproteiineja ja niiden käyttöjä
NZ756395A (en) 2017-03-16 2024-01-26 Alpine Immune Sciences Inc Cd80 variant immunomodulatory proteins and uses thereof
JP2020511144A (ja) 2017-03-16 2020-04-16 アルパイン イミューン サイエンシズ インコーポレイテッド Pd−l2バリアント免疫調節タンパク質及びその使用
US20200172628A1 (en) 2017-06-22 2020-06-04 Novartis Ag Antibody molecules to cd73 and uses thereof
MA49517A (fr) 2017-06-30 2020-05-06 Xencor Inc Protéines de fusion fc hétérodimères ciblées contenant il-15/il-15ra et domaines de liaison à l'antigène
MA50360A (fr) 2017-10-10 2020-08-19 Alpine Immune Sciences Inc Protéines immunomodulatrices de variants de ctla-4 et leurs utilisations
KR20200085777A (ko) 2017-10-18 2020-07-15 알파인 이뮨 사이언시즈, 인코포레이티드 변이체 icos 리간드 면역조절 단백질 및 관련 조성물 및 방법
US10981992B2 (en) 2017-11-08 2021-04-20 Xencor, Inc. Bispecific immunomodulatory antibodies that bind costimulatory and checkpoint receptors
JP2021502100A (ja) 2017-11-08 2021-01-28 ゼンコア インコーポレイテッド 新規抗pd−1配列を用いた二重特異性および単一特異性抗体
EP3717907A1 (fr) 2017-11-30 2020-10-07 Novartis AG Récepteur d'antigène chimérique ciblant le bcma et ses utilisations
JP2021506291A (ja) 2017-12-19 2021-02-22 ゼンコア インコーポレイテッド 改変されたil−2 fc融合タンパク質
AU2019205273B2 (en) 2018-01-03 2024-04-04 Alpine Immune Sciences, Inc. Multi-domain immunomodulatory proteins and methods of use thereof
KR20200110362A (ko) 2018-01-12 2020-09-23 암젠 인크 Pac1 항체 및 이의 용도
US20220025037A1 (en) * 2018-04-03 2022-01-27 Dragonfly Therapeutics, Inc. Antibody variable domains targeting dll3, and use thereof
CN112469477A (zh) 2018-04-04 2021-03-09 Xencor股份有限公司 与成纤维细胞活化蛋白结合的异源二聚体抗体
PE20201350A1 (es) 2018-04-09 2020-11-30 Amgen Inc Proteinas de fusion del factor de diferenciacion de crecimiento 15
EP3781599A1 (fr) 2018-04-18 2021-02-24 Xencor, Inc. Protéines de fusion hétérodimères ciblant pd-1 contenant des protéines de fusion fc d'il-15/il-15ra, domaines de liaison à l'antigène pd-1 et utilisations associées
CN112437777A (zh) 2018-04-18 2021-03-02 Xencor股份有限公司 包含IL-15/IL-15RA Fc融合蛋白和TIM-3抗原结合结构域的靶向TIM-3的异源二聚体融合蛋白
AR126019A1 (es) 2018-05-30 2023-09-06 Novartis Ag Anticuerpos frente a entpd2, terapias de combinación y métodos de uso de los anticuerpos y las terapias de combinación
CA3098420A1 (fr) 2018-06-01 2019-12-05 Novartis Ag Molecules de liaison dirigees contre bcma et leurs utilisations
WO2019241758A1 (fr) 2018-06-15 2019-12-19 Alpine Immune Sciences, Inc. Protéines immunomodulatrices de variants de pd-l1 et utilisations associées
WO2020045545A1 (fr) * 2018-08-29 2020-03-05 中外製薬株式会社 Demi-molécule d'anticorps, et procédé d'inhibition de la formation d'homodimère de la demi-molécule d'anticorps
CN113544144A (zh) 2018-09-19 2021-10-22 高山免疫科学股份有限公司 变体cd80融合蛋白和相关构建体的方法和用途
US11358999B2 (en) 2018-10-03 2022-06-14 Xencor, Inc. IL-12 heterodimeric Fc-fusion proteins
AU2019389151A1 (en) 2018-11-30 2021-06-10 Alpine Immune Sciences, Inc. CD86 variant immunomodulatory proteins and uses thereof
TW202039573A (zh) 2018-12-21 2020-11-01 美商亞維代堤生物科學公司 抗轉鐵蛋白受體抗體及其用途
CA3132185A1 (fr) 2019-03-01 2020-09-10 Xencor, Inc. Anticorps heterodimeres qui se lient a enpp3 et cd3
JP2022529059A (ja) 2019-04-17 2022-06-16 アルパイン イミューン サイエンシズ インコーポレイテッド バリアントicosリガンド(icosl)融合タンパク質の方法および使用
EP4031578A1 (fr) 2019-09-18 2022-07-27 Novartis AG Anticorps d'entpd2, polythérapies, et procédés d'utilisation des anticorps et des polythérapies
WO2021131021A1 (fr) * 2019-12-27 2021-07-01 中外製薬株式会社 Anticorps anti-ctla-4 et son utilisation
JP2023513035A (ja) * 2020-01-29 2023-03-30 コリア ユニバーシティ リサーチ アンド ビジネス ファウンデーション pH感応性FC変異体
JP2023527638A (ja) 2020-03-27 2023-06-30 アビディティー バイオサイエンシーズ,インク. 顔面肩甲上腕型筋ジストロフィーを処置するための組成物および方法
CN116903727A (zh) 2020-05-08 2023-10-20 高山免疫科学股份有限公司 具有和不具有t细胞抑制蛋白的april和baff抑制性免疫调节蛋白及其使用方法
US11919956B2 (en) 2020-05-14 2024-03-05 Xencor, Inc. Heterodimeric antibodies that bind prostate specific membrane antigen (PSMA) and CD3
KR102607909B1 (ko) 2020-08-19 2023-12-01 젠코어 인코포레이티드 항-cd28 조성물
WO2022044248A1 (fr) * 2020-08-28 2022-03-03 中外製薬株式会社 Polypeptide fc hétérodimère
US20230303665A1 (en) 2020-08-28 2023-09-28 Sana Biotechnology, Inc. Modified anti-viral binding agents
KR20230104651A (ko) 2020-11-06 2023-07-10 노파르티스 아게 Cd19 결합 분자 및 이의 용도
CA3212665A1 (fr) 2021-03-09 2022-09-15 Xencor, Inc. Anticorps heterodimeriques se liant a cd3 et a cldn6
KR20230154311A (ko) 2021-03-10 2023-11-07 젠코어 인코포레이티드 Cd3 및 gpc3에 결합하는 이종이량체 항체
KR20240019124A (ko) 2021-05-07 2024-02-14 알파인 이뮨 사이언시즈, 인코포레이티드 Taci-fc 융합 면역조절 단백질의 투여 및 치료 방법
KR20240024255A (ko) 2021-06-25 2024-02-23 추가이 세이야쿠 가부시키가이샤 항ctla-4 항체의 사용
CR20240026A (es) 2021-06-25 2024-03-14 Chugai Seiyaku Kk Anticuerpo anti-ctla-4
WO2023172883A1 (fr) 2022-03-07 2023-09-14 Alpine Immune Sciences, Inc. Protéines immunomodulatrices de polypeptides cd80 variants, thérapies cellulaires associées et méthodes et utilisations associés
WO2024077018A2 (fr) 2022-10-04 2024-04-11 Alpine Immune Sciences, Inc. Procédés et utilisations d'une protéine immunomodulatrice de fusion taci-fc

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050079184A1 (en) * 2003-08-08 2005-04-14 Immunomedics, Inc. Bispecific antibodies for inducing apoptosis of tumor and diseased cells
US20060235208A1 (en) * 2002-09-27 2006-10-19 Xencor, Inc. Fc variants with optimized properties
WO2007005612A2 (fr) * 2005-07-01 2007-01-11 Medimmune, Inc. Approche integree permettant de generer des applications therapeutiques a base de proteine multidomaine
US20080089892A1 (en) * 2004-01-12 2008-04-17 Eli Lilly And Co. Fc Region Variants
US20090042250A1 (en) * 2007-07-12 2009-02-12 Sangamo Biosciences, Inc. Methods and compositions for inactivating alpha 1,6 fucosyltransferase (FUT8) gene expression
US20100286374A1 (en) * 2008-01-07 2010-11-11 Gunasekaran Kannan Method for making antibody fc-heterodimeric molecules using electrostatic steering effects
US7951917B1 (en) * 1997-05-02 2011-05-31 Genentech, Inc. Method for making multispecific antibodies having heteromultimeric and common components
US20120244578A1 (en) * 2009-11-23 2012-09-27 Amgen Inc. Monomeric antibody fc

Family Cites Families (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1990005183A1 (fr) 1988-10-31 1990-05-17 Immunex Corporation Recepteurs d'interleukine-4
WO1991018982A1 (fr) 1990-06-05 1991-12-12 Immunex Corporation Recepteurs de l'interleukine-1 de type ii
US5731168A (en) 1995-03-01 1998-03-24 Genentech, Inc. Method for making heteromultimeric polypeptides
US20020062010A1 (en) 1997-05-02 2002-05-23 Genentech, Inc. Method for making multispecific antibodies having heteromultimeric and common components
US6737056B1 (en) * 1999-01-15 2004-05-18 Genentech, Inc. Polypeptide variants with altered effector function
US20040132101A1 (en) * 2002-09-27 2004-07-08 Xencor Optimized Fc variants and methods for their generation
JP2006524039A (ja) * 2003-01-09 2006-10-26 マクロジェニクス,インコーポレーテッド 変異型Fc領域を含む抗体の同定および作製ならびにその利用法
WO2006104989A2 (fr) * 2005-03-29 2006-10-05 Verenium Corporation Anticorps a regions fc modifiees et utilisations
US7820790B2 (en) 2005-07-13 2010-10-26 Amgen Mountain View Inc. IL-6 binding proteins
AR056142A1 (es) 2005-10-21 2007-09-19 Amgen Inc Metodos para generar el anticuerpo igg monovalente
US20080112961A1 (en) * 2006-10-09 2008-05-15 Macrogenics, Inc. Identification and Engineering of Antibodies with Variant Fc Regions and Methods of Using Same
JP6071165B2 (ja) 2007-05-31 2017-02-01 ゲンマブ エー/エス 安定なIgG4抗体
AU2009256250B2 (en) 2008-06-03 2013-05-30 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
JP6167040B2 (ja) * 2010-11-05 2017-07-19 ザイムワークス,インコーポレイテッド Fcドメイン中に突然変異を有する、安定したヘテロ二量体抗体の設計
TWI687441B (zh) * 2011-06-30 2020-03-11 中外製藥股份有限公司 異源二聚化多胜肽

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7951917B1 (en) * 1997-05-02 2011-05-31 Genentech, Inc. Method for making multispecific antibodies having heteromultimeric and common components
US20060235208A1 (en) * 2002-09-27 2006-10-19 Xencor, Inc. Fc variants with optimized properties
US20050079184A1 (en) * 2003-08-08 2005-04-14 Immunomedics, Inc. Bispecific antibodies for inducing apoptosis of tumor and diseased cells
US20080089892A1 (en) * 2004-01-12 2008-04-17 Eli Lilly And Co. Fc Region Variants
WO2007005612A2 (fr) * 2005-07-01 2007-01-11 Medimmune, Inc. Approche integree permettant de generer des applications therapeutiques a base de proteine multidomaine
US20090042250A1 (en) * 2007-07-12 2009-02-12 Sangamo Biosciences, Inc. Methods and compositions for inactivating alpha 1,6 fucosyltransferase (FUT8) gene expression
US20100286374A1 (en) * 2008-01-07 2010-11-11 Gunasekaran Kannan Method for making antibody fc-heterodimeric molecules using electrostatic steering effects
US20120244578A1 (en) * 2009-11-23 2012-09-27 Amgen Inc. Monomeric antibody fc

Cited By (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11359194B2 (en) 2008-04-11 2022-06-14 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding two or more antigen molecules repeatedly
US11371039B2 (en) 2008-04-11 2022-06-28 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding to two or more antigen molecules repeatedly
US10618965B2 (en) 2011-02-25 2020-04-14 Chugai Seiyaku Kabushiki Kaisha Method for altering plasma retention and immunogenicity of antigen-binding molecule
US11718678B2 (en) 2011-02-25 2023-08-08 Chugai Seiyaku Kabushiki Kaisha Method for altering plasma retention and immunogenicity of antigen-binding molecule
US9890218B2 (en) 2011-06-30 2018-02-13 Chugai Seiyaku Kabushiki Kaisha Heterodimerized polypeptide
US10253100B2 (en) 2011-09-30 2019-04-09 Chugai Seiyaku Kabushiki Kaisha Therapeutic antigen-binding molecule with a FcRn-binding domain that promotes antigen clearance
US11827699B2 (en) 2011-09-30 2023-11-28 Chugai Seiyaku Kabushiki Kaisha Methods for producing antibodies promoting disappearance of antigens having plurality of biological activities
US11820793B2 (en) * 2011-11-30 2023-11-21 Chugai Seiyaku Kabushiki Kaisha Drug containing carrier into cell for forming immune complex
US20150056182A1 (en) * 2011-11-30 2015-02-26 Chugai Seiyaku Kabushiki Kaisha Drug containing carrier into cell for forming immune complex
US20150166636A1 (en) * 2012-06-14 2015-06-18 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule containing modified fc region
US11142563B2 (en) * 2012-06-14 2021-10-12 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule containing modified Fc region
US10919953B2 (en) 2012-08-24 2021-02-16 Chugai Seiyaku Kabushiki Kaisha FcgammaRIIB-specific Fc region variant
US11236168B2 (en) 2012-08-24 2022-02-01 Chugai Seiyaku Kabushiki Kaisha Mouse FcγammaRII-specific Fc antibody
US10766960B2 (en) 2012-12-27 2020-09-08 Chugai Seiyaku Kabushiki Kaisha Heterodimerized polypeptide
US11267868B2 (en) 2013-04-02 2022-03-08 Chugai Seiyaku Kabushiki Kaisha Fc region variant
US11739149B2 (en) 2013-11-11 2023-08-29 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule containing modified antibody variable region
US9884921B2 (en) 2014-07-01 2018-02-06 Pfizer Inc. Bispecific heterodimeric diabodies and uses thereof
US11154615B2 (en) 2014-11-11 2021-10-26 Chugai Seiyaku Kabushiki Kaisha Library of antigen-binding molecules including modified antibody variable region
US11130808B2 (en) 2016-05-26 2021-09-28 Qilu Puget Sound Biotherapeutics Corporation Mixtures of antibodies
US11814438B2 (en) * 2016-07-06 2023-11-14 Laboratoire Français Du Fractionnement Et Des Biotechnologies Fc mutants with improved functional activity
US11834506B2 (en) 2017-02-08 2023-12-05 Dragonfly Therapeutics, Inc. Multi-specific binding proteins that bind NKG2D, CD16, and a tumor-associated antigen for activation of natural killer cells and therapeutic uses thereof to treat cancer
US11884732B2 (en) 2017-02-20 2024-01-30 Dragonfly Therapeutics, Inc. Proteins binding HER2, NKG2D and CD16
RU2788531C2 (ru) * 2017-05-23 2023-01-23 Драгонфлай Терапьютикс, Инк. Белок, связывающийся с nkg2d, cd16 и с опухолеспецифическим антигеном
CN111278460A (zh) * 2017-05-23 2020-06-12 蜻蜓疗法股份有限公司 结合nkg2d、cd16和肿瘤相关抗原的蛋白质
WO2018217945A1 (fr) * 2017-05-23 2018-11-29 Dragonfly Therapeutics, Inc. Protéine de liaison nkg2d, cd16 et antigène associé à une tumeur
RU2816716C2 (ru) * 2017-09-07 2024-04-03 Драгонфлай Терапьютикс, Инк. Белки, связывающие NKG2D, CD16 и опухолеассоциированный антиген
US11952422B2 (en) 2017-12-05 2024-04-09 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule comprising altered antibody variable region binding CD3 and CD137
US11884733B2 (en) 2018-02-08 2024-01-30 Dragonfly Therapeutics, Inc. Antibody variable domains targeting the NKG2D receptor
US11939384B1 (en) 2018-02-08 2024-03-26 Dragonfly Therapeutics, Inc. Antibody variable domains targeting the NKG2D receptor
WO2021232163A1 (fr) * 2020-05-20 2021-11-25 Zymeworks Inc. Variants de région fc d'immunoglobuline comprenant des mutations améliorant la stabilité

Also Published As

Publication number Publication date
AU2012229048B2 (en) 2016-01-21
ES2668895T3 (es) 2018-05-23
JP5972915B2 (ja) 2016-08-17
MX2013010469A (es) 2014-02-06
EP2686345B1 (fr) 2018-04-25
US11198732B2 (en) 2021-12-14
CA2830254C (fr) 2019-09-10
JP2014509857A (ja) 2014-04-24
MX348071B (es) 2017-05-26
WO2012125850A1 (fr) 2012-09-20
CA2830254A1 (fr) 2012-09-20
US20200010545A1 (en) 2020-01-09
JP2016172729A (ja) 2016-09-29
AU2012229048A1 (en) 2013-09-19
EP2686345A1 (fr) 2014-01-22

Similar Documents

Publication Publication Date Title
US11198732B2 (en) Fc variants
US20230077648A1 (en) Heterodimeric Bispecific Antibodies
US20230055407A1 (en) V-c-fc-v-c antibody
US8815237B2 (en) Aglycosylated immunoglobulin mutants
JP2023051968A (ja) 多重特異的NKp46結合タンパク質
JP6404313B2 (ja) ヘテロ二量体性二重特異性抗体
TW201843177A (zh) 具有經受限cd3結合的多重特異性多肽構築體及使用其之方法
EP3589655B1 (fr) Diabody en tandem pour l'engagement de cellules nk dirigées contre cd16a
WO2006105062A2 (fr) Régions fc d'anticorps altérées et utilisations de celles-ci
CA3070272A1 (fr) Proteines de liaison a l'antigene se liant a 5t4 et 4-1bb, compositions et procedes associes
WO2008030564A2 (fr) Anticorps aglycosylés et procédés de fabrication et d'utilisation de ces anticorps
US20230416364A1 (en) Methods of redirecting of il-2 to target cells of interest
RU2788521C2 (ru) Биспецифические иммуномодулирующие антитела, которые связывают костимуляторные рецепторы и рецепторы контрольных точек
WO2023010098A2 (fr) Nouvelles molécules multi-spécifiques

Legal Events

Date Code Title Description
AS Assignment

Owner name: AMGEN INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:LIU, ZHI;KANNAN, GUNASEKARAN;YAN, WEI;SIGNING DATES FROM 20131126 TO 20131213;REEL/FRAME:031938/0902

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION