WO2006105338A2 - Fc VARIANTS WITH OPTIMIZED PROPERTIES - Google Patents

Fc VARIANTS WITH OPTIMIZED PROPERTIES Download PDF

Info

Publication number
WO2006105338A2
WO2006105338A2 PCT/US2006/011752 US2006011752W WO2006105338A2 WO 2006105338 A2 WO2006105338 A2 WO 2006105338A2 US 2006011752 W US2006011752 W US 2006011752W WO 2006105338 A2 WO2006105338 A2 WO 2006105338A2
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
variant
polypeptide
positions
variants
Prior art date
Application number
PCT/US2006/011752
Other languages
French (fr)
Other versions
WO2006105338A3 (en
Inventor
Gregory Alan Lazar
Wei Dang
John R. 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
Priority claimed from US11/124,620 external-priority patent/US8188231B2/en
Application filed by Xencor, Inc. filed Critical Xencor, Inc.
Priority to AU2006230413A priority Critical patent/AU2006230413B8/en
Priority to JP2008504389A priority patent/JP2008537941A/en
Priority to CA002602663A priority patent/CA2602663A1/en
Priority to EP06748967A priority patent/EP1871808A2/en
Publication of WO2006105338A2 publication Critical patent/WO2006105338A2/en
Publication of WO2006105338A3 publication Critical patent/WO2006105338A3/en
Priority to IL186145A priority patent/IL186145A0/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • 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
    • C07K16/2896Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against molecules with a "CD"-designation, not provided for elsewhere
    • 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
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • C07K2317/732Antibody-dependent cellular cytotoxicity [ADCC]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • C07K2317/734Complement-dependent cytotoxicity [CDC]

Definitions

  • the present invention relates to Fc variant polypeptides with optimized properties, 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 V H -CH1-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 V H -CD1-CD2-CD3, 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 V L -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 comprising the variable region can exist in the absence of other regions of the antibody, including for example the antigen binding fragment (Fab) comprising V ⁇ -C ⁇ 1 and VH-C L , the variable fragment (Fv) comprising V H and VL, the single chain variable fragment (scFv) comprising V H 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, hereby 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 Figure 1, 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 are 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 hereby 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, hereby entirely incorporated by reference).
  • These receptors typically have an extracellular domain that mediates binding to Fc 1 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
  • Fc ⁇ Rs bind the same region on Fc, at the N-terminal end of the C ⁇ 2 domain and the preceding hinge, shown in Figure 2. This interaction is well characterized structurally (Sondermann et al., 2001 , J MoI
  • 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, hereby 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 ⁇ RII and Fc ⁇ RIII generally bind at 10 '8 and 10 '5 respectively.
  • Fc ⁇ Rllla and Fc ⁇ Rlllb are 96% identical, however Fc ⁇ Rlllb does not have a 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.
  • ITIM immunoreceptor tyrosine-based inhibition motif
  • 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, hereby 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, Stood 99:754-758, both hereby entirely incorporated by reference).
  • 80-90% of humans are poor responders, e.g., 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).
  • C1q forms a complex with the serine proteases C1r and C1s to form the C1 complex.
  • C1q 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 C1q, 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, hereby entirely incorporated by reference).
  • the binding site for FcRn on Fc 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.
  • a key 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. While not wanting to be limited to one theory, it is believed that the structural purpose of this carbohydrate may be to stabilize or solubilize Fc, determine a specific angle or level of flexibility between the C ⁇ 3 and C ⁇ 2 domains, keep the two C ⁇ 2 domains from aggregating with one another across the central axis, or a combination of these.
  • An Fc fusion is a protein wherein one or more polypeptides is operably linked to Fc.
  • An Fc fusion combines the Fc region of an antibody, and thus its favorable effector functions and pharmacokinetics, with the target-binding region of a receptor, ligand, or some other protein or protein domain. The role of the latter is to mediate target recognition, and thus it is functionally analogous to the antibody variable region. Because of the structural and functional overlap of Fc fusions with antibodies, the discussion on antibodies in the present invention extends also to Fc fusions.
  • Antibodies have found widespread application in oncology, particularly for targeting cellular antigens selectively expressed on tumor cells with the goal of cell destruction.
  • There are a number of possible mechanisms by which antibodies destroy tumor cells including anti-proliferation via blockage of needed growth pathways, intracellular signaling leading to apoptosis, enhanced down regulation and/or turnover of receptors, CDC, ADCC, ADCP, and promotion of an adaptive immune response (Cragg et al., 1999, Curr Opin Immunol 11 :541 -547; Glennie et al., 2000, Immunol Today 21 :403-410, both hereby entirely incorporated by reference).
  • Anti-tumor efficacy may be due to a combination of these mechanisms, and their relative importance in clinical therapy appears to be cancer dependent. Despite this arsenal of anti-tumor weapons, the potency of antibodies as anti-cancer agents is unsatisfactory, particularly given their high cost.
  • Patient tumor response data show that monoclonal antibodies provide only a small improvement in therapeutic success over normal single-agent cytotoxic chemotherapeutics. For example, just half of all relapsed low-grade non-Hodgkin's lymphoma patients respond to the anti-CD20 antibody rituximab (McLaughlin et al., 1998, J CHn Oncol 16:2825-2833, hereby entirely incorporated by reference).
  • trastuzumab Herceptin®, Genentech
  • trastuzumab an anti-HER2/neu antibody for treatment of metastatic breast cancer
  • the overall response rate using trastuzumab for the 222 patients tested was only 15%, with 8 complete and 26 partial responses and a median response duration and survival of 9 to 13 months (Cobleigh et al., 1999, J Clin Oncol 17:2639-2648, hereby entirely incorporated by reference).
  • any small improvement in mortality rate defines success.
  • a promising means for enhancing the anti-tumor potency of antibodies is via enhancement of their ability to mediate cytotoxic effector functions such as ADCC, ADCP, and CDC.
  • cytotoxic effector functions such as ADCC, ADCP, and CDC.
  • the importance of Fc ⁇ R-mediated effector functions for the anti-cancer activity of antibodies has been demonstrated in mice (Clynes et al., 1998, Proc Natl Acad Sci U S A 95:652-656; Clynes et al., 2000, Nat Med 6:443-446, both hereby entirely incorporated by reference), and the affinity of interaction between Fc and certain Fc ⁇ Rs correlates with targeted cytotoxicity in cell-based assays (Shields et al., 2001 , J Biol Chem 276:6591-6604; Presta et al., 2002, Biochem Soc Trans 30:487-490; Shields et al., 2002, J Biol Chem 277:26733-26740
  • Fc ⁇ Rs can mediate antigen uptake and processing by antigen presenting cells
  • optimized Fc ⁇ R affinity may also improve the capacity of antibody therapeutics to elicit an adaptive immune response (Dhodapkar & Dhodapkar, 2005, Proc Natl Acad Sci USA, 102, 6243-6244, hereby entirely incorporated by reference).
  • the importance of complement-mediated effector function for anti-cancer therapy of antibodies is not as well characterized.
  • Antibodies optimized for CDC would provide a way to investigate the role of complement in antibody clinical applications, and provide a potential mechanism for improving the tumor killing capacity of antibodies.
  • anti-CD4 antibodies For example, the ability of anti-CD4 antibodies to block CD4 receptors on T cells makes them effective anti-inflammatories, yet their ability to recruit Fc ⁇ R receptors also directs immune attack against the target cells, resulting in T cell depletion (Reddy et al., 2000, J Immunol 164:1925-1933, hereby entirely incorporated by reference). Effector function may also be a problem for radiolabeled antibodies, referred to as radioconjugates, and antibodies conjugated to toxins, referred to as immunotoxins.
  • Fc variants that reduce or ablate Fc ligand binding are also known in the art (Alegre et al., 1994, Transplantation 57:1537-1543; Hutchins et al., 1995, Proc Natl Acad Sci USA 92:11980-11984; Armour et al., 1999, Eur J Immunol 29:2613-2624; Reddy et al., 2000, J Immunol 164:1925-1933; Xu et al., 2000, Cell Immunol 200:16-26; Shields et al., 2001 , J Biol Chem 276:6591- 6604; Armour et al., 1999, Eur J Immunol 29:2613-2624; US 6,194,551 ; US 5,885,573; PCT WO 99/58572; USSN 10/267,286, all hereby entirely incorporated by reference).
  • Fc variants should be engineered that not only ablate binding to Fc ⁇ Rs and/or C1q, but also maintain antibody stability, solubility, and structural integrity, as well as ability to interact with other important Fc ligands such as FcRn and proteins A and G.
  • Fc variants obtained in these studies provide a variety of optimal enhancements in Fc ligand and effector function properties, including but not limited to selectively improved binding to Fc ⁇ Rs, enhanced ADCC, improved binding to complement protein C1q, reduced binding to Fc ⁇ Rs, reduced binding to complement protein C1q, and other optimized properties.
  • the present invention aims to further characterize the properties of select Fc variants from these studies, and to utilize the data to generate novel variants with optimized properties.
  • the present invention provides Fc variants with optimized properties.
  • Said optimized properties include altered binding to Fc ⁇ R's, altered antibody dependent cell-mediated cytotoxicity, and altered complement dependent cytotoxicity relative to a parent Fc polypeptide.
  • the Fc variants of the present invention improve binding to one or more Fc ⁇ R's relative to a parent Fc polypeptide.
  • the Fc variants of the invention improve antibody dependent cell-mediated cytotoxicity relative to a parent Fc polypeptide.
  • said Fc variants comprise an amino acid modification at one or more positions selected from the group consisting of: 227, 234, 235, 236, 239, 246, 255, 258, 260, 264, 267, 268, 272, 281 , 282, 283, 284, 293, 295, 304, 324, 327, 328, 330, 332, 335, wherein numbering is according to the EU index.
  • the Fc variants of the present invention improve complement dependent cytotoxicity relative to a parent Fc polypeptide.
  • said Fc variants comprise an amino acid modification at one or more positions selected from the group consisting of: 233, 234, 235, 239, 267, 268, 271 , 272, 274, 276, 278, 281 , 282, 284, 285, 293, 300, 320, 322, 324,
  • the Fc variants of the present invention reduce binding to one or more Fc ⁇ Rs relative to a parent Fc polypeptide. In an alternate embodiment, the Fc variants of the invention reduce antibody dependent cell-mediated cytotoxicity relative to a parent Fc polypeptide. In an alternate embodiment, the Fc variants of the invention reduce complement dependent cytotoxicity relative to a parent Fc polypeptide. In a preferred embodiment, the Fc variants of the invention reduce binding to one or more Fc ⁇ Rs, reduce antibody dependent cell-mediated cytotoxicity, and reduce complement dependent cytotoxicity relative to a parent Fc polypeptide.
  • said Fc variants comprise one or more amino acid modifications at a position selected from the group consisting of: 232, 234, 235, 236, 237, 238, 239, 265, 267, 269, 270, 297, 299, 325,
  • the present invention provides methods for engineering Fc variants with optimized properties. It is a further object of the present invention to provide experimental production and screening methods for obtaining optimized Fc variants.
  • the present invention provides isolated nucleic acids encoding the Fc variants described herein.
  • the present invention provides vectors comprising said nucleic acids, optionally, operably linked to control sequences.
  • the present invention provides host cells containing the vectors, and methods for producing and optionally recovering the Fc variants.
  • the present invention provides novel Fc polypeptides, including antibodies, Fc fusions, isolated Fc, and Fc fragments, that comprise the Fc variants disclosed herein. Said novel Fc polypeptides may find use in a therapeutic product. In a most preferred embodiment, the Fc polypeptides of the invention are antibodies.
  • compositions comprising Fc polypeptides that comprise the Fc variants described herein, and a physiologically or pharmaceutically acceptable carrier or diluent.
  • the present invention contemplates therapeutic and diagnostic uses for Fc polypeptides that comprise the Fc variants disclosed 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 1CE1 (James et al., 1999, J MoI Biol 289:293-301, hereby entirely incorporated by reference) and a human IgGI Fc structure from pdb accession code 1 DN2 (DeLano et al., 2000, Science 287:1279-1283, hereby 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 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, C1q, and proteins A and G in the Fc region.
  • FIG. 1 The Fc/Fc ⁇ Rlllb complex structure 11IS. Fc is shown as a gray ribbon diagram, and Fc ⁇ Rlllb is shown as a black ribbon. The N297 carbohydrate is shown as black sticks.
  • Figures 3a - 3b Alignment of the amino acid sequences of the human IgG immunoglobulins IgGI , lgG2, lgG3, and lgG4.
  • Figure 3a provides the sequences of the CH1 (C ⁇ 1) and hinge domains
  • Figure 3b provides the sequences of the CH2 (C ⁇ 2) and CH3 (C ⁇ 3) domains.
  • Positions are numbered according to the EU index of the IgGI sequence, and differences between IgGI and the other immunoglobulins lgG2, lgG3, and lgG4 are shown in grey.
  • Polymorphisms exist at a number of positions (Kim et al., 2001 , J. MoI. Evol. 54:1-9, hereby entirely incorporated by reference), and thus slight differences between the presented sequences and sequences in the prior art may exist.
  • the possible beginnings of the Fc region are labeled, defined herein as either EU position 226 or 230.
  • FIG. 6 Binding to human V158 Fc ⁇ Rllla ( Figure 6a) and F158 Fc ⁇ Rllla ( Figure 6b) by select PRO70769 Fc variants as determined by the competition AlphaScreen assay. In the presence of competitor antibody (Fc variant or WT) a characteristic inhibition curve is observed as a decrease in luminescence signal. The binding data were normalized to the maximum and minimum luminescence signal for each particular curve, provided by the baselines at low and high antibody concentrations respectively. The curves represent the fits of the data to a one site competition model using nonlinear regression.
  • Figure 7 Binding to human V158 Fc ⁇ Rllla and F158 Fc ⁇ Rllla by PRO70769 Fc variants as measured by competition AlphaScreen assay.
  • Figure 7a provides data for select variants
  • Figure 7b provides the IC50's and folds relative to WT PRO70769 IgGI .
  • Figure 8 Fc variants and Fc ⁇ R binding data. All Fc variants were constructed in the context of the variable region PRO70769 and either human IgGI or IgG(I /2) ELLGG.
  • Figure 8a provides the IC50's and fold IC50's relative to WT PRO70769 IgGI for binding to human activating receptors V158 and F158 Fc ⁇ Rllla, and the inhibitory receptor Fc ⁇ Rllb, as measured by competition AlphaScreen assay.
  • Figure 8b shows the AlphaScreen data for select variants.
  • Figure 9 Competition Surface Plasmon Resonance (SPR) experiment measuring binding affinities of I332E and S239D/I332E variants in the context of trastuzumab to human V158 Fc ⁇ Rllla.
  • Figure 9a provides the sensorgram raw data
  • Figure 9b provides a plot of the log of receptor concentration against the initial rate obtained at each concentration
  • Figure 9c provides the affinities obtained from the fits to these data as described in Example 1.
  • FIG. 10 Cell-based ADCC assays of select Fc variants in the context of the anti-CD20 antibody PRO70769. ADCC was measured by the release of lactose dehydrogenase using a LDH Cytotoxicity Detection Kit (Roche Diagnostic). CD20+ lymphoma WIL2-S cells were used as target cells and human PBMCs were used as effector cells. Shown is the dose-dependence of ADCC on antibody concentration for the indicated antibodies, normalized to the minimum and maximum fluorescence signal for each particular curve, provided by the baselines at low and high antibody concentrations respectively. The curves represent the fits of the data to a sigmoidal dose-response model using nonlinear regression.
  • FIG. 11 Cell-based ADCC assay of select Fc variants in the context of PRO70769 IgGI in the absence and presence of serum levels of human IgG. ADCC was measured by the release of lactose dehydrogenase using a LDH Cytotoxicity Detection Kit (Roche Diagnostic). CD20+ lymphoma WIL2-S cells were used as target cells and human PBM ⁇ s were used as effector cells.
  • FIG. 12 Residues mutated in Fc variants designed to enhance CDC.
  • the structure of the human IgGI Fc region is shown (pdb accession code 1 E4K, Sondermann et al., 2000, Nature 406:267-273, hereby entirely incorporated by reference).
  • Black ball and sticks indicate residues D270, K322, P329, and P331 , which have been shown to be important in mediating binding to complement protein C1q, and grey sticks indicate residues that were mutated in the present invention to explore variants that affect CDC.
  • FIG. 13 Fc variants screened for complement-mediated cytotoxicity (CDC) and CDC data.
  • the variable region is that of the anti-CD20 antibody PRO70769, and the heavy chain constant region is IgGI unless noted IgG(I /2) ELLGG.
  • Fold CDC provides the relative CDC activity compared to WT PRO70769 lgG1.
  • FIG. 14 CDC assays of Fc variant anti-CD20 antibodies. The dose-dependence on antibody concentration of complement-mediated lysis is shown for the indicated PRO70769 antibodies against CD20+ WIL2-S lymphoma target cells. Lysis was measured using release of Alamar Blue, and data were normalized to the minimum and maximum fluorescence signal for each particular curve, provided by the baselines at low and high antibody concentrations respectively. The curves represent the fits of the data to a sigmoidal dose-response model with variable slope using nonlinear regression.
  • Figure 15 Amino acid modifications that provide enhanced and reduced CDC, and positions that may be modified that may provide enhanced/modulated CDC. Positions are numbered according to the EU index.
  • FIG. 16 Fc variants screened for reduced Fc ⁇ R affinity, Fc ⁇ R-mediated effector function, and complement-mediated effector function.
  • the variable region is that of the anti-CD20 antibody PRO70769, and the heavy chain constant region is IgGI .
  • the figure provides the Fold IC50 for binding to human V158 Fc ⁇ Rllla and the Fold EC50 of CDC activity relative to WT PRO70769 IgGI .
  • FIG. Binding to human V158 Fc ⁇ Rllla by select PRO70769 Fc variants as determined by the competition AlphaScreen assay.
  • Figure 18 CDC assays of select Fc variant anti-CD20 antibodies against CD20+ W1L2-S lymphoma target cells. Lysis was measured by Alamar Blue release.
  • FIG. 19 Cell-based ADCC activity of select anti-CD20 Fc variants against CD20+ lymphoma WIL2-S cells. Human PBMCs were used as effector cells, and lysis was measured by LDH release.
  • FIG. 20 Fc variants screened for reduced Fc ⁇ R affinity, Fc ⁇ R-mediated effector function, and complement-mediated effector function.
  • the variable region is that of the anti-CD20 antibody PRO70769, and the heavy chain constant region is IgGL
  • the figure provides the Fold IC50 relative to WT for binding to human V158 Fc ⁇ Rllla by two separate experiments, the Fold IC50 relative to WT for binding to human Fc ⁇ RI, and the Fold EC50 relative to WT for CDC activity.
  • FIG. 21 Binding to the low affinity human activating receptor V158 Fc ⁇ Rllla and the high affinity human activating receptor Fc ⁇ RI by select PRO70769 Fc variants as determined by the competition AlphaScreen assay.
  • FIG. 23 Cell-based ADCC activity of anti-Her2 Fc variant and WT IgG antibodies against Her2/neu+ SkBr-3 breast carcinoma target cells. Human PBMCs were used as effector cells, and lysis was measured by LDH release.
  • FIG. 24 Amino acid sequences of variable light (VL) and heavy (VH) chains used in the present invention, including PRO70769 ( Figures 24a and 24b), trastuzumab ( Figures 24c and 24d), and ipilimumab ( Figures 24e and 24f).
  • Figure 25 Amino acid sequences of human constant light kappa (Figure 25a) and heavy ( Figures 25b - 25f) chains used in the present invention.
  • Figure 26 Sequences showing possible constant heavy chain sequences with reduced Fc ligand binding and effector function properties (Figure 26a), and sequences of improved anti-CTLA-4 antibodies ( Figures 26b - 26d).
  • Figure 26a shows potential Fc variant constant heavy chain sequences, with variable positions designated in bold as X1 , X2, X3, X4, X5, X6, X7, and X8.
  • the table below the sequence provides the WT amino acid and possible substitutions for these positions.
  • Improved antibody sequences may comprise one or more non-WT amino acid selected from this group of possible modifications.
  • Figure 26b provides the light chain sequence of an anti-CTLA-4 antibody
  • Figures 26c and 26d provide heavy chain sequences of anti-CTLA-4 antibodies with reduced Fc ligand binding and Fc-mediated effector function. These include an L235G/G236R IgGI heavy chain ( Figure 26c) and an lgG2 heavy chain ( Figure 26d). The positions are numbered according to the EU index as in Kabat, and thus do not correspond to the sequential order in the sequence.
  • ADCC antibody dependent cell-mediated cytotoxicity
  • ADCP antibody dependent cell-mediated phagocytosis as used herein is meant the cell-mediated reaction wherein nonspecific cytotoxic cells that express FcDRs recognize bound antibody on a target cell and subsequently cause phagocytosis of the target cell.
  • 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 L328R refers to a variant polypeptide, in this case an Fc variant, in which the leucine at position 328 is replaced with arginine.
  • amino acid insertion or “insertion” as used herein is meant the addition of an amino acid at a particular position in a parent polypeptide sequence.
  • insert G > 235-236 designates an insertion of glycine between positions 235 and 236.
  • amino acid deletion or “deletion” as used herein is meant the removal of an amino acid at a particular position in a parent polypeptide sequence.
  • G236- designates the deletion of glycine at position 236.
  • Amino acids of the invention may be further classified as either isotypic or novel.
  • CDC complement dependent cytotoxicity
  • isotypic modification as used herein is meant an amino acid modification that converts one amino acid of one isotype to the corresponding amino amino acid in a different, aligned isotype.
  • isotypic modification is meant an amino acid modification that converts one amino acid of one isotype to the corresponding amino amino acid in a different, aligned isotype.
  • IgGI has a tyrosine and lgG2 a phenylalanine at EU position 296, a F296Y substitution in lgG2 is considered an isotypic modification.
  • novel modification as used herein is meant an amino acid modification that is not isotypic. For example, because none of the IgGs has a glutamic acid at position 332, the substitution I332E in IgGI , lgG2, lgG3, or lgG4 is considered a novel 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 Fc ⁇ R- mediated effector functions such as ADCC and ADCP, and complement-mediated effector functions such as 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 DD T cells, and may be from any organism including but not limited to humans, mice, rats, rabbits, and monkeys.
  • Fab or "Fab region” as used herein is meant the polypeptides that comprise the VH, CH1 , VH, and C
  • 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.
  • Fc refers to the last two constant region immunoglobulin domains of IgA, IgD, and IgG, and 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 (C ⁇ 2 and C ⁇ 3) and the hinge between Cgammai (C ⁇ 1 ) and Cgamma2 (C ⁇ 2).
  • Fc region may vary, the human IgG heavy chain Fc region is usually defined to comprise 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.
  • Fc fusion as used herein is meant a protein wherein one or more polypeptides is operably linked to Fc.
  • 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 a/., 1997, Curr Opin Immunol 9:195- 200, both hereby entirely incorporated by reference).
  • An Fc fusion combines the Fc region of an immunoglobulin with a fusion partner, which in general may be any protein, polypeptide or small molecule.
  • a fusion partner which in general may be any protein, polypeptide 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 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, preferrably an extracellular receptor that is implicated in disease.
  • Fc gamma receptor or “FcDR” as used herein is meant any member of the family of proteins that bind the IgG antibody Fc region and are substantially encoded by the FcDR genes. In humans 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, hereby
  • An Fc ⁇ R may be from any organism, including but not limited to humans, mice, rats, rabbits, and monkeys.
  • Mouse FcDRs include but are not limited to FcDRI (CD64), FcDRII (CD32), FcDRIII (CD16), and FcDRIII-2 (CD16- 2), as well as any undiscovered mouse FcDRs or Fc ⁇ R isoforms or allotypes.
  • 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 ligands include but are not limited to Fc ⁇ Rs, Fc ⁇ Rs, Fc ⁇ Rs, FcRn, C1 q, 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 FcDRs (Davis et al., 2002, Immunological Reviews 190:123-136, hereby entirely incorporated by reference). Fc ligands may include undiscovered molecules that bind Fc.
  • FcRH Fc receptor homologs
  • full length antibodv is meant the structure that constitutes the natural biological form of an antibody, including variable and constant regions.
  • the full length antibody of the IgG isotype is a tetramer and consists of two identical pairs of two immunoglobulin chains, each pair having one light and one heavy chain, each light chain comprising immunoglobulin domains V L and C L , and each heavy chain comprising immunoglobulin domains V H , CD1 , CD2, and CD3.
  • IgG antibodies may consist of only two heavy chains, each heavy chain comprising a variable domain attached to the Fc region.
  • IgG as used herein is meant a polypeptide belonging to the class of antibodies that are substantially encoded by a recognized immunoglobulin gamma gene. In humans this IgG comprises the subclasses or isotypes IgGI , lgG2, lgG3, and lgG4. In mice IgG comprises IgGI , lgG2a, lgG2b, lgG3.
  • immunoglobulin herein is meant a protein consisting of one or more polypeptides substantially encoded by immunoglobulin genes. Immunoglobulins include but are not limited to antibodies. Immunoglobulins may have a number of structural forms, including but not limited to full length antibodies, antibody fragments, and individual immunoglobulin domains.
  • immunoglobulin domain as used herein is meant a region of an immunoglobulin that exists as a distinct structural entity as ascertained by one skilled in the art of protein structure. Ig domains typically have a characteristic G-sandwich folding topology. The known Ig domains in the IgG isotype of antibodies are V H , CD1 , CD2, CD3, V L , and C L .
  • IgG' or "IgG immunoglobulin” as used herein is meant a polypeptide belonging to the class of antibodies that are substantially encoded by a recognized immunoglobulin gamma gene. In humans this class comprises the subclasses or isotypes IgGI , lgG2, lgG3, and lgG4.
  • 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, IgM, IgD, and IgE.
  • parent polypeptide By “parent polypeptide”, “parent protein”, “precursor polypeptide”, or “precursor protein” as used herein is meant an unmodified polypeptide that is subsequently modified to generate a variant.
  • Said 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 Fc polypeptide” as used herein is meant an Fc polypeptide that is modified to generate a variant, and by “parent antibody” as used herein is meant an antibody that is modified to generate a variant antibody.
  • 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 IgGI .
  • polypeptide or "protein” as used herein is meant at least two covalently attached amino acids, which includes proteins, polypeptides, oligopeptides and peptides.
  • 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 .
  • 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.
  • 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 VD, VD, and/or V H genes that make up the kappa, lambda, and heavy chain immunoglobulin genetic loci respectively.
  • variant polypeptide polypeptide variant
  • variant polypeptide a polypeptide sequence that differs from that of a parent polypeptide sequence by virtue of at least one amino acid modification.
  • the parent polypeptide may be a naturally occurring or wild-type (WT) polypeptide, or may be a modified version of a WT polypeptide.
  • variant polypeptide may refer to the polypeptide itself, a composition comprising the polypeptide, or the amino sequence that encodes it.
  • the variant polypeptide has at least one amino acid modification compared to the parent polypeptide, 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 variant polypeptide sequence herein will preferably possess at least about 80% homology with a parent polypeptide sequence, and most preferably at least about 90% homology, more preferably at least about 95% homology. Accordingly, by "Fc variant” or “variant Fc” as used herein is meant an Fc sequence that differs from that of a parent Fc sequence by virtue of at least one amino acid modification.
  • An Fc variant may only encompass an Fc region, or may exist in the context of an antibody, Fc fusion, isolated Fc, Fc fragment, or other polypeptide that is substantially encoded by Fc.
  • Fc variant may refer to the Fc polypeptide itself, compositions comprising the Fc variant polypeptide, or the amino acid sequence that encodes it.
  • Fc polypeptide variant or “variant Fc polypeptide” as used herein is meant an Fc polypeptide that differs from a parent Fc polyeptide by virtue of at least one amino acid modification.
  • protein variant or “variant protein” as used herein is meant a protein that differs from a parent protein by virtue of at least one amino acid modification.
  • 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 immunoglobluin” 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.
  • 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, polypeptide, antibody, immunoglobulin, IgG, etc. has an amino acid sequence or a nucleotide sequence that has not been intentionally modified.
  • the present invention provides variant antibodies.
  • 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.
  • 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.
  • CDR complementarity-determining region
  • 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.
  • 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: “CH1” 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, and 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).
  • 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.
  • 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 CH1 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) 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.
  • scFv single chain Fv molecules
  • the scaffold components can be a mixture from different species.
  • the antibody 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 92/11018, Jones, 1986, Nature 321 :522-525, Verhoeyen et al., 1988, Science 239:1534-1536.
  • 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.
  • 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): 22611-22618; Rader et al., 1998, Proc. Natl. Acad. Sci.
  • 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), e.g., prepared chemically or from hybrid hybridomas.
  • the antibody is a minibody.
  • Minibodies are minimized antibody-like proteins comprising a scFv joined to a CH3 domain.
  • the scFv can be joined to the Fc region, and may include some or all of the hinge region.
  • 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").
  • antibody fusions are 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).
  • 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, that is implicated in disease.
  • 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, that 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.
  • 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).
  • 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 , hereby incorporated by reference in its entirety.
  • Covalent modifications of antibodies are included within the scope of this invention, and are generally, but not always, done post-translationally.
  • 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 also are 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.
  • 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.
  • 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-ethyI) 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 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 (Umana 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); (PotelligentTM technology [Biowa, Inc., Princeton, NJ]; GlycoMAbTM 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.
  • 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-hydroxyIysine 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 degiycosylation is described by Hakimuddin et al., 1987, Arch. Biochem. Biophys. 259:52 and by Edge et al., 1981 , Anal. Biochem. 118:131.
  • 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. 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. 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 U.S. Pat. Nos. 4,640,835; 4,496,689; 4,301 ,144; 4,670,417; 4,791 ,192 or 4,179,337.
  • 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. 2005/0114037, incorporated herein by reference in its entirety.
  • 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.
  • 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.
  • 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), FlTC, Rhodamine
  • 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.
  • 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
  • EYFP enhanced yellow fluorescent protein
  • luciferase lchiki et al., 1993, J. Immunol. 150:5408-5417
  • ⁇ galactosidase Nolan et al., 1988, Proc. Natl. Acad. Sci. U.S.A. 85:2603-2607
  • 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 are expressly incorporated herein by reference.
  • antibody can meant a protein consisting of one or more polypeptides substantially encoded by all or part of the recognized immunoglobulin genes.
  • the recognized immunoglobulin genes include the kappa (D), lambda (D), and heavy chain genetic loci, which together comprise the myriad variable region genes, and the constant region genes mu (D), delta (D), gamma (D), sigma (D), and alpha (D) which encode the IgM, IgD, IgG (IgGI , lgG2, lgG3, and lgG4), IgE, and IgA (IgAI and lgA2) isotypes respectively.
  • Antibody herein is meant to include full length antibodies and antibody fragments, and may refer to a natural antibody from any organism, an engineered antibody, or an antibody generated recombinantly for experimental, therapeutic, or other purposes.
  • An Fc variant comprises one or more amino acid modifications relative to a parent Fc polypeptide, wherein said amino acid modification(s) provide one or more optimized properties.
  • An Fc variant of the present invention differs in amino acid sequence from its parent IgG by virtue of at least one amino acid modification.
  • Fc variants of the present invention have at least one amino acid modification compared to the parent.
  • the Fc variants of the present invention may have more than one amino acid modification as compared to the parent, for example from about one to fifty amino acid modifications, preferrably from about one to ten amino acid modifications, and most preferably from about one to about five amino acid modifications compared to the parent.
  • sequences of the Fc variants and those of the parent Fc polypeptide are substantially homologous.
  • variant Fc variant sequences herein will possess about 80% homology with the parent Fc 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.
  • 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/A330L/I332E defines an Fc variant with the substitutions S239D, A330L, and I332E relative to the parent Fc polypeptide. It is noted that the order in which substitutions are provided is arbitrary, that is to say that, for example, S239D/A330L/I332E is the same Fc variant as S239D/I332E/A330L, 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).
  • Fc variants of the present invention may be substantially encoded by genes from any organism, preferably mammals, including but not limited to humans, rodents including but not limited to mice and rats, lagomorpha including but not limited to rabbits and hares, camelidae including but not limited to camels, llamas, and dromedaries, and non-human primates, including but not limited to Prosimians, Platyrrhini (New World monkeys), Cercopithecoidea (Old World monkeys), and Hominoidea including the Gibbons and Lesser and Great Apes.
  • the Fc variants of the present invention are substantially human.
  • the parent Fc polypeptide may be an antibody.
  • Parent antibodies may be fully human, obtained for example using transgenic mice (Bruggemann et al., 1997, Curr Opin Biotechnol 8:455- 458, hereby entirely incorporated by reference) or human antibody libraries coupled with selection methods (Griffiths et al., 1998, Curr Opin Biotechnol 9:102-108, hereby entirely incorporated by reference).
  • the parent antibody need not be naturally occurring.
  • the parent antibody may be an engineered antibody, including but not limited to chimeric antibodies and humanized antibodies (Clark, 2000, Immunol Today 21 :397-402, hereby entirely incorporated by reference).
  • the parent antibody may be an engineered variant of an antibody that is substantially encoded by one or more natural antibody genes.
  • the parent antibody has been affinity matured, as is known in the art.
  • the antibody has been modified in some other way, for example as described in USSN 10/339788, filed on March 3, 2003, hereby entirely incorporated by reference.
  • the Fc variants of the present invention may be substantially encoded by immunoglobulin genes belonging to any of the antibody classes.
  • the Fc variants of the present invention find use in antibodies or Fc fusions that comprise sequences belonging to the IgG class of antibodies, including IgGI, lgG2, lgG3, or lgG4.
  • Figure 3 provides an alignment of these human IgG sequences.
  • the Fc variants of the present invention find use in antibodies or Fc fusions that comprise sequences belonging to the IgA (including subclasses IgAI and lgA2), IgD, IgE, IgG, or IgM classes of antibodies.
  • the Fc variants of the present invention may comprise more than one protein chain. That is, the present invention may find use in an antibody or Fc fusion that is a monomer or an oligomer, including a homo- or hetero-oligomer.
  • Gm polymorphism is determined by the IGHG1 , IGHG2 and IGHG3 genes which have alleles encoding allotypic antigenic determinants referred to as G1m, G2m, and G3m allotypes for markers of the human IgGI , lgG2 and lgG3 molecules (no Gm allotypes have been found on the gamma 4 chain). Markers may be classified into 'allotypes' and 'isoallotypes'. These are distinguished on different serological bases dependent upon the strong sequence homologies between isotypes.
  • Allotypes are antigenic determinants specified by allelic forms of the Ig genes. Allotypes represent slight differences in the amino acid sequences of heavy or light chains of different individuals. Even a single amino acid difference can give rise to an allotypic determinant, although in many cases there are several amino acid substitutions that have occurred. Allotypes are sequence differences between alleles of a subclass whereby the antisera recognize only the allelic differences.
  • An isoallotype is an allele in one isotype which produces an epitope which is shared with a non-polymorphic homologous region of one or more other isotypes and because of this the antisera will react with both the relevant allotypes and the relevant homologous isotypes (Clark, 1997, IgG effector mechanisms, Chem Immunol. 65:88-110; Gorman & Clark, 1990, Semin Immunol 2(6):457-66, both hereby entirely incorporated by reference).
  • Gm haplotypes G1m (1 , 2, 3, 17) or G1m (a, x, f, z), G2m (23) or G2m (n), G3m (5, 6, 10, 11 , 13, 14, 15, 16, 21 , 24, 26, 27, 28) or G3m (b1 , c3, b5, b ⁇ , b3, b4, s, t, g1 , c5, u, v, g5)
  • Lefranc, et al. The human IgG subclasses: molecular analysis of structure, function and regulation. Pergamon, Oxford, pp. 43-78 (1990); Lefranc, G. et al., 1979, Hum. Genet.: 50, 199-211 , both hereby entirely incorporated by reference). Allotypes that are inherited in fixed combinations are called Gm haplotypes.
  • Figure 4 shows the allotypes and isoaliotypes of the gammal chain of human IgGI showing the positions and the relevant amino acid substitutions (Gorman & Clark, 1990, Semin Immunol 2(6):457-66, hereby entirely incorporated by reference). For comparison the amino acids found in the equivalent positions in human lgG2, lgG3 and lgG4 gamma chains are also shown.
  • the Fc variants of the present invention may be substantially encoded by any allotype or isoallotype of any immunoglobulin gene.
  • the Fc variants of the present invention find use in antibodies or Fc fusions that comprise IgGI sequences that are classified as G1m(1), G1m(2), G1m(3), G1m(17), nG1m(1), nG1 m(2), and/or nG1 m(17).
  • the Fc variants of the present invention may comprise a Lys (G1m(17)) or Arg (G1m(3)) at position 214, an Asp356/Leu358 (G1m(1)) or Glu356/Met358 (nG1m(1)), and/or a GIy (G1m(2)) or Ala (nG1m(2)) at position 431.
  • the Fc variants of the invention are 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.
  • Fc variants may also comprise sequences from other immunoglobulin classes such as IgA, IgE, IgGD, IgGM, and the like. It is contemplated that, although the Fc variants of the present invention 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 Fc 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 about 0.13 nm and preferably about 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 Fc variant does not affect the ability to transfer the Fc variants of the present invention to other parent IgGs.
  • any antigen may be targeted by the Fc variants of the present invention, including but not limited to proteins, subunits, domains, motifs, and/or epitopes belonging to the following list of targets: 17-IA, 4-1 BB, 4Dc, 6 ⁇ keto-PGF1a, 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
  • the present invention provides Fc variants that are optimized for a variety of therapeutically relevant properties.
  • An Fc variant that is engineered or predicted to display one or more optimized properties is herein referred to as an "optimized Fc variant".
  • Properties that may be optimized include but are not limited to enhanced or reduced affinity for an Fc ⁇ R.
  • the Fc variants of the present invention are optimized to possess enhanced affinity for a human activating Fc ⁇ R, preferably Fc ⁇ RI, Fc ⁇ Rlla, Fc ⁇ Rllc, Fc ⁇ Rllla, and Fc ⁇ Rlllb, most preferably Fc ⁇ Rllla.
  • the Fc variants are optimized to possess reduced affinity for the human inhibitory receptor Fc ⁇ Rllb.
  • Fc variants of the present invention are optimized to have reduced or ablated affinity for a human Fc ⁇ R, including but not limited to Fc ⁇ RI, Fc ⁇ Rlla, Fc ⁇ Rllb, Fc ⁇ Rllc, Fc ⁇ Rllla, and Fc ⁇ Rlllb. These embodiments are anticipated to provide IgG polypeptides with enhanced therapeutic properties in humans, for example reduced effector function and reduced toxicity. In other embodiments, Fc variants of the present invention provide enhanced affinity for one or more Fc ⁇ Rs, yet reduced affinity for one or more other Fc ⁇ Rs.
  • an Fc variant of the present invention may have enhanced binding to Fc ⁇ Rllla, yet reduced binding to Fc ⁇ Rllb.
  • an Fc variant of the present invention may have enhanced binding to Fc ⁇ Rlla and Fc ⁇ RI, yet reduced binding to Fc ⁇ Rllb.
  • an Fc variant of the present invention may have enhanced affinity for Fc ⁇ Rllb, yet reduced affinity to one or more activating Fc ⁇ Rs.
  • Preferred embodiments comprise optimization of binding to a human Fc ⁇ R, however in alternate embodiments the Fc variants of the present invention possess enhanced or reduced affinity for Fc ⁇ Rs from nonhuman organisms, including but not limited to rodents and non-human primates.
  • Fc variants that are optimized for binding to a nonhuman Fc ⁇ R may find use in experimentation.
  • 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.
  • cancer cells can be grafted or injected into mice to mimic a human cancer, a process referred to as xenografting.
  • Fc variants that comprise Fc variants that are optimized for one or more mouse Fc ⁇ Rs, may provide valuable information with regard to the efficacy of the protein, its mechanism of action, and the like.
  • the Fc variants of the present invention may also be optimized for enhanced functionality and/or solution properties in aglycosylated form.
  • the aglycosylated Fc variants of the present invention bind an Fc ligand with greater affinity than the aglycosylated form of the parent Fc variant.
  • Said Fc ⁇ gands include but are not limited to Fc ⁇ Rs, C1q, FcRn, 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 Fc variants are optimized to be more stable and/or more soluble than the aglycosylated form of the parent Fc variant.
  • Fc variants of the invention may comprise modifications that modulate interaction with Fc ligands other than Fc ⁇ Rs, including but not limited to complement proteins, FcRn, 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, hereby entirely incorporated by reference).
  • the Fc ligand specificity of the Fc variant of the present invention will determine its therapeutic utility.
  • the utility of a given Fc variant for therapeutic purposes will depend on the epitope or form of the Target antigen and the disease or indication being treated.
  • enhanced Fc ⁇ R-mediated effector functions may be preferable. This may be particularly favorable for anti-cancer Fc variants.
  • Fc variants may be used that comprise Fc variants that provide enhanced affinity for activating Fc ⁇ Rs and/or reduced affinity for inhibitory Fc ⁇ Rs.
  • Fc 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 ⁇ Rl, whereas in other cases, enhanced binding only to Fc ⁇ Rlla may be preferred.
  • Fc variants that enhance both Fc ⁇ R-mediated and complement-mediated effector functions, whereas for other cases it may be advantageous to utilize Fc variants that enhance either Fc ⁇ R-mediated or complement- mediated effector functions.
  • Fc 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 Fc variant is to inhibit inflammation or auto-immune disease, or modulate the immune system in some way.
  • Fc ligand selectivity or specifity of a given Fc variant will provide different properties depending on whether it composes an antibody, Fc fusion, or Fc variants with a coupled fusion or conjugate partner.
  • toxin, radionucleotide, or other conjugates may be less toxic to normal cells if the Fc variant that comprises them has reduced or ablated binding to one or more Fc ligands.
  • an Fc variant with enhanced affinity for activating Fc ⁇ Rs such as to bind these Fc ⁇ Rs and prevent their activation.
  • an Fc variant that comprises two or more Fc regions with enhanced Fc ⁇ Rllb affinity may co-engage this receptor on the surface of immune cells, thereby inhibiting proliferation of these cells.
  • an Fc variants may engage its target antigen on one cell type yet engage Fc ⁇ Rs on separate cells from the target antigen, in other cases it may be advantageous to engage Fc ⁇ Rs on the surface of the same cells as the target antigen.
  • an antibody targets an antigen on a cell that also expresses one or more Fc ⁇ Rs
  • antigen and Fc ⁇ R co-engagement on the same cell may be advantageous when the Fc variant is being used to modulate the immune system in some way, wherein co-engagement of target antigen and Fc ⁇ R provides some proliferative or anti-proliferative effect.
  • Fc variants that comprise two or more Fc regions may benefit from Fc variants that modulate Fc ⁇ R selectivity or specifity to co-engage Fc ⁇ Rs on the surface of the same cell.
  • Fc ⁇ Rs The presence of different polymorphic forms of Fc ⁇ Rs provides yet another parameter that impacts the therapeutic utility of the Fc variants of the present invention.
  • specificity and selectivity of a given Fc variant for the different classes of Fc ⁇ Rs signficantly affects the capacity of an Fc variant to target a given antigen for treatment of a given disease
  • the specificity or selectivity of an Fc variant for different polymorphic forms of these receptors may in part determine which research or pre-clinical experiments may be appropriate for testing, and ultimately which patient populations may or may not respond to treatment.
  • Fc variants of the present invention may be used to guide the selection of valid research and pre-clinical experiments, clinical trial design, patient selection, dosing dependence, and/or other aspects concerning clinical trials.
  • the Fc variants of the present invention may comprise modifications to reduce immunogenicity in humans.
  • the immunogenicity of an Fc variant of the present invention is reduced using a method described in USSN 11/004,590, filed December 3, 2004, hereby entirely incorporated by reference.
  • the Fc variants of the present invention are humanized (Clark, 2000, Immunol Today 21 :397-402, hereby entirely incorporated by reference).
  • humanized antibody as used herein is meant an antibody comprising a human framework region (FR) and one or more complementarity determining regions (CDR's) from a non-human (usually mouse or rat) antibody.
  • the non-human antibody providing the CDR's is called the “donor” and the human immunoglobulin providing the framework is called the “acceptor”.
  • Humanization relies principally on the grafting of donor CDRs onto acceptor (human) VL and VH frameworks (e.g., Winter et al, US 5225539, hereby entirely incorporated by reference). This strategy is referred to as "CDR grafting".
  • 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.
  • 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, hereby entirely incorporated by reference.
  • the parent antibody has been affinity matured, as is well known in the art. Structure-based methods may be employed for humanization and affinity maturation, for example as described in USSN 11/004,590, hereby entirely incorporated by reference.
  • 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): 22611-22618; Rader et al., 1998, Proc. Natl. Acad. Sci. USA 95: 8910-8915; Krauss et al., 2003, Protein Engineering 16(10):753-759, all hereby entirely incorporated by reference.
  • Modifications to reduce immunogenicity may include modifications that reduce binding of processed peptides derived from the parent sequence to MHC proteins.
  • amino acid modifications may 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 Fc variant of the present invention.
  • 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 hereby entirely incorporated by reference).
  • the Fc variants of the present invention comprise one or more engineered glycoforms.
  • engineered qlvcoform 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 (Umana 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 ); (PotelligentTM technology [Biowa, Inc., Princeton, NJ]; GlycoMAb® glycosylation engineering technology [GLYCART biotechnology AG, Zurich, Switzerland], all hereby entirely incorporated by reference).
  • Engineered glycoform typically refers to the different carbohydrate or oligosaccharide; thus an Fc variant, for example an antibody or Fc fusion, may comprise an engineered glycoform.
  • engineered glycoform may refer to the Fc variant that comprises the different carbohydrate or oligosaccharide.
  • the Fc variant of the present invention 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.
  • the present invention provides methods for engineering, producing, and screening Fc variants.
  • the described methods are not meant to constrain the present invention to any particular application or theory of operation. Rather, the provided methods are meant to illustrate generally that one or more Fc variants may be engineered, produced, and screened experimentally to obtain Fc variants with optimized effector function.
  • a variety of methods are described for designing, producing, and testing antibody and protein variants in USSN 10/672,280, USSN 10/822,231 , USSN 11/124,620, and USSN 11/256,060, all hereby entirely incorporated by reference.
  • a variety of protein engineering methods may be used to design Fc variants with optimized effector function.
  • a structure-based engineering method may be used, wherein available structural information is used to guide substitutions.
  • An alignment of sequences may be used to guide substitutions at the identified positions.
  • random or semi-random mutagenesis methods may be used to make amino acid modifications at the desired positions.
  • the Fc 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 the present invention 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 Fc variants of the present invention may be produced by culturing a host cell transformed with nucleic acid, preferably an expression vector, containing nucleic acid encoding the Fc variants, under the appropriate conditions to induce or cause expression of the protein.
  • a host cell transformed with nucleic acid preferably an expression vector, containing nucleic acid encoding the Fc variants
  • 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 in the present invention are described in the ATCC cell line catalog, available from the American Type Culture Collection.
  • the methods of introducing exogenous nucleic acid into host cells are well known in the art, and will vary with the host cell used.
  • Fc 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 the present invention for purification. Purification can often 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.
  • 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.
  • 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.
  • 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 immunogenicity of the Fc variants is determined experimentally using one or more cell-based assays. Several methods can be used for experimental confirmation of epitopes.
  • the biological properties of the Fc variants of the present invention 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.
  • monkeys can be suitable therapeutic models, and thus may be used to test the efficacy, toxicity, pharmacokinetics, or other property of the IgGs of the present invention. Tests of the in humans are ultimately required for approval as drugs, and thus of course these experiments are contemplated.
  • the IgGs of the present invention may be tested in humans to determine their therapeutic efficacy, toxicity, immunogenicity, pharmacokinetics, and/or other clinical properties.
  • the Fc variants of the present invention may find use in a wide range of products.
  • the Fc variant of the present invention is a therapeutic, a diagnostic, or a research reagent, preferably a therapeutic.
  • the Fc variant may find use in an antibody composition that is monoclonal or polyclonal.
  • the Fc variants of the present invention are used to kill target cells that bear the target antigen, for example cancer cells.
  • the Fc variants of the present invention are used to block, antagonize, or agonize the target antigen, for example for antagonizing a cytokine or cytokine receptor.
  • the Fc variants of the present invention are used to block, antagonize, or agonize the target antigen and kill the target cells that bear the target antigen.
  • the Fc variants of the present invention may be used for various therapeutic purposes.
  • an antibody comprising the Fc variant is administered to a patient to treat an antibody-related disorder.
  • a "patient” for the purposes of the present invention 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 Fc variant of the present invention.
  • Antibody related disorders include but are not limited to autoimmune diseases, immunological diseases, infectious diseases, inflammatory diseases, neurological diseases, pain, pulmonary diseases, hematological conditions, fibrotic conditions, 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.
  • treatment as used herein is meant to include therapeutic treatment, as well as prophylactic, or suppressive measures for the disease, condition or disorder.
  • successful administration of a pharmaceutical composition comprising an Fc variant of the present invention prior to onset of the disease results in "treatment" of the disease.
  • successful administration of a pharmaceutical composition comprising an Fc variant of the present invention after clinical manifestation of the disease to combat the symptoms of the disease comprises “treatment” of the disease.
  • Treatment also encompasses administration of a pharmaceutical composition comprising an Fc variant of the present invention after the appearance of the disease in order to eradicate the disease.
  • Successful administration of a pharmaceutical composition comprising an Fc variant of the present invention after onset and after clinical symptoms have developed, with possible abatement of clinical symptoms and perhaps amelioration of the disease comprises "treatment” of the disease.
  • Those "in need of treatment” as used herein include mammals already having the disease or disorder, as well as those prone to having the disease or disorder, including those in which the disease or disorder is to be prevented.
  • an Fc variant of the present invention is the only therapeutically active agent administered to a patient.
  • the Fc variant of the present invention 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, as well as pre- or post-surgery.
  • the IgG variants may be administered concomitantly with one or more other therapeutic regimens.
  • an Fc variant of the present invention may be administered to the patient along with surgery, chemotherapy, radiation therapy, or any or all of surgery, chemotherapy and radiation therapy.
  • the Fc variant of the present invention may be administered in conjunction with one or more antibodies, which may or may not comprise an Fc variant of the present invention.
  • the Fc variant of the present invention 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 Fc variants of the invention 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 Fc variants of the present invention are administered with 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.
  • compositions are contemplated wherein an Fc variant of the present invention and one or more therapeutically active agents are formulated.
  • Formulations of the Fc variants of the present invention are prepared for storage by mixing said 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, hereby 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 Fc 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 Fc variant in the formulation may vary from about 0.001 to 100 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 Fc variant of the present invention 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 0.001 to 100 mg/kg of body weight or greater, for example 0.1 , 1 , 10, or 50 mg/kg of body weight, with 1 to 10mg/kg being preferred.
  • Administration of the pharmaceutical composition comprising an Fc variant of the present invention may be done in a variety of ways, including, but not limited to, orally, subcutaneously, intravenously, intranasaliy, intraotically, transdermally, topically (e.g., gels, salves, lotions, creams, etc.), intraperitoneally, intramuscularly, intrapulmonary (e.g., AERx® inhalable technology commercially available from Aradigm, or Inhance® pulmonary delivery system commercially available from Inhale Therapeutics), vaginally, parenterally, rectally, or intraocularly.
  • intravenously intravenously, intranasaliy, intraotically, transdermally, topically (e.g., gels, salves, lotions, creams, etc.), intraperitoneally, intramuscularly, intrapulmonary (e.g., AERx® inhalable technology commercially available from Aradigm, or Inhance® pulmonary delivery system commercially available from Inhal
  • Figure 5 provides a list of these Fc variants.
  • this variant set comprises a number of insertions.
  • “Insert L > 235-236 / I332E” refers to a double mutant comprising the substitution I332E and an insertion of leucine between residues 235 and 236.
  • Plasmids containing heavy chain gene (VH-CH1-CH2-CH3) (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). Select Fc variants were also expressed in the context of alemtuzumab.
  • Binding affinity to human Fc ⁇ Rs by IgG antibodies was measured using a competitive AlphaScreenTM assay.
  • the AlphaScreen is a bead-based 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 AlphaScreen was applied as a competition assay for screening the antibodies. Wild-type IgGI antibody was biotinylated by standard methods for attachment to streptavidin donor beads, and tagged Fc ⁇ R was bound to glutathione chelate acceptor beads.
  • wild-type antibody and Fc ⁇ R interact and produce a signal at 520-620 nm.
  • Addition of untagged antibody competes with wild-type Fc/Fc ⁇ R interaction, reducing fluorescence quantitatively to enable determination of relative binding affinities.
  • Figures 6 provides competitive AlphaScreen data for binding of select PRO70769 Fc variants to the human activating receptors V158 Fc ⁇ Rllla ( Figure 6a) and F158 Fc ⁇ Rllla ( Figure 6b).
  • the data were fit to a one site competition model using nonlinear regression, and these fits are represented by the curves in the figure. These fits provide the inhibitory concentration 50% (IC50) (i.e. the concentration required for 50% inhibition) for each antibody, thus enabling the relative binding affinities relative to WT to be determined.
  • Figure 5 provides the IC50's and Fold IC50's relative to WT for fits to these binding curves.
  • WT and variant trastuzumab antibodies were expressed and purified as described above.
  • data were acquired on a BIAcore 3000 instrument (BIAcore).
  • V158 Fc ⁇ Rllla-His-GST was captured using immobilized anti-GST antibody, blocked with recombinant GST, and binding to antibody/receptor competition analyte was measured.
  • Anti-GST antibody was covalently coupled to a CM5 sensor using the BIAcore GST Capture Kit. Flow cell 1 of every sensor chip was coupled with ethanolamine as a control of unspecific binding and to subtract bulk refractive index changes online.
  • Running buffer was HBS-EP (0.01 M HEPES pH 7.4, 0.15 M NaCI, 3 mM EDTA, 0.005% v/v Surfactant P20, BIAcore), and chip regeneration buffer was Glycine 1.5 (10 mM glycine-HCI, pH 1.5, BIAcore).
  • Glycine 1.5 10 mM glycine-HCI, pH 1.5, BIAcore.
  • 1 ⁇ M V158 Fc ⁇ Rllla-His-GST was bound to the anti-GST CM5 chip in HBS-EP at 1 ⁇ l/min for 5 minutes. The surface was blocked with 5 ⁇ M recombinant GST (Sigma) injected at 1 ⁇ l/minute for 2 minutes.
  • V158 Fc ⁇ Rllla-His-GST 100 nM wild-type or variant trastuzumab antibody was combined with V158 Fc ⁇ Rllla-His-GST in serial dilutions between 4 and 1000 nM and incubated for at least two hours at room temperature.
  • the competition mixture was injected over the V158 Fc ⁇ Rllla-His-GST/recombinant GST surface for 30 seconds association in HBS-EP at 50 ⁇ l/minute. A cycle with antibody but no competing receptor provided a baseline response.
  • R - ⁇ -([A 0 ]- l0 x - K D ) + 4(K D 2 + 2 ⁇ W)(K D ) + (10 x ) 2 + 2[A 0 ]K 0 - 2[A 0 )IO* +[A 0 ] 2 ) 2IA 0 ] with:
  • R 0 reflects the rate of binding between antibody and immobilized receptor (in the absence of competing receptor), and because of their different receptor affinities was calculated separately for WT, I332E, and S239D/I332E antibodies.
  • the formula for the initial rate R is derived from the definition of K 0 for a single binding site:
  • ADCC was measured by the release of lactose dehydrogenase using a LDH Cytotoxicity Detection Kit (Roche Diagnostic).
  • Human PBMCs were purified from leukopacks using a ficoll gradient, and the CD20+ target lymphoma cell line WIL2-S was obtained from ATCC.
  • Target cells were seeded into 96-well plates at 10,000 cells/well, and opsonized using Fc variant or WT antibodies at the indicated final concentration. Triton X100 and PBMCs alone were run as controls.
  • the ADCC assays were repeated in the presence of a biologically relevant (1 mg/ml) concentration of IgG purified from human serum (purchased commercially from Jackson lmmunoresearch Lab, Inc.). These data are provided in Figure 11.
  • the efficacy of the WT anti-CD20 antibody is not only reduced, but completely ablated in the presence of serum level IgG.
  • the Fc variant antibodies although significantly reduced, still show substantial capacity to mediate killing against the target cell line.
  • Figure 12 shows a structure of the human IgGI Fc region with this epicenter mapped.
  • Select amino acid modifications disclosed in USSN 10/672,280, USSN 10/822,231 , USSN 11/124,620, and USSN 11/256,060, all hereby entirely incorporated by reference, that are structurally proximal to these four residues were investigated to explore variants that may mediate increased affinity for C1q and and/or provide enhanced CDC.
  • Variants that previously showed enhanced Fc ⁇ R affinity and Fc ⁇ R-mediated effector function were included in this set of variants to characterize their complement properties.
  • This variant library is provided in Figure 13.
  • variants were constructed as described above in the context of the anti-CD20 antibody PRO70769 (variable region) and either IgGI or lgG(1/2) ELLGG as the heavy chain constant region. Variants were expressed and purified as described above.
  • a cell-based assay was used to measure the capacity of the Fc variants to mediate CDC. Lysis was measured using release of Alamar Blue to monitor lysis of Fc variant and WT PRO70769 -opsonized WIL2-S lymphoma cells by human serum complement. Target cells were washed 3x in 10% FBS medium by centrifugation and resuspension, and WT or variant rituximab antibody was added at the indicated final concentrations.
  • Human serum complement (Quidel) was diluted 50% with medium and added to antibody-opsonized target cells. Final complement concentration was 1/6 th original stock. Plates were incubated for 2 hrs at 37 0 C, Alamar Blue was added, cells were cultured for two days, and fluorescence was measured. Representative data from this assay are shown in Figure 14. The binding data were normalized to the maximum and minimum luminescence signal for each particular curve, provided by the baselines at low and high antibody concentrations respectively. The data were fit to a sigmoidal dose-response with variable slope model using nonlinear regression, and these fits are represented by the curves in the figure. These fits provide the effective concentration 50% (EC50) (i.e.
  • modifications that show 0.5 fold and lower relative CDC include 235D, 239D, 284D, 322H, 322T, 322Y, 327R, 330E, 3301, 330L, 330N, 330V, 331 D, and 331 L, 332E ( Figure 15). These modifications provide further valuable structure activity relationship (SAR) information that may be used to guide further design of variants for enhanced CDC.
  • SAR structure activity relationship
  • Fc ⁇ R- and complement- binding A previously unconsidered advantage of ablated Fc ⁇ R- and complement- binding is that in cases where effector function is not needed, binding to Fc ⁇ R and complement may effectively reduce the active concentration of drug. Binding to Fc ligands may localize an antibody or Fc fusion to cell surfaces or in complex with serum proteins wherein it is less active or inactive relative to when it is free (uncomplexed).
  • Fc ⁇ R- receptors may be one mechanism of antibody turnover, and can mediate uptake and processing by antigen presenting cells such as dendritic cells and macrophages. This may affect affect the pharmacokinetics (or in vivo half-life) of the antibody or Fc fusion and its immunogenicity, both of which are critical parameters of clinical performance.
  • Figure 17 shows AlphaScreen data for binding of select Fc variants to human V158 Fc ⁇ Rllla, and Figure 16 provides their Fold IC ⁇ O's relative to WT PRO70769 IgGI .
  • the variants were also investigated for their capacity to mediate complement- mediated lysis against CD20+ WIL2-S lymphoma target cells using the CDC assay described above.
  • Figure 18 provides CDC data for select Fc variants, and Figure 16 provides their Fold EC50's relative to WT PRO70769 IgGI . Based on the results of these experiments, select Fc variants were characterized for their capacity to mediate Fc ⁇ R-mediated effector function.
  • An ADCC assay using human PBMCs as effector cells and WIL2-S lymphoma cells as target cells was carried out as described above.
  • Figure 19 shows these ADCC data for select variants.
  • modification at a number of positions provide reduced or ablated Fc ⁇ R affinity, reduced Fc ⁇ R-mediated effector function, and reduced complement-mediated effector function.
  • modifications at some positions may provide reduced CDC but WT Fc ⁇ R affinity.
  • 235D, 330L, 330N, and 330R display such behavior.
  • modification at some positions including but not limited to 236 and 299, may provide reduced Fc ⁇ R affinity but WT level CDC. For example 236I and 299A show these properties.
  • WT IgGI antibody As well as lgG2 and lgG4 antibody versions, an aglycosylated variant N297S, and two variants previously characterized as having reduced effector function: L234A/L235A (Xu et al., 2000, Cellular Immunology 200:16-26; USSN 10/267,286, hereby entirely incorporated by reference) and E233P/L234V/L235A/G236- (Armour et al., 1999, Eur J Immunol 29:2613-2624, hereby entirely incorporated by reference).
  • L234A/L235A Xu et al., 2000, Cellular Immunology 200:16-26; USSN 10/267,286, hereby entirely incorporated by reference
  • E233P/L234V/L235A/G236- Armour et al., 1999, Eur J Immunol 29:2613-2624, hereby entirely incorporated by reference).
  • trastuzumab variant region sequences provided in Figures 24c and 24d.
  • trastuzumab robustly provides a substantial signal in ADCC assays against Her2+ expressing cell lines, and therefore provides a stringent test of the Fc variants for reducing/ablating effector function.
  • L235G, G236R, G237K, N325L, N325A, L328R, L235G/G236R, G236R/G237K, G236R/N325L, G236R/L328R, G237K/N325L, L235G/G236R/G237K, and G236R/G237K/L328R were constructed in the context of trastuzumab IgGI .
  • WT IgGI ,WT lgG2, and WT lgG4 antibody versions were constructed as well.
  • An ADCC assay was carried out as described above, except the Her2+ breast carcinoma cell line SkBr-3 was used as target cells.
  • Figure 23 provides the results of the ADCC experiments. The data indicate that some of the variants completely ablate ADCC. Additionally, although lgG2 also appears to mediate no ADCC, lgG4 does show a significant level of ADCC.
  • amino acid modifications 232G, 234G, 234H, 235D, 235G, 235H, 236I, 236N, 236P, 236R, 237K, 237L, 237N, 237P, 238K, 239R, 265G, 267R, 269R, 270H, 297S, 299A, 299I, 299V, 325A, 325L, 327R, 328R, 329K, 330I, 330L, 330N, 330P, 330R, 330S, and 331 L provide significantly reduced Fc ligand binding properties and/or effector function.
  • amino acid modifications that compose these variants are capable of reducing binding to both Fc ⁇ Rllla and Fc ⁇ RI, and reducing CDC by greater than 10 fold.
  • the data show that human lgG2 has significantly reduced Fc ⁇ R-affinity, Fc ⁇ R-mediated effector function, and complement-mediated effector function relative to human lgG4.
  • reduced Fc ⁇ R affinity and/or effector function may be optimal for Fc polypeptides for which Fc ligand binding or effector function leads to toxicity and/or reduced efficacy.
  • Fc ligand binding or effector function leads to toxicity and/or reduced efficacy.
  • antibodies that target CTLA-4 block inhibition of T-celi activation and are effective at promoting anti-tumor immune response, but destruction of T cells via antibody mediated effector functions may be counterproductive to mechanism of action and/or potentially toxic. Indeed toxicity has been observed with clinical use of the anti-CTLA-4 antibody ipilimumab (Maker et al., 2005, Ann Surg Oncol 12:1005-16, hereby entirely incorporated by reference).
  • the optimized antibody sequences sequences comprise at least one non-WT amino acid selected from the group consisting of X 1 , X 2 , X 3 , X 4 , X 5 , Xe, X 7 , and X 8 .
  • an improved anti- CTLA-4 antibody sequence comprising the L235G and G236R modifications in the IgGI constant region are provided in Figures 26b and 26c.
  • lgG2 and lgG4 can also be used to reduce Fc ligand binding and Fc-mediated effector function.
  • Figures 26b and 26d provide the sequences of improved anti-CTLA-4 lgG2 antibody sequences.
  • an anti-CTLA-4 here is solely an example, and is not meant to constrain application of the Fc variants to this antibody or any other particular Fc polypeptide.
  • Other exemplary applications for reduced Fc ligand binding and/or effector function include but are not limited to anti-TNF ⁇ antibodies, including for example infliximab and adalimumab, anti-VEGF antibodies, including for example bevacizumab, anti- ⁇ 4-integrin antibodies, including for example natalizumab, and anti-CD32b antibodies, including for example those described in USSN 10/643,857, hereby entirely incorporated by reference.

Landscapes

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

Abstract

The present invention relates to Fc variants with optimized properties, methods for their generation, Fc polypeptides comprising Fc variants with optimized properties, and methods for using Fc variants with optimized properties.

Description

Fc VARIANTS WITH OPTIMIZED PROPERTIES
[1] This application claims benefit under 35 U.S.C. §119(e) to USSNs 60/667,197, filed March 31 , 2005; 60/705,378 filed August 3, 2005; 60/723,294 filed October 3, 2005; and 60/723,335 filed October 3, 2005; and is contination-in-part of USSNs 11/124,620, filed May 5, 2005, each of which is incorporated by reference herein in its entirety. The disclosures of U.S. Patent Application No. 10/822,231 , filed March 26, 2004 is also expressly incorporated by reference in their entirety.
FIELD OF THE INVENTION
[2] The present invention relates to Fc variant polypeptides with optimized properties, 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 features between these antibody classes are 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-CH1-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-CD1-CD2-CD3, 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, hereby 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, hereby 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 comprising the variable region can exist in the absence of other regions of the antibody, including for example the antigen binding fragment (Fab) comprising Vπ-Cγ1 and VH-CL, the variable fragment (Fv) comprising VH and VL, the single chain variable fragment (scFv) comprising 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, hereby 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 Figure 1, comprises Ig domains Cγ2 and Cγ3 and the N-terminal hinge leading into Cγ2. An important family of Fc receptors for the IgG class are 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 hereby 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, hereby entirely incorporated by reference). These receptors typically have an extracellular domain that mediates binding to Fc1 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, both hereby entirely incorporated by reference).
The ceil-mediated reaction wherein nonspecific cytotoxic cells that express FcγRs recognize bound antibody on a target cell and subsequently cause phagocytosis of the target ceil 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)(Sondermann et al., 2001 , J MoI Biol 309:737-749, hereby entirely incorporated by reference)
(pdb accession code 1 FCG)(Maxwell et al., 1999, Nat Struct Biol 6:437-442, hereby entirely incorporated by reference), FcγRllb (pdb accession code 2FCB)(Sondermann et al., 1999, Embo J
18:1095-1103, hereby entirely incorporated by reference); and FcγRlllb (pdb accession code
1 E4J)(Sondermann et al., 2000, Nature 406:267-273, hereby 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 2. This interaction is well characterized structurally (Sondermann et al., 2001 , J MoI
Biol 309:737-749, hereby 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)(Sondermann et al., 2000, Nature 406:267-273, hereby entirely incorporated by reference) (pdb accession codes 11IS and 11IX)(Radaev et al., 2001 , J Biol Chem 276:16469-16477, hereby entirely incorporated by reference), as well as has the structure of the human IgE Fc/FcDRlD complex (pdb accession code 1 F6A)(Garman et al., 2000, Nature 406:259-266, hereby 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, hereby 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 FcγRII and FcγRIII generally bind at 10'8 and 10'5 respectively. The extracellular domains of FcγRllla and FcγRlllb are 96% identical, however FcγRlllb does not have a 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, hereby 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, Stood 99:754-758, both hereby entirely incorporated by reference). Thus 80-90% of humans are poor responders, e.g., 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). C1q forms a complex with the serine proteases C1r and C1s to form the C1 complex. C1q 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 C1q, with IgGI and lgG3 typically binding substantially better to the FcγRs than lgG2 and lgG4 (Jefferis et al., 2002, Immunol Lett 82:57-65, hereby entirely incorporated by reference). There is currently no structure available for the Fc/C1q complex; however, mutagenesis studies have mapped the binding site on human IgG for C1q to a region involving residues D270, K322, K326, P329, and P331 , and E333 (Idusogie et al., 2000, J Immunol 164:4178-4184; Idusogie et al., 2001 , J Immunol 166:2571-2575, both hereby entirely incorporated by reference).
[8] A site on Fc between the Cγ2 and Cγ3 domains, shown in 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 hereby 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 for FcRn on Fc 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 (Martin et al., 2001 , MoI Cell 7:867-877, hereby 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 hereby entirely incorporated by reference) provide insight into the interaction of Fc with these proteins.
[9] A key 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. While not wanting to be limited to one theory, it is believed that the structural purpose of this carbohydrate may be to stabilize or solubilize Fc, determine a specific angle or level of flexibility between the Cγ3 and Cγ2 domains, keep the two Cγ2 domains from aggregating with one another across the central axis, or a combination of these. Efficient Fc binding to FcγR and C1q requires this modification, and alterations in the composition of the N297 carbohydrate or its elimination affect binding to these proteins (Umana et al., 1999, Nat Biotechnol 17:176-180; Davies et al., 2001 , Biotechnol Bioeng 74:288-294; Mimura et al., 2001 , J Biol Chem 276:45539-45547.; Radaev et al., 2001 , J Biol Chem 276:16478-16483; Shields et a/., 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, all hereby entirely incorporated by reference). Yet the carbohydrate makes little if any specific contact with FcγRs (Radaev et al., 2001 , J Biol Chem 276:16469-16477, hereby entirely incorporated by reference), indicating that the functional role of the N297 carbohydrate in mediating Fc/FcγR binding may be via the structural role it plays in determining the Fc conformation. This is supported by a collection of crystal structures of four different Fc glycoforms, which show that the composition of the oligosaccharide impacts the conformation of Cγ2 and as a result the Fc/FcγR interface (Krapp et al., 2003, J MoI Biol 325:979-989, hereby entirely incorporated by reference).
[10] The features of antibodies discussed above - specificity for target, ability to mediate immune effector mechanisms, and long half-life in serum - make antibodies powerful therapeutics. Monoclonal antibodies are used therapeutically for the treatment of a variety of conditions including cancer, inflammation, and cardiovascular disease. There are currently over ten antibody products on the market and hundreds in development. In addition to antibodies, an antibody-like protein that is finding an expanding role in research and therapy is the Fc fusion (Chamow et al., 1996, Trends Biotechnol 14:52-60; Ashkenazi et al., 1997, Curr Opin Immunol 9:195-200, both hereby entirely incorporated by reference). An Fc fusion is a protein wherein one or more polypeptides is operably linked to Fc. An Fc fusion combines the Fc region of an antibody, and thus its favorable effector functions and pharmacokinetics, with the target-binding region of a receptor, ligand, or some other protein or protein domain. The role of the latter is to mediate target recognition, and thus it is functionally analogous to the antibody variable region. Because of the structural and functional overlap of Fc fusions with antibodies, the discussion on antibodies in the present invention extends also to Fc fusions.
[11] Antibodies have found widespread application in oncology, particularly for targeting cellular antigens selectively expressed on tumor cells with the goal of cell destruction. There are a number of possible mechanisms by which antibodies destroy tumor cells, including anti-proliferation via blockage of needed growth pathways, intracellular signaling leading to apoptosis, enhanced down regulation and/or turnover of receptors, CDC, ADCC, ADCP, and promotion of an adaptive immune response (Cragg et al., 1999, Curr Opin Immunol 11 :541 -547; Glennie et al., 2000, Immunol Today 21 :403-410, both hereby entirely incorporated by reference). Anti-tumor efficacy may be due to a combination of these mechanisms, and their relative importance in clinical therapy appears to be cancer dependent. Despite this arsenal of anti-tumor weapons, the potency of antibodies as anti-cancer agents is unsatisfactory, particularly given their high cost. Patient tumor response data show that monoclonal antibodies provide only a small improvement in therapeutic success over normal single-agent cytotoxic chemotherapeutics. For example, just half of all relapsed low-grade non-Hodgkin's lymphoma patients respond to the anti-CD20 antibody rituximab (McLaughlin et al., 1998, J CHn Oncol 16:2825-2833, hereby entirely incorporated by reference). Of 166 clinical patients, 6% showed a complete response and 42% showed a partial response, with median response duration of approximately 12 months. Trastuzumab (Herceptin®, Genentech), an anti-HER2/neu antibody for treatment of metastatic breast cancer, has less efficacy. The overall response rate using trastuzumab for the 222 patients tested was only 15%, with 8 complete and 26 partial responses and a median response duration and survival of 9 to 13 months (Cobleigh et al., 1999, J Clin Oncol 17:2639-2648, hereby entirely incorporated by reference). Currently for anticancer therapy, any small improvement in mortality rate defines success. Thus there is a significant need to enhance the capacity of antibodies to destroy targeted cancer cells.
[12] A promising means for enhancing the anti-tumor potency of antibodies is via enhancement of their ability to mediate cytotoxic effector functions such as ADCC, ADCP, and CDC. The importance of FcγR-mediated effector functions for the anti-cancer activity of antibodies has been demonstrated in mice (Clynes et al., 1998, Proc Natl Acad Sci U S A 95:652-656; Clynes et al., 2000, Nat Med 6:443-446, both hereby entirely incorporated by reference), and the affinity of interaction between Fc and certain FcγRs correlates with targeted cytotoxicity in cell-based assays (Shields et al., 2001 , J Biol Chem 276:6591-6604; Presta et al., 2002, Biochem Soc Trans 30:487-490; Shields et al., 2002, J Biol Chem 277:26733-26740, all hereby entirely incorporated by reference). Additionally, a correlation has been observed between clinical efficacy in humans and their allotype of high (V158) or low (F158) affinity polymorphic forms of FcγRllla (Cartron et al., 2002, Blood 99:754-758, hereby entirely incorporated by reference). Together these data suggest that an antibody that is optimized for binding to certain FcγRs may better mediate effector functions and thereby destroy cancer cells more effectively in patients. The balance between activating and inhibiting receptors is an important consideration, and optimal effector function may result from an antibody that has enhanced affinity for activation receptors, for example FcγRI, FcγRlla/c, and FcγRllla, yet reduced affinity for the inhibitory receptor FcγRI Ib. Furthermore, because FcγRs can mediate antigen uptake and processing by antigen presenting cells, optimized FcγR affinity may also improve the capacity of antibody therapeutics to elicit an adaptive immune response (Dhodapkar & Dhodapkar, 2005, Proc Natl Acad Sci USA, 102, 6243-6244, hereby entirely incorporated by reference). The importance of complement-mediated effector function for anti-cancer therapy of antibodies is not as well characterized. Antibodies optimized for CDC would provide a way to investigate the role of complement in antibody clinical applications, and provide a potential mechanism for improving the tumor killing capacity of antibodies.
[13] In contrast antibody therapeutics and indications wherein effector functions contribute to clinical efficacy, for some antibodies and clinical applications it may be favorable to reduce or eliminate binding to one or more FcγRs, or reduce or eliminate one or more FcγR- or complement- mediated effector functions including but not limited to ADCC, ADCP, and/or CDC. This is often the case for therapeutic antibodies whose mechanism of action involves blocking or antagonism but not killing of the cells bearing target antigen. In these cases depletion of target cells is undesirable and can be considered a side effect. For example, the ability of anti-CD4 antibodies to block CD4 receptors on T cells makes them effective anti-inflammatories, yet their ability to recruit FcγR receptors also directs immune attack against the target cells, resulting in T cell depletion (Reddy et al., 2000, J Immunol 164:1925-1933, hereby entirely incorporated by reference). Effector function may also be a problem for radiolabeled antibodies, referred to as radioconjugates, and antibodies conjugated to toxins, referred to as immunotoxins. These drugs can be used to destroy cancer ceils, but the recruitment of immune cells via Fc interaction with FcγRs brings healthy immune cells in proximity to the deadly payload (radiation or toxin), resulting in depletion of normal lymphoid tissue along with targeted cancer cells (Hutchins et al., 1995, Proc Natl Acad ScI U S A 92:11980-11984; White et al., 2001 , Annu Rev Med 52:125-145, both entirely incorporated by reference). IgG isotypes that poorly recruit complement or effector cells, for example lgG2 and lgG4, can be used to address this problem in part. Fc variants that reduce or ablate Fc ligand binding are also known in the art (Alegre et al., 1994, Transplantation 57:1537-1543; Hutchins et al., 1995, Proc Natl Acad Sci USA 92:11980-11984; Armour et al., 1999, Eur J Immunol 29:2613-2624; Reddy et al., 2000, J Immunol 164:1925-1933; Xu et al., 2000, Cell Immunol 200:16-26; Shields et al., 2001 , J Biol Chem 276:6591- 6604; Armour et al., 1999, Eur J Immunol 29:2613-2624; US 6,194,551 ; US 5,885,573; PCT WO 99/58572; USSN 10/267,286, all hereby entirely incorporated by reference). However the complete Fc ligand-binding properties and effector function capacity of these variants, and their properties relative to the WT IgG isotypes, are unclear. What is needed is a general and robust means to completely ablate all FcγR binding and FcγR- and complement-mediated effector functions. A further consideration is that other important antibody properties not be perturbed. Fc variants should be engineered that not only ablate binding to FcγRs and/or C1q, but also maintain antibody stability, solubility, and structural integrity, as well as ability to interact with other important Fc ligands such as FcRn and proteins A and G.
[14] Recent success has been achieved at obtaining Fc variants with modulated binding to FcγRs and C1q, and in some cases these Fc variants have been test in for capacity to mediate FcγR- and complement-mediated effector functions (USSN 10/672,280, USSN 10/822,231 , USSN 11/124,620, filed May 5, 2005, and USSN 11/256,060, filed October 21 , 2005, all herein entirely incorporated by reference). Fc variants obtained in these studies provide a variety of optimal enhancements in Fc ligand and effector function properties, including but not limited to selectively improved binding to FcγRs, enhanced ADCC, improved binding to complement protein C1q, reduced binding to FcγRs, reduced binding to complement protein C1q, and other optimized properties. The present invention aims to further characterize the properties of select Fc variants from these studies, and to utilize the data to generate novel variants with optimized properties.
SUMMARY OF THE INVENTION
[15] The present invention provides Fc variants with optimized properties. Said optimized properties include altered binding to FcγR's, altered antibody dependent cell-mediated cytotoxicity, and altered complement dependent cytotoxicity relative to a parent Fc polypeptide.
[16] In one embodiment, the Fc variants of the present invention improve binding to one or more FcγR's relative to a parent Fc polypeptide. In an alternate embodiment, the Fc variants of the invention improve antibody dependent cell-mediated cytotoxicity relative to a parent Fc polypeptide. In a preferred embodiment, said Fc variants comprise an amino acid modification at one or more positions selected from the group consisting of: 227, 234, 235, 236, 239, 246, 255, 258, 260, 264, 267, 268, 272, 281 , 282, 283, 284, 293, 295, 304, 324, 327, 328, 330, 332, 335, wherein numbering is according to the EU index.
[17] In an alternate embodiment, the Fc variants of the present invention improve complement dependent cytotoxicity relative to a parent Fc polypeptide. In a preferred embodiment, said Fc variants comprise an amino acid modification at one or more positions selected from the group consisting of: 233, 234, 235, 239, 267, 268, 271 , 272, 274, 276, 278, 281 , 282, 284, 285, 293, 300, 320, 322, 324,
326, 327, 328, 330, 331 , 332, 333, 334, and 335, wherein numbering is according to the EU index.
[18] In an alternate embodiment, the Fc variants of the present invention reduce binding to one or more FcγRs relative to a parent Fc polypeptide. In an alternate embodiment, the Fc variants of the invention reduce antibody dependent cell-mediated cytotoxicity relative to a parent Fc polypeptide. In an alternate embodiment, the Fc variants of the invention reduce complement dependent cytotoxicity relative to a parent Fc polypeptide. In a preferred embodiment, the Fc variants of the invention reduce binding to one or more FcγRs, reduce antibody dependent cell-mediated cytotoxicity, and reduce complement dependent cytotoxicity relative to a parent Fc polypeptide. In a most preferred embodiment, said Fc variants comprise one or more amino acid modifications at a position selected from the group consisting of: 232, 234, 235, 236, 237, 238, 239, 265, 267, 269, 270, 297, 299, 325,
327, 328, 329, 330, and 331 , wherein numbering is according to the EU index.
[19] The present invention provides methods for engineering Fc variants with optimized properties. It is a further object of the present invention to provide experimental production and screening methods for obtaining optimized Fc variants.
[20] The present invention provides isolated nucleic acids encoding the Fc variants described herein.The present invention provides vectors comprising said nucleic acids, optionally, operably linked to control sequences. The present invention provides host cells containing the vectors, and methods for producing and optionally recovering the Fc variants.
[21] The present invention provides novel Fc polypeptides, including antibodies, Fc fusions, isolated Fc, and Fc fragments, that comprise the Fc variants disclosed herein. Said novel Fc polypeptides may find use in a therapeutic product. In a most preferred embodiment, the Fc polypeptides of the invention are antibodies.
[22] The present invention provides compositions comprising Fc polypeptides that comprise the Fc variants described herein, and a physiologically or pharmaceutically acceptable carrier or diluent.
[23] The present invention contemplates therapeutic and diagnostic uses for Fc polypeptides that comprise the Fc variants disclosed herein.
BRIEF DESCRIPTION OF THE DRAWINGS
[24] 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 1CE1 (James et al., 1999, J MoI Biol 289:293-301, hereby entirely incorporated by reference) and a human IgGI Fc structure from pdb accession code 1 DN2 (DeLano et al., 2000, Science 287:1279-1283, hereby 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, C1q, and proteins A and G in the Fc region.
[25] Figure 2. The Fc/FcγRlllb complex structure 11IS. Fc is shown as a gray ribbon diagram, and FcγRlllb is shown as a black ribbon. The N297 carbohydrate is shown as black sticks.
[26] Figures 3a - 3b. Alignment of the amino acid sequences of the human IgG immunoglobulins IgGI , lgG2, lgG3, and lgG4. Figure 3a provides the sequences of the CH1 (Cγ1) and hinge domains, and Figure 3b provides the sequences of the CH2 (Cγ2) and CH3 (Cγ3) domains. Positions are numbered according to the EU index of the IgGI sequence, and differences between IgGI and the other immunoglobulins lgG2, lgG3, and lgG4 are shown in grey. Polymorphisms exist at a number of positions (Kim et al., 2001 , J. MoI. Evol. 54:1-9, hereby entirely incorporated by reference), and thus slight differences between the presented sequences and sequences in the prior art may exist. The possible beginnings of the Fc region are labeled, defined herein as either EU position 226 or 230.
[27] Figure 4. Allotypes and isoallotypes of the gammal chain of human IgGI showing the positions and the relevant amino acid substitutions (Gorman & Clark, 1990, Semin Immunol 2(6):457- 66, hereby entirely incorporated by reference). For comparison the amino acids found in the equivalent positions in human lgG2, lgG3 and lgG4 gamma chains are also shown.
[28] Figure 5. Fc variants and FcγR binding data. All Fc variants were constructed in the context of the antibody PRO70769 IgGI . Fold indicates the fold IC50 relative to WT PRO70769 IgGI for binding to human V158 and F158 FcγRllla as measured by the competition AlphaScreen assay.
[29] Figure 6. Binding to human V158 FcγRllla (Figure 6a) and F158 FcγRllla (Figure 6b) by select PRO70769 Fc variants as determined by the competition AlphaScreen assay. In the presence of competitor antibody (Fc variant or WT) a characteristic inhibition curve is observed as a decrease in luminescence signal. The binding data were normalized to the maximum and minimum luminescence signal for each particular curve, provided by the baselines at low and high antibody concentrations respectively. The curves represent the fits of the data to a one site competition model using nonlinear regression.
[30] Figure 7. Binding to human V158 FcγRllla and F158 FcγRllla by PRO70769 Fc variants as measured by competition AlphaScreen assay. Figure 7a provides data for select variants, Figure 7b provides the IC50's and folds relative to WT PRO70769 IgGI .
[31] Figure 8. Fc variants and FcγR binding data. All Fc variants were constructed in the context of the variable region PRO70769 and either human IgGI or IgG(I /2) ELLGG. Figure 8a provides the IC50's and fold IC50's relative to WT PRO70769 IgGI for binding to human activating receptors V158 and F158 FcγRllla, and the inhibitory receptor FcγRllb, as measured by competition AlphaScreen assay. Figure 8b shows the AlphaScreen data for select variants.
[32] Figure 9. Competition Surface Plasmon Resonance (SPR) experiment measuring binding affinities of I332E and S239D/I332E variants in the context of trastuzumab to human V158 FcγRllla. Figure 9a provides the sensorgram raw data, Figure 9b provides a plot of the log of receptor concentration against the initial rate obtained at each concentration, and Figure 9c provides the affinities obtained from the fits to these data as described in Example 1.
[33] Figure 10. Cell-based ADCC assays of select Fc variants in the context of the anti-CD20 antibody PRO70769. ADCC was measured by the release of lactose dehydrogenase using a LDH Cytotoxicity Detection Kit (Roche Diagnostic). CD20+ lymphoma WIL2-S cells were used as target cells and human PBMCs were used as effector cells. Shown is the dose-dependence of ADCC on antibody concentration for the indicated antibodies, normalized to the minimum and maximum fluorescence signal for each particular curve, provided by the baselines at low and high antibody concentrations respectively. The curves represent the fits of the data to a sigmoidal dose-response model using nonlinear regression.
[34] Figure 11. Cell-based ADCC assay of select Fc variants in the context of PRO70769 IgGI in the absence and presence of serum levels of human IgG. ADCC was measured by the release of lactose dehydrogenase using a LDH Cytotoxicity Detection Kit (Roche Diagnostic). CD20+ lymphoma WIL2-S cells were used as target cells and human PBMβs were used as effector cells.
[35] Figure 12. Residues mutated in Fc variants designed to enhance CDC. The structure of the human IgGI Fc region is shown (pdb accession code 1 E4K, Sondermann et al., 2000, Nature 406:267-273, hereby entirely incorporated by reference). Black ball and sticks indicate residues D270, K322, P329, and P331 , which have been shown to be important in mediating binding to complement protein C1q, and grey sticks indicate residues that were mutated in the present invention to explore variants that affect CDC.
[36] Figure 13. Fc variants screened for complement-mediated cytotoxicity (CDC) and CDC data. The variable region is that of the anti-CD20 antibody PRO70769, and the heavy chain constant region is IgGI unless noted IgG(I /2) ELLGG. Fold CDC provides the relative CDC activity compared to WT PRO70769 lgG1.
[37] Figure 14. CDC assays of Fc variant anti-CD20 antibodies. The dose-dependence on antibody concentration of complement-mediated lysis is shown for the indicated PRO70769 antibodies against CD20+ WIL2-S lymphoma target cells. Lysis was measured using release of Alamar Blue, and data were normalized to the minimum and maximum fluorescence signal for each particular curve, provided by the baselines at low and high antibody concentrations respectively. The curves represent the fits of the data to a sigmoidal dose-response model with variable slope using nonlinear regression. [38] Figure 15. Amino acid modifications that provide enhanced and reduced CDC, and positions that may be modified that may provide enhanced/modulated CDC. Positions are numbered according to the EU index.
[39] Figure 16. Fc variants screened for reduced FcγR affinity, FcγR-mediated effector function, and complement-mediated effector function. The variable region is that of the anti-CD20 antibody PRO70769, and the heavy chain constant region is IgGI . The figure provides the Fold IC50 for binding to human V158 FcγRllla and the Fold EC50 of CDC activity relative to WT PRO70769 IgGI .
[40] Figure 17. Binding to human V158 FcγRllla by select PRO70769 Fc variants as determined by the competition AlphaScreen assay.
[41] Figure 18. CDC assays of select Fc variant anti-CD20 antibodies against CD20+ W1L2-S lymphoma target cells. Lysis was measured by Alamar Blue release.
[42] Figure 19. Cell-based ADCC activity of select anti-CD20 Fc variants against CD20+ lymphoma WIL2-S cells. Human PBMCs were used as effector cells, and lysis was measured by LDH release.
[43] Figure 20. Fc variants screened for reduced FcγR affinity, FcγR-mediated effector function, and complement-mediated effector function. The variable region is that of the anti-CD20 antibody PRO70769, and the heavy chain constant region is IgGL The figure provides the Fold IC50 relative to WT for binding to human V158 FcγRllla by two separate experiments, the Fold IC50 relative to WT for binding to human FcγRI, and the Fold EC50 relative to WT for CDC activity.
[44] Figure 21. Binding to the low affinity human activating receptor V158 FcγRllla and the high affinity human activating receptor FcγRI by select PRO70769 Fc variants as determined by the competition AlphaScreen assay.
[45] Figure 22. CDC activity of select PRO70769 Fc variants against CD20+ WIL2-S lymphoma target cells. Lysis was measured by release of Alamar Blue.
[46] Figure 23. Cell-based ADCC activity of anti-Her2 Fc variant and WT IgG antibodies against Her2/neu+ SkBr-3 breast carcinoma target cells. Human PBMCs were used as effector cells, and lysis was measured by LDH release.
[47] Figure 24. Amino acid sequences of variable light (VL) and heavy (VH) chains used in the present invention, including PRO70769 (Figures 24a and 24b), trastuzumab (Figures 24c and 24d), and ipilimumab (Figures 24e and 24f).
[48] Figure 25. Amino acid sequences of human constant light kappa (Figure 25a) and heavy (Figures 25b - 25f) chains used in the present invention.
[49] Figure 26. Sequences showing possible constant heavy chain sequences with reduced Fc ligand binding and effector function properties (Figure 26a), and sequences of improved anti-CTLA-4 antibodies (Figures 26b - 26d). Figure 26a shows potential Fc variant constant heavy chain sequences, with variable positions designated in bold as X1 , X2, X3, X4, X5, X6, X7, and X8. The table below the sequence provides the WT amino acid and possible substitutions for these positions. Improved antibody sequences may comprise one or more non-WT amino acid selected from this group of possible modifications. Figure 26b provides the light chain sequence of an anti-CTLA-4 antibody, and Figures 26c and 26d provide heavy chain sequences of anti-CTLA-4 antibodies with reduced Fc ligand binding and Fc-mediated effector function. These include an L235G/G236R IgGI heavy chain (Figure 26c) and an lgG2 heavy chain (Figure 26d). The positions are numbered according to the EU index as in Kabat, and thus do not correspond to the sequential order in the sequence.
DETAILED DESCRIPTION OF THE INVENTION
[50] In order that the invention may be more completely understood, several definitions are set forth below. Such definitions are meant to encompass grammatical equivalents.
[51] By "ADCC" or "antibody dependent cell-mediated cytotoxicity" as used herein is meant the cell-mediated reaction wherein nonspecific cytotoxic cells that express FcDRs recognize bound antibody on a target cell and subsequently cause lysis of the target cell.
[52] By "ADCP" or antibody dependent cell-mediated phagocytosis as used herein is meant the cell-mediated reaction wherein nonspecific cytotoxic cells that express FcDRs recognize bound antibody on a target cell and subsequently cause phagocytosis of the target cell.
[53] By "amino acid modification" herein is meant an amino acid substitution, insertion, and/or deletion in a polypeptide sequence. 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 L328R refers to a variant polypeptide, in this case an Fc variant, in which the leucine at position 328 is replaced with arginine. By "amino acid insertion" or "insertion" as used herein is meant the addition of an amino acid at a particular position in a parent polypeptide sequence. For example, insert G > 235-236 designates an insertion of glycine between positions 235 and 236. By "amino acid deletion" or "deletion" as used herein is meant the removal of an amino acid at a particular position in a parent polypeptide sequence. For example, G236- designates the deletion of glycine at position 236. Amino acids of the invention may be further classified as either isotypic or novel.
[54] By "CDC" or "complement dependent cytotoxicity" as used herein is meant the reaction wherein one or more complement protein components recognize bound antibody on a target cell and subsequently cause lysis of the target cell.
[55] By "isotypic modification" as used herein is meant an amino acid modification that converts one amino acid of one isotype to the corresponding amino amino acid in a different, aligned isotype. For example, because IgGI has a tyrosine and lgG2 a phenylalanine at EU position 296, a F296Y substitution in lgG2 is considered an isotypic modification.
[56] By "novel modification" as used herein is meant an amino acid modification that is not isotypic. For example, because none of the IgGs has a glutamic acid at position 332, the substitution I332E in IgGI , lgG2, lgG3, or lgG4 is considered a novel modification. [57] 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.
[58] 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 FcγR- mediated effector functions such as ADCC and ADCP, and complement-mediated effector functions such as CDC.
[59] 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 DD T cells, and may be from any organism including but not limited to humans, mice, rats, rabbits, and monkeys.
[60] By "Fab" or "Fab region" as used herein is meant the polypeptides that comprise the VH, CH1 , VH, and C|_ immunoglobulin domains. Fab may refer to this region in isolation, or this region in the context of a full length antibody or antibody fragment.
[61] 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. Thus Fc refers to the last two constant region immunoglobulin domains of IgA, IgD, and IgG, and 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 (Cγ2 and Cγ3) and the hinge between Cgammai (Cγ1 ) and Cgamma2 (Cγ2). Although the boundaries of the Fc region may vary, the human IgG heavy chain Fc region is usually defined to comprise 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.
[62] By "Fc fusion" as used herein is meant a protein wherein one or more polypeptides is operably linked to Fc. 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 a/., 1997, Curr Opin Immunol 9:195- 200, both hereby entirely incorporated by reference). An Fc fusion combines the Fc region of an immunoglobulin with a fusion partner, which in general may be any protein, polypeptide or small molecule. The role of the non-Fc part of an Fc fusion, i.e., the fusion partner, is to mediate target binding, and thus it is functionally analogous to the variable regions of an antibody. 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 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, preferrably an extracellular receptor that is implicated in disease.
[63] By "Fc gamma receptor" or "FcDR" as used herein is meant any member of the family of proteins that bind the IgG antibody Fc region and are substantially encoded by the FcDR genes. 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, hereby entirely incorporated by reference), as well as any undiscovered human FcDRs or FcγR isoforms or allotypes. An FcγR may be from any organism, including but not limited to humans, mice, rats, rabbits, and monkeys. Mouse FcDRs include but are not limited to FcDRI (CD64), FcDRII (CD32), FcDRIII (CD16), and FcDRIII-2 (CD16- 2), as well as any undiscovered mouse FcDRs or FcγR isoforms or allotypes.
[64] 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. Fc ligands include but are not limited to FcγRs, FcγRs, FcγRs, FcRn, C1 q, 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 FcDRs (Davis et al., 2002, Immunological Reviews 190:123-136, hereby entirely incorporated by reference). Fc ligands may include undiscovered molecules that bind Fc.
[65] By "full length antibodv"as used herein is meant the structure that constitutes the natural biological form of an antibody, including variable and constant regions. For example, in most mammals, including humans and mice, the full length antibody of the IgG isotype is a tetramer and consists of two identical pairs of two immunoglobulin chains, each pair having one light and one heavy chain, each light chain comprising immunoglobulin domains VL and CL, and each heavy chain comprising immunoglobulin domains VH, CD1 , CD2, and CD3. In some mammals, for example in camels and llamas, IgG antibodies may consist of only two heavy chains, each heavy chain comprising a variable domain attached to the Fc region.
[66] By "IgG." as used herein is meant a polypeptide belonging to the class of antibodies that are substantially encoded by a recognized immunoglobulin gamma gene. In humans this IgG comprises the subclasses or isotypes IgGI , lgG2, lgG3, and lgG4. In mice IgG comprises IgGI , lgG2a, lgG2b, lgG3.
[67] By "immunoglobulin (Ig)" herein is meant a protein consisting of one or more polypeptides substantially encoded by immunoglobulin genes. Immunoglobulins include but are not limited to antibodies. Immunoglobulins may have a number of structural forms, including but not limited to full length antibodies, antibody fragments, and individual immunoglobulin domains.
[68] By "immunoglobulin (Ig) domain"as used herein is meant a region of an immunoglobulin that exists as a distinct structural entity as ascertained by one skilled in the art of protein structure. Ig domains typically have a characteristic G-sandwich folding topology. The known Ig domains in the IgG isotype of antibodies are VH, CD1 , CD2, CD3, VL, and CL.
[69] By "IgG' or "IgG immunoglobulin" as used herein is meant a polypeptide belonging to the class of antibodies that are substantially encoded by a recognized immunoglobulin gamma gene. In humans this class comprises the subclasses or isotypes IgGI , lgG2, lgG3, and lgG4. By "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, IgM, IgD, and IgE.
[70] By "parent polypeptide", "parent protein", "precursor polypeptide", or "precursor protein" as used herein is meant an unmodified polypeptide that is subsequently modified to generate a variant. Said 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 Fc polypeptide" as used herein is meant an Fc polypeptide that is modified to generate a variant, and by "parent antibody" as used herein is meant an antibody that is modified to generate a variant antibody.
[71] 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 IgGI .
[72] By "polypeptide" or "protein" as used herein is meant at least two covalently attached amino acids, which includes proteins, polypeptides, oligopeptides and peptides.
[73] 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 .
[74] 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.
[75] By "target cell" as used herein is meant a cell that expresses a target antigen.
[76] 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 VD, VD, and/or VH genes that make up the kappa, lambda, and heavy chain immunoglobulin genetic loci respectively.
[77] By "variant polypeptide", "polypeptide variant", or "variant" as used herein is meant a polypeptide sequence that differs from that of a parent polypeptide sequence by virtue of at least one amino acid modification. The parent polypeptide may be a naturally occurring or wild-type (WT) polypeptide, or may be a modified version of a WT polypeptide. Variant polypeptide may refer to the polypeptide itself, a composition comprising the polypeptide, or the amino sequence that encodes it. Preferably, the variant polypeptide has at least one amino acid modification compared to the parent polypeptide, 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 variant polypeptide sequence herein will preferably possess at least about 80% homology with a parent polypeptide sequence, and most preferably at least about 90% homology, more preferably at least about 95% homology. Accordingly, by "Fc variant" or "variant Fc" as used herein is meant an Fc sequence that differs from that of a parent Fc sequence by virtue of at least one amino acid modification. An Fc variant may only encompass an Fc region, or may exist in the context of an antibody, Fc fusion, isolated Fc, Fc fragment, or other polypeptide that is substantially encoded by Fc. Fc variant may refer to the Fc polypeptide itself, compositions comprising the Fc variant polypeptide, or the amino acid sequence that encodes it. By "Fc polypeptide variant" or "variant Fc polypeptide" as used herein is meant an Fc polypeptide that differs from a parent Fc polyeptide by virtue of at least one amino acid modification. By "protein variant" or "variant protein" as used herein is meant a protein that differs from a parent protein by virtue of at least one amino acid modification. 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. By "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. By "immunoglobulin variant" or "variant immunoglobluin" 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.
[78] 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, polypeptide, antibody, immunoglobulin, IgG, etc. has an amino acid sequence or a nucleotide sequence that has not been intentionally modified.
Antibodies
[79]Accordingly, the present invention provides variant antibodies.
[80]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.
[81]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. [82]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, EA Kabat et al.).
[83] 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: "CH1" 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.
[84] 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.
[85]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, and 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.
[86]ln 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. [87]Altematively, 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
[88] 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.
[89]Specific antibody fragments include, but are not limited to, (i) the Fab fragment consisting of VL, VH, CL and CH1 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) 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), (viii) bispecific single chain Fv dimers (PCT/US92/09965) 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). 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).
Chimeric and Humanized Antibodies
[9O]In some embodiments, the scaffold components can be a mixture from different species. As such, if the antibody 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 92/11018, Jones, 1986, Nature 321 :522-525, Verhoeyen et al., 1988, Science 239:1534-1536. "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). 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. 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). 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 11 :321-8. 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. 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): 22611-22618; Rader et al., 1998, Proc. Natl. Acad. Sci. USA 95: 8910-8915; Krauss et al., 2003, Protein Engineering 16(10):753-759. Other humanization methods may involve the grafting of only parts of the CDRs, including but not limited to methods described in USSN 09/810,502; Tan et al., 2002, J. Immunol. 169:1119-1125; De Pascalis et al., 2002, J. Immunol. 169:3076-3084.
Bispecific Antibodies
[91] 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), e.g., prepared chemically or from hybrid hybridomas.
Minibodies
[92]ln 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. In some cases, the scFv can be joined to the Fc region, and may include some or all of the hinge region.
Human Antibodies
[93]ln 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.
Antibody Fusions
[94]ln one embodiment, the antibodies of the invention are antibody fusion proteins (sometimes referred to herein as an "antibody conjugate"). One type of antibody fusions are 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). 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, that is implicated in disease.
[95] 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, that is implicated in disease.
[96]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).
[97]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 , hereby incorporated by reference in its entirety.
Covalent modifications of Antibodies
[98]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.
[99]Cysteinyl residues most commonly are reacted with σ-haloacetates (and corresponding amines), such as chloroacetic acid or chloroacetamide, to give carboxymethyl or carboxyamidomethyl derivatives. Cysteinyl residues also are derivatized by reaction with bromotrifluoroacetone, σ-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.
[100] 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.
[101] 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.
[102] 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.
[103] 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.
[104] Carboxyl side groups (aspartyl or glutamyl) are selectively modified by reaction with carbodiimides (R' — N=C=N-R1), where R and R' are optionally different alkyl groups, such as 1- cyclohexyl-3-(2-morpholinyl-4-ethyI) 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.
[105] 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 are employed for protein immobilization.
[106] 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.
[107] Other modifications include hydroxylation of proline and lysine, phosphorylation of hydroxyl groups of seryl or threonyl residues, methylation of the σ-annino 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]), acetylation of the N-terminal amine, and amidation of any C- terminal carboxyl group.
Glycosylation
[108] 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 (Umana 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); (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 [σ1 ,6-fucosyltranserase] and/or β\ -A- 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.
[109] 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-hydroxyIysine may also be used.
[110] 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.
[111] 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 published Sep. 11 , 1987, and in Aplin and Wriston, 1981 , CRC Crit. Rev. Biochem., pp. 259-306.
[112] 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 degiycosylation is described by Hakimuddin et al., 1987, Arch. Biochem. Biophys. 259:52 and by Edge et al., 1981 , Anal. Biochem. 118:131. 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. 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. Tunicamycin blocks the formation of protein-N-glycoside linkages.
[113] 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 U.S. Pat. Nos. 4,640,835; 4,496,689; 4,301 ,144; 4,670,417; 4,791 ,192 or 4,179,337. 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. 2005/0114037, incorporated herein by reference in its entirety.
Labeled Antibodies
[114] 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.
[115] 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.
[116] 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, /?-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.
[117] 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.
[118] 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), FlTC, 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, hereby expressly incorporated by reference.
[119] 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 are expressly incorporated herein by reference.
[120] In certain variations, antibody can meant a protein consisting of one or more polypeptides substantially encoded by all or part of the recognized immunoglobulin genes. The recognized immunoglobulin genes, for example in humans, include the kappa (D), lambda (D), and heavy chain genetic loci, which together comprise the myriad variable region genes, and the constant region genes mu (D), delta (D), gamma (D), sigma (D), and alpha (D) which encode the IgM, IgD, IgG (IgGI , lgG2, lgG3, and lgG4), IgE, and IgA (IgAI and lgA2) isotypes respectively. Antibody herein is meant to include full length antibodies and antibody fragments, and may refer to a natural antibody from any organism, an engineered antibody, or an antibody generated recombinantly for experimental, therapeutic, or other purposes.
[121] An Fc variant comprises one or more amino acid modifications relative to a parent Fc polypeptide, wherein said amino acid modification(s) provide one or more optimized properties. An Fc variant of the present invention differs in amino acid sequence from its parent IgG by virtue of at least one amino acid modification. Thus Fc variants of the present invention have at least one amino acid modification compared to the parent. Alternatively, the Fc variants of the present invention may have more than one amino acid modification as compared to the parent, for example from about one to fifty amino acid modifications, preferrably from about one to ten amino acid modifications, and most preferably from about one to about five amino acid modifications compared to the parent. Thus the sequences of the Fc variants and those of the parent Fc polypeptide are substantially homologous. For example, the variant Fc variant sequences herein will possess about 80% homology with the parent Fc 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.
[122] 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/A330L/I332E defines an Fc variant with the substitutions S239D, A330L, and I332E relative to the parent Fc polypeptide. It is noted that the order in which substitutions are provided is arbitrary, that is to say that, for example, S239D/A330L/I332E is the same Fc variant as S239D/I332E/A330L, 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).
[123] Fc variants of the present invention may be substantially encoded by genes from any organism, preferably mammals, including but not limited to humans, rodents including but not limited to mice and rats, lagomorpha including but not limited to rabbits and hares, camelidae including but not limited to camels, llamas, and dromedaries, and non-human primates, including but not limited to Prosimians, Platyrrhini (New World monkeys), Cercopithecoidea (Old World monkeys), and Hominoidea including the Gibbons and Lesser and Great Apes. In a most preferred embodiment, the Fc variants of the present invention are substantially human.
[124] The parent Fc polypeptide may be an antibody. Parent antibodies may be fully human, obtained for example using transgenic mice (Bruggemann et al., 1997, Curr Opin Biotechnol 8:455- 458, hereby entirely incorporated by reference) or human antibody libraries coupled with selection methods (Griffiths et al., 1998, Curr Opin Biotechnol 9:102-108, hereby entirely incorporated by reference). The parent antibody need not be naturally occurring. For example, the parent antibody may be an engineered antibody, including but not limited to chimeric antibodies and humanized antibodies (Clark, 2000, Immunol Today 21 :397-402, hereby entirely incorporated by reference). The parent antibody may be an engineered variant of an antibody that is substantially encoded by one or more natural antibody genes. In one embodiment, the parent antibody has been affinity matured, as is known in the art. Alternatively, the antibody has been modified in some other way, for example as described in USSN 10/339788, filed on March 3, 2003, hereby entirely incorporated by reference.
[125] The Fc variants of the present invention may be substantially encoded by immunoglobulin genes belonging to any of the antibody classes. In a preferred embodiment, the Fc variants of the present invention find use in antibodies or Fc fusions that comprise sequences belonging to the IgG class of antibodies, including IgGI, lgG2, lgG3, or lgG4. Figure 3 provides an alignment of these human IgG sequences. In an alternate embodiment the Fc variants of the present invention find use in antibodies or Fc fusions that comprise sequences belonging to the IgA (including subclasses IgAI and lgA2), IgD, IgE, IgG, or IgM classes of antibodies. The Fc variants of the present invention may comprise more than one protein chain. That is, the present invention may find use in an antibody or Fc fusion that is a monomer or an oligomer, including a homo- or hetero-oligomer.
[126] As is well known in the art, immunoglobulin polymorphisms exist in the human population. Gm polymorphism is determined by the IGHG1 , IGHG2 and IGHG3 genes which have alleles encoding allotypic antigenic determinants referred to as G1m, G2m, and G3m allotypes for markers of the human IgGI , lgG2 and lgG3 molecules (no Gm allotypes have been found on the gamma 4 chain). Markers may be classified into 'allotypes' and 'isoallotypes'. These are distinguished on different serological bases dependent upon the strong sequence homologies between isotypes. Allotypes are antigenic determinants specified by allelic forms of the Ig genes. Allotypes represent slight differences in the amino acid sequences of heavy or light chains of different individuals. Even a single amino acid difference can give rise to an allotypic determinant, although in many cases there are several amino acid substitutions that have occurred. Allotypes are sequence differences between alleles of a subclass whereby the antisera recognize only the allelic differences. An isoallotype is an allele in one isotype which produces an epitope which is shared with a non-polymorphic homologous region of one or more other isotypes and because of this the antisera will react with both the relevant allotypes and the relevant homologous isotypes (Clark, 1997, IgG effector mechanisms, Chem Immunol. 65:88-110; Gorman & Clark, 1990, Semin Immunol 2(6):457-66, both hereby entirely incorporated by reference).
[127] Allelic forms of human immunoglobulins have been well-characterized (WHO Review of the notation for the allotypic and related markers of human immunoglobulins. J lmmunogen 1976, 3: 357- 362; WHO Review of the notation for the allotypic and related markers of human immunoglobulins. 1976, Eur. J. Immunol. 6, 599-601 ; Loghem E van, 1986, Allotypic markers, Monogr Allergy 19: 40-51 , all hereby entirely incorporated by reference). Additionally, other polymorphisms have been characterized (Kim et al., 2001 , J. MoI. Evol. 54:1-9, hereby entirely incorporated by reference). At present, 18 Gm allotypes are known: G1m (1 , 2, 3, 17) or G1m (a, x, f, z), G2m (23) or G2m (n), G3m (5, 6, 10, 11 , 13, 14, 15, 16, 21 , 24, 26, 27, 28) or G3m (b1 , c3, b5, bθ, b3, b4, s, t, g1 , c5, u, v, g5) (Lefranc, et al., The human IgG subclasses: molecular analysis of structure, function and regulation. Pergamon, Oxford, pp. 43-78 (1990); Lefranc, G. et al., 1979, Hum. Genet.: 50, 199-211 , both hereby entirely incorporated by reference). Allotypes that are inherited in fixed combinations are called Gm haplotypes.
[128] Figure 4 shows the allotypes and isoaliotypes of the gammal chain of human IgGI showing the positions and the relevant amino acid substitutions (Gorman & Clark, 1990, Semin Immunol 2(6):457-66, hereby entirely incorporated by reference). For comparison the amino acids found in the equivalent positions in human lgG2, lgG3 and lgG4 gamma chains are also shown.
[129] The Fc variants of the present invention may be substantially encoded by any allotype or isoallotype of any immunoglobulin gene. In a preferred embodiment, the Fc variants of the present invention find use in antibodies or Fc fusions that comprise IgGI sequences that are classified as G1m(1), G1m(2), G1m(3), G1m(17), nG1m(1), nG1 m(2), and/or nG1 m(17). Thus in the context of an IgGI isotype, the Fc variants of the present invention may comprise a Lys (G1m(17)) or Arg (G1m(3)) at position 214, an Asp356/Leu358 (G1m(1)) or Glu356/Met358 (nG1m(1)), and/or a GIy (G1m(2)) or Ala (nG1m(2)) at position 431.
[130] In the most preferred embodiment, the Fc variants of the invention are 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. Fc variants may also comprise sequences from other immunoglobulin classes such as IgA, IgE, IgGD, IgGM, and the like. It is contemplated that, although the Fc variants of the present invention 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 first and second 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 Fc 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 about 0.13 nm and preferably about 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 Fc variants discovered by the present invention may be engineered into any second parent IgG that has significant sequence or structural homology with the Fc 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 Fc variant does not affect the ability to transfer the Fc variants of the present invention to other parent IgGs.
[131] Virtually any antigen may be targeted by the Fc variants of the present invention, including but not limited to proteins, subunits, domains, motifs, and/or epitopes belonging to the following list of targets: 17-IA, 4-1 BB, 4Dc, 6~keto-PGF1a, 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 VgM , 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 XIZIP, CBL, CCI, CCK2, CCL, CCL1 , CCL11 , 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, CDHa1 CDHb, CD11c, 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)-lGF-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 Ha, 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 IHB 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-Il, IL, IL-1 , IL-I R, IL-2, IL-2R, IL-4, IL-4R, IL-5, IL-5R, IL-6, IL-6R, IL-8, lL-9, IL-10, 1L-12, IL- 13, 1L-15, IL-18, IL-18R, IL-23, interferon (lNF)-alpha, INF-beta, INF-gamma, lnhibin, 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 alphaδ, integrin betai , integrin beta2, interferon gamma, IP-10, 1-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 TGM1 Latent TGM 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-11 , 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, NT1 NTN, OB, 0GG1 , 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, PS1 PSA1 PSCA1 prostate specific membrane antigen (PSMA), PTEN1 PTHrp, Ptk, PTN, R51 , RANK, RANKL, RANTES, RANTES1 Relaxin A-chain, Relaxin B-chain, renin, respiratory syncytial virus (RSV) F, RSV Fgp, Ret, Rheumatoid factors, RLIP76, RP A2, RSK, S100, SCF/KL, SDF-1 , SERINE, Serum albumin, sFRP-3, Shh, SIGIRR, SK-1 , SLAM, SLPI, SMAC1 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, TERT1 testicular PLAP-like alkaline phosphatase, TfR, TGF, TGF-alpha, TGF-beta, TGF-beta Pan Specific, TGF-beta Rl (ALK-5), TGF-beta RII1 TGF-beta RIIb1 TGF-beta RIII1 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-RI1 TNF-RII1 TNFRSF10A (TRAIL R1 Apo-2, DR4), TNFRSF10B (TRAIL R2 DR5, KILLER, TRICK-2A, TRICK-B), TNFRSF10C (TRAIL R3 DcR1 , LIT, TRID)1 TNFRSF10D (TRAIL R4 DcR2, TRUNDD), TNFRSF11 A (RANK ODF R1 TRANCE R), TNFRSF11B (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)1 TNFRSF19 (TROY TAJ, TRADE), TNFRSF19L (RELT)1 TNFRSF1A (TNF Rl CD120a, p55-60), TNFRSF1B (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 (4-1BB CD137, ILA), TNFRSF21 (DR6), TNFRSF22 (DcTRAIL R2 TNFRH2), TNFRST23 (DcTRAIL R1 TNFRH1), TNFRSF25 (DR3 Apo-3, LARD1 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 LTa1 TNFSF1), TNFSF3 (LTb TNFC1 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 (4-1BB 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, etc.
[132] The present invention provides Fc variants that are optimized for a variety of therapeutically relevant properties. An Fc variant that is engineered or predicted to display one or more optimized properties is herein referred to as an "optimized Fc variant". Properties that may be optimized include but are not limited to enhanced or reduced affinity for an FcγR. In a preferred embodiment, the Fc variants of the present invention are optimized to possess enhanced affinity for a human activating FcγR, preferably FcγRI, FcγRlla, FcγRllc, FcγRllla, and FcγRlllb, most preferably FcγRllla. In an alternately preferred embodiment, the Fc variants are optimized to possess reduced affinity for the human inhibitory receptor FcγRllb. These preferred embodiments are anticipated to provide IgG polypeptides with enhanced therapeutic properties in humans, for example enhanced effector function and greater anti-cancer potency. In an alternate embodiment, the Fc variants of the present invention are optimized to have reduced or ablated affinity for a human FcγR, including but not limited to FcγRI, FcγRlla, FcγRllb, FcγRllc, FcγRllla, and FcγRlllb. These embodiments are anticipated to provide IgG polypeptides with enhanced therapeutic properties in humans, for example reduced effector function and reduced toxicity. In other embodiments, Fc variants of the present invention provide enhanced affinity for one or more FcγRs, yet reduced affinity for one or more other FcγRs. For example, an Fc variant of the present invention may have enhanced binding to FcγRllla, yet reduced binding to FcγRllb. Alternately, an Fc variant of the present invention may have enhanced binding to FcγRlla and FcγRI, yet reduced binding to FcγRllb. In yet another embodiment, an Fc variant of the present invention may have enhanced affinity for FcγRllb, yet reduced affinity to one or more activating FcγRs.
[133] Preferred embodiments comprise optimization of binding to a human FcγR, however in alternate embodiments the Fc variants of the present invention possess enhanced or reduced affinity for FcγRs from nonhuman organisms, including but not limited to rodents and non-human primates. Fc variants that are optimized for binding to a nonhuman FcγR 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 Fc variants that comprise Fc variants that are optimized for one or more mouse FcγRs, may provide valuable information with regard to the efficacy of the protein, its mechanism of action, and the like. The Fc variants of the present invention may also be optimized for enhanced functionality and/or solution properties in aglycosylated form. In a preferred embodiment, the aglycosylated Fc variants of the present invention bind an Fc ligand with greater affinity than the aglycosylated form of the parent Fc variant. Said Fc ϋgands include but are not limited to FcγRs, C1q, FcRn, 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 Fc variants are optimized to be more stable and/or more soluble than the aglycosylated form of the parent Fc variant.
[134] Fc variants of the invention may comprise modifications that modulate interaction with Fc ligands other than FcγRs, including but not limited to complement proteins, FcRn, 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, hereby entirely incorporated by reference).
[135] Preferably, the Fc ligand specificity of the Fc variant of the present invention will determine its therapeutic utility. The utility of a given Fc variant for therapeutic purposes will depend on the epitope or form of the Target antigen and the disease or indication being treated. For some targets and indications, enhanced FcγR-mediated effector functions may be preferable. This may be particularly favorable for anti-cancer Fc variants. Thus Fc variants may be used that comprise Fc variants that provide enhanced affinity for activating FcγRs and/or reduced affinity for inhibitory FcγRs. For some targets and indications, it may be further beneficial to utilize Fc 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 FcγRl, whereas in other cases, enhanced binding only to FcγRlla may be preferred. For certain targets and indications, it may be preferable to utilize Fc variants that enhance both FcγR-mediated and complement-mediated effector functions, whereas for other cases it may be advantageous to utilize Fc variants that enhance 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 C1q, one or more FcγR's, FcRn, or one or more other Fc ligands. For other targets and indications, it may be preferable to utilize Fc 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 Fc variant is to inhibit inflammation or auto-immune disease, or modulate the immune system in some way.
[136] Clearly an important parameter that determines the most beneficial selectivity of a given Fc variant to treat a given disease is the context of the Fc variant, e.g., what type of Fc variant is being used. Thus the Fc ligand selectivity or specifity of a given Fc variant will provide different properties depending on whether it composes an antibody, Fc fusion, or Fc variants with a coupled fusion or conjugate partner. For example, toxin, radionucleotide, or other conjugates may be less toxic to normal cells if the Fc variant that comprises them has reduced or ablated binding to one or more Fc ligands. As another example, in order to inhibit inflammation or auto-immune disease, it may be preferable to utilize an Fc variant with enhanced affinity for activating FcγRs, such as to bind these FcγRs and prevent their activation. Conversely, an Fc variant that comprises two or more Fc regions with enhanced FcγRllb affinity may co-engage this receptor on the surface of immune cells, thereby inhibiting proliferation of these cells. Whereas in some cases an Fc variants may engage its target antigen on one cell type yet engage FcγRs on separate cells from the target antigen, in other cases it may be advantageous to engage FcγRs on the surface of the same cells as the target antigen. For example, if an antibody targets an antigen on a cell that also expresses one or more FcγRs, it may be beneficial to utilize an Fc variant that enhances or reduces binding to the FcγRs on the surface of that cell. This may be the case, for example when the Fc variant is being used as an anti-cancer agent, and co-engagement of target antigen and FcγR on the surface of the same cell promote signaling events within the cell that result in growth inhibition, apoptosis, or other anti-proliferative effect. Alternatively, antigen and FcγR co-engagement on the same cell may be advantageous when the Fc variant is being used to modulate the immune system in some way, wherein co-engagement of target antigen and FcγR provides some proliferative or anti-proliferative effect. Likewise, Fc variants that comprise two or more Fc regions may benefit from Fc variants that modulate FcγR selectivity or specifity to co-engage FcγRs on the surface of the same cell.
[137] The presence of different polymorphic forms of FcγRs provides yet another parameter that impacts the therapeutic utility of the Fc variants of the present invention. Whereas the specificity and selectivity of a given Fc variant for the different classes of Fc^Rs signficantly affects the capacity of an Fc variant to target a given antigen for treatment of a given disease, the specificity or selectivity of an Fc variant for different polymorphic forms of these receptors may in part determine which research or pre-clinical experiments may be appropriate for testing, and ultimately which patient populations may or may not respond to treatment. Thus the specificity or selecitivty of Fc variants of the present invention to Fc ligand polymorphisms, including but not limited to FcγR, C1q, FcRn, and FcRH polymorphisms, may be used to guide the selection of valid research and pre-clinical experiments, clinical trial design, patient selection, dosing dependence, and/or other aspects concerning clinical trials.
[138] Modification may be made to improve the IgG stability, solubility, function, or clinical use. In a preferred embodiment, the Fc variants of the present invention may comprise modifications to reduce immunogenicity in humans. In a most preferred embodiment, the immunogenicity of an Fc variant of the present invention is reduced using a method described in USSN 11/004,590, filed December 3, 2004, hereby entirely incorporated by reference. In alternate embodiments, the Fc variants of the present invention are humanized (Clark, 2000, Immunol Today 21 :397-402, hereby entirely incorporated by reference). By "humanized" antibody as used herein is meant an antibody comprising a human framework region (FR) and one or more complementarity determining regions (CDR's) from a non-human (usually mouse or rat) antibody. The non-human antibody providing the CDR's is called the "donor" and the human immunoglobulin providing the framework is called the "acceptor". Humanization relies principally on the grafting of donor CDRs onto acceptor (human) VL and VH frameworks (e.g., Winter et al, US 5225539, hereby entirely incorporated by reference). This strategy is referred to as "CDR grafting". "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; and US 6407213, all hereby 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. 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 hereby 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 11 :321-8, all hereby 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, hereby entirely incorporated by reference. In one embodiment, the parent antibody has been affinity matured, as is well known in the art. Structure-based methods may be employed for humanization and affinity maturation, for example as described in USSN 11/004,590, hereby entirely incorporated by reference. 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): 22611-22618; Rader et al., 1998, Proc. Natl. Acad. Sci. USA 95: 8910-8915; Krauss et al., 2003, Protein Engineering 16(10):753-759, all hereby 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 09/810,502; Tan et al., 2002, J. Immunol. 169:1119-1125; De Pascalis et al., 2002, J. Immunol. 169:3076-3084, all hereby entirely incorporated by reference.
[139] Modifications to reduce immunogenicity may include modifications that reduce binding of processed peptides derived from the parent sequence to MHC proteins. For example, amino acid modifications may 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 Fc variant of the present invention. 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; Stumiolo 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 hereby 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 hereby entirely incorporated by reference).
[140] In one embodiment, the Fc variants of the present invention comprise one or more engineered glycoforms. By "engineered qlvcoform" 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 (Umana 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 ); (Potelligent™ technology [Biowa, Inc., Princeton, NJ]; GlycoMAb® glycosylation engineering technology [GLYCART biotechnology AG, Zurich, Switzerland], all hereby entirely incorporated by reference). 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 £1-4- 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 Fc variant, for example an antibody or Fc fusion, may comprise an engineered glycoform. Alternatively, engineered glycoform may refer to the Fc variant that comprises the different carbohydrate or oligosaccharide.
[141] In an alternate embodiment, the Fc variant of the present invention 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.
[142] The present invention provides methods for engineering, producing, and screening Fc variants. The described methods are not meant to constrain the present invention to any particular application or theory of operation. Rather, the provided methods are meant to illustrate generally that one or more Fc variants may be engineered, produced, and screened experimentally to obtain Fc variants with optimized effector function. A variety of methods are described for designing, producing, and testing antibody and protein variants in USSN 10/672,280, USSN 10/822,231 , USSN 11/124,620, and USSN 11/256,060, all hereby entirely incorporated by reference.
[143] A variety of protein engineering methods may be used to design Fc 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. An alignment of sequences may be used to guide substitutions at the identified positions. Alternatively, random or semi-random mutagenesis methods may be used to make amino acid modifications at the desired positions.
[144] Methods for production and screening of Fc 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, all hereby entirely incorporated by reference. Also see the methods described in USSN 10/672,280, USSN 10/822,231 , USSN 11/124,620, and USSN 11/256,060, all hereby entirely incorporated by reference.
[145] In one embodiment of the present invention, the Fc 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 the present invention 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 Fc variants of the present invention may be produced by culturing a host cell transformed with nucleic acid, preferably an expression vector, containing nucleic acid encoding the Fc 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 in the present invention are described in the ATCC cell line catalog, available from the American Type Culture Collection. The methods of introducing exogenous nucleic acid into host cells are well known in the art, and will vary with the host cell used.
[146] In a preferred embodiment, Fc 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 the present invention for purification. Purification can often 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, hereby entirely incorporated by reference.
[147] 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. [148] 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.
[149] As is known in the art, a subset 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.
[150] 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.
[151] In a preferred embodiment, the immunogenicity of the Fc variants is determined experimentally using one or more cell-based assays. Several methods can be used for experimental confirmation of epitopes.
[152] The biological properties of the Fc variants of the present invention 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 of the present invention. Tests of the in humans are ultimately required for approval as drugs, and thus of course these experiments are contemplated. Thus the IgGs of the present invention may be tested in humans to determine their therapeutic efficacy, toxicity, immunogenicity, pharmacokinetics, and/or other clinical properties.
[153] The Fc variants of the present invention may find use in a wide range of products. In one embodiment the Fc variant of the present invention is a therapeutic, a diagnostic, or a research reagent, preferably a therapeutic. The Fc variant may find use in an antibody composition that is monoclonal or polyclonal. In a preferred embodiment, the Fc variants of the present invention are used to kill target cells that bear the target antigen, for example cancer cells. In an alternate embodiment, the Fc variants of the present invention 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 Fc variants of the present invention are used to block, antagonize, or agonize the target antigen and kill the target cells that bear the target antigen.
[154] The Fc variants of the present invention may be used for various therapeutic purposes. In a preferred embodiment, an antibody comprising the Fc variant is administered to a patient to treat an antibody-related disorder. A "patient" for the purposes of the present invention 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 Fc variant of the present invention. Antibody related disorders include but are not limited to autoimmune diseases, immunological diseases, infectious diseases, inflammatory diseases, neurological diseases, pain, pulmonary diseases, hematological conditions, fibrotic conditions, 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. Other conditions that may be treated include but are not limited to rheumatoid arthritis, juvenile rheumatoid arthritis, Crohn's disease, ulcerative colitis, Sjorgren's disease, multiple sclerosis, ankylosing spondylitis, asthma, allergies and allergenic conditions, graft versus host disease, and the like. The term "treatment" as used herein is meant to include therapeutic treatment, as well as prophylactic, or suppressive measures for the disease, condition or disorder. Thus, for example, successful administration of a pharmaceutical composition comprising an Fc variant of the present invention prior to onset of the disease results in "treatment" of the disease. As another example, successful administration of a pharmaceutical composition comprising an Fc variant of the present invention after clinical manifestation of the disease to combat the symptoms of the disease comprises "treatment" of the disease. "Treatment" also encompasses administration of a pharmaceutical composition comprising an Fc variant of the present invention after the appearance of the disease in order to eradicate the disease. Successful administration of a pharmaceutical composition comprising an Fc variant of the present invention after onset and after clinical symptoms have developed, with possible abatement of clinical symptoms and perhaps amelioration of the disease, comprises "treatment" of the disease. Those "in need of treatment" as used herein include mammals already having the disease or disorder, as well as those prone to having the disease or disorder, including those in which the disease or disorder is to be prevented. [155] In one embodiment, an Fc variant of the present invention is the only therapeutically active agent administered to a patient. Alternatively, the Fc variant of the present invention 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, as well as pre- or post-surgery. The IgG variants may be administered concomitantly with one or more other therapeutic regimens. For example, an Fc variant of the present invention may be administered to the patient along with surgery, chemotherapy, radiation therapy, or any or all of surgery, chemotherapy and radiation therapy. In one embodiment, the Fc variant of the present invention may be administered in conjunction with one or more antibodies, which may or may not comprise an Fc variant of the present invention. In accordance with another embodiment of the invention, the Fc variant of the present invention 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 Fc variants of the invention can be employed in combination with still other therapeutic techniques such as surgery.
[156] A variety of other therapeutic agents may find use for administration with the Fc variants of the present invention. In one embodiment, the IgG is administered with an anti-angiogenic agent. By "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). 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 Fc variants of the present invention are administered with 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. [157] Pharmaceutical compositions are contemplated wherein an Fc variant of the present invention and one or more therapeutically active agents are formulated. Formulations of the Fc variants of the present invention are prepared for storage by mixing said 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, hereby 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 Fc variants and other therapeutically active agents disclosed herein may also be formulated as immunoliposomes, and/or entrapped in microcapsules.
[158] The concentration of the therapeutically active Fc variant in the formulation may vary from about 0.001 to 100 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 Fc variant of the present invention 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 0.001 to 100 mg/kg of body weight or greater, for example 0.1 , 1 , 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.
[159] Administration of the pharmaceutical composition comprising an Fc variant of the present invention, 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, intranasaliy, intraotically, transdermally, topically (e.g., gels, salves, lotions, creams, etc.), intraperitoneally, intramuscularly, intrapulmonary (e.g., AERx® inhalable technology commercially available from Aradigm, or Inhance® pulmonary delivery system commercially available from Inhale Therapeutics), vaginally, parenterally, rectally, or intraocularly.
EXAMPLES
[160] 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.
Example 1. Fc variants with enhanced FcyR-mediated effector function
[161] Using the methods described in USSN 10/672,280, USSN 10/822,231 , USSN 11/124,620, and USSN 11/256,060, all hereby entirely incorporated by reference, additional Fc variants were designed for enhanced binding to Fc ligands and optimized effector function, and for reduced or ablated FcγR binding and effector function. The variants were constructed in the context of the anti- CD20 antibody PRO70769 (PCT/US2003/040426, hereby entirely incorporated by reference), which is known to mediate measurable CDC and ADCC in cell-based assays. Previously characterized variants were also constructed in PRO70769, in order to further characterize their properties and provide comparators for the current set of new variants. Figure 5 provides a list of these Fc variants. Notably, this variant set comprises a number of insertions. For example, "Insert L > 235-236 / I332E" refers to a double mutant comprising the substitution I332E and an insertion of leucine between residues 235 and 236.
[162] The genes for the variable regions of PRO70769 (Figures 24a and 24b) were constructed using recursive PCR, and subcloned into the mammalian expression vector pcDNA3.1Zeo (Invitrogen) comprising the full length light kappa (CK) and heavy chain IgGI constant regions. Variants were constructed in the variable region of the antibody in the pcDNA3.1Zeo vector using quick-change mutagenesis techniques (Stratagene), expressed in 293T cells. DNA was sequenced to confirm the fidelity of the sequences. Plasmids containing heavy chain gene (VH-CH1-CH2-CH3) (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). Select Fc variants were also expressed in the context of alemtuzumab.
[163] Binding affinity to human FcγRs by IgG antibodies was measured using a competitive AlphaScreen™ assay. The AlphaScreen is a bead-based 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 AlphaScreen was applied as a competition assay for screening the antibodies. Wild-type IgGI antibody was biotinylated by standard methods for attachment to streptavidin donor beads, and tagged FcγR was bound to glutathione chelate acceptor beads. In the absence of competing Fc polypeptides, wild-type antibody and FcγR interact and produce a signal at 520-620 nm. Addition of untagged antibody competes with wild-type Fc/FcγR interaction, reducing fluorescence quantitatively to enable determination of relative binding affinities.
[164] Figures 6 provides competitive AlphaScreen data for binding of select PRO70769 Fc variants to the human activating receptors V158 FcγRllla (Figure 6a) and F158 FcγRllla (Figure 6b). The data were fit to a one site competition model using nonlinear regression, and these fits are represented by the curves in the figure. These fits provide the inhibitory concentration 50% (IC50) (i.e. the concentration required for 50% inhibition) for each antibody, thus enabling the relative binding affinities relative to WT to be determined. Figure 5 provides the IC50's and Fold IC50's relative to WT for fits to these binding curves.
[165] Select Fc variants were reexpressed and reetested using the competition AlphaScreen assay for binding to human V158 FcγRllla and F158 FcγRllla (Figure 7). Figure 7a shows the binding data for these variants, and Figure 7b provides the IC50's and Fold IC50's relative to WT for fits to these binding curves.
[166] Based on these data, a number of additional Fc variants were constructed in the context of PRO70769 IgGI . Additionally, some Fc variants were constructed in the context of a novel IgG molecule lgG(1/2) ELLGG described in USSN 11/256,060, filed October 21 , 2005, hereby entirely incorporated by reference. These variants were constructed as described above, and expressed and purified along with a number of previously characterized Fc variants. These variants are listed in Figure 8a. Binding of the variant to the human activating receptors V158 FcγRllla and F158 FcγRllla, and the inhibitory receptor FcγRllb was measured using the competition AlphaScreen assay. Figure 8b shows data for binding of select variants to these receptors, and Figure 8a provides the IC50's and Folds relative to WT PRO70769 IgGI for all of this set of Fc variants.
[167] Because of the high avidity nature of the assay, the AlphaScreen provides only relative affinities. True binding constants were obtained using a competition SPR experiment (Nieba et al., 1996, Anal Biochem 234:155-65, hereby entirely incorporated by reference) in which unbound antibody in an antibody/FcγR equilibrium was captured to an FcγRllla surface. This experiment was carried out with the I332E and S239D/I332E variants in the context of trastuzumab IgGI , constructed and characterized previously (USSN 10/672,280, USSN 10/822,231 , and USSN 1 1/124,620, all hereby entirely incorporated by reference). WT and variant trastuzumab antibodies were expressed and purified as described above. For this experiment, data were acquired on a BIAcore 3000 instrument (BIAcore). V158 FcγRllla-His-GST was captured using immobilized anti-GST antibody, blocked with recombinant GST, and binding to antibody/receptor competition analyte was measured. Anti-GST antibody was covalently coupled to a CM5 sensor using the BIAcore GST Capture Kit. Flow cell 1 of every sensor chip was coupled with ethanolamine as a control of unspecific binding and to subtract bulk refractive index changes online. Running buffer was HBS-EP (0.01 M HEPES pH 7.4, 0.15 M NaCI, 3 mM EDTA, 0.005% v/v Surfactant P20, BIAcore), and chip regeneration buffer was Glycine 1.5 (10 mM glycine-HCI, pH 1.5, BIAcore). 1 μM V158 FcγRllla-His-GST was bound to the anti-GST CM5 chip in HBS-EP at 1 μl/min for 5 minutes. The surface was blocked with 5 μM recombinant GST (Sigma) injected at 1 μl/minute for 2 minutes. 100 nM wild-type or variant trastuzumab antibody was combined with V158 FcγRllla-His-GST in serial dilutions between 4 and 1000 nM and incubated for at least two hours at room temperature. The competition mixture was injected over the V158 FcγRllla-His-GST/recombinant GST surface for 30 seconds association in HBS-EP at 50 μl/minute. A cycle with antibody but no competing receptor provided a baseline response.
[168] An earlier described "competition BIAcore" method used fitted kinetic curves to derive on- rates (Nieba et al., 1996, Anal Biochem 234:155-65, hereby entirely incorporated by reference). We found this method to be less reliable since the on-rates derived from the kinetic curves showed no linear correlation to the antibody concentration applied. The analysis used in the present study is based on the proportionality of the initial rate R to the free antibody concentration (Holwill et al., 1996, Process Control and Quality 8:133-145; Edwards & Leatherbarrow, 1997, Anal Biochem 246:1-6, all hereby entirely incorporated by reference). Response units data were exported using BIAevaluation software (BIAcore) and analyzed using Microsoft Excel with Xlfit version 3.0.5 (IDBS). Initial rate (of signal increase) values were determined from the raw data of each sensorgram using the Excel formula for slope. The equilibrium dissociation binding constant (KD) was determined by plotting the log of FcγRllla concentration against the initial rate obtained at each concentration. GraphPad Prism (GraphPad Software) was used to fit the data to the following formula:
R = -^-([A0]- l0x - KD) + 4(KD 2 + 2{W)(KD) + (10x)2 + 2[A0]K0 - 2[A0)IO* +[A0]2) 2IA0] with:
[A0] = Antibody concentration
R0 = Initial rate at antibody concentration A0, with no competing receptor present
X = log[L0], where [L0] = input receptor concentration
KD = Equilibrium dissociation constant
R0 reflects the rate of binding between antibody and immobilized receptor (in the absence of competing receptor), and because of their different receptor affinities was calculated separately for WT, I332E, and S239D/I332E antibodies.
The formula for the initial rate R is derived from the definition of K0 for a single binding site:
[A)[L0] ^ κ [A0L0] and the conservation of mass [L0] = [L] + [A0L0] with:
[L] = concentration of free receptor
[169] Initial binding rates were determined from sensorgram raw data (Figure 9a), and KD's were calculated by plotting the log of receptor concentration against the initial rate obtained at each concentration (Figure 9b, 9c) (Edwards & Leatherbarrow, 1997, Anal Biochem 246:1-6, hereby entirely incorporated by reference). The WT KD (252 nM) agrees well with published data (208 nM from SPR, 535 nM from calorimetry) (Okazaki et al., 2004 J MoI Biol 336:1239-49, hereby entirely incorporated by reference). KD's of the I332E (30 nM) and S239D/I332E (2 nM) variants indicate approximately one- and two- logs greater affinity to V158 FcγRllla respectively.
[170] To investigate the capacity of antibodies comprising the Fc variants of the present invention to carry out FcγR-mediated effector function, in vitro cell-based ADCC assays were run using human PBMCs as effector cells. ADCC was measured by the release of lactose dehydrogenase using a LDH Cytotoxicity Detection Kit (Roche Diagnostic). Human PBMCs were purified from leukopacks using a ficoll gradient, and the CD20+ target lymphoma cell line WIL2-S was obtained from ATCC. Target cells were seeded into 96-well plates at 10,000 cells/well, and opsonized using Fc variant or WT antibodies at the indicated final concentration. Triton X100 and PBMCs alone were run as controls. Effector cells were added at 25:1 PBMCs:target cells, and the plate was incubated at 37°C for 4 hrs. Cells were incubated with the LDH reaction mixture, and fluorescence was measured using a Fusion™ Alpha-FP (Perkin Elmer). Data were normalized to maximal (triton) and minimal (PBMCs alone) lysis, and fit to a sigmoidal dose-response model. Figure 10 provides these data for select Fc variant antibodies in the context of the variable region PRO70769 and either IgGI or lgG(1/2) ELLGG. The Fc variants provide clear enhancements in FcγR-mediated CD20+ target cell lysis relative to the WT PRO70769 IgGI antibody.
[171] These in vitro assays suggest that the Fc variants of the present invention may provide enhanced potency and/or efficacy in a clinical setting. In vivo performance may be affected by a number of factors, including some of which are not considered by these in vitro experiments. One such parameter is the high concentration of non-specific IgG in serum, which has been shown to impact antibody clinical potency (Vugmeyster & Howell, 2004, lnt lmmunopharmacol 4:1117-24; Preithner et al., 2005, MoI Immunol, 43(8):1183-93, all hereby entirely incorporated by reference). In order to investigate how the Fc variants of the present invention perform in a solution more closely mimicking in vivo biology, the ADCC assays were repeated in the presence of a biologically relevant (1 mg/ml) concentration of IgG purified from human serum (purchased commercially from Jackson lmmunoresearch Lab, Inc.). These data are provided in Figure 11. The efficacy of the WT anti-CD20 antibody is not only reduced, but completely ablated in the presence of serum level IgG. In contrast, the Fc variant antibodies, although significantly reduced, still show substantial capacity to mediate killing against the target cell line.
Example 2. Fc variants with enhanced complement-mediated effector function
[172] A number of variants were designed with the goal of enhancing complement dependant cytotoxicity (CDC). In the same way that Fc/FcγR binding mediates ADCC, Fc/C1q binding mediates complement dependent cytotoxicity (CDC). There is currently no structure available for the Fc/C1q complex; however, mutagenesis studies have mapped the binding site on human IgG for C1q to a region centered on residues D270, K322, P329, and P331 (Idusogie et al., 2000, J Immunol 164:4178-4184; Idusogie et al., 2001 , J Immunol 166:2571-2575, both hereby entirely incorporated by reference). Figure 12 shows a structure of the human IgGI Fc region with this epicenter mapped. Select amino acid modifications disclosed in USSN 10/672,280, USSN 10/822,231 , USSN 11/124,620, and USSN 11/256,060, all hereby entirely incorporated by reference, that are structurally proximal to these four residues were investigated to explore variants that may mediate increased affinity for C1q and and/or provide enhanced CDC. Variants that previously showed enhanced FcγR affinity and FcγR-mediated effector function were included in this set of variants to characterize their complement properties. This variant library is provided in Figure 13.
[173] The variants were constructed as described above in the context of the anti-CD20 antibody PRO70769 (variable region) and either IgGI or lgG(1/2) ELLGG as the heavy chain constant region. Variants were expressed and purified as described above. A cell-based assay was used to measure the capacity of the Fc variants to mediate CDC. Lysis was measured using release of Alamar Blue to monitor lysis of Fc variant and WT PRO70769 -opsonized WIL2-S lymphoma cells by human serum complement. Target cells were washed 3x in 10% FBS medium by centrifugation and resuspension, and WT or variant rituximab antibody was added at the indicated final concentrations. Human serum complement (Quidel) was diluted 50% with medium and added to antibody-opsonized target cells. Final complement concentration was 1/6th original stock. Plates were incubated for 2 hrs at 37 0C, Alamar Blue was added, cells were cultured for two days, and fluorescence was measured. Representative data from this assay are shown in Figure 14. The binding data were normalized to the maximum and minimum luminescence signal for each particular curve, provided by the baselines at low and high antibody concentrations respectively. The data were fit to a sigmoidal dose-response with variable slope model using nonlinear regression, and these fits are represented by the curves in the figure. These fits provide the effective concentration 50% (EC50) (i.e. the concentration required for 50% response) for each antibody, enabling the relative binding affinities of Fc variants to be quantitatively determined. By dividing the EC50 for each variant by that of WT PRO70769, the fold- enhancement or reduction relative to WT PRO70769 (Fold WT) were obtained. These values are provided in Figure 13. Here a fold above 1 indicates an enhancement in CDC EC50, and a fold below 1 indicates a reduction in CDC EC50 relative to WT PRO70769.
[174] The data in Figures 13 and 14 indicate that a number of modifications provide enhanced CDC relative to WT PRO70769 IgGI . For example, greater than 2-fold CDC enhancement is observed for modifications 239D, 267D, 267Q, 268D, 268E, 268F, 268G, 2721, 276D, 276L, 276S, 278R, 282G, 284T, 285Y, 293R, 300T, 3241, 324T, 324V, 326E, 326T, 326W, 327D, 330H, 330S, 332E, 333F, 334T, and 335D (Figure 15). Additionally, the data show that a number of modifications provide reduced CDC relative to WT PRO70769 IgGI . For example, modifications that show 0.5 fold and lower relative CDC include 235D, 239D, 284D, 322H, 322T, 322Y, 327R, 330E, 3301, 330L, 330N, 330V, 331 D, and 331 L, 332E (Figure 15). These modifications provide further valuable structure activity relationship (SAR) information that may be used to guide further design of variants for enhanced CDC. Together the data suggest that modification at positions 235, 239, 267, 268, 272, 276, 278, 282, 284, 285, 293, 300, 322, 324, 326, 327, 330, 331 , 332, 333, 334, and 335 (Figure 15) may provide enhanced CDC relative to a parent Fc polypeptide.
Example 3. Fc variants with reduced FcyR- and complement-mediated effector function
[175] As described above, in contrast antibody therapeutics and indications wherein effector functions contribute to clinical efficacy, for some antibodies and clinical applications it may be favorable to reduce or eliminate binding to one or more FcγRs, or reduce or eliminate one or more FcγR- or complement-mediated effector functions including but not limited to ADCC, ADCP, and/or CDC. This is often the case for therapeutic antibodies whose mechanism of action involves blocking or antagonism but not killing of the cells bearing target antigen. In these cases depletion of target cells is undesirable and can be considered a side effect. Effector function can also be a problem for radiolabeled antibodies, referred to as radioconjugates, and antibodies conjugated to toxins, referred to as immunotoxins. These drugs can be used to destroy cancer cells, but the recruitment of immune cells via Fc interaction with FcγRs brings healthy immune cells in proximity to the deadly payload (radiation or toxin), resulting in depletion of normal lymphoid tissue along with targeted cancer cells. [176] A previously unconsidered advantage of ablated FcγR- and complement- binding is that in cases where effector function is not needed, binding to FcγR and complement may effectively reduce the active concentration of drug. Binding to Fc ligands may localize an antibody or Fc fusion to cell surfaces or in complex with serum proteins wherein it is less active or inactive relative to when it is free (uncomplexed). This may be due to decreased effective concentration at binding sites where the antibody is desired, or perhaps Fc ligand binding may put the Fc polypeptide in a conformation in which it is less active than it would be if it were unbound. An additional consideration is that FcγR- receptors may be one mechanism of antibody turnover, and can mediate uptake and processing by antigen presenting cells such as dendritic cells and macrophages. This may affect affect the pharmacokinetics (or in vivo half-life) of the antibody or Fc fusion and its immunogenicity, both of which are critical parameters of clinical performance.
[177] Visual inspection of the Fc/FcγR structure (Figure 2) and the aforedescribed Fc/C1q interface (Figure 12), as well as data disclosed above and in USSN 10/672,280, USSN 10/822,231 , USSN 11/124,620, and USSN 11/256,060, all hereby entirely incorporated by reference, were used to guide the design of a library to screen for variants with reduced affinity for FcγRs and reduced CDC. This variant library is provided in Figure 16. The variants were constructed in the context of PRO70769 IgGI , and expressed and purified as described above. Relative FcγR affinity was measured using the competition AlphaScreen assay, as described above. Figure 17 shows AlphaScreen data for binding of select Fc variants to human V158 FcγRllla, and Figure 16 provides their Fold ICδO's relative to WT PRO70769 IgGI . The variants were also investigated for their capacity to mediate complement- mediated lysis against CD20+ WIL2-S lymphoma target cells using the CDC assay described above. Figure 18 provides CDC data for select Fc variants, and Figure 16 provides their Fold EC50's relative to WT PRO70769 IgGI . Based on the results of these experiments, select Fc variants were characterized for their capacity to mediate FcγR-mediated effector function. An ADCC assay using human PBMCs as effector cells and WIL2-S lymphoma cells as target cells was carried out as described above. Figure 19 shows these ADCC data for select variants.
[178] The data indicate that modification at a number of positions provide reduced or ablated FcγR affinity, reduced FcγR-mediated effector function, and reduced complement-mediated effector function. Furthermore, modifications at some positions, including but not limited to 235 and 330, may provide reduced CDC but WT FcγR affinity. For example 235D, 330L, 330N, and 330R display such behavior. Alternatively, modification at some positions, including but not limited to 236 and 299, may provide reduced FcγR affinity but WT level CDC. For example 236I and 299A show these properties.
[179] Based on the results of these experiments, a number of modifications that simultaneously ablate FcγR affinity and CDC were combined in multiple mutations variants in a new library of Fc variants was designed to screen for variants with completely ablated FcγR affinity, FcγR-mediated effector function, and complement-mediated effector function. These variants include modifications at positions 234, 235, 236, 267, 269, 325, and 328, and are provided in Figure 20. Included in the set are the WT IgGI antibody, as well as lgG2 and lgG4 antibody versions, an aglycosylated variant N297S, and two variants previously characterized as having reduced effector function: L234A/L235A (Xu et al., 2000, Cellular Immunology 200:16-26; USSN 10/267,286, hereby entirely incorporated by reference) and E233P/L234V/L235A/G236- (Armour et al., 1999, Eur J Immunol 29:2613-2624, hereby entirely incorporated by reference).
[180] These variants were constructed in the context of the anti-CD20 antibody PRO70769, with the heavy chain constant region IgGI except for the lgG2 and lgG4 antibodies. Antibodies were expressed and purified as described previously. The competition AlphaScreen assay was used as described previously to measure the relative FcγR affinity of the Fc variants. Figure 21 shows AlphaScreen data for binding of select variants to the low affinity human activating receptor V158 FcγRllla, as well as the high affinity human activating receptor FcγRI. The fold IC50's relative to WT are provided in Figure 20. Because of its greater binding affinity for the Fc region, FcγRI provides a more stringent test for the variants. The data in Figures 20 and 21 support this, showing that although variants may substantially reduce or completely ablate affinity to FcγRllla, FcγRI binding is more modestly affected. The Fc variants were also tested for their capacity to mediate complement- mediated lysis against CD20+ WIL2-S cells using the CDC assay described above. Figure 22 shows CDC data for select Fc variants, and Figure 20 provides the fold EC50's relative to WT PRO70769 IgGl
[181] In order to investigate the capacity of the Fc variants to mediate ADCC, select variants were subcloned into the anti-Her2/neu antibody trastuzumab (variable region sequences provided in Figures 24c and 24d). Trastuzumab robustly provides a substantial signal in ADCC assays against Her2+ expressing cell lines, and therefore provides a stringent test of the Fc variants for reducing/ablating effector function. Fc variants L235G, G236R, G237K, N325L, N325A, L328R, L235G/G236R, G236R/G237K, G236R/N325L, G236R/L328R, G237K/N325L, L235G/G236R/G237K, and G236R/G237K/L328R were constructed in the context of trastuzumab IgGI . WT IgGI ,WT lgG2, and WT lgG4 antibody versions were constructed as well. An ADCC assay was carried out as described above, except the Her2+ breast carcinoma cell line SkBr-3 was used as target cells. Figure 23 provides the results of the ADCC experiments. The data indicate that some of the variants completely ablate ADCC. Additionally, although lgG2 also appears to mediate no ADCC, lgG4 does show a significant level of ADCC.
[182] The results show that amino acid modifications at a number of positions, including but not limited to 232, 234, 235, 236, 237, 238, 239, 265, 267, 269, 270, 297, 299, 325, 327, 328, 329, 330, and 331 , provide promising candidates for improving the clinical properties of antibodies and Fc fusions wherein FcγR binding, FcγR-mediated effector functions, and/or complement-mediated effector function are undesired. For example the amino acid modifications 232G, 234G, 234H, 235D, 235G, 235H, 236I, 236N, 236P, 236R, 237K, 237L, 237N, 237P, 238K, 239R, 265G, 267R, 269R, 270H, 297S, 299A, 299I, 299V, 325A, 325L, 327R, 328R, 329K, 330I, 330L, 330N, 330P, 330R, 330S, and 331 L provide significantly reduced Fc ligand binding properties and/or effector function. Particularly effective at reducing binding to Fc ligands and effector function are variants 236R/237K, 236R/325L, 236R/328R, 237K/325L, 237K/328R, 325L/328R, 235G/236R, 267R/269R, 234G/235G, 236R/237K/325L, 236R/325L/328R, 235G/236R/237K, and 237K/325L/328R. Notably, the amino acid modifications that compose these variants, including 234G, 235G, 236R, 237K, 267R1 269R, 325L, and 328R, are capable of reducing binding to both FcγRllla and FcγRI, and reducing CDC by greater than 10 fold. Additionally, the data show that human lgG2 has significantly reduced FcγR-affinity, FcγR-mediated effector function, and complement-mediated effector function relative to human lgG4.
[183] As discussed above, reduced FcγR affinity and/or effector function may be optimal for Fc polypeptides for which Fc ligand binding or effector function leads to toxicity and/or reduced efficacy. For example, antibodies that target CTLA-4 block inhibition of T-celi activation and are effective at promoting anti-tumor immune response, but destruction of T cells via antibody mediated effector functions may be counterproductive to mechanism of action and/or potentially toxic. Indeed toxicity has been observed with clinical use of the anti-CTLA-4 antibody ipilimumab (Maker et al., 2005, Ann Surg Oncol 12:1005-16, hereby entirely incorporated by reference). The sequences for the anti-CTLA- 4 antibody ipilimumab (Mab 10D.1 , MDX010) are provided in Figure 24, taken from US 6,984,720 SEQ ID:7 (VL, Figure 24e) and SEQ ID:17 (VH, Figure 24f) , hereby entirely incorporated by reference. For illustration purposes, a number of Fc variants of the present invention have been incorporated into the sequence of an antibody targeting CTLA-4. Because combinations of Fc variants of the present invention have typically resulted in additive or synergistic binding modulations, and accordingly additive or synergistic modulations in effector function, it is anticipated that as yet unexplored combinations of the Fc variants provided in the present invention, or with other previously disclosed modifications, will also provide favorable results. Potential Fc variants are provided in Figure 26a. The optimized antibody sequences sequences comprise at least one non-WT amino acid selected from the group consisting of X1, X2, X3, X4, X5, Xe, X7, and X8. For example, an improved anti- CTLA-4 antibody sequence comprising the L235G and G236R modifications in the IgGI constant region are provided in Figures 26b and 26c. Alternatively, as the present invention shows, lgG2 and lgG4 can also be used to reduce Fc ligand binding and Fc-mediated effector function. Figures 26b and 26d provide the sequences of improved anti-CTLA-4 lgG2 antibody sequences. The use of an anti-CTLA-4 here is solely an example, and is not meant to constrain application of the Fc variants to this antibody or any other particular Fc polypeptide. Other exemplary applications for reduced Fc ligand binding and/or effector function include but are not limited to anti-TNFα antibodies, including for example infliximab and adalimumab, anti-VEGF antibodies, including for example bevacizumab, anti- α4-integrin antibodies, including for example natalizumab, and anti-CD32b antibodies, including for example those described in USSN 10/643,857, hereby entirely incorporated by reference.
[184] This list of preferred Fc variants is not meant to constrain the present invention. Indeed all combinations of the any of the Fc variants provided are embodiments of the present invention. Furthermore, combinations of any of the Fc variants of the present invention with other discovered or undiscovered Fc variants may also provide favorable properties, and these combinations are also contemplated as embodiments of the present invention. Finally, it is anticipated from these results that other substitutions at positions mutated in present invention may also provide favorable binding enhancements and specificities, and thus substitutions at all positions disclosed herein are contemplated.
[185] All cited references are herein expressly incorporated by reference in their entirety.
[186] 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.

Claims

1. An Fc variant of a parent Fc polypeptide, wherein said Fc variant exhibits altered binding to at least on FcγR or altered antibody dependent cell-mediated cytotoxicity as compared to said parent Fc polypeptide, wherein said Fc variant comprises at least one amino acid modification in the Fc region of said parent Fc polypeptide, wherein said variant is selected from the group consisting of: 227G/332E, 234D/332E, 234E/332E, 234Y/332E, 234I/332E, 234G/332E, 235I/332E, 235S/332E, 235D/332E, 235E/332E, 246H/332E, 255Y/332E, 258H/332E, 260H/332E, 267E/332E, 267D/332E, 268E/330Y, 268D/330Y, 272R/332E, 272H/332E, 283H/332E, 293R/332E, 295E/332E, 304T/332E, 324I/332E, 324G/332E, 324I/332D, 324G/332D, 327D/332E, 328F/332E, 328Y/332E, 328D/332E, 328A/332D, 328T/332D, 328V/332D, 328I/332D, 328F/332D, 328Y/332D, 328M/332D, 328D/332D, 328E/332D, 328N/332D, 328Q/332D, 335D/332E, 239D/268E/330Y, 239D/332E/268E/330Y, 239D/332E/327A, 239D/332E/268E/327A, 239D/332E/330Y/327A, 332E/330Y/268E/327A, and 239D/332E/268E/330Y/327A, wherein numbering is according to the EU index.
2. An Fc variant according to claim 1 , wherein said Fc polypeptide composes a human IgGI antibody.
3. An Fc variant according to claim 2, wherein said Fc polypeptide composes a human IgGI antibody, wherein said IgGI antibody has the allotypic residues 356D and 358L.
4. An Fc variant according to claim 2, wherein said Fc polypeptide composes a human IgGI antibody, wherein said IgGI antibody has the allotypic residues 356E and 358M.
5. An Fc variant according to claim 1 , wherein said Fc polypeptide composes a lgG(1/2) ELLGG antibody as defined by SEQ ID:12.
6. An IgGI variant of a parent IgGI polypeptide, wherein said IgGI variant exhibits improved binding to at least on FcγR, wherein said Fc variant comprises at least one amino acid insertion in the Fc region of said parent IgGI polypeptide.
7. An IgGI variant according to claim 6, wherein said insertion occurs between two positions, wherein said two positions are selected from the group consisting of 235 and 236, 297 and 298, and 326 and 327, wherein numbering is according to the EU index.
8. An IgGI variant according to claim 7, wherein said insertion is selected from the group consisting of: an insertion of G between positions 235 and 236, an insertion of A between positions 235 and 236, an insertion of S between positions 235 and 236, an insertion of T between positions 235 and 236, an insertion of N between positions 235 and 236, an insertion of D between positions 235 and 236, an insertion of V between positions 235 and 236, an insertion of L between positions 235 and 236, an insertion of G between positions 235 and 236, an insertion of A between positions 235 and 236, an insertion of S between positions 235 and 236, an insertion of T between positions 235 and 236, an insertion of N between positions 235 and 236, an insertion of D between positions 235 and 236, an insertion of V between positions 235 and 236, an insertion of L between positions 235 and 236, an insertion of G between positions 297 and 298, an insertion of A between positions 297 and 298, an insertion of S between positions 297 and 298, an insertion of D between positions 297 and 298, an insertion of G between positions 326 and 327, an insertion of A between positions 326 and 327, an insertion of T between positions 326 and 327, an insertion of D between positions 326 and 327, and an insertion of E between positions 326 and 327, wherein numbering is according to the EU index.
9. An Fc variant of a parent Fc polypeptide, wherein said Fc variant exhibits altered complement dependent cytotoxicity as compared to said parent Fc polypeptide, wherein said Fc variant comprises at least one amino acid modification in the Fc region of said parent Fc polypeptide, wherein said modification is selected from the group consisting of: 2331, 234Y, 235D, 235Y, 239D, 267D, 267E, 267Q, 268D, 268E, 268F, 268G, 271 A, 271 D, 2711, 272H, 272), 272R, 274E, 274R, 274Y, 276D, 276L, 276S, 278E, 278H, 278Q, 278R, 281 D, 282G, 284D, 284E, 284T, 285Y, 293R, 300D, 300T, 320I, 320T, 320Y, 322H, 322T, 322Y, 324D, 324H, 324I, 324L, 324T, 324V, 326L, 326T, 327A, 327D, 327H, 327R, 328Q, 330E, 330G, 330H, 330I, 330L, 330N, 330V, 330Y, 331 D, 331 L, 332E, 333F, 334T, 335D, and 335Y, wherein numbering is according to the EU index.
10. An Fc variant of a parent Fc polypeptide, wherein said FG variant exhibits improved complement dependent cytotoxicity as compared to said parent Fc polypeptide, wherein said Fc variant comprises at least one amino acid modification in the Fc region of said parent Fc polypeptide, wherein said modification is at a position selected from the group consisting of: 235, 239, 267, 268, 272, 276, 278, 282, 284, 285, 293, 300, 322, 324, 327, 330, 331 , 332, and 335, wherein numbering is according to the EU index.
11. An Fc variant according to claim 10, wherein said Fc variant comprises one or more amino acid modifications selected from the group consisting of: 239D, 267D, 267Q, 268D, 268E, 268F, 268G, 272I1 276D, 276L, 276S, 278R, 282G, 284T, 285Y, 293R, 300T, 3241, 324T, 324V, 327D, 330H, 330S, 332E, 335D.
12. An Fc variant of a parent Fc polypeptide, wherein said Fc variant exhibits reduced binding to one or more FcγRs, reduced antibody dependent cell-mediated cytotoxicity, or reduced complement dependent cytotoxicity as compared to said parent Fc polypeptide, wherein said Fc variant comprises at least one amino acid modification in the Fc region of said parent Fc polypeptide, wherein said variant is selected from the group consisting of: 236R/237K, 236R/325L, 236R/328R, 237K/325L, 237K/328R, 325L/328R, 235G/236R, 267R/269R, 234G/235G, 236R/237K/325L, 236R/325L/328R, 235G/236R/237K, and 237K/325L/328R.
13. An Fc variant of a parent Fc polypeptide, wherein said Fc variant reduces by at least 10 fold relative to said parent Fc polypeptide: affinity to human FcγRllla, affinity to human FcγRI, and complement dependent cytotoxicity.
14. An Fc variant according to claim 13, wherein said Fc polypeptide comprises at least one modification at a position selected from the group consisting of: 234G, 235G, 236R, 237K, 267R, 269R, 325L, 328R, wherein numbering is a according to the EU index.
15. An IgG antibody that targets CTLA-4, TNFα, VEGF, α4-integrin, or CD32b, wherein said antibody has reduced FcγR affinity, reduced antibody dependent cell-mediated cytotoxicity, or reduced complement dependent cytotoxicity relative to a WT antibody.
16. An IgG antibody according to claim 15, wherein said antibody comprises an amino acid modification at a position selected from the group consisting of: 234, 235, 236, 237, 267, 269, 325, and 328, relative to a parent IgG antibody, wherein numbering is according to the EU index.
17. An IgG antibody according to claim 15, wherein said antibody is a human lgG2 or lgG4 antibody.
18. An IgG antibody variant of a parent IgG antibody, wherein said IgG variant exhibits improved antibody dependent cell-mediated cytotoxicity in the presence of competing IgG.
PCT/US2006/011752 2005-03-31 2006-03-31 Fc VARIANTS WITH OPTIMIZED PROPERTIES WO2006105338A2 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
AU2006230413A AU2006230413B8 (en) 2005-03-31 2006-03-31 Fc variants with optimized properties
JP2008504389A JP2008537941A (en) 2005-03-31 2006-03-31 Fc variants with optimized properties
CA002602663A CA2602663A1 (en) 2005-03-31 2006-03-31 Fc variants with optimized properties
EP06748967A EP1871808A2 (en) 2005-03-31 2006-03-31 Fc VARIANTS WITH OPTIMIZED PROPERTIES
IL186145A IL186145A0 (en) 2005-03-31 2007-09-20 Fc VARIANTS WITH OPTIMIZED PROPERTIES

Applications Claiming Priority (10)

Application Number Priority Date Filing Date Title
US66719705P 2005-03-31 2005-03-31
US60/667,197 2005-03-31
US11/124,620 US8188231B2 (en) 2002-09-27 2005-05-05 Optimized FC variants
US11/124,620 2005-05-05
US70537805P 2005-08-03 2005-08-03
US60/705,378 2005-08-03
US72329405P 2005-10-03 2005-10-03
US72333505P 2005-10-03 2005-10-03
US60/723,294 2005-10-03
US60/723,335 2005-10-03

Publications (2)

Publication Number Publication Date
WO2006105338A2 true WO2006105338A2 (en) 2006-10-05
WO2006105338A3 WO2006105338A3 (en) 2007-03-29

Family

ID=37054149

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2006/011752 WO2006105338A2 (en) 2005-03-31 2006-03-31 Fc VARIANTS WITH OPTIMIZED PROPERTIES

Country Status (5)

Country Link
EP (1) EP1871808A2 (en)
AU (1) AU2006230413B8 (en)
CA (1) CA2602663A1 (en)
IL (1) IL186145A0 (en)
WO (1) WO2006105338A2 (en)

Cited By (132)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007008943A2 (en) * 2005-07-08 2007-01-18 Xencor, Inc. Optimized anti-ep-cam antibodies
WO2007041635A2 (en) * 2005-10-03 2007-04-12 Xencor, Inc. Fc variants with optimized fc receptor binding properties
JP2008505174A (en) * 2004-07-15 2008-02-21 ゼンコー・インコーポレイテッド Optimized Fc variant
WO2008065543A2 (en) 2006-11-28 2008-06-05 Centelion Modified soluble fgf receptor fc fusions with improved biological activity
WO2008091798A2 (en) * 2007-01-22 2008-07-31 Xencor, Inc. Optimized ca9 antibodies and methods of using the same
WO2008091954A2 (en) * 2007-01-23 2008-07-31 Xencor, Inc. Optimized cd40 antibodies and methods of using the same
WO2008092117A2 (en) * 2007-01-25 2008-07-31 Xencor, Inc. Immunoglobulins with modifications in the fcr binding region
WO2008098115A2 (en) * 2007-02-07 2008-08-14 Xencor, Inc. Optimized igf-1r antibodies and methods of using the same
WO2008121160A2 (en) * 2006-11-21 2008-10-09 Xencor, Inc. Optimized antibodies that target cd5
WO2008150494A1 (en) * 2007-05-30 2008-12-11 Xencor, Inc. Methods and compositions for inhibiting cd32b expressing cells
WO2009016164A1 (en) * 2007-08-01 2009-02-05 Glaxo Group Limited Novel antibodies
WO2009129538A2 (en) * 2008-04-18 2009-10-22 Xencor, Inc. Human equivalent monoclonal antibodies engineered from nonhuman variable regions
EP2119780A1 (en) * 2007-01-24 2009-11-18 Kyowa Hakko Kirin Co., Ltd. Genetically recombinant antibody composition having enhanced effector activity
US7740847B2 (en) 2004-08-04 2010-06-22 Applied Molecular Evolution, Inc. Variant Fc regions
WO2010146059A2 (en) 2009-06-16 2010-12-23 F. Hoffmann-La Roche Ag Biomarkers for igf-1r inhibitor therapy
WO2011039724A1 (en) 2009-10-02 2011-04-07 Sanofi-Aventis Antibodies that specifically bind to the epha2 receptor
WO2011100403A1 (en) 2010-02-10 2011-08-18 Immunogen, Inc Cd20 antibodies and uses thereof
WO2011108502A1 (en) 2010-03-02 2011-09-09 協和発酵キリン株式会社 Modified antibody composition
WO2011122011A2 (en) 2010-03-30 2011-10-06 Chugai Seiyaku Kabushiki Kaisha Antibodies with modified affinity to fcrn that promote antigen clearance
US8039595B2 (en) 2000-10-06 2011-10-18 Kyowa Hakko Kirin Co., Ltd. Glycoengineered, recombinant antibody to CCR-4 with reduced fucosylation
WO2011091078A3 (en) * 2010-01-19 2011-12-01 Xencor, Inc. Antibody fc variants with enhanced complement activity
WO2012032080A1 (en) 2010-09-07 2012-03-15 F-Star Biotechnologische Forschungs- Und Entwicklungsges.M.B.H Stabilised human fc
US8153765B2 (en) 2006-10-19 2012-04-10 Sanof Aventis Anti-CD38 antibodies for the treatment of cancer
WO2012073992A1 (en) 2010-11-30 2012-06-07 中外製薬株式会社 Antigen-binding molecule capable of binding to plurality of antigen molecules repeatedly
WO2012118813A2 (en) 2011-03-03 2012-09-07 Apexigen, Inc. Anti-il-6 receptor antibodies and methods of use
WO2012122513A2 (en) 2011-03-10 2012-09-13 Omeros Corporation Generation of anti-fn14 monoclonal antibodies by ex-vivo accelerated antibody evolution
WO2012133782A1 (en) 2011-03-30 2012-10-04 中外製薬株式会社 Retention of antigen-binding molecules in blood plasma and method for modifying immunogenicity
WO2012149356A2 (en) 2011-04-29 2012-11-01 Apexigen, Inc. Anti-cd40 antibodies and methods of use
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
WO2013046722A1 (en) 2011-09-30 2013-04-04 中外製薬株式会社 Ion concentration-dependent binding molecule library
WO2013047729A1 (en) 2011-09-30 2013-04-04 中外製薬株式会社 Antigen-binding molecule inducing immune response to target antigen
WO2013046704A2 (en) 2011-09-30 2013-04-04 Chugai Seiyaku Kabushiki Kaisha THERAPEUTIC ANTIGEN-BINDING MOLECULE WITH A FcRn-BINDING DOMAIN THAT PROMOTES ANTIGEN CLEARANCE
WO2013067098A1 (en) 2011-11-02 2013-05-10 Apexigen, Inc. Anti-kdr antibodies and methods of use
WO2013081143A1 (en) 2011-11-30 2013-06-06 中外製薬株式会社 Drug containing carrier into cell for forming immune complex
WO2013087789A1 (en) * 2011-12-13 2013-06-20 Glykos Finland Ltd. Antibody isoform arrays and methods thereof
WO2013093809A1 (en) 2011-12-23 2013-06-27 Pfizer Inc. Engineered antibody constant regions for site-specific conjugation and methods and uses therefor
WO2013100120A1 (en) 2011-12-28 2013-07-04 中外製薬株式会社 Humanized anti-epiregulin antibody, and cancer therapeutic agent comprising said antibody as active ingredient
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
US8546539B2 (en) 2007-05-14 2013-10-01 Novimmune S.A. Fc receptor-binding polypeptides with modified effector functions
WO2013147153A1 (en) 2012-03-29 2013-10-03 株式会社未来創薬研究所 Anti-lamp5 antibody and utilization thereof
WO2013180200A1 (en) 2012-05-30 2013-12-05 中外製薬株式会社 Target-tissue-specific antigen-binding molecule
WO2013180201A1 (en) 2012-05-30 2013-12-05 中外製薬株式会社 Antigen-binding molecule for eliminating aggregated antigens
US8679491B2 (en) 1999-04-09 2014-03-25 Kyowa Hakko Kirin Co., Ltd. Method of modulating the activity of functional immune molecules
WO2014072876A1 (en) 2012-11-09 2014-05-15 Pfizer Inc. Platelet-derived growth factor b specific antibodies and compositions and uses thereof
WO2014097648A1 (en) 2012-12-21 2014-06-26 中外製薬株式会社 Gpc3-targeted therapeutic agent for administration to patients for whom gpc3-targeted therapeutic agent therapy is effective
WO2014190441A1 (en) 2013-05-31 2014-12-04 Zymeworks Inc. Heteromultimers with reduced or silenced effector function
WO2014208482A1 (en) 2013-06-24 2014-12-31 中外製薬株式会社 Therapeutic agent comprising humanized anti-epiregulin antibody as active ingredient for non-small-cell lung carcinoma excluding adenocarcinoma
US8975376B2 (en) 2010-02-23 2015-03-10 Sanofi Anti-alpha2 integrin antibodies and their uses
WO2015046467A1 (en) 2013-09-27 2015-04-02 中外製薬株式会社 Method for producing polypeptide heteromultimer
US9017683B2 (en) 2009-05-01 2015-04-28 The University Of Tokyo Highly effective anti-cadherin antibody for induction of antibody-dependent cellular cytotoxicity in vivo
WO2015073721A1 (en) 2013-11-13 2015-05-21 Zymeworks Inc. Monovalent antigen binding constructs targeting egfr and/or her2 and uses thereof
WO2015073580A1 (en) 2013-11-13 2015-05-21 Pfizer Inc. Tumor necrosis factor-like ligand 1a specific antibodies and compositions and uses thereof
WO2015077891A1 (en) 2013-11-27 2015-06-04 Zymeworks Inc. Bispecific antigen-binding constructs targeting her2
WO2015083764A1 (en) 2013-12-04 2015-06-11 中外製薬株式会社 Antigen-binding molecules, the antigen-binding activity of which varies according to the concentration of compounds, and libraries of said molecules
US9079949B1 (en) 2014-03-07 2015-07-14 Alexion Pharmaceuticals, Inc. Anti-C5 antibodies having improved pharmacokinetics
US9085621B2 (en) 2010-09-10 2015-07-21 Apexigen, Inc. Anti-IL-1β antibodies
WO2015109212A1 (en) 2014-01-17 2015-07-23 Pfizer Inc. Anti-il-2 antibodies and compositions and uses thereof
US9102724B2 (en) 2011-08-12 2015-08-11 Omeros Corporation Anti-FZD10 monoclonal antibodies and methods for their use
US9234039B2 (en) 2010-08-31 2016-01-12 Sanofi Peptide or peptide complex binding to ALPHA2 integrin and methods and uses involving the same
WO2016082044A1 (en) 2014-11-27 2016-06-02 Zymeworks Inc. Methods of using bispecific antigen-binding constructs targeting her2
WO2016125495A1 (en) 2015-02-05 2016-08-11 Chugai Seiyaku Kabushiki Kaisha Antibodies comprising an ion concentration dependent antigen-binding domain, fc region variants, il-8-binding antibodies, and uses therof
WO2017015619A1 (en) 2015-07-23 2017-01-26 The Regents Of The University Of California Antibodies to coagulation factor xia and uses thereof
WO2017046994A1 (en) 2015-09-18 2017-03-23 Chugai Seiyaku Kabushiki Kaisha Il-8-binding antibodies and uses thereof
WO2017070561A1 (en) 2015-10-23 2017-04-27 Pfizer Inc. Anti-il-2 antibodies and compositions and uses thereof
US9714282B2 (en) 2003-09-26 2017-07-25 Xencor, Inc. Optimized Fc variants and methods for their generation
US9718883B2 (en) 2003-09-10 2017-08-01 Amgen Fremont Inc. Antibodies to M-CSF
WO2017159699A1 (en) 2016-03-15 2017-09-21 Chugai Seiyaku Kabushiki Kaisha Methods of treating cancers using pd-1 axis binding antagonists and anti-gpc3 antibodies
US9803023B2 (en) 2004-11-12 2017-10-31 Xencor, Inc. Fc variants with altered binding to FcRn
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
WO2018045379A1 (en) 2016-09-02 2018-03-08 Dana-Farber Cancer Institute, Inc. Composition and methods of treating b cell disorders
US9914777B2 (en) 2015-07-10 2018-03-13 Merus N.V. Human CD3 binding antibody
US10000560B2 (en) 2014-12-19 2018-06-19 Chugai Seiyaku Kabushiki Kaisha Anti-myostatin antibodies, polypeptides containing variant Fc regions, and methods of use
WO2018129029A1 (en) 2017-01-04 2018-07-12 Immunogen, Inc. Met antibodies and immunoconjugates and uses thereof
US10064952B2 (en) 2014-10-09 2018-09-04 Genzyme Corporation Glycoengineered antibody drug conjugates
EP2647707B1 (en) 2010-11-30 2018-09-12 Chugai Seiyaku Kabushiki Kaisha Cytotoxicity-inducing therapeutic agent
USRE47123E1 (en) 2006-07-18 2018-11-13 Sanofi EPHA2 receptor antagonist antibodies
US10214589B2 (en) 2013-03-11 2019-02-26 Genzyme Corporation Site-specific antibody-drug conjugation through glycoengineering
WO2019059411A1 (en) 2017-09-20 2019-03-28 Chugai Seiyaku Kabushiki Kaisha Dosage regimen for combination therapy using pd-1 axis binding antagonists and gpc3 targeting agent
WO2019090002A1 (en) * 2017-11-02 2019-05-09 Systimmune, Inc. Bispecific antibodies and methods of making and using thereof
US10336818B2 (en) 2004-11-12 2019-07-02 Xencor, Inc. Fc variants with altered binding to FcRn
US10358492B2 (en) 2012-09-27 2019-07-23 Merus N.V. Bispecific IgG antibodies as T cell engagers
EP3557260A1 (en) 2012-12-21 2019-10-23 Chugai Seiyaku Kabushiki Kaisha Gpc3-targeting drug which is administered to patient responsive to gpc3-targeting drug therapy
CN110546162A (en) * 2017-03-28 2019-12-06 礼进生物医药控股有限公司 Therapeutic agents and methods for enhancing immune response in tumor microenvironment
WO2019234576A1 (en) 2018-06-03 2019-12-12 Lamkap Bio Beta Ltd. Bispecific antibodies against ceacam5 and cd47
WO2019244107A1 (en) 2018-06-21 2019-12-26 Daiichi Sankyo Company, Limited Compositions including cd3 antigen binding fragments and uses thereof
WO2020014306A1 (en) 2018-07-10 2020-01-16 Immunogen, Inc. Met antibodies and immunoconjugates and uses 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
US20200123261A1 (en) * 2016-04-01 2020-04-23 Avidity Biosciences, Inc. Nucleic acid-polypeptide compositions and uses thereof
WO2020189748A1 (en) 2019-03-19 2020-09-24 中外製薬株式会社 Antigen-binding molecule containing antigen-binding domain of which binding activity to antigen is changed depending on mta, and library for obtaining said antigen-binding domain
US10793616B2 (en) 2015-05-04 2020-10-06 Apogenix Ag Single-chain CD40-receptor agonist proteins
US10836813B2 (en) 2012-09-12 2020-11-17 Genzyme Corporation Fc containing polypeptides with altered glycosylation and reduced effector function
US10858686B2 (en) 2010-05-27 2020-12-08 Merck Sharp & Dohme Corp. Method for preparing antibodies having improved properties
US10919953B2 (en) 2012-08-24 2021-02-16 Chugai Seiyaku Kabushiki Kaisha FcgammaRIIB-specific Fc region variant
US10934344B2 (en) 2006-03-31 2021-03-02 Chugai Seiyaku Kabushiki Kaisha Methods of modifying antibodies for purification of bispecific antibodies
US11001643B2 (en) 2014-09-26 2021-05-11 Chugai Seiyaku Kabushiki Kaisha Cytotoxicity-inducing therapeutic agent
EP3831849A1 (en) 2019-12-02 2021-06-09 LamKap Bio beta AG Bispecific antibodies against ceacam5 and cd47
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
US11072666B2 (en) 2016-03-14 2021-07-27 Chugai Seiyaku Kabushiki Kaisha Cell injury inducing therapeutic drug for use in cancer therapy
US11078283B2 (en) 2018-01-17 2021-08-03 Apexigen, Inc. Anti-PD-L1 antibodies and methods of use
CN113660952A (en) * 2019-04-04 2021-11-16 小野药品工业株式会社 Bispecific antibodies
GB2595299A (en) * 2020-05-21 2021-11-24 Mabsolve Ltd Modified immunoglobulin FC regions
US11186638B2 (en) 2011-09-12 2021-11-30 Genzyme Corporation Anti-αβTCR antibody
EP3925977A1 (en) 2012-10-30 2021-12-22 Apexigen, Inc. Anti-cd40 antibodies and methods of use
US11236168B2 (en) 2012-08-24 2022-02-01 Chugai Seiyaku Kabushiki Kaisha Mouse FcγammaRII-specific Fc antibody
US11267868B2 (en) 2013-04-02 2022-03-08 Chugai Seiyaku Kabushiki Kaisha Fc region variant
US11359009B2 (en) 2015-12-25 2022-06-14 Chugai Seiyaku Kabushiki Kaisha Anti-myostatin antibodies and methods of use
US11365256B2 (en) 2016-06-08 2022-06-21 Xencor, Inc. Methods and compositions for inhibiting CD32B expressing cells in IGG4-related diseases
US11365241B2 (en) 2017-07-27 2022-06-21 Alexion Pharmaceuticals, Inc. High concentration anti-C5 antibody formulations
WO2022130348A1 (en) 2020-12-18 2022-06-23 Lamkap Bio Beta Ag Bispecific antibodies against ceacam5 and cd47
US11401348B2 (en) 2009-09-02 2022-08-02 Xencor, Inc. Heterodimeric Fc variants
US11485790B2 (en) 2014-04-07 2022-11-01 Chugai Seiyaku Kabushiki Kaisha Immunoactivating antigen-binding molecule
US11505605B2 (en) 2014-05-13 2022-11-22 Chugai Seiyaku Kabushiki Kaisha T cell-redirected antigen-binding molecule for cells having immunosuppression function
WO2022263501A1 (en) 2021-06-18 2022-12-22 F. Hoffmann-La Roche Ag Bispecific anti-ccl2 antibodies
US11649293B2 (en) 2015-11-18 2023-05-16 Chugai Seiyaku Kabushiki Kaisha Method for enhancing humoral immune response
US11649262B2 (en) 2015-12-28 2023-05-16 Chugai Seiyaku Kabushiki Kaisha Method for promoting efficiency of purification of Fc region-containing polypeptide
US11660340B2 (en) 2015-11-18 2023-05-30 Chugai Seiyaku Kabushiki Kaisha Combination therapy using T cell redirection antigen binding molecule against cell having immunosuppressing function
US11673971B2 (en) * 2016-09-23 2023-06-13 Marengo Therapeutics, Inc. Multispecific antibody molecules comprising lambda and kappa light chains
US11697690B2 (en) 2014-03-19 2023-07-11 Genzyme Corporation Site-specific glycoengineering of targeting moieties
US11746150B2 (en) 2017-12-19 2023-09-05 Surrozen Operating, Inc. Anti-LRP5/6 antibodies and methods of use
US11760807B2 (en) 2014-05-08 2023-09-19 Chugai Seiyaku Kabushiki Kaisha GPC3-targeting drug which is administered to patient responsive to GPC3-targeting drug therapy
US11773171B2 (en) 2017-12-19 2023-10-03 Surrozen Operating, Inc. WNT surrogate molecules and uses thereof
EP4253421A2 (en) 2018-03-13 2023-10-04 Zymeworks BC Inc. Anti-her2 biparatopic antibody-drug conjugates and methods of use
WO2023242351A1 (en) 2022-06-16 2023-12-21 Lamkap Bio Beta Ag Combination therapy of bispecific antibodies against ceacam5 and cd47 and bispecific antibodies against ceacam5 and cd3
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
RU2814713C2 (en) * 2019-04-04 2024-03-04 Оно Фармасьютикал Ко., Лтд. Bispecific antibody
US11932685B2 (en) 2007-10-31 2024-03-19 Xencor, Inc. Fc variants with altered binding to FcRn
US11938194B2 (en) 2017-02-28 2024-03-26 Seagen Inc. Cysteine mutated antibodies for conjugation

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8101720B2 (en) 2004-10-21 2012-01-24 Xencor, Inc. Immunoglobulin insertions, deletions and substitutions
BR112014010257A2 (en) 2011-10-31 2017-04-18 Chugai Pharmaceutical Co Ltd antigen binding molecule having regulated conjugation between heavy and light chains
AR104368A1 (en) 2015-04-03 2017-07-19 Lilly Co Eli ANTI-CD20- / ANTI-BAFF BIESPECTIFIC ANTIBODIES

Family Cites Families (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ATE375365T1 (en) * 1998-04-02 2007-10-15 Genentech Inc ANTIBODIES VARIANTS AND FRAGMENTS THEREOF
BR0008758A (en) * 1999-01-15 2001-12-04 Genentech Inc Variants of parental polypeptides with altered effector function, polypeptides, isolated nucleic acid composition, vector, host cell, method for producing a polypeptide variant, method for treating a disorder in mammals and method for producing a variant fc region
MXPA02010011A (en) * 2000-04-11 2003-04-25 Genentech Inc Multivalent antibodies and uses therefor.
US20040002587A1 (en) 2002-02-20 2004-01-01 Watkins Jeffry D. Fc region variants
US7657380B2 (en) * 2003-12-04 2010-02-02 Xencor, Inc. Methods of generating variant antibodies with increased host string content
US20040132101A1 (en) * 2002-09-27 2004-07-08 Xencor Optimized Fc variants and methods for their generation
EP2042517B1 (en) * 2002-09-27 2012-11-14 Xencor, Inc. Optimized FC variants and methods for their generation
ES2897506T3 (en) * 2003-01-09 2022-03-01 Macrogenics Inc Identification and modification of antibodies with variant Fc regions and methods of using them
US20050142133A1 (en) * 2003-12-03 2005-06-30 Xencor, Inc. Optimized proteins that target the epidermal growth factor receptor
DE602005015542D1 (en) * 2004-01-12 2009-09-03 Applied Molecular Evolution VARIANTS OF FC REGION
AU2005227326B2 (en) * 2004-03-24 2009-12-03 Xencor, Inc. Immunoglobulin variants outside the Fc region
WO2006085967A2 (en) * 2004-07-09 2006-08-17 Xencor, Inc. OPTIMIZED ANTI-CD20 MONOCONAL ANTIBODIES HAVING Fc VARIANTS
CN103351434B (en) * 2004-07-15 2015-09-30 赞科股份有限公司 The Fc variant optimized
WO2006047350A2 (en) * 2004-10-21 2006-05-04 Xencor, Inc. IgG IMMUNOGLOBULIN VARIANTS WITH OPTIMIZED EFFECTOR FUNCTION
EP2845865A1 (en) * 2004-11-12 2015-03-11 Xencor Inc. Fc variants with altered binding to FcRn

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of EP1871808A2 *

Cited By (243)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10233247B2 (en) 1999-04-09 2019-03-19 Kyowa Hakko Kirin Co., Ltd Method of modulating the activity of functional immune molecules
US8679491B2 (en) 1999-04-09 2014-03-25 Kyowa Hakko Kirin Co., Ltd. Method of modulating the activity of functional immune molecules
US8039595B2 (en) 2000-10-06 2011-10-18 Kyowa Hakko Kirin Co., Ltd. Glycoengineered, recombinant antibody to CCR-4 with reduced fucosylation
US10233475B2 (en) 2000-10-06 2019-03-19 Kyowa Hakko Kirin Co., Ltd Antibody composition-producing cell
US8895266B2 (en) 2000-10-06 2014-11-25 Kyowa Hakko Kirin Co., Ltd Antibody composition-producing cell
US8067232B2 (en) 2000-10-06 2011-11-29 Kyowa Hakko Kirin Co., Ltd Antibody composition-producing cell with inactivated A-1,6-fusocyltransferase
US9409982B2 (en) 2000-10-06 2016-08-09 Kyowa Hakko Kirin Co., Ltd Antibody composition-producing cell
US8110195B2 (en) 2000-10-06 2012-02-07 Kyowa Hakko Kirin Co., Ltd. Antibody composition exhibiting cellular cytotoxicity due to glycosylation and containing ganglioside GM2 binding antibody
US8367407B2 (en) 2000-10-06 2013-02-05 Kyowa Hakko Kirin Co., Ltd. Cells with altered fucosylation and producing antibodies therefrom
US8329443B2 (en) 2000-10-06 2012-12-11 Kyowa Hakko Kirin Co., Ltd Antibody composition-producing cell
US8101185B2 (en) 2000-10-06 2012-01-24 Kyowa Hakko Kirin Co., Ltd. Anti-il-5 antibody composition exhibiting cellular cytotoxicity due to glycosylation
US10280219B2 (en) 2003-09-10 2019-05-07 Amgen Fremont Inc. Antibodies to M-CSF
US9718883B2 (en) 2003-09-10 2017-08-01 Amgen Fremont Inc. Antibodies to M-CSF
US9714282B2 (en) 2003-09-26 2017-07-25 Xencor, Inc. Optimized Fc variants and methods for their generation
JP2008505174A (en) * 2004-07-15 2008-02-21 ゼンコー・インコーポレイテッド Optimized Fc variant
US7740847B2 (en) 2004-08-04 2010-06-22 Applied Molecular Evolution, Inc. Variant Fc regions
US10336818B2 (en) 2004-11-12 2019-07-02 Xencor, Inc. Fc variants with altered binding to FcRn
US9803023B2 (en) 2004-11-12 2017-10-31 Xencor, Inc. Fc variants with altered binding to FcRn
US11198739B2 (en) 2004-11-12 2021-12-14 Xencor, Inc. Fc variants with altered binding to FcRn
WO2007008943A2 (en) * 2005-07-08 2007-01-18 Xencor, Inc. Optimized anti-ep-cam antibodies
WO2007008943A3 (en) * 2005-07-08 2007-12-13 Xencor Inc Optimized anti-ep-cam antibodies
WO2007041635A3 (en) * 2005-10-03 2008-11-13 Xencor Inc Fc variants with optimized fc receptor binding properties
US9040041B2 (en) 2005-10-03 2015-05-26 Xencor, Inc. Modified FC molecules
WO2007041635A2 (en) * 2005-10-03 2007-04-12 Xencor, Inc. Fc variants with optimized fc receptor binding properties
US11046784B2 (en) 2006-03-31 2021-06-29 Chugai Seiyaku Kabushiki Kaisha Methods for controlling blood pharmacokinetics of antibodies
US10934344B2 (en) 2006-03-31 2021-03-02 Chugai Seiyaku Kabushiki Kaisha Methods of modifying antibodies for purification of bispecific antibodies
USRE47123E1 (en) 2006-07-18 2018-11-13 Sanofi EPHA2 receptor antagonist antibodies
EP3498735A1 (en) 2006-10-19 2019-06-19 Sanofi Novel anti-cd38 antibodies for the treatment of leukemia
EP3909980A1 (en) 2006-10-19 2021-11-17 Sanofi Novel anti-cd38 antibodies for the treatment of cancer
US8153765B2 (en) 2006-10-19 2012-04-10 Sanof Aventis Anti-CD38 antibodies for the treatment of cancer
WO2008121160A3 (en) * 2006-11-21 2009-01-22 Xencor Inc Optimized antibodies that target cd5
WO2008121160A2 (en) * 2006-11-21 2008-10-09 Xencor, Inc. Optimized antibodies that target cd5
WO2008065543A2 (en) 2006-11-28 2008-06-05 Centelion Modified soluble fgf receptor fc fusions with improved biological activity
US8481487B2 (en) 2006-11-28 2013-07-09 Aventis Pharma S.A. Modified soluble FGF receptor Fc fusions method
US8119770B2 (en) 2006-11-28 2012-02-21 Aventis Pharma Sa Modified soluble FGF receptor Fc fusions with improved biological activity
WO2008091798A3 (en) * 2007-01-22 2008-10-02 Xencor Inc Optimized ca9 antibodies and methods of using the same
WO2008091798A2 (en) * 2007-01-22 2008-07-31 Xencor, Inc. Optimized ca9 antibodies and methods of using the same
JP2010514460A (en) * 2007-01-23 2010-05-06 ゼンコー・インコーポレイテッド Optimized CD40 antibodies and methods of using the same
WO2008091954A2 (en) * 2007-01-23 2008-07-31 Xencor, Inc. Optimized cd40 antibodies and methods of using the same
WO2008091954A3 (en) * 2007-01-23 2008-09-04 Xencor Inc Optimized cd40 antibodies and methods of using the same
EP2119780A1 (en) * 2007-01-24 2009-11-18 Kyowa Hakko Kirin Co., Ltd. Genetically recombinant antibody composition having enhanced effector activity
US8492526B2 (en) 2007-01-24 2013-07-23 Kyowa Hakko Kirin Co., Ltd. Effector function enhanced recombinant antibody composition
US7994290B2 (en) 2007-01-24 2011-08-09 Kyowa Hakko Kirin Co., Ltd Effector function enhanced recombinant antibody composition
EP2119780A4 (en) * 2007-01-24 2010-08-25 Kyowa Hakko Kirin Co Ltd Genetically recombinant antibody composition having enhanced effector activity
WO2008092117A3 (en) * 2007-01-25 2008-12-31 Xencor Inc Immunoglobulins with modifications in the fcr binding region
WO2008092117A2 (en) * 2007-01-25 2008-07-31 Xencor, Inc. Immunoglobulins with modifications in the fcr binding region
WO2008098115A3 (en) * 2007-02-07 2008-10-02 Xencor Inc Optimized igf-1r antibodies and methods of using the same
WO2008098115A2 (en) * 2007-02-07 2008-08-14 Xencor, Inc. Optimized igf-1r antibodies and methods of using the same
US9688755B2 (en) 2007-05-14 2017-06-27 Novimmune S.A. Fc receptor-binding polypeptides with modified effector functions
US8546539B2 (en) 2007-05-14 2013-10-01 Novimmune S.A. Fc receptor-binding polypeptides with modified effector functions
AU2008260498B2 (en) * 2007-05-30 2012-11-29 Xencor, Inc. Methods and compositions for inhibiting CD32b expressing cells
WO2008150494A1 (en) * 2007-05-30 2008-12-11 Xencor, Inc. Methods and compositions for inhibiting cd32b expressing cells
US9394366B2 (en) 2007-05-30 2016-07-19 Xencor, Inc. Methods and compositions for inhibiting CD32B expressing cells
US9260523B2 (en) 2007-05-30 2016-02-16 Xencor, Inc. Methods and compositions for inhibiting CD32b expressing cells
US11434295B2 (en) 2007-05-30 2022-09-06 Xencor, Inc. Methods and compositions for inhibiting CD32B expressing cells
US11447552B2 (en) 2007-05-30 2022-09-20 Xencor, Inc. Methods and compositions for inhibiting CD32B expressing cells
US9079960B2 (en) 2007-05-30 2015-07-14 Xencor, Inc. Methods and compositions for inhibiting CD32B expressing cells
US9902773B2 (en) 2007-05-30 2018-02-27 Xencor, Inc. Methods and compositions for inhibiting CD32b expressing cells
US9914778B2 (en) 2007-05-30 2018-03-13 Xencor, Inc. Methods and compositions for inhibiting CD32B expressing cells
EP3392273A1 (en) * 2007-05-30 2018-10-24 Xencor, Inc. Methods and compositions for inhibiting cd32b expressing cells
US8063187B2 (en) 2007-05-30 2011-11-22 Xencor, Inc. Methods and compositions for inhibiting CD32B expressing cells
EP2708557A1 (en) * 2007-05-30 2014-03-19 Xencor, Inc. Method and compositions for inhibiting CD32B expressing cells
WO2009016164A1 (en) * 2007-08-01 2009-02-05 Glaxo Group Limited Novel antibodies
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
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
US11932685B2 (en) 2007-10-31 2024-03-19 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
US11359194B2 (en) 2008-04-11 2022-06-14 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding 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
WO2009129538A2 (en) * 2008-04-18 2009-10-22 Xencor, Inc. Human equivalent monoclonal antibodies engineered from nonhuman variable regions
WO2009129538A3 (en) * 2008-04-18 2010-07-01 Xencor, Inc. Human equivalent monoclonal antibodies engineered from nonhuman variable regions
US8314213B2 (en) 2008-04-18 2012-11-20 Xencor, Inc. Human equivalent monoclonal antibodies engineered from nonhuman variable regions
US9017683B2 (en) 2009-05-01 2015-04-28 The University Of Tokyo Highly effective anti-cadherin antibody for induction of antibody-dependent cellular cytotoxicity in vivo
WO2010146059A2 (en) 2009-06-16 2010-12-23 F. Hoffmann-La Roche Ag Biomarkers for igf-1r inhibitor therapy
US11401348B2 (en) 2009-09-02 2022-08-02 Xencor, Inc. Heterodimeric Fc variants
US8668910B2 (en) 2009-10-02 2014-03-11 Sanofi Antibodies that specifically bind to the EphA2 receptor
US9676864B2 (en) 2009-10-02 2017-06-13 Sanofi Antibodies that specifically bind to the EphA2 receptor
WO2011039724A1 (en) 2009-10-02 2011-04-07 Sanofi-Aventis Antibodies that specifically bind to the epha2 receptor
WO2011091078A3 (en) * 2010-01-19 2011-12-01 Xencor, Inc. Antibody fc variants with enhanced complement activity
US8362210B2 (en) 2010-01-19 2013-01-29 Xencor, Inc. Antibody variants with enhanced complement activity
US9475881B2 (en) 2010-01-19 2016-10-25 Xencor, Inc. Antibody variants with enhanced complement activity
WO2011100403A1 (en) 2010-02-10 2011-08-18 Immunogen, Inc Cd20 antibodies and uses thereof
US8975376B2 (en) 2010-02-23 2015-03-10 Sanofi Anti-alpha2 integrin antibodies and their uses
US11267891B2 (en) 2010-02-23 2022-03-08 Sanofi Fc variant antibodies and their uses
WO2011108502A1 (en) 2010-03-02 2011-09-09 協和発酵キリン株式会社 Modified antibody composition
EP3181581A1 (en) 2010-03-30 2017-06-21 Chugai Seiyaku Kabushiki Kaisha Antibodies with modified affinity to fcrn that promote antigen clearance
EP3702368A1 (en) 2010-03-30 2020-09-02 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecules that promote antigen clearance
WO2011122011A2 (en) 2010-03-30 2011-10-06 Chugai Seiyaku Kabushiki Kaisha Antibodies with modified affinity to fcrn that promote antigen clearance
US10858686B2 (en) 2010-05-27 2020-12-08 Merck Sharp & Dohme Corp. Method for preparing antibodies having improved properties
US11959118B2 (en) 2010-05-27 2024-04-16 Merck Sharp & Dohme Llc Fc-containing polypeptides having improved properties and comprising mutations at positions 243 and 264 of the Fc-region
US9234039B2 (en) 2010-08-31 2016-01-12 Sanofi Peptide or peptide complex binding to ALPHA2 integrin and methods and uses involving the same
WO2012032080A1 (en) 2010-09-07 2012-03-15 F-Star Biotechnologische Forschungs- Und Entwicklungsges.M.B.H Stabilised human fc
US9085621B2 (en) 2010-09-10 2015-07-21 Apexigen, Inc. Anti-IL-1β antibodies
US11891434B2 (en) 2010-11-30 2024-02-06 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding to plurality of antigen molecules repeatedly
EP4231014A2 (en) 2010-11-30 2023-08-23 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding to plurality of antigen molecules repeatedly
WO2012073992A1 (en) 2010-11-30 2012-06-07 中外製薬株式会社 Antigen-binding molecule capable of binding to plurality of antigen molecules repeatedly
US11066483B2 (en) 2010-11-30 2021-07-20 Chugai Seiyaku Kabushiki Kaisha Cytotoxicity-inducing therapeutic agent
EP2647707B1 (en) 2010-11-30 2018-09-12 Chugai Seiyaku Kabushiki Kaisha Cytotoxicity-inducing therapeutic agent
US11718678B2 (en) 2011-02-25 2023-08-08 Chugai Seiyaku Kabushiki Kaisha Method for altering plasma retention and immunogenicity of antigen-binding molecule
US10618965B2 (en) 2011-02-25 2020-04-14 Chugai Seiyaku Kabushiki Kaisha Method for altering plasma retention and immunogenicity of antigen-binding molecule
WO2012118813A2 (en) 2011-03-03 2012-09-07 Apexigen, Inc. Anti-il-6 receptor antibodies and methods of use
WO2012122513A2 (en) 2011-03-10 2012-09-13 Omeros Corporation Generation of anti-fn14 monoclonal antibodies by ex-vivo accelerated antibody evolution
US9346888B2 (en) 2011-03-10 2016-05-24 Omeros Corporation Generation of anti-FN14 monoclonal antibodies by ex-vivo accelerated antibody evolution
WO2012133782A1 (en) 2011-03-30 2012-10-04 中外製薬株式会社 Retention of antigen-binding molecules in blood plasma and method for modifying immunogenicity
EP3825325A2 (en) 2011-03-30 2021-05-26 Chugai Seiyaku Kabushiki Kaisha Retention of antigen-binding molecules in blood plasma and method for modifying immunogenicity
EP3508500A1 (en) 2011-04-29 2019-07-10 Apexigen, Inc. Anti-cd40 antibodies and methods of use
WO2012149356A2 (en) 2011-04-29 2012-11-01 Apexigen, Inc. Anti-cd40 antibodies and methods of use
US9102724B2 (en) 2011-08-12 2015-08-11 Omeros Corporation Anti-FZD10 monoclonal antibodies and methods for their use
US9777065B2 (en) 2011-08-12 2017-10-03 Omeros Corporation Anti-FZD10 monoclonal antibodies and methods for their use
EP3392274A1 (en) 2011-08-12 2018-10-24 Omeros Corporation Anti-fzd10 monoclonal antibodies and methods for their use
EP3783028A1 (en) 2011-08-12 2021-02-24 Omeros Corporation Anti-fzd10 monoclonal antibodies and methods for their use
US11186638B2 (en) 2011-09-12 2021-11-30 Genzyme Corporation Anti-αβTCR antibody
EP3939996A1 (en) 2011-09-30 2022-01-19 Chugai Seiyaku Kabushiki Kaisha 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
EP3549956A2 (en) 2011-09-30 2019-10-09 Chugai Seiyaku Kabushiki Kaisha Therapeutic antigen-binding molecule with a fcrn-binding domain that promotes antigen clearance
WO2013046722A1 (en) 2011-09-30 2013-04-04 中外製薬株式会社 Ion concentration-dependent binding molecule library
EP4324850A2 (en) 2011-09-30 2024-02-21 Chugai Seiyaku Kabushiki Kaisha Therapeutic antigen-binding molecule with a fcrn-binding domain that promotes antigen clearance
WO2013047729A1 (en) 2011-09-30 2013-04-04 中外製薬株式会社 Antigen-binding molecule inducing immune response to target antigen
WO2013046704A2 (en) 2011-09-30 2013-04-04 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
EP3680251A1 (en) 2011-09-30 2020-07-15 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecules for promoting elimination 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
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
WO2013067098A1 (en) 2011-11-02 2013-05-10 Apexigen, Inc. Anti-kdr antibodies and methods of use
US11820793B2 (en) 2011-11-30 2023-11-21 Chugai Seiyaku Kabushiki Kaisha Drug containing carrier into cell for forming immune complex
WO2013081143A1 (en) 2011-11-30 2013-06-06 中外製薬株式会社 Drug containing carrier into cell for forming immune complex
EP3517550A1 (en) 2011-11-30 2019-07-31 Chugai Seiyaku Kabushiki Kaisha Drug containing carrier into cell for forming immune complex
WO2013087789A1 (en) * 2011-12-13 2013-06-20 Glykos Finland Ltd. Antibody isoform arrays and methods thereof
WO2013093809A1 (en) 2011-12-23 2013-06-27 Pfizer Inc. Engineered antibody constant regions for site-specific conjugation and methods and uses therefor
WO2013100120A1 (en) 2011-12-28 2013-07-04 中外製薬株式会社 Humanized anti-epiregulin antibody, and cancer therapeutic agent comprising said antibody as active ingredient
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
WO2013147153A1 (en) 2012-03-29 2013-10-03 株式会社未来創薬研究所 Anti-lamp5 antibody and utilization thereof
WO2013180200A1 (en) 2012-05-30 2013-12-05 中外製薬株式会社 Target-tissue-specific antigen-binding molecule
EP3892638A1 (en) 2012-05-30 2021-10-13 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule for eliminating aggregated antigens
WO2013180201A1 (en) 2012-05-30 2013-12-05 中外製薬株式会社 Antigen-binding molecule for eliminating aggregated antigens
EP3795215A1 (en) 2012-05-30 2021-03-24 Chugai Seiyaku Kabushiki Kaisha Target tissue-specific antigen-binding molecule
US10919953B2 (en) 2012-08-24 2021-02-16 Chugai Seiyaku Kabushiki Kaisha FcgammaRIIB-specific Fc region variant
US11236168B2 (en) 2012-08-24 2022-02-01 Chugai Seiyaku Kabushiki Kaisha Mouse FcγammaRII-specific Fc antibody
US10836813B2 (en) 2012-09-12 2020-11-17 Genzyme Corporation Fc containing polypeptides with altered glycosylation and reduced effector function
US10358492B2 (en) 2012-09-27 2019-07-23 Merus N.V. Bispecific IgG antibodies as T cell engagers
EP3925977A1 (en) 2012-10-30 2021-12-22 Apexigen, Inc. Anti-cd40 antibodies and methods of use
WO2014072876A1 (en) 2012-11-09 2014-05-15 Pfizer Inc. Platelet-derived growth factor b specific antibodies and compositions and uses thereof
WO2014097648A1 (en) 2012-12-21 2014-06-26 中外製薬株式会社 Gpc3-targeted therapeutic agent for administration to patients for whom gpc3-targeted therapeutic agent therapy is effective
EP3557260A1 (en) 2012-12-21 2019-10-23 Chugai Seiyaku Kabushiki Kaisha Gpc3-targeting drug which is administered to patient responsive to gpc3-targeting drug therapy
EP4119947A1 (en) 2012-12-21 2023-01-18 Chugai Seiyaku Kabushiki Kaisha Gpc3-targeting drug which is administered to patient responsive to gpc3-targeting drug therapy
US11130816B2 (en) 2013-03-11 2021-09-28 Genzyme Corporation Site-specific antibody-drug conjugation through glycoengineering
US10214589B2 (en) 2013-03-11 2019-02-26 Genzyme Corporation Site-specific antibody-drug conjugation through glycoengineering
US10494439B2 (en) 2013-03-11 2019-12-03 Genzyme Corporation Hyperglycosylated binding polypeptides
US11807690B2 (en) 2013-03-11 2023-11-07 Genzyme Corporation Hyperglycosylated binding polypeptides
US11267868B2 (en) 2013-04-02 2022-03-08 Chugai Seiyaku Kabushiki Kaisha Fc region variant
WO2014190441A1 (en) 2013-05-31 2014-12-04 Zymeworks Inc. Heteromultimers with reduced or silenced effector function
WO2014208482A1 (en) 2013-06-24 2014-12-31 中外製薬株式会社 Therapeutic agent comprising humanized anti-epiregulin antibody as active ingredient for non-small-cell lung carcinoma excluding adenocarcinoma
WO2015046467A1 (en) 2013-09-27 2015-04-02 中外製薬株式会社 Method for producing polypeptide heteromultimer
WO2015073580A1 (en) 2013-11-13 2015-05-21 Pfizer Inc. Tumor necrosis factor-like ligand 1a specific antibodies and compositions and uses thereof
WO2015073721A1 (en) 2013-11-13 2015-05-21 Zymeworks Inc. Monovalent antigen binding constructs targeting egfr and/or her2 and uses thereof
WO2015077891A1 (en) 2013-11-27 2015-06-04 Zymeworks Inc. Bispecific antigen-binding constructs targeting her2
EP3763813A1 (en) 2013-12-04 2021-01-13 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecules, the antigen-binding activity of which varies according to the concentration of compounds, and libraries of said molecules
WO2015083764A1 (en) 2013-12-04 2015-06-11 中外製薬株式会社 Antigen-binding molecules, the antigen-binding activity of which varies according to the concentration of compounds, and libraries of said molecules
WO2015109212A1 (en) 2014-01-17 2015-07-23 Pfizer Inc. Anti-il-2 antibodies and compositions and uses thereof
US9803007B1 (en) 2014-03-07 2017-10-31 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
US9371377B2 (en) 2014-03-07 2016-06-21 Alexion Pharmaceuticals, Inc. Anti-C5 antibodies having improved pharmacokinetics
US9079949B1 (en) 2014-03-07 2015-07-14 Alexion Pharmaceuticals, Inc. Anti-C5 antibodies having improved pharmacokinetics
US11434280B2 (en) 2014-03-07 2022-09-06 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
US9206251B2 (en) 2014-03-07 2015-12-08 Alexion Pharmaceuticals, Inc. Nucleic acids encoding 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
US9663574B2 (en) 2014-03-07 2017-05-30 Alexion Pharmaceuticals, Inc. Anti-C5 antibodies having improved pharmacokinetics
US11697690B2 (en) 2014-03-19 2023-07-11 Genzyme Corporation Site-specific glycoengineering of targeting moieties
US11485790B2 (en) 2014-04-07 2022-11-01 Chugai Seiyaku Kabushiki Kaisha Immunoactivating antigen-binding molecule
US11760807B2 (en) 2014-05-08 2023-09-19 Chugai Seiyaku Kabushiki Kaisha GPC3-targeting drug which is administered to patient responsive to GPC3-targeting drug therapy
US11505605B2 (en) 2014-05-13 2022-11-22 Chugai Seiyaku Kabushiki Kaisha T cell-redirected antigen-binding molecule for cells having immunosuppression function
US11001643B2 (en) 2014-09-26 2021-05-11 Chugai Seiyaku Kabushiki Kaisha Cytotoxicity-inducing therapeutic agent
US11160874B2 (en) 2014-10-09 2021-11-02 Genzyme Corporation Glycoengineered antibody drug conjugates
US10064952B2 (en) 2014-10-09 2018-09-04 Genzyme Corporation Glycoengineered antibody drug conjugates
WO2016082044A1 (en) 2014-11-27 2016-06-02 Zymeworks Inc. Methods of using bispecific antigen-binding constructs targeting her2
US11454633B2 (en) 2014-12-19 2022-09-27 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
US10738111B2 (en) 2014-12-19 2020-08-11 Chugai Seiyaku Kabushiki Kaisha Anti-myostatin antibodies, polypeptides containing variant Fc regions, and methods of use
WO2016125495A1 (en) 2015-02-05 2016-08-11 Chugai Seiyaku Kabushiki Kaisha Antibodies comprising an ion concentration dependent antigen-binding domain, fc region variants, il-8-binding antibodies, and uses therof
EP3816179A2 (en) 2015-02-05 2021-05-05 Chugai Seiyaku Kabushiki Kaisha Fc region variant comprising a modified fcrn-binding domain
US10519229B2 (en) 2015-02-05 2019-12-31 Chugai Seiyaku Kabushiki Kaisha Nucleic acids encoding IL-8 antibodies
US11180548B2 (en) 2015-02-05 2021-11-23 Chugai Seiyaku Kabushiki Kaisha Methods of neutralizing IL-8 biological activity
US9969800B2 (en) 2015-02-05 2018-05-15 Chugai Seiyaku Kabushiki Kaisha IL-8 antibodies
US10793616B2 (en) 2015-05-04 2020-10-06 Apogenix Ag Single-chain CD40-receptor agonist proteins
US11739148B2 (en) 2015-07-10 2023-08-29 Merus N.V. Human CD3 binding antibody
US10266593B2 (en) 2015-07-10 2019-04-23 Merus N.V. Human CD3 binding antibody
US9914777B2 (en) 2015-07-10 2018-03-13 Merus N.V. Human CD3 binding antibody
WO2017015619A1 (en) 2015-07-23 2017-01-26 The Regents Of The University Of California Antibodies to coagulation factor xia and uses thereof
WO2017046994A1 (en) 2015-09-18 2017-03-23 Chugai Seiyaku Kabushiki Kaisha Il-8-binding antibodies and uses thereof
WO2017070561A1 (en) 2015-10-23 2017-04-27 Pfizer Inc. Anti-il-2 antibodies and compositions and uses thereof
US11649293B2 (en) 2015-11-18 2023-05-16 Chugai Seiyaku Kabushiki Kaisha Method for enhancing humoral immune response
US11660340B2 (en) 2015-11-18 2023-05-30 Chugai Seiyaku Kabushiki Kaisha Combination therapy using T cell redirection antigen binding molecule against cell having immunosuppressing function
US11359009B2 (en) 2015-12-25 2022-06-14 Chugai Seiyaku Kabushiki Kaisha Anti-myostatin antibodies and methods of use
US11649262B2 (en) 2015-12-28 2023-05-16 Chugai Seiyaku Kabushiki Kaisha Method for promoting efficiency of purification of Fc region-containing polypeptide
US11072666B2 (en) 2016-03-14 2021-07-27 Chugai Seiyaku Kabushiki Kaisha Cell injury inducing therapeutic drug for use in cancer therapy
EP4112641A1 (en) 2016-03-15 2023-01-04 Chugai Seiyaku Kabushiki Kaisha Methods of treating cancers using pd-1 axis binding antagonists and anti-gpc3 antibodies
WO2017159699A1 (en) 2016-03-15 2017-09-21 Chugai Seiyaku Kabushiki Kaisha Methods of treating cancers using pd-1 axis binding antagonists and anti-gpc3 antibodies
US20200123261A1 (en) * 2016-04-01 2020-04-23 Avidity Biosciences, Inc. Nucleic acid-polypeptide compositions and uses thereof
US11365256B2 (en) 2016-06-08 2022-06-21 Xencor, Inc. Methods and compositions for inhibiting CD32B expressing cells in IGG4-related diseases
US11780912B2 (en) 2016-08-05 2023-10-10 Chugai Seiyaku Kabushiki Kaisha Composition for prophylaxis or treatment of IL-8 related diseases
US11053308B2 (en) 2016-08-05 2021-07-06 Chugai Seiyaku Kabushiki Kaisha Method for treating IL-8-related diseases
US10870694B2 (en) 2016-09-02 2020-12-22 Dana Farber Cancer Institute, Inc. Composition and methods of treating B cell disorders
WO2018045379A1 (en) 2016-09-02 2018-03-08 Dana-Farber Cancer Institute, Inc. Composition and methods of treating b cell disorders
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
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
US11673971B2 (en) * 2016-09-23 2023-06-13 Marengo Therapeutics, Inc. Multispecific antibody molecules comprising lambda and kappa light chains
WO2018129029A1 (en) 2017-01-04 2018-07-12 Immunogen, Inc. Met antibodies and immunoconjugates and uses thereof
US11938194B2 (en) 2017-02-28 2024-03-26 Seagen Inc. Cysteine mutated antibodies for conjugation
CN110546162A (en) * 2017-03-28 2019-12-06 礼进生物医药控股有限公司 Therapeutic agents and methods for enhancing immune response in tumor microenvironment
US11365241B2 (en) 2017-07-27 2022-06-21 Alexion Pharmaceuticals, Inc. High concentration anti-C5 antibody formulations
WO2019059411A1 (en) 2017-09-20 2019-03-28 Chugai Seiyaku Kabushiki Kaisha Dosage regimen for combination therapy using pd-1 axis binding antagonists and gpc3 targeting agent
AU2018358138C1 (en) * 2017-11-02 2022-12-08 Baili-Bio (Chengdu) Pharmaceutical Co., Ltd. Bispecific antibodies and methods of making and using thereof
CN111212658A (en) * 2017-11-02 2020-05-29 西雅图免疫公司 Bispecific antibodies and methods of making and using the same
AU2018358138B2 (en) * 2017-11-02 2022-06-02 Baili-Bio (Chengdu) Pharmaceutical Co., Ltd. Bispecific antibodies and methods of making and using thereof
CN111212658B (en) * 2017-11-02 2024-05-03 西雅图免疫公司 Bispecific antibodies and methods of making and using the same
WO2019090002A1 (en) * 2017-11-02 2019-05-09 Systimmune, Inc. Bispecific antibodies and methods of making and using thereof
US11746150B2 (en) 2017-12-19 2023-09-05 Surrozen Operating, Inc. Anti-LRP5/6 antibodies and methods of use
US11773171B2 (en) 2017-12-19 2023-10-03 Surrozen Operating, Inc. WNT surrogate molecules and uses thereof
US11078283B2 (en) 2018-01-17 2021-08-03 Apexigen, Inc. Anti-PD-L1 antibodies and methods of use
EP4253421A2 (en) 2018-03-13 2023-10-04 Zymeworks BC Inc. Anti-her2 biparatopic antibody-drug conjugates and methods of use
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
US11555071B2 (en) 2018-06-03 2023-01-17 Lamkap Bio Beta Ltd. Bispecific antibodies against CEACAM5 and CD47
WO2019234576A1 (en) 2018-06-03 2019-12-12 Lamkap Bio Beta Ltd. Bispecific antibodies against ceacam5 and cd47
WO2019244107A1 (en) 2018-06-21 2019-12-26 Daiichi Sankyo Company, Limited Compositions including cd3 antigen binding fragments and uses thereof
WO2020014306A1 (en) 2018-07-10 2020-01-16 Immunogen, Inc. Met antibodies and immunoconjugates and uses thereof
WO2020189748A1 (en) 2019-03-19 2020-09-24 中外製薬株式会社 Antigen-binding molecule containing antigen-binding domain of which binding activity to antigen is changed depending on mta, and library for obtaining said antigen-binding domain
RU2814713C2 (en) * 2019-04-04 2024-03-04 Оно Фармасьютикал Ко., Лтд. Bispecific antibody
CN113660952A (en) * 2019-04-04 2021-11-16 小野药品工业株式会社 Bispecific antibodies
WO2021110647A1 (en) 2019-12-02 2021-06-10 Lamkap Bio Beta Ag Bispecific antibodies against ceacam5 and cd47
EP3831849A1 (en) 2019-12-02 2021-06-09 LamKap Bio beta AG Bispecific antibodies against ceacam5 and cd47
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
GB2595299B (en) * 2020-05-21 2022-08-03 Mabsolve Ltd Modified immunoglobulin FC regions
GB2595299A (en) * 2020-05-21 2021-11-24 Mabsolve Ltd Modified immunoglobulin FC regions
WO2022130348A1 (en) 2020-12-18 2022-06-23 Lamkap Bio Beta Ag Bispecific antibodies against ceacam5 and cd47
US11753481B2 (en) 2020-12-18 2023-09-12 Lamkap Bio Beta Ltd Bispecific antibodies against CEACAM5 and CD47
WO2022263501A1 (en) 2021-06-18 2022-12-22 F. Hoffmann-La Roche Ag Bispecific anti-ccl2 antibodies
WO2023242351A1 (en) 2022-06-16 2023-12-21 Lamkap Bio Beta Ag Combination therapy of bispecific antibodies against ceacam5 and cd47 and bispecific antibodies against ceacam5 and cd3

Also Published As

Publication number Publication date
IL186145A0 (en) 2008-01-20
AU2006230413B2 (en) 2010-09-23
AU2006230413B8 (en) 2011-01-20
EP1871808A2 (en) 2008-01-02
WO2006105338A3 (en) 2007-03-29
AU2006230413A1 (en) 2006-10-05
CA2602663A1 (en) 2006-10-05

Similar Documents

Publication Publication Date Title
AU2006230413B8 (en) Fc variants with optimized properties
US9040041B2 (en) Modified FC molecules
US20080242845A1 (en) Fc variants with optimized properties
AU2016234983B2 (en) Fc variants with altered binding to FcRn
KR101573883B1 (en) Fc VARIANTS WITH ALTERED BINDING TO FcRn
US20060134105A1 (en) IgG immunoglobulin variants with optimized effector function
JP6310394B2 (en) Methods for purifying antibodies
AU2011200066B2 (en) Fc variants with altered binding to FcRn
EP3029066B1 (en) Antibodies with modified isoelectric points
US20070275460A1 (en) Fc Variants With Optimized Fc Receptor Binding Properties
JP2008537941A (en) Fc variants with optimized properties
WO2006047350A2 (en) IgG IMMUNOGLOBULIN VARIANTS WITH OPTIMIZED EFFECTOR FUNCTION
US20100093979A1 (en) Fc Polypeptides With Novel Fc Ligand Binding Sites
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: 2006748967

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 1452/MUMNP/2007

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 186145

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 2006230413

Country of ref document: AU

ENP Entry into the national phase

Ref document number: 2602663

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2008504389

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2006230413

Country of ref document: AU

Date of ref document: 20060330

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: RU

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