WO2009058492A2 - Fc variants with altered binding to fcrn - Google Patents

Fc variants with altered binding to fcrn Download PDF

Info

Publication number
WO2009058492A2
WO2009058492A2 PCT/US2008/077250 US2008077250W WO2009058492A2 WO 2009058492 A2 WO2009058492 A2 WO 2009058492A2 US 2008077250 W US2008077250 W US 2008077250W WO 2009058492 A2 WO2009058492 A2 WO 2009058492A2
Authority
WO
WIPO (PCT)
Prior art keywords
ser
vai
pro
lys
leu
Prior art date
Application number
PCT/US2008/077250
Other languages
French (fr)
Other versions
WO2009058492A3 (en
Inventor
Aaron Chamberlain
Bassil Dahiyat
John Rudolph Desjarlais
Sher Bahadur Karki
Original Assignee
Xencor, 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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=40197140&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2009058492(A2) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority to CA2703385A priority Critical patent/CA2703385C/en
Priority to EP16185391.6A priority patent/EP3138853B1/en
Priority to BRPI0818746-0A priority patent/BRPI0818746A2/en
Priority to CN200880113962.0A priority patent/CN101970492B/en
Priority to RU2010121898/10A priority patent/RU2517621C2/en
Priority to JP2010531108A priority patent/JP5542677B2/en
Priority to CN202210438504.6A priority patent/CN114891093A/en
Application filed by Xencor, Inc filed Critical Xencor, Inc
Priority to ES08846103.3T priority patent/ES2537202T3/en
Priority to AU2008319144A priority patent/AU2008319144B2/en
Priority to EP08846103.3A priority patent/EP2212354B1/en
Priority to DK08846103.3T priority patent/DK2212354T3/en
Publication of WO2009058492A2 publication Critical patent/WO2009058492A2/en
Publication of WO2009058492A3 publication Critical patent/WO2009058492A3/en
Priority to IL205050A priority patent/IL205050A0/en
Priority to IL231423A priority patent/IL231423A/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2863Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for growth factors, growth regulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/08Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses
    • C07K16/081Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses from DNA viruses
    • C07K16/082Hepadnaviridae, e.g. hepatitis B virus
    • 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
    • C07K16/22Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against growth factors ; against growth regulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2878Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30, CD40, CD95
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2893Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against CD52
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/32Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against translation products of oncogenes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype
    • C07K2317/524CH2 domain
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype
    • C07K2317/526CH3 domain
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin

Definitions

  • the present application relates to optimized IgG immunoglobulin variants, engineering methods for their generation, and their application, particularly for therapeutic purposes.
  • Antibodies are immunological proteins that bind a specific antigen. In most mammals, including humans and mice, antibodies are constructed from paired heavy and light polypeptide chains. Each chain is made up of individual immunoglobulin (Ig) domains, and thus the generic term immunoglobulin is used for such proteins. Each chain is made up of two distinct regions, referred to as the variable and constant regions. The light and heavy chain variable regions show significant sequence diversity between antibodies, and are responsible for binding the target antigen. The constant regions show less sequence diversity, and are responsible for binding a number of natural proteins to elicit important biochemical events.
  • IgA which includes subclasses IgAI and lgA2
  • IgD which includes subclasses IgAI and lgA2
  • IgE which includes subclasses IgG
  • IgM which includes subclasses IgGI , lgG2, lgG3, and lgG4
  • IgM IgM
  • Figure 1 shows an IgGI antibody, used here as an example to describe the general structural features of immunoglobulins.
  • IgG antibodies are tetrameric proteins composed of two heavy chains and two light chains.
  • the IgG heavy chain is composed of four immunoglobulin domains linked from N- to C-terminus in the order VH-CH 1-CH2-CH3, referring to the heavy chain variable domain, heavy chain constant domain 1 , heavy chain constant domain 2, and heavy chain constant domain 3 respectively (also referred to as VH-C ⁇ 1 -C ⁇ 2-C ⁇ 3, referring to the heavy chain variable domain, constant gamma 1 domain, constant gamma 2 domain, and constant gamma 3 domain respectively).
  • the IgG light chain is composed of two immunoglobulin domains linked from N- to C-terminus in the order VL-CL, referring to the light chain variable domain and the light chain constant domain respectively.
  • variable region of an antibody contains the antigen binding determinants of the molecule, and thus determines the specificity of an antibody for its target antigen.
  • the variable region is so named because it is the most distinct in sequence from other antibodies within the same class.
  • the majority of sequence variability occurs in the complementarity determining regions (CDRs).
  • CDRs complementarity determining regions
  • the variable region outside of the CDRs is referred to as the framework (FR) region.
  • FR framework
  • this characteristic architecture of antibodies provides a stable scaffold (the FR region) upon which substantial antigen binding diversity (the CDRs) can be explored by the immune system to obtain specificity for a broad array of antigens.
  • the CDRs substantial antigen binding diversity
  • a number of high-resolution structures are available for a variety of variable region fragments from different organisms, some unbound and some in complex with antigen.
  • Fragments including the variable region can exist in the absence of other regions of the antibody, including for example the antigen binding fragment (Fab) including VH-C ⁇ 1 and VH-CL, the variable fragment (Fv) including VH and VL, the single chain variable fragment (scFv) including VH and VL linked together in the same chain, as well as a variety of other variable region fragments (Little et al., 2000, Immunol Today 21 :364-370, entirely incorporated by reference).
  • Fab antigen binding fragment
  • Fv variable fragment
  • scFv single chain variable fragment
  • the Fc region of an antibody interacts with a number of Fc receptors and ligands, imparting an array of important functional capabilities referred to as effector functions.
  • the Fc region As shown in Figures 1 and 2, comprises Ig domains C ⁇ 2 and C ⁇ 3 and the N-terminal hinge leading into C ⁇ 2.
  • An important family of Fc receptors for the IgG class is the Fc gamma receptors (Fc ⁇ Rs). These receptors mediate communication between antibodies and the cellular arm of the immune system (Raghavan et al., 1996, Annu Rev Cell Dev Biol 12:181-220; Ravetch et al., 2001 , Annu Rev Immunol 19:275-290, both entirely incorporated by reference).
  • this protein family includes Fc ⁇ RI (CD64), including isoforms Fc ⁇ RIa, Fc ⁇ RIb, and Fc ⁇ RIc; Fc ⁇ RII (CD32), including isoforms Fc ⁇ Rlla (including allotypes H131 and R131 ), Fc ⁇ Rllb (including Fc ⁇ Rllb-1 and Fc ⁇ Rllb-2), and Fc ⁇ Rllc; and Fc ⁇ RIII (CD16), including isoforms Fc ⁇ Rllla (including allotypes V158 and F158) and Fc ⁇ Rlllb (including allotypes Fc ⁇ Rlllb-NA1 and Fc ⁇ Rlllb-NA2) (Jefferis et al., 2002, Immunol Lett 82:57-65, entirely incorporated by reference).
  • These receptors typically have an extracellular domain that mediates binding to Fc, a membrane spanning region, and an intracellular domain that may mediate some signaling event within the cell.
  • These receptors are expressed in a variety of immune cells including monocytes, macrophages, neutrophils, dendritic cells, eosinophils, mast cells, platelets, B cells, large granular lymphocytes, Langerhans' cells, natural killer (NK) cells, and ⁇ T cells.
  • NK natural killer
  • ADCC antibody dependent cell-mediated cytotoxicity
  • ADCP antibody dependent cell- mediated phagocytosis
  • the different IgG subclasses have different affinities for the Fc ⁇ Rs, with IgGI and lgG3 typically binding substantially better to the receptors than lgG2 and lgG4 (Jefferis et al., 2002, Immunol Lett 82:57-65, entirely incorporated by reference). All Fc ⁇ Rs bind the same region on IgG Fc, yet with different affinities: the high affinity binder Fc ⁇ RI has a Kd for IgGI of 10 "8 M "1 , whereas the low affinity receptors Fc ⁇ RI I and Fc ⁇ RI 11 generally bind at 10 "6 and 10 "5 respectively.
  • Fc ⁇ Rllla and Fc ⁇ Rlllb are 96% identical; however Fc ⁇ Rlllb does not have an intracellular signaling domain.
  • Fc ⁇ RI, Fc ⁇ Rlla/c, and Fc ⁇ Rllla are positive regulators of immune complex-triggered activation, characterized by having an intracellular domain that has an immunoreceptor tyrosine-based activation motif (ITAM)
  • Fc ⁇ Rllb has an immunoreceptor tyrosine-based inhibition motif (ITIM) and is therefore inhibitory.
  • the former are referred to as activation receptors
  • Fc ⁇ Rllb is referred to as an inhibitory receptor.
  • the receptors also differ in expression pattern and levels on different immune cells.
  • V158 allotype respond favorably to rituximab treatment; however, patients with the lower affinity F158 allotype respond poorly (Cartron et al., 2002, Blood 99:754-758, entirely incorporated by reference).
  • Approximately 10-20% of humans are V158/V158 homozygous, 45% are V158/F158 heterozygous, and 35-45% of humans are F158/F158 homozygous (Lehrnbecher et al., 1999, Blood 94:4220-4232; Cartron et al., 2002, Blood 99:754-758, all entirely incorporated by reference).
  • 80-90% of humans are poor responders, i.e., they have at least one allele of the F158 Fc ⁇ Rllla.
  • FIG. 1 An overlapping but separate site on Fc, shown in Figure 1 , serves as the interface for the complement protein C1q.
  • Fc/Fc ⁇ R binding mediates ADCC
  • Fc/C1q binding mediates complement dependent cytotoxicity (CDC).
  • C1 q forms a complex with the serine proteases C1 r and C1 s to form the C1 complex.
  • C1 q is capable of binding six antibodies, although binding to two IgGs is sufficient to activate the complement cascade.
  • IgG subclasses Similar to Fc interaction with Fc ⁇ Rs, different IgG subclasses have different affinity for C1 q, with IgGI and lgG3 typically binding substantially better to the Fc ⁇ Rs than lgG2 and lgG4 (Jefferis et al., 2002, Immunol Lett 82:57-65, entirely incorporated by reference).
  • the binding site on Fc for FcRn is also the site at which the bacterial proteins A and G bind.
  • the tight binding by these proteins is typically exploited as a means to purify antibodies by employing protein A or protein G affinity chromatography during protein purification.
  • the fidelity of this region on Fc is important for both the clinical properties of antibodies and their purification.
  • the FcRn receptor is also responsible for the transfer of IgG to the neo-natal gut and to the lumen of the intestinal epithelia in adults (Ghetie and Ward, Annu. Rev. Immunol., 2000, 18:739-766; Yoshida et al., Immunity, 2004, 20(6):769-783, both entirely incorporated by reference).
  • Studies of rat and human Fc domains have demonstrated the importance of some Fc residues to the binding of FcRn.
  • the rat and human sequences have about 64% sequence identity in the Fc regions (residues 237-443 in the numbering of Kabat et al.).
  • Hinton et al. found that some point mutants a double mutant increased the FcRn binding affinity (Hinton et al., 2004, J. Biol. Chem. 279(8): 6213-6216. Hinton et al. Journal of Immunology 2006, 176:346-356, both entirely incorporated by reference). Two mutations had increased half-lives in monkeys. Shields et al. mutated residues, almost exclusively to Ala, and studied their binding to FcRn and the FcyR's (Shields et al., 2001 , J. Biol. Chem., 276(9):6591-6604, entirely incorporated by reference).
  • Dall'Acqua et al. used phage display to select for Fc mutations that bound FcRn with increased affinity (DaN' Acqua et al. 2002, J. Immunol. 169:5171-5180, entirely incorporated by reference).
  • the DNA sequences selected for were primarily double and triple mutants.
  • the reference expressed the proteins encoded by many of their selected sequences and found some that bound to FcRn more tightly than the wild-type Fc.
  • the administration of antibodies and Fc fusion proteins as therapeutics requires injections with a prescribed frequency relating to the clearance and half-life characteristics of the protein. Longer in vivo half-lives allow more seldom injections or lower dosing, which is clearly advantageous.
  • the past mutations in the Fc domain have lead to some proteins with increased FcRn binding affinity and in vivo half-lives, these mutations have not identified the optimal mutations and enhanced in vivo half-life.
  • Fc region is the conserved N-linked glycosylation that occurs at N297, shown in Figure 1. This carbohydrate, or oligosaccharide as it is sometimes referred, plays a critical structural and functional role for the antibody, and is one of the principle reasons that antibodies must be produced using mammalian expression systems.
  • Antibodies have been developed for therapeutic use. Representative publications related to such therapies include Chamow et ai, 1996, Trends Biotechnol 14:52-60; Ashkenazi et al., 1997, Curr Opin Immunol 9: 195-200, Cragg et al., 1999, Curr Opin Immunol 1 1 :541-547; Glennie et al., 2000, Immunol Today 21 :403-410, McLaughlin et al., 1998, J Clin Oncol 16:2825-2833, and Cobleigh et al., 1999, J Clin Oncol 17:2639-2648, all entirely incorporated by reference. Currently for anticancer therapy, any small improvement in mortality rate defines success.
  • Anti-tumor potency of antibodies is via enhancement of their ability to mediate cytotoxic effector functions such as ADCC, ADCP, and CDC. Examples include Clynes et ai, 1998, Proc Natl Acad Sci U S A 95:652-656; Clynes et ai, 2000, Nat Med 6:443-446 and Cartron et al., 2002, Blood 99:754-758, both entirely incorporated by reference.
  • Human IgGI is the most commonly used antibody for therapeutic purposes, and the majority of engineering studies have been constructed in this context.
  • IgGI , lgG2, lgG3, and lgG4 have unique physical, biological, and clinical properties.
  • IgGI , lgG2, lgG3, and lgG4 variants There is a need in the art to design improved IgGI , lgG2, lgG3, and lgG4 variants.
  • design such variants to improve binding to FcRn and/or increase in vivo half-life as compared to native IgG polypeptides.
  • variants with improved pharmacokinetic properties with variants comprising modifications to improve efficacy through altered FcgammaR binding The present application meets these and other needs.
  • the present application is directed to Fc variants of a parent polypeptide including at least one modification in the Fc region of the polypeptide.
  • the variant polypeptides exhibit altered binding to FcRn as compared to a parent polypeptide.
  • the modification can be selected from the group consisting of: 246H, 246S, 247D, 247T, 248H, 248P, 248Q, 248R, 248Y, 249T, 249W, 250E, 250I, 250Q, 250V, 251 D, 251 E, 251 H, 2511, 251 K, 251 M, 251 N, 251T, 251V, 251Y, 252Q, 252Y, 253L, 253T, 253V, 254H, 254L, 254N, 254T, 254V, ⁇ 254N, 255E, 255F, 255H, 255K, 255S, 255V, 256E, 256V, 257A, 257C, 257D, 257E, 257F, 257G, 257H, 257I, 257K, 257L, 257M, 257N, 257Q, 257R, 257S, 257T, 257
  • the Fc variant includes at least two modifications selected from the group consisting of: 250Q/252Y, 250Q/256E, 250Q/286D, 250Q/308F, 250Q/308Y, 250Q/31 1A, 250Q/31 1V, 250Q/380A, 250Q/428L, 250Q/428F, 250Q/434H, 250Q/434F, 250Q/434Y, 250Q/434A, 250Q/434M, and 250Q/434S.
  • the Fc variant includes at least two modifications selected from the group consisting of: 250E/252Y, 250E/256E, 250E/286D, 250E/308F, 250E/308Y, 250E/31 1A, 250E/31 1V, 250E/380A, 250E/428L, 250E/428F, 250E/434H, 250E/434F, 250E/434Y, 250E/434A, 250E/434M, and 250E/434S.
  • the Fc variant includes at least two modifications selected from the group consisting of: 252Y/250Q, 252Y/250E, 252Y/256E, 252Y/286D, 252Y/308F, 252Y/308Y, 252Y/31 1A, 252Y/311V, 252Y/380A, 252Y/428L, 252Y/428F, 252Y/434H, 252Y/434F, 252Y/434Y, 252Y/434A, 252Y/434M, and 252Y/434S.
  • the Fc variant includes at least two modifications selected from the group consisting of: 256E/250Q, 256E/250E, 256E/252Y, 256E/286D, 256E/308F, 256E/308Y, 256E/31 1A, 256E/311V, 256E/380A, 256E/428L, 256E/428F, 256E/434H, 256E/434F, 256E/434Y, 256E/434A, 256E/434M, and 256E/434S.
  • the Fc variant includes at least two modifications selected from the group consisting of: 286D/250Q, 286D/250E, 286D/252Y, 286D/256E, 286D/308F, 286D/308Y, 286D/31 1A, 286D/31 1V, 286D/380A, 286D/428L, 286D/428F, 286D/434H,
  • the Fc variant includes at least two modifications selected from the group consisting of: 308F/250Q, 308F/250E, 308F/252Y, 308F/256E, 308F/286D,
  • the Fc variant includes at least two modifications selected from the group consisting of: 308Y/250Q, 308Y/250E, 308Y/252Y, 308Y/256E, 308Y/286D,
  • the Fc variant includes at least two modifications selected from the group consisting of: 311 A/250Q, 311 A/250E, 31 1A/252Y, 31 1A/256E, 311 A/286D,
  • the Fc variant includes at least two modifications selected from the group consisting of: 311 V/250Q, 311 V/250E, 31 1V/252Y, 31 1V/256E, 311 V/286D,
  • the Fc variant includes at least two modifications selected from the group consisting of: 380A/250Q, 380A/250E, 380A/252Y, 380A/256E, 380A/286D,
  • the Fc variant includes at least two modifications selected from the group consisting of: 428L/250Q, 428L/250E, 428L/252Y, 428L/256E, 428L/286D,
  • the Fc variant includes at least two modifications selected from the group consisting of: 434H/250Q, 434H/250E, 434H/252Y, 434H/256E, 434H/286D,
  • the Fc variant includes at least two modifications selected from the group consisting of: 434F/250Q, 434F/250E, 434F/252Y, 434F/256E, 434F/286D,
  • the Fc variant includes at least two modifications selected from the group consisting of: 434Y/250Q, 434Y/250E, 434Y/252Y, 434Y/256E, 434Y/286D,
  • the Fc variant includes at least two modifications selected from the group consisting of: 434A/250Q, 434A/250E, 434A/252Y, 434A/256E, 434A/286D, 434A/308F, 434A/308Y, 434A/31 1A, 434A/31 1V, 434A/380A, 434A/428L, and 434A/428F.
  • the Fc variant includes at least two modifications selected from the group consisting of: 434M/250Q, 434M/250E, 434M/252Y, 434M/256E, 434M/286D, 434M/308F, 434M/308Y, 434M/311A, 434M/311V, 434M/380A, 434M/428L, and 434M/428F.
  • the Fc variant includes at least two modifications selected from the group consisting of: 434S/250Q, 434S/250E, 434S/252Y, 434S/256E, 434S/286D, 434S/308F, 434S/308Y, 434S/31 1A, 434S/31 1V, 434S/380A, 434S/428L, and 434S/428F.
  • the Fc variant includes at least one modification selected from the group consisting of: Y319L, T307Q, V259I, M252Y, V259I/N434S, M428L/N434S, V308F/N434S, M252Y/S254T/T256E/N434S, M252Y/S254T/T256E/V308F, M252Y/S254T/T256E/M428L, V308F/M428L/N434S, V259I/V308F/N434S, T307Q/V308F/N434S, T250I/V308F/N434S, V308F/Y319L/N434S, V259I/V308F/M428L, V259I/T307Q/V308F, T250I/V259I/V308F, V259I/T307Q/V
  • the Fc variant includes at least one modification selected from the group consisting of: Y319L, T307Q, V259I, M252Y, V259I/N434S, M428L/N434S, V308F/N434S, V308F/M428L/N434S, V259I/V308F/N434S, T307Q/V308F/N434S, T250I/V308F/N434S, V308F/Y319L/N434S, V259I/V308F/M428L, V259I/T307Q/V308F, T250I/V259I/V308F, V259I/V308F/Y319L, T307Q/V308F/L309Y, T307Q/V308F/Y319L, and T250Q/V308F/M428L.
  • the Fc variant includes at least one modification selected from the group consisting of: 250I, 250V, 252Q, 252Y, 254T, 256V, 259I, 307P, 307Q, 307S, 308F, 309N, 309Y, 311 P, 319F, 319L, 428L, and 434S.
  • the Fc variant includes at least one modification selected from the group consisting of: 250V/308F, 250I/308F, 254T/308F, 256V/308F, 259I/308F, 307P/208F, 307Q/308F, 307S/308F, 308F/309Y, 308F/309Y, V308F/311 P, 308F/319L, 308F/319F, 308F/428L, 252Q/308F, M252Y/S254T/T256E, 259I/434S, 428L/434S, 308F/434S, 308F/428L/434S, 259I/308F/434S, 307Q/308F/434S, 250I/308F/434S, 308F/319L/434S, 259I/308F/428L, 259I/307Q/308F, 250I/259I/308F, 259I/308F/319L, 307Q/3
  • the invention includes a method of treating a patient in need of said treatment comprising administering an effective amount of an Fc variant described herein.
  • the invention includes a method of increasing the half-life of an antibody or immunoadhesin by modifying an Fc according to the modifications described herein.
  • FIG. 1 Antibody structure and function. Shown is a model of a full length human IgGI antibody, modeled using a humanized Fab structure from pdb accession code 1 CE1 (James et al., 1999, J MoI Biol 289:293-301 , entirely incorporated by reference) and a human IgGI Fc structure from pdb accession code 1 DN2 (DeLano et al., 2000, Science 287:1279-1283, entirely incorporated by reference). The flexible hinge that links the Fab and Fc regions is not shown. IgGI is a homodimer of heterodimers, made up of two light chains and two heavy chains.
  • the Ig domains that comprise the antibody are labeled, and include V L and C L for the light chain, and V H , Cgammai (C ⁇ 1 ), Cgamma2 (C ⁇ 2), and Cgamma3 (C ⁇ 3) for the heavy chain.
  • the Fc region is labeled. Binding sites for relevant proteins are labeled, including the antigen binding site in the variable region, and the binding sites for Fc ⁇ Rs, FcRn, C1 q, and proteins A and G in the Fc region.
  • FIG. 5 Example human and rodent IgG sequences (SEQ ID NOS: 5-6) used in the present invention with the EU numbering as in Kabat.
  • FIG. 4 Example human and rodent FcRn heavy chain sequences (SEQ ID NOS: 7-8) used in the present invention.
  • FIG. 9 Example human and rodent beta-2-microglobulin sequences (SEQ ID NOS: 9-10) used in the present invention.
  • Figure 6 A human Fc/FcRn complex model created from the rat structures (Burmeister et al., 1994, Nature, 372:379-383; Martin et al., 2001 , MoI Cell 7:867-877, both entirely incorporated by reference). Some histidine residues are shown in space-filling atoms on the FcRn chains (light grey) and Fc polypeptide (dark grey).
  • FIG. 11 Diagram of the vector pcDNA3.1 Zeo+, which may be used in the construct of Fc variants.
  • FIG. 12a-12b Competition FcRn binding data of wild-type Fc and Fc variants of the present invention.
  • the Fc variants of the present invention are shown as the left (red or dark grey) curve and the wild-type anti-HER2 antibody is shown as the right (blue or light grey) curve.
  • FIGs 13a - 13j Summary of FcRn binding properties of the Fc variants.
  • the columns from left to right show the FcRn binding modifications, the immunoglobulin used, other modifications, the relative FcRn affinity by AlphaScreenTM competition assays compared to wild type (median value), and the number of assays performed.
  • Relative FcRn affinity numbers greater than 1.0 demonstrate increased binding over wild type. Data were collected at pH 6.0 (0.1 M sodium phosphate, 25mM sodium chloride).
  • Figure 14a-14d FcRn binding data of Fc variants.
  • the Fc variants are in alemtuzumab or anti-HER2 antibody. Shown are the fold-increases in binding compared to wild type, that is, numbers greater than one indicate tighter binding to FcRn whereas numbers less than one indicate reduced binding to FcRn.
  • FIG. 15 Summary of surface plasmon resonance experiments of Fc variants with improved binding to FcRn.
  • the bar graph shows the fold-increase in FcRn binding affinity of each variant relative to wild-type Fc domain.
  • FIG. 16a-16b Surface plasmon resonance experiments of wild-type antibody and variants of the present invention. The traces shown are the association and dissociation of the Fc variant antibody to FcRn at pH6.0.
  • FIG. 17a-17c Binding assays of Fc variants of the present invention to FcRn. Shown are direct binding assays measured by AlphaScreenTM at pH 6.0 (a and b) and pH 7.0 (C).
  • FIG. Binding assays of Fc variants of the present invention to FcRn. Shown are the surface plasmon resonance units created upon binding of the variant Fc to surface- bound FcRn.
  • FIG. 20 Summary of surface plasmon resonance (SPR) measurements of the binding affinity of Fc variants of the present invention with human, macaque and mouse FcRn. Numbers greater than one indicate increased binding of the variant Fc to FcRn as determined by fitting SPR curves to a 1 :1 Langmuir binding model.
  • Figure 21a-21 b Summary of FcRn binding properties of the Fc variants. The columns from left to right show the FcRn binding modifications, the immunoglobulin used, other modifications, the relative FcRn affinity by AlphaScreenTM competition assays compared to wild type (median value), and the number of assays performed. Relative FcRn affinity numbers greater than 1.0 demonstrate increased binding over wild type. Data were collected at pH 6.0 (0.1 M sodium phosphate, 125mM sodium chloride).
  • Figure 22 Amino acid sequences (SEQ ID NO: 1 1-12) of the anti-HER2 antibody heavy and light chains.
  • FIG. 13 Amino acid sequences (SEQ ID NO: 13-18) of the constant regions (CH 1 to CH3) of the some IgGI heavy chains used herein.
  • FIG. 25 Fc variants binding to the human FcgammaRIIIA (V158 Allotype) as determined with AlphaScreenTM competition assays.
  • Figure 26 Fc variants binding binding to protein A as determined with AlphaScreenTM competition assays.
  • FIG. Serum concentrations of WT and variants of antibodies in human FcRn knockin mice.
  • Anti-VEGF antibodies used were the WT (open squares), V308F (closed squares), P257L (closed triagles) and P257N (crosses).
  • FIG. 28 Examples of FcRn binding variants of the present invention.
  • Anti-VEGF antibodies are listed with the volume of culture media and the yield of purified protein.
  • FIG. 29 Binding affinity of variants of the present invention to human FcRn at pH6.0. The values shown are fold increase in binding strength of the variant in question to the wild-type antibody. For example, the variant 434S binds to FcRn 4.4-fold more tightly than does the wild-type antibody.
  • FIG. 30 Binding of WT and variant antibodies to FcRn on the surface of 293T cells.
  • Figure 31 a-31 b Combination variants of the present invention comprising multiple substitutions.
  • the present invention discloses the generation of novel variants of Fc domains, including those found in antibodies, Fc fusions, and immuno-adhesions, which have an increased binding to the FcRn receptor. As noted herein, binding to FcRn results in longer serum retention in vivo.
  • An additional aspect of the invention is the increase in FcRn binding over wild type specifically at lower pH, about pH 6.0, to facilitate Fc/FcRn binding in the endosome. Also disclosed are Fc variants with altered FcRn binding and altered binding to another class of Fc receptors, the Fc ⁇ R's (sometimes written FcgammaR's) as differential binding to Fc ⁇ Rs, particularly increased binding to Fc ⁇ Rlllb and decreased binding to Fc ⁇ Rllb, has been shown to result in increased efficacy. [78] Definitions
  • ADCC antibody dependent cell-mediated cytotoxicity
  • ADCP antibody dependent cell-mediated phagocytosis
  • modification herein is meant an amino acid substitution, insertion, and/or deletion in a polypeptide sequence or an alteration to a moiety chemically linked to a protein.
  • a modification may be an altered carbohydrate or PEG structure attached to a protein.
  • amino acid modification herein is meant an amino acid substitution, insertion, and/or deletion in a polypeptide sequence.
  • amino acid substitution or “substitution” herein is meant the replacement of an amino acid at a particular position in a parent polypeptide sequence with another amino acid.
  • substitution E272Y refers to a variant polypeptide, in this case an Fc variant, in which the glutamic acid at position 272 is replaced with tyrosine.
  • amino acid insertion or “insertion” as used herein is meant the addition of an amino acid sequence at a particular position in a parent polypeptide sequence.
  • -233E or ⁇ 233E designates an insertion of glutamic acid after position 233 and before position 234.
  • -233ADE or ⁇ 233ADE designates an insertion of AlaAspGlu after position 233 and before position 234.
  • amino acid deletion or “deletion” as used herein is meant the removal of an amino acid sequence at a particular position in a parent polypeptide sequence.
  • E233- or E233# designates a deletion of glutamic acid at position 233.
  • EDA233- or EDA233# designates a deletion of the sequence GluAspAla that begins at position 233.
  • variant protein or “protein variant”, or “variant” as used herein is meant a protein that differs from that of a parent protein by virtue of at least one amino acid modification. Protein variant may refer to the protein itself, a composition comprising the protein, or the amino sequence that encodes it.
  • the protein variant has at least one amino acid modification compared to the parent protein, e.g. from about one to about ten amino acid modifications, and preferably from about one to about five amino acid modifications compared to the parent.
  • the protein variant sequence herein will preferably possess at least about 80% homology with a parent protein sequence, and most preferably at least about 90% homology, more preferably at least about 95% homology.
  • Variant protein can refer to the variant protein itself, compositions comprising the protein variant, or the DNA sequence that encodes it.
  • antibody variant or “variant antibody” as used herein is meant an antibody that differs from a parent antibody by virtue of at least one amino acid modification
  • IgG variant or “variant IgG” as used herein is meant an antibody that differs from a parent IgG by virtue of at least one amino acid modification
  • immunoglobulin variant or “variant immunoglobulin” as used herein is meant an immunoglobulin sequence that differs from that of a parent immunoglobulin sequence by virtue of at least one amino acid modification
  • Fc variant or “variant Fc” as used herein is meant a protein comprising a modification in an Fc domain. The Fc variants of the present invention are defined according to the amino acid modifications that compose them.
  • I332E is an Fc variant with the substitution I332E relative to the parent Fc polypeptide.
  • S239D/I332E/G236A defines an Fc variant with the substitutions S239D, I332E, and G236A relative to the parent Fc polypeptide.
  • the identity of the WT amino acid may be unspecified, in which case the aforementioned variant is referred to as 239D/332E/236A. It is noted that the order in which substitutions are provided is arbitrary, that is to say that, for example, S239D/I332E/G236A is the same Fc variant as G236A/S239D/I332E, and so on.
  • EU index or EU numbering is according to the EU index or EU numbering scheme (Kabat et al., 1991 , Sequences of Proteins of Immunological Interest, 5th Ed., United States Public Health Service, National Institutes of Health, Bethesda, hereby entirely incorporated by reference).
  • the EU index or EU index as in Kabat or EU numbering scheme refers to the numbering of the EU antibody (Edelman et al., 1969, Proc Natl Acad Sci USA 63:78-85, hereby entirely incorporated by reference.)
  • the modification can be an addition, deletion, or substitution. Substitutions can include naturally occurring amino acids and non-naturally occurring amino acids. Variants may comprise non-natural amino acids.
  • Examples include US6586207; WO 98/48032; WO 03/073238; US2004-0214988A1 ; WO 05/35727A2; WO 05/74524A2; J. W. Chin et al., (2002), Journal of the American Chemical Society 124:9026-9027; J. W. Chin, & P. G. Schultz, (2002), ChemBioChem 11 :1 135-1137; J. W. Chin, et al., (2002), PICAS United States of America 99:11020-1 1024; and, L. Wang, & P. G. Schultz, (2002), Chem. 1-10, all entirely incorporated by reference.
  • protein herein is meant at least two covalently attached amino acids, which includes proteins, polypeptides, oligopeptides and peptides.
  • the peptidyl group may comprise naturally occurring amino acids and peptide bonds, or synthetic peptidomimetic structures, i.e. "analogs", such as peptoids (see Simon et al., PNAS USA 89(20):9367 (1992), entirely incorporated by reference).
  • the amino acids may either be naturally occurring or non-naturally occurring; as will be appreciated by those in the art.
  • homo-phenylalanine, citrulline, and noreleucine are considered amino acids for the purposes of the invention, and both D- and L- (R or S) configured amino acids may be utilized.
  • the variants of the present invention may comprise modifications that include the use of unnatural amino acids incorporated using, for example, the technologies developed by Schultz and colleagues, including but not limited to methods described by Cropp & Shultz, 2004, Trends Genet. 20(12):625-30, Anderson et al., 2004, Proc Natl Acad Sci USA 101 (2):7566-71 , Zhang et al., 2003, 303(5656):371-3, and Chin et al., 2003, Science 301 (5635):964-7, all entirely incorporated by reference.
  • polypeptides may include synthetic derivatization of one or more side chains or termini, glycosylation, PEGylation, circular permutation, cyclization, linkers to other molecules, fusion to proteins or protein domains, and addition of peptide tags or labels.
  • residue as used herein is meant a position in a protein and its associated amino acid identity.
  • Asparagine 297 also referred to as Asn297, also referred to as N297
  • Asn297 is a residue in the human antibody IgGI .
  • Fab or "Fab region” as used herein is meant the polypeptide that comprises the VH, CH1 , VL, and CL immunoglobulin domains. Fab may refer to this region in isolation, or this region in the context of a full length antibody, antibody fragment or Fab fusion protein.
  • IqG subclass modification as used herein is meant an amino acid modification that converts one amino acid of one IgG isotype to the corresponding amino acid in a different, aligned IgG isotype. For example, because IgGI comprises a tyrosine and lgG2 a phenylalanine at EU position 296, a F296Y substitution in lgG2 is considered an IgG subclass modification.
  • non-naturally occurring modification is meant an amino acid modification that is not isotypic.
  • the substitution I332E in IgGI , lgG2, lgG3, or lgG4 is considered a non-natural occuring modification.
  • amino acid and “amino acid identity” as used herein is meant one of the 20 naturally occurring amino acids or any non-natural analogues that may be present at a specific, defined position.
  • effector function as used herein is meant a biochemical event that results from the interaction of an antibody Fc region with an Fc receptor or ligand. Effector functions include but are not limited to ADCC, ADCP, and CDC.
  • effector cell as used herein is meant a cell of the immune system that expresses one or more Fc receptors and mediates one or more effector functions. Effector cells include but are not limited to monocytes, macrophages, neutrophils, dendritic cells, eosinophils, mast cells, platelets, B cells, large granular lymphocytes, Langerhans' cells, natural killer (NK) cells, and ⁇ T cells, and may be from any organism including but not limited to humans, mice, rats, rabbits, and monkeys.
  • IgG Fc ligand as used herein is meant a molecule, preferably a polypeptide, from any organism that binds to the Fc region of an IgG antibody to form an Fc / Fc ligand complex.
  • Fc ligands include but are not limited to Fc ⁇ Rs, Fc ⁇ Rs, Fc ⁇ Rs, FcRn, C1q, C3, mannan binding lectin, mannose receptor, staphylococcal protein A, streptococcal protein G, and viral Fc ⁇ R.
  • Fc ligands also include Fc receptor homologs (FcRH), which are a family of Fc receptors that are homologous to the Fc ⁇ Rs (Davis et al., 2002, Immunological Reviews 190:123-136, entirely incorporated by reference).
  • Fc ligands may include undiscovered molecules that bind Fc. Particular IgG Fc ligands are FcRn and Fc gamma receptors.
  • Fc ligand as used herein is meant a molecule, preferably a polypeptide, from any organism that binds to the Fc region of an antibody to form an Fc / Fc ligand complex.
  • Fc gamma receptor any member of the family of proteins that bind the IgG antibody Fc region and is encoded by an Fc ⁇ R gene.
  • this family includes but is not limited to Fc ⁇ RI (CD64), including isoforms Fc ⁇ RIa, Fc ⁇ RIb, and Fc ⁇ RIc; Fc ⁇ RII (CD32), including isoforms Fc ⁇ Rlla (including allotypes H131 and R131 ), Fc ⁇ Rllb (including Fc ⁇ Rllb-1 and Fc ⁇ Rllb-2), and Fc ⁇ Rllc; and Fc ⁇ RIII (CD16), including isoforms Fc ⁇ Rllla (including allotypes V158 and F158) and Fc ⁇ Rlllb (including allotypes Fc ⁇ Rlllb-NA1 and Fc ⁇ Rlllb-NA2) (Jefferis et al., 2002, Immunol Lett 82:57-65, entirely incorporated by reference), as well as any undiscovered human Fc ⁇ Rs or Fc ⁇ R isoforms or allotypes.
  • An Fc ⁇ R may be from any organism, including but not limited to humans, mice, rats, rabbits, and monkeys.
  • Mouse Fc ⁇ Rs include but are not limited to Fc ⁇ RI (CD64), Fc ⁇ RII (CD32), Fc ⁇ RIII (CD16), and Fc ⁇ RIII-2 (CD16-2), as well as any undiscovered mouse Fc ⁇ Rs or Fc ⁇ R isoforms or allotypes.
  • FcRn or "neonatal Fc Receptor” as used herein is meant a protein that binds the IgG antibody Fc region and is encoded at least in part by an FcRn gene.
  • the FcRn may be from any organism, including but not limited to humans, mice, rats, rabbits, and monkeys.
  • the functional FcRn protein comprises two polypeptides, often referred to as the heavy chain and light chain.
  • the light chain is beta-2-microglobulin and the heavy chain is encoded by the FcRn gene.
  • FcRn or an FcRn protein refers to the complex of FcRn heavy chain with beta-2-microglobulin. Sequences of particular interest of FcRn are shown in the Figures, particularly the human species.
  • parent polypeptide as used herein is meant an unmodified polypeptide that is subsequently modified to generate a variant.
  • parent polypeptide may be a naturally occurring polypeptide, or a variant or engineered version of a naturally occurring polypeptide.
  • Parent polypeptide may refer to the polypeptide itself, compositions that comprise the parent polypeptide, or the amino acid sequence that encodes it.
  • parent immunoglobulin as used herein is meant an unmodified immunoglobulin polypeptide that is modified to generate a variant
  • parent antibody as used herein is meant an unmodified antibody that is modified to generate a variant antibody. It should be noted that "parent antibody” includes known commercial, recombinantly produced antibodies as outlined below.
  • position as used herein is meant a location in the sequence of a protein. Positions may be numbered sequentially, or according to an established format, for example the EU index as in Kabat. For example, position 297 is a position in the human antibody IgGL
  • target antigen as used herein is meant the molecule that is bound specifically by the variable region of a given antibody.
  • a target antigen may be a protein, carbohydrate, lipid, or other chemical compound.
  • target cell as used herein is meant a cell that expresses a target antigen.
  • variant region as used herein is meant the region of an immunoglobulin that comprises one or more Ig domains substantially encoded by any of the VK, V ⁇ , and/or VH genes that make up the kappa, lambda, and heavy chain immunoglobulin genetic loci respectively.
  • wild type or WT herein is meant an amino acid sequence or a nucleotide sequence that is found in nature, including allelic variations.
  • a WT protein has an amino acid sequence or a nucleotide sequence that has not been intentionally modified.
  • the present invention is directed to antibodies that exhibit moduluated binding to FcRn (modulation including increased as well as decreased binding). For example, in some instances, increased binding results in cellular recycling of the antibody and hence increased half-life, for example for therapeutic antibodies. Alternatively, decreased FcRn binding is desirable, for example for diagnostic antibodies or therapeutic antibodies that contain radiolabels. In addition, antibodies exhibiting increased binding to FcRn and altered binding to other Fc receptors, eg. FcvRs, find use in the present invention. Accordingly, the present invention provides antibodies. Antibodies
  • the present application is directed to antibodies that include amino acid modifications that modulate binding to FcRn.
  • Traditional antibody structural units typically comprise a tetramer. Each tetramer is typically composed of two identical pairs of polypeptide chains, each pair having one "light” (typically having a molecular weight of about 25 kDa) and one "heavy” chain (typically having a molecular weight of about 50-70 kDa). Human light chains are classified as kappa and lambda light chains. Heavy chains are classified as mu, delta, gamma, alpha, or epsilon, and define the antibody's isotype as IgM, IgD, IgG, IgA, and IgE, respectively.
  • IgG has several subclasses, including, but not limited to IgGI , lgG2, lgG3, and lgG4.
  • IgM has subclasses, including, but not limited to, IgMI and lgM2.
  • isotype as used herein is meant any of the subclasses of immunoglobulins defined by the chemical and antigenic characteristics of their constant regions.
  • the known human immunoglobulin isotypes are IgGI , lgG2, lgG3, lgG4, IgAI , lgA2, IgMI , lgM2, IgD, and IgE.
  • each chain includes a variable region of about 100 to 110 or more amino acids primarily responsible for antigen recognition.
  • variable region three loops are gathered for each of the V domains of the heavy chain and light chain to form an antigen-binding site.
  • Each of the loops is referred to as a complementarity- determining region (hereinafter referred to as a "CDR"), in which the variation in the amino acid sequence is most significant.
  • CDR complementarity- determining region
  • each chain defines a constant region primarily responsible for effector function.
  • Kabat et al. collected numerous primary sequences of the variable regions of heavy chains and light chains. Based on the degree of conservation of the sequences, they classified individual primary sequences into the CDR and the framework and made a list thereof (see SEQUENCES OF IMMUNOLOGICAL INTEREST, 5th edition, NIH publication, No. 91-3242, E.A. Kabat et al., entirely incorporated by reference).
  • immunoglobulin domains in the heavy chain.
  • immunoglobulin (Ig) domain herein is meant a region of an immunoglobulin having a distinct tertiary structure.
  • the heavy chain domains including, the constant heavy (CH) domains and the hinge domains.
  • the IgG isotypes each have three CH regions. Accordingly, "CH” domains in the context of IgG are as follows: “CH 1" refers to positions 118-220 according to the EU index as in Kabat. "CH2" refers to positions 237-340 according to the EU index as in Kabat, and “CH3” refers to positions 341-447 according to the EU index as in Kabat.
  • Ig domain of the heavy chain is the hinge region.
  • hinge region or “hinge region” or “antibody hinge region” or “immunoglobulin hinge region” herein is meant the flexible polypeptide comprising the amino acids between the first and second constant domains of an antibody.
  • the IgG CH1 domain ends at EU position 220, and the IgG CH2 domain begins at residue EU position 237.
  • the antibody hinge is herein defined to include positions 221 (D221 in IgGI ) to 236 (G236 in IgGI ), wherein the numbering is according to the EU index as in Kabat.
  • the lower hinge is included, with the “lower hinge” generally referring to positions 226 or 230.
  • Fc regions are the Fc regions.
  • Fc or “Fc region”, as used herein is meant the polypeptide comprising the constant region of an antibody excluding the first constant region immunoglobulin domain and in some cases, part of the hinge.
  • Fc refers to the last two constant region immunoglobulin domains of IgA, IgD, and IgG, the last three constant region immunoglobulin domains of IgE and IgM, and the flexible hinge N-terminal to these domains.
  • Fc may include the J chain.
  • Fc comprises immunoglobulin domains Cgamma2 and Cgamma3 (Cg2 and Cg3) and the lower hinge region between Cgammai (Cg1 ) and Cgamma2 (Cg2).
  • Cg2 and Cg3 immunoglobulin domains Cgamma2 and Cgamma3
  • Cg1 Cgammai
  • Cg2 and Cg3 the human IgG heavy chain Fc region
  • the human IgG heavy chain Fc region is usually defined to include residues C226 or P230 to its carboxyl-terminus, wherein the numbering is according to the EU index as in Kabat.
  • Fc may refer to this region in isolation, or this region in the context of an Fc polypeptide, as described below.
  • Fc polypeptide as used herein is meant a polypeptide that comprises all or part of an Fc region.
  • Fc polypeptides include antibodies, Fc fusions, isolated Fes, and Fc fragments.
  • the antibodies are full length.
  • full length antibody herein is meant the structure that constitutes the natural biological form of an antibody, including variable and constant regions, including one or more modifications as outlined herein.
  • the antibodies can be a variety of structures, including, but not limited to, antibody fragments, monoclonal antibodies, bispecific antibodies, minibodies, domain antibodies, synthetic antibodies (sometimes referred to herein as "antibody mimetics"), chimeric antibodies, humanized antibodies, antibody fusions (sometimes referred to as "antibody conjugates”), and fragments of each, respectively.
  • Antibody Fragments include antibodies, Fc fusions, isolated Fes, and Fc fragments.
  • the antibody is an antibody fragment.
  • antibodies that comprise Fc regions, Fc fusions, and the constant region of the heavy chain (CH1-hinge-CH2-CH3), again also including constant heavy region fusions.
  • Specific antibody fragments include, but are not limited to, (i) the Fab fragment consisting of VL, VH, CL and CH 1 domains, (ii) the Fd fragment consisting of the VH and CH1 domains, (iii) the Fv fragment consisting of the VL and VH domains of a single antibody; (iv) the dAb fragment (Ward et al., 1989, Nature 341 :544-546, entirely incorporated by reference) which consists of a single variable, (v) isolated CDR regions, (vi) F(ab')2 fragments, a bivalent fragment comprising two linked Fab fragments (vii) single chain Fv molecules (scFv), wherein a VH domain and a VL domain are linked by
  • the scaffold components can be a mixture from different species.
  • the protein is an antibody
  • such antibody may be a chimeric antibody and/or a humanized antibody.
  • both “chimeric antibodies” and “humanized antibodies” refer to antibodies that combine regions from more than one species.
  • “chimeric antibodies” traditionally comprise variable region(s) from a mouse (or rat, in some cases) and the constant region(s) from a human.
  • “Humanized antibodies” generally refer to non-human antibodies that have had the variable-domain framework regions swapped for sequences found in human antibodies.
  • a humanized antibody the entire antibody, except the CDRs, is encoded by a polynucleotide of human origin or is identical to such an antibody except within its CDRs.
  • the CDRs some or all of which are encoded by nucleic acids originating in a non-human organism, are grafted into the beta- sheet framework of a human antibody variable region to create an antibody, the specificity of which is determined by the engrafted CDRs.
  • the creation of such antibodies is described in, e.g., WO 92/1 1018, Jones, 1986, Nature 321 :522-525, Verhoeyen et al., 1988, Science 239:1534-1536, all entirely incorporated by reference.
  • the humanized antibody optimally also will comprise at least a portion of an immunoglobulin constant region, typically that of a human immunoglobulin, and thus will typically comprise a human Fc region. Humanized antibodies can also be generated using mice with a genetically engineered immune system. Roque et al., 2004, Biotechnol. Prog.
  • Humanization methods include but are not limited to methods described in Jones et al., 1986, Nature 321 :522-525; Riechmann et a/., 1988; Nature 332:323-329; Verhoeyen et al., 1988, Science, 239:1534-1536; Queen et al., 1989, Proc Natl Acad Sci, USA 86:10029-33; He et al., 1998, J. Immunol. 160: 1029-1035; Carter et al., 1992, Proc Natl Acad Sci USA 89:4285-9, Presta et al., 1997, Cancer Res.57(20):4593-9; Gorman et al., 1991 , Proc. Natl.
  • Humanization or other methods of reducing the immunogenicity of nonhuman antibody variable regions may include resurfacing methods, as described for example in Roguska et al., 1994, Proc. Natl. Acad. Sci. USA 91 :969-973, entirely incorporated by reference.
  • the parent antibody has been affinity matured, as is known in the art. Structure-based methods may be employed for humanization and affinity maturation, for example as described in USSN 11/004,590.
  • Selection based methods may be employed to humanize and/or affinity mature antibody variable regions, including but not limited to methods described in Wu et al., 1999, J. MoI. Biol. 294:151-162; Baca et al., 1997, J. Biol. Chem. 272(16):10678-10684; Rosok et al., 1996, J. Biol. Chem. 271 (37): 2261 1-22618; Rader et al., 1998, Proc. Natl. Acad. Sci. USA 95: 8910-8915; Krauss et al., 2003, Protein Engineering 16(10):753-759, all entirely incorporated by reference.
  • the antibodies of the invention multispecific antibody, and notably a bispecific antibody, also sometimes referred to as "diabodies". These are antibodies that bind to two (or more) different antigens. Diabodies can be manufactured in a variety of ways known in the art (Holliger and Winter, 1993, Current Opinion Biotechnol. 4:446-449, entirely incorporated by reference), e.g., prepared chemically or from hybrid hybridomas. [121] Minibodies
  • the antibody is a minibody. Minibodies are minimized antibody- like proteins comprising a scFv joined to a CH3 domain. Hu et al., 1996, Cancer Res. 56:3055-3061 , entirely incorporated by reference. In some cases, the scFv can be joined to the Fc region, and may include some or the entire hinge region. [123] Human Antibodies
  • the antibody is a fully human antibody with at least one modification as outlined herein.
  • Fully human antibody or “complete human antibody” refers to a human antibody having the gene sequence of an antibody derived from a human chromosome with the modifications outlined herein.
  • the antibodies of the invention are antibody fusion proteins (sometimes referred to herein as an "antibody conjugate").
  • One type of antibody fusions comprises Fc fusions, which join the Fc region with a conjugate partner.
  • Fc fusion as used herein is meant a protein wherein one or more polypeptides is operably linked to an Fc region.
  • Fc fusion is herein meant to be synonymous with the terms “immunoadhesin”, “Ig fusion”, “Ig chimera”, and “receptor globulin” (sometimes with dashes) as used in the prior art (Chamow et al., 1996, Trends Biotechnol 14:52-60; Ashkenazi et al., 1997, Curr Opin Immunol 9:195-200, both entirely incorporated by reference).
  • An Fc fusion combines the Fc region of an immunoglobulin with a fusion partner, which in general can be any protein or small molecule. Virtually any protein or small molecule may be linked to Fc to generate an Fc fusion.
  • Protein fusion partners may include, but are not limited to, the variable region of any antibody, the target-binding region of a receptor, an adhesion molecule, a ligand, an enzyme, a cytokine, a chemokine, or some other protein or protein domain.
  • Small molecule fusion partners may include any therapeutic agent that directs the Fc fusion to a therapeutic target.
  • targets may be any molecule, preferably an extracellular receptor, which is implicated in disease.
  • the IgG variants can be linked to one or more fusion partners. In one alternate embodiment, the IgG variant is conjugated or operably linked to another therapeutic compound.
  • the therapeutic compound may be a cytotoxic agent, a chemotherapeutic agent, a toxin, a radioisotope, a cytokine, or other therapeutically active agent.
  • the IgG may be linked to one of a variety of nonproteinaceous polymers, e.g., polyethylene glycol, polypropylene glycol, polyoxyalkylenes, or copolymers of polyethylene glycol and polypropylene glycol.
  • antibody fusions include the fusion of the constant region of the heavy chain with one or more fusion partners (again including the variable region of any antibody), while other antibody fusions are substantially or completely full length antibodies with fusion partners.
  • a role of the fusion partner is to mediate target binding, and thus it is functionally analogous to the variable regions of an antibody (and in fact can be).
  • Virtually any protein or small molecule may be linked to Fc to generate an Fc fusion (or antibody fusion).
  • Protein fusion partners may include, but are not limited to, the target-binding region of a receptor, an adhesion molecule, a ligand, an enzyme, a cytokine, a chemokine, or some other protein or protein domain.
  • Small molecule fusion partners may include any therapeutic agent that directs the Fc fusion to a therapeutic target.
  • targets may be any molecule, preferably an extracellular receptor, which is implicated in disease.
  • the conjugate partner can be proteinaceous or non-proteinaceous; the latter generally being generated using functional groups on the antibody and on the conjugate partner.
  • Suitable conjugates include, but are not limited to, labels as described below, drugs and cytotoxic agents including, but not limited to, cytotoxic drugs (e.g., chemotherapeutic agents) or toxins or active fragments of such toxins.
  • cytotoxic drugs e.g., chemotherapeutic agents
  • Suitable toxins and their corresponding fragments include diptheria A chain, exotoxin A chain, ricin A chain, abrin A chain, curcin, crotin, phenomycin, enomycin and the like.
  • Cytotoxic agents also include radiochemicals made by conjugating radioisotopes to antibodies, or binding of a radionuclide to a chelating agent that has been covalently attached to the antibody. Additional embodiments utilize calicheamicin, auristatins, geldanamycin, maytansine, and duocarmycins and analogs; for the latter, see U.S. 2003/0050331 A1 , entirely incorporated by reference. [130] Covalent modifications of Antibodies
  • Covalent modifications of antibodies are included within the scope of this invention, and are generally, but not always, done post-translationally. For example, several types of covalent modifications of the antibody are introduced into the molecule by reacting specific amino acid residues of the antibody with an organic derivatizing agent that is capable of reacting with selected side chains or the N- or C-terminal residues.
  • Cysteinyl residues most commonly are reacted with ⁇ -haloacetates (and corresponding amines), such as chloroacetic acid or chloroacetamide, to give carboxymethyl or carboxyamidomethyl derivatives.
  • Cysteinyl residues may also be derivatized by reaction with bromotrifluoroacetone, ⁇ -bromo- ⁇ -(5-imidozoyl)propionic acid, chloroacetyl phosphate, N-alkylmaleimides, 3-nitro-2-pyridyl disulfide, methyl 2-pyridyl disulfide, p- chloromercuribenzoate, 2-chloromercuri-4-nitrophenol, or chloro-7-nitrobenzo-2-oxa-1 ,3- diazole and the like.
  • Histidyl residues are derivatized by reaction with diethylpyrocarbonate at pH 5.5-7.0 because this agent is relatively specific for the histidyl side chain. Para-bromophenacyl bromide also is useful; the reaction is preferably performed in 0.1 M sodium cacodylate at pH 6.0.
  • Lysinyl and amino terminal residues are reacted with succinic or other carboxylic acid anhydrides. Derivatization with these agents has the effect of reversing the charge of the lysinyl residues.
  • Other suitable reagents for derivatizing alpha-amino-containing residues include imidoesters such as methyl picolinimidate; pyridoxal phosphate; pyridoxal; chloroborohydride; trinitrobenzenesulfonic acid; O-methylisourea; 2,4-pentanedione; and transaminase-catalyzed reaction with glyoxylate.
  • Arginyl residues are modified by reaction with one or several conventional reagents, among them phenylglyoxal, 2,3-butanedione, 1 ,2-cyclohexanedione, and ninhydrin. Derivatization of arginine residues requires that the reaction be performed in alkaline conditions because of the high pKa of the guanidine functional group. Furthermore, these reagents may react with the groups of lysine as well as the arginine epsilon-amino group.
  • tyrosyl residues may be made, with particular interest in introducing spectral labels into tyrosyl residues by reaction with aromatic diazonium compounds or tetranitromethane. Most commonly, N-acetylimidizole and tetranitromethane are used to form O-acetyl tyrosyl species and 3-nitro derivatives, respectively. Tyrosyl residues are iodinated using 1251 or 1311 to prepare labeled proteins for use in radioimmunoassay, the chloramine T method described above being suitable.
  • R and R' are optionally different alkyl groups, such as 1-cyclohexyl-3-(2-morpholinyl-4-ethyl) carbodiimide or 1-ethyl-3-(4-azonia-4,4- dimethylpentyl) carbodiimide.
  • aspartyl and glutamyl residues are converted to asparaginyl and glutaminyl residues by reaction with ammonium ions.
  • Derivatization with bifunctional agents is useful for crosslinking antibodies to a water- insoluble support matrix or surface for use in a variety of methods, in addition to methods described below.
  • Commonly used crosslinking agents include, e.g., 1 ,1-bis(diazoacetyl)-2- phenylethane, glutaraldehyde, N-hydroxysuccinimide esters, for example, esters with 4- azidosalicylic acid, homobifunctional imidoesters, including disuccinimidyl esters such as 3,3'-dithiobis (succinimidylpropionate), and bifunctional maleimides such as bis-N- maleimido-1 ,8-octane.
  • Derivatizing agents such as methyl-3-[(p- azidophenyl)dithio]propioimidate yield photoactivatable intermediates that are capable of forming crosslinks in the presence of light.
  • reactive water-insoluble matrices such as cyanogen bromide-activated carbohydrates and the reactive substrates described in U.S. Pat. Nos. 3,969,287; 3,691 ,016; 4,195,128; 4,247,642; 4,229,537; and 4,330,440, all entirely incorporated by reference, are employed for protein immobilization.
  • Glutaminyl and asparaginyl residues are frequently deamidated to the corresponding glutamyl and aspartyl residues, respectively. Alternatively, these residues are deamidated under mildly acidic conditions. Either form of these residues falls within the scope of this invention.
  • glycosylation Another type of covalent modification is glycosylation.
  • the IgG variants disclosed herein can be modified to include one or more engineered glycoforms.
  • engineered glycoform as used herein is meant a carbohydrate composition that is covalently attached to an IgG, wherein said carbohydrate composition differs chemically from that of a parent IgG.
  • Engineered glycoforms may be useful for a variety of purposes, including but not limited to enhancing or reducing effector function.
  • Engineered glycoforms may be generated by a variety of methods known in the art (Uma ⁇ a et al., 1999, Nat Biotechnol 17:176-180; Davies et al., 2001 , Biotechnol Bioeng 74:288-294; Shields et al., 2002, J Biol Chem 277:26733-26740; Shinkawa et al., 2003, J Biol Chem 278:3466- 3473; US 6,602,684; USSN 10/277,370; USSN 10/113,929; PCT WO 00/61739A1 ; PCT WO 01/29246A1 ; PCT WO 02/31140A1 ; PCT WO 02/30954A1 , all entirely incorporated by reference; (Potelligent® technology [Biowa, Inc., Princeton, NJ]; GlycoMAb® glycosylation engineering technology [Glycart Biotechnology AG, Zurich, Switzerland]).
  • Engineered glycoform typically refers to the different carbohydrate or oligosaccharide; thus an IgG variant, for example an antibody or Fc fusion, can include an engineered glycoform.
  • engineered glycoform may refer to the IgG variant that comprises the different carbohydrate or oligosaccharide.
  • glycosylation patterns can depend on both the sequence of the protein (e.g., the presence or absence of particular glycosylation amino acid residues, discussed below), or the host cell or organism in which the protein is produced. Particular expression systems are discussed below. [142] 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 tri-peptide 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.
  • X is any amino acid except proline
  • O-linked glycosylation refers to the attachment of one of the sugars N-acetylgalactosamine, 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 antibody is conveniently accomplished by altering the amino acid sequence such that it contains one or more of the above-described tri-peptide 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 starting sequence (for O-linked glycosylation sites).
  • the antibody amino acid sequence is preferably altered through changes at the DNA level, particularly by mutating the DNA encoding the target polypeptide at preselected bases such that codons are generated that will translate into the desired amino acids.
  • Another means of increasing the number of carbohydrate moieties on the antibody is by chemical or enzymatic coupling of glycosides to the protein. These procedures are advantageous in that they do not require production of the protein in a host cell that has glycosylation capabilities for N- and O-linked glycosylation.
  • the sugar(s) may be attached to (a) arginine and histidine, (b) free carboxyl groups, (c) free sulfhydryl groups such as those of cysteine, (d) free hydroxyl groups such as those of serine, threonine, or hydroxyproline, (e) aromatic residues such as those of phenylalanine, tyrosine, or tryptophan, or (f) the amide group of glutamine.
  • Removal of carbohydrate moieties present on the starting antibody may be accomplished chemically or enzymatically.
  • Chemical deglycosylation requires exposure of the protein to the compound trifluoromethanesulfonic acid, or an equivalent compound. This treatment results in the cleavage of most or all sugars except the linking sugar (N- acetylglucosamine or N-acetylgalactosamine), while leaving the polypeptide intact.
  • Chemical deglycosylation is described by Hakimuddin et al., 1987, Arch. Biochem. Biophys. 259:52 and by Edge et al., 1981 , Anal. Biochem. 1 18:131 , both entirely incorporated by reference.
  • Enzymatic cleavage of carbohydrate moieties on polypeptides can be achieved by the use of a variety of endo- and exo-glycosidases as described by Thotakura et al., 1987, Meth. Enzymol. 138:350, entirely incorporated by reference. Glycosylation at potential glycosylation sites may be prevented by the use of the compound tunicamycin as described by Duskin et al., 1982, J. Biol. Chem. 257:3105, entirely incorporated by reference. Tunicamycin blocks the formation of protein-N-glycoside linkages.
  • Another type of covalent modification of the antibody comprises linking the antibody to various nonproteinaceous polymers, including, but not limited to, various polyols such as polyethylene glycol, polypropylene glycol or polyoxyalkylenes, in the manner set forth in, for example, 2005-2006 PEG Catalog from Nektar Therapeutics (available at the Nektar website) US Patents 4,640,835; 4,496,689; 4,301 ,144; 4,670,417; 4,791 ,192 or 4,179,337, all entirely incorporated by reference.
  • amino acid substitutions may be made in various positions within the antibody to facilitate the addition of polymers such as PEG. See for example, U.S. Publication No. 2005/0114037A1 , entirely incorporated by reference.
  • the covalent modification of the antibodies of the invention comprises the addition of one or more labels. In some cases, these are considered antibody fusions.
  • the term "labelling group" means any detectable label.
  • the labelling group is coupled to the antibody via spacer arms of various lengths to reduce potential steric hindrance. Various methods for labelling proteins are known in the art and may be used in performing the present invention.
  • labels fall into a variety of classes, depending on the assay in which they are to be detected: a) isotopic labels, which may be radioactive or heavy isotopes; b) magnetic labels (e.g., magnetic particles); c) redox active moieties; d) optical dyes; enzymatic groups (e.g. horseradish peroxidase, ⁇ -galactosidase, luciferase, alkaline phosphatase); e) biotinylated groups; and f) predetermined polypeptide epitopes recognized by a secondary reporter (e.g., leucine zipper pair sequences, binding sites for secondary antibodies, metal binding domains, epitope tags, etc.).
  • the labelling group is coupled to the antibody via spacer arms of various lengths to reduce potential steric hindrance.
  • Various methods for labelling proteins are known in the art and may be used in performing the present invention.
  • Specific labels include optical dyes, including, but not limited to, chromophores, phosphors and fluorophores, with the latter being specific in many instances.
  • Fluorophores can be either "small molecule" fluores, or proteinaceous fluores.
  • fluorescent label any molecule that may be detected via its inherent fluorescent properties. Suitable fluorescent labels include, but are not limited to, fluorescein, rhodamine, tetramethylrhodamine, eosin, erythrosin, coumarin, methyl-coumarins, pyrene, Malacite green, stilbene, Lucifer Yellow, Cascade BlueJ, Texas Red, IAEDANS, EDANS, BODIPY FL, LC Red 640, Cy 5, Cy 5.5, LC Red 705, Oregon green, the Alexa-Fluor dyes (Alexa Fluor 350, Alexa Fluor 430, Alexa Fluor 488, Alexa Fluor 546, Alexa Fluor 568, Alexa Fluor 594, Alexa Fluor 633, Alexa Fluor 660, Alexa Fluor 680), Cascade Blue, Cascade Yellow and R-phycoerythrin (PE) (Molecular Probes, Eugene, OR), FITC, Rhod
  • Suitable optical dyes including fluorophores, are described in Molecular Probes Handbook by Richard P. Haugland, entirely incorporated by reference.
  • Suitable proteinaceous fluorescent labels also include, but are not limited to, green fluorescent protein, including a Renilla, Ptilosarcus, or Aequorea species of GFP (Chalfie et al., 1994, Science 263:802-805), EGFP (Clontech Laboratories, Inc., Genbank Accession Number U55762), blue fluorescent protein (BFP, Quantum Biotechnologies, Inc. 1801 de Maisonneuve Blvd.
  • the invention provides variant IgG proteins.
  • IgG variants comprise an antibody fragment comprising the CH2-CH3 region of the heavy chain.
  • suitable IgG variants comprise Fc domains (e.g. including the lower hinge region), as well as IgG variants comprising the constant region of the heavy chain (CH 1- hinge-CH2-CH3) also being useful in the present invention, all of which can be fused to fusion partners.
  • An IgG variant includes one or more amino acid modifications relative to a parent IgG polypeptide, in some cases relative to the wild type IgG.
  • the IgG variant can have one or more optimized properties.
  • An IgG variant differs in amino acid sequence from its parent IgG by virtue of at least one amino acid modification.
  • IgG variants have at least one amino acid modification compared to the parent.
  • the IgG variants may have more than one amino acid modification as compared to the parent, for example from about one to fifty amino acid modifications, preferably from about one to ten amino acid modifications, and most preferably from about one to about five amino acid modifications compared to the parent.
  • the sequences of the IgG variants and those of the parent Fc polypeptide are substantially homologous.
  • the variant IgG variant sequences herein will possess about 80% homology with the parent IgG variant sequence, preferably at least about 90% homology, and most preferably at least about 95% homology. Modifications may be made genetically using molecular biology, or may be made enzymatically or chemically.
  • any antigen may be targeted by the IgG variants, including but not limited to proteins, subunits, domains, motifs, and/or epitopes belonging to the following list of target antigens, which includes both soluble factors such as cytokines and membrane-bound factors, including transmembrane receptors: 17-IA, 4-1 BB, 4Dc, 6-keto-PGF1 a, 8-iso- PGF2a, 8-oxo-dG, A1 Adenosine Receptor, A33, ACE, ACE-2, Activin, Activin A, Activin AB, Activin B, Activin C, Activin RIA, Activin RIA ALK-2, Activin RIB ALK-4, Activin RIIA, Activin RIIB, ADAM, ADAM10, ADAM12, ADAM15, ADAM17/TACE, ADAM8, ADAM9, ADAMTS, ADAMTS4, ADAMTS5,
  • targets refers not only to specific proteins and biomolecules, but the biochemical pathway or pathways that comprise them.
  • CTLA-4 as a target antigen implies that the ligands and receptors that make up the T cell co-stimulatory pathway, including CTLA-4, B7- 1 , B7-2, CD28, and any other undiscovered ligands or receptors that bind these proteins, are also targets.
  • target as used herein refers not only to a specific biomolecule, but the set of proteins that interact with said target and the members of the biochemical pathway to which said target belongs.
  • any of the aforementioned target antigens, the ligands or receptors that bind them, or other members of their corresponding biochemical pathway may be operably linked to the Fc variants of the present invention in order to generate an Fc fusion.
  • an Fc fusion that targets EGFR could be constructed by operably linking an Fc variant to EGF, TGF-b, or any other ligand, discovered or undiscovered, that binds EGFR.
  • an Fc variant of the present invention could be operably linked to EGFR in order to generate an Fc fusion that binds EGF, TGF-b, or any other ligand, discovered or undiscovered, that binds EGFR.
  • any polypeptide whether a ligand, receptor, or some other protein or protein domain, including but not limited to the aforementioned targets and the proteins that compose their corresponding biochemical pathways, may be operably linked to the Fc variants of the present invention to develop an Fc fusion.
  • Suitable antigen depends on the desired application. For anti-cancer treatment it is desirable to have a target whose expression is restricted to the cancerous cells. Some targets that have proven especially amenable to antibody therapy are those with signaling functions. Other therapeutic antibodies exert their effects by blocking signaling of the receptor by inhibiting the binding between a receptor and its cognate ligand. Another mechanism of action of therapeutic antibodies is to cause receptor down regulation. Other antibodies do not work by signaling through their target antigen. In some cases, antibodies directed against infectious disease agents are used.
  • the Fc variants of the present invention are incorporated into an antibody against a cytokine.
  • the Fc variants are fused or conjugated to a cytokine.
  • cytokine as used herein is meant a generic term for proteins released by one cell population that act on another cell as intercellular mediators.
  • cytokines may be fused to antibody to provide an array of desirable properties. Examples of such cytokines are lymphokines, monokines, and traditional polypeptide hormones.
  • cytokines include growth hormone such as human growth hormone, N-methionyl human growth hormone, and bovine growth hormone; parathyroid hormone; thyroxine; insulin; proinsulin; relaxin; prorelaxin; glycoprotein hormones such as follicle stimulating hormone (FSH), thyroid stimulating hormone (TSH), and luteinizing hormone (LH); hepatic growth factor; fibroblast growth factor; prolactin; placental lactogen; tumor necrosis factor-alpha and -beta; mullerian-inhibiting substance; mouse gonadotropin- associated peptide; inhibin; activin; vascular endothelial growth factor; integrin; thrombopoietin (TPO); nerve growth factors such as NGF-beta; platelet-growth factor; transforming growth factors (TGFs) such as TGF-alpha and TGF-beta; insulin-like growth factor-l and -II; erythropoietin (EPO)
  • Cytokines and soluble targets are preferred targets for use with the variants of the present invention.
  • anti-VEGF, anti- CTLA-4, and anti-TNF antibodies, or fragments thereof are particularly good antibodies for the use of Fc variants that increase the FcRn binding.
  • Therapeutics against these targets are frequently involved in the treatment of autoimmune diseases and require multiple injections over long time periods. Therefore, longer serum half-lives and less frequent treatments, brought about from the variants of the present invention, are particularly preferred.
  • a number of antibodies and Fc fusions that are approved for use, in clinical trials, or in development may benefit from the Fc variants of the present invention.
  • the Fc polypeptides of the present invention may find use in a range of clinical products and candidates.
  • a number of antibodies that target CD20 may benefit from the Fc polypeptides of the present invention.
  • the Fc polypeptides of the present invention may find use in an antibody that is substantially similar to rituximab (Rituxan®, IDEC/Genentech/Roche) (see for example US 5,736,137), a chimeric anti-CD20 antibody approved to treat Non-Hodgkin's lymphoma; HuMax-CD20, an anti-CD20 currently being developed by Genmab, an anti-CD20 antibody described in US 5,500,362, AME-133 (Applied Molecular Evolution), hA20 (Immunomedics, Inc.), HumaLYM (Intracel), and PRO70769 (PCT/US2003/040426, entitled "Immunoglobulin Variants and Uses Thereof").
  • rituximab Rituxan®, IDEC/Genentech/Roche
  • a number of antibodies that target members of the family of epidermal growth factor receptors may benefit from the Fc polypeptides of the present invention.
  • the Fc polypeptides of the present invention may find use in an antibody that is substantially similar to trastuzumab (Herceptin®, Genentech) (see for example US 5,677,171 ), a humanized anti- Her2/neu antibody approved to treat breast cancer; pertuzumab (rhuMab-2C4, OmnitargTM), currently being developed by Genentech; an anti-Her2 antibody described in US 4,753,894; cetuximab (Erbitux®, Imclone) (US 4,943,533; PCT WO 96/40210), a chimeric anti-EGFR antibody in clinical trials for a variety of cancers; ABX-EGF (US 6,235,883), currently being developed by Abgenix-lmmunex-Amgen; HuMaX-EGFr (USSN 10/172,317), currently being developed by Genmab; 425, EMD55900, EMD62000, and EMD72000 (Merck KGaA) (US 5,55
  • the Fc polypeptides of the present invention may find use in alemtuzumab (Campath®, Millenium), a humanized monoclonal antibody currently approved for treatment of B-cell chronic lymphocytic leukemia.
  • the Fc polypeptides of the present invention may find use in a variety of antibodies or Fc fusions that are substantially similar to other clinical products and candidates, including but not limited to muromonab-CD3 (Orthoclone OKT3®), an anti-CD3 antibody developed by Ortho Biotech/Johnson & Johnson, ibritumomab tiuxetan (Zevalin®), an anti-CD20 antibody developed by IDEC/Schering AG, gemtuzumab ozogamicin (Mylotarg®), an anti-CD33 (p67 protein) antibody developed by Celltech/Wyeth, alefacept (Amevive®), an anti-LFA-3 Fc fusion developed by Biogen), abciximab (ReoPro®), developed by Centocor/Lilly, basiliximab (Simulect®), developed by Novartis, palivizumab (Synagis®), developed by Medlmmune, infliximab (Remicade®),
  • the Fc polypeptides of the present invention may be incorporated into the aforementioned clinical candidates and products, or into antibodies and Fc fusions that are substantially similar to them.
  • the Fc polypeptides of the present invention may be incorporated into versions of the aforementioned clinical candidates and products that are humanized, affinity matured, engineered, or modified in some other way.
  • the Fc polypeptides of the present invention are used for the treatment of autoimmune, inflammatory, or transplant indications.
  • Target antigens and clinical products and candidates that are relevant for such diseases include but are not limited to anti- ⁇ 4 ⁇ 7 integrin antibodies such as LDP-02, anti-beta2 integrin antibodies such as LDP-01 , anti-complement (C5) antibodies such as 5G1.1 , anti-CD2 antibodies such as BTI-322, MEDI-507, anti-CD3 antibodies such as OKT3, SMART anti-CD3, anti-CD4 antibodies such as IDEC-151 , MDX-CD4, OKT4A, anti-CD1 1a antibodies, anti-CD14 antibodies such as IC14, anti-CD18 antibodies, anti-CD23 antibodies such as IDEC 152, anti-CD25 antibodies such as Zenapax, anti-CD40L antibodies such as 5c8, Antova, IDEC- 131 , anti-CD64 antibodies such as MDX-33, anti-CD80 antibodies such as IDEC-114, anti- CD147 antibodies such as ABX-CBL, anti-E-selectin antibodies such as CDP850, anti- g
  • Fc variants of the present invention such as those with increased binding to FcRn may be utilized in TNF inhibitor molecules to provide enhanced properties.
  • Useful TNF inhibitor molecules include any molecule that inhibits the action of TNF-alpha in a mammal. Suitable examples include the Fc fusion Enbrel® (etanercept) and the antibodies Humira® (adalimumab) and Remicade® (infliximab). Monoclonal antibodies (such as Remicade and Humira) engineered using the Fc variants of the present invention to increase FcFn binding, may translate to better efficacy through an increased half-life.
  • antibodies against infectious diseases are used.
  • Antibodies against eukaryotic cells include antibodies targeting yeast cells, including but not limited to Saccharomyces cerevisiae, Hansenula polymorpha, Kluyveromyces fragilis and K. lactis, Pichia guillerimondii and P. pastoris, Schizosaccharomyces pombe, Plasmodium falciparium, and Yarrowia lipolytica.
  • Antibodies against additional fungal cells are also useful, including target antigens associated with Candida strains including Candida glabrata, Candida albicans, C. krusei, C. lusitaniaeand C. maltosa, as well as species of Aspergillus, Cryptococcus, Histoplasma, Coccidioides, Blastomyces, and Penicillium, among others
  • Antibodies directed against target antigens associated with protozoa include, but are not limited to, antibodies associated with Trypanosoma, Leishmania species including Leishmania donovanii; , Plasmodium spp., Pneumocystis carinii, Cryptosporidium parvum, Giardia lamblia, Entamoeba histolytica, and Cyclospora cayetanensis.
  • Antibodies against prokaryotic antigens are also useful, including antibodies against suitable bacteria such as pathogenic and non-pathogenic prokaryotes including but not limited to Bacillus, including Bacillus anthracis; Vibrio, e.g. V.
  • cholerae Escherichia, e.g. Enterotoxigenic E. coli, Shigella, e.g. S. dysenteriae; Salmonella, e.g. S. typhi; Mycobacterium e.g. M. tuberculosis, M. leprae; Clostridium, e.g. C. botulinum, C. tetani, C. difficile, C.perfringens; Cornyebacterium, e.g. C. diphtheriae; Streptococcus, S. pyogenes, S. pneumoniae; Staphylococcus, e.g. S. aureus; Haemophilus, e.g. H.
  • influenzae influenzae
  • Neisseria e.g. N. meningitidis, N. gonorrhoeae
  • Yersinia e.g. Y. lamblia, Y. pestis, Pseudomonas, e.g. P. aeruginosa, P. putida
  • Chlamydia e.g. C. trachomatis
  • Bordetella e.g. B. pertussis
  • Treponema e.g. T. palladium
  • B. anthracis Y. pestis
  • Brucella spp. F. tularensis, B. mallei, B .pseudomallei, B.
  • the antibodies are directed against viral infections; these viruses include, but are not limited to, including orthomyxoviruses, (e.g. influenza virus), paramyxoviruses (e.g respiratory syncytial virus, mumps virus, measles virus), adenoviruses, rhinoviruses, coronaviruses, reoviruses, togaviruses (e.g. rubella virus), parvoviruses, poxviruses (e.g. variola virus, vaccinia virus), enteroviruses (e.g. poliovirus, coxsackievirus), hepatitis viruses (including A, B and C), herpesviruses (e.g.
  • orthomyxoviruses e.g. influenza virus
  • paramyxoviruses e.g respiratory syncytial virus, mumps virus, measles virus
  • adenoviruses e.g respiratory syncytial virus
  • rhinoviruses
  • rabies virus retroviruses
  • retroviruses including HIV, HTLV-I and -II
  • papovaviruses e.g. papillomavirus
  • polyomaviruses e.g. papillomavirus
  • picornaviruses and the like.
  • the present application also provides IgG variants that are optimized for a variety of therapeutically relevant properties.
  • An IgG variant that is engineered or predicted to display one or more optimized properties is herein referred to as an "optimized IqG variant".
  • the most preferred properties that may be optimized include but are not limited to enhanced or reduced affinity for FcRn and increased or decreased in vivo half-life.
  • Suitable embodiments include antibodies that exhibit increased binding affinity to FcRn at lowered pH, such as the pH associated with endosomes, e.g. pH 6.0, while maintaining the reduced affinity at higher pH, such as 7.4., to allow increased uptake into endosomes but normal release rates.
  • these antibodies with modulated FcRn binding may optionally have other desirable properties, such as modulated FcvR binding, such as outlined in U.S.S.N.s U.S.S.N.s 11/174,287, 11/124,640, 10/822,231 , 10/672,280, 10/379,392, and the patent application entitled IgG Immunoglobulin variants with optimized effector function filed on October 21 , 2005 having application no. 1 1/256,060. That is, optimized properties also include but are not limited to enhanced or reduced affinity for an Fc ⁇ R.
  • the IgG variants are optimized to possess enhanced affinity for a human activating Fc ⁇ R, preferably Fc ⁇ Rllla in addition to the FcRn binding profile.
  • the IgG variants are optimized to possess reduced affinity for the human inhibitory receptor Fc ⁇ Rllb. That is, particular embodiments embrace the use of antibodies that show increased binding to FcRn, and increased binding to Fc ⁇ Rllla. Other embodiments utilize use of antibodies that show increased binding to FcRn, and increased binding to Fc ⁇ Rllla. These embodiments are anticipated to provide IgG polypeptides with enhanced therapeutic properties in humans, for example enhanced effector function and greater anticancer potency.
  • the IgG variants are optimized to have increased or reduced affinity for FcRn and increased or decreased affinity for a human Fc ⁇ R, including but not limited to Fc ⁇ RI, Fc ⁇ Rlla, Fc ⁇ Rllb, Fc ⁇ Rllc, Fc ⁇ Rllla, and Fc ⁇ Rlllb including their allelic variations. These embodiments are anticipated to provide IgG polypeptides with enhanced therapeutic properties in humans, for example increased serum half-life and reduced effector function. In other embodiments, IgG variants provide enhanced affinity for FcRn and enhanced affinity for one or more Fc ⁇ Rs, yet reduced affinity for one or more other Fc ⁇ Rs.
  • an IgG variant may have enhanced binding to FcRn and Fc ⁇ Rllla, yet reduced binding to Fc ⁇ Rllb.
  • an IgG variant may have reduced binding to FcRn and to Fc ⁇ R's.
  • an IgG variant may have reduced affinity for FcRn and enhanced affinity for Fc ⁇ Rllb, yet reduced affinity to one or more activating Fc ⁇ Rs.
  • an IgG variant may have increased serum half-life and reduced effector functions.
  • Preferred embodiments comprise optimization of binding to a human FcRn and Fc ⁇ R, however in alternate embodiments the IgG variants possess enhanced or reduced affinity for FcRn and Fc ⁇ R from nonhuman organisms, including but not limited to rodents and non- human primates. IgG variants that are optimized for binding to a nonhuman FcRn may find use in experimentation. For example, mouse models are available for a variety of diseases that enable testing of properties such as efficacy, toxicity, and pharmacokinetics for a given drug candidate. As is known in the art, cancer cells can be grafted or injected into mice to mimic a human cancer, a process referred to as xenografting.
  • IgG variants that comprise IgG variants that are optimized for FcRn may provide valuable information with regard to the clearance characteristics of the protein, its mechanism of clearance, and the like.
  • the IgG variants may also be optimized for enhanced functionality and/or solution properties in aglycosylated form.
  • the Fc ligands include but are not limited to FcRn, Fc ⁇ Rs, C1 q, and proteins A and G, and may be from any source including but not limited to human, mouse, rat, rabbit, or monkey, preferably human.
  • the IgG variants are optimized to be more stable and/or more soluble than the aglycosylated form of the parent IgG variant.
  • IgG variants can include modifications that modulate interaction with Fc ligands other than FcRn and Fc ⁇ Rs, including but not limited to complement proteins, and Fc receptor homologs (FcRHs).
  • FcRHs include but are not limited to FcRHI , FcRH2, FcRH3, FcRH4, FcRH5, and FcRH6 (Davis et al., 2002, Immunol. Reviews 190:123-136, entirely incorporated by reference).
  • the Fc ligand specificity of the IgG variant will determine its therapeutic utility.
  • the utility of a given IgG variant for therapeutic purposes will depend on the epitope or form of the target antigen and the disease or indication being treated.
  • enhanced FcRn binding may be preferable as the enhanced FcRn binding may result in an increase in serum half-life. Longer serum half-lives allow less frequent dosing or lower dosing of the therapeutic. This is particularly preferable when the therapeutic agent is given in response to an indication that requires repeated administration.
  • decreased FcRn affinity may be preferable.
  • IgG variants may be used that comprise IgG variants that provide enhanced affinity for FcRn with enhanced activating Fc ⁇ Rs and/or reduced affinity for inhibitory Fc ⁇ Rs.
  • IgG variants may be further beneficial to utilize IgG variants that provide differential selectivity for different activating Fc ⁇ Rs; for example, in some cases enhanced binding to Fc ⁇ Rlla and Fc ⁇ Rllla may be desired, but not Fc ⁇ RI, whereas in other cases, enhanced binding only to Fc ⁇ Rlla may be preferred.
  • IgG variants that alter FcRn binding and enhance both Fc ⁇ R- mediated and complement-mediated effector functions, whereas for other cases it may be advantageous to utilize IgG variants that enhance FcRn binding, or serum half-life, and either Fc ⁇ R-mediated or complement-mediated effector functions.
  • IgG variants that provide enhanced binding to the inhibitory Fc ⁇ Rllb, yet WT level, reduced, or ablated binding to activating Fc ⁇ Rs. This may be particularly useful, for example, when the goal of an IgG variant is to inhibit inflammation or auto-immune disease, or modulate the immune system in some way. Because auto-immune diseases are generally long-lasting and treatment is given in repeated dosing, their treatment with Fc variants with increased half-life from increased FcRn is particularly preferred.
  • the IgG variants can include modifications to reduce immunogenicity in humans.
  • the immunogenicity of an IgG variant is reduced using a method described in USSN 11/004,590, entirely incorporated by reference.
  • the IgG variants are humanized (Clark, 2000, Immunol Today 21 :397-402, entirely incorporated by reference).
  • the IgG variants can include modifications that reduce immunogenicity.
  • Modifications to reduce immunogenicity can include modifications that reduce binding of processed peptides derived from the parent sequence to MHC proteins.
  • amino acid modifications would be engineered such that there are no or a minimal number of immune epitopes that are predicted to bind, with high affinity, to any prevalent MHC alleles.
  • Several methods of identifying MHC-binding epitopes in protein sequences are known in the art and may be used to score epitopes in an IgG variant.
  • variants of the present invention may be designed by various means.
  • the variants, as described herein, may be insertions, deletions, substitutions, other modifications, or combinations of these and other changes.
  • a particularly novel embodiment of the present invention is the design of insertions and deletions that either improve or reduce the binding of an Fc polypeptide to an Fc ligand.
  • insertions or deletions may be made that increase or decrease the affinity of the Fc polypeptide for FcRn.
  • Insertions and deletions may be designed by rational approaches or by approaches that include the use or random components, such as random or semi-random library creation or screening.
  • substitutions are disclosed that increase or decrease the affinity of the Fc polypeptide for FcRn.
  • Variant Fc polypeptides may be created by substituting a variant amino acid in place of the parent amino acid at a position in the Fc polypeptide. By substituting one or more amino acids for variant amino acids in the Fc polypeptide, the side chains at those positions are altered. Most useful substitutions modify the Fc properties by altering the Fc side chains. The substituted side chains may interact directly or indirectly with an Fc binding partner that is associated with an Fc function or property. The at least one substitution alters the covalent structure of one or more side chains of the parent Fc polypeptide. [184] Alternatively, variant Fc polypeptides may be created that change the covalent structure of the backbone of the parent Fc polypeptide.
  • the backbone atoms in proteins are the peptide nitrogen, the alpha carbon, the carbonyl or peptide carbon and the carbonyl oxygen.
  • Changing the covalent structure of the backbone provides additional methods of altering the properties of the Fc polypeptides.
  • the covalent structure of the Fc backbone may be altered by the addition of atoms into the backbone, e.g. by inserting one or more amino acids, or the subtraction of atoms from the backbone, e.g. by deleting one or more amino acids.
  • the covalent structure of the backbone may also be altered by changing individual atoms of the backbone to other atoms (Deechongkit et al., J Am Chem Soc. 2004.
  • insertions or deletions of amino acids in Fc polypeptides may be done by inserting or deleting the corresponding nucleotides in the DNA encoding the Fc polypeptide. Alternatively, as is known in the art, insertions or deletions of amino acids may be done during synthesis of Fc polypeptides.
  • the design of insertions or deletions of amino acids that alter the interaction of the Fc polypeptide with one or more binding partners may be done by considering the structure of the complex of the Fc polypeptide and its binding partner.
  • the design may be done by considering the structure of the Fc polypeptide and information about the Fc region involved in binding the binding partner. This information may be obtained by mutagenesis experiments, phage display experiments, homology comparisons, computer modeling or other means.
  • positions in the amino acid sequence for insertions or deletions that affect the Fc binding interactions, but do not affect the overall structure, stability, expression or use of the Fc polypeptide, are in loops that are involved in the Fc/Fc-binding partner interactions.
  • positions 244-257, 279-284, 307-317, 383-390, and 428-435 are preferred loop locations for insertions or deletions (numbering from EU index of Kabat et al., Burffle et al., 1994, Nature, 372:379-383; Martin et al., 2001 , MoI Cell 7:867-877, all entirely incorporated by reference).
  • positions 229-239, 266-273, 294-299, and 324-331 are preferred loop locations for insertions or deletions (numbering from EU index of Kabat et al., PDB code 1 E4K.pdb Sondermann et al. Nature. 2000 406:267, all entirely incorporated by reference).
  • Loops are regions of the polypeptide not involved in alpha helical or beta sheet structure. Loops positions are positions that are not in either alpha helical or beta sheet structures (van Holde, Johnson and Ho. Principles of Physical Biochemistry. Prentice Hall, New Jersey 1998, Chapter 1 pp2-67, entirely incorporated by reference).
  • Loop positions are preferred because the backbone atoms are typically more flexible and less likely involved in hydrogen bonds compared to the backbone atoms of alpha helices and beta sheets. Therefore, the lengthening or shortening of a loop due to an insertion or deletion of one or more amino acids is less likely to lead to large, disruptive changes to the Fc polypeptide, including stability, expression or other problems.
  • Insertions and deletions may be used to alter the length of the polypeptide. For example, in loop regions, altering the loop length results in altered flexibility and conformational entropy of the loop. Insertions in a loop will generally increase the conformational entropy of the loop, which may be defined as Boltzman's constant multiplied by the natural logarithm of the number of possible conformations (van Holde, Johnson and Ho. Principles of Physical Biochemistry. Prentice Hall, New Jersey 1998, pp78, entirely incorporated by reference). By inserting at least one amino acid into a polypeptide, the total number of conformations available to the polypeptide increases.
  • additional conformations may be beneficial for forming favorable Fc/Fc-binding partner interactions because the Fc polypeptide may use one of the additional conformations in binding the Fc- binding protein.
  • the insertion may lead to stronger Fc/Fc-binding partner interactions. If the additional conformations are not used in the binding interface, then the insertion may lead to weaker Fc/Fc-binding partner interactions, because the additional conformations would compete with the binding-competent conformation.
  • deletion of a polypeptide segment may also lead to either stronger or weaker Fc/Fc binding-partner interactions.
  • deletion of a segment which reduces the possible number of backbone conformations, removes the binding-competent conformation, then the deletion may lead to weaker Fc/Fc-binding partner interactions. If the deletion does not remove the binding- competent conformation, then the deletion may lead to stronger Fc/Fc-binding partner interactions because the deletion may remove conformations that compete with the binding- competent conformation.
  • Insertions and deletions may be used to alter the positions and orientations of the amino acids in the Fc polypeptide. Because insertions and deletions cause a change in the covalent structure of the backbone, they necessarily cause a change in the positions of the backbone atoms.
  • Figure 7 compares the backbone positions at some loop segments, marked L1 to L4, in three different backbones.
  • the reference backbone structure contains four loop segments, whereas the deletion backbone lacks segment L1 and the insertion segment comprises an additional segment before, ie, N-terminal to, segment L1. Deletions and insertions cause the largest change in the backbone structure near the site of the insertion or deletion. By deleting a segment near the N-terminal end of the loop, e.g.
  • L1 the loop shortens and the remaining segments shift their position closer to the loop N-terminus. This has the effect of moving the L2 segment toward the prior location of the L1 segment and toward the loop N-terminus.
  • This change in position of the L2 segment toward the L1 segment may strengthen the binding of the Fc/Fc-binding partner complex and is preferred when there is prior information suggesting that the amino acid or amino acids located in L2 make favorable interactions with the Fc-binding partner, when located in L1.
  • L2 contains alanine and tyrosine and substitution of two L1 amino acids for alanine and tyrosine previously lead to an Fc variant with increased binding
  • deletion of L1 may create an Fc variant with increased affinity for the Fc-binding partner.
  • an insertion of a polypeptide segment into an Fc polypeptide at the N- terminal side of a loop causes the positions of the loop segments to be shifted toward the C- terminal side of the loop.
  • an insertion of one or more amino acids before, i.e. N- terminally to, segment L1 alters the backbone conformation including a shift of the L1 segment toward the C-terminal end of the loop. This type of insertion is preferred when the amino acids located in segment L1 are known to make favorable interactions when located in the L2 positions, as the insertion may lead to stronger Fc/Fc-binding partner interactions.
  • the inserted, deleted and reference segments may be one or more than one amino acid in the Fc polypeptide.
  • insertions or deletions may be used at the C-terminal end of loops in a manner analogous to the insertions or deletions at the N-terminal end of loops. Insertions at the loop C-terminus may lead to a movement of the positions N-terminal of the insertion toward the loop N-terminus. Deletions at the loop C-terminus may lead to a movement of the positions N-terminal of the deletion toward the loop C-terminus.
  • the choice of using an insertion or deletion at the N-terminal or C-terminal end of the loop is based on the amino acids located in the loop, the desire for increased or decreased Fc/Fc-binding partner affinity, and the positional shift desired.
  • Insertions or deletions may be used in any region of an Fc polypeptide, including the loops, the alpha helical, and the beta sheet regions.
  • Preferred locations for insertions and deletions include loop regions, which are those that are not alpha helical or beta sheet regions. Loops are preferred because they generally accept alterations in the backbone better than alpha helixes or beta sheets. The particularly preferred locations for insertions or deletions that result in stronger protein/protein interactions are at the N-terminal or C- terminal edges of a loop. If the loop side chains are involve in the Fc/Fc-binding partner interactions, then insertions or deletion at the edges are less likely to lead to strongly detrimental changes in the binding interactions.
  • Deletions within the exact center of the loop are more likely to remove important residues in the Fc/Fc-binding partner interface and insertions within the exact center of the loop are more likely to create unfavorable interactions in the Fc/Fc-binding partner interface.
  • the number of residues deleted or inserted may be determined by the size of the backbone change desired with insertions or deletions of 15 or less residues being preferred, insertions or deletions of 10 or less residues being more preferred, and insertions or deletions of 5 or less residues being most preferred.
  • Fc insertion variants have the additional step of designing the sequence of the at least one amino acid to be inserted. Insertions of polar residues, including Ser, Thr, Asn, GIn, Ala, GIy, His, are preferred at positions expected to be exposed in the Fc polypeptide.
  • the smaller amino acids, including Ser, Thr, and Ala, are particularly preferred as the small size is less likely to sterically interfere with the Fc/Fc-binding partner interactions.
  • Ser and Thr also have the capability to hydrogen bond with atoms on the Fc- binding partner.
  • Insertions also have the added flexibility that the inserted polypeptide may be designed to make favorable interactions with the Fc-binding partner as would be desire when stronger Fc/Fc-binding partner binding is desired.
  • the length of the backbone insertion may be determined by modeling the variant backbone with a simple, generic sequence to be inserted. For example, polyserine, polyglycine or polyalanine insertions of different lengths may be constructed and modeled. Modeling may be done by a variety of methods, including homology modeling based on known three-dimensional structures of homologues comprising the insertion, and by computer modeling including MODELLER (M.A. Marti- Renom et al. Annu. Rev. Biophys. Biomol. Struct.
  • Insertions and deletions may be made to alter the binding of Fc polypeptides to FcgammaR in an analogous manner to the described method to alter FcRn-binding properties.
  • Fc domains bind to the FcgammaR at the position indicated in Figure 1.
  • Structures of the Fc/FcgammaR complex including PDB codes 1T89 and 11 IS (Radaev S et al. J. Biol. Chem. v276, p.16469-16477 entirely incorporated by reference), demonstrate the interacting residues and loops between the two structures. Mutagenesis results such as those found in US1 1/124620 and US6737056, both entirely incorporated by reference) all have utility in determined appropriate shifts of backbone positioning.
  • Insertions and deletions may be designed in any polypeptide besides Fc polypeptides by the methods described herein.
  • insertions or deletions in the TNF superfamily member, APRIL may be designed with the aid of its three-dimensional structure (PDB code 1XU1.pdb, Hymowitz, et al. (2005) J. Biol. Chem. 280:7218, entirely incorporated by reference).
  • Insertions or deletions may be designed to increase APRIL binding to its receptor, TACI.
  • the loop residues preferred as insertion or deletion sites are residues Ser1 18-Val124, Asp164-Phe167, Pro192-Ala198, Pro221-Lys226. These loops interact with TACI in the APRI L/TACI complex and mediate binding.
  • Polypeptides incorporating variants are residues Ser1 18-Val124, Asp164-Phe167, Pro192-Ala198, Pro221-Lys226. These loops interact with TACI in the A
  • the IgG variants can be based on human IgG sequences, and thus human IgG sequences are used as the "base" sequences against which other sequences are compared, including but not limited to sequences from other organisms, for example rodent and primate sequences. IgG variants may also comprise sequences from other immunoglobulin classes such as IgA, IgE, IgD, IgM, and the like. It is contemplated that, although the IgG variants are engineered in the context of one parent IgG, the variants may be engineered in or "transferred" to the context of another, second parent IgG.
  • the amino acid sequence of a first IgG outlined herein is directly compared to the sequence of a second IgG. After aligning the sequences, using one or more of the homology alignment programs known in the art (for example using conserved residues as between species), allowing for necessary insertions and deletions in order to maintain alignment (i.e., avoiding the elimination of conserved residues through arbitrary deletion and insertion), the residues equivalent to particular amino acids in the primary sequence of the first IgG variant are defined.
  • Alignment of conserved residues preferably should conserve 100% of such residues. However, alignment of greater than 75% or as little as 50% of conserved residues is also adequate to define equivalent residues.
  • Equivalent residues may also be defined by determining structural homology between a first and second IgG that is at the level of tertiary structure for IgGs whose structures have been determined. In this case, equivalent residues are defined as those for which the atomic coordinates of two or more of the main chain atoms of a particular amino acid residue of the parent or precursor (N on N, CA on CA, C on C and O on O) are within 0.13 nm and preferably 0.1 nm after alignment.
  • the variant antibody may be engineered in another IgGI parent antibody that binds a different antigen, a human lgG2 parent antibody, a human IgA parent antibody, a mouse lgG2a or lgG2b parent antibody, and the like.
  • the context of the parent IgG variant does not affect the ability to transfer the IgG variants to other parent IgGs.
  • IgG variants may be engineered, produced, and screened experimentally to obtain IgG variants with optimized effector function.
  • a variety of methods are described for designing, producing, and testing antibody and protein variants in USSN 10/754,296, and USSN 10/672,280, both entirely incorporated by reference.
  • a variety of protein engineering methods may be used to design IgG variants with optimized effector function.
  • a structure-based engineering method may be used, wherein available structural information is used to guide substitutions, insertions or deletions.
  • a computational screening method may be used, wherein substitutions are designed based on their energetic fitness in computational calculations. See for example USSN 10/754,296 and USSN 10/672,280, and references cited therein, all entirely incorporated by reference.
  • sequences may be used to guide substitutions at the identified positions.
  • sequence information may curb the introduction of substitutions that are potentially deleterious to protein structure.
  • the source of the sequences may vary widely, and include one or more of the known databases, including but not limited to the Kabat database (Northwestern University); Johnson & Wu, 2001 , Nucleic Acids Res. 29:205-206; Johnson & Wu, 2000, Nucleic Acids Res. 28:214- 218), the IMGT database (IMGT, the international ImMunoGeneTics information system®; ; Lefranc et al., 1999, Nucleic Acids Res.
  • Antibody sequence information can be obtained, compiled, and/or generated from sequence alignments of germline sequences or sequences of naturally occurring antibodies from any organism, including but not limited to mammals.
  • sequences that are human or substantially human may further have the advantage of being less immunogenic when administered to a human.
  • Other databases which are more general nucleic acid or protein databases, i.e.
  • Aligned sequences can include VH, VL, CH, and/or CL sequences.
  • sequence- based alignment programs and methods There are numerous sequence- based alignment programs and methods known in the art, and all of these find use in the generation of sequence alignments.
  • random or semi-random mutagenesis methods may be used to make amino acid modifications at the desired positions. In these cases positions are chosen randomly, or amino acid changes are made using simplistic rules. For example all residues may be mutated to alanine, referred to as alanine scanning.
  • Such methods may be coupled with more sophisticated engineering approaches that employ selection methods to screen higher levels of sequence diversity.
  • selection technologies such as phage display, ribosome display, cell surface display, and the like, as described below.
  • Preferred variants of the present invention include those found in Figure 8.
  • Alternatively preferred variants of the present invention include those found in Figure 9.
  • Additionally alternatively preferred variants of the present invention include those found in Figure 10. These variants have shown increased binding to the Fc receptor, FcRn, as illustrated in the examples.
  • the IgG variants can be made by any method known in the art.
  • the IgG variant sequences are used to create nucleic acids that encode the member sequences, and that may then be cloned into host cells, expressed and assayed, if desired. These practices are carried out using well-known procedures, and a variety of methods that may find use in are described in Molecular Cloning - A Laboratory Manual, 3 rd Ed. (Maniatis, Cold Spring Harbor Laboratory Press, New York, 2001 ), and Current Protocols in Molecular Biology (John Wiley & Sons), both entirely incorporated by reference.
  • the nucleic acids that encode the IgG variants may be incorporated into an expression vector in order to express the protein.
  • Expression vectors typically include a protein operably linked, that is, placed in a functional relationship, with control or regulatory sequences, selectable markers, any fusion partners, and/or additional elements.
  • the IgG variants may be produced by culturing a host cell transformed with nucleic acid, preferably an expression vector, containing nucleic acid encoding the IgG variants, under the appropriate conditions to induce or cause expression of the protein.
  • nucleic acid preferably an expression vector, containing nucleic acid encoding the IgG variants
  • yeast a wide variety of appropriate host cells may be used, including but not limited to mammalian cells, bacteria, insect cells, and yeast.
  • a variety of cell lines that may find use are described in the ATCC cell line catalog, available from the American Type Culture Collection, entirely incorporated by reference.
  • the methods of introducing exogenous nucleic acid into host cells are well known in the art, and will vary with the host cell used.
  • IgG variants are purified or isolated after expression.
  • Antibodies may be isolated or purified in a variety of ways known to those skilled in the art. Standard purification methods include chromatographic techniques, electrophoretic, immunological, precipitation, dialysis, filtration, concentration, and chromatofocusing techniques. As is well known in the art, a variety of natural proteins bind antibodies, for example bacterial proteins A, G, and L, and these proteins may find use in purification. Often, purification may be enabled by a particular fusion partner.
  • proteins may be purified using glutathione resin if a GST fusion is employed, Ni +2 affinity chromatography if a His-tag is employed, or immobilized anti-flag antibody if a flag-tag is used.
  • glutathione resin if a GST fusion is employed
  • Ni +2 affinity chromatography if a His-tag is employed
  • immobilized anti-flag antibody if a flag-tag is used.
  • Fc variants may be screened using a variety of methods, including but not limited to those that use in vitro assays, in vivo and cell-based assays, and selection technologies. Automation and high-throughput screening technologies may be utilized in the screening procedures. Screening may employ the use of a fusion partner or label, for example an immune label, isotopic label, or small molecule label such as a fluorescent or colorimetric dye.
  • a fusion partner or label for example an immune label, isotopic label, or small molecule label such as a fluorescent or colorimetric dye.
  • the functional and/or biophysical properties of Fc variants are screened in an in vitro assay.
  • the protein is screened for functionality, for example its ability to catalyze a reaction or its binding affinity to its target.
  • selection methods are those that select for favorable members of a library.
  • the methods are herein referred to as "selection methods", and these methods find use in the present invention for screening Fc variants.
  • selection methods When protein libraries are screened using a selection method, only those members of a library that are favorable, that is which meet some selection criteria, are propagated, isolated, and/or observed.
  • selection methods are known in the art that may find use in the present invention for screening protein libraries.
  • Other selection methods that may find use in the present invention include methods that do not rely on display, such as in vivo methods.
  • Fc variants are screened using one or more cell-based or in vivo assays.
  • purified or unpurified proteins are typically added exogenously such that cells are exposed to individual variants or pools of variants belonging to a library.
  • These assays are typically, but not always, based on the function of the Fc polypeptide; that is, the ability of the Fc polypeptide to bind to its target and mediate some biochemical event, for example effector function, ligand/receptor binding inhibition, apoptosis, and the like.
  • Such assays often involve monitoring the response of cells to the IgG, for example cell survival, cell death, change in cellular morphology, or transcriptional activation such as cellular expression of a natural gene or reporter gene.
  • such assays may measure the ability of Fc variants to elicit ADCC, ADCP, or CDC.
  • additional cells or components that is in addition to the target cells, may need to be added, for example example serum complement, or effector cells such as peripheral blood monocytes (PBMCs), NK cells, macrophages, and the like.
  • PBMCs peripheral blood monocytes
  • NK cells macrophages, and the like.
  • additional cells may be from any organism, preferably humans, mice, rat, rabbit, and monkey.
  • Antibodies may cause apoptosis of certain cell lines expressing the target, or they may mediate attack on target cells by immune cells which have been added to the assay.
  • Methods for monitoring cell death or viability are known in the art, and include the use of dyes, immunochemical, cytochemical, and radioactive reagents. Transcriptional activation may also serve as a method for assaying function in cell-based assays.
  • cell-based screens are performed using cells that have been transformed or transfected with nucleic acids encoding the variants. That is, Fc variants are not added exogenously to the cells.
  • the biological properties of the IgG variants may be characterized in cell, tissue, and whole organism experiments.
  • drugs are often tested in animals, including but not limited to mice, rats, rabbits, dogs, cats, pigs, and monkeys, in order to measure a drug's efficacy for treatment against a disease or disease model, or to measure a drug's pharmacokinetics, toxicity, and other properties.
  • the animals may be referred to as disease models.
  • Therapeutics are often tested in mice, including but not limited to nude mice, SCID mice, xenograft mice, and transgenic mice (including knockins and knockouts). Such experimentation may provide meaningful data for determination of the potential of the protein to be used as a therapeutic. Any organism, preferably mammals, may be used for testing.
  • the IgG variants may find use in a wide range of products.
  • the IgG variant is a therapeutic, a diagnostic, or a research reagent, preferably a therapeutic.
  • the IgG variant may find use in an antibody composition that is monoclonal or polyclonal.
  • the IgG variants are used to kill target cells that bear the target antigen, for example cancer cells.
  • the IgG variants are used to block, antagonize or agonize the target antigen, for example for antagonizing a cytokine or cytokine receptor.
  • the IgG variants are used to block, antagonize or agonize the target antigen and kill the target cells that bear the target antigen.
  • the IgG variants may be used for various therapeutic purposes.
  • an antibody comprising the IgG variant is administered to a patient to treat an antibody-related disorder.
  • a "patient” for the purposes includes humans and other animals, preferably mammals and most preferably humans.
  • antibody related disorder or “antibody responsive disorder” or “condition” or “disease” herein are meant a disorder that may be ameliorated by the administration of a pharmaceutical composition comprising an IgG variant.
  • Antibody related disorders include but are not limited to autoimmune diseases, immunological diseases, infectious diseases, inflammatory diseases, neurological diseases, and oncological and neoplastic diseases including cancer.
  • cancer and “cancerous” herein 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 (including liposarcoma), neuroendocrine tumors, mesothelioma, schwanoma, meningioma, adenocarcinoma, melanoma, and leukemia and lymphoid malignancies.
  • an IgG variant is the only therapeutically active agent administered to a patient.
  • the IgG variant is administered in combination with one or more other therapeutic agents, including but not limited to cytotoxic agents, chemotherapeutic agents, cytokines, growth inhibitory agents, anti-hormonal agents, kinase inhibitors, anti-angiogenic agents, cardioprotectants, or other therapeutic agents.
  • the IgG varariants may be administered concomitantly with one or more other therapeutic regimens.
  • an IgG variant may be administered to the patient along with chemotherapy, radiation therapy, or both chemotherapy and radiation therapy.
  • the IgG variant may be administered in conjunction with one or more antibodies, which may or may not be an IgG variant.
  • the IgG variant and one or more other anti-cancer therapies are employed to treat cancer cells ex vivo. It is contemplated that such ex vivo treatment may be useful in bone marrow transplantation and particularly, autologous bone marrow transplantation. It is of course contemplated that the IgG variants can be employed in combination with still other therapeutic techniques such as surgery.
  • the IgG is administered with an anti-angiogenic agent.
  • anti-angiogenic agent as used herein is meant a compound that blocks, or interferes to some degree, the development of blood vessels.
  • the anti-angiogenic factor may, for instance, be a small molecule or a protein, for example an antibody, Fc fusion, or cytokine, that binds to a growth factor or growth factor receptor involved in promoting angiogenesis.
  • the preferred anti-angiogenic factor herein is an antibody that binds to Vascular Endothelial Growth Factor (VEGF).
  • VEGF Vascular Endothelial Growth Factor
  • the IgG is administered with a therapeutic agent that induces or enhances adaptive immune response, for example an antibody that targets CTLA-4.
  • the IgG is administered with a tyrosine kinase inhibitor.
  • tyrosine kinase inhibitor as used herein is meant a molecule that inhibits to some extent tyrosine kinase activity of a tyrosine kinase.
  • the IgG variants are administered with a cytokine.
  • compositions are contemplated wherein an IgG variant and one or more therapeutically active agents are formulated.
  • Formulations of the IgG variants are prepared for storage by mixing the IgG having the desired degree of purity with optional pharmaceutically acceptable carriers, excipients or stabilizers (Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed., 1980, entirely incorporated by reference), in the form of lyophilized formulations or aqueous solutions.
  • the formulations to be used for in vivo administration are preferably sterile. This is readily accomplished by filtration through sterile filtration membranes or other methods.
  • the IgG variants and other therapeutically active agents disclosed herein may also be formulated as immunoliposomes, and/or entrapped in microcapsules.
  • the concentration of the therapeutically active IgG variant in the formulation may vary from about 0.1 to 100% by weight. In a preferred embodiment, the concentration of the IgG is in the range of 0.003 to 1.0 molar.
  • a therapeutically effective dose of the IgG variant may be administered.
  • therapeutically effective dose herein is meant a dose that produces the effects for which it is administered. The exact dose will depend on the purpose of the treatment, and will be ascertainable by one skilled in the art using known techniques. Dosages may range from 0.01 to 100 mg/kg of body weight or greater, for example 0.01 , 0.1 , 1.0, 10, or 50 mg/kg of body weight, with 1 to 10mg/kg being preferred.
  • Administration of the pharmaceutical composition comprising an IgG variant, preferably in the form of a sterile aqueous solution may be done in a variety of ways, including, but not limited to, orally, subcutaneously, intravenously, parenterally, intranasally, intraotically, intraocularly, rectally, vaginally, transdermally, topically (e.g., gels, salves, lotions, creams, etc.), intraperitoneal ⁇ , intramuscularly, intrapulmonary (e.g., AERx® inhalable technology commercially available from Aradigm, or Inhance® pulmonary delivery system commercially available from Nektar Therapeutics, etc.).
  • Therapeutic described herein may be administered with other therapeutics concomitantly, i.e., the therapeutics described herein may be co-administered with other therapies or therapeutics, including for example, small molecules, other biologicals, radiation therapy, surgery, etc.
  • Fc variants were constructed using the human IgGI Fc domain and the variable domain of trastuzumab (Herceptin®, Genentech).
  • the Fc polypeptides were part of Alemtuzumab, An anti-HER2 antibody or AC10.
  • Alemtuzumab (Campath®, a registered trademark of Millenium) is a humanized monoclonal antibody currently approved for treatment of B-cell chronic lymphocytic leukemia (Hale et al., 1990, Tissue Antigens 35:1 18- 127, entirely incorporated by reference).
  • Trastuzumab (Herceptin®, a registered trademark of Genentech) is an anti-HER2/neu antibody for treatment of metastatic breast cancer.
  • the heavy and light chain sequences of the anti-HER2 antibody are shown in Figure 22.
  • AC10 is an anti-CD30 monoclonal antibody.
  • the Herceptin variable region was assembled using recursive PCR. This variable region was then cloned with human IgGI into the pcDNA3.1/Zeo(+) vector (Invitrogen), shown in Figure 1 1. Plasmids were propagated in One Shot TOP10 E. coli cells (Invitrogen) and purified using the Hi-Speed Plasmid Maxi Kit (Qiagen). Plasmids were sequenced to verify the presence of the cloned inserts.
  • Binding of Fc polypeptides to Fc ligands was assayed with surface plasmon resonance measurements.
  • Surface plasmon resonance (SPR) measurements were performed using a BIAcore 3000 instrument (BIAcore AB). Wild-type or variant antibody was captured using immobilized protein L (Pierce Biotechnology, Rockford, IL), and binding to receptor analyte was measured.
  • Protein L was covalently coupled to a CM5 sensor chip at a concentration of 1 uM in 10 mM sodium acetate, pH 4.5 on a CM5 sensor chip using N- hydroxysuccinimide/N-ethyl-N'-(-3-dimethylamino-propyl) carbodiimide (NHS/EDC) at a flow rate of 5 ul/min.
  • NHS/EDC N- hydroxysuccinimide/N-ethyl-N'-(-3-dimethylamino-propyl) carbodiimide
  • Running buffer was 0.01 M HEPES pH 7.4, 0.15 M NaCI, 3 mM EDTA, 0.005% v/v Surfactant P20 (HBS-EP, Biacore, Uppsala, Sweden), and chip regeneration buffer was 10 mM glycine-HCI pH 1.5. 125 nM Wild-type or variant anti-HER2 antibody was bound to the protein L CM5 chip in HBS-EP at 1 ul/min for 5 minutes.
  • FcRn-His-GST analyte a FcRn fused to a His-tag and glutathione S transferase, in serial dilutions between 1 and 250 nM, were injected for 20 minutes association, 10 minutes dissociation, in HBS-EP at 10 ul/min.
  • Response measured in resonance units (RU), was acquired at 1200 seconds after receptor injection, reflecting near steady state binding. A cycle with antibody and buffer only provided baseline response.
  • RU versus 1 / log concentration plots were generated and fit to a sigmoidal dose response using nonlinear regression with GraphPad Prism.
  • AlphaScreenTM is a bead-based non-radioactive luminescent proximity assay. Laser excitation of a donor bead excites oxygen, which if sufficiently close to the acceptor bead will generate a cascade of chemiluminescent events, ultimately leading to fluorescence emission at 520-620 nm.
  • the principal advantage of the AlphaScreenTM is its sensitivity. Because one donor bead emits up to 60,000 excited oxygen molecules per second, signal amplification is extremely high, allowing detection down to attomolar (10 "18 ) levels.
  • Wild-type antibody was biotinylated by standard methods for attachment to streptavidin donor beads, and tagged Fc ligand, for example FcRn, FcgammaR or Protein A, was bound to glutathione chelate acceptor beads.
  • the AlphaScreenTM was applied as a direct binding assay in which the Fc/Fc ligand interactions bring together the donor and acceptor beads to create the measured signal.
  • the AlphaScreenTM was applied as a competition assay for screening designed Fc polypeptides. In the absence of competing Fc polypeptides, wild-type antibody and FcRn interact and produce a signal at 520-620 nm. Untagged Fc domains compete with wild-type Fc/FcRn interaction, reducing fluorescence quantitatively to enable determination of relative binding affinities.
  • Binding affinity of IgGI Fc to FcRn was measured with variant antibodies using AlphaScreenTM.
  • the Fc polypeptides were part of Alemtuzumab or Trastuzumab.
  • Alemtuzumab (Campath®, Ilex) is a humanized monoclonal antibody currently approved for treatment of B-cell chronic lymphocytic leukemia (Hale et al., 1990, Tissue Antigens 35:1 18- 127, entirely incorporated by reference).
  • Trastuzumab (Herceptin®, Genentech) is an anti- HER2/neu antibody for treatment of metastatic breast cancer.
  • the Fc variants V308F and V08Y have a 2.9-fold and 4.3-fold increase in FcRn affinity over wild type (Figure 13). Positions 279 and 385 interact with FcRn as variants V279E, V279Q and V279Y and G385H and G385N all have stronger FcRn interactions. These variants all are to amino acids that are capable of hydrogen bonding. Sequences of the Fc regions of human IgGI comprising various modifications of the present invention are shown in Figure 23.
  • the Fc variant N434Y has particularly strong binding to FcRn at pH 6.0 as shown in figure 13.
  • the single variant N434Y has 16-fold increased binding.
  • Combinations of this variant with other modifications led to even stronger binding.
  • P257L/N434Y, ⁇ 281 S/N434Y, and V308F/N434Y show 830-fold, 180-fold, and 350-fold increases in FcRn binding.
  • EXAMPLE 4 Variants incorporating insertions and deletions.
  • Insertions and deletions that alter the strength of Fc/FcRn interactions were constructed and their binding properties to various Fc ligands were measured.
  • AlphaScreenTM data showing the improved binding of ⁇ 281 S is shown in Figure 12b and 21 a.
  • the inserted sequence which may be more than one residue, is given after the position number.
  • This Fc variant was constructed in the kappa, IgGI anti-HER2 antibody trastuzumab (Herceptin®, Genetech) using methods disclosed herein.
  • An insertion at the site between residues 281 and 282 shifts the Fc loop residues C-terminal of residue 281 toward the C-terminus of the loop and alters the side chain positioning.
  • Fc variants comprising substitutions at positions 282, 283, and 284 suggested that the C-terminal shift of this loop was beneficial (See Figure 14).
  • the AlphaScreenTM data shows the binding of the ⁇ 281S variant and other variants to FcRn.
  • This AlphaScreenTM data was collected as a direct binding assay. Higher levels of chemiluminescent signals demonstrate stronger binding. As the concentrations of the variants are raised in the assay, stronger signals are created.
  • These data at pH 6.0, in Figures 17a and 17b demonstrate the increased affinity of ⁇ 281 S, P257L, P257L/ ⁇ 281 S (a combination substitution/insertion variant) and other variants over the wild-type Fc. Also shown is a double substitution, T250Q/M428L, shown previously to have an increased serum half in monkeys (Hinton et al., 2004, J. Biol. Chem.
  • FIG. 18 shows the response units created as various Fc variant binding to FcRn on the chip surface. After allowing the variant to fully bind to the chip, the response units are recorded and shown on the ordinate. The insertion, ⁇ 281 S shows binding properties comparable to other variants shown herein to have increased affinity for FcRn over the wild type (See figures 13, 14 and 15, for examples).
  • the deletion variant comprising a deletion of N286, N286#, also shows increased affinity for FcRn over wild type.
  • This variant has a 2.0-fold increase in FcRn affinity as shown in Figure 13.
  • the data therein are also AlphaScreenTM data collected as a competition experiment at pH 6.0.
  • the variants are used to inhibit the binding of wild-type Fc, linked to the donor bead, with FcRn, linked to the acceptor beads. Two-fold less free N286# was needed than free wild-type Fc to inhibit the binding of the donor/acceptor beads through the Fc/FcRn complex. This demonstrates the 2-fold tighter binding of N286# over the wild type.
  • Fc variants comprising insertions or deletions have decreased affinity for FcRn.
  • the insertion variant, ⁇ 254N has greatly decreased FcRn binding as would be expected from the nature and positioning of the variant.
  • This variant places the insertion, an Asn, in the middle of an FcRn binding loop.
  • This insertion has only 1.1 % of the binding of the binding affinity of the wild type ( Figure 13).
  • EXAMPLE 5 Combination Variants with altered FcRn and FcgammaR characteristics.
  • the Fc variant P257L has increased affinity for FcRn relative to WT.
  • P257L gave a median of 2.6-fold increase in FcRn affinity for human FcRn, pH 6.0 in phosphate buffer with 25mM NaCI added.
  • the addition of I332E or S239D/I332E to the P257L variant yielded double and triple variants, P257L/I332E and S239D/P257L/I332E, which retain the increased affinity for FcRn.
  • the variant S239D/I332E has essentially un-altered FcRn binding compared to wild type as shown in the AlphaScreenTM assays in figure 14b. These double and triple variants had a 5- and 4-fold increased affinity.
  • the I332E and S239D/I332E variants have improved binding to FcgammaR, in particular to FcgammaRllla (See US1 1/124620, entirely incorporated by reference).
  • the FcgammaR-binding properties of some variants of the present invention are shown in Figure 25.
  • the protein A binding properties of some variant of the present invention are shown in Figure 26. Protein A binding is frequently used during purification of Fc- containing proteins.
  • V308F also improves FcRn binding at pH 6.0 ( Figure 13e).
  • V308F has 3-fold increased affinity as a single substitution in the anti-HER2 antibody trastuzumab (HerceptinTM, Genentech) and also has increased affinity when combined with substitutions that increase FcgammaR binding, such as I332E, S239D/I332E, and S298A/E333A/K334A (Lazar et al. 2006 Proc. Nat. Acad. Sci USA. 103(11 1 ):4005-4010, Shields et al. 2001 J. Biol. Chem.
  • FcRn binding of G385H is also maintained when combined with FcgammaR improving substitutions, especially in the triple-substitution variant S239D/I332E/G385H.
  • Variants with increased binding to FcRn may be combined with variants that reduce or knock-out binding to FcgammaR and the complement protein, C1 q.
  • the improved binding to FcRn increases the effect from a protecting receptor allowing for improved half-life.
  • Fc containing proteins may also be taken into cells and metabolized through their interaction with the FcgammaR and the C1q protein.
  • the Fc/FcgammaR and Fc/C1q protein interactions are not required for antibody efficacy, deletions of these interactions may be made. Deletions of these interactions may also decrease the effect of a degrading receptor, thereby also allowing for improved half-life.
  • the variants 234G, 235G, 236R, 237K, 267R, 269R, 325A, 325L, and 328R (US1 1/396,495 entirely incorporated by reference) may be combined with FcRn-improving variants to create variants with increased FcRn affinity and decreased FcgammaR or C1 q affinity.
  • variants include 235G/257C, 325A/385H, 325A/257L, 234G/308F, 234G/434Y, and 269R/308F/311V.
  • variants may be made in Fc domains from IgGI , although reduced interactions with the FcgammaR or C1 q may also be achieved by placing these mutations into proteins comprising Fc domains from lgG2, lgG4, or lgG3.
  • FcRn modifications such as 257N, 257L, 257M, 308F, 31 1V into lgG2 allows for a reduction in FcgammaR binding and increased FcRn interactions.
  • Variants with decreased binding to FcRn may be combined with variants that have increased FcgammaR or C1q binding.
  • the decreased FcRn binding combined with increased FcgammaR binding may be beneficial for increasing the amount of the Fc- containing protein available to illicit effector functions. Reducing FcRn binding may reduce the amount of the Fc-containing protein that is sequestered by FcRn and thus affect bioavailability.
  • Modifications such as I253V, S254N, S254# (deletion of 254), T255H, and H435N reduce Fc/FcRn binding ( Figure 13) and may be combined with variants with improved FcgammaR binding such as S239D, I332E, H268E, G236A.
  • the resulting Fc domains such as those comprising I253V/S239D/I332E, I332E/H435N, or S254N/H268E, have reduced FcRn binding and increased FcgammaR binding.
  • Variants with decreased binding to FcRn may be combined with variants with decreased FcgammaR binding.
  • This combination of decreased FcRn and FcgammaR binding is beneficial in applications such as imaging wherein the Fc-containing protein is labeled with a radioactive or toxic tracer.
  • the half-life of the protein comprising the radioactive tracer is similar to the half-life of the radionuclide itself. This allows clearance of the tracer from the body in the same time as the decay of the radionuclide.
  • the reduced FcgammaR interactions also allow optimal availability of the Fc-containing protein for its target. For example, if the Fc-containing protein is an antibody, then the reduce FcgammaR binding allow more antibody to be assessable to antigen.
  • Combinations of FcRn- and FcgammaR-affecting variants such as 235G/254N, 236R/435N, 269R/I253V are good for this application.
  • EXAMPLE 6 Fc variants in antibody OST577 binding to human FcRn.
  • OST577 is an anti-Hepatitis B surface antigen antibody (Ehrlich et al. (1992) Hum. Antibodies Hybridomas 3:2-7, entirely incorporated by reference).
  • Heavy and light chain sequences were taken from the Kabat Database with KADBID 000653 (heavy) and KADBID 007557(light) (Martin AC, Proteins. 1996 May;25(1 ):130-3, entirely incorporated by reference). DNA encoding the heavy and light chains were synthesized by Blue Heron Biolotechnology, Bothell, WA.
  • Wild-type and variant OST577 antibodies were expressed and purified as in the anti-HER2 (trastuzumab) variants in EXAMPLE 1.
  • BiacoreTM binding assays were performed as in EXAMPLE 2, with a human FcRn/Glutathione D transferase (GST) fusion protein attached to the chip surface.
  • GST human FcRn/Glutathione D transferase
  • Fc variants of the present invention have altered binding to human FcRn. Variants with increased binding adhere more easily to the FcRn on the surface and cause a greater rise in Response Units (RU's).
  • the variants shown with modification in the FcRn-binding region all have increased affinity for FcRn compared to the wild-type protein.
  • variants include P257L, P257N, V308F, N434Y, P257L/N434Y and P257L/V308F.
  • EXAMPLE 7 Affinity of Fc variants for human, monkey and mouse FcRn.
  • Fc variants in the anti-HER2 antibody trastuzumab were created as described in EXAMPLE 1.
  • Surface plasmon resonance (SPR) traces were collected as described in EXAMPLE 2, except that human, macaque or mouse FcRn was attached to the chip surface.
  • SPR curves were collected for each Fc variant with differing amounts of GST-FcRn attached to the surface. Each curve was fit to a 1 :1 Langmuir binding model and the two resulting Kd values were averaged to produce a representative value for each variant- receptor pair.
  • the variant V308F/Q311 V has 3.4-fold tigher binding to human FcRn than does the wild type.
  • V308F/Q311 V also has 3.7-fold and 5.1 -fold tigher binding to monkey and mouse FcRn, respectively.
  • the variant M428L has been shown to increase the antibody half-life (Hinton et al. 2004 Journal of Biological Chemistry 279(8):6213-6216, entirely incorporated by reference) and has a 2.4-, 2.0, and 2.1 -fold increased binding to the human, monkey and mouse FcRn's, respectively.
  • P257L, P257N, N434Y, Q31 1V, V308F, V308F/N434Y, P257L/V308F, and P257L/N434Y also show increased binding at pH6.0.
  • EXAMPLE 8 FcRn variants in various Fc domains.
  • Variants of the present invention may be incorporated into any constant domain, using the molecular biology and purification techniques described herein, including those in EXAMPLE 1.
  • Amino acid sequences of the IgGI , lgG2, lgG3, and lgG4 constant domains may be used as listed in Figure 2.
  • combinations of two or more different constant domains may be used.
  • Figure 24 lists some of the modifications found in the present invention incorporated into a hybrid of IgGI and lgG2. This hybrid comprises the lgG2 CH1 domain and the IgGI CH2 and CH3 domains.
  • lgG3 has a lower half-life in humans compared to IgGI , lgG2, and lgG4 (7 days vs -21 days, Janeway, Travers, Walport, Shlomchik. Immunology, 5 th ed. Garland Publishing c2001 , Figure 4-16, incorporated by reference) and is therefore desirable in certain applications.
  • EXAMPLE 9 Creation of variant in an anti-VEGF antibody.
  • Anti-VEGF antibodies with altered binding were produced using the methods described herein, including EXAMPLE 1.
  • the wild type anti-VEGF heavy chain comprises the following sequence of amino acids:
  • Figure 28 which shows the variants produced, the volume of culture media used, and the resulting yield of the antibody variants. Numbering for the variants follows the EU index as in
  • variants listed in Figure 28 were produced in either IgGI or hybrid VH comprising sequences from both IgGI and lgG2. These variants contain the variable region that binds the antigen VEGF. All proteins were judged to be >90% pure by size exclusion chromatography and SDS gel electrophoresis.
  • mice aged 9-12 weeks were injected intravenously with 2 mg/kg antibody in groups of 6 mice per antibody. Blood samples were collected at 1 hr, and days 1 , 4, 8, 11 , 15, 18, 21 , 25 and 28 from the oribital flexus. The concentration of each antiobody in serum was measured with a sandwich ELISA assay using anti-human Fc antibodies and europium detection. [249] The results of the study are shown in Figure 27, which are representative data of two separate studies. The mean and standard deviation of the mean for the four samples are shown. Clearly, the V308F variant has longer half-life, remaining at measurable concentrations out to 25 days.
  • the WT and P257L and P257N variants are cleared more quickly, only having measurable concentrations out to 15, 8, and 4 days, respectively.
  • the serum concentrations as a function of time were fit to a non-compartimental model using the software package, WinNonLin (Pharsight Inc).
  • the terminal half-life of the V308F variant was 4.9 days, whereas the terminal half-lives of the WT and P257L and P257N variants were 3.0, 1.9 and 0.9 days, respectively.
  • the area under the curves (AUC) of the V308F variant was 129 day * ug/ml, whereas those of the WT and P257L and P257N variants were 70, 38 and 38 day * ug/ml, respectively.
  • EXAMPLE 11 FcRn binding experiments at pH 6.0.
  • Anti-VEGF variants of the present invention were tested for their binding ability to human FcRn with Biacore assays as described in EXAMPLE 2 with some modifications.
  • Human FcRn was attached covalently to a CM5 chip in 10 mM sodium acetate, pH 4.5 on using N-hydroxysuccinimide/N-ethyl-N'- ⁇ S-dimethylamino-propyl) carbodiimide (NHS/EDC) at a flow rate of 5 ul/min.
  • the human FcRn used contained GST and HIS tagged version to aid in purification and other assys.
  • the wild-type anti-VEGF antibody had a Kd of 18 nM, which differs considerably from the value reported in DaII' Acqua et al (DaII' Acqua et al Journal of Immunology 2002, 169:5171-5180) because of the differences in assay design and data fitting.
  • Our assay format gave reproducible results if the FcRn chip was used soon after creation. The FcRn chip, however, degraded with use, possibly from the dissociation of either the two FcRn chains from the surface.
  • the results demonstrate the altered binding of the variants compared to wild-type anti-VEGF.
  • Figure 29 shows the fold increase in binding strength relative to the wild-type control.
  • the variant V308F for example, binds FcRn 4.5 fold more tightly than the wild-type antibody.
  • the variant V308F/M428L binds FcRn 12.3 fold more tightly and the variant T307P/V308F binds FcRn 3.16 fold more tightly than the wild-type protein.
  • No variants shown in Figure 29 have reduced affinity for FcRn compared to the wild-type (values would be less than 1.0).
  • the variant N434S has an FcRn binding affinity 4.4 fold stronger than WT, comparable to V308F.
  • FcRn alpha chain and beta-2-microglobulin cDNA was ordered from OriGene Technologies lnc (Rockville, MD) and transfected in 293T cells to express functional FcRn on the cell surface. 20 ug Fcgrt and 40 ug of beta-2-microglobulin DNA was transfected with lipofectamine (Invitrogen Inc.) and the cells were allowed to grow for 3 days in DMEM media with 10% ultra low IgG serum. Control cells not transfected with the two FcRn chains were also grown.
  • Varying amounts of anti-VEGF antibodies were bound to the cells for 30 minutes in 25mM phosphate buffer pH6.0, 15OmM NaCI, 0.5% BSA and then washed 6-9 times in 25mM phosphate buffer pH6.0, 15OmM NaCI, 0.5% BSA plus 0.003% igepal. After washing, antibodies were fixed to the surface by treatment with the binding with 1% PFA. Bound antibodies were than detected using a PE tagged Fab' 2 against human Fab domains and the mean fluorescence intensity (MFI) was measured using a BD FACS Canto II. The average of two samples per antibody are presented in Figure 30.
  • MFI mean fluorescence intensity
  • the curve fits to the data in Figure 30 do not provide interpretable EC50 values because many curves did not form an upper baseline by saturating the cells.
  • the antibodies may be listed from strongest to weakest FcRn affinity as follows: V308F/M428L, V259I/V308F, T250I/V308F, T250Q/M428L, N434S, T307Q/V308F, P257L, T307S/V308F, V308F, T256V/V308F, V308F/L309Y, and WT.
  • Antibodies comprising the modification 434S have particularly favorable properties making them preferred variants of the present invention.
  • the wild-type residue is an asparagine, Asn, at position 434 so that this variant may be referred to as N434S in the context of IgGI or other Fc domains which contain Asn, N, at position 434. More generally, this variant may be referred to simply as 434S.
  • the 434S variant has been produced successfully in both the anti-HER2 antibody trastuzumab and the anti-VEGF antibody.
  • the Ser at position 434 has the ability to hydrogen bond with FcRn either directly or indirectly, ie, mediated by water or solute molecules.
  • the gamma oxygen of Ser at position 434 is in the vicinity of the carbonyl oxygen atoms of Gly131 and Pro134 on the FcRn molecule, as shown in Figure 32.
  • Figure 32 shows a model of the human Fc domain in complex with human FcRn.
  • the Fc domain in the model comprises the 434S substitution, hence residue 434 is Ser in Figure 32.
  • the model is created with PDA® technologies (Dahiyat and Mayo Protein Sci. 1996 May;5(5):895-903), the crystal structure of the rat Fc domain bound to the rat FcRn (Martin et al. MoI Cell. 2001 Apr;7(4):867-77), and Pymol (Delano Scientific).
  • the small size of Ser is easily accomidated in the interface between the two proteins.
  • the antibody variant N434S has a 4.4-fold increased binding affinity for FcRn compared to the wild-type antibody as shown by biacoreTM measurements ( Figure 29).
  • the variant also shows increased binding to cell surface bound FcRn as shown by cell counting measurements ( Figure 30).
  • preferred variants include Y319L, T307Q, V259I, M252Y, V259I/N434S, M428L/N434S, V308F/N434S, M252Y/S254T/T256E/N434S, M252Y/S254T/T256E/V308F, M252Y/S254T/T256E/M428L, V308F/M428L/N434S, V259I/V308F/N434S, T307Q/V308F/N434S, T250I/V308F/N434S, V308F/Y319L/N434S, V259I/V308F/M428L,
  • GIu Lys Thr lie Ser Lys Ala Lys GIy GIn Pro Arg GIu Pro GIn VaI 100 105 110
  • VaI Ser GIu Leu Pro lie VaI His Arg Asp Trp Leu Asn GIy Lys Thr 65 70 75 80
  • GIn lie Asn GIy Thr Phe Thr Leu GIn GIy Leu Leu GIy Cys GIu Leu 85 90 95
  • Rattus norvegicus ⁇ 400> 10 lie GIn Lys Thr Pro GIn lie GIn VaI Tyr Ser Arg His Pro Pro GIu 1 5 10 15
  • Ala Arg lie Tyr Pro Thr Asn GIy Tyr Thr Arg Tyr Ala Asp Ser VaI 50 55 60
  • Lys GIy Arg Phe Thr lie Ser Ala Asp Thr Ser Lys Asn Thr Ala Tyr 65 70 75 80
  • Lys Thr lie Ser Lys Ala Lys GIy GIn Pro Arg GIu Pro GIn VaI Tyr 340 345 350
  • Ser Arg Ser GIy Thr Asp Phe Thr Leu Thr lie Ser Ser Leu GIn Pro 65 70 75 80
  • Lys Pro Lys Asp Thr Leu Met lie Ser Arg Thr Pro GIu VaI Thr Cys
  • Lys Ala Leu Pro Ala Pro lie GIu Lys Thr lie Ser Lys Ala Lys GIy 210 215 220
  • Lys Pro Lys Asp Thr Leu Met lie Ser Arg Thr Leu GIu VaI Thr Cys 130 135 140
  • Lys Ala Leu Pro Ala Pro lie GIu Lys Thr lie Ser Lys Ala Lys GIy 210 215 220
  • Lys Pro Lys Asp Thr Leu Met lie Ser Arg Thr Asn GIu VaI Thr Cys 130 135 140
  • Lys Ala Leu Pro Ala Pro lie GIu Lys Thr lie Ser Lys Ala Lys GIy 210 215 220
  • Lys Pro Lys Asp Thr Leu Met lie Ser Arg Thr Pro GIu VaI Thr Cys 130 135 140
  • Lys Ala Leu Pro Ala Pro lie GIu Lys Thr lie Ser Lys Ala Lys GIy 210 215 220
  • Lys Pro Lys Asp Thr Leu Met lie Ser Arg Thr Pro GIu VaI Thr Cys 130 135 140
  • Lys Ala Leu Pro Ala Pro lie GIu Lys Thr lie Ser Lys Ala Lys GIy 210 215 220
  • Lys Pro Lys Asp Thr Leu Met lie Ser Arg Thr Pro GIu VaI Thr Cys 130 135 140
  • Lys Ala Leu Pro Ala Pro lie GIu Lys Thr lie Ser Lys Ala Lys GIy 210 215 220
  • Lys Pro Lys Asp Thr Leu Met lie Ser Arg Thr Pro GIu VaI Thr Cys 130 135 140
  • Lys Ala Leu Pro Ala Pro lie GIu Lys Thr lie Ser Lys Thr Lys GIy 210 215 220
  • Lys Pro Lys Asp Thr Leu Met lie Ser Arg Thr Leu GIu VaI Thr Cys 130 135 140
  • Lys Ala Leu Pro Ala Pro lie GIu Lys Thr lie Ser Lys Thr Lys GIy 210 215 220
  • Lys Pro Lys Asp Thr Leu Met lie Ser Arg Thr Asn GIu VaI Thr Cys 130 135 140
  • Lys Pro Lys Asp Thr Leu Met lie Ser Arg Thr Pro GIu VaI Thr Cys 130 135 140
  • Lys Ala Leu Pro Ala Pro lie GIu Lys Thr lie Ser Lys Thr Lys GIy 210 215 220
  • Lys Ala Leu Pro Ala Pro lie GIu Lys Thr lie Ser Lys Thr Lys GIy 210 215 220
  • Lys Pro Lys Asp Thr Leu Met lie Ser Arg Thr Pro GIu VaI Thr Cys 130 135 140
  • Lys Ala Leu Pro Ala Pro lie GIu Lys Thr lie Ser Lys Thr Lys GIy 210 215 220

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Immunology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Virology (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Peptides Or Proteins (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

The present application relates to optimized IgG immunoglobulin variants, engineering methods for their generation, and their application, particularly for therapeutic purposes.

Description

Fc VARIANTS WITH ALTERED BINDING TO FcRn
[1] This application claims benefit under 35 U.S.C. §119(e) to USSN 11/932,151 , filed October 31 , 2007.
FIELD OF THE INVENTION
[2] The present application relates to optimized IgG immunoglobulin variants, engineering methods for their generation, and their application, particularly for therapeutic purposes.
BACKGROUND OF THE INVENTION
[3] Antibodies are immunological proteins that bind a specific antigen. In most mammals, including humans and mice, antibodies are constructed from paired heavy and light polypeptide chains. Each chain is made up of individual immunoglobulin (Ig) domains, and thus the generic term immunoglobulin is used for such proteins. Each chain is made up of two distinct regions, referred to as the variable and constant regions. The light and heavy chain variable regions show significant sequence diversity between antibodies, and are responsible for binding the target antigen. The constant regions show less sequence diversity, and are responsible for binding a number of natural proteins to elicit important biochemical events. In humans there are five different classes of antibodies including IgA (which includes subclasses IgAI and lgA2), IgD, IgE, IgG (which includes subclasses IgGI , lgG2, lgG3, and lgG4), and IgM. The distinguishing feature between these antibody classes is their constant regions, although subtler differences may exist in the V region. Figure 1 shows an IgGI antibody, used here as an example to describe the general structural features of immunoglobulins. IgG antibodies are tetrameric proteins composed of two heavy chains and two light chains. The IgG heavy chain is composed of four immunoglobulin domains linked from N- to C-terminus in the order VH-CH 1-CH2-CH3, referring to the heavy chain variable domain, heavy chain constant domain 1 , heavy chain constant domain 2, and heavy chain constant domain 3 respectively (also referred to as VH-Cγ1 -Cγ2-Cγ3, referring to the heavy chain variable domain, constant gamma 1 domain, constant gamma 2 domain, and constant gamma 3 domain respectively). The IgG light chain is composed of two immunoglobulin domains linked from N- to C-terminus in the order VL-CL, referring to the light chain variable domain and the light chain constant domain respectively. [4] The variable region of an antibody contains the antigen binding determinants of the molecule, and thus determines the specificity of an antibody for its target antigen. The variable region is so named because it is the most distinct in sequence from other antibodies within the same class. The majority of sequence variability occurs in the complementarity determining regions (CDRs). There are 6 CDRs total, three each per heavy and light chain, designated VH CDR1 , VH CDR2, VH CDR3, VL CDR1 , VL CDR2, and VL CDR3. The variable region outside of the CDRs is referred to as the framework (FR) region. Although not as diverse as the CDRs, sequence variability does occur in the FR region between different antibodies. Overall, this characteristic architecture of antibodies provides a stable scaffold (the FR region) upon which substantial antigen binding diversity (the CDRs) can be explored by the immune system to obtain specificity for a broad array of antigens. A number of high-resolution structures are available for a variety of variable region fragments from different organisms, some unbound and some in complex with antigen. The sequence and structural features of antibody variable regions are well characterized (Morea et al., 1997, Biophys Chem 68:9-16; Morea et al., 2000, Methods 20:267-279, entirely incorporated by reference), and the conserved features of antibodies have enabled the development of a wealth of antibody engineering techniques (Maynard et al., 2000, Annu Rev Biomed Eng 2:339-376, entirely incorporated by reference). For example, it is possible to graft the CDRs from one antibody, for example a murine antibody, onto the framework region of another antibody, for example a human antibody. This process, referred to in the art as "humanization", enables generation of less immunogenic antibody therapeutics from nonhuman antibodies. Fragments including the variable region can exist in the absence of other regions of the antibody, including for example the antigen binding fragment (Fab) including VH-Cγ1 and VH-CL, the variable fragment (Fv) including VH and VL, the single chain variable fragment (scFv) including VH and VL linked together in the same chain, as well as a variety of other variable region fragments (Little et al., 2000, Immunol Today 21 :364-370, entirely incorporated by reference).
[5] The Fc region of an antibody interacts with a number of Fc receptors and ligands, imparting an array of important functional capabilities referred to as effector functions. For IgG the Fc region, as shown in Figures 1 and 2, comprises Ig domains Cγ2 and Cγ3 and the N-terminal hinge leading into Cγ2. An important family of Fc receptors for the IgG class is the Fc gamma receptors (FcγRs). These receptors mediate communication between antibodies and the cellular arm of the immune system (Raghavan et al., 1996, Annu Rev Cell Dev Biol 12:181-220; Ravetch et al., 2001 , Annu Rev Immunol 19:275-290, both entirely incorporated by reference). In humans this protein family includes FcγRI (CD64), including isoforms FcγRIa, FcγRIb, and FcγRIc; FcγRII (CD32), including isoforms FcγRlla (including allotypes H131 and R131 ), FcγRllb (including FcγRllb-1 and FcγRllb-2), and FcγRllc; and FcγRIII (CD16), including isoforms FcγRllla (including allotypes V158 and F158) and FcγRlllb (including allotypes FcγRlllb-NA1 and FcγRlllb-NA2) (Jefferis et al., 2002, Immunol Lett 82:57-65, entirely incorporated by reference). These receptors typically have an extracellular domain that mediates binding to Fc, a membrane spanning region, and an intracellular domain that may mediate some signaling event within the cell. These receptors are expressed in a variety of immune cells including monocytes, macrophages, neutrophils, dendritic cells, eosinophils, mast cells, platelets, B cells, large granular lymphocytes, Langerhans' cells, natural killer (NK) cells, and γγ T cells. Formation of the Fc/FcγR complex recruits these effector cells to sites of bound antigen, typically resulting in signaling events within the cells and important subsequent immune responses such as release of inflammation mediators, B cell activation, endocytosis, phagocytosis, and cytotoxic attack. The ability to mediate cytotoxic and phagocytic effector functions is a potential mechanism by which antibodies destroy targeted cells. The cell-mediated reaction wherein nonspecific cytotoxic cells that express FcγRs recognize bound antibody on a target cell and subsequently cause lysis of the target cell is referred to as antibody dependent cell-mediated cytotoxicity (ADCC) (Raghavan et al., 1996, Annu Rev Cell Dev Biol 12:181-220; Ghetie et al., 2000, Annu Rev Immunol 18:739-766; Ravetch et al., 2001 , Annu Rev Immunol 19:275- 290, all entirely incorporated by reference). The cell-mediated reaction wherein nonspecific cytotoxic cells that express FcγRs recognize bound antibody on a target cell and subsequently cause phagocytosis of the target cell is referred to as antibody dependent cell- mediated phagocytosis (ADCP). A number of structures have been solved of the extracellular domains of human FcγRs, including FcγRlla (pdb accession code 1 H9V, entirely incorporated by reference)(Sondermann et al., 2001 , J MoI Biol 309:737-749, entirely incorporated by reference) (pdb accession code 1 FCG, entirely incorporated by reference)(Maxwell et al., 1999, Nat Struct Biol 6:437-442, entirely incorporated by reference), FcγRllb (pdb accession code 2FCB, entirely incorporated by reference)(Sondermann et al., 1999, Embo J 18:1095-1 103, entirely incorporated by reference); and FcγRlllb (pdb accession code 1 E4J, entirely incorporated by reference)(Sondermann et al., 2000, Nature 406:267-273, entirely incorporated by reference.). All FcγRs bind the same region on Fc, at the N-terminal end of the Cγ2 domain and the preceding hinge, shown in Figure 1. This interaction is well characterized structurally (Sondermann et al., 2001 , J MoI Biol 309:737-749, entirely incorporated by reference), and several structures of the human Fc bound to the extracellular domain of human FcγRlllb have been solved (pdb accession code 1 E4K, entirely incorporated by reference)(Sondermann et al., 2000, Nature 406:267-273, entirely incorporated by reference) (pdb accession codes 11 IS and 1 IIX, entirely incorporated by reference)(Radaev et al., 2001 , J Biol Chem 276:16469-16477, entirely incorporated by reference), as well as has the structure of the human IgE Fc/FcεRlα complex (pdb accession code 1 F6A, entirely incorporated by reference)(Garman et al., 2000, Nature 406:259-266, entirely incorporated by reference). The effector function response may be modified by variant in the Fc region (Lazar et al. 2006 Proc. Nat. Acad. Sci USA. 103(1 11 ):4005-4010, entirely incorporated by reference).
[6] The different IgG subclasses have different affinities for the FcγRs, with IgGI and lgG3 typically binding substantially better to the receptors than lgG2 and lgG4 (Jefferis et al., 2002, Immunol Lett 82:57-65, entirely incorporated by reference). All FcγRs bind the same region on IgG Fc, yet with different affinities: the high affinity binder FcγRI has a Kd for IgGI of 10"8 M"1, whereas the low affinity receptors FcγRI I and FcγRI 11 generally bind at 10"6 and 10"5 respectively. The extracellular domains of FcγRllla and FcγRlllb are 96% identical; however FcγRlllb does not have an intracellular signaling domain. Furthermore, whereas FcγRI, FcγRlla/c, and FcγRllla are positive regulators of immune complex-triggered activation, characterized by having an intracellular domain that has an immunoreceptor tyrosine-based activation motif (ITAM), FcγRllb has an immunoreceptor tyrosine-based inhibition motif (ITIM) and is therefore inhibitory. Thus the former are referred to as activation receptors, and FcγRllb is referred to as an inhibitory receptor. The receptors also differ in expression pattern and levels on different immune cells. Yet another level of complexity is the existence of a number of FcγR polymorphisms in the human proteome. A particularly relevant polymorphism with clinical significance is V158/F158 FcγRllla. Human IgGI binds with greater affinity to the V158 allotype than to the F158 allotype. This difference in affinity, and presumably its effect on ADCC and/or ADCP, has been shown to be a significant determinant of the efficacy of the anti-CD20 antibody rituximab (Rituxan®, Biogenldec). Patients with the V158 allotype respond favorably to rituximab treatment; however, patients with the lower affinity F158 allotype respond poorly (Cartron et al., 2002, Blood 99:754-758, entirely incorporated by reference). Approximately 10-20% of humans are V158/V158 homozygous, 45% are V158/F158 heterozygous, and 35-45% of humans are F158/F158 homozygous (Lehrnbecher et al., 1999, Blood 94:4220-4232; Cartron et al., 2002, Blood 99:754-758, all entirely incorporated by reference). Thus 80-90% of humans are poor responders, i.e., they have at least one allele of the F158 FcγRllla.
[7] An overlapping but separate site on Fc, shown in Figure 1 , serves as the interface for the complement protein C1q. In the same way that Fc/FcγR binding mediates ADCC, Fc/C1q binding mediates complement dependent cytotoxicity (CDC). C1 q forms a complex with the serine proteases C1 r and C1 s to form the C1 complex. C1 q is capable of binding six antibodies, although binding to two IgGs is sufficient to activate the complement cascade. Similar to Fc interaction with FcγRs, different IgG subclasses have different affinity for C1 q, with IgGI and lgG3 typically binding substantially better to the FcγRs than lgG2 and lgG4 (Jefferis et al., 2002, Immunol Lett 82:57-65, entirely incorporated by reference). [8] In IgG, a site on Fc between the Cg2 and Cg3 domains (Figure 1 ) mediates interaction with the neonatal receptor FcRn, the binding of which recycles endocytosed antibody from the endosome back to the bloodstream (Raghavan et al., 1996, Annu Rev Cell Dev Biol 12:181-220; Ghetie et al., 2000, Annu Rev Immunol 18:739-766, both entirely incorporated by reference). This process, coupled with preclusion of kidney filtration due to the large size of the full-length molecule, results in favorable antibody serum half-lives ranging from one to three weeks. Binding of Fc to FcRn also plays a key role in antibody transport. The binding site on Fc for FcRn is also the site at which the bacterial proteins A and G bind. The tight binding by these proteins is typically exploited as a means to purify antibodies by employing protein A or protein G affinity chromatography during protein purification. Thus the fidelity of this region on Fc is important for both the clinical properties of antibodies and their purification. Available structures of the rat Fc/FcRn complex (Burmeister et al., 1994, Nature, 372:379-383; Martin et al., 2001 , MoI Cell 7:867-877, both entirely incorporated by reference), and of the complexes of Fc with proteins A and G (Deisenhofer, 1981 , Biochemistry 20:2361-2370; Sauer-Eriksson et al., 1995, Structure 3:265-278; Tashiro et al., 1995, Curr Opin Struct Biol 5:471-481 , all entirely incorporated by reference), provide insight into the interaction of Fc with these proteins. The FcRn receptor is also responsible for the transfer of IgG to the neo-natal gut and to the lumen of the intestinal epithelia in adults (Ghetie and Ward, Annu. Rev. Immunol., 2000, 18:739-766; Yoshida et al., Immunity, 2004, 20(6):769-783, both entirely incorporated by reference). [9] Studies of rat and human Fc domains have demonstrated the importance of some Fc residues to the binding of FcRn. The rat and human sequences have about 64% sequence identity in the Fc regions (residues 237-443 in the numbering of Kabat et al.). See figures 3, 4, and 5 for the rat/human alignments of Fc, FcRn heavy chain, and FcRn light chain (beta- 2-microglobulin). A model of the human Fc/FcRn complex has been built from the existing structure of the rat Fc/FcRn complex (Martin et al., 2001 , MoI Cell 7:867-877, entirely incorporated by reference). The rat and human sequences share some residues that are critical for FcRn binding, such as H310 and H435 (Medesan et al., 1997 J. Immunol. 158(5):221-7; Shields et al., 2001 , J. Biol. Chem. 276(9):6591-6604, both entirely incorporated by reference). In many positions, however, the human and rat proteins have different amino acids, giving the residues in the human sequence different environments, and possibly a different identities, than in the rat sequence. This variability limits the ability to transfer characteristics from one homolog to the other homolog.
[10] In the murine Fc, random mutation and phage display selection at the sites, T252, T254, and T256 lead to a triple mutant, T252L/T254S/T256F, that has a 3.5-fold increase in FcRn affinity and a 1.5-fold increase in serum half-life (Ghetie et al., 1997, Nat. Biotech. 15(7): 637-640, entirely incorporated by reference). Disruption of the Fc/FcRn interaction by mutations at positions 253, 310 and 435 also lead to decreased in vivo half-life (Medesan et al J. Immunol. 1997 158(5):2211-7, entirely incorporated by reference). [11] The crystal structures of the rat Fc/FcRn complex identified important Fc residues for FcRn binding (Burmeister et al. Nature. 372:379-383 (1994); Martin et al. Molecular Cell. 7:867-877 (2001 ), both entirely incorporated by reference). The original Fc/FcRn complex structure was solved in 1994 to a resolution of 6 A (Table 2a, Burmeister et al. Nature. 372:379-383 (1994), entirely incorporated by reference). The higher resolution structure, solved in 2001 by Marin et al, showed a more detailed view of the side chains positions (Martin et al. Molecular Cell. 7:867-877 (2001 ), entirely incorporated by reference). This crystal structure of rat Fc bound to rat FcRn was solved using an Fc dimer with one monomer containing the mutations T252G/I253G/T254G/H310E/H433E/H435E, which disrupt FcRn binding, and one monomer containing a wild-type Fc monomer. [12] Mutational studies in human Fey have been done on some of the residues that are important for binding to FcRn and have demonstrated an increased serum half-life. In human Fcγ1 , Hinton et al. mutated three residues individually to the other 19 common amino acids. Hinton et al., found that some point mutants a double mutant increased the FcRn binding affinity (Hinton et al., 2004, J. Biol. Chem. 279(8): 6213-6216. Hinton et al. Journal of Immunology 2006, 176:346-356, both entirely incorporated by reference). Two mutations had increased half-lives in monkeys. Shields et al. mutated residues, almost exclusively to Ala, and studied their binding to FcRn and the FcyR's (Shields et al., 2001 , J. Biol. Chem., 276(9):6591-6604, entirely incorporated by reference).
[13] Dall'Acqua et al. used phage display to select for Fc mutations that bound FcRn with increased affinity (DaN' Acqua et al. 2002, J. Immunol. 169:5171-5180, entirely incorporated by reference). The DNA sequences selected for were primarily double and triple mutants. The reference expressed the proteins encoded by many of their selected sequences and found some that bound to FcRn more tightly than the wild-type Fc. [14] The administration of antibodies and Fc fusion proteins as therapeutics requires injections with a prescribed frequency relating to the clearance and half-life characteristics of the protein. Longer in vivo half-lives allow more seldom injections or lower dosing, which is clearly advantageous. Although the past mutations in the Fc domain have lead to some proteins with increased FcRn binding affinity and in vivo half-lives, these mutations have not identified the optimal mutations and enhanced in vivo half-life.
[15] One feature of the Fc region is the conserved N-linked glycosylation that occurs at N297, shown in Figure 1. This carbohydrate, or oligosaccharide as it is sometimes referred, plays a critical structural and functional role for the antibody, and is one of the principle reasons that antibodies must be produced using mammalian expression systems. Umaήa et al., 1999, Nat Biotechnol 17:176-180; Davies et al., 2001 , Biotechnol Bioeng 74:288-294; Mimura ef a/., 2001 , J Biol Chem 276:45539-45547.; Radaev ef a/., 2001 , J Biol Chem 276:16478-16483; Shields et al., 2001 , J Biol Chem 276:6591-6604; Shields et al., 2002, J Biol Chem 277:26733-26740; Simmons et al., 2002, J Immunol Methods 263:133-147; Radaev et ai, 2001 , J Biol Chem 276:16469-16477; and Krapp et ai, 2003, J MoI Biol 325:979-989, all entirely incorporated by reference).
[16] Antibodies have been developed for therapeutic use. Representative publications related to such therapies include Chamow et ai, 1996, Trends Biotechnol 14:52-60; Ashkenazi et al., 1997, Curr Opin Immunol 9: 195-200, Cragg et al., 1999, Curr Opin Immunol 1 1 :541-547; Glennie et al., 2000, Immunol Today 21 :403-410, McLaughlin et al., 1998, J Clin Oncol 16:2825-2833, and Cobleigh et al., 1999, J Clin Oncol 17:2639-2648, all entirely incorporated by reference. Currently for anticancer therapy, any small improvement in mortality rate defines success. Certain IgG variants disclosed herein enhance the capacity of antibodies to limit further growth or destroy at least partially, targeted cancer cells. [17] Anti-tumor potency of antibodies is via enhancement of their ability to mediate cytotoxic effector functions such as ADCC, ADCP, and CDC. Examples include Clynes et ai, 1998, Proc Natl Acad Sci U S A 95:652-656; Clynes et ai, 2000, Nat Med 6:443-446 and Cartron et al., 2002, Blood 99:754-758, both entirely incorporated by reference. [18] Human IgGI is the most commonly used antibody for therapeutic purposes, and the majority of engineering studies have been constructed in this context. The different isotypes of the IgG class however, including IgGI , lgG2, lgG3, and lgG4, have unique physical, biological, and clinical properties. There is a need in the art to design improved IgGI , lgG2, lgG3, and lgG4 variants. There is a further need to design such variants to improve binding to FcRn and/or increase in vivo half-life as compared to native IgG polypeptides. Additionally, there is a need to combine variants with improved pharmacokinetic properties with variants comprising modifications to improve efficacy through altered FcgammaR binding. The present application meets these and other needs.
SUMMARY OF THE INVENTION
[19] The present application is directed to Fc variants of a parent polypeptide including at least one modification in the Fc region of the polypeptide. In various embodiments, the variant polypeptides exhibit altered binding to FcRn as compared to a parent polypeptide. In certain variations, the modification can be selected from the group consisting of: 246H, 246S, 247D, 247T, 248H, 248P, 248Q, 248R, 248Y, 249T, 249W, 250E, 250I, 250Q, 250V, 251 D, 251 E, 251 H, 2511, 251 K, 251 M, 251 N, 251T, 251V, 251Y, 252Q, 252Y, 253L, 253T, 253V, 254H, 254L, 254N, 254T, 254V, Λ254N, 255E, 255F, 255H, 255K, 255S, 255V, 256E, 256V, 257A, 257C, 257D, 257E, 257F, 257G, 257H, 257I, 257K, 257L, 257M, 257N, 257Q, 257R, 257S, 257T, 257V, 257W, 257Y, 258R, 258V, 259I, 279A, 279D, 279C, 279F, 279G, 279H, 279I, 279K, 279M, 279N, 279P, 279Q, 279Q, 279R, 279S, 279T, 279W, 279Y, 280H, Λ281A, Λ281 D, Λ281S, Λ281T, 282D, 282F, 282H, 282I, 282T, 283F, 283I, 283L, 283Y, 284H, 284K, 284P, 284Q, 284R, 284S, 284Y, 285S, 285V, 286D, 286#, 286L, 287H, 287S, 287V, 287Y, 288H, 288Q, 288S, 305H, 305T, 306F, 306H, 306I, 306N, 306T, 306V, 306Y, 307D, 307P, 307Q, 307S, 307V, 307Y, 308C, 308D, 308E, 308F, 308G, 308H, 308I, 308K, 308L, 308M, 308N, 308Q, 308P, 308R, 308S, 308W, 308Y, 309F, 309H, 309N, 309Q, 309V, 309Y, 310K, 310N, 310T, 311A, 31 1 L, 311 P, 311T, 311V, 311W, 312H, 313Y, 315E, 315G, 315H, 315Q, 315S, 315T, 317H, 317S, 319F, 319F, 319L, 339P, 340P, 341S, 374H, 374S, 376H, 376L, 378H, 378N, 380A, 380T, 380Y, 382H, 383H, 383K, 383Q, 384E, 384G, 384H, 385A, 385C, 385F, 385H, 385I, 385K, 385L, 385M, 385N, 385P, 385Q, 385S, 385T, 385V, 385W, 385Y, 386E, 386K, 387#, 387A, 387H, 387K, 387Q, 389E, 389H, 426E, 426H, 426L, 426N, 426R, 426V, 426Y, 427I, 428F, 428L, 429D, 429F, 429K, 429N, 429Q, 429S, 429T, 429Y, 430D, 430H, 430K, 430L, 430Q, 430Y, 431 G, 431 H, 4311, 431 P, 431 P, 431 S, 432F, 432H, 432N, 432S, 432V, 433E, 433P, 433S, 434A, 434F, 434H, 434L, 434M, 434Q, 434S, 434Y, 435N, 436E, 436F, 436L, 436V, 436W, 437E, 437V, 438H, and 438K, where the numbering is according to the EU Index in Kabat et al. and Λ is an insertion after the identified position and # is a deletion of the identified position.
[20] In another variation, the Fc variant includes at least two modifications selected from the group consisting of: 250Q/252Y, 250Q/256E, 250Q/286D, 250Q/308F, 250Q/308Y, 250Q/31 1A, 250Q/31 1V, 250Q/380A, 250Q/428L, 250Q/428F, 250Q/434H, 250Q/434F, 250Q/434Y, 250Q/434A, 250Q/434M, and 250Q/434S.
[21] In another variation, the Fc variant includes at least two modifications selected from the group consisting of: 250E/252Y, 250E/256E, 250E/286D, 250E/308F, 250E/308Y, 250E/31 1A, 250E/31 1V, 250E/380A, 250E/428L, 250E/428F, 250E/434H, 250E/434F, 250E/434Y, 250E/434A, 250E/434M, and 250E/434S.
[22] In another variation, the Fc variant includes at least two modifications selected from the group consisting of: 252Y/250Q, 252Y/250E, 252Y/256E, 252Y/286D, 252Y/308F, 252Y/308Y, 252Y/31 1A, 252Y/311V, 252Y/380A, 252Y/428L, 252Y/428F, 252Y/434H, 252Y/434F, 252Y/434Y, 252Y/434A, 252Y/434M, and 252Y/434S. [23] In another variation, the Fc variant includes at least two modifications selected from the group consisting of: 256E/250Q, 256E/250E, 256E/252Y, 256E/286D, 256E/308F, 256E/308Y, 256E/31 1A, 256E/311V, 256E/380A, 256E/428L, 256E/428F, 256E/434H, 256E/434F, 256E/434Y, 256E/434A, 256E/434M, and 256E/434S. [24] In another variation, the Fc variant includes at least two modifications selected from the group consisting of: 286D/250Q, 286D/250E, 286D/252Y, 286D/256E, 286D/308F, 286D/308Y, 286D/31 1A, 286D/31 1V, 286D/380A, 286D/428L, 286D/428F, 286D/434H,
286D/434F, 286D/434Y, 286D/434A, 286D/434M, and 286D/434S.
[25] In another variation, the Fc variant includes at least two modifications selected from the group consisting of: 308F/250Q, 308F/250E, 308F/252Y, 308F/256E, 308F/286D,
308F/311A, 308F/31 1V, 308F/380A, 308F/428L, 308F/428F, 308F/434H, 308F/434F,
308F/434Y, 308F/434A, 308F/434M, and 308F/434S.
[26] In another variation, the Fc variant includes at least two modifications selected from the group consisting of: 308Y/250Q, 308Y/250E, 308Y/252Y, 308Y/256E, 308Y/286D,
308Y/31 1A, 308Y/31 1V, 308Y/380A, 308Y/428L, 308Y/428F, 308Y/434H, 308Y/434F,
308Y/434Y, 308Y/434A, 308Y/434M, and 308Y/434S.
[27] In another variation, the Fc variant includes at least two modifications selected from the group consisting of: 311 A/250Q, 311 A/250E, 31 1A/252Y, 31 1A/256E, 311 A/286D,
31 1A/308F, 31 1A/308Y, 31 1A/380A, 311A/428L, 311A/428F, 31 1A/434H, 311A/434F,
31 1A/434Y, 311A/434A, 311A/434M, and 311A/434S.
[28] In another variation, the Fc variant includes at least two modifications selected from the group consisting of: 311 V/250Q, 311 V/250E, 31 1V/252Y, 31 1V/256E, 311 V/286D,
31 1V/308F, 31 1V/308Y, 31 1V/380A, 311V/428L, 311V/428F, 31 1V/434H, 311V/434F,
31 1V/434Y, 311V/434A, 311V/434M, and 311V/434S.
[29] In another variation, the Fc variant includes at least two modifications selected from the group consisting of: 380A/250Q, 380A/250E, 380A/252Y, 380A/256E, 380A/286D,
380A/308F, 380A/308Y, 380A/31 1A, 380A/31 1V, 380A/428L, 380A/428F, 380A/434H,
380A/434F, 380A/434Y, 380A/434A, 380A/434M, and 380A/434S.
[30] In another variation, the Fc variant includes at least two modifications selected from the group consisting of: 428L/250Q, 428L/250E, 428L/252Y, 428L/256E, 428L/286D,
428L/308F, 428L/308Y, 428L/311A, 428L/311V, 428L/380A, 428L/434H, 428L/434F,
428L/434Y, 428L/434A, 428L/434M, and 428L/434S.
[31] In another variation, the Fc variant includes at least two modifications selected from the group consisting of: 434H/250Q, 434H/250E, 434H/252Y, 434H/256E, 434H/286D,
434H/308F, 434H/308Y, 434H/311A, 434H/31 1V, 434H/380A, 434H/428L, and 434H/428F.
[32] In another variation, the Fc variant includes at least two modifications selected from the group consisting of: 434F/250Q, 434F/250E, 434F/252Y, 434F/256E, 434F/286D,
434F/308F, 434F/308Y, 434F/311A, 434F/31 1V, 434F/380A, 434F/428L, and 434F/428F.
[33] In another variation, the Fc variant includes at least two modifications selected from the group consisting of: 434Y/250Q, 434Y/250E, 434Y/252Y, 434Y/256E, 434Y/286D,
434Y/308F, 434Y/308Y, 434Y/31 1A, 434Y/31 1V, 434Y/380A, 434Y/428L, and 434Y/428F. [34] In another variation, the Fc variant includes at least two modifications selected from the group consisting of: 434A/250Q, 434A/250E, 434A/252Y, 434A/256E, 434A/286D, 434A/308F, 434A/308Y, 434A/31 1A, 434A/31 1V, 434A/380A, 434A/428L, and 434A/428F. [35] In another variation, the Fc variant includes at least two modifications selected from the group consisting of: 434M/250Q, 434M/250E, 434M/252Y, 434M/256E, 434M/286D, 434M/308F, 434M/308Y, 434M/311A, 434M/311V, 434M/380A, 434M/428L, and 434M/428F.
[36] In another variation, the Fc variant includes at least two modifications selected from the group consisting of: 434S/250Q, 434S/250E, 434S/252Y, 434S/256E, 434S/286D, 434S/308F, 434S/308Y, 434S/31 1A, 434S/31 1V, 434S/380A, 434S/428L, and 434S/428F. [37] In another variation, the Fc variant includes at least one modification selected from the group consisting of: Y319L, T307Q, V259I, M252Y, V259I/N434S, M428L/N434S, V308F/N434S, M252Y/S254T/T256E/N434S, M252Y/S254T/T256E/V308F, M252Y/S254T/T256E/M428L, V308F/M428L/N434S, V259I/V308F/N434S, T307Q/V308F/N434S, T250I/V308F/N434S, V308F/Y319L/N434S, V259I/V308F/M428L, V259I/T307Q/V308F, T250I/V259I/V308F, V259I/V308F/Y319L, T307Q/V308F/L309Y, T307Q/V308F/Y319L, and T250Q/V308F/M428L.
[38] In another variation, the Fc variant includes at least one modification selected from the group consisting of: Y319L, T307Q, V259I, M252Y, V259I/N434S, M428L/N434S, V308F/N434S, V308F/M428L/N434S, V259I/V308F/N434S, T307Q/V308F/N434S, T250I/V308F/N434S, V308F/Y319L/N434S, V259I/V308F/M428L, V259I/T307Q/V308F, T250I/V259I/V308F, V259I/V308F/Y319L, T307Q/V308F/L309Y, T307Q/V308F/Y319L, and T250Q/V308F/M428L.
[39] In another variation, the Fc variant includes at least one modification selected from the group consisting of: 250I, 250V, 252Q, 252Y, 254T, 256V, 259I, 307P, 307Q, 307S, 308F, 309N, 309Y, 311 P, 319F, 319L, 428L, and 434S.
[40] In another variation, the Fc variant includes at least one modification selected from the group consisting of: 250V/308F, 250I/308F, 254T/308F, 256V/308F, 259I/308F, 307P/208F, 307Q/308F, 307S/308F, 308F/309Y, 308F/309Y, V308F/311 P, 308F/319L, 308F/319F, 308F/428L, 252Q/308F, M252Y/S254T/T256E, 259I/434S, 428L/434S, 308F/434S, 308F/428L/434S, 259I/308F/434S, 307Q/308F/434S, 250I/308F/434S, 308F/319L/434S, 259I/308F/428L, 259I/307Q/308F, 250I/259I/308F, 259I/308F/319L, 307Q/308F/309Y, 307Q/308F/319L, and 250Q/308F/428L.
[41] In another variation, the invention includes a method of treating a patient in need of said treatment comprising administering an effective amount of an Fc variant described herein. [42] In another variation, the invention includes a method of increasing the half-life of an antibody or immunoadhesin by modifying an Fc according to the modifications described herein.
BRIEF DESCRIPTION OF THE DRAWINGS
[43] Figure 1. Antibody structure and function. Shown is a model of a full length human IgGI antibody, modeled using a humanized Fab structure from pdb accession code 1 CE1 (James et al., 1999, J MoI Biol 289:293-301 , entirely incorporated by reference) and a human IgGI Fc structure from pdb accession code 1 DN2 (DeLano et al., 2000, Science 287:1279-1283, entirely incorporated by reference). The flexible hinge that links the Fab and Fc regions is not shown. IgGI is a homodimer of heterodimers, made up of two light chains and two heavy chains. The Ig domains that comprise the antibody are labeled, and include VL and CL for the light chain, and VH, Cgammai (Cγ1 ), Cgamma2 (Cγ2), and Cgamma3 (Cγ3) for the heavy chain. The Fc region is labeled. Binding sites for relevant proteins are labeled, including the antigen binding site in the variable region, and the binding sites for FcγRs, FcRn, C1 q, and proteins A and G in the Fc region.
[44] Figure 2. Human IgG sequences (SEQ ID NOS: 1-4) used in the present invention with the EU numbering as in Kabat et al.
[45] Figure 3. Example human and rodent IgG sequences (SEQ ID NOS: 5-6) used in the present invention with the EU numbering as in Kabat.
[46] Figure 4. Example human and rodent FcRn heavy chain sequences (SEQ ID NOS: 7-8) used in the present invention.
[47] Figure 5. Example human and rodent beta-2-microglobulin sequences (SEQ ID NOS: 9-10) used in the present invention.
[48] Figure 6. A human Fc/FcRn complex model created from the rat structures (Burmeister et al., 1994, Nature, 372:379-383; Martin et al., 2001 , MoI Cell 7:867-877, both entirely incorporated by reference). Some histidine residues are shown in space-filling atoms on the FcRn chains (light grey) and Fc polypeptide (dark grey). [49] Figure 7. Illustration of some concepts used in the design of variants comprising insertions or deletions.
[50] Figure 8a-8b. Variants of the present invention. [51] Figure 9a-9b. Variants of the present invention. [52] Figure 10a-1 Ob. Variants of the present invention.
[53] Figure 11. Diagram of the vector pcDNA3.1 Zeo+, which may be used in the construct of Fc variants.
[54] Figure 12a-12b. Competition FcRn binding data of wild-type Fc and Fc variants of the present invention. In each panel, the Fc variants of the present invention are shown as the left (red or dark grey) curve and the wild-type anti-HER2 antibody is shown as the right (blue or light grey) curve.
[55] Figures 13a - 13j. Summary of FcRn binding properties of the Fc variants. The columns from left to right show the FcRn binding modifications, the immunoglobulin used, other modifications, the relative FcRn affinity by AlphaScreen™ competition assays compared to wild type (median value), and the number of assays performed. Relative FcRn affinity numbers greater than 1.0 demonstrate increased binding over wild type. Data were collected at pH 6.0 (0.1 M sodium phosphate, 25mM sodium chloride). [56] Figure 14a-14d. FcRn binding data of Fc variants. The Fc variants are in alemtuzumab or anti-HER2 antibody. Shown are the fold-increases in binding compared to wild type, that is, numbers greater than one indicate tighter binding to FcRn whereas numbers less than one indicate reduced binding to FcRn.
[57] Figure 15. Summary of surface plasmon resonance experiments of Fc variants with improved binding to FcRn. The bar graph shows the fold-increase in FcRn binding affinity of each variant relative to wild-type Fc domain.
[58] Figure 16a-16b. Surface plasmon resonance experiments of wild-type antibody and variants of the present invention. The traces shown are the association and dissociation of the Fc variant antibody to FcRn at pH6.0.
[59] Figure 17a-17c. Binding assays of Fc variants of the present invention to FcRn. Shown are direct binding assays measured by AlphaScreen™ at pH 6.0 (a and b) and pH 7.0 (C).
[60] Figure 18. Binding assays of Fc variants of the present invention to FcRn. Shown are the surface plasmon resonance units created upon binding of the variant Fc to surface- bound FcRn.
[61] Figure 19. Surface plasmon resonance measurement of the binding affinity of Fc variants of the present invention to human FcRn at pH 6.0.
[62] Figure 20. Summary of surface plasmon resonance (SPR) measurements of the binding affinity of Fc variants of the present invention with human, macaque and mouse FcRn. Numbers greater than one indicate increased binding of the variant Fc to FcRn as determined by fitting SPR curves to a 1 :1 Langmuir binding model. [63] Figure 21a-21 b. Summary of FcRn binding properties of the Fc variants. The columns from left to right show the FcRn binding modifications, the immunoglobulin used, other modifications, the relative FcRn affinity by AlphaScreen™ competition assays compared to wild type (median value), and the number of assays performed. Relative FcRn affinity numbers greater than 1.0 demonstrate increased binding over wild type. Data were collected at pH 6.0 (0.1 M sodium phosphate, 125mM sodium chloride). [64] Figure 22. Amino acid sequences (SEQ ID NO: 1 1-12) of the anti-HER2 antibody heavy and light chains.
[65] Figure 23. Amino acid sequences (SEQ ID NO: 13-18) of the constant regions (CH 1 to CH3) of the some IgGI heavy chains used herein.
[66] Figure 24. Amino acid sequences (SEQ ID NO: 19-24) of the constant regions (CH 1 to CH3) of the some hybrid lgG1/2 heavy chains used herein.
[67] Figure 25. Fc variants binding to the human FcgammaRIIIA (V158 Allotype) as determined with AlphaScreen™ competition assays.
[68] Figure 26. Fc variants binding binding to protein A as determined with AlphaScreen™ competition assays.
[69] Figure 27. Serum concentrations of WT and variants of antibodies in human FcRn knockin mice. Anti-VEGF antibodies used were the WT (open squares), V308F (closed squares), P257L (closed triagles) and P257N (crosses).
[70] Figure 28. Examples of FcRn binding variants of the present invention. Anti-VEGF antibodies are listed with the volume of culture media and the yield of purified protein.
[71] Figure 29. Binding affinity of variants of the present invention to human FcRn at pH6.0. The values shown are fold increase in binding strength of the variant in question to the wild-type antibody. For example, the variant 434S binds to FcRn 4.4-fold more tightly than does the wild-type antibody.
[72] Figure 30. Binding of WT and variant antibodies to FcRn on the surface of 293T cells.
[73] Figure 31 a-31 b. Combination variants of the present invention comprising multiple substitutions.
[74] Figure 32. A picture of the intereactions of a variant human CH3 domain comprising
434S, labeled Ser434, and human FcRn.
DETAILED DESCRIPTION OF THE INVENTION
[75] The present invention discloses the generation of novel variants of Fc domains, including those found in antibodies, Fc fusions, and immuno-adhesions, which have an increased binding to the FcRn receptor. As noted herein, binding to FcRn results in longer serum retention in vivo.
[76] In order to increase the retention of the Fc proteins in vivo, the increase in binding affinity must be at around pH 6 while maintaining lower affinity at around pH 7.4. Although still under examination, Fc regions are believed to have longer half-lives in vivo, because binding to FcRn at pH 6 in an endosome sequesters the Fc (Ghetie and Ward, 1997 Immunol Today. 18(12): 592-598, entirely incorporated by reference). The endosomal compartment then recycles the Fc to the cell surface. Once the compartment opens to the extracellular space, the higher pH, -7.4, induces the release of Fc back into the blood. In mice, Dal!' Acqua et al. showed that Fc mutants with increased FcRn binding at pH 6 and pH 7.4 actually had reduced serum concentrations and the same half life as wild-type Fc (DaII' Acqua et al. 2002, J. Immunol. 169:5171-5180, entirely incorporated by reference). The increased affinity of Fc for FcRn at pH 7.4 is thought to forbid the release of the Fc back into the blood. Therefore, the Fc mutations that will increase Fc's half-life in vivo will ideally increase FcRn binding at the lower pH while still allowing release of Fc at higher pH. The amino acid histidine changes its charge state in the pH range of 6.0 to 7.4. Therefore, it is not surprising to find His residues at important positions in the Fc/FcRn complex (Figure 6.) [77] An additional aspect of the invention is the increase in FcRn binding over wild type specifically at lower pH, about pH 6.0, to facilitate Fc/FcRn binding in the endosome. Also disclosed are Fc variants with altered FcRn binding and altered binding to another class of Fc receptors, the FcγR's (sometimes written FcgammaR's) as differential binding to FcγRs, particularly increased binding to FcγRlllb and decreased binding to FcγRllb, has been shown to result in increased efficacy. [78] Definitions
[79] In order that the application may be more completely understood, several definitions are set forth below. Such definitions are meant to encompass grammatical equivalents. [80] By "ADCC" or "antibody dependent cell-mediated cytotoxicity" as used herein is meant the cell-mediated reaction wherein nonspecific cytotoxic cells that express FcγRs recognize bound antibody on a target cell and subsequently cause lysis of the target cell. [81] By "ADCP" or antibody dependent cell-mediated phagocytosis as used herein is meant the cell-mediated reaction wherein nonspecific cytotoxic cells that express FcγRs recognize bound antibody on a target cell and subsequently cause phagocytosis of the target cell.
[82] By "modification" herein is meant an amino acid substitution, insertion, and/or deletion in a polypeptide sequence or an alteration to a moiety chemically linked to a protein. For example, a modification may be an altered carbohydrate or PEG structure attached to a protein. By "amino acid modification" herein is meant an amino acid substitution, insertion, and/or deletion in a polypeptide sequence.
[83] By "amino acid substitution" or "substitution" herein is meant the replacement of an amino acid at a particular position in a parent polypeptide sequence with another amino acid. For example, the substitution E272Y refers to a variant polypeptide, in this case an Fc variant, in which the glutamic acid at position 272 is replaced with tyrosine. [84] By "amino acid insertion" or "insertion" as used herein is meant the addition of an amino acid sequence at a particular position in a parent polypeptide sequence. For example, -233E or Λ233E designates an insertion of glutamic acid after position 233 and before position 234. Additionally, -233ADE or Λ233ADE designates an insertion of AlaAspGlu after position 233 and before position 234.
[85] By "amino acid deletion" or "deletion" as used herein is meant the removal of an amino acid sequence at a particular position in a parent polypeptide sequence. For example, E233- or E233# designates a deletion of glutamic acid at position 233. Additionally, EDA233- or EDA233# designates a deletion of the sequence GluAspAla that begins at position 233. [86] By "variant protein" or "protein variant", or "variant" as used herein is meant a protein that differs from that of a parent protein by virtue of at least one amino acid modification. Protein variant may refer to the protein itself, a composition comprising the protein, or the amino sequence that encodes it. Preferably, the protein variant has at least one amino acid modification compared to the parent protein, e.g. from about one to about ten amino acid modifications, and preferably from about one to about five amino acid modifications compared to the parent. The protein variant sequence herein will preferably possess at least about 80% homology with a parent protein sequence, and most preferably at least about 90% homology, more preferably at least about 95% homology. Variant protein can refer to the variant protein itself, compositions comprising the protein variant, or the DNA sequence that encodes it. Accordingly, by "antibody variant" or "variant antibody" as used herein is meant an antibody that differs from a parent antibody by virtue of at least one amino acid modification, "IgG variant" or "variant IgG" as used herein is meant an antibody that differs from a parent IgG by virtue of at least one amino acid modification, and "immunoglobulin variant" or "variant immunoglobulin" as used herein is meant an immunoglobulin sequence that differs from that of a parent immunoglobulin sequence by virtue of at least one amino acid modification. "Fc variant" or "variant Fc" as used herein is meant a protein comprising a modification in an Fc domain. The Fc variants of the present invention are defined according to the amino acid modifications that compose them. Thus, for example, I332E is an Fc variant with the substitution I332E relative to the parent Fc polypeptide. Likewise, S239D/I332E/G236A defines an Fc variant with the substitutions S239D, I332E, and G236A relative to the parent Fc polypeptide. The identity of the WT amino acid may be unspecified, in which case the aforementioned variant is referred to as 239D/332E/236A. It is noted that the order in which substitutions are provided is arbitrary, that is to say that, for example, S239D/I332E/G236A is the same Fc variant as G236A/S239D/I332E, and so on. For all positions discussed in the present invention, numbering is according to the EU index or EU numbering scheme (Kabat et al., 1991 , Sequences of Proteins of Immunological Interest, 5th Ed., United States Public Health Service, National Institutes of Health, Bethesda, hereby entirely incorporated by reference). The EU index or EU index as in Kabat or EU numbering scheme refers to the numbering of the EU antibody (Edelman et al., 1969, Proc Natl Acad Sci USA 63:78-85, hereby entirely incorporated by reference.) The modification can be an addition, deletion, or substitution. Substitutions can include naturally occurring amino acids and non-naturally occurring amino acids. Variants may comprise non-natural amino acids. Examples include US6586207; WO 98/48032; WO 03/073238; US2004-0214988A1 ; WO 05/35727A2; WO 05/74524A2; J. W. Chin et al., (2002), Journal of the American Chemical Society 124:9026-9027; J. W. Chin, & P. G. Schultz, (2002), ChemBioChem 11 :1 135-1137; J. W. Chin, et al., (2002), PICAS United States of America 99:11020-1 1024; and, L. Wang, & P. G. Schultz, (2002), Chem. 1-10, all entirely incorporated by reference. [87] As used herein, "protein" herein is meant at least two covalently attached amino acids, which includes proteins, polypeptides, oligopeptides and peptides. The peptidyl group may comprise naturally occurring amino acids and peptide bonds, or synthetic peptidomimetic structures, i.e. "analogs", such as peptoids (see Simon et al., PNAS USA 89(20):9367 (1992), entirely incorporated by reference). The amino acids may either be naturally occurring or non-naturally occurring; as will be appreciated by those in the art. For example, homo-phenylalanine, citrulline, and noreleucine are considered amino acids for the purposes of the invention, and both D- and L- (R or S) configured amino acids may be utilized. The variants of the present invention may comprise modifications that include the use of unnatural amino acids incorporated using, for example, the technologies developed by Schultz and colleagues, including but not limited to methods described by Cropp & Shultz, 2004, Trends Genet. 20(12):625-30, Anderson et al., 2004, Proc Natl Acad Sci USA 101 (2):7566-71 , Zhang et al., 2003, 303(5656):371-3, and Chin et al., 2003, Science 301 (5635):964-7, all entirely incorporated by reference. In addition, polypeptides may include synthetic derivatization of one or more side chains or termini, glycosylation, PEGylation, circular permutation, cyclization, linkers to other molecules, fusion to proteins or protein domains, and addition of peptide tags or labels.
[88] By "residue" as used herein is meant a position in a protein and its associated amino acid identity. For example, Asparagine 297 (also referred to as Asn297, also referred to as N297) is a residue in the human antibody IgGI .
[89] By "Fab" or "Fab region" as used herein is meant the polypeptide that comprises the VH, CH1 , VL, and CL immunoglobulin domains. Fab may refer to this region in isolation, or this region in the context of a full length antibody, antibody fragment or Fab fusion protein. [90] By "IqG subclass modification" as used herein is meant an amino acid modification that converts one amino acid of one IgG isotype to the corresponding amino acid in a different, aligned IgG isotype. For example, because IgGI comprises a tyrosine and lgG2 a phenylalanine at EU position 296, a F296Y substitution in lgG2 is considered an IgG subclass modification.
[91] By "non-naturally occurring modification" as used herein is meant an amino acid modification that is not isotypic. For example, because none of the IgGs comprise a glutamic acid at position 332, the substitution I332E in IgGI , lgG2, lgG3, or lgG4 is considered a non- naturally occuring modification.
[92] By "amino acid" and "amino acid identity" as used herein is meant one of the 20 naturally occurring amino acids or any non-natural analogues that may be present at a specific, defined position.
[93] By "effector function" as used herein is meant a biochemical event that results from the interaction of an antibody Fc region with an Fc receptor or ligand. Effector functions include but are not limited to ADCC, ADCP, and CDC.
[94] By "effector cell" as used herein is meant a cell of the immune system that expresses one or more Fc receptors and mediates one or more effector functions. Effector cells include but are not limited to monocytes, macrophages, neutrophils, dendritic cells, eosinophils, mast cells, platelets, B cells, large granular lymphocytes, Langerhans' cells, natural killer (NK) cells, and γδT cells, and may be from any organism including but not limited to humans, mice, rats, rabbits, and monkeys.
[95] By "IgG Fc ligand" as used herein is meant a molecule, preferably a polypeptide, from any organism that binds to the Fc region of an IgG antibody to form an Fc / Fc ligand complex. Fc ligands include but are not limited to FcγRs, FcγRs, FcγRs, FcRn, C1q, C3, mannan binding lectin, mannose receptor, staphylococcal protein A, streptococcal protein G, and viral FcγR. Fc ligands also include Fc receptor homologs (FcRH), which are a family of Fc receptors that are homologous to the FcγRs (Davis et al., 2002, Immunological Reviews 190:123-136, entirely incorporated by reference). Fc ligands may include undiscovered molecules that bind Fc. Particular IgG Fc ligands are FcRn and Fc gamma receptors. By "Fc ligand" as used herein is meant a molecule, preferably a polypeptide, from any organism that binds to the Fc region of an antibody to form an Fc / Fc ligand complex. [96] By "Fc gamma receptor", "FcγR" or "FcgammaR" as used herein is meant any member of the family of proteins that bind the IgG antibody Fc region and is encoded by an FcγR gene. In humans this family includes but is not limited to FcγRI (CD64), including isoforms FcγRIa, FcγRIb, and FcγRIc; FcγRII (CD32), including isoforms FcγRlla (including allotypes H131 and R131 ), FcγRllb (including FcγRllb-1 and FcγRllb-2), and FcγRllc; and FcγRIII (CD16), including isoforms FcγRllla (including allotypes V158 and F158) and FcγRlllb (including allotypes FcγRlllb-NA1 and FcγRlllb-NA2) (Jefferis et al., 2002, Immunol Lett 82:57-65, entirely incorporated by reference), as well as any undiscovered human FcγRs or FcγR isoforms or allotypes. An FcγR may be from any organism, including but not limited to humans, mice, rats, rabbits, and monkeys. Mouse FcγRs include but are not limited to FcγRI (CD64), FcγRII (CD32), FcγRIII (CD16), and FcγRIII-2 (CD16-2), as well as any undiscovered mouse FcγRs or FcγR isoforms or allotypes. [97] By "FcRn" or "neonatal Fc Receptor" as used herein is meant a protein that binds the IgG antibody Fc region and is encoded at least in part by an FcRn gene. The FcRn may be from any organism, including but not limited to humans, mice, rats, rabbits, and monkeys. As is known in the art, the functional FcRn protein comprises two polypeptides, often referred to as the heavy chain and light chain. The light chain is beta-2-microglobulin and the heavy chain is encoded by the FcRn gene. Unless other wise noted herein, FcRn or an FcRn protein refers to the complex of FcRn heavy chain with beta-2-microglobulin. Sequences of particular interest of FcRn are shown in the Figures, particularly the human species. [98] By "parent polypeptide" as used herein is meant an unmodified polypeptide that is subsequently modified to generate a variant. The parent polypeptide may be a naturally occurring polypeptide, or a variant or engineered version of a naturally occurring polypeptide. Parent polypeptide may refer to the polypeptide itself, compositions that comprise the parent polypeptide, or the amino acid sequence that encodes it. Accordingly, by "parent immunoglobulin" as used herein is meant an unmodified immunoglobulin polypeptide that is modified to generate a variant, and by "parent antibody" as used herein is meant an unmodified antibody that is modified to generate a variant antibody. It should be noted that "parent antibody" includes known commercial, recombinantly produced antibodies as outlined below.
[99] By "position" as used herein is meant a location in the sequence of a protein. Positions may be numbered sequentially, or according to an established format, for example the EU index as in Kabat. For example, position 297 is a position in the human antibody IgGL
[100] By "target antigen" as used herein is meant the molecule that is bound specifically by the variable region of a given antibody. A target antigen may be a protein, carbohydrate, lipid, or other chemical compound.
[101] By "target cell" as used herein is meant a cell that expresses a target antigen. [102] By "variable region" as used herein is meant the region of an immunoglobulin that comprises one or more Ig domains substantially encoded by any of the VK, Vλ, and/or VH genes that make up the kappa, lambda, and heavy chain immunoglobulin genetic loci respectively.
[103] By "wild type or WT" herein is meant an amino acid sequence or a nucleotide sequence that is found in nature, including allelic variations. A WT protein has an amino acid sequence or a nucleotide sequence that has not been intentionally modified. [104] The present invention is directed to antibodies that exhibit moduluated binding to FcRn (modulation including increased as well as decreased binding). For example, in some instances, increased binding results in cellular recycling of the antibody and hence increased half-life, for example for therapeutic antibodies. Alternatively, decreased FcRn binding is desirable, for example for diagnostic antibodies or therapeutic antibodies that contain radiolabels. In addition, antibodies exhibiting increased binding to FcRn and altered binding to other Fc receptors, eg. FcvRs, find use in the present invention. Accordingly, the present invention provides antibodies. Antibodies
[105] The present application is directed to antibodies that include amino acid modifications that modulate binding to FcRn. Of particular interest are antibodies that minimally comprise an Fc region, or functional variant thereof, that displays increased binding affinity to FcRn at lowered pH, and do not exhibit substantially altered binding at higher pH.
[106] Traditional antibody structural units typically comprise a tetramer. Each tetramer is typically composed of two identical pairs of polypeptide chains, each pair having one "light" (typically having a molecular weight of about 25 kDa) and one "heavy" chain (typically having a molecular weight of about 50-70 kDa). Human light chains are classified as kappa and lambda light chains. Heavy chains are classified as mu, delta, gamma, alpha, or epsilon, and define the antibody's isotype as IgM, IgD, IgG, IgA, and IgE, respectively. IgG has several subclasses, including, but not limited to IgGI , lgG2, lgG3, and lgG4. IgM has subclasses, including, but not limited to, IgMI and lgM2. Thus, "isotype" as used herein is meant any of the subclasses of immunoglobulins defined by the chemical and antigenic characteristics of their constant regions. The known human immunoglobulin isotypes are IgGI , lgG2, lgG3, lgG4, IgAI , lgA2, IgMI , lgM2, IgD, and IgE.
[107] The amino-terminal portion of each chain includes a variable region of about 100 to 110 or more amino acids primarily responsible for antigen recognition. In the variable region, three loops are gathered for each of the V domains of the heavy chain and light chain to form an antigen-binding site. Each of the loops is referred to as a complementarity- determining region (hereinafter referred to as a "CDR"), in which the variation in the amino acid sequence is most significant.
[108] The carboxy-terminal portion of each chain defines a constant region primarily responsible for effector function. Kabat et al. collected numerous primary sequences of the variable regions of heavy chains and light chains. Based on the degree of conservation of the sequences, they classified individual primary sequences into the CDR and the framework and made a list thereof (see SEQUENCES OF IMMUNOLOGICAL INTEREST, 5th edition, NIH publication, No. 91-3242, E.A. Kabat et al., entirely incorporated by reference).
[109] In the IgG subclass of immunoglobulins, there are several immunoglobulin domains in the heavy chain. By "immunoglobulin (Ig) domain" herein is meant a region of an immunoglobulin having a distinct tertiary structure. Of interest in the present invention are the heavy chain domains, including, the constant heavy (CH) domains and the hinge domains. In the context of IgG antibodies, the IgG isotypes each have three CH regions. Accordingly, "CH" domains in the context of IgG are as follows: "CH 1" refers to positions 118-220 according to the EU index as in Kabat. "CH2" refers to positions 237-340 according to the EU index as in Kabat, and "CH3" refers to positions 341-447 according to the EU index as in Kabat.
[110] Another type of Ig domain of the heavy chain is the hinge region. By "hinge" or "hinge region" or "antibody hinge region" or "immunoglobulin hinge region" herein is meant the flexible polypeptide comprising the amino acids between the first and second constant domains of an antibody. Structurally, the IgG CH1 domain ends at EU position 220, and the IgG CH2 domain begins at residue EU position 237. Thus for IgG the antibody hinge is herein defined to include positions 221 (D221 in IgGI ) to 236 (G236 in IgGI ), wherein the numbering is according to the EU index as in Kabat. In some embodiments, for example in the context of an Fc region, the lower hinge is included, with the "lower hinge" generally referring to positions 226 or 230.
[111] Of particular interest in the present invention are the Fc regions. By "Fc" or "Fc region", as used herein is meant the polypeptide comprising the constant region of an antibody excluding the first constant region immunoglobulin domain and in some cases, part of the hinge. Thus Fc refers to the last two constant region immunoglobulin domains of IgA, IgD, and IgG, the last three constant region immunoglobulin domains of IgE and IgM, and the flexible hinge N-terminal to these domains. For IgA and IgM, Fc may include the J chain. For IgG, as illustrated in Figure 1 , Fc comprises immunoglobulin domains Cgamma2 and Cgamma3 (Cg2 and Cg3) and the lower hinge region between Cgammai (Cg1 ) and Cgamma2 (Cg2). Although the boundaries of the Fc region may vary, the human IgG heavy chain Fc region is usually defined to include residues C226 or P230 to its carboxyl-terminus, wherein the numbering is according to the EU index as in Kabat. Fc may refer to this region in isolation, or this region in the context of an Fc polypeptide, as described below. By "Fc polypeptide" as used herein is meant a polypeptide that comprises all or part of an Fc region. Fc polypeptides include antibodies, Fc fusions, isolated Fes, and Fc fragments. [112] In some embodiments, the antibodies are full length. By "full length antibody" herein is meant the structure that constitutes the natural biological form of an antibody, including variable and constant regions, including one or more modifications as outlined herein. [113] Alternatively, the antibodies can be a variety of structures, including, but not limited to, antibody fragments, monoclonal antibodies, bispecific antibodies, minibodies, domain antibodies, synthetic antibodies (sometimes referred to herein as "antibody mimetics"), chimeric antibodies, humanized antibodies, antibody fusions (sometimes referred to as "antibody conjugates"), and fragments of each, respectively. [114] Antibody Fragments
[115] In one embodiment, the antibody is an antibody fragment. Of particular interest are antibodies that comprise Fc regions, Fc fusions, and the constant region of the heavy chain (CH1-hinge-CH2-CH3), again also including constant heavy region fusions. [116] Specific antibody fragments include, but are not limited to, (i) the Fab fragment consisting of VL, VH, CL and CH 1 domains, (ii) the Fd fragment consisting of the VH and CH1 domains, (iii) the Fv fragment consisting of the VL and VH domains of a single antibody; (iv) the dAb fragment (Ward et al., 1989, Nature 341 :544-546, entirely incorporated by reference) which consists of a single variable, (v) isolated CDR regions, (vi) F(ab')2 fragments, a bivalent fragment comprising two linked Fab fragments (vii) single chain Fv molecules (scFv), wherein a VH domain and a VL domain are linked by a peptide linker which allows the two domains to associate to form an antigen binding site (Bird et al., 1988, Science 242:423-426, Huston et al., 1988, Proc. Natl. Acad. Sci. U.S.A. 85:5879-5883, entirely incorporated by reference), (viii) bispecific single chain Fv (WO 03/11161 , hereby incorporated by reference) and (ix) "diabodies" or "triabodies", multivalent or multispecific fragments constructed by gene fusion (Tomlinson et. al., 2000, Methods Enzymol. 326:461- 479; WO94/13804; Holliger et al., 1993, Proc. Natl. Acad. Sci. U.S.A. 90:6444-6448, all entirely incorporated by reference). The antibody fragments may be modified. For example, the molecules may be stabilized by the incorporation of disulphide bridges linking the VH and VL domains (Reiter et al., 1996, Nature Biotech. 14:1239-1245, entirely incorporated by reference).
[117] Chimeric and Humanized Antibodies
[118] In some embodiments, the scaffold components can be a mixture from different species. As such, if the protein is an antibody, such antibody may be a chimeric antibody and/or a humanized antibody. In general, both "chimeric antibodies" and "humanized antibodies" refer to antibodies that combine regions from more than one species. For example, "chimeric antibodies" traditionally comprise variable region(s) from a mouse (or rat, in some cases) and the constant region(s) from a human. "Humanized antibodies" generally refer to non-human antibodies that have had the variable-domain framework regions swapped for sequences found in human antibodies. Generally, in a humanized antibody, the entire antibody, except the CDRs, is encoded by a polynucleotide of human origin or is identical to such an antibody except within its CDRs. The CDRs, some or all of which are encoded by nucleic acids originating in a non-human organism, are grafted into the beta- sheet framework of a human antibody variable region to create an antibody, the specificity of which is determined by the engrafted CDRs. The creation of such antibodies is described in, e.g., WO 92/1 1018, Jones, 1986, Nature 321 :522-525, Verhoeyen et al., 1988, Science 239:1534-1536, all entirely incorporated by reference. "Backmutation" of selected acceptor framework residues to the corresponding donor residues is often required to regain affinity that is lost in the initial grafted construct (US 5530101 ; US 5585089; US 5693761 ; US 5693762; US 6180370; US 5859205; US 5821337; US 6054297; US 6407213, all entirely incorporated by reference). The humanized antibody optimally also will comprise at least a portion of an immunoglobulin constant region, typically that of a human immunoglobulin, and thus will typically comprise a human Fc region. Humanized antibodies can also be generated using mice with a genetically engineered immune system. Roque et al., 2004, Biotechnol. Prog. 20:639-654, entirely incorporated by reference. A variety of techniques and methods for humanizing and reshaping non-human antibodies are well known in the art (See Tsurushita & Vasquez, 2004, Humanization of Monoclonal Antibodies, Molecular Biology of B Cells, 533-545, Elsevier Science (USA), and references cited therein, all entirely incorporated by reference). Humanization methods include but are not limited to methods described in Jones et al., 1986, Nature 321 :522-525; Riechmann et a/., 1988; Nature 332:323-329; Verhoeyen et al., 1988, Science, 239:1534-1536; Queen et al., 1989, Proc Natl Acad Sci, USA 86:10029-33; He et al., 1998, J. Immunol. 160: 1029-1035; Carter et al., 1992, Proc Natl Acad Sci USA 89:4285-9, Presta et al., 1997, Cancer Res.57(20):4593-9; Gorman et al., 1991 , Proc. Natl. Acad. Sci. USA 88:4181-4185; O'Connor et al., 1998, Protein Eng 1 1 :321-8, all entirely incorporated by reference. Humanization or other methods of reducing the immunogenicity of nonhuman antibody variable regions may include resurfacing methods, as described for example in Roguska et al., 1994, Proc. Natl. Acad. Sci. USA 91 :969-973, entirely incorporated by reference. In one embodiment, the parent antibody has been affinity matured, as is known in the art. Structure-based methods may be employed for humanization and affinity maturation, for example as described in USSN 11/004,590. Selection based methods may be employed to humanize and/or affinity mature antibody variable regions, including but not limited to methods described in Wu et al., 1999, J. MoI. Biol. 294:151-162; Baca et al., 1997, J. Biol. Chem. 272(16):10678-10684; Rosok et al., 1996, J. Biol. Chem. 271 (37): 2261 1-22618; Rader et al., 1998, Proc. Natl. Acad. Sci. USA 95: 8910-8915; Krauss et al., 2003, Protein Engineering 16(10):753-759, all entirely incorporated by reference. Other humanization methods may involve the grafting of only parts of the CDRs, including but not limited to methods described in USSN 09/810,510; Tan et al., 2002, J. Immunol. 169:1 119-1 125; De Pascalis et al., 2002, J. Immunol. 169:3076- 3084, all entirely incorporated by reference. [119] Bispecific Antibodies
[120] In one embodiment, the antibodies of the invention multispecific antibody, and notably a bispecific antibody, also sometimes referred to as "diabodies". These are antibodies that bind to two (or more) different antigens. Diabodies can be manufactured in a variety of ways known in the art (Holliger and Winter, 1993, Current Opinion Biotechnol. 4:446-449, entirely incorporated by reference), e.g., prepared chemically or from hybrid hybridomas. [121] Minibodies
[122] In one embodiment, the antibody is a minibody. Minibodies are minimized antibody- like proteins comprising a scFv joined to a CH3 domain. Hu et al., 1996, Cancer Res. 56:3055-3061 , entirely incorporated by reference. In some cases, the scFv can be joined to the Fc region, and may include some or the entire hinge region. [123] Human Antibodies
[124] In one embodiment, the antibody is a fully human antibody with at least one modification as outlined herein. "Fully human antibody" or "complete human antibody" refers to a human antibody having the gene sequence of an antibody derived from a human chromosome with the modifications outlined herein. [125] Antibody Fusions
[126] In one embodiment, the antibodies of the invention are antibody fusion proteins (sometimes referred to herein as an "antibody conjugate"). One type of antibody fusions comprises Fc fusions, which join the Fc region with a conjugate partner. By "Fc fusion" as used herein is meant a protein wherein one or more polypeptides is operably linked to an Fc region. Fc fusion is herein meant to be synonymous with the terms "immunoadhesin", "Ig fusion", "Ig chimera", and "receptor globulin" (sometimes with dashes) as used in the prior art (Chamow et al., 1996, Trends Biotechnol 14:52-60; Ashkenazi et al., 1997, Curr Opin Immunol 9:195-200, both entirely incorporated by reference). An Fc fusion combines the Fc region of an immunoglobulin with a fusion partner, which in general can be any protein or small molecule. Virtually any protein or small molecule may be linked to Fc to generate an Fc fusion. Protein fusion partners may include, but are not limited to, the variable region of any antibody, the target-binding region of a receptor, an adhesion molecule, a ligand, an enzyme, a cytokine, a chemokine, or some other protein or protein domain. Small molecule fusion partners may include any therapeutic agent that directs the Fc fusion to a therapeutic target. Such targets may be any molecule, preferably an extracellular receptor, which is implicated in disease. Thus, the IgG variants can be linked to one or more fusion partners. In one alternate embodiment, the IgG variant is conjugated or operably linked to another therapeutic compound. The therapeutic compound may be a cytotoxic agent, a chemotherapeutic agent, a toxin, a radioisotope, a cytokine, or other therapeutically active agent. The IgG may be linked to one of a variety of nonproteinaceous polymers, e.g., polyethylene glycol, polypropylene glycol, polyoxyalkylenes, or copolymers of polyethylene glycol and polypropylene glycol.
[127] In addition to Fc fusions, antibody fusions include the fusion of the constant region of the heavy chain with one or more fusion partners (again including the variable region of any antibody), while other antibody fusions are substantially or completely full length antibodies with fusion partners. In one embodiment, a role of the fusion partner is to mediate target binding, and thus it is functionally analogous to the variable regions of an antibody (and in fact can be). Virtually any protein or small molecule may be linked to Fc to generate an Fc fusion (or antibody fusion). Protein fusion partners may include, but are not limited to, the target-binding region of a receptor, an adhesion molecule, a ligand, an enzyme, a cytokine, a chemokine, or some other protein or protein domain. Small molecule fusion partners may include any therapeutic agent that directs the Fc fusion to a therapeutic target. Such targets may be any molecule, preferably an extracellular receptor, which is implicated in disease. [128] The conjugate partner can be proteinaceous or non-proteinaceous; the latter generally being generated using functional groups on the antibody and on the conjugate partner. For example linkers are known in the art; for example, homo-or hetero-bifunctional linkers as are well known (see, 1994 Pierce Chemical Company catalog, technical section on cross-linkers, pages 155-200, incorporated herein by reference). [129] Suitable conjugates include, but are not limited to, labels as described below, drugs and cytotoxic agents including, but not limited to, cytotoxic drugs (e.g., chemotherapeutic agents) or toxins or active fragments of such toxins. Suitable toxins and their corresponding fragments include diptheria A chain, exotoxin A chain, ricin A chain, abrin A chain, curcin, crotin, phenomycin, enomycin and the like. Cytotoxic agents also include radiochemicals made by conjugating radioisotopes to antibodies, or binding of a radionuclide to a chelating agent that has been covalently attached to the antibody. Additional embodiments utilize calicheamicin, auristatins, geldanamycin, maytansine, and duocarmycins and analogs; for the latter, see U.S. 2003/0050331 A1 , entirely incorporated by reference. [130] Covalent modifications of Antibodies
[131] Covalent modifications of antibodies are included within the scope of this invention, and are generally, but not always, done post-translationally. For example, several types of covalent modifications of the antibody are introduced into the molecule by reacting specific amino acid residues of the antibody with an organic derivatizing agent that is capable of reacting with selected side chains or the N- or C-terminal residues. [132] Cysteinyl residues most commonly are reacted with α-haloacetates (and corresponding amines), such as chloroacetic acid or chloroacetamide, to give carboxymethyl or carboxyamidomethyl derivatives. Cysteinyl residues may also be derivatized by reaction with bromotrifluoroacetone, α-bromo-β-(5-imidozoyl)propionic acid, chloroacetyl phosphate, N-alkylmaleimides, 3-nitro-2-pyridyl disulfide, methyl 2-pyridyl disulfide, p- chloromercuribenzoate, 2-chloromercuri-4-nitrophenol, or chloro-7-nitrobenzo-2-oxa-1 ,3- diazole and the like. [133] Histidyl residues are derivatized by reaction with diethylpyrocarbonate at pH 5.5-7.0 because this agent is relatively specific for the histidyl side chain. Para-bromophenacyl bromide also is useful; the reaction is preferably performed in 0.1 M sodium cacodylate at pH 6.0.
[134] Lysinyl and amino terminal residues are reacted with succinic or other carboxylic acid anhydrides. Derivatization with these agents has the effect of reversing the charge of the lysinyl residues. Other suitable reagents for derivatizing alpha-amino-containing residues include imidoesters such as methyl picolinimidate; pyridoxal phosphate; pyridoxal; chloroborohydride; trinitrobenzenesulfonic acid; O-methylisourea; 2,4-pentanedione; and transaminase-catalyzed reaction with glyoxylate.
[135] Arginyl residues are modified by reaction with one or several conventional reagents, among them phenylglyoxal, 2,3-butanedione, 1 ,2-cyclohexanedione, and ninhydrin. Derivatization of arginine residues requires that the reaction be performed in alkaline conditions because of the high pKa of the guanidine functional group. Furthermore, these reagents may react with the groups of lysine as well as the arginine epsilon-amino group. [136] The specific modification of tyrosyl residues may be made, with particular interest in introducing spectral labels into tyrosyl residues by reaction with aromatic diazonium compounds or tetranitromethane. Most commonly, N-acetylimidizole and tetranitromethane are used to form O-acetyl tyrosyl species and 3-nitro derivatives, respectively. Tyrosyl residues are iodinated using 1251 or 1311 to prepare labeled proteins for use in radioimmunoassay, the chloramine T method described above being suitable. [137] Carboxyl side groups (aspartyl or glutamyl) are selectively modified by reaction with carbodiimides (R' — N=C=N-R'), where R and R' are optionally different alkyl groups, such as 1-cyclohexyl-3-(2-morpholinyl-4-ethyl) carbodiimide or 1-ethyl-3-(4-azonia-4,4- dimethylpentyl) carbodiimide. Furthermore, aspartyl and glutamyl residues are converted to asparaginyl and glutaminyl residues by reaction with ammonium ions. [138] Derivatization with bifunctional agents is useful for crosslinking antibodies to a water- insoluble support matrix or surface for use in a variety of methods, in addition to methods described below. Commonly used crosslinking agents include, e.g., 1 ,1-bis(diazoacetyl)-2- phenylethane, glutaraldehyde, N-hydroxysuccinimide esters, for example, esters with 4- azidosalicylic acid, homobifunctional imidoesters, including disuccinimidyl esters such as 3,3'-dithiobis (succinimidylpropionate), and bifunctional maleimides such as bis-N- maleimido-1 ,8-octane. Derivatizing agents such as methyl-3-[(p- azidophenyl)dithio]propioimidate yield photoactivatable intermediates that are capable of forming crosslinks in the presence of light. Alternatively, reactive water-insoluble matrices such as cyanogen bromide-activated carbohydrates and the reactive substrates described in U.S. Pat. Nos. 3,969,287; 3,691 ,016; 4,195,128; 4,247,642; 4,229,537; and 4,330,440, all entirely incorporated by reference, are employed for protein immobilization. [139] Glutaminyl and asparaginyl residues are frequently deamidated to the corresponding glutamyl and aspartyl residues, respectively. Alternatively, these residues are deamidated under mildly acidic conditions. Either form of these residues falls within the scope of this invention.
Other modifications include hydroxylation of proline and lysine, phosphorylation of hydroxyl groups of seryl or threonyl residues, methylation of the α-amino groups of lysine, arginine, and histidine side chains (T. E. Creighton, Proteins: Structure and Molecular Properties, W. H. Freeman & Co., San Francisco, pp. 79-86 [1983], entirely incorporated by reference), acetylation of the N-terminal amine, and amidation of any C-terminal carboxyl group. [140] Glycosylation
[141] Another type of covalent modification is glycosylation. In another embodiment, the IgG variants disclosed herein can be modified to include one or more engineered glycoforms. By "engineered glycoform" as used herein is meant a carbohydrate composition that is covalently attached to an IgG, wherein said carbohydrate composition differs chemically from that of a parent IgG. Engineered glycoforms may be useful for a variety of purposes, including but not limited to enhancing or reducing effector function. Engineered glycoforms may be generated by a variety of methods known in the art (Umaήa et al., 1999, Nat Biotechnol 17:176-180; Davies et al., 2001 , Biotechnol Bioeng 74:288-294; Shields et al., 2002, J Biol Chem 277:26733-26740; Shinkawa et al., 2003, J Biol Chem 278:3466- 3473; US 6,602,684; USSN 10/277,370; USSN 10/113,929; PCT WO 00/61739A1 ; PCT WO 01/29246A1 ; PCT WO 02/31140A1 ; PCT WO 02/30954A1 , all entirely incorporated by reference; (Potelligent® technology [Biowa, Inc., Princeton, NJ]; GlycoMAb® glycosylation engineering technology [Glycart Biotechnology AG, Zurich, Switzerland]). Many of these techniques are based on controlling the level of fucosylated and/or bisecting oligosaccharides that are covalently attached to the Fc region, for example by expressing an IgG in various organisms or cell lines, engineered or otherwise (for example Lec-13 CHO cells or rat hybridoma YB2/0 cells), by regulating enzymes involved in the glycosylation pathway (for example FUT8 [α1 ,6-fucosyltranserase] and/or β1-4- N- acetylglucosaminyltransferase III [GnTIII]), or by modifying carbohydrate(s) after the IgG has been expressed. Engineered glycoform typically refers to the different carbohydrate or oligosaccharide; thus an IgG variant, for example an antibody or Fc fusion, can include an engineered glycoform. Alternatively, engineered glycoform may refer to the IgG variant that comprises the different carbohydrate or oligosaccharide. As is known in the art, glycosylation patterns can depend on both the sequence of the protein (e.g., the presence or absence of particular glycosylation amino acid residues, discussed below), or the host cell or organism in which the protein is produced. Particular expression systems are discussed below. [142] 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 tri-peptide 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 tri- peptide sequences in a polypeptide creates a potential glycosylation site. O-linked glycosylation refers to the attachment of one of the sugars N-acetylgalactosamine, galactose, or xylose, to a hydroxyamino acid, most commonly serine or threonine, although 5-hydroxyproline or 5-hydroxylysine may also be used.
[143] Addition of glycosylation sites to the antibody is conveniently accomplished by altering the amino acid sequence such that it contains one or more of the above-described tri-peptide 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 starting sequence (for O-linked glycosylation sites). For ease, the antibody amino acid sequence is preferably altered through changes at the DNA level, particularly by mutating the DNA encoding the target polypeptide at preselected bases such that codons are generated that will translate into the desired amino acids.
[144] Another means of increasing the number of carbohydrate moieties on the antibody is by chemical or enzymatic coupling of glycosides to the protein. These procedures are advantageous in that they do not require production of the protein in a host cell that has glycosylation capabilities for N- and O-linked glycosylation. Depending on the coupling mode used, the sugar(s) may be attached to (a) arginine and histidine, (b) free carboxyl groups, (c) free sulfhydryl groups such as those of cysteine, (d) free hydroxyl groups such as those of serine, threonine, or hydroxyproline, (e) aromatic residues such as those of phenylalanine, tyrosine, or tryptophan, or (f) the amide group of glutamine. These methods are described in WO 87/05330 and in ApNn and Wriston, 1981 , CRC Crit. Rev. Biochem., pp. 259-306, both entirely incorporated by reference.
[145] Removal of carbohydrate moieties present on the starting antibody may be accomplished chemically or enzymatically. Chemical deglycosylation requires exposure of the protein to the compound trifluoromethanesulfonic acid, or an equivalent compound. This treatment results in the cleavage of most or all sugars except the linking sugar (N- acetylglucosamine or N-acetylgalactosamine), while leaving the polypeptide intact. Chemical deglycosylation is described by Hakimuddin et al., 1987, Arch. Biochem. Biophys. 259:52 and by Edge et al., 1981 , Anal. Biochem. 1 18:131 , both entirely incorporated by reference. Enzymatic cleavage of carbohydrate moieties on polypeptides can be achieved by the use of a variety of endo- and exo-glycosidases as described by Thotakura et al., 1987, Meth. Enzymol. 138:350, entirely incorporated by reference. Glycosylation at potential glycosylation sites may be prevented by the use of the compound tunicamycin as described by Duskin et al., 1982, J. Biol. Chem. 257:3105, entirely incorporated by reference. Tunicamycin blocks the formation of protein-N-glycoside linkages.
[146] Another type of covalent modification of the antibody comprises linking the antibody to various nonproteinaceous polymers, including, but not limited to, various polyols such as polyethylene glycol, polypropylene glycol or polyoxyalkylenes, in the manner set forth in, for example, 2005-2006 PEG Catalog from Nektar Therapeutics (available at the Nektar website) US Patents 4,640,835; 4,496,689; 4,301 ,144; 4,670,417; 4,791 ,192 or 4,179,337, all entirely incorporated by reference. In addition, as is known in the art, amino acid substitutions may be made in various positions within the antibody to facilitate the addition of polymers such as PEG. See for example, U.S. Publication No. 2005/0114037A1 , entirely incorporated by reference. [147] Labeled Antibodies
[148] In some embodiments, the covalent modification of the antibodies of the invention comprises the addition of one or more labels. In some cases, these are considered antibody fusions. The term "labelling group" means any detectable label. In some embodiments, the labelling group is coupled to the antibody via spacer arms of various lengths to reduce potential steric hindrance. Various methods for labelling proteins are known in the art and may be used in performing the present invention.
[149] In general, labels fall into a variety of classes, depending on the assay in which they are to be detected: a) isotopic labels, which may be radioactive or heavy isotopes; b) magnetic labels (e.g., magnetic particles); c) redox active moieties; d) optical dyes; enzymatic groups (e.g. horseradish peroxidase, β-galactosidase, luciferase, alkaline phosphatase); e) biotinylated groups; and f) predetermined polypeptide epitopes recognized by a secondary reporter (e.g., leucine zipper pair sequences, binding sites for secondary antibodies, metal binding domains, epitope tags, etc.). In some embodiments, the labelling group is coupled to the antibody via spacer arms of various lengths to reduce potential steric hindrance. Various methods for labelling proteins are known in the art and may be used in performing the present invention.
[150] Specific labels include optical dyes, including, but not limited to, chromophores, phosphors and fluorophores, with the latter being specific in many instances. Fluorophores can be either "small molecule" fluores, or proteinaceous fluores.
[151] By "fluorescent label" is meant any molecule that may be detected via its inherent fluorescent properties. Suitable fluorescent labels include, but are not limited to, fluorescein, rhodamine, tetramethylrhodamine, eosin, erythrosin, coumarin, methyl-coumarins, pyrene, Malacite green, stilbene, Lucifer Yellow, Cascade BlueJ, Texas Red, IAEDANS, EDANS, BODIPY FL, LC Red 640, Cy 5, Cy 5.5, LC Red 705, Oregon green, the Alexa-Fluor dyes (Alexa Fluor 350, Alexa Fluor 430, Alexa Fluor 488, Alexa Fluor 546, Alexa Fluor 568, Alexa Fluor 594, Alexa Fluor 633, Alexa Fluor 660, Alexa Fluor 680), Cascade Blue, Cascade Yellow and R-phycoerythrin (PE) (Molecular Probes, Eugene, OR), FITC, Rhodamine, and Texas Red (Pierce, Rockford, IL), Cy5, Cy5.5, Cy7 (Amersham Life Science, Pittsburgh, PA). Suitable optical dyes, including fluorophores, are described in Molecular Probes Handbook by Richard P. Haugland, entirely incorporated by reference. [152] Suitable proteinaceous fluorescent labels also include, but are not limited to, green fluorescent protein, including a Renilla, Ptilosarcus, or Aequorea species of GFP (Chalfie et al., 1994, Science 263:802-805), EGFP (Clontech Laboratories, Inc., Genbank Accession Number U55762), blue fluorescent protein (BFP, Quantum Biotechnologies, Inc. 1801 de Maisonneuve Blvd. West, 8th Floor, Montreal, Quebec, Canada H3H 1J9; Stauber, 1998, Biotechniques 24:462-471 ; Heim et al., 1996, Curr. Biol. 6:178-182), enhanced yellow fluorescent protein (EYFP, Clontech Laboratories, Inc.), luciferase (lchiki et al., 1993, J. Immunol. 150:5408-5417), β galactosidase (Nolan et al., 1988, Proc. Natl. Acad. Sci. U.S.A. 85:2603-2607) and Renilla (WO92/15673, WO95/07463, WO98/14605, WO98/26277, WO99/49019, U.S. Patent Nos. 5292658, 5418155, 5683888, 5741668, 5777079, 5804387, 5874304, 5876995, 5925558). All of the above-cited references in this paragraph are expressly incorporated herein by reference. [153] IgG Variants
[154] In one embodiment, the invention provides variant IgG proteins. At a minimum, IgG variants comprise an antibody fragment comprising the CH2-CH3 region of the heavy chain. In addition, suitable IgG variants comprise Fc domains (e.g. including the lower hinge region), as well as IgG variants comprising the constant region of the heavy chain (CH 1- hinge-CH2-CH3) also being useful in the present invention, all of which can be fused to fusion partners.
[155] An IgG variant includes one or more amino acid modifications relative to a parent IgG polypeptide, in some cases relative to the wild type IgG. The IgG variant can have one or more optimized properties. An IgG variant differs in amino acid sequence from its parent IgG by virtue of at least one amino acid modification. Thus IgG variants have at least one amino acid modification compared to the parent. Alternatively, the IgG variants may have more than one amino acid modification as compared to the parent, for example from about one to fifty amino acid modifications, preferably from about one to ten amino acid modifications, and most preferably from about one to about five amino acid modifications compared to the parent. [156] Thus the sequences of the IgG variants and those of the parent Fc polypeptide are substantially homologous. For example, the variant IgG variant sequences herein will possess about 80% homology with the parent IgG variant sequence, preferably at least about 90% homology, and most preferably at least about 95% homology. Modifications may be made genetically using molecular biology, or may be made enzymatically or chemically. [157] Target Antigens for Antibodies
[158] Virtually any antigen may be targeted by the IgG variants, including but not limited to proteins, subunits, domains, motifs, and/or epitopes belonging to the following list of target antigens, which includes both soluble factors such as cytokines and membrane-bound factors, including transmembrane receptors: 17-IA, 4-1 BB, 4Dc, 6-keto-PGF1 a, 8-iso- PGF2a, 8-oxo-dG, A1 Adenosine Receptor, A33, ACE, ACE-2, Activin, Activin A, Activin AB, Activin B, Activin C, Activin RIA, Activin RIA ALK-2, Activin RIB ALK-4, Activin RIIA, Activin RIIB, ADAM, ADAM10, ADAM12, ADAM15, ADAM17/TACE, ADAM8, ADAM9, ADAMTS, ADAMTS4, ADAMTS5, Addressins, aFGF, ALCAM, ALK, ALK-1 , ALK-7, alpha-1-antitrypsin, alpha-V/beta-1 antagonist, ANG, Ang, APAF-1 , APE, APJ, APP, APRIL, AR, ARC, ART, Artemin, anti-Id, ASPARTIC, Atrial natriuretic factor, av/b3 integrin, AxI, b2M, B7-1 , B7-2, B7-H, B-lymphocyte Stimulator (BIyS), BACE, BACE-1 , Bad, BAFF, BAFF-R, Bag-1 , BAK, Bax, BCA-1 , BCAM, BcI, BCMA, BDNF, b-ECGF, bFGF, BID, Bik, BIM, BLC, BL-CAM, BLK, BMP, BMP-2 BMP-2a, BMP-3 Osteogenin, BMP-4 BMP-2b, BMP-5, BMP-6 Vgr-1 , BMP-7 (OP-1 ), BMP-8 (BMP-8a, OP-2), BMPR, BMPR-IA (ALK-3), BMPR-IB (ALK-6), BRK-2, RPK- 1 , BMPR-II (BRK-3), BMPs, b-NGF, BOK, Bombesin, Bone-derived neurotrophic factor, BPDE, BPDE-DNA, BTC, complement factor 3 (C3), C3a, C4, C5, C5a, C10, CA125, CAD- 8, Calcitonin, cAMP, carcinoembryonic antigen (CEA), carcinoma-associated antigen, Cathepsin A, Cathepsin B, Cathepsin C/DPPI, Cathepsin D, Cathepsin E, Cathepsin H, Cathepsin L, Cathepsin O, Cathepsin S, Cathepsin V, Cathepsin X/Z/P, CBL, CCI, CCK2, CCL, CCL1 , CCL1 1 , CCL12, CCL13, CCL14, CCL15, CCL16, CCL17, CCL18, CCL19, CCL2, CCL20, CCL21 , CCL22, CCL23, CCL24, CCL25, CCL26, CCL27, CCL28, CCL3, CCL4, CCL5, CCL6, CCL7, CCL8, CCL9/10, CCR, CCR1 , CCR10, CCR10, CCR2, CCR3, CCR4, CCR5, CCR6, CCR7, CCR8, CCR9, CD1 , CD2, CD3, CD3E, CD4, CD5, CD6, CD7, CD8, CD10, CD11 a, CD11 b, CD11 c, CD13, CD14, CD15, CD16, CD18, CD19, CD20, CD21 , CD22, CD23, CD25, CD27L, CD28, CD29, CD30, CD30L, CD32, CD33 (p67 proteins), CD34, CD38, CD40, CD40L, CD44, CD45, CD46, CD49a, CD52, CD54, CD55, CD56, CD61 , CD64, CD66e, CD74, CD80 (B7-1 ), CD89, CD95, CD123, CD137, CD138, CD140a, CD146, CD147, CD148, CD152, CD164, CEACAM5, CFTR, cGMP, CINC, Clostridium botulinum toxin, Clostridium perfringens toxin, CKb8-1 , CLC, CMV, CMV UL, CNTF, CNTN-1 , COX, C-Ret, CRG-2, CT-1 , CTACK, CTGF, CTLA-4, CX3CL1 , CX3CR1 , CXCL, CXCL1 , CXCL2, CXCL3, CXCL4, CXCL5, CXCL6, CXCL7, CXCL8, CXCL9, CXCL10, CXCL11 , CXCL12, CXCL13, CXCL14, CXCL15, CXCL16, CXCR, CXCR1 , CXCR2, CXCR3, CXCR4, CXCR5, CXCR6, cytokeratin tumor-associated antigen, DAN, DCC, DcR3, DC-SIGN, Decay accelerating factor, des(1-3)-IGF-l (brain IGF-1 ), Dhh, digoxin, DNAM-1 , Dnase, Dpp, DPPIV/CD26, Dtk, ECAD, EDA, EDA-A1 , EDA-A2, EDAR, EGF, EGFR (ErbB-1 ), EMA, EMMPRIN, ENA, endothelin receptor, Enkephalinase, eNOS, Eot, eotaxini , EpCAM, Ephrin B2/ EphB4, EPO, ERCC, E-selectin, ET-1 , Factor Na, Factor VII, Factor VIIIc, Factor IX, fibroblast activation protein (FAP), Fas, FcR1 , FEN-1 , Ferritin, FGF, FGF-19, FGF-2, FGF3, FGF-8, FGFR, FGFR-3, Fibrin, FL, FLIP, Flt-3, Flt-4, Follicle stimulating hormone, Fractalkine, FZD1 , FZD2, FZD3, FZD4, FZD5, FZD6, FZD7, FZD8, FZD9, FZD10, G250, Gas 6, GCP-2, GCSF, GD2, GD3, GDF, GDF-1 , GDF-3 (Vgr-2), GDF- 5 (BMP-14, CDMP-1 ), GDF-6 (BMP-13, CDMP-2), GDF-7 (BMP-12, CDMP-3), GDF-8 (Myostatin), GDF-9, GDF-15 (MIC-1 ), GDNF, GDNF, GFAP, GFRa-1 , GFR-alpha1 , GFR- alpha2, GFR-alpha3, GITR, Glucagon, Glut 4, glycoprotein llb/llla (GP llb/llla), GM-CSF, gp130, gp72, GRO, Growth hormone releasing factor, Hapten (NP-cap or NIP-cap), HB- EGF, HCC, HCMV gB envelope glycoprotein, HCMV) gH envelope glycoprotein, HCMV UL, Hemopoietic growth factor (HGF), Hep B gp120, heparanase, Her2, Her2/neu (ErbB-2), Her3 (ErbB-3), Her4 (ErbB-4), herpes simplex virus (HSV) gB glycoprotein, HSV gD glycoprotein, HGFA, High molecular weight melanoma-associated antigen (HMW-MAA), HIV gp120, HIV MIB gp 120 V3 loop, HLA, HLA-DR, HM1.24, HMFG PEM, HRG, Hrk, human cardiac myosin, human cytomegalovirus (HCMV), human growth hormone (HGH), HVEM, I- 309, IAP, ICAM, ICAM-1 , ICAM-3, ICE, ICOS, IFNg, Ig, IgA receptor, IgE, IGF, IGF binding proteins, IGF-1 R, IGFBP, IGF-I, IGF-II, IL, IL-1 , IL-1 R, IL-2, IL-2R, IL-4, IL-4R, IL-5, IL-5R, IL-6, IL-6R, IL-8, IL-9, IL-10, IL-12, IL-13, IL-15, IL-18, IL-18R, IL-23, interferon (INF)-alpha, INF-beta, INF-gamma, Inhibin, iNOS, Insulin A-chain, Insulin B-chain, Insulin-like growth factor 1 , integrin alpha2, integrin alpha3, integrin alpha4, integrin alpha4/beta1 , integrin alpha4/beta7, integrin alphaδ (alphaV), integrin alpha5/beta1 , integrin alpha5/beta3, integrin alphaθ, integrin betai , integrin beta2, interferon gamma, IP-10, I-TAC, JE, Kallikrein 2, Kallikrein 5, Kallikrein 6, , Kallikrein 11 , Kallikrein 12, Kallikrein 14, Kallikrein 15, Kallikrein L1 , Kallikrein L2, Kallikrein L3, Kallikrein L4, KC, KDR, Keratinocyte Growth Factor (KGF), laminin 5, LAMP, LAP, LAP (TGF- 1 ), Latent TGF-1 , Latent TGF- 1 bp1 , LBP, LDGF, LECT2, Lefty, Lewis-Y antigen, Lewis-Y related antigen, LFA-1 , LFA-3, Lfo, LIF, LIGHT, lipoproteins, LIX, LKN, Lptn, L-Selectin, LT-a, LT-b, LTB4, LTBP-1 , Lung surfactant, Luteinizing hormone, Lymphotoxin Beta Receptor, Mac-1 , MAdCAM, MAG, MAP2, MARC, MCAM, MCAM, MCK- 2, MCP, M-CSF, MDC, Mer, METALLOPROTEASES, MGDF receptor, MGMT, MHC (HLA- DR), MIF, MIG, MIP, MIP-1-alpha, MK, MMAC1 , MMP, MMP-1 , MMP-10, MMP-1 1 , MMP-12, MMP-13, MMP-14, MMP-15, MMP-2, MMP-24, MMP-3, MMP-7, MMP-8, MMP-9, MPIF, Mpo, MSK, MSP, mucin (Mud ), MUC18, Muellerian-inhibitin substance, Mug, MuSK, NAIP, NAP, NCAD, N-Cadherin, NCA 90, NCAM, NCAM, Neprilysin, Neurotrophin-3,-4, or -6, Neurturin, Neuronal growth factor (NGF), NGFR, NGF-beta, nNOS, NO, NOS, Npn, NRG-3, NT, NTN, OB, OGG1 , OPG, OPN, OSM, OX40L, OX40R, p150, p95, PADPr, Parathyroid hormone, PARC, PARP, PBR, PBSF, PCAD, P-Cadherin, PCNA, PDGF, PDGF, PDK-1 , PECAM, PEM, PF4, PGE, PGF, PGI2, PGJ2, PIN, PLA2, placental alkaline phosphatase (PLAP), PIGF, PLP, PP14, Proinsulin, Prorelaxin, Protein C, PS, PSA, PSCA, prostate specific membrane antigen (PSMA), PTEN, PTHrp, Ptk, PTN, R51 , RANK, RANKL, RANTES, RANTES, Relaxin A-chain, Relaxin B-chain, renin, respiratory syncytial virus (RSV) F, RSV Fgp, Ret, Rheumatoid factors, RLI P76, RPA2, RSK, S100, SCF/KL, SDF-1 , SERINE, Serum albumin, sFRP-3, Shh, SIGIRR, SK-1 , SLAM, SLPI, SMAC, SMDF, SMOH, SOD, SPARC, Stat, STEAP, STEAP-II, TACE, TACI, TAG-72 (tumor-associated glycoprotein-72), TARC, TCA-3, T-cell receptors (e.g., T-cell receptor alpha/beta), TdT, TECK, TEM1 , TEM5, TEM7, TEM8, TERT, testicular PLAP-like alkaline phosphatase, TfR, TGF, TGF-alpha, TGF-beta, TGF-beta Pan Specific, TGF-beta Rl (ALK-5), TGF-beta RII, TGF-beta RIIb, TGF-beta RIII, TGF-beta1 , TGF-beta2, TGF-beta3, TGF-beta4, TGF-beta5, Thrombin, Thymus Ck-1 , Thyroid stimulating hormone, Tie, TIMP, TIQ, Tissue Factor, TMEFF2, Tmpo, TMPRSS2, TNF, TNF-alpha, TNF-alpha beta, TNF-beta2, TNFc, TNF-RI, TNF-RII, TNFRSF10A (TRAIL R1 Apo-2, DR4), TNFRSF10B (TRAIL R2 DR5, KILLER, TRICK-2A, TRICK-B), TNFRSF10C (TRAIL R3 DcR1 , LIT, TRID), TNFRSF10D (TRAIL R4 DcR2, TRUNDD), TNFRSF11 A (RANK ODF R, TRANCE R), TNFRSF1 1 B (OPG OCIF, TR1 ), TNFRSF12 (TWEAK R FN14), TNFRSF13B (TACI), TNFRSF13C (BAFF R), TNFRSF14 (HVEM ATAR, HveA, LIGHT R, TR2), TNFRSF16 (NGFR p75NTR), TNFRSF17 (BCMA), TNFRSF18 (GITR AITR), TNFRSF19 (TROY TAJ, TRADE), TNFRSF19L (RELT), TNFRSF1A (TNF Rl CD120a, p55-60), TNFRSF1 B (TNF RII CD120b, p75-80), TNFRSF26 (TNFRH3), TNFRSF3 (LTbR TNF RIII, TNFC R), TNFRSF4 (OX40 ACT35, TXGP1 R), TNFRSF5 (CD40 p50), TNFRSF6 (Fas Apo-1 , APT1 , CD95), TNFRSF6B (DcR3 M68, TR6), TNFRSF7 (CD27), TNFRSF8 (CD30), TNFRSF9 (4-1 BB CD137, ILA), TNFRSF21 (DR6), TNFRSF22 (DcTRAIL R2 TNFRH2), TNFRST23 (DcTRAIL R1 TNFRH1 ), TNFRSF25 (DR3 Apo-3, LARD, TR-3, TRAMP, WSL-1 ), TNFSF10 (TRAIL Apo-2 Ligand, TL2), TNFSF11 (TRANCE/RANK Ligand ODF, OPG Ligand), TNFSF12 (TWEAK Apo-3 Ligand, DR3 Ligand), TNFSF13 (APRIL TALL2), TNFSF13B (BAFF BLYS, TALL1 , THANK, TNFSF20), TNFSF14 (LIGHT HVEM Ligand, LTg), TNFSF15 (TL1A/VEGI), TNFSF18 (GITR Ligand AITR Ligand, TL6), TNFSF1A (TNF-a Conectin, DIF, TNFSF2), TNFSF1 B (TNF-b LTa, TNFSF1 ), TNFSF3 (LTb TNFC, p33), TNFSF4 (OX40 Ligand gp34, TXGP1 ), TNFSF5 (CD40 Ligand CD154, gp39, HIGM1 , IMD3, TRAP), TNFSF6 (Fas Ligand Apo-1 Ligand, APT1 Ligand), TNFSF7 (CD27 Ligand CD70), TNFSF8 (CD30 Ligand CD153), TNFSF9 (4- 1 BB Ligand CD137 Ligand), TP-1 , t-PA, Tpo, TRAIL, TRAIL R, TRAIL-R1 , TRAIL-R2, TRANCE, transferring receptor, TRF, Trk, TROP-2, TSG, TSLP, tumor-associated antigen CA 125, tumor-associated antigen expressing Lewis Y related carbohydrate, TWEAK, TXB2, Ung, uPAR, uPAR-1 , Urokinase, VCAM, VCAM-1 , VECAD, VE-Cadherin, VE-cadherin-2, VEFGR-1 (flt-1 ), VEGF, VEGFR, VEGFR-3 (flt-4), VEGI, VIM, Viral antigens, VLA, VLA-1 , VLA-4, VNR integrin, von Willebrands factor, WIF-1 , WNT1 , WNT2, WNT2B/13, WNT3, WNT3A, WNT4, WNT5A, WNT5B, WNT6, WNT7A, WNT7B, WNT8A, WNT8B, WNT9A, WNT9A, WNT9B, WNT10A, WNT10B, WNT11 , WNT16, XCL1 , XCL2, XCR1 , XCR1 , XEDAR, XIAP, XPD, and receptors for hormones and growth factors. [159] One skilled in the art will appreciate that the aforementioned list of targets refers not only to specific proteins and biomolecules, but the biochemical pathway or pathways that comprise them. For example, reference to CTLA-4 as a target antigen implies that the ligands and receptors that make up the T cell co-stimulatory pathway, including CTLA-4, B7- 1 , B7-2, CD28, and any other undiscovered ligands or receptors that bind these proteins, are also targets. Thus target as used herein refers not only to a specific biomolecule, but the set of proteins that interact with said target and the members of the biochemical pathway to which said target belongs. One skilled in the art will further appreciate that any of the aforementioned target antigens, the ligands or receptors that bind them, or other members of their corresponding biochemical pathway, may be operably linked to the Fc variants of the present invention in order to generate an Fc fusion. Thus for example, an Fc fusion that targets EGFR could be constructed by operably linking an Fc variant to EGF, TGF-b, or any other ligand, discovered or undiscovered, that binds EGFR. Accordingly, an Fc variant of the present invention could be operably linked to EGFR in order to generate an Fc fusion that binds EGF, TGF-b, or any other ligand, discovered or undiscovered, that binds EGFR. Thus virtually any polypeptide, whether a ligand, receptor, or some other protein or protein domain, including but not limited to the aforementioned targets and the proteins that compose their corresponding biochemical pathways, may be operably linked to the Fc variants of the present invention to develop an Fc fusion.
[160] The choice of suitable antigen depends on the desired application. For anti-cancer treatment it is desirable to have a target whose expression is restricted to the cancerous cells. Some targets that have proven especially amenable to antibody therapy are those with signaling functions. Other therapeutic antibodies exert their effects by blocking signaling of the receptor by inhibiting the binding between a receptor and its cognate ligand. Another mechanism of action of therapeutic antibodies is to cause receptor down regulation. Other antibodies do not work by signaling through their target antigen. In some cases, antibodies directed against infectious disease agents are used.
[161] In one embodiment, the Fc variants of the present invention are incorporated into an antibody against a cytokine. Alternatively, the Fc variants are fused or conjugated to a cytokine. By "cytokine" as used herein is meant a generic term for proteins released by one cell population that act on another cell as intercellular mediators. For example, as described in Penichet et al., 2001 , J Immunol Methods 248:91-101 , expressly incorporated by reference, cytokines may be fused to antibody to provide an array of desirable properties. Examples of such cytokines are lymphokines, monokines, and traditional polypeptide hormones. Included among the cytokines are growth hormone such as human growth hormone, N-methionyl human growth hormone, and bovine growth hormone; parathyroid hormone; thyroxine; insulin; proinsulin; relaxin; prorelaxin; glycoprotein hormones such as follicle stimulating hormone (FSH), thyroid stimulating hormone (TSH), and luteinizing hormone (LH); hepatic growth factor; fibroblast growth factor; prolactin; placental lactogen; tumor necrosis factor-alpha and -beta; mullerian-inhibiting substance; mouse gonadotropin- associated peptide; inhibin; activin; vascular endothelial growth factor; integrin; thrombopoietin (TPO); nerve growth factors such as NGF-beta; platelet-growth factor; transforming growth factors (TGFs) such as TGF-alpha and TGF-beta; insulin-like growth factor-l and -II; erythropoietin (EPO); osteoinductive factors; interferons such as interferon- alpha, beta, and -gamma; colony stimulating factors (CSFs) such as macrophage-CSF (M- CSF); granulocyte-macrophage-CSF (GM-CSF); and granulocyte-CSF (G-CSF); interleukins (ILs) such as IL-1 , IL-1alpha, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-11 , IL-12; IL- 15, a tumor necrosis factor such as TNF-alpha or TNF-beta; C5a; and other polypeptide factors including LIF and kit ligand (KL). As used herein, the term cytokine includes proteins from natural sources or from recombinant cell culture, and biologically active equivalents of the native sequence cytokines.
[162] Cytokines and soluble targets, such as TNF superfamily members, are preferred targets for use with the variants of the present invention. For example, anti-VEGF, anti- CTLA-4, and anti-TNF antibodies, or fragments thereof, are particularly good antibodies for the use of Fc variants that increase the FcRn binding. Therapeutics against these targets are frequently involved in the treatment of autoimmune diseases and require multiple injections over long time periods. Therefore, longer serum half-lives and less frequent treatments, brought about from the variants of the present invention, are particularly preferred. [163] A number of antibodies and Fc fusions that are approved for use, in clinical trials, or in development may benefit from the Fc variants of the present invention. These antibodies and Fc fusions are herein referred to as "clinical products and candidates". Thus in a preferred embodiment, the Fc polypeptides of the present invention may find use in a range of clinical products and candidates. For example, a number of antibodies that target CD20 may benefit from the Fc polypeptides of the present invention. For example the Fc polypeptides of the present invention may find use in an antibody that is substantially similar to rituximab (Rituxan®, IDEC/Genentech/Roche) (see for example US 5,736,137), a chimeric anti-CD20 antibody approved to treat Non-Hodgkin's lymphoma; HuMax-CD20, an anti-CD20 currently being developed by Genmab, an anti-CD20 antibody described in US 5,500,362, AME-133 (Applied Molecular Evolution), hA20 (Immunomedics, Inc.), HumaLYM (Intracel), and PRO70769 (PCT/US2003/040426, entitled "Immunoglobulin Variants and Uses Thereof"). A number of antibodies that target members of the family of epidermal growth factor receptors, including EGFR (ErbB-1 ), Her2/neu (ErbB-2), Her3 (ErbB-3), Her4 (ErbB-4), may benefit from the Fc polypeptides of the present invention. For example the Fc polypeptides of the present invention may find use in an antibody that is substantially similar to trastuzumab (Herceptin®, Genentech) (see for example US 5,677,171 ), a humanized anti- Her2/neu antibody approved to treat breast cancer; pertuzumab (rhuMab-2C4, Omnitarg™), currently being developed by Genentech; an anti-Her2 antibody described in US 4,753,894; cetuximab (Erbitux®, Imclone) (US 4,943,533; PCT WO 96/40210), a chimeric anti-EGFR antibody in clinical trials for a variety of cancers; ABX-EGF (US 6,235,883), currently being developed by Abgenix-lmmunex-Amgen; HuMaX-EGFr (USSN 10/172,317), currently being developed by Genmab; 425, EMD55900, EMD62000, and EMD72000 (Merck KGaA) (US 5,558,864; Murthy et al. 1987, Arch Biochem Biophys. 252(2):549-60; Rodeck et al., 1987, J Cell Biochem. 35(4):315-20; Kettleborough et al., 1991 , Protein Eng. 4(7):773-83); ICR62 (Institute of Cancer Research) (PCT WO 95/20045; Modjtahedi et al., 1993, J. Cell Biophys. 1993, 22(1-3):129-46; Modjtahedi et al., 1993, Br J Cancer. 1993, 67(2):247-53; Modjtahedi et al, 1996, Br J Cancer, 73(2):228-35; Modjtahedi et al, 2003, lnt J Cancer, 105(2):273-80); TheraCIM hR3 (YM Biosciences, Canada and Centro de lmmunologia Molecular, Cuba (US 5,891 ,996; US 6, 506,883; Mateo et al, 1997, Immunotechnology, 3(1 ):71-81 ); mAb-806 (Ludwig lnstitue for Cancer Research, Memorial Sloan-Kettering) (Jungbluth et al. 2003, Proc Natl Acad Sci U S A. 100(2):639-44); KSB-102 (KS Biomedix); MR1-1 (IVAX, National Cancer Institute) (PCT WO 0162931A2); and SC100 (Scancell) (PCT WO 01/88138). In another preferred embodiment, the Fc polypeptides of the present invention may find use in alemtuzumab (Campath®, Millenium), a humanized monoclonal antibody currently approved for treatment of B-cell chronic lymphocytic leukemia. The Fc polypeptides of the present invention may find use in a variety of antibodies or Fc fusions that are substantially similar to other clinical products and candidates, including but not limited to muromonab-CD3 (Orthoclone OKT3®), an anti-CD3 antibody developed by Ortho Biotech/Johnson & Johnson, ibritumomab tiuxetan (Zevalin®), an anti-CD20 antibody developed by IDEC/Schering AG, gemtuzumab ozogamicin (Mylotarg®), an anti-CD33 (p67 protein) antibody developed by Celltech/Wyeth, alefacept (Amevive®), an anti-LFA-3 Fc fusion developed by Biogen), abciximab (ReoPro®), developed by Centocor/Lilly, basiliximab (Simulect®), developed by Novartis, palivizumab (Synagis®), developed by Medlmmune, infliximab (Remicade®), an anti-TNFalpha antibody developed by Centocor, adalimumab (Humira®), an anti-TNFalpha antibody developed by Abbott, Humicade™, an anti-TNFalpha antibody developed by Celltech, etanercept (Enbrel®), an anti-TNFalpha Fc fusion developed by Immunex/Amgen, ABX-CBL, an anti-CD147 antibody being developed by Abgenix, ABX-IL8, an anti-IL8 antibody being developed by Abgenix, ABX-MA1 , an anti- MUC18 antibody being developed by Abgenix, Pemtumomab (R1549, 90Y-muHMFG1 ), an anti-MUC1 In development by Antisoma, Therex (R1550), an anti-MUC1 antibody being developed by Antisoma, AngioMab (AS1405), being developed by Antisoma, HuBC-1 , being developed by Antisoma, Thioplatin (AS1407) being developed by Antisoma, Antegren® (natalizumab), an anti-alpha-4-beta-1 (VLA-4) and alpha-4-beta-7 antibody being developed by Biogen, VLA-1 mAb, an anti-VLA-1 integrin antibody being developed by Biogen, LTBR mAb, an anti-lymphotoxin beta receptor (LTBR) antibody being developed by Biogen, CAT- 152, an anti-TGF-β2 antibody being developed by Cambridge Antibody Technology, J695, an anti-IL-12 antibody being developed by Cambridge Antibody Technology and Abbott, CAT-192, an anti-TGFβ1 antibody being developed by Cambridge Antibody Technology and Genzyme, CAT-213, an anti-Eotaxin1 antibody being developed by Cambridge Antibody Technology, LymphoStat-B™ an anti-Blys antibody being developed by Cambridge Antibody Technology and Human Genome Sciences Inc., TRAIL-RI mAb, an anti-TRAIL-R1 antibody being developed by Cambridge Antibody Technology and Human Genome Sciences, Inc., Avastin™ (bevacizumab, rhuMAb-VEGF), an anti-VEGF antibody being developed by Genentech, an anti-HER receptor family antibody being developed by Genentech, Anti- Tissue Factor (ATF), an anti-Tissue Factor antibody being developed by Genentech, Xolair™ (Omalizumab), an anti-lgE antibody being developed by Genentech, Raptiva™ (Efalizumab), an anti-CD11 a antibody being developed by Genentech and Xoma, MLN-02 Antibody (formerly LDP-02), being developed by Genentech and Millenium Pharmaceuticals, HuMax CD4, an anti-CD4 antibody being developed by Genmab, HuMax-IL15, an anti-IL15 antibody being developed by Genmab and Amgen, HuMax-lnflam, being developed by Genmab and Medarex, HuMax-Cancer, an anti-Heparanase I antibody being developed by Genmab and Medarex and Oxford GcoSciences, HuMax-Lymphoma, being developed by Genmab and Amgen, HuMax-TAC, being developed by Genmab, IDEC-131 , and anti- CD40L antibody being developed by IDEC Pharmaceuticals, IDEC-151 (Clenoliximab), an anti-CD4 antibody being developed by IDEC Pharmaceuticals, IDEC-1 14, an anti-CD80 antibody being developed by IDEC Pharmaceuticals, IDEC-152, an anti-CD23 being developed by IDEC Pharmaceuticals, anti-macrophage migration factor (MIF) antibodies being developed by IDEC Pharmaceuticals, BEC2, an anti-idiotypic antibody being developed by Imclone, IMC-1 C1 1 , an anti-KDR antibody being developed by Imclone, DC101 , an anti-flk-1 antibody being developed by Imclone, anti-VE cadherin antibodies being developed by Imclone, CEA-Cide™ (labetuzumab), an anti-carcinoembryonic antigen (CEA) antibody being developed by Immunomedics, LymphoCide™ (Epratuzumab), an anti- CD22 antibody being developed by Immunomedics, AFP-Cide, being developed by Immunomedics, MyelomaCide, being developed by Immunomedics, LkoCide, being developed by Immunomedics, ProstaCide, being developed by Immunomedics, MDX-010, an anti-CTLA4 antibody being developed by Medarex, MDX-060, an anti-CD30 antibody being developed by Medarex, MDX-070 being developed by Medarex, MDX-018 being developed by Medarex, Osidem™ (IDM-1 ), and anti-Her2 antibody being developed by Medarex and Immuno-Designed Molecules, HuMax™-CD4, an anti-CD4 antibody being developed by Medarex and Genmab, HuMax-IL15, an anti-IL15 antibody being developed by Medarex and Genmab, CNTO 148, an anti-TNFα antibody being developed by Medarex and Centocor/J&J, CNTO 1275, an anti-cytokine antibody being developed by Centocor/J&J, MOR101 and MOR102, anti-intercellular adhesion molecule-1 (ICAM-1 ) (CD54) antibodies being developed by MorphoSys, MOR201 , an anti-fibroblast growth factor receptor 3 (FGFR- 3) antibody being developed by MorphoSys, Nuvion® (visilizumab), an anti-CD3 antibody being developed by Protein Design Labs, HuZAF™, an anti-gamma interferon antibody being developed by Protein Design Labs, Anti-α5β1 Integrin, being developed by Protein Design Labs, anti-IL-12, being developed by Protein Design Labs, ING-1 , an anti-Ep-CAM antibody being developed by Xoma, and MLN01 , an anti-Beta2 integrin antibody being developed by Xoma, all of the above-cited references in this paragraph are expressly incorporated herein by reference.
[164] The Fc polypeptides of the present invention may be incorporated into the aforementioned clinical candidates and products, or into antibodies and Fc fusions that are substantially similar to them. The Fc polypeptides of the present invention may be incorporated into versions of the aforementioned clinical candidates and products that are humanized, affinity matured, engineered, or modified in some other way. [165] In one embodiment, the Fc polypeptides of the present invention are used for the treatment of autoimmune, inflammatory, or transplant indications. Target antigens and clinical products and candidates that are relevant for such diseases include but are not limited to anti-α4β7 integrin antibodies such as LDP-02, anti-beta2 integrin antibodies such as LDP-01 , anti-complement (C5) antibodies such as 5G1.1 , anti-CD2 antibodies such as BTI-322, MEDI-507, anti-CD3 antibodies such as OKT3, SMART anti-CD3, anti-CD4 antibodies such as IDEC-151 , MDX-CD4, OKT4A, anti-CD1 1a antibodies, anti-CD14 antibodies such as IC14, anti-CD18 antibodies, anti-CD23 antibodies such as IDEC 152, anti-CD25 antibodies such as Zenapax, anti-CD40L antibodies such as 5c8, Antova, IDEC- 131 , anti-CD64 antibodies such as MDX-33, anti-CD80 antibodies such as IDEC-114, anti- CD147 antibodies such as ABX-CBL, anti-E-selectin antibodies such as CDP850, anti- gpllb/llla antibodies such as ReoPro/Abcixima, anti-ICAM-3 antibodies such as ICM3, anti- ICE antibodies such as VX-740, anti-FcR1 antibodies such as MDX-33, anti-lgE antibodies such as rhuMab-E25, anti-IL-4 antibodies such as SB-240683, anti-IL-5 antibodies such as SB-240563, SCH55700, anti-IL-8 antibodies such as ABX-IL8, anti-interferon gamma antibodies, anti-TNF (TNF, TNFa, TNFa, TNF-alpha) antibodies such as CDP571 , CDP870, D2E7, Infliximab, MAK-195F, and anti-VLA-4 antibodies such as Antegren. [166] Fc variants of the present invention such as those with increased binding to FcRn may be utilized in TNF inhibitor molecules to provide enhanced properties. Useful TNF inhibitor molecules include any molecule that inhibits the action of TNF-alpha in a mammal. Suitable examples include the Fc fusion Enbrel® (etanercept) and the antibodies Humira® (adalimumab) and Remicade® (infliximab). Monoclonal antibodies (such as Remicade and Humira) engineered using the Fc variants of the present invention to increase FcFn binding, may translate to better efficacy through an increased half-life.
[167] In some embodiments, antibodies against infectious diseases are used. Antibodies against eukaryotic cells include antibodies targeting yeast cells, including but not limited to Saccharomyces cerevisiae, Hansenula polymorpha, Kluyveromyces fragilis and K. lactis, Pichia guillerimondii and P. pastoris, Schizosaccharomyces pombe, Plasmodium falciparium, and Yarrowia lipolytica.
[168] Antibodies against additional fungal cells are also useful, including target antigens associated with Candida strains including Candida glabrata, Candida albicans, C. krusei, C. lusitaniaeand C. maltosa, as well as species of Aspergillus, Cryptococcus, Histoplasma, Coccidioides, Blastomyces, and Penicillium, among others
[169] Antibodies directed against target antigens associated with protozoa include, but are not limited to, antibodies associated with Trypanosoma, Leishmania species including Leishmania donovanii; , Plasmodium spp., Pneumocystis carinii, Cryptosporidium parvum, Giardia lamblia, Entamoeba histolytica, and Cyclospora cayetanensis. [170] Antibodies against prokaryotic antigens are also useful, including antibodies against suitable bacteria such as pathogenic and non-pathogenic prokaryotes including but not limited to Bacillus, including Bacillus anthracis; Vibrio, e.g. V. cholerae; Escherichia, e.g. Enterotoxigenic E. coli, Shigella, e.g. S. dysenteriae; Salmonella, e.g. S. typhi; Mycobacterium e.g. M. tuberculosis, M. leprae; Clostridium, e.g. C. botulinum, C. tetani, C. difficile, C.perfringens; Cornyebacterium, e.g. C. diphtheriae; Streptococcus, S. pyogenes, S. pneumoniae; Staphylococcus, e.g. S. aureus; Haemophilus, e.g. H. influenzae; Neisseria, e.g. N. meningitidis, N. gonorrhoeae; Yersinia, e.g. Y. lamblia, Y. pestis, Pseudomonas, e.g. P. aeruginosa, P. putida; Chlamydia, e.g. C. trachomatis; Bordetella, e.g. B. pertussis; Treponema, e.g. T. palladium; B. anthracis, Y. pestis, Brucella spp., F. tularensis, B. mallei, B .pseudomallei, B. mallei, B.pseudomallei , C. botulinum , Salmonella spp., SEB V. cholerae toxin B, E. coli O157:H7, Listeria spp., Trichosporon beigelii, Rhodotorula species, Hansenula anomala, Enterobacter sp., Klebsiella sp., Listeria sp., Mycoplasma sp. and the like.
[171] In some aspects, the antibodies are directed against viral infections; these viruses include, but are not limited to, including orthomyxoviruses, (e.g. influenza virus), paramyxoviruses (e.g respiratory syncytial virus, mumps virus, measles virus), adenoviruses, rhinoviruses, coronaviruses, reoviruses, togaviruses (e.g. rubella virus), parvoviruses, poxviruses (e.g. variola virus, vaccinia virus), enteroviruses (e.g. poliovirus, coxsackievirus), hepatitis viruses (including A, B and C), herpesviruses (e.g. Herpes simplex virus, varicella-zoster virus, cytomegalovirus, Epstein-Barr virus), rotaviruses, Norwalk viruses, hantavirus, arenavirus, rhabdovirus (e.g. rabies virus), retroviruses (including HIV, HTLV-I and -II), papovaviruses (e.g. papillomavirus), polyomaviruses, and picornaviruses, and the like.
[172] Optimized IgG Variant Properties
[173] The present application also provides IgG variants that are optimized for a variety of therapeutically relevant properties. An IgG variant that is engineered or predicted to display one or more optimized properties is herein referred to as an "optimized IqG variant". The most preferred properties that may be optimized include but are not limited to enhanced or reduced affinity for FcRn and increased or decreased in vivo half-life. Suitable embodiments include antibodies that exhibit increased binding affinity to FcRn at lowered pH, such as the pH associated with endosomes, e.g. pH 6.0, while maintaining the reduced affinity at higher pH, such as 7.4., to allow increased uptake into endosomes but normal release rates. Similarly, these antibodies with modulated FcRn binding may optionally have other desirable properties, such as modulated FcvR binding, such as outlined in U.S.S.N.s U.S.S.N.s 11/174,287, 11/124,640, 10/822,231 , 10/672,280, 10/379,392, and the patent application entitled IgG Immunoglobulin variants with optimized effector function filed on October 21 , 2005 having application no. 1 1/256,060. That is, optimized properties also include but are not limited to enhanced or reduced affinity for an FcγR. In one optional embodiment, the IgG variants are optimized to possess enhanced affinity for a human activating FcγR, preferably FcγRllla in addition to the FcRn binding profile. In yet another optional alternate embodiment, the IgG variants are optimized to possess reduced affinity for the human inhibitory receptor FcγRllb. That is, particular embodiments embrace the use of antibodies that show increased binding to FcRn, and increased binding to FcγRllla. Other embodiments utilize use of antibodies that show increased binding to FcRn, and increased binding to FcγRllla. These embodiments are anticipated to provide IgG polypeptides with enhanced therapeutic properties in humans, for example enhanced effector function and greater anticancer potency. In an alternate embodiment, the IgG variants are optimized to have increased or reduced affinity for FcRn and increased or decreased affinity for a human FcγR, including but not limited to FcγRI, FcγRlla, FcγRllb, FcγRllc, FcγRllla, and FcγRlllb including their allelic variations. These embodiments are anticipated to provide IgG polypeptides with enhanced therapeutic properties in humans, for example increased serum half-life and reduced effector function. In other embodiments, IgG variants provide enhanced affinity for FcRn and enhanced affinity for one or more FcγRs, yet reduced affinity for one or more other FcγRs. For example, an IgG variant may have enhanced binding to FcRn and FcγRllla, yet reduced binding to FcγRllb. Alternately, an IgG variant may have reduced binding to FcRn and to FcγR's. In another embodiment, an IgG variant may have reduced affinity for FcRn and enhanced affinity for FcγRllb, yet reduced affinity to one or more activating FcγRs. In yet another embodiment, an IgG variant may have increased serum half-life and reduced effector functions.
[174] Preferred embodiments comprise optimization of binding to a human FcRn and FcγR, however in alternate embodiments the IgG variants possess enhanced or reduced affinity for FcRn and FcγR from nonhuman organisms, including but not limited to rodents and non- human primates. IgG variants that are optimized for binding to a nonhuman FcRn may find use in experimentation. For example, mouse models are available for a variety of diseases that enable testing of properties such as efficacy, toxicity, and pharmacokinetics for a given drug candidate. As is known in the art, cancer cells can be grafted or injected into mice to mimic a human cancer, a process referred to as xenografting. Testing of IgG variants that comprise IgG variants that are optimized for FcRn may provide valuable information with regard to the clearance characteristics of the protein, its mechanism of clearance, and the like. The IgG variants may also be optimized for enhanced functionality and/or solution properties in aglycosylated form. The Fc ligands include but are not limited to FcRn, FcγRs, C1 q, and proteins A and G, and may be from any source including but not limited to human, mouse, rat, rabbit, or monkey, preferably human. In an alternately preferred embodiment, the IgG variants are optimized to be more stable and/or more soluble than the aglycosylated form of the parent IgG variant.
[175] IgG variants can include modifications that modulate interaction with Fc ligands other than FcRn and FcγRs, including but not limited to complement proteins, and Fc receptor homologs (FcRHs). FcRHs include but are not limited to FcRHI , FcRH2, FcRH3, FcRH4, FcRH5, and FcRH6 (Davis et al., 2002, Immunol. Reviews 190:123-136, entirely incorporated by reference).
[176] Preferably, the Fc ligand specificity of the IgG variant will determine its therapeutic utility. The utility of a given IgG variant for therapeutic purposes will depend on the epitope or form of the target antigen and the disease or indication being treated. For most targets and indications, enhanced FcRn binding may be preferable as the enhanced FcRn binding may result in an increase in serum half-life. Longer serum half-lives allow less frequent dosing or lower dosing of the therapeutic. This is particularly preferable when the therapeutic agent is given in response to an indication that requires repeated administration. For some targets and indications, decreased FcRn affinity may be preferable. This may be particularly preferable when a variant Fc with increased clearance or decreased serum half-life is desired, for example in Fc polypeptides used as imaging agents or radio-therapeutics. [177] IgG variants may be used that comprise IgG variants that provide enhanced affinity for FcRn with enhanced activating FcγRs and/or reduced affinity for inhibitory FcγRs. For some targets and indications, it may be further beneficial to utilize IgG variants that provide differential selectivity for different activating FcγRs; for example, in some cases enhanced binding to FcγRlla and FcγRllla may be desired, but not FcγRI, whereas in other cases, enhanced binding only to FcγRlla may be preferred. For certain targets and indications, it may be preferable to utilize IgG variants that alter FcRn binding and enhance both FcγR- mediated and complement-mediated effector functions, whereas for other cases it may be advantageous to utilize IgG variants that enhance FcRn binding, or serum half-life, and either FcγR-mediated or complement-mediated effector functions. For some targets or cancer indications, it may be advantageous to reduce or ablate one or more effector functions, for example by knocking out binding to C1 q, one or more FcγR's, FcRn, or one or more other Fc ligands. For other targets and indications, it may be preferable to utilize IgG variants that provide enhanced binding to the inhibitory FcγRllb, yet WT level, reduced, or ablated binding to activating FcγRs. This may be particularly useful, for example, when the goal of an IgG variant is to inhibit inflammation or auto-immune disease, or modulate the immune system in some way. Because auto-immune diseases are generally long-lasting and treatment is given in repeated dosing, their treatment with Fc variants with increased half-life from increased FcRn is particularly preferred.
[178] Modification may be made to improve the IgG stability, solubility, function, or clinical use. In a preferred embodiment, the IgG variants can include modifications to reduce immunogenicity in humans. In a most preferred embodiment, the immunogenicity of an IgG variant is reduced using a method described in USSN 11/004,590, entirely incorporated by reference. In alternate embodiments, the IgG variants are humanized (Clark, 2000, Immunol Today 21 :397-402, entirely incorporated by reference).
[179] The IgG variants can include modifications that reduce immunogenicity. Modifications to reduce immunogenicity can include modifications that reduce binding of processed peptides derived from the parent sequence to MHC proteins. For example, amino acid modifications would be engineered such that there are no or a minimal number of immune epitopes that are predicted to bind, with high affinity, to any prevalent MHC alleles. Several methods of identifying MHC-binding epitopes in protein sequences are known in the art and may be used to score epitopes in an IgG variant. See for example WO 98/52976; WO 02/079232; WO 00/3317; USSN 09/903,378; USSN 10/039,170; USSN 60/222,697; USSN 10/754,296; PCT WO 01/21823; and PCT WO 02/00165; Mallios, 1999, Bioinformatics 15: 432-439; Mallios, 2001 , Bioinformatics 17: 942-948; Sturniolo et al., 1999, Nature Biotech. 17: 555-561 ; WO 98/59244; WO 02/069232; WO 02/77187; Marshall et al., 1995, J. Immunol. 154: 5927-5933; and Hammer et al., 1994, J. Exp. Med. 180: 2353-2358, all entirely incorporated by reference. Sequence-based information can be used to determine a binding score for a given peptide - MHC interaction (see for example Mallios, 1999, Bioinformatics 15: 432-439; Mallios, 2001 , Bioinformatics 17: p942-948; Sturniolo et. al., 1999, Nature Biotech. 17: 555-561 , all entirely incorporated by reference). [180] Engineering IgG Variants
[181] Variants of the present invention may be designed by various means. The variants, as described herein, may be insertions, deletions, substitutions, other modifications, or combinations of these and other changes. A particularly novel embodiment of the present invention is the design of insertions and deletions that either improve or reduce the binding of an Fc polypeptide to an Fc ligand. As disclosed herein, insertions or deletions may be made that increase or decrease the affinity of the Fc polypeptide for FcRn. Insertions and deletions may be designed by rational approaches or by approaches that include the use or random components, such as random or semi-random library creation or screening. In an alternative embodiment, substitutions are disclosed that increase or decrease the affinity of the Fc polypeptide for FcRn.
[182] Backbone Modifications: Insertions and deletions
[183] Variant Fc polypeptides may be created by substituting a variant amino acid in place of the parent amino acid at a position in the Fc polypeptide. By substituting one or more amino acids for variant amino acids in the Fc polypeptide, the side chains at those positions are altered. Most useful substitutions modify the Fc properties by altering the Fc side chains. The substituted side chains may interact directly or indirectly with an Fc binding partner that is associated with an Fc function or property. The at least one substitution alters the covalent structure of one or more side chains of the parent Fc polypeptide. [184] Alternatively, variant Fc polypeptides may be created that change the covalent structure of the backbone of the parent Fc polypeptide. The backbone atoms in proteins are the peptide nitrogen, the alpha carbon, the carbonyl or peptide carbon and the carbonyl oxygen. Changing the covalent structure of the backbone provides additional methods of altering the properties of the Fc polypeptides. The covalent structure of the Fc backbone may be altered by the addition of atoms into the backbone, e.g. by inserting one or more amino acids, or the subtraction of atoms from the backbone, e.g. by deleting one or more amino acids. The covalent structure of the backbone may also be altered by changing individual atoms of the backbone to other atoms (Deechongkit et al., J Am Chem Soc. 2004. 126(51 ):16762-71 , entirely incorporated by reference). As is known in the art and is illustrated herein, insertions or deletions of amino acids in Fc polypeptides may be done by inserting or deleting the corresponding nucleotides in the DNA encoding the Fc polypeptide. Alternatively, as is known in the art, insertions or deletions of amino acids may be done during synthesis of Fc polypeptides.
[185] The design of insertions or deletions of amino acids that alter the interaction of the Fc polypeptide with one or more binding partners (e.g. FcgammaR's, FcRn, C1 q) may be done by considering the structure of the complex of the Fc polypeptide and its binding partner. In a less preferred embodiment, the design may be done by considering the structure of the Fc polypeptide and information about the Fc region involved in binding the binding partner. This information may be obtained by mutagenesis experiments, phage display experiments, homology comparisons, computer modeling or other means.
[186] Preferred positions in the amino acid sequence for insertions or deletions that affect the Fc binding interactions, but do not affect the overall structure, stability, expression or use of the Fc polypeptide, are in loops that are involved in the Fc/Fc-binding partner interactions. To alter FcRn binding to the Fc polypeptide, positions 244-257, 279-284, 307-317, 383-390, and 428-435 are preferred loop locations for insertions or deletions (numbering from EU index of Kabat et al., Burmeister et al., 1994, Nature, 372:379-383; Martin et al., 2001 , MoI Cell 7:867-877, all entirely incorporated by reference). To alter the Fcgamma receptor binding to the Fc polypeptide, positions 229-239, 266-273, 294-299, and 324-331 are preferred loop locations for insertions or deletions (numbering from EU index of Kabat et al., PDB code 1 E4K.pdb Sondermann et al. Nature. 2000 406:267, all entirely incorporated by reference). Loops are regions of the polypeptide not involved in alpha helical or beta sheet structure. Loops positions are positions that are not in either alpha helical or beta sheet structures (van Holde, Johnson and Ho. Principles of Physical Biochemistry. Prentice Hall, New Jersey 1998, Chapter 1 pp2-67, entirely incorporated by reference). Loop positions are preferred because the backbone atoms are typically more flexible and less likely involved in hydrogen bonds compared to the backbone atoms of alpha helices and beta sheets. Therefore, the lengthening or shortening of a loop due to an insertion or deletion of one or more amino acids is less likely to lead to large, disruptive changes to the Fc polypeptide, including stability, expression or other problems.
[187] Insertions and deletions may be used to alter the length of the polypeptide. For example, in loop regions, altering the loop length results in altered flexibility and conformational entropy of the loop. Insertions in a loop will generally increase the conformational entropy of the loop, which may be defined as Boltzman's constant multiplied by the natural logarithm of the number of possible conformations (van Holde, Johnson and Ho. Principles of Physical Biochemistry. Prentice Hall, New Jersey 1998, pp78, entirely incorporated by reference). By inserting at least one amino acid into a polypeptide, the total number of conformations available to the polypeptide increases. These additional conformations may be beneficial for forming favorable Fc/Fc-binding partner interactions because the Fc polypeptide may use one of the additional conformations in binding the Fc- binding protein. In this case, the insertion may lead to stronger Fc/Fc-binding partner interactions. If the additional conformations are not used in the binding interface, then the insertion may lead to weaker Fc/Fc-binding partner interactions, because the additional conformations would compete with the binding-competent conformation. Similarly, deletion of a polypeptide segment may also lead to either stronger or weaker Fc/Fc binding-partner interactions. If deletion of a segment, which reduces the possible number of backbone conformations, removes the binding-competent conformation, then the deletion may lead to weaker Fc/Fc-binding partner interactions. If the deletion does not remove the binding- competent conformation, then the deletion may lead to stronger Fc/Fc-binding partner interactions because the deletion may remove conformations that compete with the binding- competent conformation.
[188] Insertions and deletions may be used to alter the positions and orientations of the amino acids in the Fc polypeptide. Because insertions and deletions cause a change in the covalent structure of the backbone, they necessarily cause a change in the positions of the backbone atoms. Figure 7 compares the backbone positions at some loop segments, marked L1 to L4, in three different backbones. The reference backbone structure contains four loop segments, whereas the deletion backbone lacks segment L1 and the insertion segment comprises an additional segment before, ie, N-terminal to, segment L1. Deletions and insertions cause the largest change in the backbone structure near the site of the insertion or deletion. By deleting a segment near the N-terminal end of the loop, e.g. segment L1 , the loop shortens and the remaining segments shift their position closer to the loop N-terminus. This has the effect of moving the L2 segment toward the prior location of the L1 segment and toward the loop N-terminus. This change in position of the L2 segment toward the L1 segment may strengthen the binding of the Fc/Fc-binding partner complex and is preferred when there is prior information suggesting that the amino acid or amino acids located in L2 make favorable interactions with the Fc-binding partner, when located in L1. For example, if L2 contains alanine and tyrosine and substitution of two L1 amino acids for alanine and tyrosine previously lead to an Fc variant with increased binding, then deletion of L1 may create an Fc variant with increased affinity for the Fc-binding partner. [189] Similarly, an insertion of a polypeptide segment into an Fc polypeptide at the N- terminal side of a loop causes the positions of the loop segments to be shifted toward the C- terminal side of the loop. In Figure 7, an insertion of one or more amino acids before, i.e. N- terminally to, segment L1 alters the backbone conformation including a shift of the L1 segment toward the C-terminal end of the loop. This type of insertion is preferred when the amino acids located in segment L1 are known to make favorable interactions when located in the L2 positions, as the insertion may lead to stronger Fc/Fc-binding partner interactions. If weaker Fc/Fc-binding partner interactions are desired, then the insertion may be used to shift unfavorable amino acid into a new position. The inserted, deleted and reference segments (L1 to L4 in Figure 7) may be one or more than one amino acid in the Fc polypeptide.
[190] Alternatively, insertions or deletions may be used at the C-terminal end of loops in a manner analogous to the insertions or deletions at the N-terminal end of loops. Insertions at the loop C-terminus may lead to a movement of the positions N-terminal of the insertion toward the loop N-terminus. Deletions at the loop C-terminus may lead to a movement of the positions N-terminal of the deletion toward the loop C-terminus. The choice of using an insertion or deletion at the N-terminal or C-terminal end of the loop is based on the amino acids located in the loop, the desire for increased or decreased Fc/Fc-binding partner affinity, and the positional shift desired.
[191] Insertions or deletions may be used in any region of an Fc polypeptide, including the loops, the alpha helical, and the beta sheet regions. Preferred locations for insertions and deletions include loop regions, which are those that are not alpha helical or beta sheet regions. Loops are preferred because they generally accept alterations in the backbone better than alpha helixes or beta sheets. The particularly preferred locations for insertions or deletions that result in stronger protein/protein interactions are at the N-terminal or C- terminal edges of a loop. If the loop side chains are involve in the Fc/Fc-binding partner interactions, then insertions or deletion at the edges are less likely to lead to strongly detrimental changes in the binding interactions. Deletions within the exact center of the loop are more likely to remove important residues in the Fc/Fc-binding partner interface and insertions within the exact center of the loop are more likely to create unfavorable interactions in the Fc/Fc-binding partner interface. The number of residues deleted or inserted may be determined by the size of the backbone change desired with insertions or deletions of 15 or less residues being preferred, insertions or deletions of 10 or less residues being more preferred, and insertions or deletions of 5 or less residues being most preferred. [192] Once the position and size of an Fc deletion variant is designed, the entire polypeptide sequence is completely determined and the polypeptide may be constructed by methods known in the art. [193] Fc insertion variants, however, have the additional step of designing the sequence of the at least one amino acid to be inserted. Insertions of polar residues, including Ser, Thr, Asn, GIn, Ala, GIy, His, are preferred at positions expected to be exposed in the Fc polypeptide. The smaller amino acids, including Ser, Thr, and Ala, are particularly preferred as the small size is less likely to sterically interfere with the Fc/Fc-binding partner interactions. Ser and Thr also have the capability to hydrogen bond with atoms on the Fc- binding partner.
[194] Insertions also have the added flexibility that the inserted polypeptide may be designed to make favorable interactions with the Fc-binding partner as would be desire when stronger Fc/Fc-binding partner binding is desired. The length of the backbone insertion may be determined by modeling the variant backbone with a simple, generic sequence to be inserted. For example, polyserine, polyglycine or polyalanine insertions of different lengths may be constructed and modeled. Modeling may be done by a variety of methods, including homology modeling based on known three-dimensional structures of homologues comprising the insertion, and by computer modeling including MODELLER (M.A. Marti- Renom et al. Annu. Rev. Biophys. Biomol. Struct. 29, 291-325, 2000) and ROSETTA (Kuhlman et al. (2003). Science 302, 1364-8), both entirely incorporated by reference. Typically, various backbone conformations are initially generated and the final backbone structure may be determined after the identities of the side chain are established. The side chains may be designed by PDA® algorithms (US 6,188,965; 6,269,312; 6,403,312; 6,801 ,861 ; 6,804,611 ; 6,792,356, 6,950,754, and USSN 09/782,004; 09/927,790; 10/101 ,499; 10/666,307; 10/666311 ; 10/218,102, all entirely incorporated by reference). [195] Insertions and deletions may be made to alter the binding of Fc polypeptides to FcgammaR in an analogous manner to the described method to alter FcRn-binding properties. Fc domains bind to the FcgammaR at the position indicated in Figure 1. Structures of the Fc/FcgammaR complex, including PDB codes 1T89 and 11 IS (Radaev S et al. J. Biol. Chem. v276, p.16469-16477 entirely incorporated by reference), demonstrate the interacting residues and loops between the two structures. Mutagenesis results such as those found in US1 1/124620 and US6737056, both entirely incorporated by reference) all have utility in determined appropriate shifts of backbone positioning. [196] Insertions and deletions may be designed in any polypeptide besides Fc polypeptides by the methods described herein. For example, insertions or deletions in the TNF superfamily member, APRIL, may be designed with the aid of its three-dimensional structure (PDB code 1XU1.pdb, Hymowitz, et al. (2005) J. Biol. Chem. 280:7218, entirely incorporated by reference). Insertions or deletions may be designed to increase APRIL binding to its receptor, TACI. The loop residues preferred as insertion or deletion sites are residues Ser1 18-Val124, Asp164-Phe167, Pro192-Ala198, Pro221-Lys226. These loops interact with TACI in the APRI L/TACI complex and mediate binding. [197] Polypeptides incorporating variants
[198] The IgG variants can be based on human IgG sequences, and thus human IgG sequences are used as the "base" sequences against which other sequences are compared, including but not limited to sequences from other organisms, for example rodent and primate sequences. IgG variants may also comprise sequences from other immunoglobulin classes such as IgA, IgE, IgD, IgM, and the like. It is contemplated that, although the IgG variants are engineered in the context of one parent IgG, the variants may be engineered in or "transferred" to the context of another, second parent IgG. This is done by determining the "equivalent" or "corresponding" residues and substitutions between the first and second IgG, typically based on sequence or structural homology between the sequences of the IgGs. In order to establish homology, the amino acid sequence of a first IgG outlined herein is directly compared to the sequence of a second IgG. After aligning the sequences, using one or more of the homology alignment programs known in the art (for example using conserved residues as between species), allowing for necessary insertions and deletions in order to maintain alignment (i.e., avoiding the elimination of conserved residues through arbitrary deletion and insertion), the residues equivalent to particular amino acids in the primary sequence of the first IgG variant are defined. Alignment of conserved residues preferably should conserve 100% of such residues. However, alignment of greater than 75% or as little as 50% of conserved residues is also adequate to define equivalent residues. Equivalent residues may also be defined by determining structural homology between a first and second IgG that is at the level of tertiary structure for IgGs whose structures have been determined. In this case, equivalent residues are defined as those for which the atomic coordinates of two or more of the main chain atoms of a particular amino acid residue of the parent or precursor (N on N, CA on CA, C on C and O on O) are within 0.13 nm and preferably 0.1 nm after alignment. Alignment is achieved after the best model has been oriented and positioned to give the maximum overlap of atomic coordinates of non-hydrogen protein atoms of the proteins. Regardless of how equivalent or corresponding residues are determined, and regardless of the identity of the parent IgG in which the IgGs are made, what is meant to be conveyed is that the IgG variants discovered by can be engineered into any second parent IgG that has significant sequence or structural homology with the IgG variant. Thus for example, if a variant antibody is generated wherein the parent antibody is human IgGI , by using the methods described above or other methods for determining equivalent residues, the variant antibody may be engineered in another IgGI parent antibody that binds a different antigen, a human lgG2 parent antibody, a human IgA parent antibody, a mouse lgG2a or lgG2b parent antibody, and the like. Again, as described above, the context of the parent IgG variant does not affect the ability to transfer the IgG variants to other parent IgGs. [199] Methods for engineering, producing, and screening IgG variants are provided. The described methods are not meant to constrain to any particular application or theory of operation. Rather, the provided methods are meant to illustrate generally that one or more IgG variants may be engineered, produced, and screened experimentally to obtain IgG variants with optimized effector function. A variety of methods are described for designing, producing, and testing antibody and protein variants in USSN 10/754,296, and USSN 10/672,280, both entirely incorporated by reference.
[200] A variety of protein engineering methods may be used to design IgG variants with optimized effector function. In one embodiment, a structure-based engineering method may be used, wherein available structural information is used to guide substitutions, insertions or deletions. In a preferred embodiment, a computational screening method may be used, wherein substitutions are designed based on their energetic fitness in computational calculations. See for example USSN 10/754,296 and USSN 10/672,280, and references cited therein, all entirely incorporated by reference.
[201] An alignment of sequences may be used to guide substitutions at the identified positions. One skilled in the art will appreciate that the use of sequence information may curb the introduction of substitutions that are potentially deleterious to protein structure. The source of the sequences may vary widely, and include one or more of the known databases, including but not limited to the Kabat database (Northwestern University); Johnson & Wu, 2001 , Nucleic Acids Res. 29:205-206; Johnson & Wu, 2000, Nucleic Acids Res. 28:214- 218), the IMGT database (IMGT, the international ImMunoGeneTics information system®; ; Lefranc et al., 1999, Nucleic Acids Res. 27:209-212; Ruiz et al., 2000 Nucleic Acids Res. 28:219-221 ; Lefranc et al., 2001 , Nucleic Acids Res. 29:207-209; Lefranc et al., 2003, Nucleic Acids Res. 31 :307-310), and VBASE, all entirely incorporated by reference. Antibody sequence information can be obtained, compiled, and/or generated from sequence alignments of germline sequences or sequences of naturally occurring antibodies from any organism, including but not limited to mammals. One skilled in the art will appreciate that the use of sequences that are human or substantially human may further have the advantage of being less immunogenic when administered to a human. Other databases which are more general nucleic acid or protein databases, i.e. not particular to antibodies, include but are not limited to SwissProt, GenBank Entrez, and EMBL Nucleotide Sequence Database. Aligned sequences can include VH, VL, CH, and/or CL sequences. There are numerous sequence- based alignment programs and methods known in the art, and all of these find use in the generation of sequence alignments. [202] Alternatively, random or semi-random mutagenesis methods may be used to make amino acid modifications at the desired positions. In these cases positions are chosen randomly, or amino acid changes are made using simplistic rules. For example all residues may be mutated to alanine, referred to as alanine scanning. Such methods may be coupled with more sophisticated engineering approaches that employ selection methods to screen higher levels of sequence diversity. As is well known in the art, there are a variety of selection technologies that may be used for such approaches, including, for example, display technologies such as phage display, ribosome display, cell surface display, and the like, as described below.
[203] Methods for production and screening of IgG variants are well known in the art. General methods for antibody molecular biology, expression, purification, and screening are described in Antibody Engineering, edited by Duebel & Kontermann, Springer-Verlag, Heidelberg, 2001 ; and Hayhurst & Georgiou, 2001 , Curr Opin Chem Biol 5:683-689; Maynard & Georgiou, 2000, Annu Rev Biomed Eng 2:339-76. Also see the methods described in USSN 10/754,296; USSN 10/672,280; and USSN 10/822,231 ; and 11/124,620, all entirely incorporated by reference.
Preferred variants of the present invention include those found in Figure 8. Alternatively preferred variants of the present invention include those found in Figure 9. Additionally alternatively preferred variants of the present invention include those found in Figure 10. These variants have shown increased binding to the Fc receptor, FcRn, as illustrated in the examples.
[204] Making IgG Variants
[205] The IgG variants can be made by any method known in the art. In one embodiment, the IgG variant sequences are used to create nucleic acids that encode the member sequences, and that may then be cloned into host cells, expressed and assayed, if desired. These practices are carried out using well-known procedures, and a variety of methods that may find use in are described in Molecular Cloning - A Laboratory Manual, 3rd Ed. (Maniatis, Cold Spring Harbor Laboratory Press, New York, 2001 ), and Current Protocols in Molecular Biology (John Wiley & Sons), both entirely incorporated by reference. The nucleic acids that encode the IgG variants may be incorporated into an expression vector in order to express the protein. Expression vectors typically include a protein operably linked, that is, placed in a functional relationship, with control or regulatory sequences, selectable markers, any fusion partners, and/or additional elements. The IgG variants may be produced by culturing a host cell transformed with nucleic acid, preferably an expression vector, containing nucleic acid encoding the IgG variants, under the appropriate conditions to induce or cause expression of the protein. A wide variety of appropriate host cells may be used, including but not limited to mammalian cells, bacteria, insect cells, and yeast. For example, a variety of cell lines that may find use are described in the ATCC cell line catalog, available from the American Type Culture Collection, entirely incorporated by reference. The methods of introducing exogenous nucleic acid into host cells are well known in the art, and will vary with the host cell used.
[206] In a preferred embodiment, IgG variants are purified or isolated after expression. Antibodies may be isolated or purified in a variety of ways known to those skilled in the art. Standard purification methods include chromatographic techniques, electrophoretic, immunological, precipitation, dialysis, filtration, concentration, and chromatofocusing techniques. As is well known in the art, a variety of natural proteins bind antibodies, for example bacterial proteins A, G, and L, and these proteins may find use in purification. Often, purification may be enabled by a particular fusion partner. For example, proteins may be purified using glutathione resin if a GST fusion is employed, Ni+2 affinity chromatography if a His-tag is employed, or immobilized anti-flag antibody if a flag-tag is used. For general guidance in suitable purification techniques, see Antibody Purification: Principles and Practice, 3rd Ed., Scopes, Springer-Verlag, NY, 1994, entirely incorporated by reference. [207] Screening IgG Variants
[208] Fc variants may be screened using a variety of methods, including but not limited to those that use in vitro assays, in vivo and cell-based assays, and selection technologies. Automation and high-throughput screening technologies may be utilized in the screening procedures. Screening may employ the use of a fusion partner or label, for example an immune label, isotopic label, or small molecule label such as a fluorescent or colorimetric dye.
[209] In a preferred embodiment, the functional and/or biophysical properties of Fc variants are screened in an in vitro assay. In a preferred embodiment, the protein is screened for functionality, for example its ability to catalyze a reaction or its binding affinity to its target.
[210] As is known in the art, subsets of screening methods are those that select for favorable members of a library. The methods are herein referred to as "selection methods", and these methods find use in the present invention for screening Fc variants. When protein libraries are screened using a selection method, only those members of a library that are favorable, that is which meet some selection criteria, are propagated, isolated, and/or observed. A variety of selection methods are known in the art that may find use in the present invention for screening protein libraries. Other selection methods that may find use in the present invention include methods that do not rely on display, such as in vivo methods. A subset of selection methods referred to as "directed evolution" methods are those that include the mating or breading of favorable sequences during selection, sometimes with the incorporation of new mutations. [211] In a preferred embodiment, Fc variants are screened using one or more cell-based or in vivo assays. For such assays, purified or unpurified proteins are typically added exogenously such that cells are exposed to individual variants or pools of variants belonging to a library. These assays are typically, but not always, based on the function of the Fc polypeptide; that is, the ability of the Fc polypeptide to bind to its target and mediate some biochemical event, for example effector function, ligand/receptor binding inhibition, apoptosis, and the like. Such assays often involve monitoring the response of cells to the IgG, for example cell survival, cell death, change in cellular morphology, or transcriptional activation such as cellular expression of a natural gene or reporter gene. For example, such assays may measure the ability of Fc variants to elicit ADCC, ADCP, or CDC. For some assays additional cells or components, that is in addition to the target cells, may need to be added, for example example serum complement, or effector cells such as peripheral blood monocytes (PBMCs), NK cells, macrophages, and the like. Such additional cells may be from any organism, preferably humans, mice, rat, rabbit, and monkey. Antibodies may cause apoptosis of certain cell lines expressing the target, or they may mediate attack on target cells by immune cells which have been added to the assay. Methods for monitoring cell death or viability are known in the art, and include the use of dyes, immunochemical, cytochemical, and radioactive reagents. Transcriptional activation may also serve as a method for assaying function in cell-based assays. Alternatively, cell-based screens are performed using cells that have been transformed or transfected with nucleic acids encoding the variants. That is, Fc variants are not added exogenously to the cells. [212] The biological properties of the IgG variants may be characterized in cell, tissue, and whole organism experiments. As is known in the art, drugs are often tested in animals, including but not limited to mice, rats, rabbits, dogs, cats, pigs, and monkeys, in order to measure a drug's efficacy for treatment against a disease or disease model, or to measure a drug's pharmacokinetics, toxicity, and other properties. The animals may be referred to as disease models. Therapeutics are often tested in mice, including but not limited to nude mice, SCID mice, xenograft mice, and transgenic mice (including knockins and knockouts). Such experimentation may provide meaningful data for determination of the potential of the protein to be used as a therapeutic. Any organism, preferably mammals, may be used for testing. For example because of their genetic similarity to humans, monkeys can be suitable therapeutic models, and thus may be used to test the efficacy, toxicity, pharmacokinetics, or other property of the IgGs. Tests of the in humans are ultimately required for approval as drugs, and thus of course these experiments are contemplated. Thus the IgGs may be tested in humans to determine their therapeutic efficacy, toxicity, immunogenicity, pharmacokinetics, and/or other clinical properties. [213] Methods of Using IgG Variants [214] The IgG variants may find use in a wide range of products. In one embodiment the IgG variant is a therapeutic, a diagnostic, or a research reagent, preferably a therapeutic. The IgG variant may find use in an antibody composition that is monoclonal or polyclonal. In a preferred embodiment, the IgG variants are used to kill target cells that bear the target antigen, for example cancer cells. In an alternate embodiment, the IgG variants are used to block, antagonize or agonize the target antigen, for example for antagonizing a cytokine or cytokine receptor. In an alternately preferred embodiment, the IgG variants are used to block, antagonize or agonize the target antigen and kill the target cells that bear the target antigen.
[215] The IgG variants may be used for various therapeutic purposes. In a preferred embodiment, an antibody comprising the IgG variant is administered to a patient to treat an antibody-related disorder. A "patient" for the purposes includes humans and other animals, preferably mammals and most preferably humans. By "antibody related disorder" or "antibody responsive disorder" or "condition" or "disease" herein are meant a disorder that may be ameliorated by the administration of a pharmaceutical composition comprising an IgG variant. Antibody related disorders include but are not limited to autoimmune diseases, immunological diseases, infectious diseases, inflammatory diseases, neurological diseases, and oncological and neoplastic diseases including cancer. By "cancer" and "cancerous" herein 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 (including liposarcoma), neuroendocrine tumors, mesothelioma, schwanoma, meningioma, adenocarcinoma, melanoma, and leukemia and lymphoid malignancies.
[216] In one embodiment, an IgG variant is the only therapeutically active agent administered to a patient. Alternatively, the IgG variant is administered in combination with one or more other therapeutic agents, including but not limited to cytotoxic agents, chemotherapeutic agents, cytokines, growth inhibitory agents, anti-hormonal agents, kinase inhibitors, anti-angiogenic agents, cardioprotectants, or other therapeutic agents. The IgG varariants may be administered concomitantly with one or more other therapeutic regimens. For example, an IgG variant may be administered to the patient along with chemotherapy, radiation therapy, or both chemotherapy and radiation therapy. In one embodiment, the IgG variant may be administered in conjunction with one or more antibodies, which may or may not be an IgG variant. In accordance with another embodiment, the IgG variant and one or more other anti-cancer therapies are employed to treat cancer cells ex vivo. It is contemplated that such ex vivo treatment may be useful in bone marrow transplantation and particularly, autologous bone marrow transplantation. It is of course contemplated that the IgG variants can be employed in combination with still other therapeutic techniques such as surgery.
[217] A variety of other therapeutic agents may find use for administration with the IgG variants. In one embodiment, the IgG is administered with an anti-angiogenic agent. By "anti-anqioqenic agent" as used herein is meant a compound that blocks, or interferes to some degree, the development of blood vessels. The anti-angiogenic factor may, for instance, be a small molecule or a protein, for example an antibody, Fc fusion, or cytokine, that binds to a growth factor or growth factor receptor involved in promoting angiogenesis. The preferred anti-angiogenic factor herein is an antibody that binds to Vascular Endothelial Growth Factor (VEGF). In an alternate embodiment, the IgG is administered with a therapeutic agent that induces or enhances adaptive immune response, for example an antibody that targets CTLA-4. In an alternate embodiment, the IgG is administered with a tyrosine kinase inhibitor. By "tyrosine kinase inhibitor" as used herein is meant a molecule that inhibits to some extent tyrosine kinase activity of a tyrosine kinase. In an alternate embodiment, the IgG variants are administered with a cytokine.
[218] Pharmaceutical compositions are contemplated wherein an IgG variant and one or more therapeutically active agents are formulated. Formulations of the IgG variants are prepared for storage by mixing the IgG having the desired degree of purity with optional pharmaceutically acceptable carriers, excipients or stabilizers (Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed., 1980, entirely incorporated by reference), in the form of lyophilized formulations or aqueous solutions. The formulations to be used for in vivo administration are preferably sterile. This is readily accomplished by filtration through sterile filtration membranes or other methods. The IgG variants and other therapeutically active agents disclosed herein may also be formulated as immunoliposomes, and/or entrapped in microcapsules.
[219] The concentration of the therapeutically active IgG variant in the formulation may vary from about 0.1 to 100% by weight. In a preferred embodiment, the concentration of the IgG is in the range of 0.003 to 1.0 molar. In order to treat a patient, a therapeutically effective dose of the IgG variant may be administered. By "therapeutically effective dose" herein is meant a dose that produces the effects for which it is administered. The exact dose will depend on the purpose of the treatment, and will be ascertainable by one skilled in the art using known techniques. Dosages may range from 0.01 to 100 mg/kg of body weight or greater, for example 0.01 , 0.1 , 1.0, 10, or 50 mg/kg of body weight, with 1 to 10mg/kg being preferred. As is known in the art, adjustments for protein degradation, systemic versus localized delivery, and rate of new protease synthesis, as well as the age, body weight, general health, sex, diet, time of administration, drug interaction and the severity of the condition may be necessary, and will be ascertainable with routine experimentation by those skilled in the art.
[220] Administration of the pharmaceutical composition comprising an IgG variant, preferably in the form of a sterile aqueous solution, may be done in a variety of ways, including, but not limited to, orally, subcutaneously, intravenously, parenterally, intranasally, intraotically, intraocularly, rectally, vaginally, transdermally, topically (e.g., gels, salves, lotions, creams, etc.), intraperitoneal^, intramuscularly, intrapulmonary (e.g., AERx® inhalable technology commercially available from Aradigm, or Inhance® pulmonary delivery system commercially available from Nektar Therapeutics, etc.). Therapeutic described herein may be administered with other therapeutics concomitantly, i.e., the therapeutics described herein may be co-administered with other therapies or therapeutics, including for example, small molecules, other biologicals, radiation therapy, surgery, etc.
EXAMPLES
[221] Examples are provided below to illustrate the present invention. These examples are not meant to constrain the present invention to any particular application or theory of operation. For all positions discussed in the present invention, numbering is according to the EU index as in Kabat (Kabat et al., 1991 , Sequences of Proteins of Immunological Interest, 5th Ed., United States Public Health Service, National Institutes of Health, Bethesda, entirely incorporated by reference). Those skilled in the art of antibodies will appreciate that this convention consists of nonsequential numbering in specific regions of an immunoglobulin sequence, enabling a normalized reference to conserved positions in immunoglobulin families. Accordingly, the positions of any given immunoglobulin as defined by the EU index will not necessarily correspond to its sequential sequence.
EXAMPLE 1 : DNA construction, expression, and purification of Fc variants [222] Fc variants were constructed using the human IgGI Fc domain and the variable domain of trastuzumab (Herceptin®, Genentech). The Fc polypeptides were part of Alemtuzumab, An anti-HER2 antibody or AC10. Alemtuzumab (Campath®, a registered trademark of Millenium) is a humanized monoclonal antibody currently approved for treatment of B-cell chronic lymphocytic leukemia (Hale et al., 1990, Tissue Antigens 35:1 18- 127, entirely incorporated by reference). Trastuzumab (Herceptin®, a registered trademark of Genentech) is an anti-HER2/neu antibody for treatment of metastatic breast cancer. The heavy and light chain sequences of the anti-HER2 antibody are shown in Figure 22. AC10 is an anti-CD30 monoclonal antibody. The Herceptin variable region was assembled using recursive PCR. This variable region was then cloned with human IgGI into the pcDNA3.1/Zeo(+) vector (Invitrogen), shown in Figure 1 1. Plasmids were propagated in One Shot TOP10 E. coli cells (Invitrogen) and purified using the Hi-Speed Plasmid Maxi Kit (Qiagen). Plasmids were sequenced to verify the presence of the cloned inserts. [223] Site-directed mutagenesis was done using the Quikchange™ method (Stratagene). Plasmids containing the desired substitutions, insertions, and deletions were propagated in One Shot TOP10 E. coli cells (Invitrogen) and purified using the Hi-Speed Plasmid Maxi Kit (Qiagen). DNA was sequenced to confirm the fidelity of the sequences. [224] Plasmids containing heavy chain gene (VH-Cγ1-Cγ2-Cγ3) (wild-type or variants) were co-transfected with plasmid containing light chain gene (VL-CK) into 293T cells. Media were harvested 5 days after transfection, and antibodies were purified from the supernatant using protein A affinity chromatography (Pierce). Protein A binding characteristics of some modified Fc's are shown in Figure 26. Antibody concentrations were determined by bicinchoninic acid (BCA) assay (Pierce).
EXAMPLE 2: Binding affinity measurements
[225] Binding of Fc polypeptides to Fc ligands was assayed with surface plasmon resonance measurements. Surface plasmon resonance (SPR) measurements were performed using a BIAcore 3000 instrument (BIAcore AB). Wild-type or variant antibody was captured using immobilized protein L (Pierce Biotechnology, Rockford, IL), and binding to receptor analyte was measured. Protein L was covalently coupled to a CM5 sensor chip at a concentration of 1 uM in 10 mM sodium acetate, pH 4.5 on a CM5 sensor chip using N- hydroxysuccinimide/N-ethyl-N'-(-3-dimethylamino-propyl) carbodiimide (NHS/EDC) at a flow rate of 5 ul/min. Flow cell 1 of every sensor chip was mocked with NHS/EDC as a negative control of binding. Running buffer was 0.01 M HEPES pH 7.4, 0.15 M NaCI, 3 mM EDTA, 0.005% v/v Surfactant P20 (HBS-EP, Biacore, Uppsala, Sweden), and chip regeneration buffer was 10 mM glycine-HCI pH 1.5. 125 nM Wild-type or variant anti-HER2 antibody was bound to the protein L CM5 chip in HBS-EP at 1 ul/min for 5 minutes. FcRn-His-GST analyte, a FcRn fused to a His-tag and glutathione S transferase, in serial dilutions between 1 and 250 nM, were injected for 20 minutes association, 10 minutes dissociation, in HBS-EP at 10 ul/min. Response, measured in resonance units (RU), was acquired at 1200 seconds after receptor injection, reflecting near steady state binding. A cycle with antibody and buffer only provided baseline response. RU versus 1 / log concentration plots were generated and fit to a sigmoidal dose response using nonlinear regression with GraphPad Prism. [226] Binding of Fc polypeptides to Fc ligands was also done with AlphaScreen™ (Amplified Luminescent Proximity Homogeneous Assay). AlphaScreen™ is a bead-based non-radioactive luminescent proximity assay. Laser excitation of a donor bead excites oxygen, which if sufficiently close to the acceptor bead will generate a cascade of chemiluminescent events, ultimately leading to fluorescence emission at 520-620 nm. The principal advantage of the AlphaScreen™ is its sensitivity. Because one donor bead emits up to 60,000 excited oxygen molecules per second, signal amplification is extremely high, allowing detection down to attomolar (10"18) levels. Wild-type antibody was biotinylated by standard methods for attachment to streptavidin donor beads, and tagged Fc ligand, for example FcRn, FcgammaR or Protein A, was bound to glutathione chelate acceptor beads. The AlphaScreen™ was applied as a direct binding assay in which the Fc/Fc ligand interactions bring together the donor and acceptor beads to create the measured signal. Addtionally, the AlphaScreen™ was applied as a competition assay for screening designed Fc polypeptides. In the absence of competing Fc polypeptides, wild-type antibody and FcRn interact and produce a signal at 520-620 nm. Untagged Fc domains compete with wild-type Fc/FcRn interaction, reducing fluorescence quantitatively to enable determination of relative binding affinities.
EXAMPLE 3: FcRn-binding properties of Fc variants.
[227] Binding affinity of IgGI Fc to FcRn was measured with variant antibodies using AlphaScreen™. The Fc polypeptides were part of Alemtuzumab or Trastuzumab. Alemtuzumab (Campath®, Ilex) is a humanized monoclonal antibody currently approved for treatment of B-cell chronic lymphocytic leukemia (Hale et al., 1990, Tissue Antigens 35:1 18- 127, entirely incorporated by reference). Trastuzumab (Herceptin®, Genentech) is an anti- HER2/neu antibody for treatment of metastatic breast cancer.
[228] Competitive AlphaScreen™ data were collected to measure the relative binding of the Fc variants compared to the wild-type antibody in 0.1 M sodium phosphate pH6.0 with 25mM sodium chloride. Examples of the AlphaScreen™ signal as a function of competitor antibody are shown in Figure 12. The 12 variant curves shown, those of P257L, P257N, V279E, V279Q, V279Y, Λ281S, E283F, V284E, L306Y, T307V, V308F, and Q311V, demonstrate increased affinity as each variant curve is shifted to the left of the wild-type curve in their box. Competition AphaScreen™ data for Fc variants of the present invention are summarized in Figures 13 and 14. Additional competition AlphaScreen™ data in 0.1 M sodium phosphate pH 6.0 with 125mM sodium chloride are summarized in Figure 21. The relative FcRn binding of the variant compared to wild type are listed. Values greater than one demonstrate improved binding of the Fc variant to FcRn compared to the wild type. For example, the variant E283L and V284E have 9.5-fold and 26-fold stronger binding than the wild type, respectively. Surface plasmon resonance measurements of many variants also show increased binding to FcRn as shown in Figure 15 and 16.
[229] At position 257, all variants that remove the imino acid, proline, and substitute an amino acid without the backbone N to side chain covalent bond, allow the backbone more flexibility which allows more freedom for the Fc domain to better bind FcRn. In particular, variants at position 257 to L and N have strong FcRn binding at pH 6, demonstrating that the four atom side chain and gamma branching pattern of the side chain helps the Fc domain make productive, ie strong, FcRn interactions. Position 308 interacts with position 257. Both of these positions in turn interact with H310, which is directly involved in the Fc/FcRn interactions (Table 2, Burmeister et al (1994) Nature 372:379-383, entirely incorporated by reference). The Fc variants V308F and V08Y have a 2.9-fold and 4.3-fold increase in FcRn affinity over wild type (Figure 13). Positions 279 and 385 interact with FcRn as variants V279E, V279Q and V279Y and G385H and G385N all have stronger FcRn interactions. These variants all are to amino acids that are capable of hydrogen bonding. Sequences of the Fc regions of human IgGI comprising various modifications of the present invention are shown in Figure 23.
[230] The Fc variant N434Y has particularly strong binding to FcRn at pH 6.0 as shown in figure 13. The single variant N434Y has 16-fold increased binding. Combinations of this variant with other modifications led to even stronger binding. For example, P257L/N434Y, Λ281 S/N434Y, and V308F/N434Y show 830-fold, 180-fold, and 350-fold increases in FcRn binding.
EXAMPLE 4: Variants incorporating insertions and deletions. [231] Insertions and deletions that alter the strength of Fc/FcRn interactions were constructed and their binding properties to various Fc ligands were measured. An Fc variant with an inserted Ser residue between residues 281 and 282, using the EU numbering of Kabat et al, was designed to increase the FcRn binding properties of the Fc domain. This variant is referred to as Λ281S with "Λ" meaning an insertion following the position given. AlphaScreen™ data showing the improved binding of Λ281 S is shown in Figure 12b and 21 a. The inserted sequence, which may be more than one residue, is given after the position number. This Fc variant was constructed in the kappa, IgGI anti-HER2 antibody trastuzumab (Herceptin®, Genetech) using methods disclosed herein. An insertion at the site between residues 281 and 282 shifts the Fc loop residues C-terminal of residue 281 toward the C-terminus of the loop and alters the side chain positioning. Fc variants comprising substitutions at positions 282, 283, and 284 suggested that the C-terminal shift of this loop was beneficial (See Figure 14). Another variant, a deletion of N286, sometimes referred to as N286#, was also constructed to shift the position of this FcRn-binding loop. This variant shows increased binding to FcRn at pH6.0 (Figure 14b). [232] The AlphaScreen™ data shows the binding of the Λ281S variant and other variants to FcRn. This AlphaScreen™ data was collected as a direct binding assay. Higher levels of chemiluminescent signals demonstrate stronger binding. As the concentrations of the variants are raised in the assay, stronger signals are created. These data at pH 6.0, in Figures 17a and 17b, demonstrate the increased affinity of Λ281 S, P257L, P257L/Λ281 S (a combination substitution/insertion variant) and other variants over the wild-type Fc. Also shown is a double substitution, T250Q/M428L, shown previously to have an increased serum half in monkeys (Hinton et al., 2004, J. Biol. Chem. 279(8): 6213-6216, entirely incorporated by reference). The insertion, Λ281S, alone increases the Fc/FcRn binding. Additionally, Λ281 S further increases the binding of P257L when the two modifications are combined in the variant P257L/Λ281 S as shown in the ~40nM data points. The data in figure 17c demonstrate that these variants do not show increased FcRn binding at pH 7.0. The reduced affinity at pH 7.0 is desired for increased half-life in vivo, because it allows the release of Fc polypeptides from FcRn into the extracellular space, an important step in Fc recycling.
[233] Surface plasmon resonance experiments also demonstrate the improved binding of Λ281 S to FcRn. Figure 18 shows the response units created as various Fc variant binding to FcRn on the chip surface. After allowing the variant to fully bind to the chip, the response units are recorded and shown on the ordinate. The insertion, Λ281 S shows binding properties comparable to other variants shown herein to have increased affinity for FcRn over the wild type (See figures 13, 14 and 15, for examples).
[234] The deletion variant comprising a deletion of N286, N286#, also shows increased affinity for FcRn over wild type. This variant has a 2.0-fold increase in FcRn affinity as shown in Figure 13. The data therein are also AlphaScreen™ data collected as a competition experiment at pH 6.0. The variants are used to inhibit the binding of wild-type Fc, linked to the donor bead, with FcRn, linked to the acceptor beads. Two-fold less free N286# was needed than free wild-type Fc to inhibit the binding of the donor/acceptor beads through the Fc/FcRn complex. This demonstrates the 2-fold tighter binding of N286# over the wild type. [235] Other Fc variants comprising insertions or deletions have decreased affinity for FcRn. The insertion variant, Λ254N has greatly decreased FcRn binding as would be expected from the nature and positioning of the variant. This variant places the insertion, an Asn, in the middle of an FcRn binding loop. This insertion has only 1.1 % of the binding of the binding affinity of the wild type (Figure 13).
EXAMPLE 5: Combination Variants with altered FcRn and FcgammaR characteristics. [236] As shown in Figure 13b for the anti-HER2 antibody, the Fc variant P257L has increased affinity for FcRn relative to WT. P257L gave a median of 2.6-fold increase in FcRn affinity for human FcRn, pH 6.0 in phosphate buffer with 25mM NaCI added. The addition of I332E or S239D/I332E to the P257L variant yielded double and triple variants, P257L/I332E and S239D/P257L/I332E, which retain the increased affinity for FcRn. The variant S239D/I332E has essentially un-altered FcRn binding compared to wild type as shown in the AlphaScreen™ assays in figure 14b. These double and triple variants had a 5- and 4-fold increased affinity. The I332E and S239D/I332E variants have improved binding to FcgammaR, in particular to FcgammaRllla (See US1 1/124620, entirely incorporated by reference). The FcgammaR-binding properties of some variants of the present invention are shown in Figure 25. The protein A binding properties of some variant of the present invention are shown in Figure 26. Protein A binding is frequently used during purification of Fc- containing proteins. The substitution V308F also improves FcRn binding at pH 6.0 (Figure 13e). V308F has 3-fold increased affinity as a single substitution in the anti-HER2 antibody trastuzumab (Herceptin™, Genentech) and also has increased affinity when combined with substitutions that increase FcgammaR binding, such as I332E, S239D/I332E, and S298A/E333A/K334A (Lazar et al. 2006 Proc. Nat. Acad. Sci USA. 103(11 1 ):4005-4010, Shields et al. 2001 J. Biol. Chem. 276:6591-6604, both entirely incorporated by reference.) The increased FcRn binding of G385H is also maintained when combined with FcgammaR improving substitutions, especially in the triple-substitution variant S239D/I332E/G385H. [237] Variants with increased binding to FcRn may be combined with variants that reduce or knock-out binding to FcgammaR and the complement protein, C1 q. The improved binding to FcRn increases the effect from a protecting receptor allowing for improved half-life. Fc containing proteins may also be taken into cells and metabolized through their interaction with the FcgammaR and the C1q protein. If the Fc/FcgammaR and Fc/C1q protein interactions are not required for antibody efficacy, deletions of these interactions may be made. Deletions of these interactions may also decrease the effect of a degrading receptor, thereby also allowing for improved half-life. In particular the variants 234G, 235G, 236R, 237K, 267R, 269R, 325A, 325L, and 328R (US1 1/396,495 entirely incorporated by reference) may be combined with FcRn-improving variants to create variants with increased FcRn affinity and decreased FcgammaR or C1 q affinity. These variants include 235G/257C, 325A/385H, 325A/257L, 234G/308F, 234G/434Y, and 269R/308F/311V. These variants may be made in Fc domains from IgGI , although reduced interactions with the FcgammaR or C1 q may also be achieved by placing these mutations into proteins comprising Fc domains from lgG2, lgG4, or lgG3. Putting FcRn modifications, such as 257N, 257L, 257M, 308F, 31 1V into lgG2 allows for a reduction in FcgammaR binding and increased FcRn interactions.
[238] Variants with decreased binding to FcRn may be combined with variants that have increased FcgammaR or C1q binding. The decreased FcRn binding combined with increased FcgammaR binding may be beneficial for increasing the amount of the Fc- containing protein available to illicit effector functions. Reducing FcRn binding may reduce the amount of the Fc-containing protein that is sequestered by FcRn and thus affect bioavailability. Modifications such as I253V, S254N, S254# (deletion of 254), T255H, and H435N reduce Fc/FcRn binding (Figure 13) and may be combined with variants with improved FcgammaR binding such as S239D, I332E, H268E, G236A. The resulting Fc domains, such as those comprising I253V/S239D/I332E, I332E/H435N, or S254N/H268E, have reduced FcRn binding and increased FcgammaR binding. [239] Variants with decreased binding to FcRn may be combined with variants with decreased FcgammaR binding. This combination of decreased FcRn and FcgammaR binding is beneficial in applications such as imaging wherein the Fc-containing protein is labeled with a radioactive or toxic tracer. Ideally the half-life of the protein comprising the radioactive tracer is similar to the half-life of the radionuclide itself. This allows clearance of the tracer from the body in the same time as the decay of the radionuclide. The reduced FcgammaR interactions also allow optimal availability of the Fc-containing protein for its target. For example, if the Fc-containing protein is an antibody, then the reduce FcgammaR binding allow more antibody to be assessable to antigen. Combinations of FcRn- and FcgammaR-affecting variants, such as 235G/254N, 236R/435N, 269R/I253V are good for this application.
EXAMPLE 6: Fc variants in antibody OST577 binding to human FcRn. [240] OST577 is an anti-Hepatitis B surface antigen antibody (Ehrlich et al. (1992) Hum. Antibodies Hybridomas 3:2-7, entirely incorporated by reference). Heavy and light chain sequences were taken from the Kabat Database with KADBID 000653 (heavy) and KADBID 007557(light) (Martin AC, Proteins. 1996 May;25(1 ):130-3, entirely incorporated by reference). DNA encoding the heavy and light chains were synthesized by Blue Heron Biolotechnology, Bothell, WA. Wild-type and variant OST577 antibodies were expressed and purified as in the anti-HER2 (trastuzumab) variants in EXAMPLE 1. Biacore™ binding assays were performed as in EXAMPLE 2, with a human FcRn/Glutathione D transferase (GST) fusion protein attached to the chip surface. As shown in Figure 19, Fc variants of the present invention have altered binding to human FcRn. Variants with increased binding adhere more easily to the FcRn on the surface and cause a greater rise in Response Units (RU's). The variants shown with modification in the FcRn-binding region all have increased affinity for FcRn compared to the wild-type protein. These variants include P257L, P257N, V308F, N434Y, P257L/N434Y and P257L/V308F. The variant with the 3rd most RU's at 975 seconds, T250Q/M428L, has been shown to increase the half life of OST577 antibodies in macaques (Hinton et al. 2004 Journal of Biological Chemistry 279(8):6213-6216, Hinton et al. 2006 Jounal of Immunology 176:346-356, both entirely incorporated by reference). Included in this data set is an antibody with a hybrid lgG1/lgG2 heavy chain constant region containing the substitutions S239D/I332E. As described in EXAMPLE 5, these substitutions increase the antibody affinity for FcgammaR. As shown in Figure 19, these substitutions do not alter the FcRn-binding properties, as the hybrid S239D/I332E Biacore™ traces overlay the wild-type traces containing kappa or lambda CL1 domains.
EXAMPLE 7: Affinity of Fc variants for human, monkey and mouse FcRn. [241] Fc variants in the anti-HER2 antibody trastuzumab were created as described in EXAMPLE 1. Surface plasmon resonance (SPR) traces were collected as described in EXAMPLE 2, except that human, macaque or mouse FcRn was attached to the chip surface. Two SPR curves were collected for each Fc variant with differing amounts of GST-FcRn attached to the surface. Each curve was fit to a 1 :1 Langmuir binding model and the two resulting Kd values were averaged to produce a representative value for each variant- receptor pair. The results are presented in Figure 20 as the fold-improvement in Kd compared to the wild-type trastuzumab. For example, the variant V308F/Q311 V has 3.4-fold tigher binding to human FcRn than does the wild type. V308F/Q311 V also has 3.7-fold and 5.1 -fold tigher binding to monkey and mouse FcRn, respectively. The variant M428L has been shown to increase the antibody half-life (Hinton et al. 2004 Journal of Biological Chemistry 279(8):6213-6216, entirely incorporated by reference) and has a 2.4-, 2.0, and 2.1 -fold increased binding to the human, monkey and mouse FcRn's, respectively. Other variants, including P257L, P257N, N434Y, Q31 1V, V308F, V308F/N434Y, P257L/V308F, and P257L/N434Y, also show increased binding at pH6.0.
EXAMPLE 8: FcRn variants in various Fc domains.
[242] Variants of the present invention may be incorporated into any constant domain, using the molecular biology and purification techniques described herein, including those in EXAMPLE 1. Amino acid sequences of the IgGI , lgG2, lgG3, and lgG4 constant domains may be used as listed in Figure 2. In addition, combinations of two or more different constant domains may be used. For example, Figure 24 lists some of the modifications found in the present invention incorporated into a hybrid of IgGI and lgG2. This hybrid comprises the lgG2 CH1 domain and the IgGI CH2 and CH3 domains. lgG3 has a lower half-life in humans compared to IgGI , lgG2, and lgG4 (7 days vs -21 days, Janeway, Travers, Walport, Shlomchik. Immunology, 5th ed. Garland Publishing c2001 , Figure 4-16, incorporated by reference) and is therefore desirable in certain applications. EXAMPLE 9: Creation of variant in an anti-VEGF antibody.
[243] Anti-VEGF antibodies with altered binding were produced using the methods described herein, including EXAMPLE 1. The wild type anti-VEGF heavy chain comprises the following sequence of amino acids:
[244] EVQLVESGGGLVQPGGSLRLSCAASGYTFTNYGMNWVRQAPGKGLEWVGWINTY TGEPTYAADFKRRFTFSLDTSKSTAYLQMNSLRAEDTAVYYCAKYPHYYGSSHWYFDVWG QGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHT FPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCP APELLGGPSVFLFPPKPKDTLMISRTPEVTCVWDVSHEDPEVKFNWYVDGVEVHNAKTKP REEQYNSTYRWSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLP PSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVD KSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK [245] The wild-type anti-VEGF light chain comprises the following sequence of amino acids:
[246] DIQMTQSPSSLSASVGDRVTITCSASQDISNYLNWYQQKPGKAPKVLIYFTSSLHSG
VPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQYSTVPWTFGQGTKVEI KRTVAAPSVFI FP
PSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSKDSTYSLSSTL
TLSKADYEKHKVYACEVTHQGLSSPVTKSFNRGEC
[247] Single and combination variants with altered binding include the variants shown in
Figure 28, which shows the variants produced, the volume of culture media used, and the resulting yield of the antibody variants. Numbering for the variants follows the EU index as in
Kabat et al. The variants listed in Figure 28 were produced in either IgGI or hybrid VH comprising sequences from both IgGI and lgG2. These variants contain the variable region that binds the antigen VEGF. All proteins were judged to be >90% pure by size exclusion chromatography and SDS gel electrophoresis.
EXAMPLE 10: In vivo half-life of antibody variants
[248] The pharmacokinetics of wild-type and variant antibodies were studied in mice. The mice used were defiecient in the expression of mouse FcRn (B6-FcgrtTm1Dcr mice) and were heterozygous for the knock-in of human FcRn (hFcRn Tg - transgene) as described in Petkova et al. International Immunology 2006 Dec;18(12):1759-69. Petkova et al showed that the variant N434A has an increased half-life in these human FcRn knock-in mice, which agrees with earilier results showing that the N434A variant has increased half life in monkeys (US Appl No. 1 1/208422, publication number US26067930A1 ). Female mice aged 9-12 weeks were injected intravenously with 2 mg/kg antibody in groups of 6 mice per antibody. Blood samples were collected at 1 hr, and days 1 , 4, 8, 11 , 15, 18, 21 , 25 and 28 from the oribital flexus. The concentration of each antiobody in serum was measured with a sandwich ELISA assay using anti-human Fc antibodies and europium detection. [249] The results of the study are shown in Figure 27, which are representative data of two separate studies. The mean and standard deviation of the mean for the four samples are shown. Clearly, the V308F variant has longer half-life, remaining at measurable concentrations out to 25 days. The WT and P257L and P257N variants are cleared more quickly, only having measurable concentrations out to 15, 8, and 4 days, respectively. The serum concentrations as a function of time were fit to a non-compartimental model using the software package, WinNonLin (Pharsight Inc). The terminal half-life of the V308F variant was 4.9 days, whereas the terminal half-lives of the WT and P257L and P257N variants were 3.0, 1.9 and 0.9 days, respectively. The area under the curves (AUC) of the V308F variant was 129 day*ug/ml, whereas those of the WT and P257L and P257N variants were 70, 38 and 38 day*ug/ml, respectively.
EXAMPLE 11 : FcRn binding experiments at pH 6.0. [250] Anti-VEGF variants of the present invention were tested for their binding ability to human FcRn with Biacore assays as described in EXAMPLE 2 with some modifications. Human FcRn was attached covalently to a CM5 chip in 10 mM sodium acetate, pH 4.5 on using N-hydroxysuccinimide/N-ethyl-N'-^S-dimethylamino-propyl) carbodiimide (NHS/EDC) at a flow rate of 5 ul/min. The human FcRn used contained GST and HIS tagged version to aid in purification and other assys. Approximately 3300 RU of FcRn was attached to the chip. Flow cell 1 was mocked with NHS/EDC as a negative control of binding. Running buffer was 25mM phosphate buffer pH6.0, 15OmM NaCI, 3mM EDTA and 0.005% (v/v) Surfactant P20. Antibodies were washed off the FcRn chip with the same buffer at pH7.4, which quickly removed all variants tested. The biacore association and dissociation traces were fit to a conformational exchange model to calculate an apparent equilibrium binding constant, Kd. [251] The results demonstrate that the V308F variant and many other variants have improved binding to FcRn. The wild-type anti-VEGF antibody had a Kd of 18 nM, which differs considerably from the value reported in DaII' Acqua et al (DaII' Acqua et al Journal of Immunology 2002, 169:5171-5180) because of the differences in assay design and data fitting. Our assay format gave reproducible results if the FcRn chip was used soon after creation. The FcRn chip, however, degraded with use, possibly from the dissociation of either the two FcRn chains from the surface. The results demonstrate the altered binding of the variants compared to wild-type anti-VEGF. Figure 29 shows the fold increase in binding strength relative to the wild-type control. Values greater than one show that the variant antibody has has higher affinity for FcRn than the wild-type protein. The variant V308F, for example, binds FcRn 4.5 fold more tightly than the wild-type antibody. The variant V308F/M428L binds FcRn 12.3 fold more tightly and the variant T307P/V308F binds FcRn 3.16 fold more tightly than the wild-type protein. No variants shown in Figure 29 have reduced affinity for FcRn compared to the wild-type (values would be less than 1.0). The variant N434S has an FcRn binding affinity 4.4 fold stronger than WT, comparable to V308F.
EXAMPLE 12: Binding experiments to transmembrane FcRn. [252] FcRn alpha chain and beta-2-microglobulin cDNA was ordered from OriGene Technologies lnc (Rockville, MD) and transfected in 293T cells to express functional FcRn on the cell surface. 20 ug Fcgrt and 40 ug of beta-2-microglobulin DNA was transfected with lipofectamine (Invitrogen Inc.) and the cells were allowed to grow for 3 days in DMEM media with 10% ultra low IgG serum. Control cells not transfected with the two FcRn chains were also grown. Varying amounts of anti-VEGF antibodies (WT and variants) were bound to the cells for 30 minutes in 25mM phosphate buffer pH6.0, 15OmM NaCI, 0.5% BSA and then washed 6-9 times in 25mM phosphate buffer pH6.0, 15OmM NaCI, 0.5% BSA plus 0.003% igepal. After washing, antibodies were fixed to the surface by treatment with the binding with 1% PFA. Bound antibodies were than detected using a PE tagged Fab'2 against human Fab domains and the mean fluorescence intensity (MFI) was measured using a BD FACS Canto II. The average of two samples per antibody are presented in Figure 30. The curve fits to the data in Figure 30 do not provide interpretable EC50 values because many curves did not form an upper baseline by saturating the cells. The antibodies may be ranked in order of their binding affinity, however, by reporting the log[variant] at which the MFI equals 3000, EC(MFI=3000). Using this metric, the antibodies may be listed from strongest to weakest FcRn affinity as follows: V308F/M428L, V259I/V308F, T250I/V308F, T250Q/M428L, N434S, T307Q/V308F, P257L, T307S/V308F, V308F, T256V/V308F, V308F/L309Y, and WT.
EXAMPLE 13: Characteristics of the variant, 434S.
[253] Antibodies comprising the modification 434S have particularly favorable properties making them preferred variants of the present invention. In human IgGI , the wild-type residue is an asparagine, Asn, at position 434 so that this variant may be referred to as N434S in the context of IgGI or other Fc domains which contain Asn, N, at position 434. More generally, this variant may be referred to simply as 434S. Herein, the 434S variant has been produced successfully in both the anti-HER2 antibody trastuzumab and the anti-VEGF antibody.
[254] The Ser at position 434 has the ability to hydrogen bond with FcRn either directly or indirectly, ie, mediated by water or solute molecules. The gamma oxygen of Ser at position 434 is in the vicinity of the carbonyl oxygen atoms of Gly131 and Pro134 on the FcRn molecule, as shown in Figure 32. Figure 32 shows a model of the human Fc domain in complex with human FcRn. The Fc domain in the model comprises the 434S substitution, hence residue 434 is Ser in Figure 32. The model is created with PDA® technologies (Dahiyat and Mayo Protein Sci. 1996 May;5(5):895-903), the crystal structure of the rat Fc domain bound to the rat FcRn (Martin et al. MoI Cell. 2001 Apr;7(4):867-77), and Pymol (Delano Scientific). The small size of Ser is easily accomidated in the interface between the two proteins.
[255] The antibody variant N434S has a 4.4-fold increased binding affinity for FcRn compared to the wild-type antibody as shown by biacore™ measurements (Figure 29). The variant also shows increased binding to cell surface bound FcRn as shown by cell counting measurements (Figure 30).
[256] Based on the results shown in Figures 29 and 30, preferred variants comprising 434S and other modifications are expected to include V308F/434S, 428L/434S, 252Y/434S, 259I/308F/434S, 250I/308F/434S, and 307Q/308F/434S.
EXAMPLE 14: Additional Variants
[257] Additional variants may be based on the data contained herein and in the literature (DaII' Acqua et al Journal of Biological Chemistry 2006 Aug 18;281 (33):23514-24; Petkova et al. International Immunity 2006 Dec;18(12):1759-69; DaII' Acqua et al Journal of Immunology 2002, 169:5171-5180; Hinton et al, Journal of Biological Chemistry 2004 279(8): 6213-6216; Shields et al. Journal of Biological Chemistry 2001 276(9):6591-6604; Hinton et al. Journal of Immunology 2006, 176:346-356, all incorporated by reference). These variants include those found in Figure 31.
[258] Based on the results in Figures 29 and 30 and the results of DaN' Acqua et al (Journal of Biological Chemistry 2006 Aug 18;281 (33):23514-24, incorporated by reference), preferred variants include Y319L, T307Q, V259I, M252Y, V259I/N434S, M428L/N434S, V308F/N434S, M252Y/S254T/T256E/N434S, M252Y/S254T/T256E/V308F, M252Y/S254T/T256E/M428L, V308F/M428L/N434S, V259I/V308F/N434S, T307Q/V308F/N434S, T250I/V308F/N434S, V308F/Y319L/N434S, V259I/V308F/M428L, V259I/T307Q/V308F, T250I/V259I/V308F, V259I/V308F/Y319L, T307Q/V308F/L309Y, T307Q/V308F/Y319L, and T250Q/V308F/M428L.
[259] Based on the results in Figures 29 and 30 more preferred variants include Y319L, T307Q, V259I, M252Y, V259I/N434S, M428L/N434S, V308F/N434S, V308F/M428L/N434S, V259I/V308F/N434S, T307Q/V308F/N434S, T250I/V308F/N434S, V308F/Y319L/N434S, V259I/V308F/M428L, V259I/T307Q/V308F, T250I/V259I/V308F, V259I/V308F/Y319L, T307Q/V308F/L309Y, T307Q/V308F/Y319L, and T250Q/V308F/M428L. [260] Whereas particular embodiments of the invention have been described above for purposes of illustration, it will be appreciated by those skilled in the art that numerous variations of the details may be made without departing from the invention as described in the appended claims. All references cited herein are incorporated in their entirety.
SEQUENCE LISTING
<110> CHAMBERLAIN, Aaron Keith DAHIYAT, Bassil I. DESJARLAIS, John Rudolph KARKI, Sher Bahadur
<120> Fc Variants with Altered Binding to FcRn
<130> 5026-US02
<140> 11/932151
<141> 2007-10-31
<150> 60/951,536
<151> 2007-07-24
<150> 11/436,266
<151> 2006-05-17
<150> 11/274,065
<151> 2005-11-14
<150> 60/627,763
<151> 2004-11-12
<150> 60/642,886
<151> 2005-01-11
<150> 60/649,508
<151> 2005-02-02
<150> 60/662,468
<151> 2005-03-15
<150> 60/681,607
<151> 2005-05-16
<150> 60/690,200
<151> 2005-06-13
<150> 60/696,609
<151> 2005-07-05
<150> 60/703,018
<151> 2005-07-27
<150> 60/726,453
<151> 2005-10-12
<160> 24
<170> Patentln version 3.5
<210> 1
71 <211> 227 <212> PRT <213> Homo sapiens
<400> 1
Asp Lys Thr His Thr Cys Pro Pro Cys Pro Ala Pro GIu Leu Leu GIy 1 5 10 15
GIy Pro Ser VaI Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met 20 25 30
He Ser Arg Thr Pro GIu VaI Thr Cys VaI VaI VaI Asp VaI Ser His 35 40 45
GIu Asp Pro GIu VaI Lys Phe Asn Trp Tyr VaI Asp GIy VaI GIu VaI 50 55 60
His Asn Ala Lys Thr Lys Pro Arg GIu GIu GIn Tyr Asn Ser Thr Tyr 65 70 75 80
Arg VaI VaI Ser VaI Leu Thr VaI Leu His GIn Asp Trp Leu Asn GIy 85 90 95
Lys GIu Tyr Lys Cys Lys VaI Ser Asn Lys Ala Leu Pro Ala Pro He 100 105 HO
GIu Lys Thr He Ser Lys Ala Lys GIy GIn Pro Arg GIu Pro GIn VaI 115 120 125
Tyr Thr Leu Pro Pro Ser Arg Asp GIu Leu Thr Lys Asn GIn VaI Ser 130 135 140
Leu Thr Cys Leu VaI Lys GIy Phe Tyr Pro Ser Asp He Ala VaI GIu 145 150 155 160
Trp GIu Ser Asn GIy GIn Pro GIu Asn Asn Tyr Lys Thr Thr Pro Pro 165 170 175
VaI Leu Asp Ser Asp GIy Ser Phe Phe Leu Tyr Ser Lys Leu Thr VaI 180 185 190
Asp Lys Ser Arg Trp GIn Gin GIy Asn VaI Phe Ser Cys Ser VaI Met
72 195 200 205
His GIu Ala Leu His Asn His Tyr Thr GIn Lys Ser Leu Ser Leu Ser 210 215 220
Pro GIy Lys 225
<210> 2
<211> 223
<212> PRT
<213> Homo sapiens
<400> 2
VaI GIu Cys Pro Pro Cys Pro Ala Pro Pro VaI Ala GIy Pro Ser VaI 1 5 10 15
Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met lie Ser Arg Thr 20 25 30
Pro GIu VaI Thr Cys VaI VaI VaI Asp VaI Ser His GIu Asp Pro GIu 35 40 45
VaI GIn Phe Asn Trp Tyr VaI Asp GIy VaI GIu VaI His Asn Ala Lys 50 55 60
Thr Lys Pro Arg GIu GIu Gin Phe Asn Ser Thr Phe Arg VaI VaI Ser 65 70 75 80
VaI Leu Thr VaI VaI His Gin Asp Trp Leu Asn GIy Lys GIu Tyr Lys 85 90 95
Cys Lys VaI Ser Asn Lys GIy Leu Pro Ala Pro lie GIu Lys Thr lie 100 105 110
Ser Lys Thr Lys GIy GIn Pro Arg GIu Pro GIn VaI Tyr Thr Leu Pro 115 120 125
Pro Ser Arg GIu GIu Met Thr Lys Asn GIn VaI Ser Leu Thr Cys Leu 130 135 140
VaI Lys GIy Phe Tyr Pro Ser Asp lie Ala VaI GIu Trp GIu Ser Asn
73 145 150 155 160
GIy GIn Pro GIu Asn Asn Tyr Lys Thr Thr Pro Pro Met Leu Asp Ser 165 170 175
Asp GIy Ser Phe Phe Leu Tyr Ser Lys Leu Thr VaI Asp Lys Ser Arg 180 185 190
Trp GIn GIn GIy Asn VaI Phe Ser Cys Ser VaI Met His GIu Ala Leu 195 200 205
His Asn His Tyr Thr GIn Lys Ser Leu Ser Leu Ser Pro GIy Lys 210 215 220
<210> 3
<211> 229
<212> PRT
<213> Homo sapiens
<400> 3
Leu GIy Asp Thr Thr His Thr Cys Pro Arg Cys Pro Ala Pro GIu Leu 1 5 10 15
Leu GIy GIy Pro Ser VaI Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr 20 25 30
Leu Met He Ser Arg Thr Pro GIu VaI Thr Cys VaI VaI VaI Asp VaI 35 40 45
Ser His GIu Asp Pro GIu VaI GIn Phe Lys Trp Tyr VaI Asp GIy VaI 50 55 60
GIu VaI His Asn Ala Lys Thr Lys Pro Arg GIu GIu GIn Tyr Asn Ser 65 70 75 80
Thr Phe Arg VaI VaI Ser VaI Leu Thr VaI Leu His GIn Asp Trp Leu 85 90 95
Asn GIy Lys GIu Tyr Lys Cys Lys VaI Ser Asn Lys Ala Leu Pro Ala 100 105 HO
Pro He GIu Lys Thr He Ser Lys Thr Lys GIy GIn Pro Arg GIu Pro
74 115 120 125
GIn VaI Tyr Thr Leu Pro Pro Ser Arg GIu GIu Met Thr Lys Asn GIn 130 135 140
VaI Ser Leu Thr Cys Leu VaI Lys GIy Phe Tyr Pro Ser Asp lie Ala 145 150 155 160
VaI GIu Trp GIu Ser Ser GIy GIn Pro GIu Asn Asn Tyr Asn Thr Thr 165 170 175
Pro Pro Met Leu Asp Ser Asp GIy Ser Phe Phe Leu Tyr Ser Lys Leu 180 185 190
Thr VaI Asp Lys Ser Arg Trp GIn GIn GIy Asn lie Phe Ser Cys Ser 195 200 205
VaI Met His GIu Ala Leu His Asn Arg Phe Thr GIn Lys Ser Leu Ser 210 215 220
Leu Ser Pro GIy Lys 225
<210> 4
<211> 224
<212> PRT
<213> Homo sapiens
<400> 4
Pro Pro Cys Pro Ser Cys Pro Ala Pro GIu Phe Leu GIy GIy Pro Ser 1 5 10 15
VaI Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met lie Ser Arg 20 25 30
Thr Pro GIu VaI Thr Cys VaI VaI VaI Asp VaI Ser GIn GIu Asp Pro 35 40 45
GIu VaI GIn Phe Asn Trp Tyr VaI Asp GIy VaI GIu VaI His Asn Ala 50 55 60
Lys Thr Lys Pro Arg GIu GIu GIn Phe Asn Ser Thr Tyr Arg VaI VaI
75 65 70 75 80
Ser VaI Leu Thr VaI Leu His GIn Asp Trp Leu Asn GIy Lys GIu Tyr 85 90 95
Lys Cys Lys VaI Ser Asn Lys GIy Leu Pro Ser Ser lie GIu Lys Thr 100 105 110
lie Ser Lys Ala Lys GIy Gin Pro Arg GIu Pro GIn VaI Tyr Thr Leu 115 120 125
Pro Pro Ser GIn GIu GIu Met Thr Lys Asn GIn VaI Ser Leu Thr Cys 130 135 140
Leu VaI Lys GIy Phe Tyr Pro Ser Asp lie Ala VaI GIu Trp GIu Ser 145 150 155 160
Asn GIy GIn Pro GIu Asn Asn Tyr Lys Thr Thr Pro Pro VaI Leu Asp 165 170 175
Ser Asp GIy Ser Phe Phe Leu Tyr Ser Arg Leu Thr VaI Asp Lys Ser 180 185 190
Arg Trp GIn GIu GIy Asn VaI Phe Ser Cys Ser VaI Met His GIu Ala 195 200 205
Leu His Asn His Tyr Thr Gin Lys Ser Leu Ser Leu Ser Leu GIy Lys 210 215 220
<210> 5
<211> 207
<212> PRT
<213> Homo sapiens
<400> 5
GIy Pro Ser VaI Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met 1 5 10 15
lie Ser Arg Thr Pro GIu VaI Thr Cys VaI VaI VaI Asp VaI Ser His 20 25 30
GIu Asn Pro GIu VaI Lys Phe Asn Trp Tyr VaI Asp GIy VaI GIu VaI
76 35 40 45
His Asn Ala Lys Thr Lys Pro Arg GIu GIu GIn Tyr Asn Ser Thr Tyr 50 55 60
Arg VaI VaI Ser VaI Leu Thr VaI Leu His GIn Asp Trp Leu Asn GIy 65 70 75 80
Lys GIu Tyr Lys Cys Lys VaI Ser Asn Lys Ala Leu Pro Ala Pro lie 85 90 95
GIu Lys Thr lie Ser Lys Ala Lys GIy GIn Pro Arg GIu Pro GIn VaI 100 105 110
Tyr Thr Leu Pro Pro Ser Arg GIu GIu Met Thr Lys Asn GIn VaI Ser 115 120 125
Leu Thr Cys Leu VaI Lys GIy Phe Tyr Pro Ser Asp lie Ala VaI GIu 130 135 140
Trp GIu Ser Asn GIy GIn Pro GIu Asn Asn Tyr Lys Thr Thr Pro Pro 145 150 155 160
VaI Leu Asp Ser Asp GIy Ser Phe Phe Leu Tyr Ser Lys Leu Thr VaI 165 170 175
Asp Lys Ser Arg Trp GIn GIn GIy Asn VaI Phe Ser Cys Ser VaI Met 180 185 190
His GIu Ala Leu His Asn His Tyr Thr GIn Lys Ser Leu Ser Leu 195 200 205
<210> 6
<211> 205
<212> PRT
<213> Rattus norvegicus
<400> 6
Ser VaI Phe lie Phe Pro Pro Lys Thr Lys Asp VaI Leu Thr lie Thr 1 5 10 15
Leu Thr Pro Lys VaI Thr Cys VaI VaI VaI Asp lie Ser GIn Asn Asp
77 20 25 30
Pro GIu VaI Arg Phe Ser Trp Phe lie Asp Asp VaI GIu VaI His Thr 35 40 45
Ala GIn Thr His Ala Pro GIu Lys GIn Ser Asn Ser Thr Leu Arg Ser 50 55 60
VaI Ser GIu Leu Pro lie VaI His Arg Asp Trp Leu Asn GIy Lys Thr 65 70 75 80
Phe Lys Cys Lys VaI Asn Ser GIy Ala Phe Pro Ala Pro lie GIu Lys 85 90 95
Ser lie Ser Lys Pro GIu GIy Thr Pro Arg GIy Pro GIn VaI Tyr Thr 100 105 110
Met Ala Pro Pro Lys GIu GIu Met Thr GIn Ser GIn VaI Ser lie Thr 115 120 125
Cys Met VaI Lys GIy Phe Tyr Pro Pro Asp lie Tyr Thr GIu Trp Lys 130 135 140
Met Asn GIy GIn Pro GIn GIu Asn Tyr Lys Asn Thr Pro Pro Thr Met 145 150 155 160
Asp Thr Asp GIy Ser Tyr Phe Leu Tyr Ser Lys Leu Asn VaI Lys Lys 165 170 175
GIu Thr Trp GIn GIn GIy Asn Thr Phe Thr Cys Ser VaI Leu His GIu 180 185 190
GIy Leu His Asn His His Thr GIu Lys Ser Leu Ser His 195 200 205
<210> 7
<211> 256
<212> PRT
<213> Homo sapiens
<400> 7
His Leu Ser Leu Leu Tyr His Leu Thr Ala VaI Ser Ser Pro Ala Pro
78 10 15
GIy Thr Pro Ala Phe Trp VaI Ser GIy Trp Leu GIy Pro GIn GIn Tyr 20 25 30
Leu Ser Tyr Asn Ser Leu Arg GIy GIu Ala GIu Pro Cys GIy Ala Trp 35 40 45
Tyr Trp GIu Lys GIu Thr Thr Asp Leu Arg lie Lys GIu Lys Leu Phe 50 55 60
Leu GIu Ala Phe Lys Ala Leu GIy GIy Lys GIy Pro Tyr Thr Leu GIn 65 70 75 80
GIy Leu Leu GIy Cys GIu Leu GIy Pro Asp Asn Thr Ser VaI Pro Thr 85 90 95
Ala Lys Phe Ala Leu Asn GIy GIu GIu Phe Met Asn Phe Asp Leu Lys 100 105 110
GIn GIy Thr Trp GIy GIy Asp Trp Pro GIu Ala Leu Ala lie Ser GIn 115 120 125
Arg Trp GIn GIn GIn Asp Lys Ala Ala Asn Lys GIu Leu Thr Phe Leu 130 135 140
Leu Phe Ser Cys Pro His Arg Leu Arg GIu His Leu GIu Arg GIy Arg 145 150 155 160
GIy Asn Leu GIu Trp Lys GIu Pro Pro Ser Met Arg Leu Lys Ala Arg 165 170 175
Pro Ser Ser Pro GIy Phe Ser VaI Leu Thr Cys Ser Ala Phe Ser Phe 180 185 190
Tyr Pro Pro GIu Leu GIn Leu Arg Phe Leu Arg Asn GIy Leu Ala Ala 195 200 205
GIy Thr GIy GIn GIy Asp Phe GIy Pro Asn Ser Asp GIy Ser Phe His 210 215 220
79 Ala Ser Ser Ser Leu Thr VaI Lys Ser GIy Asp GIu His His Tyr Cys 225 230 235 240
Cys lie VaI GIn His Ala GIy Leu Ala GIn Pro Leu Arg VaI GIu Leu 245 250 255
<210> 8
<211> 265
<212> PRT
<213> Rattus norvegicus
<400> 8
Leu Pro Leu Met Tyr His Leu Ala Ala VaI Ser Asp Leu Ser Thr GIy 1 5 10 15
Leu Pro Ser Phe Trp Ala Thr GIy Trp Leu GIy Ala GIn GIn Tyr Leu 20 25 30
Thr Tyr Asn Asn Leu Arg GIn GIu Ala Asp Pro Cys GIy Ala Trp lie 35 40 45
Trp GIu Asn GIn VaI Ser Trp Tyr Trp GIu Lys GIu Thr Thr Asp Leu 50 55 60
Lys Ser Lys GIu GIn Leu Phe Leu GIu Ala lie Arg Thr Leu GIu Asn 65 70 75 80
GIn lie Asn GIy Thr Phe Thr Leu GIn GIy Leu Leu GIy Cys GIu Leu 85 90 95
Ala Pro Asp Asn Ser Ser Leu Pro Thr Ala VaI Phe Ala Leu Asn GIy 100 105 110
GIu GIu Phe Met Arg Phe Asn Pro Arg Thr GIy Asn Trp Ser GIy GIu 115 120 125
Trp Pro GIu Thr Asp lie VaI GIy Asn Leu Trp Met Lys GIn Pro GIu 130 135 140
Ala Ala Arg Lys GIu Ser GIu Phe Leu Leu Thr Ser Cys Pro GIu Arg 145 150 155 160
80 Leu Leu GIy His Leu GIu Arg GIy Arg GIn Asn Leu GIu Trp Lys GIu 165 170 175
Pro Pro Ser Met Arg Leu Lys Ala Arg Pro GIy Asn Ser GIy Ser Ser 180 185 190
VaI Leu Thr Cys Ala Ala Phe Ser Phe Tyr Pro Pro GIu Leu Lys Phe 195 200 205
Arg Phe Leu Arg Asn GIy Leu Ala Ser GIy Ser GIy Asn Cys Ser Thr 210 215 220
GIy Pro Asn GIy Asp GIy Ser Phe His Ala Trp Ser Leu Leu GIu VaI 225 230 235 240
Lys Arg GIy Asp GIu His His Tyr GIn Cys GIn VaI GIu His GIu GIy 245 250 255
Leu Ala GIn Pro Leu Thr VaI Asp Leu 260 265
<210> 9
<211> 99
<212> PRT
<213> Homo sapiens <400> 9 lie GIn Arg Thr Pro Lys lie GIn VaI Tyr Ser Arg His Pro Ala GIu 1 5 10 15
Asn GIy Lys Ser Asn Phe Leu Asn Cys Tyr VaI Ser GIy Phe His Pro 20 25 30
Ser Asp lie GIu VaI Asp Leu Leu Lys Asn GIy GIu Arg lie GIu Lys 35 40 45
VaI GIu His Ser Asp Leu Ser Phe Ser Lys Asp Trp Ser Phe Tyr Leu 50 55 60
Leu Tyr Tyr Thr GIu Phe Thr Pro Thr GIu Lys Asp GIu Tyr Ala Cys 65 70 75 80
81 Arg VaI Asn His VaI Thr Leu Ser GIn Pro Lys lie VaI Lys Trp Asp 85 90 95
Arg Asp Met
<210> 10
<211> 99
<212> PRT
<213> Rattus norvegicus <400> 10 lie GIn Lys Thr Pro GIn lie GIn VaI Tyr Ser Arg His Pro Pro GIu 1 5 10 15
Asn GIy Lys Pro Asn Phe Leu Asn Cys Tyr VaI Ser GIn Phe His Pro 20 25 30
Pro GIn lie GIu lie GIu Leu Leu Lys Asn GIy Lys Lys lie Pro Asn 35 40 45
lie GIu Met Ser Asp Leu Ser Phe Ser Lys Asp Trp Ser Phe Tyr lie 50 55 60
Leu Ala His Thr GIu Phe Thr Pro Thr GIu Thr Asp VaI Tyr Ala Cys 65 70 75 80
Arg VaI Lys His VaI Thr Leu Lys GIu Pro Lys Thr VaI Thr Trp Asp 85 90 95
Arg Asp Met
<210> 11
<211> 450
<212> PRT
<213> Artificial Sequence
<220>
<223> Trastuzumab Heavy chain
<400> 11
GIu VaI GIn Leu VaI GIu Ser GIy GIy GIy Leu VaI GIn Pro GIy GIy 1 5 10 15
82 Ser Leu Arg Leu Ser Cys Ala Ala Ser GIy Phe Asn lie Lys Asp Thr 20 25 30
Tyr lie His Trp VaI Arg Gin Ala Pro GIy Lys GIy Leu GIu Trp VaI 35 40 45
Ala Arg lie Tyr Pro Thr Asn GIy Tyr Thr Arg Tyr Ala Asp Ser VaI 50 55 60
Lys GIy Arg Phe Thr lie Ser Ala Asp Thr Ser Lys Asn Thr Ala Tyr 65 70 75 80
Leu GIn Met Asn Ser Leu Arg Ala GIu Asp Thr Ala VaI Tyr Tyr Cys 85 90 95
Ser Arg Trp GIy GIy Asp GIy Phe Tyr Ala Met Asp Tyr Trp GIy GIn 100 105 110
GIy Thr Leu VaI Thr VaI Ser Ser Ala Ser Thr Lys GIy Pro Ser VaI 115 120 125
Phe Pro Leu Ala Pro Ser Ser Lys Ser Thr Ser GIy GIy Thr Ala Ala 130 135 140
Leu GIy Cys Leu VaI Lys Asp Tyr Phe Pro GIu Pro VaI Thr VaI Ser 145 150 155 160
Trp Asn Ser GIy Ala Leu Thr Ser GIy VaI His Thr Phe Pro Ala VaI 165 170 175
Leu GIn Ser Ser GIy Leu Tyr Ser Leu Ser Ser VaI VaI Thr VaI Pro 180 185 190
Ser Ser Ser Leu GIy Thr Gin Thr Tyr lie Cys Asn VaI Asn His Lys 195 200 205
Pro Ser Asn Thr Lys VaI Asp Lys Lys VaI GIu Pro Lys Ser Cys Asp 210 215 220
Lys Thr His Thr Cys Pro Pro Cys Pro Ala Pro GIu Leu Leu GIy GIy
83 225 230 235 240
Pro Ser VaI Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met lie 245 250 255
Ser Arg Thr Pro GIu VaI Thr Cys VaI VaI VaI Asp VaI Ser His GIu 260 265 270
Asp Pro GIu VaI Lys Phe Asn Trp Tyr VaI Asp GIy VaI GIu VaI His 275 280 285
Asn Ala Lys Thr Lys Pro Arg GIu GIu GIn Tyr Asn Ser Thr Tyr Arg 290 295 300
VaI VaI Ser VaI Leu Thr VaI Leu His GIn Asp Trp Leu Asn GIy Lys 305 310 315 320
GIu Tyr Lys Cys Lys VaI Ser Asn Lys Ala Leu Pro Ala Pro lie GIu 325 330 335
Lys Thr lie Ser Lys Ala Lys GIy GIn Pro Arg GIu Pro GIn VaI Tyr 340 345 350
Thr Leu Pro Pro Ser Arg GIu GIu Met Thr Lys Asn GIn VaI Ser Leu 355 360 365
Thr Cys Leu VaI Lys GIy Phe Tyr Pro Ser Asp lie Ala VaI GIu Trp 370 375 380
GIu Ser Asn GIy GIn Pro GIu Asn Asn Tyr Lys Thr Thr Pro Pro VaI 385 390 395 400
Leu Asp Ser Asp GIy Ser Phe Phe Leu Tyr Ser Lys Leu Thr VaI Asp 405 410 415
Lys Ser Arg Trp GIn GIn GIy Asn VaI Phe Ser Cys Ser VaI Met His 420 425 430
GIu Ala Leu His Asn His Tyr Thr GIn Lys Ser Leu Ser Leu Ser Pro 435 440 445
84 GIy Lys 450
<210> 12
<211> 214
<212> PRT
<213> Artificial Sequence
<220>
<223> Trastuzumab light chain
<400> 12
Asp lie GIn Met Thr GIn Ser Pro Ser Ser Leu Ser Ala Ser VaI GIy 1 5 10 15
Asp Arg VaI Thr lie Thr Cys Arg Ala Ser GIn Asp VaI Asn Thr Ala 20 25 30
VaI Ala Trp Tyr GIn GIn Lys Pro GIy Lys Ala Pro Lys Leu Leu lie 35 40 45
Tyr Ser Ala Ser Phe Leu Tyr Ser GIy VaI Pro Ser Arg Phe Ser GIy 50 55 60
Ser Arg Ser GIy Thr Asp Phe Thr Leu Thr lie Ser Ser Leu GIn Pro 65 70 75 80
GIu Asp Phe Ala Thr Tyr Tyr Cys GIn GIn His Tyr Thr Thr Pro Pro 85 90 95
Thr Phe GIy GIn GIy Thr Lys VaI GIu He Lys Arg Thr VaI Ala Ala 100 105 HO
Pro Ser VaI Phe He Phe Pro Pro Ser Asp GIu GIn Leu Lys Ser GIy 115 120 125
Thr Ala Ser VaI VaI Cys Leu Leu Asn Asn Phe Tyr Pro Arg GIu Ala 130 135 140
Lys VaI GIn Trp Lys VaI Asp Asn Ala Leu GIn Ser GIy Asn Ser GIn 145 150 155 160
GIu Ser VaI Thr GIu GIn Asp Ser Lys Asp Ser Thr Tyr Ser Leu Ser
85 165 170 175
Ser Thr Leu Thr Leu Ser Lys Ala Asp Tyr GIu Lys His Lys VaI Tyr 180 185 190
Ala Cys GIu VaI Thr His Gin GIy Leu Ser Ser Pro VaI Thr Lys Ser 195 200 205
Phe Asn Arg GIy GIu Cys 210
<210 > 13
<211 > 330
<212 > PRT
<213 > Homo sapiens
<400 > 13
Ala Ser Thr Lys GIy Pro Ser VaI Phe Pro Leu Ala Pro Ser Ser Lys 1 5 10 15
Ser Thr Ser GIy GIy Thr Ala Ala Leu GIy Cys Leu VaI Lys Asp Tyr 20 25 30
Phe Pro GIu Pro VaI Thr VaI Ser Trp Asn Ser GIy Ala Leu Thr Ser 35 40 45
GIy VaI His Thr Phe Pro Ala VaI Leu GIn Ser Ser GIy Leu Tyr Ser 50 55 60
Leu Ser Ser VaI VaI Thr VaI Pro Ser Ser Ser Leu GIy Thr GIn Thr 65 70 75 80
Tyr lie Cys Asn VaI Asn His Lys Pro Ser Asn Thr Lys VaI Asp Lys 85 90 95
Lys VaI GIu Pro Lys Ser Cys Asp Lys Thr His Thr Cys Pro Pro Cys 100 105 110
Pro Ala Pro GIu Leu Leu GIy GIy Pro Ser VaI Phe Leu Phe Pro Pro 115 120 125
Lys Pro Lys Asp Thr Leu Met lie Ser Arg Thr Pro GIu VaI Thr Cys
86 130 135 140
VaI VaI VaI Asp VaI Ser His GIu Asp Pro GIu VaI Lys Phe Asn Trp 145 150 155 160
Tyr VaI Asp GIy VaI GIu VaI His Asn Ala Lys Thr Lys Pro Arg GIu 165 170 175
GIu GIn Tyr Asn Ser Thr Tyr Arg VaI VaI Ser VaI Leu Thr VaI Leu 180 185 190
His GIn Asp Trp Leu Asn GIy Lys GIu Tyr Lys Cys Lys VaI Ser Asn 195 200 205
Lys Ala Leu Pro Ala Pro lie GIu Lys Thr lie Ser Lys Ala Lys GIy 210 215 220
GIn Pro Arg GIu Pro GIn VaI Tyr Thr Leu Pro Pro Ser Arg GIu GIu 225 230 235 240
Met Thr Lys Asn GIn VaI Ser Leu Thr Cys Leu VaI Lys GIy Phe Tyr 245 250 255
Pro Ser Asp lie Ala VaI GIu Trp GIu Ser Asn GIy GIn Pro GIu Asn 260 265 270
Asn Tyr Lys Thr Thr Pro Pro VaI Leu Asp Ser Asp GIy Ser Phe Phe 275 280 285
Leu Tyr Ser Lys Leu Thr VaI Asp Lys Ser Arg Trp GIn GIn GIy Asn 290 295 300
VaI Phe Ser Cys Ser VaI Met His GIu Ala Leu His Asn His Tyr Thr 305 310 315 320
GIn Lys Ser Leu Ser Leu Ser Pro GIy Lys 325 330
<210> 14
<211> 330
<212> PRT
<213> Artificial Sequence
87 <220>
<223> P257L IgGl
<400> 14
Ala Ser Thr Lys GIy Pro Ser VaI Phe Pro Leu Ala Pro Ser Ser Lys 1 5 10 15
Ser Thr Ser GIy GIy Thr Ala Ala Leu GIy Cys Leu VaI Lys Asp Tyr 20 25 30
Phe Pro GIu Pro VaI Thr VaI Ser Trp Asn Ser GIy Ala Leu Thr Ser 35 40 45
GIy VaI His Thr Phe Pro Ala VaI Leu GIn Ser Ser GIy Leu Tyr Ser 50 55 60
Leu Ser Ser VaI VaI Thr VaI Pro Ser Ser Ser Leu GIy Thr GIn Thr 65 70 75 80
Tyr lie Cys Asn VaI Asn His Lys Pro Ser Asn Thr Lys VaI Asp Lys 85 90 95
Lys VaI GIu Pro Lys Ser Cys Asp Lys Thr His Thr Cys Pro Pro Cys 100 105 110
Pro Ala Pro GIu Leu Leu GIy GIy Pro Ser VaI Phe Leu Phe Pro Pro 115 120 125
Lys Pro Lys Asp Thr Leu Met lie Ser Arg Thr Leu GIu VaI Thr Cys 130 135 140
VaI VaI VaI Asp VaI Ser His GIu Asp Pro GIu VaI Lys Phe Asn Trp 145 150 155 160
Tyr VaI Asp GIy VaI GIu VaI His Asn Ala Lys Thr Lys Pro Arg GIu 165 170 175
GIu GIn Tyr Asn Ser Thr Tyr Arg VaI VaI Ser VaI Leu Thr VaI Leu 180 185 190
His GIn Asp Trp Leu Asn GIy Lys GIu Tyr Lys Cys Lys VaI Ser Asn
88 195 200 205
Lys Ala Leu Pro Ala Pro lie GIu Lys Thr lie Ser Lys Ala Lys GIy 210 215 220
GIn Pro Arg GIu Pro GIn VaI Tyr Thr Leu Pro Pro Ser Arg GIu GIu 225 230 235 240
Met Thr Lys Asn GIn VaI Ser Leu Thr Cys Leu VaI Lys GIy Phe Tyr 245 250 255
Pro Ser Asp lie Ala VaI GIu Trp GIu Ser Asn GIy GIn Pro GIu Asn 260 265 270
Asn Tyr Lys Thr Thr Pro Pro VaI Leu Asp Ser Asp GIy Ser Phe Phe 275 280 285
Leu Tyr Ser Lys Leu Thr VaI Asp Lys Ser Arg Trp GIn GIn GIy Asn 290 295 300
VaI Phe Ser Cys Ser VaI Met His GIu Ala Leu His Asn His Tyr Thr 305 310 315 320
GIn Lys Ser Leu Ser Leu Ser Pro GIy Lys 325 330
<210> 15
<211> 330
<212> PRT
<213> Artificial Sequence
<220>
<223> P257N IgGl
<400> 15
Ala Ser Thr Lys GIy Pro Ser VaI Phe Pro Leu Ala Pro Ser Ser Lys 1 5 10 15
Ser Thr Ser GIy GIy Thr Ala Ala Leu GIy Cys Leu VaI Lys Asp Tyr 20 25 30
Phe Pro GIu Pro VaI Thr VaI Ser Trp Asn Ser GIy Ala Leu Thr Ser 35 40 45
89 GIy VaI His Thr Phe Pro Ala VaI Leu GIn Ser Ser GIy Leu Tyr Ser 50 55 60
Leu Ser Ser VaI VaI Thr VaI Pro Ser Ser Ser Leu GIy Thr GIn Thr 65 70 75 80
Tyr lie Cys Asn VaI Asn His Lys Pro Ser Asn Thr Lys VaI Asp Lys 85 90 95
Lys VaI GIu Pro Lys Ser Cys Asp Lys Thr His Thr Cys Pro Pro Cys 100 105 110
Pro Ala Pro GIu Leu Leu GIy GIy Pro Ser VaI Phe Leu Phe Pro Pro 115 120 125
Lys Pro Lys Asp Thr Leu Met lie Ser Arg Thr Asn GIu VaI Thr Cys 130 135 140
VaI VaI VaI Asp VaI Ser His GIu Asp Pro GIu VaI Lys Phe Asn Trp 145 150 155 160
Tyr VaI Asp GIy VaI GIu VaI His Asn Ala Lys Thr Lys Pro Arg GIu 165 170 175
GIu GIn Tyr Asn Ser Thr Tyr Arg VaI VaI Ser VaI Leu Thr VaI Leu 180 185 190
His GIn Asp Trp Leu Asn GIy Lys GIu Tyr Lys Cys Lys VaI Ser Asn 195 200 205
Lys Ala Leu Pro Ala Pro lie GIu Lys Thr lie Ser Lys Ala Lys GIy 210 215 220
GIn Pro Arg GIu Pro GIn VaI Tyr Thr Leu Pro Pro Ser Arg GIu GIu 225 230 235 240
Met Thr Lys Asn GIn VaI Ser Leu Thr Cys Leu VaI Lys GIy Phe Tyr 245 250 255
Pro Ser Asp lie Ala VaI GIu Trp GIu Ser Asn GIy GIn Pro GIu Asn
90 260 265 270
Asn Tyr Lys Thr Thr Pro Pro VaI Leu Asp Ser Asp GIy Ser Phe Phe 275 280 285
Leu Tyr Ser Lys Leu Thr VaI Asp Lys Ser Arg Trp GIn GIn GIy Asn 290 295 300
VaI Phe Ser Cys Ser VaI Met His GIu Ala Leu His Asn His Tyr Thr 305 310 315 320
GIn Lys Ser Leu Ser Leu Ser Pro GIy Lys 325 330
<210> 16
<211> 330
<212> PRT
<213> Artificial Sequence
<220>
<223> V308F IgGl
<400> 16
Ala Ser Thr Lys GIy Pro Ser VaI Phe Pro Leu Ala Pro Ser Ser Lys 1 5 10 15
Ser Thr Ser GIy GIy Thr Ala Ala Leu GIy Cys Leu VaI Lys Asp Tyr 20 25 30
Phe Pro GIu Pro VaI Thr VaI Ser Trp Asn Ser GIy Ala Leu Thr Ser 35 40 45
GIy VaI His Thr Phe Pro Ala VaI Leu GIn Ser Ser GIy Leu Tyr Ser 50 55 60
Leu Ser Ser VaI VaI Thr VaI Pro Ser Ser Ser Leu GIy Thr GIn Thr 65 70 75 80
Tyr lie Cys Asn VaI Asn His Lys Pro Ser Asn Thr Lys VaI Asp Lys 85 90 95
Lys VaI GIu Pro Lys Ser Cys Asp Lys Thr His Thr Cys Pro Pro Cys 100 105 110
91 Pro Ala Pro GIu Leu Leu GIy GIy Pro Ser VaI Phe Leu Phe Pro Pro 115 120 125
Lys Pro Lys Asp Thr Leu Met lie Ser Arg Thr Pro GIu VaI Thr Cys 130 135 140
VaI VaI VaI Asp VaI Ser His GIu Asp Pro GIu VaI Lys Phe Asn Trp 145 150 155 160
Tyr VaI Asp GIy VaI GIu VaI His Asn Ala Lys Thr Lys Pro Arg GIu 165 170 175
GIu GIn Tyr Asn Ser Thr Tyr Arg VaI VaI Ser VaI Leu Thr Phe Leu 180 185 190
His GIn Asp Trp Leu Asn GIy Lys GIu Tyr Lys Cys Lys VaI Ser Asn 195 200 205
Lys Ala Leu Pro Ala Pro lie GIu Lys Thr lie Ser Lys Ala Lys GIy 210 215 220
GIn Pro Arg GIu Pro GIn VaI Tyr Thr Leu Pro Pro Ser Arg GIu GIu 225 230 235 240
Met Thr Lys Asn GIn VaI Ser Leu Thr Cys Leu VaI Lys GIy Phe Tyr 245 250 255
Pro Ser Asp lie Ala VaI GIu Trp GIu Ser Asn GIy GIn Pro GIu Asn 260 265 270
Asn Tyr Lys Thr Thr Pro Pro VaI Leu Asp Ser Asp GIy Ser Phe Phe 275 280 285
Leu Tyr Ser Lys Leu Thr VaI Asp Lys Ser Arg Trp GIn GIn GIy Asn 290 295 300
VaI Phe Ser Cys Ser VaI Met His GIu Ala Leu His Asn His Tyr Thr 305 310 315 320
GIn Lys Ser Leu Ser Leu Ser Pro GIy Lys
92 325 330
<210> 17
<211> 330
<212> PRT
<213> Artificial Sequence
<220>
<223> Q311V IgGl
<400> 17
Ala Ser Thr Lys GIy Pro Ser VaI Phe Pro Leu Ala Pro Ser Ser Lys 1 5 10 15
Ser Thr Ser GIy GIy Thr Ala Ala Leu GIy Cys Leu VaI Lys Asp Tyr 20 25 30
Phe Pro GIu Pro VaI Thr VaI Ser Trp Asn Ser GIy Ala Leu Thr Ser 35 40 45
GIy VaI His Thr Phe Pro Ala VaI Leu GIn Ser Ser GIy Leu Tyr Ser 50 55 60
Leu Ser Ser VaI VaI Thr VaI Pro Ser Ser Ser Leu GIy Thr GIn Thr 65 70 75 80
Tyr lie Cys Asn VaI Asn His Lys Pro Ser Asn Thr Lys VaI Asp Lys 85 90 95
Lys VaI GIu Pro Lys Ser Cys Asp Lys Thr His Thr Cys Pro Pro Cys 100 105 110
Pro Ala Pro GIu Leu Leu GIy GIy Pro Ser VaI Phe Leu Phe Pro Pro 115 120 125
Lys Pro Lys Asp Thr Leu Met lie Ser Arg Thr Pro GIu VaI Thr Cys 130 135 140
VaI VaI VaI Asp VaI Ser His GIu Asp Pro GIu VaI Lys Phe Asn Trp 145 150 155 160
Tyr VaI Asp GIy VaI GIu VaI His Asn Ala Lys Thr Lys Pro Arg GIu 165 170 175
93 GIu GIn Tyr Asn Ser Thr Tyr Arg VaI VaI Ser VaI Leu Thr VaI Leu 180 185 190
His VaI Asp Trp Leu Asn GIy Lys GIu Tyr Lys Cys Lys VaI Ser Asn 195 200 205
Lys Ala Leu Pro Ala Pro lie GIu Lys Thr lie Ser Lys Ala Lys GIy 210 215 220
GIn Pro Arg GIu Pro GIn VaI Tyr Thr Leu Pro Pro Ser Arg GIu GIu 225 230 235 240
Met Thr Lys Asn GIn VaI Ser Leu Thr Cys Leu VaI Lys GIy Phe Tyr 245 250 255
Pro Ser Asp lie Ala VaI GIu Trp GIu Ser Asn GIy GIn Pro GIu Asn 260 265 270
Asn Tyr Lys Thr Thr Pro Pro VaI Leu Asp Ser Asp GIy Ser Phe Phe 275 280 285
Leu Tyr Ser Lys Leu Thr VaI Asp Lys Ser Arg Trp GIn GIn GIy Asn 290 295 300
VaI Phe Ser Cys Ser VaI Met His GIu Ala Leu His Asn His Tyr Thr 305 310 315 320
GIn Lys Ser Leu Ser Leu Ser Pro GIy Lys 325 330
<210> 18
<211> 330
<212> PRT
<213> Artificial Sequence
<220>
<223> G385H IgGl
<400> 18
Ala Ser Thr Lys GIy Pro Ser VaI Phe Pro Leu Ala Pro Ser Ser Lys 1 5 10 15
94 Ser Thr Ser GIy GIy Thr Ala Ala Leu GIy Cys Leu VaI Lys Asp Tyr 20 25 30
Phe Pro GIu Pro VaI Thr VaI Ser Trp Asn Ser GIy Ala Leu Thr Ser 35 40 45
GIy VaI His Thr Phe Pro Ala VaI Leu GIn Ser Ser GIy Leu Tyr Ser 50 55 60
Leu Ser Ser VaI VaI Thr VaI Pro Ser Ser Ser Leu GIy Thr GIn Thr 65 70 75 80
Tyr lie Cys Asn VaI Asn His Lys Pro Ser Asn Thr Lys VaI Asp Lys 85 90 95
Lys VaI GIu Pro Lys Ser Cys Asp Lys Thr His Thr Cys Pro Pro Cys 100 105 110
Pro Ala Pro GIu Leu Leu GIy GIy Pro Ser VaI Phe Leu Phe Pro Pro 115 120 125
Lys Pro Lys Asp Thr Leu Met lie Ser Arg Thr Pro GIu VaI Thr Cys 130 135 140
VaI VaI VaI Asp VaI Ser His GIu Asp Pro GIu VaI Lys Phe Asn Trp 145 150 155 160
Tyr VaI Asp GIy VaI GIu VaI His Asn Ala Lys Thr Lys Pro Arg GIu 165 170 175
GIu GIn Tyr Asn Ser Thr Tyr Arg VaI VaI Ser VaI Leu Thr VaI Leu 180 185 190
His GIn Asp Trp Leu Asn GIy Lys GIu Tyr Lys Cys Lys VaI Ser Asn 195 200 205
Lys Ala Leu Pro Ala Pro lie GIu Lys Thr lie Ser Lys Ala Lys GIy 210 215 220
GIn Pro Arg GIu Pro GIn VaI Tyr Thr Leu Pro Pro Ser Arg GIu GIu 225 230 235 240
95 Met Thr Lys Asn GIn VaI Ser Leu Thr Cys Leu VaI Lys GIy Phe Tyr 245 250 255
Pro Ser Asp lie Ala VaI GIu Trp GIu Ser Asn His GIn Pro GIu Asn 260 265 270
Asn Tyr Lys Thr Thr Pro Pro VaI Leu Asp Ser Asp GIy Ser Phe Phe 275 280 285
Leu Tyr Ser Lys Leu Thr VaI Asp Lys Ser Arg Trp GIn GIn GIy Asn 290 295 300
VaI Phe Ser Cys Ser VaI Met His GIu Ala Leu His Asn His Tyr Thr 305 310 315 320
GIn Lys Ser Leu Ser Leu Ser Pro GIy Lys 325 330
<210> 19
<211> 330
<212> PRT
<213> Artificial Sequence
<220>
<223> WT hybrid
<400> 19
Ala Ser Thr Lys GIy Pro Ser VaI Phe Pro Leu Ala Pro Ser Ser Lys 1 5 10 15
Ser Thr Ser GIy GIy Thr Ala Ala Leu GIy Cys Leu VaI Lys Asp Tyr 20 25 30
Phe Pro GIu Pro VaI Thr VaI Ser Trp Asn Ser GIy Ala Leu Thr Ser 35 40 45
GIy VaI His Thr Phe Pro Ala VaI Leu GIn Ser Ser GIy Leu Tyr Ser 50 55 60
Leu Ser Ser VaI VaI Thr VaI Pro Ser Ser Ser Leu GIy Thr GIn Thr 65 70 75 80
96 Tyr lie Cys Asn VaI Asn His Lys Pro Ser Asn Thr Lys VaI Asp Lys 85 90 95
Lys VaI GIu Pro Lys Ser Cys Asp Lys Thr His Thr Cys Pro Pro Cys 100 105 110
Pro Ala Pro GIu Leu Leu GIy GIy Pro Ser VaI Phe Leu Phe Pro Pro 115 120 125
Lys Pro Lys Asp Thr Leu Met lie Ser Arg Thr Pro GIu VaI Thr Cys 130 135 140
VaI VaI VaI Asp VaI Ser His GIu Asp Pro GIu VaI GIn Phe Asn Trp 145 150 155 160
Tyr VaI Asp GIy VaI GIu VaI His Asn Ala Lys Thr Lys Pro Arg GIu 165 170 175
GIu GIn Phe Asn Ser Thr Phe Arg VaI VaI Ser VaI Leu Thr VaI VaI 180 185 190
His GIn Asp Trp Leu Asn GIy Lys GIu Tyr Lys Cys Lys VaI Ser Asn 195 200 205
Lys Ala Leu Pro Ala Pro lie GIu Lys Thr lie Ser Lys Thr Lys GIy 210 215 220
GIn Pro Arg GIu Pro GIn VaI Tyr Thr Leu Pro Pro Ser Arg GIu GIu 225 230 235 240
Met Thr Lys Asn GIn VaI Ser Leu Thr Cys Leu VaI Lys GIy Phe Tyr 245 250 255
Pro Ser Asp lie Ala VaI GIu Trp GIu Ser Asn GIy GIn Pro GIu Asn 260 265 270
Asn Tyr Lys Thr Thr Pro Pro Met Leu Asp Ser Asp GIy Ser Phe Phe 275 280 285
Leu Tyr Ser Lys Leu Thr VaI Asp Lys Ser Arg Trp GIn GIn GIy Asn 290 295 300
97 VaI Phe Ser Cys Ser VaI Met His GIu Ala Leu His Asn His Tyr Thr 305 310 315 320
GIn Lys Ser Leu Ser Leu Ser Pro GIy Lys 325 330
<210> 20
<211> 330
<212> PRT
<213> Artificial Sequence
<220>
<223> P257L Hybrid
<400> 20
Ala Ser Thr Lys GIy Pro Ser VaI Phe Pro Leu Ala Pro Ser Ser Lys 1 5 10 15
Ser Thr Ser GIy GIy Thr Ala Ala Leu GIy Cys Leu VaI Lys Asp Tyr 20 25 30
Phe Pro GIu Pro VaI Thr VaI Ser Trp Asn Ser GIy Ala Leu Thr Ser 35 40 45
GIy VaI His Thr Phe Pro Ala VaI Leu GIn Ser Ser GIy Leu Tyr Ser 50 55 60
Leu Ser Ser VaI VaI Thr VaI Pro Ser Ser Ser Leu GIy Thr GIn Thr 65 70 75 80
Tyr lie Cys Asn VaI Asn His Lys Pro Ser Asn Thr Lys VaI Asp Lys 85 90 95
Lys VaI GIu Pro Lys Ser Cys Asp Lys Thr His Thr Cys Pro Pro Cys 100 105 110
Pro Ala Pro GIu Leu Leu GIy GIy Pro Ser VaI Phe Leu Phe Pro Pro 115 120 125
Lys Pro Lys Asp Thr Leu Met lie Ser Arg Thr Leu GIu VaI Thr Cys 130 135 140
98 VaI VaI VaI Asp VaI Ser His GIu Asp Pro GIu VaI GIn Phe Asn Trp 145 150 155 160
Tyr VaI Asp GIy VaI GIu VaI His Asn Ala Lys Thr Lys Pro Arg GIu 165 170 175
GIu GIn Phe Asn Ser Thr Phe Arg VaI VaI Ser VaI Leu Thr VaI VaI 180 185 190
His GIn Asp Trp Leu Asn GIy Lys GIu Tyr Lys Cys Lys VaI Ser Asn 195 200 205
Lys Ala Leu Pro Ala Pro lie GIu Lys Thr lie Ser Lys Thr Lys GIy 210 215 220
GIn Pro Arg GIu Pro GIn VaI Tyr Thr Leu Pro Pro Ser Arg GIu GIu 225 230 235 240
Met Thr Lys Asn GIn VaI Ser Leu Thr Cys Leu VaI Lys GIy Phe Tyr 245 250 255
Pro Ser Asp lie Ala VaI GIu Trp GIu Ser Asn GIy GIn Pro GIu Asn 260 265 270
Asn Tyr Lys Thr Thr Pro Pro Met Leu Asp Ser Asp GIy Ser Phe Phe 275 280 285
Leu Tyr Ser Lys Leu Thr VaI Asp Lys Ser Arg Trp GIn GIn GIy Asn 290 295 300
VaI Phe Ser Cys Ser VaI Met His GIu Ala Leu His Asn His Tyr Thr 305 310 315 320
GIn Lys Ser Leu Ser Leu Ser Pro GIy Lys 325 330
<210> 21
<211> 330
<212> PRT
<213> Artificial Sequence
<220>
99 <223> P257N Hybrid <400> 21
Ala Ser Thr Lys GIy Pro Ser VaI Phe Pro Leu Ala Pro Ser Ser Lys 1 5 10 15
Ser Thr Ser GIy GIy Thr Ala Ala Leu GIy Cys Leu VaI Lys Asp Tyr 20 25 30
Phe Pro GIu Pro VaI Thr VaI Ser Trp Asn Ser GIy Ala Leu Thr Ser 35 40 45
GIy VaI His Thr Phe Pro Ala VaI Leu GIn Ser Ser GIy Leu Tyr Ser 50 55 60
Leu Ser Ser VaI VaI Thr VaI Pro Ser Ser Ser Leu GIy Thr GIn Thr 65 70 75 80
Tyr lie Cys Asn VaI Asn His Lys Pro Ser Asn Thr Lys VaI Asp Lys 85 90 95
Lys VaI GIu Pro Lys Ser Cys Asp Lys Thr His Thr Cys Pro Pro Cys 100 105 110
Pro Ala Pro GIu Leu Leu GIy GIy Pro Ser VaI Phe Leu Phe Pro Pro 115 120 125
Lys Pro Lys Asp Thr Leu Met lie Ser Arg Thr Asn GIu VaI Thr Cys 130 135 140
VaI VaI VaI Asp VaI Ser His GIu Asp Pro GIu VaI GIn Phe Asn Trp 145 150 155 160
Tyr VaI Asp GIy VaI GIu VaI His Asn Ala Lys Thr Lys Pro Arg GIu 165 170 175
GIu GIn Phe Asn Ser Thr Phe Arg VaI VaI Ser VaI Leu Thr VaI VaI 180 185 190
His GIn Asp Trp Leu Asn GIy Lys GIu Tyr Lys Cys Lys VaI Ser Asn 195 200 205
100 Lys Ala Leu Pro Ala Pro lie GIu Lys Thr lie Ser Lys Thr Lys GIy 210 215 220
GIn Pro Arg GIu Pro GIn VaI Tyr Thr Leu Pro Pro Ser Arg GIu GIu 225 230 235 240
Met Thr Lys Asn GIn VaI Ser Leu Thr Cys Leu VaI Lys GIy Phe Tyr 245 250 255
Pro Ser Asp lie Ala VaI GIu Trp GIu Ser Asn GIy GIn Pro GIu Asn 260 265 270
Asn Tyr Lys Thr Thr Pro Pro Met Leu Asp Ser Asp GIy Ser Phe Phe 275 280 285
Leu Tyr Ser Lys Leu Thr VaI Asp Lys Ser Arg Trp GIn GIn GIy Asn 290 295 300
VaI Phe Ser Cys Ser VaI Met His GIu Ala Leu His Asn His Tyr Thr 305 310 315 320
GIn Lys Ser Leu Ser Leu Ser Pro GIy Lys 325 330
<210> 22
<211> 330
<212> PRT
<213> Artificial Sequence
<220>
<223> V308F Hybrid
<400> 22
Ala Ser Thr Lys GIy Pro Ser VaI Phe Pro Leu Ala Pro Ser Ser Lys 1 5 10 15
Ser Thr Ser GIy GIy Thr Ala Ala Leu GIy Cys Leu VaI Lys Asp Tyr 20 25 30
Phe Pro GIu Pro VaI Thr VaI Ser Trp Asn Ser GIy Ala Leu Thr Ser 35 40 45
101 GIy VaI His Thr Phe Pro Ala VaI Leu GIn Ser Ser GIy Leu Tyr Ser 50 55 60
Leu Ser Ser VaI VaI Thr VaI Pro Ser Ser Ser Leu GIy Thr GIn Thr 65 70 75 80
Tyr lie Cys Asn VaI Asn His Lys Pro Ser Asn Thr Lys VaI Asp Lys 85 90 95
Lys VaI GIu Pro Lys Ser Cys Asp Lys Thr His Thr Cys Pro Pro Cys 100 105 110
Pro Ala Pro GIu Leu Leu GIy GIy Pro Ser VaI Phe Leu Phe Pro Pro 115 120 125
Lys Pro Lys Asp Thr Leu Met lie Ser Arg Thr Pro GIu VaI Thr Cys 130 135 140
VaI VaI VaI Asp VaI Ser His GIu Asp Pro GIu VaI GIn Phe Asn Trp 145 150 155 160
Tyr VaI Asp GIy VaI GIu VaI His Asn Ala Lys Thr Lys Pro Arg GIu 165 170 175
GIu GIn Phe Asn Ser Thr Phe Arg VaI VaI Ser VaI Leu Thr Phe VaI 180 185 190
His GIn Asp Trp Leu Asn GIy Lys GIu Tyr Lys Cys Lys VaI Ser Asn 195 200 205
Lys Ala Leu Pro Ala Pro lie GIu Lys Thr lie Ser Lys Thr Lys GIy 210 215 220
GIn Pro Arg GIu Pro GIn VaI Tyr Thr Leu Pro Pro Ser Arg GIu GIu 225 230 235 240
Met Thr Lys Asn GIn VaI Ser Leu Thr Cys Leu VaI Lys GIy Phe Tyr 245 250 255
Pro Ser Asp lie Ala VaI GIu Trp GIu Ser Asn GIy GIn Pro GIu Asn 260 265 270
102 Asn Tyr Lys Thr Thr Pro Pro Met Leu Asp Ser Asp GIy Ser Phe Phe 275 280 285
Leu Tyr Ser Lys Leu Thr VaI Asp Lys Ser Arg Trp GIn GIn GIy Asn 290 295 300
VaI Phe Ser Cys Ser VaI Met His GIu Ala Leu His Asn His Tyr Thr 305 310 315 320
GIn Lys Ser Leu Ser Leu Ser Pro GIy Lys 325 330
<210> 23
<211> 330
<212> PRT
<213> Homo sapiens
<400> 23
Ala Ser Thr Lys GIy Pro Ser VaI Phe Pro Leu Ala Pro Ser Ser Lys 1 5 10 15
Ser Thr Ser GIy GIy Thr Ala Ala Leu GIy Cys Leu VaI Lys Asp Tyr 20 25 30
Phe Pro GIu Pro VaI Thr VaI Ser Trp Asn Ser GIy Ala Leu Thr Ser 35 40 45
GIy VaI His Thr Phe Pro Ala VaI Leu GIn Ser Ser GIy Leu Tyr Ser 50 55 60
Leu Ser Ser VaI VaI Thr VaI Pro Ser Ser Ser Leu GIy Thr GIn Thr 65 70 75 80
Tyr lie Cys Asn VaI Asn His Lys Pro Ser Asn Thr Lys VaI Asp Lys 85 90 95
Lys VaI GIu Pro Lys Ser Cys Asp Lys Thr His Thr Cys Pro Pro Cys 100 105 110
Pro Ala Pro GIu Leu Leu GIy GIy Pro Ser VaI Phe Leu Phe Pro Pro 115 120 125
103 Lys Pro Lys Asp Thr Leu Met lie Ser Arg Thr Pro GIu VaI Thr Cys 130 135 140
VaI VaI VaI Asp VaI Ser His GIu Asp Pro GIu VaI GIn Phe Asn Trp 145 150 155 160
Tyr VaI Asp GIy VaI GIu VaI His Asn Ala Lys Thr Lys Pro Arg GIu 165 170 175
GIu GIn Phe Asn Ser Thr Phe Arg VaI VaI Ser VaI Leu Thr VaI VaI 180 185 190
His VaI Asp Trp Leu Asn GIy Lys GIu Tyr Lys Cys Lys VaI Ser Asn 195 200 205
Lys Ala Leu Pro Ala Pro lie GIu Lys Thr lie Ser Lys Thr Lys GIy 210 215 220
GIn Pro Arg GIu Pro GIn VaI Tyr Thr Leu Pro Pro Ser Arg GIu GIu 225 230 235 240
Met Thr Lys Asn GIn VaI Ser Leu Thr Cys Leu VaI Lys GIy Phe Tyr 245 250 255
Pro Ser Asp lie Ala VaI GIu Trp GIu Ser Asn GIy GIn Pro GIu Asn 260 265 270
Asn Tyr Lys Thr Thr Pro Pro Met Leu Asp Ser Asp GIy Ser Phe Phe 275 280 285
Leu Tyr Ser Lys Leu Thr VaI Asp Lys Ser Arg Trp GIn GIn GIy Asn 290 295 300
VaI Phe Ser Cys Ser VaI Met His GIu Ala Leu His Asn His Tyr Thr 305 310 315 320
GIn Lys Ser Leu Ser Leu Ser Pro GIy Lys 325 330
<210> 24 <211> 330
104 <212> PRT
<213> Artificial Sequence
<220>
<223> G385H Hybrid
<400> 24
Ala Ser Thr Lys GIy Pro Ser VaI Phe Pro Leu Ala Pro Ser Ser Lys 1 5 10 15
Ser Thr Ser GIy GIy Thr Ala Ala Leu GIy Cys Leu VaI Lys Asp Tyr 20 25 30
Phe Pro GIu Pro VaI Thr VaI Ser Trp Asn Ser GIy Ala Leu Thr Ser 35 40 45
GIy VaI His Thr Phe Pro Ala VaI Leu GIn Ser Ser GIy Leu Tyr Ser 50 55 60
Leu Ser Ser VaI VaI Thr VaI Pro Ser Ser Ser Leu GIy Thr GIn Thr 65 70 75 80
Tyr lie Cys Asn VaI Asn His Lys Pro Ser Asn Thr Lys VaI Asp Lys 85 90 95
Lys VaI GIu Pro Lys Ser Cys Asp Lys Thr His Thr Cys Pro Pro Cys 100 105 110
Pro Ala Pro GIu Leu Leu GIy GIy Pro Ser VaI Phe Leu Phe Pro Pro 115 120 125
Lys Pro Lys Asp Thr Leu Met lie Ser Arg Thr Pro GIu VaI Thr Cys 130 135 140
VaI VaI VaI Asp VaI Ser His GIu Asp Pro GIu VaI GIn Phe Asn Trp 145 150 155 160
Tyr VaI Asp GIy VaI GIu VaI His Asn Ala Lys Thr Lys Pro Arg GIu 165 170 175
GIu GIn Phe Asn Ser Thr Phe Arg VaI VaI Ser VaI Leu Thr VaI VaI 180 185 190
105 His GIn Asp Trp Leu Asn GIy Lys GIu Tyr Lys Cys Lys VaI Ser Asn 195 200 205
Lys Ala Leu Pro Ala Pro lie GIu Lys Thr lie Ser Lys Thr Lys GIy 210 215 220
GIn Pro Arg GIu Pro GIn VaI Tyr Thr Leu Pro Pro Ser Arg GIu GIu 225 230 235 240
Met Thr Lys Asn GIn VaI Ser Leu Thr Cys Leu VaI Lys GIy Phe Tyr 245 250 255
Pro Ser Asp lie Ala VaI GIu Trp GIu Ser Asn His GIn Pro GIu Asn 260 265 270
Asn Tyr Lys Thr Thr Pro Pro Met Leu Asp Ser Asp GIy Ser Phe Phe 275 280 285
Leu Tyr Ser Lys Leu Thr VaI Asp Lys Ser Arg Trp GIn GIn GIy Asn 290 295 300
VaI Phe Ser Cys Ser VaI Met His GIu Ala Leu His Asn His Tyr Thr 305 310 315 320
GIn Lys Ser Leu Ser Leu Ser Pro GIy Lys 325 330
106

Claims

1. An antibody or immunoadhesin of a parent Fc polypeptide, said antibody or immunoadhesin comprising at least one modification in the Fc region, wherein said antibody or immunoadhesin exhibits altered binding to FcRn as compared to said parent antibody or immunoadhesin, wherein said modification is selected from the group consisting of: 259I, 307S, 319F, 319L, and 434M, and wherein numbering is according to the EU Index in Kabat et al.
2. An antibody or immunoadhesin according to claim 1 , wherein said Fc region further comprises at least one additional modification selected from the group consisting of: 246H, 246S, 247D, 247T, 248H, 248P, 248Q, 248R, 248Y, 249T, 249W, 250E, 250I, 250Q, 250V, 251 D, 251 E, 251 H, 2511, 251 K, 251 M, 251 N, 251T, 251V, 251Y, 252Q, 252Y, 253L, 253T, 253V, 254H, 254L, 254N, 254T, 254V, Λ254N, 255E, 255F, 255H, 255K, 255S, 255V, 256E, 256V, 257A, 257C, 257D, 257E, 257F, 257G, 257H, 257I, 257K, 257L, 257M, 257N, 257Q, 257R, 257S, 257T, 257V, 257W, 257Y, 258R, 258V, 279A, 279D, 279C, 279F, 279G, 279H, 279I, 279K, 279M, 279N, 279P, 279Q, 279Q, 279R, 279S, 279T, 279W, 279Y, 280H, Λ281A, Λ281 D, Λ281 S, Λ281T, 282D, 282F, 282H, 282I, 282T, 283F, 283I, 283L, 283Y, 284H, 284K, 284P, 284Q, 284R, 284S, 284Y, 285S, 285V, 286D, 286#, 286L, 287H, 287S, 287V, 287Y, 288H, 288Q, 288S, 305H, 305T, 306F, 306H, 306I, 306N, 306T, 306V, 306Y, 307D, 307P, 307Q, 307S, 307V, 307Y, 308C, 308D, 308E, 308F, 308G, 308H, 308I, 308K, 308L, 308M, 308N, 308Q, 308P, 308R, 308S, 308W, 308Y, 309F, 309H, 309N, 309Q, 309V, 309Y, 310K, 310N, 310T, 31 1 L, 311 P, 311T, 311V, 311W, 312H, 313Y, 315E, 315G, 315H, 315Q, 315S, 315T, 317H, 317S, 339P, 340P, 341 S, 374H, 374S, 376H, 376L, 378H, 378N, 380A, 380T, 380Y, 382H, 383H, 383K, 383Q, 384E, 384G, 384H, 385A, 385C, 385F, 385H, 385I, 385K, 385L, 385M, 385N, 385P, 385Q, 385S, 385T, 385V, 385W, 385Y, 386E, 386K, 387#, 387A, 387H, 387K, 387Q, 389E, 389H, 426E, 426H, 426L, 426N, 426R, 426V, 426Y, 427I, 428F, 428L, 429D, 429F, 429K, 429N, 429Q, 429S, 429T, 429Y, 430D, 430H, 430K, 430L, 430Q, 430Y, 431 G, 431 H, 4311, 431 P, 431 P, 431 S, 432F, 432H, 432N, 432S, 432V, 433E, 433P, 433S, 434A, 434F, 434H, 434L, 434M, 434Q, 434S, 434Y, 435N, 436E, 436F, 436L, 436V, 436W, 437E, 437V, 438H, and 438K, wherein Λ is an insertion after the indicated position and # is a deletion of the indicated position.
66
3. An antibody or immunoadhesin according to claim 1 or 2, wherein said Fc region comprises at least two modifications selected from the group consisting of: 250Q/252Y, 250Q/256E, 250Q/286D, 250Q/308F, 250Q/308Y, 250Q/311A, 250Q/31 1V, 250Q/380A, 250Q/428L, 250Q/428F, 250Q/434H, 250Q/434F, 250Q/434Y, 250Q/434A, 250Q/434M, θfid-250Q/434S, 250E/252Y, 250E/256E, 250E/286D, 250E/308F, 250E/308Y, 250E/311A, 250E/311V, 250E/380A, 250E/428L, 250E/428F, 250E/434H, 250E/434F, 250E/434Y, 250E/434A, 250E/434M, and 250E/434S.
4. An antibody or immunoadhesin according to claim 1 or 2, wherein said Fc region comprises at least two modifications selected from the group consisting of: 252Y/250Q, 252Y/250E, 252Y/256E, 252Y/286D, 252Y/308F, 252Y/308Y, 252Y/31 1A, 252Y/31 1V, 252Y/380A, 252Y/428L, 252Y/428F, 252Y/434H, 252Y/434F, 252Y/434Y, 252Y/434A, 252Y/434M, and 252Y/434S.
5. An antibody or immunoadhesin according to claim 1 or 2, wherein said Fc region comprises at least two modifications selected from the group consisting of: 256E/250Q, 256E/250E, 256E/252Y, 256E/286D, 256E/308F, 256E/308Y, 256E/31 1A, 256E/31 1V, 256E/380A, 256E/428L, 256E/428F, 256E/434H, 256E/434F, 256E/434Y, 256E/434A, 256E/434M, and 256E/434S.
6. An antibody or immunoadhesin according to claim 1 or 2, wherein said Fc region comprises at least two modifications selected from the group consisting of: 286D/250Q, 286D/250E, 286D/252Y, 286D/256E, 286D/308F, 286D/308Y, 286D/311A, 286D/31 1V, 286D/380A, 286D/428L, 286D/428F, 286D/434H, 286D/434F, 286D/434Y, 286D/434A, 286D/434M, and 286D/434S.
7. An antibody or immunoadhesin according to claim 1or 2, wherein said Fc region comprises at least two modifications selected from the group consisting of: 308F/250Q, 308F/250E, 308F/252Y, 308F/256E, 308F/286D, 308F/31 1A, 308F/311V, 308F/380A, 308F/428L, 308F/428F, 308F/434H, 308F/434F, 308F/434Y, 308F/434A, 308F/434M, 308F/434S, 308Y/250Q, 308Y/250E, 308Y/252Y, 308Y/256E, 308Y/286D, 308Y/31 1A, 308Y/31 1V, 308Y/380A, 308Y/428L, 308Y/428F, 308Y/434H, 308Y/434F, 308Y/434Y, 308Y/434A, 308Y/434M, and 308Y/434S.
67
8. An antibody or immunoadhesin according to claim 1or 2, wherein said Fc region comprises at least two modifications selected from the group consisting of: 31 1A/250Q, 311A/250E, 311A/252Y, 311A/256E, 31 1A/286D, 311A/308F, 31 1A/308Y, 311A/380A, 311A/428L, 311A/428F, 311A/434H, 31 1A/434F, 31 1A/434Y, 311A/434A, 311A/434M, 311A/434S, 311V/250Q, 31 1V/250E, 31 1V/252Y, 311V/256E, 311V/286D, 311V/308F, 31 1V/308Y, 31 1V/380A, 31 1V/428L, 311V/428F, 31 1V/434H, 311V/434F, 311V/434Y, 311V/434A, 31 1V/434M, and 311V/434S.
9. An antibody or immunoadhesin according to claim 1 or 2, wherein said Fc region comprises at least two modifications selected from the group consisting of: 380A/250Q, 380A/250E, 380A/252Y, 380A/256E, 380A/286D, 380A/308F, 380A/308Y, 380A/31 1A, 380A/311V, 380A/428L, 380A/428F, 380A/434H, 380A/434F, 380A/434Y, 380A/434A, 380A/434M, and 380A/434S.
10. An antibody or immunoadhesin according to claim 1 or 2, wherein said Fc region comprises at least two modifications selected from the group consisting of: 428L/250Q, 428L/250E, 428L/252Y, 428L/256E, 428L/286D, 428L/308F, 428L/308Y, 428L/31 1A, 428L/31 1V, 428L/380A, 428L/434H, 428L/434F, 428L/434Y, 428L/434A, 428L/434M, 428L/434S, 428F/250Q, 428F/250E, 428F/252Y, 428F/256E, 428F/286D, 428F/308F, 428F/308Y, 428F/311A, 428F/31 1V, 428F/380A, 428F/434H, 428F/434F, 428F/434Y, 428F/434A, 428F/434M, and 428F/434S.
11. An antibody or immunoadhesin according to claim 1 or 2, wherein said Fc region comprises at least two modifications selected from the group consisting of: 434H/250Q, 434H/250E, 434H/252Y, 434H/256E, 434H/286D, 434H/308F, 434H/308Y, 434H/31 1A, 434H/31 1V, 434H/380A, 434H/428L, 34H/428F434F/250Q, 434F/250E, 434F/252Y, 434F/256E, 434F/286D, 434F/308F, 434F/308Y, 434F/311A, 434F/31 1V, 434F/380A, 434F/428L, 434F/428F, 434Y/250Q, 434Y/250E, 434Y/252Y, 434Y/256E, 434Y/286D, 434Y/308F, 434Y/308Y, 434Y/31 1A, 434Y/31 1V, 434Y/380A, 434Y/428L, 434Y/428F, 434A/250Q, 434A/250E, 434A/252Y, 434A/256E, 434A/286D, 434A/308F, 434A/308Y, 434A/31 1A, 434A/31 1V, 434A/380A, 434A/428L, 434A/428F, 434M/250Q, 434M/250E, 434M/252Y, 434M/256E, 434M/286D, 434M/308F, 434M/308Y, 434M/311A, 434M/311V, 434M/380A, 434M/428L, 434M/428F434S/250Q, 434S/250E, 434S/252Y, 434S/256E, 434S/286D, 434S/308F, 434S/308Y, 434S/31 1A, 434S/31 1V, 434S/380A, 434S/428L, and 434S/428F.
68
12. An antibody or immunoadhesin according to claim 1 or 2, wherein said Fc region comprises modifications selected from the group consisting of: 319L, 307Q, 259I, 252Y, 259I/434S, 428L/434S, 308F/434S, 252Y/254T/256E/434S, 252Y/254T/256E/308F, 252Y/254T/256E/428L, 308F/428L/434S, 259I/308F/434S, 307Q/308F/434S, 250I/308F/434S, 308F/319L/434S, 259I/308F/428L, 259I/307Q/308F, 250I/259I/308F, 259I/308F/319L, 307Q/308F/309Y, 307Q/308F/319L, and 250Q/308F/428L.
13. An antibody or immunoadhesin according to claim 1 or 2, wherein said Fc region comprises modifications selected from the group consisting of: 250I, 250V, 252Q, 252Y, 254T, 256V, 259I, 307P, 307Q, 307S, 308F, 309N, 309Y, 311 P, 319F, 319L, 428L, and 434S.
14. An antibody or immunoadhesin according to claim 1 or 2, wherein said Fc region comprises modifications selected from the group consisting of: 250V/308F, 250I/308F, 254T/308F, 256V/308F, 259I/308F, 307P/208F, 307Q/308F, 307S/308F, 308F/309Y, 308F/309Y, V308F/311 P, 308F/319L, 308F/319F, 308F/428L, 252Q/308F, M252Y/S254T/T256E, 259I/434S, 428L/434S, 308F/434S, 308F/428L/434S, 259I/308F/434S, 307Q/308F/434S, 250I/308F/434S, 308F/319L/434S, 259I/308F/428L, 259I/307Q/308F, 250I/259I/308F, 259I/308F/319L, 307Q/308F/309Y, 307Q/308F/319L, and 250Q/308F/428L.
15. A method of treating a patient in need of said treatment comprising administering an effective amount of an antibody or immunoadhesin of claim 1 or 2.
16. A method increasing the half-life of an antibody or immunoadhesin comprising modifying an antibody or immunoadhesin of claim 1 or 2.
17. An antibody or immunoadhesin according to claim 1 or 2, wherein said Fc region comprises 259I.
18. An antibody or immunoadhesin according to claim 1 or 2, wherein said Fc region comprises 319L.
19. An antibody or immunoadhesin according to claim 1 or 2, wherein said Fc region comprises 307S.
69
0. An antibody of immunoadhesin according to claim 1 or 2, wherein said Fc region comprises a modification selected from the group consisting of: 252Y, 254T, 256E, 259I, 307Q, 308F, 311V, 380A, 428L, 434A, and 434S.
70
PCT/US2008/077250 2007-10-31 2008-09-22 Fc variants with altered binding to fcrn WO2009058492A2 (en)

Priority Applications (13)

Application Number Priority Date Filing Date Title
DK08846103.3T DK2212354T3 (en) 2007-10-31 2008-09-22 Fc variants with modified binding to FcRn
CN202210438504.6A CN114891093A (en) 2007-10-31 2008-09-22 Fc variants with altered binding to FcRn
BRPI0818746-0A BRPI0818746A2 (en) 2007-10-31 2008-09-22 Fc variants with connection changed to fcrn
CN200880113962.0A CN101970492B (en) 2007-10-31 2008-09-22 With the Fc variants of FcRn Binding changes
ES08846103.3T ES2537202T3 (en) 2007-10-31 2008-09-22 Fc variants with altered FcRn binding
JP2010531108A JP5542677B2 (en) 2007-10-31 2008-09-22 Fc variant having mutant binding to FcRn
EP16185391.6A EP3138853B1 (en) 2007-10-31 2008-09-22 Fc variants with altered binding to fcrn
CA2703385A CA2703385C (en) 2007-10-31 2008-09-22 Fc variants with altered binding to fcrn
RU2010121898/10A RU2517621C2 (en) 2007-10-31 2008-09-22 Fc-VERSIONS WITH ALTERED BINDING WITH FcRn
AU2008319144A AU2008319144B2 (en) 2007-10-31 2008-09-22 Fc variants with altered binding to FcRn
EP08846103.3A EP2212354B1 (en) 2007-10-31 2008-09-22 Fc variants with altered binding to fcrn
IL205050A IL205050A0 (en) 2007-10-31 2010-04-13 Fc VARIANTS WITH ALTERED BINDING TO FcRn
IL231423A IL231423A (en) 2007-10-31 2014-03-09 Fc variants with altered binding to fcrn

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US11/932,151 US8802820B2 (en) 2004-11-12 2007-10-31 Fc variants with altered binding to FcRn
US11/932,151 2007-10-31

Publications (2)

Publication Number Publication Date
WO2009058492A2 true WO2009058492A2 (en) 2009-05-07
WO2009058492A3 WO2009058492A3 (en) 2009-07-23

Family

ID=40197140

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2008/077250 WO2009058492A2 (en) 2007-10-31 2008-09-22 Fc variants with altered binding to fcrn

Country Status (13)

Country Link
US (4) US8802820B2 (en)
EP (4) EP3138853B1 (en)
JP (2) JP5542677B2 (en)
KR (1) KR101573883B1 (en)
CN (3) CN101970492B (en)
AU (1) AU2008319144B2 (en)
BR (1) BRPI0818746A2 (en)
CA (1) CA2703385C (en)
DK (2) DK2444423T3 (en)
ES (2) ES2537282T3 (en)
IL (2) IL205050A0 (en)
RU (2) RU2517621C2 (en)
WO (1) WO2009058492A2 (en)

Cited By (86)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010045193A1 (en) * 2008-10-14 2010-04-22 Genentech, Inc. Immunoglobulin variants and uses thereof
WO2010070346A2 (en) 2008-12-18 2010-06-24 Medimmune Limited BINDING MEMBERS FOR INTERLEUKIN-4 RECEPTOR ALPHA (IL-4Ra) - 836
WO2012020096A1 (en) 2010-08-13 2012-02-16 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
EP2444423A1 (en) 2007-10-31 2012-04-25 Xencor Inc. Fc variants with altered binding to FcRn
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
WO2013047752A1 (en) 2011-09-30 2013-04-04 中外製薬株式会社 Antigen-binding molecule for promoting loss of antigens
WO2013070565A1 (en) 2011-11-07 2013-05-16 Medimmune, Llc Multispecific and multivalent binding proteins and uses thereof
WO2013078191A1 (en) 2011-11-23 2013-05-30 Medimmune, Llc Binding molecules specific for her3 and uses thereof
WO2013096221A1 (en) * 2011-12-21 2013-06-27 Amgen Inc. Variant fc-polypeptides with enhanced binding to the neonatal fc receptor
WO2013125667A1 (en) 2012-02-24 2013-08-29 中外製薬株式会社 ANTIGEN-BINDING MOLECULE FOR PROMOTING DISAPPEARANCE OF ANTIGEN VIA FcγRIIB
WO2014030728A1 (en) 2012-08-24 2014-02-27 中外製薬株式会社 Fcγriib-specific fc region variant
US8674083B2 (en) 1999-01-15 2014-03-18 Genentech, Inc. Polypeptide variants with altered effector function
WO2014163101A1 (en) 2013-04-02 2014-10-09 中外製薬株式会社 Fc region variant
US8969526B2 (en) 2011-03-29 2015-03-03 Roche Glycart Ag Antibody Fc variants
EP2845865A1 (en) * 2004-11-12 2015-03-11 Xencor Inc. Fc variants with altered binding to FcRn
EP2854851A2 (en) 2012-06-01 2015-04-08 Momenta Pharmaceuticals, Inc. Methods related to trastuzumab
EP2855745A2 (en) 2012-06-01 2015-04-08 Momenta Pharmaceuticals, Inc. Methods related to adalimumab
EP2856159A2 (en) 2012-06-01 2015-04-08 Momenta Pharmaceuticals, Inc. Methods related to denosumab
EP2856158A2 (en) 2012-06-01 2015-04-08 Momenta Pharmaceuticals, Inc. Methods related to rituximab
WO2015050959A1 (en) 2013-10-01 2015-04-09 Yale University Anti-kit antibodies and methods of use thereof
WO2015051010A1 (en) 2013-10-02 2015-04-09 Medimmune, Llc Neutralizing anti-influenza a antibodies and uses thereof
EP2861068A2 (en) 2012-06-01 2015-04-22 Momenta Pharmaceuticals, Inc. Methods related to bevacizumab
US9079949B1 (en) 2014-03-07 2015-07-14 Alexion Pharmaceuticals, Inc. Anti-C5 antibodies having improved pharmacokinetics
WO2015157592A1 (en) 2014-04-11 2015-10-15 Medimmune, Llc Bispecific her2 antibodies
AU2013203391B2 (en) * 2008-10-14 2016-03-03 Genentech, Inc. Immunoglobulin variants and uses thereof
WO2016050867A1 (en) 2014-10-01 2016-04-07 Medimmune Limited Antibodies to ticagrelor and methods of use
WO2017019846A1 (en) 2015-07-30 2017-02-02 Macrogenics, Inc. Pd-1-binding molecules and methods use thereof
WO2017106061A1 (en) 2015-12-14 2017-06-22 Macrogenics, Inc. Bispecific molecules having immunoreactivity with pd-1 and ctla-4, 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
WO2017142928A1 (en) 2016-02-17 2017-08-24 Macrogenics, Inc. Ror1-binding molecules, and methods of use thereof
WO2017180813A1 (en) 2016-04-15 2017-10-19 Macrogenics, Inc. Novel b7-h3 binding molecules, antibody drug conjugates thereof and methods of use thereof
US9828429B2 (en) 2007-09-26 2017-11-28 Chugai Seiyaku Kabushiki Kaisha Method of modifying isoelectric point of antibody via amino acid substitution in CDR
US9868948B2 (en) 2008-04-11 2018-01-16 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding to two or more antigen molecules repeatedly
US9879249B2 (en) 2009-02-17 2018-01-30 Redwood Bioscience, Inc. Aldehyde-tagged protein-based drug carriers and methods of use
WO2018052556A1 (en) * 2016-08-02 2018-03-22 Visterra, Inc. Engineered polypeptides and uses thereof
EP3142700A4 (en) * 2014-05-16 2018-05-02 Medimmune, LLC Molecules with altered neonate fc receptor binding having enhanced therapeutic and diagnostic properties
US9969800B2 (en) 2015-02-05 2018-05-15 Chugai Seiyaku Kabushiki Kaisha IL-8 antibodies
US10000560B2 (en) 2014-12-19 2018-06-19 Chugai Seiyaku Kabushiki Kaisha Anti-myostatin antibodies, polypeptides containing variant Fc regions, and methods of use
WO2018119196A1 (en) 2016-12-23 2018-06-28 Immunogen, Inc. Immunoconjugates targeting adam9 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
US10253100B2 (en) 2011-09-30 2019-04-09 Chugai Seiyaku Kabushiki Kaisha Therapeutic antigen-binding molecule with a FcRn-binding domain that promotes antigen clearance
JP2019178134A (en) * 2010-03-30 2019-10-17 中外製薬株式会社 ANTIBODIES WITH MODIFIED AFFINITY TO FcRn THAT PROMOTE ANTIGEN CLEARANCE
EP3575317A1 (en) * 2007-12-26 2019-12-04 Xencor, Inc. Fc variants with altered binding to fcrn
WO2020005945A1 (en) 2018-06-26 2020-01-02 Immunogen, Inc. Immunoconjugates targeting adam9 and methods of use thereof
US10538583B2 (en) 2017-10-31 2020-01-21 Staten Biotechnology B.V. Anti-APOC3 antibodies and compositions thereof
US10604561B2 (en) 2016-09-16 2020-03-31 Chugai Seiyaku Kabushiki Kaisha Anti-dengue virus antibodies, polypeptides containing variant Fc regions, and methods of use
US10618965B2 (en) 2011-02-25 2020-04-14 Chugai Seiyaku Kabushiki Kaisha Method for altering plasma retention and immunogenicity of antigen-binding molecule
WO2020086665A1 (en) 2018-10-26 2020-04-30 Immunogen, Inc. Epcam antibodies, activatable antibodies, and immunoconjugates, and uses thereof
US10669344B2 (en) 2016-08-12 2020-06-02 Janssen Biotech, Inc. Engineered antibodies and other Fc-domain containing molecules with enhanced agonism and effector functions
US10689447B2 (en) 2011-02-04 2020-06-23 Genentech, Inc. Fc variants and methods for their production
EP3608339A4 (en) * 2017-04-07 2020-12-23 Kookmin University Industry Academy Cooperation Foundation Antibody fc variants for improving blood half-life
US10899846B2 (en) 2014-11-06 2021-01-26 Hoffmann-La Roche Inc. Fc-region variants with modified FcRn- and protein A-binding properties
EP3634486A4 (en) * 2017-06-05 2021-03-31 Janssen Biotech, Inc. Engineered multispecific antibodies and other multimeric proteins with asymmetrical ch2-ch3 region mutations
WO2021122733A1 (en) 2019-12-18 2021-06-24 F. Hoffmann-La Roche Ag Bispecific anti-ccl2 antibodies
US11046784B2 (en) 2006-03-31 2021-06-29 Chugai Seiyaku Kabushiki Kaisha Methods for controlling blood pharmacokinetics of antibodies
US11053308B2 (en) 2016-08-05 2021-07-06 Chugai Seiyaku Kabushiki Kaisha Method for treating IL-8-related diseases
US11072653B2 (en) 2015-06-08 2021-07-27 Macrogenics, Inc. LAG-3-binding molecules and methods of use thereof
US11091541B2 (en) 2013-04-29 2021-08-17 Hoffmann-La Roche Inc. Human FcRn-binding modified antibodies and methods of use
US11091539B2 (en) 2016-07-08 2021-08-17 Staten Biotechnology B.V. Anti-ApoC3 antibodies and methods of use thereof
US11174315B2 (en) 2015-10-08 2021-11-16 Macrogenics, Inc. Combination therapy for the treatment of cancer
WO2021234160A2 (en) 2020-05-22 2021-11-25 Formycon Ag Ace2 fusion proteins and uses thereof
US11208632B2 (en) 2016-04-26 2021-12-28 R.P. Scherer Technologies, Llc Antibody conjugates and methods of making and using the same
WO2022010798A1 (en) 2020-07-06 2022-01-13 Kiromic BioPharma, Inc. Mesothelin isoform binding molecules and chimeric pd1 receptor molecules, cells containing the same and uses thereof
US11236168B2 (en) 2012-08-24 2022-02-01 Chugai Seiyaku Kabushiki Kaisha Mouse FcγammaRII-specific Fc antibody
US11242381B2 (en) 2017-04-21 2022-02-08 Staten Biotechnology B.V. Anti-ApoC3 antibodies and methods of use thereof
US11242402B2 (en) 2016-12-23 2022-02-08 Macrogenics, Inc. ADAM9-binding molecules, and methods of use thereof
US11248041B2 (en) 2017-10-31 2022-02-15 Staten Biotechnology B.V. Anti-ApoC3 antibodies
WO2022090469A2 (en) 2020-10-29 2022-05-05 Formycon Ag Ace2 fusion proteins and uses thereof
US11332533B2 (en) 2007-09-26 2022-05-17 Chugai Seiyaku Kabushiki Kaisha Modified antibody constant region
WO2022108627A1 (en) 2020-11-18 2022-05-27 Kiromic Biopharma, Inc.Kiromic Biopharma, Inc. Gamma-delta t cell manufacturing processes and chimeric pd1 receptor molecules
US11359009B2 (en) 2015-12-25 2022-06-14 Chugai Seiyaku Kabushiki Kaisha Anti-myostatin antibodies and methods of use
US11365241B2 (en) 2017-07-27 2022-06-21 Alexion Pharmaceuticals, Inc. High concentration anti-C5 antibody formulations
WO2022184854A2 (en) 2021-03-03 2022-09-09 Formycon Ag Formulations of ace2 fc fusion proteins
US11459394B2 (en) 2017-02-24 2022-10-04 Macrogenics, Inc. Bispecific binding molecules that are capable of binding CD137 and tumor antigens, and uses thereof
WO2022263501A1 (en) 2021-06-18 2022-12-22 F. Hoffmann-La Roche Ag Bispecific anti-ccl2 antibodies
US11555067B2 (en) 2014-01-15 2023-01-17 Hoffmann-La Roche Inc. Fc-region variants with improved protein A-binding
WO2023094571A1 (en) 2021-11-25 2023-06-01 Formycon Ag Stabilization of ace2 fusion proteins
WO2023094507A1 (en) 2021-11-24 2023-06-01 Formycon Ag Improved ace2 fusion proteins
US11685781B2 (en) 2018-02-15 2023-06-27 Macrogenics, Inc. Variant CD3-binding domains and their use in combination therapies for the treatment of disease
EP3994176A4 (en) * 2019-07-02 2023-09-20 Telix International Pty Ltd Antibodies against caix with reduced affinity for the neonatal fc receptor
US11795226B2 (en) 2017-12-12 2023-10-24 Macrogenics, Inc. Bispecific CD16-binding molecules and their use in the treatment of disease
US11820793B2 (en) 2011-11-30 2023-11-21 Chugai Seiyaku Kabushiki Kaisha Drug containing carrier into cell for forming immune complex
WO2024015830A1 (en) 2022-07-12 2024-01-18 Cytomx Therapeutics, Inc. Epcam immunoconjugates and uses thereof
US11891434B2 (en) 2010-11-30 2024-02-06 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding to plurality of antigen molecules repeatedly
US11891432B2 (en) 2018-03-15 2024-02-06 Chugai Seiyaku Kabushiki Kaisha Anti-dengue virus antibodies having cross-reactivity to Zika virus and methods of use

Families Citing this family (85)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003035835A2 (en) * 2001-10-25 2003-05-01 Genentech, Inc. Glycoprotein compositions
US20090010920A1 (en) 2003-03-03 2009-01-08 Xencor, Inc. Fc Variants Having Decreased Affinity for FcyRIIb
PT2137655E (en) * 2007-04-16 2012-09-14 Momenta Pharmaceuticals Inc Defined glycoprotein products and related methods
EP2233500A1 (en) * 2009-03-20 2010-09-29 LFB Biotechnologies Optimized Fc variants
AU2015200990B2 (en) * 2009-03-20 2016-07-21 Laboratoire Français Du Fractionnement Et Des Biotechnologies Optimized Fc variants
AU2010268690B2 (en) 2009-07-03 2011-11-10 Avipep Pty Ltd Immuno-conjugates and methods for producing them
WO2011028952A1 (en) 2009-09-02 2011-03-10 Xencor, Inc. Compositions and methods for simultaneous bivalent and monovalent co-engagement of antigens
EP2507381A4 (en) 2009-12-04 2016-07-20 Hoffmann La Roche Multispecific antibodies, antibody analogs, compositions, and methods
US9315581B2 (en) 2009-12-23 2016-04-19 A Vipep Pty Limited Immuno-conjugates and methods for producing them
ES2826894T3 (en) * 2010-02-19 2021-05-19 Xencor Inc New immunoadhesins CTLA4-IG
CN102869784A (en) 2010-04-07 2013-01-09 动量制药公司 High mannose glycans
JP5972871B2 (en) 2010-07-20 2016-08-17 テバ・ファーマシューティカルズ・オーストラリア・ピーティワイ・リミテッド Anti-IL-23 heterodimer specific antibody
WO2012106578A1 (en) * 2011-02-04 2012-08-09 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services HIV NEUTRALIZING ANTIBODIES HAVING MUTATIONS IN CONSTANT DOMAIN (Fc)
WO2012125553A2 (en) 2011-03-12 2012-09-20 Momenta Pharmaceuticals, Inc. N-acetylhexosamine-containing n-glycans in glycoprotein products
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
RU2641256C2 (en) 2011-06-30 2018-01-16 Чугаи Сейяку Кабусики Кайся Heterodimerizated polypeptide
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
US9481724B2 (en) * 2011-12-19 2016-11-01 The Rockefeller University hDC-sign binding peptides
EP3539982A3 (en) * 2011-12-23 2020-01-15 Pfizer Inc Engineered antibody constant regions for site-specific conjugation and methods and uses therefor
WO2013102123A2 (en) * 2011-12-28 2013-07-04 Novelmed Therapeutics, Inc. Aglycosylated human antibody and fusion protein and uses thereof
KR102041412B1 (en) * 2011-12-30 2019-11-11 한미사이언스 주식회사 Derivatives of Immunglobulin Fc fragment
AU2013251309B2 (en) 2012-04-27 2017-06-22 Bioatla, Llc Modified antibody regions and uses thereof
AU2013342163B2 (en) * 2012-11-08 2018-08-16 F. Hoffmann-La Roche Ltd IL-6 antagonists and uses thereof
AR094147A1 (en) * 2012-12-27 2015-07-15 Bayer Pharma Aktiengellschaft FUSION POLIPEPTIDES WITH RELAXIN ACTIVITY AND ITS USES
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
US9840559B2 (en) 2013-02-01 2017-12-12 The Regents Of The University Of California Anti-CD83 antibodies and use thereof
TW201444867A (en) * 2013-03-08 2014-12-01 Lilly Co Eli Anti-TNF-anti-IL-17 bispecific antibodies
WO2014149067A1 (en) 2013-03-15 2014-09-25 Momenta Pharmaceuticals, Inc. Methods related to ctla4-fc fusion proteins
RU2019108429A (en) * 2013-04-29 2019-05-06 Ф. Хоффманн-Ля Рош Аг MODIFIED ASYMMETRIC ANTIBODIES CONNECTING FC-RECEPTOR AND METHODS OF THEIR APPLICATION
WO2014186310A1 (en) 2013-05-13 2014-11-20 Momenta Pharmaceuticals, Inc. Methods for the treatment of neurodegeneration
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
WO2015057622A1 (en) 2013-10-16 2015-04-23 Momenta Pharmaceuticals, Inc. Sialylated glycoproteins
EP3074039A4 (en) 2013-11-26 2017-10-11 The Brigham and Women's Hospital, Inc. Compositions and methods for modulating an immune response
BR112016012248A2 (en) 2013-11-28 2017-10-17 B Creative Sweden Ab nephropathy treatment method
WO2015089585A1 (en) 2013-12-18 2015-06-25 Csl Limited Method of treating wounds
SI3088419T1 (en) 2013-12-25 2019-01-31 Daiichi Sankyo Company Anti-trop2 antibody-drug conjugate
EP2891664A1 (en) * 2014-01-07 2015-07-08 Technische Universität München Novel methods for the stabilisation of immunoglobulin constant domains
KR20230162159A (en) 2014-01-31 2023-11-28 다이이찌 산쿄 가부시키가이샤 Anti-her2 antibody-drug conjugate
EP3130608B1 (en) * 2014-04-10 2019-09-04 Daiichi Sankyo Co., Ltd. (anti-her2 antibody)-drug conjugate
WO2015175375A1 (en) 2014-05-13 2015-11-19 Short Jay M Conditionally active biological proteins
EA202193002A2 (en) 2014-09-03 2022-03-31 Бёрингер Ингельхайм Интернациональ Гмбх COMPOUND TARGETED TO IL-23A AND TNF-ALFA AND ITS APPLICATION
USD801353S1 (en) * 2014-09-11 2017-10-31 Express Scripts, Inc. Display screen with a graphical user interface
USD791797S1 (en) * 2014-09-11 2017-07-11 Express Scripts, Inc. Display screen with a graphical user interface
USD791796S1 (en) * 2014-09-11 2017-07-11 Express Scripts, Inc. Display screen with a graphical user interface
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
NZ731491A (en) 2014-10-23 2021-12-24 Kira Biotech Pty Ltd Cd83 binding proteins and uses thereof
SG11201703390SA (en) * 2014-10-27 2017-05-30 Inhibrx Lp Serpin fusion polypeptides and methods of use thereof
EP3632931A1 (en) 2014-11-07 2020-04-08 Sesen Bio, Inc. Improved il-6 antibodies
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
US10196445B1 (en) * 2015-03-17 2019-02-05 Bristol-Myers Squibb Company Ipilimumab variant with enhanced ADCC
CA2979708A1 (en) 2015-03-19 2016-09-22 Duke University Hiv-1 neutralizing antibodies and uses thereof
WO2016149698A2 (en) 2015-03-19 2016-09-22 Duke University Hiv-1 neutralizing antibodies and uses thereof (v3 antibodies)
US10450368B2 (en) 2015-03-19 2019-10-22 Duke University HIV-1 neutralizing antibodies and uses thereof (CD4bs antibodies)
WO2016149710A2 (en) 2015-03-19 2016-09-22 Duke University Hiv-1 neutralizing antibodies and uses thereof
CN116059395A (en) 2015-06-29 2023-05-05 第一三共株式会社 Method for selectively producing antibody-drug conjugates
SG11201801984TA (en) * 2015-10-01 2018-04-27 Heat Biologics Inc Compositions and methods for adjoining type i and type ii extracellular domains as heterologous chimeric proteins
JP2019506363A (en) 2015-11-25 2019-03-07 ビステラ, インコーポレイテッド Antibody molecules against APRIL and uses thereof
EP3395835B1 (en) * 2015-12-25 2021-02-03 Chugai Seiyaku Kabushiki Kaisha Antibody having enhanced activity, and method for modifying same
EP3243836A1 (en) * 2016-05-11 2017-11-15 F. Hoffmann-La Roche AG C-terminally fused tnf family ligand trimer-containing antigen binding molecules
WO2017210684A1 (en) * 2016-06-03 2017-12-07 New York University Methods and reagents for modulating macrophage phenotype
TW202300515A (en) * 2016-10-20 2023-01-01 法商賽諾菲公司 Anti-chikv antibodies and uses thereof
CA3046293A1 (en) 2016-12-12 2018-06-21 Daiichi Sankyo Company, Limited Combination of antibody-drug conjugate and immune checkpoint inhibitor
WO2018129713A1 (en) * 2017-01-13 2018-07-19 杭州翰思生物医药有限公司 Method for improving binding affinity of igg antibody to fcrn and prolonging serum half-life period thereof
TW202330036A (en) 2017-05-15 2023-08-01 日商第一三共股份有限公司 Manufacturing method of antibody-drug conjugates
EP3634994A4 (en) 2017-06-05 2021-06-30 Janssen Biotech, Inc. Methods of engineering surface charge for bispecific antibody production
AU2018327171B2 (en) 2017-08-31 2023-03-09 Daiichi Sankyo Company, Limited Improved method for producing antibody-drug conjugate
BR112020003466B1 (en) 2017-08-31 2023-12-12 Daiichi Sankyo Company, Limited COMPOST PRODUCTION METHODS, AND, COMPOST
US11364303B2 (en) 2017-09-29 2022-06-21 Pfizer Inc. Cysteine engineered antibody drug conjugates
TW201940512A (en) * 2018-01-26 2019-10-16 美商健臻公司 Fc variants with enhanced binding to FcRn and prolonged half-life
JP2018138022A (en) * 2018-02-23 2018-09-06 ゲンマブ ビー.ブイ. Human igg1 fc region variants and uses thereof
CN112236448A (en) * 2018-04-17 2021-01-15 海德堡生物技术有限公司 Means and methods for treating angiogenesis, fibrosis and cancer related diseases with protein oligomers comprising NC-1-Fc
BR112020023373A2 (en) 2018-05-18 2021-02-09 Daiichi Sankyo Company, Limited conjugate, composition, and, use of a conjugate or composition
WO2019232323A1 (en) * 2018-05-31 2019-12-05 Board Of Regents, The University Of Texas System Bi-specific antibodies and use thereof
AU2019403245A1 (en) * 2018-12-19 2021-07-22 Humabs Biomed Sa Antibodies that neutralize hepatitis B virus and uses thereof
WO2020167957A1 (en) * 2019-02-12 2020-08-20 Board Of Regents, The University Of Texas System High affinity engineered t-cell receptors targeting cmv infected cells
WO2020221450A1 (en) * 2019-04-30 2020-11-05 Humabs Biomed Sa Antibodies and methods for treatment of influenza a infection
WO2021000018A1 (en) * 2019-07-02 2021-01-07 Telix International Pty Ltd Antibodies for binding psma with reduced affinity for the neonatal fc receptor
WO2021154046A1 (en) * 2020-01-29 2021-08-05 고려대학교 산학협력단 Ph-sensitive fc variant
KR102382593B1 (en) * 2020-01-29 2022-04-05 고려대학교 산학협력단 Ph-sensitive fc variants
CA3188448A1 (en) * 2020-07-10 2022-01-13 William Brondyk Compositions for increasing half-life of a therapeutic agent in felines and methods of use
WO2023173084A1 (en) * 2022-03-11 2023-09-14 University Of Rochester Cyclopeptibodies and uses thereof
CN114853909A (en) * 2022-05-13 2022-08-05 南京吉盛澳玛生物医药有限公司 Design, preparation and application of novel IL-2 and INF alpha and Fc fusion protein

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050037000A1 (en) * 2003-01-09 2005-02-17 Macrogenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
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
WO2006053301A2 (en) * 2004-11-12 2006-05-18 Xencor, Inc. Fc variants with altered binding to fcrn

Family Cites Families (265)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3773919A (en) 1969-10-23 1973-11-20 Du Pont Polylactide-drug mixtures
US3691016A (en) 1970-04-17 1972-09-12 Monsanto Co Process for the preparation of insoluble enzymes
US3917002A (en) 1971-10-15 1975-11-04 Massey Ferguson Inc Draft control linkage for a tractor
CU22545A1 (en) * 1994-11-18 1999-03-31 Centro Inmunologia Molecular OBTAINING A CHEMICAL AND HUMANIZED ANTIBODY AGAINST THE RECEPTOR OF THE EPIDERMAL GROWTH FACTOR FOR DIAGNOSTIC AND THERAPEUTIC USE
CA1023287A (en) 1972-12-08 1977-12-27 Boehringer Mannheim G.M.B.H. Process for the preparation of carrier-bound proteins
US4179337A (en) 1973-07-20 1979-12-18 Davis Frank F Non-immunogenic polypeptides
US4195128A (en) 1976-05-03 1980-03-25 Bayer Aktiengesellschaft Polymeric carrier bound ligands
US4330440A (en) 1977-02-08 1982-05-18 Development Finance Corporation Of New Zealand Activated matrix and method of activation
CA1093991A (en) 1977-02-17 1981-01-20 Hideo Hirohara Enzyme immobilization with pullulan gel
US4229537A (en) 1978-02-09 1980-10-21 New York University Preparation of trichloro-s-triazine activated supports for coupling ligands
JPS6023084B2 (en) 1979-07-11 1985-06-05 味の素株式会社 blood substitute
US4485045A (en) 1981-07-06 1984-11-27 Research Corporation Synthetic phosphatidyl cholines useful in forming liposomes
US4640835A (en) 1981-10-30 1987-02-03 Nippon Chemiphar Company, Ltd. Plasminogen activator derivatives
GB8308235D0 (en) * 1983-03-25 1983-05-05 Celltech Ltd Polypeptides
US4816567A (en) * 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4544545A (en) 1983-06-20 1985-10-01 Trustees University Of Massachusetts Liposomes containing modified cholesterol for organ targeting
US4496689A (en) 1983-12-27 1985-01-29 Miles Laboratories, Inc. Covalently attached complex of alpha-1-proteinase inhibitor with a water soluble polymer
US4753894A (en) * 1984-02-08 1988-06-28 Cetus Corporation Monoclonal anti-human breast cancer antibodies
US4943533A (en) * 1984-03-01 1990-07-24 The Regents Of The University Of California Hybrid cell lines that produce monoclonal antibodies to epidermal growth factor receptor
EP0206448B1 (en) 1985-06-19 1990-11-14 Ajinomoto Co., Inc. Hemoglobin combined with a poly(alkylene oxide)
EP0272253A4 (en) 1986-03-07 1990-02-05 Massachusetts Inst Technology Method for enhancing glycoprotein stability.
US6548640B1 (en) * 1986-03-27 2003-04-15 Btg International Limited Altered antibodies
US5225539A (en) * 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
ATE147432T1 (en) 1986-05-29 1997-01-15 Ilexus Pty Ltd POLYNUCLEOTIDE SEQUENCES CODING FOR THE HUMAN FC RECEPTOR FOR IMMUNOGLOBULIN
US4791192A (en) 1986-06-26 1988-12-13 Takeda Chemical Industries, Ltd. Chemically modified protein with polyethyleneglycol
IL85035A0 (en) * 1987-01-08 1988-06-30 Int Genetic Eng Polynucleotide molecule,a chimeric antibody with specificity for human b cell surface antigen,a process for the preparation and methods utilizing the same
AU600575B2 (en) 1987-03-18 1990-08-16 Sb2, Inc. Altered antibodies
US4975278A (en) 1988-02-26 1990-12-04 Bristol-Myers Company Antibody-enzyme conjugates in combination with prodrugs for the delivery of cytotoxic agents to tumor cells
PT88641B (en) 1987-10-02 1993-04-30 Genentech Inc METHOD FOR PREPARING A VARIETY OF ADHESION
JP3040121B2 (en) 1988-01-12 2000-05-08 ジェネンテク,インコーポレイテッド Methods of treating tumor cells by inhibiting growth factor receptor function
US5576184A (en) 1988-09-06 1996-11-19 Xoma Corporation Production of chimeric mouse-human antibodies with specificity to human tumor antigens
US5681566A (en) 1988-10-24 1997-10-28 3I Research Exploitation Limited Antibody conjugates with two or more covalently linked FC regions
US20040049014A1 (en) 1988-12-28 2004-03-11 Protein Design Labs, Inc. Humanized immunoglobulins
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
JP3051411B2 (en) * 1989-03-14 2000-06-12 持田製薬株式会社 Novel DNA and expression plasmid containing it
US5266491A (en) 1989-03-14 1993-11-30 Mochida Pharmaceutical Co., Ltd. DNA fragment and expression plasmid containing the DNA fragment
US5683888A (en) 1989-07-22 1997-11-04 University Of Wales College Of Medicine Modified bioluminescent proteins and their use
US5208020A (en) * 1989-10-25 1993-05-04 Immunogen Inc. Cytotoxic agents comprising maytansinoids and their therapeutic use
EP0462246A4 (en) 1989-11-07 1992-11-25 Bristol-Myers Squibb Company Oligomeric immunoglobulins
US5859205A (en) * 1989-12-21 1999-01-12 Celltech Limited Humanised antibodies
US5292658A (en) 1989-12-29 1994-03-08 University Of Georgia Research Foundation, Inc. Boyd Graduate Studies Research Center Cloning and expressions of Renilla luciferase
WO1991019515A1 (en) 1990-06-21 1991-12-26 The Board Of Trustees Of The Leland Stanford Junior University Oligomeric immunoglobulin constant domain mutant with enhanced complement-mediated cytolytic activity
US5198342A (en) * 1990-07-05 1993-03-30 Immunex Corporation DNA encoding IgA Fc receptors
EP0547137A4 (en) 1990-08-31 1993-12-08 Bristol-Myers Squibb Company Homoconjugated immunoglobulins
GB9022545D0 (en) * 1990-10-17 1990-11-28 Wellcome Found Culture medium
CZ282603B6 (en) 1991-03-06 1997-08-13 Merck Patent Gesellschaft Mit Beschränkter Haftun G Humanized and chimeric monoclonal antibody, expression vector and pharmaceutical preparation
DK0575319T3 (en) 1991-03-11 2000-07-10 Univ Georgia Res Found Cloning and expression of Renilla luciferase
GB9105245D0 (en) 1991-03-12 1991-04-24 Lynxvale Ltd Binding molecules
US6797492B2 (en) 1991-05-17 2004-09-28 Merck & Co., Inc. Method for reducing the immunogenicity of antibody variable domains
WO1994004679A1 (en) * 1991-06-14 1994-03-03 Genentech, Inc. Method for making humanized antibodies
EP0590067A1 (en) 1991-06-14 1994-04-06 Xoma Corporation Microbially-produced antibody fragments and their conjugates
LU91067I2 (en) * 1991-06-14 2004-04-02 Genentech Inc Trastuzumab and its variants and immunochemical derivatives including immotoxins
US5264586A (en) 1991-07-17 1993-11-23 The Scripps Research Institute Analogs of calicheamicin gamma1I, method of making and using the same
MX9204374A (en) 1991-07-25 1993-03-01 Idec Pharma Corp RECOMBINANT ANTIBODY AND METHOD FOR ITS PRODUCTION.
US6329509B1 (en) * 1991-08-14 2001-12-11 Genentech, Inc. Anti-IgE antibodies
EP0604580A1 (en) 1991-09-19 1994-07-06 Genentech, Inc. EXPRESSION IN E. COLI OF ANTIBODY FRAGMENTS HAVING AT LEAST A CYSTEINE PRESENT AS A FREE THIOL, USE FOR THE PRODUCTION OF BIFUNCTIONAL F(ab') 2? ANTIBODIES
US5623053A (en) * 1992-01-10 1997-04-22 California Institute Of Technology Soluble mammal-derived Fc receptor which binds at a pH ranging from about 5.5 to 6.5 and releases at a pH ranging from about 7.5 to 8.5
US5714350A (en) 1992-03-09 1998-02-03 Protein Design Labs, Inc. Increasing antibody affinity by altering glycosylation in the immunoglobulin variable region
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
AU690528B2 (en) 1992-12-04 1998-04-30 Medical Research Council Multivalent and multispecific binding proteins, their manufacture and use
US20030108548A1 (en) * 1993-06-01 2003-06-12 Bluestone Jeffrey A. Methods and materials for modulation of the immunosuppressive activity and toxicity of monoclonal antibodies
US5885573A (en) * 1993-06-01 1999-03-23 Arch Development Corporation Methods and materials for modulation of the immunosuppressive activity and toxicity of monoclonal antibodies
EP0714409A1 (en) 1993-06-16 1996-06-05 Celltech Therapeutics Limited Antibodies
GB9316989D0 (en) 1993-08-16 1993-09-29 Lynxvale Ltd Binding molecules
WO1995007463A1 (en) 1993-09-10 1995-03-16 The Trustees Of Columbia University In The City Of New York Uses of green fluorescent protein
GB9401182D0 (en) 1994-01-21 1994-03-16 Inst Of Cancer The Research Antibodies to EGF receptor and their antitumour effect
WO1995021191A1 (en) 1994-02-04 1995-08-10 William Ward Bioluminescent indicator based upon the expression of a gene for a modified green-fluorescent protein
US6204007B1 (en) 1994-03-29 2001-03-20 Celltech Therapeutics Limited Antibodies against E-selectin
US5773001A (en) * 1994-06-03 1998-06-30 American Cyanamid Company Conjugates of methyltrithio antitumor agents and intermediates for their synthesis
US5541087A (en) * 1994-09-14 1996-07-30 Fuji Immunopharmaceuticals Corporation Expression and export technology of proteins as immunofusins
US5804396A (en) 1994-10-12 1998-09-08 Sugen, Inc. Assay for agents active in proliferative disorders
US5777079A (en) 1994-11-10 1998-07-07 The Regents Of The University Of California Modified green fluorescent proteins
ATE238668T1 (en) 1995-01-17 2003-05-15 Brigham & Womens Hospital RECEPTOR-SPECIFIC TRANSEPITHELIAL TRANSPORT OF IMMUNOGENS
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
US6485726B1 (en) 1995-01-17 2002-11-26 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
US6121022A (en) 1995-04-14 2000-09-19 Genentech, Inc. Altered polypeptides with increased half-life
US6096871A (en) * 1995-04-14 2000-08-01 Genentech, Inc. Polypeptides altered to contain an epitope from the Fc region of an IgG molecule for increased half-life
US6444789B1 (en) 1995-05-03 2002-09-03 Applied Research Systems Ars Holding N.V. CD16-II variants
US6365161B1 (en) * 1995-06-07 2002-04-02 Medarex, Inc. Therapeutic compounds comprised of anti-FC receptor binding agents
US5714586A (en) * 1995-06-07 1998-02-03 American Cyanamid Company Methods for the preparation of monomeric calicheamicin derivative/carrier conjugates
US5712374A (en) * 1995-06-07 1998-01-27 American Cyanamid Company Method for the preparation of substantiallly monomeric calicheamicin derivative/carrier conjugates
JPH11507535A (en) 1995-06-07 1999-07-06 イムクローン システムズ インコーポレイテッド Antibodies and antibody fragments that suppress tumor growth
US5874304A (en) 1996-01-18 1999-02-23 University Of Florida Research Foundation, Inc. Humanized green fluorescent protein genes and methods
US5804387A (en) 1996-02-01 1998-09-08 The Board Of Trustees Of The Leland Stanford Junior University FACS-optimized mutants of the green fluorescent protein (GFP)
US6750334B1 (en) 1996-02-02 2004-06-15 Repligen Corporation CTLA4-immunoglobulin fusion proteins having modified effector functions and uses therefor
US5876995A (en) 1996-02-06 1999-03-02 Bryan; Bruce Bioluminescent novelty items
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
JP3904238B2 (en) 1996-03-20 2007-04-11 イムノメディクス, インコーポレイテッド Glycosylated humanized B cell specific antibody
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
US5925558A (en) 1996-07-16 1999-07-20 The Regents Of The University Of California Assays for protein kinases using fluorescent protein substrates
EP0918872B1 (en) 1996-08-02 2008-02-20 Bristol-Myers Squibb Company A method for inhibiting immunoglobulin-induced toxicity resulting from the use of immunoglobulins in therapy and in vivo diagnosis
US5976796A (en) 1996-10-04 1999-11-02 Loma Linda University Construction and expression of renilla luciferase and green fluorescent protein fusion genes
WO1998023289A1 (en) 1996-11-27 1998-06-04 The General Hospital Corporation MODULATION OF IgG BINDING TO FcRn
IL129767A0 (en) 1996-12-12 2000-02-29 Prolume Ltd Apparatus and method for detecting and identifying infectious agents
WO1998031806A2 (en) 1997-01-21 1998-07-23 Human Genome Sciences, Inc. Fc RECEPTORS AND POLYPEPTIDES
US6277375B1 (en) 1997-03-03 2001-08-21 Board Of Regents, The University Of Texas System Immunoglobulin-like domains with increased half-lives
JP2002510966A (en) 1997-04-11 2002-04-09 カリフォルニア・インスティテュート・オブ・テクノロジー Apparatus and method for automatic protein design
EP1255209A3 (en) 1997-04-11 2009-02-11 California Institute Of Technology Apparatus and method for automated protein design
WO1998048032A2 (en) 1997-04-21 1998-10-29 Donlar Corporation POLY-(α-L-ASPARTIC ACID), POLY-(α-L-GLUTAMIC ACID) AND COPOLYMERS OF L-ASP AND L-GLU, METHOD FOR THEIR PRODUCTION AND THEIR USE
US20020062010A1 (en) * 1997-05-02 2002-05-23 Genentech, Inc. Method for making multispecific antibodies having heteromultimeric and common components
US6235883B1 (en) * 1997-05-05 2001-05-22 Abgenix, Inc. Human monoclonal antibodies to epidermal growth factor receptor
GB2339430A (en) 1997-05-21 2000-01-26 Biovation Ltd Method for the production of non-immunogenic proteins
GB9712892D0 (en) 1997-06-20 1997-08-20 Eclagen Ltd Identification of mhc binding peptides
US20040191256A1 (en) 1997-06-24 2004-09-30 Genentech, Inc. Methods and compositions for galactosylated glycoproteins
US20030105294A1 (en) * 1998-02-25 2003-06-05 Stephen Gillies Enhancing the circulating half life of antibody-based fusion proteins
US6232107B1 (en) 1998-03-27 2001-05-15 Bruce J. Bryan Luciferases, fluorescent proteins, nucleic acids encoding the luciferases and fluorescent proteins and the use thereof in diagnostics, high throughput screening and novelty items
US6528624B1 (en) 1998-04-02 2003-03-04 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
US6194551B1 (en) 1998-04-02 2001-02-27 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.
AU3655899A (en) 1998-04-20 1999-11-08 Regents Of The University Of California, The Modified immunoglobulin molecules and methods for use thereof
AU3871099A (en) 1998-05-06 1999-11-23 Temple University - Of The Commonwealth System Of Higher Education Reversal of proinflammatory response by ligating the macrophage fcgammari receptor
GB9809951D0 (en) 1998-05-08 1998-07-08 Univ Cambridge Tech Binding molecules
GB9815157D0 (en) 1998-07-13 1998-09-09 Metron Designs Ltd High resolution pulse width setting from relatively low frequency clocks
CA2341029A1 (en) 1998-08-17 2000-02-24 Abgenix, Inc. Generation of modified molecules with increased serum half-lives
DE19845202A1 (en) * 1998-10-01 2000-04-06 Wella Ag Hair restorer
US6306926B1 (en) 1998-10-07 2001-10-23 3M Innovative Properties Company Radiopaque cationically polymerizable compositions comprising a radiopacifying filler, and method for polymerizing same
US7315786B2 (en) * 1998-10-16 2008-01-01 Xencor Protein design automation for protein libraries
WO2000023564A2 (en) 1998-10-16 2000-04-27 Xencor, Inc. Protein design automation for protein libraries
US20020048772A1 (en) * 2000-02-10 2002-04-25 Dahiyat Bassil I. Protein design automation for protein libraries
US20030049654A1 (en) * 1998-10-16 2003-03-13 Xencor Protein design automation for protein libraries
US6403312B1 (en) * 1998-10-16 2002-06-11 Xencor Protein design automatic for protein libraries
US6660843B1 (en) 1998-10-23 2003-12-09 Amgen Inc. Modified peptides as therapeutic agents
US6737056B1 (en) 1999-01-15 2004-05-18 Genentech, Inc. Polypeptide variants with altered effector function
WO2000042072A2 (en) * 1999-01-15 2000-07-20 Genentech, Inc. Polypeptide variants with altered effector function
US6221556B1 (en) 1999-03-05 2001-04-24 General Electric Company Article for optical data storage device
EP1176195B1 (en) 1999-04-09 2013-05-22 Kyowa Hakko Kirin Co., Ltd. Method for controlling the activity of immunologically functional molecule
US6649165B2 (en) 1999-09-07 2003-11-18 Walter Schubert Method of blocking cytotoxic activity in patients with amyotrophic lateral sclerosis using soluble FcγRIII receptors
GB9922283D0 (en) 1999-09-22 1999-11-17 Kennedy Rheumatology Inst Adenoviral vectors
JP4668498B2 (en) 1999-10-19 2011-04-13 協和発酵キリン株式会社 Method for producing polypeptide
ES2581239T3 (en) 1999-11-29 2016-09-02 The Trustees Of Columbia University In The City Of New York Isolation of five novel genes encoding new Fc receptor-type melanomas involved in the pathogenesis of lymphoma / melanoma
CN1163506C (en) 1999-12-10 2004-08-25 上海华晨生物技术研究所 New human cell factor and its code sequence and use
WO2001057088A1 (en) 2000-02-03 2001-08-09 Hammarstroem Lennart RUMINANT MHC CLASS I-LIKE Fc RECEPTORS
EP1255826B1 (en) 2000-02-10 2005-09-14 Xencor Protein design automation for protein libraries
EP1259544B1 (en) * 2000-02-11 2011-08-24 Biogen Idec MA Inc. Heterologous polypeptide of the tnf family
US7129332B2 (en) 2000-02-25 2006-10-31 The United States Of America As Represented By The Department Of Health And Human Services Anti-EGFRvIII scFvs with improved cytotoxicity and yield, immunotoxins based thereon, and methods of use thereof
US7687461B2 (en) 2000-03-02 2010-03-30 Xencor, Inc. Treatment of TNF-α related disorders with TNF-α variant proteins
US7101974B2 (en) 2000-03-02 2006-09-05 Xencor TNF-αvariants
US7056695B2 (en) * 2000-03-02 2006-06-06 Xencor TNF-α variants
US7244823B2 (en) 2000-03-02 2007-07-17 Xencor TNF-alpha variants proteins for the treatment of TNF-alpha related disorders
US7449443B2 (en) 2000-03-23 2008-11-11 California Institute Of Technology Method for stabilization of proteins using non-natural amino acids
LT2857516T (en) * 2000-04-11 2017-09-11 Genentech, Inc. Multivalent antibodies and uses therefor
AU5345901A (en) 2000-04-13 2001-10-30 Univ Rockefeller Enhancement of antibody-mediated immune responses
US6358733B1 (en) * 2000-05-19 2002-03-19 Apolife, Inc. Expression of heterologous multi-domain proteins in yeast
AU2001258567A1 (en) 2000-05-19 2001-11-26 Scancell Limited Humanised antibodies to the epidermal growth factor receptor
US6586207B2 (en) 2000-05-26 2003-07-01 California Institute Of Technology Overexpression of aminoacyl-tRNA synthetases for efficient production of engineered proteins containing amino acid analogues
EA004875B1 (en) 2000-06-28 2004-08-26 Хамар Бакулиш Мафатлал Use an agent for reversal of drug resistance in mycobacterium tuberculosis
JP2004502946A (en) 2000-07-10 2004-01-29 ゼンコー Protein design automation for designing protein libraries with altered immunogenicity
US20030068649A1 (en) * 2000-09-14 2003-04-10 Doberstein Stephen K. Methods and compositions for the construction and use of fusion libraries
MXPA00009407A (en) 2000-09-26 2004-12-08 Mexicano Inst Petrol Process for the preparation of a zeolitic type monometallic catalyst for obtaining high octane gasoline by means of naphtha reformation.
US6946292B2 (en) 2000-10-06 2005-09-20 Kyowa Hakko Kogyo Co., Ltd. Cells producing antibody compositions with increased antibody dependent cytotoxic activity
PL218428B1 (en) 2000-10-06 2014-12-31 Kyowa Hakko Kogyo Kk Cells producing antibody compositions
US7064191B2 (en) 2000-10-06 2006-06-20 Kyowa Hakko Kogyo Co., Ltd. Process for purifying antibody
AU2001294175A1 (en) 2000-10-06 2002-04-22 Kyowa Hakko Kogyo Co. Ltd. Method of purifying antibody
US7465790B2 (en) 2000-10-09 2008-12-16 Isis Innovation, Inc. Therapeutic antibodies
WO2002043658A2 (en) * 2000-11-06 2002-06-06 The Jackson Laboratory Fcrn-based therapeutics for the treatment of auto-immune disorders
US7235643B2 (en) 2000-11-07 2007-06-26 Morphotek, Inc. Antibodies and methods for generating genetically altered antibodies with high affinity
GB0029407D0 (en) 2000-12-01 2001-01-17 Affitech As Product
US20040253242A1 (en) 2000-12-05 2004-12-16 Bowdish Katherine S. Rationally designed antibodies
EP2341060B1 (en) 2000-12-12 2019-02-20 MedImmune, LLC Molecules with extended half-lives, compositions and uses thereof
WO2002066653A2 (en) 2000-12-14 2002-08-29 Xencor Procaryotic libraries and uses
EP1356052B1 (en) 2000-12-14 2008-08-20 Genentech, Inc. Production of full antibodies in procaryotic cells
US6979556B2 (en) * 2000-12-14 2005-12-27 Genentech, Inc. Separate-cistron contructs for secretion of aglycosylated antibodies from prokaryotes
US7754208B2 (en) * 2001-01-17 2010-07-13 Trubion Pharmaceuticals, Inc. Binding domain-immunoglobulin fusion proteins
US20030133939A1 (en) 2001-01-17 2003-07-17 Genecraft, Inc. Binding domain-immunoglobulin fusion proteins
EP1358337A1 (en) 2001-01-30 2003-11-05 Degussa AG Nucleotide sequences which code for the otsa gene of c. glutamicum
ATE400030T1 (en) 2001-02-19 2008-07-15 Merck Patent Gmbh METHOD FOR IDENTIFYING T CELL PITOPES AND ITS APPLICATION TO PRODUCING MOLECULES WITH REDUCED IMMUNOGENICITY
US20020168640A1 (en) 2001-02-22 2002-11-14 Min Li Biochips comprising nucleic acid/protein conjugates
CN100489094C (en) 2001-03-23 2009-05-20 金克克国际有限公司 Proteins with altered immunogenicity and methods of making and using the same
US6992174B2 (en) 2001-03-30 2006-01-31 Emd Lexigen Research Center Corp. Reducing the immunogenicity of fusion proteins
CA2442801A1 (en) * 2001-04-02 2002-10-10 Idec Pharmaceutical Corporation Recombinant antibodies coexpressed with gntiii
US7117096B2 (en) 2001-04-17 2006-10-03 Abmaxis, Inc. Structure-based selection and affinity maturation of antibody library
CN1463270A (en) 2001-05-31 2003-12-24 梅达莱克斯公司 Disulfide prodrugs and linkers and stablizers useful therefore
AU2002304389A1 (en) 2001-06-11 2002-12-23 Francis Archibald Holtzkampf Fishing accessory, method of securing a lure to a fishing hook, and method of attaching a body to a fishing line
CN100497389C (en) 2001-06-13 2009-06-10 根马布股份公司 Human monoclonal antibodies to epidermal growth factor receptor (EGFR)
GB0118662D0 (en) * 2001-07-31 2001-09-19 Univ Southampton Binding agents
ES2271321T3 (en) 2001-08-03 2007-04-16 Tyco Healthcare Group Lp MARKER TO BE USED WITH A FABRIC MARKING DEVICE.
EP2180044A1 (en) 2001-08-03 2010-04-28 GlycArt Biotechnology AG Antibody glycosylation variants having increased anti-body-dependent cellular cytotoxicity
WO2003014325A2 (en) 2001-08-10 2003-02-20 Xencor Protein design automation for protein libraries
US7255858B2 (en) 2001-08-10 2007-08-14 University Of Virginia Patent Foundation Enhancing the efficacy of immunotherapies by supplementing with complement
WO2003035835A2 (en) 2001-10-25 2003-05-01 Genentech, Inc. Glycoprotein compositions
US6911321B2 (en) 2001-12-19 2005-06-28 Genentech, Inc. Non-human primate Fc receptors and methods of use
US20040093621A1 (en) * 2001-12-25 2004-05-13 Kyowa Hakko Kogyo Co., Ltd Antibody composition which specifically binds to CD20
AU2003216250A1 (en) 2002-02-11 2003-09-04 Genentech, Inc. Antibody variants with faster antigen association rates
US20040002587A1 (en) 2002-02-20 2004-01-01 Watkins Jeffry D. Fc region variants
WO2003073238A2 (en) 2002-02-27 2003-09-04 California Institute Of Technology Computational method for designing enzymes for incorporation of amino acid analogs into proteins
US7662925B2 (en) * 2002-03-01 2010-02-16 Xencor, Inc. Optimized Fc variants and methods for their generation
AU2003217912A1 (en) * 2002-03-01 2003-09-16 Xencor Antibody optimization
US7317091B2 (en) * 2002-03-01 2008-01-08 Xencor, Inc. Optimized Fc variants
US20040132101A1 (en) 2002-09-27 2004-07-08 Xencor Optimized Fc variants and methods for their generation
WO2003077834A2 (en) 2002-03-15 2003-09-25 The Brigham And Women's Hospital, Inc. Central airway administration for systemic delivery of therapeutics
CA2480404A1 (en) 2002-03-25 2003-10-30 Uab Research Foundation Fc receptor homolog, reagents, and uses thereof
US20040259150A1 (en) 2002-04-09 2004-12-23 Kyowa Hakko Kogyo Co., Ltd. Method of enhancing of binding activity of antibody composition to Fcgamma receptor IIIa
JP2006513139A (en) 2002-07-03 2006-04-20 ザ・ブリガーム・アンド・ウーメンズ・ホスピタル・インコーポレーテッド Central airway administration for systemic delivery of therapeutic agents
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
EP2371389A3 (en) 2002-08-14 2012-04-18 MacroGenics, Inc. FcgammaRIIB-specific antibodies and methods of use thereof
WO2004022717A2 (en) 2002-09-05 2004-03-18 The Government Of The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Minimally immunogenic variants of humanized col-1 antibody against carcinoembryonic antigen
EP1539811A4 (en) 2002-09-16 2006-05-24 Elusys Therapeutics Inc Production of bispecific molecules using polyethylene glycol linkers
US20060235208A1 (en) 2002-09-27 2006-10-19 Xencor, Inc. Fc variants with optimized properties
US7365168B2 (en) 2002-10-15 2008-04-29 Pdl Biopharma, Inc. Alteration of FcRn binding affinities or serum half-lives of antibodies by mutagenesis
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
US7361740B2 (en) * 2002-10-15 2008-04-22 Pdl Biopharma, Inc. Alteration of FcRn binding affinities or serum half-lives of antibodies by mutagenesis
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
US8388955B2 (en) 2003-03-03 2013-03-05 Xencor, Inc. Fc variants
US7610156B2 (en) 2003-03-31 2009-10-27 Xencor, Inc. Methods for rational pegylation of proteins
EP1613649A2 (en) * 2003-04-04 2006-01-11 Ingenium Pharmaceuticals AG Spinster-like protein genes, expression products, non-human animal model: uses in human metabolic disorders
TWI353991B (en) 2003-05-06 2011-12-11 Syntonix Pharmaceuticals Inc Immunoglobulin chimeric monomer-dimer hybrids
AR044388A1 (en) 2003-05-20 2005-09-07 Applied Molecular Evolution CD20 UNION MOLECULES
AU2004257142A1 (en) 2003-05-30 2005-01-27 Alexion Pharmaceuticals, Inc. Antibodies and fusion proteins that include engineered constant regions
DK1641483T3 (en) 2003-06-12 2008-06-02 Lilly Co Eli Fusion Proteins
US20040254108A1 (en) 2003-06-13 2004-12-16 Jing Ma Preparation and application of anti-tumor bifunctional fusion proteins
WO2005000899A2 (en) 2003-06-27 2005-01-06 Biogen Idec Ma Inc. Modified binding molecules comprising connecting peptides
CA2531482A1 (en) * 2003-06-30 2005-01-20 Centocor, Inc. Engineered anti-target immunoglobulin derived proteins, compositions, methods and uses
BRPI0412890B8 (en) * 2003-07-24 2021-05-25 Innate Pharma method of selecting an anti-kir2dl1 antibody or antigen-binding antibody fragment
EP1653801A4 (en) 2003-07-26 2007-05-30 Biogen Idec Inc Altered antibodies having improved antigen-binding affinity
EP1660970A4 (en) 2003-08-01 2007-02-14 Dna Twopointo Inc Systems and methods for biopolymer engineering
CA2536408A1 (en) * 2003-08-22 2005-03-03 Biogen Idec Ma Inc. Improved antibodies having altered effector function and methods for making the same
US7652843B2 (en) * 2003-08-28 2010-01-26 Hitachi Global Storage Technologies Netherlands B.V. Completely circumferential motor bracket shroud for motor hub flange outside diameter for hard disk drive
WO2005027966A2 (en) 2003-09-05 2005-03-31 Genentech, Inc. Antibodies with altered effector functions
CA2579635A1 (en) 2003-09-10 2005-03-17 Baxter International Inc. Peptides that inhibit complement activation
ES2831379T3 (en) 2003-10-09 2021-06-08 Ambrx Inc Polymeric derivatives for selective protein modification
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
GB0324368D0 (en) 2003-10-17 2003-11-19 Univ Cambridge Tech Polypeptides including modified constant regions
WO2005063815A2 (en) 2003-11-12 2005-07-14 Biogen Idec Ma Inc. Fcϝ receptor-binding polypeptide variants and methods related thereto
WO2005056759A2 (en) * 2003-12-04 2005-06-23 Xencor, Inc. Methods of generating variant proteins with increased host string content and compositions thereof
BRPI0506771A (en) * 2004-01-12 2007-05-22 Applied Molecular Evolution antibody and pharmaceutical composition
US8778880B2 (en) 2004-02-02 2014-07-15 Ambrx, Inc. Human growth hormone modified at position 35
AU2005227326B2 (en) 2004-03-24 2009-12-03 Xencor, Inc. Immunoglobulin variants outside the Fc region
WO2005123780A2 (en) 2004-04-09 2005-12-29 Protein Design Labs, Inc. Alteration of fcrn binding affinities or serum half-lives of antibodies by mutagenesis
RU2006140377A (en) * 2004-04-16 2008-05-27 Дженентек, Инк. (Us) METHOD FOR STRENGTHENING B-CELL DESTRUCTION
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
WO2006012500A2 (en) 2004-07-23 2006-02-02 Genentech, Inc. Crystallization of antibodies or fragments thereof
EP2213683B1 (en) 2004-08-04 2013-06-05 Mentrik Biotech, LLC Variant Fc regions
AU2005285347A1 (en) * 2004-08-19 2006-03-23 Genentech, Inc. Polypeptide variants with altered effector function
US7632497B2 (en) * 2004-11-10 2009-12-15 Macrogenics, Inc. Engineering Fc Antibody regions to confer effector function
US8802820B2 (en) 2004-11-12 2014-08-12 Xencor, Inc. Fc variants with altered binding to FcRn
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
US8546543B2 (en) * 2004-11-12 2013-10-01 Xencor, Inc. Fc variants that extend antibody half-life
EP1858925A2 (en) * 2005-01-12 2007-11-28 Xencor, Inc. Antibodies and fc fusion proteins with altered immunogenicity
ES2523666T3 (en) * 2005-05-31 2014-11-28 Board Of Regents, The University Of Texas System IgG1 antibodies with the mutated Fc part for increased binding to the FcRn receptor and uses thereof
WO2007008943A2 (en) * 2005-07-08 2007-01-18 Xencor, Inc. Optimized anti-ep-cam antibodies
DK1919503T3 (en) * 2005-08-10 2014-12-15 Macrogenics Inc Identification and preparation of antibodies with variant fc regions and methods of use thereof
CA2624189A1 (en) 2005-10-03 2007-04-12 Xencor, Inc. Fc variants with optimized fc receptor binding properties
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
AU2007212147A1 (en) * 2006-02-03 2007-08-16 Medimmune, Llc Protein formulations
US20100166741A1 (en) 2006-07-13 2010-07-01 Genentech , Inc. Altered br-3 binding polypeptides
US20080112961A1 (en) * 2006-10-09 2008-05-15 Macrogenics, Inc. Identification and Engineering of Antibodies with Variant Fc Regions and Methods of Using Same
EP2064335A4 (en) * 2006-10-16 2011-03-30 Medimmune Llc Molecules with reduced half-lives, compositions and uses thereof
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
TW200833711A (en) * 2006-12-22 2008-08-16 Genentech Inc Antibodies to insulin-like growth factor receptor
EP4098661A1 (en) * 2007-12-26 2022-12-07 Xencor, Inc. Fc variants with altered binding to fcrn
EP3029066B1 (en) * 2010-07-29 2019-02-20 Xencor, Inc. Antibodies with modified isoelectric points
EP3119890A4 (en) 2014-03-19 2017-12-27 Duke University Swarm immunization with envelopes from ch505
US11392902B2 (en) 2017-06-06 2022-07-19 United Parcel Service Of America, Inc. Systems, methods, apparatuses and computer program products for providing notification of items for pickup and delivery

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050037000A1 (en) * 2003-01-09 2005-02-17 Macrogenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
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
WO2006053301A2 (en) * 2004-11-12 2006-05-18 Xencor, Inc. Fc variants with altered binding to fcrn

Non-Patent Citations (7)

* Cited by examiner, † Cited by third party
Title
BERNSTEIN K E ET AL: "Nucleotide sequence of a rabbit IgG heavy chain from the recombinant F-I haplotype" IMMUNOGENETICS, SPRINGER VERLAG, BERLIN, DE, vol. 18, no. 4, 1 January 1983 (1983-01-01), pages 387-397, XP009116384 ISSN: 0093-7711 *
CLARKSON C A ET AL: "SEQUENCE OF OVINE IG GAMMA-2 CONSTANT REGION HEAVY CHAIN CDNA AND MOLECULAR MODELLING OF RUMINANT IGG ISOTYPES" MOLECULAR IMMUNOLOGY, PERGAMON, GB, vol. 30, no. 13, 1 January 1993 (1993-01-01), pages 1195-1204, XP009028022 ISSN: 0161-5890 *
DALL'ACQUA W F ET AL: "Increasing the affinity of a human IgG1 for the neonatal Fc receptor: biological consequences" JOURNAL OF IMMUNOLOGY, AMERICAN ASSOCIATION OF IMMUNOLOGISTS, US, vol. 169, no. 9, 1 November 2002 (2002-11-01), pages 5171-5180, XP002384463 ISSN: 0022-1767 *
LANCE W L ET AL: "Crystal structure at 2.8 ANG of an FcRn/heterodimeric Fc complex: Mechanism of pH-dependent binding" MOLECULAR CELL, CELL PRESS, CAMBRIDGE, MA, US, vol. 7, no. 4, 1 April 2001 (2001-04-01), pages 867-877, XP002305812 ISSN: 1097-2765 *
OBER RAIMUND J ET AL: "Differences in promiscuity for antibody-FcRn interactions across species: Implications for therapeutic antibodies" INTERNATIONAL IMMUNOLOGY, vol. 13, no. 12, December 2001 (2001-12), pages 1551-1559, XP002526759 ISSN: 0953-8178 *
ROOPENIAN DERRY C ET AL: "FcRn: the neonatal Fc receptor comes of age." NATURE REVIEWS. IMMUNOLOGY SEP 2007, vol. 7, no. 9, September 2007 (2007-09), pages 715-725, XP002510057 ISSN: 1474-1741 *
See also references of EP2212354A2 *

Cited By (158)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8674083B2 (en) 1999-01-15 2014-03-18 Genentech, Inc. Polypeptide variants with altered effector function
EP2845865A1 (en) * 2004-11-12 2015-03-11 Xencor Inc. Fc variants with altered binding to FcRn
US11046784B2 (en) 2006-03-31 2021-06-29 Chugai Seiyaku Kabushiki Kaisha Methods for controlling blood pharmacokinetics of antibodies
US11248053B2 (en) 2007-09-26 2022-02-15 Chugai Seiyaku Kabushiki Kaisha Method of modifying isoelectric point of antibody via amino acid substitution in CDR
US9828429B2 (en) 2007-09-26 2017-11-28 Chugai Seiyaku Kabushiki Kaisha Method of modifying isoelectric point of antibody via amino acid substitution in CDR
US11332533B2 (en) 2007-09-26 2022-05-17 Chugai Seiyaku Kabushiki Kaisha Modified antibody constant region
EP2937361A2 (en) 2007-10-31 2015-10-28 Xencor Inc. Fc variants with altered binding to fcRn
EP3138853A1 (en) 2007-10-31 2017-03-08 Xencor, Inc. Fc variants with altered binding to fcrn
EP2444423A1 (en) 2007-10-31 2012-04-25 Xencor Inc. Fc variants with altered binding to FcRn
EP4269443A3 (en) * 2007-12-26 2023-12-27 Xencor, Inc. Fc variants with altered binding to fcrn
EP4098661A1 (en) * 2007-12-26 2022-12-07 Xencor, Inc. Fc variants with altered binding to fcrn
EP3575317A1 (en) * 2007-12-26 2019-12-04 Xencor, Inc. Fc variants with altered binding to fcrn
US10472623B2 (en) 2008-04-11 2019-11-12 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding two or more antigen molecules repeatedly
US9868948B2 (en) 2008-04-11 2018-01-16 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding to two or more antigen molecules repeatedly
US11371039B2 (en) 2008-04-11 2022-06-28 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding to two or more antigen molecules repeatedly
US9890377B2 (en) 2008-04-11 2018-02-13 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding to two or more antigen molecules repeatedly
US11359194B2 (en) 2008-04-11 2022-06-14 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding two or more antigen molecules repeatedly
WO2010045193A1 (en) * 2008-10-14 2010-04-22 Genentech, Inc. Immunoglobulin variants and uses thereof
AU2013203391B2 (en) * 2008-10-14 2016-03-03 Genentech, Inc. Immunoglobulin variants and uses thereof
JP2012505833A (en) * 2008-10-14 2012-03-08 ジェネンテック, インコーポレイテッド Immunoglobulin variants and uses thereof
JP2016026144A (en) * 2008-10-14 2016-02-12 ジェネンテック, インコーポレイテッド Immunoglobulin variants and uses thereof
AU2009303526B2 (en) * 2008-10-14 2015-01-15 Genentech, Inc. Immunoglobulin variants and uses thereof
EP3524620A1 (en) * 2008-10-14 2019-08-14 Genentech, Inc. Immunoglobulin variants and uses thereof
WO2010070346A2 (en) 2008-12-18 2010-06-24 Medimmune Limited BINDING MEMBERS FOR INTERLEUKIN-4 RECEPTOR ALPHA (IL-4Ra) - 836
US9879249B2 (en) 2009-02-17 2018-01-30 Redwood Bioscience, Inc. Aldehyde-tagged protein-based drug carriers and methods of use
JP2019178134A (en) * 2010-03-30 2019-10-17 中外製薬株式会社 ANTIBODIES WITH MODIFIED AFFINITY TO FcRn THAT PROMOTE ANTIGEN CLEARANCE
JP7186813B2 (en) 2010-03-30 2022-12-09 中外製薬株式会社 Antibodies with altered affinity to FcRn that promote antigen clearance
JP2021091725A (en) * 2010-03-30 2021-06-17 中外製薬株式会社 ANTIBODIES WITH MODIFIED AFFINITY TO FcRn THAT PROMOTE ANTIGEN CLEARANCE
WO2012020096A1 (en) 2010-08-13 2012-02-16 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
US11891434B2 (en) 2010-11-30 2024-02-06 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding to plurality of antigen molecules repeatedly
US10183998B2 (en) 2011-01-14 2019-01-22 Redwood Bioscience, Inc. Aldehyde-tagged immunoglobulin polypeptides and methods of use thereof
US11912773B2 (en) 2011-02-04 2024-02-27 Genentech, Inc. Fc variants and methods for their production
US10689447B2 (en) 2011-02-04 2020-06-23 Genentech, Inc. Fc variants and methods for their production
US10618965B2 (en) 2011-02-25 2020-04-14 Chugai Seiyaku Kabushiki Kaisha Method for altering plasma retention and immunogenicity of antigen-binding molecule
US11718678B2 (en) 2011-02-25 2023-08-08 Chugai Seiyaku Kabushiki Kaisha Method for altering plasma retention and immunogenicity of antigen-binding molecule
US8969526B2 (en) 2011-03-29 2015-03-03 Roche Glycart Ag Antibody Fc variants
EP3680251A1 (en) 2011-09-30 2020-07-15 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecules for promoting elimination of antigens
US11827699B2 (en) 2011-09-30 2023-11-28 Chugai Seiyaku Kabushiki Kaisha Methods for producing antibodies promoting disappearance of antigens having plurality of biological activities
WO2013047752A1 (en) 2011-09-30 2013-04-04 中外製薬株式会社 Antigen-binding molecule for promoting loss of antigens
US10253100B2 (en) 2011-09-30 2019-04-09 Chugai Seiyaku Kabushiki Kaisha Therapeutic antigen-binding molecule with a FcRn-binding domain that promotes antigen clearance
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
EP3939996A1 (en) 2011-09-30 2022-01-19 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule promoting disappearance of antigens having plurality of biological activities
US9580509B2 (en) 2011-11-07 2017-02-28 Medimmune, Llc Multispecific and multivalent binding proteins and uses thereof
WO2013070565A1 (en) 2011-11-07 2013-05-16 Medimmune, Llc Multispecific and multivalent binding proteins and uses thereof
EP3608340A1 (en) 2011-11-23 2020-02-12 Medlmmune, LLC Binding molecules specific for her3 and uses thereof
WO2013078191A1 (en) 2011-11-23 2013-05-30 Medimmune, Llc Binding molecules specific for her3 and uses thereof
US11820793B2 (en) 2011-11-30 2023-11-21 Chugai Seiyaku Kabushiki Kaisha Drug containing carrier into cell for forming immune complex
US9688756B2 (en) 2011-12-21 2017-06-27 Amgen Inc. Variant Fc-polypeptides with enhanced binding to the neonatal Fc receptor
CN104302665A (en) * 2011-12-21 2015-01-21 安姆根有限公司 Variant Fc-polypeptides with enhanced binding to the neonatal Fc receptor
EA032985B1 (en) * 2011-12-21 2019-08-30 Амген Инк. VARIANT Fc-POLYPEPTIDES WITH ENHANCED BINDING TO THE NEONATAL Fc RECEPTOR
KR20140114833A (en) * 2011-12-21 2014-09-29 암젠 인크 Variant fc-polypeptides with enhanced binding to the neonatal fc receptor
KR102208698B1 (en) * 2011-12-21 2021-01-29 암젠 인크 Variant fc-polypeptides with enhanced binding to the neonatal fc receptor
WO2013096221A1 (en) * 2011-12-21 2013-06-27 Amgen Inc. Variant fc-polypeptides with enhanced binding to the neonatal fc receptor
WO2013125667A1 (en) 2012-02-24 2013-08-29 中外製薬株式会社 ANTIGEN-BINDING MOLECULE FOR PROMOTING DISAPPEARANCE OF ANTIGEN VIA FcγRIIB
EP3738980A1 (en) 2012-02-24 2020-11-18 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule for promoting disappearance of antigen via fc gamma riib
EP2861068A2 (en) 2012-06-01 2015-04-22 Momenta Pharmaceuticals, Inc. Methods related to bevacizumab
US9695244B2 (en) 2012-06-01 2017-07-04 Momenta Pharmaceuticals, Inc. Methods related to denosumab
EP2856158A2 (en) 2012-06-01 2015-04-08 Momenta Pharmaceuticals, Inc. Methods related to rituximab
EP2856159A2 (en) 2012-06-01 2015-04-08 Momenta Pharmaceuticals, Inc. Methods related to denosumab
EP2855745A2 (en) 2012-06-01 2015-04-08 Momenta Pharmaceuticals, Inc. Methods related to adalimumab
EP2854851A2 (en) 2012-06-01 2015-04-08 Momenta Pharmaceuticals, Inc. Methods related to trastuzumab
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
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
US11236168B2 (en) 2012-08-24 2022-02-01 Chugai Seiyaku Kabushiki Kaisha Mouse FcγammaRII-specific Fc antibody
US10919953B2 (en) 2012-08-24 2021-02-16 Chugai Seiyaku Kabushiki Kaisha FcgammaRIIB-specific Fc region variant
WO2014030728A1 (en) 2012-08-24 2014-02-27 中外製薬株式会社 Fcγriib-specific fc region variant
EP3721900A1 (en) 2012-08-24 2020-10-14 Chugai Seiyaku Kabushiki Kaisha Fcgammariib-specific fc region variant
EP3783017A1 (en) 2013-04-02 2021-02-24 Chugai Seiyaku Kabushiki Kaisha Fc region variant
US11267868B2 (en) 2013-04-02 2022-03-08 Chugai Seiyaku Kabushiki Kaisha Fc region variant
WO2014163101A1 (en) 2013-04-02 2014-10-09 中外製薬株式会社 Fc region variant
US11091541B2 (en) 2013-04-29 2021-08-17 Hoffmann-La Roche Inc. Human FcRn-binding modified antibodies and methods of use
WO2015050959A1 (en) 2013-10-01 2015-04-09 Yale University Anti-kit antibodies and methods of use thereof
EP3733244A1 (en) 2013-10-02 2020-11-04 Medlmmune, LLC Neutralizing anti-influenza a antibodies and uses thereof
WO2015051010A1 (en) 2013-10-02 2015-04-09 Medimmune, Llc Neutralizing anti-influenza a antibodies and uses thereof
US11555067B2 (en) 2014-01-15 2023-01-17 Hoffmann-La Roche Inc. Fc-region variants with improved protein A-binding
US9663574B2 (en) 2014-03-07 2017-05-30 Alexion Pharmaceuticals, Inc. Anti-C5 antibodies having improved pharmacokinetics
US9206251B2 (en) 2014-03-07 2015-12-08 Alexion Pharmaceuticals, Inc. Nucleic acids encoding anti-C5 antibodies having improved pharmacokinetics
US9371377B2 (en) 2014-03-07 2016-06-21 Alexion Pharmaceuticals, Inc. Anti-C5 antibodies having improved pharmacokinetics
US10584164B2 (en) 2014-03-07 2020-03-10 Alexion Pharmaceuticals, Inc. Methods of treating atypical hemolytic uremic syndrome and paroxysmal nocturnal hemoglobinuria with anti-C5 antibodies
US9803007B1 (en) 2014-03-07 2017-10-31 Alexion Pharmaceuticals, Inc. Anti-C5 antibodies having improved pharmacokinetics
US10227400B2 (en) 2014-03-07 2019-03-12 Alexion Pharmaceuticals, Inc. Methods of treating atypical hemolytic uremic syndrome with anti-C5 antibodies
US11434280B2 (en) 2014-03-07 2022-09-06 Alexion Pharmaceuticals, Inc. Anti-C5 antibodies having improved pharmacokinetics
US9107861B1 (en) 2014-03-07 2015-08-18 Alexion Pharmaceuticals, Inc. Methods of treating C5 mediated complement-associated conditions with anti-C5 antibodies having improved pharmacokinetics
US9079949B1 (en) 2014-03-07 2015-07-14 Alexion Pharmaceuticals, Inc. Anti-C5 antibodies having improved pharmacokinetics
WO2015157592A1 (en) 2014-04-11 2015-10-15 Medimmune, Llc Bispecific her2 antibodies
US11767369B2 (en) 2014-05-16 2023-09-26 Medimmune, Llc Molecules with altered neonate Fc receptor binding having enhanced therapeutic and diagnostic properties
EP3142700A4 (en) * 2014-05-16 2018-05-02 Medimmune, LLC Molecules with altered neonate fc receptor binding having enhanced therapeutic and diagnostic properties
WO2016050867A1 (en) 2014-10-01 2016-04-07 Medimmune Limited Antibodies to ticagrelor and methods of use
US10487154B2 (en) 2014-10-01 2019-11-26 Medimmune Limited Antibodies to ticagrelor and methods of use
US10954308B2 (en) 2014-10-01 2021-03-23 Medimmune Limited Antibodies to ticagrelor and methods of use
US9982061B2 (en) 2014-10-01 2018-05-29 Medimmune Limited Antibodies to ticagrelor and methods of use
US11773186B2 (en) 2014-10-01 2023-10-03 Medimmune Limited Antibodies to ticagrelor and methods of use
US10899846B2 (en) 2014-11-06 2021-01-26 Hoffmann-La Roche Inc. Fc-region variants with modified FcRn- and protein A-binding properties
US10738111B2 (en) 2014-12-19 2020-08-11 Chugai Seiyaku Kabushiki Kaisha Anti-myostatin antibodies, polypeptides containing variant Fc regions, and methods of use
US10000560B2 (en) 2014-12-19 2018-06-19 Chugai Seiyaku Kabushiki Kaisha Anti-myostatin antibodies, polypeptides containing variant Fc regions, and methods of use
US11454633B2 (en) 2014-12-19 2022-09-27 Chugai Seiyaku Kabushiki Kaisha Anti-myostatin antibodies, polypeptides containing variant Fc regions, and methods of use
US10519229B2 (en) 2015-02-05 2019-12-31 Chugai Seiyaku Kabushiki Kaisha Nucleic acids encoding IL-8 antibodies
US9969800B2 (en) 2015-02-05 2018-05-15 Chugai Seiyaku Kabushiki Kaisha IL-8 antibodies
US11180548B2 (en) 2015-02-05 2021-11-23 Chugai Seiyaku Kabushiki Kaisha Methods of neutralizing IL-8 biological activity
EP4303235A2 (en) 2015-06-08 2024-01-10 MacroGenics, Inc. Lag-3-binding moleculkes and methods of use thereof
US11072653B2 (en) 2015-06-08 2021-07-27 Macrogenics, Inc. LAG-3-binding molecules and methods of use thereof
US11858991B2 (en) 2015-06-08 2024-01-02 Macrogenics, Inc. LAG-3-binding molecules and methods of use thereof
US10577422B2 (en) 2015-07-30 2020-03-03 Macrogenics, Inc. PD-1-binding molecules and methods of use thereof
EP3456346A1 (en) 2015-07-30 2019-03-20 MacroGenics, Inc. Pd-1 and lag-3 binding molecules and methods of use thereof
EP3981792A1 (en) 2015-07-30 2022-04-13 MacroGenics, Inc. Pd-1-binding molecules and methods of use thereof
WO2017019846A1 (en) 2015-07-30 2017-02-02 Macrogenics, Inc. Pd-1-binding molecules and methods use thereof
US11623959B2 (en) 2015-07-30 2023-04-11 Macrogenics, Inc. PD-1-binding molecules and methods of use thereof
US11174315B2 (en) 2015-10-08 2021-11-16 Macrogenics, Inc. Combination therapy for the treatment of cancer
US10954301B2 (en) 2015-12-14 2021-03-23 Macrogenics, Inc. Bispecific molecules having immunoreactivity with PD-1 and CTLA-4, and methods of use thereof
WO2017106061A1 (en) 2015-12-14 2017-06-22 Macrogenics, Inc. Bispecific molecules having immunoreactivity with pd-1 and ctla-4, and methods of use thereof
US11840571B2 (en) 2015-12-14 2023-12-12 Macrogenics, Inc. Methods of using bispecific molecules having immunoreactivity with PD-1 and CTLA-4
US11359009B2 (en) 2015-12-25 2022-06-14 Chugai Seiyaku Kabushiki Kaisha Anti-myostatin antibodies and methods of use
WO2017142928A1 (en) 2016-02-17 2017-08-24 Macrogenics, Inc. Ror1-binding molecules, and methods of use thereof
US10961311B2 (en) 2016-04-15 2021-03-30 Macrogenics, Inc. B7-H3 binding molecules, antibody drug conjugates thereof and methods of use thereof
WO2017180813A1 (en) 2016-04-15 2017-10-19 Macrogenics, Inc. Novel b7-h3 binding molecules, antibody drug conjugates thereof and methods of use thereof
US11591400B2 (en) 2016-04-15 2023-02-28 Macrogenics, Inc. B7-H3 directed antibody drug conjugates
US11788066B2 (en) 2016-04-26 2023-10-17 R.P. Scherer Technologies, Llc Antibody conjugates and methods of making and using the same
US11208632B2 (en) 2016-04-26 2021-12-28 R.P. Scherer Technologies, Llc Antibody conjugates and methods of making and using the same
US11091539B2 (en) 2016-07-08 2021-08-17 Staten Biotechnology B.V. Anti-ApoC3 antibodies and methods of use thereof
US11858980B2 (en) 2016-08-02 2024-01-02 Visterra, Inc. Engineered polypeptides and uses thereof
WO2018052556A1 (en) * 2016-08-02 2018-03-22 Visterra, Inc. Engineered polypeptides and uses thereof
US11053308B2 (en) 2016-08-05 2021-07-06 Chugai Seiyaku Kabushiki Kaisha Method for treating IL-8-related diseases
US11780912B2 (en) 2016-08-05 2023-10-10 Chugai Seiyaku Kabushiki Kaisha Composition for prophylaxis or treatment of IL-8 related diseases
US10669344B2 (en) 2016-08-12 2020-06-02 Janssen Biotech, Inc. Engineered antibodies and other Fc-domain containing molecules with enhanced agonism and effector functions
US10604561B2 (en) 2016-09-16 2020-03-31 Chugai Seiyaku Kabushiki Kaisha Anti-dengue virus antibodies, polypeptides containing variant Fc regions, and methods of use
US11780908B2 (en) 2016-09-16 2023-10-10 Chugai Seiyaku Kabushiki Kaisha Anti-dengue virus antibodies, polypeptides containing variant FC regions, and methods of use
US10844113B2 (en) 2016-09-16 2020-11-24 Chugai Seiyaku Kabushiki Kaisha Anti-dengue virus antibodies, polypeptides containing variant Fc regions, and methods of use
US11242402B2 (en) 2016-12-23 2022-02-08 Macrogenics, Inc. ADAM9-binding molecules, and methods of use thereof
WO2018119196A1 (en) 2016-12-23 2018-06-28 Immunogen, Inc. Immunoconjugates targeting adam9 and methods of use thereof
US11942149B2 (en) 2017-02-24 2024-03-26 Macrogenics, Inc. Bispecific binding molecules that are capable of binding CD137 and tumor antigens, and uses thereof
US11459394B2 (en) 2017-02-24 2022-10-04 Macrogenics, Inc. Bispecific binding molecules that are capable of binding CD137 and tumor antigens, and uses thereof
EP3608339A4 (en) * 2017-04-07 2020-12-23 Kookmin University Industry Academy Cooperation Foundation Antibody fc variants for improving blood half-life
US11242381B2 (en) 2017-04-21 2022-02-08 Staten Biotechnology B.V. Anti-ApoC3 antibodies and methods of use thereof
EP3634486A4 (en) * 2017-06-05 2021-03-31 Janssen Biotech, Inc. Engineered multispecific antibodies and other multimeric proteins with asymmetrical ch2-ch3 region mutations
US11149094B2 (en) 2017-06-05 2021-10-19 Janssen Biotech, Inc. Engineered multispecific antibodies and other multimeric proteins with asymmetrical CH2-CH3 region mutations
US11365241B2 (en) 2017-07-27 2022-06-21 Alexion Pharmaceuticals, Inc. High concentration anti-C5 antibody formulations
US10538583B2 (en) 2017-10-31 2020-01-21 Staten Biotechnology B.V. Anti-APOC3 antibodies and compositions thereof
US11248042B2 (en) 2017-10-31 2022-02-15 Staten Biotechnology B.V. Polynucleotides encoding anti-ApoC3 antibodies
US11248041B2 (en) 2017-10-31 2022-02-15 Staten Biotechnology B.V. Anti-ApoC3 antibodies
US11795226B2 (en) 2017-12-12 2023-10-24 Macrogenics, Inc. Bispecific CD16-binding molecules and their use in the treatment of disease
US11685781B2 (en) 2018-02-15 2023-06-27 Macrogenics, Inc. Variant CD3-binding domains and their use in combination therapies for the treatment of disease
US11891432B2 (en) 2018-03-15 2024-02-06 Chugai Seiyaku Kabushiki Kaisha Anti-dengue virus antibodies having cross-reactivity to Zika virus and methods of use
WO2020005945A1 (en) 2018-06-26 2020-01-02 Immunogen, Inc. Immunoconjugates targeting adam9 and methods of use thereof
WO2020086665A1 (en) 2018-10-26 2020-04-30 Immunogen, Inc. Epcam antibodies, activatable antibodies, and immunoconjugates, and uses thereof
EP3994176A4 (en) * 2019-07-02 2023-09-20 Telix International Pty Ltd Antibodies against caix with reduced affinity for the neonatal fc receptor
US11739142B2 (en) 2019-12-18 2023-08-29 Hoffmann-La Roche Inc. Bispecific anti-CCL2 antibodies
WO2021122733A1 (en) 2019-12-18 2021-06-24 F. Hoffmann-La Roche Ag Bispecific anti-ccl2 antibodies
WO2021234160A2 (en) 2020-05-22 2021-11-25 Formycon Ag Ace2 fusion proteins and uses thereof
WO2022010798A1 (en) 2020-07-06 2022-01-13 Kiromic BioPharma, Inc. Mesothelin isoform binding molecules and chimeric pd1 receptor molecules, cells containing the same and uses thereof
WO2022090469A2 (en) 2020-10-29 2022-05-05 Formycon Ag Ace2 fusion proteins and uses thereof
WO2022090469A3 (en) * 2020-10-29 2022-07-21 Formycon Ag Ace2 fusion proteins and uses thereof
WO2022108627A1 (en) 2020-11-18 2022-05-27 Kiromic Biopharma, Inc.Kiromic Biopharma, Inc. Gamma-delta t cell manufacturing processes and chimeric pd1 receptor molecules
WO2022184854A2 (en) 2021-03-03 2022-09-09 Formycon Ag Formulations of ace2 fc fusion proteins
WO2022263501A1 (en) 2021-06-18 2022-12-22 F. Hoffmann-La Roche Ag Bispecific anti-ccl2 antibodies
WO2023094507A1 (en) 2021-11-24 2023-06-01 Formycon Ag Improved ace2 fusion proteins
WO2023094571A1 (en) 2021-11-25 2023-06-01 Formycon Ag Stabilization of ace2 fusion proteins
WO2024015830A1 (en) 2022-07-12 2024-01-18 Cytomx Therapeutics, Inc. Epcam immunoconjugates and uses thereof

Also Published As

Publication number Publication date
CN114891093A (en) 2022-08-12
ES2537282T3 (en) 2015-06-05
RU2517621C2 (en) 2014-05-27
CA2703385C (en) 2016-06-28
CN101970492A (en) 2011-02-09
EP2937361A3 (en) 2016-02-24
AU2008319144A1 (en) 2009-05-07
JP5542677B2 (en) 2014-07-09
ES2537202T3 (en) 2015-06-03
US8802820B2 (en) 2014-08-12
JP2014193168A (en) 2014-10-09
EP2444423A1 (en) 2012-04-25
WO2009058492A3 (en) 2009-07-23
IL231423A (en) 2017-06-29
IL205050A0 (en) 2010-11-30
DK2212354T3 (en) 2015-05-26
IL231423A0 (en) 2014-04-30
JP2011502126A (en) 2011-01-20
EP3138853B1 (en) 2021-11-24
EP2212354B1 (en) 2015-04-01
EP2212354A2 (en) 2010-08-04
JP5927231B2 (en) 2016-06-01
US20090041770A1 (en) 2009-02-12
US20180162946A1 (en) 2018-06-14
DK2444423T3 (en) 2015-05-26
US20150152183A1 (en) 2015-06-04
US20210115147A1 (en) 2021-04-22
CN107857814A (en) 2018-03-30
EP2444423B1 (en) 2015-03-04
BRPI0818746A2 (en) 2015-05-05
AU2008319144B2 (en) 2014-01-16
RU2010121898A (en) 2011-12-10
CN101970492B (en) 2018-01-05
CA2703385A1 (en) 2009-05-07
EP3138853A1 (en) 2017-03-08
RU2013158731A (en) 2015-07-10
KR20100090266A (en) 2010-08-13
EP2937361A2 (en) 2015-10-28
KR101573883B1 (en) 2015-12-02

Similar Documents

Publication Publication Date Title
US11932685B2 (en) Fc variants with altered binding to FcRn
US20210115147A1 (en) Fc VARIANTS WITH ALTERED BINDING TO FcRn
US11198739B2 (en) Fc variants with altered binding to FcRn
US10336818B2 (en) Fc variants with altered binding to FcRn
US20070135620A1 (en) Fc variants with altered binding to FcRn
US20220281964A1 (en) Fc VARIANTS WITH ALTERED BINDING TO FcRn
AU2014201371A1 (en) Fc variants with altered binding to FcRn
AU2016203334A1 (en) Fc variants with altered binding to FcRn

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 200880113962.0

Country of ref document: CN

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

Ref document number: 08846103

Country of ref document: EP

Kind code of ref document: A2

WWE Wipo information: entry into national phase

Ref document number: 2010531108

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 205050

Country of ref document: IL

Ref document number: 2008319144

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2703385

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2906/DELNP/2010

Country of ref document: IN

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2008319144

Country of ref document: AU

Date of ref document: 20080922

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 20107010782

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2008846103

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2010121898

Country of ref document: RU

WWE Wipo information: entry into national phase

Ref document number: 231423

Country of ref document: IL

ENP Entry into the national phase

Ref document number: PI0818746

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20100430