US20110293632A1 - Polypeptide variants with altered effector function - Google Patents

Polypeptide variants with altered effector function Download PDF

Info

Publication number
US20110293632A1
US20110293632A1 US11/947,982 US94798207A US2011293632A1 US 20110293632 A1 US20110293632 A1 US 20110293632A1 US 94798207 A US94798207 A US 94798207A US 2011293632 A1 US2011293632 A1 US 2011293632A1
Authority
US
United States
Prior art keywords
region
binding
antibody
amino acid
polypeptide
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
US11/947,982
Inventor
Leonard Presta
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.)
Genentech Inc
Original Assignee
Genentech 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 Genentech Inc filed Critical Genentech Inc
Priority to US11/947,982 priority Critical patent/US20110293632A1/en
Publication of US20110293632A1 publication Critical patent/US20110293632A1/en
Priority to US13/860,462 priority patent/US20140342404A1/en
Priority to US13/946,920 priority patent/US20140147436A1/en
Priority to US14/835,494 priority patent/US20160060330A1/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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/42Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against immunoglobulins
    • C07K16/4283Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against immunoglobulins against an allotypic or isotypic determinant on Ig
    • C07K16/4291Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against immunoglobulins against an allotypic or isotypic determinant on Ig against IgE
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • 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
    • 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
    • C07K16/2839Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the integrin superfamily
    • 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
    • C07K16/2887Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against CD20
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/10Immunoglobulins specific features characterized by their source of isolation or production
    • C07K2317/14Specific host cells or culture conditions, e.g. components, pH or temperature
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • 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
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/71Decreased effector function due to an Fc-modification
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • C07K2317/732Antibody-dependent cellular cytotoxicity [ADCC]
    • 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
    • C07K2317/734Complement-dependent cytotoxicity [CDC]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/30Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2799/00Uses of viruses
    • C12N2799/02Uses of viruses as vector
    • C12N2799/021Uses of viruses as vector for the expression of a heterologous nucleic acid
    • C12N2799/022Uses of viruses as vector for the expression of a heterologous nucleic acid where the vector is derived from an adenovirus

Definitions

  • the present invention concerns polypeptides comprising a variant Fc region. More particularly, the present invention concerns Fc region-containing polypeptides that have altered effector function as a consequence of one or more amino acid modifications in the Fc region thereof.
  • Antibodies are proteins which exhibit binding specificity to a specific antigen.
  • Native antibodies are usually heterotetrameric glycoproteins of about 150,000 daltons, composed of two identical light (L) chains and two identical heavy (H) chains. Each light chain is linked to a heavy chain by one covalent disulfide bond, while the number of disulfide linkages varies between the heavy chains of different immunoglobulin isotypes. Each heavy and light chain also has regularly spaced intrachain disulfide bridges. Each heavy chain has at one end a variable domain (V H ) followed by a number of constant domains.
  • V H variable domain
  • Each light chain has a variable domain at one end (V L ) and a constant domain at its other end; the constant domain of the light chain is aligned with the first constant domain of the heavy chain, and the light chain variable domain is aligned with the variable domain of the heavy chain. Particular amino acid residues are believed to form an interlace between the light and heavy chain variable domains.
  • variable refers to the fact that certain portions of the variable domains differ extensively in sequence among antibodies and are responsible for the binding specificity of each particular antibody for its particular antigen. However, the variability is not evenly distributed through the variable domains of antibodies. It is concentrated in three segments called complementarity determining regions (CDRs) both in the light chain and the heavy chain variable domains. The more highly conserved portions of the variable domains are called the framework regions (FRs).
  • CDRs complementarity determining regions
  • FRs framework regions
  • the variable domains of native heavy and light chains each comprise four FRs, largely adopting a ⁇ -sheet configuration, connected by three CDRs, which form loops connecting, and in some cases forming part of, the ⁇ -sheet structure.
  • the CDRs in each chain are held together in close proximity by the FRs and, with the CDRs from the other chain, contribute to the formation of the antigen binding site of antibodies (see Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md. (1991)).
  • the constant domains are not involved directly in binding an antibody to an antigen, but exhibit various effector functions.
  • antibodies or immunoglobulins can be assigned to different classes.
  • the heavy chain constant regions that correspond to the different classes of immunoglobulins are called ⁇ , ⁇ , ⁇ , ⁇ , and ⁇ , respectively.
  • human immunoglobulin classes only human IgG1, IgG2, IgG3 and IgM are known to activate complement; and human IgG1 and IgG3 mediate ADCC more effectively than IgG2 and IgG4.
  • FIG. 1 A schematic representation of the native IgG1 structure is shown in FIG. 1 , where the various portions of the native antibody molecule are indicated.
  • Papain digestion of antibodies produces two identical antigen binding fragments, called Fab fragments, each with a single antigen binding site, and a residual “Fc” fragment, whose name reflects its ability to crystallize readily.
  • the crystal structure of the human IgG Fc region has been determined (Deisenhofer, Biochemistiy 20:2361-2370 (1981)).
  • the Fc region is generated by papain cleavage N-terminal to Cys 226.
  • the Fc region is central to the effector functions of antibodies.
  • effector functions mediated by the antibody Fc region can be divided into two categories: (1) effector functions that operate after the binding of antibody to an antigen (these functions involve the participation of the complement cascade or Fc receptor (FcR)-bearing cells); and (2) effector functions that operate independently of antigen binding (these functions confer persistence in the circulation and the ability to be transferred across cellular barriers by transcytosis).
  • effector functions that operate after the binding of antibody to an antigen these functions involve the participation of the complement cascade or Fc receptor (FcR)-bearing cells
  • effector functions that operate independently of antigen binding these functions confer persistence in the circulation and the ability to be transferred across cellular barriers by transcytosis.
  • an antibody While binding of an antibody to the requisite antigen has a neutralizing effect that might prevent the binding of a foreign antigen to its endogenous target (e.g. receptor or ligand), binding alone may not remove the foreign antigen.
  • an antibody To be efficient in removing and/or destructing foreign antigens, an antibody should be endowed with both high affinity binding to its antigen, and efficient effector functions.
  • ADCC antibody-dependent cell-mediated cytotoxicity
  • CDC complement dependent cytotoxicity
  • Fc receptors which bind the Fc region of an antibody.
  • FcRs are defined by their specificity for immunoglobulin isotypes; Fc receptors for IgG antibodies are referred to as Fc ⁇ R, for IgE as FcER, for IgA as FcaR and so on.
  • Fc ⁇ RI CD64
  • Fc ⁇ RII CD32
  • Fc ⁇ RIII CD16
  • the three genes encoding the Fc ⁇ RI subclass are clustered in region 1q21.1 of the long arm of chromosome 1; the genes encoding Fc ⁇ RII isoforms (Fc ⁇ RIIA, Fc ⁇ RIIB and Fc ⁇ RIIC) and the two genes encoding Fc ⁇ RIII (Fc ⁇ RIIIA and Fc ⁇ RIIIB) are all clustered in region 1q22.
  • Fc ⁇ RIIA, Fc ⁇ RIIB and Fc ⁇ RIIC the genes encoding Fc ⁇ RIII
  • Fc ⁇ RIIIA and Fc ⁇ RIIIB are all clustered in region 1q22.
  • Fc ⁇ RIIIB is found only on neutrophils
  • Fc ⁇ RIIIA is found on macrophages, monocytes, natural killer (NK) cells, and a subpopulation of T-cells.
  • Fc ⁇ RIIIA is the only FcR present on NK cells, one of the cell types implicated in ADCC.
  • Fc ⁇ RI, Fc ⁇ RII and Fc ⁇ RIII are immunoglobulin superfamily (IgSF) receptors; Fc ⁇ RI has three IgSF domains in its extracellular domain, while Fc ⁇ RII and Fc ⁇ RIII have only two IgSF domains in their extracellular domains.
  • IgSF immunoglobulin superfamily
  • FcRn neonatal Fc receptor
  • MHC major histocompatibility complex
  • G316-K338 human IgG for human Fc ⁇ RI (by sequence comparison only; no substitution mutants were evaluated) (Woof et al. Molec. Immunol. 23:319-330 (1986)); K274-R301 (human IgG1) for human Fc ⁇ RIII (based on peptides) (Sarmay et al. Molec. Immunol. 21:43-51 (1984)); Y407-R416 (human IgG) for human Fc ⁇ RIII (based on peptides) (Gergely et al., Biochem. Soc. Trans.
  • Pro331 in IgG3 was changed to Ser, and the affinity of this variant to target cells analyzed.
  • the affinity was found to be six-fold lower than that of unmutated IgG3, indicating the involvement of Pro331 in Fc ⁇ RI binding.
  • C1q is a hexavalent molecule with a molecular weight of approximately 460,000 and a structure likened to a bouquet of tulips in which six collagenous “stalks” are connected to six globular head regions.
  • CDC complement dependent cytotoxicity
  • the residue Pro331 has been implicated in C1q binding by analysis of the ability of human IgG subclasses to carry out complement mediated cell lysis. Mutation of Ser331 to Pro331 in IgG4 conferred the ability to activate complement. (Tao et al., J. Exp. Med., 178:661-667 (1993); Brekke et al., Eur. J. Immunol., 24:2542-47 (1994)).
  • IgG ability of IgG to bind C1q and activate the complement cascade also depends on the presence, absence, or modification of the carbohydrate moiety positioned between the two CH2 domains (which is normally anchored at Asn297). Ward and Ghetie, Therapeutic Immunology 2:77-94 (1995) at page 81.
  • the present invention provides a variant of a parent polypeptide comprising an Fc region, which variant mediates antibody-dependent cell-mediated cytotoxicity (ADCC) in the presence of human effector cells more effectively, or binds an Fc gamma receptor (Fc ⁇ R) with better affinity, than the parent polypeptide and comprises at least one amino acid modification in the Fc region.
  • the polypeptide variant may, for example, comprise an antibody or an immunoadhesin.
  • the Fc region of the parent polypeptide preferably comprises a human Fc region; e.g., a human IgG1, IgG2, IgG3 or IgG4 Fc region.
  • the polypeptide variant preferably comprises an amino acid modification (e.g.
  • the invention provides a polypeptide comprising a variant Fc region with altered Fc gamma receptor (Fc ⁇ R) binding affinity, which polypeptide comprises an amino acid modification at any one or more of amino acid positions 238, 239, 248, 249, 252, 254, 255, 256, 258, 265, 267, 268, 269, 270, 272, 276, 278, 280, 283, 285, 286, 289, 290, 292, 293, 294, 295, 296, 298, 301, 303, 305, 307, 309, 312, 315, 320, 322, 324, 326, 327, 329, 330, 331, 333, 334, 335, 337, 338, 340, 360, 373, 376, 378, 382, 388, 389, 398, 414, 416, 419, 430, 434, 435, 437, 438 or 439 of the Fc region, wherein the numbering of the residues in the Fc region is that of
  • the variant Fc region preferably comprises a variant human IgG Fc region, e.g., a variant human IgG1, IgG2, IgG3 or IgG4 Fc region.
  • a variant human IgG Fc region e.g., a variant human IgG1, IgG2, IgG3 or IgG4 Fc region.
  • different residues from those identified herein were thought to impact FcR binding.
  • IgG E318 was found to be important for binding (Lund et al., Molec. Immunol. 27(1):53-59 (1992)), whereas E318A had no effect in the human IgG/human Fc ⁇ RII system (Table 6 below).
  • the polypeptide variant with altered Fc ⁇ R binding activity displays reduced binding to an Fc ⁇ R and comprises an amino acid modification at any one or more of amino acid positions 238, 239, 248, 249, 252, 254, 265, 268, 269, 270, 272, 278, 289, 292, 293, 294, 295, 296, 298, 301, 303, 322, 324, 327, 329, 333, 335, 338, 340, 373, 376, 382, 388, 389, 414, 416, 419, 434, 435, 437, 438 or 439 of the Fc region, wherein the numbering of the residues in the Fc region is that of the EU index as in Kabat.
  • the polypeptide variant may display reduced binding to an Fc ⁇ RI and comprise an amino acid modification at any one or more of amino acid positions 238, 265, 269, 270, 327 or 329 of the Fc region, wherein the numbering of the residues in the Fc region is that of the EU index as in Kabat.
  • the polypeptide variant may display reduced binding to an Fc ⁇ RII and comprise an amino acid modification at any one or more of amino acid positions 238, 265, 269, 270, 292, 294, 295, 298, 303, 324, 327, 329, 333, 335, 338, 373, 376, 414, 416, 419, 435, 438 or 439 of the Fc region, wherein the numbering of the residues in the Fc region is that of the EU index as in Kabat.
  • the polypeptide variant of interest may display reduced binding to an Fc ⁇ RIII and comprise an amino acid modification at one or more of amino acid positions 238, 239, 248, 249, 252, 254, 265, 268, 269, 270, 272, 278, 289, 293, 294, 295, 296, 301, 303, 322, 327, 329, 338, 340, 373, 376, 382, 388, 389, 416, 434, 435 or 437 of the Fc region, wherein the numbering of the residues in the Fc region is that of the EU index as in Kabat.
  • the polypeptide variant with altered Fc ⁇ R binding affinity displays improved binding to the Fc ⁇ R and comprises an amino acid modification at any one or more of amino acid positions 255, 256, 258, 267, 268, 272, 276, 280, 283, 285, 286, 290, 298, 301, 305, 307, 309, 312, 315, 320, 322, 326, 330, 331, 333, 334, 337, 340, 360, 378, 398 or 430 of the Fc region, wherein the numbering of the residues in the Fc region is that of the EU index as in Kabat.
  • polypeptide variant may display increased binding to an Fc ⁇ RIII and, optionally, may further display decreased binding to an Fc ⁇ RII.
  • An exemplary such variant comprises amino acid modification(s) at position(s) 298 and/or 333 of the Fc region, wherein the numbering of the residues in the Fc region is that of the EU index as in Kabat.
  • the polypeptide variant may display increased binding to an Fc ⁇ RII and comprise an amino acid modification at any one or more of amino acid positions 255, 256, 258, 267, 268, 272, 276, 280, 283, 285, 286, 290, 301, 305, 307, 309, 312, 315, 320, 322, 326, 330, 331, 337, 340, 378, 398 or 430 of the Fc region, wherein the numbering of the residues in the Fc region is that of the EU index as in Kabat.
  • Such polypeptide variants with increased binding to an Fc ⁇ RII may optionally further display decreased binding to an Fc ⁇ RIII and may, for example, comprise an amino acid modification at any one or more of amino acid positions 268, 272, 298, 301, 322 or 340 of the Fc region, wherein the numbering of the residues in the Fc region is that of the EU index as in Kabat.
  • the invention further provides a polypeptide comprising a variant Fc region with altered neonatal Fc receptor (FcRn) binding affinity, which polypeptide comprises an amino acid modification at any one or more of amino acid positions 238, 252, 253, 254, 255, 256, 265, 272, 286, 288, 303, 305, 307, 309, 311, 312, 317, 340, 356, 360, 362, 376, 378, 380, 382, 386, 388, 400, 413, 415, 424, 433, 434, 435, 436, 439 or 447 of the Fc region, wherein the numbering of the residues in the Fc region is that of the EU index as in Kabat.
  • FcRn neonatal Fc receptor
  • Such polypeptide variants with reduced binding to an FcRn may comprise an amino acid modification at any one or more of amino acid positions 252, 253, 254, 255, 288, 309, 386, 388, 400, 415, 433, 435, 436, 439 or 447 of the Fc region, wherein the numbering of the residues in the Fc region is that of the EU index as in Kabat.
  • polypeptide variants may, alternatively, display increased binding to FcRn and comprise an amino acid modification at any one or more of amino acid positions 238, 256, 265, 272, 286, 303, 305, 307, 311, 312, 317, 340, 356, 360, 362, 376, 378, 380, 382, 413, 424 or 434 of the Fc region, wherein the numbering of the residues in the Fc region is that of the EU index as in Kabat.
  • the invention also provides a composition comprising the polypeptide variant and a physiologically or pharmaceutically acceptable carrier or diluent.
  • This composition for potential therapeutic use is sterile and may be lyophilized.
  • the invention provides a method for determining the presence of an antigen of interest comprising exposing a sample suspected of containing the antigen to the polypeptide variant and determining binding of the polypeptide variant to the sample.
  • the invention provides a method of treating a mammal suffering from or predisposed to a disease or disorder, comprising administering to the mammal a therapeutically effective amount of a polypeptide variant as disclosed herein, or of a composition comprising the polypeptide variant and a pharmaceutically acceptable carrier.
  • the invention further provides: isolated nucleic acid encoding the polypeptide variant; a vector comprising the nucleic acid, optionally, operably linked to control sequences recognized by a host cell transformed with the vector; a host cell containing the vector; a method for producing the polypeptide variant comprising culturing this host cell so that the nucleic acid is expressed and, optionally, recovering the polypeptide variant from the host cell culture (e.g. from the host cell culture medium).
  • the invention further provides a method for making a variant Fc region with altered Fc receptor (FcR) binding affinity, or altered antibody-dependent cell-mediated cytotoxicity (ADCC) activity, comprising:
  • Step (b) of the method may comprise determining binding of the variant Fc region to one or more FcRs in vitro. Moreover, the method may result in the identification of a variant Fc region with improved FcR binding affinity, or with improved ADCC activity, in step (b) thereof.
  • the FcR may, for example, be human Fc gamma receptor III (Fc ⁇ RIII).
  • step (b) comprises determining binding of the variant Fc region to at least two different FcRs
  • the FcRs tested preferably include human Fc gamma receptor II (Fc ⁇ RII) and human Fc gamma receptor III (Fc ⁇ RIII).
  • FIG. 1 is a schematic representation of a native IgG. Disulfide bonds are represented by heavy lines between CH1 and CL domains and the two CH2 domains. V is variable domain; C is constant domain; L stands for light chain and H stands for heavy chain.
  • FIG. 2 shows C1q binding of wild type (wt) C2B8 antibody; C2B8 antibody with a human IgG2 constant region (IgG2); and variants K322A, K320A and E318A.
  • FIG. 3 depicts C1q binding of variants P331A, P329A and K322A.
  • FIGS. 4A and 4B depict the amino acid sequences of E27 anti-IgE antibody light chain ( FIG. 4A ; SEQ ID NO:1) and heavy chain ( FIG. 4B ; SEQ ID NO:2).
  • FIG. 5 is a schematic diagram of the “immune complex” prepared for use in the FcR assay described in Example 1.
  • the hexamer comprising three anti-IgE antibody molecules (the “Fc region-containing polypeptide”) and three IgE molecules (the “first target molecule”) is shown.
  • IgE has two “binding sites” for the anti-IgE antibody (E27) in the Fc region thereof.
  • Each IgE molecule in the complex is further able to bind two VEGF molecules (“the second target polypeptide”).
  • VEGF has two “binding sites” for IgE.
  • FIG. 6 shows C1q binding results obtained for variants D270K and D270V compared to wild type C2B8.
  • FIG. 7 depicts complement dependent cytotoxicity (CDC) of variants D270K and D270V, compared to wild type C2B8.
  • FIG. 8 shows C1q binding ELISA results for 293 cell-produced wild type C2B8 antibody (293-Wt-C2B8), CHO-produced wild type C2B8 antibody (CHO-Wt-C2B8) and various variant antibodies.
  • FIG. 9 shows C1q binding ELISA results obtained for wild type (wt) C2B8 and various variant antibodies as determined in Example 3.
  • FIG. 10 depicts the three-dimensional structure of a human IgG Fc region, highlighting residues: Asp270, Lys326, Pro329, Pro331, Lys322 and Glu333.
  • FIG. 11 shows C1q binding ELISA results obtained for wild type C2B8 and various variant antibodies as determined in Example 3.
  • FIG. 12 shows C1q binding ELISA results obtained for wild type C2B8 and double variants, K326M-E333S and K326A-E333A.
  • FIG. 13 shows CDC of wild type C2B8 and double variants, K326M-E333S and K326A-E333A.
  • FIG. 14 depicts C1q binding ELISA results obtained for C2B8 with a human IgG4 (IgG4), wild type C2B8 (Wt-C2B8), C2B8 with a human IgG2 constant region (IgG2), and variant antibodies as described in Example 3.
  • IgG4 human IgG4
  • Wt-C2B8 wild type C2B8
  • IgG2 constant region IgG2 constant region
  • variant antibodies as described in Example 3.
  • FIGS. 15A and 15B show binding patterns for parent antibody (E27) to Fc ⁇ RIIB and Fc ⁇ RIIIA.
  • FIG. 15A shows the binding pattern for the humanized anti-IgE E27 IgG1 as a monomer (open circles), hexamer (closed squares), and immune complex consisting of multiple hexamers (closed triangles) to a recombinant GST fusion protein of the human Fc ⁇ RIIB (CD32) receptor ⁇ subunit.
  • the hexameric complex (closed squares) was formed by the mixture of equal molar concentrations of E27 (which binds to the Fc region of human IgE) and a human myeloma IgE.
  • the hexamer is a stable 1.1 kD complex consisting of 3 IgG molecules (150 kD each) and 3 IgE molecules (200 kD each).
  • the immune complex (closed triangles) was formed sequentially by first mixing equal molar concentrations of E27 and recombinant anti-VEGF IgE (human IgE with Fab variable domains that bind human VEGF) to form the hexamer. Hexamers were then linked to form an immune complex by the addition of 2 ⁇ molar concentration of human VEGF, a 44 kD homodimer which has two binding sites for the anti-VEGF IgE per mole of VEGF.
  • FIG. 15B shows the binding pattern to a recombinant GST fusion protein of the human Fc ⁇ RIIIA (CD16) receptor ⁇ subunit.
  • FIG. 16A shows the binding of immune complexes using different antigen-antibody pairs to recombinant GST fusion protein of the Fc ⁇ RIIA receptor ⁇ subunit.
  • FIG. 16B shows the binding of the same antigen-antibody pairs to the GST fusion protein of the Fc ⁇ RIIIA receptor ⁇ subunit. Closed circles represent binding of human IgE:anti-IgE E27 IgG1; open circles represent binding of human VEGF:humanized anti-VEGF IgG1.
  • FIG. 17 summarizes differences in binding selectivity of some alanine variants between the different Fc ⁇ Rs. Binding of alanine variants at residues in the CH2 domain of anti-IgE E27 IgG1 are shown to Fc ⁇ RIIA, Fc ⁇ RIIB, and Fc ⁇ RIIIA. Type 1 abrogates binding to all three receptors: D278A (265 in EU numbering). Type 2 improves binding to Fc ⁇ RIIA and Fc ⁇ RIIB, while binding to Fc ⁇ RIIIA is unaffected: S280A (267 in EU numbering). Type 3 improves binding to Fc ⁇ RIIA and Fc ⁇ RIIB, but reduces binding to Fc ⁇ RIIIA: H281A (268 in EU numbering).
  • Type 4 reduces binding to Fc ⁇ RIIA and Fc ⁇ RIIB, while improving binding to Fc ⁇ RIIIA: S317A (298 in EU numbering).
  • Type 5 improves binding to Fc ⁇ RIIIA, but does not affect binding to Fc ⁇ RIIA and Fc ⁇ RIIB: E352A, K353A (333 and 334 in EU numbering).
  • FIGS. 18A and 18B compare the Fc ⁇ RIIIA protein/protein assay and CHO GPI-Fc ⁇ RIIIA cell based assay, respectively.
  • FIG. 18A illustrates binding of selected alanine variants to Fc ⁇ RIIIA-GST fusion protein. S317A (298 in EU numbering) and S317A/K353A (298 and 334 in EU numbering) bind better than E27 wildtype, while D278A (265 in EU numbering) almost completely abrogates binding.
  • FIG. 18B illustrates that a similar pattern of binding is found on CHO cells expressing a recombinant GPI-linked form of Fc ⁇ RIIIA.
  • FIGS. 19A and 19B compare the Fc ⁇ RIIB protein/protein assay and CHO GPI-Fc ⁇ RIIB cell based assay, respectively.
  • FIG. 19A illustrates binding of selected alanine variants to Fc ⁇ RIIB-GST fusion protein. H281A (268 in EU numbering) binds better than E27 wildtype while S317A (298 in EU numbering) shows reduced binding.
  • FIG. 19B illustrates that a similar pattern of binding is found on CHO cells expressing a recombinant membrane bound form of Fc ⁇ RIIB.
  • FIG. 20 shows single alanine substitutions in the CH2 domain of anti-HER2 IgG1 (HERCEPTIN®) that influence Fc ⁇ RIIIA binding in both the protein-protein and cell-based assays alter the ability to bind to Fc ⁇ RIIIA on peripheral blood mononuclear cell (PBMC) effector cells.
  • PBMC peripheral blood mononuclear cell
  • a single alanine mutation that only slightly increased binding to Fc ⁇ RIIIA, variant G30 K307A (290 in EU numbering), also showed slightly improved ADCC (i.e., a 1.1 fold improvement in ADCC activity, calculated as area under the curve) at 1.25 ng/ml at all E:T ratios (filled diamonds) compared to wildtype antibody at 1.25 ng/ml (filled square).
  • a single alanine mutation that decreased binding to Fc ⁇ RIIIA, variant G34 Q312A (295 in EU numbering), also showed decreased ADCC activity (filled inverted triangles).
  • G36 displayed a 1.7 fold improvement in ADCC activity, calculated as area under the curve.
  • the effector cells were PBMCs.
  • FIG. 22A depicts alignments of native sequence IgG Fc regions.
  • Native sequence human IgG Fc region sequences hum IgG1 (non-A and A allotypes) (SEQ ID NOs: 3 and 4, respectively), humlgG2 (SEQ ID NO:5), hum IgG3 (SEQ ID NO:6) and humlgG4 (SEQ ID NO:7), are shown.
  • the human IgG1 sequence is the non-A allotype, and differences between this sequence and the A allotype (at positions 356 and 358; EU numbering system) are shown below the human IgG1 sequence.
  • FIG. 22B shows percent identity among the Fc region sequences of FIG. 22A .
  • FIG. 23 depicts alignments of native sequence human IgG Fc region sequences, humIgG1 (non-A and A allotypes; SEQ ID NOs:3 and 4, respectively), humIgG2 (SEQ ID NO:5), humIgG3 (SEQ ID NO:6) and humIgG4 (SEQ ID NO:7) with differences between the sequences marked with asterisks.
  • FIG. 24 shows area under curve (AUC) for selected variants compared to anti-HER2 IgG1 (HERCEPTIN®) in a 4 hour ADCC assay.
  • Variant G36 (S317A; 298 in Eu numbering) with improved binding to Fc ⁇ RIIIA showed improved ADCC activity; variant G31 (R309A; 292 in Eu numbering) which did not display altered Fc ⁇ RIIIA binding, also had unaltered ADCC activity; and G14 (D265A; 278 in Eu numbering) which had reduced Fc ⁇ RIIIA binding, also had reduced ADCC activity.
  • the numbering of the residues in an immunoglobulin heavy chain is that of the EU index as in Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md. (1991), expressly incorporated herein by reference.
  • the “EU index as in Kabat” refers to the residue numbering of the human IgG1 EU antibody.
  • a “parent polypeptide” is a polypeptide comprising an amino acid sequence which lacks one or more of the Fc region modifications disclosed herein and which differs in effector function compared to a polypeptide variant as herein disclosed.
  • the parent polypeptide may comprise a native sequence Fc region or an Fc region with pre-existing amino acid sequence modifications (such as additions, deletions and/or substitutions).
  • the term “Fc region” is used to define a C-terminal region of an immunoglobulin heavy chain, e.g., as shown in FIG. 1 .
  • the “Fc region” may be a native sequence Fc region or a variant Fc region.
  • the human IgG heavy chain Fc region is usually defined to stretch from an amino acid residue at position Cys226, or from Pro230, to the carboxyl-terminus thereof.
  • the Fc region of an immunoglobulin generally comprises two constant domains, CH2 and CH3, as shown, for example, in FIG. 1 .
  • the “CH2 domain” of a human IgG Fc region usually extends from about amino acid 231 to about amino acid 340.
  • the CH2 domain is unique in that it is not closely paired with another domain. Rather, two N-linked branched carbohydrate chains are interposed between the two CH2 domains of an intact native IgG molecule. It has been speculated that the carbohydrate may provide a substitute for the domain-domain pairing and help stabilize the CH2 domain.
  • the “CH3 domain” comprises the stretch of residues C-terminal to a CH2 domain in an Fc region (i.e. from about amino acid residue 341 to about amino acid residue 447 of an IgG)
  • a “functional Fc region” possesses an “effector function” of a native sequence Fc region.
  • effector functions include C1q binding; complement dependent cytotoxicity; Fc receptor binding; antibody-dependent cell-mediated cytotoxicity (ADCC); phagocytosis; down regulation of cell surface receptors (e.g. B cell receptor; BCR), etc.
  • ADCC antibody-dependent cell-mediated cytotoxicity
  • phagocytosis down regulation of cell surface receptors (e.g. B cell receptor; BCR), etc.
  • Such effector functions generally require the Fc region to be combined with a binding domain (e.g. an antibody variable domain) and can be assessed using various assays as herein disclosed, for example.
  • a “native sequence Fc region” comprises an amino acid sequence identical to the amino acid sequence of an Fc region found in nature.
  • Native sequence human Fc regions are shown in FIG. 23 and include a native sequence human IgG1 Fc region (non-A and A allotypes); native sequence human IgG2 Fc region; native sequence human IgG3 Fc region; and native sequence human IgG4 Fc region as well as naturally occurring variants thereof.
  • Native sequence murine Fc regions are shown in FIG. 22A .
  • a “variant Fc region” comprises an amino acid sequence which differs from that of a native sequence Fc region by virtue of at least one “amino acid modification” as herein defined.
  • the variant Fc region has at least one amino acid substitution compared to a native sequence Fc region or to the Fc region of a parent polypeptide, e.g. from about one to about ten amino acid substitutions, and preferably from about one to about five amino acid substitutions in a native sequence Fc region or in the Fc region of the parent polypeptide.
  • the variant Fc region herein will preferably possess at least about 80% homology with a native sequence Fc region and/or with an Fc region of a parent polypeptide, and most preferably at least about 90% homology therewith, more preferably at least about 95% homology therewith.
  • “Homology” is defined as the percentage of residues in the amino acid sequence variant that are identical after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent homology. Methods and computer programs for the alignment are well known in the art. One such computer program is “Align 2”, authored by Genentech, Inc., which was filed with user documentation in the United States Copyright Office, Washington, D.C. 20559, on Dec. 10, 1991.
  • Fc region-containing polypeptide refers to a polypeptide, such as an antibody or immunoadhesin (see definitions below), which comprises an Fc region.
  • Fc receptor or “FcR” are used to describe a receptor that binds to the Fc region of an antibody.
  • the preferred FcR is a native sequence human FcR.
  • a preferred FcR is one which binds an IgG antibody (a gamma receptor) and includes receptors of the Fc ⁇ RI, Fc ⁇ RII, and Fc ⁇ RIII subclasses, including allelic variants and alternatively spliced forms of these receptors.
  • Fc ⁇ RII receptors include Fc ⁇ RIIA (an “activating receptor”) and Fc ⁇ RIIB (an “inhibiting receptor”), which have similar amino acid sequences that differ primarily in the cytoplasmic domains thereof.
  • Activating receptor Fc ⁇ RIIA contains an immunoreceptor tyrosine-based activation motif (ITAM) in its cytoplasmic domain.
  • Inhibiting receptor Fc ⁇ RIIB contains an immunoreceptor tyrosine-based inhibition motif (ITIM) in its cytoplasmic domain.
  • ITAM immunoreceptor tyrosine-based activation motif
  • ITIM immunoreceptor tyrosine-based inhibition motif
  • FcR FcR
  • FcRn neonatal receptor
  • Antibody-dependent cell-mediated cytotoxicity and “ADCC” refer to a cell-mediated reaction in which nonspecific cytotoxic cells that express FcRs (e.g. Natural Killer (NK) cells, neutrophils, and macrophages) recognize bound antibody on a target cell and subsequently cause lysis of the target cell.
  • FcRs e.g. Natural Killer (NK) cells, neutrophils, and macrophages
  • the primary cells for mediating ADCC NK cells, express Fc ⁇ RIII only, whereas monocytes express Fc ⁇ RI, Fc ⁇ RII and Fc ⁇ RIII.
  • FcR expression on hematopoietic cells is summarized in Table 3 on page 464 of Ravetch and Kinet, Annu. Rev. Immunol. 9:457-92 (1991).
  • Human effector cells are leukocytes which express one or more FcRs and perform effector functions. Preferably, the cells express at least Fc ⁇ RIII and perform ADCC effector function. Examples of human leukocytes which mediate ADCC include peripheral blood mononuclear cells (PBMC), natural killer (NK) cells, monocytes, cytotoxic T cells and neutrophils; with PBMCs and NK cells being preferred.
  • PBMC peripheral blood mononuclear cells
  • NK natural killer cells
  • monocytes cytotoxic T cells and neutrophils
  • the effector cells may be isolated from a native source thereof, e.g. from blood or PBMCs as described herein.
  • a polypeptide variant with “altered” FcR binding affinity or ADCC activity is one which has either enhanced or diminished FcR binding activity and/or ADCC activity compared to a parent polypeptide or to a polypeptide comprising a native sequence Fc region.
  • the polypeptide variant which “displays increased binding” to an FcR binds at least one FcR with better affinity than the parent polypeptide.
  • the polypeptide variant which “displays decreased binding” to an FcR binds at least one FcR with worse affinity than a parent polypeptide.
  • Such variants which display decreased binding to an FcR may possess little or no appreciable binding to an FcR, e.g., 0-20% binding to the FcR compared to a native sequence IgG Fc region, e.g. as determined in the Examples herein.
  • the polypeptide variant which binds an FcR with “better affinity' than a parent polypeptide is one which binds any one or more of the above identified FcRs with substantially better binding affinity than the parent antibody, when the amounts of polypeptide variant and parent polypeptide in the binding assay are essentially the same.
  • the polypeptide variant with improved FcR binding affinity may display from about 1.15 fold to about 100 fold, e.g. from about 1.2 fold to about 50 fold improvement in FcR binding affinity compared to the parent polypeptide, where FcR binding affinity is determined, for example, as disclosed in the Examples herein.
  • the polypeptide variant which “mediates antibody-dependent cell-mediated cytotoxicity (ADCC) in the presence of human effector cells more effectively” than a parent antibody is one which in vitro or in vivo is substantially more effective at mediating ADCC, when the amounts of polypeptide variant and parent antibody used in the assay are essentially the same.
  • ADCC antibody-dependent cell-mediated cytotoxicity
  • Such variants will be identified using the in vitro ADCC assay as herein disclosed, but other assays or methods for determining ADCC activity, e.g. in an animal model etc, are contemplated.
  • the preferred variant is from about 1.5 fold to about 100 fold, e.g. from about two fold to about fifty fold, more effective at mediating ADCC than the parent, e.g. in the in vitro assay disclosed herein.
  • amino acid modification refers to a change in the amino acid sequence of a predetermined amino acid sequence.
  • exemplary modifications include an amino acid substitution, insertion and/or deletion.
  • the preferred amino acid modification herein is a substitution.
  • amino acid modification at a specified position, e.g. of the Fc region, refers to the substitution or deletion of the specified residue, or the insertion of at least one amino acid residue adjacent the specified residue.
  • insertion “adjacent” a specified residue is meant insertion within one to two residues thereof. The insertion may be N-terminal or C-terminal to the specified residue.
  • amino acid substitution refers to the replacement of at least one existing amino acid residue in a predetermined amino acid sequence with another different “replacement” amino acid residue.
  • the replacement residue or residues may be “naturally occurring amino acid residues” (i.e. encoded by the genetic code) and selected from the group consisting of: alanine (Ala); arginine (Arg); asparagine (Asn); aspartic acid (Asp); cysteine (Cys); glutamine (Gln); glutamic acid (Glu); glycine (Gly); histidine (His); isoleucine (Ile): leucine (Leu); lysine (Lys); methionine (Met); phenylalanine (Phe); proline (Pro); serine (Ser); threonine (Thr); tryptophan (Trp); tyrosine (Tyr); and valine (Val).
  • the replacement residue is not cysteine.
  • substitution with one or more non-naturally occurring amino acid residues is also encompassed by the definition of an amino acid substitution herein.
  • a “non-naturally occurring amino acid residue” refers to a residue, other than those naturally occurring amino acid residues listed above, which is able to covalently bind adjacent amino acid residues(s) in a polypeptide chain. Examples of non-naturally occurring amino acid residues include norleucine, ornithine, norvaline, homoserine and other amino acid residue analogues such as those described in Ellman et al. Meth. Enzym. 202:301-336 (1991).
  • amino acid insertion refers to the incorporation of at least one amino acid into a predetermined amino acid sequence. While the insertion will usually consist of the insertion of one or two amino acid residues, the present application contemplates larger “peptide insertions”, e.g. insertion of about three to about five or even up to about ten amino acid residues.
  • the inserted residue(s) may be naturally occurring or non-naturally occurring as disclosed above.
  • amino acid deletion refers to the removal of at least one amino acid residue from a predetermined amino acid sequence.
  • Hinge region is generally defined as stretching from Glu216 to Pro230 of human IgG1 (Burton, Molec. immunol. 22:161-206 (1985)). Hinge regions of other IgG isotypes may be aligned with the IgG1 sequence by placing the first and last cysteine residues forming inter-heavy chain S—S bonds in the same positions.
  • the “lower hinge region” of an Fc region is normally defined as the stretch of residues immediately C-terminal to the hinge region, i.e. residues 233 to 239 of the Fc region. Prior to the present invention, Fc ⁇ R binding was generally attributed to amino acid residues in the lower hinge region of an IgG Fc region.
  • C1q is a polypeptide that includes a binding site for the Fc region of an immunoglobulin. C1q together with two serine proteases, C1r and C1s, forms the complex C1; the first component of the complement dependent cytotoxicity (CDC) pathway. Human C1q can be purchased commercially from, e.g. Quidel, San Diego, Calif.
  • binding domain refers to the region of a polypeptide that binds to another molecule.
  • the binding domain can comprise a portion of a polypeptide chain thereof (e.g. the a chain thereof) which is responsible for binding an Fc region.
  • One useful binding domain is the extracellular domain of an FcR a chain.
  • antibody is used in the broadest sense and specifically covers monoclonal antibodies (including full length monoclonal antibodies), polyclonal antibodies, multispecific antibodies (e.g., bispecific antibodies), and antibody fragments so long as they exhibit the desired biological activity.
  • Antibody fragments comprise a portion of an intact antibody, generally including the antigen binding or variable region of the intact antibody or the Fc region of an antibody which retains FcR binding capability.
  • antibody fragments include linear antibodies; single-chain antibody molecules; and multispecific antibodies formed from antibody fragments.
  • the antibody fragments preferably retain at least part of the hinge and optionally the CH1 region of an IgG heavy chain. More preferably, the antibody fragments retain the entire constant region of an IgG heavy chain, and include an IgG light chain.
  • the term “monoclonal antibody” as used herein refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally occurring mutations that may be present in minor amounts. Monoclonal antibodies are highly specific, being directed against a single antigenic site. Furthermore, in contrast to conventional (polyclonal) antibody preparations that typically include different antibodies directed against different determinants (epitopes), each monoclonal antibody is directed against a single determinant on the antigen.
  • the modifier “monoclonal” indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method.
  • the monoclonal antibodies to be used in accordance with the present invention may be made by the hybridoma method first described by Kohler et al., Nature 256:495 (1975), or may be made by recombinant DNA methods (see, e.g., U.S. Pat. No. 4,816,567).
  • the “monoclonal antibodies” may also be isolated from phage antibody libraries using the techniques described in Clackson et al., Nature 352:624-628 (1991) and Marks et al., J. Mol. Biol. 222:581-597 (1991), for example.
  • the monoclonal antibodies herein specifically include “chimeric” antibodies (immunoglobulins) in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is identical with or homologous to corresponding sequences in antibodies derived from another species or belonging to another antibody class or subclass, as well as fragments of such antibodies, so long as they exhibit the desired biological activity (U.S. Pat. No. 4,816,567; and Morrison et al., Proc. Natl. Acad. Sci. USA 81:6851-6855 (1984)).
  • chimeric antibodies immunoglobulins in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is identical with or homologous to corresponding sequences
  • “Humanized” forms of non-human (e.g., murine) antibodies are chimeric antibodies that contain minimal sequence derived from non-human immunoglobulin.
  • humanized antibodies are human immunoglobulins (recipient antibody) in which residues from a hypervariable region of the recipient are replaced by residues from a hypervariable region of a non-human species (donor antibody) such as mouse, rat, rabbit or nonhuman primate having the desired specificity, affinity, and capacity.
  • donor antibody such as mouse, rat, rabbit or nonhuman primate having the desired specificity, affinity, and capacity.
  • Fv framework region (FR) residues of the human immunoglobulin are replaced by corresponding non-human residues.
  • humanized antibodies may comprise residues that are not found in the recipient antibody or in the donor antibody. These modifications are made to further refine antibody performance.
  • the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the hypervariable loops correspond to those of a non-human immunoglobulin and all or substantially all of the FR regions are those of a human immunoglobulin sequence.
  • the humanized antibody optionally also will comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin.
  • Fc immunoglobulin constant region
  • hypervariable region when used herein refers to the amino acid residues of an antibody which are responsible for antigen-binding.
  • the hypervariable region comprises amino acid residues from a “complementarity determining region” or “CDR” (Le. residues 24-34 (L1), 50-56 (L2) and 89-97 (L3) in the light chain variable domain and 31-35 (H1), 50-65 (H2) and 95-102 (H3) in the heavy chainvariable domain; Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md. (1991)) and/or those residues from a “hypervariable loop” (i.e.
  • “Framework” or “FR” residues are those variable domain residues other than the hypervariable region residues as herein defined.
  • immunoadhesin designates antibody-like molecules which combine the “binding domain” of a heterologous “adhesin” protein (e.g. a receptor, ligand or enzyme) with an immunoglobulin constant domain.
  • adhesin protein e.g. a receptor, ligand or enzyme
  • the immunoadhesins comprise a fusion of the adhesin amino acid sequence with the desired binding specificity which is other than the antigen recognition and binding site (antigen combining site) of an antibody (i.e. is “heterologous”) and an immunoglobulin constant domain sequence.
  • ligand binding domain refers to any native cell-surface receptor or any region or derivative thereof retaining at least a qualitative ligand binding ability of a corresponding native receptor.
  • the receptor is from a cell-surface polypeptide having an extracellular domain that is homologous to a member of the immunoglobulin supergenefamily.
  • Other receptors which are not members of the immunoglobulin supergenefamily but are nonetheless specifically covered by this definition, are receptors for cytokines, and in particular receptors with tyrosine kinase activity (receptor tyrosine kinases), members of the hematopoietin and nerve growth factor receptor superfamilies, and cell adhesion molecules, e.g. (E-, L- and P-) selectins.
  • receptor binding domain is used to designate any native ligand for a receptor, including cell adhesion molecules, or any region or derivative of such native ligand retaining at least a qualitative receptor binding ability of a corresponding native ligand. This definition, among others, specifically includes binding sequences from ligands for the above-mentioned receptors.
  • an “antibody-immunoadhesin chimera” comprises a molecule that combines at least one binding domain of an antibody (as herein defined) with at least one immunoadhesin (as defined in this application).
  • Exemplary antibody-immunoadhesin chimeras are the bispecific CD4-IgG chimeras described in Berg et al., PNAS ( USA ) 88:4723-4727 (1991) and Chamow et al., J. Immunol. 153:4268 (1994).
  • an “isolated” polypeptide is one that has been identified and separated and/or recovered from a component of its natural environment. Contaminant components of its natural environment are materials that would interfere with diagnostic or therapeutic uses for the polypeptide, and may include enzymes, hormones, and other proteinaceous or nonproteinaceous solutes.
  • the polypeptide will be purified (1) to greater than 95% by weight of polypeptide as determined by the Lowry method, and most preferably more than 99% by weight, (2) to a degree sufficient to obtain at least 15 residues of N-terminal or internal amino acid sequence by use of a spinning cup sequenator, or (3) to homogeneity by SDS-PAGE under reducing or nonreducing conditions using Coomassie blue or, preferably, silver stain.
  • Isolated polypeptide includes the polypeptide in situ within recombinant cells since at least one component of the polypeptide's natural environment will not be present. Ordinarily, however, isolated polypeptide will be prepared by at least one purification step.
  • Treatment refers to both therapeutic treatment and prophylactic or preventative measures. Those in need of treatment include those already with the disorder as well as those in which the disorder is to be prevented.
  • a “disorder is any condition that would benefit from treatment with the polypeptide variant. This includes chronic and acute disorders or diseases including those pathological conditions which predispose the mammal to the disorder in question.
  • the disorder is cancer.
  • cancer and “cancerous” refer to or describe the physiological condition in mammals that is typically characterized by unregulated cell growth.
  • Examples of cancer include but are not limited to, carcinoma, lymphoma, blastoma, sarcoma, and leukemia.
  • cancers include squamous cell cancer, small-cell lung cancer, non-small cell lung cancer, adenocarcinoma of the lung, squamous carcinoma of the lung, cancer of the peritoneum, hepatocellular cancer, gastrointestinal cancer, pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, hepatoma, breast cancer, colon cancer, colorectal cancer, endometrial or uterine carcinoma, salivary gland carcinoma, kidney cancer, liver cancer, prostate cancer, vulval cancer, thyroid cancer, hepatic carcinoma and various types of head and neck cancer.
  • a “HER2-expressing cancer” is one comprising cells which have HER2 receptor protein (Semba et al., PNAS ( USA ) 82:6497-6501 (1985) and Yamamoto et al., Nature 319:230-234 (1986) (Genebank accession number X03363)) present at their cell surface, such that an anti-HER2 antibody is able to bind to the cancer.
  • label when used herein refers to a detectable compound or composition which is conjugated directly or indirectly to the polypeptide.
  • the label may be itself be detectable (e.g., radioisotope labels or fluorescent labels) or, in the case of an enzymatic label, may catalyze chemical alteration of a substrate compound or composition which is detectable.
  • an “isolated” nucleic acid molecule is a nucleic acid molecule that is identified and separated from at least one contaminant nucleic acid molecule with which it is ordinarily associated in the natural source of the polypeptide nucleic acid.
  • An isolated nucleic acid molecule is other than in the form or setting in which it is found in nature. Isolated nucleic acid molecules therefore are distinguished from the nucleic acid molecule as it exists in natural cells.
  • an isolated nucleic acid molecule includes a nucleic acid molecule contained in cells that ordinarily express the polypeptide where, for example, the nucleic acid molecule is in a chromosomal location different from that of natural cells.
  • control sequences refers to DNA sequences necessary for the expression of an operably linked coding sequence in a particular host organism.
  • the control sequences that are suitable for prokaryotes include a promoter, optionally an operator sequence, and a ribosome binding site.
  • Eukaryotic cells are known to utilize promoters, polyadenylation signals, and enhancers.
  • Nucleic acid is “operably linked” when it is placed into a functional relationship with another nucleic acid sequence.
  • DNA for a presequence or secretory leader is operably linked to DNA for a polypeptide if it is expressed as a preprotein that participates in the secretion of the polypeptide;
  • a promoter or enhancer is operably linked to a coding sequence if it affects the transcription of the sequence; or
  • a ribosome binding site is operably linked to a coding sequence if it is positioned so as to facilitate translation.
  • “operably linked” means that the DNA sequences being linked are contiguous, and, in the case of a secretory leader, contiguous and in reading phase. However, enhancers do not have to be contiguous. Linking is accomplished by ligation at convenient restriction sites. If such sites do not exist, the synthetic oligonucleotide adaptors or linkers are used in accordance with conventional practice.
  • the expressions “cell,” “cell line,” and “cell culture” are used interchangeably and all such designations include progeny.
  • the words “transformants” and “transformed cells” include the primary subject cell and cultures derived therefrom without regard for the number of transfers. It is also understood that all progeny may not be precisely identical in DNA content, due to deliberate or inadvertent mutations. Mutant progeny that have the same function or biological activity as screened for in the originally transformed cell are included. Where distinct designations are intended, it will be clear from the context.
  • molecular complex when used herein refers to the relatively stable structure which forms when two or more heterologous molecules (e.g. polypeptides) bind (preferably noncovalently) to one another.
  • heterologous molecules e.g. polypeptides
  • the preferred molecular complex herein is an immune complex.
  • Immuno complex refers to the relatively stable structure which forms when at least one target molecule and at least one heterologous Fc region-containing polypeptide bind to one another forming a larger molecular weight complex.
  • immune complexes are antigen-antibody aggregates and target molecule-immunoadhesin aggregates.
  • target molecule refers to a molecule, usually a polypeptide, which is capable of being bound by a heterologous molecule and has one or more binding sites for the heterologous molecule.
  • binding site refers to a region of a molecule to which another molecule can bind.
  • the “first target molecule” herein comprises at least two distinct binding sites (for example, two to five separate binding sites) for an analyte (e.g. an Fc region-containing polypeptide) such that at least two analyte molecules can bind to the first target molecule.
  • the two or more binding sites are identical (e.g. having the same amino acid sequence, where the target molecule is a polypeptide).
  • the first target molecule was IgE and had two separate binding sites in the Fc region thereof to which the Fc region-containing polypeptide (an anti-IgE antibody, E27) could bind.
  • Other first target molecules include dimers of substantially identical monomors (e.g. neurotrophins, IL8 and VEGF) or are polypeptides comprising two or more substantially identical polypeptide chains (e.g. antibodies or immunoadhesins).
  • the “second target molecule” comprises at least two distinct binding sites (for example, two to five separate binding sites) for the first target molecule such that at least two first target molecules can bind to the second target molecule.
  • the two or more binding sites are identical (e.g.
  • the second target molecule was VEGF, which has a pair of distinct binding sites to which the variable domain of the IgE antibody could bind.
  • Other second target molecules are contemplated, e.g. other dimers of substantially identical monomers (e.g. neurotrophins or IL8) or polypeptides comprising two or more substantially identical domains (e.g. antibodies or immunoadhesins).
  • an “analyte” is a substance that is to be analyzed.
  • the preferred analyte is an Fc region-containing polypeptide that is to be analyzed for its ability to bind to an Fc receptor.
  • a “receptor” is a polypeptide capable of binding at least one ligand.
  • the preferred receptor is a cell-surface receptor having an extracellular ligand-binding domain and, optionally, other domains (e.g. transmembrane domain, intracellular domain and/or membrane anchor).
  • the receptor to be evaluated in the assay described herein may be an intact receptor or a fragment or derivative thereof (e.g. a fusion protein comprising the binding domain of the receptor fused to one or more heterologous polypeptides).
  • the receptor to be evaluated for its binding properties may be present in a cell or isolated and optionally coated on an assay plate or some other solid phase.
  • low affinity receptor denotes a receptor that has a weak binding affinity for a ligand of interest, e.g. having a binding constant of about 50 nM or worse affinity.
  • exemplary low affinity receptors include Fc ⁇ RII and Fc ⁇ RIII.
  • the invention herein relates to a method for making a polypeptide variant.
  • the “parent”, “starting” or “nonvariant” polypeptide is prepared using techniques available in the art for generating polypeptides comprising an Fc region.
  • the parent polypeptide is an antibody and exemplary methods for generating antibodies are described in more detail in the following sections.
  • the parent polypeptide may, however, be any other polypeptide comprising an Fc region, e.g. an immunoadhesin. Methods for making immunoadhesins are elaborated in more detail hereinbelow.
  • a variant Fc region may be generated according to the methods herein disclosed and this “variant Fc region” can be fused to a heterologous polypeptide of choice, such as an antibody variable domain or binding domain of a receptor or ligand.
  • the parent polypeptide comprises an Fc region.
  • the Fc region of the parent polypeptide will comprise a native sequence Fc region, and preferably a human native sequence Fc region.
  • the Fc region of the parent polypeptide may have one or more pre-existing amino acid sequence alterations or modifications from a native sequence Fc region.
  • the C1q binding activity of the Fc region may have been previously altered (other types of Fc region modifications are described in more detail below).
  • the parent polypeptide Fc region is “conceptual” and, while it does not physically exist, the antibody engineer may decide upon a desired variant Fc region amino acid sequence and generate a polypeptide comprising that sequence or a DNA encoding the desired variant Fc region amino acid sequence.
  • nucleic acid encoding an Fc region of a parent polypeptide is available and this nucleic acid sequence is altered to generate a variant nucleic acid sequence encoding the Fc region variant.
  • DNA encoding an amino acid sequence variant of the starting polypeptide is prepared by a variety of methods known in the art. These methods include, but are not limited to, preparation by site-directed (or oligonucleotide-mediated) mutagenesis, PCR mutagenesis, and cassette mutagenesis of an earlier prepared DNA encoding the polypeptide
  • Site-directed mutagenesis is a preferred method for preparing substitution variants. This technique is well known in the art (see, e.g., Carter et al., Nucleic Acids Res. 13:4431-4443 (1985) and Kunkel et al., Proc. Natl. Acad. Sci. USA 82:488-492 (1985)). Briefly, in carrying out site-directed mutagenesis of DNA, the starting DNA is altered by first hybridizing an oligonucleotide encoding the desired mutation to a single strand of such starting DNA.
  • a DNA polymerase is used to synthesize an entire second strand, using the hybridized oligonucleotide as a primer, and using the single strand of the starting DNA as a template.
  • the oligonucleotide encoding the desired mutation is incorporated in the resulting double-stranded DNA.
  • PCR mutagenesis is also suitable for making amino acid sequence variants of the starting polypeptide. See Higuchi, in PCR Protocols, pp. 177-183 (Academic Press, 1990); and Vallette et al., Nuc. Acids Res. 17:723-733 (1989). Briefly, when small amounts of template DNA are used as starting material in a PCR, primers that differ slightly in sequence from the corresponding region in a template DNA can be used to generate relatively large quantities of a specific DNA fragment that differs from the template sequence only at the positions where the primers differ from the template.
  • the starting material is the plasmid (or other vector) comprising the starting polypeptide DNA to be mutated.
  • the codon(s) in the starting DNA to be mutated are identified. There must be a unique restriction endonuclease site on each side of the identified mutation site(s). If no such restriction sites exist, they may be generated using the above-described oligonucleotide-mediated mutagenesis method to introduce them at appropriate locations in the starting polypeptide DNA.
  • the plasmid DNA is cut at these sites to linearize it.
  • a double-stranded oligonucleotide encoding the sequence of the DNA between the restriction sites but containing the desired mutation(s) is synthesized using standard procedures, wherein the two strands of the oligonucleotide are synthesized separately and then hybridized together using standard techniques.
  • This double-stranded oligonucleotide is referred to as the cassette.
  • This cassette is designed to have 5′ and 3′ ends that are compatible with the ends of the linearized plasmid, such that it can be directly ligated to the plasmid.
  • This plasmid now contains the mutated DNA sequence.
  • the desired amino acid sequence encoding a polypeptide variant can be determined, and a nucleic acid sequence encoding such amino acid sequence variant can be generated synthetically.
  • the amino acid sequence of the parent polypeptide is modified in order to generate a variant Fc region with altered Fc receptor binding affinity or activity in vitro and/or in vivo and/or altered antibody-dependent cell-mediated cytotoxicity (ADCC) activity in vitro and/or in vivo.
  • ADCC antibody-dependent cell-mediated cytotoxicity
  • the modification entails one or more amino acid substitutions.
  • the replacement residue does not correspond to a residue present in the same position in any of the native sequence Fc regions in FIG. 22A .
  • Pro331 of a human IgG3 or IgG1 Fc region is replaced with a residue other than Ser (the corresponding aligned residue found in native sequence human IgG4).
  • the residue in the parent polypeptide which is substituted with a replacement residue is not an alanine and/or is not residue A1a339 of an Fc region.
  • an amino acid substitution preferably the residue in the parent polypeptide is replaced with an alanine residue.
  • the present invention contemplates replacement of the residue of the parent polypeptide with any other amino acid residue.
  • the substitution may, for example, be a “conservative substitution”. Such conservative substitutions are shown in Table 1 under the heading of “preferred substitution”. More substantial changes may be achieved by making one or more “exemplary substitutions” which are not the preferred substitution in Table 1.
  • Substantial modifications in the biological properties of the Fc region may be accomplished by selecting substitutions that differ significantly in their effect on maintaining (a) the structure of the polypeptide backbone in the area of the substitution, for example, as a sheet or helical conformation, (b) the charge or hydrophobicity of the molecule at the target site, or (c) the bulk of the side chain.
  • Naturally occurring residues are divided into groups based on common side-chain properties:
  • hydrophobic norleucine, met, ala, val, leu, ile
  • Non-conservative substitutions will entail exchanging a member of one of these classes for a member of another class.
  • Conservative and non-conservative amino acid substitutions are exemplified in Table 8 hereinbelow.
  • Example 4 one can engineer an Fc region variant with altered binding affinity for one or more FcRs.
  • different classes of Fc region variants can be made e.g., as summarized in the following table. Where the variant Fc region has more than one amino acid substitution, generally, but not necessarily, amino acid substitutions in the same class are combined to achieve the desired result.
  • one will delete one or more of the Fc region residues identified herein as effecting FcR binding (see Example 4 below) in order to generate such an Fc region variant.
  • no more than one to about ten Fc region residues will be deleted according to this embodiment of the invention.
  • the Fc region herein comprising one or more amino acid deletions will preferably retain at least about 80%, and preferably at least about 90%, and most preferably at least about 95%, of the parent Fc region or of a native sequence human Fc region.
  • variant Fc region which (a) mediates antibody-dependent cell-mediated cytotoxicity (ADCC) in the presence of human effector cells more effectively and/or (b) binds an Fc gamma receptor (Fc ⁇ R) with better affinity than the parent polypeptide.
  • ADCC antibody-dependent cell-mediated cytotoxicity
  • Fc ⁇ R Fc gamma receptor
  • Such Fc region variants will generally comprise at least one amino acid modification in the Fc region. Combining amino acid modifications is thought to be particularly desirable.
  • the variant Fc region may include two, three, four, five, etc substitutions therein, e.g. of the specific Fc region positions identified herein.
  • the parent polypeptide Fc region is a human Fc region, e.g. a native sequence human Fc region human IgG1 (A and non-A allotypes), IgG2, IgG3 or IgG4 Fc region. Such sequences are shown in FIG. 23 .
  • the parent polypeptide preferably has pre-existing ADCC activity, e.g., it comprises a human IgG1 or human IgG3 Fc region.
  • the variant with improved ADCC mediates ADCC substantially more effectively than an antibody with a native sequence IgG1 or IgG3 Fc region and the antigen-binding region of the variant.
  • the variant comprises, or consists essentially of, substitutions of two or three of the residues at positions 298, 333 and 334 of the Fc region. Most preferably, residues at positions 298, 333 and 334 are substituted, (e.g. with alanine residues).
  • an Fc region variant with improved binding affinity for Fc ⁇ RIII which is thought to be an important FcR for mediating ADCC.
  • Fc ⁇ RIII an amino acid modification (e.g. a substitution) into the parent Fc region at any one or more of amino acid positions 256, 290, 298, 312, 326, 330, 333, 334, 360, 378 or 430 to generate such a variant.
  • the variant with improved binding affinity for Fc ⁇ RIII may further have reduced binding affinity for Fc ⁇ RII, especially reduced affinity for the inhibiting Fc ⁇ RIIB receptor.
  • the amino acid modification(s) are preferably introduced into the CH2 domain of a Fc region, since the experiments herein indicate that the CH2 domain is important for FcR binding activity. Moreover, unlike the teachings of the above-cited art, the instant application contemplates the introduction of a modification into a part of the Fc region other than in the lower hinge region thereof.
  • Useful amino acid positions for modification in order to generate a variant IgG Fc region with altered Fc gamma receptor (Fc ⁇ R) binding affinity or activity include any one or more of amino acid positions 238, 239, 248, 249, 252, 254, 255, 256, 258, 265, 267, 268, 269, 270, 272, 276, 278, 280, 283, 285, 286, 289, 290, 292, 293, 294, 295, 296, 298, 301, 303, 305, 307, 309, 312, 315, 320, 322, 324, 326, 327, 329, 330, 331, 333, 334, 335, 337, 338, 340, 360, 373, 376, 378, 382, 388, 389, 398, 414, 416, 419, 430, 434, 435, 437, 438 or 439 of the Fc region.
  • Fc ⁇ R Fc gamma receptor
  • the parent Fc region used as the template to generate such variants comprises a human IgG Fc region.
  • the parent Fc region is preferably not human native sequence IgG3, or the variant Fc region comprising a substitution at position 331 preferably displays increased FcR binding, e.g. to Fc ⁇ RII.
  • Variants which display reduced binding to Fc ⁇ RI include those comprising an Fc region amino acid modification at any one or more of amino acid positions 238, 265, 269, 270, 327 or 329.
  • Variants which display reduced binding to Fc ⁇ RII include those comprising an Fc region amino acid modification at any one or more of amino acid positions 238, 265, 269, 270, 292, 294, 295, 298, 303, 324, 327, 329, 333, 335, 338, 373, 376, 414, 416, 419, 435, 438 or 439.
  • Fc region variants which display reduced binding to Fc ⁇ RIII include those comprising an Fc region amino acid modification at any one or more of amino acid positions 238, 239, 248, 249, 252, 254, 265, 268, 269, 270, 272, 278, 289, 293, 294, 295, 296, 301, 303, 322, 327, 329, 338, 340, 373, 376, 382, 388, 389, 416, 434, 435 or 437.
  • Fc region variants may comprise an amino acid modification at any one or more of amino acid positions 255, 256, 258, 267, 268, 272, 276, 280, 283, 285, 286, 290, 298, 301, 305, 307, 309, 312, 315, 320, 322, 326, 330, 331, 333, 334, 337, 340, 360, 378, 398 or 430 of the Fc region.
  • the variant with improved Fc ⁇ R binding activity may display increased binding to Fc ⁇ RIII, and optionally may further display decreased binding to Fc ⁇ RII; e.g. the variant may comprise an amino acid modification at position 298 and/or 333 of an Fc region.
  • Variants with increased binding to Fc ⁇ RII include those comprising an amino acid modification at any one or more of amino acid positions 255, 256, 258, 267, 268, 272, 276, 280, 283, 285, 286, 290, 301, 305, 307, 309, 312, 315, 320, 322, 326, 330, 331, 337, 340, 378, 398 or 430 of an Fc region.
  • Such variants may further display decreased binding to Fc ⁇ RIII.
  • they may include an Fc region amino acid modification at any one or more of amino acid positions 268, 272, 298, 301, 322 or 340.
  • Fc region variants with altered binding affinity for the neonatal receptor are also contemplated herein.
  • Fc region variants with improved affinity for FcRn are anticipated to have longer serum half-lives, and such molecules will have useful applications in methods of treating mammals where long half-life of the administered polypeptide is desired, e.g., to treat a chronic disease or disorder.
  • Fc region variants with decreased FcRn binding affinity are expected to have shorter half-lives, and such molecules may, for example, be administered to a mammal where a shortened circulation time may be advantageous, e.g.
  • Fc region variants with decreased FcRn binding affinity are anticipated to be less likely to cross the placenta, and thus may be utilized in the treatment of diseases or disorders in pregnant women.
  • Fc region variants with altered binding affinity for FcRn include those comprising an Fc region amino acid modification at any one or more of amino acid positions 238, 252, 253, 254, 255, 256, 265, 272, 286, 288, 303, 305, 307, 309, 311, 312, 317, 340, 356, 360, 362, 376, 378, 380, 382, 386, 388, 400, 413, 415, 424, 433, 434, 435, 436, 439 or 447.
  • Those which display reduced binding to FcRn will generally comprise an Fc region amino acid modification at any one or more of amino acid positions 252, 253, 254, 255, 288, 309, 386, 388, 400, 415, 433, 435, 436, 439 or 447; and those with increased binding to FcRn will usually comprise an Fc region amino acid modification at any one or more of amino acid positions 238, 256, 265, 272, 286, 303, 305, 307, 311, 312, 317, 340, 356, 360, 362, 376, 378, 380, 382, 413, 424 or 434.
  • polypeptide variant(s) prepared as described above may be subjected to further modifications, oftentimes depending on the intended use of the polypeptide. Such modifications may involve further alteration of the amino acid sequence (substitution, insertion and/or deletion of amino acid residues), fusion to heterologous polypeptide(s) and/or covalent modifications. Such “further modifications” may be made prior to, simultaneously with, or following, the amino acid modification(s) disclosed above which result in an alteration of Fc receptor binding and/or ADCC activity. In one embodiment, one may combine the Fc region modification herein with Fc region substitutions disclosed in the references cited in the “Related Art” section of this application.
  • the starting polypeptide of particular interest herein is usually one that binds to C1q and displays complement dependent cytotoxicity (CDC).
  • CDC complement dependent cytotoxicity
  • the further amino acid substitutions described herein will generally serve to alter the ability of the starting polypeptide to bind to C1q and/or modify its complement dependent cytotoxicity function, e.g. to reduce and preferably abolish these effector functions.
  • polypeptides comprising substitutions at one or more of the described positions with improved C1q binding and/or complement dependent cytotoxicity (CDC) function are contemplated herein.
  • the starting polypeptide may be unable to bind C1q and/or mediate CDC and may be modified according to the teachings herein such that it acquires these further effector functions.
  • polypeptides with pre-existing C1q binding activity, optionally further having the ability to mediate CDC may be modified such that one or both of these activities are enhanced.
  • the amino acid positions to be modified are generally selected from heavy chain positions 270, 322, 326, 327, 329, 331, 333, and 334, where the numbering of the residues in an IgG heavy chain is that of the EU index as in Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md. (1991). In one embodiment, only one of the eight above-identified positions is altered in order to generate the polypeptide variant region with altered C1q binding and/or complement dependent cytotoxicity (CDC) function.
  • residue 270, 329 or 322 is altered if this is the case.
  • two or more of the above-identified positions are modified. If substitutions are to be combined, generally substitutions which enhance human C1q binding (e.g. at residue positions 326, 327, 333 and 334) or those which diminish human C1q binding (e.g., at residue positions 270, 322, 329 and 331) are combined. In the latter embodiment, all four positions (i.e., 270, 322, 329 and 331) may be substituted.
  • Proline is conserved at position 329 in human IgG's. This residue is preferably replaced with alanine, however substitution with any other amino acid is contemplated, e.g., serine, threonine, asparagine, glycine or valine.
  • Proline is conserved at position 331 in human IgG1, IgG2 and IgG3, but not IgG4 (which has a serine residue at position 331).
  • Residue 331 is preferably replaced by alanine or another amino acid, e.g. serine (for IgG regions other than IgG4), glycine or valine.
  • Lysine 322 is conserved in human IgGs, and this residue is preferably replaced by an alanine residue, but substitution with any other amino acid residue is contemplated, e.g. serine, threonine, glycine or valine.
  • D270 is conserved in human IgGs, and this residue may be replaced by another amino acid residue, e.g. alanine, serine, threonine, glycine, valine, or lysine.
  • K326 is also conserved in human IgGs. This residue may be substituted with another residue including, but not limited to, valine, glutamic acid, alanine, glycine, aspartic acid, methionine or tryptophan, with tryptophan being preferred.
  • E333 is also conserved in human IgGs.
  • E333 is preferably replaced by an amino acid residue with a smaller side chain volume, such as valine, glycine, alanine or serine, with serine being preferred.
  • K334 is conserved in human IgGs and may be substituted with another residue such as alanine or other residue.
  • residue 327 is an alanine. In order to generate a variant with improved C1q binding, this alanine may be substituted with another residue such as glycine. In IgG2 and IgG4, residue 327 is a glycine and this may be replaced by alanine (or another residue) to diminish C1q binding.
  • one can design an Fc region with altered effector function e.g., by modifying C1q binding and/or FcR binding and thereby changing CDC activity and/or ADCC activity.
  • one may increase only one of these activities, and optionally also reduce the other activity, e.g. to generate an Fc region variant with improved ADCC activity, but reduced CDC activity and vice versa.
  • any cysteine residue not involved in maintaining the proper conformation of the polypeptide variant also may be substituted, generally with serine, to improve the oxidative stability of the molecule and prevent aberrant cross linking.
  • Another type of amino acid substitution serves to alter the glycosylation pattern of the polypeptide. This may be achieved by deleting one or more carbohydrate moieties found in the polypeptide, and/or adding one or more glycosylation sites that are not present in the polypeptide.
  • Glycosylation of polypeptides is typically either N-linked or O-linked. N-linked refers to the attachment of the carbohydrate moiety to the side chain of an asparagine residue.
  • the tripeptide sequences asparagine-X-serine and asparagine-X-threonine, where X is any amino acid except proline, are the recognition sequences for enzymatic attachment of the carbohydrate moiety to the asparagine side chain.
  • O-linked glycosylation refers to the attachment of one of the sugars N-aceylgalactosamine, galactose, or xylose to a hydroxyamino acid, most commonly serine or threonine, although 5-hydroxyproline or 5-hydroxylysine may also be used.
  • Addition of glycosylation sites to the polypeptide is conveniently accomplished by altering the amino acid sequence such that it contains one or more of the above-described tripeptide sequences (for N-linked glycosylation sites).
  • the alteration may also be made by the addition of, or substitution by, one or more serine or threonine residues to the sequence of the original polypeptide (for O-linked glycosylation sites).
  • An exemplary glycosylation variant has an amino acid substitution of residue Asn 297 of the heavy chain.
  • the class, subclass or allotype of the Fc region may be altered by one or more further amino acid substitutions to generate an Fc region with an amino acid sequence more homologous to a different class, subclass or allotype as desired.
  • a murine Fc region may be altered to generate an amino acid sequence more homologous to a human Fc region; a human non-A allotype IgG1 Fc region may be modified to achieve a human A allotype IgG1 Fc region etc.
  • the amino modification(s) herein which alter FcR binding and/or ADCC activity are made in the CH2 domain of the Fc region and the CH3 domain is deleted or replaced with another dimerization domain. Preferably, however, the CH3 domain is retained (aside from amino acid modifications therein which alter effector function as herein disclosed).
  • the polypeptide variant may be subjected to one or more assays to evaluate any change in biological activity compared to the starting polypeptide.
  • the polypeptide variant essentially retains the ability to bind antigen compared to the nonvariant polypeptide, i.e. the binding capability is no worse than about 20 fold, e.g. no worse than about 5 fold of that of the nonvariant polypeptide.
  • the binding capability of the polypeptide variant may be determined using techniques such as fluorescence activated cell sorting (FACS) analysis or radioimmunoprecipitation (RIA), for example.
  • the ability of the polypeptide variant to bind an FcR may be evaluated.
  • the FcR is a high affinity Fc receptor, such as Fc ⁇ RI, FcRn or Fc ⁇ RIIIA-V158
  • binding can be measured by titrating monomeric polypeptide variant and measuring bound polypeptide variant using an antibody which specifically binds to the polypeptide variant in a standard ELISA format (see Example 2 below).
  • Another FcR binding assay for low affinity FcRs is described in Examples 1 and 4.
  • ADCC activity of the polypeptide variant may be assessed in vivo, e.g., in a animal model such as that disclosed in Clynes et al., PNAS ( USA ) 95:652-656 (1998).
  • the ability of the variant to bind C1q and mediate complement dependent cytotoxicity (CDC) may be assessed.
  • a C1q binding ELISA may be performed. Briefly, assay plates may be coated overnight at 4° C. with polypeptide variant or starting polypeptide (control) in coating buffer. The plates may then be washed and blocked. Following washing, an aliquot of human C1q may be added to each well and incubated for 2 hrs at room temperature. Following a further wash, 100 ⁇ l of a sheep anti-complement C1q peroxidase conjugated antibody may be added to each well and incubated for 1 hour at room temperature. The plate may again be washed with wash buffer and 100 ⁇ l of substrate buffer containing OPD (O-phenylenediamine dihydrochloride (Sigma)) may be added to each well. The oxidation reaction, observed by the appearance of a yellow color, may be allowed to proceed for 30 minutes and stopped by the addition of 100 ⁇ l of 4.5 N H 2 SO 4 . The absorbance may then read at (492-405) nm.
  • OPD O-phenylenediamine dihydroch
  • An exemplary polypeptide variant is one that displays a “significant reduction in C1q binding” in this assay. This means that about 100 ⁇ g/ml of the polypeptide variant displays about 50 fold or more reduction in C1q binding compared to 100 ⁇ g/ml of a control antibody having a nonmutated IgG1 Fc region. In the most preferred embodiment, the polypeptide variant “does not bind C1q”, i.e. 100 ⁇ g/ml of the polypeptide variant displays about 100 fold or more reduction in C1q binding compared to 100 ⁇ g/ml of the control antibody.
  • Another exemplary variant is one which “has a better binding affinity for human C1q than the parent polypeptide”.
  • a molecule may display, for example, about two-fold or more, and preferably about five-fold or more, improvement in human C1q binding compared to the parent polypeptide (e.g. at the IC 50 values for these two molecules).
  • human C1q binding may be about two-fold to about 500-fold, and preferably from about two-fold or from about five-fold to about 1000-fold improved compared to the parent polypeptide.
  • a complement dependent cytotoxicity (CDC) assay may be performed, e.g. as described in Gazzano-Santoro et al., J. Immunol. Methods 202:163-171 (1997). Briefly, various concentrations of the polypeptide variant and human complement may be diluted with buffer. Cells which express the antigen to which the polypeptide variant binds may be diluted to a density of ⁇ 1 ⁇ 10 6 cells/ml. Mixtures of polypeptide variant, diluted human complement and cells expressing the antigen may be added to a flat bottom tissue culture 96 well plate and allowed to incubate for 2 hrs at 37° C. and 5% CO 2 to facilitate complement mediated cell lysis.
  • CDC complement dependent cytotoxicity
  • alamar blue (Accumed International) may then be added to each well and incubated overnight at 37° C.
  • the absorbance is measured using a 96-well fluorometer with excitation at 530 nm and emission at 590 nm.
  • the results may be expressed in relative fluorescence units (RFU).
  • the sample concentrations may be computed from a standard curve and the percent activity as compared to nonvariant polypeptide is reported for the polypeptide variant of interest.
  • Yet another exemplary variant “does not activate complement”.
  • 0.6 ⁇ g/ml of the polypeptide variant displays about 0-10% CDC activity in this assay compared to a 0.6 ⁇ g/ml of a control antibody having a nonmutated IgG1 Fc region.
  • the variant does not appear to have any CDC activity in the above CDC assay.
  • the invention also pertains to a polypeptide variant with enhanced CDC compared to a parent polypeptide, e.g., displaying about two-fold to about 100-fold improvement in CDC activity in vitro or in vivo (e.g. at the IC 50 values for each molecule being compared).
  • a receptor binding assay has been developed herein which is particularly useful for determining binding of an analyte of interest to a receptor where the affinity of the analyte for the receptor is relatively weak, e.g. in the micromolar range as is the case for Fc ⁇ RIIA, Fc ⁇ RIIB, Fc ⁇ RIIIA and Fc ⁇ RIIIB.
  • the method involves the formation of a molecular complex that has an improved avidity for the receptor of interest compared to the noncomplexed analyte.
  • the preferred molecular complex is an immune complex comprising: (a) an Fc region-containing polypeptide (such as an antibody or an immunoadhesin); (b) a first target molecule which comprises at least two binding sites for the Fc region-containing polypeptide; and (c) a second target molecule which comprises at least two binding sites for the first target molecule.
  • an Fc region-containing polypeptide such as an antibody or an immunoadhesin
  • a first target molecule which comprises at least two binding sites for the Fc region-containing polypeptide
  • a second target molecule which comprises at least two binding sites for the first target molecule.
  • the Fc region-containing polypeptide is an anti-IgE antibody, such as the E27 antibody ( FIGS. 4A-4B ).
  • E27 when mixed with human IgE at an 1:1 molar ratio, forms a stable hexamer consisting of three E27 molecules and three IgE molecules.
  • the “first target molecule” is a chimeric form of IgE in which the Fab portion of an anti-VEGF antibody is fused to the human IgE Fc portion and the “second target molecule” is the antigen to which the Fab binds (i.e. VEGF).
  • VEGF antigen to which the Fab binds
  • VEGF also binds two molecules of IgE per molecule of VEGF.
  • recombinant human VEGF was added at a 2:1 molar ratio to IgE:E27 hexamers, the hexamers were linked into larger molecular weight complexes via the IgE:VEGF interaction ( FIG. 5 ).
  • the Fc region of the anti-IgE antibody of the resultant immune complex binds to FcR with higher avidity than either uncomplexed anti-IgE or anti-IgE:IgE hexamers.
  • Examples comprising only an Fc region-containing polypeptide:first target molecule combination include an immunoadhesin:ligand combination such as VEGF receptor (KDR)-immunoadhesin:VEGF and a full-length bispecific antibody (bsAb):first target molecule.
  • an Fc region-containing polypeptide:first target molecule:second target molecule combination include a nonblocking antibody:soluble receptor:ligand combination such as anti-Trk antibody:soluble Trk receptor:neurotrophin (Urfer et al. J. Biol. Chem. 273(10):5829-5840 (1998)).
  • the immune complexes described above have further uses including evaluation of Fc region-containing polypeptide function and immune complex clearance in vivo.
  • the immune complex may be administered to a mammal (e.g. in a pre-clinical animal study) and evaluated for its half-life etc.
  • a polypeptide comprising at least the binding domain of the receptor of interest may be coated on solid phase, such as an assay plate.
  • the binding domain of the receptor alone or a receptor-fusion protein may be coated on the plate using standard procedures.
  • receptor-fusion proteins include receptor-glutathione S-transferase (GST) fusion protein, receptor-chitin binding domain fusion protein, receptor-hexaHis tag fusion protein (coated on glutathione, chitin, and nickel coated plates, respectively).
  • GST receptor-glutathione S-transferase
  • a capture molecule may be coated on the assay plate and used to bind the receptor-fusion protein via the non-receptor portion of the fusion protein.
  • Examples include anti-hexaHis F(ab′) 2 coated on the assay plate used to capture receptor-hexaHis tail fusion or anti-GST antibody coated on the assay plate used to capture a receptor-GST fusion.
  • binding to cells expressing at least the binding domain of the receptor may be evaluated.
  • the cells may be naturally occurring hematopoietic cells that express the FcR of interest or may be transformed with nucleic acid encoding the FcR or a binding domain thereof such that the binding domain is expressed at the surface of the cell to be tested.
  • the immune complex described hereinabove is added to the receptor-coated plates and incubated for a sufficient period of time such that the analyte binds to the receptor. Plates may then be washed to remove unbound complexes, and binding of the analyte may be detected according to known methods. For example, binding may be detected using a reagent (e.g. an antibody or fragment thereof) which binds specifically to the analyte, and which is optionally conjugated with a detectable label (detectable labels and methods for conjugating them to polypeptides are described below in the section entitled “Non-Therapeutic Uses for the Polypeptide Variant”).
  • a reagent e.g. an antibody or fragment thereof
  • the reagents can be provided in an assay kit, i.e., a packaged combination of reagents, for combination with the analyte in assaying the ability of the analyte to bind to a receptor of interest.
  • the components of the kit will generally be provided in predetermined ratios.
  • the kit may provide the first target molecule and/or the second target molecule, optionally complexed together.
  • the kit may further include assay plates coated with the receptor or a binding domain thereof (e.g. the extracellular domain of the ⁇ subunit of an FcR).
  • kits such as an antibody that binds specifically to the analyte to be assayed, labeled directly or indirectly with an enzymatic label
  • the detectable label is an enzyme
  • the kit will include substrates and cofactors required by the enzyme (e.g. a substrate precursor which provides the detectable chromophore or fluorophore).
  • other additives may be included such as stabilizers, buffers (e.g. assay and/or wash lysis buffer) and the like.
  • the relative amounts of the various reagents may be varied widely to provide for concentrations in solution of the reagents that substantially optimize the sensitivity of the assay.
  • the reagents may be provided as dry powders, usually lyophilized, including excipients that on dissolution will provide a reagent solution having the appropriate concentration.
  • the kit also suitably includes instructions for carrying out the assay.
  • the Fc region-containing polypeptide which is modified according to the teachings herein is an antibody. Techniques for producing antibodies follow:
  • the polypeptide is an antibody
  • it is directed against an antigen of interest.
  • the antigen is a biologically important polypeptide and administration of the antibody to a mammal suffering from a disease or disorder can result in a therapeutic benefit in that mammal.
  • antibodies directed against nonpolypeptide antigens are also contemplated.
  • the antigen is a polypeptide, it may be a transmembrane molecule (e.g. receptor) or ligand such as a growth factor.
  • exemplary antigens include molecules such as renin; a growth hormone, including human growth hormone and bovine growth hormone; growth hormone releasing factor; parathyroid hormone; thyroid stimulating hormone; lipoproteins; alpha-1-antitrypsin; insulin A-chain; insulin B-chain; proinsulin; follicle stimulating hormone; calcitonin; luteinizing hormone; glucagon; clotting factors such as factor VIIIC, factor IX, tissue factor (TF), and von Willebrands factor; anti-clotting factors such as Protein C; atrial natriuretic factor; lung surfactant; a plasminogen activator, such as urokinase or human urine or tissue-type plasminogen activator (t-PA); bombesin; thrombin; hemopoietic growth factor; tumor necrosis factor-alpha and
  • CD proteins such as CD3, CD4, CD8, CD19, CD20 and CD34
  • members of the ErbB receptor family such as the EGF receptor, HER2, HER3 or HER4 receptor
  • cell adhesion molecules such as LFA-1, Mac1, p 150.95, VLA-4, ICAM-1, VCAM, ⁇ 4/ ⁇ 7 integrin, and ⁇ v/ ⁇ 3 integrin including either ⁇ or ⁇ subunits thereof (e.g.
  • anti-CD11a, anti-CD18 or anti-CD11b antibodies growth factors such as VEGF; tissue factor (TF); alpha interferon ( ⁇ -IFN); an interleukin, such as IL-8; IgE; blood group antigens; flk2/flt3 receptor; obesity (OB) receptor; mpl receptor; CTLA-4; protein C etc.
  • growth factors such as VEGF; tissue factor (TF); alpha interferon ( ⁇ -IFN); an interleukin, such as IL-8; IgE; blood group antigens; flk2/flt3 receptor; obesity (OB) receptor; mpl receptor; CTLA-4; protein C etc.
  • Soluble antigens or fragments thereof, optionally conjugated to other molecules, can be used as immunogens for generating antibodies.
  • immunogens for transmembrane molecules, such as receptors, fragments of these (e.g. the extracellular domain of a receptor) can be used as the immunogen.
  • transmembrane molecules such as receptors
  • fragments of these e.g. the extracellular domain of a receptor
  • cells expressing the transmembrane molecule can be used as the immunogen.
  • Such cells can be derived from a natural source (e.g. cancer cell lines) or may be cells which have been transformed by recombinant techniques to express the transmembrane molecule.
  • Other antigens and forms thereof useful for preparing antibodies will be apparent to those in the art.
  • Polyclonal antibodies are preferably raised in animals by multiple subcutaneous (sc) or intraperitoneal (ip) injections of the relevant antigen and an adjuvant. It may be useful to conjugate the relevant antigen to a protein that is immunogenic in the species to be immunized, e.g., keyhole limpet hemocyanin, serum albumin, bovine thyroglobulin, or soybean trypsin inhibitor using a bifunctional or derivatizing agent, for example, maleimidobenzoyl sulfosuccinimide ester (conjugation through cysteine residues), N-hydroxysuccinimide (through lysine residues), glutaraldehyde, succinic anhydride, SOCl 2 , or R 1 N ⁇ C ⁇ NR, where R and R 1 are different alkyl groups.
  • a protein that is immunogenic in the species to be immunized e.g., keyhole limpet hemocyanin, serum albumin, bovine thy
  • Animals are immunized against the antigen, immunogenic conjugates, or derivatives by combining, e.g., 100 ⁇ g or 5 ⁇ g of the protein or conjugate (for rabbits or mice, respectively) with 3 volumes of Freund's complete adjuvant and injecting the solution intradermally at multiple sites.
  • the animals are boosted with 1 ⁇ 5 to 1/10 the original amount of peptide or conjugate in Freund's complete adjuvant by subcutaneous injection at multiple sites.
  • Seven to 14 days later the animals are bled and the serum is assayed for antibody titer. Animals are boosted until the titer plateaus.
  • the animal is boosted with the conjugate of the same antigen, but conjugated to a different protein and/or through a different cross-linking reagent.
  • Conjugates also can be made in recombinant cell culture as protein fusions.
  • aggregating agents such as alum are suitably used to enhance the immune response.
  • Monoclonal antibodies may be made using the hybridoma method first described by Kohler et al., Nature, 256:495 (1975), or may be made by recombinant DNA methods (U.S. Pat. No. 4,816,567).
  • a mouse or other appropriate host animal such as a hamster or macaque monkey
  • lymphocytes that produce or are capable of producing antibodies that will specifically bind to the protein used for immunization.
  • lymphocytes may be immunized in vitro. Lymphocytes then are fused with myeloma cells using a suitable fusing agent, such as polyethylene glycol, to form a hybridoma cell (Goding, Monoclonal Antibodies: Principles and Practice, pp. 59-103 (Academic Press, 1986)).
  • the hybridoma cells thus prepared are seeded and grown in a suitable culture medium that preferably contains one or more substances that inhibit the growth or survival of the unfused, parental myeloma cells.
  • a suitable culture medium that preferably contains one or more substances that inhibit the growth or survival of the unfused, parental myeloma cells.
  • the culture medium for the hybridomas typically will include hypoxanthine, aminopterin, and thymidine (HAT medium), which substances prevent the growth of HGPRT-deficient cells.
  • Preferred myeloma cells are those that fuse efficiently, support stable high-level production of antibody by the selected antibody-producing cells, and are sensitive to a medium such as HAT medium.
  • preferred myeloma cell lines are murine myeloma lines, such as those derived from MOPC-21 and MPC-11 mouse tumors available from the Salk Institute Cell Distribution Center, San Diego, Calif. USA, and SP-2 or X63-Ag8-653 cells available from the American Type Culture Collection, Rockville, Md. USA.
  • Human myeloma and mouse-human heteromyeloma cell lines also have been described for the production of human monoclonal antibodies (Kozbor, J. Immunol., 133:3001 (1984); Brodeur et al., Monoclonal Antibody Production Techniques and Applications, pp. 51-63 (Marcel Dekker, Inc., New York, 1987)).
  • Culture medium in which hybridoma cells are growing is assayed for production of monoclonal antibodies directed against the antigen.
  • the binding specificity of monoclonal antibodies produced by hybridoma cells is determined by immunoprecipitation or by an in vitro binding assay, such as radioimmunoassay (RIA) or enzyme-linked immunoabsorbent assay (ELISA).
  • RIA radioimmunoassay
  • ELISA enzyme-linked immunoabsorbent assay
  • the clones may be subcloned by limiting dilution procedures and grown by standard methods (Goding, Monoclonal Antibodies: Principles and Practice, pp. 59-103 (Academic Press, 1986)). Suitable culture media for this purpose include, for example, D-MEM or RPMI-1640 medium.
  • the hybridoma cells may be grown in vivo as ascites tumors in an animal.
  • the monoclonal antibodies secreted by the subclones are suitably separated from the culture medium, ascites fluid, or serum by conventional immunoglobulin purification procedures such as, for example, protein A-Sepharose, hydroxylapatite chromatography, gel electrophoresis, dialysis, or affinity chromatography.
  • DNA encoding the monoclonal antibodies is readily isolated and sequenced using conventional procedures (e.g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of the monoclonal antibodies).
  • the hybridoma cells serve as a preferred source of such DNA.
  • the DNA may be placed into expression vectors, which are then transfected into host cells such as E. coli cells, simian COS cells, Chinese hamster ovary (CHO) cells, or myeloma cells that do not otherwise produce immunoglobulin protein, to obtain the synthesis of monoclonal antibodies in the recombinant host cells. Recombinant production of antibodies will be described in more detail below.
  • antibodies or antibody fragments can be isolated from antibody phage libraries generated using the techniques described in McCafferty et al., Nature, 348:552-554 (1990). Clackson et al., Nature, 352:624-628 (1991) and Marks et al., J. Mol. Biol., 222:581-597 (1991) describe the isolation of murine and human antibodies, respectively, using phage libraries.
  • the DNA also may be modified, for example, by substituting the coding sequence for human heavy- and light-chain constant domains in place of the homologous murine sequences (U.S. Pat. No. 4,816,567; Morrison, et al., Proc. Natl Acad. Sci. USA, 81:6851 (1984)), or by covalently joining to the immunoglobulin coding sequence all or part of the coding sequence for a non-immunoglobulin polypeptide.
  • non-immunoglobulin polypeptides are substituted for the constant domains of an antibody, or they are substituted for the variable domains of one antigen-combining site of an antibody to create a chimeric bivalent antibody comprising one antigen-combining site having specificity for an antigen and another antigen-combining site having specificity for a different antigen.
  • a humanized antibody has one or more amino acid residues introduced into it from a source which is non-human. These non-human amino acid residues are often referred to as “import” residues, which are typically taken from an “import” variable domain. Humanization can be essentially performed following the method of Winter and co-workers (Jones et al., Nature, 321:522-525 (1986); Riechmann et al., Nature, 332:323-327 (1988); Verhoeyen et al., Science, 239:1534-1536 (1988)), by substituting rodent CDRs or CDR sequences for the corresponding sequences of a human antibody. Accordingly, such “humanized” antibodies are chimeric antibodies (U.S. Pat. No.
  • humanized antibodies are typically human antibodies in which some CDR residues and possibly some FR residues are substituted by residues from analogous sites in rodent antibodies.
  • variable domains both light and heavy
  • the choice of human variable domains, both light and heavy, to be used in making the humanized antibodies is very important to reduce antigenicity.
  • the sequence of the variable domain of a rodent antibody is screened against the entire library of known human variable-domain sequences.
  • the human sequence which is closest to that of the rodent is then accepted as the human framework (FR) for the humanized antibody (Sims et al., J. Immunol, 151:2296 (1993); Chothia et al., J. Mol. Biol., 196:901 (1987)).
  • Another method uses a particular framework derived from the consensus sequence of all human antibodies of a particular subgroup of light or heavy chains.
  • the same framework may be used for several different humanized antibodies (Carter et al., Proc. Natl. Acad. Sci. USA, 89:4285 (1992); Presta et al., J. Immunol., 151:2623 (1993)).
  • humanized antibodies are prepared by a process of analysis of the parental sequences and various conceptual humanized products using three-dimensional models of the parental and humanized sequences.
  • Three-dimensional immunoglobulin models are commonly available and are familiar to those skilled in the art.
  • Computer programs are available which illustrate and display probable three-dimensional conformational structures of selected candidate immunoglobulin sequences. Inspection of these displays permits analysis of the likely role of the residues in the functioning of the candidate immunoglobulin sequence, i.e., the analysis of residues that influence the ability of the candidate immunoglobulin to bind its antigen.
  • FR residues can be selected and combined from the recipient and import sequences so that the desired antibody characteristic, such as increased affinity for the target antigen(s), is achieved.
  • the CDR residues are directly and most substantially involved in influencing antigen binding.
  • transgenic animals e.g., mice
  • transgenic animals e.g., mice
  • J H antibody heavy-chain joining region
  • Human antibodies can also be derived from phage-display libraries (Hoogenboom et al., J. Mol. Biol., 227:381 (1991); Marks et al., J. Mol. Biol., 222:581-597 (1991); Vaughan et al. Nature Biotech 14:309 (1996)).
  • Multispecific antibodies have binding specificities for at least two different antigens. While such molecules normally will only bind two antigens (i.e. bispecific antibodies, BsAbs), antibodies with additional specificities such as trispecific antibodies are encompassed by this expression when used herein.
  • BsAbs include those with one arm directed against a tumor cell antigen and the other arm directed against a cytotoxic trigger molecule such as anti-Fc ⁇ RI/anti-CD15, anti-p185 HER2 /Fc ⁇ RIII (CD16), anti-CD3/anti-malignant B-cell (1D10), anti-CD3/anti-p185 HER2 , anti-CD3/anti-p97, anti-CD3/anti-renal cell carcinoma, anti-CD3/anti-OVCAR-3, anti-CD3/L-D1 (anti-colon carcinoma), anti-CD3/anti-melanocyte stimulating hormone analog, anti-EGF receptor/anti-CD3, anti-CD3/anti-CAMA1, anti-CD3/anti-CD19, anti-CD3/MoV18, anti-neural cell ahesion molecule (NCAM)/anti-CD3, anti-folate binding protein (FBP)/anti-CD3, anti-pan carcinoma associated antigen (AMOC-31)/anti-CD3; BsAbs with one arm which binds
  • BsAbs for use in therapy of infectious diseases such as anti-CD3/anti-herpes simplex virus (HSV), anti-T-cell receptor:CD3 complex/anti-influenza, anti-Fc ⁇ R/anti-HIV; BsAbs for tumor detection in vitro or in vivo such as anti-CEA/anti-EOTUBE, anti-CEA/anti-DPTA, anti-p185 HER2 /anti-hapten; BsAbs as vaccine adjuvants; and BsAbs as diagnostic tools such as anti-rabbit IgG/anti-ferritin, anti-horse radish peroxidase (HRP)/anti-hormone, anti-somatostatin/anti-substance P, anti-HRP/anti-FITC, anti-CEA/anti- ⁇ -galactosidase.
  • HRP anti-horse radish peroxidase
  • HRP anti-somatostatin/anti-substance P
  • trispecific antibodies examples include anti-CD3/anti-CD4/anti-CD37, anti-CD3/anti-CD5/anti-CD37 and anti-CD3/anti-CD8/anti-CD37.
  • Bispecific antibodies can be prepared as full length antibodies or antibody fragments (e.g. F(ab′) 2 bispecific antibodies).
  • bispecific antibodies are known in the art. Traditional production of full length bispecific antibodies is based on the coexpression of two immunoglobulin heavy chain-light chain pairs, where the two chains have different specificities (Millstein et al., Nature, 305:537-539 (1983)). Because of the random assortment of immunoglobulin heavy and light chains, these hybridomas (quadromas) produce a potential mixture of 10 different antibody molecules, of which only one has the correct bispecific structure. Purification of the correct molecule, which is usually done by affinity chromatography steps, is rather cumbersome, and the product yields are low. Similar procedures are disclosed in WO 93/08829, and in Traunecker et al., EMBO J., 10:3655-3659 (1991).
  • antibody variable domains with the desired binding specificities are fused to immunoglobulin constant domain sequences.
  • the fusion preferably is with an immunoglobulin heavy chain constant domain, comprising at least part of the hinge, CH2, and CH3 regions. It is preferred to have the first heavy-chain constant region (CH1) containing the site necessary for light chain binding, present in at least one of the fusions.
  • DNAs encoding the immunoglobulin heavy chain fusions and, if desired, the immunoglobulin light chain are inserted into separate expression vectors, and are co-transfected into a suitable host organism.
  • the bispecific antibodies are composed of a hybrid immunoglobulin heavy chain with a first binding specificity in one arm, and a hybrid immunoglobulin heavy chain-light chain pair (providing a second binding specificity) in the other arm. It was found that this asymmetric structure facilitates the separation of the desired bispecific compound from unwanted immunoglobulin chain combinations, as the presence of an immunoglobulin light chain in only one half of the bispecific molecule provides for a facile way of separation. This approach is disclosed in WO 94/04690. For further details of generating bispecific antibodies see, for example, Suresh et al., Methods in Enzymology, 121:210 (1986).
  • the interface between a pair of antibody molecules can be engineered to maximize the percentage of heterodimers which are recovered from recombinant cell culture.
  • the preferred interface comprises at least a part of the C H 3 domain of an antibody constant domain.
  • one or more small amino acid side chains from the interface of the first antibody molecule are replaced with larger side chains (e.g. tyrosine or tryptophan).
  • Compensatory “cavities” of identical or similar size to the large side chain(s) are created on the interface of the second antibody molecule by replacing large amino acid side chains with smaller ones (e.g. alanine or threonine). This provides a mechanism for increasing the yield of the heterodimer over other unwanted end-products such as homodimers.
  • Bispecific antibodies include cross-linked or “heteroconjugate” antibodies.
  • one of the antibodies in the heteroconjugate can be coupled to avidin, the other to biotin.
  • Such antibodies have, for example, been proposed to target immune system cells to unwanted cells (U.S. Pat. No. 4,676,980), and for treatment of HIV infection (WO 91/00360, WO 92/200373, and EP 03089).
  • Heteroconjugate antibodies may be made using any convenient cross-linking methods. Suitable cross-linking agents are well known in the art, and are disclosed in U.S. Pat. No. 4,676,980, along with a number of cross-linking techniques.
  • Antibodies with more than two valencies are contemplated.
  • trispecific antibodies can be prepared. Tutt et al., J. Immunol. 147: 60 (1991).
  • polypeptide of interest herein is preferably an antibody
  • Fc region-containing polypeptides which can be modified according to the methods described herein are contemplated.
  • An example of such a molecule is an immunoadhesin.
  • the simplest and most straightforward immunoadhesin design combines the binding domain(s) of the adhesin (e.g. the extracellular domain (ECD) of a receptor) with the Fc region of an immunoglobulin heavy chain.
  • ECD extracellular domain
  • nucleic acid encoding the binding domain of the adhesin will be fused C-terminally to nucleic acid encoding the N-terminus of an immunoglobulin constant domain sequence, however N-terminal fusions are also possible.
  • the encoded chimeric polypeptide will retain at least functionally active hinge, C H 2 and C H 3 domains of the constant region of an immunoglobulin heavy chain. Fusions are also made to the C-terminus of the Fc portion of a constant domain, or immediately N-terminal to the C H 1 of the heavy chain or the corresponding region of the light chain.
  • the precise site at which the fusion is made is not critical; particular sites are well known and may be selected in order to optimize the biological activity, secretion, or binding characteristics of the immunoadhesin.
  • the adhesin sequence is fused to the N-terminus of the Fc region of immunoglobulin G 1 (IgG 1 ). It is possible to fuse the entire heavy chain constant region to the adhesin sequence. However, more preferably, a sequence beginning in the hinge region just upstream of the papain cleavage site which defines IgG Fc chemically (i.e. residue 216, taking the first residue of heavy chain constant region to be 114), or analogous sites of other immunoglobulins is used in the fusion.
  • the adhesin amino acid sequence is fused to (a) the hinge region and C H 2 and C H 3 or (b) the C H 1, hinge, C H 2 and C H 3 domains, of an IgG heavy chain.
  • the immunoadhesins are assembled as multimers, and particularly as heterodimers or heterotetramers.
  • these assembled immunoglobulins will have known unit structures.
  • a basic four chain structural unit is the form in which IgG, IgD, and IgE exist.
  • a four chain unit is repeated in the higher molecular weight immunoglobulins; IgM generally exists as a pentamer of four basic units held together by disulfide bonds.
  • IgA globulin, and occasionally IgG globulin may also exist in multimeric form in serum. In the case of multimer, each of the four units may be the same or different.
  • each A represents identical or different adhesin amino acid sequences
  • V L is an immunoglobulin light chain variable domain
  • V H is an immunoglobulin heavy chain variable domain
  • C L is an immunoglobulin light chain constant domain
  • C H is an immunoglobulin heavy chain constant domain
  • n is an integer greater than 1;
  • Y designates the residue of a covalent cross-linking agent.
  • the adhesin sequences can be inserted between immunoglobulin heavy chain and light chain sequences, such that an immunoglobulin comprising a chimeric heavy chain is obtained.
  • the adhesin sequences are fused to the 3′ end of an immunoglobulin heavy chain in each arm of an immunoglobulin, either between the hinge and the C H 2 domain, or between the C H 2 and C H 3 domains. Similar constructs have been reported by Hoogenboom, et al., Mol. Immunol. 28:1027-1037 (1991).
  • an immunoglobulin light chain might be present either covalently associated to an adhesin-immunoglobulin heavy chain fusion polypeptide, or directly fused to the adhesin.
  • DNA encoding an immunoglobulin light chain is typically coexpressed with the DNA encoding the adhesin-immunoglobulin heavy chain fusion protein.
  • the hybrid heavy chain and the light chain will be covalently associated to provide an immunoglobulin-like structure comprising two disulfide-linked immunoglobulin heavy chain-light chain pairs.
  • Immunoadhesins are most conveniently constructed by fusing the cDNA sequence encoding the adhesin portion in-frame to an immunoglobulin cDNA sequence.
  • fusion to genomic immunoglobulin fragments can also be used (see, e.g. Aruffo et al., Cell 61:1303-1313 (1990); and Stamenkovic et al., Cell 66:1133-1144 (1991)).
  • the latter type of fusion requires the presence of Ig regulatory sequences for expression.
  • cDNAs encoding IgG heavy-chain constant regions can be isolated based on published sequences from cDNA libraries derived from spleen or peripheral blood lymphocytes, by hybridization or by polymerase chain reaction (PCR) techniques.
  • the cDNAs encoding the “adhesin” and the immunoglobulin parts of the immunoadhesin are inserted in tandem into a plasmid vector that directs efficient expression in the chosen host cells.
  • the invention also provides isolated nucleic acid encoding a polypeptide variant as disclosed herein, vectors and host cells comprising the nucleic acid, and recombinant techniques for the production of the polypeptide variant.
  • the nucleic acid encoding it is isolated and inserted into a replicable vector for further cloning (amplification of the DNA) or for expression.
  • DNA encoding the polypeptide variant is readily isolated and sequenced using conventional procedures (e.g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the polypeptide variant).
  • Many vectors are available.
  • the vector components generally include, but are not limited to, one or more of the following: a signal sequence, an origin of replication, one or more marker genes, an enhancer element, a promoter, and a transcription termination sequence.
  • polypeptide variant of this invention may be produced recombinantly not only directly, but also as a fusion polypeptide with a heterologous polypeptide, which is preferably a signal sequence or other polypeptide having a specific cleavage site at the N-terminus of the mature protein or polypeptide.
  • a heterologous polypeptide which is preferably a signal sequence or other polypeptide having a specific cleavage site at the N-terminus of the mature protein or polypeptide.
  • the heterologous signal sequence selected preferably is one that is recognized and processed (i.e., cleaved by a signal peptidase) by the host cell.
  • the signal sequence is substituted by a prokaryotic signal sequence selected, for example, from the group of the alkaline phosphatase, penicillinase, Ipp, or heat-stable enterotoxin II leaders.
  • a prokaryotic signal sequence selected, for example, from the group of the alkaline phosphatase, penicillinase, Ipp, or heat-stable enterotoxin II leaders.
  • yeast secretion the native signal sequence may be substituted by, e.g., the yeast invertase leader, a factor leader (including Saccharomyces and Kluyveromyces ⁇ -factor leaders), or acid phosphatase leader, the C. albicans glucoamylase leader, or the signal described in WO 90/13646.
  • mammalian signal sequences as well as viral secretory leaders for example, the herpes simplex gD signal, are available.
  • the DNA for such precursor region is ligated in reading frame to DNA encoding the polypeptide variant.
  • Both expression and cloning vectors contain a nucleic acid sequence that enables the vector to replicate in one or more selected host cells.
  • this sequence is one that enables the vector to replicate independently of the host chromosomal DNA, and includes origins of replication or autonomously replicating sequences.
  • origins of replication or autonomously replicating sequences are well known for a variety of bacteria, yeast, and viruses.
  • the origin of replication from the plasmid pBR322 is suitable for most Gram-negative bacteria, the 2 ⁇ plasmid origin is suitable for yeast, and various viral origins (SV40, polyoma, adenovirus, VSV or BPV) are useful for cloning vectors in mammalian cells.
  • the origin of replication component is not needed for mammalian expression vectors (the SV40 origin may typically be used only because it contains the early promoter).
  • Selection genes may contain a selection gene, also termed a selectable marker.
  • Typical selection genes encode proteins that (a) confer resistance to antibiotics or other toxins, e.g., ampicillin, neomycin, methotrexate, or tetracycline, (b) complement auxotrophic deficiencies, or (c) supply critical nutrients not available from complex media, e.g., the gene encoding D-alanine racemase for Bacilli.
  • One example of a selection scheme utilizes a drug to arrest growth of a host cell. Those cells that are successfully transformed with a heterologous gene produce a protein conferring drug resistance and thus survive the selection regimen. Examples of such dominant selection use the drugs neomycin, mycophenolic acid and hygromycin.
  • suitable selectable markers for mammalian cells are those that enable the identification of cells competent to take up the polypeptide variant nucleic acid, such as DHFR, thymidine kinase, metallothionein-I and -II, preferably primate metallothionein genes, adenosine deaminase, ornithine decarboxylase, etc.
  • cells transformed with the DHFR selection gene are first identified by culturing all of the transformants in a culture medium that contains methotrexate (Mtx), a competitive antagonist of DHFR.
  • Mtx methotrexate
  • An appropriate host cell when wild-type DHFR is employed is the Chinese hamster ovary (CHO) cell line deficient in DHFR activity.
  • host cells transformed or co-transformed with DNA sequences encoding polypeptide variant, wild-type DHFR protein, and another selectable marker such as aminoglycoside 3′-phosphotransferase (APH) can be selected by cell growth in medium containing a selection agent for the selectable marker such as an aminoglycosidic antibiotic, e.g., kanamycin, neomycin, or G418. See U.S. Pat. No. 4,965,199.
  • APH aminoglycoside 3′-phosphotransferase
  • a suitable selection gene for use in yeast is the trp1 gene present in the yeast plasmid YRp7 (Stinchcomb et al., Nature, 282:39 (1979)).
  • the trp1 gene provides a selection marker for a mutant strain of yeast lacking the ability to grow in tryptophan, for example, ATCC No. 44076 or PEP4-1. Jones, Genetics, 85:12 (1977).
  • the presence of the trpl lesion in the yeast host cell genome then provides an effective environment for detecting transformation by growth in the absence of tryptophan.
  • Leu2-deficient yeast strains (ATCC 20,622 or 38,626) are complemented by known plasmids bearing the Leu2 gene.
  • vectors derived from the 1.6 ⁇ m circular plasmid pKD1 can be used for transformation of Kluyveromyces yeasts.
  • an expression system for large-scale production of recombinant calf chymosin was reported for K. lactis. Van den Berg, Bio/Technology, 8:135 (1990).
  • Stable multi-copy expression vectors for secretion of mature recombinant human serum albumin by industrial strains of Kluyveromyces have also been disclosed. Fleer et al., Bio/Technology, 9:968-975 (1991).
  • Expression and cloning vectors usually contain a promoter that is recognized by the host organism and is operably linked to the polypeptide variant nucleic acid.
  • Promoters suitable for use with prokaryotic hosts include the phoA promoter, ⁇ -lactamase and lactose promoter systems, alkaline phosphatase, a tryptophan (trp) promoter system, and hybrid promoters such as the tac promoter.
  • trp tryptophan
  • Other known bacterial promoters are suitable. Promoters for use in bacterial systems also will contain a Shine-Dalgarno (S.D.) sequence operably linked to the DNA encoding the polypeptide variant.
  • S.D. Shine-Dalgarno
  • Promoter sequences are known for eukaryotes. Virtually all eukaryotic genes have an AT-rich region located approximately 25 to 30 bases upstream from the site where transcription is initiated. Another sequence found 70 to 80 bases upstream from the start of transcription of many genes is a CNCAAT region where N may be any nucleotide. At the 3′ end of most eukaryotic genes is an AATAAA sequence that may be the signal for addition of the poly A tail to the 3′ end of the coding sequence. All of these sequences are suitably inserted into eukaryotic expression vectors.
  • suitable promoting sequences for use with yeast hosts include the promoters for 3-phosphoglycerate kinase or other glycolytic enzymes, such as enolase, glyceraldehyde-3-phosphate dehydrogenase, hexokinase, pyruvate decarboxylase, phosphofructokinase, glucose-6-phosphate isomerase, 3-phosphoglycerate mutase, pyruvate kinase, triosephosphate isomerase, phosphoglucose isomerase, and glucokinase.
  • 3-phosphoglycerate kinase or other glycolytic enzymes such as enolase, glyceraldehyde-3-phosphate dehydrogenase, hexokinase, pyruvate decarboxylase, phosphofructokinase, glucose-6-phosphate isomerase, 3-phosphoglycerate mutase, pyruvate
  • yeast promoters which are inducible promoters having the additional advantage of transcription controlled by growth conditions, are the promoter regions for alcohol dehydrogenase 2, isocytochrome C, acid phosphatase, degradative enzymes associated with nitrogen metabolism, metallothionein, glyceraldehyde-3-phosphate dehydrogenase, and enzymes responsible for maltose and galactose utilization.
  • Suitable vectors and promoters for use in yeast expression are further described in EP 73,657.
  • Yeast enhancers also are advantageously used with yeast promoters.
  • Polypeptide variant transcription from vectors in mammalian host cells is controlled, for example, by promoters obtained from the genomes of viruses such as polyoma virus, fowlpox virus, adenovirus (such as Adenovirus 2), bovine papilloma virus, avian sarcoma virus, cytomegalovirus, a retrovirus, hepatitis-B virus and most preferably Simian Virus 40 (SV40), from heterologous mammalian promoters, e.g., the actin promoter or an immunoglobulin promoter, from heat-shock promoters, provided such promoters are compatible with the host cell systems.
  • viruses such as polyoma virus, fowlpox virus, adenovirus (such as Adenovirus 2), bovine papilloma virus, avian sarcoma virus, cytomegalovirus, a retrovirus, hepatitis-B virus and most preferably Simian Virus 40 (
  • the early and late promoters of the SV40 virus are conveniently obtained as an SV40 restriction fragment that also contains the SV40 viral origin of replication.
  • the immediate early promoter of the human cytomegalovirus is conveniently obtained as a HindIII E restriction fragment.
  • a system for expressing DNA in mammalian hosts using the bovine papilloma virus as a vector is disclosed in U.S. Pat. No. 4,419,446. A modification of this system is described in U.S. Pat. No. 4,601,978. See also Reyes et al., Nature 297:598-601 (1982) on expression of human ⁇ -interferon cDNA in mouse cells under the control of a thymidine kinase promoter from herpes simplex virus. Alternatively, the rous sarcoma virus long terminal repeat can be used as the promoter.
  • Enhancer sequences are now known from mammalian genes (globin, elastase, albumin, ⁇ -fetoprotein, and insulin). Typically, however, one will use an enhancer from a eukaryotic cell virus. Examples include the SV40 enhancer on the late side of the replication origin (bp 100-270), the cytomegalovirus early promoter enhancer, the polyoma enhancer on the late side of the replication origin, and adenovirus enhancers.
  • the enhancer may be spliced into the vector at a position 5′ or 3′ to the polypeptide variant-encoding sequence, but is preferably located at a site 5′ from the promoter.
  • Expression vectors used in eukaryotic host cells will also contain sequences necessary for the termination of transcription and for stabilizing the mRNA. Such sequences are commonly available from the 5′ and, occasionally 3′, untranslated regions of eukaryotic or viral DNAs or cDNAs. These regions contain nucleotide segments transcribed as polyadenylated fragments in the untranslated portion of the mRNA encoding the polypeptide variant.
  • One useful transcription termination component is the bovine growth hormone polyadenylation region. See WO94/11026 and the expression vector disclosed therein.
  • Suitable host cells for cloning or expressing the DNA in the vectors herein are the prokaryote, yeast, or higher eukaryote cells described above.
  • Suitable prokaryotes for this purpose include eubacteria, such as Gram-negative or Gram-positive organisms, for example, Enterobacteriaceae such as Escherichia, e.g., E. coli, Enterobacter, Erwinia, Klebsiella, Proteus, Salmonella, e.g., Salmonella typhimurium, Serratia, e.g., Serratia marcescans, and Shigella, as well as Bacilli such as B. subtilis and B. licheniformis (e.g., B.
  • E. coli 294 ATCC 31,446
  • E. coli B E. coli X1776
  • E. coli W3110 ATCC 27,325
  • eukaryotic microbes such as filamentous fungi or yeast are suitable cloning or expression hosts for polypeptide variant-encoding vectors.
  • Saccharomyces cerevisiae or common baker's yeast, is the most commonly used among lower eukaryotic host microorganisms.
  • a number of other genera, species, and strains are commonly available and useful herein, such as Schizosaccharomyces pombe; Kluyveromyces hosts such as, e.g., K. lactis, K. fragilis (ATCC 12,424), K. bulgaricus (ATCC 16,045), K. wickeramii (ATCC 24,178), K.
  • waltii ATCC 56,500
  • K. drosophilarum ATCC 36,906
  • K. thermotolerans K. marxianus
  • yarrowia EP 402,226
  • Pichia pastoris EP 183,070
  • Candida Trichoderma reesia
  • EP 244,234 Neurospora crassa
  • Schwanniomyces such as Schwanniomyces occidentalis
  • filamentous fungi such as, e.g., Neurospora, Penicillium, Tolypocladium , and Aspergillus hosts such as A. nidulans and A. niger.
  • Suitable host cells for the expression of glycosylated polypeptide variant are derived from multicellular organisms.
  • invertebrate cells include plant and insect cells.
  • Numerous baculoviral strains and variants and corresponding permissive insect host cells from hosts such as Spodoptera frugiperda (caterpillar), Aedes aegypti (mosquito), Aedes albopictus (mosquito), Drosophila melanogaster (fruitfly), and Bombyx mori have been identified.
  • a variety of viral strains for transfection are publicly available, e.g., the L-1 variant of Autographa californica NPV and the Bm-5 strain of Bombyx mori NPV, and such viruses may be used as the virus herein according to the present invention, particularly for transfection of Spodoptera frugiperda cells.
  • Plant cell cultures of cotton, corn, potato, soybean, petunia, tomato, and tobacco can also be utilized as hosts.
  • vertebrate cells have been greatest in vertebrate cells, and propagation of vertebrate cells in culture (tissue culture) has become a routine procedure.
  • useful mammalian host cell lines are monkey kidney CV1 line transformed by SV40 (COS-7, ATCC CRL 1651); human embryonic kidney line (293 or 293 cells subcloned for growth in suspension culture, Graham et al., J. Gen Virol. 36:59 (1977)); baby hamster kidney cells (BHK, ATCC CCL 10); Chinese hamster ovary cells/-DHFR (CHO, Urlaub et al., Proc. Natl. Acad. Sci. USA 77:4216 (1980)); mouse sertoli cells (TM4, Mather, Biol. Reprod.
  • monkey kidney cells (CV1 ATCC CCL 70); African green monkey kidney cells (VERO-76, ATCC CRL-1587); human cervical carcinoma cells (HELA, ATCC CCL 2); canine kidney cells (MDCK, ATCC CCL 34); buffalo rat liver cells (BRL 3A, ATCC CRL 1442); human lung cells (W138, ATCC CCL 75); human liver cells (Hep G2, HB 8065); mouse mammary tumor (MMT 060562, ATCC CCL51); TRI cells (Mather et al., Annals N.Y. Acad. Sci. 383:44-68 (1982)); MRC 5 cells; FS4 cells; and a human hepatoma line (Hep G2).
  • Host cells are transformed with the above-described expression or cloning vectors for polypeptide variant production and cultured in conventional nutrient media modified as appropriate for inducing promoters, selecting transformants, or amplifying the genes encoding the desired sequences.
  • the host cells used to produce the polypeptide variant of this invention may be cultured in a variety of media.
  • Commercially available media such as Ham's F10 (Sigma), Minimal Essential Medium ((MEM), (Sigma), RPMI-1640 (Sigma), and Dulbecco's Modified Eagle's Medium ((DMEM), Sigma) are suitable for culturing the host cells.
  • any of these media may be supplemented as necessary with hormones and/or other growth factors (such as insulin, transferrin, or epidermal growth factor), salts (such as sodium chloride, calcium, magnesium, and phosphate), buffers (such as HEPES), nucleotides (such as adenosine and thymidine), antibiotics (such as GENTAMYCINTM drug), trace elements (defined as inorganic compounds usually present at final concentrations in the micromolar range), and glucose or an equivalent energy source. Any other necessary supplements may also be included at appropriate concentrations that would be known to those skilled in the art.
  • the culture conditions such as temperature, pH, and the like, are those previously used with the host cell selected for expression, and will be apparent to the ordinarily skilled artisan.
  • the polypeptide variant can be produced intracellularly, in the periplasmic space, or directly secreted into the medium. If the polypeptide variant is produced intracellularly, as a first step, the particulate debris, either host cells or lysed fragments, is removed, for example, by centrifugation or ultrafiltration. Carter et al., Bio/Technology 10:163-167 (1992) describe a procedure for isolating antibodies which are secreted to the periplasmic space of E. coli . Briefly, cell paste is thawed in the presence of sodium acetate (pH 3.5), EDTA, and phenylmethylsulfonylfluoride (PMSF) over about 30 min.
  • sodium acetate pH 3.5
  • EDTA EDTA
  • PMSF phenylmethylsulfonylfluoride
  • Cell debris can be removed by centrifugation.
  • supernatants from such expression systems are generally first concentrated using a commercially available protein concentration filter, for example, an Amicon or Millipore Pellicon ultrafiltration unit.
  • a protease inhibitor such as PMSF may be included in any of the foregoing steps to inhibit proteolysis and antibiotics may be included to prevent the growth of adventitious contaminants.
  • the polypeptide variant composition prepared from the cells can be purified using, for example, hydroxylapatite chromatography, gel electrophoresis, dialysis, and affinity chromatography, with affinity chromatography being the preferred purification technique.
  • affinity chromatography is the preferred purification technique.
  • the suitability of protein A as an affinity ligand depends on the species and isotype of any immunoglobulin Fc region that is present in the polypeptide variant.
  • Protein A can be used to purify polypeptide variants that are based on human ⁇ 1, ⁇ 2, or ⁇ 4 heavy chains (Lindmark et al., J. lmmunol. Meth. 62:1-13 (1983)). Protein G is recommended for all mouse isotypes and for human ⁇ 3 (Guss et al., EMBO J.
  • the matrix to which the affinity ligand is attached is most often agarose, but other matrices are available. Mechanically stable matrices such as controlled pore glass or poly(styrenedivinyl)benzene allow for faster flow rates and shorter processing times than can be achieved with agarose. Where the polypeptide variant comprises a C H 3 domain, the Bakerbond ABXTM resin (J. T. Baker, Phillipsburg, N.J.) is useful for purification.
  • the mixture comprising the polypeptide variant of interest and contaminants may be subjected to low pH hydrophobic interaction chromatography using an elution buffer at a pH between about 2.5-4.5, preferably performed at low salt concentrations (e.g.,from about 0-0.25M salt).
  • Therapeutic formulations of the polypeptide variant are prepared for storage by mixing the polypeptide variant having the desired degree of purity with optional physiologically acceptable carriers, excipients or stabilizers ( Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980)), in the form of lyophilized formulations or aqueous solutions.
  • Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptide; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine, arg
  • the formulation herein may also contain more than one active compound as necessary for the particular indication being treated, preferably those with complementary activities that do not adversely affect each other.
  • Such molecules are suitably present in combination in amounts that are effective for the purpose intended.
  • the active ingredients may also be entrapped in microcapsule prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsule and poly-(methylmethacylate) microcapsule, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) or in macroemulsions.
  • colloidal drug delivery systems for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules
  • the formulations to be used for in vivo administration must be sterile. This is readily accomplished by filtration through sterile filtration membranes.
  • Sustained-release preparations may be prepared. Suitable examples of sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing the polypeptide variant, which matrices are in the form of shaped articles, e.g., films, or microcapsule. Examples of sustained-release matrices include polyesters, hydrogels (for example, poly(2-hydroxyethyl-methacrylate), or poly(vinylalcohol)), polylactides (U.S. Pat. No.
  • copolymers of L-glutamic acid and ⁇ ethyl-L-glutamate copolymers of L-glutamic acid and ⁇ ethyl-L-glutamate, non-degradable ethylene-vinyl acetate, degradable lactic acid-glycolic acid copolymers such as the LUPRON DEPOTTM (injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate), and poly-D-( ⁇ )-3-hydroxybutyric acid. While polymers such as ethylene-vinyl acetate and lactic acid-glycolic acid enable release of molecules for over 100 days, certain hydrogels release proteins for shorter time periods.
  • encapsulated antibodies When encapsulated antibodies remain in the body for a long time, they may denature or aggregate as a result of exposure to moisture at 37° C., resulting in a loss of biological activity and possible changes in immunogenicity. Rational strategies can be devised for stabilization depending on the mechanism involved. For example, if the aggregation mechanism is discovered to be intermolecular S—S bond formation through thio-disulfide interchange, stabilization may be achieved by modifying sulfhydryl residues, lyophilizing from acidic solutions, controlling moisture content, using appropriate additives, and developing specific polymer matrix compositions.
  • the polypeptide variant of the invention may be used as an affinity purification agent.
  • the polypeptide variant is immobilized on a solid phase such a Sephadex resin or filter paper, using methods well known in the art.
  • the immobilized polypeptide variant is contacted with a sample containing the antigen to be purified, and thereafter the support is washed with a suitable solvent that will remove substantially all the material in the sample except the antigen to be purified, which is bound to the immobilized polypeptide variant. Finally, the support is washed with another suitable solvent, such as glycine buffer, pH 5.0, that will release the antigen from the polypeptide variant.
  • the polypeptide variant may also be useful in diagnostic assays, e.g., for detecting expression of an antigen of interest in specific cells, tissues, or serum.
  • polypeptide variant typically will be labeled with a detectable moiety.
  • labels are available which can be generally grouped into the following categories:
  • Radioisotopes such as 35 S, 14 C, 125 I, 3 H, and 131 I.
  • the polypeptide variant can be labeled with the radioisotope using the techniques described in Current Protocols in Immunology, Volumes 1 and 2, Coligen et al., Ed. Wiley-Interscience, New York, N.Y., Pubs. (1991) for example and radioactivity can be measured using scintillation counting.
  • Fluorescent labels such as rare earth chelates (europium chelates) or fluorescein and its derivatives, rhodamine and its derivatives, dansyl, Lissamine, phycoerythrin and Texas Red are available.
  • the fluorescent labels can be conjugated to the polypeptide variant using the techniques disclosed in Current Protocols in Immunology, supra, for example. Fluorescence can be quantified using a fluorimeter.
  • the enzyme generally catalyzes a chemical alteration of the chromogenic substrate that can be measured using various techniques. For example, the enzyme may catalyze a color change in a substrate, which can be measured spectrophotometrically. Alternatively, the enzyme may alter the fluorescence or chemiluminescence of the substrate. Techniques for quantifying a change in fluorescence are described above.
  • the chemiluminescent substrate becomes electronically excited by a chemical reaction and may then emit light which can be measured (using a chemiluminometer, for example) or donates energy to a fluorescent acceptor.
  • enzymatic labels include luciferases (e.g., firefly luciferase and bacterial luciferase; U.S. Pat. No. 4,737,456), luciferin, 2,3-dihydrophthalazinediones, malate dehydrogenase, urease, peroxidase such as horseradish peroxidase (HRPO), alkaline phosphatase, ⁇ -galactosidase, glucoamylase, lysozyme, saccharide oxidases (e.g., glucose oxidase, galactose oxidase, and glucose-6-phosphate dehydrogenase), heterocyclic oxidases (such as uricase and xanthine oxidase), lactoperoxidase, microperoxidase, and the like.
  • luciferases e.g., firefly luciferase and bacterial lucifera
  • enzyme-substrate combinations include, for example:
  • HRPO Horseradish peroxidase
  • OPD orthophenylene diamine
  • TMB 3,3′,5,5′-tetramethyl benzidine hydrochloride
  • ⁇ -D-galactosidase ( ⁇ -D-Gal) with a chromogenic substrate (e.g., p-nitrophenyl- ⁇ -D-galactosidase) or fluorogenic substrate 4-methylumbelliferyl- ⁇ -D-galactosidase.
  • a chromogenic substrate e.g., p-nitrophenyl- ⁇ -D-galactosidase
  • fluorogenic substrate 4-methylumbelliferyl- ⁇ -D-galactosidase
  • the label is indirectly conjugated with the polypeptide variant.
  • the polypeptide variant can be conjugated with biotin and any of the three broad categories of labels mentioned above can be conjugated with avidin, or vice versa. Biotin binds selectively to avidin and thus, the label can be conjugated with the polypeptide variant in this indirect manner.
  • the polypeptide variant is conjugated with a small hapten (e.g., digoxin) and one of the different types of labels mentioned above is conjugated with an anti-hapten polypeptide variant (e.g., anti-digoxin antibody).
  • a small hapten e.g., digoxin
  • an anti-hapten polypeptide variant e.g., anti-digoxin antibody
  • the polypeptide variant need not be labeled, and the presence thereof can be detected using a labeled antibody which binds to the polypeptide variant.
  • polypeptide variant of the present invention may be employed in any known assay method, such as competitive binding assays, direct and indirect sandwich assays, and immunoprecipitation assays. Zola, Monoclonal Antibodies: A Manual of Techniques, pp. 147-158 (CRC Press, Inc. 1987).
  • the polypeptide variant may also be used for in vivo diagnostic assays.
  • the polypeptide variant is labeled with a radionuclide (such as 111 In, 99 Tc, 14 C, 131 I, 125 I, 3 H, 32 P or 35 S) so that the antigen or cells expressing it can be localized using immunoscintiography.
  • a radionuclide such as 111 In, 99 Tc, 14 C, 131 I, 125 I, 3 H, 32 P or 35 S
  • the polypeptide variant of the present invention may be used to treat a mammal e.g. a patient suffering from, or predisposed to, a disease or disorder who could benefit from administration of the polypeptide variant.
  • the conditions which can be treated with the polypeptide variant are many and include cancer (e.g. where the polypeptide variant binds the HER2 receptor, CD20 or vascular endothelial growth factor (VEGF)); allergic conditions such as asthma (with an anti-IgE antibody); and LFA-1-mediated disorders (e.g. where the polypeptide variant is an anti-LFA-1 or anti-ICAM-1 antibody) etc.
  • cancer e.g. where the polypeptide variant binds the HER2 receptor, CD20 or vascular endothelial growth factor (VEGF)
  • allergic conditions such as asthma (with an anti-IgE antibody)
  • LFA-1-mediated disorders e.g. where the polypeptide variant is an anti-LFA-1 or anti-ICAM-1 antibody
  • the disorder preferably is HER2-expressing cancer, e.g. a benign or malignant tumor characterized by overexpression of the HER2 receptor.
  • cancers include, but are not limited to, breast cancer, squamous cell cancer, small-cell lung cancer, non-small cell lung cancer, gastrointestinal cancer, pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, bladder cancer, hepatoma, colon cancer, colorectal cancer, endometrial carcinoma, salivary gland carcinoma, kidney cancer, liver cancer, prostate cancer, vulval cancer, thyroid cancer, hepatic carcinoma and various types of head and neck cancer.
  • a polypeptide with a variant Fc region which has improved, or diminished, ADCC activity. Such molecules will find applications in the treatment of different disorders.
  • the polypeptide variant with improved ADCC activity may be employed in the treatment of diseases or disorders where destruction or elimination of tissue or foreign micro-organisms is desired.
  • the polypeptide may be used to treat cancer; inflammatory disorders; infections (e.g. bacterial, viral, fungal or yeast infections); and other conditions (such as goiter) where removal of tissue is desired, etc.
  • the polypeptide variant may be used to treat diseases or disorders where a Fc region-containing polypeptide with long half-life is desired, but the polypeptide preferably does not have undesirable effector function(s).
  • the Fc region-containing polypeptide may be an anti-tissue factor (TF) antibody; anti-IgE antibody; and anti-integrin antibody (e.g. an anti- ⁇ 4 ⁇ 7 antibody).
  • TF tissue factor
  • anti-IgE antibody anti-integrin antibody
  • the desired mechanism of action of such Fc region-containing polypeptides may be to block ligand-receptor binding pairs.
  • the Fc-region containing polypeptide with diminished ADCC activity may be an agonist antibody.
  • the polypeptide variant is administered by any suitable means, including parenteral, subcutaneous, intraperitoneal, intrapulmonary, and intranasal, and, if desired for local immunosuppressive treatment, intralesional administration.
  • Parenteral infusions include intramuscular, intravenous, intraarterial, intraperitoneal, or subcutaneous administration.
  • the polypeptide variant is suitably administered by pulse infusion, particularly with declining doses of the polypeptide variant.
  • the dosing is given by injections, most preferably intravenous or subcutaneous injections, depending in part on whether the administration is brief or chronic.
  • polypeptide variant For the prevention or treatment of disease, the appropriate dosage of polypeptide variant will depend on the type of disease to be treated, the severity and course of the disease, whether the polypeptide variant is administered for preventive or therapeutic purposes, previous therapy, the patient's clinical history and response to the polypeptide variant, and the discretion of the attending physician.
  • the polypeptide variant is suitably administered to the patient at one time or over a series of treatments.
  • polypeptide variant is an initial candidate dosage for administration to the patient, whether, for example, by one or more separate administrations, or by continuous infusion.
  • a typical daily dosage might range from about 1 ⁇ g/kg to 100 mg/kg or more, depending on the factors mentioned above.
  • the treatment is sustained until a desired suppression of disease symptoms occurs.
  • other dosage regimens may be useful. The progress of this therapy is easily monitored by conventional techniques and assays.
  • the polypeptide variant composition will be formulated, dosed, and administered in a fashion consistent with good medical practice. Factors for consideration in this context include the particular disorder being treated, the particular mammal being treated, the clinical condition of the individual patient, the cause of the disorder, the site of delivery of the agent, the method of administration, the scheduling of administration, and other factors known to medical practitioners.
  • the “therapeutically effective amount” of the polypeptide variant to be administered will be governed by such considerations, and is the minimum amount necessary to prevent, ameliorate, or treat a disease or disorder.
  • the polypeptide variant need not be, but is optionally formulated with one or more agents currently used to prevent or treat the disorder in question.
  • the effective amount of such other agents depends on the amount of polypeptide variant present in the formulation, the type of disorder or treatment, and other factors discussed above. These are generally used in the same dosages and with administration routes as used hereinbefore or about from 1 to 99% of the heretofore employed dosages.
  • This assay determines binding of an IgG Fc region to recombinant Fc ⁇ RIIA, Fc ⁇ RIIB and Fc ⁇ RIIIA ⁇ subunits expressed as His6-glutathione S transferase (GST)-tagged fusion proteins. Since the affinity of the Fc region of IgG1 for the Fc ⁇ RI is in the nanomolar range, the binding of IgG1 Fc variants can be measured by titrating monomeric IgG and measuring bound IgG with a polyclonal anti-IgG in a standard ELISA format (Example 2 below). The affinity of the other members of the Fc ⁇ R family, i.e. Fc ⁇ RIIA, Fc ⁇ RIIB and Fc ⁇ RIIIA for IgG is however in the micromolar range and binding of monomeric IgG1 for these receptors can not be reliably measured in an ELISA format.
  • the following assay utilizes Fc variants of recombinant anti-IgE E27 ( FIGS. 4A and 4B ) which, when mixed with human IgE at a 1:1 molar ratio, forms a stable hexamer consisting of three anti-IgE molecules and three IgE molecules.
  • a recombinant chimeric form of IgE (chimeric IgE) was engineered and consists of a human IgE Fc region and the Fab of an anti-VEGF antibody (Presta et al., Cancer Research 57:4593-4599 (1997)) which binds two VEGF molecules per mole of anti-VEGF.
  • VEGF vascular endothelial growth factor
  • hexamers When recombinant human VEGF is added at a 2:1 molar ratio to chimeric IgE:E27 hexamers, the hexamers are linked into larger molecular weight complexes via the chimeric IgE Fab:VEGF interaction.
  • the E27 component of this complex binds to the Fc ⁇ RIIA, Fc ⁇ RIIB and Fc ⁇ RIIIA ⁇ subunits with higher avidity to permit detection in an ELISA format.
  • Receptor Coat Fc ⁇ receptor ⁇ subunits were expressed as GST fusions of His6 tagged extracellular domains (ECDs) in 293 cells resulting in an ECD-6His-GST fusion protein (Graham et al., J. Gen. Virol. 36:59-74 (1977) and Gorman et al., DNA Prot. Eng. Tech. 2:3-10 (1990)) and purified by Ni-NTA column chromatography (Qiagen, Australia) and buffer exchanged into phosphate buffered saline (PBS). Concentrations were determined by absorption at 280nm using extinction coefficients derived by amino acid composition analysis. Receptors were coated onto Nunc F96 maxisorb plates (cat no.
  • E27:chimeric IgE:VEGF: (1:1:2 molar ratio) complexes are added to Fc ⁇ R ⁇ subunit coated plates at E27 concentrations of 5 ⁇ g and 1 ⁇ g total IgG in quadruplicate in assay buffer and incubated for 120 minutes at 25° C. on an orbital shaker.
  • HRP horse radish peroxidase conjugated goat anti-human IgG ( ⁇ ) heavy chain specific
  • Plates are washed 5 ⁇ with wash buffer to remove unbound HRP goat anti-human IgG and bound anti-IgG is detected by adding 100 ⁇ l of substrate solution (0.4 mg/ml o-phenylenedaimine dihydrochloride, Sigma P6912, 6 mM H 2 O 2 in PBS) and incubating for 8 min at 25° C. Enzymatic reaction is stopped by the addition of 100 ⁇ l 4.5N H 2 SO 4 and colorimetric product is measured at 490 nm on a 96 well plate densitometer (Molecular Devices). Binding of E27 variant complexes is expressed as a percent of the wild type E27 containing complex.
  • C2B8 Variants The chimeric light and heavy chains of anti-CD20 antibody C2B8 (Reff et al., Blood 83:435 (1994)) subcloned separately into previously described PRK vectors (Gorman et al., DNA Protein Eng. Tech. 2:3 (1990)) were used. By site directed mutagenesis (Kunkel et al., Proc. Natl. Acad. Sci. USA 82:488-492 (1985)), alanine scan variants of the Fc region in the heavy chain were constructed. The heavy and light chain plasmids were co-transfected into an adenovirus transformed human embryonic kidney cell line as previously described (Werther et al., J.
  • the media was changed to serum-free 24 hours after transfection and the secreted antibody was harvested after five days.
  • the antibodies were purified using Protein A-SEPHAROSE CL-4BTM (Pharmacia), buffer exchanged and concentrated to 0.5 ml with PBS using a Centricon-30 (Amicon), and stored at 4° C. The concentration of the antibody was determined using total Ig-binding ELISA.
  • C1q Binding ELISA Costar 96 well plates were coated overnight at 4° C. with the indicated concentrations of C2B8 in coating buffer (0.05 M sodium carbonate buffer), pH 9. The plates were then washed 3 ⁇ with PBS/0.05% TWEEN 20TM, pH 7.4 and blocked with 200 ⁇ l of ELISA diluent without thimerosal (0.1 M NaPO4/0.1 M NaCl/0.1% gelatin/0.05% TWEENTM 0.05% ProClin300) for 1 hr at room temperature.
  • the plate was washed 3 ⁇ with wash buffer, an aliquot of 100 ⁇ l of 2 ⁇ g/ml C1q (Quidel, San Diego, Calif.) was added to each well and incubated for 2 hrs at room temperature. The plate was then washed 6 ⁇ with wash buffer. 100 ⁇ l of a 1:1000 dilution of sheep anti-complement C1q peroxidase conjugated antibody (Biodesign) was added to each well and incubated for 1 hour at room temperature. The plate was again washed 6 ⁇ with wash buffer and 100 ⁇ l of substrate buffer (PBS/0.012% H 2 O 2 ) containing OPD (O-phenylenediamine dihydrochloride (Sigma)) was added to each well.
  • substrate buffer PBS/0.012% H 2 O 2
  • OPD O-phenylenediamine dihydrochloride
  • the oxidation reaction observed by the appearance of a yellow color, was allowed to proceed for 30 minutes and stopped by the addition of 100 ⁇ l of 4.5 N H 2 SO 4 .
  • the absorbance was then read at (492-405) nm using a microplate reader (SPECTRA MAX 250TM, Molecular Devices Corp.).
  • the appropriate controls were run in parallel (i.e. the ELISA was performed without C1q for each concentration of C2B8 used and also the ELISA was performed without C2B8).
  • C1q binding was measured by plotting the absorbance (492-405) nm versus concentration of C2B8 in ⁇ g/ml using a 4-parameter curve fitting program (KALEIDAGRAPHTM) and comparing EC 50 values.
  • CDC Complement Dependent Cytotoxicity Assay. This assay was performed essentially as previously described (Gazzano-Santoro et al., J. Immunol. Methods 202:163-171 (1997)). Various concentrations of C2B8 (0.08-20 ⁇ g/ml) were diluted with RHB buffer (RPMI 1640/20 mM HEPES (pH 7.2)/2 mM Glutamine/0.1% BSA/100 ⁇ g/ml Gentamicin).
  • RHB buffer RPMI 1640/20 mM HEPES (pH 7.2)/2 mM Glutamine/0.1% BSA/100 ⁇ g/ml Gentamicin.
  • Human complement (Quidel) was diluted 1:3 in RHB buffer and WIL2-S cells (available from the ATCC, Manassas, Va.) which express the CD20 antigen were diluted to a density of 1 ⁇ 10 6 cells /ml with RHB buffer.
  • Mixtures of 150 ⁇ l containing equal volumes of C2B8, diluted human complement and WIL2-S cells were added to a flat bottom tissue culture 96 well plate and allowed to incubate for 2 hrs at 37° C. and 5% CO 2 to facilitate complement mediated cell lysis. 50 ⁇ l of alamar blue (Accumed International) was then added to each well and incubated overnight at 37° C.
  • the absorbance was measured using a 96-well fluorometer with excitation at 530 nm and emission at 590 nm. As described by Gazzano-Santoro et al., the results are expressed in relative fluorescence units (RFU). The sample concentrations were computed from a C2B8 standard curve and the percent activity as compared to wild type C2B8 is reported for each variant.
  • CD20 Binding Potency of the C2B8 Variants The binding of C2B8 and variants to the CD20 antigen were assessed by a method previously described (Reff et al., (1994), supra; reviewed in Gazzano-Santoro et al., (1997), supra). WIL2-S cells were grown for 3-4 days to a cell density of 1 ⁇ 10 6 cells/ml. The cells were washed and spun twice in FACS buffer (PBS/0.1% BSA/0.02% NaN 3 ) and resuspended to a cell density of 5 ⁇ 10 6 cells/ml.
  • FACS buffer PBS/0.1% BSA/0.02% NaN 3
  • Fc ⁇ R Binding ELISAs Fc ⁇ RI ⁇ subunit-GST fusion was coated onto Nunc F96 maxisorb plates (cat no. 439454) by adding 100 ⁇ l of receptor-GST fusion at 1 ⁇ g/ml in PBS and incubated for 48 hours at 4° C. Prior to assay, plates are washed 3 ⁇ with 250 ⁇ l of wash buffer (PBS pH 7.4 containing 0.5% TWEEN 20TM) and blocked with 250 ⁇ l of assay buffer (50 mM Tris buffered saline, 0.05% TWEEN 20TM, 0.5% RIA grade bovine albumin (Sigma A7888), and 2mM EDTA pH 7.4).
  • wash buffer PBS pH 7.4 containing 0.5% TWEEN 20TM
  • assay buffer 50 mM Tris buffered saline, 0.05% TWEEN 20TM, 0.5% RIA grade bovine albumin (Sigma A7888), and 2mM EDTA pH 7.4
  • Plates are washed 5 ⁇ with wash buffer to remove unbound HRP goat anti-human IgG and bound anti-IgG is detected by adding 100 ⁇ l of substrate solution (0.4 mg/ml o-phenylenedaimine dihydrochloride, Sigma P6912, 6 mM H 2 O 2 in PBS) and incubating for 8 min at 25° C. Enzymatic reaction is stopped by the addition of 100 ⁇ l 4.5N H 2 SO 4 and colorimetric product is measured at 490 nm on a 96 well plate densitometer (Molecular Devices). Binding of variant is expressed as a percent of the wild type molecule.
  • Fc ⁇ RII and III binding ELISAs were performed as described in Example 1 above.
  • ELISA plates were coated with 2 ⁇ g/ml streptavidin (Zymed, South San Francisco) in 50 mM carbonate buffer, pH 9.6, at 4° C. overnight and blocked with PBS-0.5% BSA, pH 7.2 at room temperature for one hour.
  • Biotinylated FcRn prepared using biotin-X-NHS from Research Organics, Cleveland, Ohio and used at 1-2 ⁇ g/ml
  • PBS-0.5% BSA 0.05% polysorbate 20, pH 7.2
  • the IgG2 variant appears to have a much lower affinity for C1q than the E318A and K320A variants ( FIG. 2 ).
  • the results demonstrate that E318 and K320 do not constitute the core C1q binding sites for human IgG1.
  • the K322A substitution had a significant effect on both complement activity and C1q binding.
  • the K322A variant did not have CDC activity when tested in the above CDC assay and was more than a 100 fold lower than wild type C2B8 in binding to C1q ( FIG. 2 ).
  • K322 is the only residue of the proposed core C1q binding sites that appeared to have a significant effect on complement activation and C1q binding.
  • Variants constructed, K274A, N276A, Y278A, S324A, P329A, P331 A, K334A, and T335A were assessed for their ability to bind C1q and also to activate complement. Many of these substitutions had little or no effect on C1q binding or complement activation.
  • the P329A and the P331A variants did not activate complement and had decreased binding to C1q.
  • the mutation P329A results in an antibody that does not activate complement and is more than a 100 fold lower in binding to C1q ( FIG. 3 ) when compared to wild type C2B8 ( FIG. 2 ).
  • Variants that did not bind to C1q and hence did not activate complement were examined for their ability to bind to the Fc receptors: Fc ⁇ RI, Fc ⁇ RIIA, Fc ⁇ RIIB, Fc ⁇ RIIIA and FcRn.
  • This particular study was performed using a humanized anti-IgE antibody, an IgG1 antibody with these mutations (see Example 1 above).
  • a further residue involved in binding human C1q was identified using the methods described in the present example.
  • the residue D270 was replaced with lysine and valine to generate variants D270K and D270V, respectively. These variants both showed decreased binding to human C1q ( FIG. 6 ) and were non-lytic ( FIG. 7 ).
  • the two variants bound the CD20 antigen normally and recruited ADCC.
  • K326, A327, E333 and K334 are potential sites for improving the efficacy of antibodies by way of the CDC pathway.
  • the aim of this study was to improve CDC activity of an antibody by increasing binding to C1q.
  • K326M, K326D, K326E and E333S were constructed with at least a two-fold increase in binding to C1q when compared to wild type.
  • Variant K326W displayed about a five-fold increase in binding to C1q.
  • Variants of the wild type C2B8 antibody were prepared as described above in Example 2.
  • the C1q binding ELISA, CDC assay, and CD20 binding potency assay in this example were performed as described in Example 2 above.
  • K326A, K326D, K326E, K326G, K326V, K326M and K326W were all bound to C1q with a better affinity than the wild type antibody.
  • K326W, K326M, K326D and K326E showed at least a two-fold increase in binding to C1q (Table 5).
  • K326W had the best affinity for C1q.
  • E333 was also substituted with other amino acid residues.
  • E333S, E333G, E333V, E333D, and E333Q all had increased binding to C1q when compared to the wild type ( FIG. 11 ).
  • the order of binding affinity for C1q was as follows: E333S>E333A>E333G>E333V>E333D>E333Q.
  • Substitutions with amino acid residues with small side chain volumes, i.e. serine, alanine and glycine resulted in variants with higher affinity for C1q in comparison to the other variants, E333V, E333D and E333Q, with larger side chain volumes.
  • the variant E333S had the highest affinity for C1q, showing a two-fold increase in binding when compared to the wild type. Without being bound to any one theory, this indicates the effect on C1q binding at 333 may also be due in part to the polarity of the residue.
  • Double variants were also generated. As shown in FIGS. 12 and 13 , double variants K326M-E333S and K326A-E333A were at least three-fold better at binding human C1q than wild type C2B8 ( FIG. 12 ) and at least two-fold better at mediating CDC compared to wild type C2B8 ( FIG. 13 ). Additivity indicates these are independently acting variants.
  • IgG1 Variants Recombinant anti-IgE E27 having the light chain and heavy chain sequences in FIGS. 4A and 4B , respectively, was used as the parent antibody in the following experiments.
  • This antibody binds the antigen IgE and has a non-A allotype IgG1 Fc region.
  • site directed mutagenesis By site directed mutagenesis (Kunkel et al., Proc. Natl. Acad. Sci. USA 82:488-492 (1985)), variants of the Fc region in the heavy chain of the above parent antibody were constructed.
  • the heavy and light chain plasmids were co-transfected into an adenovirus transformed human embryonic kidney cell line as previously described (Werther et al., J. Immunol.
  • the media was changed to serum-free 24 hours after transfection and the secreted antibody was harvested after five days.
  • the antibodies were purified by Protein G SEPHAROSE® (Pharmacia), buffer exchanged and concentrated to 0.5 ml with PBS using a Centricon-30 (Amicon), and stored at 4° C. Concentration was determined by adsorption at 280 nm using extinction coefficients derived by amino acid composition analysis.
  • Fc ⁇ RIA was expressed as a GST fusion of His6 tagged extracellular domain in 293 cells and purified by Ni-NTA column chromatography.
  • Purified receptors were coated onto Nunc F96 maxisorb plates (cat no. 439545) at approximately 150 ng per well by adding 100 ⁇ L of receptor at 1.5 ⁇ g/mL in PBS and incubated for 24 hours at 4° C. Prior to assay, plates were washed 3 ⁇ with 250 ⁇ L of wash buffer (phosphate buffered saline pH 7.4 containing 0.5% TWEEN 20®) and blocked with 250 ⁇ L of assay buffer (50 mM tris buffered saline, 0.05% TWEEN 20®, 0.5% RIA grade bovine albumin (Sigma A7888), and 2mM EDTA pH 7.4).
  • wash buffer phosphate buffered saline pH 7.4 containing 0.5% TWEEN 20®
  • assay buffer 50 mM tris buffered saline, 0.05% TWEEN 20®, 0.5% RIA grade bovine albumin (Sigma A7888), and 2mM EDTA pH 7.4
  • E27 100 ⁇ L of E27 was added to the first four wells of the Fc ⁇ RIA subunit coated plated at a concentration of 10 ⁇ g/mL. 80 ⁇ L of assay buffer was added to the next four well followed by 20 ⁇ L of the 10 ⁇ g/mL E27 IgG to give a final concentration of 2 ⁇ g/mL. Plates were incubated at 25° C. for 2 hours on an orbital shaker.
  • HRP horse radish peroxidase conjugated protein G
  • BIORAD horse radish peroxidase conjugated protein G
  • HRP conjugates were incubated for 1.5 hours at 25° C. on an orbital shaker. Plates were washed ⁇ 5 with wash buffer to remove unbound HRP conjugate. Binding was detected by adding 100 ⁇ L of substrate solution (0.4 mg/mL o-phenylenedaimine dihydrochloride, Sigma P6912, 6 mM H 2 O 2 in PBS) and incubating for 10 minutes at 25° C. Enzymatic reaction was stopped by the addition of 100 ⁇ L of 4.5 N H 2 SO 4 and colorimetric product was measured at 490 nm on a 96 well plate densitometer (Molecular Devices).
  • Binding of E27 variants at IgG concentration of 2 ⁇ g/mL was expressed as a ratio of wild type E27.
  • Fc ⁇ RIA THP-1 Assay 100 ⁇ L of E27 was added to the first three wells of a serocluster plate (Costar) at a concentration of 20 ⁇ g/mL in assay buffer (1 ⁇ PBS, 0.1% BSA, 0.01% NaN 3 ). 92.5 of assay buffer was added to the next three wells followed by 7.5 ⁇ L of the 20 ⁇ g/mL E27 IgG to give a final concentration of 1.5 ⁇ g/mL. To each well, 100 ⁇ L of THP-1 cells were added at a concentration of 5 million cells/mL in FACS assay buffer. The plate is incubated on ice for 30 minutes
  • IgG binding Fc ⁇ RIA was detected by adding 100 ⁇ L FITC conjugated F(ab′) 2 fragment of goat anti-human IgG heavy chain specific. (Jackson Immunoresearch) at 1:200. FITC conjugates were incubated with cells for 30 minutes on ice. Cells were washed ⁇ 3 with assay buffer to remove unbound FITC conjugate. Cells were stained with P.I. (SIGMA) at 2.5 ⁇ g/mL and analyzed by flow cytometry.
  • SIGMA P.I.
  • Binding of E27 variants at IgG concentration of 1.5 ⁇ g/mL was expressed as a ratio of wild type E27.
  • Fc ⁇ RIIA, Fc ⁇ RIIB and Fc ⁇ RIIIA binding ELISAs were performed as described in Example 1 above, with detection of the stable hexamer (consisting of three anti-IgE molecules and three IgE molecules).
  • FcRn Binding ELISA For measuring FcRn binding activity of IgG variants, ELISA plates were coated with 2 ⁇ g/ml streptavidin (Zymed, South San Francisco) in 50 mM carbonate buffer, pH 9.6, at 4° C. overnight and blocked with PBS-0.5% BSA, pH 7.2 at room temperature for one hour. Biotinylated FcRn (prepared using biotin-X-NHS from Research Organics, Cleveland, Ohio and used at 1-2 ⁇ g/ml) in PBS-0.5% BSA, 0.05% polysorbate 20, pH 7.2, was added to the plate and incubated for one hour.
  • IgG standard 1.6-100 ng/ml
  • PBS-0.5% BSA, 0.05% polysorbate 20, pH 6.0 Two fold serial dilutions of IgG standard (1.6-100 ng/ml) or variants in PBS-0.5% BSA, 0.05% polysorbate 20, pH 6.0, were added to the plate and incubated for two hours. Bound IgG was detected using peroxidase labeled goat F(ab') 2 anti-human IgG F(ab′) 2 in the above pH 6.0 buffer (Jackson ImmunoResearch, West Grove, Pa.) followed by 3,3′,5,5′-tetramethyl benzidine (Kirgaard & Perry Laboratories) as the substrate. Plates were washed between steps with PBS-0.05% TWEEN 20® at either pH 7.2 or 6.0.
  • chromium 51-labeled target cells tumor cell lines were grown in tissue culture plates and harvested using sterile 10 mM EDTA in PBS. SK-BR-3 cells, a 3 + HER2-overexpressing human breast cancer cell line, were used as targets in all assays. The detached cells were washed twice with cell culture medium. Cells (5 ⁇ 10 6 ) were labeled with 200 ⁇ Ci of chromium51 (New England Nuclear/DuPont) at 37° C. for one hour with occasional mixing. Labeled cells were washed three times with cell culture medium, then were resuspended to a concentration of 1 ⁇ 10 5 cells/mL.
  • Cells were used either without opsonization, or were opsonized prior to the assay by incubation with rhuMAb HER2 wildtype (HERCEPTIN®) or seven Fc mutants (G14, G18, G17, G36, G30, G31 and G34) at 100 ng/mL and 1.25 ng/mL in PBMC assay or 20 ng/mL and 1 ng/mL in NK assay.
  • rhuMAb HER2 wildtype HERCEPTIN®
  • Fc mutants G14, G18, G17, G36, G30, G31 and G34
  • Peripheral blood mononuclear cells were prepared by collecting blood on heparin from normal healthy donors and dilution with an equal volume of phosphate buffered saline (PBS). The blood was then layered over LYMPHOCYTE SEPARATION MEDIUM® (LSM: Organon Teknika) and centrifuged according to the manufacturer's instructions. Mononuclear cells were collected from the LSM-plasma interface and were washed three times with PBS. Effector cells were suspended in cell culture medium to a final concentration of 1 ⁇ 10 7 cells/mL.
  • PBS phosphate buffered saline
  • NK cells natural killer cells were isolated from PBMCs by negative selection using an NK cell isolation kit and a magnetic column (Miltenyi Biotech) according to the manufacturer's instructions. Isolated NK cells were collected, washed and resuspended in cell culture medium to a concentration of 2 ⁇ 10 6 cells/mL. The identity of the NK cells was confirmed by flow cytometric analysis.
  • Varying effector:target ratios were prepared by serially diluting the effector (either PBMC or NK) cells two-fold along the rows of a microtiter plate (100 ⁇ L final volume) in cell culture medium.
  • the concentration of effector cells ranged from 1.0 ⁇ 10 7 /mL to 2.0 ⁇ 10 4 /mL for PBMC and from 2.0 ⁇ 10 6 /mL to 3.9 ⁇ 10 3 /mL for NK.
  • 100 ⁇ L of chromium 51-labeled target cells (opsonized or nonoponsonized) at 1 ⁇ 10 5 cells/mL were added to each well of the plate.
  • a variety of antibody variants were generated which had FcR binding activity that differed from the parent antibody.
  • the FcR binding data for the variants generated is shown in Tables 6 and 7 below.
  • Variants with increased binding to FcRn generally had binding values ⁇ 1.30 as determined in this Example and those with reduced binding to FcRn generally had binding values ⁇ 0.70 as determined in this Example.
  • various non-alanine substitution variants were made, and the FcR binding activity of those variants is summarized in the following table.
  • This study includes a complete mapping of human IgG1 for human Fc ⁇ RI, Fc ⁇ RIIA, Fc ⁇ RIIB, Fc ⁇ RIIIA, and FcRn.
  • Fc ⁇ RI and FcRn are high affinity receptors and monomeric IgG could be evaluated in the assays for these two receptors.
  • Fc ⁇ RIIA, Fc ⁇ RIIB and Fc ⁇ RIIIA are low affinity receptors and required use of an immune complex.
  • an ELISA-type assay was used for Fc ⁇ RIIA, Fc ⁇ RIIB, and Fc ⁇ RIIIA, in which pre-formed hexamers, consisting of three anti-IgE E27 and three IgE molecules were bound to the Fc ⁇ R and either anti-human IgG Fc-HRP or protein G-HRP used as detection reagent.
  • these hexamers could be linked into multimers by addition of human VEGF (using anti-VEGF IgE).
  • the hexameric complexes were used since these provided sufficient binding and required less IgG. Complexes formed using other antibody:antigen combinations are also possible reagents, as long as the antigen contains at least two identical binding sites per molecule for the antibody.
  • VEGF contains two binding sites per VEGF dimer for anti-VEGF A.4.6.1 (Kim et al., Growth Factors 7:53 (1992) and Kim et al., Nature 362:841 (1993)). VEGF:anti-VEGF multimers also bound to the low affinity Fc ⁇ RIIA and Fc ⁇ RIIIA ( FIGS. 16A and 16B ).
  • alanine variants were found. Some variants exhibited reduced binding to all Fc ⁇ R (G14, FIG. 17 ), while other variants showed reduced binding only to one Fc ⁇ R (G36, FIG. 17 ), improved binding only to one Fc ⁇ R (G15, G54, G55, FIG. 17 ), or simultaneous reduction to one Fc ⁇ R with improvement to another (G16, FIG. 17 ).
  • One application of these variants is to improve the ADCC effector function of an antibody. This can be achieved by modifying Fc region amino acids at one or more residues which would lead to improved binding to Fc ⁇ RIIIA. Improved Fc ⁇ RIIIA binding would lead to improved binding by NK cells, which carry only Fc ⁇ RIIIA and can mediate ADCC.
  • Selected alanine variants which were either reduced in binding to Fc ⁇ RIIIA (variants 17, 18, 34; Table 6), had no effect on Fc ⁇ RIIIA binding (variant 31; Table 6), or had improved binding to Fc ⁇ RIIIA (variants 30, 36; Table 6) were tested in an in vitro ADCC assay using human PBMCs as effector cells.
  • the IgG Fc variants used in this assay were generated by substituting the V H N L domains of anti-IgE E27 with those from anti-HER2 antibody; HERCEPTIN® (humAb4D5-8 in Table 1 of Carter et al., PNAS ( USA ) 89:4285-4289 (1992)).
  • the pattern of ADCC exhibited by the variants correlated well with the pattern of binding to Fc ⁇ RIIIA ( FIGS. 20 and 21 ).
  • variant 36 S298(317)A also showed improvement in ADCC compared to wildtype HERCEPTIN® at 1.25 ng/ml ( FIG. 21 ).
  • allelic variants of several of the human Fc ⁇ R have been found in the human population. These allelic variant forms have been shown to exhibit differences in binding of human and murine IgG and a number of association studies have correlated clinical outcomes with the presence of specific allelic forms (reviewed in Lehrnbecher et al., Blood 94(12):4220-4232 (1999)). Several studies have investigated two forms of Fc ⁇ RIIA, R131 and H131, and their association with clinical outcomes (Hatta et al., Genes and Immunity 1:53-60 (1999); Yap et al., Lupus 8:305-310 (1999); and Lorenz et al. European J. Immunogenetics 22:397-401 (1995)).
  • the pattern of binding of the selected IgG1 variants to the relatively higher affinity Fc ⁇ RIIIA-V158 was the same as for the relatively lower affinity Fc ⁇ RIIIA-F158 (the F158 form was used in assaying all variants).
  • IgG1 variants which showed improved binding to the Fc ⁇ RIIIA-F158 form also showed improved binding to the Fc ⁇ RIIIA-V158 form though the improvement was not as pronounced.
  • Fc ⁇ RIIA-R131 used in assaying all variants
  • Fc ⁇ RIIA-H131 the binding pattern of the selected IgG1 variants did show some distinct differences.
  • S267(280)A, H268(281)A, and S267(280)A/H268(281)A exhibited improved binding to Fc ⁇ RIIA-R131, compared to native IgG1, but not to Fc ⁇ RIIA-H131.
  • S267(280)G showed improved binding to Fc ⁇ RIIA-R131 but reduced binding to Fc ⁇ RIIA-H131 (Table 10).

Abstract

The present invention concerns polypeptides comprising a variant Fc region. More particularly, the present invention concerns Fc region-containing polypeptides that have altered effector function as a consequence of one or more amino acid modifications in the Fc region thereof.

Description

  • This is a continuation application claiming priority to divisional application Ser. No. 11/158,839 filed Jun. 22, 2005, which claims priority to divisional application Ser. No. 10/757,863 filed Jan. 15, 2004, which claims priority to non-provisional application Ser. No. 09/483,588 filed Jan. 14, 2000 (now U.S. Pat. No. 6,737,056), which claims priority under 35 USC §119 to provisional application No. 60/116,023 filed Jan. 15, 1999, the entire disclosures of which are incorporated herein by reference.
  • BACKGROUND OF THE INVENTION
  • 1. Field of the Invention
  • The present invention concerns polypeptides comprising a variant Fc region. More particularly, the present invention concerns Fc region-containing polypeptides that have altered effector function as a consequence of one or more amino acid modifications in the Fc region thereof.
  • 2. Description of Related Art
  • Antibodies are proteins which exhibit binding specificity to a specific antigen. Native antibodies are usually heterotetrameric glycoproteins of about 150,000 daltons, composed of two identical light (L) chains and two identical heavy (H) chains. Each light chain is linked to a heavy chain by one covalent disulfide bond, while the number of disulfide linkages varies between the heavy chains of different immunoglobulin isotypes. Each heavy and light chain also has regularly spaced intrachain disulfide bridges. Each heavy chain has at one end a variable domain (VH) followed by a number of constant domains. Each light chain has a variable domain at one end (VL) and a constant domain at its other end; the constant domain of the light chain is aligned with the first constant domain of the heavy chain, and the light chain variable domain is aligned with the variable domain of the heavy chain. Particular amino acid residues are believed to form an interlace between the light and heavy chain variable domains.
  • The term “variable” refers to the fact that certain portions of the variable domains differ extensively in sequence among antibodies and are responsible for the binding specificity of each particular antibody for its particular antigen. However, the variability is not evenly distributed through the variable domains of antibodies. It is concentrated in three segments called complementarity determining regions (CDRs) both in the light chain and the heavy chain variable domains. The more highly conserved portions of the variable domains are called the framework regions (FRs). The variable domains of native heavy and light chains each comprise four FRs, largely adopting a β-sheet configuration, connected by three CDRs, which form loops connecting, and in some cases forming part of, the β-sheet structure. The CDRs in each chain are held together in close proximity by the FRs and, with the CDRs from the other chain, contribute to the formation of the antigen binding site of antibodies (see Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md. (1991)).
  • The constant domains are not involved directly in binding an antibody to an antigen, but exhibit various effector functions. Depending on the amino acid sequence of the constant region of their heavy chains, antibodies or immunoglobulins can be assigned to different classes. There are five major classes of immunoglobulins: IgA, IgD, IgE, IgG and IgM, and several of these may be further divided into subclasses (isotypes), e.g. IgG1, IgG2, IgG3, and IgG4; IgA1 and IgA2. The heavy chain constant regions that correspond to the different classes of immunoglobulins are called α, δ, ε, γ, and μ, respectively. Of the various human immunoglobulin classes, only human IgG1, IgG2, IgG3 and IgM are known to activate complement; and human IgG1 and IgG3 mediate ADCC more effectively than IgG2 and IgG4.
  • A schematic representation of the native IgG1 structure is shown in FIG. 1, where the various portions of the native antibody molecule are indicated. Papain digestion of antibodies produces two identical antigen binding fragments, called Fab fragments, each with a single antigen binding site, and a residual “Fc” fragment, whose name reflects its ability to crystallize readily. The crystal structure of the human IgG Fc region has been determined (Deisenhofer, Biochemistiy 20:2361-2370 (1981)). In human IgG molecules, the Fc region is generated by papain cleavage N-terminal to Cys 226. The Fc region is central to the effector functions of antibodies.
  • The effector functions mediated by the antibody Fc region can be divided into two categories: (1) effector functions that operate after the binding of antibody to an antigen (these functions involve the participation of the complement cascade or Fc receptor (FcR)-bearing cells); and (2) effector functions that operate independently of antigen binding (these functions confer persistence in the circulation and the ability to be transferred across cellular barriers by transcytosis). Ward and Ghetie, Therapeutic Immunology2:77-94 (1995).
  • While binding of an antibody to the requisite antigen has a neutralizing effect that might prevent the binding of a foreign antigen to its endogenous target (e.g. receptor or ligand), binding alone may not remove the foreign antigen. To be efficient in removing and/or destructing foreign antigens, an antibody should be endowed with both high affinity binding to its antigen, and efficient effector functions.
  • Fc Receptor (FcR) Binding
  • The interaction of antibodies and antibody-antigen complexes with cells of the immune system effects a variety of responses, including antibody-dependent cell-mediated cytotoxicity (ADCC) and complement dependent cytotoxicity (CDC) (reviewed in Daëron, Annu. Rev. lmmunol. 15:203-234 (1997); Ward and Ghetie, Therapeutic lmmunol. 2:77-94 (1995); as well as Ravetch and Kinet, Annu. Rev. lmmunol. 9:457-492 (1991)).
  • Several antibody effector functions are mediated by Fc receptors (FcRs), which bind the Fc region of an antibody. FcRs are defined by their specificity for immunoglobulin isotypes; Fc receptors for IgG antibodies are referred to as FcγR, for IgE as FcER, for IgA as FcaR and so on. Three subclasses of FcγR have been identified: FcγRI (CD64), FcγRII (CD32) and FcγRIII (CD16). Because each FcγR subclass is encoded by two or three genes, and alternative RNA spicing leads to multiple transcripts, a broad diversity in FcγR isoforms exists. The three genes encoding the FcγRI subclass (FcγRIA, FcγRIB and FcγRIC) are clustered in region 1q21.1 of the long arm of chromosome 1; the genes encoding FcγRII isoforms (FcγRIIA, FcγRIIB and FcγRIIC) and the two genes encoding FcγRIII (FcγRIIIA and FcγRIIIB) are all clustered in region 1q22. These different FcR subtypes are expressed on different cell types (reviewed in Ravetch and Kinet, Annu. Rev. Immunol. 9:457-492 (1991)). For example, in humans, FcγRIIIB is found only on neutrophils, whereas FcγRIIIA is found on macrophages, monocytes, natural killer (NK) cells, and a subpopulation of T-cells. Notably, FcγRIIIA is the only FcR present on NK cells, one of the cell types implicated in ADCC.
  • FcγRI, FcγRII and FcγRIII are immunoglobulin superfamily (IgSF) receptors; FcγRI has three IgSF domains in its extracellular domain, while FcγRII and FcγRIII have only two IgSF domains in their extracellular domains.
  • Another type of Fc receptor is the neonatal Fc receptor (FcRn). FcRn is structurally similar to major histocompatibility complex (MHC) and consists of an a-chain noncovalently bound to 2-microglobulin.
  • The binding site on human and murine antibodies for FcγR have been previously mapped to the so-called “lower hinge region” consisting of residues 233-239 (EU index numbering as in Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md. (1991)). Woof et al., Molec. Immunol. 23:319-330 (1986); Duncan et al., Nature 332:563 (1988); Canfield and Morrison, J. Exp. Med. 173:1483-1491 (1991); Chappel et al., Proc. Natl. Acad. Sci USA 88:9036-9040 (1991). Of residues 233-239, P238 and S239 have been cited as possibly being involved in binding, but these two residues have never been evaluated by substitution or deletion.
  • Other previously cited areas possibly involved in binding to FcγR are: G316-K338 (human IgG) for human FcγRI (by sequence comparison only; no substitution mutants were evaluated) (Woof et al. Molec. Immunol. 23:319-330 (1986)); K274-R301 (human IgG1) for human FcγRIII (based on peptides) (Sarmay et al. Molec. Immunol. 21:43-51 (1984)); Y407-R416 (human IgG) for human FcγRIII (based on peptides) (Gergely et al., Biochem. Soc. Trans. 12:739-743 (1984)); as well as N297 and E318 (murine IgG2b) for murine FcγRII (Lund et al., Molec. Immunol., 29:53-59 (1992)).
  • Pro331 in IgG3 was changed to Ser, and the affinity of this variant to target cells analyzed. The affinity was found to be six-fold lower than that of unmutated IgG3, indicating the involvement of Pro331 in FcγRI binding. Morrison et al., Immunologist, 2:119-124 (1994); and Canfield and Morrison, J. Exp. Med. 173:1483-91 (1991).
  • C1q Binding
  • C1q and two serine proteases, C1r and C1s, form the complex C1, the first component of the complement dependent cytotoxicity (CDC) pathway. C1q is a hexavalent molecule with a molecular weight of approximately 460,000 and a structure likened to a bouquet of tulips in which six collagenous “stalks” are connected to six globular head regions. Burton and Woof, Advances in Immunol. 51:1-84 (1992). To activate the complement cascade, it is necessary for C1q to bind to at least two molecules of IgG1, IgG2, or IgG3 (the consensus is that IgG4 does not activate complement), but only one molecule of IgM, attached to the antigenic target. Ward and Ghetie, Therapeutic Immunology 2:77-94 (1995) at page 80.
  • Based upon the results of chemical modifications and crystallographic studies, Burton et al. (Nature, 288:338-344 (1980)) proposed that the binding site for the complement subcomponent C1q on IgG involves the last two (C-terminal) β-strands of the CH2 domain. Burton later suggested (Molec. Immunol., 22(3):161-206 (1985)) that the region comprising amino acid residues 318 to 337 might be involved in complement fixation.
  • Duncan and Winter (Nature 332:738-40 (1988)), using site directed mutagenesis, reported that Glu318, Lys320 and Lys322 form the binding site to C1q. The data of Duncan and Winter were generated by testing the binding of a mouse IgG2b isotype to guinea pig C1q. The role of Glu318, Lys320 and Lys322 residues in the binding of C1q was confirmed by the ability of a short synthetic peptide containing these residues to inhibit complement mediated lysis. Similar results are disclosed in U.S. Pat. No. 5,648,260 issued on Jul. 15, 1997, and U.S. Pat. No. 5,624,821 issued on Apr. 29, 1997.
  • The residue Pro331 has been implicated in C1q binding by analysis of the ability of human IgG subclasses to carry out complement mediated cell lysis. Mutation of Ser331 to Pro331 in IgG4 conferred the ability to activate complement. (Tao et al., J. Exp. Med., 178:661-667 (1993); Brekke et al., Eur. J. Immunol., 24:2542-47 (1994)).
  • From the comparison of the data of the Winter group, and the Tao et al., and Brekke et al., papers, Ward and Ghetie concluded in their review article that there are at least two different regions involved in the binding of C1q: one on the β-strand of the CH2 domain bearing the Glu318, Lys320 and Lys322 residues, and the other on a turn located in close proximity to the same β-strand, and containing a key amino acid residue at position 331.
  • Other reports suggested that human IgG1 residues Leu235, and Gly237, located in the lower hinge region, play a critical role in complement fixation and activation. Xu et al., J. Immunol. 150:152A (Abstract) (1993). WO94/29351 published Dec. 22, 1994 reports that amino acid residues necessary for C1q and FcR binding of human IgG1 are located in the N-terminal region of the CH2 domain, i.e. residues 231 to 238.
  • It has further been proposed that the ability of IgG to bind C1q and activate the complement cascade also depends on the presence, absence, or modification of the carbohydrate moiety positioned between the two CH2 domains (which is normally anchored at Asn297). Ward and Ghetie, Therapeutic Immunology 2:77-94 (1995) at page 81.
  • SUMMARY OF THE INVENTION
  • The present invention provides a variant of a parent polypeptide comprising an Fc region, which variant mediates antibody-dependent cell-mediated cytotoxicity (ADCC) in the presence of human effector cells more effectively, or binds an Fc gamma receptor (FcγR) with better affinity, than the parent polypeptide and comprises at least one amino acid modification in the Fc region. The polypeptide variant may, for example, comprise an antibody or an immunoadhesin. The Fc region of the parent polypeptide preferably comprises a human Fc region; e.g., a human IgG1, IgG2, IgG3 or IgG4 Fc region. The polypeptide variant preferably comprises an amino acid modification (e.g. a substitution) at any one or more of amino acid positions 256, 290, 298, 312, 326, 330, 333, 334, 360, 378 or 430 of the Fc region, wherein the numbering of the residues in the Fc region is that of the EU index as in Kabat.
  • In addition, the invention provides a polypeptide comprising a variant Fc region with altered Fc gamma receptor (FcγR) binding affinity, which polypeptide comprises an amino acid modification at any one or more of amino acid positions 238, 239, 248, 249, 252, 254, 255, 256, 258, 265, 267, 268, 269, 270, 272, 276, 278, 280, 283, 285, 286, 289, 290, 292, 293, 294, 295, 296, 298, 301, 303, 305, 307, 309, 312, 315, 320, 322, 324, 326, 327, 329, 330, 331, 333, 334, 335, 337, 338, 340, 360, 373, 376, 378, 382, 388, 389, 398, 414, 416, 419, 430, 434, 435, 437, 438 or 439 of the Fc region, wherein the numbering of the residues in the Fc region is that of the EU index as in Kabat. The variant Fc region preferably comprises a variant human IgG Fc region, e.g., a variant human IgG1, IgG2, IgG3 or IgG4 Fc region. In this respect, it is noted that, in the work in the above-cited art where the parent polypeptide had a non-human murine Fc region, different residues from those identified herein were thought to impact FcR binding. For example, in the murine IgG2b/murine FcγRII system, IgG E318 was found to be important for binding (Lund et al., Molec. Immunol. 27(1):53-59 (1992)), whereas E318A had no effect in the human IgG/human FcγRII system (Table 6 below).
  • In one embodiment, the polypeptide variant with altered FcγR binding activity displays reduced binding to an FcγR and comprises an amino acid modification at any one or more of amino acid positions 238, 239, 248, 249, 252, 254, 265, 268, 269, 270, 272, 278, 289, 292, 293, 294, 295, 296, 298, 301, 303, 322, 324, 327, 329, 333, 335, 338, 340, 373, 376, 382, 388, 389, 414, 416, 419, 434, 435, 437, 438 or 439 of the Fc region, wherein the numbering of the residues in the Fc region is that of the EU index as in Kabat.
  • For example, the polypeptide variant may display reduced binding to an FcγRI and comprise an amino acid modification at any one or more of amino acid positions 238, 265, 269, 270, 327 or 329 of the Fc region, wherein the numbering of the residues in the Fc region is that of the EU index as in Kabat.
  • The polypeptide variant may display reduced binding to an FcγRII and comprise an amino acid modification at any one or more of amino acid positions 238, 265, 269, 270, 292, 294, 295, 298, 303, 324, 327, 329, 333, 335, 338, 373, 376, 414, 416, 419, 435, 438 or 439 of the Fc region, wherein the numbering of the residues in the Fc region is that of the EU index as in Kabat.
  • The polypeptide variant of interest may display reduced binding to an FcγRIII and comprise an amino acid modification at one or more of amino acid positions 238, 239, 248, 249, 252, 254, 265, 268, 269, 270, 272, 278, 289, 293, 294, 295, 296, 301, 303, 322, 327, 329, 338, 340, 373, 376, 382, 388, 389, 416, 434, 435 or 437 of the Fc region, wherein the numbering of the residues in the Fc region is that of the EU index as in Kabat.
  • In another embodiment, the polypeptide variant with altered FcγR binding affinity displays improved binding to the FcγR and comprises an amino acid modification at any one or more of amino acid positions 255, 256, 258, 267, 268, 272, 276, 280, 283, 285, 286, 290, 298, 301, 305, 307, 309, 312, 315, 320, 322, 326, 330, 331, 333, 334, 337, 340, 360, 378, 398 or 430 of the Fc region, wherein the numbering of the residues in the Fc region is that of the EU index as in Kabat.
  • For example, the polypeptide variant may display increased binding to an FcγRIII and, optionally, may further display decreased binding to an FcγRII. An exemplary such variant comprises amino acid modification(s) at position(s) 298 and/or 333 of the Fc region, wherein the numbering of the residues in the Fc region is that of the EU index as in Kabat.
  • The polypeptide variant may display increased binding to an FcγRII and comprise an amino acid modification at any one or more of amino acid positions 255, 256, 258, 267, 268, 272, 276, 280, 283, 285, 286, 290, 301, 305, 307, 309, 312, 315, 320, 322, 326, 330, 331, 337, 340, 378, 398 or 430 of the Fc region, wherein the numbering of the residues in the Fc region is that of the EU index as in Kabat. Such polypeptide variants with increased binding to an FcγRII may optionally further display decreased binding to an FcγRIII and may, for example, comprise an amino acid modification at any one or more of amino acid positions 268, 272, 298, 301, 322 or 340 of the Fc region, wherein the numbering of the residues in the Fc region is that of the EU index as in Kabat.
  • The invention further provides a polypeptide comprising a variant Fc region with altered neonatal Fc receptor (FcRn) binding affinity, which polypeptide comprises an amino acid modification at any one or more of amino acid positions 238, 252, 253, 254, 255, 256, 265, 272, 286, 288, 303, 305, 307, 309, 311, 312, 317, 340, 356, 360, 362, 376, 378, 380, 382, 386, 388, 400, 413, 415, 424, 433, 434, 435, 436, 439 or 447 of the Fc region, wherein the numbering of the residues in the Fc region is that of the EU index as in Kabat. Such polypeptide variants with reduced binding to an FcRn may comprise an amino acid modification at any one or more of amino acid positions 252, 253, 254, 255, 288, 309, 386, 388, 400, 415, 433, 435, 436, 439 or 447 of the Fc region, wherein the numbering of the residues in the Fc region is that of the EU index as in Kabat. The above-mentioned polypeptide variants may, alternatively, display increased binding to FcRn and comprise an amino acid modification at any one or more of amino acid positions 238, 256, 265, 272, 286, 303, 305, 307, 311, 312, 317, 340, 356, 360, 362, 376, 378, 380, 382, 413, 424 or 434 of the Fc region, wherein the numbering of the residues in the Fc region is that of the EU index as in Kabat.
  • The invention also provides a composition comprising the polypeptide variant and a physiologically or pharmaceutically acceptable carrier or diluent. This composition for potential therapeutic use is sterile and may be lyophilized.
  • Diagnostic and therapeutic uses for the polypeptide variants disclosed herein are contemplated. In one diagnostic application, the invention provides a method for determining the presence of an antigen of interest comprising exposing a sample suspected of containing the antigen to the polypeptide variant and determining binding of the polypeptide variant to the sample. In one therapeutic application, the invention provides a method of treating a mammal suffering from or predisposed to a disease or disorder, comprising administering to the mammal a therapeutically effective amount of a polypeptide variant as disclosed herein, or of a composition comprising the polypeptide variant and a pharmaceutically acceptable carrier.
  • The invention further provides: isolated nucleic acid encoding the polypeptide variant; a vector comprising the nucleic acid, optionally, operably linked to control sequences recognized by a host cell transformed with the vector; a host cell containing the vector; a method for producing the polypeptide variant comprising culturing this host cell so that the nucleic acid is expressed and, optionally, recovering the polypeptide variant from the host cell culture (e.g. from the host cell culture medium).
  • The invention further provides a method for making a variant Fc region with altered Fc receptor (FcR) binding affinity, or altered antibody-dependent cell-mediated cytotoxicity (ADCC) activity, comprising:
    • (a) introducing one or more amino acid modifications into an Fc region of a parent polypeptide in order to generate a variant Fc region;
    • (b) determining binding of the variant Fc region to an FcR, or determining ADCC activity of the variant Fc region.
  • Step (b) of the method may comprise determining binding of the variant Fc region to one or more FcRs in vitro. Moreover, the method may result in the identification of a variant Fc region with improved FcR binding affinity, or with improved ADCC activity, in step (b) thereof. Where step (b) comprises determining binding of the Fc region to an FcR, the FcR may, for example, be human Fc gamma receptor III (FcγRIII). Where step (b) comprises determining binding of the variant Fc region to at least two different FcRs, the FcRs tested preferably include human Fc gamma receptor II (FcγRII) and human Fc gamma receptor III (FcγRIII).
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 is a schematic representation of a native IgG. Disulfide bonds are represented by heavy lines between CH1 and CL domains and the two CH2 domains. V is variable domain; C is constant domain; L stands for light chain and H stands for heavy chain.
  • FIG. 2 shows C1q binding of wild type (wt) C2B8 antibody; C2B8 antibody with a human IgG2 constant region (IgG2); and variants K322A, K320A and E318A.
  • FIG. 3 depicts C1q binding of variants P331A, P329A and K322A.
  • FIGS. 4A and 4B depict the amino acid sequences of E27 anti-IgE antibody light chain (FIG. 4A; SEQ ID NO:1) and heavy chain (FIG. 4B; SEQ ID NO:2).
  • FIG. 5 is a schematic diagram of the “immune complex” prepared for use in the FcR assay described in Example 1. The hexamer comprising three anti-IgE antibody molecules (the “Fc region-containing polypeptide”) and three IgE molecules (the “first target molecule”) is shown. IgE has two “binding sites” for the anti-IgE antibody (E27) in the Fc region thereof. Each IgE molecule in the complex is further able to bind two VEGF molecules (“the second target polypeptide”). VEGF has two “binding sites” for IgE.
  • FIG. 6 shows C1q binding results obtained for variants D270K and D270V compared to wild type C2B8.
  • FIG. 7 depicts complement dependent cytotoxicity (CDC) of variants D270K and D270V, compared to wild type C2B8.
  • FIG. 8 shows C1q binding ELISA results for 293 cell-produced wild type C2B8 antibody (293-Wt-C2B8), CHO-produced wild type C2B8 antibody (CHO-Wt-C2B8) and various variant antibodies.
  • FIG. 9 shows C1q binding ELISA results obtained for wild type (wt) C2B8 and various variant antibodies as determined in Example 3.
  • FIG. 10 depicts the three-dimensional structure of a human IgG Fc region, highlighting residues: Asp270, Lys326, Pro329, Pro331, Lys322 and Glu333.
  • FIG. 11 shows C1q binding ELISA results obtained for wild type C2B8 and various variant antibodies as determined in Example 3.
  • FIG. 12 shows C1q binding ELISA results obtained for wild type C2B8 and double variants, K326M-E333S and K326A-E333A.
  • FIG. 13 shows CDC of wild type C2B8 and double variants, K326M-E333S and K326A-E333A.
  • FIG. 14 depicts C1q binding ELISA results obtained for C2B8 with a human IgG4 (IgG4), wild type C2B8 (Wt-C2B8), C2B8 with a human IgG2 constant region (IgG2), and variant antibodies as described in Example 3.
  • FIGS. 15A and 15B show binding patterns for parent antibody (E27) to FcγRIIB and FcγRIIIA. FIG. 15A shows the binding pattern for the humanized anti-IgE E27 IgG1 as a monomer (open circles), hexamer (closed squares), and immune complex consisting of multiple hexamers (closed triangles) to a recombinant GST fusion protein of the human FcγRIIB (CD32) receptor α subunit. The hexameric complex (closed squares) was formed by the mixture of equal molar concentrations of E27 (which binds to the Fc region of human IgE) and a human myeloma IgE. The hexamer is a stable 1.1 kD complex consisting of 3 IgG molecules (150 kD each) and 3 IgE molecules (200 kD each). The immune complex (closed triangles) was formed sequentially by first mixing equal molar concentrations of E27 and recombinant anti-VEGF IgE (human IgE with Fab variable domains that bind human VEGF) to form the hexamer. Hexamers were then linked to form an immune complex by the addition of 2× molar concentration of human VEGF, a 44 kD homodimer which has two binding sites for the anti-VEGF IgE per mole of VEGF. FIG. 15B shows the binding pattern to a recombinant GST fusion protein of the human FcγRIIIA (CD16) receptor α subunit.
  • FIG. 16A shows the binding of immune complexes using different antigen-antibody pairs to recombinant GST fusion protein of the FcγRIIA receptor α subunit. FIG. 16B shows the binding of the same antigen-antibody pairs to the GST fusion protein of the FcγRIIIA receptor α subunit. Closed circles represent binding of human IgE:anti-IgE E27 IgG1; open circles represent binding of human VEGF:humanized anti-VEGF IgG1.
  • FIG. 17 summarizes differences in binding selectivity of some alanine variants between the different FcγRs. Binding of alanine variants at residues in the CH2 domain of anti-IgE E27 IgG1 are shown to FcγRIIA, FcγRIIB, and FcγRIIIA. Type 1 abrogates binding to all three receptors: D278A (265 in EU numbering). Type 2 improves binding to FcγRIIA and FcγRIIB, while binding to FcγRIIIA is unaffected: S280A (267 in EU numbering). Type 3 improves binding to FcγRIIA and FcγRIIB, but reduces binding to FcγRIIIA: H281A (268 in EU numbering). Type 4 reduces binding to FcγRIIA and FcγRIIB, while improving binding to FcγRIIIA: S317A (298 in EU numbering). Type 5 improves binding to FcγRIIIA, but does not affect binding to FcγRIIA and FcγRIIB: E352A, K353A (333 and 334 in EU numbering).
  • FIGS. 18A and 18B compare the FcγRIIIA protein/protein assay and CHO GPI-FcγRIIIA cell based assay, respectively. FIG. 18A illustrates binding of selected alanine variants to FcγRIIIA-GST fusion protein. S317A (298 in EU numbering) and S317A/K353A (298 and 334 in EU numbering) bind better than E27 wildtype, while D278A (265 in EU numbering) almost completely abrogates binding. FIG. 18B illustrates that a similar pattern of binding is found on CHO cells expressing a recombinant GPI-linked form of FcγRIIIA.
  • FIGS. 19A and 19B compare the FcγRIIB protein/protein assay and CHO GPI-FcγRIIB cell based assay, respectively. FIG. 19A illustrates binding of selected alanine variants to FcγRIIB-GST fusion protein. H281A (268 in EU numbering) binds better than E27 wildtype while S317A (298 in EU numbering) shows reduced binding. FIG. 19B illustrates that a similar pattern of binding is found on CHO cells expressing a recombinant membrane bound form of FcγRIIB.
  • FIG. 20 shows single alanine substitutions in the CH2 domain of anti-HER2 IgG1 (HERCEPTIN®) that influence FcγRIIIA binding in both the protein-protein and cell-based assays alter the ability to bind to FcγRIIIA on peripheral blood mononuclear cell (PBMC) effector cells. Recombinant humanized anti-HER2 (HERCEPTIN®), which binds to HER2-expressing SK-BR-3 breast tumor cells, was preincubated with 51Cr-labeled SK-BR-3 cells for 30 minutes (opsonization) at 100 ng/ml (filled circles) and 1.25 ng/ml (filled squares). Keeping the SK-BR-3 tumor target cell concentration constant, the ratio of effector cells was increased from 0 to 100. The spontaneous cytotoxicity in the absence of antibody (hatched squares) was 20% at an effector:target (E:T) ratio of 100:1. A single alanine mutation that did not affect FcγRIIIA binding, variant G31=R309A (292 in EU numbering), did not effect ADCC (filled triangles). A single alanine mutation that only slightly increased binding to FcγRIIIA, variant G30=K307A (290 in EU numbering), also showed slightly improved ADCC (i.e., a 1.1 fold improvement in ADCC activity, calculated as area under the curve) at 1.25 ng/ml at all E:T ratios (filled diamonds) compared to wildtype antibody at 1.25 ng/ml (filled square). A single alanine mutation that decreased binding to FcγRIIIA, variant G34=Q312A (295 in EU numbering), also showed decreased ADCC activity (filled inverted triangles).
  • FIG. 21 illustrates that a single alanine mutation which had the most improved binding to FcγRIIIA, variant G36=S317A (298 in EU numbering), in the protein-protein and cell-based assays also showed the most improvement in ADCC (filled triangles) among the variants compared to wildtype (closed squares) at 1.25 ng/ml. G36 displayed a 1.7 fold improvement in ADCC activity, calculated as area under the curve. Variants G17=E282A (269 in EU numbering) and G18=D283A (270 in EU numbering) both showed reduced binding to FcγRIIIA as well as reduced efficacy in ADCC. The effector cells were PBMCs.
  • FIG. 22A depicts alignments of native sequence IgG Fc regions. Native sequence human IgG Fc region sequences, hum IgG1 (non-A and A allotypes) (SEQ ID NOs: 3 and 4, respectively), humlgG2 (SEQ ID NO:5), hum IgG3 (SEQ ID NO:6) and humlgG4 (SEQ ID NO:7), are shown. The human IgG1 sequence is the non-A allotype, and differences between this sequence and the A allotype (at positions 356 and 358; EU numbering system) are shown below the human IgG1 sequence. Native sequence murine IgG Fc region sequences, murIgG1 (SEQ ID NO:8), murlgG2A (SEQ ID NO:9), murlgG2B (SEQ ID NO:10) and murlgG3 (SEQ ID NO:11), are also shown. FIG. 22B shows percent identity among the Fc region sequences of FIG. 22A.
  • FIG. 23 depicts alignments of native sequence human IgG Fc region sequences, humIgG1 (non-A and A allotypes; SEQ ID NOs:3 and 4, respectively), humIgG2 (SEQ ID NO:5), humIgG3 (SEQ ID NO:6) and humIgG4 (SEQ ID NO:7) with differences between the sequences marked with asterisks.
  • FIG. 24 shows area under curve (AUC) for selected variants compared to anti-HER2 IgG1 (HERCEPTIN®) in a 4 hour ADCC assay. The effector cells were PBMCs (N=5). Variant G36 (S317A; 298 in Eu numbering) with improved binding to FcγRIIIA showed improved ADCC activity; variant G31 (R309A; 292 in Eu numbering) which did not display altered FcγRIIIA binding, also had unaltered ADCC activity; and G14 (D265A; 278 in Eu numbering) which had reduced FcγRIIIA binding, also had reduced ADCC activity.
  • DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENTS I. Definitions
  • Throughout the present specification and claims, the numbering of the residues in an immunoglobulin heavy chain is that of the EU index as in Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md. (1991), expressly incorporated herein by reference. The “EU index as in Kabat” refers to the residue numbering of the human IgG1 EU antibody.
  • A “parent polypeptide” is a polypeptide comprising an amino acid sequence which lacks one or more of the Fc region modifications disclosed herein and which differs in effector function compared to a polypeptide variant as herein disclosed. The parent polypeptide may comprise a native sequence Fc region or an Fc region with pre-existing amino acid sequence modifications (such as additions, deletions and/or substitutions).
  • The term “Fc region” is used to define a C-terminal region of an immunoglobulin heavy chain, e.g., as shown in FIG. 1. The “Fc region” may be a native sequence Fc region or a variant Fc region. Although the boundaries of the Fc region of an immunoglobulin heavy chain might vary, the human IgG heavy chain Fc region is usually defined to stretch from an amino acid residue at position Cys226, or from Pro230, to the carboxyl-terminus thereof. The Fc region of an immunoglobulin generally comprises two constant domains, CH2 and CH3, as shown, for example, in FIG. 1.
  • The “CH2 domain” of a human IgG Fc region (also referred to as “Cγ2” domain) usually extends from about amino acid 231 to about amino acid 340. The CH2 domain is unique in that it is not closely paired with another domain. Rather, two N-linked branched carbohydrate chains are interposed between the two CH2 domains of an intact native IgG molecule. It has been speculated that the carbohydrate may provide a substitute for the domain-domain pairing and help stabilize the CH2 domain. Burton, Molec. Immunol. 22:161-206 (1985).
  • The “CH3 domain” comprises the stretch of residues C-terminal to a CH2 domain in an Fc region (i.e. from about amino acid residue 341 to about amino acid residue 447 of an IgG)
  • A “functional Fc region” possesses an “effector function” of a native sequence Fc region. Exemplary “effector functions” include C1q binding; complement dependent cytotoxicity; Fc receptor binding; antibody-dependent cell-mediated cytotoxicity (ADCC); phagocytosis; down regulation of cell surface receptors (e.g. B cell receptor; BCR), etc. Such effector functions generally require the Fc region to be combined with a binding domain (e.g. an antibody variable domain) and can be assessed using various assays as herein disclosed, for example.
  • A “native sequence Fc region” comprises an amino acid sequence identical to the amino acid sequence of an Fc region found in nature. Native sequence human Fc regions are shown in FIG. 23 and include a native sequence human IgG1 Fc region (non-A and A allotypes); native sequence human IgG2 Fc region; native sequence human IgG3 Fc region; and native sequence human IgG4 Fc region as well as naturally occurring variants thereof. Native sequence murine Fc regions are shown in FIG. 22A.
  • A “variant Fc region” comprises an amino acid sequence which differs from that of a native sequence Fc region by virtue of at least one “amino acid modification” as herein defined. Preferably, the variant Fc region has at least one amino acid substitution compared to a native sequence Fc region or to the Fc region of a parent polypeptide, e.g. from about one to about ten amino acid substitutions, and preferably from about one to about five amino acid substitutions in a native sequence Fc region or in the Fc region of the parent polypeptide. The variant Fc region herein will preferably possess at least about 80% homology with a native sequence Fc region and/or with an Fc region of a parent polypeptide, and most preferably at least about 90% homology therewith, more preferably at least about 95% homology therewith.
  • “Homology” is defined as the percentage of residues in the amino acid sequence variant that are identical after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent homology. Methods and computer programs for the alignment are well known in the art. One such computer program is “Align 2”, authored by Genentech, Inc., which was filed with user documentation in the United States Copyright Office, Washington, D.C. 20559, on Dec. 10, 1991.
  • The term “Fc region-containing polypeptide” refers to a polypeptide, such as an antibody or immunoadhesin (see definitions below), which comprises an Fc region.
  • The terms “Fc receptor” or “FcR” are used to describe a receptor that binds to the Fc region of an antibody. The preferred FcR is a native sequence human FcR. Moreover, a preferred FcR is one which binds an IgG antibody (a gamma receptor) and includes receptors of the FcγRI, FcγRII, and FcγRIII subclasses, including allelic variants and alternatively spliced forms of these receptors. FcγRII receptors include FcγRIIA (an “activating receptor”) and FcγRIIB (an “inhibiting receptor”), which have similar amino acid sequences that differ primarily in the cytoplasmic domains thereof. Activating receptor FcγRIIA contains an immunoreceptor tyrosine-based activation motif (ITAM) in its cytoplasmic domain. Inhibiting receptor FcγRIIB contains an immunoreceptor tyrosine-based inhibition motif (ITIM) in its cytoplasmic domain. (see review M. in Daëron, Annu. Rev. Immunol. 15:203-234 (1997)). FcRs are reviewed in Ravetch and Kinet, Annu. Rev. Immunol 9:457-92 (1991); Capel et al., Immunomethods 4:25-34 (1994); and de Haas et al., J. Lab. Clin. Med. 126:330-41 (1995). Other FcRs, including those to be identified in the future, are encompassed by the term “FcR” herein. The term also includes the neonatal receptor, FcRn, which is responsible for the transfer of maternal IgGs to the fetus (Guyer et al., J. Immunol. 117:587 (1976) and Kim et al., J. Immunol. 24:249 (1994)).
  • “Antibody-dependent cell-mediated cytotoxicity” and “ADCC” refer to a cell-mediated reaction in which nonspecific cytotoxic cells that express FcRs (e.g. Natural Killer (NK) cells, neutrophils, and macrophages) recognize bound antibody on a target cell and subsequently cause lysis of the target cell. The primary cells for mediating ADCC, NK cells, express FcγRIII only, whereas monocytes express FcγRI, FcγRII and FcγRIII. FcR expression on hematopoietic cells is summarized in Table 3 on page 464 of Ravetch and Kinet, Annu. Rev. Immunol. 9:457-92 (1991).
  • “Human effector cells” are leukocytes which express one or more FcRs and perform effector functions. Preferably, the cells express at least FcγRIII and perform ADCC effector function. Examples of human leukocytes which mediate ADCC include peripheral blood mononuclear cells (PBMC), natural killer (NK) cells, monocytes, cytotoxic T cells and neutrophils; with PBMCs and NK cells being preferred. The effector cells may be isolated from a native source thereof, e.g. from blood or PBMCs as described herein.
  • A polypeptide variant with “altered” FcR binding affinity or ADCC activity is one which has either enhanced or diminished FcR binding activity and/or ADCC activity compared to a parent polypeptide or to a polypeptide comprising a native sequence Fc region. The polypeptide variant which “displays increased binding” to an FcR binds at least one FcR with better affinity than the parent polypeptide. The polypeptide variant which “displays decreased binding” to an FcR, binds at least one FcR with worse affinity than a parent polypeptide. Such variants which display decreased binding to an FcR may possess little or no appreciable binding to an FcR, e.g., 0-20% binding to the FcR compared to a native sequence IgG Fc region, e.g. as determined in the Examples herein.
  • The polypeptide variant which binds an FcR with “better affinity' than a parent polypeptide, is one which binds any one or more of the above identified FcRs with substantially better binding affinity than the parent antibody, when the amounts of polypeptide variant and parent polypeptide in the binding assay are essentially the same. For example, the polypeptide variant with improved FcR binding affinity may display from about 1.15 fold to about 100 fold, e.g. from about 1.2 fold to about 50 fold improvement in FcR binding affinity compared to the parent polypeptide, where FcR binding affinity is determined, for example, as disclosed in the Examples herein.
  • The polypeptide variant which “mediates antibody-dependent cell-mediated cytotoxicity (ADCC) in the presence of human effector cells more effectively” than a parent antibody is one which in vitro or in vivo is substantially more effective at mediating ADCC, when the amounts of polypeptide variant and parent antibody used in the assay are essentially the same. Generally, such variants will be identified using the in vitro ADCC assay as herein disclosed, but other assays or methods for determining ADCC activity, e.g. in an animal model etc, are contemplated. The preferred variant is from about 1.5 fold to about 100 fold, e.g. from about two fold to about fifty fold, more effective at mediating ADCC than the parent, e.g. in the in vitro assay disclosed herein.
  • An “amino acid modification” refers to a change in the amino acid sequence of a predetermined amino acid sequence. Exemplary modifications include an amino acid substitution, insertion and/or deletion. The preferred amino acid modification herein is a substitution.
  • An “amino acid modification at” a specified position, e.g. of the Fc region, refers to the substitution or deletion of the specified residue, or the insertion of at least one amino acid residue adjacent the specified residue. By insertion “adjacent” a specified residue is meant insertion within one to two residues thereof. The insertion may be N-terminal or C-terminal to the specified residue.
  • An “amino acid substitution” refers to the replacement of at least one existing amino acid residue in a predetermined amino acid sequence with another different “replacement” amino acid residue. The replacement residue or residues may be “naturally occurring amino acid residues” (i.e. encoded by the genetic code) and selected from the group consisting of: alanine (Ala); arginine (Arg); asparagine (Asn); aspartic acid (Asp); cysteine (Cys); glutamine (Gln); glutamic acid (Glu); glycine (Gly); histidine (His); isoleucine (Ile): leucine (Leu); lysine (Lys); methionine (Met); phenylalanine (Phe); proline (Pro); serine (Ser); threonine (Thr); tryptophan (Trp); tyrosine (Tyr); and valine (Val). Preferably, the replacement residue is not cysteine. Substitution with one or more non-naturally occurring amino acid residues is also encompassed by the definition of an amino acid substitution herein. A “non-naturally occurring amino acid residue” refers to a residue, other than those naturally occurring amino acid residues listed above, which is able to covalently bind adjacent amino acid residues(s) in a polypeptide chain. Examples of non-naturally occurring amino acid residues include norleucine, ornithine, norvaline, homoserine and other amino acid residue analogues such as those described in Ellman et al. Meth. Enzym. 202:301-336 (1991). To generate such non-naturally occurring amino acid residues, the procedures of Noren et al., Science 244:182 (1989) and Ellman et al., supra, can be used. Briefly, these procedures involve chemically activating a suppressor tRNA with a non-naturally occurring amino acid residue followed by in vitro transcription and translation of the RNA.
  • An “amino acid insertion” refers to the incorporation of at least one amino acid into a predetermined amino acid sequence. While the insertion will usually consist of the insertion of one or two amino acid residues, the present application contemplates larger “peptide insertions”, e.g. insertion of about three to about five or even up to about ten amino acid residues. The inserted residue(s) may be naturally occurring or non-naturally occurring as disclosed above.
  • An “amino acid deletion” refers to the removal of at least one amino acid residue from a predetermined amino acid sequence.
  • “Hinge region” is generally defined as stretching from Glu216 to Pro230 of human IgG1 (Burton, Molec. immunol. 22:161-206 (1985)). Hinge regions of other IgG isotypes may be aligned with the IgG1 sequence by placing the first and last cysteine residues forming inter-heavy chain S—S bonds in the same positions.
  • The “lower hinge region” of an Fc region is normally defined as the stretch of residues immediately C-terminal to the hinge region, i.e. residues 233 to 239 of the Fc region. Prior to the present invention, FcγR binding was generally attributed to amino acid residues in the lower hinge region of an IgG Fc region.
  • “C1q” is a polypeptide that includes a binding site for the Fc region of an immunoglobulin. C1q together with two serine proteases, C1r and C1s, forms the complex C1; the first component of the complement dependent cytotoxicity (CDC) pathway. Human C1q can be purchased commercially from, e.g. Quidel, San Diego, Calif.
  • The term “binding domain” refers to the region of a polypeptide that binds to another molecule. In the case of an FcR, the binding domain can comprise a portion of a polypeptide chain thereof (e.g. the a chain thereof) which is responsible for binding an Fc region. One useful binding domain is the extracellular domain of an FcR a chain.
  • The term “antibody” is used in the broadest sense and specifically covers monoclonal antibodies (including full length monoclonal antibodies), polyclonal antibodies, multispecific antibodies (e.g., bispecific antibodies), and antibody fragments so long as they exhibit the desired biological activity.
  • “Antibody fragments”, as defined for the purpose of the present invention, comprise a portion of an intact antibody, generally including the antigen binding or variable region of the intact antibody or the Fc region of an antibody which retains FcR binding capability. Examples of antibody fragments include linear antibodies; single-chain antibody molecules; and multispecific antibodies formed from antibody fragments. The antibody fragments preferably retain at least part of the hinge and optionally the CH1 region of an IgG heavy chain. More preferably, the antibody fragments retain the entire constant region of an IgG heavy chain, and include an IgG light chain.
  • The term “monoclonal antibody” as used herein refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally occurring mutations that may be present in minor amounts. Monoclonal antibodies are highly specific, being directed against a single antigenic site. Furthermore, in contrast to conventional (polyclonal) antibody preparations that typically include different antibodies directed against different determinants (epitopes), each monoclonal antibody is directed against a single determinant on the antigen. The modifier “monoclonal” indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method. For example, the monoclonal antibodies to be used in accordance with the present invention may be made by the hybridoma method first described by Kohler et al., Nature 256:495 (1975), or may be made by recombinant DNA methods (see, e.g., U.S. Pat. No. 4,816,567). The “monoclonal antibodies” may also be isolated from phage antibody libraries using the techniques described in Clackson et al., Nature 352:624-628 (1991) and Marks et al., J. Mol. Biol. 222:581-597 (1991), for example.
  • The monoclonal antibodies herein specifically include “chimeric” antibodies (immunoglobulins) in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is identical with or homologous to corresponding sequences in antibodies derived from another species or belonging to another antibody class or subclass, as well as fragments of such antibodies, so long as they exhibit the desired biological activity (U.S. Pat. No. 4,816,567; and Morrison et al., Proc. Natl. Acad. Sci. USA 81:6851-6855 (1984)).
  • “Humanized” forms of non-human (e.g., murine) antibodies are chimeric antibodies that contain minimal sequence derived from non-human immunoglobulin. For the most part, humanized antibodies are human immunoglobulins (recipient antibody) in which residues from a hypervariable region of the recipient are replaced by residues from a hypervariable region of a non-human species (donor antibody) such as mouse, rat, rabbit or nonhuman primate having the desired specificity, affinity, and capacity. In some instances, Fv framework region (FR) residues of the human immunoglobulin are replaced by corresponding non-human residues. Furthermore, humanized antibodies may comprise residues that are not found in the recipient antibody or in the donor antibody. These modifications are made to further refine antibody performance. In general, the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the hypervariable loops correspond to those of a non-human immunoglobulin and all or substantially all of the FR regions are those of a human immunoglobulin sequence. The humanized antibody optionally also will comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin. For further details, see Jones et al., Nature 321:522-525 (1986); Riechmann et al., Nature 332:323-329 (1988); and Presta, Curr. Op. Struct. Biol. 2:593-596 (1992).
  • The term “hypervariable region” when used herein refers to the amino acid residues of an antibody which are responsible for antigen-binding. The hypervariable region comprises amino acid residues from a “complementarity determining region” or “CDR” (Le. residues 24-34 (L1), 50-56 (L2) and 89-97 (L3) in the light chain variable domain and 31-35 (H1), 50-65 (H2) and 95-102 (H3) in the heavy chainvariable domain; Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md. (1991)) and/or those residues from a “hypervariable loop” (i.e. residues 26-32 (L1), 50-52 (L2) and 91-96 (L3) in the light chain variable domain and 26-32 (H1), 53-55 (H2) and 96-101 (H3) in the heavy chain variable domain; Chothia and Lesk J. Mol. Biol. 196:901-917 (1987)). “Framework” or “FR” residues are those variable domain residues other than the hypervariable region residues as herein defined.
  • As used herein, the term “immunoadhesin” designates antibody-like molecules which combine the “binding domain” of a heterologous “adhesin” protein (e.g. a receptor, ligand or enzyme) with an immunoglobulin constant domain. Structurally, the immunoadhesins comprise a fusion of the adhesin amino acid sequence with the desired binding specificity which is other than the antigen recognition and binding site (antigen combining site) of an antibody (i.e. is “heterologous”) and an immunoglobulin constant domain sequence.
  • The term “ligand binding domain” as used herein refers to any native cell-surface receptor or any region or derivative thereof retaining at least a qualitative ligand binding ability of a corresponding native receptor. In a specific embodiment, the receptor is from a cell-surface polypeptide having an extracellular domain that is homologous to a member of the immunoglobulin supergenefamily. Other receptors, which are not members of the immunoglobulin supergenefamily but are nonetheless specifically covered by this definition, are receptors for cytokines, and in particular receptors with tyrosine kinase activity (receptor tyrosine kinases), members of the hematopoietin and nerve growth factor receptor superfamilies, and cell adhesion molecules, e.g. (E-, L- and P-) selectins.
  • The term “receptor binding domain” is used to designate any native ligand for a receptor, including cell adhesion molecules, or any region or derivative of such native ligand retaining at least a qualitative receptor binding ability of a corresponding native ligand. This definition, among others, specifically includes binding sequences from ligands for the above-mentioned receptors.
  • An “antibody-immunoadhesin chimera” comprises a molecule that combines at least one binding domain of an antibody (as herein defined) with at least one immunoadhesin (as defined in this application). Exemplary antibody-immunoadhesin chimeras are the bispecific CD4-IgG chimeras described in Berg et al., PNAS (USA) 88:4723-4727 (1991) and Chamow et al., J. Immunol. 153:4268 (1994).
  • An “isolated” polypeptide is one that has been identified and separated and/or recovered from a component of its natural environment. Contaminant components of its natural environment are materials that would interfere with diagnostic or therapeutic uses for the polypeptide, and may include enzymes, hormones, and other proteinaceous or nonproteinaceous solutes. In preferred embodiments, the polypeptide will be purified (1) to greater than 95% by weight of polypeptide as determined by the Lowry method, and most preferably more than 99% by weight, (2) to a degree sufficient to obtain at least 15 residues of N-terminal or internal amino acid sequence by use of a spinning cup sequenator, or (3) to homogeneity by SDS-PAGE under reducing or nonreducing conditions using Coomassie blue or, preferably, silver stain. Isolated polypeptide includes the polypeptide in situ within recombinant cells since at least one component of the polypeptide's natural environment will not be present. Ordinarily, however, isolated polypeptide will be prepared by at least one purification step.
  • “Treatment” refers to both therapeutic treatment and prophylactic or preventative measures. Those in need of treatment include those already with the disorder as well as those in which the disorder is to be prevented.
  • A “disorder is any condition that would benefit from treatment with the polypeptide variant. This includes chronic and acute disorders or diseases including those pathological conditions which predispose the mammal to the disorder in question. In one embodiment, the disorder is cancer.
  • The terms “cancer” and “cancerous” refer to or describe the physiological condition in mammals that is typically characterized by unregulated cell growth. Examples of cancer include but are not limited to, carcinoma, lymphoma, blastoma, sarcoma, and leukemia. More particular examples of such cancers include squamous cell cancer, small-cell lung cancer, non-small cell lung cancer, adenocarcinoma of the lung, squamous carcinoma of the lung, cancer of the peritoneum, hepatocellular cancer, gastrointestinal cancer, pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, hepatoma, breast cancer, colon cancer, colorectal cancer, endometrial or uterine carcinoma, salivary gland carcinoma, kidney cancer, liver cancer, prostate cancer, vulval cancer, thyroid cancer, hepatic carcinoma and various types of head and neck cancer.
  • A “HER2-expressing cancer” is one comprising cells which have HER2 receptor protein (Semba et al., PNAS (USA) 82:6497-6501 (1985) and Yamamoto et al., Nature 319:230-234 (1986) (Genebank accession number X03363)) present at their cell surface, such that an anti-HER2 antibody is able to bind to the cancer.
  • The word “label” when used herein refers to a detectable compound or composition which is conjugated directly or indirectly to the polypeptide. The label may be itself be detectable (e.g., radioisotope labels or fluorescent labels) or, in the case of an enzymatic label, may catalyze chemical alteration of a substrate compound or composition which is detectable.
  • An “isolated” nucleic acid molecule is a nucleic acid molecule that is identified and separated from at least one contaminant nucleic acid molecule with which it is ordinarily associated in the natural source of the polypeptide nucleic acid. An isolated nucleic acid molecule is other than in the form or setting in which it is found in nature. Isolated nucleic acid molecules therefore are distinguished from the nucleic acid molecule as it exists in natural cells. However, an isolated nucleic acid molecule includes a nucleic acid molecule contained in cells that ordinarily express the polypeptide where, for example, the nucleic acid molecule is in a chromosomal location different from that of natural cells.
  • The expression “control sequences” refers to DNA sequences necessary for the expression of an operably linked coding sequence in a particular host organism. The control sequences that are suitable for prokaryotes, for example, include a promoter, optionally an operator sequence, and a ribosome binding site. Eukaryotic cells are known to utilize promoters, polyadenylation signals, and enhancers.
  • Nucleic acid is “operably linked” when it is placed into a functional relationship with another nucleic acid sequence. For example, DNA for a presequence or secretory leader is operably linked to DNA for a polypeptide if it is expressed as a preprotein that participates in the secretion of the polypeptide; a promoter or enhancer is operably linked to a coding sequence if it affects the transcription of the sequence; or a ribosome binding site is operably linked to a coding sequence if it is positioned so as to facilitate translation. Generally, “operably linked” means that the DNA sequences being linked are contiguous, and, in the case of a secretory leader, contiguous and in reading phase. However, enhancers do not have to be contiguous. Linking is accomplished by ligation at convenient restriction sites. If such sites do not exist, the synthetic oligonucleotide adaptors or linkers are used in accordance with conventional practice.
  • As used herein, the expressions “cell,” “cell line,” and “cell culture” are used interchangeably and all such designations include progeny. Thus, the words “transformants” and “transformed cells” include the primary subject cell and cultures derived therefrom without regard for the number of transfers. It is also understood that all progeny may not be precisely identical in DNA content, due to deliberate or inadvertent mutations. Mutant progeny that have the same function or biological activity as screened for in the originally transformed cell are included. Where distinct designations are intended, it will be clear from the context.
  • The term “molecular complex” when used herein refers to the relatively stable structure which forms when two or more heterologous molecules (e.g. polypeptides) bind (preferably noncovalently) to one another. The preferred molecular complex herein is an immune complex.
  • “Immune complex” refers to the relatively stable structure which forms when at least one target molecule and at least one heterologous Fc region-containing polypeptide bind to one another forming a larger molecular weight complex. Examples of immune complexes are antigen-antibody aggregates and target molecule-immunoadhesin aggregates. The term “immune complex” as used herein, unless indicated otherwise, refers to an ex vivo complex (i.e. other than the form or setting in which it may be found in nature). However, the immune complex may be administered to a mammal, e.g. to evaluate clearance of the immune complex in the mammal.
  • The term “target molecule” refers to a molecule, usually a polypeptide, which is capable of being bound by a heterologous molecule and has one or more binding sites for the heterologous molecule. The term “binding site” refers to a region of a molecule to which another molecule can bind. The “first target molecule” herein comprises at least two distinct binding sites (for example, two to five separate binding sites) for an analyte (e.g. an Fc region-containing polypeptide) such that at least two analyte molecules can bind to the first target molecule. In the preferred embodiment of the invention, the two or more binding sites are identical (e.g. having the same amino acid sequence, where the target molecule is a polypeptide). In Example 1 below, the first target molecule was IgE and had two separate binding sites in the Fc region thereof to which the Fc region-containing polypeptide (an anti-IgE antibody, E27) could bind. Other first target molecules include dimers of substantially identical monomors (e.g. neurotrophins, IL8 and VEGF) or are polypeptides comprising two or more substantially identical polypeptide chains (e.g. antibodies or immunoadhesins). The “second target molecule” comprises at least two distinct binding sites (for example, two to five separate binding sites) for the first target molecule such that at least two first target molecules can bind to the second target molecule. Preferably, the two or more binding sites are identical (e.g. having the same amino acid sequence, where the target molecule is a polypeptide). In Example 2, the second target molecule was VEGF, which has a pair of distinct binding sites to which the variable domain of the IgE antibody could bind. Other second target molecules are contemplated, e.g. other dimers of substantially identical monomers (e.g. neurotrophins or IL8) or polypeptides comprising two or more substantially identical domains (e.g. antibodies or immunoadhesins).
  • An “analyte” is a substance that is to be analyzed. The preferred analyte is an Fc region-containing polypeptide that is to be analyzed for its ability to bind to an Fc receptor.
  • A “receptor” is a polypeptide capable of binding at least one ligand. The preferred receptor is a cell-surface receptor having an extracellular ligand-binding domain and, optionally, other domains (e.g. transmembrane domain, intracellular domain and/or membrane anchor). The receptor to be evaluated in the assay described herein may be an intact receptor or a fragment or derivative thereof (e.g. a fusion protein comprising the binding domain of the receptor fused to one or more heterologous polypeptides). Moreover, the receptor to be evaluated for its binding properties may be present in a cell or isolated and optionally coated on an assay plate or some other solid phase.
  • The phrase “low affinity receptor” denotes a receptor that has a weak binding affinity for a ligand of interest, e.g. having a binding constant of about 50 nM or worse affinity. Exemplary low affinity receptors include FcγRII and FcγRIII.
  • II. Modes for Carrying Out the Invention
  • The invention herein relates to a method for making a polypeptide variant. The “parent”, “starting” or “nonvariant” polypeptide is prepared using techniques available in the art for generating polypeptides comprising an Fc region. In the preferred embodiment of the invention, the parent polypeptide is an antibody and exemplary methods for generating antibodies are described in more detail in the following sections. The parent polypeptide may, however, be any other polypeptide comprising an Fc region, e.g. an immunoadhesin. Methods for making immunoadhesins are elaborated in more detail hereinbelow.
  • In an alternative embodiment, a variant Fc region may be generated according to the methods herein disclosed and this “variant Fc region” can be fused to a heterologous polypeptide of choice, such as an antibody variable domain or binding domain of a receptor or ligand.
  • The parent polypeptide comprises an Fc region. Generally the Fc region of the parent polypeptide will comprise a native sequence Fc region, and preferably a human native sequence Fc region. However, the Fc region of the parent polypeptide may have one or more pre-existing amino acid sequence alterations or modifications from a native sequence Fc region. For example, the C1q binding activity of the Fc region may have been previously altered (other types of Fc region modifications are described in more detail below). In a further embodiment the parent polypeptide Fc region is “conceptual” and, while it does not physically exist, the antibody engineer may decide upon a desired variant Fc region amino acid sequence and generate a polypeptide comprising that sequence or a DNA encoding the desired variant Fc region amino acid sequence.
  • In the preferred embodiment of the invention, however, a nucleic acid encoding an Fc region of a parent polypeptide is available and this nucleic acid sequence is altered to generate a variant nucleic acid sequence encoding the Fc region variant.
  • DNA encoding an amino acid sequence variant of the starting polypeptide is prepared by a variety of methods known in the art. These methods include, but are not limited to, preparation by site-directed (or oligonucleotide-mediated) mutagenesis, PCR mutagenesis, and cassette mutagenesis of an earlier prepared DNA encoding the polypeptide
  • Site-directed mutagenesis is a preferred method for preparing substitution variants. This technique is well known in the art (see, e.g., Carter et al., Nucleic Acids Res. 13:4431-4443 (1985) and Kunkel et al., Proc. Natl. Acad. Sci. USA 82:488-492 (1985)). Briefly, in carrying out site-directed mutagenesis of DNA, the starting DNA is altered by first hybridizing an oligonucleotide encoding the desired mutation to a single strand of such starting DNA. After hybridization, a DNA polymerase is used to synthesize an entire second strand, using the hybridized oligonucleotide as a primer, and using the single strand of the starting DNA as a template. Thus, the oligonucleotide encoding the desired mutation is incorporated in the resulting double-stranded DNA.
  • PCR mutagenesis is also suitable for making amino acid sequence variants of the starting polypeptide. See Higuchi, in PCR Protocols, pp. 177-183 (Academic Press, 1990); and Vallette et al., Nuc. Acids Res. 17:723-733 (1989). Briefly, when small amounts of template DNA are used as starting material in a PCR, primers that differ slightly in sequence from the corresponding region in a template DNA can be used to generate relatively large quantities of a specific DNA fragment that differs from the template sequence only at the positions where the primers differ from the template.
  • Another method for preparing variants, cassette mutagenesis, is based on the technique described by Wells et al., Gene 34:315-323 (1985). The starting material is the plasmid (or other vector) comprising the starting polypeptide DNA to be mutated. The codon(s) in the starting DNA to be mutated are identified. There must be a unique restriction endonuclease site on each side of the identified mutation site(s). If no such restriction sites exist, they may be generated using the above-described oligonucleotide-mediated mutagenesis method to introduce them at appropriate locations in the starting polypeptide DNA. The plasmid DNA is cut at these sites to linearize it. A double-stranded oligonucleotide encoding the sequence of the DNA between the restriction sites but containing the desired mutation(s) is synthesized using standard procedures, wherein the two strands of the oligonucleotide are synthesized separately and then hybridized together using standard techniques. This double-stranded oligonucleotide is referred to as the cassette. This cassette is designed to have 5′ and 3′ ends that are compatible with the ends of the linearized plasmid, such that it can be directly ligated to the plasmid. This plasmid now contains the mutated DNA sequence.
  • Alternatively, or additionally, the desired amino acid sequence encoding a polypeptide variant can be determined, and a nucleic acid sequence encoding such amino acid sequence variant can be generated synthetically.
  • The amino acid sequence of the parent polypeptide is modified in order to generate a variant Fc region with altered Fc receptor binding affinity or activity in vitro and/or in vivo and/or altered antibody-dependent cell-mediated cytotoxicity (ADCC) activity in vitro and/or in vivo.
  • Generally, the modification entails one or more amino acid substitutions. In one embodiment, the replacement residue does not correspond to a residue present in the same position in any of the native sequence Fc regions in FIG. 22A. For example, according to this embodiment of the invention, Pro331 of a human IgG3 or IgG1 Fc region is replaced with a residue other than Ser (the corresponding aligned residue found in native sequence human IgG4). In one embodiment, the residue in the parent polypeptide which is substituted with a replacement residue is not an alanine and/or is not residue A1a339 of an Fc region. In the case of an amino acid substitution, preferably the residue in the parent polypeptide is replaced with an alanine residue. However, the present invention contemplates replacement of the residue of the parent polypeptide with any other amino acid residue. The substitution may, for example, be a “conservative substitution”. Such conservative substitutions are shown in Table 1 under the heading of “preferred substitution”. More substantial changes may be achieved by making one or more “exemplary substitutions” which are not the preferred substitution in Table 1.
  • TABLE 1
    Original Exemplary Preferred
    Residue Substitutions Substitution
    Ala (A) val; leu; ile val
    Arg (R) lys; gln; asn Lys
    Asn (N) gln; his; lys; arg Gln
    Asp (D) glu Glu
    Cys (C) ser Ser
    Gln (Q) asn Asn
    Glu (E) asp Asp
    Gly (G) pro; ala Ala
    His (H) asn; gln; lys; arg Arg
    Ile (I) leu; val; met; ala; Leu
    phe; norleucine
    Leu (L) norleucine; ile; val; Ile
    met; ala; phe
    Lys (K) arg; gln; asn Arg
    Met (M) leu; phe; ile Leu
    Phe (F) leu; val; ile; ala; tyr leu
    Pro (P) ala ala
    Ser (S) thr thr
    Thr (T) ser ser
    Trp (W) tyr; phe tyr
    Tyr (Y) trp; phe; thr; ser phe
    Val (V) ile; leu; met; phe; leu
    ala; norleucine
  • Substantial modifications in the biological properties of the Fc region may be accomplished by selecting substitutions that differ significantly in their effect on maintaining (a) the structure of the polypeptide backbone in the area of the substitution, for example, as a sheet or helical conformation, (b) the charge or hydrophobicity of the molecule at the target site, or (c) the bulk of the side chain. Naturally occurring residues are divided into groups based on common side-chain properties:
  • (1) hydrophobic: norleucine, met, ala, val, leu, ile;
  • (2) neutral hydrophilic: cys, ser, thr;
  • (3) acidic: asp, glu;
  • (4) basic: asn, gln, his, lys, arg;
  • (5) residues that influence chain orientation: gly, pro; and
  • (6) aromatic: trp, tyr, phe.
  • Non-conservative substitutions will entail exchanging a member of one of these classes for a member of another class. Conservative and non-conservative amino acid substitutions are exemplified in Table 8 hereinbelow.
  • As is demonstrated in Example 4 herein, one can engineer an Fc region variant with altered binding affinity for one or more FcRs. As was shown in that Example, different classes of Fc region variants can be made e.g., as summarized in the following table. Where the variant Fc region has more than one amino acid substitution, generally, but not necessarily, amino acid substitutions in the same class are combined to achieve the desired result.
  • TABLE 2
    CLASSES OF Fc REGION VARIANTS
    Class FcR binding property Position of Fc region substitution(s)
    1A reduced binding to all FcγR 238, 265, 269, 270, 297*, 327, 329
    1B reduced binding to both FcγRII and 239, 294, 295, 303, 338, 373, 376, 416, 435
    FcγRIII
    2 improved binding to both FcγRII and 256, 290, 312, 326, 330, 339#, 378, 430
    FcγRIII
    3 improved binding to FcγRII and no effect 255, 258, 267, 276, 280, 283, 285, 286, 305, 307,
    on FcγRIII binding 309, 315, 320, 331, 337, 398
    4 improved binding to FcγRII and reduced 268, 272, 301, 322, 340
    binding to FcγRIII
    5 reduced binding to FcγRII and no effect 292, 324, 335, 414, 419, 438, 439
    on FcγRIII binding
    6 reduced binding to FcγRII and improved 298, 333
    binding to FcγRIII
    7 no effect on FcγRII binding and reduced 248, 249, 252, 254, 278, 289, 293, 296, 338, 382,
    binding to FcγRIII 388, 389, 434, 437
    8 no effect on FcγRII binding and improved 334, 360
    binding to FcγRIII
    *deglycosylated version
    #Preferably combined with other Fc modification(s), (e.g. as disclosed herein)
    Aside from amino acid substitutions, the present invention contemplates other modifications of the parent Fc region amino acid sequence in order to generate an Fc region variant with altered effector function.
  • One may, for example, delete one or more amino acid residues of the Fc region in order to reduce binding to an FcR. Generally, one will delete one or more of the Fc region residues identified herein as effecting FcR binding (see Example 4 below) in order to generate such an Fc region variant. Generally, no more than one to about ten Fc region residues will be deleted according to this embodiment of the invention. The Fc region herein comprising one or more amino acid deletions will preferably retain at least about 80%, and preferably at least about 90%, and most preferably at least about 95%, of the parent Fc region or of a native sequence human Fc region.
  • One may also make amino acid insertion Fc region variants, which variants have altered effector function. For example, one may introduce at least one amino acid residue (e.g. one to two amino acid residues and generally no more than ten residues) adjacent to one or more of the Fc region positions identified herein as impacting FcR binding. By “adjacent” is meant within one to two amino acid residues of a Fc region residue identified herein. Such Fc region variants may display enhanced or diminished FcR binding and/or ADCC activity. In order to generate such insertion variants, one may evaluate a co-crystal structure of a polypeptide comprising a binding region of an FcR (e.g. the extracellular domain of the FcR of interest) and the Fc region into which the amino acid residue(s) are to be inserted (see, for example, Deisenhofer, Biochemistry 20(9):2361-2370 (1981); and Burmeister et al., Nature 342:379-383, (1994) order to rationally design an Fc region variant with, e.g., improved FcR binding ability. Such insertion(s) will generally be made in an Fc region loop, but not in the secondary structure (i.e. in a β-strand) of the Fc region.
  • By introducing the appropriate amino acid sequence modifications in a parent Fc region, one can generate a variant Fc region which (a) mediates antibody-dependent cell-mediated cytotoxicity (ADCC) in the presence of human effector cells more effectively and/or (b) binds an Fc gamma receptor (FcγR) with better affinity than the parent polypeptide. Such Fc region variants will generally comprise at least one amino acid modification in the Fc region. Combining amino acid modifications is thought to be particularly desirable. For example, the variant Fc region may include two, three, four, five, etc substitutions therein, e.g. of the specific Fc region positions identified herein.
  • Preferably, the parent polypeptide Fc region is a human Fc region, e.g. a native sequence human Fc region human IgG1 (A and non-A allotypes), IgG2, IgG3 or IgG4 Fc region. Such sequences are shown in FIG. 23.
  • To generate an Fc region with improved ADCC activity, the parent polypeptide preferably has pre-existing ADCC activity, e.g., it comprises a human IgG1 or human IgG3 Fc region. In one embodiment, the variant with improved ADCC mediates ADCC substantially more effectively than an antibody with a native sequence IgG1 or IgG3 Fc region and the antigen-binding region of the variant. Preferably, the variant comprises, or consists essentially of, substitutions of two or three of the residues at positions 298, 333 and 334 of the Fc region. Most preferably, residues at positions 298, 333 and 334 are substituted, (e.g. with alanine residues). Moreover, in order to generate the Fc region variant with improved ADCC activity, one will generally engineer an Fc region variant with improved binding affinity for FcγRIII, which is thought to be an important FcR for mediating ADCC. For example, one may introduce an amino acid modification (e.g. a substitution) into the parent Fc region at any one or more of amino acid positions 256, 290, 298, 312, 326, 330, 333, 334, 360, 378 or 430 to generate such a variant. The variant with improved binding affinity for FcγRIII may further have reduced binding affinity for FcγRII, especially reduced affinity for the inhibiting FcγRIIB receptor.
  • The amino acid modification(s) are preferably introduced into the CH2 domain of a Fc region, since the experiments herein indicate that the CH2 domain is important for FcR binding activity. Moreover, unlike the teachings of the above-cited art, the instant application contemplates the introduction of a modification into a part of the Fc region other than in the lower hinge region thereof.
  • Useful amino acid positions for modification in order to generate a variant IgG Fc region with altered Fc gamma receptor (FcγR) binding affinity or activity include any one or more of amino acid positions 238, 239, 248, 249, 252, 254, 255, 256, 258, 265, 267, 268, 269, 270, 272, 276, 278, 280, 283, 285, 286, 289, 290, 292, 293, 294, 295, 296, 298, 301, 303, 305, 307, 309, 312, 315, 320, 322, 324, 326, 327, 329, 330, 331, 333, 334, 335, 337, 338, 340, 360, 373, 376, 378, 382, 388, 389, 398, 414, 416, 419, 430, 434, 435, 437, 438 or 439 of the Fc region. Preferably, the parent Fc region used as the template to generate such variants comprises a human IgG Fc region. Where residue 331 is substituted, the parent Fc region is preferably not human native sequence IgG3, or the variant Fc region comprising a substitution at position 331 preferably displays increased FcR binding, e.g. to FcγRII.
  • To generate an Fc region variant with reduced binding to the FcγR one may introduce an amino acid modification at any one or more of amino acid positions 238, 239, 248, 249, 252, 254, 265, 268, 269, 270, 272, 278, 289, 292, 293, 294, 295, 296, 298, 301, 303, 322, 324, 327, 329, 333, 335, 338, 340, 373, 376, 382, 388, 389, 414, 416, 419, 434, 435, 437, 438 or 439 of the Fc region.
  • Variants which display reduced binding to FcγRI, include those comprising an Fc region amino acid modification at any one or more of amino acid positions 238, 265, 269, 270, 327 or 329.
  • Variants which display reduced binding to FcγRII include those comprising an Fc region amino acid modification at any one or more of amino acid positions 238, 265, 269, 270, 292, 294, 295, 298, 303, 324, 327, 329, 333, 335, 338, 373, 376, 414, 416, 419, 435, 438 or 439.
  • Fc region variants which display reduced binding to FcγRIII include those comprising an Fc region amino acid modification at any one or more of amino acid positions 238, 239, 248, 249, 252, 254, 265, 268, 269, 270, 272, 278, 289, 293, 294, 295, 296, 301, 303, 322, 327, 329, 338, 340, 373, 376, 382, 388, 389, 416, 434, 435 or 437.
  • Variants with improved binding to one or more FcγRs may also be made. Such Fc region variants may comprise an amino acid modification at any one or more of amino acid positions 255, 256, 258, 267, 268, 272, 276, 280, 283, 285, 286, 290, 298, 301, 305, 307, 309, 312, 315, 320, 322, 326, 330, 331, 333, 334, 337, 340, 360, 378, 398 or 430 of the Fc region.
  • For example, the variant with improved FcγR binding activity may display increased binding to FcγRIII, and optionally may further display decreased binding to FcγRII; e.g. the variant may comprise an amino acid modification at position 298 and/or 333 of an Fc region.
  • Variants with increased binding to FcγRII include those comprising an amino acid modification at any one or more of amino acid positions 255, 256, 258, 267, 268, 272, 276, 280, 283, 285, 286, 290, 301, 305, 307, 309, 312, 315, 320, 322, 326, 330, 331, 337, 340, 378, 398 or 430 of an Fc region. Such variants may further display decreased binding to FcγRIII. For example, they may include an Fc region amino acid modification at any one or more of amino acid positions 268, 272, 298, 301, 322 or 340.
  • While it is preferred to alter binding to a FcγR, Fc region variants with altered binding affinity for the neonatal receptor (FcRn) are also contemplated herein. Fc region variants with improved affinity for FcRn are anticipated to have longer serum half-lives, and such molecules will have useful applications in methods of treating mammals where long half-life of the administered polypeptide is desired, e.g., to treat a chronic disease or disorder. Fc region variants with decreased FcRn binding affinity, on the contrary, are expected to have shorter half-lives, and such molecules may, for example, be administered to a mammal where a shortened circulation time may be advantageous, e.g. for in vivo diagnostic imaging or for polypeptides which have toxic side effects when left circulating in the blood stream for extended periods, etc. Fc region variants with decreased FcRn binding affinity are anticipated to be less likely to cross the placenta, and thus may be utilized in the treatment of diseases or disorders in pregnant women.
  • Fc region variants with altered binding affinity for FcRn include those comprising an Fc region amino acid modification at any one or more of amino acid positions 238, 252, 253, 254, 255, 256, 265, 272, 286, 288, 303, 305, 307, 309, 311, 312, 317, 340, 356, 360, 362, 376, 378, 380, 382, 386, 388, 400, 413, 415, 424, 433, 434, 435, 436, 439 or 447. Those which display reduced binding to FcRn will generally comprise an Fc region amino acid modification at any one or more of amino acid positions 252, 253, 254, 255, 288, 309, 386, 388, 400, 415, 433, 435, 436, 439 or 447; and those with increased binding to FcRn will usually comprise an Fc region amino acid modification at any one or more of amino acid positions 238, 256, 265, 272, 286, 303, 305, 307, 311, 312, 317, 340, 356, 360, 362, 376, 378, 380, 382, 413, 424 or 434.
  • The polypeptide variant(s) prepared as described above may be subjected to further modifications, oftentimes depending on the intended use of the polypeptide. Such modifications may involve further alteration of the amino acid sequence (substitution, insertion and/or deletion of amino acid residues), fusion to heterologous polypeptide(s) and/or covalent modifications. Such “further modifications” may be made prior to, simultaneously with, or following, the amino acid modification(s) disclosed above which result in an alteration of Fc receptor binding and/or ADCC activity. In one embodiment, one may combine the Fc region modification herein with Fc region substitutions disclosed in the references cited in the “Related Art” section of this application.
  • Alternatively or additionally, it may be useful to combine the above amino acid modifications with one or more further amino acid modifications that alter C1q binding and/or complement dependent cytoxicity function of the Fc region.
  • The starting polypeptide of particular interest herein is usually one that binds to C1q and displays complement dependent cytotoxicity (CDC). The further amino acid substitutions described herein will generally serve to alter the ability of the starting polypeptide to bind to C1q and/or modify its complement dependent cytotoxicity function, e.g. to reduce and preferably abolish these effector functions. However, polypeptides comprising substitutions at one or more of the described positions with improved C1q binding and/or complement dependent cytotoxicity (CDC) function are contemplated herein. For example, the starting polypeptide may be unable to bind C1q and/or mediate CDC and may be modified according to the teachings herein such that it acquires these further effector functions. Moreover, polypeptides with pre-existing C1q binding activity, optionally further having the ability to mediate CDC may be modified such that one or both of these activities are enhanced.
  • To generate an Fc region with altered C1q binding and/or complement dependent cytotoxicity (CDC) function, the amino acid positions to be modified are generally selected from heavy chain positions 270, 322, 326, 327, 329, 331, 333, and 334, where the numbering of the residues in an IgG heavy chain is that of the EU index as in Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md. (1991). In one embodiment, only one of the eight above-identified positions is altered in order to generate the polypeptide variant region with altered C1q binding and/or complement dependent cytotoxicity (CDC) function. Preferably only residue 270, 329 or 322 is altered if this is the case. Alternatively, two or more of the above-identified positions are modified. If substitutions are to be combined, generally substitutions which enhance human C1q binding (e.g. at residue positions 326, 327, 333 and 334) or those which diminish human C1q binding (e.g., at residue positions 270, 322, 329 and 331) are combined. In the latter embodiment, all four positions (i.e., 270, 322, 329 and 331) may be substituted. Preferably, further substitutions at two, three or all of positions 326, 327, 333 or 334 are combined, optionally with other Fc region substitutions, to generate a polypeptide with improved human C1q binding and preferably improved CDC activity in vitro or in vivo.
  • Proline is conserved at position 329 in human IgG's. This residue is preferably replaced with alanine, however substitution with any other amino acid is contemplated, e.g., serine, threonine, asparagine, glycine or valine.
  • Proline is conserved at position 331 in human IgG1, IgG2 and IgG3, but not IgG4 (which has a serine residue at position 331). Residue 331 is preferably replaced by alanine or another amino acid, e.g. serine (for IgG regions other than IgG4), glycine or valine.
  • Lysine 322 is conserved in human IgGs, and this residue is preferably replaced by an alanine residue, but substitution with any other amino acid residue is contemplated, e.g. serine, threonine, glycine or valine.
  • D270 is conserved in human IgGs, and this residue may be replaced by another amino acid residue, e.g. alanine, serine, threonine, glycine, valine, or lysine.
  • K326 is also conserved in human IgGs. This residue may be substituted with another residue including, but not limited to, valine, glutamic acid, alanine, glycine, aspartic acid, methionine or tryptophan, with tryptophan being preferred.
  • Likewise, E333 is also conserved in human IgGs. E333 is preferably replaced by an amino acid residue with a smaller side chain volume, such as valine, glycine, alanine or serine, with serine being preferred.
  • K334 is conserved in human IgGs and may be substituted with another residue such as alanine or other residue.
  • In human IgG1 and IgG3, residue 327 is an alanine. In order to generate a variant with improved C1q binding, this alanine may be substituted with another residue such as glycine. In IgG2 and IgG4, residue 327 is a glycine and this may be replaced by alanine (or another residue) to diminish C1q binding.
  • As disclosed above, one can design an Fc region with altered effector function, e.g., by modifying C1q binding and/or FcR binding and thereby changing CDC activity and/or ADCC activity. For example, one can generate a variant Fc region with improved C1q binding and improved FcγRIII binding; e.g. having both improved ADCC activity and improved CDC activity. Alternatively, where one desires that effector function be reduced or ablated, one may engineer a variant Fc region with reduced CDC activity and/or reduced ADCC activity. In other embodiments, one may increase only one of these activities, and optionally also reduce the other activity, e.g. to generate an Fc region variant with improved ADCC activity, but reduced CDC activity and vice versa.
  • With respect to further amino acid sequence alterations, any cysteine residue not involved in maintaining the proper conformation of the polypeptide variant also may be substituted, generally with serine, to improve the oxidative stability of the molecule and prevent aberrant cross linking.
  • Another type of amino acid substitution serves to alter the glycosylation pattern of the polypeptide. This may be achieved by deleting one or more carbohydrate moieties found in the polypeptide, and/or adding one or more glycosylation sites that are not present in the polypeptide. Glycosylation of polypeptides is typically either N-linked or O-linked. N-linked refers to the attachment of the carbohydrate moiety to the side chain of an asparagine residue. The tripeptide sequences asparagine-X-serine and asparagine-X-threonine, where X is any amino acid except proline, are the recognition sequences for enzymatic attachment of the carbohydrate moiety to the asparagine side chain. Thus, the presence of either of these tripeptide sequences in a polypeptide creates a potential glycosylation site. O-linked glycosylation refers to the attachment of one of the sugars N-aceylgalactosamine, galactose, or xylose to a hydroxyamino acid, most commonly serine or threonine, although 5-hydroxyproline or 5-hydroxylysine may also be used. Addition of glycosylation sites to the polypeptide is conveniently accomplished by altering the amino acid sequence such that it contains one or more of the above-described tripeptide sequences (for N-linked glycosylation sites). The alteration may also be made by the addition of, or substitution by, one or more serine or threonine residues to the sequence of the original polypeptide (for O-linked glycosylation sites). An exemplary glycosylation variant has an amino acid substitution of residue Asn 297 of the heavy chain.
  • Moreover, the class, subclass or allotype of the Fc region may be altered by one or more further amino acid substitutions to generate an Fc region with an amino acid sequence more homologous to a different class, subclass or allotype as desired. For example, a murine Fc region may be altered to generate an amino acid sequence more homologous to a human Fc region; a human non-A allotype IgG1 Fc region may be modified to achieve a human A allotype IgG1 Fc region etc. In one embodiment, the amino modification(s) herein which alter FcR binding and/or ADCC activity are made in the CH2 domain of the Fc region and the CH3 domain is deleted or replaced with another dimerization domain. Preferably, however, the CH3 domain is retained (aside from amino acid modifications therein which alter effector function as herein disclosed).
  • The polypeptide variant may be subjected to one or more assays to evaluate any change in biological activity compared to the starting polypeptide.
  • Preferably the polypeptide variant essentially retains the ability to bind antigen compared to the nonvariant polypeptide, i.e. the binding capability is no worse than about 20 fold, e.g. no worse than about 5 fold of that of the nonvariant polypeptide. The binding capability of the polypeptide variant may be determined using techniques such as fluorescence activated cell sorting (FACS) analysis or radioimmunoprecipitation (RIA), for example.
  • The ability of the polypeptide variant to bind an FcR may be evaluated. Where the FcR is a high affinity Fc receptor, such as FcγRI, FcRn or FcγRIIIA-V158, binding can be measured by titrating monomeric polypeptide variant and measuring bound polypeptide variant using an antibody which specifically binds to the polypeptide variant in a standard ELISA format (see Example 2 below). Another FcR binding assay for low affinity FcRs is described in Examples 1 and 4.
  • To assess ADCC activity of the polypeptide variant, an in vitro ADCC assay, such as that described in Example 4 may be performed using varying effector:target ratios. Useful “effector cells” for such assays include peripheral blood mononuclear cells (PBMC) and Natural Killer (NK) cells. Alternatively, or additionally, ADCC activity of the polypeptide variant may be assessed in vivo, e.g., in a animal model such as that disclosed in Clynes et al., PNAS (USA) 95:652-656 (1998).
  • The ability of the variant to bind C1q and mediate complement dependent cytotoxicity (CDC) may be assessed.
  • To determine C1q binding, a C1q binding ELISA may be performed. Briefly, assay plates may be coated overnight at 4° C. with polypeptide variant or starting polypeptide (control) in coating buffer. The plates may then be washed and blocked. Following washing, an aliquot of human C1q may be added to each well and incubated for 2 hrs at room temperature. Following a further wash, 100μl of a sheep anti-complement C1q peroxidase conjugated antibody may be added to each well and incubated for 1 hour at room temperature. The plate may again be washed with wash buffer and 100 μl of substrate buffer containing OPD (O-phenylenediamine dihydrochloride (Sigma)) may be added to each well. The oxidation reaction, observed by the appearance of a yellow color, may be allowed to proceed for 30 minutes and stopped by the addition of 100 μl of 4.5 N H2SO4. The absorbance may then read at (492-405) nm.
  • An exemplary polypeptide variant is one that displays a “significant reduction in C1q binding” in this assay. This means that about 100 μg/ml of the polypeptide variant displays about 50 fold or more reduction in C1q binding compared to 100 μg/ml of a control antibody having a nonmutated IgG1 Fc region. In the most preferred embodiment, the polypeptide variant “does not bind C1q”, i.e. 100 μg/ml of the polypeptide variant displays about 100 fold or more reduction in C1q binding compared to 100 μg/ml of the control antibody.
  • Another exemplary variant is one which “has a better binding affinity for human C1q than the parent polypeptide”. Such a molecule may display, for example, about two-fold or more, and preferably about five-fold or more, improvement in human C1q binding compared to the parent polypeptide (e.g. at the IC50 values for these two molecules). For example, human C1q binding may be about two-fold to about 500-fold, and preferably from about two-fold or from about five-fold to about 1000-fold improved compared to the parent polypeptide.
  • To assess complement activation, a complement dependent cytotoxicity (CDC) assay may be performed, e.g. as described in Gazzano-Santoro et al., J. Immunol. Methods 202:163-171 (1997). Briefly, various concentrations of the polypeptide variant and human complement may be diluted with buffer. Cells which express the antigen to which the polypeptide variant binds may be diluted to a density of ˜1×106 cells/ml. Mixtures of polypeptide variant, diluted human complement and cells expressing the antigen may be added to a flat bottom tissue culture 96 well plate and allowed to incubate for 2 hrs at 37° C. and 5% CO2 to facilitate complement mediated cell lysis. 50 μl of alamar blue (Accumed International) may then be added to each well and incubated overnight at 37° C. The absorbance is measured using a 96-well fluorometer with excitation at 530 nm and emission at 590 nm. The results may be expressed in relative fluorescence units (RFU). The sample concentrations may be computed from a standard curve and the percent activity as compared to nonvariant polypeptide is reported for the polypeptide variant of interest.
  • Yet another exemplary variant “does not activate complement”. For example, 0.6 μg/ml of the polypeptide variant displays about 0-10% CDC activity in this assay compared to a 0.6 μg/ml of a control antibody having a nonmutated IgG1 Fc region. Preferably the variant does not appear to have any CDC activity in the above CDC assay.
  • The invention also pertains to a polypeptide variant with enhanced CDC compared to a parent polypeptide, e.g., displaying about two-fold to about 100-fold improvement in CDC activity in vitro or in vivo (e.g. at the IC50 values for each molecule being compared).
  • A. Receptor Binding Assay and Immune Complex
  • A receptor binding assay has been developed herein which is particularly useful for determining binding of an analyte of interest to a receptor where the affinity of the analyte for the receptor is relatively weak, e.g. in the micromolar range as is the case for FcγRIIA, FcγRIIB, FcγRIIIA and FcγRIIIB. The method involves the formation of a molecular complex that has an improved avidity for the receptor of interest compared to the noncomplexed analyte. The preferred molecular complex is an immune complex comprising: (a) an Fc region-containing polypeptide (such as an antibody or an immunoadhesin); (b) a first target molecule which comprises at least two binding sites for the Fc region-containing polypeptide; and (c) a second target molecule which comprises at least two binding sites for the first target molecule.
  • In Example 1 below, the Fc region-containing polypeptide is an anti-IgE antibody, such as the E27 antibody (FIGS. 4A-4B). E27, when mixed with human IgE at an 1:1 molar ratio, forms a stable hexamer consisting of three E27 molecules and three IgE molecules. In Example 1 below, the “first target molecule” is a chimeric form of IgE in which the Fab portion of an anti-VEGF antibody is fused to the human IgE Fc portion and the “second target molecule” is the antigen to which the Fab binds (i.e. VEGF). Each molecule of IgE binds two molecules of VEGF. VEGF also binds two molecules of IgE per molecule of VEGF. When recombinant human VEGF was added at a 2:1 molar ratio to IgE:E27 hexamers, the hexamers were linked into larger molecular weight complexes via the IgE:VEGF interaction (FIG. 5). The Fc region of the anti-IgE antibody of the resultant immune complex binds to FcR with higher avidity than either uncomplexed anti-IgE or anti-IgE:IgE hexamers.
  • Other forms of molecular complexes for use in the receptor assay are contemplated. Examples comprising only an Fc region-containing polypeptide:first target molecule combination include an immunoadhesin:ligand combination such as VEGF receptor (KDR)-immunoadhesin:VEGF and a full-length bispecific antibody (bsAb):first target molecule. A further example of an Fc region-containing polypeptide:first target molecule:second target molecule combination include a nonblocking antibody:soluble receptor:ligand combination such as anti-Trk antibody:soluble Trk receptor:neurotrophin (Urfer et al. J. Biol. Chem. 273(10):5829-5840 (1998)).
  • Aside from use in a receptor binding assay, the immune complexes described above have further uses including evaluation of Fc region-containing polypeptide function and immune complex clearance in vivo. Hence, the immune complex may be administered to a mammal (e.g. in a pre-clinical animal study) and evaluated for its half-life etc.
  • To determine receptor binding, a polypeptide comprising at least the binding domain of the receptor of interest (e.g. the extracellular domain of an α subunit of an FcR) may be coated on solid phase, such as an assay plate. The binding domain of the receptor alone or a receptor-fusion protein may be coated on the plate using standard procedures. Examples of receptor-fusion proteins include receptor-glutathione S-transferase (GST) fusion protein, receptor-chitin binding domain fusion protein, receptor-hexaHis tag fusion protein (coated on glutathione, chitin, and nickel coated plates, respectively). Alternatively, a capture molecule may be coated on the assay plate and used to bind the receptor-fusion protein via the non-receptor portion of the fusion protein. Examples include anti-hexaHis F(ab′)2 coated on the assay plate used to capture receptor-hexaHis tail fusion or anti-GST antibody coated on the assay plate used to capture a receptor-GST fusion. In other embodiments, binding to cells expressing at least the binding domain of the receptor may be evaluated. The cells may be naturally occurring hematopoietic cells that express the FcR of interest or may be transformed with nucleic acid encoding the FcR or a binding domain thereof such that the binding domain is expressed at the surface of the cell to be tested.
  • The immune complex described hereinabove is added to the receptor-coated plates and incubated for a sufficient period of time such that the analyte binds to the receptor. Plates may then be washed to remove unbound complexes, and binding of the analyte may be detected according to known methods. For example, binding may be detected using a reagent (e.g. an antibody or fragment thereof) which binds specifically to the analyte, and which is optionally conjugated with a detectable label (detectable labels and methods for conjugating them to polypeptides are described below in the section entitled “Non-Therapeutic Uses for the Polypeptide Variant”).
  • As a matter of convenience, the reagents can be provided in an assay kit, i.e., a packaged combination of reagents, for combination with the analyte in assaying the ability of the analyte to bind to a receptor of interest. The components of the kit will generally be provided in predetermined ratios. The kit may provide the first target molecule and/or the second target molecule, optionally complexed together. The kit may further include assay plates coated with the receptor or a binding domain thereof (e.g. the extracellular domain of the α subunit of an FcR). Usually, other reagents, such as an antibody that binds specifically to the analyte to be assayed, labeled directly or indirectly with an enzymatic label, will also be provided in the kit. Where the detectable label is an enzyme, the kit will include substrates and cofactors required by the enzyme (e.g. a substrate precursor which provides the detectable chromophore or fluorophore). In addition, other additives may be included such as stabilizers, buffers (e.g. assay and/or wash lysis buffer) and the like. The relative amounts of the various reagents may be varied widely to provide for concentrations in solution of the reagents that substantially optimize the sensitivity of the assay. Particularly, the reagents may be provided as dry powders, usually lyophilized, including excipients that on dissolution will provide a reagent solution having the appropriate concentration. The kit also suitably includes instructions for carrying out the assay.
  • B. Antibody Preparation
  • In the preferred embodiment of the invention, the Fc region-containing polypeptide which is modified according to the teachings herein is an antibody. Techniques for producing antibodies follow:
  • Antigen Selection and Preparation
  • Where the polypeptide is an antibody, it is directed against an antigen of interest. Preferably, the antigen is a biologically important polypeptide and administration of the antibody to a mammal suffering from a disease or disorder can result in a therapeutic benefit in that mammal. However, antibodies directed against nonpolypeptide antigens (such as tumor-associated glycolipid antigens; see U.S. Pat. No. 5,091,178) are also contemplated.
  • Where the antigen is a polypeptide, it may be a transmembrane molecule (e.g. receptor) or ligand such as a growth factor. Exemplary antigens include molecules such as renin; a growth hormone, including human growth hormone and bovine growth hormone; growth hormone releasing factor; parathyroid hormone; thyroid stimulating hormone; lipoproteins; alpha-1-antitrypsin; insulin A-chain; insulin B-chain; proinsulin; follicle stimulating hormone; calcitonin; luteinizing hormone; glucagon; clotting factors such as factor VIIIC, factor IX, tissue factor (TF), and von Willebrands factor; anti-clotting factors such as Protein C; atrial natriuretic factor; lung surfactant; a plasminogen activator, such as urokinase or human urine or tissue-type plasminogen activator (t-PA); bombesin; thrombin; hemopoietic growth factor; tumor necrosis factor-alpha and -beta; enkephalinase; RANTES (regulated on activation normally T-cell expressed and secreted); human macrophage inflammatory protein (MIP-1-alpha); a serum albumin such as human serum albumin; Muellerian-inhibiting substance; relaxin A-chain; relaxin B-chain; prorelaxin; mouse gonadotropin-associated peptide; a microbial protein, such as beta-lactamase; DNase; IgE; a cytotoxic T-lymphocyte associated antigen (CTLA), such as CTLA-4; inhibin; activin; vascular endothelial growth factor (VEGF); receptors for hormones or growth factors; protein A or D; rheumatoid factors; a neurotrophic factor such as bone-derived neurotrophic factor (BDNF), neurotrophin-3, -4, -5, or -6 (NT-3, NT-4, NT-5, or NT-6), or a nerve growth factor such as NGF-β; platelet-derived growth factor (PDGF); fibroblast growth factor such as aFGF and bFGF; epidermal growth factor (EGF); transforming growth factor (TGF) such as TGF-alpha and TGF-beta, including TGF-β1, TGF-β2, TGF-β3, TGF-β4, or TGF-β5; insulin-like growth factor-I and -II (IGF-I and IGF-II); des(1-3)-IGF-I (brain IGF-I), insulin-like growth factor binding proteins; CD proteins such as CD3, CD4, CD8, CD19 and CD20; erythropoietin; osteoinductive factors; immunotoxins; a bone morphogenetic protein (BMP); an interferon such as interferon-alpha, -beta, and -gamma; colony stimulating factors (CSFs), e.g., M-CSF, GM-CSF, and G-CSF; interleukins (ILs), e.g., IL-1 to IL-10; superoxide dismutase; T-cell receptors; surface membrane proteins; decay accelerating factor; viral antigen such as, for example, a portion of the AIDS envelope; transport proteins; homing receptors; addressins; regulatory proteins; integrins such as CD11a, CD11b, CD11c, CD18, an ICAM, VLA-4 and VCAM; a tumor associated antigen such as HER2, HER3 or HER4 receptor; and fragments of any of the above-listed polypeptides.
  • Preferred molecular targets for antibodies encompassed by the present invention include CD proteins such as CD3, CD4, CD8, CD19, CD20 and CD34; members of the ErbB receptor family such as the EGF receptor, HER2, HER3 or HER4 receptor; cell adhesion molecules such as LFA-1, Mac1, p 150.95, VLA-4, ICAM-1, VCAM, α4/β7 integrin, and αv/β3 integrin including either α or β subunits thereof (e.g. anti-CD11a, anti-CD18 or anti-CD11b antibodies); growth factors such as VEGF; tissue factor (TF); alpha interferon (α-IFN); an interleukin, such as IL-8; IgE; blood group antigens; flk2/flt3 receptor; obesity (OB) receptor; mpl receptor; CTLA-4; protein C etc.
  • Soluble antigens or fragments thereof, optionally conjugated to other molecules, can be used as immunogens for generating antibodies. For transmembrane molecules, such as receptors, fragments of these (e.g. the extracellular domain of a receptor) can be used as the immunogen. Alternatively, cells expressing the transmembrane molecule can be used as the immunogen. Such cells can be derived from a natural source (e.g. cancer cell lines) or may be cells which have been transformed by recombinant techniques to express the transmembrane molecule. Other antigens and forms thereof useful for preparing antibodies will be apparent to those in the art.
  • (ii) Polyclonal Antibodies
  • Polyclonal antibodies are preferably raised in animals by multiple subcutaneous (sc) or intraperitoneal (ip) injections of the relevant antigen and an adjuvant. It may be useful to conjugate the relevant antigen to a protein that is immunogenic in the species to be immunized, e.g., keyhole limpet hemocyanin, serum albumin, bovine thyroglobulin, or soybean trypsin inhibitor using a bifunctional or derivatizing agent, for example, maleimidobenzoyl sulfosuccinimide ester (conjugation through cysteine residues), N-hydroxysuccinimide (through lysine residues), glutaraldehyde, succinic anhydride, SOCl2, or R1N═C═NR, where R and R1 are different alkyl groups.
  • Animals are immunized against the antigen, immunogenic conjugates, or derivatives by combining, e.g., 100 μg or 5 μg of the protein or conjugate (for rabbits or mice, respectively) with 3 volumes of Freund's complete adjuvant and injecting the solution intradermally at multiple sites. One month later the animals are boosted with ⅕ to 1/10 the original amount of peptide or conjugate in Freund's complete adjuvant by subcutaneous injection at multiple sites. Seven to 14 days later the animals are bled and the serum is assayed for antibody titer. Animals are boosted until the titer plateaus. Preferably, the animal is boosted with the conjugate of the same antigen, but conjugated to a different protein and/or through a different cross-linking reagent. Conjugates also can be made in recombinant cell culture as protein fusions. Also, aggregating agents such as alum are suitably used to enhance the immune response.
  • (iii) Monoclonal Antibodies
  • Monoclonal antibodies may be made using the hybridoma method first described by Kohler et al., Nature, 256:495 (1975), or may be made by recombinant DNA methods (U.S. Pat. No. 4,816,567).
  • In the hybridoma method, a mouse or other appropriate host animal, such as a hamster or macaque monkey, is immunized as hereinabove described to elicit lymphocytes that produce or are capable of producing antibodies that will specifically bind to the protein used for immunization. Alternatively, lymphocytes may be immunized in vitro. Lymphocytes then are fused with myeloma cells using a suitable fusing agent, such as polyethylene glycol, to form a hybridoma cell (Goding, Monoclonal Antibodies: Principles and Practice, pp. 59-103 (Academic Press, 1986)).
  • The hybridoma cells thus prepared are seeded and grown in a suitable culture medium that preferably contains one or more substances that inhibit the growth or survival of the unfused, parental myeloma cells. For example, if the parental myeloma cells lack the enzyme hypoxanthine guanine phosphoribosyl transferase (HGPRT or HPRT), the culture medium for the hybridomas typically will include hypoxanthine, aminopterin, and thymidine (HAT medium), which substances prevent the growth of HGPRT-deficient cells.
  • Preferred myeloma cells are those that fuse efficiently, support stable high-level production of antibody by the selected antibody-producing cells, and are sensitive to a medium such as HAT medium. Among these, preferred myeloma cell lines are murine myeloma lines, such as those derived from MOPC-21 and MPC-11 mouse tumors available from the Salk Institute Cell Distribution Center, San Diego, Calif. USA, and SP-2 or X63-Ag8-653 cells available from the American Type Culture Collection, Rockville, Md. USA. Human myeloma and mouse-human heteromyeloma cell lines also have been described for the production of human monoclonal antibodies (Kozbor, J. Immunol., 133:3001 (1984); Brodeur et al., Monoclonal Antibody Production Techniques and Applications, pp. 51-63 (Marcel Dekker, Inc., New York, 1987)).
  • Culture medium in which hybridoma cells are growing is assayed for production of monoclonal antibodies directed against the antigen. Preferably, the binding specificity of monoclonal antibodies produced by hybridoma cells is determined by immunoprecipitation or by an in vitro binding assay, such as radioimmunoassay (RIA) or enzyme-linked immunoabsorbent assay (ELISA).
  • After hybridoma cells are identified that produce antibodies of the desired specificity, affinity, and/or activity, the clones may be subcloned by limiting dilution procedures and grown by standard methods (Goding, Monoclonal Antibodies: Principles and Practice, pp. 59-103 (Academic Press, 1986)). Suitable culture media for this purpose include, for example, D-MEM or RPMI-1640 medium. In addition, the hybridoma cells may be grown in vivo as ascites tumors in an animal.
  • The monoclonal antibodies secreted by the subclones are suitably separated from the culture medium, ascites fluid, or serum by conventional immunoglobulin purification procedures such as, for example, protein A-Sepharose, hydroxylapatite chromatography, gel electrophoresis, dialysis, or affinity chromatography.
  • DNA encoding the monoclonal antibodies is readily isolated and sequenced using conventional procedures (e.g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of the monoclonal antibodies). The hybridoma cells serve as a preferred source of such DNA. Once isolated, the DNA may be placed into expression vectors, which are then transfected into host cells such as E. coli cells, simian COS cells, Chinese hamster ovary (CHO) cells, or myeloma cells that do not otherwise produce immunoglobulin protein, to obtain the synthesis of monoclonal antibodies in the recombinant host cells. Recombinant production of antibodies will be described in more detail below.
  • In a further embodiment, antibodies or antibody fragments can be isolated from antibody phage libraries generated using the techniques described in McCafferty et al., Nature, 348:552-554 (1990). Clackson et al., Nature, 352:624-628 (1991) and Marks et al., J. Mol. Biol., 222:581-597 (1991) describe the isolation of murine and human antibodies, respectively, using phage libraries. Subsequent publications describe the production of high affinity (nM range) human antibodies by chain shuffling (Marks et al., Bio/Technology, 10:779-783 (1992)), as well as combinatorial infection and in vivo recombination as a strategy for constructing very large phage libraries (Waterhouse et al., Nuc. Acids. Res., 21:2265-2266 (1993)). Thus, these techniques are viable alternatives to traditional monoclonal antibody hybridoma techniques for isolation of monoclonal antibodies.
  • The DNA also may be modified, for example, by substituting the coding sequence for human heavy- and light-chain constant domains in place of the homologous murine sequences (U.S. Pat. No. 4,816,567; Morrison, et al., Proc. Natl Acad. Sci. USA, 81:6851 (1984)), or by covalently joining to the immunoglobulin coding sequence all or part of the coding sequence for a non-immunoglobulin polypeptide.
  • Typically such non-immunoglobulin polypeptides are substituted for the constant domains of an antibody, or they are substituted for the variable domains of one antigen-combining site of an antibody to create a chimeric bivalent antibody comprising one antigen-combining site having specificity for an antigen and another antigen-combining site having specificity for a different antigen.
  • (iv) Humanized and Human Antibodies
  • A humanized antibody has one or more amino acid residues introduced into it from a source which is non-human. These non-human amino acid residues are often referred to as “import” residues, which are typically taken from an “import” variable domain. Humanization can be essentially performed following the method of Winter and co-workers (Jones et al., Nature, 321:522-525 (1986); Riechmann et al., Nature, 332:323-327 (1988); Verhoeyen et al., Science, 239:1534-1536 (1988)), by substituting rodent CDRs or CDR sequences for the corresponding sequences of a human antibody. Accordingly, such “humanized” antibodies are chimeric antibodies (U.S. Pat. No. 4,816,567) wherein substantially less than an intact human variable domain has been substituted by the corresponding sequence from a non-human species. In practice, humanized antibodies are typically human antibodies in which some CDR residues and possibly some FR residues are substituted by residues from analogous sites in rodent antibodies.
  • The choice of human variable domains, both light and heavy, to be used in making the humanized antibodies is very important to reduce antigenicity. According to the so-called “best-fit” method, the sequence of the variable domain of a rodent antibody is screened against the entire library of known human variable-domain sequences. The human sequence which is closest to that of the rodent is then accepted as the human framework (FR) for the humanized antibody (Sims et al., J. Immunol, 151:2296 (1993); Chothia et al., J. Mol. Biol., 196:901 (1987)). Another method uses a particular framework derived from the consensus sequence of all human antibodies of a particular subgroup of light or heavy chains. The same framework may be used for several different humanized antibodies (Carter et al., Proc. Natl. Acad. Sci. USA, 89:4285 (1992); Presta et al., J. Immunol., 151:2623 (1993)).
  • It is further important that antibodies be humanized with retention of high affinity for the antigen and other favorable biological properties. To achieve this goal, according to a preferred method, humanized antibodies are prepared by a process of analysis of the parental sequences and various conceptual humanized products using three-dimensional models of the parental and humanized sequences. Three-dimensional immunoglobulin models are commonly available and are familiar to those skilled in the art. Computer programs are available which illustrate and display probable three-dimensional conformational structures of selected candidate immunoglobulin sequences. Inspection of these displays permits analysis of the likely role of the residues in the functioning of the candidate immunoglobulin sequence, i.e., the analysis of residues that influence the ability of the candidate immunoglobulin to bind its antigen. In this way, FR residues can be selected and combined from the recipient and import sequences so that the desired antibody characteristic, such as increased affinity for the target antigen(s), is achieved. In general, the CDR residues are directly and most substantially involved in influencing antigen binding.
  • Alternatively, it is now possible to produce transgenic animals (e.g., mice) that are capable, upon immunization, of producing a full repertoire of human antibodies in the absence of endogenous immunoglobulin production. For example, it has been described that the homozygous deletion of the antibody heavy-chain joining region (JH) gene in chimeric and germ-line mutant mice results in complete inhibition of endogenous antibody production. Transfer of the human germ-line immunoglobulin gene array in such germ-line mutant mice will result in the production of human antibodies upon antigen challenge. See, e.g., Jakobovits et al., Proc. Natl. Acad. Sci. USA, 90:2551 (1993); Jakobovits et al., Nature, 362:255-258 (1993); Bruggermann et al., Year in Immuno., 7:33 (1993); and Duchosal et al., Nature 355:258 (1992). Human antibodies can also be derived from phage-display libraries (Hoogenboom et al., J. Mol. Biol., 227:381 (1991); Marks et al., J. Mol. Biol., 222:581-597 (1991); Vaughan et al. Nature Biotech 14:309 (1996)).
  • (v) Multispecific Antibodies
  • Multispecific antibodies have binding specificities for at least two different antigens. While such molecules normally will only bind two antigens (i.e. bispecific antibodies, BsAbs), antibodies with additional specificities such as trispecific antibodies are encompassed by this expression when used herein. Examples of BsAbs include those with one arm directed against a tumor cell antigen and the other arm directed against a cytotoxic trigger molecule such as anti-FcγRI/anti-CD15, anti-p185HER2/FcγRIII (CD16), anti-CD3/anti-malignant B-cell (1D10), anti-CD3/anti-p185HER2, anti-CD3/anti-p97, anti-CD3/anti-renal cell carcinoma, anti-CD3/anti-OVCAR-3, anti-CD3/L-D1 (anti-colon carcinoma), anti-CD3/anti-melanocyte stimulating hormone analog, anti-EGF receptor/anti-CD3, anti-CD3/anti-CAMA1, anti-CD3/anti-CD19, anti-CD3/MoV18, anti-neural cell ahesion molecule (NCAM)/anti-CD3, anti-folate binding protein (FBP)/anti-CD3, anti-pan carcinoma associated antigen (AMOC-31)/anti-CD3; BsAbs with one arm which binds specifically to a tumor antigen and one arm which binds to a toxin such as anti-saporin/anti-Id-1, anti-CD22/anti-saporin, anti-CD7/anti-saporin, anti-CD38/anti-saporin, anti-CEA/anti-ricin A chain, anti-interferon-α (IFN-α)/anti-hybridoma idiotype, anti-CEA/anti-vinca alkaloid; BsAbs for converting enzyme activated prodrugs such as anti-CD30/anti-alkaline phosphatase (which catalyzes conversion of mitomycin phosphate prodrug to mitomycin alcohol); BsAbs which can be used as fibrinolytic agents such as anti-fibrin/anti-tissue plasminogen activator (tPA), anti-fibrin/anti-urokinase-type plasminogen activator (uPA); BsAbs for targeting immune complexes to cell surface receptors such as anti-low density lipoprotein (LDL)/anti-Fc receptor (e.g. FcγRI, FcγRII or FcγRIII); BsAbs for use in therapy of infectious diseases such as anti-CD3/anti-herpes simplex virus (HSV), anti-T-cell receptor:CD3 complex/anti-influenza, anti-FcγR/anti-HIV; BsAbs for tumor detection in vitro or in vivo such as anti-CEA/anti-EOTUBE, anti-CEA/anti-DPTA, anti-p185HER2/anti-hapten; BsAbs as vaccine adjuvants; and BsAbs as diagnostic tools such as anti-rabbit IgG/anti-ferritin, anti-horse radish peroxidase (HRP)/anti-hormone, anti-somatostatin/anti-substance P, anti-HRP/anti-FITC, anti-CEA/anti-β-galactosidase. Examples of trispecific antibodies include anti-CD3/anti-CD4/anti-CD37, anti-CD3/anti-CD5/anti-CD37 and anti-CD3/anti-CD8/anti-CD37. Bispecific antibodies can be prepared as full length antibodies or antibody fragments (e.g. F(ab′)2 bispecific antibodies).
  • Methods for making bispecific antibodies are known in the art. Traditional production of full length bispecific antibodies is based on the coexpression of two immunoglobulin heavy chain-light chain pairs, where the two chains have different specificities (Millstein et al., Nature, 305:537-539 (1983)). Because of the random assortment of immunoglobulin heavy and light chains, these hybridomas (quadromas) produce a potential mixture of 10 different antibody molecules, of which only one has the correct bispecific structure. Purification of the correct molecule, which is usually done by affinity chromatography steps, is rather cumbersome, and the product yields are low. Similar procedures are disclosed in WO 93/08829, and in Traunecker et al., EMBO J., 10:3655-3659 (1991).
  • According to a different approach, antibody variable domains with the desired binding specificities (antibody-antigen combining sites) are fused to immunoglobulin constant domain sequences. The fusion preferably is with an immunoglobulin heavy chain constant domain, comprising at least part of the hinge, CH2, and CH3 regions. It is preferred to have the first heavy-chain constant region (CH1) containing the site necessary for light chain binding, present in at least one of the fusions. DNAs encoding the immunoglobulin heavy chain fusions and, if desired, the immunoglobulin light chain, are inserted into separate expression vectors, and are co-transfected into a suitable host organism. This provides for great flexibility in adjusting the mutual proportions of the three polypeptide fragments in embodiments when unequal ratios of the three polypeptide chains used in the construction provide the optimum yields. It is, however, possible to insert the coding sequences for two or all three polypeptide chains in one expression vector when the expression of at least two polypeptide chains in equal ratios results in high yields or when the ratios are of no particular significance.
  • In a preferred embodiment of this approach, the bispecific antibodies are composed of a hybrid immunoglobulin heavy chain with a first binding specificity in one arm, and a hybrid immunoglobulin heavy chain-light chain pair (providing a second binding specificity) in the other arm. It was found that this asymmetric structure facilitates the separation of the desired bispecific compound from unwanted immunoglobulin chain combinations, as the presence of an immunoglobulin light chain in only one half of the bispecific molecule provides for a facile way of separation. This approach is disclosed in WO 94/04690. For further details of generating bispecific antibodies see, for example, Suresh et al., Methods in Enzymology, 121:210 (1986). According to another approach described in WO96/27011, the interface between a pair of antibody molecules can be engineered to maximize the percentage of heterodimers which are recovered from recombinant cell culture. The preferred interface comprises at least a part of the C H3 domain of an antibody constant domain. In this method, one or more small amino acid side chains from the interface of the first antibody molecule are replaced with larger side chains (e.g. tyrosine or tryptophan). Compensatory “cavities” of identical or similar size to the large side chain(s) are created on the interface of the second antibody molecule by replacing large amino acid side chains with smaller ones (e.g. alanine or threonine). This provides a mechanism for increasing the yield of the heterodimer over other unwanted end-products such as homodimers.
  • Bispecific antibodies include cross-linked or “heteroconjugate” antibodies. For example, one of the antibodies in the heteroconjugate can be coupled to avidin, the other to biotin. Such antibodies have, for example, been proposed to target immune system cells to unwanted cells (U.S. Pat. No. 4,676,980), and for treatment of HIV infection (WO 91/00360, WO 92/200373, and EP 03089). Heteroconjugate antibodies may be made using any convenient cross-linking methods. Suitable cross-linking agents are well known in the art, and are disclosed in U.S. Pat. No. 4,676,980, along with a number of cross-linking techniques.
  • Antibodies with more than two valencies are contemplated. For example, trispecific antibodies can be prepared. Tutt et al., J. Immunol. 147: 60 (1991).
  • While the polypeptide of interest herein is preferably an antibody, other Fc region-containing polypeptides which can be modified according to the methods described herein are contemplated. An example of such a molecule is an immunoadhesin.
  • C. Immunoadhesin Preparation
  • The simplest and most straightforward immunoadhesin design combines the binding domain(s) of the adhesin (e.g. the extracellular domain (ECD) of a receptor) with the Fc region of an immunoglobulin heavy chain. Ordinarily, when preparing the immunoadhesins of the present invention, nucleic acid encoding the binding domain of the adhesin will be fused C-terminally to nucleic acid encoding the N-terminus of an immunoglobulin constant domain sequence, however N-terminal fusions are also possible.
  • Typically, in such fusions the encoded chimeric polypeptide will retain at least functionally active hinge, C H2 and C H3 domains of the constant region of an immunoglobulin heavy chain. Fusions are also made to the C-terminus of the Fc portion of a constant domain, or immediately N-terminal to the C H1 of the heavy chain or the corresponding region of the light chain. The precise site at which the fusion is made is not critical; particular sites are well known and may be selected in order to optimize the biological activity, secretion, or binding characteristics of the immunoadhesin.
  • In a preferred embodiment, the adhesin sequence is fused to the N-terminus of the Fc region of immunoglobulin G1 (IgG1). It is possible to fuse the entire heavy chain constant region to the adhesin sequence. However, more preferably, a sequence beginning in the hinge region just upstream of the papain cleavage site which defines IgG Fc chemically (i.e. residue 216, taking the first residue of heavy chain constant region to be 114), or analogous sites of other immunoglobulins is used in the fusion. In a particularly preferred embodiment, the adhesin amino acid sequence is fused to (a) the hinge region and C H2 and C H3 or (b) the C H1, hinge, C H2 and C H3 domains, of an IgG heavy chain.
  • For bispecific immunoadhesins, the immunoadhesins are assembled as multimers, and particularly as heterodimers or heterotetramers. Generally, these assembled immunoglobulins will have known unit structures. A basic four chain structural unit is the form in which IgG, IgD, and IgE exist. A four chain unit is repeated in the higher molecular weight immunoglobulins; IgM generally exists as a pentamer of four basic units held together by disulfide bonds. IgA globulin, and occasionally IgG globulin, may also exist in multimeric form in serum. In the case of multimer, each of the four units may be the same or different.
  • Various exemplary assembled immunoadhesins within the scope herein are schematically diagrammed below:
  • (a) ACL-ACL;
  • (b) ACH-(ACH, ACL-ACH, ACL-VHCH, or VLCL-ACH);
  • (c) ACL-ACH-(ACL-ACH, ACL-VHCH, VLCL-ACH, or VLCL-VHCH)
  • (d) ACL-VHCH-(ACH, or ACL-VHCH, or VLCL-ACH);
  • (e) VLCL-ACH-(ACL-VHCH, or VLCL-ACH); and
  • (f) (A-Y)n-(VLCL-VHCH)2,
  • wherein each A represents identical or different adhesin amino acid sequences;
  • VL is an immunoglobulin light chain variable domain;
  • VH is an immunoglobulin heavy chain variable domain;
  • CL is an immunoglobulin light chain constant domain;
  • CH is an immunoglobulin heavy chain constant domain;
  • n is an integer greater than 1;
  • Y designates the residue of a covalent cross-linking agent.
  • In the interests of brevity, the foregoing structures only show key features; they do not indicate joining (J) or other domains of the immunoglobulins, nor are disulfide bonds shown. However, where such domains are required for binding activity, they shall be constructed to be present in the ordinary locations which they occupy in the immunoglobulin molecules.
  • Alternatively, the adhesin sequences can be inserted between immunoglobulin heavy chain and light chain sequences, such that an immunoglobulin comprising a chimeric heavy chain is obtained. In this embodiment, the adhesin sequences are fused to the 3′ end of an immunoglobulin heavy chain in each arm of an immunoglobulin, either between the hinge and the C H2 domain, or between the C H2 and C H3 domains. Similar constructs have been reported by Hoogenboom, et al., Mol. Immunol. 28:1027-1037 (1991).
  • Although the presence of an immunoglobulin light chain is not required in the immunoadhesins of the present invention, an immunoglobulin light chain might be present either covalently associated to an adhesin-immunoglobulin heavy chain fusion polypeptide, or directly fused to the adhesin. In the former case, DNA encoding an immunoglobulin light chain is typically coexpressed with the DNA encoding the adhesin-immunoglobulin heavy chain fusion protein. Upon secretion, the hybrid heavy chain and the light chain will be covalently associated to provide an immunoglobulin-like structure comprising two disulfide-linked immunoglobulin heavy chain-light chain pairs. Methods suitable for the preparation of such structures are, for example, disclosed in U.S. Pat. No. 4,816,567, issued 28 Mar. 1989.
  • Immunoadhesins are most conveniently constructed by fusing the cDNA sequence encoding the adhesin portion in-frame to an immunoglobulin cDNA sequence. However, fusion to genomic immunoglobulin fragments can also be used (see, e.g. Aruffo et al., Cell 61:1303-1313 (1990); and Stamenkovic et al., Cell 66:1133-1144 (1991)). The latter type of fusion requires the presence of Ig regulatory sequences for expression. cDNAs encoding IgG heavy-chain constant regions can be isolated based on published sequences from cDNA libraries derived from spleen or peripheral blood lymphocytes, by hybridization or by polymerase chain reaction (PCR) techniques. The cDNAs encoding the “adhesin” and the immunoglobulin parts of the immunoadhesin are inserted in tandem into a plasmid vector that directs efficient expression in the chosen host cells.
  • D. Vectors, Host Cells and Recombinant Methods
  • The invention also provides isolated nucleic acid encoding a polypeptide variant as disclosed herein, vectors and host cells comprising the nucleic acid, and recombinant techniques for the production of the polypeptide variant.
  • For recombinant production of the polypeptide variant, the nucleic acid encoding it is isolated and inserted into a replicable vector for further cloning (amplification of the DNA) or for expression. DNA encoding the polypeptide variant is readily isolated and sequenced using conventional procedures (e.g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the polypeptide variant). Many vectors are available. The vector components generally include, but are not limited to, one or more of the following: a signal sequence, an origin of replication, one or more marker genes, an enhancer element, a promoter, and a transcription termination sequence.
  • Signal Sequence Component
  • The polypeptide variant of this invention may be produced recombinantly not only directly, but also as a fusion polypeptide with a heterologous polypeptide, which is preferably a signal sequence or other polypeptide having a specific cleavage site at the N-terminus of the mature protein or polypeptide. The heterologous signal sequence selected preferably is one that is recognized and processed (i.e., cleaved by a signal peptidase) by the host cell. For prokaryotic host cells that do not recognize and process the native polypeptide variant signal sequence, the signal sequence is substituted by a prokaryotic signal sequence selected, for example, from the group of the alkaline phosphatase, penicillinase, Ipp, or heat-stable enterotoxin II leaders. For yeast secretion the native signal sequence may be substituted by, e.g., the yeast invertase leader, a factor leader (including Saccharomyces and Kluyveromyces α-factor leaders), or acid phosphatase leader, the C. albicans glucoamylase leader, or the signal described in WO 90/13646. In mammalian cell expression, mammalian signal sequences as well as viral secretory leaders, for example, the herpes simplex gD signal, are available.
  • The DNA for such precursor region is ligated in reading frame to DNA encoding the polypeptide variant.
  • (ii) Origin of Replication Component
  • Both expression and cloning vectors contain a nucleic acid sequence that enables the vector to replicate in one or more selected host cells. Generally, in cloning vectors this sequence is one that enables the vector to replicate independently of the host chromosomal DNA, and includes origins of replication or autonomously replicating sequences. Such sequences are well known for a variety of bacteria, yeast, and viruses. The origin of replication from the plasmid pBR322 is suitable for most Gram-negative bacteria, the 2μ plasmid origin is suitable for yeast, and various viral origins (SV40, polyoma, adenovirus, VSV or BPV) are useful for cloning vectors in mammalian cells. Generally, the origin of replication component is not needed for mammalian expression vectors (the SV40 origin may typically be used only because it contains the early promoter).
  • (iii) Selection Gene Component
  • Expression and cloning vectors may contain a selection gene, also termed a selectable marker. Typical selection genes encode proteins that (a) confer resistance to antibiotics or other toxins, e.g., ampicillin, neomycin, methotrexate, or tetracycline, (b) complement auxotrophic deficiencies, or (c) supply critical nutrients not available from complex media, e.g., the gene encoding D-alanine racemase for Bacilli.
  • One example of a selection scheme utilizes a drug to arrest growth of a host cell. Those cells that are successfully transformed with a heterologous gene produce a protein conferring drug resistance and thus survive the selection regimen. Examples of such dominant selection use the drugs neomycin, mycophenolic acid and hygromycin.
  • Another example of suitable selectable markers for mammalian cells are those that enable the identification of cells competent to take up the polypeptide variant nucleic acid, such as DHFR, thymidine kinase, metallothionein-I and -II, preferably primate metallothionein genes, adenosine deaminase, ornithine decarboxylase, etc.
  • For example, cells transformed with the DHFR selection gene are first identified by culturing all of the transformants in a culture medium that contains methotrexate (Mtx), a competitive antagonist of DHFR. An appropriate host cell when wild-type DHFR is employed is the Chinese hamster ovary (CHO) cell line deficient in DHFR activity.
  • Alternatively, host cells (particularly wild-type hosts that contain endogenous DHFR) transformed or co-transformed with DNA sequences encoding polypeptide variant, wild-type DHFR protein, and another selectable marker such as aminoglycoside 3′-phosphotransferase (APH) can be selected by cell growth in medium containing a selection agent for the selectable marker such as an aminoglycosidic antibiotic, e.g., kanamycin, neomycin, or G418. See U.S. Pat. No. 4,965,199.
  • A suitable selection gene for use in yeast is the trp1 gene present in the yeast plasmid YRp7 (Stinchcomb et al., Nature, 282:39 (1979)). The trp1 gene provides a selection marker for a mutant strain of yeast lacking the ability to grow in tryptophan, for example, ATCC No. 44076 or PEP4-1. Jones, Genetics, 85:12 (1977). The presence of the trpl lesion in the yeast host cell genome then provides an effective environment for detecting transformation by growth in the absence of tryptophan. Similarly, Leu2-deficient yeast strains (ATCC 20,622 or 38,626) are complemented by known plasmids bearing the Leu2 gene.
  • In addition, vectors derived from the 1.6 μm circular plasmid pKD1 can be used for transformation of Kluyveromyces yeasts. Alternatively, an expression system for large-scale production of recombinant calf chymosin was reported for K. lactis. Van den Berg, Bio/Technology, 8:135 (1990). Stable multi-copy expression vectors for secretion of mature recombinant human serum albumin by industrial strains of Kluyveromyces have also been disclosed. Fleer et al., Bio/Technology, 9:968-975 (1991).
  • (iv) Promoter Component
  • Expression and cloning vectors usually contain a promoter that is recognized by the host organism and is operably linked to the polypeptide variant nucleic acid. Promoters suitable for use with prokaryotic hosts include the phoA promoter, β-lactamase and lactose promoter systems, alkaline phosphatase, a tryptophan (trp) promoter system, and hybrid promoters such as the tac promoter. However, other known bacterial promoters are suitable. Promoters for use in bacterial systems also will contain a Shine-Dalgarno (S.D.) sequence operably linked to the DNA encoding the polypeptide variant.
  • Promoter sequences are known for eukaryotes. Virtually all eukaryotic genes have an AT-rich region located approximately 25 to 30 bases upstream from the site where transcription is initiated. Another sequence found 70 to 80 bases upstream from the start of transcription of many genes is a CNCAAT region where N may be any nucleotide. At the 3′ end of most eukaryotic genes is an AATAAA sequence that may be the signal for addition of the poly A tail to the 3′ end of the coding sequence. All of these sequences are suitably inserted into eukaryotic expression vectors.
  • Examples of suitable promoting sequences for use with yeast hosts include the promoters for 3-phosphoglycerate kinase or other glycolytic enzymes, such as enolase, glyceraldehyde-3-phosphate dehydrogenase, hexokinase, pyruvate decarboxylase, phosphofructokinase, glucose-6-phosphate isomerase, 3-phosphoglycerate mutase, pyruvate kinase, triosephosphate isomerase, phosphoglucose isomerase, and glucokinase.
  • Other yeast promoters, which are inducible promoters having the additional advantage of transcription controlled by growth conditions, are the promoter regions for alcohol dehydrogenase 2, isocytochrome C, acid phosphatase, degradative enzymes associated with nitrogen metabolism, metallothionein, glyceraldehyde-3-phosphate dehydrogenase, and enzymes responsible for maltose and galactose utilization. Suitable vectors and promoters for use in yeast expression are further described in EP 73,657. Yeast enhancers also are advantageously used with yeast promoters.
  • Polypeptide variant transcription from vectors in mammalian host cells is controlled, for example, by promoters obtained from the genomes of viruses such as polyoma virus, fowlpox virus, adenovirus (such as Adenovirus 2), bovine papilloma virus, avian sarcoma virus, cytomegalovirus, a retrovirus, hepatitis-B virus and most preferably Simian Virus 40 (SV40), from heterologous mammalian promoters, e.g., the actin promoter or an immunoglobulin promoter, from heat-shock promoters, provided such promoters are compatible with the host cell systems.
  • The early and late promoters of the SV40 virus are conveniently obtained as an SV40 restriction fragment that also contains the SV40 viral origin of replication. The immediate early promoter of the human cytomegalovirus is conveniently obtained as a HindIII E restriction fragment. A system for expressing DNA in mammalian hosts using the bovine papilloma virus as a vector is disclosed in U.S. Pat. No. 4,419,446. A modification of this system is described in U.S. Pat. No. 4,601,978. See also Reyes et al., Nature 297:598-601 (1982) on expression of human β-interferon cDNA in mouse cells under the control of a thymidine kinase promoter from herpes simplex virus. Alternatively, the rous sarcoma virus long terminal repeat can be used as the promoter.
  • (v) Enhancer Element Component
  • Transcription of a DNA encoding the polypeptide variant of this invention by higher eukaryotes is often increased by inserting an enhancer sequence into the vector. Many enhancer sequences are now known from mammalian genes (globin, elastase, albumin, α-fetoprotein, and insulin). Typically, however, one will use an enhancer from a eukaryotic cell virus. Examples include the SV40 enhancer on the late side of the replication origin (bp 100-270), the cytomegalovirus early promoter enhancer, the polyoma enhancer on the late side of the replication origin, and adenovirus enhancers. See also Yaniv, Nature 297:17-18 (1982) on enhancing elements for activation of eukaryotic promoters. The enhancer may be spliced into the vector at a position 5′ or 3′ to the polypeptide variant-encoding sequence, but is preferably located at a site 5′ from the promoter.
  • (vi) Transcription Termination Component
  • Expression vectors used in eukaryotic host cells (yeast, fungi, insect, plant, animal, human, or nucleated cells from other multicellular organisms) will also contain sequences necessary for the termination of transcription and for stabilizing the mRNA. Such sequences are commonly available from the 5′ and, occasionally 3′, untranslated regions of eukaryotic or viral DNAs or cDNAs. These regions contain nucleotide segments transcribed as polyadenylated fragments in the untranslated portion of the mRNA encoding the polypeptide variant. One useful transcription termination component is the bovine growth hormone polyadenylation region. See WO94/11026 and the expression vector disclosed therein.
  • (vii) Selection and Transformation of Host Cells
  • Suitable host cells for cloning or expressing the DNA in the vectors herein are the prokaryote, yeast, or higher eukaryote cells described above. Suitable prokaryotes for this purpose include eubacteria, such as Gram-negative or Gram-positive organisms, for example, Enterobacteriaceae such as Escherichia, e.g., E. coli, Enterobacter, Erwinia, Klebsiella, Proteus, Salmonella, e.g., Salmonella typhimurium, Serratia, e.g., Serratia marcescans, and Shigella, as well as Bacilli such as B. subtilis and B. licheniformis (e.g., B. licheniformis 41P disclosed in DD 266,710 published 12 Apr. 1989), Pseudomonas such as P. aeruginosa, and Streptomyces. One preferred E. coli cloning host is E. coli 294 (ATCC 31,446), although other strains such as E. coli B, E. coli X1776 (ATCC 31,537), and E. coli W3110 (ATCC 27,325) are suitable. These examples are illustrative rather than limiting.
  • In addition to prokaryotes, eukaryotic microbes such as filamentous fungi or yeast are suitable cloning or expression hosts for polypeptide variant-encoding vectors. Saccharomyces cerevisiae, or common baker's yeast, is the most commonly used among lower eukaryotic host microorganisms. However, a number of other genera, species, and strains are commonly available and useful herein, such as Schizosaccharomyces pombe; Kluyveromyces hosts such as, e.g., K. lactis, K. fragilis (ATCC 12,424), K. bulgaricus (ATCC 16,045), K. wickeramii (ATCC 24,178), K. waltii (ATCC 56,500), K. drosophilarum (ATCC 36,906), K. thermotolerans, and K. marxianus; yarrowia (EP 402,226); Pichia pastoris (EP 183,070); Candida; Trichoderma reesia; (EP 244,234); Neurospora crassa; Schwanniomyces such as Schwanniomyces occidentalis; and filamentous fungi such as, e.g., Neurospora, Penicillium, Tolypocladium, and Aspergillus hosts such as A. nidulans and A. niger.
  • Suitable host cells for the expression of glycosylated polypeptide variant are derived from multicellular organisms. Examples of invertebrate cells include plant and insect cells. Numerous baculoviral strains and variants and corresponding permissive insect host cells from hosts such as Spodoptera frugiperda (caterpillar), Aedes aegypti (mosquito), Aedes albopictus (mosquito), Drosophila melanogaster (fruitfly), and Bombyx mori have been identified. A variety of viral strains for transfection are publicly available, e.g., the L-1 variant of Autographa californica NPV and the Bm-5 strain of Bombyx mori NPV, and such viruses may be used as the virus herein according to the present invention, particularly for transfection of Spodoptera frugiperda cells.
  • Plant cell cultures of cotton, corn, potato, soybean, petunia, tomato, and tobacco can also be utilized as hosts.
  • However, interest has been greatest in vertebrate cells, and propagation of vertebrate cells in culture (tissue culture) has become a routine procedure. Examples of useful mammalian host cell lines are monkey kidney CV1 line transformed by SV40 (COS-7, ATCC CRL 1651); human embryonic kidney line (293 or 293 cells subcloned for growth in suspension culture, Graham et al., J. Gen Virol. 36:59 (1977)); baby hamster kidney cells (BHK, ATCC CCL 10); Chinese hamster ovary cells/-DHFR (CHO, Urlaub et al., Proc. Natl. Acad. Sci. USA 77:4216 (1980)); mouse sertoli cells (TM4, Mather, Biol. Reprod. 23:243-251 (1980)); monkey kidney cells (CV1 ATCC CCL 70); African green monkey kidney cells (VERO-76, ATCC CRL-1587); human cervical carcinoma cells (HELA, ATCC CCL 2); canine kidney cells (MDCK, ATCC CCL 34); buffalo rat liver cells (BRL 3A, ATCC CRL 1442); human lung cells (W138, ATCC CCL 75); human liver cells (Hep G2, HB 8065); mouse mammary tumor (MMT 060562, ATCC CCL51); TRI cells (Mather et al., Annals N.Y. Acad. Sci. 383:44-68 (1982)); MRC 5 cells; FS4 cells; and a human hepatoma line (Hep G2).
  • Host cells are transformed with the above-described expression or cloning vectors for polypeptide variant production and cultured in conventional nutrient media modified as appropriate for inducing promoters, selecting transformants, or amplifying the genes encoding the desired sequences.
  • (viii) Culturing the Host Cells
  • The host cells used to produce the polypeptide variant of this invention may be cultured in a variety of media. Commercially available media such as Ham's F10 (Sigma), Minimal Essential Medium ((MEM), (Sigma), RPMI-1640 (Sigma), and Dulbecco's Modified Eagle's Medium ((DMEM), Sigma) are suitable for culturing the host cells. In addition, any of the media described in Ham et al., Meth. Enz. 58:44 (1979), Barnes et al., Anal. Biochem. 102:255 (1980), U.S. Pat. Nos. 4,767,704; 4,657,866; 4,927,762; 4,560,655; or 5,122,469; WO 90/03430; WO 87/00195; or U.S. Pat. Re. 30,985 may be used as culture media for the host cells. Any of these media may be supplemented as necessary with hormones and/or other growth factors (such as insulin, transferrin, or epidermal growth factor), salts (such as sodium chloride, calcium, magnesium, and phosphate), buffers (such as HEPES), nucleotides (such as adenosine and thymidine), antibiotics (such as GENTAMYCIN™ drug), trace elements (defined as inorganic compounds usually present at final concentrations in the micromolar range), and glucose or an equivalent energy source. Any other necessary supplements may also be included at appropriate concentrations that would be known to those skilled in the art. The culture conditions, such as temperature, pH, and the like, are those previously used with the host cell selected for expression, and will be apparent to the ordinarily skilled artisan.
  • (ix) Polypeptide Variant Purification
  • When using recombinant techniques, the polypeptide variant can be produced intracellularly, in the periplasmic space, or directly secreted into the medium. If the polypeptide variant is produced intracellularly, as a first step, the particulate debris, either host cells or lysed fragments, is removed, for example, by centrifugation or ultrafiltration. Carter et al., Bio/Technology 10:163-167 (1992) describe a procedure for isolating antibodies which are secreted to the periplasmic space of E. coli. Briefly, cell paste is thawed in the presence of sodium acetate (pH 3.5), EDTA, and phenylmethylsulfonylfluoride (PMSF) over about 30 min. Cell debris can be removed by centrifugation. Where the polypeptide variant is secreted into the medium, supernatants from such expression systems are generally first concentrated using a commercially available protein concentration filter, for example, an Amicon or Millipore Pellicon ultrafiltration unit. A protease inhibitor such as PMSF may be included in any of the foregoing steps to inhibit proteolysis and antibiotics may be included to prevent the growth of adventitious contaminants.
  • The polypeptide variant composition prepared from the cells can be purified using, for example, hydroxylapatite chromatography, gel electrophoresis, dialysis, and affinity chromatography, with affinity chromatography being the preferred purification technique. The suitability of protein A as an affinity ligand depends on the species and isotype of any immunoglobulin Fc region that is present in the polypeptide variant. Protein A can be used to purify polypeptide variants that are based on human γ1, γ2, or γ4 heavy chains (Lindmark et al., J. lmmunol. Meth. 62:1-13 (1983)). Protein G is recommended for all mouse isotypes and for human γ3 (Guss et al., EMBO J. 5:15671575 (1986)). The matrix to which the affinity ligand is attached is most often agarose, but other matrices are available. Mechanically stable matrices such as controlled pore glass or poly(styrenedivinyl)benzene allow for faster flow rates and shorter processing times than can be achieved with agarose. Where the polypeptide variant comprises a C H3 domain, the Bakerbond ABX™ resin (J. T. Baker, Phillipsburg, N.J.) is useful for purification. Other techniques for protein purification such as fractionation on an ion-exchange column, ethanol precipitation, Reverse Phase HPLC, chromatography on silica, chromatography on heparin SEPHAROSE™ chromatography on an anion or cation exchange resin (such as a polyaspartic acid column), chromatofocusing, SDS-PAGE, and ammonium sulfate precipitation are also available depending on the polypeptide variant to be recovered.
  • Following any preliminary purification step(s), the mixture comprising the polypeptide variant of interest and contaminants may be subjected to low pH hydrophobic interaction chromatography using an elution buffer at a pH between about 2.5-4.5, preferably performed at low salt concentrations (e.g.,from about 0-0.25M salt).
  • E. Pharmaceutical Formulations
  • Therapeutic formulations of the polypeptide variant are prepared for storage by mixing the polypeptide variant having the desired degree of purity with optional physiologically acceptable carriers, excipients or stabilizers (Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980)), in the form of lyophilized formulations or aqueous solutions. Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptide; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine, arginine, or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugars such as sucrose, mannitol, trehalose or sorbitol; salt-forming counter-ions such as sodium; metal complexes (e.g., Zn-protein complexes); and/or non-ionic surfactants such as TWEEN™, PLURONICS™ or polyethylene glycol (PEG).
  • The formulation herein may also contain more than one active compound as necessary for the particular indication being treated, preferably those with complementary activities that do not adversely affect each other. Such molecules are suitably present in combination in amounts that are effective for the purpose intended.
  • The active ingredients may also be entrapped in microcapsule prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsule and poly-(methylmethacylate) microcapsule, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) or in macroemulsions. Such techniques are disclosed in Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980).
  • The formulations to be used for in vivo administration must be sterile. This is readily accomplished by filtration through sterile filtration membranes.
  • Sustained-release preparations may be prepared. Suitable examples of sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing the polypeptide variant, which matrices are in the form of shaped articles, e.g., films, or microcapsule. Examples of sustained-release matrices include polyesters, hydrogels (for example, poly(2-hydroxyethyl-methacrylate), or poly(vinylalcohol)), polylactides (U.S. Pat. No. 3,773,919), copolymers of L-glutamic acid and γ ethyl-L-glutamate, non-degradable ethylene-vinyl acetate, degradable lactic acid-glycolic acid copolymers such as the LUPRON DEPOT™ (injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate), and poly-D-(−)-3-hydroxybutyric acid. While polymers such as ethylene-vinyl acetate and lactic acid-glycolic acid enable release of molecules for over 100 days, certain hydrogels release proteins for shorter time periods. When encapsulated antibodies remain in the body for a long time, they may denature or aggregate as a result of exposure to moisture at 37° C., resulting in a loss of biological activity and possible changes in immunogenicity. Rational strategies can be devised for stabilization depending on the mechanism involved. For example, if the aggregation mechanism is discovered to be intermolecular S—S bond formation through thio-disulfide interchange, stabilization may be achieved by modifying sulfhydryl residues, lyophilizing from acidic solutions, controlling moisture content, using appropriate additives, and developing specific polymer matrix compositions.
  • F. Non-Therapeutic Uses for the Polypeptide Variant
  • The polypeptide variant of the invention may be used as an affinity purification agent. In this process, the polypeptide variant is immobilized on a solid phase such a Sephadex resin or filter paper, using methods well known in the art. The immobilized polypeptide variant is contacted with a sample containing the antigen to be purified, and thereafter the support is washed with a suitable solvent that will remove substantially all the material in the sample except the antigen to be purified, which is bound to the immobilized polypeptide variant. Finally, the support is washed with another suitable solvent, such as glycine buffer, pH 5.0, that will release the antigen from the polypeptide variant.
  • The polypeptide variant may also be useful in diagnostic assays, e.g., for detecting expression of an antigen of interest in specific cells, tissues, or serum.
  • For diagnostic applications, the polypeptide variant typically will be labeled with a detectable moiety. Numerous labels are available which can be generally grouped into the following categories:
  • (a) Radioisotopes, such as 35S, 14C, 125I, 3H, and 131I. The polypeptide variant can be labeled with the radioisotope using the techniques described in Current Protocols in Immunology, Volumes 1 and 2, Coligen et al., Ed. Wiley-Interscience, New York, N.Y., Pubs. (1991) for example and radioactivity can be measured using scintillation counting.
  • (b) Fluorescent labels such as rare earth chelates (europium chelates) or fluorescein and its derivatives, rhodamine and its derivatives, dansyl, Lissamine, phycoerythrin and Texas Red are available. The fluorescent labels can be conjugated to the polypeptide variant using the techniques disclosed in Current Protocols in Immunology, supra, for example. Fluorescence can be quantified using a fluorimeter.
  • (c) Various enzyme-substrate labels are available and U.S. Pat. No. 4,275,149 provides a review of some of these. The enzyme generally catalyzes a chemical alteration of the chromogenic substrate that can be measured using various techniques. For example, the enzyme may catalyze a color change in a substrate, which can be measured spectrophotometrically. Alternatively, the enzyme may alter the fluorescence or chemiluminescence of the substrate. Techniques for quantifying a change in fluorescence are described above. The chemiluminescent substrate becomes electronically excited by a chemical reaction and may then emit light which can be measured (using a chemiluminometer, for example) or donates energy to a fluorescent acceptor. Examples of enzymatic labels include luciferases (e.g., firefly luciferase and bacterial luciferase; U.S. Pat. No. 4,737,456), luciferin, 2,3-dihydrophthalazinediones, malate dehydrogenase, urease, peroxidase such as horseradish peroxidase (HRPO), alkaline phosphatase, β-galactosidase, glucoamylase, lysozyme, saccharide oxidases (e.g., glucose oxidase, galactose oxidase, and glucose-6-phosphate dehydrogenase), heterocyclic oxidases (such as uricase and xanthine oxidase), lactoperoxidase, microperoxidase, and the like. Techniques for conjugating enzymes to antibodies are described in O'Sullivan et al., Methods for the Preparation of Enzyme-Antibody Conjugates for use in Enzyme Immunoassay, in Methods in Enzym. (ed J. Langone & H. Van Vunakis), Academic press, New York, 73:147-166 (1981).
  • Examples of enzyme-substrate combinations include, for example:
  • (i) Horseradish peroxidase (HRPO) with hydrogen peroxidase as a substrate, wherein the hydrogen peroxidase oxidizes a dye precursor (e.g.,orthophenylene diamine (OPD) or 3,3′,5,5′-tetramethyl benzidine hydrochloride (TMB));
  • (ii) alkaline phosphatase (AP) with para-Nitrophenyl phosphate as chromogenic substrate; and
  • (iii) β-D-galactosidase (β-D-Gal) with a chromogenic substrate (e.g., p-nitrophenyl-β-D-galactosidase) or fluorogenic substrate 4-methylumbelliferyl-β-D-galactosidase.
  • Numerous other enzyme-substrate combinations are available to those skilled in the art. For a general review of these, see U.S. Pat. Nos. 4,275,149 and 4,318,980.
  • Sometimes, the label is indirectly conjugated with the polypeptide variant. The skilled artisan will be aware of various techniques for achieving this. For example, the polypeptide variant can be conjugated with biotin and any of the three broad categories of labels mentioned above can be conjugated with avidin, or vice versa. Biotin binds selectively to avidin and thus, the label can be conjugated with the polypeptide variant in this indirect manner. Alternatively, to achieve indirect conjugation of the label with the polypeptide variant, the polypeptide variant is conjugated with a small hapten (e.g., digoxin) and one of the different types of labels mentioned above is conjugated with an anti-hapten polypeptide variant (e.g., anti-digoxin antibody). Thus, indirect conjugation of the label with the polypeptide variant can be achieved.
  • In another embodiment of the invention, the polypeptide variant need not be labeled, and the presence thereof can be detected using a labeled antibody which binds to the polypeptide variant.
  • The polypeptide variant of the present invention may be employed in any known assay method, such as competitive binding assays, direct and indirect sandwich assays, and immunoprecipitation assays. Zola, Monoclonal Antibodies: A Manual of Techniques, pp. 147-158 (CRC Press, Inc. 1987).
  • The polypeptide variant may also be used for in vivo diagnostic assays. Generally, the polypeptide variant is labeled with a radionuclide (such as 111In, 99Tc, 14C, 131I, 125I, 3H, 32P or 35S) so that the antigen or cells expressing it can be localized using immunoscintiography.
  • G. In Vivo uses for the Polypeptide Variant
  • It is contemplated that the polypeptide variant of the present invention may be used to treat a mammal e.g. a patient suffering from, or predisposed to, a disease or disorder who could benefit from administration of the polypeptide variant. The conditions which can be treated with the polypeptide variant are many and include cancer (e.g. where the polypeptide variant binds the HER2 receptor, CD20 or vascular endothelial growth factor (VEGF)); allergic conditions such as asthma (with an anti-IgE antibody); and LFA-1-mediated disorders (e.g. where the polypeptide variant is an anti-LFA-1 or anti-ICAM-1 antibody) etc.
  • Where the antibody binds the HER2 receptor, the disorder preferably is HER2-expressing cancer, e.g. a benign or malignant tumor characterized by overexpression of the HER2 receptor. Such cancers include, but are not limited to, breast cancer, squamous cell cancer, small-cell lung cancer, non-small cell lung cancer, gastrointestinal cancer, pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, bladder cancer, hepatoma, colon cancer, colorectal cancer, endometrial carcinoma, salivary gland carcinoma, kidney cancer, liver cancer, prostate cancer, vulval cancer, thyroid cancer, hepatic carcinoma and various types of head and neck cancer. According to the teachings herein, one may prepare a polypeptide with a variant Fc region which has improved, or diminished, ADCC activity. Such molecules will find applications in the treatment of different disorders.
  • For example, the polypeptide variant with improved ADCC activity may be employed in the treatment of diseases or disorders where destruction or elimination of tissue or foreign micro-organisms is desired. For example, the polypeptide may be used to treat cancer; inflammatory disorders; infections (e.g. bacterial, viral, fungal or yeast infections); and other conditions (such as goiter) where removal of tissue is desired, etc.
  • Where the polypeptide variant has diminished ADCC activity, such variants may be used to treat diseases or disorders where a Fc region-containing polypeptide with long half-life is desired, but the polypeptide preferably does not have undesirable effector function(s). For example, the Fc region-containing polypeptide may be an anti-tissue factor (TF) antibody; anti-IgE antibody; and anti-integrin antibody (e.g. an anti-α4β7 antibody). The desired mechanism of action of such Fc region-containing polypeptides may be to block ligand-receptor binding pairs. Moreover, the Fc-region containing polypeptide with diminished ADCC activity may be an agonist antibody.
  • The polypeptide variant is administered by any suitable means, including parenteral, subcutaneous, intraperitoneal, intrapulmonary, and intranasal, and, if desired for local immunosuppressive treatment, intralesional administration. Parenteral infusions include intramuscular, intravenous, intraarterial, intraperitoneal, or subcutaneous administration. In addition, the polypeptide variant is suitably administered by pulse infusion, particularly with declining doses of the polypeptide variant. Preferably the dosing is given by injections, most preferably intravenous or subcutaneous injections, depending in part on whether the administration is brief or chronic.
  • For the prevention or treatment of disease, the appropriate dosage of polypeptide variant will depend on the type of disease to be treated, the severity and course of the disease, whether the polypeptide variant is administered for preventive or therapeutic purposes, previous therapy, the patient's clinical history and response to the polypeptide variant, and the discretion of the attending physician. The polypeptide variant is suitably administered to the patient at one time or over a series of treatments.
  • Depending on the type and severity of the disease, about 1 μg/kg to 15 mg/kg (e.g., 0.1-20 mg/kg) of polypeptide variant is an initial candidate dosage for administration to the patient, whether, for example, by one or more separate administrations, or by continuous infusion. A typical daily dosage might range from about 1 μg/kg to 100 mg/kg or more, depending on the factors mentioned above. For repeated administrations over several days or longer, depending on the condition, the treatment is sustained until a desired suppression of disease symptoms occurs. However, other dosage regimens may be useful. The progress of this therapy is easily monitored by conventional techniques and assays.
  • The polypeptide variant composition will be formulated, dosed, and administered in a fashion consistent with good medical practice. Factors for consideration in this context include the particular disorder being treated, the particular mammal being treated, the clinical condition of the individual patient, the cause of the disorder, the site of delivery of the agent, the method of administration, the scheduling of administration, and other factors known to medical practitioners. The “therapeutically effective amount” of the polypeptide variant to be administered will be governed by such considerations, and is the minimum amount necessary to prevent, ameliorate, or treat a disease or disorder. The polypeptide variant need not be, but is optionally formulated with one or more agents currently used to prevent or treat the disorder in question. The effective amount of such other agents depends on the amount of polypeptide variant present in the formulation, the type of disorder or treatment, and other factors discussed above. These are generally used in the same dosages and with administration routes as used hereinbefore or about from 1 to 99% of the heretofore employed dosages.
  • The invention will be more fully understood by reference to the following examples. They should not, however, be construed as limiting the scope of this invention. All literature and patent citations mentioned herein are expressly incorporated by reference.
  • EXAMPLE 1 Low Affinity Receptor Binding Assay
  • This assay determines binding of an IgG Fc region to recombinant FcγRIIA, FcγRIIB and FcγRIIIA α subunits expressed as His6-glutathione S transferase (GST)-tagged fusion proteins. Since the affinity of the Fc region of IgG1 for the FcγRI is in the nanomolar range, the binding of IgG1 Fc variants can be measured by titrating monomeric IgG and measuring bound IgG with a polyclonal anti-IgG in a standard ELISA format (Example 2 below). The affinity of the other members of the FcγR family, i.e. FcγRIIA, FcγRIIB and FcγRIIIA for IgG is however in the micromolar range and binding of monomeric IgG1 for these receptors can not be reliably measured in an ELISA format.
  • The following assay utilizes Fc variants of recombinant anti-IgE E27 (FIGS. 4A and 4B) which, when mixed with human IgE at a 1:1 molar ratio, forms a stable hexamer consisting of three anti-IgE molecules and three IgE molecules. A recombinant chimeric form of IgE (chimeric IgE) was engineered and consists of a human IgE Fc region and the Fab of an anti-VEGF antibody (Presta et al., Cancer Research 57:4593-4599 (1997)) which binds two VEGF molecules per mole of anti-VEGF. When recombinant human VEGF is added at a 2:1 molar ratio to chimeric IgE:E27 hexamers, the hexamers are linked into larger molecular weight complexes via the chimeric IgE Fab:VEGF interaction. The E27 component of this complex binds to the FcγRIIA, FcγRIIB and FcγRIIIA α subunits with higher avidity to permit detection in an ELISA format.
  • Materials and Methods
  • Receptor Coat Fcγreceptor α subunits were expressed as GST fusions of His6 tagged extracellular domains (ECDs) in 293 cells resulting in an ECD-6His-GST fusion protein (Graham et al., J. Gen. Virol. 36:59-74 (1977) and Gorman et al., DNA Prot. Eng. Tech. 2:3-10 (1990)) and purified by Ni-NTA column chromatography (Qiagen, Australia) and buffer exchanged into phosphate buffered saline (PBS). Concentrations were determined by absorption at 280nm using extinction coefficients derived by amino acid composition analysis. Receptors were coated onto Nunc F96 maxisorb plates (cat no. 439454) at 100 ng per well by adding 100 μl of receptor-GST fusion at 1 μg/ml in PBS and incubated for 48 hours at 4° C. Prior to assay, plates are washed 3× with 250 μl of wash buffer (PBS pH 7.4 containing 0.5% TWEEN™) and blocked with 250 μl of assay buffer (50 mM Tris buffered saline, 0.05% TWEEN 20™, 0.5% RIA grade bovine albumin (Sigma A7888), and 2 mM EDTA pH 7.4).
  • Immune Complex Formation: Equal molar amounts (1:1) of E27 and recombinant chimeric IgE which binds two moles recombinant human VEGF per mole of chimeric IgE are added to a 12×75 mm polypropylene tube in PBS and mixed by rotation for 30 minutes at 25° C. E27 (anti-IgE) /chimeric IgE (IgE) hexamers are formed during this incubation. Recombinant human VEGF (165 form, MW 44,000) is added at a 2:1 molar ratio to the IgE concentration and mixed by rotation an additional 30 minutes at 25° C. VEGF-chimeric IgE binding links E27: chimeric IgE hexamers into larger molecular weight complexes which bind FcγR α subunit ECD coated plates via the Fc region of the E27 antibody.
  • E27:chimeric IgE:VEGF: (1:1:2 molar ratio) complexes are added to FcγR α subunit coated plates at E27 concentrations of 5 μg and 1 μg total IgG in quadruplicate in assay buffer and incubated for 120 minutes at 25° C. on an orbital shaker.
  • Complex Detection: Plates are washed 5× with wash buffer to remove unbound complexes and IgG binding is detected by adding 100 μl horse radish peroxidase (HRP) conjugated goat anti-human IgG (γ) heavy chain specific (Boehringer Mannheim 1814249) at 1:10,000 in assay buffer and incubated for 90 min at 25° C. on an orbital shaker. Plates are washed 5× with wash buffer to remove unbound HRP goat anti-human IgG and bound anti-IgG is detected by adding 100 μl of substrate solution (0.4 mg/ml o-phenylenedaimine dihydrochloride, Sigma P6912, 6 mM H2O2 in PBS) and incubating for 8 min at 25° C. Enzymatic reaction is stopped by the addition of 100 μl 4.5N H2SO4 and colorimetric product is measured at 490 nm on a 96 well plate densitometer (Molecular Devices). Binding of E27 variant complexes is expressed as a percent of the wild type E27 containing complex.
  • EXAMPLE 2 Identification of Unique C1q Binding Sites in a Human IgG Antibody
  • In the present study, mutations were identified in the CH2 domain of a human IgG1 antibody, “C2B8” (Reff et al., Blood 83:435 (1994)), that ablated binding of the antibody to C1q but did not alter the conformation of the antibody nor affect binding to each of the FcγRs. By alanine scanning mutagenesis, five variants in human IgG1 were identified, D270K, D270V, K322A P329A, and P331, that were non-lytic and had decreased binding to C1q. The data suggested that the core C1q binding sites in human IgG1 is different from that of murine IgG2b. In addition, K322A, P329A and P331A were found to bind normally to the CD20 antigen, and to four Fc receptors, FcγRI, FcγRII, FcγRIII and FcRn.
  • Materials and Methods
  • Construction of C2B8 Variants: The chimeric light and heavy chains of anti-CD20 antibody C2B8 (Reff et al., Blood 83:435 (1994)) subcloned separately into previously described PRK vectors (Gorman et al., DNA Protein Eng. Tech. 2:3 (1990)) were used. By site directed mutagenesis (Kunkel et al., Proc. Natl. Acad. Sci. USA 82:488-492 (1985)), alanine scan variants of the Fc region in the heavy chain were constructed. The heavy and light chain plasmids were co-transfected into an adenovirus transformed human embryonic kidney cell line as previously described (Werther et al., J. Immunol. 157:4986 (1996)). The media was changed to serum-free 24 hours after transfection and the secreted antibody was harvested after five days. The antibodies were purified using Protein A-SEPHAROSE CL-4B™ (Pharmacia), buffer exchanged and concentrated to 0.5 ml with PBS using a Centricon-30 (Amicon), and stored at 4° C. The concentration of the antibody was determined using total Ig-binding ELISA.
  • C1q Binding ELISA: Costar 96 well plates were coated overnight at 4° C. with the indicated concentrations of C2B8 in coating buffer (0.05 M sodium carbonate buffer), pH 9. The plates were then washed 3× with PBS/0.05% TWEEN 20™, pH 7.4 and blocked with 200 μl of ELISA diluent without thimerosal (0.1 M NaPO4/0.1 M NaCl/0.1% gelatin/0.05% TWEEN™ 0.05% ProClin300) for 1 hr at room temperature. The plate was washed 3× with wash buffer, an aliquot of 100 μl of 2 μg/ml C1q (Quidel, San Diego, Calif.) was added to each well and incubated for 2 hrs at room temperature. The plate was then washed 6× with wash buffer. 100 μl of a 1:1000 dilution of sheep anti-complement C1q peroxidase conjugated antibody (Biodesign) was added to each well and incubated for 1 hour at room temperature. The plate was again washed 6× with wash buffer and 100 μl of substrate buffer (PBS/0.012% H2O2) containing OPD (O-phenylenediamine dihydrochloride (Sigma)) was added to each well. The oxidation reaction, observed by the appearance of a yellow color, was allowed to proceed for 30 minutes and stopped by the addition of 100 μl of 4.5 N H2SO4. The absorbance was then read at (492-405) nm using a microplate reader (SPECTRA MAX 250™, Molecular Devices Corp.). The appropriate controls were run in parallel (i.e. the ELISA was performed without C1q for each concentration of C2B8 used and also the ELISA was performed without C2B8). For each variant, C1q binding was measured by plotting the absorbance (492-405) nm versus concentration of C2B8 in μg/ml using a 4-parameter curve fitting program (KALEIDAGRAPH™) and comparing EC50 values.
  • Complement Dependent Cytotoxicity (CDC) Assay. This assay was performed essentially as previously described (Gazzano-Santoro et al., J. Immunol. Methods 202:163-171 (1997)). Various concentrations of C2B8 (0.08-20 μg/ml) were diluted with RHB buffer (RPMI 1640/20 mM HEPES (pH 7.2)/2 mM Glutamine/0.1% BSA/100 μg/ml Gentamicin). Human complement (Quidel) was diluted 1:3 in RHB buffer and WIL2-S cells (available from the ATCC, Manassas, Va.) which express the CD20 antigen were diluted to a density of 1×106 cells /ml with RHB buffer. Mixtures of 150 μl containing equal volumes of C2B8, diluted human complement and WIL2-S cells were added to a flat bottom tissue culture 96 well plate and allowed to incubate for 2 hrs at 37° C. and 5% CO2 to facilitate complement mediated cell lysis. 50 μl of alamar blue (Accumed International) was then added to each well and incubated overnight at 37° C. The absorbance was measured using a 96-well fluorometer with excitation at 530 nm and emission at 590 nm. As described by Gazzano-Santoro et al., the results are expressed in relative fluorescence units (RFU). The sample concentrations were computed from a C2B8 standard curve and the percent activity as compared to wild type C2B8 is reported for each variant.
  • CD20 Binding Potency of the C2B8 Variants: The binding of C2B8 and variants to the CD20 antigen were assessed by a method previously described (Reff et al., (1994), supra; reviewed in Gazzano-Santoro et al., (1997), supra). WIL2-S cells were grown for 3-4 days to a cell density of 1×106 cells/ml. The cells were washed and spun twice in FACS buffer (PBS/0.1% BSA/0.02% NaN3) and resuspended to a cell density of 5×106 cells/ml. 200 μl of cells (5×106 cells/ml) and 20 μl of diluted C2B8 samples were added to a 5 ml tube and incubated at room temperature for 30 minutes with agitation. The mixture was then washed with 2 ml of cold FACS buffer, spun down and resuspended in 200 μl of cold FACS buffer. To the suspension, 10 μl of goat anti-human IgG-FITC (American Qualex Labs.) was added and the mixture was incubated in the dark at room temperature for 30 minutes with agitation. After incubation, the mixture was washed with 2 ml of FACS buffer, spun down and resuspended in 1 ml of cold fixative buffer (1% formaldehyde in PBS). The samples were analyzed by flow cytometry and the results expressed as relative fluorescence units (RFU) were plotted against antibody concentrations using a 4-parameter curve fitting program (KALEIDAGRAPH™). The EC50 values are reported as a percentage of that of the C2B8 reference material.
  • FcγR Binding ELISAs: FcγRI α subunit-GST fusion was coated onto Nunc F96 maxisorb plates (cat no. 439454) by adding 100 μl of receptor-GST fusion at 1 μg/ml in PBS and incubated for 48 hours at 4° C. Prior to assay, plates are washed 3× with 250 μl of wash buffer (PBS pH 7.4 containing 0.5% TWEEN 20™) and blocked with 250 μl of assay buffer (50 mM Tris buffered saline, 0.05% TWEEN 20™, 0.5% RIA grade bovine albumin (Sigma A7888), and 2mM EDTA pH 7.4). Samples diluted to 10 μg/ml in 1 ml of assay buffer are added to FcγRI α subunit coated plates and incubated for 120 minutes at 25° C. on an orbital shaker. Plates are washed 5× with wash buffer to remove unbound complexes and IgG binding is detected by adding 100 μl horse radish peroxidase (HRP) conjugated goat anti-human IgG (γ) heavy chain specific (Boehringer Mannheim 1814249) at 1:10,000 in assay buffer and incubated for 90 min at 25° C. on an orbital shaker. Plates are washed 5× with wash buffer to remove unbound HRP goat anti-human IgG and bound anti-IgG is detected by adding 100 μl of substrate solution (0.4 mg/ml o-phenylenedaimine dihydrochloride, Sigma P6912, 6 mM H2O2 in PBS) and incubating for 8 min at 25° C. Enzymatic reaction is stopped by the addition of 100 μl 4.5N H2SO4 and colorimetric product is measured at 490 nm on a 96 well plate densitometer (Molecular Devices). Binding of variant is expressed as a percent of the wild type molecule.
  • FcγRII and III binding ELISAs were performed as described in Example 1 above.
  • For measuring FcRn binding activity of IgG variants, ELISA plates were coated with 2 μg/ml streptavidin (Zymed, South San Francisco) in 50 mM carbonate buffer, pH 9.6, at 4° C. overnight and blocked with PBS-0.5% BSA, pH 7.2 at room temperature for one hour. Biotinylated FcRn (prepared using biotin-X-NHS from Research Organics, Cleveland, Ohio and used at 1-2 μg/ml) in PBS-0.5% BSA, 0.05% polysorbate 20, pH 7.2, was added to the plate and incubated for one hour. Two fold serial dilutions of IgG standard (1.6-100 ng/ml) or variants in PBS-0.5% BSA, 0.05% polysorbate 20, pH 6.0, were added to the plate and incubated for two hours. Bound IgG was detected using peroxidase labeled goat F(ab′)2 anti-human IgG F(ab′)2 in the above pH 6.0 buffer (Jackson ImmunoResearch, West Grove, Pa.) followed by 3,3′,5,5′-tetramethyl benzidine (Kirgaard & Perry Laboratories) as the substrate. Plates were washed between steps with PBS-0.05% polysorbate 20 at either pH 7.2 or 6.0. Absorbance was read at 450 nm on a Vmax plate reader (Molecular Devices, Menlo Park, Calif.). Titration curves were fit with a four-parameter nonlinear regression curve-fitting program (KaleidaGraph, Synergy software, Reading, Pa.). Concentrations of IgG variants corresponding to the mid-point absorbance of the titration curve of the standard were calculated and then divided by the concentration of the standard corresponding to the mid-point absorbance of the standard titration curve.
  • Results and Discussion
  • By alanine scanning mutagenesis, several single point mutations were constructed in the CH2 domain of C2B8 beginning with E318A, K320A and K322A. All the variants constructed bound normally to the CD20 antigen (Table 3).
  • TABLE 3
    wt E318A K320A K322A P329A P331A
    FcRn + + + +
    CD20 + + + + + +
    FcγRI + + + + + +
    FcγRII + + + + + +
    FcγRIII + + + + + +
    *C1q +++ ++ +++
    CDC + + +
    (+) indicates binding and
    (−) signifies binding abolished
    *With respect to C1q binding, each + sign is equivalent to approximately 33% binding.
  • Where binding of human complement to an antibody with a human Fc was analyzed, the ability of E318A and K320A to activate complement was essentially identical to that of wild type C2B8 (Table 3). When compared to wild type C2B8, there appears to be little difference in the binding of E318A and K320A to C1q. There is only a 10% decrease in the binding of K320A and about a 30% decrease in the binding of E318A to C1q (FIG. 2). The results indicate that the effect of the E318A and the K320A substitution on complement activation and C1q binding is minimal. Also, the human IgG1 of C2B8 was substituted for human IgG2 and used as a negative control in the C1q binding studies. The IgG2 variant appears to have a much lower affinity for C1q than the E318A and K320A variants (FIG. 2). Thus, the results demonstrate that E318 and K320 do not constitute the core C1q binding sites for human IgG1. Conversely, the K322A substitution had a significant effect on both complement activity and C1q binding. The K322A variant did not have CDC activity when tested in the above CDC assay and was more than a 100 fold lower than wild type C2B8 in binding to C1q (FIG. 2). In the human system, K322 is the only residue of the proposed core C1q binding sites that appeared to have a significant effect on complement activation and C1q binding.
  • Since the Duncan and Winter study was performed using mouse IgG2b and the above results reveal that K320 and E318 in human IgG1 are not involved in C1q binding, and without being bound to any one theory, the above data suggest that the C1q binding region in murine IgGs is different from that of the human. To investigate this further and also to identify additional variants that do not bind to C1q and hence do not activate complement, several more point mutations in the vicinity of K322 were constructed as assessed from the three dimensional structure of the C2B8 Fc. Variants constructed, K274A, N276A, Y278A, S324A, P329A, P331 A, K334A, and T335A, were assessed for their ability to bind C1q and also to activate complement. Many of these substitutions had little or no effect on C1q binding or complement activation. In the above assays, the P329A and the P331A variants did not activate complement and had decreased binding to C1q. The P331A variant did not activate complement and was 60 fold lower in binding to C1q (FIG. 3) when compared to wild type C2B8 (FIG. 2). The concentration range of the antibody variants used in FIG. 3 is expanded to 100 μg/ml in order to observe saturation of C1q binding to the P331 A variant. The mutation P329A results in an antibody that does not activate complement and is more than a 100 fold lower in binding to C1q (FIG. 3) when compared to wild type C2B8 (FIG. 2).
  • Variants that did not bind to C1q and hence did not activate complement were examined for their ability to bind to the Fc receptors: FcγRI, FcγRIIA, FcγRIIB, FcγRIIIA and FcRn. This particular study was performed using a humanized anti-IgE antibody, an IgG1 antibody with these mutations (see Example 1 above). The results revealed the variants, K322A and P329A, bind to all the Fc receptors to the same extent as the wild type protein (Table 4). However, there was a slight decrease in the binding of P331A to FcγRIIB.
  • In conclusion, two amino acid substitutions in the COON terminal region of the CH2 domain of human IgG1, K322A and P329A were identified that result in more than 100 fold decrease in C1q binding and do not activate the CDC pathway. These two variants, K322A and P329A, bind to all Fc receptors with the same affinity as the wild type antibody. Based on the results, summarized in Table 4, and without being bound to any one theory, it is proposed that the C1q binding epicenter of human IgG1 is centered around K322, P329 and P331 and is different from the murine IgG2b epicenter which constitutes E318, K320 and K322.
  • TABLE 4
    wt E318A K320A K322A P329A P331A
    CD20
    100 89 102 86 112 103
    a FcγRI 100 93 102 90 104 74
    a FcγRIIA 100 113 94 109 111 86
    a FcγRIIB 100 106 83 101 96 58
    a FcγRIII 100 104 72 90 85 73
    CDC 100 108 108 none none none
    aFor binding to the FcγRs the variants were made in the E27 background (anti-IgE).
    The results are presented as a percentage of the wild type.
  • A further residue involved in binding human C1q was identified using the methods described in the present example. The residue D270 was replaced with lysine and valine to generate variants D270K and D270V, respectively. These variants both showed decreased binding to human C1q (FIG. 6) and were non-lytic (FIG. 7). The two variants bound the CD20 antigen normally and recruited ADCC.
  • EXAMPLE 3 Variants with Improved C1q Binding
  • The following study shows that substitution of residues at positions K326, A327, E333 and K334 resulted in variants with at least about a 30% increase in binding to C1q when compared to the wild type antibody. This indicated K326, A327, E333 and K334 are potential sites for improving the efficacy of antibodies by way of the CDC pathway. The aim of this study was to improve CDC activity of an antibody by increasing binding to C1q. By site directed mutagenesis at K326 and E333, several variants with increased binding to C1q were constructed. The residues in order of increased binding at K326 are K<V<E<A<G<D<M<W, and the residues in order of increased binding at E333 are E<Q<D<V<G<A<S. Four variants, K326M, K326D, K326E and E333S were constructed with at least a two-fold increase in binding to C1q when compared to wild type. Variant K326W displayed about a five-fold increase in binding to C1q.
  • Variants of the wild type C2B8 antibody were prepared as described above in Example 2. A further control antibody, wild type C2B8 produced in Chinese hamster ovary (CHO) cells essentially as described in U.S. Pat. No. 5,736,137, was included in a C1q binding ELISA to confirm that wt C2B8 produced in the 293 kidney cell line had the same C1q binding activity as the CHO-produced antibody (see “CHO-wt-C2B8” in FIG. 8). The C1q binding ELISA, CDC assay, and CD20 binding potency assay in this example were performed as described in Example 2 above.
  • As shown in FIG. 8, alanine substitution at K326 and E333 in C2B8 resulted in variants with about a 30% increase in binding to C1q.
  • Several other single point variants at K326 and E333 were constructed and assessed for their ability to bind C1q and activate complement. All the variants constructed bound normally to the CD20 antigen.
  • With respect to K326, the other single point variants constructed were K326A, K326D, K326E, K326G, K326V, K326M and K326W. As shown in FIG. 9, these variants all bound to C1q with a better affinity than the wild type antibody. K326W, K326M, K326D and K326E showed at least a two-fold increase in binding to C1q (Table 5). Among the K326 variants, K326W had the best affinity for C1q.
  • TABLE 5
    Variant EC50 value
    Wild type 1.53
    K326V 1.30
    K326A 1.03
    K326E 1.08
    K326G 0.95
    K326D 0.76
    K326M 0.67
    K326W 0.47
    E333S 0.81
    E333A 0.98
    E333G 1.14
    E333V 1.18
    E333D 1.22
    E333Q 1.52
    K334A 1.07
  • Substitutions with hydrophobic as well as charged residues resulted in variants with increased binding to C1 q. Even substitution with glycine which is known to impart flexibility to a chain and is well conserved in nature, resulted in a variant with higher affinity for C1q when compared to the wild type. It would appear that any amino acid substitution at this site would result in a variant with higher affinity for C1q. As assessed from the three-dimensional structure, K326 and E333 are in the vicinity of the core C1q binding sites (FIG. 10).
  • In addition to alanine, E333 was also substituted with other amino acid residues. These variants, E333S, E333G, E333V, E333D, and E333Q, all had increased binding to C1q when compared to the wild type (FIG. 11). As shown in Table 5, the order of binding affinity for C1q was as follows: E333S>E333A>E333G>E333V>E333D>E333Q. Substitutions with amino acid residues with small side chain volumes, i.e. serine, alanine and glycine, resulted in variants with higher affinity for C1q in comparison to the other variants, E333V, E333D and E333Q, with larger side chain volumes. The variant E333S had the highest affinity for C1q, showing a two-fold increase in binding when compared to the wild type. Without being bound to any one theory, this indicates the effect on C1q binding at 333 may also be due in part to the polarity of the residue.
  • Double variants were also generated. As shown in FIGS. 12 and 13, double variants K326M-E333S and K326A-E333A were at least three-fold better at binding human C1q than wild type C2B8 (FIG. 12) and at least two-fold better at mediating CDC compared to wild type C2B8 (FIG. 13). Additivity indicates these are independently acting variants.
  • As shown in FIG. 14, a further variant with improved C1q binding (50% increase) was made by changing A327 in a human IgG1 constant region to glycine. Conversely, in a human IgG2 constant region, changing G327 to alanine reduced C1q binding of the IgG2 antibody.
  • EXAMPLE 4 Identification of FcR Binding Sites in Human IgG Antibodies
  • In the present study, the effect of mutating various Fc region residues of an IgG1 antibody with respect to binding FcγRI, FcγRIIA, FcγRIIB and FcγRIIIIA as well as FcRn was evaluated. Antibody variants with improved as well as diminished FcR binding were identified.
  • Materials and Methods
  • Construction of IgG1 Variants: Recombinant anti-IgE E27 having the light chain and heavy chain sequences in FIGS. 4A and 4B, respectively, was used as the parent antibody in the following experiments. This antibody binds the antigen IgE and has a non-A allotype IgG1 Fc region. By site directed mutagenesis (Kunkel et al., Proc. Natl. Acad. Sci. USA 82:488-492 (1985)), variants of the Fc region in the heavy chain of the above parent antibody were constructed. The heavy and light chain plasmids were co-transfected into an adenovirus transformed human embryonic kidney cell line as previously described (Werther et al., J. Immunol. 157:4986 (1996)). The media was changed to serum-free 24 hours after transfection and the secreted antibody was harvested after five days. The antibodies were purified by Protein G SEPHAROSE® (Pharmacia), buffer exchanged and concentrated to 0.5 ml with PBS using a Centricon-30 (Amicon), and stored at 4° C. Concentration was determined by adsorption at 280 nm using extinction coefficients derived by amino acid composition analysis.
  • High Affinity FcRIA Binding ELISA: FcγRIA was expressed as a GST fusion of His6 tagged extracellular domain in 293 cells and purified by Ni-NTA column chromatography.
  • To purify FcγRIA, supernatant from transfected 293 cells was removed after three days. Protease inhibitors were added; 50 μL Aprotinin (Sigma)/50 mL supernatant, and PMSF (1 mM). Supernatants were concentrated to 10 mL in a stirred cell (Amicon), and dialyzed overnight at 4° C. against 1 liter column buffer (50 mM Tris pH 8.0, 20 mM Imidazole, 300 mM NaCl). Additional dialysis was done the following morning against fresh column buffer for 4 hours at 4° C. The solution was loaded on to a 1 mL Ni++ column (NTA super flow resin, Qiagen) previously equilibrated with 10 mL column buffer. Columns were washed with 10 mL column buffer, and protein was eluted with 2.5 mL elution buffer (50 mM Tris pH 8.0, 250 mM Imidazole, 300 mM NaCl). Protein was concentrated to 0.5 mL and buffer exchanged into PBS. Concentrations were determined by adsorption at 280 nm using an extinction coefficient derived by amino acid composition analysis.
  • Purified receptors were coated onto Nunc F96 maxisorb plates (cat no. 439545) at approximately 150 ng per well by adding 100 μL of receptor at 1.5 μg/mL in PBS and incubated for 24 hours at 4° C. Prior to assay, plates were washed 3× with 250 μL of wash buffer (phosphate buffered saline pH 7.4 containing 0.5% TWEEN 20®) and blocked with 250 μL of assay buffer (50 mM tris buffered saline, 0.05% TWEEN 20®, 0.5% RIA grade bovine albumin (Sigma A7888), and 2mM EDTA pH 7.4).
  • 100 μL of E27 was added to the first four wells of the FcγRIA subunit coated plated at a concentration of 10 μg/mL. 80 μL of assay buffer was added to the next four well followed by 20 μL of the 10 μg/mL E27 IgG to give a final concentration of 2 μg/mL. Plates were incubated at 25° C. for 2 hours on an orbital shaker.
  • For detection, plates were washed 5× with wash buffer to remove unbound antibody. IgG binding to GST-FcγRIA was detected by adding 100 μL horse radish peroxidase (HRP) conjugated protein G (BIORAD) at 1:5000. HRP conjugates were incubated for 1.5 hours at 25° C. on an orbital shaker. Plates were washed ×5 with wash buffer to remove unbound HRP conjugate. Binding was detected by adding 100 μL of substrate solution (0.4 mg/mL o-phenylenedaimine dihydrochloride, Sigma P6912, 6 mM H2O2 in PBS) and incubating for 10 minutes at 25° C. Enzymatic reaction was stopped by the addition of 100 μL of 4.5 N H2SO4 and colorimetric product was measured at 490 nm on a 96 well plate densitometer (Molecular Devices).
  • Binding of E27 variants at IgG concentration of 2 μg/mL was expressed as a ratio of wild type E27.
  • FcγRIA THP-1 Assay. 100 μL of E27 was added to the first three wells of a serocluster plate (Costar) at a concentration of 20 μg/mL in assay buffer (1× PBS, 0.1% BSA, 0.01% NaN3). 92.5 of assay buffer was added to the next three wells followed by 7.5 μL of the 20 μg/mL E27 IgG to give a final concentration of 1.5 μg/mL. To each well, 100 μL of THP-1 cells were added at a concentration of 5 million cells/mL in FACS assay buffer. The plate is incubated on ice for 30 minutes
  • For detection, cells were washed 2× with assay buffer to remove unbound antibody. IgG binding FcγRIA was detected by adding 100 μL FITC conjugated F(ab′)2 fragment of goat anti-human IgG heavy chain specific. (Jackson Immunoresearch) at 1:200. FITC conjugates were incubated with cells for 30 minutes on ice. Cells were washed ×3 with assay buffer to remove unbound FITC conjugate. Cells were stained with P.I. (SIGMA) at 2.5 μg/mL and analyzed by flow cytometry.
  • Binding of E27 variants at IgG concentration of 1.5 μg/mL was expressed as a ratio of wild type E27.
  • Data from the plate assay (FcγRIA ELISA) and cell-based assay (FcγRIA THP-1 assay) was averaged to arrive at an FcγRIA-binding activity.
  • Low Affinity FcγR Binding ELISAs: FcγRIIA, FcγRIIB and FcγRIIIA binding ELISAs were performed as described in Example 1 above, with detection of the stable hexamer (consisting of three anti-IgE molecules and three IgE molecules).
  • FcRn Binding ELISA: For measuring FcRn binding activity of IgG variants, ELISA plates were coated with 2 μg/ml streptavidin (Zymed, South San Francisco) in 50 mM carbonate buffer, pH 9.6, at 4° C. overnight and blocked with PBS-0.5% BSA, pH 7.2 at room temperature for one hour. Biotinylated FcRn (prepared using biotin-X-NHS from Research Organics, Cleveland, Ohio and used at 1-2 μg/ml) in PBS-0.5% BSA, 0.05% polysorbate 20, pH 7.2, was added to the plate and incubated for one hour. Two fold serial dilutions of IgG standard (1.6-100 ng/ml) or variants in PBS-0.5% BSA, 0.05% polysorbate 20, pH 6.0, were added to the plate and incubated for two hours. Bound IgG was detected using peroxidase labeled goat F(ab')2 anti-human IgG F(ab′)2 in the above pH 6.0 buffer (Jackson ImmunoResearch, West Grove, Pa.) followed by 3,3′,5,5′-tetramethyl benzidine (Kirgaard & Perry Laboratories) as the substrate. Plates were washed between steps with PBS-0.05% TWEEN 20® at either pH 7.2 or 6.0. Absorbance was read at 450 nm on a Vmax plate reader (Molecular Devices, Menlo Park, Calif.). Titration curves were fit with a four-parameter nonlinear regression curve-fitting program (KaleidaGraph, Synergy software, Reading, Pa.). Concentrations of IgG variants corresponding to the mid-point absorbance of the titration curve of the standard were calculated and then divided by the concentration of the standard corresponding to the mid-point absorbance of the standard titration curve.
  • In Vitro ADCC Assay: To prepare chromium 51-labeled target cells, tumor cell lines were grown in tissue culture plates and harvested using sterile 10 mM EDTA in PBS. SK-BR-3 cells, a 3+HER2-overexpressing human breast cancer cell line, were used as targets in all assays. The detached cells were washed twice with cell culture medium. Cells (5×106) were labeled with 200 μCi of chromium51 (New England Nuclear/DuPont) at 37° C. for one hour with occasional mixing. Labeled cells were washed three times with cell culture medium, then were resuspended to a concentration of 1×105 cells/mL. Cells were used either without opsonization, or were opsonized prior to the assay by incubation with rhuMAb HER2 wildtype (HERCEPTIN®) or seven Fc mutants (G14, G18, G17, G36, G30, G31 and G34) at 100 ng/mL and 1.25 ng/mL in PBMC assay or 20 ng/mL and 1 ng/mL in NK assay.
  • Peripheral blood mononuclear cells were prepared by collecting blood on heparin from normal healthy donors and dilution with an equal volume of phosphate buffered saline (PBS). The blood was then layered over LYMPHOCYTE SEPARATION MEDIUM® (LSM: Organon Teknika) and centrifuged according to the manufacturer's instructions. Mononuclear cells were collected from the LSM-plasma interface and were washed three times with PBS. Effector cells were suspended in cell culture medium to a final concentration of 1×107 cells/mL.
  • After purification through LSM, natural killer (NK) cells were isolated from PBMCs by negative selection using an NK cell isolation kit and a magnetic column (Miltenyi Biotech) according to the manufacturer's instructions. Isolated NK cells were collected, washed and resuspended in cell culture medium to a concentration of 2×106cells/mL. The identity of the NK cells was confirmed by flow cytometric analysis.
  • Varying effector:target ratios were prepared by serially diluting the effector (either PBMC or NK) cells two-fold along the rows of a microtiter plate (100 μL final volume) in cell culture medium. The concentration of effector cells ranged from 1.0×107/mL to 2.0×104/mL for PBMC and from 2.0×106/mL to 3.9×103/mL for NK. After titration of effector cells, 100 μL of chromium 51-labeled target cells (opsonized or nonoponsonized) at 1×105 cells/mL were added to each well of the plate. This resulted in an initial effector:target ratio of 100:1 for PBMC and 20:1 for NK cells. All assays were run in duplicate, and each plate contained controls for both spontaneous lysis (no effector cells) and total lysis (target cells plus 100 μL) 1% sodium dodecyl sulfate, 1 N sodium hydroxide). The plates were incubated at 37° C. for 18 hours, after which the cell culture supernatants were harvested using a supernatant collection system (Skatron Instrument, Inc.) and counted in a Minaxi auto-gamma 5000 series gamma counter (Packard) for one minute. Results were then expressed as percent cytotoxicity using the formula:

  • % Cytotoxicity=(sample cpm-spontaneous lysis)/(total lysis-spontaneous lysis)×100
  • Four-parameter curve-fitting was then used to evaluate the data (KaleidaGraph 3.0.5).
  • Results
  • A variety of antibody variants were generated which had FcR binding activity that differed from the parent antibody. The FcR binding data for the variants generated is shown in Tables 6 and 7 below. An additional variant, T307Q, also displayed improved FcRn binding compared to E27 parent antibody.
  • TABLE 6
    CH2 DOMAIN VARIANTS
    Res#EU FcRn FcγRI FcγRIIA FcγRIIB FcγRIIIA
    IG2 (Kabat) mean sd n mean sd n mean sd mean sd mean sd
    REDUCED BINDING TO ALL FcγR
     1 233-236 0.54 (0.20) 3 0.12 (0.06) 6 0.08 (0.01) 0.12 (0.01) 0.04 (0.02) n = 2
    ELLG > PVA-
     2 P238A(251) 1.49 (0.17) 3 0.60 (0.05) 5 0.38 (0.14) 0.36 (0.15) 0.07 (0.05) n = 4
    14 D265A(278) 1.23 (0.14) 4 0.14 (0.04) 6 0.07 (0.01) 0.13 (0.05) 0.09 (0.06) n = 4
    17 E269A(282) 1.05 0.52 (0.03) 6 0.65 (0.18) 0.75 (0.29) 0.45 (0.13) n = 5
    18 D270A(283) 1.05 0.76 (0.12) 6 0.06 (0.01) 0.11 (0.05) 0.14 (0.04) n = 5
    58 N297A(314) 0.80 (0.18) 8 0.15 (0.06) 7 0.05 (0.00) 0.10 (0.02) 0.03 (0.01) n = 3
    52 A327Q(346) 0.97 0.63 (0.15) 7 0.13 (0.03) 0.14 (0.03) 0.06 (0.01) n = 4
    64 P329A(348) 0.80 0.48 (0.10) 6 0.08 (0.02) 0.12 (0.08) 0.21 (0.03) n = 4
    REDUCED BINDING TO FcγRII & FcγRIII
     3 S239A(252) 1.06 0.81 (0.09) 7 0.73 (0.25) 0.76 (0.36) 0.26 (0.08) n = 3
    33 E294A(311) 0.75 0.90 (0.08) 4 0.87 (0.19) 0.63 (0.17) 0.66 (0.14) n = 5
    34 Q295A(312) 0.79 1.00 (0.11) 4 0.62 (0.20) 0.50 (0.24) 0.25 (0.09) n = 5
    39 V303A(322) 1.26 (0.21) 3 0.91 (0.11) 5 0.86 (0.10) 0.65 (0.17) 0.33 (0.09) n = 8
    IMPROVED BINDING TO FcγRII & FcγRIII
    11 T256A(269) 1.91 (0.43) 6 1.14 (0.14) 4 1.41 (0.27) 2.06 (0.66) 1.32 (0.18) n = 9
    30 K290A(307) 0.79 (0.14) 3 1.01 (0.08) 4 1.29 (0.21) 1.40 (0.18) 1.28 (0.21) n = 7
    44 D312A(331) 1.50 (0.06) 4 1.01 (0.12) 5 1.20 (0.24) 1.19 (0.07) 1.23 (0.14) n = 3
    51 K326A(345) 1.03 1.04 (0.05) 4 1.26 (0.21) 1.49 (0.27) 1.22 (0.28) n = 5
    197  A330(349)K 1.28 1.25 1.28 n = 1
    273  A339(359)T 1.23 1.11 1.23 1.42 n = 1
    EFFECT FcγRII
    10 R255A(268) 0.59 (0.19) 4 1.26 (0.26) 8 1.30 (0.20) 1.59 (0.42) 0.98 (0.18) n = 5
    12 E258A(271) 1.18 1.18 (0.13) 4 1.33 (0.22) 1.65 (0.38) 1.12 (0.12) n = 5
    15 S267A(280) 1.08 1.20 (0.14) 4 1.64 (0.18) 2.06 (0.35) 1.14 (0.25) n = 7
    16 H268A(281) 1.02 (0.22) 3 1.05 (0.11) 4 1.22 (0.14) 1.45 (0.23) 0.52 (0.09) n = 12
    19 E272A(285) 1.34 (0.24) 4 1.04 (0.06) 4 1.24 (0.11) 1.58 (0.19) 0.74 (0.12) n = 4
    21 N276A(289) 1.15 (0.21) 3 1.05 (0.14) 4 1.29 (0.20) 1.34 (0.40) 0.95 (0.04) n = 4
    23 D280A(295) 0.82 0.97 (0.06) 4 1.34 (0.14) 1.60 (0.31) 1.09 (0.20) n = 10
    25 E283A(300) 0.71 0.97 (0.03) 4 1.24 (0.23) 1.20 (0.17) 1.01 (0.14) n = 5
    26 H285A(302) 0.85 0.96 (0.07) 4 1.26 (0.12) 1.23 (0.15) 0.87 (0.04) n = 4
    27 N286A(303) 1.24 (0.04) 2 0.94 (0.20) 13 1.28 (0.23) 1.39 (0.14) 1.03 (0.08) n = 5
    31 R292A(309) 0.81 (0.18) 4 0.93 (0.02) 4 0.27 (0.14) 0.18 (0.07) 0.90 (0.18) n = 9
    36 S298A(317) 0.80 1.10 (0.04) 3 0.40 (0.08) 0.21 (0.11) 1.30 (0.18) n = 12
    38 R301A(320) 0.86 1.06 (0.10) 4 1.12 (0.12) 1.26 (0.14) 0.21 (0.08) n = 6
    38B R301M(320) 0.88 1.06 (0.12) 4 1.29 (0.17) 1.56 (0.12) 0.48 (0.21) n = 4
    40 V305A(324) 1.46 (0.48) 6 1.04 (0.19) 10 1.12 (0.12) 1.23 (0.22) 0.84 (0.15) n = 4
    41 T307A(326) 1.81 (0.32) 6 0.99 (0.14) 4 1.19 (0.37) 1.35 (0.33) 1.12 (0.18) n = 12
    42 L309A(328) 0.63 (0.18) 4 0.93 (0.18) 6 1.13 (0.08) 1.26 (0.12) 1.07 (0.20) n = 3
    45 N315A(334) 0.76 (0.14) 3 1.27 (0.36) 6 1.15 (0.06) 1.30 (0.17) 1.07 (0.21) n = 5
    48 K320A(339) 1.10 0.98 (0.09) 5 1.12 (0.11) 1.22 (0.05) 0.87 (0.17) n = 4
    49 K322A(341) 0.98 0.94 (0.05) 6 1.15 (0.11) 1.27 (0.24) 0.61 (0.14) n = 5
    50 S324A(343) 1.08 0.95 (0.05) 4 0.82 (0.22) 0.70 (0.12) 1.12 (0.17) n = 4
    65 P331A(350) 0.85 1.30 (0.34) 8 1.29 (0.14) 1.47 (0.28) 1.03 (0.19) n = 3
    54 E333A(352) 1.03 (0.01) 2 0.98 (0.15) 5 0.92 (0.12) 0.76 (0.11) 1.27 (0.17) n = 10
    56 T335A(354) 0.98 1.00 (0.05) 4 0.79 (0.22) 0.65 (0.26) 0.92 (0.54) n = 3
    57 S337A(356) 1.03 1.17 (0.23) 3 1.22 (0.30) 1.26 (0.06) 0.94 (0.18) n = 4
    EFFECT FcγRIII
     5 K248A(261) 0.87 0.95 (0.05) 5 1.06 (0.12) 1.01 (0.12) 0.71 (0.05) n = 4
     6 D249A(262) 0.93 1.04 (0.10) 4 1.02 (0.12) 0.94 (0.02) 0.66 (0.07) n = 5
     7 M252A(265) 0.64 (0.13) 4 0.99 (0.10) 5 1.01 (0.18) 1.15 (0.22) 0.65 (0.17) n = 6
     9 S254A(267) <0.10 0.96 (0.08) 4 0.97 (0.24) 1.15 (0.38) 0.73 (0.14) n = 3
    16 H268A(281) 1.02 (0.22) 3 1.05 (0.11) 4 1.22 (0.14) 1.45 (0.23) 0.52 (0.09) n = 12
    19 E272A(285) 1.34 (0.24) 4 1.04 (0.06) 4 1.24 (0.11) 1.58 (0.19) 0.74 (0.12) n = 4
    22 Y278A(291) 0.90 0.96 (0.02) 4 1.11 (0.08) 1.10 (0.16) 0.67 (0.11) n = 4
    29 T289A(306) 0.86 0.93 (0.03) 4 0.96 (0.33) 0.83 (0.22) 0.62 (0.19) n = 7
    32 E293A(310) 0.85 1.11 (0.07) 4 1.08 (0.19) 1.07 (0.20) 0.31 (0.13) n = 6
    35 Y296F(313) 0.79 1.07 (0.12) 4 0.97 (0.26) 0.84 (0.18) 0.52 (0.09) n = 5
    36 S298A(317) 0.80 1.10 (0.04) 3 0.40 (0.08) 0.21 (0.11) 1.30 (0.18) n = 12
    38 R301A(320) 0.86 1.06 (0.10) 4 1.12 (0.12) 1.26 (0.14) 0.21 (0.08) n = 6
    38B R301M(320) 0.88 1.06 (0.12) 4 1.29 (0.17) 1.56 (0.12) 0.48 (0.21) n = 4
    49 K322A(341) 0.98 0.94 (0.05) 6 1.15 (0.11) 1.27 (0.24) 0.61 (0.14) n = 5
    54 E333A(352) 1.03 (0.01) 2 0.98 (0.15) 5 0.92 (0.12) 0.76 (0.11) 1.27 (0.17) n = 10
    55 K334A(353) 1.05 (0.03) 2 1.10 (0.06) 4 1.01 (0.15) 0.90 (0.12) 1.39 (0.19) n = 17
    NO EFFECT ON FcγR
     4 K246A(259) 1.03 0.94 (0.06) 4 1.02 (0.10) 0.92 (0.15) 1.14 (0.38) n = 4
     4B K246M(259) 0.69 0.83 (0.05) 5 0.83 (0.06) 0.76 (0.05) 0.95 (0.09) n = 3
      5B K248M(261) 0.79 0.95 (0.06) 4 0.89 (0.09) 0.83 (0.04) 1.01 (0.23) n = 3
     8 I253A(266) <0.10 0.96 (0.05) 4 1.14 (0.02) 1.18 (0.06) 1.08 (0.14) n = 3
    13 T260A(273) 1.09 0.93 (0.09) 4 0.89 (0.14) 0.87 (0.10) 0.89 (0.08) n = 4
    20 K274A(287) 1.18 1.02 (0.04) 4 0.86 (0.09) 0.96 (0.10) 1.11 (0.08) n = 3
    24 V282A(299) 1.13 (0.07) 2 0.96 (0.02) 4 1.15 (0.13) 1.15 (0.20) 1.00 (0.18) n = 4
    28 K288A(305) 0.38 (0.12) 5 0.88 (0.15) 15 1.15 (0.26) 1.14 (0.20) 1.06 (0.04) n = 4
    37 Y300F(319) 0.74 (0.10) 2 1.07 (0.15) 4 1.11 (0.04) 1.09 (0.09) 1.01 (0.10) n = 3
    43 Q311A(330) 1.62 (0.25) 4 0.93 (0.05) 4 1.11 (0.06) 1.19 (0.13) 0.93 (0.17) n = 3
    46 K317A(336) 1.44 (0.18) 4 0.92 (0.17) 6 1.13 (0.05) 1.18 (0.27) 1.10 (0.23) n = 4
    47 E318A(337) 0.85 0.92 (0.07) 4 1.04 (0.10) 1.17 (0.23) 1.01 (0.05) n = 3
    53 A330Q(349) 0.76 0.96 (0.10) 4 1.01 (0.12) 1.02 (0.02) 0.75 (0.18) n = 3
  • TABLE 7
    CH3 DOMAIN VARIANTS
    Res#EU FcRn FcγRI FcγRIIA FcγRIIB FcγRIIIA
    IG2 (Kabat) mean sd n mean sd n mean sd mean sd mean sd
    B1 K338(358)A 1.14 0.90 (0.05) 3 0.78 (0.09) 0.63 (0.08) 0.15 (0.01) n = 2
    B1A K338(358)M 0.78 0.99 (0.08) 3 0.99 (0.13) 0.93 (0.15) 0.49 (0.04) n = 2
    B2 K340(360)A 1.02 1.04 (0.07) 3 1.05 (0.18) 0.96 (0.20) 0.84 (0.11) n = 2
    B2A K340(360)M 1.20 1.17 (0.11) 3 1.10 (0.12) 1.20 (0.19) 0.75 (0.12) n = 2
    B3 Q342(363)A 1.09 1.13 (0.11) 3 1.01 (0.10) 1.09 (0.23) 0.98 (0.10) n = 2
    B4 R344(365)A 0.77 1.04 (0.08) 3 0.89 (0.14) 0.91 (0.04) 0.97 (0.07) n = 4
    B5 E345(366)A 1.18 1.06 (0.05) 3 1.03 (0.10) 0.98 (0.10) 0.97 (0.13) n = 4
    B6 Q347(368)A 0.95 1.04 (0.06) 3 1.00 (0.03) 0.92 (0.02) 1.04 (0.12) n = 4
    B7 R355(376)A 1.06 1.09 (0.07) 3 0.84 (0.09) 0.87 (0.11) 0.98 (0.09) n = 4
    B8 E356(377)A 1.21 (0.11) 2 1.05 (0.04) 3 0.90 (0.02) 0.99 (0.13) 0.92 (0.03) n = 3
    B9 M358(381)A 0.96 1.06 (0.07) 3 1.11 (0.06) 1.16 (0.25) 0.91 (0.09) n = 3
    B10 T359(382) 1.04 1.04 (0.05) 3 1.13 (0.10) 1.15 (0.04) 1.23 (0.26) n = 3
    B11 K360(383)A 1.30 (0.08) 4 1.02 (0.04) 3 1.12 (0.10) 1.12 (0.08) 1.23 (0.16) n = 6
    B12 N361(384)A 1.16 1.00 (0.03) 3 0.82 (0.07) 0.82 (0.12) 1.08 (0.06) n = 3
    B13 Q362(385)A 1.25 (0.24) 3 1.00 (0.04) 3 1.03 (0.10) 1.02 (0.03) 1.03 (0.16) n = 4
    B14 Y373(396)A 0.86 0.98 (0.07) 3 0.84 (0.11) 0.75 (0.08) 0.67 (0.04) n = 5
    B15 S375(398)A 1.17 (0.19) 5 0.95 (0.02) 3 1.08 (0.06) 1.14 (0.11) 1.04 (0.05) n = 6
    B16 D376(399)A 1.45 (0.36) 4 1.00 (0.05) 3 0.80 (0.16) 0.68 (0.14) 0.55 (0.10) n = 5
    B17 A378(401)Q 1.32 (0.13) 3 1.06 (0.05) 3 1.40 (0.17) 1.45 (0.17) 1.19 (0.17) n = 5
    B18 E380(405)A 2.19 (0.29) 6 1.04 (0.06) 3 1.18 (0.01) 1.07 (0.05) 0.92 (0.12) n = 2
    B19 E382(407)A 1.51 (0.18) 4 1.06 (0.03) 3 0.95 (0.11) 0.84 (0.04) 0.76 (0.17) n = 3
    B20 S383(408)A 0.74 1.03 (0.03) 3 0.92 (0.04) 0.94 (0.05) 0.88 (0.07) n = 3
    B21 N384(410)A 0.88 1.00 (0.01) 3 1.05 (0.19) 1.10 (0.18) 0.96 (0.18) n = 8
    B22 Q386(414)A 0.70 (0.10) 2 1.14 (0.08) 3 1.08 (0.13) 1.19 (0.25) 0.98 (0.14) n = 9
    B23 E388(416)A 0.64 (0.12) 2 1.15 (0.09) 3 0.87 (0.03) 0.94 (0.09) 0.62 (0.04) n = 3
    B24 N389(417)A 0.73 1.00 (0.02) 3 0.98 (0.15) 0.81 (0.04) 0.75 (0.02) n = 3
    B25 N390(418)A 0.87 1.06 (0.04) 3 0.99 (0.10) 0.94 (0.02) 0.87 (0.09) n = 3
    B26A Y391(419)A 1.14 1.00 (0.08) 3 0.97 (0.10) 0.94 (0.02) 0.86 (0.05) n = 3
    B26B Y391(419)F 0.81 (0.10) 2 1.00 (0.01) 3 1.05 (0.12) 1.11 (0.08) 1.01 (0.15) n = 5
    B27 K392(420)A 0.97 1.01 (0.08) 3 0.92 (0.20) 0.94 (0.01) 0.79 (0.22) n = 3
    B28 L398(426)A 0.94 (0.04) 2 1.13 (0.15) 6 1.17 (0.11) 1.20 (0.08) 0.94 (0.04) n = 3
    B29 S400(428)A 0.64 (0.07) 3 1.10 (0.09) 3 0.95 (0.04) 0.99 (0.08) 0.83 (0.07) n = 2
    B30 D401(430)A 1.10 (0.09) 3 1.13 (0.16) 6 1.11 (0.12) 1.19 (0.11) 0.97 (0.10) n = 5
    B31 D413(444)A 1.21 (0.07) 2 1.00 (0.01) 3 0.83 (0.08) 0.84 (0.06) 0.90 (0.16) n = 2
    B32 K414(445)A 1.02 1.00 (0.04) 3 0.64 (0.15) 0.58 (0.18) 0.82 (0.27) n = 3
    B33 S415(446)A 0.44 1.04 (0.03) 3 0.90 (0.11) 0.88 (0.05) 0.86 (0.18) n = 2
    B34 R416(447)A 1.08 0.96 (0.04) 3 0.68 (0.05) 0.80 (0.05) 0.71 (0.08) n = 2
    B35 Q418(449)A 0.77 (0.03) 2 0.98 (0.01) 3 1.00 (0.01) 0.96 (0.02) 0.96 (0.05) n = 2
    B36 Q419(450)A 0.76 (0.01) 2 0.97 (0.02) 3 0.68 (0.09) 0.63 (0.07) 0.86 (0.08) n = 3
    B37 N421(452)A 0.98 0.99 (0.01) 3 0.90 (0.03) 0.81 (0.0) 0.87 (0.12) n = 2
    B38 V422(453)A 1.01 0.98 (0.02) 3 0.89 (0.0) 0.83 (0.05) 0.83 (0.12) n = 2
    B39 S424(455)A 1.41 (0.14) 3 0.98 (0.03) 3 1.04 (0.06) 1.02 (0.02) 0.88 (0.09) n = 2
    B40 E430(461)A 0.93 (0.03) 2 1.05 (0.02) 3 1.24 (0.11) 1.28 (0.10) 1.20 (0.18) n = 5
    B41 H433(464)A 0.41 (0.14) 2 0.98 (0.03) 3 0.92 (0.18) 0.79 (0.18) 1.02 (0.15) n = 3
    B42 N434(465)A 3.46 (0.37) 7 1.00 (0.04) 3 0.97 (0.07) 0.98 (0.13) 0.74 (0.12) n = 5
    B43 H435(466)A <0.10 4 1.25 (0.09) 3 0.77 (0.05) 0.72 (0.05) 0.78 (0.03) n = 3
    B44 Y436(467)A <0.10 2 0.99 (0.02) 2 0.93 (0.05) 0.91 (0.06) 0.91 (0.15) n = 3
    B45 T437(468)A 0.99 (0.07) 1.00 (0.02) 3 1.12 (0.18) 1.00 (0.22) 0.77 (0.19) n = 5
    B46 Q438(469)A 0.79 (0.05) 2 1.02 (0.05) 3 0.80 (0.10) 0.72 (0.16) 1.01 (0.17) n = 5
    B47 K439(470)A 0.70 (0.04) 2 0.98 (0.04) 3 0.78 (0.16) 0.68 (0.22) 0.86 (0.19) n = 4
    B48 S440(471)A 0.99 1.01 (0.02) 3 1.10 (0.15) 1.11 (0.26) 0.93 (0.01) n = 3
    B49 S442(473)A 0.86 1.02 (0.02) 3 0.98 (0.08) 0.91 (0.11) 0.95 (0.10) n = 5
    B50 S444(475)A 0.80 1.01 (0.02) 3 1.07 (0.03) 1.03 (0.03) 0.88 (0.12) n = 2
    B51 K447(478)A 0.62 (0.12) 3 1.02 (0.03) 3 0.95 (0.05) 0.91 (0.05) 0.84 (0.09) n = 2
    Variants with increased binding to a FcγR generally had binding values ≧1.20 as determined in this Example and those with reduced binding to a FcγR generally had binding values ≦0.80 as determined in this Example. Variants with increased binding to FcRn generally had binding values ≧1.30 as determined in this Example and those with reduced binding to FcRn generally had binding values ≦0.70 as determined in this Example.
    Aside from alanine variants, various non-alanine substitution variants were made, and the FcR binding activity of those variants is summarized in the following table.
  • TABLE 8
    NON-ALANINE VARIANTS
    Res#EU FcRn FcγRI FcγRIIA FcγRIIB FcγRIIIA
    IG2 (Kabat) mean sd n mean sd n mean sd mean sd mean sd
    222 D249(262)E 0.97 0.99 0.84 n = 1
    176 T256(269)G 1.10 (0.03) 1.06 (0.07) 0.96 (0.27) n = 2
    254 T256(269)N 1.03 0.89 1.13 n = 1
    157 D265(278)N 0.02 (0.01) 0.03 (0.01) 0.02 (0.01) n = 3
    158 D265(278)E 0.11 (0.04) 0.03 (0.01) 0.02 (0.01) n = 3
    189 S267(280)G R131 1.21 (0.05) 0.97 (0.16) 0.09 (0.02) n = 3
    H131 0.59 (0.09) n = 3
    84 H268(281)N 1.33 1.41 0.56 n = 1
    85 H268(281)S 1.35 1.38 0.81 n = 1
    87 H268(281)Y 1.19 1.29 0.76 n = 1
    168 E269(282)D 0.89 (0.10) 0.73 (0.07) 1.13 (0.21) n = 2
    169 E269(282)Q 0.08 (0.01) 0.16 (0.00) 0.28 (0.03) n = 2
    92 D270(283)N 0.06 (0.01) 0.10 (0.02) 0.04 (0.00) n = 2
    93 D270(283)E 0.55 (0.05) 0.38 (0.05) 1.17 (0.01) n = 2
    223 E272(285)Q 1.93 1.81 0.82 n = 1
    224 E272(285)N 0.43 0.23 0.50 n = 1
    167 K274(287)Q 0.86 0.94 0.62 n = 1
    165 N276(289)K 0.81 0.77 0.61 n = 1
    233 N276(289)Q 1.09 0.79 0.91 n = 1
    79 D280(295)N 1.26 (0.07) 1.38 (0.04) 1.13 (0.13) n = 2
    149 D280(295)S 1.07 (0.06) 1.04 (0.08) 1.09 (0.06) n = 2
    226 E283(300)Q 1.12 1.24 1.19 n = 1
    227 E283(300)S 1.03 1.07 0.85 n = 1
    228 E283(300)N 1.18 1.28 0.94 n = 1
    229 E283(300)D 1.14 1.23 0.95 n = 1
    23 N286(303)Q 1.52 1.13 0.96 n = 1
    237 N286(303)S 1.72 1.38 1.32 n = 1
    238 N286(303)D 1.41 1.23 0.98 n = 1
    73 K290(307)Q 1.17 1.26 1.40 n = 1
    75 K290(307)S 1.27 1.34 1.26 n = 1
    77 K290(307)E 1.14 1.10 1.20 1.30 n = 1
    78 K290(307)R 1.25 1.05 1.15 1.08 n = 1
    177 K290(307)G 1.07 1.21 1.23 n = 1
    80 R292(309)K 0.71 (0.17) 0.75 (0.10) 1.15 (0.18) n = 3
    81 R292(309)H 0.21 (0.09) 0.12 (0.01) 0.92 (0.08) n = 2
    82 R292(309)Q 0.47 (0.12) 0.25 (0.06) 0.45 (0.09) n = 3
    83 R292(309)N 0.54 (0.16) 0.29 (0.07) 0.88 (0.02) n = 3
    144 E293(310)Q 0.85 (0.03) 0.77 (0.13) 0.99 (0.04) n = 2
    145 E293(310)D 0.90 (0.02) 0.88 (0.07) 0.37 (0.07) n = 2
    147 E293(310)K 1.13 (0.04) 1.31 (0.17) 0.72 (0.08) n = 4
    173 E294(311)Q 1.01 0.95 0.84 n = 1
    174 E294(311)D 0.37 0.26 0.14 n = 1
    185 Y296(313)H 0.90 0.81 0.92 n = 1
    186 Y296(313)W 0.96 0.93 1.38 n = 1
    70 S298(317)G 0.87 (0.17) 0.63 (0.33) 0.46 (0.09) n = 4
    71 S298(317)T 0.41 (0.21) 0.40 (0.19) 0.89 (0.20) n = 3
    72 S298(317)N 0.08 (0.01) 0.16 (0.03) 0.06 (0.01) n = 2
    218 S298(317)V 0.11 (0.06) 0.17 (0.01) 0.33 (0.19) n = 3
    219 S298(317)L 1.14 (0.12) 1.42 (0.31) 0.34 (0.04) n = 3
    150 V303(322)L 0.89 (0.05) 0.73 (0.10) 0.76 (0.09) n = 4
    151 V303(322)T 0.64 (0.11) 0.34 (0.05) 0.20 (0.05) n = 4
    217 E318(337)K 1.03 1.08 0.72 n = 1
    172 K320(339)R 0.71 0.66 0.68 n = 1
    202 K320(339)M 1.34 1.40 1.27 n = 1
    204 K320(339)Q 1.23 1.12 1.17 n = 1
    205 K320(339)E 1.29 1.34 1.12 n = 1
    235 K320(339)R 1.24 0.95 0.86 n = 1
    155 K322(341)R 0.87 (0.07) 0.87 (0.21) 0.92 (0.15) n = 3
    156 K322(341)Q 0.87 (0.02) 0.92 (0.23) 0.78 (0.18) n = 3
    206 K322(341)E 1.38 1.34 0.81 n = 1
    207 K322(341)N 0.57 0.36 0.04 n = 1
    213 S324(343)N 1.15 1.09 0.97 n = 1
    214 S324(343)Q 0.82 0.83 0.78 n = 1
    215 S324(343)K 0.66 0.37 0.77 n = 1
    216 S324(343)E 0.82 0.73 0.81 n = 1
    208 K326(345)S 1.44 1.62 1.37 n = 1
    209 K326(345)N 1.04 1.00 1.27 n = 1
    210 K326(345)Q 1.36 1.41 1.15 n = 1
    211 K326(345)D 1.68 2.01 1.36 n = 1
    212 K326(345)E 1.34 (0.27) 1.47 (0.33) 1.26 (0.04) n = 2
    131 A327(346)S 0.23 (0.06) 0.22 (0.05) 0.06 (0.01) n = 4
    159 A327(346)G 0.92 (0.09) 0.83 (0.10) 0.36 (0.05) n = 3
    196 A330(349)D 0.18 0.08 0.07 n = 1
    197 A330(349)K 1.28 1.25 1.28 n = 1
    198 P331(350)S 1.00 0.86 0.86 n = 1
    199 P331(350)N 0.86 0.65 0.23 n = 1
    200 P331(350)E 1.06 0.91 0.42 n = 1
    203 P331(350)K 0.94 0.71 0.33 n = 1
    141 E333(352)Q 0.70 (0.05) 0.64 (0.09) 1.10 (0.03) n = 2
    142 E333(352)N 0.59 (0.04) 0.52 (0.07) 0.56 (0.10) n = 2
    143 E333(352)S 0.94 n = 1
    152 E333(352)K 0.85 (0.14) n = 3
    153 E333(352)R 0.75 (0.04) 0.66 (0.03) 0.84 (0.05) n = 2
    154 E333(352)D 1.26 (0.04) n = 3
    178 E333(352)G 0.87 0.76 1.05 n = 1
    179 K334(353)G 0.76 (0.08) 0.60 (0.13) 0.88 (0.22) n = 5
    135 K334(353)R 1.15 (0.09) 1.33 (0.18) 0.68 (0.07) n = 5
    136 K334(353)Q 1.08 (0.11) 1.10 (0.21) 1.31 (0.26) n = 7
    137 K334(353)N 1.16 (0.11) 1.29 (0.30) 1.15 (0.16) n = 7
    138 K334(353)S 1.01 (0.11) 1.03 ( ).05) 1.19 (0.08) n = 3
    139 K334(353)E 0.74 (0.15) 0.72 (0.12) 1.30 (0.09) n = 4
    140 K334(353)D 0.51 (0.09) 0.40 (0.03) 1.13 (0.09) n = 4
    190 K334(353)M 1.18 1.06 1.01 1.35 n = 1
    191 K334(353)Y 1.15 1.08 1.05 1.31 n = 1
    192 K334(353)W 1.16 0.94 0.91 1.07 n = 1
    193 K334(353)H 1.11 1.09 1.07 1.26 n = 1
    220 K334(353)V 1.13 (0.11) 1.09 (0.15) 1.34 (0.18) n = 3
    221 K334(353)L 1.05 1.09 1.38 n = 1
    171 T335(354)Q 0.86 0.79 0.84 n = 1
    194 T335(354)E 1.24 1.30 1.19 n = 1
    195 T335(354)K 1.19 1.14 1.30 n = 1
    273 A339(359)T 1.23 1.11 1.23 1.42 n = 1
  • The following table summarizes the FcR binding activity of various combination variants.
  • TABLE 9
    COMBINATION VARIANTS
    Res#EU FcRn FcγRI FcγRIIA FcγRIIB FcγRIIIA
    IG2 (Kabat) mean sd n mean sd n mean sd mean sd mean sd
    96 S267(280)A 1.41 1.72 0.84 n = 1
    H268(281)A
    134 E333(352)A 0.72 (0.08) 0.63 (0.13) 1.30 (0.12) n = 5
    K334(353)A
    1059 T256(269)A 0.44 (0.03) 0.22 (0.04) 1.41 (0.06) n = 2
    S298(317)A
    1051 T256(269)A 0.47 (0.01) 0.30 (0.03) 1.21 (0.26) n = 2
    D280(295)A
    S298(317)A
    T307(326)A
    106 T256(269)A 0.11 0.08 0.90 n = 1
    D280(295)A
    R292(309)A
    S298(317)A
    T307(326)A
    107 S298(317)A 0.34 (0.05) 0.16 (0.08) 1.53 (0.24) n = 5
    E333(352)A
    109 S298(317)A 0.41 (0.07) 0.19 (0.08) 1.62 (0.34) n = 6
    K334(353)A
    110 S298(317)A 0.35 (0.13) 0.18 (0.08) 1.66 (0.42) n = 11
    E333(352)A
    K334(353)A
    246 S267(280)A 1.62 (0.15) 2.01 (0.45) 1.04 (0.12) n = 2
    E258(271)A
    247 S267(280)A 1.60 (0.18) 1.72 (0.13) 0.88 (0.07) n = 3
    R255(268)A
    248 S267(280)A 1.54 (0.08) 1.96 (0.37) 1.13 (0.07) n = 2
    D280(295)A
    250 S267(280)A 1.51 (0.13) 1.82 (0.32) 0.95 (0.05) n = 3
    E272(285)A
    251 S267(280)A 1.67 (0.11) 1.85 (0.10) 0.92 (0.09) n = 3
    E293(310)A
    264 S267(280)A 1.48 (0.12) 2.03 (0.30) 0.89 (0.04) n = 2
    E258(271)A
    D280(295)A
    R255(268)A
    269 E380(405)A 8.55 (0.94) 3 1.02 (0.07) 1.05 (0.11) 1.02 n = 2
    N434(465)A
    270 E380(405)A 12.6 (1.7) 0.99 (0.06) 0.99 (0.11) 0.96 n = 2
    N434(465)A
    T307(326)A
    271 E380(405)A 1.01 (0.01) 2 0.98 1.04 0.92 n = 1
    L309(328)A
    272 N434(465)A 3.15 (0.42) 2 0.94 (0.11) 0.96 (0.17) 0.88 n = 2
    K288(305)A
  • Discussion
  • This study includes a complete mapping of human IgG1 for human FcγRI, FcγRIIA, FcγRIIB, FcγRIIIA, and FcRn. An alanine-scan of all amino acids in human IgG1 Fc (CH2 and CH3 domains) exposed to solvent, based on the crystal structure of human Fc (Deisenhofer, Biochemistry 20:2361-2370 (1981)), was performed. Each exposed amino acid in CH2 and CH3 was individually changed to alanine and the variant IgG assayed against all five human receptors; all variants were evaluated using humanized anti-IgE E27 IgG1 as the parent polypeptide. FcγRI and FcRn are high affinity receptors and monomeric IgG could be evaluated in the assays for these two receptors. FcγRIIA, FcγRIIB and FcγRIIIA are low affinity receptors and required use of an immune complex. Hence, an ELISA-type assay was used for FcγRIIA, FcγRIIB, and FcγRIIIA, in which pre-formed hexamers, consisting of three anti-IgE E27 and three IgE molecules were bound to the FcγR and either anti-human IgG Fc-HRP or protein G-HRP used as detection reagent. In order to increase binding, these hexamers could be linked into multimers by addition of human VEGF (using anti-VEGF IgE). The hexamers bound to the low affinity FcγR significantly better than the IgG monomers; the multimers bound better than the hexamers (FIGS. 15A and 15B). The hexameric complexes were used since these provided sufficient binding and required less IgG. Complexes formed using other antibody:antigen combinations are also possible reagents, as long as the antigen contains at least two identical binding sites per molecule for the antibody. As an example, VEGF contains two binding sites per VEGF dimer for anti-VEGF A.4.6.1 (Kim et al., Growth Factors 7:53 (1992) and Kim et al., Nature 362:841 (1993)). VEGF:anti-VEGF multimers also bound to the low affinity FcγRIIA and FcγRIIIA (FIGS. 16A and 16B).
  • Once the complete alanine-scan was performed, several classes of alanine variants were found. Some variants exhibited reduced binding to all FcγR (G14, FIG. 17), while other variants showed reduced binding only to one FcγR (G36, FIG. 17), improved binding only to one FcγR (G15, G54, G55, FIG. 17), or simultaneous reduction to one FcγR with improvement to another (G16, FIG. 17).
  • Individual alanine variants were also combined in a single variant Fc region; e.g. combining S298(317)A with K334(353)A improved binding to FcγRIIIA more than either S298(317)A or K334(353)A alone (FIGS. 18A and B; and compare variants 36, 55, and 109 in Tables 6 and 9) (residue numbers in parentheses are those of the EU index as in Kabat). Similarly, combining S298(317)A with E333(352)A improved binding to FcγRIIIA more than either S298(317)A or E333(352)A alone (compare variants 36, 54, and 107 in Tables 6 and 9).
  • Selected IgG variants were also tested for their binding to FcγR transfected into mammalian cells. The α-chain extracellular portion of human FcγRIIIA was transfected into CHO cells using a GPI-link, whereas for human FcγRIIB the full-length receptor was transfected into CHO cells. For the variants tested, the pattern of binding to the cells was the same as the pattern of binding in the protein:protein (ELISA) assay (FIGS. 18A-B and 19A-B).
  • One application of these variants is to improve the ADCC effector function of an antibody. This can be achieved by modifying Fc region amino acids at one or more residues which would lead to improved binding to FcγRIIIA. Improved FcγRIIIA binding would lead to improved binding by NK cells, which carry only FcγRIIIA and can mediate ADCC. Selected alanine variants which were either reduced in binding to FcγRIIIA (variants 17, 18, 34; Table 6), had no effect on FcγRIIIA binding (variant 31; Table 6), or had improved binding to FcγRIIIA (variants 30, 36; Table 6) were tested in an in vitro ADCC assay using human PBMCs as effector cells. Since the target cells were HER2-overexpressing SKBR3 cells, the IgG Fc variants used in this assay were generated by substituting the VHNL domains of anti-IgE E27 with those from anti-HER2 antibody; HERCEPTIN® (humAb4D5-8 in Table 1 of Carter et al., PNAS (USA) 89:4285-4289 (1992)). The pattern of ADCC exhibited by the variants correlated well with the pattern of binding to FcγRIIIA (FIGS. 20 and 21). Notably the variant which showed the best improvement in binding to FcγRIIIA in protein:protein assays, variant 36 S298(317)A, also showed improvement in ADCC compared to wildtype HERCEPTIN® at 1.25 ng/ml (FIG. 21).
  • EXAMPLE 5 Bind of Fc Variants to Polymorphic Fc Receptors
  • Allelic variants of several of the human FcγR have been found in the human population. These allelic variant forms have been shown to exhibit differences in binding of human and murine IgG and a number of association studies have correlated clinical outcomes with the presence of specific allelic forms (reviewed in Lehrnbecher et al., Blood 94(12):4220-4232 (1999)). Several studies have investigated two forms of FcγRIIA, R131 and H131, and their association with clinical outcomes (Hatta et al., Genes and Immunity 1:53-60 (1999); Yap et al., Lupus 8:305-310 (1999); and Lorenz et al. European J. Immunogenetics 22:397-401 (1995)). Two allelic forms of FcγRIIIA, F158 and V158, are only now being investigated (Lehrnbecher et al., supra; and Wu et al., J. Clin. Invest. 100(5):1059-1070 (1997)). In this example, selected IgG variants were tested against both allelic forms of FcγRIIA or FcγRIIIA. Fc receptor binding assays were performed essentially as described in the above examples. However, for FcγRIIIA-V158, both (a) the low affinity receptor binding assay of Example 1 (which analyzes binding of the IgG complex to FcγRIIIA-V158); and (b) the high affinity FcγR binding assay of Example 4 (which analyzes binding of IgG monomer to FcγRIIIA-V158) were carried out. The results of these studies are summarized in Table 10 below.
  • TABLE 10
    Binding of Variants to FcγRIIA and FcγRIIIA Polymorphic Receptors
    IgG Complex IgG Complex IgG Complex IgG Complex IgG Monomer
    Res#EU FcγRIIA-R131 FcγRIIA-H131 FcγRIIIA-F158 FcγRIIIA-V158 FcγRIIIA-V158
    IG2 (Kabat) mean sd n mean sd n mean sd n mean sd n mean sd n
    11 T256(269)A 1.41 (0.27) 9 1.32 (0.18) 9 0.97 (0.03) 2 1.20 1
    254 T256(269)N 1.03 1 1.13 1 0.95 1 0.88 1
    14 D265(278)A 0.07 (0.01) 4 0.09 (0.06) 4 0.01 1
    15 S267(280)A 1.64 (0.18) 7 1.05 (0.03) 2 1.14 (0.25) 7
    189 S267(280)G 1.21 (0.05) 3 0.59 (0.09) 3 0.09 (0.02) 3
    16 H268(281)A 1.22 (0.14) 12 1.09 (0.01) 2 0.52 (0.09) 12
    25 E283(300)A 1.24 (0.23) 5 1.01 (0.14) 5 0.78 1
    226 E283(300)Q 1.12 1 1.19 1 0.89 1
    227 E283(300)S 1.03 1 0.85 1 0.83 1
    228 E283(300)N 1.18 1 0.94 1 0.63 1
    229 E283(300)D 1.14 1 0.95 1 0.67 1
    30 K290(307)A 1.29 (0.21) 7 1.28 (0.21) 7 1.12 (0.05) 2 1.13 1
    73 K290(307)Q 1.17 1 1.40 1 1.02 1 1.30 1
    75 K290(307)S 1.27 1 1.26 1 1.05 1 1.62 1
    77 K290(307)E 1.10 1 1.30 1 0.98 1 1.50 1
    78 K290(307)R 1.05 1 1.08 1 1.07 1 1.24 1
    177 K290(307)G 1.07 1 1.23 1 1.11 1 2.29 1
    31 R292(309)A 0.27 (0.14) 9 0.90 (0.18) 9 0.94 1
    80 R292(309)K 0.71 (0.17) 3 1.15 (0.18) 3 1.64 1
    81 R292(309)H 0.21 (0.09) 2 0.92 (0.08) 2 1.21 1
    82 R292(309)Q 0.47 (0.12) 3 0.45 (0.09) 3 0.56 1
    83 R292(309)N 0.54 (0.16) 3 0.88 (0.02) 3 0.91 1
    144 E293(310)Q 0.85 (0.03) 2 0.99 (0.04) 2 1.00 1 0.97 1
    33 E294(311)A 0.87 (0.19) 5 0.66 (0.14) 5 0.68 1
    173 E294(311)Q 1.01 1 0.84 1 0.79 1
    174 E294(311)D 0.37 1 0.14 1 0.26 1
    36 S298(317)A 0.40 (0.08) 12 1.30 (0.18) 12 1.02 (0.04) 2 1.96 1
    70 S298(317)G 0.87 (0.17) 4 0.46 (0.09) 4 0.88 1 1.88 1
    71 S298(317)T 0.41 (0.21) 3 0.89 (0.20) 3 0.96 1 0.75 1
    72 S298(317)N 0.08 (0.01) 2 0.06 (0.01) 2 0.66 1 0.17 1
    218 S298(317)V 0.11 (0.06) 3 0.33 (0.19) 3 0.88 1 0.39 1
    219 S298(317)L 1.14 (0.12) 3 0.34 (0.04) 3 0.83 1 0.67 1
    40 V305(324)A 1.12 (0.12) 4 1.04 1 0.84 (0.15) 4
    41 T307(326)A 1.19 (0.37) 12 1.37 (0.13) 2 1.12 (0.18) 12
    45 N315(334)A 1.15 (0.06) 5 1.11 (0.06) 2 1.07 (0.21) 5
    46 K317(336)A 1.13 (0.05) 4 1.04 1 1.10 (0.23) 4
    48 K320(339)A 1.12 (0.11) 4 1.16 1 0.87 (0.17) 4
    54 E333(352)A 0.92 (0.12) 10 1.27 (0.17) 10 1.10 (0.10) 2 1.29 1
    141 E333(352)Q 0.70 (0.05) 2 1.10 (0.03) 2 1.05 1 1.00 1
    142 E333(352)N 0.59 (0.04) 2 0.56 (0.10) 2 0.64 1 0.56 1
    143 E333(352)S 0.94 1 0.99 1 1.07 1
    152 E333(352)K 0.85 (0.14) 3 0.88 1 0.81 1
    153 E333(352)R 0.75 (0.04) 2 0.84 (0.05) 2 0.93 1 0.83 1
    154 E333(352)D 1.26 (0.04) 3 1.00 1 1.70 1
    178 E333(352)G 0.87 1 1.05 1 1.23 1
    55 K334(353)A 1.01 (0.15) 17 1.39 (0.19) 17 1.07 (0.09) 3 1.60 (0.01) 2
    135 K334(353)R 1.15 (0.09) 5 0.68 (0.07) 5 0.88 1
    136 K334(353)Q 1.08 (0.11) 7 1.31 (0.26) 7 1.27 (0.01) 2 1.92 1
    137 K334(353)N 1.16 (0.11) 7 1.15 (0.16) 7 1.19 (0.06) 2 1.70 1
    138 K334(353)S 1.01 (0.11) 3 1.19 (0.08) 3 1.25 1 1.82 1
    139 K334(353)E 0.74 (0.15) 4 1.30 (0.09) 4 1.17 1 2.75 1
    140 K334(353)D 0.51 (0.09) 4 1.13 (0.09) 4 1.07 1
    179 K334(353)G 0.76 (0.08) 5 0.88 (0.22) 5 0.94 1 1.28 1
    190 K334(353)M 1.06 1 1.35 1 0.99 1 2.08 1
    191 K334(353)Y 1.08 1 1.31 1 0.98 1 1.72 1
    192 K334(353)W 0.94 1 1.07 1 0.96 1 1.53 1
    193 K334(353)H 1.09 1 1.26 1 0.97 1 2.06 1
    220 K334(353)V 1.13 (0.11) 3 1.34 (0.18) 3 1.00 1 2.89 1
    221 K334(353)L 1.05 1 1.38 1 0.96 1 3.59 1
    65 P331(350)A 1.29 (0.14) 3 1.03 (0.19) 3 0.96 1 0.78 1
    198 P331(350)S 1.00 1 0.86 1 0.54 1
    199 P331(350)N 0.86 1 0.23 1 0.24 1
    200 P331(350)E 1.06 1 0.42 1 0.36 1
    203 P331(350)K 0.94 1 0.33 1 0.26 1
    96 S267(280)A 1.54 (0.12) 3 1.07 (0.06) 2 0.84 1
    H268(281)A
    110 S298(317)A 0.35 (0.13) 11 1.66 (0.42) 11 1.19 (0.18) 3
    E333(352)A
    K334(353)A
    271 E380(405)A 0.98 1 0.92 1 1.10 1
    L309(328)A
  • For FcγRIIIA, the pattern of binding of the selected IgG1 variants to the relatively higher affinity FcγRIIIA-V158 was the same as for the relatively lower affinity FcγRIIIA-F158 (the F158 form was used in assaying all variants). IgG1 variants which showed improved binding to the FcγRIIIA-F158 form also showed improved binding to the FcγRIIIA-V158 form though the improvement was not as pronounced. For FcγRIIA-R131 (used in assaying all variants) and FcγRIIA-H131, the binding pattern of the selected IgG1 variants did show some distinct differences. S267(280)A, H268(281)A, and S267(280)A/H268(281)A exhibited improved binding to FcγRIIA-R131, compared to native IgG1, but not to FcγRIIA-H131. In contrast, S267(280)G showed improved binding to FcγRIIA-R131 but reduced binding to FcγRIIA-H131 (Table 10). Other variants bound similarly to both allelic FcγRIIA forms: V305(324)A, T307(326)A, N315(324)A, K317(336)A, and K320(339)A.

Claims (25)

1. An antibody comprising a variant human IgG1 Fc region, which antibody comprises an amino acid substitution at position 265 of the Fc region, wherein the numbering of the residues in the Fc region is that of the EU index as in Kabat.
2. The antibody of claim 1 which comprises one amino acid substitution in the Fc region.
3. The antibody of claim 1 which consists of one amino acid substitution in the Fc region.
4. The antibody of claim 1 which comprises two amino acid substitutions in the Fc region.
5. The antibody of claim 1 which consists of two amino acid substitutions in the Fc region.
6. The antibody of claim 1 wherein the amino acid at position 265 is substituted with Ala.
7. The antibody of claim 1 wherein the amino acid at position 265 is substituted with Asn or Glu.
8. The antibody of claim 1 further comprising an amino acid substitution at position 297 of the Fc region.
9. The antibody of claim 8 wherein the amino acid at position 297 is substituted with Ala.
10. The antibody of claim 1 which binds to an integrin.
11. The antibody of claim 1 which binds a surface membrane protein or fragment thereof.
12. A host cell comprising nucleic acid encoding an antibody comprising a variant human IgG1 Fc region, which antibody comprises an amino acid substitution at position 265 of the Fc region, wherein the numbering of the residues in the Fc region is that of the EU index as in Kabat.
13. A method for producing an antibody comprising culturing the host cell of claim 12 so that the nucleic acid is expressed.
14. The method of claim 13 further comprising recovering the antibody from the host cell culture.
15. The method of claim 13 wherein the antibody comprises one amino acid substitution in the Fc region.
16. The method of claim 13 wherein the antibody consists of one amino acid substitution in the Fc region.
17. The method of claim 13 wherein the antibody comprises two amino acid substitutions in the Fc region.
18. The method of claim 13 wherein the antibody consists of two amino acid substitutions in the Fc region.
19. The method of claim 13 wherein the amino acid at position 265 of the antibody is substituted with Ala.
20. The method of claim 13 wherein the amino acid at position 265 of the antibody is substituted with Asn or Glu.
21. The method of claim 13 wherein the antibody further comprises an amino acid substitution at position 297 of the Fc region.
22. The method of claim 21 wherein the amino acid at position 297 is substituted with Ala.
23. The method of claim 13 wherein the antibody binds to an integrin.
24. The method of claim 13 wherein the antibody binds a surface membrane protein or fragment thereof.
25. A method for treating a disorder in a mammal comprising administering to the mammal a therapeutically effective amount of the antibody of claim 1.
US11/947,982 1999-01-15 2007-11-30 Polypeptide variants with altered effector function Abandoned US20110293632A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
US11/947,982 US20110293632A1 (en) 1999-01-15 2007-11-30 Polypeptide variants with altered effector function
US13/860,462 US20140342404A1 (en) 1999-01-15 2013-04-10 Polypeptide variants with altered effector function
US13/946,920 US20140147436A1 (en) 1999-01-15 2013-07-19 Polypeptide variants with altered effector function
US14/835,494 US20160060330A1 (en) 1999-01-15 2015-08-25 Polypeptide variants with altered effector function

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US11602399P 1999-01-15 1999-01-15
US09/483,588 US6737056B1 (en) 1999-01-15 2000-01-14 Polypeptide variants with altered effector function
US10/757,863 US7416727B2 (en) 1999-01-15 2004-01-15 Polypeptide variants with altered effector function
US11/158,839 US7332581B2 (en) 1999-01-15 2005-06-22 Polypeptide variants with altered effector function
US11/947,982 US20110293632A1 (en) 1999-01-15 2007-11-30 Polypeptide variants with altered effector function

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US11/158,839 Continuation US7332581B2 (en) 1999-01-15 2005-06-22 Polypeptide variants with altered effector function

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US13/860,462 Division US20140342404A1 (en) 1999-01-15 2013-04-10 Polypeptide variants with altered effector function

Publications (1)

Publication Number Publication Date
US20110293632A1 true US20110293632A1 (en) 2011-12-01

Family

ID=32302071

Family Applications (11)

Application Number Title Priority Date Filing Date
US09/483,588 Expired - Lifetime US6737056B1 (en) 1999-01-15 2000-01-14 Polypeptide variants with altered effector function
US10/757,863 Expired - Fee Related US7416727B2 (en) 1999-01-15 2004-01-15 Polypeptide variants with altered effector function
US10/835,642 Expired - Lifetime US7122637B2 (en) 1999-01-15 2004-04-30 Polypeptide variants with altered effector function
US10/982,470 Abandoned US20050118174A1 (en) 1999-01-15 2004-11-05 Polypeptide variants with altered effector function
US11/158,839 Expired - Fee Related US7332581B2 (en) 1999-01-15 2005-06-22 Polypeptide variants with altered effector function
US11/429,786 Expired - Lifetime US7335742B2 (en) 1999-01-15 2006-05-08 Polypeptide variants with altered effector function
US11/947,982 Abandoned US20110293632A1 (en) 1999-01-15 2007-11-30 Polypeptide variants with altered effector function
US12/171,888 Abandoned US20080274108A1 (en) 1999-01-15 2008-07-11 Polypeptide variants with altered effector function
US13/860,462 Abandoned US20140342404A1 (en) 1999-01-15 2013-04-10 Polypeptide variants with altered effector function
US13/946,920 Abandoned US20140147436A1 (en) 1999-01-15 2013-07-19 Polypeptide variants with altered effector function
US14/835,494 Abandoned US20160060330A1 (en) 1999-01-15 2015-08-25 Polypeptide variants with altered effector function

Family Applications Before (6)

Application Number Title Priority Date Filing Date
US09/483,588 Expired - Lifetime US6737056B1 (en) 1999-01-15 2000-01-14 Polypeptide variants with altered effector function
US10/757,863 Expired - Fee Related US7416727B2 (en) 1999-01-15 2004-01-15 Polypeptide variants with altered effector function
US10/835,642 Expired - Lifetime US7122637B2 (en) 1999-01-15 2004-04-30 Polypeptide variants with altered effector function
US10/982,470 Abandoned US20050118174A1 (en) 1999-01-15 2004-11-05 Polypeptide variants with altered effector function
US11/158,839 Expired - Fee Related US7332581B2 (en) 1999-01-15 2005-06-22 Polypeptide variants with altered effector function
US11/429,786 Expired - Lifetime US7335742B2 (en) 1999-01-15 2006-05-08 Polypeptide variants with altered effector function

Family Applications After (4)

Application Number Title Priority Date Filing Date
US12/171,888 Abandoned US20080274108A1 (en) 1999-01-15 2008-07-11 Polypeptide variants with altered effector function
US13/860,462 Abandoned US20140342404A1 (en) 1999-01-15 2013-04-10 Polypeptide variants with altered effector function
US13/946,920 Abandoned US20140147436A1 (en) 1999-01-15 2013-07-19 Polypeptide variants with altered effector function
US14/835,494 Abandoned US20160060330A1 (en) 1999-01-15 2015-08-25 Polypeptide variants with altered effector function

Country Status (1)

Country Link
US (11) US6737056B1 (en)

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080274108A1 (en) * 1999-01-15 2008-11-06 Genentech, Inc. Polypeptide variants with altered effector function
US20100098730A1 (en) * 2008-10-14 2010-04-22 Lowman Henry B Immunoglobulin variants and uses thereof
US8674083B2 (en) 1999-01-15 2014-03-18 Genentech, Inc. Polypeptide variants with altered effector function
US8969526B2 (en) 2011-03-29 2015-03-03 Roche Glycart Ag Antibody Fc variants
US9238878B2 (en) 2009-02-17 2016-01-19 Redwood Bioscience, Inc. Aldehyde-tagged protein-based drug carriers and methods of use
US9260527B2 (en) 2013-03-15 2016-02-16 Sdix, Llc Anti-human CXCR4 antibodies and methods of making same
US9540438B2 (en) 2011-01-14 2017-01-10 Redwood Bioscience, Inc. Aldehyde-tagged immunoglobulin polypeptides and methods of use thereof
US9695233B2 (en) 2012-07-13 2017-07-04 Roche Glycart Ag Bispecific anti-VEGF/anti-ANG-2 antibodies and their use in the treatment of ocular vascular diseases
US10654916B2 (en) 2011-04-21 2020-05-19 The Regents Of The University Of California, A California Corporation Compositions and methods for the treatment of neuromyelitis optica
US11066459B2 (en) 2014-03-14 2021-07-20 Biomolecular Holdings Llc Hybrid immunoglobulin containing non-peptidyl linkage
US11208632B2 (en) 2016-04-26 2021-12-28 R.P. Scherer Technologies, Llc Antibody conjugates and methods of making and using the same
US11220556B2 (en) 2013-03-15 2022-01-11 Biomolecular Holdings Llc Hybrid immunoglobulin containing non-peptidyl linkage

Families Citing this family (1743)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6528624B1 (en) * 1998-04-02 2003-03-04 Genentech, Inc. Polypeptide variants
US6194551B1 (en) * 1998-04-02 2001-02-27 Genentech, Inc. Polypeptide variants
WO2000042072A2 (en) * 1999-01-15 2000-07-20 Genentech, Inc. Polypeptide variants with altered effector function
EP1176195B1 (en) 1999-04-09 2013-05-22 Kyowa Hakko Kirin Co., Ltd. Method for controlling the activity of immunologically functional molecule
IL146005A0 (en) 1999-05-07 2002-07-25 Genentech Inc Treatment of autoimmune diseases with antagonists which bind to b cell surface markers
DE60142614D1 (en) * 2000-01-27 2010-09-02 Medimmune Inc INITÄT
DK1259547T3 (en) * 2000-03-01 2012-10-15 Medimmune Inc HIGH POTENTIAL, RECOMBINANT ANTIBODIES AND PROCEDURE FOR PRODUCING THEM
AU5345901A (en) * 2000-04-13 2001-10-30 Univ Rockefeller Enhancement of antibody-mediated immune responses
EP1296140A4 (en) * 2000-06-27 2005-06-29 Chugai Pharmaceutical Co Ltd Method of evaluating binding activity of ligand to ligand-binding protein
US20060029604A1 (en) * 2000-06-28 2006-02-09 Gerngross Tillman U Immunoglobulins comprising predominantly a GlcNAc2Man3GlcNAc2 glycoform
US20060034830A1 (en) * 2000-06-28 2006-02-16 Gerngross Tillman U Immunoglobulins comprising predominantly a GalGlcNAcMan5GLcNAc2 glycoform
US20060034828A1 (en) * 2000-06-28 2006-02-16 Gerngross Tillman U Immunoglobulins comprising predominantly a GlcNAcMAN5GLCNAC2 glycoform
US20060024304A1 (en) * 2000-06-28 2006-02-02 Gerngross Tillman U Immunoglobulins comprising predominantly a Man5GlcNAc2 glycoform
CA2422076A1 (en) * 2000-09-18 2002-03-21 Idec Pharmaceutical Corporation Combination therapy for treatment of autoimmune diseases using b cell depleting/immunoregulatory antibody combination
US6946292B2 (en) 2000-10-06 2005-09-20 Kyowa Hakko Kogyo Co., Ltd. Cells producing antibody compositions with increased antibody dependent cytotoxic activity
US6855493B2 (en) 2000-11-28 2005-02-15 Medimmune, Inc. Methods of administering/dosing anti-RSV antibodies for prophylaxis and treatment
US7179900B2 (en) * 2000-11-28 2007-02-20 Medimmune, Inc. Methods of administering/dosing anti-RSV antibodies for prophylaxis and treatment
US20060057651A1 (en) * 2000-12-08 2006-03-16 Bowdish Katherine S Polypeptides and antibodies derived from chronic lymphocytic leukemia cells and uses thereof
JP4280803B2 (en) * 2000-12-08 2009-06-17 アレクシオン ファーマシューティカルズ, インコーポレイテッド Chronic lymphocytic leukemia cell line and its use for producing antibodies
US9249229B2 (en) * 2000-12-08 2016-02-02 Alexion Pharmaceuticals, Inc. Polypeptides and antibodies derived from chronic lymphocytic leukemia cells and uses thereof
US7408041B2 (en) * 2000-12-08 2008-08-05 Alexion Pharmaceuticals, Inc. Polypeptides and antibodies derived from chronic lymphocytic leukemia cells and uses thereof
US7658921B2 (en) * 2000-12-12 2010-02-09 Medimmune, Llc Molecules with extended half-lives, compositions and uses thereof
EP2341060B1 (en) * 2000-12-12 2019-02-20 MedImmune, LLC Molecules with extended half-lives, compositions and uses thereof
US7270960B2 (en) * 2001-08-29 2007-09-18 Pacific Northwest Research Institute Diagnosis of ovarian carcinomas
WO2003035835A2 (en) * 2001-10-25 2003-05-01 Genentech, Inc. Glycoprotein compositions
US7737260B2 (en) 2003-11-13 2010-06-15 Hanmi Pharm. Co., Ltd Protein complex using an immunoglobulin fragment and method for the preparation thereof
US20060034829A1 (en) * 2001-12-27 2006-02-16 Gerngross Tillman U Immunoglobulins comprising predominantly a MAN3GLCNAC2 glycoform
US20060024292A1 (en) * 2001-12-27 2006-02-02 Gerngross Tillman U Immunoglobulins comprising predominantly a Gal2GlcNAc2Man3GlcNAc2 glycoform
US20040002587A1 (en) * 2002-02-20 2004-01-01 Watkins Jeffry D. Fc region variants
US7317091B2 (en) * 2002-03-01 2008-01-08 Xencor, Inc. Optimized Fc variants
US20090042291A1 (en) * 2002-03-01 2009-02-12 Xencor, Inc. Optimized Fc variants
US20100311954A1 (en) * 2002-03-01 2010-12-09 Xencor, Inc. Optimized Proteins that Target Ep-CAM
US20040132101A1 (en) * 2002-09-27 2004-07-08 Xencor Optimized Fc variants and methods for their generation
US7662925B2 (en) * 2002-03-01 2010-02-16 Xencor, Inc. Optimized Fc variants and methods for their generation
US8188231B2 (en) 2002-09-27 2012-05-29 Xencor, Inc. Optimized FC variants
US20080260731A1 (en) * 2002-03-01 2008-10-23 Bernett Matthew J Optimized antibodies that target cd19
US20080199471A1 (en) * 2002-03-01 2008-08-21 Bernett Matthew J Optimized cd40 antibodies and methods of using the same
US20080254027A1 (en) * 2002-03-01 2008-10-16 Bernett Matthew J Optimized CD5 antibodies and methods of using the same
US20070148171A1 (en) * 2002-09-27 2007-06-28 Xencor, Inc. Optimized anti-CD30 antibodies
WO2003084569A1 (en) 2002-04-09 2003-10-16 Kyowa Hakko Kogyo Co., Ltd. Drug containing antibody composition
DE60334453D1 (en) * 2002-05-30 2010-11-18 Macrogenics Inc CD16A BINDING PROTEINS AND USE FOR THE TREATMENT OF IMMUNE DISEASES
US7132100B2 (en) * 2002-06-14 2006-11-07 Medimmune, Inc. Stabilized liquid anti-RSV antibody formulations
US8187593B2 (en) * 2002-08-14 2012-05-29 Macrogenics, Inc. FcγRIIB specific antibodies and methods of use thereof
EP2371389A3 (en) * 2002-08-14 2012-04-18 MacroGenics, Inc. FcgammaRIIB-specific antibodies and methods of use thereof
US8193318B2 (en) * 2002-08-14 2012-06-05 Macrogenics, Inc. FcγRIIB specific antibodies and methods of use thereof
US8044180B2 (en) * 2002-08-14 2011-10-25 Macrogenics, Inc. FcγRIIB specific antibodies and methods of use thereof
US8968730B2 (en) 2002-08-14 2015-03-03 Macrogenics Inc. FcγRIIB specific antibodies and methods of use thereof
US8946387B2 (en) * 2002-08-14 2015-02-03 Macrogenics, Inc. FcγRIIB specific antibodies and methods of use thereof
US8530627B2 (en) * 2002-08-14 2013-09-10 Macrogenics, Inc. FcγRIIB specific antibodies and methods of use thereof
US20060177439A1 (en) * 2004-12-15 2006-08-10 Scott Koenig FcgammaRIIB-specific antibodies and methods of use thereof
FR2844455B1 (en) * 2002-09-13 2007-12-14 Lab Francais Du Fractionnement TREATMENT OF PATHOLOGIES EXCLUDING IMMUNE RESPONSE BY OPTIMIZED ANTIBODIES
US20060235208A1 (en) * 2002-09-27 2006-10-19 Xencor, Inc. Fc variants with optimized properties
AR042485A1 (en) * 2002-12-16 2005-06-22 Genentech Inc HUMANIZED ANTIBODY THAT JOINS THE HUMAN CD20
US7960512B2 (en) * 2003-01-09 2011-06-14 Macrogenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
EP2368578A1 (en) 2003-01-09 2011-09-28 Macrogenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
GB0300571D0 (en) * 2003-01-10 2003-02-12 Imp College Innovations Ltd Modification of feeding behaviour
EP2272533A1 (en) * 2003-01-13 2011-01-12 MacroGenics, Inc. Soluble FcyR fusion proteins and methods of use thereof
US8084582B2 (en) 2003-03-03 2011-12-27 Xencor, Inc. Optimized anti-CD20 monoclonal antibodies having Fc variants
US20090010920A1 (en) * 2003-03-03 2009-01-08 Xencor, Inc. Fc Variants Having Decreased Affinity for FcyRIIb
US20070275460A1 (en) * 2003-03-03 2007-11-29 Xencor.Inc. Fc Variants With Optimized Fc Receptor Binding Properties
US8388955B2 (en) * 2003-03-03 2013-03-05 Xencor, Inc. Fc variants
US20050233390A1 (en) * 2003-04-09 2005-10-20 Allen John W Device including a proteinaceous factor, a recombinant proteinaceous factor, and a nucleotide sequence encoding the proteinaceous factor
US9051373B2 (en) 2003-05-02 2015-06-09 Xencor, Inc. Optimized Fc variants
TWI353991B (en) 2003-05-06 2011-12-11 Syntonix Pharmaceuticals Inc Immunoglobulin chimeric monomer-dimer hybrids
US7709610B2 (en) 2003-05-08 2010-05-04 Facet Biotech Corporation Therapeutic use of anti-CS1 antibodies
CA2897608C (en) * 2003-05-09 2018-07-31 Duke University Cd20-specific antibodies and methods employing same
WO2005003175A2 (en) * 2003-06-13 2005-01-13 Biogen Idec Ma Inc. Aglycosyl anti-cd154 (cd40 ligand) antibodies and uses thereof
WO2005000899A2 (en) * 2003-06-27 2005-01-06 Biogen Idec Ma Inc. Modified binding molecules comprising connecting peptides
CA2536408A1 (en) 2003-08-22 2005-03-03 Biogen Idec Ma Inc. Improved antibodies having altered effector function and methods for making the same
US8101720B2 (en) * 2004-10-21 2012-01-24 Xencor, Inc. Immunoglobulin insertions, deletions and substitutions
US9714282B2 (en) * 2003-09-26 2017-07-25 Xencor, Inc. Optimized Fc variants and methods for their generation
GB0324368D0 (en) * 2003-10-17 2003-11-19 Univ Cambridge Tech Polypeptides including modified constant regions
WO2005044854A2 (en) 2003-11-04 2005-05-19 Chiron Corporation Antagonist anti-cd40 monoclonal antibodies and methods for their use
US20070218060A1 (en) 2003-11-04 2007-09-20 Chiron Corporation Use of Antagonist Anti-Cd40 Monoclonal Antibodies for Treatment of Multiple Myeloma
US20070098717A1 (en) 2003-11-04 2007-05-03 Chiron Corporation Methods of therapy for solid tumors expressing the cd40 cell-surface antigen
AU2004287480B2 (en) 2003-11-04 2011-09-15 Novartis Vaccines And Diagnostics, Inc. Use of antagonist anti-CD40 antibodies for treatment of chronic lymphocytic leukemia
CA2544368C (en) 2003-11-04 2014-04-01 Chiron Corporation Methods of therapy for b cell-related cancers
EA036531B1 (en) * 2003-11-05 2020-11-19 Роше Гликарт Аг Type ii anti-cd20 humanized antibody (variants), pharmaceutical composition comprising these antibody variants, and use thereof
WO2005063815A2 (en) * 2003-11-12 2005-07-14 Biogen Idec Ma Inc. Fcϝ receptor-binding polypeptide variants and methods related thereto
WO2005047327A2 (en) * 2003-11-12 2005-05-26 Biogen Idec Ma Inc. NEONATAL Fc RECEPTOR (FcRn)-BINDING POLYPEPTIDE VARIANTS, DIMERIC Fc BINDING PROTEINS AND METHODS RELATED THERETO
CA2546580A1 (en) * 2003-11-18 2005-06-09 Iconic Therapeutics, Inc. Homogeneous preparations of chimeric proteins
WO2005056759A2 (en) * 2003-12-04 2005-06-23 Xencor, Inc. Methods of generating variant proteins with increased host string content and compositions thereof
WO2005077981A2 (en) * 2003-12-22 2005-08-25 Xencor, Inc. Fc POLYPEPTIDES WITH NOVEL Fc LIGAND BINDING SITES
PT2311873T (en) 2004-01-07 2018-11-20 Novartis Vaccines & Diagnostics Inc M-csf-specific monoclonal antibody and uses thereof
BRPI0506771A (en) * 2004-01-12 2007-05-22 Applied Molecular Evolution antibody and pharmaceutical composition
AU2005227326B2 (en) * 2004-03-24 2009-12-03 Xencor, Inc. Immunoglobulin variants outside the Fc region
KR20070038453A (en) * 2004-04-16 2007-04-10 마크로제닉스, 인크. FCgamma;RIIB-SPECIFIC ANTIBODIES AND METHODS OF USE THEREOF
BRPI0510224A (en) * 2004-05-05 2007-10-23 Genentech Inc autoimmune disease prevention methods and industrialized article
WO2005110474A2 (en) * 2004-05-10 2005-11-24 Macrogenics, Inc. HUMANIZED FcϜRIIB SPECIFIC ANTIBODIES AND METHODS OF USE THEREOF
KR100545720B1 (en) * 2004-05-31 2006-01-24 메덱스젠 주식회사 Glycosylated Immunoglobulin and Immunoadhesin comprising the same
PA8635501A1 (en) * 2004-06-04 2006-06-02 Genentech Inc USE OF AN ANTIBODY FOR THE TREATMENT OF LUPUS
BRPI0510915A (en) * 2004-06-04 2007-11-13 Genentech Inc Method for treating multiple sclerosis and manufactured article
AR049390A1 (en) * 2004-06-09 2006-07-26 Wyeth Corp ANTIBODIES AGAINST HUMAN INTERLEUQUINE-13 AND USES OF THE SAME
US7501121B2 (en) * 2004-06-17 2009-03-10 Wyeth IL-13 binding agents
RU2427588C2 (en) * 2004-07-09 2011-08-27 Чугаи Сейяку Кабусики Кайся Glypican-3 antibody
BRPI0510674A (en) 2004-07-15 2007-12-26 Xencor Inc optimized fc variants
US20150010550A1 (en) * 2004-07-15 2015-01-08 Xencor, Inc. OPTIMIZED Fc VARIANTS
KR20070036187A (en) * 2004-07-22 2007-04-02 제넨테크, 인크. Method of treating sjogren's syndrome
KR20070047327A (en) 2004-07-26 2007-05-04 비오겐 아이덱 엠에이 아이엔씨. Anti-cd154 antibodies
EP2213683B1 (en) 2004-08-04 2013-06-05 Mentrik Biotech, LLC Variant Fc regions
CA2577329A1 (en) * 2004-08-16 2006-03-02 Medimmune, Inc. Eph receptor fc variants with enhanced antibody dependent cell-mediated cytotoxicity activity
AU2005285347A1 (en) * 2004-08-19 2006-03-23 Genentech, Inc. Polypeptide variants with altered effector function
EP1800693B1 (en) * 2004-08-24 2013-07-17 Chugai Seiyaku Kabushiki Kaisha Adjuvant therapy with the use of anti-glypican 3 antibody
US20060074225A1 (en) * 2004-09-14 2006-04-06 Xencor, Inc. Monomeric immunoglobulin Fc domains
CA2580271A1 (en) * 2004-10-05 2006-04-20 Genentech, Inc. Method for treating vasculitis
WO2006041934A2 (en) * 2004-10-05 2006-04-20 Neuralab Limited Methods and compositions for improving recombinant protein production
JO3000B1 (en) 2004-10-20 2016-09-05 Genentech Inc Antibody Formulations.
RU2451030C2 (en) * 2004-10-26 2012-05-20 Чугаи Сейяку Кабусики Кайся Anti-glypican 3-antibody having modified sugar chain
EP1812068A4 (en) * 2004-10-29 2010-06-09 Medimmune Inc Methods of preventing and treating rsv infections and related conditions
WO2007001448A2 (en) 2004-11-04 2007-01-04 Massachusetts Institute Of Technology Coated controlled release polymer particles as efficient oral delivery vehicles for biopharmaceuticals
US7632497B2 (en) * 2004-11-10 2009-12-15 Macrogenics, Inc. Engineering Fc Antibody regions to confer effector function
US8367805B2 (en) * 2004-11-12 2013-02-05 Xencor, Inc. Fc variants with altered binding to FcRn
US20070135620A1 (en) * 2004-11-12 2007-06-14 Xencor, Inc. Fc variants with altered binding to FcRn
US8802820B2 (en) 2004-11-12 2014-08-12 Xencor, Inc. Fc variants with altered binding to FcRn
DK2325207T3 (en) 2004-11-12 2017-06-06 Xencor Inc Fc variants with altered binding to FcRn
WO2007001457A2 (en) * 2004-11-12 2007-01-04 Xencor, Inc. Antibodies operably linked to selected chemoattractants
US8546543B2 (en) 2004-11-12 2013-10-01 Xencor, Inc. Fc variants that extend antibody half-life
EP1674479A1 (en) * 2004-12-22 2006-06-28 Memorial Sloan-Kettering Cancer Center Modulation of Fc Gamma receptors for optimizing immunotherapy
BRPI0518994A2 (en) * 2004-12-31 2008-12-02 Biogen Idec Inc br3-binding antibodies or polypeptides, their uses and liquid formulations
WO2006074399A2 (en) * 2005-01-05 2006-07-13 Biogen Idec Ma Inc. Multispecific binding molecules comprising connecting peptides
EP1858925A2 (en) * 2005-01-12 2007-11-28 Xencor, Inc. Antibodies and fc fusion proteins with altered immunogenicity
WO2006082515A2 (en) 2005-02-07 2006-08-10 Glycart Biotechnology Ag Antigen binding molecules that bind egfr, vectors encoding same, and uses thereof
DOP2006000029A (en) * 2005-02-07 2006-08-15 Genentech Inc ANTIBODY VARIANTS AND USES THEREOF. (VARIATIONS OF AN ANTIBODY AND USES OF THE SAME)
US7700099B2 (en) * 2005-02-14 2010-04-20 Merck & Co., Inc. Non-immunostimulatory antibody and compositions containing the same
US20060263357A1 (en) 2005-05-05 2006-11-23 Tedder Thomas F Anti-CD19 antibody therapy for autoimmune disease
KR101289537B1 (en) 2005-02-15 2013-07-31 듀크 유니버시티 Anti-cd19 antibodies and uses in oncology
WO2006093794A1 (en) * 2005-02-28 2006-09-08 Centocor, Inc. Heterodimeric protein binding compositions
EP1871882A1 (en) * 2005-03-25 2008-01-02 GlycArt Biotechnology AG Antigen binding molecules directed to mcsp and having increased fc receptor binding affinity and effector function
WO2006104989A2 (en) * 2005-03-29 2006-10-05 Verenium Corporation Altered antibody fc regions and uses thereof
ES2592271T3 (en) 2005-03-31 2016-11-29 Chugai Seiyaku Kabushiki Kaisha Polypeptide production methods by regulating the association of polypeptides
US9284375B2 (en) * 2005-04-15 2016-03-15 Macrogenics, Inc. Covalent diabodies and uses thereof
US9296816B2 (en) * 2005-04-15 2016-03-29 Macrogenics, Inc. Covalent diabodies and uses thereof
US9963510B2 (en) * 2005-04-15 2018-05-08 Macrogenics, Inc. Covalent diabodies and uses thereof
US11254748B2 (en) 2005-04-15 2022-02-22 Macrogenics, Inc. Covalent diabodies and uses thereof
WO2006116260A2 (en) 2005-04-26 2006-11-02 Medimmune, Inc. Modulation of antibody effector function by hinge domain engineering
CN101228189A (en) * 2005-05-09 2008-07-23 格黎卡特生物技术股份公司 Antigen binding molecules having modified FC regions and altered binding to FC receptors
JP5421590B2 (en) 2005-05-18 2014-02-19 ノバルティス アーゲー Methods for diagnosis and treatment of diseases with autoimmune and / or inflammatory components
KR20080046135A (en) * 2005-05-20 2008-05-26 제넨테크, 인크. Pretreatment of a biological sample from an autoimmune disease subject
US20070110757A1 (en) 2005-06-23 2007-05-17 Ziping Wei Antibody formulations having optimized aggregation and fragmentation profiles
US8309690B2 (en) * 2005-07-01 2012-11-13 Medimmune, Llc Integrated approach for generating multidomain protein therapeutics
WO2007008943A2 (en) 2005-07-08 2007-01-18 Xencor, Inc. Optimized anti-ep-cam antibodies
SG164379A1 (en) * 2005-07-21 2010-09-29 Genmab As Potency assays for antibody drug substance binding to an fc receptor
WO2007011041A1 (en) * 2005-07-22 2007-01-25 Kyowa Hakko Kogyo Co., Ltd. Genetically modified antibody composition
US7923538B2 (en) 2005-07-22 2011-04-12 Kyowa Hakko Kirin Co., Ltd Recombinant antibody composition
JP5457671B2 (en) * 2005-07-28 2014-04-02 ノバルティス アーゲー M-CSF specific monoclonal antibody and use thereof
DK1919503T3 (en) * 2005-08-10 2014-12-15 Macrogenics Inc Identification and preparation of antibodies with variant fc regions and methods of use thereof
WO2007022520A2 (en) * 2005-08-19 2007-02-22 Cerus Corporation Antibody-mediated enhancement of immune response
US20070190029A1 (en) * 2005-08-19 2007-08-16 Cerus Corporation Listeria-induced immunorecruitment and activation, and methods of use thereof
BRPI0615397B1 (en) * 2005-08-26 2023-10-03 Roche Glycart Ag ANTI-CD20 ANTIBODY, PHARMACEUTICAL COMPOSITION CONTAINING IT AND USE THEREOF
CA2623197A1 (en) * 2005-09-22 2007-03-29 Prosci Incorporated Glycosylated polypeptides produced in yeast mutants and methods of use thereof
CA2624189A1 (en) * 2005-10-03 2007-04-12 Xencor, Inc. Fc variants with optimized fc receptor binding properties
JP4860703B2 (en) 2005-10-06 2012-01-25 ゼンコー・インコーポレイテッド Optimized anti-CD30 antibody
KR20080073293A (en) 2005-10-14 2008-08-08 메디뮨 엘엘씨 Cell display of antibody libraries
US20070087005A1 (en) * 2005-10-14 2007-04-19 Lazar Gregory A Anti-glypican-3 antibody
ATE439598T1 (en) * 2005-10-17 2009-08-15 Pls Design Gmbh CHIMERA BETWEEN A HUMAN IGE RECEPTOR ALPHA CHAIN AND BIRD CONSTANT IMMUNOGLOBULIN DOMAIN FOR THE DETERMINATION OF SERUM IGE.
DK1945665T3 (en) * 2005-10-21 2012-02-06 Genzyme Corp Antibody-based therapy agents with elevated ADCC activity
MY153249A (en) 2005-11-14 2015-01-29 Rinat Neuroscience Corp Antagonist antibodies directed against calcitonin gene-related peptide and methods using same
MY149159A (en) 2005-11-15 2013-07-31 Hoffmann La Roche Method for treating joint damage
PT2361638E (en) * 2005-12-12 2014-04-17 Ac Immune Sa A beta 1-42 specific monoclonal antibodies with therapeutic properties
WO2007070682A2 (en) * 2005-12-15 2007-06-21 Massachusetts Institute Of Technology System for screening particles
FR2894982A1 (en) * 2005-12-16 2007-06-22 Lab Francais Du Fractionnement Preparation of antibodies selective for activating Fc receptors, useful for treatment of tumors and viral or bacterial infections, by replacing specific histidine residues in the Fc region of a monoclonal antibody
BRPI0620795A2 (en) 2005-12-30 2011-11-22 Merck Patent Ges Mit Beschronkter Haftung anti-cd19 antibodies reduced immunogenicity
DK1973950T3 (en) 2006-01-05 2014-12-15 Genentech Inc ANTI-EphB4 ANTIBODIES AND METHODS FOR USING SAME
DK2463305T3 (en) 2006-01-12 2016-08-29 Alexion Pharma Inc Antibodies to OX-2 / CD200 and uses thereof
AU2007212147A1 (en) * 2006-02-03 2007-08-16 Medimmune, Llc Protein formulations
EP2540741A1 (en) 2006-03-06 2013-01-02 Aeres Biomedical Limited Humanized anti-CD22 antibodies and their use in treatment of oncology, transplantation and autoimmune disease
CA2644903A1 (en) * 2006-03-10 2007-09-20 Macrogenics, Inc. Identification and engineering of antibodies with variant heavy chains and methods of using same
AR059851A1 (en) 2006-03-16 2008-04-30 Genentech Inc ANTIBODIES OF EGFL7 AND METHODS OF USE
US8278421B2 (en) 2006-03-20 2012-10-02 Xoma Techolology Ltd. Human antibodies specific for gastrin materials and methods
EP4001409A1 (en) 2006-03-31 2022-05-25 Chugai Seiyaku Kabushiki Kaisha Methods for controlling blood pharmacokinetics of antibodies
WO2007114325A1 (en) 2006-03-31 2007-10-11 Chugai Seiyaku Kabushiki Kaisha Antibody modification method for purifying bispecific antibody
WO2008105773A2 (en) 2006-03-31 2008-09-04 Massachusetts Institute Of Technology System for targeted delivery of therapeutic agents
JP2009538273A (en) * 2006-04-14 2009-11-05 トルビオン ファーマシューティカルズ, インコーポレイテッド Binding protein with immunoglobulin hinge region and Fc region with modified FC effector function
WO2007119796A1 (en) * 2006-04-14 2007-10-25 Medical And Biological Laboratories Co., Ltd. Mutant polypeptide having effector function
CN101426817B (en) 2006-04-21 2013-07-10 诺华有限公司 Antagonist anti-cd40 antibody pharmaceutical compositions
CA2652280C (en) 2006-05-15 2014-01-28 Massachusetts Institute Of Technology Polymers for functional particles
ES2489646T3 (en) 2006-05-26 2014-09-02 Macrogenics, Inc. Humanized antibodies specific to Fc gamma RIIB and its methods of use
RS53168B (en) 2006-05-30 2014-06-30 Genentech Inc. Antibodies and immunoconjugates and uses therefor
EP2023955A4 (en) * 2006-06-06 2009-10-28 Tolerrx Inc Administration of anti-cd3 antibodies in the treatment of autoimmune diseases
CL2007001623A1 (en) * 2006-06-06 2008-01-18 Genentech Inc Anti-dll4 antibody; polynucleotide that encodes it; vector and host cell comprising said polynucleotide; method for making the antibody and immunojugate; method of detection of dll4 and diagnostic method of a disorder associated with dll4; composition comprising the antibody.
TW200817435A (en) * 2006-06-06 2008-04-16 Genentech Inc Compositions and methods for modulating vascular development
WO2007150030A2 (en) * 2006-06-23 2007-12-27 Massachusetts Institute Of Technology Microfluidic synthesis of organic nanoparticles
EP2505209A1 (en) 2006-06-26 2012-10-03 MacroGenics, Inc. Fcgamma-RIIB-specific antibodies and methods of the use thereof
EP2032159B1 (en) * 2006-06-26 2015-01-07 MacroGenics, Inc. Combination of fcgammariib antibodies and cd20-specific antibodies and methods of use thereof
US20100166741A1 (en) * 2006-07-13 2010-07-01 Genentech , Inc. Altered br-3 binding polypeptides
CA2657681C (en) * 2006-07-14 2019-03-19 Ac Immune S.A. Humanized antibodies against beta amyloid protein
AU2007275654A1 (en) 2006-07-19 2008-01-24 The Trustees Of The University Of Pennsylvania WSX-1/p28 as a target for anti-inflammatory responses
CN101626783A (en) * 2006-08-04 2010-01-13 诺华有限公司 EPHB3-specific antibody and its application
WO2008019142A2 (en) * 2006-08-04 2008-02-14 Massachusetts Institute Of Technology Oligonucleotide systems for targeted intracellular delivery
AR062223A1 (en) 2006-08-09 2008-10-22 Glycart Biotechnology Ag MOLECULES OF ADHESION TO THE ANTIGEN THAT ADHER TO EGFR, VECTORS THAT CODE THEM, AND THEIR USES OF THESE
ES2457072T3 (en) 2006-08-14 2014-04-24 Xencor, Inc. Optimized antibodies that select as target CD19
EP3415532A1 (en) 2006-08-18 2018-12-19 XOMA Technology Ltd. Prlr-specific antibody and uses thereof
DK2066349T3 (en) 2006-09-08 2012-07-09 Medimmune Llc HUMANIZED ANTI-CD19 ANTIBODIES AND USE THEREOF IN TREATMENT OF TUMORS, TRANSPLANTATION AND AUTOIMMUNE DISEASES
EP2061906B1 (en) 2006-09-12 2011-08-31 Genentech, Inc. Methods and compositions for the diagnosis and treatment of lung cancer using pdgfra, kit or kdr gene as genetic marker
EP2083017A4 (en) * 2006-09-14 2011-01-12 Med & Biological Lab Co Ltd Antibody having enhanced adcc activity and method for production thereof
CA2660795C (en) * 2006-09-18 2014-11-18 Xencor, Inc. Optimized antibodies that target hm1.24
US20080112961A1 (en) * 2006-10-09 2008-05-15 Macrogenics, Inc. Identification and Engineering of Antibodies with Variant Fc Regions and Methods of Using Same
CA2665644A1 (en) * 2006-10-12 2008-05-29 Genentech, Inc. Antibodies to lymphotoxin-alpha
EP2064335A4 (en) * 2006-10-16 2011-03-30 Medimmune Llc Molecules with reduced half-lives, compositions and uses thereof
BRPI0717601A2 (en) * 2006-10-24 2013-10-22 Trubion Pharmaceuticals Inc IMPROVED IMMUNGLYPROTEINS MATERIALS AND METHODS
US7846434B2 (en) 2006-10-24 2010-12-07 Trubion Pharmaceuticals, Inc. Materials and methods for improved immunoglycoproteins
RS53942B1 (en) 2006-10-27 2015-08-31 Genentech Inc. Antibodies and immunoconjugates and uses therefor
US20100303723A1 (en) * 2006-11-20 2010-12-02 Massachusetts Institute Of Technology Drug delivery systems using fc fragments
CA2671264C (en) 2006-11-30 2015-11-24 Research Development Foundation Improved immunoglobulin libraries
TWI447124B (en) 2006-12-01 2014-08-01 Medarex Llc Human antibodies that bind cd22 and uses thereof
ES2523915T5 (en) * 2006-12-01 2022-05-26 Seagen Inc Variant Target Binding Agents and Uses Thereof
US20080127996A1 (en) * 2006-12-04 2008-06-05 Weinhold Dennis G Method and apparatus to remediate an acid and/or liquid spill
WO2008070780A1 (en) 2006-12-07 2008-06-12 Novartis Ag Antagonist antibodies against ephb3
US8652466B2 (en) * 2006-12-08 2014-02-18 Macrogenics, Inc. Methods for the treatment of disease using immunoglobulins having Fc regions with altered affinities for FcγRactivating and FcγRinhibiting
CL2007003622A1 (en) 2006-12-13 2009-08-07 Medarex Inc Human anti-cd19 monoclonal antibody; composition comprising it; and tumor cell growth inhibition method.
KR20090088946A (en) 2006-12-14 2009-08-20 메다렉스, 인코포레이티드 Human antibodies that bind cd70 and uses thereof
TW200833711A (en) * 2006-12-22 2008-08-16 Genentech Inc Antibodies to insulin-like growth factor receptor
WO2008090959A1 (en) * 2007-01-24 2008-07-31 Kyowa Hakko Kirin Co., Ltd. Genetically recombinant antibody composition having enhanced effector activity
EP2125013A4 (en) * 2007-01-26 2010-04-07 Bioinvent Int Ab Dll4 signaling inhibitors and uses thereof
WO2008098165A2 (en) * 2007-02-09 2008-08-14 Massachusetts Institute Of Technology Oscillating cell culture bioreactor
WO2008109440A2 (en) 2007-03-02 2008-09-12 Genentech, Inc. Predicting response to a her dimerisation inhibitor based on low her3 expression
WO2008112192A2 (en) 2007-03-08 2008-09-18 Kalobios Pharmaceuticals, Inc. Epha3 antibodies for the treatment of solid tumors
MX2009010120A (en) 2007-03-22 2009-10-19 Ucb Pharma Sa Binding proteins, including antibodies, antibody derivatives and antibody fragments, that specifically bind cd154 and uses thereof.
NZ614857A (en) 2007-03-29 2015-04-24 Genmab As Bispecific antibodies and methods for production thereof
EP2144600A4 (en) * 2007-04-04 2011-03-16 Massachusetts Inst Technology Poly (amino acid) targeting moieties
US20090068195A1 (en) * 2007-04-23 2009-03-12 Wyeth Methods and compositions for treating and monitoring treatment of il-13-associated disorders
FR2915398B1 (en) * 2007-04-25 2012-12-28 Lab Francais Du Fractionnement "SET OF MEANS FOR THE TREATMENT OF MALIGNANT PATHOLOGY, AUTOIMMUNE DISEASE OR INFECTIOUS DISEASE"
US8629245B2 (en) 2007-05-01 2014-01-14 Research Development Foundation Immunoglobulin Fc libraries
EP2703011A3 (en) * 2007-05-07 2014-03-26 MedImmune, LLC Anti-icos antibodies and their use in treatment of oncology, transplantation and autoimmune disease
ES2441189T3 (en) 2007-05-14 2014-02-03 Novimmune Sa Fc receptor binding polypeptides with modified effector functions
AU2012244353B2 (en) * 2007-05-30 2016-10-20 Xencor, Inc. Methods and compositions for inhibiting CD32b expressing cells
AU2013204284B2 (en) * 2007-05-30 2017-02-02 Xencor, Inc. Methods and compositions for inhibiting CD32b expressing cells
DK2176298T3 (en) * 2007-05-30 2018-02-12 Xencor Inc Methods and compositions for inhibiting CD32B-expressing cells
EP2167669A2 (en) * 2007-05-31 2010-03-31 Genmab A/S Transgenic animals producing monovalent human antibodies and antibodies obtainable from these animals
US20100267934A1 (en) 2007-05-31 2010-10-21 Genmab A/S Stable igg4 antibodies
EP1997830A1 (en) 2007-06-01 2008-12-03 AIMM Therapeutics B.V. RSV specific binding molecules and means for producing them
US8048420B2 (en) * 2007-06-12 2011-11-01 Ac Immune S.A. Monoclonal antibody
US8613923B2 (en) 2007-06-12 2013-12-24 Ac Immune S.A. Monoclonal antibody
TWI557136B (en) 2007-06-12 2016-11-11 Ac免疫公司 Antibody specifically binding to beta amyloid and relevant nucleic acid molecule, expression vector, cell, composition, kit, method, and use
US7580304B2 (en) * 2007-06-15 2009-08-25 United Memories, Inc. Multiple bus charge sharing
PT2170956E (en) 2007-06-15 2015-02-05 Medigene Ag Treatment of tumors using specific anti-l1 antibody
US20110077383A1 (en) * 2007-07-03 2011-03-31 Medimmune, Llc Hinge domain engineering
MX2010000979A (en) * 2007-07-25 2010-03-26 Alexion Pharma Inc Methods and compositions for treating autoimmune disease.
CA2694488A1 (en) 2007-07-31 2009-02-05 Medimmune, Llc Multispecific epitope binding proteins and uses thereof
CN101874042B9 (en) 2007-09-26 2019-01-01 中外制药株式会社 Method for changing isoelectric point of antibody by using amino acid substitution of CDR
TWI547503B (en) * 2007-09-26 2016-09-01 建南德克公司 Anti-α5β1 antibodies, nucleic acids encoding and compositions comprising the same, and methods of production and use thereof
MX342551B (en) * 2007-09-26 2016-10-04 Chugai Pharmaceutical Co Ltd Modified antibody constant region.
ES2609918T3 (en) * 2007-10-05 2017-04-25 Genentech, Inc. Use of anti-amyloid beta antibody in eye diseases
EP2238166B1 (en) * 2007-10-05 2013-11-27 Genentech, Inc. Use of anti-amyloid beta antibody in ocular diseases
NZ601858A (en) * 2007-10-05 2014-03-28 Genentech Inc Methods and compositions for diagnosis and treatment of amyloidosis
ES2627233T3 (en) 2007-10-12 2017-07-27 Massachusetts Institute Of Technology Vaccine Nanotechnology
JP5620106B2 (en) * 2007-10-24 2014-11-05 株式会社糖鎖工学研究所 Polypeptide having enhanced effector function
AU2008323701B2 (en) 2007-11-07 2015-03-26 Genentech, Inc Methods and compositions for assessing responsiveness of B-cell lymphoma to treatment with anti-CD40 antibodies
LT3002298T (en) 2007-11-21 2019-12-10 Univ Oregon Health & Science Anti-factor xi monoclonal antibodies and methods of use thereof
TWI468417B (en) 2007-11-30 2015-01-11 Genentech Inc Anti-vegf antibodies
EP2229410A4 (en) 2007-12-05 2012-07-04 Massachusetts Inst Technology Aglycosylated immunoglobulin mutants
US20110129459A1 (en) 2007-12-05 2011-06-02 Chugai Seiyaku Kabushiki Kaisha Anti-nr10 antibody and use thereof
WO2009117030A2 (en) 2007-12-19 2009-09-24 Macrogenics, Inc. Improved compositions for the prevention and treatment of smallpox
US8092804B2 (en) 2007-12-21 2012-01-10 Medimmune Limited Binding members for interleukin-4 receptor alpha (IL-4Rα)-173
MX343594B (en) 2007-12-21 2016-11-11 Medimmune Ltd BINDING MEMBERS FOR INTERLEUKIN-4 RECEPTOR ALPHA (IL-4Ra) - 173.
EP4098661A1 (en) 2007-12-26 2022-12-07 Xencor, Inc. Fc variants with altered binding to fcrn
BRPI0907046A2 (en) 2008-01-18 2015-07-28 Medimmune Llc Engineered cysteine antibody, isolated nucleic acid, vector, host cell, antibody conjugate, pharmaceutical composition, methods of detecting cancer, autoimmune, inflammatory or infectious disorders in an individual and inhibiting proliferation of a target cell
ES2618292T3 (en) 2008-01-31 2017-06-21 Inserm - Institut National De La Sante Et De La Recherche Medicale Antibodies against and use of human CD39 to inhibit the activity of regulatory T cells
BRPI0906387A2 (en) 2008-02-05 2015-07-07 Bristol Myers Squibb Co Alpha 5 - beta 1 antibodies and their uses
NZ587292A (en) 2008-03-04 2012-09-28 Pfizer Ltd Use of anti-CGRP (calcitonin gene-related peptide) antagonists to treat chronic pain
SG2014014138A (en) * 2008-03-31 2014-07-30 Genentech Inc Compositions and methods for treating and diagnosing asthma
DK2247304T3 (en) 2008-04-02 2016-09-26 Macrogenics Inc Her2 / neu-specific antibodies and methods of use thereof
MX2010010387A (en) 2008-04-02 2010-10-15 Macrogenics Inc Bcr-complex-specific antibodies and methods of using same.
NZ602884A (en) 2008-04-11 2014-08-29 Chugai Pharmaceutical Co Ltd Antigen-binding molecule capable of binding to two or more antigen molecules repeatedly
AU2008201871A1 (en) * 2008-04-16 2009-11-26 Deutsches Krebsforschungszentrum Stiftung Des Oeffentlichen Rechts Inhibition of angiogenesis and tumor metastasis
ES2828627T3 (en) 2008-04-25 2021-05-27 Kyowa Kirin Co Ltd Stable multivalent antibody
US20110081347A1 (en) * 2008-06-04 2011-04-07 Macrogenics, Inc. Antibodies with Altered Binding to FcRn and Methods of Using Same
JP5425775B2 (en) 2008-06-30 2014-02-26 協和発酵キリン株式会社 Anti-CD27 antibody
US20100021460A1 (en) * 2008-07-15 2010-01-28 Genentech, Inc. Methods of Treating Autoimmune Diseases Using CD4 Antibodies
AR073295A1 (en) 2008-09-16 2010-10-28 Genentech Inc METHODS TO TREAT PROGRESSIVE MULTIPLE SCLEROSIS. MANUFACTURING ARTICLE.
WO2010032060A1 (en) 2008-09-19 2010-03-25 Medimmune Llc Antibodies directed to dll4 and uses thereof
JP2012503656A (en) * 2008-09-26 2012-02-09 エウレカ セラピューティクス,インコーポレイテッド Cell lines and proteins with mutant glycosylation patterns
TWI440469B (en) 2008-09-26 2014-06-11 Chugai Pharmaceutical Co Ltd Improved antibody molecules
US20100082438A1 (en) * 2008-10-01 2010-04-01 Ronnie Jack Garmon Methods and systems for customer performance scoring
US8343498B2 (en) 2008-10-12 2013-01-01 Massachusetts Institute Of Technology Adjuvant incorporation in immunonanotherapeutics
US8591905B2 (en) 2008-10-12 2013-11-26 The Brigham And Women's Hospital, Inc. Nicotine immunonanotherapeutics
US8343497B2 (en) 2008-10-12 2013-01-01 The Brigham And Women's Hospital, Inc. Targeting of antigen presenting cells with immunonanotherapeutics
US8277812B2 (en) 2008-10-12 2012-10-02 Massachusetts Institute Of Technology Immunonanotherapeutics that provide IgG humoral response without T-cell antigen
US8298533B2 (en) 2008-11-07 2012-10-30 Medimmune Limited Antibodies to IL-1R1
US20110293605A1 (en) 2008-11-12 2011-12-01 Hasige Sathish Antibody formulation
GB0821100D0 (en) * 2008-11-18 2008-12-24 Hansa Medical Ab Antibodies
US20110229471A1 (en) 2008-11-26 2011-09-22 Cedars-Sinai Medical Center Methods of determining responsiveness to anti-tnf alpha therapy in inflammatory bowel disease
US8775090B2 (en) 2008-12-12 2014-07-08 Medimmune, Llc Crystals and structure of a human IgG Fc variant with enhanced FcRn binding
AR074777A1 (en) 2008-12-19 2011-02-09 Glaxo Group Ltd PROTEINS OF UNION TO ANTIGEN
WO2010075249A2 (en) 2008-12-22 2010-07-01 Genentech, Inc. A method for treating rheumatoid arthritis with b-cell antagonists
JP2012513194A (en) 2008-12-23 2012-06-14 アストラゼネカ アクチボラグ Targeted binding agents directed to α5β1 and uses thereof
PT2374883T (en) 2008-12-26 2016-10-20 Kyowa Hakko Kirin Co Ltd Anti-cd4 antibody
WO2010078526A1 (en) 2008-12-31 2010-07-08 Biogen Idec Ma Inc. Anti-lymphotoxin antibodies
CA2749539C (en) 2009-01-21 2022-07-19 Amgen Inc. Compositions and methods comprising interleukin-2 mutants for treating inflammatory and autoimmune diseases
AR075715A1 (en) * 2009-03-05 2011-04-20 Novartis Ag FORMULATION OF LIOFILIZED ANTIBODY
CA2753995A1 (en) 2009-03-06 2010-09-10 Kalobios Pharmaceuticals, Inc. Treatment of leukemias and chronic myeloproliferative diseases with antibodies to epha3
JP5717624B2 (en) 2009-03-19 2015-05-13 中外製薬株式会社 Antibody constant region variants
JP5787446B2 (en) 2009-03-19 2015-09-30 中外製薬株式会社 Antibody constant region variants
MA33198B1 (en) 2009-03-20 2012-04-02 Genentech Inc ANTI-HER DI-SPECIFIC ANTIBODIES
CN102378767B (en) 2009-03-25 2015-01-14 健泰科生物技术公司 Anti-fgfr3 antibodies and methods using same
US8124740B2 (en) 2009-03-25 2012-02-28 Genentech, Inc. Anti- α5 β1 antibodies and uses thereof
WO2010112413A1 (en) 2009-03-31 2010-10-07 Roche Glycart Ag Treatment of cancer with a humanized anti-egfr igg1 antibody and irinotecan
US20100256340A1 (en) 2009-04-07 2010-10-07 Ulrich Brinkmann Trivalent, bispecific antibodies
US20100297127A1 (en) 2009-04-08 2010-11-25 Ghilardi Nico P Use of il-27 antagonists to treat lupus
US8647623B2 (en) 2009-04-10 2014-02-11 Kyowa Hakko Kirin Co., Ltd Method for treatment of blood tumor using anti-TIM-3 antibody
BRPI1011394A8 (en) 2009-04-18 2018-06-26 Genentech Inc method for assessing B cell linforma responsiveness to anti-cd40 antibody treatment
CN102459342B (en) 2009-04-27 2015-01-07 协和发酵麒麟株式会社 Anti-il-3ra antibody for use in treatment of blood tumor
WO2010129248A1 (en) 2009-05-06 2010-11-11 Centocor Ortho Biotech Inc. Melanocortin receptor binding conjugates
EP3320920A1 (en) 2009-05-08 2018-05-16 Vaccinex, Inc. Anti-cd100 antibodies and methods for using the same
KR20120024763A (en) 2009-05-15 2012-03-14 추가이 세이야쿠 가부시키가이샤 Anti-axl antibody
WO2010141329A1 (en) 2009-06-01 2010-12-09 Medimmune, Llc Molecules with extended half-lives and uses thereof
AU2010259533B2 (en) 2009-06-11 2015-03-12 Kyowa Kirin Co., Ltd. Process for production of protein
US20100316639A1 (en) 2009-06-16 2010-12-16 Genentech, Inc. Biomarkers for igf-1r inhibitor therapy
US9676845B2 (en) 2009-06-16 2017-06-13 Hoffmann-La Roche, Inc. Bispecific antigen binding proteins
JP5918129B2 (en) * 2009-06-22 2016-05-18 メディミューン,エルエルシー Engineered Fc region for site-specific conjugation
CN102549016B (en) * 2009-06-30 2015-05-06 研究发展基金会 Immunoglobulin FC polypeptides
AU2010273585B2 (en) 2009-07-13 2015-04-23 Genentech, Inc. Diagnostic methods and compositions for treatment of cancer
TW201106972A (en) 2009-07-27 2011-03-01 Genentech Inc Combination treatments
DK2463368T3 (en) 2009-08-07 2018-01-22 Kyowa Hakko Kirin Co Ltd HUMANIZED ANTI-AMYLOID-BETA OLIGOMER ANTIBODY
EP2463369A4 (en) 2009-08-07 2013-09-18 Kyowa Hakko Kirin Co Ltd Humanized anti-amyloid-b oligomer antibody
MX2012001716A (en) 2009-08-14 2012-04-02 Genentech Inc Biological markers for monitoring patient response to vegf antagonists.
US8221753B2 (en) * 2009-09-30 2012-07-17 Tracon Pharmaceuticals, Inc. Endoglin antibodies
KR101553740B1 (en) 2009-08-17 2015-09-17 트라콘 파마수티칼즈, 인코포레이티드 Combination therapy of cancer with anti-endoglin antibodies and anti-vegf agents
CA2772051C (en) 2009-08-24 2020-08-18 Amunix Operating Inc. Coagulation factor ix compositions and methods of making and using same
ES2912569T3 (en) 2009-08-28 2022-05-26 Teva Pharmaceuticals Int Gmbh Methods of treating visceral pain by administering antagonistic antibodies directed against calcitonin gene-related peptide
RU2570554C2 (en) 2009-08-31 2015-12-10 Роше Гликарт Аг Affinity-matured humanised anti-cea monoclonal antibodies
DK2473522T3 (en) 2009-09-02 2016-11-28 Genentech Inc Smoothened MUTANT AND METHODS OF USING THE SAME
WO2011028952A1 (en) 2009-09-02 2011-03-10 Xencor, Inc. Compositions and methods for simultaneous bivalent and monovalent co-engagement of antigens
MX353186B (en) 2009-09-03 2018-01-05 Genentech Inc Methods for treating, diagnosing, and monitoring rheumatoid arthritis.
CA2773662A1 (en) 2009-09-11 2011-03-17 Genentech, Inc. Method to identify a patient with an increased likelihood of responding to an anti-cancer agent
NZ701769A (en) 2009-09-16 2016-06-24 Genentech Inc Coiled coil and/or tether containing protein complexes and uses thereof
EP2481752B1 (en) 2009-09-24 2016-11-09 Chugai Seiyaku Kabushiki Kaisha Modified antibody constant regions
EP2480888B1 (en) 2009-09-25 2016-11-30 XOMA Technology Ltd. Screening methods
US8926976B2 (en) 2009-09-25 2015-01-06 Xoma Technology Ltd. Modulators
US8568726B2 (en) 2009-10-06 2013-10-29 Medimmune Limited RSV specific binding molecule
EP2486141B1 (en) 2009-10-07 2018-01-10 MacroGenics, Inc. Fc region-containing polypeptides that exhibit improved effector function due to alterations of the extent of fucosylation, and methods for their use
PL2488554T3 (en) 2009-10-14 2020-03-31 Humanigen, Inc. Antibodies to epha3
KR20120105447A (en) 2009-10-22 2012-09-25 제넨테크, 인크. Anti-hepsin antibodies and methods using same
WO2011056497A1 (en) 2009-10-26 2011-05-12 Genentech, Inc. Activin receptor type iib compositions and methods of use
WO2011056494A1 (en) 2009-10-26 2011-05-12 Genentech, Inc. Activin receptor-like kinase-1 antagonist and vegfr3 antagonist combinations
WO2011056502A1 (en) 2009-10-26 2011-05-12 Genentech, Inc. Bone morphogenetic protein receptor type ii compositions and methods of use
WO2011053982A2 (en) 2009-11-02 2011-05-05 University Of Washington Therapeutic nuclease compositions and methods
WO2011057198A1 (en) * 2009-11-09 2011-05-12 Carson Cantwell G Vaccine testing system
JP2013511281A (en) * 2009-11-23 2013-04-04 アムジェン インコーポレイテッド Monomeric antibody Fc
NZ628923A (en) 2009-11-24 2016-02-26 Medimmune Ltd Targeted binding agents against b7-h1
LT2510012T (en) 2009-12-09 2017-07-25 Bayer Pharma Aktiengesellschaft Anti-c4.4a antibodies and uses thereof
TWI505836B (en) 2009-12-11 2015-11-01 Genentech Inc Anti-vegf-c antibodies and methods using same
PL2516465T3 (en) 2009-12-23 2016-11-30 Anti-bv8 antibodies and uses thereof
HUE028629T2 (en) 2009-12-23 2016-12-28 Synimmune Gmbh Anti-flt3 antibodies and methods of using the same
US8362210B2 (en) 2010-01-19 2013-01-29 Xencor, Inc. Antibody variants with enhanced complement activity
US9074192B2 (en) 2010-01-22 2015-07-07 The Board Of Trustees Of The Leland Stanford Junior University Inhibition of AXL signaling in anti-metastatic therapy
WO2011089211A1 (en) 2010-01-22 2011-07-28 Synimmune Gmbh Anti-cd133 antibodies and methods of using the same
CA3056999A1 (en) 2010-01-22 2011-07-28 Amato J. Giaccia Inhibition of axl signaling in anti-metastatic therapy
PE20170687A1 (en) 2010-01-28 2017-06-13 Glaxo Group Ltd BINDING PROTEINS TO CD127
CA2788758A1 (en) 2010-02-09 2011-08-18 Andrew Ian Bayliffe Treatment of a metabolic disorder
RU2573994C2 (en) 2010-02-10 2016-01-27 Иммьюноджен, Инк Anti-cd20 antibodies and thereof application
MX2012008958A (en) 2010-02-18 2012-08-23 Genentech Inc Neuregulin antagonists and use thereof in treating cancer.
EP3590966A1 (en) 2010-02-23 2020-01-08 Sanofi Anti-alpha2 integrin antibodies and their uses
US9260529B2 (en) 2010-02-24 2016-02-16 The University Of Washington Through Its Center For Commercialization Molecules that bind CD180, compositions and methods of use
UA108227C2 (en) 2010-03-03 2015-04-10 ANTIGENCY PROTEIN
NZ705128A (en) 2010-03-04 2015-04-24 Macrogenics Inc Antibodies reactive with b7-h3, immunologically active fragments thereof and uses thereof
WO2011108714A1 (en) 2010-03-04 2011-09-09 中外製薬株式会社 Antibody constant region variant
US8802091B2 (en) 2010-03-04 2014-08-12 Macrogenics, Inc. Antibodies reactive with B7-H3 and uses thereof
BR112012022044A2 (en) 2010-03-24 2020-08-25 Genentech Inc ''antibody, immunoconjugate, pharmaceutical formulation, antibody use, treatment method, isolated bispecific antibody and host cell''.
TW201138821A (en) 2010-03-26 2011-11-16 Roche Glycart Ag Bispecific antibodies
CA2794745A1 (en) * 2010-03-29 2011-10-06 Zymeworks, Inc. Antibodies with enhanced or suppressed effector function
AU2011244282A1 (en) * 2010-04-20 2012-11-15 Genmab A/S Heterodimeric antibody Fc-containing proteins and methods for production thereof
WO2011133931A1 (en) 2010-04-22 2011-10-27 Genentech, Inc. Use of il-27 antagonists for treating inflammatory bowel disease
WO2011146568A1 (en) 2010-05-19 2011-11-24 Genentech, Inc. Predicting response to a her inhibitor
WO2011147834A1 (en) 2010-05-26 2011-12-01 Roche Glycart Ag Antibodies against cd19 and uses thereof
CA2800785C (en) 2010-05-27 2019-09-24 Genmab A/S Monoclonal antibodies against her2
WO2011153224A2 (en) 2010-06-02 2011-12-08 Genentech, Inc. Diagnostic methods and compositions for treatment of cancer
AR081556A1 (en) 2010-06-03 2012-10-03 Glaxo Group Ltd HUMANIZED ANTIGEN UNION PROTEINS
AU2011261362B2 (en) 2010-06-03 2016-06-09 Genentech, Inc. Immuno-pet imaging of antibodies and immunoconjugates and uses therefor
SG10201600791TA (en) 2010-06-08 2016-03-30 Genentech Inc Cysteine engineered antibodies and conjugates
TWI629483B (en) 2010-06-11 2018-07-11 協和醱酵麒麟有限公司 anti-TIM-3 antibody
CN103080136B (en) 2010-06-18 2015-08-12 霍夫曼-拉罗奇有限公司 Anti-Axl antibody and using method
WO2011161119A1 (en) 2010-06-22 2011-12-29 F. Hoffmann-La Roche Ag Antibodies against insulin-like growth factor i receptor and uses thereof
WO2011161189A1 (en) 2010-06-24 2011-12-29 F. Hoffmann-La Roche Ag Anti-hepsin antibodies and methods of use
AU2011274423B2 (en) 2010-07-09 2016-02-11 Bioverativ Therapeutics Inc. Chimeric clotting factors
NZ605449A (en) 2010-07-09 2015-03-27 Genentech Inc Anti-neuropilin antibodies and methods of use
EP2409712A1 (en) 2010-07-19 2012-01-25 International-Drug-Development-Biotech Anti-CD19 antibody having ADCC and CDC functions and improved glycosylation profile
WO2012010582A1 (en) 2010-07-21 2012-01-26 Roche Glycart Ag Anti-cxcr5 antibodies and methods of use
RU2598711C2 (en) 2010-07-22 2016-09-27 Те Риджентс Оф Те Юниверсити Оф Калифорния Anti-tumour antigen antibody and methods of application
EP2598882B1 (en) 2010-07-30 2017-07-26 AC Immune S.A. Safe and functional humanized antibodies for use in treating an amyloidosis
EP2601216B1 (en) 2010-08-02 2018-01-03 MacroGenics, Inc. Covalent diabodies and uses thereof
BR112013002535A2 (en) 2010-08-03 2019-09-24 Hoffmann La Roche biomarkers of chronic lymphocytic leukemia (cll)
RU2013106217A (en) 2010-08-05 2014-09-10 Ф. Хоффманн-Ля Рош Аг HYBRID PROTEIN FROM ANTIBODIES AGAINST MHC AND ANTIVIRAL CYTOKINE
WO2012019061A2 (en) 2010-08-05 2012-02-09 Stem Centrx, Inc. Novel effectors and methods of use
AU2011288487B2 (en) 2010-08-13 2015-10-01 Roche Glycart Ag Anti-FAP antibodies and methods of use
MX2013001336A (en) 2010-08-13 2013-03-08 Roche Glycart Ag Anti-tenascin-c a2 antibodies and methods of use.
AU2011288412A1 (en) 2010-08-13 2013-02-21 Medimmune Limited Monomeric polypeptides comprising variant Fc regions and methods of use
WO2012022734A2 (en) 2010-08-16 2012-02-23 Medimmune Limited Anti-icam-1 antibodies and methods of use
NZ604510A (en) 2010-08-17 2013-10-25 Csl Ltd Dilutable biocidal compositions and methods of use
EP2609117B1 (en) 2010-08-25 2015-09-16 F.Hoffmann-La Roche Ag Antibodies against il-18r1 and uses thereof
SG187965A1 (en) 2010-08-27 2013-04-30 Stem Centrx Inc Notum protein modulators and methods of use
TWI560199B (en) 2010-08-31 2016-12-01 Sanofi Sa Peptide or peptide complex binding to α2 integrin and methods and uses involving the same
DK2612151T3 (en) 2010-08-31 2017-10-02 Genentech Inc BIOMARKETS AND METHODS OF TREATMENT
CA2810217C (en) 2010-09-02 2019-03-12 Vaccinex, Inc. Anti-cxcl13 antibodies and methods of using the same
WO2012028683A1 (en) 2010-09-02 2012-03-08 Novartis Ag Antibody gel system for sustained drug delivery
BR112013005116A2 (en) 2010-09-03 2019-09-24 Stem Centrx Inc modulators and methods of use
US9150655B2 (en) 2010-09-03 2015-10-06 Academia Sinica Anti-C-met antibody and methods of use thereof
TW201302793A (en) 2010-09-03 2013-01-16 Glaxo Group Ltd Novel antigen binding proteins
WO2012045085A1 (en) 2010-10-01 2012-04-05 Oxford Biotherapeutics Ltd. Anti-rori antibodies
AU2011312205B2 (en) 2010-10-05 2015-08-13 Curis, Inc. Mutant smoothened and methods of using the same
AU2011325833C1 (en) 2010-11-05 2017-07-13 Zymeworks Bc Inc. Stable heterodimeric antibody design with mutations in the Fc domain
US20130330350A1 (en) * 2010-11-09 2013-12-12 Medimmune, Llc Antibody Scaffold For Homogenous Conjugation
JP6163429B2 (en) 2010-11-10 2017-07-12 ジェネンテック, インコーポレイテッド Methods and compositions for neurological disease immunotherapy
KR102385507B1 (en) * 2010-11-30 2022-04-12 추가이 세이야쿠 가부시키가이샤 Antigen-binding molecule capable of binding to plurality of antigen molecules repeatedly
WO2012075111A1 (en) 2010-11-30 2012-06-07 Novartis Ag Uses of anti-cd40 antibodies in combination therapy for b cell-related cancers
CN103429737B (en) 2010-11-30 2020-07-14 中外制药株式会社 Cytotoxic induction therapeutic agent
CN106279415A (en) 2010-12-08 2017-01-04 施特姆森特克斯股份有限公司 Novel modulator and using method
EP2653541B1 (en) 2010-12-15 2017-11-08 Kyowa Hakko Kirin Co., Ltd. Method for producing proteins
CN107312796A (en) 2010-12-15 2017-11-03 大学共同利用机关法人情报·系统研究机构 Method for producing protein
NZ609493A (en) 2010-12-16 2015-11-27 Genentech Inc Diagnosis and treatments relating to th2 inhibition
EA028744B1 (en) 2010-12-20 2017-12-29 Дженентек, Инк. Anti-mesothelin antibodies and immunoconjugates
KR20130118925A (en) 2010-12-22 2013-10-30 제넨테크, 인크. Anti-pcsk9 antibodies and methods of use
JOP20210044A1 (en) 2010-12-30 2017-06-16 Takeda Pharmaceuticals Co Anti-cd38 antibodies
CA2826467C (en) 2011-02-07 2019-11-12 Research Development Foundation Engineered immunoglobulin fc polypeptides
EP2672999A2 (en) 2011-02-10 2013-12-18 Roche Glycart AG Improved immunotherapy
SA112330278B1 (en) 2011-02-18 2015-10-09 ستيم سينتركس، انك. Novel modulators and methods of use
AR085302A1 (en) * 2011-02-24 2013-09-18 Sanofi Sa METHOD OF PRODUCTION OF STIRATED ANTIBODIES
US20140093496A1 (en) * 2011-02-25 2014-04-03 Chugai Seiyaku Kabushiki Kaisha Fc-gamma-RIIb-SPECIFIC Fc ANTIBODY
CA2824824A1 (en) 2011-02-28 2012-09-07 F. Hoffmann-La Roche Ag Monovalent antigen binding proteins
RU2013140975A (en) 2011-02-28 2015-04-10 Дженентек, Инк. BIOLOGICAL MARKERS AND METHODS FOR PREDICTING SUSCEPTIBILITY TO B-CELL ANTAGONISTS
CA2825081A1 (en) 2011-02-28 2012-09-07 Birgit Bossenmaier Antigen binding proteins
PL2681244T3 (en) 2011-03-02 2018-04-30 Roche Glycart Ag Cea antibodies
US20140112926A1 (en) * 2011-03-16 2014-04-24 Amgen Inc. Fc VARIANTS
RU2013143358A (en) 2011-04-07 2015-05-20 Дженентек, Инк. ANTI-FGFR4 ANTIBODIES AND WAYS OF THEIR APPLICATION
JP5977814B2 (en) 2011-04-08 2016-08-24 アムジエン・インコーポレーテツド Method for treating or ameliorating metabolic disorders using growth differentiation factor 15 (GDF-15)
ES2608835T3 (en) 2011-04-13 2017-04-17 Bristol-Myers Squibb Company Fc fusion proteins comprising new linkers or arrangements
EA201892619A1 (en) 2011-04-29 2019-04-30 Роше Гликарт Аг IMMUNOCONJUGATES CONTAINING INTERLEUKIN-2 MUTANT POLYPETIPS
SI2704737T1 (en) 2011-04-29 2018-06-29 University Of Washington Therapeutic nuclease compositions and methods
WO2012146630A1 (en) 2011-04-29 2012-11-01 F. Hoffmann-La Roche Ag N-terminal acylated polypeptides, methods for their production and uses thereof
MX2013013054A (en) 2011-05-12 2014-02-20 Genentech Inc Multiple reaction monitoring lc-ms/ms method to detect therapeutic antibodies in animal samples using framework signature peptides.
PE20140995A1 (en) 2011-05-16 2014-08-23 Genentech Inc FGFR1 AGONISTS AND THEIR METHODS OF USE
US9745373B2 (en) 2011-05-20 2017-08-29 Alderbio Holdings Llc Anti-CGRP compositions and use thereof
JP6189832B2 (en) 2011-05-20 2017-08-30 アルダーバイオ・ホールディングズ・エルエルシー Use of anti-CGRP or anti-CGRP-R antibodies or antibody fragments to treat or prevent chronic and acute diarrhea
CN103702685B (en) 2011-05-20 2017-12-15 奥尔德生物控股有限责任公司 Anti- CGRP antibody and antibody fragment are used to prevent or suppress photophobia in subject in need, especially migraineur or detest the purposes of light
EP2714733B1 (en) 2011-05-21 2019-01-23 MacroGenics, Inc. Cd3-binding molecules capable of binding to human and non-human cd3
AU2012259162C1 (en) 2011-05-21 2020-05-21 Macrogenics, Inc. Deimmunized serum-binding domains and their use for extending serum half-life
CA2837169C (en) 2011-05-24 2021-11-09 Zyngenia, Inc. Multispecific complexes comprising angiopoietin-2-binding peptide and their uses
SG10201902706VA (en) 2011-06-03 2019-04-29 Xoma Technology Ltd Antibodies specific for tgf-beta
WO2012170740A2 (en) 2011-06-07 2012-12-13 University Of Hawaii Biomarker of asbestos exposure and mesothelioma
US9561274B2 (en) 2011-06-07 2017-02-07 University Of Hawaii Treatment and prevention of cancer with HMGB1 antagonists
CA2838833A1 (en) 2011-06-10 2012-12-13 Biogen Idec Ma Inc. Pro-coagulant compounds and methods of use thereof
BR112013032235A2 (en) 2011-06-15 2016-11-22 Hoffmann La Roche anti-human epo receptor antibodies and methods of use
WO2012176779A1 (en) 2011-06-20 2012-12-27 協和発酵キリン株式会社 Anti-erbb3 antibody
MX354663B (en) 2011-06-22 2018-03-14 Hoffmann La Roche Removal of target cells by circulating virus-specific cytotoxic t-cells using mhc class i comprising complexes.
WO2013003641A2 (en) 2011-06-28 2013-01-03 Inhibrx Llc Serpin fusion polypeptides and methods of use thereof
US10400029B2 (en) 2011-06-28 2019-09-03 Inhibrx, Lp Serpin fusion polypeptides and methods of use thereof
US8986688B2 (en) 2011-06-28 2015-03-24 Inhibrx, Llc WAP domain fusion polypeptides and methods of use thereof
AR086823A1 (en) 2011-06-30 2014-01-22 Genentech Inc ANTI-C-MET ANTIBODY FORMULATIONS, METHODS
RU2641256C2 (en) 2011-06-30 2018-01-16 Чугаи Сейяку Кабусики Кайся Heterodimerizated polypeptide
UA117901C2 (en) * 2011-07-06 2018-10-25 Ґенмаб Б.В. Antibody variants and uses thereof
US9738707B2 (en) * 2011-07-15 2017-08-22 Biogen Ma Inc. Heterodimeric Fc regions, binding molecules comprising same, and methods relating thereto
CA2842375A1 (en) 2011-08-17 2013-02-21 Erica Jackson Neuregulin antibodies and uses thereof
BR112014003999A2 (en) 2011-08-23 2017-06-13 Roche Glycart Ag isolated antibody that binds to a near epitope near the human mcsp membrane, isolated nucleic acid, host cell, method of producing an antibody, immunoconjugate, pharmaceutical formulation, use of the antibody, method of treating cancer patients, unduction method of cell lysis in individuals and mcsp immunohistochemical test
LT2748202T (en) 2011-08-23 2018-09-25 Roche Glycart Ag Bispecific antigen binding molecules
WO2013026839A1 (en) 2011-08-23 2013-02-28 Roche Glycart Ag Bispecific antibodies specific for t-cell activating antigens and a tumor antigen and methods of use
UA116192C2 (en) 2011-08-23 2018-02-26 Рош Глікарт Аг Bispecific t cell activating antigen binding molecules
US20130078250A1 (en) 2011-08-23 2013-03-28 Oliver Ast Bispecific t cell activating antigen binding molecules
US20130058947A1 (en) 2011-09-02 2013-03-07 Stem Centrx, Inc Novel Modulators and Methods of Use
UY34317A (en) 2011-09-12 2013-02-28 Genzyme Corp T cell antireceptor antibody (alpha) / ß
WO2013039954A1 (en) 2011-09-14 2013-03-21 Sanofi Anti-gitr antibodies
MX2014003094A (en) 2011-09-15 2014-04-25 Genentech Inc Methods of promoting differentiation.
GB201116092D0 (en) 2011-09-16 2011-11-02 Bioceros B V Antibodies and uses thereof
CN103930111A (en) 2011-09-19 2014-07-16 霍夫曼-拉罗奇有限公司 Combination treatments comprising C-MET antagonists and B-RAF antagonists
JP2014530009A (en) * 2011-09-29 2014-11-17 エーピーオー‐ティー ビー.ヴイ. Multispecific binding molecules targeting abnormal cells
CN104114577A (en) 2011-09-30 2014-10-22 特瓦制药澳大利亚私人有限公司 Antibodies against TL1a and uses thereof
CA2850322C (en) * 2011-09-30 2023-10-10 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule inducing immune response to target antigen
BR112014007487A2 (en) 2011-09-30 2017-04-04 Dana Farber Cancer Inst Inc therapeutic peptides
US20140335089A1 (en) * 2011-09-30 2014-11-13 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule for promoting elimination of antigens
TW201817745A (en) 2011-09-30 2018-05-16 日商中外製藥股份有限公司 Therapeutic antigen-binding molecule with a FcRn-binding domain that promotes antigen clearance
WO2013047748A1 (en) 2011-09-30 2013-04-04 中外製薬株式会社 Antigen-binding molecule promoting disappearance of antigens having plurality of biological activities
US9663573B2 (en) 2011-10-05 2017-05-30 Genentech, Inc. Methods of treating liver conditions using Notch2 antagonists
SI2771022T1 (en) 2011-10-11 2020-12-31 Viela Bio, Inc. Cd40l-specific tn3-derived scaffolds and methods of use thereof
ES2669209T3 (en) 2011-10-11 2018-05-24 Vaccinex, Inc. Use of semaforin-4D binding molecules for the modulation of the blood brain barrier permeability
US9738727B2 (en) 2011-10-14 2017-08-22 Genentech, Inc. Anti-HtrA1 antibodies and methods of use
WO2013056148A2 (en) 2011-10-15 2013-04-18 Genentech, Inc. Methods of using scd1 antagonists
WO2013059531A1 (en) 2011-10-20 2013-04-25 Genentech, Inc. Anti-gcgr antibodies and uses thereof
SG10201805291TA (en) 2011-10-27 2018-08-30 Genmab As Production of heterodimeric proteins
CA2852709A1 (en) 2011-10-28 2013-05-02 Patrys Limited Pat-lm1 epitopes and methods for using same
SG11201401815XA (en) 2011-10-28 2014-05-29 Genentech Inc Therapeutic combinations and methods of treating melanoma
CN104011207B (en) 2011-10-31 2018-09-18 中外制药株式会社 Control the antigen binding molecules of the association of heavy chain and light chain
EP2773667A1 (en) 2011-11-01 2014-09-10 Bionomics, Inc. Anti-gpr49 antibodies
EP2773664A1 (en) 2011-11-01 2014-09-10 Bionomics, Inc. Anti-gpr49 antibodies
AU2012332587B2 (en) 2011-11-01 2017-02-23 Bionomics, Inc. Antibodies and methods of treating cancer
AU2012332588B2 (en) 2011-11-01 2017-09-07 Bionomics, Inc. Methods of blocking cancer stem cell growth
KR102052774B1 (en) 2011-11-04 2019-12-04 자임워크스 인코포레이티드 Stable heterodimeric antibody design with mutations in the fc domain
US9580509B2 (en) 2011-11-07 2017-02-28 Medimmune, Llc Multispecific and multivalent binding proteins and uses thereof
CA2854720C (en) 2011-11-11 2018-12-18 Rinat Neuroscience Corp. Antibodies specific for trop-2 and their uses
RU2014124842A (en) 2011-11-21 2015-12-27 Дженентек, Инк. CLEANING ANTI-C-MET ANTIBODIES
RU2014124530A (en) 2011-11-23 2015-12-27 Идженика, Инк. ANTIBODIES TO CD98 AND WAYS OF THEIR APPLICATION
AR088941A1 (en) 2011-11-23 2014-07-16 Bayer Ip Gmbh ANTI-FGFR2 ANTIBODIES AND THEIR USES
KR20140100532A (en) 2011-11-30 2014-08-14 추가이 세이야쿠 가부시키가이샤 Drug containing carrier into cell for forming immune complex
US9416179B2 (en) 2011-12-05 2016-08-16 X-Body, Inc. PDGF receptor beta binding polypeptides
EP2788024A1 (en) 2011-12-06 2014-10-15 F.Hoffmann-La Roche Ag Antibody formulation
WO2013090776A1 (en) 2011-12-15 2013-06-20 The Board Of Trustees Of The Leland Stanford Junior University Inhibition of axl/gas6 signaling in the treatment of disease
US9527927B2 (en) 2011-12-20 2016-12-27 Medimmune, Llc Modified polypeptides for bispecific antibody scaffolds
AR089434A1 (en) 2011-12-23 2014-08-20 Genentech Inc PROCEDURE TO PREPARE FORMULATIONS WITH HIGH CONCENTRATION OF PROTEINS
EP3539982A3 (en) 2011-12-23 2020-01-15 Pfizer Inc Engineered antibody constant regions for site-specific conjugation and methods and uses therefor
KR102041412B1 (en) * 2011-12-30 2019-11-11 한미사이언스 주식회사 Derivatives of Immunglobulin Fc fragment
AU2013208003B2 (en) 2012-01-09 2017-12-14 The Scripps Research Institute Ultralong complementarity determining regions and uses thereof
CN104411717A (en) 2012-01-09 2015-03-11 斯克利普斯研究所 Humanized antibodies with ultralong CDR3S
HUE046396T2 (en) 2012-01-12 2020-02-28 Bioverativ Therapeutics Inc Chimeric factor viii polypeptides and uses thereof
AU2013207778B2 (en) 2012-01-13 2017-10-12 Genentech, Inc. Biological markers for identifying patients for treatment with VEGF antagonists
JP2015504895A (en) 2012-01-13 2015-02-16 エーピーオー‐ティー ビー.ヴイ. Abnormal cell-restricted immunoglobulin with a toxic moiety
WO2013109856A2 (en) 2012-01-18 2013-07-25 Genentech, Inc. Methods of using fgf19 modulators
JP6242813B2 (en) 2012-01-18 2017-12-06 ジェネンテック, インコーポレイテッド Anti-LRP5 antibody and method of use
EP2807266B1 (en) 2012-01-26 2020-01-15 Amgen Inc. Growth differentiation factor 15 (gdf-15) polypeptides
WO2013117647A1 (en) 2012-02-07 2013-08-15 Innate Pharma Mica binding agents
CN113398268A (en) 2012-02-11 2021-09-17 霍夫曼-拉罗奇有限公司 R-spondin translocations and methods of use thereof
KR102008190B1 (en) 2012-02-15 2019-08-07 바이오버라티브 테라퓨틱스 인크. Recombinant factor viii proteins
WO2013120929A1 (en) 2012-02-15 2013-08-22 F. Hoffmann-La Roche Ag Fc-receptor based affinity chromatography
ES2935489T3 (en) 2012-02-15 2023-03-07 Bioverativ Therapeutics Inc Factor VIII compositions and methods of preparation and use thereof
SI2817338T1 (en) 2012-02-24 2017-11-30 Abbvie Stemcentrx Llc Dll3 modulators and methods of use
JP6193275B2 (en) 2012-03-02 2017-09-06 ヴァクシネックス, インコーポレイテッド Methods for treating B cell mediated inflammatory diseases
AU2013240261A1 (en) 2012-03-27 2014-09-18 Genentech, Inc. Diagnosis and treatments relating to HER3 inhibitors
UA117097C2 (en) 2012-03-28 2018-06-25 Санофі Antibodies to bradykinin b1 receptor ligands
AR090549A1 (en) 2012-03-30 2014-11-19 Genentech Inc ANTI-LGR5 AND IMMUNOCATE PLAYERS
CA2867588A1 (en) 2012-03-30 2013-10-03 Genentech, Inc. Diagnostic methods and compositions for treatment of cancer
EP2834270B1 (en) 2012-04-05 2019-10-30 AC Immune S.A. Humanized tau antibody
US10385395B2 (en) 2012-04-11 2019-08-20 The Regents Of The University Of California Diagnostic tools for response to 6-thiopurine therapy
AU2013251309B2 (en) 2012-04-27 2017-06-22 Bioatla, Llc Modified antibody regions and uses thereof
WO2013165690A1 (en) * 2012-04-30 2013-11-07 Medimmune, Llc Molecules with reduced effector function and extended half-lives, compositions, and uses thereof
WO2013165940A1 (en) 2012-05-01 2013-11-07 Genentech, Inc. Anti-pmel17 antibodies and immunoconjugates
CA2872856A1 (en) 2012-05-07 2013-11-14 Sanofi Methods for preventing biofilm formation
US20130336973A1 (en) 2012-05-10 2013-12-19 Zymeworks Inc. Heteromultimer Constructs of Immunoglobulin Heavy Chains with Mutations in the Fc Domain
WO2013170191A1 (en) 2012-05-11 2013-11-14 Genentech, Inc. Methods of using antagonists of nad biosynthesis from nicotinamide
SG11201407512VA (en) 2012-05-18 2014-12-30 Genentech Inc High-concentration monoclonal antibody formulations
MX2014014166A (en) 2012-05-21 2015-03-03 Genentech Inc Methods for improving safety of blood-brain barrier transport.
US9844582B2 (en) 2012-05-22 2017-12-19 Massachusetts Institute Of Technology Synergistic tumor treatment with extended-PK IL-2 and therapeutic agents
EP2852840B1 (en) 2012-05-23 2019-10-09 F.Hoffmann-La Roche Ag Selection method for therapeutic agents
WO2013175276A1 (en) 2012-05-23 2013-11-28 Argen-X B.V Il-6 binding molecules
CN104427995A (en) 2012-06-08 2015-03-18 比奥根艾迪克Ma公司 Chimeric clotting factors
AU2013270682A1 (en) 2012-06-08 2014-12-11 Biogen Ma Inc. Procoagulant compounds
JP6628966B2 (en) 2012-06-14 2020-01-15 中外製薬株式会社 Antigen binding molecule containing an altered Fc region
WO2013188855A1 (en) 2012-06-15 2013-12-19 Genentech, Inc. Anti-pcsk9 antibodies, formulations, dosing, and methods of use
US20140051834A1 (en) 2012-06-21 2014-02-20 Hoffmann-La Roche, Inc. Incretin Receptor Ligand Polypeptide Fc-Region Fusion Polypeptides And Conjugates With Altered Fc-Effector Function
US9499634B2 (en) 2012-06-25 2016-11-22 Zymeworks Inc. Process and methods for efficient manufacturing of highly pure asymmetric antibodies in mammalian cells
RU2630296C2 (en) 2012-07-04 2017-09-06 Ф. Хоффманн-Ля Рош Аг Antibodies to biotin and application methods
RU2630664C2 (en) 2012-07-04 2017-09-11 Ф. Хоффманн-Ля Рош Аг Theophylline antibodies and methods for their application
SG11201407420RA (en) 2012-07-04 2015-02-27 Hoffmann La Roche Covalently linked antigen-antibody conjugates
CA3188124A1 (en) 2012-07-05 2014-01-09 Genentech, Inc. Expression and secretion system
EP2870973B1 (en) 2012-07-06 2022-08-31 St. Marianna University School of Medicine Remedy for htlv-1-associated myelopathy patients
EP3404105A1 (en) 2012-07-06 2018-11-21 Bioverativ Therapeutics Inc. Cell line expressing single chain factor viii polypeptides and uses thereof
AU2013285355A1 (en) 2012-07-06 2015-01-29 Genmab B.V. Dimeric protein with triple mutations
EP3632462A1 (en) 2012-07-06 2020-04-08 Genmab B.V. Dimeric protein with triple mutations
AU2013288929A1 (en) 2012-07-09 2014-12-04 Genentech, Inc. Immunoconjugates comprising anti-CD22 antibodies
PE20150325A1 (en) 2012-07-09 2015-03-05 Genentech Inc IMMUNOCONJUGATES INCLUDING ANTI-CD22 ANTIBODIES AND DERIVATIVES OF NEMORUBICIN.
EA201590172A1 (en) 2012-07-09 2015-09-30 Дженентек, Инк. IMMUNOCONGATES CONTAINING ANTIBODIES TO CD79b
IN2014DN10652A (en) 2012-07-09 2015-09-11 Genentech Inc
JP6603128B2 (en) 2012-07-11 2019-11-06 バイオベラティブ セラピューティクス インコーポレイテッド Complex of factor VIII with XTEN and von Willebrand factor protein and uses thereof
US20140154253A1 (en) * 2012-07-13 2014-06-05 Zymeworks Inc. Bispecific Asymmetric Heterodimers Comprising Anti-CD3 Constructs
BR112015001459B1 (en) 2012-07-25 2023-02-14 Celldex Therapeutics, Inc ISOLATED ANTIBODY OR FRAGMENT THEREOF, CONJUGATE, USES THEREOF, PHARMACEUTICAL COMPOSITION, POLYNUCLEOTIDE, VECTOR, HOST CELL, ISOLATED CELL, KIT, IN VITRO METHOD TO INHIBIT KIT ACTIVITY, METHOD TO PRODUCE AN ANTIBODY
JP6388581B2 (en) 2012-08-02 2018-09-12 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft Method for producing monomer and multimeric molecules and uses thereof
DK2880170T3 (en) 2012-08-02 2016-10-24 Hoffmann La Roche PROCEDURE FOR PREPARING SOLUBLE FcR AS Fc FUSION WITH INERT IMMUNOGLOBULIN Fc REGION AND APPLICATIONS THEREOF
MY175687A (en) 2012-08-07 2020-07-06 Roche Glycart Ag Composition comprising two antibodies engineered to have reduced and increased effector function
CN104540848B (en) 2012-08-08 2019-05-31 罗切格利卡特公司 Interleukin-10 fusion protein and application thereof
WO2014023709A1 (en) 2012-08-09 2014-02-13 Roche Glycart Ag Asgpr antibodies and uses thereof
US20140044675A1 (en) 2012-08-10 2014-02-13 Roche Glycart Ag Interleukin-2 fusion proteins and uses thereof
RU2729831C2 (en) 2012-08-24 2020-08-12 Чугаи Сейяку Кабусики Кайся Versions of fcγriib-specific fc-region
JP6774164B2 (en) 2012-08-24 2020-10-21 中外製薬株式会社 Mouse FcγRII specific Fc antibody
UA115789C2 (en) 2012-09-05 2017-12-26 Трейкон Фармасутікалз, Інк. Antibody formulations and uses thereof
US9790268B2 (en) 2012-09-12 2017-10-17 Genzyme Corporation Fc containing polypeptides with altered glycosylation and reduced effector function
AU2013315499B2 (en) 2012-09-12 2018-08-09 Genzyme Corporation Fc containing polypeptides with altered glycosylation and reduced effector function
JP6273205B2 (en) 2012-10-05 2018-01-31 協和発酵キリン株式会社 Heterodimeric protein composition
EP2904016B1 (en) 2012-10-08 2018-11-14 Roche Glycart AG Fc-free antibodies comprising two fab-fragments and methods of use
CA2887129A1 (en) 2012-10-09 2014-04-17 Igenica, Inc. Anti-c16orf54 antibodies and methods of use thereof
EP2914621B1 (en) 2012-11-05 2023-06-07 Foundation Medicine, Inc. Novel ntrk1 fusion molecules and uses thereof
AR093378A1 (en) 2012-11-08 2015-06-03 Hoffmann La Roche BINDING PROTEINS OF ANTIGEN HER3 OF UNION TO THE FORK b OF HER3
MX2015005874A (en) 2012-11-09 2015-09-10 Pfizer Platelet-derived growth factor b specific antibodies and compositions and uses thereof.
EA201892509A1 (en) 2012-11-13 2019-04-30 Дженентек, Инк. ANTIBODIES TO HEMAGGLUTININ AND METHODS OF APPLICATION
SI3447069T1 (en) 2012-11-21 2021-02-26 Janssen Biotech, Inc. Bispecific egfr/c-met antibodies
US9914785B2 (en) 2012-11-28 2018-03-13 Zymeworks Inc. Engineered immunoglobulin heavy chain-light chain pairs and uses thereof
WO2014088040A1 (en) 2012-12-06 2014-06-12 国立大学法人 金沢大学 Method for treating mesothelioma
EP2930240B1 (en) 2012-12-07 2018-08-01 Kyowa Hakko Kirin Co., Ltd. Anti-folr1 antibody
EP3686218A1 (en) 2012-12-10 2020-07-29 Biogen MA Inc. Anti-blood dendritic cell antigen 2 antibodies and uses thereof
FI2931265T6 (en) * 2012-12-14 2023-05-23 Modified axl peptides and their use in inhibition of axl signaling in anti-metastatic therapy
TWI693073B (en) 2012-12-21 2020-05-11 日商中外製藥股份有限公司 Therapeutic agent for GPC3 target is the effective therapeutic agent for GPC3 target administered to patients
WO2014096015A1 (en) 2012-12-21 2014-06-26 F. Hoffmann-La Roche Ag Disulfide-linked multivalent mhc class i comprising multi-function proteins
US10766960B2 (en) * 2012-12-27 2020-09-08 Chugai Seiyaku Kabushiki Kaisha Heterodimerized polypeptide
KR20220156667A (en) 2013-01-10 2022-11-25 젠맵 비. 브이 Human igg1 fc region variants and uses thereof
AU2014207549B2 (en) 2013-01-15 2018-12-06 Xencor, Inc. Rapid clearance of antigen complexes using novel antibodies
CA2898326C (en) 2013-01-18 2022-05-17 Foundation Medicine, Inc. Methods of treating cholangiocarcinoma
WO2014116749A1 (en) 2013-01-23 2014-07-31 Genentech, Inc. Anti-hcv antibodies and methods of using thereof
US9840559B2 (en) 2013-02-01 2017-12-12 The Regents Of The University Of California Anti-CD83 antibodies and use thereof
CN110551217B (en) 2013-02-07 2023-12-15 Csl有限公司 IL-11R binding proteins and uses thereof
CN115925957A (en) 2013-02-08 2023-04-07 Irm责任有限公司 Specific sites for modifying antibodies to make immunoconjugates
ES2813501T3 (en) 2013-02-12 2021-03-24 Bristol Myers Squibb Co Protein Refolding Methods Based on Tangential Flow Filtration
US10065987B2 (en) 2013-02-12 2018-09-04 Bristol-Myers Squibb Company High pH protein refolding methods
ES2747920T3 (en) 2013-02-14 2020-03-12 Innate Pharma Anti-NKP46 antibody for diagnosis of peripheral non-cutaneous T-cell lymphoma (PTCL)
EP4223772A3 (en) 2013-02-15 2023-10-18 Bioverativ Therapeutics Inc. Optimized factor viii gene
CN105026427B (en) 2013-02-20 2019-12-24 依奈特制药公司 Compounds that specifically bind to KIR3DL2 for use in the treatment of peripheral T cell lymphoma
DK2958944T3 (en) 2013-02-22 2019-06-24 Abbvie Stemcentrx Llc ANTI-DLL3 ANTISTOF PBD CONJUGATES AND USES THEREOF
CA2900097A1 (en) 2013-02-22 2014-08-28 F. Hoffmann-La Roche Ag Methods of treating cancer and preventing drug resistance
WO2014131694A1 (en) 2013-02-26 2014-09-04 Roche Glycart Ag Bispecific t cell activating antigen binding molecules
US20140242083A1 (en) 2013-02-26 2014-08-28 Roche Glycart Ag Anti-mcsp antibodies
JP2016512421A (en) 2013-02-26 2016-04-28 ロシュ グリクアート アーゲー Bispecific T cell activation antigen binding molecule
EP2961771B1 (en) * 2013-02-26 2020-01-01 Roche Glycart AG Bispecific t cell activating antigen binding molecules specific to cd3 and cea
US9487587B2 (en) 2013-03-05 2016-11-08 Macrogenics, Inc. Bispecific molecules that are immunoreactive with immune effector cells of a companion animal that express an activating receptor and cells that express B7-H3 and uses thereof
EP2964260A2 (en) 2013-03-06 2016-01-13 F. Hoffmann-La Roche AG Methods of treating and preventing cancer drug resistance
US20160002325A1 (en) 2013-03-08 2016-01-07 Vaccinex, Inc. Anti-cxcl13 antibodies and associated epitope sequences
KR102420934B1 (en) 2013-03-11 2022-07-15 젠자임 코포레이션 Hyperglycosylated binding polypeptides
EP4218409A3 (en) 2013-03-13 2023-08-30 Regeneron Pharmaceuticals, Inc. Mice expressing a limited immunoglobulin light chain repertoire
JP2016519568A (en) 2013-03-13 2016-07-07 リジェネロン・ファーマシューティカルズ・インコーポレイテッドRegeneron Pharmaceuticals, Inc. Common light chain mouse
US9580486B2 (en) 2013-03-14 2017-02-28 Amgen Inc. Interleukin-2 muteins for the expansion of T-regulatory cells
AP2015008740A0 (en) 2013-03-14 2015-09-30 Macrogenics Inc Bispecific molecules that are immunoreactive with immune effector cells that express an activating receptor and an antigen expressed by a cell infected by a virus and uses thereof
CN105307683A (en) 2013-03-14 2016-02-03 基因泰克公司 Methods of treating cancer and preventing cancer drug resistance
US9562099B2 (en) 2013-03-14 2017-02-07 Genentech, Inc. Anti-B7-H4 antibodies and immunoconjugates
JP6436965B2 (en) 2013-03-14 2018-12-12 ジェネンテック, インコーポレイテッド Anti-B7-H4 antibody and immunoconjugate
BR112015022576A2 (en) 2013-03-14 2017-10-24 Genentech Inc pharmaceutical product and its use, kit and method for treating hyperproliferative dysfunction
WO2014143739A2 (en) 2013-03-15 2014-09-18 Biogen Idec Ma Inc. Anti-alpha v beta 6 antibodies and uses thereof
KR20150128707A (en) 2013-03-15 2015-11-18 제넨테크, 인크. Compositions and methods for diagnosis and treatment of hepatic cancers
BR112015023752B1 (en) 2013-03-15 2023-11-14 Zyngenia, Inc. MODULAR RECOGNITION DOMAIN (MRD), COMPLEX COMPRISING MRD AND CETUXIMAB, USES OF THE COMPLEX TO INHIBIT ANGIOGENESIS AND TREAT CANCER AND PHARMACEUTICAL COMPOSITION COMPRISING SAID COMPLEX
US10035859B2 (en) 2013-03-15 2018-07-31 Biogen Ma Inc. Anti-alpha V beta 6 antibodies and uses thereof
CA2903772A1 (en) 2013-03-15 2014-09-25 Novartis Ag Antibody drug conjugates
BR112015023120A2 (en) 2013-03-15 2017-11-21 Genentech Inc method for identifying an individual with a disease or dysfunction, method for predicting the responsiveness of an individual with a disease or dysfunction, method for determining the likelihood that an individual with a disease or dysfunction will exhibit benefit from treatment, method for selecting a therapy, Uses of a pd-11 Axis Binding Antagonist, Assay to Identify an Individual with a Disease, Diagnostic Kit, Method to Evaluate a Treatment Response, and Method to Monitor the Response of a Treated Individual
US9598485B2 (en) 2013-03-15 2017-03-21 Ac Immune S.A. Anti-tau antibodies and methods of use
CN105143265A (en) 2013-03-15 2015-12-09 豪夫迈·罗氏有限公司 Anti-crth2 antibodies and their use
SG11201505926VA (en) 2013-03-15 2015-09-29 Biogen Ma Inc Factor ix polypeptide formulations
CN105263514B (en) 2013-03-15 2019-04-26 本质生命科学有限公司 Anti- hepcidin antibody and application thereof
BR112015023203A8 (en) 2013-03-15 2018-01-23 Constellation Pharmaceuticals Inc methods for treating cancer, method for increasing the efficiency of cancer treatment, method for delaying and / or preventing cancer development, method for treating an individual with cancer, method for increasing sensitivity for a cancer therapy agent, method for extending a sensitivity period and method for extending the duration of response to cancer therapy.
PE20151673A1 (en) 2013-03-15 2015-11-27 Genentech Inc IL-22 POLYPEPTIDES AND IL-22 Fc FUSION PROTEINS AND METHODS OF USE
BR112015021576A2 (en) 2013-03-15 2017-10-10 Dana Farber Cancer Inst Inc therapeutic peptides
WO2014151680A1 (en) 2013-03-15 2014-09-25 Biogen Idec Ma Inc. Treatment and prevention of acute kidney injury using anti-alpha v beta 5 antibodies
US20140302037A1 (en) 2013-03-15 2014-10-09 Amgen Inc. BISPECIFIC-Fc MOLECULES
ES2687282T3 (en) 2013-03-18 2018-10-24 Biocerox Products B.V. ANTI-CD134 (OX40) humanized antibodies and their uses
AU2014241162A1 (en) 2013-03-27 2015-10-22 Cedars-Sinai Medical Center Mitigation and reversal of fibrosis and inflammation by inhibition of TL1A function and related signaling pathways
EP3783017A1 (en) 2013-04-02 2021-02-24 Chugai Seiyaku Kabushiki Kaisha Fc region variant
UA118028C2 (en) 2013-04-03 2018-11-12 Рош Глікарт Аг Bispecific antibodies specific for fap and dr5, antibodies specific for dr5 and methods of use
KR20150140752A (en) 2013-04-12 2015-12-16 모르포시스 아게 Antibodies targeting m-csf
AU2014261630B2 (en) 2013-04-29 2019-05-09 F. Hoffmann-La Roche Ag Human FcRn-binding modified antibodies and methods of use
RU2019108429A (en) 2013-04-29 2019-05-06 Ф. Хоффманн-Ля Рош Аг MODIFIED ASYMMETRIC ANTIBODIES CONNECTING FC-RECEPTOR AND METHODS OF THEIR APPLICATION
CN105164158A (en) 2013-04-29 2015-12-16 豪夫迈·罗氏有限公司 Fcrn-binding abolished ANTI-IGF-1R antibodies and their use in the treatment of vascular eye diseases
CA2910945A1 (en) 2013-05-08 2014-11-13 Zymeworks Inc. Bispecific her2 and her3 antigen binding constructs
JP6612214B2 (en) 2013-05-20 2019-11-27 ジェネンテック, インコーポレイテッド Anti-transferrin receptor antibodies and methods of use
RU2655439C2 (en) 2013-05-31 2018-05-28 Займворкс Инк. Heteromultimers with reduced or silenced effector function
JP2016521715A (en) 2013-06-14 2016-07-25 バイエル ファーマ アクチエンゲゼルシャフト Anti-TWEAKR antibody and use thereof
BR112015032690B1 (en) 2013-06-25 2020-03-10 Vaccinex, Inc. USE OF SEMAPHORIN-4D INHIBITOR MOLECULES IN COMBINATION WITH AN IMMUNOMODULATIVE THERAPY TO INHIBIT TUMORAL GROWTH AND METASTASIS
AU2014290361B2 (en) 2013-07-18 2019-04-18 Taurus Biosciences, Llc Humanized antibodies with ultralong complementarity determining regions
EP3022224A2 (en) 2013-07-18 2016-05-25 Fabrus, Inc. Antibodies with ultralong complementarity determining regions
EP4105236A1 (en) 2013-07-19 2022-12-21 Cedars-Sinai Medical Center Anti-tl1a (tnfsf15) antibody for treatment of inflammatory bowel disease
CA2918624C (en) * 2013-07-31 2024-02-13 Amgen Inc. Growth differentiation factor 15 (gdf-15) constructs
CA2919583C (en) 2013-07-31 2018-09-11 Rinat Neuroscience Corp. Engineered polypeptide conjugates
LT3708583T (en) 2013-08-01 2022-04-11 Five Prime Therapeutics, Inc. Afucosylated anti-fgfr2iiib antibodies
MY190252A (en) 2013-08-02 2022-04-08 Pfizer Anti-cxcr4 antibodies and antibody-drug conjugates
US20150038682A1 (en) * 2013-08-02 2015-02-05 Jn Biosciences Llc Antibodies or fusion proteins multimerized via homomultimerizing peptide
EP3875106A1 (en) 2013-08-08 2021-09-08 Bioverativ Therapeutics Inc. Purification of chimeric fviii molecules
ES2704909T3 (en) 2013-08-09 2019-03-20 Toray Industries Pharmaceutical composition for the treatment and / or prevention of cancer
UA116479C2 (en) 2013-08-09 2018-03-26 Макродженікс, Інк. Bi-specific monovalent fc diabodies that are capable of binding cd32b and cd79b and uses thereof
US11384149B2 (en) 2013-08-09 2022-07-12 Macrogenics, Inc. Bi-specific monovalent Fc diabodies that are capable of binding CD32B and CD79b and uses thereof
US9845363B2 (en) 2013-08-13 2017-12-19 Sanofi Antibodies to plasminogen activator inhibitor-1 (PAI-1) and uses thereof
TWI592426B (en) 2013-08-13 2017-07-21 賽諾菲公司 Antibodies to plasminogen activator inhibitor-1 (pai-1) and uses thereof
US10548953B2 (en) 2013-08-14 2020-02-04 Bioverativ Therapeutics Inc. Factor VIII-XTEN fusions and uses thereof
EP2840091A1 (en) 2013-08-23 2015-02-25 MacroGenics, Inc. Bi-specific diabodies that are capable of binding gpA33 and CD3 and uses thereof
EP2839842A1 (en) 2013-08-23 2015-02-25 MacroGenics, Inc. Bi-specific monovalent diabodies that are capable of binding CD123 and CD3 and uses thereof
MX2016002574A (en) 2013-08-28 2016-06-14 Stemcentrx Inc Novel sez6 modulators and methods of use.
MX2016002547A (en) 2013-08-28 2016-06-17 Stemcentrx Inc Site-specific antibody conjugation methods and compositions.
EP3041863A4 (en) * 2013-09-05 2017-08-16 Amgen Inc. Fc-containing molecules exhibiting predictable, consistent, and reproducible glycoform profiles
HUE049281T2 (en) 2013-09-13 2020-09-28 Beigene Switzerland Gmbh Anti-pd1 antibodies and their use as therapeutics and diagnostics
MX2016003248A (en) 2013-09-17 2016-06-07 Genentech Inc Methods of using anti-lgr5 antibodies.
EP3048112B1 (en) * 2013-09-18 2020-03-11 Tosoh Corporation Fc-BINDING PROTEIN, METHOD FOR PRODUCING SAID PROTEIN, AND ANTIBODY ADSORBENT USING SAID PROTEIN, AND METHODS FOR PURIFYING AND IDENTIFYING ANTIBODY USING SAID ADSORBENT
EP3903599A1 (en) 2013-09-25 2021-11-03 Bioverativ Therapeutics Inc. On-column viral inactivation methods
SG11201602261VA (en) 2013-09-27 2016-04-28 Chugai Pharmaceutical Co Ltd Method for producing polypeptide heteromultimer
US10376583B2 (en) 2013-09-30 2019-08-13 Beth Israel Deaconess Medical Center, Inc. Human immunodeficiency virus therapies utilizing N332-glycan-dependent antibodies and a reservoir activator
WO2015050959A1 (en) 2013-10-01 2015-04-09 Yale University Anti-kit antibodies and methods of use thereof
BR112016006999B1 (en) 2013-10-02 2023-11-14 Medimmune, Llc ANTI-INFLUENZA NEUTRALIZING ANTIBODIES AND THEIR USES, THEIR PRODUCTION METHOD, COMPOSITION THAT COMPRISES THEM, NUCLEIC ACID AND VECTOR
NZ630881A (en) 2013-10-10 2016-03-31 Vaccinex Inc Use of semaphorin-4d binding molecules for treatment of atherosclerosis
MX2016003593A (en) 2013-10-11 2016-06-02 Hoffmann La Roche Multispecific domain exchanged common variable light chain antibodies.
US10081682B2 (en) 2013-10-11 2018-09-25 Oxford Bio Therapeutics Ltd. Conjugated antibodies against LY75 for the treatment of cancer
BR112016007635A2 (en) 2013-10-11 2017-09-12 Genentech Inc nsp4 inhibitors and methods of use
CA2925598A1 (en) 2013-10-18 2015-04-23 Genentech, Inc. Anti-rspo antibodies and methods of use
WO2015057939A1 (en) 2013-10-18 2015-04-23 Biogen Idec Ma Inc. Anti-s1p4 antibodies and uses thereof
NZ630892A (en) 2013-10-21 2016-03-31 Vaccinex Inc Use of semaphorin-4d binding molecules for treating neurodegenerative disorders
MX2016005159A (en) 2013-10-23 2016-07-05 Genentech Inc Methods of diagnosing and treating eosinophilic disorders.
ES2759252T3 (en) 2013-10-31 2020-05-08 Resolve Therapeutics Llc Nuclease-albumin fusions and therapeutic methods
KR102331054B1 (en) 2013-11-06 2021-11-24 얀센 바이오테크 인코포레이티드 Anti-ccl17 antibodies
SG11201603397QA (en) 2013-11-06 2016-05-30 Stemcentrx Inc Novel anti-claudin antibodies and methods of use
CN104623637A (en) 2013-11-07 2015-05-20 健能隆医药技术(上海)有限公司 Application of IL-22 dimer in preparation of intravenous injection drugs
EP3065769A4 (en) 2013-11-08 2017-05-31 Biogen MA Inc. Procoagulant fusion compound
TWI736515B (en) 2013-11-13 2021-08-21 美商輝瑞大藥廠 Tumor necrosis factor-like ligand 1a specific antibodies and compositions and uses thereof
WO2015073721A1 (en) 2013-11-13 2015-05-21 Zymeworks Inc. Monovalent antigen binding constructs targeting egfr and/or her2 and uses thereof
EP3071599A4 (en) * 2013-11-18 2017-07-12 University of Maryland, Baltimore Hyper-glycosylated antibodies with selective fc receptor binding
SI3071597T1 (en) 2013-11-21 2020-11-30 F. Hoffmann-La Roche Ag Anti-alpha-synuclein antibodies and methods of use
CA2931356A1 (en) 2013-11-27 2015-06-04 Zymeworks Inc. Bispecific antigen-binding constructs targeting her2
EP3077504B1 (en) 2013-12-06 2019-08-14 Dana-Farber Cancer Institute, Inc. Therapeutic peptides
CN114044825A (en) 2013-12-09 2022-02-15 爱乐科斯公司 anti-Siglec-8 antibodies and methods of use thereof
CA2931340A1 (en) 2013-12-13 2015-06-18 Genentech, Inc. Anti-cd33 antibodies and immunoconjugates
US20150190506A1 (en) 2013-12-17 2015-07-09 Genentech, Inc. Combination therapy comprising ox40 binding agonists and pd-1 axis binding antagonists
AU2014364601A1 (en) 2013-12-17 2016-07-07 Genentech, Inc. Methods of treating HER2-positive cancers using PD-1 axis binding antagonists and anti-HER2 antibodies
PE20210107A1 (en) 2013-12-17 2021-01-19 Genentech Inc ANTI-CD3 ANTIBODIES AND METHODS OF USE
US8980273B1 (en) 2014-07-15 2015-03-17 Kymab Limited Method of treating atopic dermatitis or asthma using antibody to IL4RA
US8986691B1 (en) 2014-07-15 2015-03-24 Kymab Limited Method of treating atopic dermatitis or asthma using antibody to IL4RA
US20150210772A1 (en) 2013-12-17 2015-07-30 Genentech, Inc. Methods of treating cancer using pd-1 axis binding antagonists and an anti-cd20 antibody
DE202014010499U1 (en) 2013-12-17 2015-10-20 Kymab Limited Targeting of human PCSK9 for cholesterol treatment
AU2014368696A1 (en) 2013-12-20 2016-06-02 F. Hoffmann-La Roche Ag Humanized anti-Tau(pS422) antibodies and methods of use
TWI728373B (en) 2013-12-23 2021-05-21 美商建南德克公司 Antibodies and methods of use
CN112142843A (en) 2013-12-24 2020-12-29 阿尔金克斯有限公司 FcRn antagonists and methods of use
CN112079923A (en) 2013-12-26 2020-12-15 田边三菱制药株式会社 Human anti-IL-33 neutralizing monoclonal antibodies
CN105873615B (en) 2014-01-03 2020-12-25 豪夫迈·罗氏有限公司 Covalently linked helicar-anti-helicar antibody conjugates and uses thereof
KR102278429B1 (en) 2014-01-03 2021-07-16 에프. 호프만-라 로슈 아게 Covalently linked polypeptide toxin-antibody conjugates
KR20160104628A (en) 2014-01-03 2016-09-05 에프. 호프만-라 로슈 아게 Bispecific anti-hapten/anti-blood brain barrier receptor antibodies, complexes thereof and their use as blood brain barrier shuttles
CA2932547C (en) 2014-01-06 2023-05-23 F. Hoffmann-La Roche Ag Monovalent blood brain barrier shuttle modules
SG11201605242YA (en) 2014-01-10 2016-07-28 Biogen Ma Inc Factor viii chimeric proteins and uses thereof
KR20160107190A (en) 2014-01-15 2016-09-13 에프. 호프만-라 로슈 아게 Fc-region variants with modified fcrn- and maintained protein a-binding properties
WO2015109212A1 (en) 2014-01-17 2015-07-23 Pfizer Inc. Anti-il-2 antibodies and compositions and uses thereof
KR20160111469A (en) 2014-01-24 2016-09-26 제넨테크, 인크. Methods of using anti-steap1 antibodies and immunoconjugates
DK3097122T3 (en) 2014-01-24 2020-08-10 Ngm Biopharmaceuticals Inc ANTIBODIES BINDING BETA-KLOTHO DOMAIN 2 AND METHODS OF USING IT
CA2937539A1 (en) 2014-02-04 2015-08-13 Genentech, Inc. Mutant smoothened and methods of using the same
HUE041530T2 (en) 2014-02-06 2019-05-28 Hoffmann La Roche Interleukin-2 fusion proteins and uses thereof
ES2873248T3 (en) 2014-02-08 2021-11-03 Hoffmann La Roche Methods to treat Alzheimer's disease
TWI705824B (en) 2014-02-08 2020-10-01 美商建南德克公司 Methods of treating alzheimer's disease
SG10201808259TA (en) 2014-02-12 2018-10-30 Genentech Inc Anti-jagged1 antibodies and methods of use
JP6533534B2 (en) 2014-02-14 2019-06-19 マクロジェニクス,インコーポレーテッド Composition for use in the treatment of glioblastoma and use thereof
EP3107574A2 (en) 2014-02-21 2016-12-28 F. Hoffmann-La Roche AG Anti-il-13/il-17 bispecific antibodies and uses thereof
US9603927B2 (en) * 2014-02-28 2017-03-28 Janssen Biotech, Inc. Combination therapies with anti-CD38 antibodies
DK3110446T3 (en) 2014-02-28 2022-02-28 Allakos Inc Methods and compositions for treating Siglec-8-associated diseases
US9732154B2 (en) 2014-02-28 2017-08-15 Janssen Biotech, Inc. Anti-CD38 antibodies for treatment of acute lymphoblastic leukemia
UY36021A (en) * 2014-03-05 2015-09-30 Ucb Biopharma Sprl FC MULTIMÈRIC PROTEINS
NZ631007A (en) 2014-03-07 2015-10-30 Alexion Pharma Inc Anti-c5 antibodies having improved pharmacokinetics
MX2016011748A (en) 2014-03-14 2016-12-12 Innate Pharma Humanized antibodies with increased stability.
CN113491766A (en) 2014-03-14 2021-10-12 达纳-法伯癌症研究所公司 Vaccine compositions and methods for restoring function of the NKG2D pathway against cancer
CN106103730B (en) 2014-03-14 2021-06-08 豪夫迈·罗氏有限公司 Methods and compositions for secretion of heterologous polypeptides
SI3129067T1 (en) 2014-03-19 2023-06-30 Genzyme Corporation Site-specific glycoengineering of targeting moieties
CN106164094B (en) 2014-03-21 2021-05-14 X博迪公司 Bispecific antigen binding polypeptides
IL247853B (en) 2014-03-21 2022-08-01 Teva Pharmaceuticals Int Gmbh Antagonist antibodies directed against calcitonin gene-related peptide and methods using same
US10556945B2 (en) 2014-03-21 2020-02-11 Teva Pharmaceuticals International Gmbh Antagonist antibodies directed against calcitonin gene-related peptide and methods using same
WO2015140591A1 (en) 2014-03-21 2015-09-24 Nordlandssykehuset Hf Anti-cd14 antibodies and uses thereof
JP2017516458A (en) 2014-03-24 2017-06-22 ジェネンテック, インコーポレイテッド Cancer treatment with c-met antagonist and correlation with HGF expression of c-met antagonist
KR20160145624A (en) 2014-03-31 2016-12-20 제넨테크, 인크. Anti-ox40 antibodies and methods of use
EP3126386A1 (en) 2014-03-31 2017-02-08 F. Hoffmann-La Roche AG Combination therapy comprising anti-angiogenesis agents and ox40 binding agonists
JP6614582B2 (en) 2014-04-04 2019-12-04 バイオノミクス インコーポレイテッド Humanized antibody that binds to LGR5
TWI726842B (en) 2014-04-07 2021-05-11 日商中外製藥股份有限公司 Immune activation antigen binding molecule
CN106232139A (en) 2014-04-11 2016-12-14 免疫医疗有限责任公司 Bispecific HER2 antibody
JP6649895B2 (en) 2014-04-18 2020-02-19 アクセルロン ファーマ, インコーポレイテッド Method for increasing red blood cell levels and treating sickle cell disease
WO2015164615A1 (en) 2014-04-24 2015-10-29 University Of Oslo Anti-gluten antibodies and uses thereof
EP3134127B1 (en) 2014-04-25 2020-02-26 Rinat Neuroscience Corp. Antibody-drug conjugates with high drug loading
ES2962694T3 (en) 2014-05-02 2024-03-20 Momenta Pharmaceuticals Inc Compositions and procedures related to manipulated Fc constructs
SG11201609014TA (en) 2014-05-08 2016-12-29 Chugai Pharmaceutical Co Ltd Gpc3 -targeting drug which is administered to patient responsive to gpc3-targeting drug therapy
EP3144388B1 (en) 2014-05-13 2020-07-01 Chugai Seiyaku Kabushiki Kaisha T cell-redirecting antigen-binding molecule for cells having immunosuppression function
WO2015175375A1 (en) 2014-05-13 2015-11-19 Short Jay M Conditionally active biological proteins
RU2016150370A (en) 2014-05-22 2018-06-26 Дженентек, Инк. Antibodies and immunoconjugates against GPC3
WO2015179835A2 (en) 2014-05-23 2015-11-26 Genentech, Inc. Mit biomarkers and methods using the same
US10155818B2 (en) 2014-05-28 2018-12-18 Agenus Inc. Anti-GITR antibodies and methods of use thereof
ES2905777T3 (en) 2014-05-30 2022-04-12 Shanghai Henlius Biotech Inc Anti-epidermal growth factor receptor (EGFR) antibodies
PE20170441A1 (en) 2014-06-06 2017-04-26 Bristol Myers Squibb Co ANTIBODIES AGAINST THE GLUCOCORTICOID-INDUCED TUMOR NECROSIS FACTOR RECEPTOR (GITR) AND ITS USES
EP3155015A1 (en) 2014-06-11 2017-04-19 F. Hoffmann-La Roche AG Anti-lgr5 antibodies and uses thereof
US20230190750A1 (en) 2014-06-13 2023-06-22 Genentech, Inc. Methods of treating and preventing cancer drug resistance
TN2016000553A1 (en) 2014-06-13 2018-04-04 Acceleron Pharma Inc Methods and compositions for treating ulcers
TWI713453B (en) 2014-06-23 2020-12-21 美商健生生物科技公司 Interferon alpha and omega antibody antagonists
EP3164419A1 (en) 2014-06-26 2017-05-10 F. Hoffmann-La Roche AG Anti-brdu antibodies and methods of use
AR100978A1 (en) 2014-06-26 2016-11-16 Hoffmann La Roche ANTI-Tau HUMANIZED ANTIBODY BRAIN LAUNCHERS (pS422) AND USES OF THE SAME
WO2015197598A2 (en) 2014-06-27 2015-12-30 Innate Pharma Multispecific antigen binding proteins
US10519234B2 (en) 2014-06-27 2019-12-31 Innate Pharma NKp46 binding proteins
US11008561B2 (en) 2014-06-30 2021-05-18 Bioverativ Therapeutics Inc. Optimized factor IX gene
WO2016000619A1 (en) 2014-07-03 2016-01-07 Beigene, Ltd. Anti-pd-l1 antibodies and their use as therapeutics and diagnostics
EP3166974A1 (en) 2014-07-11 2017-05-17 Genentech, Inc. Anti-pd-l1 antibodies and diagnostic uses thereof
RU2017103289A (en) 2014-07-11 2018-08-14 Дженентек, Инк. INHIBITING THE NOTCH WAY
RU2711141C2 (en) 2014-07-22 2020-01-15 СиБи ТЕРЕПЬЮТИКС, ИНК. Anti-pd-1 antibodies
WO2016014984A1 (en) 2014-07-24 2016-01-28 Xencor, Inc. Rapid clearance of antigen complexes using novel antibodies
EP3174894B1 (en) 2014-07-30 2021-06-23 NGM Biopharmaceuticals, Inc. Compositions and methods of use for treating metabolic disorders
NZ766556A (en) 2014-08-04 2024-02-23 Hoffmann La Roche Bispecific t cell activating antigen binding molecules
NZ728749A (en) 2014-08-05 2023-06-30 Cb Therapeutics Inc Anti-pd-l1 antibodies
WO2016020791A1 (en) 2014-08-05 2016-02-11 Novartis Ag Ckit antibody drug conjugates
US10238748B2 (en) 2014-08-12 2019-03-26 Novartis Ag Anti-CDH6 antibody drug conjugates
WO2016023916A1 (en) 2014-08-12 2016-02-18 Kymab Limited Treatment of disease using ligand binding to targets of interest
WO2016025645A1 (en) 2014-08-12 2016-02-18 Massachusetts Institute Of Technology Synergistic tumor treatment with il-2, a therapeutic antibody, and an immune checkpoint blocker
WO2016030488A1 (en) 2014-08-27 2016-03-03 Innate Pharma Treatment of celiac disease
RU2764074C2 (en) 2014-08-28 2022-01-13 Байоатла, Ллк Conditionally active chimeric antigen receptors for modified t-cells
US10967068B2 (en) 2014-08-28 2021-04-06 Pfizer Inc. Stability-modulating linkers for use with antibody drug conjugates
EA202193002A2 (en) 2014-09-03 2022-03-31 Бёрингер Ингельхайм Интернациональ Гмбх COMPOUND TARGETED TO IL-23A AND TNF-ALFA AND ITS APPLICATION
JP6805130B2 (en) 2014-09-05 2020-12-23 ヤンセン ファーマシューティカ エヌ.ベー. CD123 binder and its use
TW201617368A (en) 2014-09-05 2016-05-16 史坦森特瑞斯公司 Novel anti-MFI2 antibodies and methods of use
CN108064182B (en) 2014-09-09 2021-09-03 詹森生物科技公司 Combination therapy with anti-CD 38 antibodies
JP2017528468A (en) 2014-09-10 2017-09-28 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft Galactose engineered immunoglobulin 1 antibody
AU2015314826A1 (en) 2014-09-12 2017-03-02 Genentech, Inc. Cysteine engineered antibodies and conjugates
CN113698488A (en) 2014-09-12 2021-11-26 基因泰克公司 anti-B7-H4 antibodies and immunoconjugates
AU2015314744A1 (en) 2014-09-12 2017-03-02 Genentech, Inc. Anti-CLL-1 antibodies and immunoconjugates
MA40576B1 (en) 2014-09-12 2020-11-30 Genentech Inc Anti-her2 antibodies and immunoconjugates
EP3197500A1 (en) 2014-09-17 2017-08-02 Genentech, Inc. Immunoconjugates comprising anti-her2 antibodies and pyrrolobenzodiazepines
CA2959428A1 (en) 2014-09-19 2016-03-24 Regeneron Pharmaceuticals, Inc. Chimeric antigen receptors
NZ730186A (en) 2014-09-22 2020-04-24 Intrinsic Lifesciences Llc Humanized anti-hepcidin antibodies and uses thereof
PL3262071T3 (en) 2014-09-23 2020-08-10 F. Hoffmann-La Roche Ag Method of using anti-cd79b immunoconjugates
SG11201701803XA (en) 2014-09-26 2017-04-27 Bayer Pharma AG Stabilized adrenomedullin derivatives and use thereof
MA40764A (en) 2014-09-26 2017-08-01 Chugai Pharmaceutical Co Ltd THERAPEUTIC AGENT INDUCING CYTOTOXICITY
BR112017006515A8 (en) 2014-09-29 2018-02-27 Univ Duke bispecific molecules, compositions, method for treating or preventing HIV-1 infection in an individual with this need, and vector
AU2015330869B2 (en) 2014-10-09 2021-07-08 Genzyme Corporation Glycoengineered antibody drug conjugates
WO2016131950A1 (en) 2015-02-20 2016-08-25 Innate Pharma Cd73 blockade
EP3736294A3 (en) 2014-10-10 2021-02-17 Innate Pharma Cd73 blockade
ES2753391T3 (en) 2014-10-14 2020-04-08 Halozyme Inc Adenosine deaminase 2 (ADA2) compositions, variants thereof and methods of use thereof
WO2016059602A2 (en) 2014-10-16 2016-04-21 Glaxo Group Limited Methods of treating cancer and related compositions
US9732148B2 (en) 2014-10-16 2017-08-15 Genentech, Inc. Anti-α-synuclein antibodies and methods of use
US10137173B2 (en) 2014-10-20 2018-11-27 Aravive Biologics, Inc. Antiviral activity of Gas6 inhibitor
EP3209697A4 (en) 2014-10-23 2018-05-30 La Trobe University Fn14-binding proteins and uses thereof
NZ731491A (en) 2014-10-23 2021-12-24 Kira Biotech Pty Ltd Cd83 binding proteins and uses thereof
JP6889660B2 (en) 2014-10-23 2021-06-18 イナート・ファルマ・ソシエテ・アノニムInnate Pharma Pharma S.A. Treatment of cancer with anti-NKG2A agents
NZ730054A (en) 2014-10-31 2023-11-24 Ngm Biopharmaceuticals Inc Compositions and methods of use for treating metabolic disorders
WO2016070001A1 (en) 2014-10-31 2016-05-06 Jounce Therapeutics, Inc. Methods of treating conditions with antibodies that bind b7-h4
US20160160290A1 (en) 2014-11-03 2016-06-09 Genentech, Inc. Methods and biomarkers for predicting efficacy and evaluation of an ox40 agonist treatment
CA2966523A1 (en) 2014-11-03 2016-05-12 Genentech, Inc. Assays for detecting t cell immune subsets and methods of use thereof
AU2015342961B2 (en) 2014-11-05 2021-08-12 Genentech, Inc. Methods of producing two chain proteins in bacteria
BR112017008666A2 (en) 2014-11-05 2018-01-30 Genentech, Inc. anti-fgfr2 / 3 antibodies and methods of use
RU2739500C2 (en) 2014-11-05 2020-12-25 Дженентек, Инк. Methods for producing double-stranded proteins in bacteria
SG11201703376QA (en) 2014-11-06 2017-05-30 Genentech Inc Combination therapy comprising ox40 binding agonists and tigit inhibitors
CN107074966A (en) 2014-11-06 2017-08-18 豪夫迈·罗氏有限公司 The Fc region variants of FCRN and albumin A binding property with change
WO2016073157A1 (en) 2014-11-06 2016-05-12 Genentech, Inc. Anti-ang2 antibodies and methods of use thereof
EP3215528B1 (en) 2014-11-06 2019-08-07 F.Hoffmann-La Roche Ag Fc-region variants with modified fcrn-binding and methods of use
WO2016071701A1 (en) 2014-11-07 2016-05-12 Kymab Limited Treatment of disease using ligand binding to targets of interest
GB2537445A (en) 2014-11-10 2016-10-19 Medimmune Ltd Binding molecules specific for CD73 and uses thereof
CN107172879B (en) 2014-11-10 2021-11-05 豪夫迈·罗氏有限公司 Anti-interleukin-33 antibodies and uses thereof
CN107105632A (en) 2014-11-10 2017-08-29 豪夫迈·罗氏有限公司 Nephrosis animal model and its therapeutic agent
JP6847037B2 (en) 2014-11-11 2021-03-24 メディミューン リミテッド Concomitant therapeutic agents containing anti-CD73 antibody and A2A receptor inhibitor and their use
US10155820B2 (en) 2014-11-12 2018-12-18 Tracon Pharmaceuticals, Inc. Anti-endoglin antibodies and uses thereof
US9926375B2 (en) 2014-11-12 2018-03-27 Tracon Pharmaceuticals, Inc. Anti-endoglin antibodies and uses thereof
MY191428A (en) 2014-11-14 2022-06-27 Hoffmann La Roche Antigen binding molecules comprising a tnf family ligand trimer
CN106999583A (en) 2014-11-17 2017-08-01 豪夫迈·罗氏有限公司 Combination treatment comprising OX40 combinations activator and the axle binding antagonists of PD 1
EP3845565A3 (en) 2014-11-19 2021-09-08 Genentech, Inc. Antibodies against bace1 and use thereof for neural disease immunotherapy
JP6779876B2 (en) 2014-11-19 2020-11-04 ジェネンテック, インコーポレイテッド Anti-transferrin receptor antibody and how to use it
EP3221361B1 (en) 2014-11-19 2021-04-21 Genentech, Inc. Anti-transferrin receptor / anti-bace1 multispecific antibodies and methods of use
SI3789402T1 (en) 2014-11-20 2022-10-28 F. Hoffmann-La Roche Ag Combination therapy of t cell activating bispecific antigen binding molecules and pd-1 axis binding antagonists
CA2968162A1 (en) 2014-11-20 2016-05-26 F. Hoffmann-La Roche Ag Common light chains and methods of use
MX2017006323A (en) 2014-11-21 2017-08-21 Bristol Myers Squibb Co Antibodies comprising modified heavy constant regions.
CN113773388A (en) 2014-11-21 2021-12-10 百时美施贵宝公司 anti-CD 73 antibodies and uses thereof
WO2016087416A1 (en) 2014-12-03 2016-06-09 F. Hoffmann-La Roche Ag Multispecific antibodies
MA41119A (en) 2014-12-03 2017-10-10 Acceleron Pharma Inc METHODS OF TREATMENT OF MYELODYSPLASIC SYNDROMES AND SIDEROBLASTIC ANEMIA
AU2015358615B2 (en) 2014-12-04 2021-08-05 Janssen Biotech, Inc. Anti-CD38 antibodies for treatment of acute myeloid leukemia
LT3227336T (en) 2014-12-05 2019-09-25 F. Hoffmann-La Roche Ag Anti-cd79b antibodies and methods of use
KR20170085595A (en) 2014-12-10 2017-07-24 제넨테크, 인크. Blood brain barrier receptor antibodies and methods of use
ES2834618T3 (en) * 2014-12-18 2021-06-18 Aravive Biologics Inc GAS6 inhibitor antifibrotic activity
AU2015365167B2 (en) 2014-12-19 2021-07-29 Chugai Seiyaku Kabushiki Kaisha Anti-C5 antibodies and methods of use
CN113563468A (en) 2014-12-19 2021-10-29 雷根尼桑斯公司 Antibodies that bind to human C6 and uses thereof
EP3233907B1 (en) 2014-12-19 2021-03-03 Genmab A/S Rodent bispecific heterodimeric proteins
SG11201700841QA (en) 2014-12-19 2017-03-30 Chugai Pharmaceutical Co Ltd Anti-myostatin antibodies, polypeptides containing variant fc regions, and methods of use
JP7211703B2 (en) 2014-12-22 2023-01-24 ザ ロックフェラー ユニバーシティー Anti-MERTK agonist antibody and use thereof
KR102644115B1 (en) 2014-12-23 2024-03-05 브리스톨-마이어스 스큅 컴퍼니 Antibodies to tigit
WO2016111947A2 (en) 2015-01-05 2016-07-14 Jounce Therapeutics, Inc. Antibodies that inhibit tim-3:lilrb2 interactions and uses thereof
SG10201906471PA (en) 2015-01-14 2019-09-27 Brigham & Womens Hospital Inc Treatment of cancer with anti-lap monoclonal antibodies
JP2018511557A (en) 2015-01-22 2018-04-26 中外製薬株式会社 Combination and use of two or more anti-C5 antibodies
JP2018512597A (en) 2015-02-04 2018-05-17 ジェネンテック, インコーポレイテッド Mutant smoothened and method of using the same
CN114773469A (en) 2015-02-05 2022-07-22 中外制药株式会社 Antibodies comprising an ion concentration-dependent antigen-binding domain, FC region variants, IL-8-binding antibodies and uses thereof
CA2976236A1 (en) * 2015-02-09 2016-08-18 Research Development Foundation Engineered immunoglobulin fc polypeptides displaying improved complement activation
US10227408B2 (en) 2015-02-19 2019-03-12 Compugen Ltd. Anti-PVRIG antibodies and methods of use
EP3259597B1 (en) 2015-02-19 2022-04-06 Compugen Ltd. Pvrig polypeptides and methods of treatment
JP6130983B2 (en) 2015-02-27 2017-05-17 中外製薬株式会社 Composition for treating IL-6 related diseases
MY188761A (en) * 2015-03-09 2021-12-29 Argenx Bvba Method of reducing serum levels of fc-containing agents using fcrn antagonists
CA2977675A1 (en) 2015-03-12 2016-09-15 Medimmune, Llc Method of purifying albumin-fusion proteins
AU2016233398A1 (en) 2015-03-16 2017-09-07 F. Hoffmann-La Roche Ag Methods of detecting and quantifying IL-13 and uses in diagnosing and treating Th2-associated diseases
WO2016146833A1 (en) 2015-03-19 2016-09-22 F. Hoffmann-La Roche Ag Biomarkers for nad(+)-diphthamide adp ribosyltransferase resistance
PL3273992T3 (en) 2015-03-23 2020-11-16 Jounce Therapeutics, Inc. Antibodies to icos
DK3274370T3 (en) 2015-03-23 2020-02-17 Bayer Pharma AG ANTI-CEACAM6 ANTIBODIES AND APPLICATIONS THEREOF
RS61438B1 (en) 2015-03-31 2021-03-31 Medimmune Ltd A novel il33 form, mutated forms of il33, antibodies, assays and methods of using the same
JP6996979B2 (en) 2015-03-31 2022-02-04 エフ・ホフマン-ラ・ロシュ・アクチェンゲゼルシャフト Antigen-binding molecule containing trimer TNF family ligand
WO2016159213A1 (en) 2015-04-01 2016-10-06 中外製薬株式会社 Method for producing polypeptide hetero-oligomer
CN114456272A (en) 2015-04-03 2022-05-10 优瑞科生物技术公司 Constructs targeting AFP peptide/MHC complexes and uses thereof
AR104368A1 (en) 2015-04-03 2017-07-19 Lilly Co Eli ANTI-CD20- / ANTI-BAFF BIESPECTIFIC ANTIBODIES
US10167334B2 (en) 2015-04-03 2019-01-01 Xoma Technology Ltd. Treatment of cancer using anti-TGF-BETA and PD-1 antibodies
CN114736307A (en) 2015-04-06 2022-07-12 阿塞勒隆制药公司 TGF-beta superfamily type I and type II receptor heteromultimers and uses thereof
MA41919A (en) 2015-04-06 2018-02-13 Acceleron Pharma Inc ALK4 HETEROMULTIMERS: ACTRIIB AND THEIR USES
CN107709364A (en) 2015-04-07 2018-02-16 豪夫迈·罗氏有限公司 Antigen binding complex and application method with agonist activity
UA126270C2 (en) 2015-04-10 2022-09-14 Емджен Інк. Interleukin-2 muteins for the expansion of t-regulatory cells
DK3283106T3 (en) 2015-04-13 2022-01-10 Pfizer Therapeutic antibodies and uses thereof
LT3283508T (en) 2015-04-17 2021-07-12 Alpine Immune Sciences, Inc. Immunomodulatory proteins with tunable affinities
JP7044553B2 (en) 2015-04-24 2022-03-30 ジェネンテック, インコーポレイテッド How to identify bacteria containing bound polypeptides
MX2017013894A (en) 2015-04-28 2018-03-01 Mitsubishi Tanabe Pharma Corp RGMa BINDING PROTEIN AND USE THEREOF.
JP2018520642A (en) 2015-05-01 2018-08-02 ジェネンテック, インコーポレイテッド Mask anti-CD3 antibody and method of use thereof
WO2016179194A1 (en) 2015-05-04 2016-11-10 Jounce Therapeutics, Inc. Lilra3 and method of using the same
FR3035879A1 (en) 2015-05-07 2016-11-11 Lab Francais Du Fractionnement MUTANTS FC WITH MODIFIED FUNCTIONAL ACTIVITY
CN116196414A (en) 2015-05-11 2023-06-02 豪夫迈·罗氏有限公司 Compositions and methods for treating lupus nephritis
IL295002A (en) 2015-05-12 2022-09-01 Genentech Inc Therapeutic and diagnostic methods for cancer comprising a pd-l1 binding antagonist
CA2986594A1 (en) 2015-05-20 2016-11-24 Tufts Medical Center, Inc. Anti-cd38 antibodies for treatment of light chain amyloidosis and other cd38-positive hematological malignancies
WO2016196343A1 (en) 2015-05-29 2016-12-08 Genentech, Inc. Humanized anti-ebola virus glycoprotein antibodies and methods of use
HUE061253T2 (en) 2015-05-29 2023-06-28 Bristol Myers Squibb Co Antibodies against ox40 and uses thereof
KR20180012753A (en) 2015-05-29 2018-02-06 제넨테크, 인크. Treatment and Diagnosis Methods for Cancer
EP3303619B1 (en) 2015-05-29 2020-06-10 H. Hoffnabb-La Roche Ag Pd-l1 promoter methylation in cancer
JP2018516933A (en) 2015-06-02 2018-06-28 ジェネンテック, インコーポレイテッド Compositions and methods for treating neurological disorders using anti-IL-34 antibodies
SG10201911349YA (en) 2015-06-05 2020-01-30 Genentech Inc Anti-tau antibodies and methods of use
AU2016274585A1 (en) 2015-06-08 2017-12-14 Genentech, Inc. Methods of treating cancer using anti-OX40 antibodies
KR20180025888A (en) 2015-06-08 2018-03-09 제넨테크, 인크. Methods for treating cancer using anti-OX40 antibodies and PD-1 axis-binding antagonists
US10017577B2 (en) 2015-06-15 2018-07-10 Genentech, Inc. Antibodies and immunoconjugates
WO2016204966A1 (en) 2015-06-16 2016-12-22 Genentech, Inc. Anti-cd3 antibodies and methods of use
CN107847568B (en) 2015-06-16 2022-12-20 豪夫迈·罗氏有限公司 anti-CLL-1 antibodies and methods of use
PE20180330A1 (en) 2015-06-16 2018-02-13 Genentech Inc AFFINITY MATURED AND HUMANIZED ANTIBODIES FOR FCRH5 AND METHODS FOR THEIR USE
JP2018524312A (en) 2015-06-17 2018-08-30 ジェネンテック, インコーポレイテッド Anti-HER2 antibody and method of use
WO2016205567A1 (en) 2015-06-17 2016-12-22 Allakos Inc. Methods and compositions for treating fibrotic diseases
KR20180018538A (en) 2015-06-17 2018-02-21 제넨테크, 인크. Methods for the treatment of locally advanced or metastatic breast cancer using PD-1 axis-binding antagonists and taxanes
EA036789B1 (en) 2015-06-22 2020-12-22 Янссен Байотек, Инк. Combination therapies for heme malignancies with anti-cd38 antibodies and survivin inhibitors
CA2990518A1 (en) 2015-06-23 2016-12-29 Innate Pharma Multispecific nk engager proteins
AU2016284866B2 (en) 2015-06-23 2022-09-29 Innate Pharma Multispecific antigen binding proteins
CN113999312A (en) 2015-06-24 2022-02-01 豪夫迈·罗氏有限公司 Anti-transferrin receptor antibodies with tailored affinity
EP3313877B1 (en) 2015-06-24 2020-06-03 H. Hoffnabb-La Roche Ag Humanized anti-tau(ps422) antibodies and methods of use
US20170044265A1 (en) 2015-06-24 2017-02-16 Janssen Biotech, Inc. Immune Modulation and Treatment of Solid Tumors with Antibodies that Specifically Bind CD38
MA42270A (en) 2015-06-24 2018-05-02 Janssen Biotech Inc IMMUNE MODULATION AND SOLID TUMOR TREATMENT WITH ANTIBODIES BINDING SPECIFICALLY TO CD38
EP3514174B1 (en) 2015-06-29 2021-03-31 Ventana Medical Systems, Inc. Materials and methods for performing histochemical assays for human pro-epiregulin and amphiregulin
CN108473573A (en) 2015-06-29 2018-08-31 豪夫迈·罗氏有限公司 II type anti-CD 20 antibodies are used in organ transplant
WO2017002934A1 (en) 2015-07-01 2017-01-05 中外製薬株式会社 Gpc3-targeted therapeutic agent administered to patient in whom gpc3-targetd therapeutic ag
WO2017009712A1 (en) 2015-07-13 2017-01-19 Compugen Ltd. Hide1 compositions and methods
WO2017015433A2 (en) 2015-07-23 2017-01-26 Boehringer Ingelheim International Gmbh Compound targeting il-23a and b-cell activating factor (baff) and uses thereof
US11066481B2 (en) 2015-07-23 2021-07-20 The Regents Of The University Of California Antibodies to coagulation factor XIa and uses thereof
EP3328427A4 (en) 2015-07-27 2018-12-12 The General Hospital Corporation Antibody derivatives with conditionally enabled effector function
EP3328399B1 (en) 2015-07-31 2023-12-27 Regents of the University of Minnesota Modified cells and methods of therapy
EA201890423A1 (en) 2015-08-03 2018-07-31 Биовератив Терапьютикс Инк. SLIGHT PROTEINS OF THE FACTOR IX, METHODS OF THEIR RECEPTION AND APPLICATION
EP4218792A1 (en) 2015-08-04 2023-08-02 Acceleron Pharma Inc. Composition for treating myeloproliferative disorders
CN105384825B (en) 2015-08-11 2018-06-01 南京传奇生物科技有限公司 A kind of bispecific chimeric antigen receptor and its application based on single domain antibody
ES2814550T3 (en) 2015-08-17 2021-03-29 Janssen Pharmaceutica Nv Anti-BCMA antibodies, bispecific antigen-binding molecules that bind BCMA and CD3, and uses thereof
CN108026180B (en) 2015-08-28 2022-06-07 豪夫迈·罗氏有限公司 Anti-hypusine antibodies and uses thereof
US10526408B2 (en) * 2015-08-28 2020-01-07 Research Development Foundation Engineered antibody FC variants
PE20181322A1 (en) 2015-09-01 2018-08-14 Agenus Inc ANTI-PD1 ANTIBODY AND ITS METHODS OF USE
US20190022092A1 (en) 2015-09-15 2019-01-24 Acerta Pharma B.V. Therapeutic Combinations of a BTK Inhibitor and a GITR Binding Molecule, a 4-1BB Agonist, or an OX40 Agonist
KR20180054701A (en) 2015-09-16 2018-05-24 아블렉시스, 엘엘씨 Anti-CD115 antibody
US9862760B2 (en) 2015-09-16 2018-01-09 Novartis Ag Polyomavirus neutralizing antibodies
CA2993423C (en) 2015-09-18 2024-03-12 Chugai Seiyaku Kabushiki Kaisha Il-8-binding antibodies and uses thereof
CA2999369C (en) 2015-09-22 2023-11-07 Spring Bioscience Corporation Anti-ox40 antibodies and diagnostic uses thereof
AU2016326666B2 (en) 2015-09-23 2023-06-15 Genentech, Inc. Optimized variants of anti-VEGF antibodies
CN108289954B (en) 2015-09-24 2022-05-31 阿布维特罗有限责任公司 HIV antibody compositions and methods of use
WO2017058859A1 (en) 2015-09-29 2017-04-06 Celgene Corporation Pd-1 binding proteins and methods of use thereof
AU2016329057A1 (en) 2015-09-30 2018-04-12 Janssen Biotech, Inc. Antagonistic antibodies specifically binding human CD40 and methods of use
AR106188A1 (en) 2015-10-01 2017-12-20 Hoffmann La Roche ANTI-CD19 HUMANIZED HUMAN ANTIBODIES AND METHODS OF USE
WO2017055393A1 (en) 2015-10-02 2017-04-06 F. Hoffmann-La Roche Ag Anti-cd3xtim-3 bispecific t cell activating antigen binding molecules
BR112018002570A2 (en) 2015-10-02 2018-10-16 Hoffmann La Roche bispecific antigen binding molecule, bispecific antibody, polynucleotides, ox40-specific binding antibody, pharmaceutical composition and method for inhibiting tumor cell growth in an individual
DK3356404T3 (en) 2015-10-02 2021-10-25 Hoffmann La Roche Anti-pd1 antibodies and methods of use
WO2017055385A1 (en) 2015-10-02 2017-04-06 F. Hoffmann-La Roche Ag Anti-cd3xgd2 bispecific t cell activating antigen binding molecules
MA43345A (en) 2015-10-02 2018-08-08 Hoffmann La Roche PYRROLOBENZODIAZEPINE ANTIBODY-DRUG CONJUGATES AND METHODS OF USE
AR106189A1 (en) 2015-10-02 2017-12-20 Hoffmann La Roche BIESPECTIFIC ANTIBODIES AGAINST HUMAN A-b AND THE HUMAN TRANSFERRINE RECEIVER AND METHODS OF USE
EP3150636A1 (en) 2015-10-02 2017-04-05 F. Hoffmann-La Roche AG Tetravalent multispecific antibodies
EP3356407B1 (en) 2015-10-02 2021-11-03 F. Hoffmann-La Roche AG Bispecific anti-cd19xcd3 t cell activating antigen binding molecules
KR102146319B1 (en) 2015-10-02 2020-08-25 에프. 호프만-라 로슈 아게 Bispecific antibodies specific for PD1 and TIM3
AU2016333512B2 (en) 2015-10-02 2022-11-17 F. Hoffmann-La Roche Ag Bispecific anti-human CD20/human transferrin receptor antibodies and methods of use
CR20180162A (en) 2015-10-02 2018-05-25 Hoffmann La Roche BISPECIFIC MOLECULES OF ANTIGEN BINDING TO T-CELL ACTIVATORS
AR106199A1 (en) 2015-10-02 2017-12-20 Hoffmann La Roche T-CELL ACTIVATING ANTIGEN BINDING MOLECULES OF ANTIGEN
WO2017055392A1 (en) 2015-10-02 2017-04-06 F. Hoffmann-La Roche Ag Anti-cd3xcd44v6 bispecific t cell activating antigen binding molecules
CN108026179A (en) 2015-10-02 2018-05-11 豪夫迈·罗氏有限公司 With reference to mesothelin and the bispecific T cell activation antigen binding molecules of CD3
US20180282410A1 (en) 2015-10-02 2018-10-04 Hoffmann-La Roche Inc. Anti-cd3xrob04 bispecific t cell activating antigen binding molecules
JP2018529747A (en) 2015-10-06 2018-10-11 ジェネンテック, インコーポレイテッド Methods for treating multiple sclerosis
WO2017060144A1 (en) 2015-10-07 2017-04-13 F. Hoffmann-La Roche Ag Bispecific antibodies with tetravalency for a costimulatory tnf receptor
US11130817B2 (en) 2015-10-12 2021-09-28 Innate Pharma CD73 blocking agents
MA43354A (en) 2015-10-16 2018-08-22 Genentech Inc CONJUGATE DRUG CONJUGATES WITH CLOUDY DISULPHIDE
MA45326A (en) 2015-10-20 2018-08-29 Genentech Inc CALICHEAMICIN-ANTIBODY-DRUG CONJUGATES AND METHODS OF USE
US10604577B2 (en) 2015-10-22 2020-03-31 Allakos Inc. Methods and compositions for treating systemic mastocytosis
TW201723190A (en) 2015-10-22 2017-07-01 永斯醫療股份有限公司 Gene signatures for determining ICOS expression
WO2017070561A1 (en) 2015-10-23 2017-04-27 Pfizer Inc. Anti-il-2 antibodies and compositions and uses thereof
WO2017072210A1 (en) 2015-10-29 2017-05-04 F. Hoffmann-La Roche Ag Anti-variant fc-region antibodies and methods of use
EP3184547A1 (en) 2015-10-29 2017-06-28 F. Hoffmann-La Roche AG Anti-tpbg antibodies and methods of use
KR102440160B1 (en) 2015-10-30 2022-09-02 제넨테크, 인크. Anti-HtrA1 antibodies and methods of use thereof
TW201730211A (en) 2015-10-30 2017-09-01 建南德克公司 Anti-Factor D antibodies and conjugates
KR20180072820A (en) 2015-11-02 2018-06-29 얀센 파마슈티카 엔.브이. A bispecific antigen binding molecule that binds to an anti-IL1RAP antibody, IL1RAP and CD3,
US10781261B2 (en) 2015-11-03 2020-09-22 Janssen Biotech, Inc. Subcutaneous formulations of anti-CD38 antibodies and their uses
TWI724048B (en) 2015-11-03 2021-04-11 美商健生生物科技公司 Subcutaneous formulations of anti-cd38 antibodies and their uses
RS63125B1 (en) 2015-11-03 2022-05-31 Janssen Biotech Inc Antibodies specifically binding pd-1 and their uses
WO2017079768A1 (en) 2015-11-08 2017-05-11 Genentech, Inc. Methods of screening for multispecific antibodies
JP6931329B2 (en) 2015-11-18 2021-09-01 中外製薬株式会社 Combination therapy using T cell redirection antigen-binding molecule for cells with immunosuppressive function
JP6925278B2 (en) 2015-11-18 2021-08-25 中外製薬株式会社 Method of enhancing humoral immune response
KR20180082563A (en) 2015-11-19 2018-07-18 브리스톨-마이어스 스큅 컴퍼니 Antibodies to glucocorticoid-induced tumor necrosis factor receptor (GITR) and uses thereof
RU2021107536A (en) 2015-11-23 2021-07-02 Файв Прайм Терапьютикс, Инк. FGFR2 INHIBITORS SEPARATELY OR IN COMBINATION WITH IMMUNOSTIMULATING AGENTS IN THE TREATMENT OF CANCER
AU2016359695A1 (en) 2015-11-23 2018-06-14 Acceleron Pharma Inc. Methods for treating eye disorders
MX2018006477A (en) 2015-12-02 2018-09-03 Agenus Inc Antibodies and methods of use thereof.
EP3178848A1 (en) 2015-12-09 2017-06-14 F. Hoffmann-La Roche AG Type ii anti-cd20 antibody for reducing formation of anti-drug antibodies
AU2016368469B2 (en) 2015-12-09 2023-11-02 F. Hoffmann-La Roche Ag Type II anti-CD20 antibody for reducing formation of anti-drug antibodies
KR20180087430A (en) 2015-12-17 2018-08-01 얀센 바이오테크 인코포레이티드 Antibodies specifically binding to HLA-DR and uses thereof
KR102467124B1 (en) 2015-12-18 2022-11-15 추가이 세이야쿠 가부시키가이샤 Anti-c5 antibodies and methods of use
US11359009B2 (en) 2015-12-25 2022-06-14 Chugai Seiyaku Kabushiki Kaisha Anti-myostatin antibodies and methods of use
CA3004288A1 (en) 2015-12-28 2017-07-06 Nobuyuki Tanaka Method for promoting efficiency of purification of fc region-containing polypeptide
EP3862365A1 (en) 2016-01-08 2021-08-11 F. Hoffmann-La Roche AG Methods of treating cea-positive cancers using pd-1 axis binding antagonists and anti-cea/anti-cd3 bispecific antibodies
CN108602883A (en) 2016-01-20 2018-09-28 基因泰克公司 High-dose therapy for Alzheimer's disease
CN109219618B (en) 2016-01-21 2022-08-09 辉瑞大药厂 Monospecific and bispecific antibodies against epidermal growth factor receptor variants III and CD3 and uses thereof
BR112018015659A2 (en) 2016-02-01 2018-12-26 Bioverativ Therapeutics Inc optimized factor viii genes
CN109071625A (en) 2016-02-04 2018-12-21 柯瑞斯公司 Smooth mutant and its application method
AU2017225854B2 (en) 2016-02-29 2020-11-19 Foundation Medicine, Inc. Therapeutic and diagnostic methods for cancer
CN108699156A (en) 2016-03-01 2018-10-23 豪夫迈·罗氏有限公司 The outstanding trastuzumab in shore difficult to understand and Rituximab variant of ADCP with reduction
AU2017228470A1 (en) 2016-03-04 2018-08-30 Bristol-Myers Squibb Company Combination therapy with anti-CD73 antibodies
JP7270379B2 (en) 2016-03-08 2023-05-10 イナート・ファルマ・ソシエテ・アノニム Siglec neutralizing antibody
AU2017229575A1 (en) 2016-03-08 2018-09-27 Janssen Biotech, Inc. GITR antibodies, methods, and uses
SG11201807523PA (en) 2016-03-10 2018-09-27 Viela Bio Inc Ilt7 binding molecules and methods of using the same
WO2017157948A1 (en) 2016-03-14 2017-09-21 Innate Pharma Anti-cd39 antibodies
RU2746754C2 (en) 2016-03-14 2021-04-20 Чугаи Сейяку Кабусики Кайся Cell damage inducing therapeutic medicinal product for anticancer therapy
KR20180127320A (en) * 2016-03-14 2018-11-28 유니버시티에트 이 오슬로 A modified immunoglobulin with altered FcRn binding
JP6430025B2 (en) 2016-03-15 2018-11-28 中外製薬株式会社 Methods of treating cancer using PD-1 binding antagonists and anti-GPC3 antibodies
AU2017235274A1 (en) 2016-03-15 2018-08-16 Innate Pharma Anti-mica antibodies
JP2019518713A (en) 2016-03-16 2019-07-04 メリマック ファーマシューティカルズ インコーポレーティッド Modified TRAIL for Cancer Therapy
CN109462996A (en) 2016-03-17 2019-03-12 西达-赛奈医疗中心 The method for diagnosing inflammatory bowel disease by RNASET2
CN108697799A (en) 2016-03-22 2018-10-23 生态学有限公司 The application of anti-LGR5 monoclonal antibodies
CR20180453A (en) 2016-03-22 2018-12-05 Hoffmann La Roche Bispecific MOLECULES OF T-CELLS ACTIVATED BY PROTEASES
JP7015244B2 (en) 2016-03-22 2022-02-02 エフ・ホフマン-ラ・ロシュ・アクチェンゲゼルシャフト Protease-activated T cell bispecific molecule
EP4273551A3 (en) 2016-03-25 2024-01-17 F. Hoffmann-La Roche AG Multiplexed total antibody and antibody-conjugated drug quantification assay
CN109563158B (en) 2016-04-04 2022-08-09 比奥贝拉蒂美国公司 Anti-complement factor BB antibodies and uses thereof
EP3865511A1 (en) 2016-04-14 2021-08-18 F. Hoffmann-La Roche AG Anti-rspo3 antibodies and methods of use
AU2017248766A1 (en) 2016-04-15 2018-11-01 Genentech, Inc. Methods for monitoring and treating cancer
EP3442562B1 (en) 2016-04-15 2022-09-21 Evive Biotechnology (Shanghai) Ltd An il-22 dimer for use in treating necrotizing enterocolitis
SG11201808783XA (en) 2016-04-15 2018-11-29 Alpine Immune Sciences Inc Cd80 variant immunomodulatory proteins and uses thereof
EP3443012A4 (en) 2016-04-15 2019-09-11 Bioatla, LLC Anti-axl antibodies, antibody fragments and their immunoconjugates and uses thereof
WO2017181148A2 (en) 2016-04-15 2017-10-19 Alpine Immune Sciences, Inc. Icos ligand variant immunomodulatory proteins and uses thereof
MX2018012492A (en) 2016-04-15 2019-06-06 Genentech Inc Methods for monitoring and treating cancer.
CA3020864A1 (en) 2016-04-15 2017-10-19 Macrogenics, Inc. Novel b7-h3-binding molecules, antibody drug conjugates thereof and methods of use thereof
AU2017254674A1 (en) 2016-04-21 2018-11-01 Abbvie Stemcentrx Llc Novel anti-BMPR1B antibodies and methods of use
AU2017259869A1 (en) 2016-05-02 2018-09-27 F. Hoffmann-La Roche Ag The contorsbody - a single chain target binder
EA039084B1 (en) 2016-05-09 2021-12-01 Бристол-Майерс Сквибб Компани Tl1a antibodies and uses thereof
JP7285076B2 (en) 2016-05-11 2023-06-01 エフ・ホフマン-ラ・ロシュ・アクチェンゲゼルシャフト Antigen-binding molecule comprising a TNF family ligand trimer and a tenascin-binding portion
EP3243836A1 (en) 2016-05-11 2017-11-15 F. Hoffmann-La Roche AG C-terminally fused tnf family ligand trimer-containing antigen binding molecules
JP7089483B2 (en) 2016-05-11 2022-06-22 エフ・ホフマン-ラ・ロシュ・アクチェンゲゼルシャフト Modified anti-tenascin antibody and usage
RS63698B1 (en) 2016-05-13 2022-11-30 Bioatla Inc Anti-ror2 antibodies, antibody fragments, their immunoconjugates and uses thereof
EP3243832A1 (en) 2016-05-13 2017-11-15 F. Hoffmann-La Roche AG Antigen binding molecules comprising a tnf family ligand trimer and pd1 binding moiety
ES2858151T3 (en) 2016-05-20 2021-09-29 Hoffmann La Roche PROTAC-Antibody Conjugates and Procedures for Use
BR112018074032A2 (en) * 2016-05-23 2019-02-26 Momenta Pharmaceuticals, Inc. compositions and methods related to manipulated fc constructs
EP3464362B1 (en) 2016-05-27 2020-12-09 AbbVie Biotherapeutics Inc. Anti-4-1bb antibodies and their uses
EP3465221B1 (en) 2016-05-27 2020-07-22 H. Hoffnabb-La Roche Ag Bioanalytical method for the characterization of site-specific antibody-drug conjugates
KR102543118B1 (en) 2016-05-27 2023-06-14 아게누스 인코포레이티드 Anti-tim-3 antibodies and methods of use thereof
EP3252078A1 (en) 2016-06-02 2017-12-06 F. Hoffmann-La Roche AG Type ii anti-cd20 antibody and anti-cd20/cd3 bispecific antibody for treatment of cancer
CN110603266A (en) 2016-06-02 2019-12-20 豪夫迈·罗氏有限公司 Type II anti-CD 20 and anti-CD 20/CD3 bispecific antibodies for the treatment of cancer
DK3462853T3 (en) 2016-06-03 2023-04-03 Regeneron Pharma Rodents expressing exogenous terminal deoxynucleotidyl transferase
JP7043425B2 (en) 2016-06-06 2022-03-29 ジェネンテック, インコーポレイテッド Silvestrol Antibodies-Drug Conjugates and Usage
JP7148504B2 (en) 2016-06-08 2022-10-05 ゼンコー,インコーポレイティド Treatment of IgG4-related diseases with anti-CD19 antibodies cross-linked to CD32B
TW201808987A (en) 2016-06-08 2018-03-16 健生生物科技公司 GM-CSF variants and methods of use
EP3475298A1 (en) 2016-06-24 2019-05-01 H. Hoffnabb-La Roche Ag Anti-polyubiquitin multispecific antibodies
WO2018002181A1 (en) 2016-06-28 2018-01-04 Umc Utrecht Holding B.V. TREATMENT OF IgE-MEDIATED DISEASES WITH ANTIBODIES THAT SPECIFICALLY BIND CD38
MA45554A (en) 2016-07-01 2019-05-08 Resolve Therapeutics Llc OPTIMIZED BINUCLEASE FUSIONS.
JP6983824B2 (en) 2016-07-04 2021-12-17 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft New antibody format
EP3481393B1 (en) 2016-07-05 2021-04-14 Beigene, Ltd. Combination of a pd-1 antagonist and a raf inhibitor for treating cancer
EP3481864A1 (en) 2016-07-08 2019-05-15 Staten Biotechnology B.V. Anti-apoc3 antibodies and methods of use thereof
CN109757103B (en) 2016-07-14 2024-01-02 百时美施贵宝公司 Antibodies against TIM3 and uses thereof
CN117330747A (en) 2016-07-15 2024-01-02 武田药品工业株式会社 Methods and materials for assessing response to plasmacytoid and plasma cell depleting therapies
PT3496739T (en) 2016-07-15 2021-06-21 Acceleron Pharma Inc Compositions and methods for treating pulmonary hypertension
WO2018014260A1 (en) 2016-07-20 2018-01-25 Nanjing Legend Biotech Co., Ltd. Multispecific antigen binding proteins and methods of use thereof
TWI781108B (en) 2016-07-20 2022-10-21 比利時商健生藥品公司 Anti- gprc5d antibodies, bispecific antigen binding molecules that bind gprc5d and cd3, and uses thereof
WO2018022479A1 (en) 2016-07-25 2018-02-01 Biogen Ma Inc. Anti-hspa5 (grp78) antibodies and uses thereof
KR20190040972A (en) 2016-07-27 2019-04-19 악셀레론 파마 인코포레이티드 Methods and compositions for treatment of osteoporosis
US11471488B2 (en) 2016-07-28 2022-10-18 Alpine Immune Sciences, Inc. CD155 variant immunomodulatory proteins and uses thereof
CN110088127A (en) 2016-07-28 2019-08-02 高山免疫科学股份有限公司 CD155 variant immune modulator and application thereof
US11834490B2 (en) 2016-07-28 2023-12-05 Alpine Immune Sciences, Inc. CD112 variant immunomodulatory proteins and uses thereof
MX2018015721A (en) 2016-07-29 2019-05-27 Chugai Pharmaceutical Co Ltd Bispecific antibody exhibiting increased alternative fviii-cofactor-function activity.
CA3031734A1 (en) 2016-07-29 2018-02-01 Juno Therapeutics, Inc. Anti-idiotypic antibodies against anti-cd19 antibodies
JP7148493B2 (en) 2016-08-01 2022-10-05 ゾーマ (ユーエス) リミテッド ライアビリティ カンパニー Parathyroid hormone receptor 1 (PTH1R) antibodies and uses thereof
MX2019001458A (en) * 2016-08-02 2019-07-04 Visterra Inc Engineered polypeptides and uses thereof.
JP2019530434A (en) 2016-08-05 2019-10-24 ジェネンテック, インコーポレイテッド Multivalent and multi-epitope antibodies with agonist activity and methods of use
SG11201801024XA (en) 2016-08-05 2018-05-30 Chugai Pharmaceutical Co Ltd Therapeutic or preventive compositions for il-8-related diseases
WO2018027124A1 (en) 2016-08-05 2018-02-08 Medimmune, Llc Anti-o2 antibodies and uses thereof
WO2018029124A1 (en) 2016-08-08 2018-02-15 F. Hoffmann-La Roche Ag Therapeutic and diagnostic methods for cancer
US10696722B2 (en) 2016-08-10 2020-06-30 Ajou University Industry-Academic Cooperation Foundation Heterodimeric Fc-fused cytokine and pharmaceutical composition comprising the same
CN109689111B (en) 2016-08-11 2024-04-05 基因泰克公司 Pyrrolobenzodiazepine prodrugs and antibody conjugates thereof
JP7009448B2 (en) 2016-08-12 2022-02-10 ヤンセン バイオテツク,インコーポレーテツド Fc genetically engineered anti-TNFR superfamily member antibody with enhanced agonist activity and how to use it
CA3033661A1 (en) 2016-08-12 2018-02-15 Janssen Biotech, Inc. Engineered antibodies and other fc-domain containing molecules with enhanced agonism and effector functions
KR102340687B1 (en) * 2016-08-15 2021-12-16 국립대학법인 홋가이도 다이가쿠 anti-LAG-3 antibody
AU2017313405A1 (en) 2016-08-17 2019-02-28 Compugen Ltd. Anti-TIGIT antibodies, anti-PVRIG antibodies and combinations thereof
FI3500299T3 (en) 2016-08-19 2024-02-14 Beigene Switzerland Gmbh Combination of zanubrutinib with an anti-cd20 or an anti-pd-1 antibody for use in treating cancer
WO2018044970A1 (en) 2016-08-31 2018-03-08 University Of Rochester Human monoclonal antibodies to human endogenous retrovirus k envelope (herv-k) and uses thereof
EP3510046A4 (en) 2016-09-07 2020-05-06 The Regents of the University of California Antibodies to oxidation-specific epitopes
US20190270821A1 (en) 2016-09-13 2019-09-05 Humanigen, Inc. Epha3 antibodies for the treatment of pulmonary fibrosis
BR112019005129A2 (en) 2016-09-16 2019-06-04 Shanghai Henlius Biotech Inc anti-pd-1 antibodies
SG10201607778XA (en) 2016-09-16 2018-04-27 Chugai Pharmaceutical Co Ltd Anti-Dengue Virus Antibodies, Polypeptides Containing Variant Fc Regions, And Methods Of Use
JP2019534859A (en) 2016-09-19 2019-12-05 セルジーン コーポレイション Method for treating vitiligo using PD-1 binding protein
SG11201901950TA (en) 2016-09-19 2019-04-29 Celgene Corp Methods of treating immune disorders using pd-1 binding proteins
WO2018050878A1 (en) 2016-09-19 2018-03-22 F. Hoffmann-La Roche Ag Complement factor based affinity chromatography
UA124269C2 (en) 2016-09-23 2021-08-18 Дженентек, Інк. Uses of il-13 antagonists for treating atopic dermatitis
CN109952314A (en) 2016-09-23 2019-06-28 泰瓦制药国际有限公司 Treat intractable migraine
MX2019003543A (en) 2016-09-28 2019-06-17 Xoma Us Llc Antibodies that bind interleukin-2 and uses thereof.
CN109843926B (en) 2016-09-30 2023-01-20 豪夫迈·罗氏有限公司 Bispecific antibodies against CD3
EP3522933B1 (en) 2016-10-05 2021-12-15 F. Hoffmann-La Roche AG Methods for preparing antibody drug conjugates
AU2017340504A1 (en) 2016-10-05 2019-04-11 Acceleron Pharma, Inc. Compositions and method for treating kidney disease
KR20190072528A (en) 2016-10-06 2019-06-25 제넨테크, 인크. Treatment and Diagnosis Methods for Cancer
WO2018065552A1 (en) 2016-10-06 2018-04-12 Innate Pharma Anti-cd39 antibodies
US10844119B2 (en) 2016-10-11 2020-11-24 Agenus Inc. Anti-LAG-3 antibodies and methods of use thereof
WO2018068201A1 (en) 2016-10-11 2018-04-19 Nanjing Legend Biotech Co., Ltd. Single-domain antibodies and variants thereof against ctla-4
JP7142630B2 (en) 2016-10-14 2022-09-27 ゼンコア インコーポレイテッド IL15/IL15Rα heterodimeric FC-fusion protein
JP7181862B2 (en) 2016-10-18 2022-12-01 リージェンツ オブ ザ ユニバーシティ オブ ミネソタ Tumor-infiltrating lymphocytes and methods of treatment
KR20190064636A (en) 2016-10-19 2019-06-10 메디뮨 엘엘씨 Anti-O1 antibodies and uses thereof
TW202300515A (en) 2016-10-20 2023-01-01 法商賽諾菲公司 Anti-chikv antibodies and uses thereof
WO2018073363A1 (en) 2016-10-21 2018-04-26 Innate Pharma Treatment with anti-kir3dl2 agents
MY191324A (en) 2016-10-26 2022-06-15 Cedars Sinai Medical Center Neutralizing anti-tl1a monoclonal antibodies
GB2571036B (en) * 2016-10-27 2022-09-07 Univ Kookmin Ind Acad Coop Found Aglycosylated antibody Fc region for treating cancer
EP3533466A4 (en) 2016-10-28 2020-06-10 Toray Industries, Inc. Pharmaceutical composition for cancer treatment and/or prevention
EP3532091A2 (en) 2016-10-29 2019-09-04 H. Hoffnabb-La Roche Ag Anti-mic antibidies and methods of use
TWI788307B (en) 2016-10-31 2023-01-01 美商艾歐凡斯生物治療公司 Engineered artificial antigen presenting cells for tumor infiltrating lymphocyte expansion
MD3535298T2 (en) 2016-11-02 2022-01-31 Jounce Therapeutics Inc Antibodies to pd-1 and uses thereof
WO2018083080A2 (en) 2016-11-04 2018-05-11 Innate Pharma Nkp46 ligand
CA3042435A1 (en) 2016-11-15 2018-05-24 Genentech, Inc. Dosing for treatment with anti-cd20/anti-cd3 bispecific antibodies
TW201829463A (en) 2016-11-18 2018-08-16 瑞士商赫孚孟拉羅股份公司 Anti-hla-g antibodies and use thereof
JOP20190100A1 (en) 2016-11-19 2019-05-01 Potenza Therapeutics Inc Anti-gitr antigen-binding proteins and methods of use thereof
AU2017361887B2 (en) 2016-11-21 2019-08-15 Cureab Gmbh Anti-GP73 antibodies and immunoconjugates
MA46893A (en) 2016-11-23 2019-10-02 Bioverativ Therapeutics Inc BISPECIFIC ANTIBODIES BINDING TO COAGULATION FACTOR IX AND COAGULATION FACTOR X
TW201825119A (en) 2016-11-30 2018-07-16 日商協和醱酵麒麟有限公司 Method of treating cancer using anti-ccr4 antibody and anti-pd-1 antibody
MX2019006446A (en) 2016-12-02 2019-12-11 Bioverativ Therapeutics Inc Methods of inducing immune tolerance to clotting factors.
MA46967A (en) 2016-12-02 2019-10-09 Bioverativ Therapeutics Inc METHODS OF TREATMENT OF HEMOPHILIC ARTHROPATHY USING CHEMERICAL COAGULATION FACTORS
WO2018106776A2 (en) 2016-12-07 2018-06-14 Genentech, Inc. Anti-tau antibodies and methods of use
IL307242A (en) 2016-12-07 2023-11-01 Agenus Inc Anti-ctla-4 antibodies and methods of use thereof
CN110300599A (en) 2016-12-07 2019-10-01 艾吉纳斯公司 Antibody and its application method
WO2018106781A1 (en) 2016-12-07 2018-06-14 Genentech, Inc Anti-tau antibodies and methods of use
JP2020500856A (en) 2016-12-09 2020-01-16 グリックニック インコーポレイテッド Method for treating inflammatory disease using polyvalent FC compound
KR20190095921A (en) 2016-12-12 2019-08-16 제넨테크, 인크. How to Treat Cancer Using Anti-PD-L1 Antibody and Antiandrogen
TWI829628B (en) 2016-12-19 2024-01-21 瑞士商赫孚孟拉羅股份公司 Combination therapy with targeted 4-1bb (cd137) agonists
CA3039446A1 (en) 2016-12-20 2018-06-28 F. Hoffmann-La Roche Ag Combination therapy of anti-cd20/anti-cd3 bispecific antibodies and 4-1bb (cd137) agonists
MX2019007433A (en) 2016-12-22 2019-08-16 Genentech Inc Methods and formulations for reducing reconstitution time of lyophilized polypeptides.
EP3559032A1 (en) 2016-12-23 2019-10-30 Innate Pharma Heterodimeric antigen binding proteins
JOP20190134A1 (en) 2016-12-23 2019-06-02 Potenza Therapeutics Inc Anti-neuropilin antigen-binding proteins and methods of use thereof
RU2019123613A (en) 2017-01-03 2021-02-05 Ф. Хоффманн-Ля Рош Аг BISPECIFIC ANTIGENBINDING MOLECULES CONTAINING ANTIBODY K4-1BB, CLONE 20N4.9
US20180230218A1 (en) 2017-01-04 2018-08-16 Immunogen, Inc. Met antibodies and immunoconjugates and uses thereof
JP2020503351A (en) 2017-01-06 2020-01-30 アイオバンス バイオセラピューティクス,インコーポレイテッド Proliferation of tumor infiltrating lymphocytes by potassium channel agonist and its therapeutic use
MX2019007963A (en) 2017-01-06 2019-10-21 Iovance Biotherapeutics Inc Expansion of tumor infiltrating lymphocytes (tils) with tumor necrosis factor receptor superfamily (tnfrsf) agonists and therapeutic combinations of tils and tnfrsf agonists.
BR112019013955A2 (en) 2017-01-06 2020-02-11 Momenta Pharmaceuticals, Inc. COMPOSITIONS AND METHODS RELATED TO HANDLED FC CONSTRUCTS
WO2018132597A1 (en) 2017-01-12 2018-07-19 Eureka Therapeutics, Inc. Constructs targeting histone h3 peptide/mhc complexes and uses thereof
WO2018139404A1 (en) 2017-01-24 2018-08-02 協和発酵キリン株式会社 Therapeutic or prophylactic agent and treatment or prevention method for radiation sickness
WO2018138032A2 (en) 2017-01-24 2018-08-02 Innate Pharma NKp46 BINDING AGENTS
US11555038B2 (en) 2017-01-25 2023-01-17 Beigene, Ltd. Crystalline forms of (S)-7-(1-(but-2-ynoyl)piperidin-4-yl)-2-(4-phenoxyphenyl)-4,5,6,7-tetrahydropyrazolo[1,5-a]pyrimidine-3-carboxamide, preparation, and uses thereof
WO2018139623A1 (en) 2017-01-30 2018-08-02 Chugai Seiyaku Kabushiki Kaisha Anti-sclerostin antibodies and methods of use
SG10201908697XA (en) 2017-01-31 2019-10-30 Chugai Pharmaceutical Co Ltd A pharmaceutical composition for use in the treatment or prevention of a c5-related disease and a method for treating or preventing a c5-related disease
WO2018141959A1 (en) 2017-02-06 2018-08-09 Innate Pharma Immunomodulatory antibody drug conjugates binding to a human mica polypeptide
KR20240023449A (en) 2017-02-08 2024-02-21 드래곤플라이 쎄라퓨틱스, 인크. Multi-specific binding proteins for activation of natural killer cells and therapeutic uses thereof to treat cancer
JP6995127B2 (en) 2017-02-10 2022-02-04 ジェネンテック, インコーポレイテッド Anti-tryptase antibody, its composition, and its use
CA3051839A1 (en) 2017-02-17 2018-08-23 Bristol-Myers Squibb Company Antibodies to alpha-synuclein and uses thereof
RS63818B1 (en) 2017-02-17 2023-01-31 Denali Therapeutics Inc Engineered transferrin receptor binding polypeptides
EP3589320A4 (en) 2017-02-28 2020-12-23 Seagen Inc. Cysteine mutated antibodies for conjugation
CA3052670A1 (en) 2017-03-01 2018-09-07 Genentech, Inc. Diagnostic and therapeutic methods for cancer
WO2018159845A1 (en) 2017-03-02 2018-09-07 学校法人 聖マリアンナ医科大学 Preventive or therapeutic agent for htlv-1-associated myelopathy using low-dose anti-ccr4 antibody
EP3589657A1 (en) 2017-03-03 2020-01-08 Rinat Neuroscience Corp. Anti-gitr antibodies and methods of use thereof
GB201703876D0 (en) 2017-03-10 2017-04-26 Berlin-Chemie Ag Pharmaceutical combinations
ES2963638T3 (en) 2017-03-16 2024-04-01 Alpine Immune Sciences Inc Immunomodulatory proteins of PD-L1 variants and their uses
NZ756395A (en) 2017-03-16 2024-01-26 Alpine Immune Sciences Inc Cd80 variant immunomodulatory proteins and uses thereof
MX2019010848A (en) 2017-03-16 2019-10-30 Innate Pharma Compositions and methods for treating cancer.
WO2018170023A1 (en) 2017-03-16 2018-09-20 Alpine Immune Sciences, Inc. Pd-l2 variant immunomodulatory proteins and uses thereof
KR102627372B1 (en) 2017-03-20 2024-01-19 백시넥스 인코포레이티드 Treatment of cancer using semaphorin-4D antibody in combination with epigenetic modulators
JP7227151B2 (en) 2017-03-22 2023-02-21 ジェネンテック, インコーポレイテッド Antibody Compositions Optimized for Treatment of Eye Disorders
CN108623686A (en) 2017-03-25 2018-10-09 信达生物制药(苏州)有限公司 Anti- OX40 antibody and application thereof
AU2018241624B2 (en) 2017-03-27 2024-01-25 F. Hoffmann-La Roche Ag Improved antigen binding receptors
AU2018244224A1 (en) 2017-03-28 2019-09-19 Genentech, Inc. Methods of treating neurodegenerative diseases
EP3601346A1 (en) 2017-03-29 2020-02-05 H. Hoffnabb-La Roche Ag Bispecific antigen binding molecule for a costimulatory tnf receptor
CN110573528B (en) 2017-03-29 2023-06-09 豪夫迈·罗氏有限公司 Bispecific antigen binding molecules to costimulatory TNF receptors
JOP20190203A1 (en) 2017-03-30 2019-09-03 Potenza Therapeutics Inc Anti-tigit antigen-binding proteins and methods of use thereof
CA3053357A1 (en) 2017-04-03 2018-10-11 F. Hoffmann-La Roche Ag Immunoconjugates of an anti-pd-1 antibody with a mutant il-2 or with il-15
WO2018187191A1 (en) 2017-04-03 2018-10-11 Jounce Therapeutics, Inc Compositions and methods for the treatment of cancer
CN110382525B (en) 2017-04-03 2023-10-20 豪夫迈·罗氏有限公司 Immunoconjugates
WO2018185618A1 (en) 2017-04-03 2018-10-11 Novartis Ag Anti-cdh6 antibody drug conjugates and anti-gitr antibody combinations and methods of treatment
JP7247101B2 (en) 2017-04-03 2023-03-28 エフ・ホフマン-ラ・ロシュ・アクチェンゲゼルシャフト Antibody that binds to STEAP-1
MX2019011907A (en) 2017-04-04 2020-01-09 Hoffmann La Roche Novel bispecific antigen binding molecules capable of specific binding to cd40 and to fap.
KR102408873B1 (en) 2017-04-05 2022-06-15 에프. 호프만-라 로슈 아게 Bispecific antibodies specifically binding to pd1 and lag3
PL3606954T3 (en) 2017-04-05 2022-10-17 F. Hoffmann-La Roche Ag Anti-lag3 antibodies
MX2019012187A (en) 2017-04-13 2019-11-25 Hoffmann La Roche An interleukin-2 immunoconjugate, a cd40 agonist, and optionally a pd-1 axis binding antagonist for use in methods of treating cancer.
JP7297672B2 (en) 2017-04-13 2023-06-26 アジェナス インコーポレイテッド ANTI-CD137 ANTIBODY AND METHODS OF USE THEREOF
CN110621787A (en) 2017-04-14 2019-12-27 豪夫迈·罗氏有限公司 Methods for diagnosis and treatment of cancer
PE20200150A1 (en) 2017-04-21 2020-01-17 Genentech Inc USE OF KLK5 ANTAGONISTS FOR THE TREATMENT OF A DISEASE
WO2018193427A1 (en) 2017-04-21 2018-10-25 Staten Biotechnology B.V. Anti-apoc3 antibodies and methods of use thereof
SG11201909728XA (en) 2017-04-26 2019-11-28 Eureka Therapeutics Inc Constructs specifically recognizing glypican 3 and uses thereof
JP2020517695A (en) 2017-04-27 2020-06-18 テサロ, インコーポレイテッド Antibody drugs against lymphocyte activation gene-3 (LAG-3) and their use
HUE062927T2 (en) 2017-05-01 2023-12-28 Agenus Inc Anti-tigit antibodies and methods of use thereof
JP7185884B2 (en) 2017-05-02 2022-12-08 国立研究開発法人国立精神・神経医療研究センター METHOD FOR PREDICTING AND DETERMINING THERAPEUTIC EFFECT OF IL-6 AND NEUTROPHIL-RELATED DISEASE
BR112019023138A2 (en) 2017-05-05 2020-07-28 Vaccinex, Inc. 4d human anti-semaphorin antibody
MX2019013137A (en) 2017-05-05 2020-07-14 Allakos Inc Methods and compositions for treating allergic ocular diseases.
WO2019103857A1 (en) 2017-11-22 2019-05-31 Iovance Biotherapeutics, Inc. Expansion of peripheral blood lymphocytes (pbls) from peripheral blood
WO2018209115A1 (en) 2017-05-10 2018-11-15 Iovance Biotherapeutics, Inc. Expansion of tumor infiltrating lymphocytes from liquid tumors and therapeutic uses thereof
US11168129B2 (en) 2017-05-15 2021-11-09 University Of Rochester Broadly neutralizing anti-influenza human monoclonal antibody and uses thereof
CN110621336A (en) 2017-05-16 2019-12-27 戊瑞治疗有限公司 Combination of an anti-FGFR 2 antibody with a chemotherapeutic agent in the treatment of cancer
CN116333129A (en) 2017-05-25 2023-06-27 百时美施贵宝公司 Antibodies comprising modified heavy chain constant regions
EP3630831B1 (en) 2017-05-25 2022-06-15 Bristol-Myers Squibb Company Antagonistic cd40 monoclonal antibodies and uses thereof
AU2018276140A1 (en) 2017-06-01 2019-12-19 Compugen Ltd. Triple combination antibody therapies
KR20230035421A (en) 2017-06-02 2023-03-13 화이자 인코포레이티드 Antibodies specific for flt3 and their uses
EP3642239A1 (en) 2017-06-20 2020-04-29 Amgen Inc. Method of treating or ameliorating metabolic disorders using binding proteins for gastric inhibitory peptide receptor (gipr) in combination with glp-1 agonists
US11597768B2 (en) 2017-06-26 2023-03-07 Beigene, Ltd. Immunotherapy for hepatocellular carcinoma
JP2020530307A (en) 2017-06-30 2020-10-22 インティマ・バイオサイエンス,インコーポレーテッド Adeno-associated virus vector for gene therapy
US11084863B2 (en) 2017-06-30 2021-08-10 Xencor, Inc. Targeted heterodimeric Fc fusion proteins containing IL-15 IL-15alpha and antigen binding domains
US11623954B2 (en) 2017-07-10 2023-04-11 Innate Pharma Siglec-9-neutralizing antibodies
US11447545B2 (en) 2017-07-10 2022-09-20 Innate Pharma Combination therapy using antibody to human Siglec-9 and antibody to human NKG2A for treating cancer
EP3652206A1 (en) 2017-07-10 2020-05-20 International-Drug-Development-Biotech Treatment of b cell malignancies using afucosylated pro-apoptotic anti-cd19 antibodies in combination with anti cd20 antibodies or chemotherapeutics
KR20200029469A (en) 2017-07-18 2020-03-18 쿄와 기린 가부시키가이샤 Anti-human CCR1 monoclonal antibody
TWI823859B (en) 2017-07-21 2023-12-01 美商建南德克公司 Therapeutic and diagnostic methods for cancer
AU2018308364C1 (en) 2017-07-26 2023-02-16 Forty Seven, Inc. Anti-SIRP-alpha antibodies and related methods
SI3658184T1 (en) 2017-07-27 2024-01-31 Alexion Pharmaceuticals, Inc., High concentration anti-c5 antibody formulations
JP7374883B2 (en) 2017-08-09 2023-11-07 バイオベラティブ セラピューティクス インコーポレイテッド Nucleic acid molecules and their uses
KR20200035972A (en) * 2017-08-11 2020-04-06 리서치 디벨럽먼트 파운데이션 Engineered antibody FC variants for enhanced serum half-life
CA3070297A1 (en) 2017-08-11 2019-02-14 Genentech, Inc. Anti-cd8 antibodies and uses thereof
WO2019036605A2 (en) 2017-08-17 2019-02-21 Massachusetts Institute Of Technology Multiple specificity binders of cxc chemokines and uses thereof
MX2020002070A (en) 2017-08-22 2020-03-24 Sanabio Llc Soluble interferon receptors and uses thereof.
MA49950A (en) 2017-08-25 2020-07-01 Five Prime Therapeutics Inc ANTI-B7-H4 ANTIBODIES AND PROCEDURES FOR USE
EP3456737B1 (en) * 2017-09-19 2024-02-14 Tillotts Pharma Ag Antibody variants
WO2019059411A1 (en) 2017-09-20 2019-03-28 Chugai Seiyaku Kabushiki Kaisha Dosage regimen for combination therapy using pd-1 axis binding antagonists and gpc3 targeting agent
WO2019067499A1 (en) 2017-09-27 2019-04-04 Alexion Pharmaceuticals, Inc. Biomarker signature for predicting tumor response to anti-cd200 therapy
EP3690050A4 (en) 2017-09-29 2021-06-16 Chugai Seiyaku Kabushiki Kaisha Multispecific antigen-binding molecule having blood coagulation factor viii (fviii) cofactor function-substituting activity, and pharmaceutical formulation containing said molecule as active ingredient
WO2019075090A1 (en) 2017-10-10 2019-04-18 Tilos Therapeutics, Inc. Anti-lap antibodies and uses thereof
CA3198255A1 (en) 2017-10-10 2019-04-18 Alpine Immune Sciences, Inc. Ctla-4 variant immunomodulatory proteins and uses thereof
WO2019075270A1 (en) 2017-10-12 2019-04-18 Amesino Llc Vegfr-antibody light chain fusion protein
BR112020007309A2 (en) 2017-10-14 2020-09-29 Cytomx Therapeutics, Inc. antibodies, activable antibodies, bispecific antibodies and bispecific activable antibodies and methods of using them
WO2019079520A2 (en) 2017-10-18 2019-04-25 Alpine Immune Sciences, Inc. Variant icos ligand immunomodulatory proteins and related compositions and methods
TW201932142A (en) 2017-10-20 2019-08-16 瑞士商赫孚孟拉羅股份公司 Method for generating multispecific antibodies from monospecific antibodies
MX2020004100A (en) 2017-10-30 2020-07-24 Hoffmann La Roche Method for in vivo generation of multispecific antibodies from monospecific antibodies.
US11618787B2 (en) 2017-10-31 2023-04-04 Janssen Biotech, Inc. Methods of treating high risk multiple myeloma
JP7039694B2 (en) 2017-10-31 2022-03-22 スターテン・バイオテクノロジー・ベー・フェー Anti-APOC3 antibody and how to use it
US10538583B2 (en) 2017-10-31 2020-01-21 Staten Biotechnology B.V. Anti-APOC3 antibodies and compositions thereof
KR20200079492A (en) 2017-11-01 2020-07-03 에프. 호프만-라 로슈 아게 Bispecific 2+1 connector
EP3704155A2 (en) 2017-11-01 2020-09-09 F. Hoffmann-La Roche AG Combination therapy with targeted ox40 agonists
WO2019086499A1 (en) 2017-11-01 2019-05-09 F. Hoffmann-La Roche Ag Novel tnf family ligand trimer-containing antigen binding molecules
AU2018359967A1 (en) 2017-11-06 2020-04-23 Genentech, Inc. Diagnostic and therapeutic methods for cancer
JP7311425B2 (en) 2017-11-08 2023-07-19 協和キリン株式会社 Bispecific antibodies that bind to CD40 and EpCAM
CA3079363A1 (en) 2017-11-21 2019-05-31 Innate Pharma Multispecific antigen binding proteins
US11786529B2 (en) 2017-11-29 2023-10-17 Beigene Switzerland Gmbh Treatment of indolent or aggressive B-cell lymphomas using a combination comprising BTK inhibitors
SG11202003930YA (en) 2017-12-01 2020-05-28 Pfizer Anti-cxcr5 antibodies and compositions and uses thereof
JP2021508439A (en) 2017-12-01 2021-03-11 ノバルティス アーゲー Polyomavirus neutralizing antibody
US11952422B2 (en) 2017-12-05 2024-04-09 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule comprising altered antibody variable region binding CD3 and CD137
EP3720877A1 (en) 2017-12-08 2020-10-14 Argenx BVBA Use of fcrn antagonists for treatment of generalized myasthenia gravis
WO2019118873A2 (en) 2017-12-15 2019-06-20 Iovance Biotherapeutics, Inc. Systems and methods for determining the beneficial administration of tumor infiltrating lymphocytes, and methods of use thereof and beneficial administration of tumor infiltrating lymphocytes, and methods of use thereof
WO2019126536A1 (en) 2017-12-20 2019-06-27 Alexion Pharmaceuticals Inc. Humanized anti-cd200 antibodies and uses thereof
WO2019126133A1 (en) 2017-12-20 2019-06-27 Alexion Pharmaceuticals, Inc. Liquid formulations of anti-cd200 antibodies
WO2019122046A1 (en) 2017-12-21 2019-06-27 F. Hoffmann-La Roche Ag Universal reporter cell assay for specificity test of novel antigen binding moieties
EP3502140A1 (en) 2017-12-21 2019-06-26 F. Hoffmann-La Roche AG Combination therapy of tumor targeted icos agonists with t-cell bispecific molecules
CN111527107A (en) 2017-12-21 2020-08-11 豪夫迈·罗氏有限公司 Antibodies that bind HLA-A2/WT1
JP7391868B2 (en) 2017-12-22 2023-12-05 ジョウンセ セラピューティクス, インク. Antibody against LILRB2
WO2019126472A1 (en) 2017-12-22 2019-06-27 Genentech, Inc. Use of pilra binding agents for treatment of a disease
WO2019129136A1 (en) 2017-12-27 2019-07-04 信达生物制药(苏州)有限公司 Anti-pd-l1 antibody and uses thereof
CN115925943A (en) 2017-12-27 2023-04-07 信达生物制药(苏州)有限公司 Anti-PD-L1 antibodies and uses thereof
WO2019129137A1 (en) 2017-12-27 2019-07-04 信达生物制药(苏州)有限公司 Anti-lag-3 antibody and uses thereof
JP7314146B2 (en) 2017-12-28 2023-07-25 中外製薬株式会社 Cytotoxicity-inducing therapeutic agent
KR20200104886A (en) 2017-12-28 2020-09-04 난징 레전드 바이오테크 씨오., 엘티디. Antibodies and variants against PD-L1
US11905327B2 (en) 2017-12-28 2024-02-20 Nanjing Legend Biotech Co., Ltd. Single-domain antibodies and variants thereof against TIGIT
EP3731864A1 (en) 2017-12-29 2020-11-04 F. Hoffmann-La Roche SA Anti-vegf antibodies and methods of use
CA3087149A1 (en) 2018-01-03 2019-07-11 Alpine Immune Sciences, Inc. Multi-domain immunomodulatory proteins and methods of use thereof
KR20200118029A (en) 2018-01-04 2020-10-14 아이코닉 테라퓨틱스, 인코포레이티드 Anti-tissue factor antibodies, antibody-drug conjugates, and related methods
KR20200106525A (en) 2018-01-05 2020-09-14 에이씨 이뮨 에스.에이. Misfolded TDP-43 binding molecule
BR112020014052A2 (en) 2018-01-12 2020-12-08 Takeda Pharmaceutical Company Limited SUBCUTANEOUS DOSAGE OF ANTI-CD38 ANTIBODIES
US20220089720A1 (en) 2018-01-12 2022-03-24 Bristol-Myers Squibb Company Antibodies against tim3 and uses thereof
SG11202004233UA (en) 2018-01-15 2020-06-29 Nanjing Legend Biotech Co Ltd Single-domain antibodies and variants thereof against pd-1
US20200339686A1 (en) 2018-01-16 2020-10-29 Lakepharma, Inc. Bispecific antibody that binds cd3 and another target
US11402380B2 (en) 2018-01-25 2022-08-02 Emory University Assays for detecting antibodies capable of mediating antibody-dependent cell-mediated cytotoxicity
LT3743088T (en) 2018-01-26 2022-12-27 F. Hoffmann-La Roche Ag Il-22 fc compositions and methods of use
RU2020127792A (en) 2018-01-26 2022-02-28 Дженентек, Инк. IL-22-Fc fusion proteins AND METHODS OF APPLICATION
CN111918674A (en) 2018-02-01 2020-11-10 比奥维拉迪维治疗股份有限公司 Use of lentiviral vectors expressing factor VIII
EP3746483A1 (en) 2018-02-01 2020-12-09 Pfizer Inc Chimeric antigen receptors targeting cd70
TW201934580A (en) 2018-02-01 2019-09-01 美商輝瑞大藥廠 Antibodies specific for CD70 and their uses
EP3749346A4 (en) 2018-02-08 2021-12-15 Dragonfly Therapeutics, Inc. Antibody variable domains targeting the nkg2d receptor
MA51793A (en) 2018-02-08 2020-12-16 Hoffmann La Roche BISPECIFIC ANTIGEN BINDING MOLECULES AND METHODS OF USE
JP7418337B2 (en) 2018-02-09 2024-01-19 ジェネンテック, インコーポレイテッド Treatment and diagnosis of mast cell-mediated inflammatory diseases
TWI829667B (en) 2018-02-09 2024-01-21 瑞士商赫孚孟拉羅股份公司 Antibodies binding to gprc5d
JP2021512962A (en) 2018-02-13 2021-05-20 アイオバンス バイオセラピューティクス,インコーポレイテッド Expansion culture of tumor-infiltrating lymphocytes (TIL) with adenosine A2A receptor antagonist and therapeutic combination of TIL and adenosine A2A receptor antagonist
JP7350756B2 (en) 2018-02-14 2023-09-26 アバ セラピューティクス アーゲー Anti-human PD-L2 antibody
SG11202007694UA (en) 2018-02-21 2020-09-29 Genentech Inc DOSING FOR TREATMENT WITH IL-22 Fc FUSION PROTEINS
TW202000702A (en) 2018-02-26 2020-01-01 美商建南德克公司 Dosing for treatment with anti-TIGIT and anti-PD-L1 antagonist antibodies
SG11202007518RA (en) 2018-02-28 2020-09-29 Pfizer Il-15 variants and uses thereof
WO2019169212A1 (en) 2018-03-02 2019-09-06 Five Prime Therapeutics, Inc. B7-h4 antibodies and methods of use thereof
EP3765489A1 (en) 2018-03-13 2021-01-20 F. Hoffmann-La Roche AG Therapeutic combination of 4-1 bb agonists with anti-cd20 antibodies
TW202003561A (en) 2018-03-13 2020-01-16 瑞士商赫孚孟拉羅股份公司 Combination therapy with targeted 4-1BB (CD137) agonists
RU2020128013A (en) 2018-03-14 2022-04-15 Бейцзин Сюаньи Фармасайенсиз Ко., Лтд. ANTIBODIES AGAINST CLAUDIN 18.2
US20200040103A1 (en) 2018-03-14 2020-02-06 Genentech, Inc. Anti-klk5 antibodies and methods of use
CN112119090B (en) 2018-03-15 2023-01-13 中外制药株式会社 Anti-dengue virus antibodies cross-reactive to Zika virus and methods of use
SG11202008593PA (en) 2018-03-21 2020-10-29 Five Prime Therapeutics Inc ANTIBODIES BINDING TO VISTA AT ACIDIC pH
AU2019236865A1 (en) 2018-03-23 2020-10-01 Bristol-Myers Squibb Company Antibodies against MICA and/or MICB and uses thereof
EP3772926A1 (en) 2018-03-26 2021-02-17 Regeneron Pharmaceuticals, Inc. Humanized rodents for testing therapeutic agents
CA3095086A1 (en) 2018-03-28 2019-10-03 Takeda Pharmaceutical Company Limited Subcutaneous dosing of anti-cd38 antibodies
EA202092316A1 (en) 2018-03-28 2021-05-25 Бристол-Маерс Сквибб Компани FUSION PROTEINS OF INTERLEUKIN-2 / ALPHA-RECEPTOR OF INTERLEUKIN-2 AND METHODS OF APPLICATION
WO2019190984A1 (en) 2018-03-29 2019-10-03 Pfizer Inc. Lfa3 variants and compositions and uses thereof
CN111936625A (en) 2018-03-29 2020-11-13 豪夫迈·罗氏有限公司 Modulating lactogenic activity in mammalian cells
MX2020009862A (en) 2018-03-30 2020-10-08 Toray Industries Pharmaceutical composition for treatment and/or prevention of cancer.
SG11202006674QA (en) 2018-03-30 2020-08-28 Nanjing Legend Biotech Co Ltd Single-domain antibodies against lag-3 and uses thereof
SG11202009625WA (en) 2018-04-02 2020-10-29 Bristol Myers Squibb Co Anti-trem-1 antibodies and uses thereof
TW202011029A (en) 2018-04-04 2020-03-16 美商建南德克公司 Methods for detecting and quantifying FGF21
KR20200140878A (en) 2018-04-09 2020-12-16 암젠 인크 Growth differentiation factor 15 fusion protein
CN113880952A (en) * 2018-04-13 2022-01-04 艾菲默德有限责任公司 Natural killer cell-conjugated antibody fusion constructs
TW202012440A (en) 2018-04-13 2020-04-01 瑞士商赫孚孟拉羅股份公司 Her2-targeting antigen binding molecules comprising 4-1bbl
CA3097741A1 (en) 2018-04-18 2019-10-24 Xencor, Inc. Tim-3 targeted heterodimeric fusion proteins containing il-15/il-15ra fc-fusion proteins and tim-3 antigen binding domains
AR115052A1 (en) 2018-04-18 2020-11-25 Hoffmann La Roche MULTI-SPECIFIC ANTIBODIES AND THE USE OF THEM
AR114789A1 (en) 2018-04-18 2020-10-14 Hoffmann La Roche ANTI-HLA-G ANTIBODIES AND THE USE OF THEM
JP2021521784A (en) 2018-04-18 2021-08-30 ゼンコア インコーポレイテッド PD-1 targeted heterodimer fusion proteins containing IL-15 / IL-15RaFc fusion proteins and PD-1 antigen binding domains and their use
IT201800004853A1 (en) 2018-04-24 2019-10-24 Methods of treating cancer
US20210070871A1 (en) 2018-04-25 2021-03-11 Prometheus Biosciences, Inc. Optimized anti-tl1a antibodies
US11958895B2 (en) 2018-05-03 2024-04-16 University Of Rochester Anti-influenza neuraminidase monoclonal antibodies and uses thereof
CN110464842B (en) 2018-05-11 2022-10-14 信达生物制药(苏州)有限公司 Formulations comprising anti-PCSK 9 antibodies and uses thereof
BR112020022164A2 (en) 2018-05-18 2021-02-02 Bioverativ Therapeutics Inc. methods of treating hemophilia a
WO2019226658A1 (en) 2018-05-21 2019-11-28 Compass Therapeutics Llc Multispecific antigen-binding compositions and methods of use
WO2019226829A1 (en) 2018-05-22 2019-11-28 Beth Israel Deaconess Medical Center, Inc. Antibody therapies for human immunodeficiency virus (hiv)
JOP20190116A1 (en) 2018-05-24 2019-11-24 Janssen Biotech Inc Anti-cd33 antibodies, anti-cd33/anti-cd3 bispecific antibodies and uses thereof
CN112165974A (en) 2018-05-31 2021-01-01 诺华股份有限公司 Hepatitis B antibodies
WO2019232484A1 (en) 2018-06-01 2019-12-05 Compugen Ltd Anti-pvrig/anti-tigit bispecific antibodies and methods of use
KR20210027352A (en) 2018-06-04 2021-03-10 바이오젠 엠에이 인코포레이티드 Anti-VLA-4 antibody with reduced effector function
WO2019235426A1 (en) 2018-06-04 2019-12-12 中外製薬株式会社 Antigen-binding molecule showing changed half-life in cytoplasm
AU2019282264A1 (en) 2018-06-05 2020-11-26 Amgen Inc. Modulating antibody dependent cellular phagocytosis
GB201809746D0 (en) 2018-06-14 2018-08-01 Berlin Chemie Ag Pharmaceutical combinations
US20210363219A1 (en) 2018-06-15 2021-11-25 Alpine Immune Sciences, Inc. Pd-1 variant immunomodulatory proteins and uses thereof
CA3099893A1 (en) 2018-06-18 2019-12-26 Innate Pharma Compositions and methods for treating cancer
US20200030443A1 (en) 2018-06-23 2020-01-30 Genentech, Inc. Methods of treating lung cancer with a pd-1 axis binding antagonist, a platinum agent, and a topoisomerase ii inhibitor
AU2019295279A1 (en) 2018-06-26 2021-01-21 Kagoshima University Antibody binding to cell adhesion molecule 3
CA3104997A1 (en) 2018-06-26 2020-01-02 Kyowa Kirin Co., Ltd. Antibody binding to chondroitin sulfate proteoglycan 5
BR112020026512A2 (en) 2018-07-03 2021-04-06 Bristol-Myers Squibb Company FGF-21 FORMULATIONS
JP2021528988A (en) 2018-07-04 2021-10-28 エフ・ホフマン−ラ・ロシュ・アクチェンゲゼルシャフト New bispecific agonist 4-1BB antigen-binding molecule
BR112020027095A2 (en) 2018-07-09 2021-03-30 Five Prime Therapeutics, Inc. ILT4 BINDING ANTIBODIES
WO2020014306A1 (en) 2018-07-10 2020-01-16 Immunogen, Inc. Met antibodies and immunoconjugates and uses thereof
KR20210031722A (en) 2018-07-11 2021-03-22 파이브 프라임 테라퓨틱스, 인크. Antibodies that bind to VISTA at acidic pH
US20220275043A1 (en) 2018-07-17 2022-09-01 Massachusetts Institute Of Technology Soluble multimeric immunoglobulin-scaffold based fusion proteins and uses thereof
KR20210034622A (en) 2018-07-18 2021-03-30 제넨테크, 인크. Lung cancer treatment method using PD-1 axis binding antagonist, anti-metabolite, and platinum agent
EP3823664A1 (en) 2018-07-19 2021-05-26 Regeneron Pharmaceuticals, Inc. Bispecific anti-bcma x anti-cd3 antibodies and uses thereof
JP7072715B2 (en) 2018-07-20 2022-05-20 サーフィス オンコロジー インコーポレイテッド Anti-CD112R Compositions and Methods
CN112512563A (en) 2018-08-01 2021-03-16 中外制药株式会社 Pharmaceutical composition for treating or preventing C5-related diseases and method for treating or preventing C5-related diseases
KR20210040989A (en) 2018-08-03 2021-04-14 추가이 세이야쿠 가부시키가이샤 Antigen-binding molecule comprising two antigen-binding domains linked to each other
JP2021533753A (en) 2018-08-09 2021-12-09 バイオベラティブ セラピューティクス インコーポレイテッド Nucleic acid molecules and their use for non-viral gene therapy
MA50586A (en) 2018-08-09 2020-09-16 Regeneron Pharma METHODS FOR EVALUATING THE BINDING AFFINITY OF AN ANTIBODY VARIANT TO THE NEONATAL FC RECEPTOR
EA202190451A1 (en) 2018-08-10 2021-07-13 Чугаи Сейяку Кабусики Кайся ANTI-CD137 ANTIGEN-BINDING MOLECULES AND THEIR APPLICATION
TW202021618A (en) 2018-08-17 2020-06-16 美商23與我有限公司 Anti-il1rap antibodies and methods of use thereof
JP7177543B2 (en) 2018-08-21 2022-11-24 エービーエル バイオ インコーポレイテッド Anti-PD-L1/anti-LAG3 bispecific antibodies and uses thereof
TW202031273A (en) 2018-08-31 2020-09-01 美商艾歐凡斯生物治療公司 Treatment of nsclc patients refractory for anti-pd-1 antibody
GB201814281D0 (en) 2018-09-03 2018-10-17 Femtogenix Ltd Cytotoxic agents
SG11202100888WA (en) 2018-09-07 2021-02-25 Pfizer Anti-avb8 antibodies and compositions and uses thereof
WO2020053742A2 (en) 2018-09-10 2020-03-19 Novartis Ag Anti-hla-hbv peptide antibodies
JP2022500371A (en) 2018-09-11 2022-01-04 アムジエン・インコーポレーテツド How to regulate antibody-dependent cellular cytotoxicity
JP2022501332A (en) 2018-09-19 2022-01-06 ジェネンテック, インコーポレイテッド How to treat and diagnose bladder cancer
KR20210089146A (en) 2018-09-19 2021-07-15 알파인 이뮨 사이언시즈, 인코포레이티드 Methods and uses of variant CD80 proteins and related constructs
ES2955032T3 (en) 2018-09-21 2023-11-28 Hoffmann La Roche Diagnostic methods for triple negative breast cancer
CN112513088B (en) 2018-09-26 2023-05-16 江苏恒瑞医药股份有限公司 anti-OX 40 antibodies, antigen binding fragments thereof, and medical uses thereof
EP3856763A1 (en) 2018-09-28 2021-08-04 Massachusetts Institute of Technology Collagen-localized immunomodulatory molecules and methods thereof
TW202028467A (en) 2018-09-28 2020-08-01 日商協和麒麟股份有限公司 Antibody composition
CN112654641A (en) 2018-10-01 2021-04-13 豪夫迈·罗氏有限公司 Bispecific antigen binding molecules with trivalent binding to CD40
WO2020070041A1 (en) 2018-10-01 2020-04-09 F. Hoffmann-La Roche Ag Bispecific antigen binding molecules comprising anti-fap clone 212
AU2019355995A1 (en) 2018-10-05 2021-04-08 Five Prime Therapeutics, Inc. Anti-FGFR2 antibody formulations
WO2020076969A2 (en) 2018-10-10 2020-04-16 Tilos Therapeutics, Inc. Anti-lap antibody variants and uses thereof
MA53862A (en) 2018-10-12 2022-01-19 Xencor Inc FC FUSION PROTEINS OF IL-15/IL-15RALPHA TARGETTING PD-1 AND USES IN COMBINATION THERAPIES INVOLVING THE SAME
WO2020081493A1 (en) 2018-10-16 2020-04-23 Molecular Templates, Inc. Pd-l1 binding proteins
WO2020081767A1 (en) 2018-10-18 2020-04-23 Genentech, Inc. Diagnostic and therapeutic methods for sarcomatoid kidney cancer
PE20211279A1 (en) 2018-10-23 2021-07-19 Dragonfly Therapeutics Inc HETERODIMERIC PROTEINS FUSED WITH FC
US20210395390A1 (en) 2018-10-31 2021-12-23 Bayer Aktiengesellschaft Reversal agents for neutralizing the therapeutic activity of anti-fxia antibodies
JP2022506156A (en) 2018-11-05 2022-01-17 ジェネンテック, インコーポレイテッド Method for producing double-stranded protein in prokaryotic host cell
TW202039831A (en) 2018-11-05 2020-11-01 美商艾歐凡斯生物治療公司 Treatment of nsclc patients refractory for anti-pd-1 antibody
MX2021005751A (en) 2018-11-16 2021-10-01 Memorial Sloan Kettering Cancer Center Antibodies to mucin-16 and methods of use thereof.
AR117091A1 (en) 2018-11-19 2021-07-07 Bristol Myers Squibb Co MONOCLONAL ANTIBODIES ANTAGONISTS AGAINST CD40 AND THEIR USES
TWI779253B (en) 2018-11-27 2022-10-01 大陸商信達生物製藥(蘇州)有限公司 ANTI-IL-23p19 ANTIBODY AND USE THEREOF
WO2020112781A1 (en) 2018-11-28 2020-06-04 Bristol-Myers Squibb Company Antibodies comprising modified heavy constant regions
BR112021010402A2 (en) 2018-11-30 2021-08-24 Abl Bio Inc. Bispecific anti-pd-l1/anti-4-1bb antibodies and their use
JP2022510276A (en) 2018-11-30 2022-01-26 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 CD86 variant immunomodulatory protein and its use
CA3121265A1 (en) 2018-12-05 2020-06-11 Genentech, Inc. Diagnostic methods and compositions for cancer immunotherapy
WO2020117257A1 (en) 2018-12-06 2020-06-11 Genentech, Inc. Combination therapy of diffuse large b-cell lymphoma comprising an anti-cd79b immunoconjugates, an alkylating agent and an anti-cd20 antibody
WO2020118011A1 (en) 2018-12-06 2020-06-11 Alexion Pharmaceuticals, Inc. Anti-alk2 antibodies and uses thereof
WO2020114616A1 (en) 2018-12-07 2020-06-11 Tillotts Pharma Ag Topical treatment of immune checkpoint inhibitor induced diarrhoea, colitis or enterocolitis using antibodies and fragments thereof
EP3894427A1 (en) 2018-12-10 2021-10-20 Genentech, Inc. Photocrosslinking peptides for site specific conjugation to fc-containing proteins
TW202039554A (en) 2018-12-19 2020-11-01 瑞士商諾華公司 Anti-tnf-alpha antibodies
JP2022514290A (en) 2018-12-20 2022-02-10 ジェネンテック, インコーポレイテッド Modified antibody FC and usage
AR117327A1 (en) 2018-12-20 2021-07-28 23Andme Inc ANTI-CD96 ANTIBODIES AND METHODS OF USE OF THEM
WO2020132646A1 (en) 2018-12-20 2020-06-25 Xencor, Inc. Targeted heterodimeric fc fusion proteins containing il-15/il-15ra and nkg2d antigen binding domains
KR20210107721A (en) 2018-12-21 2021-09-01 23앤드미 인코포레이티드 Anti-IL-36 antibodies and methods of use thereof
CN111349162A (en) 2018-12-21 2020-06-30 神州细胞工程有限公司 Humanized anti-PD-1 antibodies and uses thereof
CA3123493A1 (en) 2018-12-21 2020-06-25 F. Hoffmann-La Roche Ag Tumor-targeted agonistic cd28 antigen binding molecules
SG11202106116QA (en) 2018-12-21 2021-07-29 Genentech Inc Methods of producing polypeptides using a cell line resistant to apoptosis
PE20211603A1 (en) 2018-12-21 2021-08-18 Hoffmann La Roche ANTIBODIES THAT JOIN CD3
WO2020127628A1 (en) 2018-12-21 2020-06-25 F. Hoffmann-La Roche Ag Tumor-targeted superagonistic cd28 antigen binding molecules
US11130804B2 (en) 2018-12-21 2021-09-28 Hoffmann-La Roche Inc. Antibody that binds to VEGF and IL-1beta and methods of use
CN113330033A (en) 2018-12-26 2021-08-31 先天制药公司 Leukocyte immunoglobulin-like receptor 2 neutralizing antibodies
KR20210141447A (en) 2018-12-26 2021-11-23 실리오 디벨럽먼트, 인크. Anti-CTLA4 antibodies and methods of use thereof
MX2021007797A (en) 2018-12-28 2021-10-26 Kyowa Kirin Co Ltd BISPECIFIC ANTIBODY BINDING TO TfR.
JP2022516505A (en) 2018-12-28 2022-02-28 スパークス・セラピューティクス・インコーポレイテッド Claudin 18.2 specific binding molecule, composition and method thereof for the treatment of cancer and other diseases.
WO2020141145A1 (en) 2018-12-30 2020-07-09 F. Hoffmann-La Roche Ag Anti-rabbit cd19 antibodies and methods of use
CN113597319A (en) 2019-01-04 2021-11-02 分解治疗有限责任公司 Treatment of xerosis with nuclease fusion proteins
US11639380B2 (en) 2019-01-08 2023-05-02 H. Lundbeck A/S Acute treatment and rapid treatment of headache using anti-CGRP antibodies
MX2021008434A (en) 2019-01-14 2021-09-23 Genentech Inc Methods of treating cancer with a pd-1 axis binding antagonist and an rna vaccine.
EP3914291A2 (en) 2019-01-22 2021-12-01 F. Hoffmann-La Roche AG Immunoglobulin a antibodies and methods of production and use
BR112021014236A2 (en) 2019-01-22 2021-09-28 Innate Pharma ANTIBODY, PHARMACEUTICAL COMPOSITION, KIT AND METHOD TO PREDICT OR EVALUATE EFFECTIVENESS
WO2020154293A1 (en) 2019-01-22 2020-07-30 Bristol-Myers Squibb Company Antibodies against il-7r alpha subunit and uses thereof
WO2020154540A1 (en) 2019-01-23 2020-07-30 Millennium Pharmaceuticals, Inc. Anti-cd38 antibodies
CA3124515A1 (en) 2019-01-23 2020-07-30 Genentech, Inc. Methods of producing multimeric proteins in eukaryotic host cells
EP3915581A4 (en) 2019-01-24 2023-03-22 Chugai Seiyaku Kabushiki Kaisha Novel cancer antigens and antibodies of said antigens
JP2020117502A (en) 2019-01-28 2020-08-06 ファイザー・インク Method of treating signs and symptoms of osteoarthritis
GB201901197D0 (en) 2019-01-29 2019-03-20 Femtogenix Ltd G-A Crosslinking cytotoxic agents
US20220135663A1 (en) 2019-02-18 2022-05-05 Pfizer Inc. Method of treatment of Chronic Low Back Pain
MA55080A (en) 2019-02-26 2022-01-05 Inspirna Inc HIGH AFFINITY ANTI-MERTK ANTIBODIES AND ASSOCIATED USES
MX2021010313A (en) 2019-02-27 2021-09-23 Genentech Inc Dosing for treatment with anti-tigit and anti-cd20 or anti-cd38 antibodies.
BR112021016875A2 (en) 2019-03-01 2022-01-04 Iovance Biotherapeutics Inc Process for expansion of peripheral blood lymphocytes
AU2020238811A1 (en) 2019-03-08 2021-07-22 Genentech, Inc. Methods for detecting and quantifying membrane-associated proteins on extracellular vesicles
MA55305A (en) 2019-03-11 2022-01-19 Janssen Biotech Inc ANTI-V BETA 17/ANTI-CD123 BISPECIFIC ANTIBODIES
US20220135682A1 (en) 2019-03-11 2022-05-05 Jounce Therapeutics, Inc. Anti-ICOS Antibodies for the Treatment of Cancer
CA3130446A1 (en) 2019-03-14 2020-09-17 Genentech, Inc. Treatment of cancer with her2xcd3 bispecific antibodies in combination with anti-her2 mab
MA55529A (en) 2019-04-03 2022-02-09 Genzyme Corp REDUCED FRAGMENTATION ANTI-ALPHA BETA TCR BINDING POLYPEPTIDES
MX2021012160A (en) 2019-04-08 2022-01-06 Biogen Ma Inc Anti-integrin antibodies and uses thereof.
GB2589049C (en) 2019-04-11 2024-02-21 argenx BV Anti-IgE antibodies
WO2020208049A1 (en) 2019-04-12 2020-10-15 F. Hoffmann-La Roche Ag Bispecific antigen binding molecules comprising lipocalin muteins
CA3136816A1 (en) 2019-04-17 2020-10-22 Alpine Immune Sciences, Inc. Methods and uses of variant icos ligand (icosl) fusion proteins
JP2022529344A (en) 2019-04-18 2022-06-21 エイシー イミューン ソシエテ アノニム New molecules for treatment and diagnosis
AU2020258480A1 (en) 2019-04-19 2021-10-21 Genentech, Inc. Anti-mertk antibodies and their methods of use
KR20220002959A (en) 2019-04-23 2022-01-07 이나뜨 파르마 에스.에이. CD73 blocking antibody
WO2020226986A2 (en) 2019-05-03 2020-11-12 Genentech, Inc. Methods of treating cancer with an anti-pd-l1 antibody
BR112021022089A2 (en) 2019-05-08 2022-02-08 Janssen Biotech Inc Materials and methods for modulating t-cell-mediated immunity
AU2020275415A1 (en) 2019-05-14 2021-11-25 Genentech, Inc. Methods of using anti-CD79B immunoconjugates to treat follicular lymphoma
JPWO2020230901A1 (en) 2019-05-15 2020-11-19
KR20220008820A (en) 2019-05-15 2022-01-21 쿄와 기린 가부시키가이샤 Bispecific antibody that binds to CD40 and FAP
AU2020278907A1 (en) 2019-05-23 2022-01-20 Ac Immune Sa Anti-TDP-43 binding molecules and uses thereof
MX2021014756A (en) 2019-06-07 2022-01-18 Argenx Bvba PHARMACEUTICAL FORMULATIONS OF FcRn INHIBITORS SUITABLE FOR SUBCUTANEOUS ADMINISTRATION.
US20220241413A1 (en) 2019-06-10 2022-08-04 Takeda Pharmaceutical Company Limited Combination therapies using cd-38 antibodies
AU2020291300A1 (en) 2019-06-10 2022-01-06 Chugai Seiyaku Kabushiki Kaisha Anti-T cell antigen-binding molecule to be used in combination with cytokine inhibitor
CR20210607A (en) 2019-06-12 2022-01-21 Novartis Ag Natriuretic peptide receptor 1 antibodies and methods of use
EP3986918A1 (en) 2019-06-18 2022-04-27 Bayer Aktiengesellschaft Adrenomedullin-analogues for long-term stabilization and their use
CA3141378A1 (en) 2019-06-26 2020-12-30 F. Hoffmann-La Roche Ag Fusion of an antibody binding cea and 4-1bbl
JP2022538974A (en) 2019-06-26 2022-09-07 マサチューセッツ インスチテュート オブ テクノロジー Immunomodulatory fusion protein-metal hydroxide complexes and methods thereof
CN114531878A (en) 2019-06-27 2022-05-24 豪夫迈·罗氏有限公司 Novel ICOS antibodies and tumor-targeted antigen-binding molecules comprising same
TW202115115A (en) 2019-07-02 2021-04-16 瑞士商赫孚孟拉羅股份公司 Immunoconjugates
CA3139250A1 (en) 2019-07-10 2021-01-14 Naoki Kimura Claudin-6 binding molecules and uses thereof
AR119382A1 (en) 2019-07-12 2021-12-15 Hoffmann La Roche PRE-TARGETING ANTIBODIES AND METHODS OF USE
JP2022540904A (en) 2019-07-15 2022-09-20 ブリストル-マイヤーズ スクイブ カンパニー Antibodies against human TREM-1 and uses thereof
WO2021011678A1 (en) 2019-07-15 2021-01-21 Bristol-Myers Squibb Company Anti-trem-1 antibodies and uses thereof
AR119393A1 (en) 2019-07-15 2021-12-15 Hoffmann La Roche ANTIBODIES THAT BIND NKG2D
WO2021013065A1 (en) 2019-07-19 2021-01-28 神州细胞工程有限公司 Humanized anti-vegf fab antibody fragment and use thereof
BR112022001017A2 (en) 2019-07-19 2022-04-12 Sinocelltech Ltd Humanized anti-vegf monoclonal antibody
KR20220071179A (en) 2019-07-26 2022-05-31 사이노셀테크 엘티디. Humanized anti-IL17A antibodies and uses thereof
CN112300279A (en) 2019-07-26 2021-02-02 上海复宏汉霖生物技术股份有限公司 Methods and compositions directed to anti-CD 73 antibodies and variants
MX2022001146A (en) 2019-07-29 2022-03-17 Compugen Ltd Anti-pvrig antibodies formulations and uses thereof.
JP2022543551A (en) 2019-07-31 2022-10-13 エフ・ホフマン-ラ・ロシュ・アクチェンゲゼルシャフト Antibody that binds to GPRC5D
TW202120124A (en) 2019-07-31 2021-06-01 瑞士商赫孚孟拉羅股份公司 Dosage and administration regimen for the treatment or prevention of c5-related diseases by the use of the anti-c5 antibody crovalimab
CR20220019A (en) 2019-07-31 2022-02-11 Hoffmann La Roche Antibodies binding to gprc5d
MX2022001154A (en) 2019-07-31 2022-02-22 Hoffmann La Roche Dosage and administration regimen for the treatment or prevention of c5-related diseases by the use of the anti-c5 antibody crovalimab.
WO2021021991A1 (en) 2019-08-01 2021-02-04 Vaccinex,Inc. Combined inhibition of semaphorin-4d and tgfb and compositions therefor
KR102509648B1 (en) 2019-08-06 2023-03-15 아프리노이아 테라퓨틱스 리미티드 Antibodies that bind to pathological Tau species and uses thereof
EP4010023A1 (en) 2019-08-07 2022-06-15 Rakuten Medical, Inc. Cetuximab-ir700 conjugate compositions
MX2022001882A (en) 2019-08-12 2022-05-30 Aptevo Res & Development Llc 4-1bb and ox40 binding proteins and related compositions and methods, antibodies against 4-1bb, antibodies against ox40.
KR20220053007A (en) 2019-08-30 2022-04-28 아게누스 인코포레이티드 Anti-CD96 antibodies and methods of use thereof
CN114340675A (en) 2019-09-12 2022-04-12 豪夫迈·罗氏有限公司 Compositions and methods for treating lupus nephritis
PE20221906A1 (en) 2019-09-18 2022-12-23 Genentech Inc ANTI-KLK7 ANTIBODIES, ANTI-KLK5 ANTIBODIES, ANTI-KLK5/KLK7 MULTISPECIFIC ANTIBODIES AND METHODS OF USE
PE20221416A1 (en) 2019-09-18 2022-09-20 Novartis Ag NKG2D FUSION PROTEINS AND THEIR USES
CA3150265A1 (en) 2019-09-18 2021-03-25 Sara MAJOCCHI Bispecific antibodies against ceacam5 and cd3
MX2022003204A (en) 2019-09-19 2022-04-18 Bristol Myers Squibb Co Antibodies binding to vista at acidic ph.
CR20220149A (en) 2019-09-20 2022-05-23 Genentech Inc Dosing for anti-tryptase antibodies
TWI764291B (en) 2019-09-24 2022-05-11 財團法人工業技術研究院 Anti-tigit antibodies and methods of use
WO2021059075A1 (en) 2019-09-27 2021-04-01 Janssen Biotech, Inc. Anti-ceacam antibodies and uses thereof
JP2022548978A (en) 2019-09-27 2022-11-22 ジェネンテック, インコーポレイテッド Dosing for Treatment with Drugs Anti-TIGIT and Anti-PD-L1 Antagonist Antibodies
EP4038182A1 (en) 2019-09-30 2022-08-10 Bioverativ Therapeutics Inc. Lentiviral vector formulations
US20240058466A1 (en) 2019-10-04 2024-02-22 TAE Life Sciences Antibody Compositions Comprising Fc Mutations and Site-Specific Conjugation Properties For Use In Treating Cancer, Immunological Disorders, and Methods Thereof
WO2021072244A1 (en) 2019-10-11 2021-04-15 Beth Israel Deaconess Medical Center, Inc. Anti-tn antibodies and uses thereof
TW202128757A (en) 2019-10-11 2021-08-01 美商建南德克公司 Pd-1 targeted il-15/il-15ralpha fc fusion proteins with improved properties
JP7413519B2 (en) 2019-10-18 2024-01-15 ジェネンテック, インコーポレイテッド Methods of using anti-CD79B immunoconjugates to treat diffuse large B-cell lymphoma
US11459389B2 (en) 2019-10-24 2022-10-04 Massachusetts Institute Of Technology Monoclonal antibodies that bind human CD161
IL292419A (en) 2019-10-24 2022-06-01 Prometheus Biosciences Inc Humanized antibodies to tnf-like ligand 1a (tl1a) and uses thereof
MX2022005132A (en) 2019-11-04 2022-08-15 Compugen Ltd Combination therapy with anti-pvrig antibodies formulations and anti-pd-1 antibodies.
US20220378909A1 (en) 2019-11-05 2022-12-01 Jounce Therapeutics, Inc. Methods of Treating Cancer with Anti-PD-1 Antibodies
IL292458A (en) 2019-11-06 2022-06-01 Genentech Inc Diagnostic and therapeutic methods for treatment of hematologic cancers
MX2022006676A (en) 2019-12-04 2022-07-05 Ac Immune Sa Novel molecules for therapy and diagnosis.
US20230057899A1 (en) 2019-12-05 2023-02-23 Compugen Ltd. Anti-pvrig and anti-tigit antibodies for enhanced nk-cell based tumor killing
KR20220122656A (en) 2019-12-06 2022-09-02 주노 쎄러퓨티크스 인코퍼레이티드 Anti-idiotypic Antibodies and Related Compositions and Methods Against GPDAC5D-Targeting Binding Domain
CA3163897A1 (en) 2019-12-06 2021-06-10 Juno Therapeutics, Inc. Anti-idiotypic antibodies to bcma-targeted binding domains and related compositions and methods
AU2020403145A1 (en) 2019-12-13 2022-07-07 Genentech, Inc. Anti-Ly6G6D antibodies and methods of use
US20210230278A1 (en) 2019-12-18 2021-07-29 Hoffmann-La Roche Inc. Antibodies binding to HLA-A2/MAGE-A4
JP2023506956A (en) 2019-12-20 2023-02-20 エフ・ホフマン-ラ・ロシュ・アクチェンゲゼルシャフト IL-37 fusion proteins and uses thereof
AR120898A1 (en) 2019-12-26 2022-03-30 Univ Osaka AGENT TO TREAT OR PREVENT ACUTE NEUROMYELITIS OPTICA
AU2019479791A1 (en) 2019-12-27 2022-07-14 Chugai Seiyaku Kabushiki Kaisha Anti-CTLA-4 antibody and use thereof
CN113045655A (en) 2019-12-27 2021-06-29 高诚生物医药(香港)有限公司 anti-OX 40 antibodies and uses thereof
AU2021205877A1 (en) 2020-01-06 2022-06-09 Vaccinex, Inc. Anti-CCR8 antibodies and uses thereof
CN115052663A (en) 2020-01-08 2022-09-13 辛瑟斯治疗股份有限公司 ALK5 inhibitor conjugates and uses thereof
US20210236596A1 (en) 2020-01-08 2021-08-05 argenx BV Methods for treating pemphigus disorders
KR20220107251A (en) 2020-01-09 2022-08-02 에프. 호프만-라 로슈 아게 Novel 4-1BBL Trimer-Containing Antigen Binding Molecules
CN110818795B (en) 2020-01-10 2020-04-24 上海复宏汉霖生物技术股份有限公司 anti-TIGIT antibodies and methods of use
WO2021145432A1 (en) 2020-01-15 2021-07-22 国立大学法人大阪大学 Agent for prevention or treatment of diabetic autonomic neuropathy
US20230090965A1 (en) 2020-01-15 2023-03-23 Osaka University Prophylactic or therapeutic agent for dementia
KR20220131293A (en) 2020-01-24 2022-09-27 화이자 인코포레이티드 Anti-E-Selectin Antibodies, Compositions and Methods of Use
WO2022050954A1 (en) 2020-09-04 2022-03-10 Genentech, Inc. Dosing for treatment with anti-tigit and anti-pd-l1 antagonist antibodies
WO2021194481A1 (en) 2020-03-24 2021-09-30 Genentech, Inc. Dosing for treatment with anti-tigit and anti-pd-l1 antagonist antibodies
EP4096708A1 (en) 2020-01-31 2022-12-07 Genentech, Inc. Methods of inducing neoepitope-specific t cells with a pd-1 axis binding antagonist and an rna vaccine
US20230087600A1 (en) 2020-02-06 2023-03-23 Bristol-Myers Squibb Company Il-10 and uses thereof
TW202144395A (en) 2020-02-12 2021-12-01 日商中外製藥股份有限公司 Anti-CD137 antigen-binding molecule for use in cancer treatment
WO2021163064A2 (en) 2020-02-14 2021-08-19 Jounce Therapeutics, Inc. Antibodies and fusion proteins that bind to ccr8 and uses thereof
TW202140561A (en) 2020-02-14 2021-11-01 日商協和麒麟股份有限公司 Bispecific antibody that binds to CD3
MX2022010604A (en) 2020-02-27 2022-09-09 Novartis Ag Methods of making chimeric antigen receptor-expressing cells.
CA3169910A1 (en) 2020-02-28 2021-09-02 Shanghai Henlius Biotech, Inc. Anti-cd137 constructs, multispecific antibody and uses thereof
JP2023516952A (en) 2020-02-28 2023-04-21 ジェンザイム・コーポレーション Modified binding polypeptides for optimized drug conjugation
CN115066440A (en) 2020-02-28 2022-09-16 上海复宏汉霖生物技术股份有限公司 anti-CD 137 constructs and uses thereof
AU2021232158A1 (en) 2020-03-06 2022-09-29 Ona Therapeutics, S.L. Anti-CD36 antibodies and their use to treat cancer
KR20220153615A (en) 2020-03-12 2022-11-18 도레이 카부시키가이샤 Medicines for the treatment and/or prevention of cancer
BR112022018157A2 (en) 2020-03-12 2022-10-25 Toray Industries DRUG FOR THE TREATMENT AND/OR PREVENTION OF CANCER, AGENTS THAT INCREASE THE EFFICACY OF A DRUG AND METHOD FOR THE TREATMENT AND/OR PREVENTION OF CANCER
KR20220153621A (en) 2020-03-12 2022-11-18 도레이 카부시키가이샤 Medicines for the treatment and/or prevention of cancer
US20230165957A1 (en) 2020-03-12 2023-06-01 Toray Industries, Inc. Medicament for treatment and/or prevention of cancer
WO2021182574A1 (en) 2020-03-12 2021-09-16 東レ株式会社 Medicament for treatment and/or prevention of cancer
IL296256A (en) 2020-03-13 2022-11-01 Genentech Inc Anti-interleukin-33 antibodies and uses thereof
WO2021180205A1 (en) 2020-03-13 2021-09-16 江苏恒瑞医药股份有限公司 Pvrig binding protein and its medical uses
CN117510630A (en) 2020-03-19 2024-02-06 基因泰克公司 Isotype selective anti-TGF-beta antibodies and methods of use
WO2021194913A1 (en) 2020-03-24 2021-09-30 Genentech, Inc. Tie2-binding agents and methods of use
EP4127153A2 (en) 2020-03-26 2023-02-08 Genentech, Inc. Modified mammalian cells having reduced host cell proteins
CN115397850A (en) 2020-03-30 2022-11-25 豪夫迈·罗氏有限公司 Antibodies that bind to VEGF and PDGF-B and methods of use thereof
AR121706A1 (en) 2020-04-01 2022-06-29 Hoffmann La Roche OX40 AND FAP-TARGETED BSPECIFIC ANTIGEN-BINDING MOLECULES
WO2021201236A1 (en) 2020-04-01 2021-10-07 協和キリン株式会社 Antibody composition
CA3170570A1 (en) 2020-04-01 2021-10-07 James J. KOBIE Monoclonal antibodies against the hemagglutinin (ha) and neuraminidase (na) of influenza h3n2 viruses
WO2021202959A1 (en) 2020-04-03 2021-10-07 Genentech, Inc. Therapeutic and diagnostic methods for cancer
WO2021207449A1 (en) 2020-04-09 2021-10-14 Merck Sharp & Dohme Corp. Affinity matured anti-lap antibodies and uses thereof
TW202204622A (en) 2020-04-09 2022-02-01 大陸商蘇州艾博生物科技有限公司 Nucleic acid vaccines for coronavirus
WO2021207662A1 (en) 2020-04-10 2021-10-14 Genentech, Inc. Use of il-22fc for the treatment or prevention of pneumonia, acute respiratory distress syndrome, or cytokine release syndrome
US20230192795A1 (en) 2020-04-15 2023-06-22 Hoffmann-La Roche Inc. Immunoconjugates
AU2021261257A1 (en) 2020-04-20 2022-12-22 Genzyme Corporation Humanized anti-complement factor Bb antibodies and uses thereof
IL297313A (en) 2020-04-20 2022-12-01 Jounce Therapeutics Inc Compositions and methods for vaccination and the treatment of infectious diseases
IL297541A (en) 2020-04-24 2022-12-01 Genentech Inc Methods of using anti-cd79b immunoconjugates
CN115335411A (en) 2020-04-28 2022-11-11 神州细胞工程有限公司 TGF beta R2 extracellular region truncated molecule, fusion protein of TGF beta R2 extracellular region truncated molecule and anti-EGFR antibody and anti-tumor application of fusion protein
SG11202112792WA (en) 2020-04-28 2021-12-30 Univ Rockefeller Neutralizing anti-sars-cov-2 antibodies and methods of use thereof
WO2021222167A1 (en) 2020-04-28 2021-11-04 Genentech, Inc. Methods and compositions for non-small cell lung cancer immunotherapy
EP4146283A1 (en) 2020-05-03 2023-03-15 Levena (Suzhou) Biopharma Co., Ltd. Antibody-drug conjugates (adcs) comprising an anti-trop-2 antibody, compositions comprising such adcs, as well as methods of making and using the same
WO2021226551A1 (en) 2020-05-08 2021-11-11 Alpine Immune Sciences, Inc. April and baff inhibitory immunomodulatory proteins and methods of use thereof
CN111995681B (en) 2020-05-09 2022-03-08 华博生物医药技术(上海)有限公司 anti-TIGIT antibody, preparation method and application thereof
IL298046A (en) 2020-05-11 2023-01-01 Janssen Biotech Inc Methods for treating multiple myeloma
WO2021231732A1 (en) 2020-05-15 2021-11-18 Bristol-Myers Squibb Company Antibodies to garp
WO2021233834A1 (en) 2020-05-17 2021-11-25 Astrazeneca Uk Limited Sars-cov-2 antibodies and methods of selecting and using the same
GB2595299B (en) 2020-05-21 2022-08-03 Mabsolve Ltd Modified immunoglobulin FC regions
KR20230015997A (en) * 2020-05-26 2023-01-31 베링거 인겔하임 인터내셔날 게엠베하 Anti-PD-1 antibody
EP4157881A1 (en) 2020-05-27 2023-04-05 Staidson (Beijing) Biopharmaceuticals Co., Ltd. Antibodies specifically recognizing nerve growth factor and uses thereof
AU2021281417A1 (en) 2020-05-29 2022-12-08 23Andme, Inc. Anti-CD200R1 antibodies and methods of use thereof
CN116529260A (en) 2020-06-02 2023-08-01 当康生物技术有限责任公司 anti-CD 93 constructs and uses thereof
IL298735A (en) 2020-06-02 2023-02-01 Dynamicure Biotechnology Llc Anti-cd93 constructs and uses thereof
AR122569A1 (en) 2020-06-08 2022-09-21 Hoffmann La Roche ANTI-HBV ANTIBODIES AND METHODS OF USE
WO2021252977A1 (en) 2020-06-12 2021-12-16 Genentech, Inc. Methods and compositions for cancer immunotherapy
KR20230025691A (en) 2020-06-16 2023-02-22 제넨테크, 인크. Methods and compositions for treating triple negative breast cancer
KR20230024368A (en) 2020-06-18 2023-02-20 제넨테크, 인크. Treatment with anti-TIGIT antibodies and PD-1 axis binding antagonists
US20230235056A1 (en) 2020-06-19 2023-07-27 Chugai Seiyaku Kabushiki Kaisha Anti-t cell antigen-binding molecule for use in combination with angiogenesis inhibitor
IL296429A (en) 2020-06-19 2022-11-01 Hoffmann La Roche Protease-activated t cell bispecific antibodies
TW202216766A (en) 2020-06-19 2022-05-01 瑞士商赫孚孟拉羅股份公司 Antibodies binding to cd3
WO2021255146A1 (en) 2020-06-19 2021-12-23 F. Hoffmann-La Roche Ag Antibodies binding to cd3 and cea
AU2021291011A1 (en) 2020-06-19 2023-01-05 F. Hoffmann-La Roche Ag Antibodies binding to CD3 and CD19
WO2021255143A1 (en) 2020-06-19 2021-12-23 F. Hoffmann-La Roche Ag Antibodies binding to cd3 and folr1
CR20220629A (en) 2020-06-19 2023-02-17 Hoffmann La Roche Immune activating fc domain binding molecules
US20230312741A1 (en) 2020-06-22 2023-10-05 Innovent Biologics (Suzhou) Co., Ltd. Anti-cd73 antibody and uses thereof
WO2021259880A1 (en) 2020-06-22 2021-12-30 Almirall, S.A. Anti-il-36 antibodies and methods of use thereof
EP4168448A1 (en) 2020-06-23 2023-04-26 F. Hoffmann-La Roche AG Agonistic cd28 antigen binding molecules targeting her2
EP4169949A1 (en) 2020-06-23 2023-04-26 Jiangsu Kanion Pharmaceutical Co., Ltd. Anti-cd38 antibody and use thereof
JP2023533217A (en) 2020-06-24 2023-08-02 ジェネンテック, インコーポレイテッド Apoptosis resistant cell line
KR20230135556A (en) 2020-06-25 2023-09-25 백시넥스 인코포레이티드 Use of semaphorin-4D binding molecules for the treatment of Rett syndrome
EP4172203A1 (en) 2020-06-25 2023-05-03 F. Hoffmann-La Roche AG Anti-cd3/anti-cd28 bispecific antigen binding molecules
CA3165342A1 (en) 2020-06-29 2022-01-06 James Arthur Posada Treatment of sjogren's syndrome with nuclease fusion proteins
MX2023000339A (en) 2020-07-10 2023-02-09 Hoffmann La Roche Antibodies which bind to cancer cells and target radionuclides to said cells.
PE20231300A1 (en) 2020-07-17 2023-08-24 Genentech Inc ANTI-NOTCH2 ANTIBODIES AND METHODS OF USE
CN116323668A (en) 2020-07-17 2023-06-23 辉瑞公司 Therapeutic antibodies and uses thereof
WO2022027037A1 (en) 2020-07-27 2022-02-03 Single Cell Technology, Inc. Anti-sars corona virus-2 spike protein antibodies
GB2597532A (en) 2020-07-28 2022-02-02 Femtogenix Ltd Cytotoxic compounds
CA3190328A1 (en) 2020-07-29 2022-02-03 Dynamicure Biotechnology Llc Anti-cd93 constructs and uses thereof
JP2023536602A (en) 2020-08-03 2023-08-28 ジェネンテック, インコーポレイテッド Diagnostic and therapeutic methods for lymphoma
BR112023001740A2 (en) 2020-08-03 2023-02-28 Hoffmann La Roche ANTIGEN-BINDING RECEPTORS, TRANSDUCED T-CELL, ISOLATED POLYNUCLEOTIDE, VECTOR, KIT, METHOD FOR TREAT A DISEASE IN A SUBJECT AND INDUCE LYSIS OF A TARGET CELL AND USE OF ANTIGEN-BINDING RECEPTOR
JP2023536326A (en) 2020-08-05 2023-08-24 ジュノー セラピューティクス インコーポレイテッド Anti-idiotypic antibodies against ROR1 target binding domains and related compositions and methods
JP2023536904A (en) 2020-08-06 2023-08-30 バイオベラティブ・ユーエスエイ・インコーポレイテッド Inflammatory Cytokines and Fatigue in Subjects With Complement-Mediated Disease
WO2022034044A1 (en) 2020-08-10 2022-02-17 Astrazeneca Uk Limited Sars-cov-2 antibodies for treatment and prevention of covid-19
KR20230050378A (en) 2020-08-13 2023-04-14 이나뜨 파르마 에스.에이. Methods of Treating Cancer Using Anti-CD73 Antibodies
AR123254A1 (en) 2020-08-14 2022-11-16 Ac Immune Sa HUMANIZED ANTI-TDP-43 BINDING MOLECULES AND THEIR USES
BR112023003036A2 (en) * 2020-08-19 2023-04-25 Astellas Pharma Inc HUMAN NON NATURALLY OCCURRING MODIFIED FC REGION SPECIFICALLY BINDING TO NON NATURALLY OCCURRING MODIFIED FC RECEIVER
WO2022040586A2 (en) 2020-08-21 2022-02-24 Novartis Ag Compositions and methods for in vivo generation of car expressing cells
EP4204448A2 (en) 2020-08-27 2023-07-05 cureab GmbH Anti-golph2 antibodies for macrophage and dendritic cell differentiation
CA3192344A1 (en) 2020-08-28 2022-03-03 Genentech, Inc. Crispr/cas9 multiplex knockout of host cell proteins
EP4204447A1 (en) 2020-08-28 2023-07-05 Sana Biotechnology, Inc. Modified anti-viral binding agents
WO2022044248A1 (en) 2020-08-28 2022-03-03 中外製薬株式会社 Heterodimer fc polypeptide
KR20230061458A (en) 2020-09-04 2023-05-08 에프. 호프만-라 로슈 아게 Antibodies that bind to VEGF-A and ANG2 and methods of use
MX2023002948A (en) 2020-09-11 2023-05-22 Janssen Biotech Inc Methods and compositions for modulating beta chain mediated immunity.
WO2022053715A1 (en) 2020-09-14 2022-03-17 Ichnos Sciences SA Antibodies that bind to il1rap and uses thereof
EP4222172A1 (en) 2020-09-30 2023-08-09 Compugen Ltd. Combination therapy with anti-pvrig antibodies formulations, anti-tigit antibodies, and anti-pd-1 antibodies
CN116406291A (en) 2020-10-05 2023-07-07 基因泰克公司 Administration of treatment with anti-FCRH 5/anti-CD 3 bispecific antibodies
WO2022076606A1 (en) 2020-10-06 2022-04-14 Iovance Biotherapeutics, Inc. Treatment of nsclc patients with tumor infiltrating lymphocyte therapies
JP2023546359A (en) 2020-10-06 2023-11-02 アイオバンス バイオセラピューティクス,インコーポレイテッド Treatment of NSCLC patients with tumor-infiltrating lymphocyte therapy
AR123767A1 (en) 2020-10-13 2023-01-11 Janssen Biotech Inc IMMUNITY MEDIATED BY T CELLS BY BIOENGINEERING, MATERIALS AND OTHER METHODS TO MODULATE THE CLUSTER OF DIFFERENTIATION IV AND/OR VIII
EP4229082A1 (en) 2020-10-16 2023-08-23 AC Immune SA Antibodies binding to alpha-synuclein for therapy and diagnosis
AR123855A1 (en) 2020-10-20 2023-01-18 Genentech Inc PEG-CONJUGATED ANTI-MERTK ANTIBODIES AND METHODS OF USE
WO2022090801A2 (en) 2020-10-26 2022-05-05 Compugen Ltd. Pvrl2 and/or pvrig as biomarkers for treatment
CN116507640A (en) 2020-10-28 2023-07-28 豪夫迈·罗氏有限公司 Improved antigen binding receptors
WO2022093981A1 (en) 2020-10-28 2022-05-05 Genentech, Inc. Combination therapy comprising ptpn22 inhibitors and pd-l1 binding antagonists
CA3196539A1 (en) 2020-11-04 2022-05-12 Chi-Chung Li Dosing for treatment with anti-cd20/anti-cd3 bispecific antibodies
CA3196076A1 (en) 2020-11-04 2022-05-12 Chi-Chung Li Subcutaneous dosing of anti-cd20/anti-cd3 bispecific antibodies
AU2021374594A1 (en) 2020-11-04 2023-06-01 Genentech, Inc. Dosing for treatment with anti-cd20/anti-cd3 bispecific antibodies and anti-cd79b antibody drug conjugates
WO2022098870A1 (en) 2020-11-04 2022-05-12 The Rockefeller University Neutralizing anti-sars-cov-2 antibodies
CN116472288A (en) 2020-11-06 2023-07-21 诺华股份有限公司 Antibody Fc variants
CN117916261A (en) 2020-11-16 2024-04-19 豪夫迈·罗氏有限公司 Combination therapy with FAP-targeted CD40 agonists
CN114539418A (en) 2020-11-26 2022-05-27 上海华奥泰生物药业股份有限公司 Bispecific antibodies and uses thereof
EP4255574A1 (en) 2020-12-01 2023-10-11 Aptevo Research and Development LLC Heterodimeric psma and cd3-binding bispecific antibodies
CA3199648A1 (en) 2020-12-02 2022-06-09 Gerben BOUMA Il-7 binding proteins and their use in medical therapy
MX2023006650A (en) 2020-12-07 2023-06-21 UCB Biopharma SRL Multi-specific antibodies and antibody combinations.
CN116802211A (en) 2020-12-07 2023-09-22 Ucb生物制药有限责任公司 Antibodies against interleukin-22
TW202241468A (en) 2020-12-11 2022-11-01 美商艾歐凡斯生物治療公司 Treatment of cancer patients with tumor infiltrating lymphocyte therapies in combination with braf inhibitors and/or mek inhibitors
WO2022130182A1 (en) 2020-12-14 2022-06-23 Novartis Ag Reversal binding agents for anti-natriuretic peptide receptor 1 (npr1) antibodies and uses thereof
JP2023554395A (en) 2020-12-17 2023-12-27 アイオバンス バイオセラピューティクス,インコーポレイテッド Treatment with tumor-infiltrating lymphocyte therapy in combination with CTLA-4 and PD-1 inhibitors
EP4262827A1 (en) 2020-12-17 2023-10-25 Iovance Biotherapeutics, Inc. Treatment of cancers with tumor infiltrating lymphocytes
JP7326584B2 (en) 2020-12-17 2023-08-15 エフ・ホフマン-ラ・ロシュ・アクチェンゲゼルシャフト Anti-HLA-G antibody and use thereof
US20240059763A1 (en) 2020-12-18 2024-02-22 Zhuhai Trinomab Pharmaceutical Co., Ltd. Respiratory syncytial virus-specific binding molecule
WO2022140797A1 (en) 2020-12-23 2022-06-30 Immunowake Inc. Immunocytokines and uses thereof
KR20230124959A (en) 2020-12-23 2023-08-28 이노벤트 바이오로직스 (쑤저우) 컴퍼니, 리미티드 Anti-B7-H3 Antibodies and Uses Thereof
JP2024501845A (en) 2020-12-31 2024-01-16 アイオバンス バイオセラピューティクス,インコーポレイテッド Devices and processes for automated production of tumor-infiltrating lymphocytes
CN116829598A (en) 2021-01-06 2023-09-29 豪夫迈·罗氏有限公司 Combination therapy with PD1-LAG3 bispecific antibodies and CD20T cell bispecific antibodies
WO2022148853A1 (en) 2021-01-11 2022-07-14 F. Hoffmann-La Roche Ag Immunoconjugates
EP4277705A1 (en) 2021-01-12 2023-11-22 F. Hoffmann-La Roche AG Split antibodies which bind to cancer cells and target radionuclides to said cells
CA3204291A1 (en) 2021-01-13 2022-07-21 F. Hoffmann-La Roche Ag Combination therapy
EP4277926A1 (en) 2021-01-15 2023-11-22 The Rockefeller University Neutralizing anti-sars-cov-2 antibodies
US20240082397A1 (en) 2021-01-28 2024-03-14 Compugen Ltd. Anti-pvrig antibodies formulations and uses thereof
US20240076373A1 (en) 2021-01-28 2024-03-07 Compugen Ltd. Combination therapy with anti-pvrig antibodies formulations and anti-pd-1 antibodies
US20240059789A1 (en) 2021-01-28 2024-02-22 Janssen Biotech, Inc. Psma binding proteins and uses thereof
EP4284919A1 (en) 2021-01-29 2023-12-06 Iovance Biotherapeutics, Inc. Methods of making modified tumor infiltrating lymphocytes and their use in adoptive cell therapy
WO2022169872A1 (en) 2021-02-03 2022-08-11 Genentech, Inc. Multispecific binding protein degrader platform and methods of use
AU2022218137A1 (en) 2021-02-03 2023-08-24 Mozart Therapeutics, Inc. Binding agents and methods of using the same
BR112023016121A2 (en) 2021-02-16 2023-11-28 Janssen Pharmaceutica Nv TRIESPECIFIC ANTIBODY TARGETING BCMA, GPRC5D AND CD3
AR124914A1 (en) 2021-02-18 2023-05-17 Mitsubishi Tanabe Pharma Corp NEW ANTI-PAD4 ANTIBODY
WO2022187270A1 (en) 2021-03-01 2022-09-09 Xilio Development, Inc. Combination of ctla4 and pd1/pdl1 antibodies for treating cancer
CA3209364A1 (en) 2021-03-01 2022-09-09 Jennifer O'neil Combination of masked ctla4 and pd1/pdl1 antibodies for treating cancer
WO2022184082A1 (en) 2021-03-03 2022-09-09 Sorrento Therapeutics, Inc. Antibody-drug conjugates comprising an anti-bcma antibody
JP2024509191A (en) 2021-03-05 2024-02-29 ダイナミキュア バイオテクノロジー エルエルシー Anti-VISTA constructs and their uses
CA3210755A1 (en) 2021-03-05 2022-09-09 Kenneth ONIMUS Tumor storage and cell culture compositions
EP4294927A2 (en) 2021-03-10 2023-12-27 Immunowake Inc. Immunomodulatory molecules and uses thereof
WO2022192647A1 (en) 2021-03-12 2022-09-15 Genentech, Inc. Anti-klk7 antibodies, anti-klk5 antibodies, multispecific anti-klk5/klk7 antibodies, and methods of use
WO2022198192A1 (en) 2021-03-15 2022-09-22 Genentech, Inc. Compositions and methods of treating lupus nephritis
CN117203232A (en) * 2021-03-17 2023-12-08 瑞塞普托斯有限责任公司 Methods of treating atopic dermatitis with anti-IL-13 antibodies
CA3213636A1 (en) 2021-03-18 2022-09-22 Seagen Inc. Selective drug release from internalized conjugates of biologically active compounds
WO2022195504A1 (en) 2021-03-19 2022-09-22 Pfizer Inc. Method of treating osteoarthritis pain with an anti ngf antibody
WO2022197877A1 (en) 2021-03-19 2022-09-22 Genentech, Inc. Methods and compositions for time delayed bio-orthogonal release of cytotoxic agents
WO2022198141A1 (en) 2021-03-19 2022-09-22 Iovance Biotherapeutics, Inc. Methods for tumor infiltrating lymphocyte (til) expansion related to cd39/cd69 selection and gene knockout in tils
CA3208944A1 (en) 2021-03-22 2022-09-29 Edith NALBANDIAN Method to assess potency of viral vector particles
CA3210581A1 (en) 2021-03-22 2022-09-29 Neil HAIG Methods of determining potency of a therapeutic cell composition
EP4313122A1 (en) 2021-03-23 2024-02-07 Iovance Biotherapeutics, Inc. Cish gene editing of tumor infiltrating lymphocytes and uses of same in immunotherapy
CA3214594A1 (en) 2021-03-24 2022-09-29 Janssen Biotech, Inc. Trispecific antibody targeting cd79b, cd20, and cd3
EP4314049A1 (en) 2021-03-25 2024-02-07 Dynamicure Biotechnology LLC Anti-igfbp7 constructs and uses thereof
EP4314253A2 (en) 2021-03-25 2024-02-07 Iovance Biotherapeutics, Inc. Methods and compositions for t-cell coculture potency assays and use with cell therapy products
BR112023019138A2 (en) 2021-03-26 2023-10-24 Innate Pharma MULTI-SPECIFIC PROTEIN, PHARMACEUTICAL COMPOSITION, RECOMBINANT CELL, METHOD OF PREPARING AN NK CELL COMPOSITION, NK CELL COMPOSITION, USE OF A PROTEIN OR COMPOSITION, METHODS AND USE
JP2024513837A (en) 2021-03-31 2024-03-27 バイオベラティブ・ユーエスエイ・インコーポレイテッド Reducing surgery-related hemolysis in patients with cold agglutinin disease
TW202304994A (en) 2021-04-02 2023-02-01 美商泰尼歐生物公司 Agonistic anti-il-2r antibodies and methods of use
EP4314068A1 (en) 2021-04-02 2024-02-07 The Regents Of The University Of California Antibodies against cleaved cdcp1 and uses thereof
CN117279664A (en) 2021-04-10 2023-12-22 普方生物制药美国公司 FOLR1 binding agents, conjugates thereof, and methods of use thereof
BR112023021665A2 (en) 2021-04-19 2023-12-19 Iovance Biotherapeutics Inc METHOD FOR TREATING A CANCER, AND, COMPOSITION
JP2024514222A (en) 2021-04-19 2024-03-28 ジェネンテック, インコーポレイテッド Modified Mammalian Cells
JP2024514281A (en) 2021-04-23 2024-04-01 エフ・ホフマン-ラ・ロシュ・アクチェンゲゼルシャフト Prevention or reduction of adverse effects related to NK cell-engaging agents
EP4326768A1 (en) 2021-04-23 2024-02-28 Profoundbio Us Co. Anti-cd70 antibodies, conjugates thereof and methods of using the same
AU2021443863A1 (en) 2021-04-30 2023-10-26 F. Hoffmann-La Roche Ag Dosing for treatment with anti-cd20/anti-cd3 bispecific antibody
KR20240005691A (en) 2021-04-30 2024-01-12 에프. 호프만-라 로슈 아게 Dosage for combination therapy with anti-CD20/anti-CD3 bispecific antibody and anti-CD79B antibody drug conjugate
KR20240004694A (en) 2021-05-03 2024-01-11 유씨비 바이오파마 에스알엘 antibody
EP4334343A2 (en) 2021-05-06 2024-03-13 The Rockefeller University Neutralizing anti-sars- cov-2 antibodies and methods of use thereof
IL308336A (en) 2021-05-07 2024-01-01 Alpine Immune Sciences Inc Methods of dosing and treatment with a taci-fc fusion immunomodulatory protein
CA3218170A1 (en) 2021-05-12 2022-11-17 Jamie Harue HIRATA Methods of using anti-cd79b immunoconjugates to treat diffuse large b-cell lymphoma
WO2022241082A1 (en) 2021-05-14 2022-11-17 Genentech, Inc. Agonists of trem2
US20230115257A1 (en) 2021-05-17 2023-04-13 Curia Ip Holdings, Llc Sars-cov-2 spike protein antibodies
US20220372114A1 (en) 2021-05-17 2022-11-24 Curia Ip Holdings, Llc Sars-cov-2 spike protein antibodies
WO2022245754A1 (en) 2021-05-17 2022-11-24 Iovance Biotherapeutics, Inc. Pd-1 gene-edited tumor infiltrating lymphocytes and uses of same in immunotherapy
WO2022243261A1 (en) 2021-05-19 2022-11-24 F. Hoffmann-La Roche Ag Agonistic cd40 antigen binding molecules targeting cea
WO2022246259A1 (en) 2021-05-21 2022-11-24 Genentech, Inc. Modified cells for the production of a recombinant product of interest
BR112023024804A2 (en) 2021-05-28 2024-02-15 Glaxosmithkline Ip Dev Ltd COMBINATION THERAPIES TO TREAT CANCER
AR126009A1 (en) 2021-06-02 2023-08-30 Hoffmann La Roche CD28 ANTIGEN-BINDING AGONIST MOLECULES THAT TARGET EPCAM
TW202306994A (en) 2021-06-04 2023-02-16 日商中外製藥股份有限公司 Anti-ddr2 antibodies and uses thereof
CN117616050A (en) 2021-06-09 2024-02-27 先天制药公司 Multispecific proteins that bind to NKP46, cytokine receptor, tumor antigen and CD16A
WO2022258673A1 (en) 2021-06-09 2022-12-15 Innate Pharma Multispecific antibodies binding to cd20, nkp46, cd16 and conjugated to il-2
WO2022258678A1 (en) 2021-06-09 2022-12-15 Innate Pharma Multispecific proteins binding to nkp30, a cytokine receptor, a tumour antigen and cd16a
WO2022258691A1 (en) 2021-06-09 2022-12-15 Innate Pharma Multispecific proteins binding to nkg2d, a cytokine receptor, a tumour antigen and cd16a
AU2022293999A1 (en) 2021-06-14 2023-11-30 argenx BV Anti-il-9 antibodies and methods of use thereof
EP4355785A1 (en) 2021-06-17 2024-04-24 Amberstone Biosciences, Inc. Anti-cd3 constructs and uses thereof
EP4360648A1 (en) 2021-06-23 2024-05-01 Toray Industries, Inc. Medicament for treatment and/or prevention of cancer
EP4360649A1 (en) 2021-06-23 2024-05-01 Toray Industries, Inc. Medicament for treatment and/or prevention of cancer
EP4361273A1 (en) 2021-06-25 2024-05-01 Chugai Seiyaku Kabushiki Kaisha Anti-ctla-4 antibody
EP4361176A1 (en) 2021-06-25 2024-05-01 Chugai Seiyaku Kabushiki Kaisha Use of anti-ctla-4 antibody
WO2023275621A1 (en) 2021-07-01 2023-01-05 Compugen Ltd. Anti-tigit and anti-pvrig in monotherapy and combination treatments
TW202309078A (en) 2021-07-02 2023-03-01 美商建南德克公司 Methods and compositions for treating cancer
TW202320857A (en) 2021-07-06 2023-06-01 美商普方生物製藥美國公司 Linkers, drug linkers and conjugates thereof and methods of using the same
TW202317633A (en) 2021-07-08 2023-05-01 美商舒泰神(加州)生物科技有限公司 Antibodies specifically recognizing tnfr2 and uses thereof
WO2023288182A1 (en) 2021-07-12 2023-01-19 Genentech, Inc. Structures for reducing antibody-lipase binding
IL309856A (en) 2021-07-14 2024-02-01 Genentech Inc Anti-c-c motif chemokine receptor 8 (ccr8) antibodies and methods of use
WO2023284714A1 (en) 2021-07-14 2023-01-19 舒泰神(北京)生物制药股份有限公司 Antibody that specifically recognizes cd40 and application thereof
WO2023001884A1 (en) 2021-07-22 2023-01-26 F. Hoffmann-La Roche Ag Heterodimeric fc domain antibodies
WO2023004074A2 (en) 2021-07-22 2023-01-26 Iovance Biotherapeutics, Inc. Method for cryopreservation of solid tumor fragments
WO2023004386A1 (en) 2021-07-22 2023-01-26 Genentech, Inc. Brain targeting compositions and methods of use thereof
WO2023008461A1 (en) 2021-07-27 2023-02-02 東レ株式会社 Medicament for treatment and/or prevention of cancer
WO2023008459A1 (en) 2021-07-27 2023-02-02 東レ株式会社 Medicament for treatment and/or prevention of cancer
AU2022320304A1 (en) 2021-07-27 2024-02-29 Toray Industries, Inc. Medicament for treatment and/or prevention of cancer
WO2023007374A1 (en) 2021-07-27 2023-02-02 Pfizer Inc. Method of treatment of cancer pain with tanezumab
CA3226942A1 (en) 2021-07-28 2023-02-02 Iovance Biotherapeutics, Inc. Treatment of cancer patients with tumor infiltrating lymphocyte therapies in combination with kras inhibitors
CN117715936A (en) 2021-07-28 2024-03-15 豪夫迈·罗氏有限公司 Methods and compositions for treating cancer
AU2022320051A1 (en) 2021-07-30 2024-01-25 ONA Therapeutics S.L. Anti-cd36 antibodies and their use to treat cancer
AU2022324406A1 (en) 2021-08-02 2024-03-14 Innovent Biologics (Suzhou) Co., Ltd. Anti-cd79b×cd3 bispecific antibody and use thereof
CN117794953A (en) 2021-08-03 2024-03-29 豪夫迈·罗氏有限公司 Bispecific antibodies and methods of use
WO2023019092A1 (en) 2021-08-07 2023-02-16 Genentech, Inc. Methods of using anti-cd79b immunoconjugates to treat diffuse large b-cell lymphoma
WO2023019239A1 (en) 2021-08-13 2023-02-16 Genentech, Inc. Dosing for anti-tryptase antibodies
WO2023021055A1 (en) 2021-08-19 2023-02-23 F. Hoffmann-La Roche Ag Multivalent anti-variant fc-region antibodies and methods of use
GB202111905D0 (en) 2021-08-19 2021-10-06 UCB Biopharma SRL Antibodies
AR126838A1 (en) 2021-08-20 2023-11-22 Novartis Ag METHODS FOR PREPARING CELLS THAT EXPRESS CHIMERIC ANTIGEN RECEPTORS
CA3229448A1 (en) 2021-08-23 2023-03-02 Immunitas Therapeutics, Inc. Anti-cd161 antibodies and uses thereof
TW202317637A (en) 2021-08-26 2023-05-01 日商協和麒麟股份有限公司 Bispecific antibody that binds to CD116 and CD131
AU2022332303A1 (en) 2021-08-27 2024-02-01 Genentech, Inc. Methods of treating tau pathologies
TW202325727A (en) 2021-08-30 2023-07-01 美商建南德克公司 Anti-polyubiquitin multispecific antibodies
AU2022338463A1 (en) 2021-09-03 2024-03-21 Toray Industries, Inc. Pharmaceutical composition for cancer treatment and/or prevention
WO2023039488A1 (en) 2021-09-09 2023-03-16 Iovance Biotherapeutics, Inc. Processes for generating til products using pd-1 talen knockdown
CA3232700A1 (en) 2021-09-24 2023-03-30 Rafael CUBAS Expansion processes and agents for tumor infiltrating lymphocytes
WO2023048726A1 (en) 2021-09-27 2023-03-30 Vaccinex, Inc. Predictive outcome profiling for use of an anti-semaphorin-4d binding molecule to treat neurodegenerative disorders
WO2023056069A1 (en) 2021-09-30 2023-04-06 Angiex, Inc. Degrader-antibody conjugates and methods of using same
CA3232806A1 (en) 2021-09-30 2023-04-06 Seagen Inc. B7-h4 antibody-drug conjugates for the treatment of cancer
WO2023056403A1 (en) 2021-09-30 2023-04-06 Genentech, Inc. Methods for treatment of hematologic cancers using anti-tigit antibodies, anti-cd38 antibodies, and pd-1 axis binding antagonists
WO2023057893A1 (en) 2021-10-05 2023-04-13 Glaxosmithkline Intellectual Property Development Limited Combination therapies for treating cancer
CN116064598B (en) 2021-10-08 2024-03-12 苏州艾博生物科技有限公司 Nucleic acid vaccine for coronavirus
WO2023062048A1 (en) 2021-10-14 2023-04-20 F. Hoffmann-La Roche Ag Alternative pd1-il7v immunoconjugates for the treatment of cancer
US20230192843A1 (en) 2021-10-14 2023-06-22 Teneobio, Inc. Mesothelin binding proteins and uses thereof
WO2023062050A1 (en) 2021-10-14 2023-04-20 F. Hoffmann-La Roche Ag New interleukin-7 immunoconjugates
WO2023064958A1 (en) 2021-10-15 2023-04-20 Compugen Ltd. Combination therapy with anti-pvrig antibodies formulations, anti-tigit antibodies, and anti-pd-1 antibodies
WO2023076876A1 (en) 2021-10-26 2023-05-04 Mozart Therapeutics, Inc. Modulation of immune responses to viral vectors
TW202331735A (en) 2021-10-27 2023-08-01 美商艾歐凡斯生物治療公司 Systems and methods for coordinating manufacturing of cells for patient-specific immunotherapy
WO2023073599A1 (en) 2021-10-28 2023-05-04 Novartis Ag Engineered fc variants
WO2023077155A1 (en) 2021-11-01 2023-05-04 Janssen Biotech, Inc. Compositions and methods for the modulation of beta chain-mediated immunity
TW202342095A (en) 2021-11-05 2023-11-01 英商阿斯特捷利康英國股份有限公司 Composition for treatment and prevention of covid-19
WO2023081818A1 (en) 2021-11-05 2023-05-11 American Diagnostics & Therapy, Llc (Adxrx) Monoclonal antibodies against carcinoembryonic antigens, and their uses
WO2023086807A1 (en) 2021-11-10 2023-05-19 Genentech, Inc. Anti-interleukin-33 antibodies and uses thereof
WO2023086803A1 (en) 2021-11-10 2023-05-19 Iovance Biotherapeutics, Inc. Methods of expansion treatment utilizing cd8 tumor infiltrating lymphocytes
TW202337494A (en) 2021-11-16 2023-10-01 美商建南德克公司 Methods and compositions for treating systemic lupus erythematosus (sle) with mosunetuzumab
CA3235206A1 (en) 2021-11-16 2023-05-25 Davide BASCO Novel molecules for therapy and diagnosis
WO2023091968A1 (en) 2021-11-17 2023-05-25 Disc Medicine, Inc. Methods for treating anemia of kidney disease
WO2023094413A1 (en) 2021-11-25 2023-06-01 F. Hoffmann-La Roche Ag Improved antigen binding receptors
AR127887A1 (en) 2021-12-10 2024-03-06 Hoffmann La Roche ANTIBODIES THAT BIND CD3 AND PLAP
WO2023114829A1 (en) 2021-12-15 2023-06-22 Genentech, Inc. Stabilized il-18 polypeptides and uses thereof
TW202340248A (en) 2021-12-20 2023-10-16 瑞士商赫孚孟拉羅股份公司 Agonistic ltbr antibodies and bispecific antibodies comprising them
WO2023131901A1 (en) 2022-01-07 2023-07-13 Johnson & Johnson Enterprise Innovation Inc. Materials and methods of il-1beta binding proteins
WO2023139107A1 (en) 2022-01-18 2023-07-27 argenx BV Galectin-10 antibodies
TW202340251A (en) 2022-01-19 2023-10-16 美商建南德克公司 Anti-notch2 antibodies and conjugates and methods of use
WO2023147399A1 (en) 2022-01-27 2023-08-03 The Rockefeller University Broadly neutralizing anti-sars-cov-2 antibodies targeting the n-terminal domain of the spike protein and methods of use thereof
WO2023147486A1 (en) 2022-01-28 2023-08-03 Iovance Biotherapeutics, Inc. Tumor infiltrating lymphocytes engineered to express payloads
WO2023147488A1 (en) 2022-01-28 2023-08-03 Iovance Biotherapeutics, Inc. Cytokine associated tumor infiltrating lymphocytes compositions and methods
TW202342519A (en) 2022-02-16 2023-11-01 瑞士商Ac 免疫有限公司 Humanized anti-tdp-43 binding molecules and uses thereof
WO2023172883A1 (en) 2022-03-07 2023-09-14 Alpine Immune Sciences, Inc. Immunomodulatory proteins of variant cd80 polypeptides, cell therapies thereof and related methods and uses
WO2023173026A1 (en) 2022-03-10 2023-09-14 Sorrento Therapeutics, Inc. Antibody-drug conjugates and uses thereof
WO2023178192A1 (en) 2022-03-15 2023-09-21 Compugen Ltd. Il-18bp antagonist antibodies and their use in monotherapy and combination therapy in the treatment of cancer
US20240103010A1 (en) 2022-03-18 2024-03-28 Compugen Ltd. Pvrl2 and/or pvrig as biomarkers for treatment
WO2023178357A1 (en) 2022-03-18 2023-09-21 Evolveimmune Therapeutics, Inc. Bispecific antibody fusion molecules and methods of use thereof
TW202346365A (en) 2022-03-23 2023-12-01 瑞士商赫孚孟拉羅股份公司 Combination treatment of an anti-cd20/anti-cd3 bispecific antibody and chemotherapy
WO2023186756A1 (en) 2022-03-28 2023-10-05 F. Hoffmann-La Roche Ag Interferon gamma variants and antigen binding molecules comprising these
TW202402794A (en) 2022-03-28 2024-01-16 瑞士商赫孚孟拉羅股份公司 Improved folr1 protease-activatable t cell bispecific antibodies
WO2023187657A1 (en) 2022-03-30 2023-10-05 Novartis Ag Methods of treating disorders using anti-natriuretic peptide receptor 1 (npr1) antibodies
WO2023191816A1 (en) 2022-04-01 2023-10-05 Genentech, Inc. Dosing for treatment with anti-fcrh5/anti-cd3 bispecific antibodies
WO2023196877A1 (en) 2022-04-06 2023-10-12 Iovance Biotherapeutics, Inc. Treatment of nsclc patients with tumor infiltrating lymphocyte therapies
WO2023194565A1 (en) 2022-04-08 2023-10-12 Ac Immune Sa Anti-tdp-43 binding molecules
WO2023201299A1 (en) 2022-04-13 2023-10-19 Genentech, Inc. Pharmaceutical compositions of therapeutic proteins and methods of use
WO2023198727A1 (en) 2022-04-13 2023-10-19 F. Hoffmann-La Roche Ag Pharmaceutical compositions of anti-cd20/anti-cd3 bispecific antibodies and methods of use
WO2023201369A1 (en) 2022-04-15 2023-10-19 Iovance Biotherapeutics, Inc. Til expansion processes using specific cytokine combinations and/or akti treatment
US20230357381A1 (en) 2022-04-26 2023-11-09 Novartis Ag Multispecific antibodies targeting il-13 and il-18
WO2023209177A1 (en) 2022-04-29 2023-11-02 Astrazeneca Uk Limited Sars-cov-2 antibodies and methods of using the same
WO2023215737A1 (en) 2022-05-03 2023-11-09 Genentech, Inc. Anti-ly6e antibodies, immunoconjugates, and uses thereof
WO2023220608A1 (en) 2022-05-10 2023-11-16 Iovance Biotherapeutics, Inc. Treatment of cancer patients with tumor infiltrating lymphocyte therapies in combination with an il-15r agonist
WO2023217933A1 (en) 2022-05-11 2023-11-16 F. Hoffmann-La Roche Ag Antibody that binds to vegf-a and il6 and methods of use
WO2023218320A1 (en) 2022-05-11 2023-11-16 Pfizer Inc. Anti-lymphotoxin beta receptor antibodies and methods of use thereof
WO2023219613A1 (en) 2022-05-11 2023-11-16 Genentech, Inc. Dosing for treatment with anti-fcrh5/anti-cd3 bispecific antibodies
WO2023222557A1 (en) 2022-05-17 2023-11-23 Bayer Aktiengesellschaft Radiopharmaceutical complexes targeting prostate-specific membrane antigen and its combinations
EP4279092A1 (en) 2022-05-17 2023-11-22 Bayer AG Radiopharmaceutical complexes targeting prostate-specific membrane antigen
WO2023227660A1 (en) 2022-05-25 2023-11-30 Innate Pharma Nectin-4 binding agents
WO2023228082A1 (en) 2022-05-26 2023-11-30 Pfizer Inc. Anti-tnfr2 antibodies and methods of use thereof
WO2023227641A1 (en) 2022-05-27 2023-11-30 Glaxosmithkline Intellectual Property Development Limited Use of tnf-alpha binding proteins and il-7 binding proteins in medical treatment
WO2023235699A1 (en) 2022-05-31 2023-12-07 Jounce Therapeutics, Inc. Antibodies to lilrb4 and uses thereof
WO2023233330A1 (en) 2022-05-31 2023-12-07 Pfizer Inc. Anti-bmp9 antibodies and methods of use thereof
WO2023240058A2 (en) 2022-06-07 2023-12-14 Genentech, Inc. Prognostic and therapeutic methods for cancer
WO2023239803A1 (en) 2022-06-08 2023-12-14 Angiex, Inc. Anti-tm4sf1 antibody-drug conjugates comprising cleavable linkers and methods of using same
WO2023245048A1 (en) 2022-06-15 2023-12-21 Bioverativ Usa Inc. Anti-complement c1s antibody formulation
WO2023242361A1 (en) 2022-06-15 2023-12-21 argenx BV Fcrn binding molecules and methods of use
WO2023242769A1 (en) 2022-06-17 2023-12-21 Pfizer Inc. Il-12 variants, anti-pd1 antibodies, fusion proteins, and uses thereof
WO2023250507A1 (en) 2022-06-24 2023-12-28 Bioverativ Usa Inc. Methods for treating complement-mediated diseases
WO2024011114A1 (en) 2022-07-06 2024-01-11 Iovance Biotherapeutics, Inc. Devices and processes for automated production of tumor infiltrating lymphocytes
WO2024015897A1 (en) 2022-07-13 2024-01-18 Genentech, Inc. Dosing for treatment with anti-fcrh5/anti-cd3 bispecific antibodies
WO2024020432A1 (en) 2022-07-19 2024-01-25 Genentech, Inc. Dosing for treatment with anti-fcrh5/anti-cd3 bispecific antibodies
WO2024020407A1 (en) 2022-07-19 2024-01-25 Staidson Biopharma Inc. Antibodies specifically recognizing b- and t-lymphocyte attenuator (btla) and uses thereof
WO2024020579A1 (en) 2022-07-22 2024-01-25 Bristol-Myers Squibb Company Antibodies binding to human pad4 and uses thereof
WO2024020564A1 (en) 2022-07-22 2024-01-25 Genentech, Inc. Anti-steap1 antigen-binding molecules and uses thereof
WO2024026358A1 (en) 2022-07-27 2024-02-01 Teneobio, Inc. Mesothelin binding proteins and uses thereof
WO2024026496A1 (en) 2022-07-28 2024-02-01 Compugen Ltd. Combination therapy with anti-pvrig antibodies formulations and anti-pd-1 antibodies
WO2024030758A1 (en) 2022-08-01 2024-02-08 Iovance Biotherapeutics, Inc. Chimeric costimulatory receptors, chemokine receptors, and the use of same in cellular immunotherapies
US20240052047A1 (en) 2022-08-03 2024-02-15 Pfizer Inc. Anti- il27r antibodies and methods of use thereof
WO2024030956A2 (en) 2022-08-03 2024-02-08 Mozart Therapeutics, Inc. Cd39-specific binding agents and methods of using the same
WO2024028731A1 (en) 2022-08-05 2024-02-08 Janssen Biotech, Inc. Transferrin receptor binding proteins for treating brain tumors
WO2024028732A1 (en) 2022-08-05 2024-02-08 Janssen Biotech, Inc. Cd98 binding constructs for treating brain tumors
WO2024037633A2 (en) 2022-08-19 2024-02-22 Evive Biotechnology (Shanghai) Ltd Formulations comprising g-csf and uses thereof
WO2024044547A1 (en) * 2022-08-22 2024-02-29 Abdera Therapeutics Inc. Kidney targeting antibodies
WO2024049949A1 (en) 2022-09-01 2024-03-07 Genentech, Inc. Therapeutic and diagnostic methods for bladder cancer
WO2024054929A1 (en) 2022-09-07 2024-03-14 Dynamicure Biotechnology Llc Anti-vista constructs and uses thereof
WO2024056809A1 (en) 2022-09-15 2024-03-21 Novartis Ag Treatment of autoimmune disorders using chimeric antigen receptor therapy
US20240101691A1 (en) 2022-09-21 2024-03-28 Sanofi Biotechnology Humanized anti-il-1r3 antibody and methods of use
WO2024068572A1 (en) 2022-09-28 2024-04-04 F. Hoffmann-La Roche Ag Improved protease-activatable t cell bispecific antibodies
WO2024077018A2 (en) 2022-10-04 2024-04-11 Alpine Immune Sciences, Inc. Methods and uses of taci-fc fusion immunomodulatory protein
WO2024077044A1 (en) 2022-10-05 2024-04-11 Amgen Inc. Combination therapies comprising t-cell redirecting therapies and agonistic anti-il-2r antibodies or fragments thereof
WO2024077239A1 (en) 2022-10-07 2024-04-11 Genentech, Inc. Methods of treating cancer with anti-c-c motif chemokine receptor 8 (ccr8) antibodies
WO2024079015A1 (en) 2022-10-10 2024-04-18 F. Hoffmann-La Roche Ag Combination therapy of a gprc5d tcb and imids
WO2024079009A1 (en) 2022-10-10 2024-04-18 F. Hoffmann-La Roche Ag Combination therapy of a gprc5d tcb and proteasome inhibitors
WO2024079010A1 (en) 2022-10-10 2024-04-18 F. Hoffmann-La Roche Ag Combination therapy of a gprc5d tcb and cd38 antibodies

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999022764A1 (en) * 1997-10-31 1999-05-14 Genentech, Inc. Methods and compositions comprising glycoprotein glycoforms
WO2003074679A2 (en) * 2002-03-01 2003-09-12 Xencor Antibody optimization

Family Cites Families (73)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US98193A (en) * 1869-12-21 Improvement in corn-planters
US158389A (en) * 1875-01-05 Improvement in gas-regulators
US2587A (en) * 1842-04-29 Cooking-stove
US4490473A (en) 1983-03-28 1984-12-25 Panab Labeled antibodies and methods
US4752601A (en) 1983-08-12 1988-06-21 Immunetech Pharmaceuticals Method of blocking immune complex binding to immunoglobulin FC receptors
US5985599A (en) 1986-05-29 1999-11-16 The Austin Research Institute FC receptor for immunoglobulin
AU600575B2 (en) 1987-03-18 1990-08-16 Sb2, Inc. Altered antibodies
US5204244A (en) 1987-10-27 1993-04-20 Oncogen Production of chimeric antibodies by homologous recombination
US5576184A (en) * 1988-09-06 1996-11-19 Xoma Corporation Production of chimeric mouse-human antibodies with specificity to human tumor antigens
US5047335A (en) * 1988-12-21 1991-09-10 The Regents Of The University Of Calif. Process for controlling intracellular glycosylation of proteins
GB8916400D0 (en) 1989-07-18 1989-09-06 Dynal As Modified igg3
US5364930A (en) 1990-10-16 1994-11-15 Northwestern University Synthetic C1q peptide fragments
US5419904A (en) 1990-11-05 1995-05-30 The Regents Of The University Of California Human B-lymphoblastoid cell line secreting anti-ganglioside antibody
US5278299A (en) * 1991-03-18 1994-01-11 Scripps Clinic And Research Foundation Method and composition for synthesizing sialylated glycosyl compounds
US6136310A (en) 1991-07-25 2000-10-24 Idec Pharmaceuticals Corporation Recombinant anti-CD4 antibodies for human therapy
AU3144193A (en) * 1991-11-21 1993-06-15 Board Of Trustees Of The Leland Stanford Junior University Controlling degradation of glycoprotein oligosaccharides by extracellular glycosisases
GB9206422D0 (en) 1992-03-24 1992-05-06 Bolt Sarah L Antibody preparation
EP0640094A1 (en) 1992-04-24 1995-03-01 The Board Of Regents, The University Of Texas System Recombinant production of immunoglobulin-like domains in prokaryotic cells
US5736137A (en) 1992-11-13 1998-04-07 Idec Pharmaceuticals Corporation Therapeutic application of chimeric and radiolabeled antibodies to human B lymphocyte restricted differentiation antigen for treatment of B cell lymphoma
US6491916B1 (en) 1994-06-01 2002-12-10 Tolerance Therapeutics, Inc. Methods and materials for modulation of the immunosuppresive activity and toxicity of monoclonal antibodies
EP0714409A1 (en) 1993-06-16 1996-06-05 Celltech Therapeutics Limited Antibodies
US6485726B1 (en) * 1995-01-17 2002-11-26 The Brigham And Women's Hospital, Inc. Receptor specific transepithelial transport of therapeutics
US6086875A (en) * 1995-01-17 2000-07-11 The Brigham And Women's Hospital, Inc. Receptor specific transepithelial transport of immunogens
US6030613A (en) * 1995-01-17 2000-02-29 The Brigham And Women's Hospital, Inc. Receptor specific transepithelial transport of therapeutics
US5731168A (en) 1995-03-01 1998-03-24 Genentech, Inc. Method for making heteromultimeric polypeptides
US5730977A (en) 1995-08-21 1998-03-24 Mitsui Toatsu Chemicals, Inc. Anti-VEGF human monoclonal antibody
US6750334B1 (en) 1996-02-02 2004-06-15 Repligen Corporation CTLA4-immunoglobulin fusion proteins having modified effector functions and uses therefor
JP4046354B2 (en) 1996-03-18 2008-02-13 ボード オブ リージェンツ,ザ ユニバーシティ オブ テキサス システム Immunoglobulin-like domain with increased half-life
US5834597A (en) 1996-05-20 1998-11-10 Protein Design Labs, Inc. Mutated nonactivating IgG2 domains and anti CD3 antibodies incorporating the same
JP2000516452A (en) * 1996-07-16 2000-12-12 プリュックテュン,アンドレアス Immunoglobulin superfamily domains and fragments with increased solubility
WO1998023289A1 (en) 1996-11-27 1998-06-04 The General Hospital Corporation MODULATION OF IgG BINDING TO FcRn
US6277375B1 (en) 1997-03-03 2001-08-21 Board Of Regents, The University Of Texas System Immunoglobulin-like domains with increased half-lives
US6323321B1 (en) * 1997-03-05 2001-11-27 Eberhard-Karls-Universitat Tubingen Universitatsklinikum Antibody 97A6
AU7467898A (en) * 1997-04-21 1998-11-13 Arch Development Corporation Fc receptor non-binding anti-cd3 monoclonal antibodies deliver a partial cr signal and induce clonal anergy
DE19721700C1 (en) 1997-05-23 1998-11-19 Deutsches Krebsforsch Mutant OKT3 antibody
US6172213B1 (en) * 1997-07-02 2001-01-09 Genentech, Inc. Anti-IgE antibodies and method of improving polypeptides
PL199659B1 (en) 1998-02-25 2008-10-31 Merck Patent Gmbh Antibody-based fusion protein featured by prolonged residence in a cireculatory system and method of prolonging its residence time in a circulatory system
US6194551B1 (en) 1998-04-02 2001-02-27 Genentech, Inc. Polypeptide variants
IL138608A0 (en) 1998-04-02 2001-10-31 Genentech Inc Antibody variants and fragments thereof
US6242195B1 (en) * 1998-04-02 2001-06-05 Genentech, Inc. Methods for determining binding of an analyte to a receptor
US6528624B1 (en) 1998-04-02 2003-03-04 Genentech, Inc. Polypeptide variants
ES2340112T3 (en) * 1998-04-20 2010-05-28 Glycart Biotechnology Ag ANTIBODY GLICOSILATION ENGINEERING FOR THE IMPROVEMENT OF DEPENDENT CELLULAR CYTOTOXICITY OF ANTIBODIES.
GB9809951D0 (en) 1998-05-08 1998-07-08 Univ Cambridge Tech Binding molecules
CA2341029A1 (en) 1998-08-17 2000-02-24 Abgenix, Inc. Generation of modified molecules with increased serum half-lives
WO2000042072A2 (en) * 1999-01-15 2000-07-20 Genentech, Inc. Polypeptide variants with altered effector function
US6737056B1 (en) 1999-01-15 2004-05-18 Genentech, Inc. Polypeptide variants with altered effector function
US7183387B1 (en) 1999-01-15 2007-02-27 Genentech, Inc. Polypeptide variants with altered effector function
US6676927B1 (en) 1999-01-20 2004-01-13 The Rockefeller University Animal model and methods for its use in the selection of cytotoxic antibodies
EP1176195B1 (en) * 1999-04-09 2013-05-22 Kyowa Hakko Kirin Co., Ltd. Method for controlling the activity of immunologically functional molecule
DE60122286T2 (en) 2000-02-11 2007-08-02 Merck Patent Gmbh INCREASING THE CIRCULATORY HALF-TIME OF ANTIBODY-BASED FUSION PROTEINS
AU5345901A (en) * 2000-04-13 2001-10-30 Univ Rockefeller Enhancement of antibody-mediated immune responses
US7598055B2 (en) * 2000-06-28 2009-10-06 Glycofi, Inc. N-acetylglucosaminyltransferase III expression in lower eukaryotes
US6946292B2 (en) * 2000-10-06 2005-09-20 Kyowa Hakko Kogyo Co., Ltd. Cells producing antibody compositions with increased antibody dependent cytotoxic activity
EP2341060B1 (en) 2000-12-12 2019-02-20 MedImmune, LLC Molecules with extended half-lives, compositions and uses thereof
ATE507839T1 (en) * 2001-04-02 2011-05-15 Genentech Inc COMBINATION THERAPY
WO2003035835A2 (en) * 2001-10-25 2003-05-01 Genentech, Inc. Glycoprotein compositions
US20040002587A1 (en) 2002-02-20 2004-01-01 Watkins Jeffry D. Fc region variants
US20040132101A1 (en) 2002-09-27 2004-07-08 Xencor Optimized Fc variants and methods for their generation
US7317091B2 (en) * 2002-03-01 2008-01-08 Xencor, Inc. Optimized Fc variants
FR2840779B1 (en) * 2002-06-18 2004-09-17 Lafina Man Sa SIMPLE MANEUVER KEY RING
JP2006500009A (en) 2002-07-09 2006-01-05 ジェネンテック・インコーポレーテッド Compositions and methods for tumor diagnosis and treatment
BRPI0314814C1 (en) 2002-09-27 2021-07-27 Xencor Inc antibody comprising an fc variant
US7217797B2 (en) 2002-10-15 2007-05-15 Pdl Biopharma, Inc. Alteration of FcRn binding affinities or serum half-lives of antibodies by mutagenesis
CA2502904C (en) 2002-10-15 2013-05-28 Protein Design Labs, Inc. Alteration of fcrn binding affinities or serum half-lives of antibodies by mutagenesis
US7608260B2 (en) 2003-01-06 2009-10-27 Medimmune, Llc Stabilized immunoglobulins
EP2368578A1 (en) 2003-01-09 2011-09-28 Macrogenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
JP2007531707A (en) 2003-10-15 2007-11-08 ピーディーエル バイオファーマ, インコーポレイテッド Modification of Fc fusion protein serum half-life by mutagenesis of heavy chain constant region positions 250, 314 and / or 428 of IG
AU2005227326B2 (en) * 2004-03-24 2009-12-03 Xencor, Inc. Immunoglobulin variants outside the Fc region
AU2005285347A1 (en) 2004-08-19 2006-03-23 Genentech, Inc. Polypeptide variants with altered effector function
DK2325207T3 (en) * 2004-11-12 2017-06-06 Xencor Inc Fc variants with altered binding to FcRn
EP1858925A2 (en) * 2005-01-12 2007-11-28 Xencor, Inc. Antibodies and fc fusion proteins with altered immunogenicity
DOP2006000029A (en) 2005-02-07 2006-08-15 Genentech Inc ANTIBODY VARIANTS AND USES THEREOF. (VARIATIONS OF AN ANTIBODY AND USES OF THE SAME)
NZ569957A (en) * 2006-02-10 2012-03-30 Genentech Inc Anti-FGF19 antibodies and methods using same

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999022764A1 (en) * 1997-10-31 1999-05-14 Genentech, Inc. Methods and compositions comprising glycoprotein glycoforms
WO2003074679A2 (en) * 2002-03-01 2003-09-12 Xencor Antibody optimization

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
Jefferis et al. Immunology Letters, 1995, 44:111-117. *
Macher et al. Molecular BioSystems, 2007, 3:705-713. *
Murray et al. Harper's Biochemistry, 23rd Edition, By Appleton & Lange, Chapter 4, pages 24-28. *

Cited By (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080274108A1 (en) * 1999-01-15 2008-11-06 Genentech, Inc. Polypeptide variants with altered effector function
US8674083B2 (en) 1999-01-15 2014-03-18 Genentech, Inc. Polypeptide variants with altered effector function
US20100098730A1 (en) * 2008-10-14 2010-04-22 Lowman Henry B Immunoglobulin variants and uses thereof
US9879249B2 (en) 2009-02-17 2018-01-30 Redwood Bioscience, Inc. Aldehyde-tagged protein-based drug carriers and methods of use
US9238878B2 (en) 2009-02-17 2016-01-19 Redwood Bioscience, Inc. Aldehyde-tagged protein-based drug carriers and methods of use
US9540438B2 (en) 2011-01-14 2017-01-10 Redwood Bioscience, Inc. Aldehyde-tagged immunoglobulin polypeptides and methods of use thereof
US10183998B2 (en) 2011-01-14 2019-01-22 Redwood Bioscience, Inc. Aldehyde-tagged immunoglobulin polypeptides and methods of use thereof
US8969526B2 (en) 2011-03-29 2015-03-03 Roche Glycart Ag Antibody Fc variants
US10654916B2 (en) 2011-04-21 2020-05-19 The Regents Of The University Of California, A California Corporation Compositions and methods for the treatment of neuromyelitis optica
US11390667B2 (en) 2011-04-21 2022-07-19 The Regents Of The University Of California Compositions and methods for the treatment of neuromyelitis optica
US9695233B2 (en) 2012-07-13 2017-07-04 Roche Glycart Ag Bispecific anti-VEGF/anti-ANG-2 antibodies and their use in the treatment of ocular vascular diseases
US10683345B2 (en) 2012-07-13 2020-06-16 Roche Glycart Ag Bispecific anti-VEGF/anti-ANG-2 antibodies and their use in the treatment of ocular vascular diseases
US11220556B2 (en) 2013-03-15 2022-01-11 Biomolecular Holdings Llc Hybrid immunoglobulin containing non-peptidyl linkage
US9260527B2 (en) 2013-03-15 2016-02-16 Sdix, Llc Anti-human CXCR4 antibodies and methods of making same
US11066459B2 (en) 2014-03-14 2021-07-20 Biomolecular Holdings Llc Hybrid immunoglobulin containing non-peptidyl linkage
US11208632B2 (en) 2016-04-26 2021-12-28 R.P. Scherer Technologies, Llc Antibody conjugates and methods of making and using the same
US11788066B2 (en) 2016-04-26 2023-10-17 R.P. Scherer Technologies, Llc Antibody conjugates and methods of making and using the same

Also Published As

Publication number Publication date
US7122637B2 (en) 2006-10-17
US6737056B1 (en) 2004-05-18
US20080274108A1 (en) 2008-11-06
US20040228856A1 (en) 2004-11-18
US20050118174A1 (en) 2005-06-02
US20160060330A1 (en) 2016-03-03
US7332581B2 (en) 2008-02-19
US20050233382A1 (en) 2005-10-20
US20060194957A1 (en) 2006-08-31
US20040191244A1 (en) 2004-09-30
US20140342404A1 (en) 2014-11-20
US7335742B2 (en) 2008-02-26
US7416727B2 (en) 2008-08-26
US20140147436A1 (en) 2014-05-29

Similar Documents

Publication Publication Date Title
US7335742B2 (en) Polypeptide variants with altered effector function
US7790858B2 (en) Polypeptide variants with altered effector function
US8674083B2 (en) Polypeptide variants with altered effector function
AU2004233493B2 (en) Polypeptide variants with altered effector function

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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