US20100256340A1 - Trivalent, bispecific antibodies - Google Patents

Trivalent, bispecific antibodies Download PDF

Info

Publication number
US20100256340A1
US20100256340A1 US12/752,216 US75221610A US2010256340A1 US 20100256340 A1 US20100256340 A1 US 20100256340A1 US 75221610 A US75221610 A US 75221610A US 2010256340 A1 US2010256340 A1 US 2010256340A1
Authority
US
United States
Prior art keywords
antibody
domain
trivalent
chain variable
variable domain
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US12/752,216
Inventor
Ulrich Brinkmann
Rebecca Croasdale
Eike Hoffmann
Christian Klein
Ekkehard Moessner
Juergen Michael Schanzer
Claudio Sustmann
Pablo Umana
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
F Hoffmann La Roche AG
Roche Glycart AG
Original Assignee
F Hoffmann La Roche AG
Roche Glycart AG
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by F Hoffmann La Roche AG, Roche Glycart AG filed Critical F Hoffmann La Roche AG
Assigned to ROCHE GLYCART AG reassignment ROCHE GLYCART AG ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: MOESSNER, EKKEHARD, UMANA, PABLO
Assigned to ROCHE GLYCART AG reassignment ROCHE GLYCART AG ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: F. HOFFMANN-LA ROCHE AG
Assigned to F. HOFFMANN-LA ROCHE AG reassignment F. HOFFMANN-LA ROCHE AG ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BRINKMANN, ULRICH, CROASDALE, REBECCA, HOFFMANN, EIKE, SCHANZER, JUERGEN MICHAEL, SUSTMANN, CLAUDIO
Assigned to F. HOFFMANN-LA ROCHE AG reassignment F. HOFFMANN-LA ROCHE AG ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: KLEIN, CHRISTIAN
Publication of US20100256340A1 publication Critical patent/US20100256340A1/en
Priority to US13/568,224 priority Critical patent/US9890204B2/en
Priority to US15/857,473 priority patent/US20180282399A1/en
Priority to US16/413,469 priority patent/US11993642B2/en
Abandoned legal-status Critical Current

Links

Images

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
    • C07K16/46Hybrid immunoglobulins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2863Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for growth factors, growth regulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/32Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against translation products of oncogenes
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/31Immunoglobulins specific features characterized by aspects of specificity or valency multispecific
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide

Definitions

  • the present invention relates to trivalent, bispecific antibodies, methods for their production, pharmaceutical compositions containing the antibodies, and uses thereof.
  • a wide variety of multispecific recombinant antibody formats have been developed in the recent past, e.g. tetravalent bispecific antibodies by fusion of, e.g., an IgG antibody format and single chain domains (see e.g. Coloma, M. J., et al., Nature Biotech 15 (1997) 159-163; WO 2001/077342; and Morrison, S. L., Nature Biotech 25 (2007) 1233-1234).
  • All such formats use linkers either to fuse the antibody core (IgA, IgD, IgE, IgG or IgM) to a further binding protein (e.g. scFv) or to fuse e.g. two Fab fragments or scFvs (Fischer, N., and Léger, O., Pathobiology 74 (2007) 3-14). It has to be kept in mind that one may want to retain effector functions, such as e.g. complement-dependent cytotoxicity (CDC) or antibody dependent cellular cytotoxicity (ADCC), which are mediated through the Fc receptor binding, by maintaining a high degree of similarity to naturally occurring antibodies.
  • CDC complement-dependent cytotoxicity
  • ADCC antibody dependent cellular cytotoxicity
  • WO 2007/024715 are reported dual variable domain immunoglobulins as engineered multivalent and multispecific binding proteins.
  • a process for the preparation of biologically active antibody dimers is reported in U.S. Pat. No. 6,897,044.
  • Multivalent F V antibody construct having at least four variable domains which are linked with each over via peptide linkers are reported in U.S. Pat. No. 7,129,330.
  • Dimeric and multimeric antigen binding structures are reported in US 2005/0079170.
  • Tri- or tetra-valent monospecific antigen-binding protein comprising three or four Fab fragments bound to each other covalently by a connecting structure, which protein is not a natural immunoglobulin are reported in U.S. Pat. No. 6,511,663.
  • bispecific antibodies are reported that can be efficiently expressed in prokaryotic and eukaryotic cells, and are useful in therapeutic and diagnostic methods.
  • a method of separating or preferentially synthesizing dimers which are linked via at least one interchain disulfide linkage from dimers which are not linked via at least one interchain disulfide linkage from a mixture comprising the two types of polypeptide dimers is reported in US 2005/0163782.
  • Bispecific tetravalent receptors are reported in U.S. Pat. No. 5,959,083.
  • Engineered antibodies with three or more functional antigen binding sites are reported in WO 2001/077342.
  • Multispecific and multivalent antigen-binding polypeptides are reported in WO 1997/001580.
  • WO 1992/004053 reports homoconjugates, typically prepared from monoclonal antibodies of the IgG class which bind to the same antigenic determinant are covalently linked by synthetic cross-linking
  • Oligomeric monoclonal antibodies with high avidity for antigen are reported in WO 1991/06305 whereby the oligomers, typically of the IgG class, are secreted having two or more immunoglobulin monomers associated together to form tetravalent or hexavalent IgG molecules.
  • Sheep-derived antibodies and engineered antibody constructs are reported in U.S. Pat. No.
  • a first aspect of the current invention is a trivalent, bispecific antibody comprising
  • a further aspect of the invention is a nucleic acid molecule encoding a trivalent, bispecific antibody according to the invention.
  • Still further aspects of the invention are a pharmaceutical composition comprising the trivalent, bispecific antibody.
  • the trivalent, bispecific antibodies according to the invention one the one hand show new properties due to their binding to different antigens, and on the other hand are suitable for production and pharmaceutical formulation due to their stability, low aggregation and pharmacokinetic and biological properties. Due to their Ig core they still retain the properties of natural antibodies like ADCC and CDC.
  • One aspect of the invention is trivalent, bispecific antibody comprising
  • the antibody heavy chain variable domain (VH) of the polypeptide under b) and the antibody light chain variable domain (VL) of the polypeptide under c) are linked and stabilized via a interchain disulfide bridge by introduction of a disulfide bond between the following positions:
  • heavy chain variable domain position 44 to light chain variable domain position 100
  • heavy chain variable domain position 105 to light chain variable domain position 43
  • heavy chain variable domain position 101 to light chain variable domain position 100 (numbering always according to EU index of Kabat).
  • the optional disulfide bond between the variable domains of the polypeptides under b) and c) is between heavy chain variable domain position 105 and light chain variable domain position 43. (numbering always according to EU index of Kabat)
  • a trivalent, bispecific antibody without the optional disulfide stabilization between the variable domains VH and VL of the single chain Fab fragments is preferred.
  • full length antibody denotes an antibody consisting of two “full length antibody heavy chains” and two “full length antibody light chains” (see FIG. 1 ).
  • a “full length antibody heavy chain” is a polypeptide consisting in N-terminal to C-terminal direction of an antibody heavy chain variable domain (VH), an antibody constant heavy chain domain 1 (CH1), an antibody hinge region (HR), an antibody heavy chain constant domain 2 (CH2), and an antibody heavy chain constant domain 3 (CH3), abbreviated as VH-CH1-HR-CH2-CH3; and optionally an antibody heavy chain constant domain 4 (CH4) in case of an antibody of the subclass IgE.
  • VH antibody heavy chain variable domain
  • CH1 antibody constant heavy chain domain 1
  • HR antibody hinge region
  • CH2 antibody heavy chain constant domain 2
  • CH3 antibody heavy chain constant domain 3
  • the “full length antibody heavy chain” is a polypeptide consisting in N-terminal to C-terminal direction of VH, CH1, HR, CH2 and CH3.
  • a “full length antibody light chain” is a polypeptide consisting in N-terminal to C-terminal direction of an antibody light chain variable domain (VL), and an antibody light chain constant domain (CL), abbreviated as VL-CL.
  • the antibody light chain constant domain (CL) can be ⁇ (kappa) or ⁇ (lambda).
  • the two full length antibody chains are linked together via inter-polypeptide disulfide bonds between the CL domain and the CH1 domain and between the hinge regions of the full length antibody heavy chains. Examples of typical full length antibodies are natural antibodies like IgG (e.g.
  • the full length antibodies according to the invention can be from a single species e.g. human, or they can be chimerized or humanized antibodies.
  • the full length antibodies according to the invention comprise two antigen binding sites each formed by a pair of VH and VL, which both specifically bind to the same antigen.
  • the C-terminus of the heavy or light chain of the full length antibody denotes the last amino acid at the C-terminus of the heavy or light chain.
  • the N-terminus of the antibody heavy chain variable domain (VH) of the polypeptide under b) and the antibody light chain variable domain (VL) of the polypeptide under c) denotes the last amino acid at the N-terminus of VH or VL domain.
  • the CH3 domains of the full length antibody according to the invention can be altered by the “knob-into-holes” technology which is described in detail with several examples in e.g. WO 96/027011, Ridgway, J. B., et al., Protein Eng 9 (1996) 617-621; and Merchant, A. M., et al., Nat Biotechnol 16 (1998) 677-681.
  • this method the interaction surfaces of the two CH3 domains are altered to increase the heterodimerisation of both heavy chains containing these two CH3 domains.
  • Each of the two CH3 domains (of the two heavy chains) can be the “knob”, while the other is the “hole”.
  • the trivalent, bispecific antibody is further is characterized in that the CH3 domain of one heavy chain of the full length antibody and the CH3 domain of the other heavy chain of the full length antibody each meet at an interface which comprises an original interface between the antibody CH3 domains;
  • the interface is altered to promote the formation of the bivalent, bispecific antibody, wherein the alteration is characterized in that:
  • an amino acid residue is replaced with an amino acid residue having a larger side chain volume, thereby generating a protuberance within the interface of the CH3 domain of one heavy chain which is positionable in a cavity within the interface of the CH3 domain of the other heavy chain and
  • amino acid residue having a larger side chain volume is selected from the group consisting of arginine (R), phenylalanine (F), tyrosine (Y), tryptophan (W).
  • amino acid residue having a smaller side chain volume is selected from the group consisting of alanine (A), serine (S), threonine (T), valine (V).
  • both CH3 domains are further altered by the introduction of cysteine (C) as amino acid in the corresponding positions of each CH3 domain such that a disulfide bridge between both CH3 domains can be formed.
  • C cysteine
  • the trivalent, bispecific comprises a T366W mutation in the CH3 domain of the “knobs chain” and T366S, L368A, Y407V mutations in the CH3 domain of the “hole chain”.
  • An additional interchain disulfide bridge between the CH3 domains can also be used (Merchant, A. M., et al., Nature Biotech 16 (1998) 677-681) e.g. by introducing a Y349C mutation into the CH3 domain of the “knobs chain” and a E356C mutation or a S354C mutation into the CH3 domain of the “hole chain”.
  • the trivalent, bispecific antibody comprises Y349C, T366W mutations in one of the two CH3 domains and E356C, T366S, L368A, Y407V mutations in the other of the two CH3 domains or the trivalent, bispecific antibody comprises Y349C, T366W mutations in one of the two CH3 domains and S354C, T366S, L368A, Y407V mutations in the other of the two CH3 domains (the additional Y349C mutation in one CH3 domain and the additional E356C or S354C mutation in the other CH3 domain forming a interchain disulfide bridge) (numbering always according to EU index of Kabat).
  • knobs-in-holes technologies as described by EP 1 870 459A1, can be used alternatively or additionally.
  • a preferred example for the trivalent, bispecific antibody are R409D; K370E mutations in the CH3 domain of the “knobs chain” and D399K; E357K mutations in the CH3 domain of the “hole chain” (numbering always according to EU index of Kabat).
  • the trivalent, bispecific antibody comprises a T366W mutation in the CH3 domain of the “knobs chain” and T366S, L368A, Y407V mutations in the CH3 domain of the “hole chain” and additionally R409D; K370E mutations in the CH3 domain of the “knobs chain” and D399K; E357K mutations in the CH3 domain of the “hole chain”.
  • the trivalent, bispecific antibody comprises Y349C, T366W mutations in one of the two CH3 domains and S354C, T366S, L368A, Y407V mutations in the other of the two CH3 domains or the trivalent, bispecific antibody comprises Y349C, T366W mutations in one of the two CH3 domains and S354C, T366S, L368A, Y407V mutations in the other of the two CH3 domains and additionally R409D; K370E mutations in the CH3 domain of the “knobs chain” and D399K; E357K mutations in the CH3 domain of the “hole chain”.
  • the bispecific antibody to the invention comprises three antigen-binding sites (A) the full length antibody according comprises two identical antigen-binding sites specifically binding to a first antigen, and B) the antibody heavy chain variable domain (VH) of the polypeptide under b) and the antibody light chain variable domain (VL) of the polypeptide under c) form together one antigen binding site specifically binding to a second antigen).
  • binding site or “antigen-binding site” as used herein denotes the region(s) of the bispecific antibody according to the invention to which the respective antigen actually specifically binds.
  • the antigen binding sites either in the full length antibody or by the antibody heavy chain variable domain (VH) of the polypeptide under b) and the antibody light chain variable domain (VL) of the polypeptide under c) are formed each by a pair consisting of an antibody light chain variable domain (VL) and an antibody heavy chain variable domain (VH).
  • the antigen-binding sites that specifically bind to the desired antigen can be derived a) from known antibodies to the antigen or b) from new antibodies or antibody fragments obtained by de novo immunization methods using inter alia either the antigen protein or nucleic acid or fragments thereof or by phage display.
  • An antigen-binding site of an antibody of the invention contains six complementarity determining regions (CDRs) which contribute in varying degrees to the affinity of the binding site for antigen. There are three heavy chain variable domain CDRs (CDRH1, CDRH2 and CDRH3) and three light chain variable domain CDRs (CDRL1, CDRL2 and CDRL3). The extent of CDR and framework regions (FRs) is determined by comparison to a compiled database of amino acid sequences in which those regions have been defined according to variability among the sequences.
  • Antibody specificity refers to selective recognition of the antibody for a particular epitope of an antigen.
  • Natural antibodies for example, are monospecific.
  • “Bispecific antibodies” according to the invention are antibodies which have two different antigen-binding specificities. Where an antibody has more than one specificity, the recognized epitopes may be associated with a single antigen or with more than one antigen.
  • the term “monospecific” antibody as used herein denotes an antibody that has one or more binding sites each of which bind to the same epitope of the same antigen.
  • the term “valent” as used within the current application denotes the presence of a specified number of binding sites in an antibody molecule.
  • a natural antibody for example or a full length antibody according to the invention has two binding sites and is bivalent.
  • trivalent denote the presence of three binding sites in an antibody molecule.
  • the bispecific antibodies according to the invention are “trivalent”.
  • the term “trivalent, bispecific” antibody as used herein denotes an antibody that has three antigen-binding sites of which two bind to the same antigen (or the same epitope of the antigen) and the third binds to a different antigen or a different epitope of the same antigen.
  • Antibodies of the present invention have three binding sites and are bispecific.
  • Another embodiment of the current invention is a trivalent, bispecific antibody comprising:
  • a full length antibody specifically binding to a first antigen consisting of: aa) two antibody heavy chains consisting in N-terminal to C-terminal direction of an antibody heavy chain variable domain (VH), an antibody constant heavy chain domain 1 (CH1), an antibody hinge region (HR), an antibody heavy chain constant domain 2 (CH2), and an antibody heavy chain constant domain 3 (CH3); and ab) two antibody light chains consisting in N-terminal to C-terminal direction of an antibody light chain variable domain (VL), and an antibody light chain constant domain (CL); and b) a polypeptide consisting of ba) an antibody heavy chain variable domain (VH); or bb) an antibody heavy chain variable domain (VH) and an antibody constant domain 1 (CH1), wherein the polypeptide is fused with the N-terminus of the VH domain via a peptide connector to the C-terminus of one of the two heavy chains of the full length antibody wherein the peptide connector is a peptide of at least 5 amino acids, preferably between 25 and 50
  • the trivalent, bispecific antibody comprises a T366W mutation in one of the two CH3 domains of and T366S, L368A, Y407V mutations in the other of the two CH3 domains and more preferably the trivalent, bispecific antibody comprises Y349C, T366W mutations in one of the two CH3 domains of and D356C, T366S, L368A, Y407V mutations in the other of the two CH3 domains (the additional Y349C mutation in one CH3 domain and the additional D356C mutation in the other CH3 domain forming a interchain disulfide bridge).
  • the trivalent, bispecific antibody according to the invention is characterized in that
  • the full length antibody is specifically binding to ErbB-3 comprises as heavy chain variable domain the sequence of SEQ ID NO: 1, and as light chain variable domain the sequence of SEQ ID NO: 2
  • the polypeptide under b) comprises as the heavy chain variable domain the sequence of SEQ ID NO: 3
  • the polypeptide under c) comprises as the light chain variable domain the sequence of SEQ ID NO: 4.
  • the trivalent, bispecific antibody according to the invention comprises
  • Another embodiment of the current invention is a trivalent, bispecific antibody comprising
  • a full length antibody specifically binding to human ErbB-3 consisting of: aa) two antibody heavy chains consisting in N-terminal to C-terminal direction of an antibody heavy chain variable domain (VH), an antibody constant heavy chain domain 1 (CH1), an antibody hinge region (HR), an antibody heavy chain constant domain 2 (CH2), and an antibody heavy chain constant domain 3 (CH3); and ab) two antibody light chains consisting in N-terminal to C-terminal direction of an antibody light chain variable domain (VL), and an antibody light chain constant domain (CL) (VL-CL); and b) one single chain Fv fragment specifically binding to human c-Met), wherein the single chain Fv fragment under b) is fused to the full length antibody under a) via a peptide connector at the C- or N-terminus of the heavy or light chain (preferably at the C-terminus of the heavy chain) of the full length antibody; wherein the peptide connector is a peptide of at least 5 amino acids, preferably between 25 and 50 amino acids.
  • such trivalent, bispecific antibody further comprises Y349C, T366W mutations in one of the two CH3 domains of the full length antibody and S354C (or E356C), T366S, L368A, Y407V mutations in the other of the two CH3 domains of the full length antibody.
  • Another embodiment of the current invention is a trivalent, bispecific antibody comprising
  • the full length antibodies of the invention comprise immunoglobulin constant regions of one or more immunoglobulin classes.
  • Immunoglobulin classes include IgG, IgM, IgA, IgD, and IgE isotypes and, in the case of IgG and IgA, their subtypes.
  • a full length antibody of the invention has a constant domain structure of an IgG type antibody.
  • monoclonal antibody or “monoclonal antibody composition” as used herein refer to a preparation of antibody molecules of a single amino acid composition.
  • chimeric antibody refers to an antibody comprising a variable region, i.e., binding region, from one source or species and at least a portion of a constant region derived from a different source or species, usually prepared by recombinant DNA techniques. Chimeric antibodies comprising a murine variable region and a human constant region are preferred. Other preferred forms of “chimeric antibodies” encompassed by the present invention are those in which the constant region has been modified or changed from that of the original antibody to generate the properties according to the invention, especially in regard to C1q binding and/or Fc receptor (FcR) binding. Such chimeric antibodies are also referred to as “class-switched antibodies.”.
  • Chimeric antibodies are the product of expressed immunoglobulin genes comprising DNA segments encoding immunoglobulin variable regions and DNA segments encoding immunoglobulin constant regions. Methods for producing chimeric antibodies involve conventional recombinant DNA and gene transfection techniques are well known in the art. See, e.g., Morrison, S. L., et al., Proc. Natl. Acad. Sci. USA 81 (1984) 6851-6855; U.S. Pat. No. 5,202,238 and U.S. Pat. No. 5,204,244.
  • humanized antibody refers to antibodies in which the framework or “complementarity determining regions” (CDR) have been modified to comprise the CDR of an immunoglobulin of different specificity as compared to that of the parent immunoglobulin.
  • CDR complementarity determining regions
  • a murine CDR is grafted into the framework region of a human antibody to prepare the “humanized antibody.” See, e.g., Riechmann, L., et al., Nature 332 (1988) 323-327; and Neuberger, M. S., et al., Nature 314 (1985) 268-270.
  • Particularly preferred CDRs correspond to those representing sequences recognizing the antigens noted above for chimeric antibodies.
  • humanized antibodies encompassed by the present invention are those in which the constant region has been additionally modified or changed from that of the original antibody to generate the properties according to the invention, especially in regard to C1q binding and/or Fc receptor (FcR) binding.
  • FcR Fc receptor
  • human antibody is intended to include antibodies having variable and constant regions derived from human germ line immunoglobulin sequences.
  • Human antibodies are well-known in the state of the art (van Dijk, M. A., and van de Winkel, J. G., Curr. Opin. Chem. Biol. 5 (2001) 368-374).
  • Human antibodies can also be produced in transgenic animals (e.g., mice) that are capable, upon immunization, of producing a full repertoire or a selection of human antibodies in the absence of endogenous immunoglobulin production.
  • Human antibodies can also be produced in phage display libraries (Hoogenboom, H. R., and Winter, G. J., Mol. Biol. 227 (1992) 381-388; Marks, J.
  • human antibody as used herein also comprises such antibodies which are modified in the constant region to generate the properties according to the invention, especially in regard to C1q binding and/or FcR binding, e.g. by “class switching” i.e. change or mutation of Fc parts (e.g. from IgG1 to IgG4 and/or IgG1/IgG4 mutation.)
  • recombinant human antibody is intended to include all human antibodies that are prepared, expressed, created or isolated by recombinant means, such as antibodies isolated from a host cell such as a NS0 or CHO cell or from an animal (e.g. a mouse) that is transgenic for human immunoglobulin genes or antibodies expressed using a recombinant expression vector transfected into a host cell.
  • recombinant human antibodies have variable and constant regions in a rearranged form.
  • the recombinant human antibodies according to the invention have been subjected to in vivo somatic hypermutation.
  • the amino acid sequences of the VH and VL regions of the recombinant antibodies are sequences that, while derived from and related to human germ line VH and VL sequences, may not naturally exist within the human antibody germ line repertoire in vivo.
  • variable domain (variable domain of a light chain (VL), variable region of a heavy chain (VH) as used herein denotes each of the pair of light and heavy chains which is involved directly in binding the antibody to the antigen.
  • the domains of variable human light and heavy chains have the same general structure and each domain comprises four framework (FR) regions whose sequences are widely conserved, connected by three “hypervariable regions” (or complementarity determining regions, CDRs).
  • the framework regions adopt a 13-sheet conformation and the CDRs may form loops connecting the 13-sheet structure.
  • the CDRs in each chain are held in their three-dimensional structure by the framework regions and form together with the CDRs from the other chain the antigen binding site.
  • the antibody heavy and light chain CDR3 regions play a particularly important role in the binding specificity/affinity of the antibodies according to the invention and therefore provide a further object of the invention.
  • hypervariable region or “antigen-binding portion of an antibody” when used herein refer to the amino acid residues of an antibody which are responsible for antigen-binding.
  • the hypervariable region comprises amino acid residues from the “complementarity determining regions” or “CDRs”.
  • “Framework” or “FR” regions are those variable domain regions other than the hypervariable region residues as herein defined. Therefore, the light and heavy chains of an antibody comprise from N- to C-terminus the domains FR1, CDR1, FR2, CDR2, FR3, CDR3, and FR4. CDRs on each chain are separated by such framework amino acids. Especially, CDR3 of the heavy chain is the region which contributes most to antigen binding.
  • CDR and FR regions are determined according to the standard definition of Kabat, E. A., et al., Sequences of Proteins of Immunological Interest, 5th ed., Public Health Service, National Institutes of Health, Bethesda, Md. (1991).
  • binding refers to the binding of the antibody to an epitope of the antigen in an in vitro assay, preferably in an plasmon resonance assay (BIAcore, GE-Healthcare Uppsala, Sweden) with purified wild-type antigen.
  • the affinity of the binding is defined by the terms ka (rate constant for the association of the antibody from the antibody/antigen complex), k D (dissociation constant), and K D (k D /ka).
  • Binding or specifically binding means a binding affinity (K D ) of 10 ⁇ 8 mol/l or less, preferably 10 ⁇ 9 M to 10 ⁇ 13 mol/l.
  • an trivalent, bispecific antibody according to the invention is specifically binding to each antigen for which it is specific with a binding affinity (K D ) of 10 ⁇ 8 mol/l or less, preferably 10 ⁇ 9 M to 10 ⁇ 13 mol/l.
  • Binding of the antibody to the Fc ⁇ RIII can be investigated by a BIAcore assay (GE-Healthcare Uppsala, Sweden).
  • the affinity of the binding is defined by the terms ka (rate constant for the association of the antibody from the antibody/antigen complex), k D (dissociation constant), and K D (k D /ka).
  • epitope includes any polypeptide determinant capable of specific binding to an antibody.
  • epitope determinant include chemically active surface groupings of molecules such as amino acids, sugar side chains, phosphoryl, or sulfonyl, and, in certain embodiments, may have specific three dimensional structural characteristics, and or specific charge characteristics.
  • An epitope is a region of an antigen that is bound by an antibody.
  • an antibody is the to specifically bind an antigen when it preferentially recognizes its target antigen in a complex mixture of proteins and/or macromolecules.
  • peptide connector denotes a peptide with amino acid sequences, which is preferably of synthetic origin. These peptide connectors according to invention are used to fuse the polypeptides under b) and c) to the heavy chain C-termini of the full length antibody to form the trivalent, bispecific antibody according to the invention.
  • the peptide connectors are peptides with an amino acid sequence with a length of at least 5 amino acids, preferably with a length of 10 to 100 amino acids, more preferably with a length of 25 to 50 amino acids.
  • the trivalent, bispecific antibody according to the invention is characterized in that the full length antibody is of human IgG1 subclass, or of human IgG1 subclass with the mutations L234A and L235A.
  • the trivalent, bispecific antibody according to the invention is characterized in that the full length antibody is of human IgG2 subclass.
  • the trivalent, bispecific antibody according to the invention is characterized in that the full length antibody is of human IgG3 subclass.
  • the trivalent, bispecific antibody according to the invention is characterized in that the full length antibody is of human IgG4 subclass or, of human IgG4 subclass with the additional mutation S228P.
  • the trivalent, bispecific antibody according to the invention is characterized in that the full length antibody is of human IgG1 subclass, of human IgG4 subclass with the additional mutation S228P.
  • the trivalent, bispecific antibodies according to the invention have improved characteristics such as biological or pharmacological activity, pharmacokinetic properties or toxicity. They can be used e.g. for the treatment of diseases such as cancer.
  • the trivalent, bispecific antibody according to the invention is characterized in specifically binding to ErbB3 and c-Met.
  • the term “constant region” as used within the current applications denotes the sum of the domains of an antibody other than the variable region.
  • the constant region is not involved directly in binding of an antigen, but exhibit various effector functions.
  • antibodies are divided in the classes: IgA, IgD, IgE, IgG and IgM, and several of these may be further divided into subclasses, such as IgG1, IgG2, IgG3, and IgG4, IgA1 and IgA2.
  • the heavy chain constant regions that correspond to the different classes of antibodies are called ⁇ , ⁇ , ⁇ , ⁇ , and ⁇ , respectively.
  • the light chain constant regions (CL) which can be found in all five antibody classes are called ⁇ (kappa) and ⁇ (lambda).
  • constant region derived from human origin denotes a constant heavy chain region of a human antibody of the subclass IgG1, IgG2, IgG3, or IgG4 and/or a constant light chain kappa or lambda region.
  • constant regions are well known in the state of the art and e.g. described by Kabat, E. A., (see e.g. Johnson, G., and Wu, T. T., Nucleic Acids Res. 28 (2000) 214-218; Kabat, E. A., et al., Proc. Natl. Acad. Sci. USA 72 (1975) 2785-2788).
  • an antibody according to the invention has a reduced FcR binding compared to an IgG1 antibody and the full length parent antibody is in regard to FcR binding of IgG4 subclass or of IgG1 or IgG2 subclass with a mutation in S228, L234, L235 and/or D265, and/or contains the PVA236 mutation.
  • the mutations in the full length parent antibody are S228P, L234A, L235A, L235E and/or PVA236.
  • the mutations in the full length parent antibody are in IgG4 S228P and in IgG1 L234A and L235A.
  • ADCC antibody-dependent cell-mediated cytotoxicity
  • CDC complement-dependent cytotoxicity
  • C1q complement factor C1q
  • binding site IgG antibody subclasses
  • binding site IgG antibody subclasses
  • Such constant region binding sites are known in the state of the art and described e.g. by Lukas, T. J., et al., J. Immunol. 127 (1981) 2555-2560; Brunhouse, R. and Cebra, J. J., Mol. Immunol. 16 (1979) 907-917; Burton, D.
  • Such constant region binding sites are, e.g., characterized by the amino acids L234, L235, D270, N297, E318, K320, K322, P331, and P329 (numbering according to EU index of Kabat).
  • ADCC antibody-dependent cellular cytotoxicity
  • complement-dependent cytotoxicity denotes a process initiated by binding of complement factor C1q to the Fc part of most IgG antibody subclasses. Binding of C1q to an antibody is caused by defined protein-protein interactions at the so called binding site.
  • Fc part binding sites are known in the state of the art (see above). Such Fc part binding sites are, e.g., characterized by the amino acids L234, L235, D270, N297, E318, K320, K322, P331, and P329 (numbering according to EU index of Kabat).
  • Antibodies of subclass IgG1, IgG2, and IgG3 usually show complement activation including C1q and C3 binding, whereas IgG4 does not activate the complement system and does not bind C1q and/or C3.
  • IgG1 type antibodies the most commonly used therapeutic antibodies, are glycoproteins that have a conserved N-linked glycosylation site at Asn297 in each CH2 domain.
  • the two complex biantennary oligosaccharides attached to Asn297 are buried between the CH2 domains, forming extensive contacts with the polypeptide backbone, and their presence is essential for the antibody to mediate effector functions such as antibody dependent cellular cytotoxicity (ADCC) (Lifely, M.
  • ADCC antibody dependent cellular cytotoxicity
  • the bispecific ⁇ ErbB3-c-Met> antibodies which are one embodiment of the invention show reduced downregulation and internalization of target antigen compared to their parent ⁇ ErbB3> and/or ⁇ c-Met> antibodies. Therefore in one preferred embodiment of the invention, the bispecific antibody is glycosylated (if it comprises an Fc part of IgG1, IgG2, IgG3 or IgG4 subclass, preferably of IgG1 or IgG3 subclass) with a sugar chain at Asn297 whereby the amount of fucose within the sugar chain is 65% or lower (Numbering according to Kabat). In another embodiment is the amount of fucose within the sugar chain is between 5% and 65%, preferably between 20% and 40%.
  • “Asn297” according to the invention means amino acid asparagine located at about position 297 in the Fc region. Based on minor sequence variations of antibodies, Asn297 can also be located some amino acids (usually not more than +3 amino acids) upstream or downstream of position 297, i.e. between position 294 and 300.
  • the glycosylated antibody according to the invention the IgG subclass is of human IgG1 subclass, of human IgG1 subclass with the mutations L234A and L235A or of IgG3 subclass.
  • the amount of N-glycolylneuraminic acid (NGNA) is 1% or less and/or the amount of N-terminal alpha-1,3-galactose is 1% or less within the sugar chain.
  • the sugar chain show preferably the characteristics of N-linked glycans attached to Asn297 of an antibody recombinantly expressed in a CHO cell.
  • the sugar chains show characteristics of N-linked glycans attached to Asn297 of an antibody recombinantly expressed in a CHO cell” denotes that the sugar chain at Asn297 of the full length parent antibody according to the invention has the same structure and sugar residue sequence except for the fucose residue as those of the same antibody expressed in unmodified CHO cells, e.g. as those reported in WO 2006/103100.
  • NGNA as used within this application denotes the sugar residue N-glycolylneuraminic acid.
  • CHO type glycosylation of antibody Fc parts is e.g. described by Routier, F. H., Glycoconjugate J. 14 (1997) 201-207.
  • Antibodies which are recombinantly expressed in non-glycomodified CHO host cells usually are fucosylated at Asn297 in an amount of at least 85%.
  • the modified oligosaccharides of the full length parent antibody may be hybrid or complex.
  • the bisected, reduced/not-fucosylated oligosaccharides are hybrid.
  • the bisected, reduced/not-fucosylated oligosaccharides are complex.
  • amount of fucose means the amount of the sugar within the sugar chain at Asn297, related to the sum of all glycostructures attached to Asn297 (e.g. complex, hybrid and high mannose structures) measured by MALDI-TOF mass spectrometry and calculated as average value.
  • the relative amount of fucose is the percentage of fucose-containing structures related to all glycostructures identified in an N-Glycosidase F treated sample (e.g. complex, hybrid and oligo- and high-mannose structures, resp.) by MALDI-TOF.
  • the antibody according to the invention is produced by recombinant means.
  • one aspect of the current invention is a nucleic acid encoding the antibody according to the invention and a further aspect is a cell comprising the nucleic acid encoding an antibody according to the invention.
  • Methods for recombinant production are widely known in the state of the art and comprise protein expression in prokaryotic and eukaryotic cells with subsequent isolation of the antibody and usually purification to a pharmaceutically acceptable purity.
  • nucleic acids encoding the respective modified light and heavy chains are inserted into expression vectors by standard methods.
  • the trivalent, bispecific antibodies according to the invention are suitably separated from the culture medium by conventional immunoglobulin purification procedures such as, for example, protein A-Sepharose, hydroxylapatite chromatography, gel electrophoresis, dialysis, or affinity chromatography.
  • DNA and RNA encoding the monoclonal antibodies is readily isolated and sequenced using conventional procedures.
  • the hybridoma cells can serve as a source of such DNA and RNA.
  • the DNA may be inserted into expression vectors, which are then transfected into host cells such as HEK 293 cells, CHO cells, or myeloma cells that do not otherwise produce immunoglobulin protein, to obtain the synthesis of recombinant monoclonal antibodies in the host cells.
  • Amino acid sequence variants (or mutants) of the trivalent, bispecific antibody are prepared by introducing appropriate nucleotide changes into the antibody DNA, or by nucleotide synthesis. Such modifications can be performed, however, only in a very limited range, e.g. as described above. For example, the modifications do not alter the above mentioned antibody characteristics such as the IgG isotype and antigen binding, but may improve the yield of the recombinant production, protein stability or facilitate the purification.
  • host cell denotes any kind of cellular system which can be engineered to generate the antibodies according to the current invention.
  • HEK293 cells and CHO cells are used as host cells.
  • the expressions “cell,” “cell line,” and “cell culture” are used interchangeably and all such designations include progeny.
  • the words “transformants” and “transformed cells” include the primary subject cell and cultures derived therefrom without regard for the number of transfers. It is also understood that all progeny may not be precisely identical in DNA content, due to deliberate or inadvertent mutations. Variant progeny that have the same function or biological activity as screened for in the originally transformed cell are included.
  • NS0 cells Expression in NS0 cells is described by, e.g., Barnes, L. M., et al., Cytotechnology 32 (2000) 109-123; Barnes, L. M., et al., Biotech. Bioeng. 73 (2001) 261-270.
  • Transient expression is described by, e.g., Durocher, Y., et al., Nucl. Acids. Res. 30 (2002) E9.
  • Cloning of variable domains is described by Orlandi, R., et al., Proc. Natl. Acad. Sci. USA 86 (1989) 3833-3837; Carter, P., et al., Proc. Natl. Acad. Sci.
  • HEK 293 A preferred transient expression system (HEK 293) is described by Schlaeger, E.-J., and Christensen, K., in Cytotechnology 30 (1999) 71-83 and by Schlaeger, E.-J., in J. Immunol. Methods 194 (1996) 191-199.
  • control sequences that are suitable for prokaryotes include a promoter, optionally an operator sequence, and a ribosome binding site.
  • Eukaryotic cells are known to utilize promoters, enhancers and polyadenylation signals.
  • a nucleic acid is “operably linked” when it is placed in a functional relationship with another nucleic acid sequence.
  • DNA for a pre-sequence or secretory leader is operably linked to DNA for a polypeptide if it is expressed as a pre-protein that participates in the secretion of the polypeptide;
  • a promoter or enhancer is operably linked to a coding sequence if it affects the transcription of the sequence; or a ribosome binding site is operably linked to a coding sequence if it is positioned so as to facilitate translation.
  • “operably linked” means that the DNA sequences being linked are contiguous, and, in the case of a secretory leader, contiguous and in reading frame. However, enhancers do not have to be contiguous. Linking is accomplished by ligation at convenient restriction sites. If such sites do not exist, the synthetic oligonucleotide adaptors or linkers are used in accordance with conventional practice.
  • Purification of antibodies is performed in order to eliminate cellular components or other contaminants, e.g. other cellular nucleic acids or proteins, by standard techniques, including alkaline/SDS treatment, CsCl banding, column chromatography, agarose gel electrophoresis, and others well known in the art. See Ausubel, F., et al., ed. Current Protocols in Molecular Biology, Greene Publishing and Wiley Interscience, New York (1987). Different methods are well established and widespread used for protein purification, such as affinity chromatography with microbial proteins (e.g. protein A or protein G affinity chromatography), ion exchange chromatography (e.g.
  • cation exchange (carboxymethyl resins), anion exchange (amino ethyl resins) and mixed-mode exchange), thiophilic adsorption (e.g. with beta-mercaptoethanol and other SH ligands), hydrophobic interaction or aromatic adsorption chromatography (e.g. with phenyl-sepharose, aza-arenophilic resins, or m-aminophenylboronic acid), metal chelate affinity chromatography (e.g. with Ni(II)- and Cu(II)-affinity material), size exclusion chromatography, and electrophoretical methods (such as gel electrophoresis, capillary electrophoresis) (Vijayalakshmi, M. A., Appl. Biochem. Biotech. 75 (1998) 93-102).
  • One aspect of the invention is a pharmaceutical composition comprising an antibody according to the invention.
  • Another aspect of the invention is the use of an antibody according to the invention for the manufacture of a pharmaceutical composition.
  • a further aspect of the invention is a method for the manufacture of a pharmaceutical composition comprising an antibody according to the invention.
  • the present invention provides a composition, e.g. a pharmaceutical composition, containing an antibody according to the present invention, formulated together with a pharmaceutical carrier.
  • One embodiment of the invention is the trivalent, bispecific antibody according to the invention for the treatment of cancer.
  • Another aspect of the invention is the pharmaceutical composition for the treatment of cancer.
  • Another aspect of the invention is the use of an antibody according to the invention for the manufacture of a medicament for the treatment of cancer.
  • Another aspect of the invention is method of treatment of patient suffering from cancer by administering an antibody according to the invention to a patient in the need of such treatment.
  • “pharmaceutical carrier” includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible.
  • the carrier is suitable for intravenous, intramuscular, subcutaneous, parenteral, spinal or epidermal administration (e.g. by injection or infusion).
  • a composition of the present invention can be administered by a variety of methods known in the art. As will be appreciated by the skilled artisan, the route and/or mode of administration will vary depending upon the desired results. To administer a compound of the invention by certain routes of administration, it may be necessary to coat the compound with, or co-administer the compound with, a material to prevent its inactivation.
  • the compound may be administered to a subject in an appropriate carrier, for example, liposomes, or a diluent.
  • Pharmaceutically acceptable diluents include saline and aqueous buffer solutions.
  • Pharmaceutical carriers include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion. The use of such media and agents for pharmaceutically active substances is known in the art.
  • parenteral administration and “administered parenterally” as used herein means modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intra-arterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intra-articular, subcapsular, subarachnoid, intraspinal, epidural and intrasternal injection and infusion.
  • cancer refers to proliferative diseases, such as lymphomas, lymphocytic leukemias, lung cancer, non small cell lung (NSCL) cancer, bronchioloalviolar cell lung cancer, bone cancer, pancreatic cancer, skin cancer, cancer of the head or neck, cutaneous or intraocular melanoma, uterine cancer, ovarian cancer, rectal cancer, cancer of the anal region, stomach cancer, gastric cancer, colon cancer, breast cancer, uterine cancer, carcinoma of the fallopian tubes, carcinoma of the endometrium, carcinoma of the cervix, carcinoma of the vagina, carcinoma of the vulva, Hodgkin's Disease, cancer of the esophagus, cancer of the small intestine, cancer of the endocrine system, cancer of the thyroid gland, cancer of the parathyroid gland, cancer of the adrenal gland, sarcoma of soft tissue, cancer of the urethra, cancer of the penis, prostate cancer, cancer of the bladder, cancer of the kidney or ure
  • compositions may also contain adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents. Prevention of presence of microorganisms may be ensured both by sterilization procedures, supra, and by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol, sorbic acid, and the like. It may also be desirable to include isotonic agents, such as sugars, sodium chloride, and the like into the compositions. In addition, prolonged absorption of the injectable pharmaceutical form may be brought about by the inclusion of agents which delay absorption such as aluminum monostearate and gelatin.
  • the compounds of the present invention which may be used in a suitable hydrated form, and/or the pharmaceutical compositions of the present invention, are formulated into pharmaceutically acceptable dosage forms by conventional methods known to those of skill in the art.
  • Actual dosage levels of the active ingredients in the pharmaceutical compositions of the present invention may be varied so as to obtain an amount of the active ingredient which is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient.
  • the selected dosage level will depend upon a variety of pharmacokinetic factors including the activity of the particular compositions of the present invention employed, the route of administration, the time of administration, the rate of excretion of the particular compound being employed, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular compositions employed, the age, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors well known in the medical arts.
  • composition must be sterile and fluid to the extent that the composition is deliverable by syringe.
  • carrier preferably is an isotonic buffered saline solution.
  • Proper fluidity can be maintained, for example, by use of coating such as lecithin, by maintenance of required particle size in the case of dispersion and by use of surfactants.
  • isotonic agents for example, sugars, polyalcohols such as mannitol or sorbitol, and sodium chloride in the composition.
  • the expressions “cell,” “cell line,” and “cell culture” are used interchangeably and all such designations include progeny.
  • the words “transformants” and “transformed cells” include the primary subject cell and cultures derived therefrom without regard for the number of transfers. It is also understood that all progeny may not be precisely identical in DNA content, due to deliberate or inadvertent mutations. Variant progeny that have the same function or biological activity as screened for in the originally transformed cell are included. Where distinct designations are intended, it will be clear from the context.
  • transfection refers to process of transfer of a vectors/nucleic acid into a host cell. If cells without daunting cell wall barriers are used as host cells, transfection is carried out e.g. by the calcium phosphate precipitation method as described by Graham, F. L., and van der Eb, A. J., Virology 52 (1973) 456-467. However, other methods for introducing DNA into cells such as by nuclear injection or by protoplast fusion may also be used. If prokaryotic cells or cells which contain substantial cell wall constructions are used, e.g. one method of transfection is calcium treatment using calcium chloride as described by Cohen, S. N, et al., PNAS 69 (1972) 2110-2114.
  • expression refers to the process by which a nucleic acid is transcribed into mRNA and/or to the process by which the transcribed mRNA (also referred to as transcript) is subsequently being translated into peptides, polypeptides, or proteins.
  • the transcripts and the encoded polypeptides are collectively referred to as gene product. If the polynucleotide is derived from genomic DNA, expression in a eukaryotic cell may include splicing of the mRNA.
  • a “vector” is a nucleic acid molecule, in particular self-replicating, which transfers an inserted nucleic acid molecule into and/or between host cells.
  • the term includes vectors that function primarily for insertion of DNA or RNA into a cell (e.g., chromosomal integration), replication of vectors that function primarily for the replication of DNA or RNA, and expression vectors that function for transcription and/or translation of the DNA or RNA. Also included are vectors that provide more than one of the functions as described.
  • an “expression vector” is a polynucleotide which, when introduced into an appropriate host cell, can be transcribed and translated into a polypeptide.
  • An “expression system” usually refers to a suitable host cell comprised of an expression vector that can function to yield a desired expression product.
  • FIG. 1 Schematic structure of a full length antibody without CH4 domain specifically binding to a first antigen 1 with two pairs of heavy and light chain which comprise variable and constant domains in a typical order.
  • FIG. 2 Schematic representation of a trivalent, bispecific antibody according to the invention, comprising a full length antibody specifically binding to a first antigen 1 to which
  • FIG. 2 a two polypeptides VH and VL are fused (the VH and VL domains of both together forming a antigen binding site specifically binding to a second antigen 2; b) FIG. 2 b two polypeptides VH-CH1 and VL-CL are fused (the VH and VL domains of both together forming a antigen binding site specifically binding to a second antigen 2)
  • FIG. 3 Schematic representation of a trivalent, bispecific antibody according to the invention, comprising a full length antibody specifically binding to a first antigen 1 to which two polypeptides VH and VL are fused (the VH and VL domains of both together forming a antigen binding site specifically binding to a second antigen 2) with “knobs and holes”.
  • FIG. 4 Schematic representation of a trivalent, bispecific antibody according to the invention, comprising a full length antibody specifically binding to a first antigen 1 to which two polypeptides VH and VL are fused (the VH and VL domains of both together forming a antigen binding site specifically binding to a second antigen 2, wherein these VH and VL domains comprise an interchain disulfide bridge between positions VH44 and VL100) with “knobs and holes”.
  • FIG. 5 Binding of bispecific antibodies to the cell surface of cancer cells
  • FIG. 6 Inhibition of HGF-induced c-Met receptor phosphorylation by bispecific Her3/c-Met antibody formats
  • FIG. 7 Inhibition of HRG-induced Her3 receptor phosphorylation by bispecific Her3/c-Met antibody formats.
  • FIG. 8 Inhibition of HGF-induced HUVEC proliferation by bispecific Her3/c-Met antibody formats
  • FIG. 9 Inhibition of proliferation in the cancer cell line A431 by bispecific Her3/c-Met antibody formats.
  • FIG. 10 Analysis of inhibition of HGF-induced cell-cell dissemination (scattering) in the cancer cell line A431 by bispecific Her3/c-Met antibody formats.
  • DNA sequences were determined by double strand sequencing performed at SequiServe (Vaterstetten, Germany) and Geneart AG (Regensburg, Germany).
  • Desired gene segments were prepared by Geneart AG (Regensburg, Germany) from synthetic oligonucleotides and PCR products by automated gene synthesis.
  • the gene segments which are flanked by singular restriction endonuclease cleavage sites were cloned into pGA18 (ampR) plasmids.
  • the plasmid DNA was purified from transformed bacteria and concentration determined by UV spectroscopy. The DNA sequence of the subcloned gene fragments was confirmed by DNA sequencing.
  • DNA sequences coding “knobs-into-hole” Her3 (clone 29) antibody heavy chain carrying S354C and T366W mutations in the CH3 domain with a C-terminal 5D5 VH region linked by a (G 4 S) n peptide connector as well as “knobs-into-hole” Her3 (clone 29) antibody heavy chain carrying Y349C, T366S, L368A and Y407V mutations with a C-terminal 5D5 VL region linked by a (G 4 S) n peptide connector were prepared by gene synthesis with flanking BamHI and XbaI restriction sites.
  • DNA sequenes encoding unmodified heavy and light chains of the Her3 (clone 29) and 5D5 antibody were synthesized with flanking BamHI and XbaI restriction sites. All constructs were designed with a 5′-end DNA sequence coding for a leader peptide (MGWSCIILFLVATATGVHS), which targets proteins for secretion in eukaryotic cells.
  • MGWSCIILFLVATATGVHS leader peptide
  • a Roche expression vector was used for the construction of all heavy VH/or VL fusion protein and light chain protein encoding expression plasmids.
  • the vector is composed of the following elements:
  • the immunoglobulin fusion genes comprising the heavy or light chain constructs as well as “knobs-into-hole” constructs with C-terminal VH and VL domains were prepared by gene synthesis and cloned into pGA18 (ampR) plasmids as described.
  • the pG18 (ampR) plasmids carrying the synthesized DNA segments and the Roche expression vector were digested with BamHI and XbaI restriction enzymes (Roche Molecular Biochemicals) and subjected to agarose gel electrophoresis. Purified heavy and light chain coding DNA segments were then ligated to the isolated Roche expression vector BamHI/XbaI fragment resulting in the final expression vectors. The final expression vectors were transformed into E.
  • Recombinant immunoglobulin variants were expressed by transient transfection of human embryonic kidney 293-F cells using the FreeStyleTM 293 Expression System according to the manufacturer's instruction (Invitrogen, USA). Briefly, suspension FreeStyleTM 293-F cells were cultivated in FreeStyleTM 293 Expression medium at 37° C./8% CO 2 and the cells were seeded in fresh medium at a density of 1-2 ⁇ 10 6 viable cells/ml on the day of transfection.
  • DNA-293FectinTM complexes were prepared in Opti-MEM® I medium (Invitrogen, USA) using 325 ⁇ A of 293FectinTM (Invitrogen, Germany) and 250 ⁇ g of heavy and light chain plasmid DNA in a 1:1 molar ratio for a 250 ml final transfection volume.
  • “Knobs-into-hole” DNA-293fectin complexes were prepared in Opti-MEM® I medium (Invitrogen, USA) using 325 ⁇ A of 293FectinTM (Invitrogen, Germany) and 250 ⁇ g of “Knobs-into-hole” heavy chain 1 and 2 and light chain plasmid DNA in a 1:1:2 molar ratio for a 250 ml final transfection volume.
  • Antibody containing cell culture supernatants were harvested 7 days after transfection by centrifugation at 14000 g for 30 minutes and filtered through a sterile filter (0.22 ⁇ m). Supernatants were stored at ⁇ 20° C. until purification.
  • Trivalent bispecific and control antibodies were purified from cell culture supernatants by affinity chromatography using Protein A-SepharoseTM (GE Healthcare, Sweden) and Superdex200 size exclusion chromatography. Briefly, sterile filtered cell culture supernatants were applied on a HiTrap ProteinA HP (5 ml) column equilibrated with PBS buffer (10 mM Na 2 HPO 4 , 1 mM KH 2 PO 4 , 137 mM NaCl and 2.7 mM KCl, pH 7.4). Unbound proteins were washed out with equilibration buffer.
  • Antibody and antibody variants were eluted with 0.1 M citrate buffer, pH 2.8, and the protein containing fractions were neutralized with 0.1 ml 1 M Tris, pH 8.5. Then, the eluted protein fractions were pooled, concentrated with an Amicon Ultra centrifugal filter device (MWCO: 30 K, Millipore) to a volume of 3 ml and loaded on a Superdex200 HiLoad 120 ml 16/60 gel filtration column (GE Healthcare, Sweden) equilibrated with 20 mM Histidin, 140 mM NaCl, pH 6.0.
  • MWCO Amicon Ultra centrifugal filter device
  • Fab fragments were generated by a Papain digest of the purified 5D5 monoclonal antibody and subsequent removal of contaminating Fc domains by Protein A chromatography. Unbound Fab fragments were further purified on a Superdex200 HiLoad 120 ml 16/60 gel filtration column (GE Healthcare, Sweden) equilibrated with 20 mM Histidin, 140 mM NaCl, pH 6.0, pooled and stored as 1.0 mg/ml aliquots at ⁇ 80° C.
  • the protein concentration of purified protein samples was determined by measuring the optical density (OD) at 280 nm, using the molar extinction coefficient calculated on the basis of the amino acid sequence.
  • Purity and molecular weight of bispecific and control antibodies were analyzed by SDS-PAGE in the presence and absence of a reducing agent (5 mM 1,4-dithiotreitol) and staining with Coomassie brilliant blue).
  • the NuPAGE® Pre-Cast gel system (Invitrogen, USA) was used according to the manufacturer's instruction (4-20% Tris-Glycine gels).
  • the aggregate content of bispecific and control antibody samples was analyzed by high-performance SEC using a Superdex 200 analytical size-exclusion column (GE Healthcare, Sweden) in 200 mM KH 2 PO 4 , 250 mM KCl, pH 7.0 running buffer at 25° C. 25 ⁇ g protein were injected on the column at a flow rate of 0.5 ml/min and eluted isocratic over 50 minutes.
  • concentrations of 1 mg/ml of purified proteins were incubated at 4° C. and 40° C. for 7 days and then evaluated by high-performance SEC.
  • 5 ⁇ 10e5 A549 cells were seeded per well of a 6-well plate the day prior HGF stimulation in RPMI with 0.5% FCS (fetal calf serum). The next day, growth medium was replaced for one hour with RPMI containing 0.2% BSA (bovine serum albumine). 5 ⁇ g/mL of the bispecific antibody was then added to the medium and cells were incubated for 10 minutes upon which HGF was added for further 10 minutes in a final concentration of 50 ⁇ g/mL.
  • FCS fetal calf serum
  • BSA bovine serum albumine
  • Cells were washed once with ice cold PBS containing 1 mM sodium vanadate upon which they were placed on ice and lysed in the cell culture plate with 100 ⁇ L lysis buffer (50 mM Tris-Cl pH7.5, 150 mM NaCl, 1% NP40, 0.5% DOC, aprotinine, 0.5 mM PMSF, 1 mM sodium-vanadate). Cell lysates were transferred to eppendorf tubes and lysis was allowed to proceed for 30 minutes on ice. Protein concentration was determined using the BCA method (Pierce).
  • 2 ⁇ 10e5 MCF7 cells were seeded per well of a 12-well plate in complete growth medium (RPMI 1640, 10% FCS). Cells were allowed to grow to 90% confluency within two days. Medium was then replaced with starvation medium containing 0.5% FCS. The next day the respective antibodies were supplemented at the indicated concentrations 1 hour prior addition of 500 ng/mL Heregulin (R&D). Upon addition of Heregulin cells were cultivated further 10 minutes before the cells were harvested and lysed. Protein concentration was determined using the BCA method (Pierce). 30-50 ⁇ g of the lysate was separated on a 4-12% Bis-Tris NuPage gel (Invitrogen) and proteins on the gel were transferred to a nitrocellulose membrane. Membranes were blocked for one hour with TBS-T containing 5% BSA and developed with a phospho-specific Her3/ErbB3 antibody specifically recognizing Tyr1289 (4791, Cell Signaling).
  • A549 (4000 cells per well) or A431 (8000 cells per well) were seeded the day prior compound treatment in a total volume of 200 ⁇ L in 96-well E-Plates (Roche, 05232368001) in RPMI with 0.5% FCS. Adhesion and cell growth was monitored over night with the Real Time Cell Analyzer machine with sweeps every 15 min monitoring the impedance. The next day, cells were pre-incubated with 5 ⁇ L of the respective antibody dilutions in PBS with sweeps every five minutes. After 30 minutes 2.5 ⁇ L of a HGF solution yielding a final concentration of 20 ng/mL were added and the experiment was allowed to proceed for further 72 hours. Immediate changes were monitored with sweeps every minute for 180 minutes followed by sweeps every 15 minutes for the remainder of the time.
  • A431 were detached and counted. 1.5 ⁇ 10e5 cells were seeded per well of a conical 96-well plate. Cells were spun down (1500 rpm, 4° C., 5 min) and incubated for 30 min on ice in 50 ⁇ L of a dilution series of the respective bispecific antibody in PBS with 2% FCS (fetal calf serum). Cells were again spun down and washed once with 200 ⁇ L PBS containing 2% FCS followed by a second incubation of 30 min with a phycoerythrin-coupled antibody directed against human Fc which was diluted in PBS containing 2% FCS (Jackson Immunoresearch, 109116098).
  • mfi mean fluorescence intensity of the cells was determined by flow cytometry (FACS Canto, BD). Mfi was determined at least in duplicates of two independent stainings Flow cytometry spectra were further processed using the FlowJo software (TreeStar). Half-maximal binding was determined using XLFit 4.0 (IDBS) and the dose response one site model 205.
  • HT29 cells were detached counted and split in two populations which were individually stained with PKH26 and PKH67 (Sigma) according to the manufacturer's instructions. Of each of the stained populations 5 ⁇ 10e5 cells were taken, combined and incubated for 30 and 60 minutes with 10 ⁇ g/mL of the respective bispecific antibody in complete medium. After the indicated time points cells were stored on ice until the time course was completed. Cells were spun down (1500 rpm, 4° C., 5 min), washed with PBS+2% FCS and fluorescence intensity was determined by flow cytometry (FACS Canto, BD).
  • Cell viability and proliferation was quantified using the cell titer glow assay (Promega). The assay was performed according to the manufacturer's instructions. Briefly, cells were cultured in 96-well plates in a total volume of 100 ⁇ L for the desired period of time. For the proliferation assay, cells were removed from the incubator and placed at room temperature for 30 min. 100 ⁇ L of cell titer glow reagent were added and multi-well plates were placed on an orbital shaker for 2 min. Luminescence was quantified after 15 min on a microplate reader (Tecan).
  • Wst-1 viability and cell proliferation assay was performed as endpoint analysis, detecting the number of metabolic active cells. Briefly, 20 ⁇ L of Wst-1 reagent (Roche, 11644807001) were added to 200 ⁇ L of culture medium. 96-well plates were further incubated for 30 min to 1 h until robust development of the dye. Staining intensity was quantified on a microplate reader (Tecan) at a wavelength of 450 nm.
  • Table 1 Trivalent, bispecific ⁇ ErbB3-c-Met> antibodies based on a full length ErbB-3 antibody (HER3 clone29) and the VH and VL domain from a C-met antibody (c-Met 5D5) with the respective features shown in Table 1 one were expressed and purified according to the general methods described above.
  • the corresponding VH and VL of HER3 clone29 and c-Met 5D5 are given in the sequence listing.
  • the binding properties of the bispecific antibodies to their respective receptor on the cell surface was analyzed on A431 cancer cells in a flow cytometry based assay.
  • Cells were incubated with the mono- or bispecific primary antibodies and binding of these antibodies to their cognate receptors was detected with a secondary antibody coupled to a fluorophore binding specifically to the Fc of the primary antibody.
  • the mean fluorescence intensity of a dilution series of the primary antibodies was plotted against the concentration of the antibody to obtain a sigmoidal binding curve.
  • Cell surface expression of c-Met and Her3 was validated by incubation with the bivalent 5D5 and Her3 clone 29 antibody only.
  • the Her3/c-Met_KHSS antibody readily binds to the cell surface of A431. Under these experimental settings, the antibody can only bind via its Her3 part and consequently the mean fluorescence intensity does not exceed the staining for Her3 clone 29 alone.
  • a c-Met phosphorylation assay was performed.
  • A549 lung cancer cells or HT29 colorectal cancer cells were treated with the bispecific antibodies or control antibodies prior exposure to HGF.
  • Cells were then lysed and phosphorylation of the c-Met receptor was examined. Both cell lines can be stimulated with HGF as can be observed by the occurrence of a phospho-c-Met specific band in the immunoblot.
  • Her3 phosphorylation assay was performed.
  • MCF7 cells were treated with the bispecific antibodies or control antibodies prior exposure to HRG (Heregulin). Cells were then lysed and phosphorylation of the Her3 receptor was examined.
  • Her3/c-Met_KHSS inhibit Her3 receptor phosphorylation to the same extent as the parental Her3 clone29 indicating that Her3 binding and functionality of the antibody are not compromised by the trivalent antibody format.
  • HUVEC proliferation assays were performed to demonstrate the mitogenic effect of HGF. Addition of HGF to HUVEC leads to a twofold increase in proliferation. Addition of human IgG control antibody in the same concentration range as the bispecific antibodies has no impact on cellular proliferation while the 5D5 Fab fragment inhibits HGF-induced proliferation. Titration of Her3/c-Met_KHSS demonstrate a weak inhibitory effect of the antibody ( FIG. 8 ). The effect is more pronounced for the Her3/Met-6C antibody indicating that a longer connector improves efficacy of the antibody. This demonstrates the functionality of the c-Met component in the trivalent antibody format.
  • HGF-induced scattering includes morphological changes of the cell, resulting in rounding of the cells, filopodia-like protrusions, spindle-like structures and a certain motility of the cells.
  • the Real Time Cell Analyzer (Roche) measures the impedance of a given cell culture well and can therefore indirectly monitor changes in cellular morphology and proliferation. Addition of HGF to A431 and A549 cells resulted in changes of the impedance which was monitored as function of time.
  • Her3/c-Met_KHSS and Her3/Met-6C inhibited HGF-induced scattering with Her3/Met-6C being more efficacious (20.7% and 43.7% scatter inhibition) ( FIG. 10 ).

Landscapes

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

Abstract

The present invention relates to trivalent, bispecific antibodies, methods for their production, pharmaceutical compositions containing the antibodies, and uses thereof.

Description

    PRIORITY TO RELATED APPLICATION(S)
  • This application claims the benefit of European Patent Application No. 09005108.7, filed Apr. 7, 2009, which is hereby incorporated by reference in its entirety.
  • SEQUENCE LISTING
  • The instant application contains a Sequence Listing which has been submitted via EFS-Web and is hereby incorporated by reference in its entirety. Said ASCII copy, created on Mar. 1, 2010 is named 26063.txt, and is 93,010 bytes in size.
  • The present invention relates to trivalent, bispecific antibodies, methods for their production, pharmaceutical compositions containing the antibodies, and uses thereof.
  • BACKGROUND OF THE INVENTION
  • A wide variety of multispecific recombinant antibody formats have been developed in the recent past, e.g. tetravalent bispecific antibodies by fusion of, e.g., an IgG antibody format and single chain domains (see e.g. Coloma, M. J., et al., Nature Biotech 15 (1997) 159-163; WO 2001/077342; and Morrison, S. L., Nature Biotech 25 (2007) 1233-1234).
  • Also several other new formats wherein the antibody core structure (IgA, IgD, IgE, IgG or IgM) is no longer retained such as dia-, tria- or tetrabodies, minibodies, several single chain formats (scFv, Bis-scFv), which are capable of binding two or more antigens, have been developed (Holliger, P., et al, Nature Biotech 23 (2005) 1126-1136; Fischer, N., and Léger, O., Pathobiology 74 (2007) 3-14; Shen, J., et al., Journal of Immunological Methods 318 (2007) 65-74; Wu, C., et al., Nature Biotech. 25 (2007) 1290-1297).
  • All such formats use linkers either to fuse the antibody core (IgA, IgD, IgE, IgG or IgM) to a further binding protein (e.g. scFv) or to fuse e.g. two Fab fragments or scFvs (Fischer, N., and Léger, O., Pathobiology 74 (2007) 3-14). It has to be kept in mind that one may want to retain effector functions, such as e.g. complement-dependent cytotoxicity (CDC) or antibody dependent cellular cytotoxicity (ADCC), which are mediated through the Fc receptor binding, by maintaining a high degree of similarity to naturally occurring antibodies.
  • In WO 2007/024715 are reported dual variable domain immunoglobulins as engineered multivalent and multispecific binding proteins. A process for the preparation of biologically active antibody dimers is reported in U.S. Pat. No. 6,897,044. Multivalent FV antibody construct having at least four variable domains which are linked with each over via peptide linkers are reported in U.S. Pat. No. 7,129,330. Dimeric and multimeric antigen binding structures are reported in US 2005/0079170. Tri- or tetra-valent monospecific antigen-binding protein comprising three or four Fab fragments bound to each other covalently by a connecting structure, which protein is not a natural immunoglobulin are reported in U.S. Pat. No. 6,511,663. In WO 2006/020258 tetravalent bispecific antibodies are reported that can be efficiently expressed in prokaryotic and eukaryotic cells, and are useful in therapeutic and diagnostic methods. A method of separating or preferentially synthesizing dimers which are linked via at least one interchain disulfide linkage from dimers which are not linked via at least one interchain disulfide linkage from a mixture comprising the two types of polypeptide dimers is reported in US 2005/0163782. Bispecific tetravalent receptors are reported in U.S. Pat. No. 5,959,083. Engineered antibodies with three or more functional antigen binding sites are reported in WO 2001/077342.
  • Multispecific and multivalent antigen-binding polypeptides are reported in WO 1997/001580. WO 1992/004053 reports homoconjugates, typically prepared from monoclonal antibodies of the IgG class which bind to the same antigenic determinant are covalently linked by synthetic cross-linking Oligomeric monoclonal antibodies with high avidity for antigen are reported in WO 1991/06305 whereby the oligomers, typically of the IgG class, are secreted having two or more immunoglobulin monomers associated together to form tetravalent or hexavalent IgG molecules. Sheep-derived antibodies and engineered antibody constructs are reported in U.S. Pat. No. 6,350,860, which can be used to treat diseases wherein interferon gamma activity is pathogenic. In US 2005/0100543 are reported targetable constructs that are multivalent carriers of bi-specific antibodies, i.e., each molecule of a targetable construct can serve as a carrier of two or more bi-specific antibodies. Genetically engineered bispecific tetravalent antibodies are reported in WO 1995/009917. In WO 2007/109254 stabilized binding molecules that consist of or comprise a stabilized scFv are reported.
  • SUMMARY OF THE INVENTION
  • A first aspect of the current invention is a trivalent, bispecific antibody comprising
  • a) a full length antibody specifically binding to a first antigen and consisting of two antibody heavy chains and two antibody light chains;
    b) a polypeptide consisting of
    ba) an antibody heavy chain variable domain (VH); or
    bb) an antibody heavy chain variable domain (VH) and an antibody constant domain 1 (CH1), wherein the polypeptide is fused with the N-terminus of the VH domain via a peptide connector to the C-terminus of one of the two heavy chains of the full length antibody
    c) a polypeptide consisting of
    ca) an antibody light chain variable domain (VL), or
    cb) an antibody light chain variable domain (VL) and an antibody light chain constant domain (CL);
    wherein the polypeptide is fused with the N-terminus of the VL domain via a peptide connector to the C-terminus of the other of the two heavy chains of the full length antibody; and wherein the antibody heavy chain variable domain (VH) of the polypeptide under b) and the antibody light chain variable domain (VL) of the polypeptide under c) together form an antigen-binding site specifically binding to a second antigen
  • A further aspect of the invention is a nucleic acid molecule encoding a trivalent, bispecific antibody according to the invention.
  • Still further aspects of the invention are a pharmaceutical composition comprising the trivalent, bispecific antibody.
  • The trivalent, bispecific antibodies according to the invention one the one hand show new properties due to their binding to different antigens, and on the other hand are suitable for production and pharmaceutical formulation due to their stability, low aggregation and pharmacokinetic and biological properties. Due to their Ig core they still retain the properties of natural antibodies like ADCC and CDC.
  • DETAILED DESCRIPTION OF THE INVENTION
  • One aspect of the invention is trivalent, bispecific antibody comprising
  • a) a full length antibody specifically binding to a first antigen and consisting of two antibody heavy chains and two antibody light chains;
    b) a polypeptide consisting of
    ba) an antibody heavy chain variable domain (VH); or
    bb) an antibody heavy chain variable domain (VH) and an antibody constant domain 1 (CH1), wherein the polypeptide is fused with the N-terminus of the VH domain via a peptide connector to the C-terminus of one of the two heavy chains of the full length antibody
    c) a polypeptide consisting of
    ca) an antibody light chain variable domain (VL), or
    cb) an antibody light chain variable domain (VL) and an antibody light chain constant domain (CL);
    wherein the polypeptide is fused with the N-terminus of the VL domain via a peptide connector to the C-terminus of the other of the two heavy chains of the full length antibody; and wherein the antibody heavy chain variable domain (VH) of the polypeptide under b) and the antibody light chain variable domain (VL) of the polypeptide under c) together form an antigen-binding site specifically binding to a second antigen
  • Optionally the antibody heavy chain variable domain (VH) of the polypeptide under b) and the antibody light chain variable domain (VL) of the polypeptide under c) are linked and stabilized via a interchain disulfide bridge by introduction of a disulfide bond between the following positions:
  • i) heavy chain variable domain position 44 to light chain variable domain position 100,
    ii) heavy chain variable domain position 105 to light chain variable domain position 43, or
    iii) heavy chain variable domain position 101 to light chain variable domain position 100 (numbering always according to EU index of Kabat).
  • Techniques to introduce unnatural disulfide bridges for stabilization are described e.g. in WO 94/029350, Rajagopal, V., et al., Prot. Engin. (1997) 1453-59; Kobayashi, H., et al., Nuclear Medicine & Biology, Vol. 25, (1998) 387-393; or Schmidt, M., et al., Oncogene (1999) 18 1711-1721. In one embodiment the optional disulfide bond between the variable domains of the polypeptides under b) and c) is between heavy chain variable domain position 44 and light chain variable domain position 100. In one embodiment the optional disulfide bond between the variable domains of the polypeptides under b) and c) is between heavy chain variable domain position 105 and light chain variable domain position 43. (numbering always according to EU index of Kabat) In one embodiment a trivalent, bispecific antibody without the optional disulfide stabilization between the variable domains VH and VL of the single chain Fab fragments is preferred.
  • The term “full length antibody” denotes an antibody consisting of two “full length antibody heavy chains” and two “full length antibody light chains” (see FIG. 1). A “full length antibody heavy chain” is a polypeptide consisting in N-terminal to C-terminal direction of an antibody heavy chain variable domain (VH), an antibody constant heavy chain domain 1 (CH1), an antibody hinge region (HR), an antibody heavy chain constant domain 2 (CH2), and an antibody heavy chain constant domain 3 (CH3), abbreviated as VH-CH1-HR-CH2-CH3; and optionally an antibody heavy chain constant domain 4 (CH4) in case of an antibody of the subclass IgE. Preferably the “full length antibody heavy chain” is a polypeptide consisting in N-terminal to C-terminal direction of VH, CH1, HR, CH2 and CH3. A “full length antibody light chain” is a polypeptide consisting in N-terminal to C-terminal direction of an antibody light chain variable domain (VL), and an antibody light chain constant domain (CL), abbreviated as VL-CL. The antibody light chain constant domain (CL) can be κ (kappa) or λ (lambda). The two full length antibody chains are linked together via inter-polypeptide disulfide bonds between the CL domain and the CH1 domain and between the hinge regions of the full length antibody heavy chains. Examples of typical full length antibodies are natural antibodies like IgG (e.g. IgG1 and IgG2), IgM, IgA, IgD, and IgE.) The full length antibodies according to the invention can be from a single species e.g. human, or they can be chimerized or humanized antibodies. The full length antibodies according to the invention comprise two antigen binding sites each formed by a pair of VH and VL, which both specifically bind to the same antigen. The C-terminus of the heavy or light chain of the full length antibody denotes the last amino acid at the C-terminus of the heavy or light chain.
  • The N-terminus of the antibody heavy chain variable domain (VH) of the polypeptide under b) and the antibody light chain variable domain (VL) of the polypeptide under c) denotes the last amino acid at the N-terminus of VH or VL domain.
  • The CH3 domains of the full length antibody according to the invention can be altered by the “knob-into-holes” technology which is described in detail with several examples in e.g. WO 96/027011, Ridgway, J. B., et al., Protein Eng 9 (1996) 617-621; and Merchant, A. M., et al., Nat Biotechnol 16 (1998) 677-681. In this method the interaction surfaces of the two CH3 domains are altered to increase the heterodimerisation of both heavy chains containing these two CH3 domains. Each of the two CH3 domains (of the two heavy chains) can be the “knob”, while the other is the “hole”. The introduction of a disulfide bridge further stabilizes the heterodimers (Merchant, A. M., et al., Nature Biotech 16 (1998) 677-681; Atwell, S., et al., J. Mol. Biol. 270 (1997) 26-35) and increases the yield.
  • Thus in one aspect of the invention the trivalent, bispecific antibody is further is characterized in that the CH3 domain of one heavy chain of the full length antibody and the CH3 domain of the other heavy chain of the full length antibody each meet at an interface which comprises an original interface between the antibody CH3 domains;
  • wherein the interface is altered to promote the formation of the bivalent, bispecific antibody, wherein the alteration is characterized in that:
  • a) the CH3 domain of one heavy chain is altered,
  • so that within the original interface the CH3 domain of one heavy chain that meets the original interface of the CH3 domain of the other heavy chain within the bivalent, bispecific antibody, an amino acid residue is replaced with an amino acid residue having a larger side chain volume, thereby generating a protuberance within the interface of the CH3 domain of one heavy chain which is positionable in a cavity within the interface of the CH3 domain of the other heavy chain and
  • b) the CH3 domain of the other heavy chain is altered,
  • so that within the original interface of the second CH3 domain that meets the original interface of the first CH3 domain within the trivalent, bispecific antibody
    an amino acid residue is replaced with an amino acid residue having a smaller side chain volume, thereby generating a cavity within the interface of the second CH3 domain within which a protuberance within the interface of the first CH3 domain is positionable.
  • Preferably the amino acid residue having a larger side chain volume is selected from the group consisting of arginine (R), phenylalanine (F), tyrosine (Y), tryptophan (W).
  • Preferably the amino acid residue having a smaller side chain volume is selected from the group consisting of alanine (A), serine (S), threonine (T), valine (V).
  • In one aspect of the invention both CH3 domains are further altered by the introduction of cysteine (C) as amino acid in the corresponding positions of each CH3 domain such that a disulfide bridge between both CH3 domains can be formed.
  • In a preferred embodiment, the trivalent, bispecific comprises a T366W mutation in the CH3 domain of the “knobs chain” and T366S, L368A, Y407V mutations in the CH3 domain of the “hole chain”. An additional interchain disulfide bridge between the CH3 domains can also be used (Merchant, A. M., et al., Nature Biotech 16 (1998) 677-681) e.g. by introducing a Y349C mutation into the CH3 domain of the “knobs chain” and a E356C mutation or a S354C mutation into the CH3 domain of the “hole chain”. Thus in a another preferred embodiment, the trivalent, bispecific antibody comprises Y349C, T366W mutations in one of the two CH3 domains and E356C, T366S, L368A, Y407V mutations in the other of the two CH3 domains or the trivalent, bispecific antibody comprises Y349C, T366W mutations in one of the two CH3 domains and S354C, T366S, L368A, Y407V mutations in the other of the two CH3 domains (the additional Y349C mutation in one CH3 domain and the additional E356C or S354C mutation in the other CH3 domain forming a interchain disulfide bridge) (numbering always according to EU index of Kabat). But also other knobs-in-holes technologies as described by EP 1 870 459A1, can be used alternatively or additionally. A preferred example for the trivalent, bispecific antibody are R409D; K370E mutations in the CH3 domain of the “knobs chain” and D399K; E357K mutations in the CH3 domain of the “hole chain” (numbering always according to EU index of Kabat).
  • In another preferred embodiment the trivalent, bispecific antibody comprises a T366W mutation in the CH3 domain of the “knobs chain” and T366S, L368A, Y407V mutations in the CH3 domain of the “hole chain” and additionally R409D; K370E mutations in the CH3 domain of the “knobs chain” and D399K; E357K mutations in the CH3 domain of the “hole chain”.
  • In another preferred embodiment the trivalent, bispecific antibody comprises Y349C, T366W mutations in one of the two CH3 domains and S354C, T366S, L368A, Y407V mutations in the other of the two CH3 domains or the trivalent, bispecific antibody comprises Y349C, T366W mutations in one of the two CH3 domains and S354C, T366S, L368A, Y407V mutations in the other of the two CH3 domains and additionally R409D; K370E mutations in the CH3 domain of the “knobs chain” and D399K; E357K mutations in the CH3 domain of the “hole chain”.
  • The bispecific antibody to the invention comprises three antigen-binding sites (A) the full length antibody according comprises two identical antigen-binding sites specifically binding to a first antigen, and B) the antibody heavy chain variable domain (VH) of the polypeptide under b) and the antibody light chain variable domain (VL) of the polypeptide under c) form together one antigen binding site specifically binding to a second antigen). The terms “binding site” or “antigen-binding site” as used herein denotes the region(s) of the bispecific antibody according to the invention to which the respective antigen actually specifically binds. The antigen binding sites either in the full length antibody or by the antibody heavy chain variable domain (VH) of the polypeptide under b) and the antibody light chain variable domain (VL) of the polypeptide under c) are formed each by a pair consisting of an antibody light chain variable domain (VL) and an antibody heavy chain variable domain (VH).
  • The antigen-binding sites that specifically bind to the desired antigen can be derived a) from known antibodies to the antigen or b) from new antibodies or antibody fragments obtained by de novo immunization methods using inter alia either the antigen protein or nucleic acid or fragments thereof or by phage display.
  • An antigen-binding site of an antibody of the invention contains six complementarity determining regions (CDRs) which contribute in varying degrees to the affinity of the binding site for antigen. There are three heavy chain variable domain CDRs (CDRH1, CDRH2 and CDRH3) and three light chain variable domain CDRs (CDRL1, CDRL2 and CDRL3). The extent of CDR and framework regions (FRs) is determined by comparison to a compiled database of amino acid sequences in which those regions have been defined according to variability among the sequences.
  • Antibody specificity refers to selective recognition of the antibody for a particular epitope of an antigen. Natural antibodies, for example, are monospecific. “Bispecific antibodies” according to the invention are antibodies which have two different antigen-binding specificities. Where an antibody has more than one specificity, the recognized epitopes may be associated with a single antigen or with more than one antigen. The term “monospecific” antibody as used herein denotes an antibody that has one or more binding sites each of which bind to the same epitope of the same antigen. The term “valent” as used within the current application denotes the presence of a specified number of binding sites in an antibody molecule. A natural antibody for example or a full length antibody according to the invention has two binding sites and is bivalent. As such, the terms “trivalent”, denote the presence of three binding sites in an antibody molecule. The bispecific antibodies according to the invention are “trivalent”. The term “trivalent, bispecific” antibody as used herein denotes an antibody that has three antigen-binding sites of which two bind to the same antigen (or the same epitope of the antigen) and the third binds to a different antigen or a different epitope of the same antigen. Antibodies of the present invention have three binding sites and are bispecific.
  • Another embodiment of the current invention is a trivalent, bispecific antibody comprising:
  • a) a full length antibody specifically binding to a first antigen and consisting of:
    aa) two antibody heavy chains consisting in N-terminal to C-terminal direction of an antibody heavy chain variable domain (VH), an antibody constant heavy chain domain 1 (CH1), an antibody hinge region (HR), an antibody heavy chain constant domain 2 (CH2), and an antibody heavy chain constant domain 3 (CH3); and
    ab) two antibody light chains consisting in N-terminal to C-terminal direction of an antibody light chain variable domain (VL), and an antibody light chain constant domain (CL); and
    b) a polypeptide consisting of
    ba) an antibody heavy chain variable domain (VH); or
    bb) an antibody heavy chain variable domain (VH) and an antibody constant domain 1 (CH1), wherein the polypeptide is fused with the N-terminus of the VH domain via a peptide connector to the C-terminus of one of the two heavy chains of the full length antibody wherein the peptide connector is a peptide of at least 5 amino acids, preferably between 25 and 50 amino acids;
    c) a polypeptide consisting of
    ca) an antibody light chain variable domain (VL), or
    cb) an antibody light chain variable domain (VL) and an antibody light chain constant domain (CL);
    wherein the polypeptide is fused with the N-terminus of the VL domain via a peptide connector to the C-terminus of the other of the two heavy chains of the full length antibody;
    wherein the peptide connector is identical to the peptide connector under b); and wherein the antibody heavy chain variable domain (VH) of the polypeptide under b) and the antibody light chain variable domain (VL) of the polypeptide under c) together form an antigen-binding site specifically binding to a second antigen.
  • Within this embodiment, preferably the trivalent, bispecific antibody comprises a T366W mutation in one of the two CH3 domains of and T366S, L368A, Y407V mutations in the other of the two CH3 domains and more preferably the trivalent, bispecific antibody comprises Y349C, T366W mutations in one of the two CH3 domains of and D356C, T366S, L368A, Y407V mutations in the other of the two CH3 domains (the additional Y349C mutation in one CH3 domain and the additional D356C mutation in the other CH3 domain forming a interchain disulfide bridge).
  • In one embodiment of the invention the trivalent, bispecific antibody according to the invention is characterized in that
  • a) the full length antibody is specifically binding to ErbB-3 comprises as heavy chain variable domain the sequence of SEQ ID NO: 1, and as light chain variable domain the sequence of SEQ ID NO: 2
    b) the polypeptide under b) comprises as the heavy chain variable domain the sequence of SEQ ID NO: 3; and
    c) the polypeptide under c) comprises as the light chain variable domain the sequence of SEQ ID NO: 4.
  • In another aspect of the current invention the trivalent, bispecific antibody according to the invention comprises
  • a) a full length antibody binding to a first antigen consisting of two antibody heavy chains VH-CH1-HR-CH2-CH3 and two antibody light chains VL-CL;
    (wherein preferably one of the two CH3 domains comprises Y349C, T366W mutations and the other of the two CH3 domains comprises S354C, T366S, L368A, Y407V mutations);
    b) a polypeptide consisting of
    ba) an antibody heavy chain variable domain (VH); or
    bb) an antibody heavy chain variable domain (VH) and an antibody constant domain 1 (CH1), wherein the polypeptide is fused with the N-terminus of the VH domain via a peptide connector to the C-terminus of one of the two heavy chains of the full length antibody
    c) a polypeptide consisting of
    ca) an antibody light chain variable domain (VL), or
    cb) an antibody light chain variable domain (VL) and an antibody light chain constant domain (CL);
    wherein the polypeptide is fused with the N-terminus of the VL domain via a peptide connector to the C-terminus of the other of the two heavy chains of the full length antibody;
    and wherein the antibody heavy chain variable domain (VH) of the polypeptide under b) and the antibody light chain variable domain (VL) of the polypeptide under c) together form an antigen-binding site specifically binding to a second antigen.
  • Another embodiment of the current invention is a trivalent, bispecific antibody comprising
  • a) a full length antibody specifically binding to human ErbB-3 and consisting of:
    aa) two antibody heavy chains consisting in N-terminal to C-terminal direction of an antibody heavy chain variable domain (VH), an antibody constant heavy chain domain 1 (CH1), an antibody hinge region (HR), an antibody heavy chain constant domain 2 (CH2), and an antibody heavy chain constant domain 3 (CH3); and
    ab) two antibody light chains consisting in N-terminal to C-terminal direction of an antibody light chain variable domain (VL), and an antibody light chain constant domain (CL) (VL-CL); and
    b) one single chain Fv fragment specifically binding to human c-Met),
    wherein the single chain Fv fragment under b) is fused to the full length antibody under a) via a peptide connector at the C- or N-terminus of the heavy or light chain (preferably at the C-terminus of the heavy chain) of the full length antibody;
    wherein the peptide connector is a peptide of at least 5 amino acids, preferably between 25 and 50 amino acids.
  • Preferably such trivalent, bispecific antibody further comprises Y349C, T366W mutations in one of the two CH3 domains of the full length antibody and S354C (or E356C), T366S, L368A, Y407V mutations in the other of the two CH3 domains of the full length antibody.
  • Another embodiment of the current invention is a trivalent, bispecific antibody comprising
  • a) a full length antibody specifically binding to human ErbB-3 and consisting of:
    aa) two antibody heavy chains consisting in N-terminal to C-terminal direction of an antibody heavy chain variable domain (VH), an antibody constant heavy chain domain 1 (CH1), an antibody hinge region (HR), an antibody heavy chain constant domain 2 (CH2), and an antibody heavy chain constant domain 3 (CH3); and
    ab) two antibody light chains consisting in N-terminal to C-terminal direction of an antibody light chain variable domain (VL), and an antibody light chain constant domain (CL); and
    b) a polypeptide consisting of
    ba) an antibody heavy chain variable domain (VH); or
    bb) an antibody heavy chain variable domain (VH) and an antibody constant domain 1 (CH1),
    wherein the polypeptide is fused with the N-terminus of the VH domain via a peptide connector to the C-terminus of one of the two heavy chains of the full length antibody wherein the peptide connector is a peptide of at least 5 amino acids, preferably between 25 and 50 amino acids;
    c) a polypeptide consisting of
    ca) an antibody light chain variable domain (VL), or
    cb) an antibody light chain variable domain (VL) and an antibody light chain constant domain (CL);
    wherein the polypeptide is fused with the N-terminus of the VL domain via a peptide connector to the C-terminus of the other of the two heavy chains of the full length antibody;
    wherein the peptide connector is identical to the peptide connector under b);
    and wherein the antibody heavy chain variable domain (VH) of the polypeptide under b) and the antibody light chain variable domain (VL) of the polypeptide under c) together form an antigen-binding site specifically binding to human c-Met
  • The full length antibodies of the invention comprise immunoglobulin constant regions of one or more immunoglobulin classes. Immunoglobulin classes include IgG, IgM, IgA, IgD, and IgE isotypes and, in the case of IgG and IgA, their subtypes. In a preferred embodiment, a full length antibody of the invention has a constant domain structure of an IgG type antibody.
  • The terms “monoclonal antibody” or “monoclonal antibody composition” as used herein refer to a preparation of antibody molecules of a single amino acid composition.
  • The term “chimeric antibody” refers to an antibody comprising a variable region, i.e., binding region, from one source or species and at least a portion of a constant region derived from a different source or species, usually prepared by recombinant DNA techniques. Chimeric antibodies comprising a murine variable region and a human constant region are preferred. Other preferred forms of “chimeric antibodies” encompassed by the present invention are those in which the constant region has been modified or changed from that of the original antibody to generate the properties according to the invention, especially in regard to C1q binding and/or Fc receptor (FcR) binding. Such chimeric antibodies are also referred to as “class-switched antibodies.”. Chimeric antibodies are the product of expressed immunoglobulin genes comprising DNA segments encoding immunoglobulin variable regions and DNA segments encoding immunoglobulin constant regions. Methods for producing chimeric antibodies involve conventional recombinant DNA and gene transfection techniques are well known in the art. See, e.g., Morrison, S. L., et al., Proc. Natl. Acad. Sci. USA 81 (1984) 6851-6855; U.S. Pat. No. 5,202,238 and U.S. Pat. No. 5,204,244.
  • The term “humanized antibody” refers to antibodies in which the framework or “complementarity determining regions” (CDR) have been modified to comprise the CDR of an immunoglobulin of different specificity as compared to that of the parent immunoglobulin. In a preferred embodiment, a murine CDR is grafted into the framework region of a human antibody to prepare the “humanized antibody.” See, e.g., Riechmann, L., et al., Nature 332 (1988) 323-327; and Neuberger, M. S., et al., Nature 314 (1985) 268-270. Particularly preferred CDRs correspond to those representing sequences recognizing the antigens noted above for chimeric antibodies. Other forms of “humanized antibodies” encompassed by the present invention are those in which the constant region has been additionally modified or changed from that of the original antibody to generate the properties according to the invention, especially in regard to C1q binding and/or Fc receptor (FcR) binding.
  • The term “human antibody”, as used herein, is intended to include antibodies having variable and constant regions derived from human germ line immunoglobulin sequences. Human antibodies are well-known in the state of the art (van Dijk, M. A., and van de Winkel, J. G., Curr. Opin. Chem. Biol. 5 (2001) 368-374). Human antibodies can also be produced in transgenic animals (e.g., mice) that are capable, upon immunization, of producing a full repertoire or a selection of human antibodies in the absence of endogenous immunoglobulin production. Transfer of the human germ-line immunoglobulin gene array in such germ-line mutant mice will result in the production of human antibodies upon antigen challenge (see, e.g., Jakobovits, A., et al., Proc. Natl. Acad. Sci. USA 90 (1993) 2551-2555; Jakobovits, A., et al., Nature 362 (1993) 255-258; Brüggemann, M., et al., Year Immunol. 7 (1993) 33-40). Human antibodies can also be produced in phage display libraries (Hoogenboom, H. R., and Winter, G. J., Mol. Biol. 227 (1992) 381-388; Marks, J. D., et al., J. Mol. Biol. 222 (1991) 581-597). The techniques of Cole, S. P. C., et al., and Boerner, P., et al., are also available for the preparation of human monoclonal antibodies (Cole, S. P. C., et al., Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, p. 77-96 (1985); and Boerner, P., et al., J. Immunol. 147 (1991) 86-95). As already mentioned for chimeric and humanized antibodies according to the invention the term “human antibody” as used herein also comprises such antibodies which are modified in the constant region to generate the properties according to the invention, especially in regard to C1q binding and/or FcR binding, e.g. by “class switching” i.e. change or mutation of Fc parts (e.g. from IgG1 to IgG4 and/or IgG1/IgG4 mutation.)
  • The term “recombinant human antibody”, as used herein, is intended to include all human antibodies that are prepared, expressed, created or isolated by recombinant means, such as antibodies isolated from a host cell such as a NS0 or CHO cell or from an animal (e.g. a mouse) that is transgenic for human immunoglobulin genes or antibodies expressed using a recombinant expression vector transfected into a host cell. Such recombinant human antibodies have variable and constant regions in a rearranged form. The recombinant human antibodies according to the invention have been subjected to in vivo somatic hypermutation. Thus, the amino acid sequences of the VH and VL regions of the recombinant antibodies are sequences that, while derived from and related to human germ line VH and VL sequences, may not naturally exist within the human antibody germ line repertoire in vivo.
  • The “variable domain” (variable domain of a light chain (VL), variable region of a heavy chain (VH) as used herein denotes each of the pair of light and heavy chains which is involved directly in binding the antibody to the antigen. The domains of variable human light and heavy chains have the same general structure and each domain comprises four framework (FR) regions whose sequences are widely conserved, connected by three “hypervariable regions” (or complementarity determining regions, CDRs). The framework regions adopt a 13-sheet conformation and the CDRs may form loops connecting the 13-sheet structure. The CDRs in each chain are held in their three-dimensional structure by the framework regions and form together with the CDRs from the other chain the antigen binding site. The antibody heavy and light chain CDR3 regions play a particularly important role in the binding specificity/affinity of the antibodies according to the invention and therefore provide a further object of the invention.
  • The terms “hypervariable region” or “antigen-binding portion of an antibody” when used herein refer to the amino acid residues of an antibody which are responsible for antigen-binding. The hypervariable region comprises amino acid residues from the “complementarity determining regions” or “CDRs”. “Framework” or “FR” regions are those variable domain regions other than the hypervariable region residues as herein defined. Therefore, the light and heavy chains of an antibody comprise from N- to C-terminus the domains FR1, CDR1, FR2, CDR2, FR3, CDR3, and FR4. CDRs on each chain are separated by such framework amino acids. Especially, CDR3 of the heavy chain is the region which contributes most to antigen binding. CDR and FR regions are determined according to the standard definition of Kabat, E. A., et al., Sequences of Proteins of Immunological Interest, 5th ed., Public Health Service, National Institutes of Health, Bethesda, Md. (1991).
  • As used herein, the term “binding” or “specifically binding” refers to the binding of the antibody to an epitope of the antigen in an in vitro assay, preferably in an plasmon resonance assay (BIAcore, GE-Healthcare Uppsala, Sweden) with purified wild-type antigen. The affinity of the binding is defined by the terms ka (rate constant for the association of the antibody from the antibody/antigen complex), kD (dissociation constant), and KD (kD/ka). Binding or specifically binding means a binding affinity (KD) of 10−8 mol/l or less, preferably 10−9 M to 10−13 mol/l. Thus, an trivalent, bispecific antibody according to the invention is specifically binding to each antigen for which it is specific with a binding affinity (KD) of 10−8 mol/l or less, preferably 10−9 M to 10−13 mol/l.
  • Binding of the antibody to the FcγRIII can be investigated by a BIAcore assay (GE-Healthcare Uppsala, Sweden). The affinity of the binding is defined by the terms ka (rate constant for the association of the antibody from the antibody/antigen complex), kD (dissociation constant), and KD (kD/ka).
  • The term “epitope” includes any polypeptide determinant capable of specific binding to an antibody. In certain embodiments, epitope determinant include chemically active surface groupings of molecules such as amino acids, sugar side chains, phosphoryl, or sulfonyl, and, in certain embodiments, may have specific three dimensional structural characteristics, and or specific charge characteristics. An epitope is a region of an antigen that is bound by an antibody.
  • In certain embodiments, an antibody is the to specifically bind an antigen when it preferentially recognizes its target antigen in a complex mixture of proteins and/or macromolecules.
  • The term “peptide connector” as used within the invention denotes a peptide with amino acid sequences, which is preferably of synthetic origin. These peptide connectors according to invention are used to fuse the polypeptides under b) and c) to the heavy chain C-termini of the full length antibody to form the trivalent, bispecific antibody according to the invention. Preferably the peptide connectors are peptides with an amino acid sequence with a length of at least 5 amino acids, preferably with a length of 10 to 100 amino acids, more preferably with a length of 25 to 50 amino acids. Preferably the peptide connector under b) and c) are identical peptides with a length of at least 25 amino acids, preferably with a length between 25 and 50 amino acids and more preferably the peptide connector is (G×S)n or (G×S)nGm with G=glycine, S=serine, and (x=3, n=6, 7 or 8, and m=0, 1, 2 or 3) or (x=4, n=3, 4, 5, 6, or 7 and m=0, 1, 2 or 3), preferably x=4 and n=5, 6, or 7.
  • In a further embodiment the trivalent, bispecific antibody according to the invention is characterized in that the full length antibody is of human IgG1 subclass, or of human IgG1 subclass with the mutations L234A and L235A.
  • In a further embodiment the trivalent, bispecific antibody according to the invention is characterized in that the full length antibody is of human IgG2 subclass.
  • In a further embodiment the trivalent, bispecific antibody according to the invention is characterized in that the full length antibody is of human IgG3 subclass.
  • In a further embodiment the trivalent, bispecific antibody according to the invention is characterized in that the full length antibody is of human IgG4 subclass or, of human IgG4 subclass with the additional mutation S228P.
  • Preferably the trivalent, bispecific antibody according to the invention is characterized in that the full length antibody is of human IgG1 subclass, of human IgG4 subclass with the additional mutation S228P.
  • It has now been found that the trivalent, bispecific antibodies according to the invention have improved characteristics such as biological or pharmacological activity, pharmacokinetic properties or toxicity. They can be used e.g. for the treatment of diseases such as cancer.
  • In a further embodiment the trivalent, bispecific antibody according to the invention is characterized in specifically binding to ErbB3 and c-Met. The term “constant region” as used within the current applications denotes the sum of the domains of an antibody other than the variable region. The constant region is not involved directly in binding of an antigen, but exhibit various effector functions. Depending on the amino acid sequence of the constant region of their heavy chains, antibodies are divided in the classes: IgA, IgD, IgE, IgG and IgM, and several of these may be further divided into subclasses, such as IgG1, IgG2, IgG3, and IgG4, IgA1 and IgA2. The heavy chain constant regions that correspond to the different classes of antibodies are called α, δ, ε, γ, and μ, respectively. The light chain constant regions (CL) which can be found in all five antibody classes are called κ (kappa) and λ (lambda).
  • The term “constant region derived from human origin” as used in the current application denotes a constant heavy chain region of a human antibody of the subclass IgG1, IgG2, IgG3, or IgG4 and/or a constant light chain kappa or lambda region. Such constant regions are well known in the state of the art and e.g. described by Kabat, E. A., (see e.g. Johnson, G., and Wu, T. T., Nucleic Acids Res. 28 (2000) 214-218; Kabat, E. A., et al., Proc. Natl. Acad. Sci. USA 72 (1975) 2785-2788).
  • While antibodies of the IgG4 subclass show reduced Fc receptor (FcγRIIIa) binding, antibodies of other IgG subclasses show strong binding. However Pro238, Asp265, Asp270, Asn297 (loss of Fc carbohydrate), Pro329, Leu234, Leu235, Gly236, Gly237, Ile253, Ser254, Lys288, Thr307, Gln311, Asn434, and His435 are residues which, if altered, provide also reduced Fc receptor binding (Shields, R. L., et al., J. Biol. Chem. 276 (2001) 6591-6604; Lund, J., et al., FASEB J. 9 (1995) 115-119; Morgan, A., et al., Immunology 86 (1995) 319-324; EP 0 307 434).
  • In one embodiment an antibody according to the invention has a reduced FcR binding compared to an IgG1 antibody and the full length parent antibody is in regard to FcR binding of IgG4 subclass or of IgG1 or IgG2 subclass with a mutation in S228, L234, L235 and/or D265, and/or contains the PVA236 mutation. In one embodiment the mutations in the full length parent antibody are S228P, L234A, L235A, L235E and/or PVA236. In another embodiment the mutations in the full length parent antibody are in IgG4 S228P and in IgG1 L234A and L235A.
  • The constant region of an antibody is directly involved in ADCC (antibody-dependent cell-mediated cytotoxicity) and CDC (complement-dependent cytotoxicity). Complement activation (CDC) is initiated by binding of complement factor C1q to the constant region of most IgG antibody subclasses. Binding of C1q to an antibody is caused by defined protein-protein interactions at the so called binding site. Such constant region binding sites are known in the state of the art and described e.g. by Lukas, T. J., et al., J. Immunol. 127 (1981) 2555-2560; Brunhouse, R. and Cebra, J. J., Mol. Immunol. 16 (1979) 907-917; Burton, D. R., et al., Nature 288 (1980) 338-344; Thommesen, J. E., et al., Mol. Immunol. 37 (2000) 995-1004; Idusogie, E. E., et al., J. Immunol. 164 (2000) 4178-4184; Hezareh, M., et al., J. Virol. 75 (2001) 12161-12168; Morgan, A., et al., Immunology 86 (1995) 319-324; and EP 0 307 434. Such constant region binding sites are, e.g., characterized by the amino acids L234, L235, D270, N297, E318, K320, K322, P331, and P329 (numbering according to EU index of Kabat).
  • The term “antibody-dependent cellular cytotoxicity (ADCC)” refers to lysis of human target cells by an antibody according to the invention in the presence of effector cells. ADCC is measured preferably by the treatment of a preparation of antigen expressing cells with an antibody according to the invention in the presence of effector cells such as freshly isolated PBMC or purified effector cells from buffy coats, like monocytes or natural killer (NK) cells or a permanently growing NK cell line.
  • The term “complement-dependent cytotoxicity (CDC)” denotes a process initiated by binding of complement factor C1q to the Fc part of most IgG antibody subclasses. Binding of C1q to an antibody is caused by defined protein-protein interactions at the so called binding site. Such Fc part binding sites are known in the state of the art (see above). Such Fc part binding sites are, e.g., characterized by the amino acids L234, L235, D270, N297, E318, K320, K322, P331, and P329 (numbering according to EU index of Kabat). Antibodies of subclass IgG1, IgG2, and IgG3 usually show complement activation including C1q and C3 binding, whereas IgG4 does not activate the complement system and does not bind C1q and/or C3.
  • Cell-mediated effector functions of monoclonal antibodies can be enhanced by engineering their oligosaccharide component as described in Umana, P., et al., Nature Biotechnol. 17 (1999) 176-180, and U.S. Pat. No. 6,602,684. IgG1 type antibodies, the most commonly used therapeutic antibodies, are glycoproteins that have a conserved N-linked glycosylation site at Asn297 in each CH2 domain. The two complex biantennary oligosaccharides attached to Asn297 are buried between the CH2 domains, forming extensive contacts with the polypeptide backbone, and their presence is essential for the antibody to mediate effector functions such as antibody dependent cellular cytotoxicity (ADCC) (Lifely, M. R., et al., Glycobiology 5 (1995) 813-822; Jefferis, R., et al., Immunol. Rev. 163 (1998) 59-76; Wright, A., and Morrison, S. L., Trends Biotechnol. 15 (1997) 26-32). Umana, P., et al. Nature Biotechnol. 17 (1999) 176-180 and WO 99/54342 showed that overexpression in Chinese hamster ovary (CHO) cells of β(1,4)-N-acetylglucosaminyltransferase III (“GnTIII”), a glycosyltransferase catalyzing the formation of bisected oligosaccharides, significantly increases the in vitro ADCC activity of antibodies. Alterations in the composition of the Asn297 carbohydrate or its elimination affect also binding to FcγR and C1q (Umana, P., et al., Nature Biotechnol. 17 (1999) 176-180; Davies, J., et al., Biotechnol. Bioeng. 74 (2001) 288-294; Mimura, Y., et al., J. Biol. Chem. 276 (2001) 45539-45547; Radaev, S., et al., J. Biol. Chem. 276 (2001) 16478-16483; Shields, R. L., et al., J. Biol. Chem. 276 (2001) 6591-6604; Shields, R. L., et al., J. Biol. Chem. 277 (2002) 26733-26740; Simmons, L. C., et al., J. Immunol. Methods 263 (2002) 133-147).
  • Methods to enhance cell-mediated effector functions of monoclonal antibodies are reported e.g. in WO 2005/018572, WO 2006/116260, WO 2006/114700, WO 2004/065540, WO 2005/011735, WO 2005/027966, WO 1997/028267, US 2006/0134709, US 2005/0054048, US 2005/0152894, WO 2003/035835, WO 2000/061739.
  • Surprisingly the bispecific <ErbB3-c-Met> antibodies which are one embodiment of the invention show reduced downregulation and internalization of target antigen compared to their parent <ErbB3> and/or <c-Met> antibodies. Therefore in one preferred embodiment of the invention, the bispecific antibody is glycosylated (if it comprises an Fc part of IgG1, IgG2, IgG3 or IgG4 subclass, preferably of IgG1 or IgG3 subclass) with a sugar chain at Asn297 whereby the amount of fucose within the sugar chain is 65% or lower (Numbering according to Kabat). In another embodiment is the amount of fucose within the sugar chain is between 5% and 65%, preferably between 20% and 40%. “Asn297” according to the invention means amino acid asparagine located at about position 297 in the Fc region. Based on minor sequence variations of antibodies, Asn297 can also be located some amino acids (usually not more than +3 amino acids) upstream or downstream of position 297, i.e. between position 294 and 300. In one embodiment the glycosylated antibody according to the invention the IgG subclass is of human IgG1 subclass, of human IgG1 subclass with the mutations L234A and L235A or of IgG3 subclass. In a further embodiment the amount of N-glycolylneuraminic acid (NGNA) is 1% or less and/or the amount of N-terminal alpha-1,3-galactose is 1% or less within the sugar chain. The sugar chain show preferably the characteristics of N-linked glycans attached to Asn297 of an antibody recombinantly expressed in a CHO cell.
  • The term “the sugar chains show characteristics of N-linked glycans attached to Asn297 of an antibody recombinantly expressed in a CHO cell” denotes that the sugar chain at Asn297 of the full length parent antibody according to the invention has the same structure and sugar residue sequence except for the fucose residue as those of the same antibody expressed in unmodified CHO cells, e.g. as those reported in WO 2006/103100.
  • The term “NGNA” as used within this application denotes the sugar residue N-glycolylneuraminic acid.
  • Glycosylation of human IgG1 or IgG3 occurs at Asn297 as core fucosylated biantennary complex oligosaccharide glycosylation terminated with up to two Gal residues. Human constant heavy chain regions of the IgG1 or IgG3 subclass are reported in detail by Kabat, E. A., et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md. (1991), and by Brüggemann, M., et al., J. Exp. Med. 166 (1987) 1351-1361; Love, T. W., et al., Methods Enzymol. 178 (1989) 515-527. These structures are designated as G0, G1 (α-1,6- or α-1,3-), or G2 glycan residues, depending from the amount of terminal Gal residues (Raju, T. S., Bioprocess Int. 1 (2003) 44-53). CHO type glycosylation of antibody Fc parts is e.g. described by Routier, F. H., Glycoconjugate J. 14 (1997) 201-207. Antibodies which are recombinantly expressed in non-glycomodified CHO host cells usually are fucosylated at Asn297 in an amount of at least 85%. The modified oligosaccharides of the full length parent antibody may be hybrid or complex. Preferably the bisected, reduced/not-fucosylated oligosaccharides are hybrid. In another embodiment, the bisected, reduced/not-fucosylated oligosaccharides are complex.
  • According to the invention “amount of fucose” means the amount of the sugar within the sugar chain at Asn297, related to the sum of all glycostructures attached to Asn297 (e.g. complex, hybrid and high mannose structures) measured by MALDI-TOF mass spectrometry and calculated as average value. The relative amount of fucose is the percentage of fucose-containing structures related to all glycostructures identified in an N-Glycosidase F treated sample (e.g. complex, hybrid and oligo- and high-mannose structures, resp.) by MALDI-TOF.
  • The antibody according to the invention is produced by recombinant means. Thus, one aspect of the current invention is a nucleic acid encoding the antibody according to the invention and a further aspect is a cell comprising the nucleic acid encoding an antibody according to the invention. Methods for recombinant production are widely known in the state of the art and comprise protein expression in prokaryotic and eukaryotic cells with subsequent isolation of the antibody and usually purification to a pharmaceutically acceptable purity. For the expression of the antibodies as aforementioned in a host cell, nucleic acids encoding the respective modified light and heavy chains are inserted into expression vectors by standard methods. Expression is performed in appropriate prokaryotic or eukaryotic host cells like CHO cells, NS0 cells, SP2/0 cells, HEK293 cells, COS cells, PER.C6 cells, yeast, or E. coli cells, and the antibody is recovered from the cells (supernatant or cells after lysis). General methods for recombinant production of antibodies are well-known in the state of the art and described, for example, in the review articles of Makrides, S. C., Protein Expr. Purif. 17 (1999) 183-202; Geisse, S., et al., Protein Expr. Purif. 8 (1996) 271-282; Kaufman, R. J., Mol. Biotechnol. 16 (2000) 151-160; Werner, R. G., Drug Res. 48 (1998) 870-880.
  • The trivalent, bispecific antibodies according to the invention are suitably separated from the culture medium by conventional immunoglobulin purification procedures such as, for example, protein A-Sepharose, hydroxylapatite chromatography, gel electrophoresis, dialysis, or affinity chromatography. DNA and RNA encoding the monoclonal antibodies is readily isolated and sequenced using conventional procedures. The hybridoma cells can serve as a source of such DNA and RNA. Once isolated, the DNA may be inserted into expression vectors, which are then transfected into host cells such as HEK 293 cells, CHO cells, or myeloma cells that do not otherwise produce immunoglobulin protein, to obtain the synthesis of recombinant monoclonal antibodies in the host cells.
  • Amino acid sequence variants (or mutants) of the trivalent, bispecific antibody are prepared by introducing appropriate nucleotide changes into the antibody DNA, or by nucleotide synthesis. Such modifications can be performed, however, only in a very limited range, e.g. as described above. For example, the modifications do not alter the above mentioned antibody characteristics such as the IgG isotype and antigen binding, but may improve the yield of the recombinant production, protein stability or facilitate the purification.
  • The term “host cell” as used in the current application denotes any kind of cellular system which can be engineered to generate the antibodies according to the current invention. In one embodiment HEK293 cells and CHO cells are used as host cells. As used herein, the expressions “cell,” “cell line,” and “cell culture” are used interchangeably and all such designations include progeny. Thus, the words “transformants” and “transformed cells” include the primary subject cell and cultures derived therefrom without regard for the number of transfers. It is also understood that all progeny may not be precisely identical in DNA content, due to deliberate or inadvertent mutations. Variant progeny that have the same function or biological activity as screened for in the originally transformed cell are included.
  • Expression in NS0 cells is described by, e.g., Barnes, L. M., et al., Cytotechnology 32 (2000) 109-123; Barnes, L. M., et al., Biotech. Bioeng. 73 (2001) 261-270. Transient expression is described by, e.g., Durocher, Y., et al., Nucl. Acids. Res. 30 (2002) E9. Cloning of variable domains is described by Orlandi, R., et al., Proc. Natl. Acad. Sci. USA 86 (1989) 3833-3837; Carter, P., et al., Proc. Natl. Acad. Sci. USA 89 (1992) 4285-4289; and Norderhaug, L., et al., J. Immunol. Methods 204 (1997) 77-87. A preferred transient expression system (HEK 293) is described by Schlaeger, E.-J., and Christensen, K., in Cytotechnology 30 (1999) 71-83 and by Schlaeger, E.-J., in J. Immunol. Methods 194 (1996) 191-199.
  • The control sequences that are suitable for prokaryotes, for example, include a promoter, optionally an operator sequence, and a ribosome binding site. Eukaryotic cells are known to utilize promoters, enhancers and polyadenylation signals.
  • A nucleic acid is “operably linked” when it is placed in a functional relationship with another nucleic acid sequence. For example, DNA for a pre-sequence or secretory leader is operably linked to DNA for a polypeptide if it is expressed as a pre-protein that participates in the secretion of the polypeptide; a promoter or enhancer is operably linked to a coding sequence if it affects the transcription of the sequence; or a ribosome binding site is operably linked to a coding sequence if it is positioned so as to facilitate translation. Generally, “operably linked” means that the DNA sequences being linked are contiguous, and, in the case of a secretory leader, contiguous and in reading frame. However, enhancers do not have to be contiguous. Linking is accomplished by ligation at convenient restriction sites. If such sites do not exist, the synthetic oligonucleotide adaptors or linkers are used in accordance with conventional practice.
  • Purification of antibodies is performed in order to eliminate cellular components or other contaminants, e.g. other cellular nucleic acids or proteins, by standard techniques, including alkaline/SDS treatment, CsCl banding, column chromatography, agarose gel electrophoresis, and others well known in the art. See Ausubel, F., et al., ed. Current Protocols in Molecular Biology, Greene Publishing and Wiley Interscience, New York (1987). Different methods are well established and widespread used for protein purification, such as affinity chromatography with microbial proteins (e.g. protein A or protein G affinity chromatography), ion exchange chromatography (e.g. cation exchange (carboxymethyl resins), anion exchange (amino ethyl resins) and mixed-mode exchange), thiophilic adsorption (e.g. with beta-mercaptoethanol and other SH ligands), hydrophobic interaction or aromatic adsorption chromatography (e.g. with phenyl-sepharose, aza-arenophilic resins, or m-aminophenylboronic acid), metal chelate affinity chromatography (e.g. with Ni(II)- and Cu(II)-affinity material), size exclusion chromatography, and electrophoretical methods (such as gel electrophoresis, capillary electrophoresis) (Vijayalakshmi, M. A., Appl. Biochem. Biotech. 75 (1998) 93-102).
  • One aspect of the invention is a pharmaceutical composition comprising an antibody according to the invention. Another aspect of the invention is the use of an antibody according to the invention for the manufacture of a pharmaceutical composition. A further aspect of the invention is a method for the manufacture of a pharmaceutical composition comprising an antibody according to the invention. In another aspect, the present invention provides a composition, e.g. a pharmaceutical composition, containing an antibody according to the present invention, formulated together with a pharmaceutical carrier.
  • One embodiment of the invention is the trivalent, bispecific antibody according to the invention for the treatment of cancer.
  • Another aspect of the invention is the pharmaceutical composition for the treatment of cancer.
  • Another aspect of the invention is the use of an antibody according to the invention for the manufacture of a medicament for the treatment of cancer.
  • Another aspect of the invention is method of treatment of patient suffering from cancer by administering an antibody according to the invention to a patient in the need of such treatment.
  • As used herein, “pharmaceutical carrier” includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible. Preferably, the carrier is suitable for intravenous, intramuscular, subcutaneous, parenteral, spinal or epidermal administration (e.g. by injection or infusion).
  • A composition of the present invention can be administered by a variety of methods known in the art. As will be appreciated by the skilled artisan, the route and/or mode of administration will vary depending upon the desired results. To administer a compound of the invention by certain routes of administration, it may be necessary to coat the compound with, or co-administer the compound with, a material to prevent its inactivation. For example, the compound may be administered to a subject in an appropriate carrier, for example, liposomes, or a diluent. Pharmaceutically acceptable diluents include saline and aqueous buffer solutions. Pharmaceutical carriers include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion. The use of such media and agents for pharmaceutically active substances is known in the art.
  • The phrases “parenteral administration” and “administered parenterally” as used herein means modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intra-arterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intra-articular, subcapsular, subarachnoid, intraspinal, epidural and intrasternal injection and infusion.
  • The term cancer as used herein refers to proliferative diseases, such as lymphomas, lymphocytic leukemias, lung cancer, non small cell lung (NSCL) cancer, bronchioloalviolar cell lung cancer, bone cancer, pancreatic cancer, skin cancer, cancer of the head or neck, cutaneous or intraocular melanoma, uterine cancer, ovarian cancer, rectal cancer, cancer of the anal region, stomach cancer, gastric cancer, colon cancer, breast cancer, uterine cancer, carcinoma of the fallopian tubes, carcinoma of the endometrium, carcinoma of the cervix, carcinoma of the vagina, carcinoma of the vulva, Hodgkin's Disease, cancer of the esophagus, cancer of the small intestine, cancer of the endocrine system, cancer of the thyroid gland, cancer of the parathyroid gland, cancer of the adrenal gland, sarcoma of soft tissue, cancer of the urethra, cancer of the penis, prostate cancer, cancer of the bladder, cancer of the kidney or ureter, renal cell carcinoma, carcinoma of the renal pelvis, mesothelioma, hepatocellular cancer, biliary cancer, neoplasms of the central nervous system (CNS), spinal axis tumors, brain stem glioma, glioblastoma multiforme, astrocytomas, schwanomas, ependymonas, medulloblastomas, meningiomas, squamous cell carcinomas, pituitary adenoma and Ewings sarcoma, including refractory versions of any of the above cancers, or a combination of one or more of the above cancers.
  • These compositions may also contain adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents. Prevention of presence of microorganisms may be ensured both by sterilization procedures, supra, and by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol, sorbic acid, and the like. It may also be desirable to include isotonic agents, such as sugars, sodium chloride, and the like into the compositions. In addition, prolonged absorption of the injectable pharmaceutical form may be brought about by the inclusion of agents which delay absorption such as aluminum monostearate and gelatin.
  • Regardless of the route of administration selected, the compounds of the present invention, which may be used in a suitable hydrated form, and/or the pharmaceutical compositions of the present invention, are formulated into pharmaceutically acceptable dosage forms by conventional methods known to those of skill in the art.
  • Actual dosage levels of the active ingredients in the pharmaceutical compositions of the present invention may be varied so as to obtain an amount of the active ingredient which is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient. The selected dosage level will depend upon a variety of pharmacokinetic factors including the activity of the particular compositions of the present invention employed, the route of administration, the time of administration, the rate of excretion of the particular compound being employed, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular compositions employed, the age, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors well known in the medical arts.
  • The composition must be sterile and fluid to the extent that the composition is deliverable by syringe. In addition to water, the carrier preferably is an isotonic buffered saline solution.
  • Proper fluidity can be maintained, for example, by use of coating such as lecithin, by maintenance of required particle size in the case of dispersion and by use of surfactants. In many cases, it is preferable to include isotonic agents, for example, sugars, polyalcohols such as mannitol or sorbitol, and sodium chloride in the composition.
  • As used herein, the expressions “cell,” “cell line,” and “cell culture” are used interchangeably and all such designations include progeny. Thus, the words “transformants” and “transformed cells” include the primary subject cell and cultures derived therefrom without regard for the number of transfers. It is also understood that all progeny may not be precisely identical in DNA content, due to deliberate or inadvertent mutations. Variant progeny that have the same function or biological activity as screened for in the originally transformed cell are included. Where distinct designations are intended, it will be clear from the context.
  • The term “transformation” as used herein refers to process of transfer of a vectors/nucleic acid into a host cell. If cells without formidable cell wall barriers are used as host cells, transfection is carried out e.g. by the calcium phosphate precipitation method as described by Graham, F. L., and van der Eb, A. J., Virology 52 (1973) 456-467. However, other methods for introducing DNA into cells such as by nuclear injection or by protoplast fusion may also be used. If prokaryotic cells or cells which contain substantial cell wall constructions are used, e.g. one method of transfection is calcium treatment using calcium chloride as described by Cohen, S. N, et al., PNAS 69 (1972) 2110-2114.
  • As used herein, “expression” refers to the process by which a nucleic acid is transcribed into mRNA and/or to the process by which the transcribed mRNA (also referred to as transcript) is subsequently being translated into peptides, polypeptides, or proteins. The transcripts and the encoded polypeptides are collectively referred to as gene product. If the polynucleotide is derived from genomic DNA, expression in a eukaryotic cell may include splicing of the mRNA.
  • A “vector” is a nucleic acid molecule, in particular self-replicating, which transfers an inserted nucleic acid molecule into and/or between host cells. The term includes vectors that function primarily for insertion of DNA or RNA into a cell (e.g., chromosomal integration), replication of vectors that function primarily for the replication of DNA or RNA, and expression vectors that function for transcription and/or translation of the DNA or RNA. Also included are vectors that provide more than one of the functions as described.
  • An “expression vector” is a polynucleotide which, when introduced into an appropriate host cell, can be transcribed and translated into a polypeptide. An “expression system” usually refers to a suitable host cell comprised of an expression vector that can function to yield a desired expression product.
  • Description of the Amino acid Sequences
    SEQ ID NO: 1 heavy chain variable domain <ErbB3> HER3 clone 29
    SEQ ID NO: 2 light chain variable domain <ErbB3> HER3 clone 29
    SEQ ID NO: 3 heavy chain variable domain <c-Met> Mab 5D5
    SEQ ID NO: 4 light chain variable domain <c-Met> Mab 5D5
    SEQ ID NO: 5 heavy chain <ErbB3> HER3 clone 29
    SEQ ID NO: 6 light chain <ErbB3> HER3 clone 29
    SEQ ID NO: 7 heavy chain <c-Met> Mab 5D5
    SEQ ID NO: 8 light chain <c-Met> Mab 5D5
    SEQ ID NO: 9 heavy chain <c-Met> Fab 5D5
    SEQ ID NO: 10 light chain <c-Met> Fab 5D5
    SEQ ID NO: 11 heavy chain 1 <ErbB3-c-Met> Her3/Met_KHSS
    SEQ ID NO: 12 heavy chain 2 <ErbB3-c-Met> Her3/Met_KHSS
    SEQ ID NO: 13 light chain <ErbB3-c-Met> Her3/Met_KHSS
    SEQ ID NO: 14 heavy chain 1 <ErbB3-c-Met> Her3/Met_SSKH
    SEQ ID NO: 15 heavy chain 2 <ErbB3-c-Met> Her3/Met_SSKH
    SEQ ID NO: 16 light chain <ErbB3-c-Met> Her3/Met_SSKH
    SEQ ID NO: 17 heavy chain 1 <ErbB3-c-Met> Her3/Met_SSKHSS
    SEQ ID NO: 18 heavy chain 2 <ErbB3-c-Met> Her3/Met_SSKHSS
    SEQ ID NO: 19 light chain <ErbB3-c-Met> Her3/Met_SSKHSS
    SEQ ID NO: 20 heavy chain 1 <ErbB3-c-Met> Her3/Met_1C
    SEQ ID NO: 21 heavy chain 2 <ErbB3-c-Met> Her3/Met_1C
    SEQ ID NO: 22 light chain <ErbB3-c-Met> Her3/Met_1C
    SEQ ID NO: 23 heavy chain 1 <ErbB3-c-Met> Her3/Met_6C
    SEQ ID NO: 24 heavy chain 2 <ErbB3-c-Met> Her3/Met_6C
    SEQ ID NO: 25 light chain <ErbB3-c-Met> Her3/Met_6C
    SEQ ID NO: 26 heavy chain constant region of human IgG1
    SEQ ID NO: 27 heavy chain constant region of human IgG1
    SEQ ID NO: 28 human light chain kappa constant region
    SEQ ID NO: 29 human light chain lambda constant region
  • The following examples, sequence listing and figures are provided to aid the understanding of the present invention, the true scope of which is set forth in the appended claims. It is understood that modifications can be made in the procedures set forth without departing from the spirit of the invention.
  • DESCRIPTION OF THE FIGURES
  • FIG. 1 Schematic structure of a full length antibody without CH4 domain specifically binding to a first antigen 1 with two pairs of heavy and light chain which comprise variable and constant domains in a typical order.
  • FIG. 2 Schematic representation of a trivalent, bispecific antibody according to the invention, comprising a full length antibody specifically binding to a first antigen 1 to which
  • a) FIG. 2 a two polypeptides VH and VL are fused (the VH and VL domains of both together forming a antigen binding site specifically binding to a second antigen 2;
    b) FIG. 2 b two polypeptides VH-CH1 and VL-CL are fused (the VH and VL domains of both together forming a antigen binding site specifically binding to a second antigen 2)
  • FIG. 3 Schematic representation of a trivalent, bispecific antibody according to the invention, comprising a full length antibody specifically binding to a first antigen 1 to which two polypeptides VH and VL are fused (the VH and VL domains of both together forming a antigen binding site specifically binding to a second antigen 2) with “knobs and holes”.
  • FIG. 4 Schematic representation of a trivalent, bispecific antibody according to the invention, comprising a full length antibody specifically binding to a first antigen 1 to which two polypeptides VH and VL are fused (the VH and VL domains of both together forming a antigen binding site specifically binding to a second antigen 2, wherein these VH and VL domains comprise an interchain disulfide bridge between positions VH44 and VL100) with “knobs and holes”.
  • FIG. 5 Binding of bispecific antibodies to the cell surface of cancer cells
  • FIG. 6 Inhibition of HGF-induced c-Met receptor phosphorylation by bispecific Her3/c-Met antibody formats
  • FIG. 7 Inhibition of HRG-induced Her3 receptor phosphorylation by bispecific Her3/c-Met antibody formats.
  • FIG. 8 Inhibition of HGF-induced HUVEC proliferation by bispecific Her3/c-Met antibody formats
  • FIG. 9 Inhibition of proliferation in the cancer cell line A431 by bispecific Her3/c-Met antibody formats.
  • FIG. 10 Analysis of inhibition of HGF-induced cell-cell dissemination (scattering) in the cancer cell line A431 by bispecific Her3/c-Met antibody formats.
  • EXAMPLES Experimental Procedure Materials & Methods
  • Recombinant DNA techniques
  • Standard methods were used to manipulate DNA as described in Sambrook, J. et al., Molecular cloning: A laboratory manual; Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1989. The molecular biological reagents were used according to the manufacturer's instructions.
  • DNA and Protein Sequence Analysis and Sequence Data Management
  • General information regarding the nucleotide sequences of human immunoglobulins light and heavy chains is given in: Kabat, E. A. et al., (1991) Sequences of Proteins of Immunological Interest, Fifth Ed., NIH Publication No 91-3242. Amino acids of antibody chains are numbered according to EU numbering (Edelman, G. M., et al., PNAS 63 (1969) 78-85; Kabat, E. A., et al., (1991) Sequences of Proteins of Immunological Interest, Fifth Ed., NIH Publication No 91-3242). The GCG's (Genetics Computer Group, Madison, Wis.) software package version 10.2 and Infomax's Vector NTI Advance suite version 8.0 was used for sequence creation, mapping, analysis, annotation and illustration.
  • DNA Sequencing
  • DNA sequences were determined by double strand sequencing performed at SequiServe (Vaterstetten, Germany) and Geneart AG (Regensburg, Germany).
  • Gene Synthesis
  • Desired gene segments were prepared by Geneart AG (Regensburg, Germany) from synthetic oligonucleotides and PCR products by automated gene synthesis. The gene segments which are flanked by singular restriction endonuclease cleavage sites were cloned into pGA18 (ampR) plasmids. The plasmid DNA was purified from transformed bacteria and concentration determined by UV spectroscopy. The DNA sequence of the subcloned gene fragments was confirmed by DNA sequencing. Gene Segments coding “knobs-into-hole” Her3 (clone 29) antibody heavy chain carrying a T366W mutation in the CH3 domain with a C-terminal 5D5 VH region linked by a (G4S)n peptide connector as well as “knobs-into-hole” Her3 (clone 29) antibody heavy chain carrying T366S, L368A and Y407V mutations with a C-terminal 5D5 VL region linked by a (G4S)n peptide connector were synthesized with 5′-BamHI and 3′-XbaI restriction sites. In a similar manner, DNA sequences coding “knobs-into-hole” Her3 (clone 29) antibody heavy chain carrying S354C and T366W mutations in the CH3 domain with a C-terminal 5D5 VH region linked by a (G4S)n peptide connector as well as “knobs-into-hole” Her3 (clone 29) antibody heavy chain carrying Y349C, T366S, L368A and Y407V mutations with a C-terminal 5D5 VL region linked by a (G4S)n peptide connector were prepared by gene synthesis with flanking BamHI and XbaI restriction sites. Finally, DNA sequenes encoding unmodified heavy and light chains of the Her3 (clone 29) and 5D5 antibody were synthesized with flanking BamHI and XbaI restriction sites. All constructs were designed with a 5′-end DNA sequence coding for a leader peptide (MGWSCIILFLVATATGVHS), which targets proteins for secretion in eukaryotic cells.
  • Construction of the Expression Plasmids
  • A Roche expression vector was used for the construction of all heavy VH/or VL fusion protein and light chain protein encoding expression plasmids. The vector is composed of the following elements:
      • a hygromycin resistance gene as a selection marker,
      • an origin of replication, oriP, of Epstein-Barr virus (EBV),
      • an origin of replication from the vector pUC18 which allows replication of this plasmid in E. coli
      • a beta-lactamase gene which confers ampicillin resistance in E. coli,
      • the immediate early enhancer and promoter from the human cytomegalovirus (HCMV),
      • the human 1-immunoglobulin polyadenylation (“poly A”) signal sequence, and
      • unique BamHI and XbaI restriction sites.
  • The immunoglobulin fusion genes comprising the heavy or light chain constructs as well as “knobs-into-hole” constructs with C-terminal VH and VL domains were prepared by gene synthesis and cloned into pGA18 (ampR) plasmids as described. The pG18 (ampR) plasmids carrying the synthesized DNA segments and the Roche expression vector were digested with BamHI and XbaI restriction enzymes (Roche Molecular Biochemicals) and subjected to agarose gel electrophoresis. Purified heavy and light chain coding DNA segments were then ligated to the isolated Roche expression vector BamHI/XbaI fragment resulting in the final expression vectors. The final expression vectors were transformed into E. coli cells, expression plasmid DNA was isolated (Miniprep) and subjected to restriction enzyme analysis and DNA sequencing. Correct clones were grown in 150 ml LB-Amp medium, again plasmid DNA was isolated (Maxiprep) and sequence integrity confirmed by DNA sequencing.
  • Transient Expression of Immunoglobulin Variants in HEK293 Cells
  • Recombinant immunoglobulin variants were expressed by transient transfection of human embryonic kidney 293-F cells using the FreeStyle™ 293 Expression System according to the manufacturer's instruction (Invitrogen, USA). Briefly, suspension FreeStyle™ 293-F cells were cultivated in FreeStyle™ 293 Expression medium at 37° C./8% CO2 and the cells were seeded in fresh medium at a density of 1-2×106 viable cells/ml on the day of transfection. DNA-293Fectin™ complexes were prepared in Opti-MEM® I medium (Invitrogen, USA) using 325 μA of 293Fectin™ (Invitrogen, Germany) and 250 μg of heavy and light chain plasmid DNA in a 1:1 molar ratio for a 250 ml final transfection volume. “Knobs-into-hole” DNA-293fectin complexes were prepared in Opti-MEM® I medium (Invitrogen, USA) using 325 μA of 293Fectin™ (Invitrogen, Germany) and 250 μg of “Knobs-into-hole” heavy chain 1 and 2 and light chain plasmid DNA in a 1:1:2 molar ratio for a 250 ml final transfection volume. Antibody containing cell culture supernatants were harvested 7 days after transfection by centrifugation at 14000 g for 30 minutes and filtered through a sterile filter (0.22 μm). Supernatants were stored at −20° C. until purification.
  • Purification of Trivalent Bispecific and Control Antibodies
  • Trivalent bispecific and control antibodies were purified from cell culture supernatants by affinity chromatography using Protein A-Sepharose™ (GE Healthcare, Sweden) and Superdex200 size exclusion chromatography. Briefly, sterile filtered cell culture supernatants were applied on a HiTrap ProteinA HP (5 ml) column equilibrated with PBS buffer (10 mM Na2HPO4, 1 mM KH2PO4, 137 mM NaCl and 2.7 mM KCl, pH 7.4). Unbound proteins were washed out with equilibration buffer. Antibody and antibody variants were eluted with 0.1 M citrate buffer, pH 2.8, and the protein containing fractions were neutralized with 0.1 ml 1 M Tris, pH 8.5. Then, the eluted protein fractions were pooled, concentrated with an Amicon Ultra centrifugal filter device (MWCO: 30 K, Millipore) to a volume of 3 ml and loaded on a Superdex200 HiLoad 120 ml 16/60 gel filtration column (GE Healthcare, Sweden) equilibrated with 20 mM Histidin, 140 mM NaCl, pH 6.0. Fractions containing purified bispecific and control antibodies with less than 5% high molecular weight aggregates were pooled and stored as 1.0 mg/ml aliquots at −80° C. Fab fragments were generated by a Papain digest of the purified 5D5 monoclonal antibody and subsequent removal of contaminating Fc domains by Protein A chromatography. Unbound Fab fragments were further purified on a Superdex200 HiLoad 120 ml 16/60 gel filtration column (GE Healthcare, Sweden) equilibrated with 20 mM Histidin, 140 mM NaCl, pH 6.0, pooled and stored as 1.0 mg/ml aliquots at −80° C.
  • Analysis of Purified Proteins
  • The protein concentration of purified protein samples was determined by measuring the optical density (OD) at 280 nm, using the molar extinction coefficient calculated on the basis of the amino acid sequence. Purity and molecular weight of bispecific and control antibodies were analyzed by SDS-PAGE in the presence and absence of a reducing agent (5 mM 1,4-dithiotreitol) and staining with Coomassie brilliant blue). The NuPAGE® Pre-Cast gel system (Invitrogen, USA) was used according to the manufacturer's instruction (4-20% Tris-Glycine gels). The aggregate content of bispecific and control antibody samples was analyzed by high-performance SEC using a Superdex 200 analytical size-exclusion column (GE Healthcare, Sweden) in 200 mM KH2PO4, 250 mM KCl, pH 7.0 running buffer at 25° C. 25 μg protein were injected on the column at a flow rate of 0.5 ml/min and eluted isocratic over 50 minutes. For stability analysis, concentrations of 1 mg/ml of purified proteins were incubated at 4° C. and 40° C. for 7 days and then evaluated by high-performance SEC. The integrity of the amino acid backbone of reduced bispecific antibody light and heavy chains was verified by NanoElectrospray Q-TOF mass spectrometry after removal of N-glycans by enzymatic treatment with Peptide-N-Glycosidase F (Roche Molecular Biochemicals).
  • c-Met Phosphorylation Assay
  • 5×10e5 A549 cells were seeded per well of a 6-well plate the day prior HGF stimulation in RPMI with 0.5% FCS (fetal calf serum). The next day, growth medium was replaced for one hour with RPMI containing 0.2% BSA (bovine serum albumine). 5 μg/mL of the bispecific antibody was then added to the medium and cells were incubated for 10 minutes upon which HGF was added for further 10 minutes in a final concentration of 50 μg/mL. Cells were washed once with ice cold PBS containing 1 mM sodium vanadate upon which they were placed on ice and lysed in the cell culture plate with 100 μL lysis buffer (50 mM Tris-Cl pH7.5, 150 mM NaCl, 1% NP40, 0.5% DOC, aprotinine, 0.5 mM PMSF, 1 mM sodium-vanadate). Cell lysates were transferred to eppendorf tubes and lysis was allowed to proceed for 30 minutes on ice. Protein concentration was determined using the BCA method (Pierce). 30-50 μg of the lysate was separated on a 4-12% Bis-Tris NuPage gel (Invitrogen) and proteins on the gel were transferred to a nitrocellulose membrane. Membranes were blocked for one hour with TBS-T containing 5% BSA and developed with a phospho-specific c-Met antibody directed against Y1230,1234,1235 (44-888, Biosource) according to the manufacturer's instructions. Immunoblots were reprobed with an antibody binding to unphosphorylated c-Met (AF276, R&D).
  • Her3 (ErbB3) Phosphorylation Assay
  • 2×10e5 MCF7 cells were seeded per well of a 12-well plate in complete growth medium (RPMI 1640, 10% FCS). Cells were allowed to grow to 90% confluency within two days. Medium was then replaced with starvation medium containing 0.5% FCS. The next day the respective antibodies were supplemented at the indicated concentrations 1 hour prior addition of 500 ng/mL Heregulin (R&D). Upon addition of Heregulin cells were cultivated further 10 minutes before the cells were harvested and lysed. Protein concentration was determined using the BCA method (Pierce). 30-50 μg of the lysate was separated on a 4-12% Bis-Tris NuPage gel (Invitrogen) and proteins on the gel were transferred to a nitrocellulose membrane. Membranes were blocked for one hour with TBS-T containing 5% BSA and developed with a phospho-specific Her3/ErbB3 antibody specifically recognizing Tyr1289 (4791, Cell Signaling).
  • Scatter Assay
  • A549 (4000 cells per well) or A431 (8000 cells per well) were seeded the day prior compound treatment in a total volume of 200 μL in 96-well E-Plates (Roche, 05232368001) in RPMI with 0.5% FCS. Adhesion and cell growth was monitored over night with the Real Time Cell Analyzer machine with sweeps every 15 min monitoring the impedance. The next day, cells were pre-incubated with 5 μL of the respective antibody dilutions in PBS with sweeps every five minutes. After 30 minutes 2.5 μL of a HGF solution yielding a final concentration of 20 ng/mL were added and the experiment was allowed to proceed for further 72 hours. Immediate changes were monitored with sweeps every minute for 180 minutes followed by sweeps every 15 minutes for the remainder of the time.
  • FACS a) Binding Assay
  • A431 were detached and counted. 1.5×10e5 cells were seeded per well of a conical 96-well plate. Cells were spun down (1500 rpm, 4° C., 5 min) and incubated for 30 min on ice in 50 μL of a dilution series of the respective bispecific antibody in PBS with 2% FCS (fetal calf serum). Cells were again spun down and washed once with 200 μL PBS containing 2% FCS followed by a second incubation of 30 min with a phycoerythrin-coupled antibody directed against human Fc which was diluted in PBS containing 2% FCS (Jackson Immunoresearch, 109116098). Cells were spun down washed twice with 200 μL PBS containing 2% FCS, resuspended in BD CellFix solution (BD Biosciences) and incubated for at least 10 min on ice. Mean fluorescence intensity (mfi) of the cells was determined by flow cytometry (FACS Canto, BD). Mfi was determined at least in duplicates of two independent stainings Flow cytometry spectra were further processed using the FlowJo software (TreeStar). Half-maximal binding was determined using XLFit 4.0 (IDBS) and the dose response one site model 205.
  • b) Internalization Assay
  • Cells were detached and counted. 5×10e5 cells were placed in 50 μL complete medium in an eppendorf tube and incubated with 5 μg/mL of the respective bispecific antibody at 37° C. After the indicated time points cells were stored on ice until the time course was completed. Afterwards, cells were transferred to FACS tubes, spun down (1500 rpm, 4° C., 5 min), washed with PBS+2% FCS and incubated for 30 minutes in 50 μL phycoerythrin-coupled secondary antibody directed against human Fc which was diluted in PBS containing 2% FCS (Jackson Immunoresearch, 109116098). Cells were again spun down, washed with PBS+2% FCS and fluorescence intensity was determined by flow cytometry (FACS Canto, BD).
  • c) Crosslinking Experiment
  • HT29 cells were detached counted and split in two populations which were individually stained with PKH26 and PKH67 (Sigma) according to the manufacturer's instructions. Of each of the stained populations 5×10e5 cells were taken, combined and incubated for 30 and 60 minutes with 10 μg/mL of the respective bispecific antibody in complete medium. After the indicated time points cells were stored on ice until the time course was completed. Cells were spun down (1500 rpm, 4° C., 5 min), washed with PBS+2% FCS and fluorescence intensity was determined by flow cytometry (FACS Canto, BD).
  • Cell Titer Glow Assay
  • Cell viability and proliferation was quantified using the cell titer glow assay (Promega). The assay was performed according to the manufacturer's instructions. Briefly, cells were cultured in 96-well plates in a total volume of 100 μL for the desired period of time. For the proliferation assay, cells were removed from the incubator and placed at room temperature for 30 min. 100 μL of cell titer glow reagent were added and multi-well plates were placed on an orbital shaker for 2 min. Luminescence was quantified after 15 min on a microplate reader (Tecan).
  • Wst-1 Assay
  • A Wst-1 viability and cell proliferation assay was performed as endpoint analysis, detecting the number of metabolic active cells. Briefly, 20 μL of Wst-1 reagent (Roche, 11644807001) were added to 200 μL of culture medium. 96-well plates were further incubated for 30 min to 1 h until robust development of the dye. Staining intensity was quantified on a microplate reader (Tecan) at a wavelength of 450 nm.
  • Design of Expressed and Purified Trivalent, Bispecific <ErbB3-c-Met> Antibodies
  • In Table 1: Trivalent, bispecific <ErbB3-c-Met> antibodies based on a full length ErbB-3 antibody (HER3 clone29) and the VH and VL domain from a C-met antibody (c-Met 5D5) with the respective features shown in Table 1 one were expressed and purified according to the general methods described above. The corresponding VH and VL of HER3 clone29 and c-Met 5D5 are given in the sequence listing.
  • TABLE 1
    Trivalent, bispecific antibody <ErbB3-c-Met> with the VHVL-Ab-
    nomenclature in Table 1 were expressed and purified (see also in the Examples
    below and FIG. 3c)
    Molecule Name
    VHVL-Ab-
    nomenclature
    for bispecific
    antibodies Her3/Met_KHSS Her3/Met_SSKH Her3/Met_SSKHSS Her3/Met_1C Her3/Met_6C
    Features:
    Knobs-in-hole S354C:T366W/ T366W/ S354C:T366W/ S354C:T366W/ S354C:T366W/
    mutations Y349′C:T366′S: T366′S:L368′A: Y349′C:T366′S: Y349′C:T366′S: Y349′C:T366′S:
    L368′A:Y407′V Y407′V L368′A:Y407′V L368′A:Y407′V L368′A:Y407′V
    Full length Her3 Her3 Her3 Her3 Her3
    antibody clone 29 clone 29 clone 29 clone 29 clone 29
    backbone (chimeric) (chimeric) (chimeric) (chimeric) (chimeric)
    derived from
    VHVL cMet 5D5 cMet 5D5 cMet 5D5 cMet 5D5 cMet 5D5
    fragment (humanized) (humanized) (humanized) (humanized) (humanized)
    derived from
    Position of VH C-terminus C-terminus C-terminus C-terminus knob C-terminus
    attached to knob heavy knob heavy knob heavy heavy chain knob heavy
    antibody chain chain chain chain
    Position of VL C-terminus C-terminus C-terminus C-terminus hole C-terminus
    attached to hole heavy hole heavy hole heavy heavy chain hole heavy
    antibody chain chain chain chain
    Peptide (G4S)3 (G4S)3 (G4S)3 (G4S)1 (G4S)6
    connector
    VHVL disulfide + +
    VH44/VL100
    stabilized
  • Example 1 FIG. 5 Binding of Bispecific Antibodies to the Cell Surface of Cancer Cells
  • The binding properties of the bispecific antibodies to their respective receptor on the cell surface was analyzed on A431 cancer cells in a flow cytometry based assay. Cells were incubated with the mono- or bispecific primary antibodies and binding of these antibodies to their cognate receptors was detected with a secondary antibody coupled to a fluorophore binding specifically to the Fc of the primary antibody. The mean fluorescence intensity of a dilution series of the primary antibodies was plotted against the concentration of the antibody to obtain a sigmoidal binding curve. Cell surface expression of c-Met and Her3 was validated by incubation with the bivalent 5D5 and Her3 clone 29 antibody only. The Her3/c-Met_KHSS antibody readily binds to the cell surface of A431. Under these experimental settings, the antibody can only bind via its Her3 part and consequently the mean fluorescence intensity does not exceed the staining for Her3 clone 29 alone.
  • Example 2 FIG. 6 Inhibition of HGF-Induced c-Met Receptor Phosphorylation by Bispecific Her3/c-Met antibody formats
  • To confirm functionality of the c-Met part in the bispecific antibodies a c-Met phosphorylation assay was performed. In this experiment A549 lung cancer cells or HT29 colorectal cancer cells were treated with the bispecific antibodies or control antibodies prior exposure to HGF. Cells were then lysed and phosphorylation of the c-Met receptor was examined. Both cell lines can be stimulated with HGF as can be observed by the occurrence of a phospho-c-Met specific band in the immunoblot.
  • Example 3 FIG. 6 Inhibition of HRG-Induced Her3 Receptor Phosphorylation by Bispecific Her3/c-Met Antibody Formats
  • To confirm functionality of the Her3 part in the bispecific antibodies a Her3 phosphorylation assay was performed. In this experiment MCF7 cells were treated with the bispecific antibodies or control antibodies prior exposure to HRG (Heregulin). Cells were then lysed and phosphorylation of the Her3 receptor was examined. Her3/c-Met_KHSS inhibit Her3 receptor phosphorylation to the same extent as the parental Her3 clone29 indicating that Her3 binding and functionality of the antibody are not compromised by the trivalent antibody format.
  • Example 4 FIG. 8 Inhibition of HGF-Induced HUVEC Proliferation by Bispecific Her3/c-Met Antibody Formats
  • HUVEC proliferation assays were performed to demonstrate the mitogenic effect of HGF. Addition of HGF to HUVEC leads to a twofold increase in proliferation. Addition of human IgG control antibody in the same concentration range as the bispecific antibodies has no impact on cellular proliferation while the 5D5 Fab fragment inhibits HGF-induced proliferation. Titration of Her3/c-Met_KHSS demonstrate a weak inhibitory effect of the antibody (FIG. 8). The effect is more pronounced for the Her3/Met-6C antibody indicating that a longer connector improves efficacy of the antibody. This demonstrates the functionality of the c-Met component in the trivalent antibody format.
  • Example 5 FIG. 9 Inhibition of Proliferation in the Cancer Cell Line A431 by Bispecific Her3/c-Met Antibody Formats
  • If A431 were seeded in serum reduced medium, addition of HGF induces apart from scattering a weak mitogenic effect. This was exploited to analyze the impact of Her3/c-Met_KHSS on HGF treated A431 proliferation. Indeed, the bispecific antibodies can largely inhibit the HGF-induced increase of proliferation (15%). A control human IgG1 antibody has no influence on HGF promoted A431 cell growth.
  • Example 6 FIG. 10 Analysis of Inhibition of HGF-Induced Cell-Cell Dissemination (Scattering) in the Cancer Cell Line A431 by Bispecific Her3/c-Met Antibody Formats
  • HGF-induced scattering includes morphological changes of the cell, resulting in rounding of the cells, filopodia-like protrusions, spindle-like structures and a certain motility of the cells. The Real Time Cell Analyzer (Roche) measures the impedance of a given cell culture well and can therefore indirectly monitor changes in cellular morphology and proliferation. Addition of HGF to A431 and A549 cells resulted in changes of the impedance which was monitored as function of time. Her3/c-Met_KHSS and Her3/Met-6C inhibited HGF-induced scattering with Her3/Met-6C being more efficacious (20.7% and 43.7% scatter inhibition) (FIG. 10).

Claims (18)

1. A trivalent, bispecific antibody comprising
a) a full length antibody that specifically binds to a first antigen wherein the full length antibody consists of two antibody heavy chains and two antibody light chains;
b) a polypeptide consisting of
ba) an antibody heavy chain variable domain (VH); or
bb) an antibody heavy chain variable domain (VH) and an antibody constant domain 1 (CH1), wherein the N-terminus of the VH domain of the polypeptide is fused via a peptide connector to the C-terminus of one of the two heavy chains of the full length antibody;
c) a polypeptide consisting of
ca) an antibody light chain variable domain (VL), or
cb) an antibody light chain variable domain (VL) and an antibody light chain constant domain (CL);
wherein N-terminus of the VL domain of the polypeptide is fused via a peptide connector to the C-terminus of the other of the two heavy chains of the full length antibody;
and wherein the antibody heavy chain variable domain (VH) of the polypeptide under b) and the antibody light chain variable domain (VL) of the polypeptide under c) together form an antigen-binding site specifically binding to a second antigen.
2. The trivalent, bispecific antibody according to claim 1, wherein
the CH3 domain of one heavy chain and the CH3 domain of the other heavy chain each meet at an interface which comprises an alteration in the original interface between the antibody CH3 domains;
wherein i) in the CH3 domain of one heavy chain
an amino acid residue is replaced with an amino acid residue having a larger side chain volume, thereby generating a protuberance within the interface of the CH3 domain of one heavy chain which is positionable in a cavity within the interface of the CH3 domain of the other heavy chain and wherein
ii) in the CH3 domain of the other heavy chain
an amino acid residue is replaced with an amino acid residue having a smaller side chain volume, thereby generating a cavity within the interface of the second CH3 domain within which a protuberance within the interface of the first CH3 domain is positionable.
3. The trivalent, bispecific antibody according to claim 2, wherein
i) the amino acid residue having a larger side chain volume is selected from the group consisting of arginine (R), phenylalanine (F), tyrosine (Y), tryptophan (W); and
ii) the amino acid residue having a smaller side chain volume is selected from the group consisting of alanine (A), serine (S), threonine (T), valine (V).
4. The trivalent, bispecific antibody according to claim 3, wherein
both CH3 domains are further altered by the introduction of cysteine as an amino acid in each CH3 domain such that a disulfide bridge between both CH3 domains can be formed.
5. The trivalent, bispecific antibody according to claim 4, wherein
the CH3 domain under i) comprises a T366W mutation; and
the CH3 domain under ii) comprises T366S, L368A, and Y407V mutations.
6. The trivalent, bispecific antibody according to claim 5, wherein
the CH3 domain under i) comprises Y349C and T366W mutations; and
the CH3 domain under ii) comprises S354C, T366S, L368A, and Y407V mutations.
7. The trivalent, bispecific antibody according to claim 4, wherein
the antibody heavy chain variable domain (VH) of the polypeptide under b) and the antibody light chain variable domain (VL) of the polypeptide under c) are linked and stabilized via a interchain disulfide bridge by introduction of a disulfide bond between the following positions:
i) heavy chain variable domain position 44 to light chain variable domain position 100,
ii) heavy chain variable domain position 105 to light chain variable domain position 43, or
iii) heavy chain variable domain position 101 to light chain variable domain position 100.
8. The trivalent, bispecific antibody according to claim 7, wherein
the antibody heavy chain variable domain (VH) of the polypeptide under b) and the antibody light chain variable domain (VL) of the polypeptide under c) are linked and stabilized via a interchain disulfide bridge by introduction of a disulfide bond between the following positions:
i) heavy chain variable domain position 44 to light chain variable domain position 100.
9. The trivalent, bispecific antibody according to claim 6, wherein the peptide connectors under b) and c) are identical peptides with a length between 25 and 50 amino acids.
10. A pharmaceutical composition comprising a trivalent, bispecific antibody according to claim 1.
11. A nucleic acid encoding a trivalent, bispecific antibody according to claim 2.
12. A nucleic acid encoding a trivalent, bispecific antibody according to claim 3.
13. A nucleic acid encoding a trivalent, bispecific antibody according to claim 4.
14. A nucleic acid encoding a trivalent, bispecific antibody according to claim 5.
15. A nucleic acid encoding a trivalent, bispecific antibody according to claim 6.
16. A nucleic acid encoding a trivalent, bispecific antibody according to claim 7.
17. A nucleic acid encoding a trivalent, bispecific antibody according to claim 8.
18. A method for treating a human suffering from cancer by administering an effective amount of the trivalent, bispecific antibody according to claim 8 to a human in need of such treatment.
US12/752,216 2009-04-07 2010-04-01 Trivalent, bispecific antibodies Abandoned US20100256340A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US13/568,224 US9890204B2 (en) 2009-04-07 2012-08-07 Trivalent, bispecific antibodies
US15/857,473 US20180282399A1 (en) 2009-04-07 2017-12-28 Trivalent, bispecific antibodies
US16/413,469 US11993642B2 (en) 2009-04-07 2019-05-15 Trivalent, bispecific antibodies

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP09005108 2009-04-07
EP09005108.7 2009-04-07

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US13/568,224 Continuation US9890204B2 (en) 2009-04-07 2012-08-07 Trivalent, bispecific antibodies

Publications (1)

Publication Number Publication Date
US20100256340A1 true US20100256340A1 (en) 2010-10-07

Family

ID=40942481

Family Applications (4)

Application Number Title Priority Date Filing Date
US12/752,216 Abandoned US20100256340A1 (en) 2009-04-07 2010-04-01 Trivalent, bispecific antibodies
US13/568,224 Active US9890204B2 (en) 2009-04-07 2012-08-07 Trivalent, bispecific antibodies
US15/857,473 Abandoned US20180282399A1 (en) 2009-04-07 2017-12-28 Trivalent, bispecific antibodies
US16/413,469 Active 2031-05-27 US11993642B2 (en) 2009-04-07 2019-05-15 Trivalent, bispecific antibodies

Family Applications After (3)

Application Number Title Priority Date Filing Date
US13/568,224 Active US9890204B2 (en) 2009-04-07 2012-08-07 Trivalent, bispecific antibodies
US15/857,473 Abandoned US20180282399A1 (en) 2009-04-07 2017-12-28 Trivalent, bispecific antibodies
US16/413,469 Active 2031-05-27 US11993642B2 (en) 2009-04-07 2019-05-15 Trivalent, bispecific antibodies

Country Status (21)

Country Link
US (4) US20100256340A1 (en)
EP (1) EP2417156B1 (en)
JP (2) JP5616428B2 (en)
KR (1) KR101456326B1 (en)
CN (1) CN102369214B (en)
AR (1) AR076193A1 (en)
AU (1) AU2010234031B2 (en)
BR (1) BRPI1010297A2 (en)
CA (1) CA2757931C (en)
CY (1) CY1116376T1 (en)
DK (1) DK2417156T3 (en)
ES (1) ES2537100T3 (en)
HR (1) HRP20150444T1 (en)
IL (1) IL214884A (en)
MX (1) MX2011010168A (en)
PL (1) PL2417156T3 (en)
PT (1) PT2417156E (en)
SG (1) SG175077A1 (en)
SI (1) SI2417156T1 (en)
TW (1) TW201039850A (en)
WO (1) WO2010115589A1 (en)

Cited By (70)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100256339A1 (en) * 2009-04-07 2010-10-07 Birgit Bossenmaier Bispecific Anti ErbB3 / Anti cMet Antibodies
US20100322934A1 (en) * 2009-06-18 2010-12-23 Sabine Imhof-Jung Bispecific, Tetravalent Antigen Binding Proteins
WO2012059858A1 (en) * 2010-11-01 2012-05-10 Symphogen A/S Anti-her3 antibodies and compositions
WO2012123949A1 (en) 2011-03-17 2012-09-20 Ramot At Tel-Aviv University Ltd. Bi- and monospecific, asymmetric antibodies and methods of generating the same
EP2722343A1 (en) * 2011-06-20 2014-04-23 Kyowa Hakko Kirin Co., Ltd. ANTI-erbB3 ANTIBODY
WO2014108854A1 (en) 2013-01-09 2014-07-17 Fusimab Ltd. Monospecific anti-hgf and anti-ang2 antibodies and bispecific anti-hgf/anti-ang2 antibodies
EP2794905A4 (en) * 2011-12-20 2015-11-18 Medimmune Llc Modified polypeptides for bispecific antibody scaffolds
WO2015175375A1 (en) 2014-05-13 2015-11-19 Short Jay M Conditionally active biological proteins
WO2015173756A3 (en) * 2014-05-16 2016-01-28 Pfizer Inc. Bispecific antibodies with engineered ch1-cl interfaces
US9382323B2 (en) 2009-04-02 2016-07-05 Roche Glycart Ag Multispecific antibodies comprising full length antibodies and single chain fab fragments
US9493578B2 (en) 2009-09-02 2016-11-15 Xencor, Inc. Compositions and methods for simultaneous bivalent and monovalent co-engagement of antigens
US9605061B2 (en) 2010-07-29 2017-03-28 Xencor, Inc. Antibodies with modified isoelectric points
US9605084B2 (en) 2013-03-15 2017-03-28 Xencor, Inc. Heterodimeric proteins
US9650446B2 (en) 2013-01-14 2017-05-16 Xencor, Inc. Heterodimeric proteins
US9676845B2 (en) 2009-06-16 2017-06-13 Hoffmann-La Roche, Inc. Bispecific antigen binding proteins
US9688758B2 (en) 2012-02-10 2017-06-27 Genentech, Inc. Single-chain antibodies and other heteromultimers
US9701759B2 (en) 2013-01-14 2017-07-11 Xencor, Inc. Heterodimeric proteins
US9738722B2 (en) 2013-01-15 2017-08-22 Xencor, Inc. Rapid clearance of antigen complexes using novel antibodies
US9822186B2 (en) 2014-03-28 2017-11-21 Xencor, Inc. Bispecific antibodies that bind to CD38 and CD3
US9850320B2 (en) 2014-11-26 2017-12-26 Xencor, Inc. Heterodimeric antibodies to CD3 X CD20
US9856327B2 (en) 2014-11-26 2018-01-02 Xencor, Inc. Heterodimeric antibodies to CD3 X CD123
WO2018002358A1 (en) 2016-06-30 2018-01-04 F. Hoffmann-La Roche Ag Improved adoptive t-cell therapy
US9879095B2 (en) 2010-08-24 2018-01-30 Hoffman-La Roche Inc. Bispecific antibodies comprising a disulfide stabilized-Fv fragment
US9890204B2 (en) 2009-04-07 2018-02-13 Hoffmann-La Roche Inc. Trivalent, bispecific antibodies
US9975966B2 (en) 2014-09-26 2018-05-22 Chugai Seiyaku Kabushiki Kaisha Cytotoxicity-inducing theraputic agent
US9982036B2 (en) 2011-02-28 2018-05-29 Hoffmann-La Roche Inc. Dual FC antigen binding proteins
US9994646B2 (en) 2009-09-16 2018-06-12 Genentech, Inc. Coiled coil and/or tether containing protein complexes and uses thereof
US10106612B2 (en) 2012-06-27 2018-10-23 Hoffmann-La Roche Inc. Method for selection and production of tailor-made highly selective and multi-specific targeting entities containing at least two different binding entities and uses thereof
US10106600B2 (en) 2010-03-26 2018-10-23 Roche Glycart Ag Bispecific antibodies
US10106624B2 (en) 2013-03-15 2018-10-23 Xencor, Inc. Heterodimeric proteins
US10131710B2 (en) 2013-01-14 2018-11-20 Xencor, Inc. Optimized antibody variable regions
US10138293B2 (en) 2007-12-21 2018-11-27 Hoffmann-La Roche, Inc. Bivalent, bispecific antibodies
US10227410B2 (en) 2015-12-07 2019-03-12 Xencor, Inc. Heterodimeric antibodies that bind CD3 and PSMA
US10227411B2 (en) 2015-03-05 2019-03-12 Xencor, Inc. Modulation of T cells with bispecific antibodies and FC fusions
US10316088B2 (en) 2016-06-28 2019-06-11 Xencor, Inc. Heterodimeric antibodies that bind somatostatin receptor 2
US10323099B2 (en) 2013-10-11 2019-06-18 Hoffmann-La Roche Inc. Multispecific domain exchanged common variable light chain antibodies
US10428155B2 (en) 2014-12-22 2019-10-01 Xencor, Inc. Trispecific antibodies
WO2019199916A1 (en) * 2018-04-13 2019-10-17 Eli Lilly And Company Fab-based trispecific antibodies
US10487155B2 (en) 2013-01-14 2019-11-26 Xencor, Inc. Heterodimeric proteins
US10501543B2 (en) 2016-10-14 2019-12-10 Xencor, Inc. IL15/IL15Rα heterodimeric Fc-fusion proteins
US10513699B2 (en) 2014-09-03 2019-12-24 Bioatla, Llc Discovering and producing conditionally active biologic proteins in the same eukaryotic cell production hosts
US10519242B2 (en) 2013-03-15 2019-12-31 Xencor, Inc. Targeting regulatory T cells with heterodimeric proteins
US10526413B2 (en) 2015-10-02 2020-01-07 Hoffmann-La Roche Inc. Bispecific antibodies specific for OX40
US10526417B2 (en) 2014-11-26 2020-01-07 Xencor, Inc. Heterodimeric antibodies that bind CD3 and CD38
US10544187B2 (en) 2013-03-15 2020-01-28 Xencor, Inc. Targeting regulatory T cells with heterodimeric proteins
US10611825B2 (en) 2011-02-28 2020-04-07 Hoffmann La-Roche Inc. Monovalent antigen binding proteins
US10633457B2 (en) 2014-12-03 2020-04-28 Hoffmann-La Roche Inc. Multispecific antibodies
US10633458B2 (en) 2018-04-10 2020-04-28 Y-Biologics Inc. Cell engaging binding molecules
US10787518B2 (en) 2016-06-14 2020-09-29 Xencor, Inc. Bispecific checkpoint inhibitor antibodies
US10793632B2 (en) 2016-08-30 2020-10-06 Xencor, Inc. Bispecific immunomodulatory antibodies that bind costimulatory and checkpoint receptors
US10851178B2 (en) 2011-10-10 2020-12-01 Xencor, Inc. Heterodimeric human IgG1 polypeptides with isoelectric point modifications
US10858417B2 (en) 2013-03-15 2020-12-08 Xencor, Inc. Heterodimeric proteins
US10968276B2 (en) 2013-03-12 2021-04-06 Xencor, Inc. Optimized anti-CD3 variable regions
US10981992B2 (en) 2017-11-08 2021-04-20 Xencor, Inc. Bispecific immunomodulatory antibodies that bind costimulatory and checkpoint receptors
US10982006B2 (en) 2018-04-04 2021-04-20 Xencor, Inc. Heterodimeric antibodies that bind fibroblast activation protein
WO2021113748A1 (en) * 2019-12-05 2021-06-10 Arbele Corp. Composition of triaxial antibodies and method of making and using thereof
US11053316B2 (en) 2013-01-14 2021-07-06 Xencor, Inc. Optimized antibody variable regions
US11084863B2 (en) 2017-06-30 2021-08-10 Xencor, Inc. Targeted heterodimeric Fc fusion proteins containing IL-15 IL-15alpha and antigen binding domains
US11312770B2 (en) 2017-11-08 2022-04-26 Xencor, Inc. Bispecific and monospecific antibodies using novel anti-PD-1 sequences
US11319355B2 (en) 2017-12-19 2022-05-03 Xencor, Inc. Engineered IL-2 Fc fusion proteins
US11358999B2 (en) 2018-10-03 2022-06-14 Xencor, Inc. IL-12 heterodimeric Fc-fusion proteins
US11421022B2 (en) 2012-06-27 2022-08-23 Hoffmann-La Roche Inc. Method for making antibody Fc-region conjugates comprising at least one binding entity that specifically binds to a target and uses thereof
US11472890B2 (en) 2019-03-01 2022-10-18 Xencor, Inc. Heterodimeric antibodies that bind ENPP3 and CD3
US11505595B2 (en) 2018-04-18 2022-11-22 Xencor, Inc. TIM-3 targeted heterodimeric fusion proteins containing IL-15/IL-15RA Fc-fusion proteins and TIM-3 antigen binding domains
US11524991B2 (en) 2018-04-18 2022-12-13 Xencor, Inc. PD-1 targeted heterodimeric fusion proteins containing IL-15/IL-15Ra Fc-fusion proteins and PD-1 antigen binding domains and uses thereof
US11591401B2 (en) 2020-08-19 2023-02-28 Xencor, Inc. Anti-CD28 compositions
US11618790B2 (en) 2010-12-23 2023-04-04 Hoffmann-La Roche Inc. Polypeptide-polynucleotide-complex and its use in targeted effector moiety delivery
US11739144B2 (en) 2021-03-09 2023-08-29 Xencor, Inc. Heterodimeric antibodies that bind CD3 and CLDN6
US11859012B2 (en) 2021-03-10 2024-01-02 Xencor, Inc. Heterodimeric antibodies that bind CD3 and GPC3
US11919956B2 (en) 2020-05-14 2024-03-05 Xencor, Inc. Heterodimeric antibodies that bind prostate specific membrane antigen (PSMA) and CD3

Families Citing this family (215)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2592271T3 (en) 2005-03-31 2016-11-29 Chugai Seiyaku Kabushiki Kaisha Polypeptide production methods by regulating the association of polypeptides
EP4342995A3 (en) 2006-03-31 2024-05-15 Chugai Seiyaku Kabushiki Kaisha Methods for controlling blood pharmacokinetics of antibodies
EP4218801A3 (en) 2006-03-31 2023-08-23 Chugai Seiyaku Kabushiki Kaisha Antibody modification method for purifying bispecific antibody
AR068563A1 (en) 2007-09-26 2009-11-18 Chugai Pharmaceutical Co Ltd CONSTANT MUTANT ANTIBODY REGION
WO2009041643A1 (en) 2007-09-26 2009-04-02 Chugai Seiyaku Kabushiki Kaisha Method of modifying isoelectric point of antibody via amino acid substitution in cdr
WO2010107110A1 (en) 2009-03-19 2010-09-23 中外製薬株式会社 Antibody constant region variant
EP3674317A1 (en) 2009-03-19 2020-07-01 Chugai Seiyaku Kabushiki Kaisha Antibody constant region variant
WO2011037158A1 (en) 2009-09-24 2011-03-31 中外製薬株式会社 Modified antibody constant regions
EP2543730B1 (en) 2010-03-04 2018-10-31 Chugai Seiyaku Kabushiki Kaisha Antibody constant region variant
WO2011147834A1 (en) 2010-05-26 2011-12-01 Roche Glycart Ag Antibodies against cd19 and uses thereof
KR20150013949A (en) 2010-07-19 2015-02-05 에프. 호프만-라 로슈 아게 Method to identify a patient with an increased likelihood of responding to an anti-cancer therapy
EP2596362A1 (en) 2010-07-19 2013-05-29 F. Hoffmann-La Roche AG Method to identify a patient with an increased likelihood of responding to an anti-cancer therapy
WO2012010582A1 (en) 2010-07-21 2012-01-26 Roche Glycart Ag Anti-cxcr5 antibodies and methods of use
BR112013002532A2 (en) 2010-08-05 2016-05-31 Hoffmann La Roche anti-mhc antibody anti-viral cytokine fusion protein
PT2644698T (en) 2010-11-17 2018-01-31 Chugai Pharmaceutical Co Ltd Multi-specific antigen-binding molecule having alternative function to function of blood coagulation factor viii
US10689447B2 (en) 2011-02-04 2020-06-23 Genentech, Inc. Fc variants and methods for their production
MX355255B (en) * 2011-02-04 2018-04-11 Genentech Inc Fc VARIANTS AND METHODS FOR THEIR PRODUCTION.
CA2828662A1 (en) 2011-04-20 2012-10-26 Roche Glycart Ag Method and constructs for the ph dependent passage of the blood-brain-barrier
AR086924A1 (en) 2011-06-15 2014-01-29 Hoffmann La Roche HUMAN EPO ANTI-RECEIVER ANTIBODIES AND THE METHODS FOR USE
AR086982A1 (en) 2011-06-22 2014-02-05 Hoffmann La Roche ELIMINATION OF DIANA CELLS BY SPECIFIC CYTOTOXIC T-CELLS OF VIRUSES USING COMPLEXES THAT INCLUDE MHC CLASS I
EP2748202B1 (en) * 2011-08-23 2018-07-04 Roche Glycart AG Bispecific antigen binding molecules
MX356947B (en) 2011-08-23 2018-06-20 Roche Glycart Ag Bispecific antibodies specific for t-cell activating antigens and a tumor antigen and methods of use.
US20130078250A1 (en) * 2011-08-23 2013-03-28 Oliver Ast Bispecific t cell activating antigen binding molecules
LT2748201T (en) * 2011-08-23 2018-02-26 Roche Glycart Ag Bispecific t cell activating antigen binding molecules
RU2014114119A (en) 2011-09-23 2015-10-27 Рош Гликарт Аг BSPECIFIC ANTI-EGFR / ANTI IGF-1R-ANTIBODIES
ES2732712T3 (en) 2011-10-31 2019-11-25 Chugai Pharmaceutical Co Ltd Antigen binding molecule that has a regulated conjugation between the heavy chain and the light chain
MX349003B (en) 2011-12-19 2017-07-06 Hoffmann La Roche Method for the detection of free binding partner of a multispecific binder.
US9963511B2 (en) 2011-12-22 2018-05-08 Hoffmann-La Roche Inc. Expression vector organization, novel production cell generation methods and their use for the recombinant production of polypeptides
EP2794651B1 (en) 2011-12-22 2022-09-21 F. Hoffmann-La Roche AG Expression vector element combinations, novel production cell generation methods and their use for the recombinant production of polypeptides
SG11201403445YA (en) 2011-12-22 2014-07-30 Hoffmann La Roche Full length antibody display system for eukaryotic cells and its use
JP2015507193A (en) 2012-02-01 2015-03-05 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft Method for detecting binding partners of multispecific binders
RU2693264C2 (en) 2012-02-03 2019-07-01 Ф.Хоффман-Ля Рош Аг Bispecific antibody molecules and antigen transfected t-cells, and their use in medicine
ES2676031T3 (en) 2012-02-15 2018-07-16 F. Hoffmann-La Roche Ag Affinity chromatography based on the Fc receptor
WO2013150043A1 (en) 2012-04-05 2013-10-10 F. Hoffmann-La Roche Ag Bispecific antibodies against human tweak and human il17 and uses thereof
HUE029435T2 (en) 2012-07-04 2017-02-28 Hoffmann La Roche Anti-theophylline antibodies and methods of use
KR20150030755A (en) 2012-07-04 2015-03-20 에프. 호프만-라 로슈 아게 Anti-biotin antibodies and methods of use
WO2014006124A1 (en) 2012-07-04 2014-01-09 F. Hoffmann-La Roche Ag Covalently linked antigen-antibody conjugates
WO2014009474A1 (en) 2012-07-13 2014-01-16 F. Hoffmann-La Roche Ag Method for the detection of a multispecific binder
BR112015003938A2 (en) 2012-09-14 2018-09-04 Hoffmann La Roche polypeptide production methods, multispecific binder production, multispecific binder selection, bispecific antibody selection and combination determination, bispecific antibody, pharmaceutical formulation and use of bispecific antibody
WO2014056783A1 (en) 2012-10-08 2014-04-17 Roche Glycart Ag Fc-free antibodies comprising two fab-fragments and methods of use
SG11201502538TA (en) 2012-11-08 2015-05-28 Hoffmann La Roche Her3 antigen binding proteins binding to the beta-hairpin of her3
GB201223276D0 (en) 2012-12-21 2013-02-06 Ucb Pharma Sa Antibodies and methods of producing same
DK2961771T3 (en) 2013-02-26 2020-03-02 Roche Glycart Ag Bispecific, T cell-activating, antigen-binding molecules specific for CD3 and CEA
EP2961773B1 (en) 2013-02-26 2019-03-20 Roche Glycart AG Bispecific t cell activating antigen binding molecules
CA2903056A1 (en) * 2013-03-15 2014-09-18 Merck Patent Gmbh Tetravalent bispecific antibodies
JP2016528168A (en) 2013-04-29 2016-09-15 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft Anti-IGF-1R antibodies with ineffective FcRn binding and their use in the treatment of vascular ocular diseases
CA2904806C (en) 2013-04-29 2021-11-23 F. Hoffmann-La Roche Ag Human fcrn-binding modified antibodies and methods of use
EP2992010B1 (en) 2013-04-29 2021-03-24 F.Hoffmann-La Roche Ag Fc-receptor binding modified asymmetric antibodies and methods of use
US20160115231A1 (en) 2013-05-21 2016-04-28 Yissum Research Development Company Of The Hebrew University Of Jerusalem Ltd. Treatment of mast cell related pathologies
WO2015025054A1 (en) 2013-08-22 2015-02-26 Medizinische Universität Wien Dye-specific antibodies for prestained molecular weight markers and methods producing the same
AU2014325063B2 (en) 2013-09-27 2019-10-31 Chugai Seiyaku Kabushiki Kaisha Method for producing polypeptide heteromultimer
SI3071597T1 (en) 2013-11-21 2020-11-30 F. Hoffmann-La Roche Ag Anti-alpha-synuclein antibodies and methods of use
MX2016007208A (en) 2013-12-20 2016-07-21 Hoffmann La Roche HUMANIZED ANTI-Tau(pS422) ANTIBODIES AND METHODS OF USE.
BR112016015140A2 (en) 2013-12-30 2018-01-23 Epimab Biotherapeutics Inc. tandem-fab immunoglobulin and its uses
CA2930046A1 (en) 2014-01-03 2015-07-09 F. Hoffmann-La Roche Ag Covalently linked polypeptide toxin-antibody conjugates
JP6602304B2 (en) 2014-01-03 2019-11-06 エフ.ホフマン−ラ ロシュ アーゲー Covalently linked helicer-anti-helicer antibody conjugates and uses thereof
BR112016013849A2 (en) 2014-01-03 2017-10-10 Hoffmann La Roche bispecific antihapten / blood-brain barrier receptor conjugates, their uses, and pharmaceutical formulation
CN111057151B (en) 2014-01-06 2022-05-03 豪夫迈·罗氏有限公司 Monovalent blood brain barrier shuttle modules
MX2016008539A (en) 2014-01-15 2016-09-26 Hoffmann La Roche Fc-region variants with modified fcrn- and maintained protein a-binding properties.
EP3126389A1 (en) 2014-04-02 2017-02-08 F. Hoffmann-La Roche AG Method for detecting multispecific antibody light chain mispairing
AR100978A1 (en) 2014-06-26 2016-11-16 Hoffmann La Roche ANTI-Tau HUMANIZED ANTIBODY BRAIN LAUNCHERS (pS422) AND USES OF THE SAME
RU2705299C2 (en) 2014-06-26 2019-11-06 Ф. Хоффманн-Ля Рош Аг Antibodies against 5-bromo-2'-deoxyuridine and methods of use
TW201623329A (en) 2014-06-30 2016-07-01 亞佛瑞司股份有限公司 Vaccines and monoclonal antibodies targeting truncated variants of osteopontin and uses thereof
CA2954687A1 (en) 2014-07-10 2016-01-14 Affiris Ag Substances and methods for the use in prevention and/or treatment in huntington's disease
MA50584A (en) 2014-08-04 2020-09-16 Hoffmann La Roche BISPECIFIC T-LYMPHOCYTE ACTIVATE ANTIGEN-BINDING MOLECULES
EP3191523B1 (en) 2014-09-08 2019-08-07 Yeda Research and Development Co., Ltd. Compositions and methods for treating cancer resistant to a tyrosine kinase inhibitor (tki)
CN107074966A (en) 2014-11-06 2017-08-18 豪夫迈·罗氏有限公司 The Fc region variants of FCRN and albumin A binding property with change
PL3215528T3 (en) 2014-11-06 2020-01-31 F.Hoffmann-La Roche Ag Fc-region variants with modified fcrn-binding and methods of use
CA2966566C (en) 2014-11-20 2024-03-19 F. Hoffmann-La Roche Ag Combination therapy of t cell activating bispecific antigen binding molecules cd3 and folate receptor 1 (folr1) and pd-1 axis binding antagonists
WO2016087514A1 (en) 2014-12-02 2016-06-09 Cemm - Forschungszentrum Für Molekulare Medizin Gmbh Anti-mutant calreticulin antibodies and their use in the diagnosis and therapy of myeloid malignancies
WO2016096788A1 (en) 2014-12-18 2016-06-23 F. Hoffmann-La Roche Ag Assay and method for determining cdc eliciting antibodies
WO2016159213A1 (en) * 2015-04-01 2016-10-06 中外製薬株式会社 Method for producing polypeptide hetero-oligomer
US10501545B2 (en) 2015-06-16 2019-12-10 Genentech, Inc. Anti-CLL-1 antibodies and methods of use
EP3744732A1 (en) 2015-06-24 2020-12-02 F. Hoffmann-La Roche AG Humanized anti-tau(ps422) antibodies and methods of use
AR106188A1 (en) 2015-10-01 2017-12-20 Hoffmann La Roche ANTI-CD19 HUMANIZED HUMAN ANTIBODIES AND METHODS OF USE
WO2017055399A1 (en) 2015-10-02 2017-04-06 F. Hoffmann-La Roche Ag Cellular based fret assay for the determination of simultaneous binding
JP7044700B2 (en) 2015-10-02 2022-03-30 エフ・ホフマン-ラ・ロシュ・アクチェンゲゼルシャフト Bispecific anti-CEAXCD3 T cell activating antigen binding molecule
PE20181092A1 (en) 2015-10-02 2018-07-09 Hoffmann La Roche ANTI-PD1 ANTIBODIES AND METHODS OF USE
EP4015533A1 (en) 2015-10-29 2022-06-22 F. Hoffmann-La Roche AG Anti-variant fc-region antibodies and methods of use
EP3184547A1 (en) 2015-10-29 2017-06-28 F. Hoffmann-La Roche AG Anti-tpbg antibodies and methods of use
EP3176183A1 (en) 2015-12-02 2017-06-07 Yeda Research and Development Co. Ltd Compositions and methods for treating cancer not resistant to a tyrosine kinase inhibitor (tki)
CN115920030A (en) 2015-12-09 2023-04-07 豪夫迈·罗氏有限公司 Use of type II anti-CD 20 antibodies for reducing anti-drug antibody formation
EP3398965A4 (en) 2015-12-28 2019-09-18 Chugai Seiyaku Kabushiki Kaisha Method for promoting efficiency of purification of fc region-containing polypeptide
CN108368179B (en) 2016-01-08 2022-08-23 豪夫迈·罗氏有限公司 Methods of treating CEA positive cancers using PD-1 axis binding antagonists and anti-CEA/anti-CD 3 bispecific antibodies
MX2018009581A (en) 2016-02-06 2019-05-06 Epimab Biotherapeutics Inc Fabs-in-tandem immunoglobulin and uses thereof.
SG11201807936VA (en) 2016-03-14 2018-10-30 Chugai Pharmaceutical Co Ltd Cell injury inducing therapeutic drug for use in cancer therapy
EP3433281A1 (en) 2016-03-21 2019-01-30 Elstar Therapeutics, Inc. Multispecific and multifunctional molecules and uses thereof
LT3433280T (en) 2016-03-22 2023-07-10 F. Hoffmann-La Roche Ag Protease-activated t cell bispecific molecules
WO2017165681A1 (en) 2016-03-24 2017-09-28 Gensun Biopharma Inc. Trispecific inhibitors for cancer treatment
UA123323C2 (en) * 2016-05-02 2021-03-17 Ф. Хоффманн-Ля Рош Аг The contorsbody - a single chain target binder
EP3472177A2 (en) 2016-06-17 2019-04-24 F. Hoffmann-La Roche AG Purification of multispecific antibodies
CA3030841A1 (en) 2016-07-14 2018-01-18 Fred Hutchinson Cancer Research Center Multiple bi-specific binding domain constructs with different epitope binding to treat cancer
CN116731197A (en) 2016-09-19 2023-09-12 豪夫迈·罗氏有限公司 Complement factor based affinity chromatography
WO2018060035A1 (en) 2016-09-30 2018-04-05 F. Hoffmann-La Roche Ag Spr-based dual-binding assay for the functional analysis of multispecific molecules
PL3519437T3 (en) 2016-09-30 2022-01-17 F. Hoffmann-La Roche Ag Bispecific antibodies against p95her2
TW201829463A (en) 2016-11-18 2018-08-16 瑞士商赫孚孟拉羅股份公司 Anti-hla-g antibodies and use thereof
WO2018098363A2 (en) 2016-11-23 2018-05-31 Bioverativ Therapeutics Inc. Bispecific antibodies binding to coagulation factor ix and coagulation factor x
MX2019007411A (en) 2016-12-21 2019-08-29 Hoffmann La Roche Re-use of enzymes in in vitro glycoengineering of antibodies.
CN117887794A (en) 2016-12-21 2024-04-16 豪夫迈·罗氏有限公司 Methods for in vitro glycoengineering antibodies
MX2019006266A (en) 2016-12-21 2019-08-21 Hoffmann La Roche In vitro glycoengineering of antibodies.
WO2018128939A1 (en) 2017-01-05 2018-07-12 Gensun Biopharma Inc. Checkpoint regulator antagonists
WO2018151820A1 (en) 2017-02-16 2018-08-23 Elstar Therapeutics, Inc. Multifunctional molecules comprising a trimeric ligand and uses thereof
JP7247101B2 (en) 2017-04-03 2023-03-28 エフ・ホフマン-ラ・ロシュ・アクチェンゲゼルシャフト Antibody that binds to STEAP-1
IL268527B2 (en) 2017-04-05 2024-03-01 Hoffmann La Roche Bispecific antibodies specifically binding to pd1 and lag3
LT3606954T (en) 2017-04-05 2022-09-26 F. Hoffmann-La Roche Ag Anti-lag3 antibodies
US20210246227A1 (en) 2017-05-31 2021-08-12 Elstar Therapeutics, Inc. Multispecific molecules that bind to myeloproliferative leukemia (mpl) protein and uses thereof
WO2019035938A1 (en) 2017-08-16 2019-02-21 Elstar Therapeutics, Inc. Multispecific molecules that bind to bcma and uses thereof
EP3684801A1 (en) * 2017-09-22 2020-07-29 H. Hoffnabb-La Roche Ag Multivalent mono- or bispecific recombinant antibodies for analytic purpose
EP3704146B1 (en) * 2017-11-01 2021-12-15 F. Hoffmann-La Roche AG Trifab-contorsbody
PE20210844A1 (en) * 2017-11-01 2021-05-10 Hoffmann La Roche CONTORSBODIES 2 + BI-SPECIFIC
CA3081801C (en) 2017-11-29 2022-12-20 F. Hoffman-La Roche Ag Target interference suppressed anti-drug antibody assay
AU2018390881A1 (en) 2017-12-21 2020-07-02 F. Hoffmann-La Roche Ag Antibodies binding to HLA-A2/WT1
WO2019129679A1 (en) 2017-12-29 2019-07-04 F. Hoffmann-La Roche Ag Method for improving vegf-receptor blocking selectivity of an anti-vegf antibody
JP2021511782A (en) 2018-01-31 2021-05-13 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft Stabilized immunoglobulin domain
WO2019149716A1 (en) 2018-01-31 2019-08-08 F. Hoffmann-La Roche Ag Bispecific antibodies comprising an antigen-binding site binding to lag3
EP3749361A1 (en) 2018-02-08 2020-12-16 F. Hoffmann-La Roche AG Bispecific antigen-binding molecules and methods of use
TWI829667B (en) 2018-02-09 2024-01-21 瑞士商赫孚孟拉羅股份公司 Antibodies binding to gprc5d
WO2019178364A2 (en) 2018-03-14 2019-09-19 Elstar Therapeutics, Inc. Multifunctional molecules and uses thereof
WO2019178362A1 (en) 2018-03-14 2019-09-19 Elstar Therapeutics, Inc. Multifunctional molecules that bind to calreticulin and uses thereof
MX2020010028A (en) 2018-03-29 2020-10-14 Genentech Inc Modulating lactogenic activity in mammalian cells.
AR114789A1 (en) 2018-04-18 2020-10-14 Hoffmann La Roche ANTI-HLA-G ANTIBODIES AND THE USE OF THEM
AR115052A1 (en) 2018-04-18 2020-11-25 Hoffmann La Roche MULTI-SPECIFIC ANTIBODIES AND THE USE OF THEM
KR20210028222A (en) 2018-06-29 2021-03-11 젠선 바이오파마, 인코포레이티드 Anti-tumor immune checkpoint control antagonists
EP3818083A2 (en) 2018-07-03 2021-05-12 Elstar Therapeutics, Inc. Anti-tcr antibody molecules and uses thereof
WO2020014542A2 (en) * 2018-07-11 2020-01-16 Momenta Pharmaceuticals, Inc. Compositions and methods related to engineered fc-antigen binding domain constructs
TWI754157B (en) 2018-07-25 2022-02-01 大陸商信達生物製藥(蘇州)有限公司 Anti-tigit antibody and use thereof
US20200047085A1 (en) 2018-08-09 2020-02-13 Regeneron Pharmaceuticals, Inc. Methods for assessing binding affinity of an antibody variant to the neonatal fc receptor
CA3108434A1 (en) 2018-08-24 2020-02-27 Yeda Research And Development Co. Ltd. Methods of modulating m2 macrophage polarization and use of same in therapy
SG11202102859TA (en) 2018-12-21 2021-04-29 Hoffmann La Roche Antibodies binding to cd3
JP2022515424A (en) 2018-12-24 2022-02-18 サノフイ Multispecific binding protein with mutated fab domain
EP3902560A1 (en) 2018-12-28 2021-11-03 F. Hoffmann-La Roche AG A peptide-mhc-i-antibody fusion protein for therapeutic use in a patient with amplified immune response
EP3903102B1 (en) 2018-12-30 2023-04-12 F. Hoffmann-La Roche AG Ph-gradient spr-based binding assay
AU2020226904A1 (en) 2019-02-21 2021-09-16 Marengo Therapeutics, Inc. Anti-TCR antibody molecules and uses thereof
CN114127112A (en) 2019-02-21 2022-03-01 马伦戈治疗公司 Multifunctional molecules that bind to T cells and their use to treat autoimmune disorders
JP2022521937A (en) 2019-02-21 2022-04-13 マレンゴ・セラピューティクス,インコーポレーテッド Antibody molecules that bind to NKp30 and their use
SG11202109056TA (en) 2019-02-21 2021-09-29 Marengo Therapeutics Inc Multifunctional molecules that bind to calreticulin and uses thereof
CN114026122A (en) 2019-02-21 2022-02-08 马伦戈治疗公司 Multifunctional molecules that bind to T cell-associated cancer cells and uses thereof
EP3947440A1 (en) 2019-03-29 2022-02-09 F. Hoffmann-La Roche AG Method for generating avid-binding multispecific antibodies
JP7249432B2 (en) 2019-03-29 2023-03-30 エフ. ホフマン-ラ ロシュ アーゲー SPR-based binding assays for functional analysis of multivalent molecules
JP7273195B2 (en) 2019-05-13 2023-05-12 エフ. ホフマン-ラ ロシュ アーゲー Interference suppression pharmacokinetic immunoassay
AU2020296247A1 (en) 2019-06-19 2021-12-23 F. Hoffmann-La Roche Ag Method for the generation of a trivalent antibody expressing cell by targeted integration of multiple expression cassettes in a defined organization
AU2020297940A1 (en) 2019-06-19 2021-12-16 F. Hoffmann-La Roche Ag Method for the generation of a multivalent, bispecific antibody expressing cell by targeted integration of multiple expression cassettes in a defined organization
AU2020294878A1 (en) 2019-06-19 2021-12-23 F. Hoffmann-La Roche Ag Method for the generation of a multivalent, multispecific antibody expressing cell by targeted integration of multiple expression cassettes in a defined organization
CA3140318A1 (en) 2019-06-19 2020-12-24 Johannes Auer Method for the generation of a bivalent, bispecific antibody expressing cell by targeted integration of multiple expression cassettes in a defined organization
CN114080451B (en) 2019-06-19 2024-03-22 豪夫迈·罗氏有限公司 Method for generating protein expressing cells by targeted integration using Cre mRNA
MX2021015823A (en) 2019-06-26 2022-02-03 Hoffmann La Roche Mammalian cell lines with sirt-1 gene knockout.
WO2020263312A1 (en) 2019-06-28 2020-12-30 Gensun Biopharma, Inc. ANTITUMOR ANTAGONIST CONSISTING OF A MUTATED TGFβ1 - RII EXTRACELLULAR DOMAIN AND AN IMMUNOGLOBULIN SCAFFOLD
AR119393A1 (en) 2019-07-15 2021-12-15 Hoffmann La Roche ANTIBODIES THAT BIND NKG2D
CN114174338A (en) 2019-07-31 2022-03-11 豪夫迈·罗氏有限公司 Antibodies that bind to GPRC5D
EP4003526A2 (en) 2019-07-31 2022-06-01 F. Hoffmann-La Roche AG Antibodies binding to gprc5d
AU2020349509A1 (en) 2019-09-18 2022-03-31 Genentech, Inc. Anti-KLK7 antibodies, anti-KLK5 antibodies, multispecific anti-KLK5/KLK7 antibodies, and methods of use
AU2020384917A1 (en) 2019-11-15 2022-03-31 F. Hoffmann-La Roche Ag Prevention of visible particle formation in aqueous protein solutions
JP7415005B2 (en) 2019-12-18 2024-01-16 エフ・ホフマン-ラ・ロシュ・アクチェンゲゼルシャフト Bispecific anti-CCL2 antibody
KR20220100963A (en) 2019-12-18 2022-07-18 에프. 호프만-라 로슈 아게 Antibodies that bind to HLA-A2/MAGE-A4
JP2023509390A (en) 2019-12-23 2023-03-08 ジェネンテック, インコーポレイテッド Apolipoprotein L1-specific antibody and method of use thereof
US11913945B2 (en) 2020-01-02 2024-02-27 Hoffmann-La Roche Inc. Method for determining the amount of a therapeutic antibody in the brain
WO2021138407A2 (en) 2020-01-03 2021-07-08 Marengo Therapeutics, Inc. Multifunctional molecules that bind to cd33 and uses thereof
WO2021144422A1 (en) 2020-01-15 2021-07-22 F. Hoffmann-La Roche Ag Methods to decrease impurities from recombinant protein manufacturing processes
JP2023518841A (en) 2020-03-26 2023-05-08 ジェネンテック, インコーポレイテッド Modified mammalian cells with reduced host cell proteins
WO2021217085A1 (en) 2020-04-24 2021-10-28 Marengo Therapeutics, Inc. Multifunctional molecules that bind to t cell related cancer cells and uses thereof
WO2021228917A1 (en) 2020-05-15 2021-11-18 F. Hoffmann-La Roche Ag Prevention of visible particle formation in parenteral protein solutions
CN115605185A (en) 2020-05-19 2023-01-13 豪夫迈·罗氏有限公司(Ch) Use of a chelating agent to prevent the formation of visible particles in parenteral protein solutions
JP2023527918A (en) 2020-06-08 2023-06-30 エフ・ホフマン-ラ・ロシュ・アクチェンゲゼルシャフト Anti-HBV antibody and method of use
CA3183441A1 (en) 2020-06-16 2021-12-23 F. Hoffmann-La Roche Ag Method for determining the free antigen of an antibody in a sample
WO2021255146A1 (en) 2020-06-19 2021-12-23 F. Hoffmann-La Roche Ag Antibodies binding to cd3 and cea
KR20230025665A (en) 2020-06-19 2023-02-22 에프. 호프만-라 로슈 아게 Antibodies that bind to CD3
JP2023538716A (en) 2020-06-19 2023-09-11 エフ・ホフマン-ラ・ロシュ・アクチェンゲゼルシャフト Antibodies that bind to CD3 and CD19
BR112022025809A2 (en) 2020-06-19 2023-01-10 Hoffmann La Roche ANTIBODIES, ISOLATED POLYNUCLEOTIDE, HOST CELL, METHOD FOR PRODUCING AN ANTIBODY, USE OF THE ANTIBODY, METHOD FOR TREATING A DISEASE AND INVENTION
IL299161A (en) 2020-06-24 2023-02-01 Genentech Inc Apoptosis resistant cell lines
WO2022008468A1 (en) 2020-07-07 2022-01-13 F. Hoffmann-La Roche Ag Alternative surfactants as stabilizers for therapeutic protein formulations
PE20231300A1 (en) 2020-07-17 2023-08-24 Genentech Inc ANTI-NOTCH2 ANTIBODIES AND METHODS OF USE
EP4204458A1 (en) 2020-08-26 2023-07-05 Marengo Therapeutics, Inc. Methods of detecting trbc1 or trbc2
WO2022046922A2 (en) 2020-08-26 2022-03-03 Marengo Therapeutics, Inc. Antibody molecules that bind to nkp30 and uses thereof
WO2022046920A2 (en) 2020-08-26 2022-03-03 Marengo Therapeutics, Inc. Multifunctional molecules that bind to calreticulin and uses thereof
CA3192344A1 (en) 2020-08-28 2022-03-03 Genentech, Inc. Crispr/cas9 multiplex knockout of host cell proteins
WO2022061214A1 (en) 2020-09-21 2022-03-24 Genentech, Inc. Purification of multispecific antibodies
MX2023003328A (en) 2020-09-24 2023-03-27 Hoffmann La Roche Mammalian cell lines with gene knockout.
TW202233671A (en) 2020-10-20 2022-09-01 美商建南德克公司 Peg-conjugated anti-mertk antibodies and methods of use
WO2022124866A1 (en) 2020-12-10 2022-06-16 주식회사 유틸렉스 Anti-pd-1 antibody and uses thereof
CR20230263A (en) 2020-12-17 2023-08-21 Hoffmann La Roche Anti-hla-g antibodies and use thereof
WO2022136140A1 (en) 2020-12-22 2022-06-30 F. Hoffmann-La Roche Ag Oligonucleotides targeting xbp1
EP4269435A1 (en) 2020-12-23 2023-11-01 Innovent Biologics (Singapore) Pte. Ltd. Anti-b7-h3 antibody and uses thereof
CN114716548A (en) 2021-01-05 2022-07-08 (株)爱恩德生物 anti-FGFR 3 antibodies and uses thereof
EP4288458A1 (en) 2021-02-03 2023-12-13 Genentech, Inc. Multispecific binding protein degrader platform and methods of use
JP2024512240A (en) 2021-02-18 2024-03-19 エフ. ホフマン-ラ ロシュ アーゲー Methods for elucidating complex multistep antibody interactions
JP2024512377A (en) 2021-03-12 2024-03-19 ジェネンテック, インコーポレイテッド Anti-KLK7 antibodies, anti-KLK5 antibodies, multispecific anti-KLK5/KLK7 antibodies, and methods of use
KR20240004462A (en) 2021-04-08 2024-01-11 마렝고 테라퓨틱스, 인크. Multifunctional molecules that bind to TCR and their uses
WO2022214565A1 (en) 2021-04-09 2022-10-13 F. Hoffmann-La Roche Ag Process for selecting cell clones expressing a heterologous polypeptide
KR20230173164A (en) 2021-04-19 2023-12-26 제넨테크, 인크. modified mammalian cells
WO2022246259A1 (en) 2021-05-21 2022-11-24 Genentech, Inc. Modified cells for the production of a recombinant product of interest
CN113278071B (en) 2021-05-27 2021-12-21 江苏荃信生物医药股份有限公司 Anti-human interferon alpha receptor1 monoclonal antibody and application thereof
WO2022256820A1 (en) 2021-06-03 2022-12-08 Gensun Biopharma Inc. Multispecific antagonists
EP4355775A1 (en) 2021-06-18 2024-04-24 F. Hoffmann-La Roche AG Bispecific anti-ccl2 antibodies
US20230197278A1 (en) 2021-07-13 2023-06-22 Genentech, Inc. Multi-variate model for predicting cytokine release syndrome
WO2023001884A1 (en) 2021-07-22 2023-01-26 F. Hoffmann-La Roche Ag Heterodimeric fc domain antibodies
CA3226947A1 (en) 2021-08-03 2023-02-09 Muhammad YASSIN Engineered tcr complex and methods of using same
CN117794953A (en) 2021-08-03 2024-03-29 豪夫迈·罗氏有限公司 Bispecific antibodies and methods of use
EP4130028A1 (en) 2021-08-03 2023-02-08 Rhazes Therapeutics Ltd Engineered tcr complex and methods of using same
CN113603775B (en) 2021-09-03 2022-05-20 江苏荃信生物医药股份有限公司 Anti-human interleukin-33 monoclonal antibody and application thereof
CN113683694B (en) 2021-09-03 2022-05-13 江苏荃信生物医药股份有限公司 Anti-human TSLP monoclonal antibody and application thereof
WO2023094282A1 (en) 2021-11-25 2023-06-01 F. Hoffmann-La Roche Ag Quantification of low amounts of antibody sideproducts
AR127887A1 (en) 2021-12-10 2024-03-06 Hoffmann La Roche ANTIBODIES THAT BIND CD3 AND PLAP
WO2023117325A1 (en) 2021-12-21 2023-06-29 F. Hoffmann-La Roche Ag Method for the determination of hydrolytic activity
AU2022424002A1 (en) 2021-12-29 2024-06-13 Bristol-Myers Squibb Company Generation of landing pad cell lines
TW202340251A (en) 2022-01-19 2023-10-16 美商建南德克公司 Anti-notch2 antibodies and conjugates and methods of use
WO2023175171A1 (en) 2022-03-18 2023-09-21 Inserm (Institut National De La Sante Et De La Recherche Medicale) Bk polyomavirus antibodies and uses thereof
WO2023202967A1 (en) 2022-04-19 2023-10-26 F. Hoffmann-La Roche Ag Improved production cells
WO2023232961A1 (en) 2022-06-03 2023-12-07 F. Hoffmann-La Roche Ag Improved production cells
WO2024020564A1 (en) 2022-07-22 2024-01-25 Genentech, Inc. Anti-steap1 antigen-binding molecules and uses thereof
WO2024079015A1 (en) 2022-10-10 2024-04-18 F. Hoffmann-La Roche Ag Combination therapy of a gprc5d tcb and imids
WO2024079009A1 (en) 2022-10-10 2024-04-18 F. Hoffmann-La Roche Ag Combination therapy of a gprc5d tcb and proteasome inhibitors
WO2024079010A1 (en) 2022-10-10 2024-04-18 F. Hoffmann-La Roche Ag Combination therapy of a gprc5d tcb and cd38 antibodies
WO2024079069A1 (en) 2022-10-12 2024-04-18 F. Hoffmann-La Roche Ag Method for classifying cells
WO2024102948A1 (en) 2022-11-11 2024-05-16 Celgene Corporation Fc receptor-homolog 5 (fcrh5) specific binding molecules and bispecific t-cell engaging antibodies including same and related methods
WO2024110426A1 (en) 2022-11-23 2024-05-30 F. Hoffmann-La Roche Ag Method for increasing recombinant protein expression

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994009131A1 (en) * 1992-10-15 1994-04-28 Scotgen Limited Recombinant specific binding protein
WO1999066951A2 (en) * 1998-06-22 1999-12-29 Immunomedics, Inc. Use of bi-specific antibodies for pre-targeting diagnosis and therapy
US6982321B2 (en) * 1986-03-27 2006-01-03 Medical Research Council Altered antibodies
US20070071742A1 (en) * 1998-11-30 2007-03-29 Fang Fang Humanized antibodies

Family Cites Families (261)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4150149A (en) 1976-11-29 1979-04-17 Professional Staff Association Of The Los Angeles County Harbor General Hospital Method and means for the early detection and diagnosis of certain types of cancers
US4444744A (en) 1980-03-03 1984-04-24 Goldenberg Milton David Tumor localization and therapy with labeled antibodies to cell surface antigens
US4361544A (en) 1980-03-03 1982-11-30 Goldenberg Milton David Tumor localization and therapy with labeled antibodies specific to intracellular tumor-associated markers
US4737456A (en) 1985-05-09 1988-04-12 Syntex (U.S.A.) Inc. Reducing interference in ligand-receptor binding assays
DE3883899T3 (en) 1987-03-18 1999-04-22 Sb2 Inc CHANGED ANTIBODIES.
US5204244A (en) 1987-10-27 1993-04-20 Oncogen Production of chimeric antibodies by homologous recombination
US5202238A (en) 1987-10-27 1993-04-13 Oncogen Production of chimeric antibodies by homologous recombination
WO1990005144A1 (en) 1988-11-11 1990-05-17 Medical Research Council Single domain ligands, receptors comprising said ligands, methods for their production, and use of said ligands and receptors
US5959177A (en) 1989-10-27 1999-09-28 The Scripps Research Institute Transgenic plants expressing assembled secretory antibodies
JPH04505709A (en) 1989-11-07 1992-10-08 ブリストル―マイアーズ スクイブ カンパニー oligomeric immunoglobulin
KR930702029A (en) 1990-08-31 1993-09-08 스티븐 체스노프 Homozygous Immunoglobulins
US5571894A (en) 1991-02-05 1996-11-05 Ciba-Geigy Corporation Recombinant antibodies specific for a growth factor receptor
DE4118120A1 (en) 1991-06-03 1992-12-10 Behringwerke Ag TETRAVALENT BISPECIFIC RECEPTORS, THEIR PRODUCTION AND USE
US6511663B1 (en) 1991-06-11 2003-01-28 Celltech R&D Limited Tri- and tetra-valent monospecific antigen-binding proteins
GB9114948D0 (en) 1991-07-11 1991-08-28 Pfizer Ltd Process for preparing sertraline intermediates
EP0861893A3 (en) 1991-09-19 1999-11-10 Genentech, Inc. High level expression of immunoglobulin polypeptides
US5587458A (en) 1991-10-07 1996-12-24 Aronex Pharmaceuticals, Inc. Anti-erbB-2 antibodies, combinations thereof, and therapeutic and diagnostic uses thereof
EP0614375A1 (en) 1991-11-26 1994-09-14 Alkermes, Inc. Process for the preparation of transferrin receptor specific antibody-neuropharmaceutical or diagnostic agent conjugates
FR2684822B1 (en) 1991-12-06 1994-03-25 Nicolas Vaugnier METHOD AND APPARATUS FOR CONVERTING REGULAR CODES INTO VARIABLE SIZE CODES AND FOR PROVIDING SERIAL COMMUNICATION OF THE CODES OBTAINED.
DE69334255D1 (en) 1992-02-06 2009-02-12 Novartis Vaccines & Diagnostic Marker for cancer and biosynthetic binding protein for it
DE69232539T3 (en) 1992-10-28 2007-01-04 Genentech, Inc., South San Francisco Use of anti-VEGF antibodies for the treatment of cancer
US5747654A (en) 1993-06-14 1998-05-05 The United States Of America As Represented By The Department Of Health And Human Services Recombinant disulfide-stabilized polypeptide fragments having binding specificity
CA2163345A1 (en) 1993-06-16 1994-12-22 Susan Adrienne Morgan Antibodies
US6476198B1 (en) 1993-07-13 2002-11-05 The Scripps Research Institute Multispecific and multivalent antigen-binding polypeptide molecules
UA40577C2 (en) 1993-08-02 2001-08-15 Мерк Патент Гмбх Bispecific antigen molecule for lysis of tumor cells, method for preparing of bispecific antigen molecule, monoclonal antibody (variants), pharmaceutical preparation, pharmaceutical kit for lysis of tumor cells (variants), method of lysis of tumor cells
WO1995009917A1 (en) 1993-10-07 1995-04-13 The Regents Of The University Of California Genetically engineered bispecific tetravalent antibodies
US5814464A (en) 1994-10-07 1998-09-29 Regeneron Pharma Nucleic acids encoding TIE-2 ligand-2
US5789199A (en) 1994-11-03 1998-08-04 Genentech, Inc. Process for bacterial production of polypeptides
US6485943B2 (en) 1995-01-17 2002-11-26 The University Of Chicago Method for altering antibody light chain interactions
US5840523A (en) 1995-03-01 1998-11-24 Genetech, Inc. Methods and compositions for secretion of heterologous polypeptides
US5731168A (en) 1995-03-01 1998-03-24 Genentech, Inc. Method for making heteromultimeric polypeptides
GB9504344D0 (en) 1995-03-03 1995-04-19 Unilever Plc Antibody fragment production
US5869046A (en) 1995-04-14 1999-02-09 Genentech, Inc. Altered polypeptides with increased half-life
US6267958B1 (en) 1995-07-27 2001-07-31 Genentech, Inc. Protein formulation
CN1173878A (en) 1995-10-16 1998-02-18 尤尼利弗公司 Bifunctional or bivalent antibody fragment analogue
US6750334B1 (en) 1996-02-02 2004-06-15 Repligen Corporation CTLA4-immunoglobulin fusion proteins having modified effector functions and uses therefor
GB9603256D0 (en) 1996-02-16 1996-04-17 Wellcome Found Antibodies
WO1997038102A1 (en) * 1996-04-04 1997-10-16 Unilever Plc Multivalent and multispecific antigen-binding protein
DE69841815D1 (en) 1997-04-07 2010-09-16 Genentech Inc Anti-VEGF antibody
SI1325932T1 (en) 1997-04-07 2005-08-31 Genentech Inc
WO1998048032A2 (en) 1997-04-21 1998-10-29 Donlar Corporation POLY-(α-L-ASPARTIC ACID), POLY-(α-L-GLUTAMIC ACID) AND COPOLYMERS OF L-ASP AND L-GLU, METHOD FOR THEIR PRODUCTION AND THEIR USE
US6171586B1 (en) 1997-06-13 2001-01-09 Genentech, Inc. Antibody formulation
EP0994903B1 (en) 1997-06-24 2005-05-25 Genentech, Inc. Methods and compositions for galactosylated glycoproteins
US6040498A (en) 1998-08-11 2000-03-21 North Caroline State University Genetically engineered duckweed
AU9262598A (en) 1997-08-18 1999-03-08 Innogenetics N.V. Interferon-gamma-binding molecules for treating septic shock, cachexia, immune diseases and skin disorders
AU759779B2 (en) 1997-10-31 2003-05-01 Genentech Inc. Methods and compositions comprising glycoprotein glycoforms
WO1999037791A1 (en) 1998-01-23 1999-07-29 Vlaams Interuniversitair Instituut Voor Biotechnologie Multipurpose antibody derivatives
ATE375365T1 (en) 1998-04-02 2007-10-15 Genentech Inc ANTIBODIES VARIANTS AND FRAGMENTS THEREOF
US6194551B1 (en) 1998-04-02 2001-02-27 Genentech, Inc. Polypeptide variants
DK2180007T4 (en) 1998-04-20 2017-11-27 Roche Glycart Ag Glycosylation technique for antibodies to enhance antibody-dependent cell cytotoxicity
DE19819846B4 (en) 1998-05-05 2016-11-24 Deutsches Krebsforschungszentrum Stiftung des öffentlichen Rechts Multivalent antibody constructs
US7138103B2 (en) * 1998-06-22 2006-11-21 Immunomedics, Inc. Use of bi-specific antibodies for pre-targeting diagnosis and therapy
WO2000035956A1 (en) 1998-12-16 2000-06-22 Kyowa Hakko Kogyo Co., Ltd. Antihuman vegf monoclonal antibody
KR101077001B1 (en) 1999-01-15 2011-10-26 제넨테크, 인크. Polypeptide Variants with Altered Effector Function
US6737056B1 (en) 1999-01-15 2004-05-18 Genentech, Inc. Polypeptide variants with altered effector function
US6897044B1 (en) 1999-01-28 2005-05-24 Biogen Idec, Inc. Production of tetravalent antibodies
CN1232039A (en) 1999-04-02 1999-10-20 中国人民解放军海军总医院 Genetic engineering double specific antibody and its use
PT1176195E (en) 1999-04-09 2013-07-18 Kyowa Hakko Kirin Co Ltd Method for controlling the activity of immunologically functional molecule
US7125978B1 (en) 1999-10-04 2006-10-24 Medicago Inc. Promoter for regulating expression of foreign genes
KR100797667B1 (en) 1999-10-04 2008-01-23 메디카고 인코포레이티드 Method for regulating transcription of foreign genes
AU7950400A (en) 1999-10-19 2001-04-30 Kyowa Hakko Kogyo Co. Ltd. Process for producing polypeptide
US7449443B2 (en) 2000-03-23 2008-11-11 California Institute Of Technology Method for stabilization of proteins using non-natural amino acids
AU2001260153B2 (en) 2000-03-24 2006-08-17 Micromet Ag Multifunctional polypeptides comprising a binding site to an epitope of the NKG2D receptor complex
DK2857516T3 (en) 2000-04-11 2017-08-07 Genentech Inc Multivalent antibodies and uses thereof
FR2807767B1 (en) 2000-04-12 2005-01-14 Lab Francais Du Fractionnement MONOCLONAL ANTIBODIES ANTI-D
DE10021678A1 (en) 2000-05-05 2002-04-18 Stefan Duebel Recombinant polyspecific antibody constructs, useful for diagnosis and treatment of cancer, comprises three antibody fragments,where at least one comprises a disulfide bridge
US20020103345A1 (en) 2000-05-24 2002-08-01 Zhenping Zhu Bispecific immunoglobulin-like antigen binding proteins and method of production
US6586207B2 (en) 2000-05-26 2003-07-01 California Institute Of Technology Overexpression of aminoacyl-tRNA synthetases for efficient production of engineered proteins containing amino acid analogues
WO2002002781A1 (en) 2000-06-30 2002-01-10 Vlaams Interuniversitair Instituut Voor Biotechnologie Vzw Heterodimeric fusion proteins
US6946292B2 (en) 2000-10-06 2005-09-20 Kyowa Hakko Kogyo Co., Ltd. Cells producing antibody compositions with increased antibody dependent cytotoxic activity
US7064191B2 (en) 2000-10-06 2006-06-20 Kyowa Hakko Kogyo Co., Ltd. Process for purifying antibody
EP3263702A1 (en) 2000-10-06 2018-01-03 Kyowa Hakko Kirin Co., Ltd. Cells producing antibody compositions
RU2295537C2 (en) 2000-10-20 2007-03-20 Тугаи Сейяку Кабусики Кайся Modified antagonistic antibody
US7319139B2 (en) 2001-01-29 2008-01-15 Biogen Idec, Inc. TAG-72 specific CH2 domain deleted antibodies
MXPA04001072A (en) 2001-08-03 2005-02-17 Glycart Biotechnology Ag Antibody glycosylation variants having increased antibody-dependent cellular cytotoxicity.
AU2002359244A1 (en) 2001-09-05 2003-04-28 The Government Of The United States Of America, As Represented By The Secretary Of The Department Of Imaging the activity of extracellular proteases in cells using mutant anthrax toxin protective antigens that are cleaved by specific extracellular proteases
DE60124912T2 (en) 2001-09-14 2007-06-14 Affimed Therapeutics Ag Multimeric, single chain, tandem Fv antibodies
US7521053B2 (en) 2001-10-11 2009-04-21 Amgen Inc. Angiopoietin-2 specific binding agents
US7138370B2 (en) 2001-10-11 2006-11-21 Amgen Inc. Specific binding agents of human angiopoietin-2
US7658924B2 (en) 2001-10-11 2010-02-09 Amgen Inc. Angiopoietin-2 specific binding agents
ATE481109T1 (en) 2001-10-16 2010-10-15 Us Gov Health & Human Serv NEUTRALIZING ANTIBODIES AGAINST HIV WITH BROAD CROSS-REACTION SELECTED USING ENV-CD4 CO-RECEPTOR COMPLEXES
US7053202B2 (en) 2001-10-19 2006-05-30 Millennium Pharmaceuticals, Inc. Immunoglobulin DNA cassette molecules, monobody constructs, methods of production, and methods of use therefor
KR100988949B1 (en) 2001-10-25 2010-10-20 제넨테크, 인크. Glycoprotein compositions
US20040093621A1 (en) 2001-12-25 2004-05-13 Kyowa Hakko Kogyo Co., Ltd Antibody composition which specifically binds to CD20
US7139665B2 (en) 2002-02-27 2006-11-21 California Institute Of Technology Computational method for designing enzymes for incorporation of non natural amino acids into proteins
EP1487879B1 (en) 2002-03-01 2012-12-26 Immunomedics, Inc. Bispecific antibody point mutations for enhancing rate of clearance
US7332585B2 (en) 2002-04-05 2008-02-19 The Regents Of The California University Bispecific single chain Fv antibody molecules and methods of use thereof
DE60336548D1 (en) 2002-04-09 2011-05-12 Kyowa Hakko Kirin Co Ltd CELL WITH REDUCED OR DELETED ACTIVITY OF A PROTEIN INVOLVED IN GDP FUCOSET TRANSPORT
EP1498490A4 (en) 2002-04-09 2006-11-29 Kyowa Hakko Kogyo Kk Process for producing antibody composition
PL373256A1 (en) 2002-04-09 2005-08-22 Kyowa Hakko Kogyo Co, Ltd. Cells with modified genome
US20050031613A1 (en) 2002-04-09 2005-02-10 Kazuyasu Nakamura Therapeutic agent for patients having human FcgammaRIIIa
EP1500400A4 (en) 2002-04-09 2006-10-11 Kyowa Hakko Kogyo Kk Drug containing antibody composition
EP1498491A4 (en) 2002-04-09 2006-12-13 Kyowa Hakko Kogyo Kk METHOD OF ENHANCING ACTIVITY OF ANTIBODY COMPOSITION OF BINDING TO Fc GAMMA RECEPTOR IIIa
US20070274998A1 (en) 2002-04-29 2007-11-29 Genpatzz Pharmacogentetics Ag Novel Bispecific Molecules For Use In Therapy And Diagnosis
US6658672B2 (en) 2002-05-06 2003-12-09 Elvis Hsieh Hinge of toilet bowl seat and toilet bowl cover
US7081443B2 (en) 2002-05-21 2006-07-25 Korea Advanced Institutes Of Science And Technology (Kaist) Chimeric comp-ang1 molecule
SE0201863D0 (en) 2002-06-18 2002-06-18 Cepep Ab Cell penetrating peptides
EP2366718A3 (en) 2002-06-28 2012-05-02 Domantis Limited Ligand
EP1549345A1 (en) * 2002-10-10 2005-07-06 MERCK PATENT GmbH Bispecific anti-erb-b antibodies and their use in tumor therapy
US7361740B2 (en) 2002-10-15 2008-04-22 Pdl Biopharma, Inc. Alteration of FcRn binding affinities or serum half-lives of antibodies by mutagenesis
SI2289936T1 (en) 2002-12-16 2017-10-30 Genentech, Inc. Immunoglobulin variants and uses thereof
US7534427B2 (en) 2002-12-31 2009-05-19 Immunomedics, Inc. Immunotherapy of B cell malignancies and autoimmune diseases using unconjugated antibodies and conjugated antibodies and antibody combinations and fusion proteins
WO2004063351A2 (en) 2003-01-09 2004-07-29 Macrogenics, Inc. IDENTIFICATION AND ENGINEERING OF ANTIBODIES WITH VARIANT Fc REGIONS AND METHODS OF USING SAME
EP2264151B1 (en) 2003-01-22 2016-04-20 Roche Glycart AG Fusion constructs and use of same to produce antibodies with increased FC receptor binding affinity and effector function
MXPA05008521A (en) * 2003-02-13 2005-10-20 Pharmacia Corp Antibodies to c-met for the treatment of cancers.
US7871607B2 (en) 2003-03-05 2011-01-18 Halozyme, Inc. Soluble glycosaminoglycanases and methods of preparing and using soluble glycosaminoglycanases
US20060104968A1 (en) 2003-03-05 2006-05-18 Halozyme, Inc. Soluble glycosaminoglycanases and methods of preparing and using soluble glycosaminogly ycanases
TWI353991B (en) * 2003-05-06 2011-12-11 Syntonix Pharmaceuticals Inc Immunoglobulin chimeric monomer-dimer hybrids
KR20060006975A (en) 2003-05-30 2006-01-20 제넨테크, 인크. Treatment with anti-vegf antibodies
CA2527694C (en) 2003-05-30 2015-07-14 Hendricus Renerus Jacobus Mattheus Hoogenboom Fab library for the preparation of anti vegf and anti rabies virus fabs
WO2005000898A2 (en) 2003-06-27 2005-01-06 Biogen Idec Ma Inc. Use of hydrophobic-interaction-chromatography or hinge-region modifications for the production of homogeneous antibody-solutions
AU2004255216B2 (en) 2003-07-01 2010-08-19 Immunomedics, Inc. Multivalent carriers of bi-specific antibodies
AR046071A1 (en) 2003-07-10 2005-11-23 Hoffmann La Roche ANTIBODIES AGAINST RECEIVER I OF THE INSULINAL TYPE GROWTH FACTOR AND THE USES OF THE SAME
EP1648511A1 (en) 2003-07-29 2006-04-26 Morphotek, Inc. Antibodies and methods for generating genetically altered antibodies with enhanced effector function
US20050106667A1 (en) 2003-08-01 2005-05-19 Genentech, Inc Binding polypeptides with restricted diversity sequences
WO2005044853A2 (en) 2003-11-01 2005-05-19 Genentech, Inc. Anti-vegf antibodies
JP2007503206A (en) 2003-08-22 2007-02-22 バイオジェン・アイデック・エムエイ・インコーポレイテッド Improved antibody with altered effector function and method for producing the antibody
EP1663306A2 (en) 2003-09-05 2006-06-07 Genentech, Inc. Antibodies with altered effector functions
US20050064509A1 (en) 2003-09-23 2005-03-24 The Regents Of The University Of California Use of templated self assembly to create novel multifunctional species
CN1326881C (en) 2003-09-29 2007-07-18 中国人民解放军军事医学科学院基础医学研究所 Trivalent bispecific antibody and its preparation process and use
WO2005035586A1 (en) 2003-10-08 2005-04-21 Kyowa Hakko Kogyo Co., Ltd. Fused protein composition
AU2004280065A1 (en) 2003-10-09 2005-04-21 Kyowa Hakko Kirin Co., Ltd. Process for producing antibody composition by using RNA inhibiting the function of alpha1,6-fucosyltransferase
EP2322569B1 (en) 2003-10-09 2020-08-26 Ambrx, Inc. Polymer derivatives for the selective modification of proteins
LT2380911T (en) 2003-11-05 2018-07-10 Roche Glycart Ag Antigen binding molecules with increased Fc receptor binding affinity and effector function
WO2005051976A2 (en) 2003-11-20 2005-06-09 Ansata Therapeutics, Inc. Protein and peptide ligation processes and one-step purification processes
WO2005053742A1 (en) 2003-12-04 2005-06-16 Kyowa Hakko Kogyo Co., Ltd. Medicine containing antibody composition
PL1718677T3 (en) 2003-12-19 2012-09-28 Genentech Inc Monovalent antibody fragments useful as therapeutics
MXPA06008496A (en) 2004-02-02 2007-01-30 Ambrx Inc Modified human interferon polypeptides and their uses.
WO2005075514A2 (en) 2004-03-10 2005-08-18 Lonza Ltd. Method for producing antibodies
EP2360186B1 (en) 2004-04-13 2017-08-30 F. Hoffmann-La Roche AG Anti-P-selectin antibodies
EP1786918A4 (en) 2004-07-17 2009-02-11 Imclone Systems Inc Novel tetravalent bispecific antibody
EP1778728A2 (en) 2004-08-19 2007-05-02 Genentech, Inc. Polypeptide variants with altered effector function
TWI309240B (en) 2004-09-17 2009-05-01 Hoffmann La Roche Anti-ox40l antibodies
PL1791565T3 (en) 2004-09-23 2016-10-31 Cysteine engineered antibodies and conjugates
JO3000B1 (en) 2004-10-20 2016-09-05 Genentech Inc Antibody Formulations.
CA2587766A1 (en) 2004-11-10 2007-03-01 Macrogenics, Inc. Engineering fc antibody regions to confer effector function
PL1838733T3 (en) 2004-12-21 2012-02-29 Medimmune Ltd Antibodies directed to angiopoietin-2 and uses thereof
PL1871805T3 (en) 2005-02-07 2020-03-31 Roche Glycart Ag Antigen binding molecules that bind egfr, vectors encoding same, and uses thereof
RU2007135216A (en) * 2005-02-23 2009-03-27 Мерримак Фармасьютикалз, Инк. (Us) BSPECIFIC BINDING AGENTS FOR MODULATION OF BIOLOGICAL ACTIVITY
WO2006093794A1 (en) 2005-02-28 2006-09-08 Centocor, Inc. Heterodimeric protein binding compositions
ES2592271T3 (en) 2005-03-31 2016-11-29 Chugai Seiyaku Kabushiki Kaisha Polypeptide production methods by regulating the association of polypeptides
TW200720289A (en) 2005-04-01 2007-06-01 Hoffmann La Roche Antibodies against CCR5 and uses thereof
WO2006113665A2 (en) 2005-04-15 2006-10-26 Macrogenics, Inc. Covalent diabodies and uses thereof
US9963510B2 (en) * 2005-04-15 2018-05-08 Macrogenics, Inc. Covalent diabodies and uses thereof
US20090215639A1 (en) 2005-04-26 2009-08-27 Bioren, Inc. Method of Producing Human IgG Antibodies with Enhanced Effector Functions
US8008443B2 (en) 2005-04-26 2011-08-30 Medimmune, Llc Modulation of antibody effector function by hinge domain engineering
WO2007014901A1 (en) 2005-08-02 2007-02-08 Basf Se Process for preparing xylylenediamine by continuous hydrogenation of phthalonitrile
US8008453B2 (en) * 2005-08-12 2011-08-30 Amgen Inc. Modified Fc molecules
US7612181B2 (en) * 2005-08-19 2009-11-03 Abbott Laboratories Dual variable domain immunoglobulin and uses thereof
BRPI0615026A8 (en) 2005-08-19 2018-03-06 Abbott Lab double variable domain immunoglobulin and its uses
CA2619298C (en) 2005-08-26 2017-07-04 Glycart Biotechnology Ag Modified antigen binding molecules with altered cell signaling activity
US8053569B2 (en) 2005-10-07 2011-11-08 Armagen Technologies, Inc. Nucleic acids encoding and methods of producing fusion proteins
WO2007044887A2 (en) 2005-10-11 2007-04-19 Transtarget, Inc. Method for producing a population of homogenous tetravalent bispecific antibodies
US7666622B2 (en) * 2005-10-19 2010-02-23 Regeneron Pharmaceuticals, Inc. Monomeric self-associating fusion polypeptides and therapeutic uses thereof
TW200732350A (en) 2005-10-21 2007-09-01 Amgen Inc Methods for generating monovalent IgG
FR2894959B1 (en) 2005-12-15 2008-02-29 Galderma Res & Dev RAR-GAMMA RECEPTOR SELECTIVE AGONIST BIPHENYL DERIVATIVES
KR101371773B1 (en) 2005-12-15 2014-03-07 아스트라제네카 아베 Combination of angiopoietin-2 antagonist and of vegf-a, kdr and/or flt1 antagonist for treating cancer
GB0601513D0 (en) 2006-01-25 2006-03-08 Univ Erasmus Medical Ct Binding molecules 3
AR059066A1 (en) 2006-01-27 2008-03-12 Amgen Inc COMBINATIONS OF THE ANGIOPOYETINE INHIBITOR -2 (ANG2) AND THE VASCULAR ENDOTELIAL GROWTH FACTOR INHIBITOR (VEGF)
AU2007215013A1 (en) 2006-02-15 2007-08-23 Imclone Systems Incorporated Functional antibodies
JP5374359B2 (en) * 2006-03-17 2013-12-25 バイオジェン・アイデック・エムエイ・インコーポレイテッド Stabilized polypeptide compounds
EP1996236A2 (en) 2006-03-22 2008-12-03 National Institute of Immunology Novel bioconjugates as therapeutic agent and synthesis thereof
AR060070A1 (en) 2006-03-24 2008-05-21 Merck Patent Gmbh HETERODYMERIC PROTEIN DOMAINS OBTAINED BY ENGINEERING
US20070274985A1 (en) 2006-05-26 2007-11-29 Stefan Dubel Antibody
EP2395021A1 (en) 2006-06-06 2011-12-14 Oleg Iliich Epshtein Medicinal agent for treating fatness, diabetes, and diseases associated with impaired glucose tolerance
WO2008005828A2 (en) 2006-06-30 2008-01-10 Novo Nordisk A/S PHARMACEUTICALLY ACCEPTABLE COMPOSITIONS COMPRISING ANTIBODY MOLECULES SPECIFIC TO LAMININ-5 α3 CHAIN DOMAINS G1G2 AND USE THEREOF
AR062223A1 (en) 2006-08-09 2008-10-22 Glycart Biotechnology Ag MOLECULES OF ADHESION TO THE ANTIGEN THAT ADHER TO EGFR, VECTORS THAT CODE THEM, AND THEIR USES OF THESE
US8759297B2 (en) 2006-08-18 2014-06-24 Armagen Technologies, Inc. Genetically encoded multifunctional compositions bidirectionally transported between peripheral blood and the cns
CN101205255A (en) 2006-12-14 2008-06-25 上海中信国健药业有限公司 Anti CD20 tetravalent antibody, preparation method and uses thereof
MY183404A (en) 2006-12-19 2021-02-18 Genentech Inc Vegf-specific antagonists for adjuvant and neoadjuvant therapy and the treatment of early stage tumors
US20080226635A1 (en) 2006-12-22 2008-09-18 Hans Koll Antibodies against insulin-like growth factor I receptor and uses thereof
PT2129396E (en) * 2007-02-16 2013-11-18 Merrimack Pharmaceuticals Inc Antibodies against erbb3 and uses thereof
US10259860B2 (en) 2007-02-27 2019-04-16 Aprogen Inc. Fusion proteins binding to VEGF and angiopoietin
US20090155275A1 (en) * 2007-07-31 2009-06-18 Medimmune, Llc Multispecific epitope binding proteins and uses thereof
AU2008286759B2 (en) 2007-08-15 2014-05-08 Isp Investments Inc. Polyvinylamide polymers containing polymerizable functionalities
US9624309B2 (en) 2007-08-15 2017-04-18 Bayer Intellectual Property Gmbh Monospecific and multispecific antibodies and method of use
DE102007038753A1 (en) 2007-08-16 2009-02-19 Giesecke & Devrient Gmbh Device and method for the calibration of a sensor system
US20090130105A1 (en) 2007-08-28 2009-05-21 Biogen Idec Ma Inc. Compositions that bind multiple epitopes of igf-1r
EP2050764A1 (en) 2007-10-15 2009-04-22 sanofi-aventis Novel polyvalent bispecific antibody format and uses thereof
US9266967B2 (en) 2007-12-21 2016-02-23 Hoffmann-La Roche, Inc. Bivalent, bispecific antibodies
US20090162359A1 (en) 2007-12-21 2009-06-25 Christian Klein Bivalent, bispecific antibodies
US8242247B2 (en) 2007-12-21 2012-08-14 Hoffmann-La Roche Inc. Bivalent, bispecific antibodies
US8227577B2 (en) 2007-12-21 2012-07-24 Hoffman-La Roche Inc. Bivalent, bispecific antibodies
EP2235064B1 (en) 2008-01-07 2015-11-25 Amgen Inc. Method for making antibody fc-heterodimeric molecules using electrostatic steering effects
JP2009181819A (en) 2008-01-31 2009-08-13 Hitachi High-Technologies Corp Charged particle beam device
JP4438875B2 (en) 2008-02-27 2010-03-24 三菱自動車工業株式会社 Vehicle fuel storage amount estimation device
CA2719924C (en) 2008-04-11 2017-10-03 Philip Tan Cd37 immunotherapeutic and combination with bifunctional chemotherapeutic thereof
BRPI0918947A2 (en) 2008-09-26 2015-12-01 Ucb Pharma Sa antibody fusion protein
AU2009296297A1 (en) 2008-09-26 2010-04-01 Roche Glycart Ag Bispecific anti-EGFR/anti-IGF-1R antibodies
US8268314B2 (en) 2008-10-08 2012-09-18 Hoffmann-La Roche Inc. Bispecific anti-VEGF/anti-ANG-2 antibodies
EP3524620A1 (en) 2008-10-14 2019-08-14 Genentech, Inc. Immunoglobulin variants and uses thereof
SG171812A1 (en) 2008-12-04 2011-07-28 Abbott Lab Dual variable domain immunoglobulins and uses thereof
WO2010087994A2 (en) 2009-01-30 2010-08-05 Whitehead Institute For Biomedical Research Methods for ligation and uses thereof
JP5501439B2 (en) 2009-04-02 2014-05-21 ロシュ グリクアート アクチェンゲゼルシャフト Multispecific antibody comprising a full-length antibody and a single chain Fab fragment
WO2010112194A1 (en) 2009-04-02 2010-10-07 F. Hoffmann-La Roche Ag Antigen-binding polypeptides and multispecific antibodies comprising them
JP5616428B2 (en) 2009-04-07 2014-10-29 ロシュ グリクアート アクチェンゲゼルシャフト Trivalent bispecific antibody
DE102009016373A1 (en) 2009-04-07 2010-10-21 V. KRÜTTEN MEDIZINISCHE EINMALGERÄTE GmbH Connector for the probe tube of an enteral feeding tube and assembly of an enteral feeding tube and an enteral transfer system
BRPI1014449A2 (en) 2009-04-07 2017-06-27 Roche Glycart Ag bispecific antibodies anti-erbb-2 / anti-c-met.
MX2011010166A (en) 2009-04-07 2011-10-11 Roche Glycart Ag Bispecific anti-erbb-3/anti-c-met antibodies.
EP2424567B1 (en) 2009-04-27 2018-11-21 OncoMed Pharmaceuticals, Inc. Method for making heteromultimeric molecules
EP2435473B1 (en) 2009-05-27 2013-10-02 F.Hoffmann-La Roche Ag Tri- or tetraspecific antibodies
US9676845B2 (en) 2009-06-16 2017-06-13 Hoffmann-La Roche, Inc. Bispecific antigen binding proteins
US8703132B2 (en) 2009-06-18 2014-04-22 Hoffmann-La Roche, Inc. Bispecific, tetravalent antigen binding proteins
WO2011003780A1 (en) 2009-07-06 2011-01-13 F. Hoffmann-La Roche Ag Bi-specific digoxigenin binding antibodies
US9493578B2 (en) 2009-09-02 2016-11-15 Xencor, Inc. Compositions and methods for simultaneous bivalent and monovalent co-engagement of antigens
WO2011034605A2 (en) 2009-09-16 2011-03-24 Genentech, Inc. Coiled coil and/or tether containing protein complexes and uses thereof
PT2483310E (en) 2009-09-29 2014-10-07 Roche Glycart Ag Bispecific death receptor agonistic antibodies
AU2010343057B2 (en) 2009-12-29 2017-02-23 Aptevo Research And Development Llc Heterodimer binding proteins and uses thereof
WO2011143545A1 (en) 2010-05-14 2011-11-17 Rinat Neuroscience Corporation Heterodimeric proteins and methods for producing and purifying them
BR112013002167A2 (en) 2010-08-24 2016-05-31 Roche Glycart Ag bispecific antibody, pharmaceutical composition, use, method of treatment of a cancer patient and a patient suffering from inflammation
WO2012025530A1 (en) 2010-08-24 2012-03-01 F. Hoffmann-La Roche Ag Bispecific antibodies comprising a disulfide stabilized - fv fragment
BR112013011811A2 (en) 2010-11-05 2023-02-23 Zymeworks Inc STABLE HETERODIMERIC ANTIBODY MODEL WITH MUTATIONS IN THE FC DOMAIN
PT2647707T (en) 2010-11-30 2018-11-15 Chugai Pharmaceutical Co Ltd Cytotoxicity-inducing therapeutic agent
EP2646470B1 (en) 2010-11-30 2017-03-01 F. Hoffmann-La Roche AG Low affinity anti-transferrin receptor antibodies and their use to transfer therapeutic scfv across the blood brain barrier
MX342034B (en) 2011-02-28 2016-09-12 Hoffmann La Roche Monovalent antigen binding proteins.
RU2607038C2 (en) 2011-02-28 2017-01-10 Ф. Хоффманн-Ля Рош Аг Antigen-binding proteins
EP2688909A2 (en) 2011-03-25 2014-01-29 Glenmark Pharmaceuticals S.A. Hetero-dimeric immunoglobulins
CA2828662A1 (en) 2011-04-20 2012-10-26 Roche Glycart Ag Method and constructs for the ph dependent passage of the blood-brain-barrier
WO2013003555A1 (en) 2011-06-28 2013-01-03 Whitehead Institute For Biomedical Research Using sortases to install click chemistry handles for protein ligation
WO2013012733A1 (en) 2011-07-15 2013-01-24 Biogen Idec Ma Inc. Heterodimeric fc regions, binding molecules comprising same, and methods relating thereto
US20130060011A1 (en) 2011-08-23 2013-03-07 Peter Bruenker Fc-free antibodies comprising two fab fragments and methods of use
LT2748201T (en) 2011-08-23 2018-02-26 Roche Glycart Ag Bispecific t cell activating antigen binding molecules
EP2748202B1 (en) 2011-08-23 2018-07-04 Roche Glycart AG Bispecific antigen binding molecules
ES2732712T3 (en) 2011-10-31 2019-11-25 Chugai Pharmaceutical Co Ltd Antigen binding molecule that has a regulated conjugation between the heavy chain and the light chain
LT2794905T (en) 2011-12-20 2020-07-10 Medimmune, Llc Modified polypeptides for bispecific antibody scaffolds
KR20140104963A (en) 2011-12-21 2014-08-29 에프. 호프만-라 로슈 아게 Rapid method for cloning and expression of cognate antibody variable region gene segments
WO2013150043A1 (en) 2012-04-05 2013-10-10 F. Hoffmann-La Roche Ag Bispecific antibodies against human tweak and human il17 and uses thereof
KR102382304B1 (en) 2012-04-20 2022-04-04 메뤼스 엔.페. Methods and means for the production of ig-like molecules
CA2869529A1 (en) 2012-05-24 2013-11-28 Raffaella CASTOLDI Multispecific antibodies
CA2871386A1 (en) 2012-06-27 2014-01-03 F. Hoffmann-La Roche Ag Method for the selection and production of tailor-made, selective and multi-specific therapeutic molecules comprising at least two different targeting entities and uses thereof
WO2014001325A1 (en) 2012-06-27 2014-01-03 F. Hoffmann-La Roche Ag Method for making antibody fc-region conjugates comprising at least one binding entity that specifically binds to a target and uses thereof
US20140154253A1 (en) 2012-07-13 2014-06-05 Zymeworks Inc. Bispecific Asymmetric Heterodimers Comprising Anti-CD3 Constructs
BR112015003938A2 (en) 2012-09-14 2018-09-04 Hoffmann La Roche polypeptide production methods, multispecific binder production, multispecific binder selection, bispecific antibody selection and combination determination, bispecific antibody, pharmaceutical formulation and use of bispecific antibody
EP3401337A1 (en) 2012-09-25 2018-11-14 Glenmark Pharmaceuticals S.A. Purification of hetero-dimeric immunoglobulins
UY35148A (en) 2012-11-21 2014-05-30 Amgen Inc HETERODIMERIC IMMUNOGLOBULINS
CA2903056A1 (en) 2013-03-15 2014-09-18 Merck Patent Gmbh Tetravalent bispecific antibodies
UA118028C2 (en) 2013-04-03 2018-11-12 Рош Глікарт Аг Bispecific antibodies specific for fap and dr5, antibodies specific for dr5 and methods of use
CN111057151B (en) 2014-01-06 2022-05-03 豪夫迈·罗氏有限公司 Monovalent blood brain barrier shuttle modules
EP3126389A1 (en) 2014-04-02 2017-02-08 F. Hoffmann-La Roche AG Method for detecting multispecific antibody light chain mispairing
EP3174897B1 (en) 2014-07-29 2020-02-12 F.Hoffmann-La Roche Ag Multispecific antibodies
EP2982692A1 (en) 2014-08-04 2016-02-10 EngMab AG Bispecific antibodies against CD3epsilon and BCMA
RU2017116020A (en) 2014-10-08 2018-11-12 Ф.Хоффманн-Ля Рош Аг COMBINED THERAPY BY SPECIFIC ANTIBODIES SPECIFIC TO FAP AND DR5, AND CHEMOTHERAPEUTIC AGENTS
AU2015329965A1 (en) 2014-10-09 2017-04-27 Engmab Sàrl Bispecific antibodies against CD3epsilon and ROR1
SG11201703426PA (en) 2014-11-10 2017-05-30 Hoffmann La Roche Anti-pdgf-b antibodies and methods of use
AU2015345321A1 (en) 2014-11-10 2017-04-20 F. Hoffmann-La Roche Ag Anti-ANG2 antibodies and methods of use
EP3218399A1 (en) 2014-11-10 2017-09-20 F. Hoffmann-La Roche AG Bispecific antibodies and methods of use in ophthalmology
CA2963719A1 (en) 2014-11-10 2016-05-19 F. Hoffmann-La Roche Ag Anti-il-1beta antibodies and methods of use
PL3227332T3 (en) * 2014-12-03 2020-06-15 F. Hoffmann-La Roche Ag Multispecific antibodies
RU2020124105A (en) 2015-06-24 2020-09-18 Ф. Хоффманн-Ля Рош Аг ANTIBODIES AGAINST TRANSFERRINE RECEPTOR WITH INDIVIDUAL AFFINITY
EP3350216A1 (en) 2015-09-15 2018-07-25 Amgen Inc. Tetravalent bispecific and tetraspecific antigen binding proteins and uses thereof
AR106188A1 (en) 2015-10-01 2017-12-20 Hoffmann La Roche ANTI-CD19 HUMANIZED HUMAN ANTIBODIES AND METHODS OF USE
JP7239320B2 (en) 2015-10-02 2023-03-14 エフ. ホフマン-ラ ロシュ アーゲー Multispecific antibody
EP3150636A1 (en) 2015-10-02 2017-04-05 F. Hoffmann-La Roche AG Tetravalent multispecific antibodies
WO2018162517A1 (en) 2017-03-10 2018-09-13 F. Hoffmann-La Roche Ag Method for producing multispecific antibodies
EP3704146B1 (en) 2017-11-01 2021-12-15 F. Hoffmann-La Roche AG Trifab-contorsbody
SG11202102859TA (en) 2018-12-21 2021-04-29 Hoffmann La Roche Antibodies binding to cd3
TW202106714A (en) 2019-04-25 2021-02-16 瑞士商赫孚孟拉羅股份公司 Generation of antibody-derived polypeptides by polypeptide chain exchange
AR119393A1 (en) 2019-07-15 2021-12-15 Hoffmann La Roche ANTIBODIES THAT BIND NKG2D
KR20220100963A (en) 2019-12-18 2022-07-18 에프. 호프만-라 로슈 아게 Antibodies that bind to HLA-A2/MAGE-A4
KR20230025665A (en) 2020-06-19 2023-02-22 에프. 호프만-라 로슈 아게 Antibodies that bind to CD3
JP2023538716A (en) 2020-06-19 2023-09-11 エフ・ホフマン-ラ・ロシュ・アクチェンゲゼルシャフト Antibodies that bind to CD3 and CD19

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6982321B2 (en) * 1986-03-27 2006-01-03 Medical Research Council Altered antibodies
WO1994009131A1 (en) * 1992-10-15 1994-04-28 Scotgen Limited Recombinant specific binding protein
WO1999066951A2 (en) * 1998-06-22 1999-12-29 Immunomedics, Inc. Use of bi-specific antibodies for pre-targeting diagnosis and therapy
US20070071742A1 (en) * 1998-11-30 2007-03-29 Fang Fang Humanized antibodies

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Merchant et al (Nat. Biotech., 16: 677-681, 1998) *

Cited By (118)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10927163B2 (en) 2007-12-21 2021-02-23 Hoffmann-La Roche, Inc. Bivalent, bispecific antibodies
US10138293B2 (en) 2007-12-21 2018-11-27 Hoffmann-La Roche, Inc. Bivalent, bispecific antibodies
US9382323B2 (en) 2009-04-02 2016-07-05 Roche Glycart Ag Multispecific antibodies comprising full length antibodies and single chain fab fragments
US20100256339A1 (en) * 2009-04-07 2010-10-07 Birgit Bossenmaier Bispecific Anti ErbB3 / Anti cMet Antibodies
US11993642B2 (en) 2009-04-07 2024-05-28 Hoffmann-La Roche Inc. Trivalent, bispecific antibodies
US9890204B2 (en) 2009-04-07 2018-02-13 Hoffmann-La Roche Inc. Trivalent, bispecific antibodies
US9676845B2 (en) 2009-06-16 2017-06-13 Hoffmann-La Roche, Inc. Bispecific antigen binding proteins
US10640555B2 (en) 2009-06-16 2020-05-05 Hoffmann-La Roche Inc. Bispecific antigen binding proteins
US11673945B2 (en) 2009-06-16 2023-06-13 Hoffmann-La Roche Inc. Bispecific antigen binding proteins
US8703132B2 (en) 2009-06-18 2014-04-22 Hoffmann-La Roche, Inc. Bispecific, tetravalent antigen binding proteins
US20100322934A1 (en) * 2009-06-18 2010-12-23 Sabine Imhof-Jung Bispecific, Tetravalent Antigen Binding Proteins
US9493578B2 (en) 2009-09-02 2016-11-15 Xencor, Inc. Compositions and methods for simultaneous bivalent and monovalent co-engagement of antigens
US9994646B2 (en) 2009-09-16 2018-06-12 Genentech, Inc. Coiled coil and/or tether containing protein complexes and uses thereof
US10106600B2 (en) 2010-03-26 2018-10-23 Roche Glycart Ag Bispecific antibodies
US9605061B2 (en) 2010-07-29 2017-03-28 Xencor, Inc. Antibodies with modified isoelectric points
US9879095B2 (en) 2010-08-24 2018-01-30 Hoffman-La Roche Inc. Bispecific antibodies comprising a disulfide stabilized-Fv fragment
US9217039B2 (en) 2010-11-01 2015-12-22 Symphogen A/S Anti-HER3 antibodies and compositions
WO2012059858A1 (en) * 2010-11-01 2012-05-10 Symphogen A/S Anti-her3 antibodies and compositions
AU2011324871A1 (en) * 2010-11-01 2013-05-09 Symphogen A/S Anti-HER3 antibodies and compositions
US11618790B2 (en) 2010-12-23 2023-04-04 Hoffmann-La Roche Inc. Polypeptide-polynucleotide-complex and its use in targeted effector moiety delivery
US9982036B2 (en) 2011-02-28 2018-05-29 Hoffmann-La Roche Inc. Dual FC antigen binding proteins
US10793621B2 (en) 2011-02-28 2020-10-06 Hoffmann-La Roche Inc. Nucleic acid encoding dual Fc antigen binding proteins
US10611825B2 (en) 2011-02-28 2020-04-07 Hoffmann La-Roche Inc. Monovalent antigen binding proteins
US9624291B2 (en) 2011-03-17 2017-04-18 Ramot At Tel-Aviv University Ltd. Bi- and monospecific, asymmetric antibodies and methods of generating the same
WO2012123949A1 (en) 2011-03-17 2012-09-20 Ramot At Tel-Aviv University Ltd. Bi- and monospecific, asymmetric antibodies and methods of generating the same
US20170174791A1 (en) * 2011-03-17 2017-06-22 Ramot At Tel-Aviv University Ltd. Bi- and monospecific, asymmetric antibodies and methods of generating the same
US10822428B2 (en) * 2011-03-17 2020-11-03 Ramot At Tel-Aviv University Ltd. Bi-and monospecific, asymmetric antibodies and methods of generating the same
EP2722343A1 (en) * 2011-06-20 2014-04-23 Kyowa Hakko Kirin Co., Ltd. ANTI-erbB3 ANTIBODY
US9034328B2 (en) 2011-06-20 2015-05-19 Kyowa Hakko Kirin Co., Ltd Anti-erbB3 antibody
EP2722343A4 (en) * 2011-06-20 2014-12-17 Kyowa Hakko Kirin Co Ltd ANTI-erbB3 ANTIBODY
US10851178B2 (en) 2011-10-10 2020-12-01 Xencor, Inc. Heterodimeric human IgG1 polypeptides with isoelectric point modifications
US9527927B2 (en) 2011-12-20 2016-12-27 Medimmune, Llc Modified polypeptides for bispecific antibody scaffolds
EP2794905A4 (en) * 2011-12-20 2015-11-18 Medimmune Llc Modified polypeptides for bispecific antibody scaffolds
US9688758B2 (en) 2012-02-10 2017-06-27 Genentech, Inc. Single-chain antibodies and other heteromultimers
US10106612B2 (en) 2012-06-27 2018-10-23 Hoffmann-La Roche Inc. Method for selection and production of tailor-made highly selective and multi-specific targeting entities containing at least two different binding entities and uses thereof
US11407836B2 (en) 2012-06-27 2022-08-09 Hoffmann-La Roche Inc. Method for selection and production of tailor-made highly selective and multi-specific targeting entities containing at least two different binding entities and uses thereof
US11421022B2 (en) 2012-06-27 2022-08-23 Hoffmann-La Roche Inc. Method for making antibody Fc-region conjugates comprising at least one binding entity that specifically binds to a target and uses thereof
WO2014108854A1 (en) 2013-01-09 2014-07-17 Fusimab Ltd. Monospecific anti-hgf and anti-ang2 antibodies and bispecific anti-hgf/anti-ang2 antibodies
US10472427B2 (en) 2013-01-14 2019-11-12 Xencor, Inc. Heterodimeric proteins
US10487155B2 (en) 2013-01-14 2019-11-26 Xencor, Inc. Heterodimeric proteins
US11718667B2 (en) 2013-01-14 2023-08-08 Xencor, Inc. Optimized antibody variable regions
US10131710B2 (en) 2013-01-14 2018-11-20 Xencor, Inc. Optimized antibody variable regions
US10738133B2 (en) 2013-01-14 2020-08-11 Xencor, Inc. Heterodimeric proteins
US11634506B2 (en) 2013-01-14 2023-04-25 Xencor, Inc. Heterodimeric proteins
US11053316B2 (en) 2013-01-14 2021-07-06 Xencor, Inc. Optimized antibody variable regions
US9701759B2 (en) 2013-01-14 2017-07-11 Xencor, Inc. Heterodimeric proteins
US10738132B2 (en) 2013-01-14 2020-08-11 Xencor, Inc. Heterodimeric proteins
US9650446B2 (en) 2013-01-14 2017-05-16 Xencor, Inc. Heterodimeric proteins
US9738722B2 (en) 2013-01-15 2017-08-22 Xencor, Inc. Rapid clearance of antigen complexes using novel antibodies
US10968276B2 (en) 2013-03-12 2021-04-06 Xencor, Inc. Optimized anti-CD3 variable regions
US10106624B2 (en) 2013-03-15 2018-10-23 Xencor, Inc. Heterodimeric proteins
US10519242B2 (en) 2013-03-15 2019-12-31 Xencor, Inc. Targeting regulatory T cells with heterodimeric proteins
US11299554B2 (en) 2013-03-15 2022-04-12 Xencor, Inc. Heterodimeric proteins
US10544187B2 (en) 2013-03-15 2020-01-28 Xencor, Inc. Targeting regulatory T cells with heterodimeric proteins
US10287364B2 (en) 2013-03-15 2019-05-14 Xencor, Inc. Heterodimeric proteins
US9605084B2 (en) 2013-03-15 2017-03-28 Xencor, Inc. Heterodimeric proteins
US11814423B2 (en) 2013-03-15 2023-11-14 Xencor, Inc. Heterodimeric proteins
US10858417B2 (en) 2013-03-15 2020-12-08 Xencor, Inc. Heterodimeric proteins
US10323099B2 (en) 2013-10-11 2019-06-18 Hoffmann-La Roche Inc. Multispecific domain exchanged common variable light chain antibodies
US9822186B2 (en) 2014-03-28 2017-11-21 Xencor, Inc. Bispecific antibodies that bind to CD38 and CD3
US10858451B2 (en) 2014-03-28 2020-12-08 Xencor, Inc. Bispecific antibodies that bind to CD38 and CD3
US11840579B2 (en) 2014-03-28 2023-12-12 Xencor, Inc. Bispecific antibodies that bind to CD38 and CD3
US10329556B2 (en) 2014-05-13 2019-06-25 Bioatla, Llc Conditionally active biological proteins
WO2015175375A1 (en) 2014-05-13 2015-11-19 Short Jay M Conditionally active biological proteins
WO2015173756A3 (en) * 2014-05-16 2016-01-28 Pfizer Inc. Bispecific antibodies with engineered ch1-cl interfaces
US10392438B2 (en) 2014-05-16 2019-08-27 Pfizer Inc. Bispecific antibodies
US10513699B2 (en) 2014-09-03 2019-12-24 Bioatla, Llc Discovering and producing conditionally active biologic proteins in the same eukaryotic cell production hosts
US11001643B2 (en) 2014-09-26 2021-05-11 Chugai Seiyaku Kabushiki Kaisha Cytotoxicity-inducing therapeutic agent
US9975966B2 (en) 2014-09-26 2018-05-22 Chugai Seiyaku Kabushiki Kaisha Cytotoxicity-inducing theraputic agent
US9856327B2 (en) 2014-11-26 2018-01-02 Xencor, Inc. Heterodimeric antibodies to CD3 X CD123
US10526417B2 (en) 2014-11-26 2020-01-07 Xencor, Inc. Heterodimeric antibodies that bind CD3 and CD38
US11945880B2 (en) 2014-11-26 2024-04-02 Xencor, Inc. Heterodimeric antibodies that bind CD3 and tumor antigens
US9850320B2 (en) 2014-11-26 2017-12-26 Xencor, Inc. Heterodimeric antibodies to CD3 X CD20
US11859011B2 (en) 2014-11-26 2024-01-02 Xencor, Inc. Heterodimeric antibodies that bind CD3 and tumor antigens
US11673972B2 (en) 2014-11-26 2023-06-13 Xencor, Inc. Heterodimeric antibodies that bind CD3 and tumor antigens
US10889653B2 (en) 2014-11-26 2021-01-12 Xencor, Inc. Heterodimeric antibodies that bind CD3 and tumor antigens
US10913803B2 (en) 2014-11-26 2021-02-09 Xencor, Inc. Heterodimeric antibodies that bind CD3 and tumor antigens
US10259887B2 (en) 2014-11-26 2019-04-16 Xencor, Inc. Heterodimeric antibodies that bind CD3 and tumor antigens
US11352442B2 (en) 2014-11-26 2022-06-07 Xencor, Inc. Heterodimeric antibodies that bind CD3 and CD38
US11225528B2 (en) 2014-11-26 2022-01-18 Xencor, Inc. Heterodimeric antibodies that bind CD3 and tumor antigens
US11111315B2 (en) 2014-11-26 2021-09-07 Xencor, Inc. Heterodimeric antibodies that bind CD3 and tumor antigens
US10633457B2 (en) 2014-12-03 2020-04-28 Hoffmann-La Roche Inc. Multispecific antibodies
US11999801B2 (en) 2014-12-03 2024-06-04 Hoffman-La Roche Inc. Multispecific antibodies
US10428155B2 (en) 2014-12-22 2019-10-01 Xencor, Inc. Trispecific antibodies
US10227411B2 (en) 2015-03-05 2019-03-12 Xencor, Inc. Modulation of T cells with bispecific antibodies and FC fusions
US11091548B2 (en) 2015-03-05 2021-08-17 Xencor, Inc. Modulation of T cells with bispecific antibodies and Fc fusions
US10526413B2 (en) 2015-10-02 2020-01-07 Hoffmann-La Roche Inc. Bispecific antibodies specific for OX40
US10227410B2 (en) 2015-12-07 2019-03-12 Xencor, Inc. Heterodimeric antibodies that bind CD3 and PSMA
US11623957B2 (en) 2015-12-07 2023-04-11 Xencor, Inc. Heterodimeric antibodies that bind CD3 and PSMA
US11492407B2 (en) 2016-06-14 2022-11-08 Xencor, Inc. Bispecific checkpoint inhibitor antibodies
US10787518B2 (en) 2016-06-14 2020-09-29 Xencor, Inc. Bispecific checkpoint inhibitor antibodies
US11236170B2 (en) 2016-06-14 2022-02-01 Xencor, Inc. Bispecific checkpoint inhibitor antibodies
US10316088B2 (en) 2016-06-28 2019-06-11 Xencor, Inc. Heterodimeric antibodies that bind somatostatin receptor 2
US11225521B2 (en) 2016-06-28 2022-01-18 Xencor, Inc. Heterodimeric antibodies that bind somatostatin receptor 2
WO2018002358A1 (en) 2016-06-30 2018-01-04 F. Hoffmann-La Roche Ag Improved adoptive t-cell therapy
US10793632B2 (en) 2016-08-30 2020-10-06 Xencor, Inc. Bispecific immunomodulatory antibodies that bind costimulatory and checkpoint receptors
US10501543B2 (en) 2016-10-14 2019-12-10 Xencor, Inc. IL15/IL15Rα heterodimeric Fc-fusion proteins
US10550185B2 (en) 2016-10-14 2020-02-04 Xencor, Inc. Bispecific heterodimeric fusion proteins containing IL-15-IL-15Rα Fc-fusion proteins and PD-1 antibody fragments
US11084863B2 (en) 2017-06-30 2021-08-10 Xencor, Inc. Targeted heterodimeric Fc fusion proteins containing IL-15 IL-15alpha and antigen binding domains
US10981992B2 (en) 2017-11-08 2021-04-20 Xencor, Inc. Bispecific immunomodulatory antibodies that bind costimulatory and checkpoint receptors
US11312770B2 (en) 2017-11-08 2022-04-26 Xencor, Inc. Bispecific and monospecific antibodies using novel anti-PD-1 sequences
US11319355B2 (en) 2017-12-19 2022-05-03 Xencor, Inc. Engineered IL-2 Fc fusion proteins
US10982006B2 (en) 2018-04-04 2021-04-20 Xencor, Inc. Heterodimeric antibodies that bind fibroblast activation protein
US10633458B2 (en) 2018-04-10 2020-04-28 Y-Biologics Inc. Cell engaging binding molecules
US10640576B2 (en) 2018-04-10 2020-05-05 Y-Biologics Inc. Cell engaging binding molecules
US10654944B2 (en) 2018-04-10 2020-05-19 Y-Biologics Inc. Cell engaging binding molecules
US10836833B2 (en) 2018-04-10 2020-11-17 Y-Biologics Inc. Cell engaging binding molecules
WO2019199916A1 (en) * 2018-04-13 2019-10-17 Eli Lilly And Company Fab-based trispecific antibodies
US11524991B2 (en) 2018-04-18 2022-12-13 Xencor, Inc. PD-1 targeted heterodimeric fusion proteins containing IL-15/IL-15Ra Fc-fusion proteins and PD-1 antigen binding domains and uses thereof
US11505595B2 (en) 2018-04-18 2022-11-22 Xencor, Inc. TIM-3 targeted heterodimeric fusion proteins containing IL-15/IL-15RA Fc-fusion proteins and TIM-3 antigen binding domains
US11358999B2 (en) 2018-10-03 2022-06-14 Xencor, Inc. IL-12 heterodimeric Fc-fusion proteins
US11472890B2 (en) 2019-03-01 2022-10-18 Xencor, Inc. Heterodimeric antibodies that bind ENPP3 and CD3
WO2021113748A1 (en) * 2019-12-05 2021-06-10 Arbele Corp. Composition of triaxial antibodies and method of making and using thereof
US11919956B2 (en) 2020-05-14 2024-03-05 Xencor, Inc. Heterodimeric antibodies that bind prostate specific membrane antigen (PSMA) and CD3
US11919958B2 (en) 2020-08-19 2024-03-05 Xencor, Inc. Anti-CD28 compositions
US11591401B2 (en) 2020-08-19 2023-02-28 Xencor, Inc. Anti-CD28 compositions
US11739144B2 (en) 2021-03-09 2023-08-29 Xencor, Inc. Heterodimeric antibodies that bind CD3 and CLDN6
US11859012B2 (en) 2021-03-10 2024-01-02 Xencor, Inc. Heterodimeric antibodies that bind CD3 and GPC3

Also Published As

Publication number Publication date
PT2417156E (en) 2015-04-29
PL2417156T3 (en) 2015-07-31
US9890204B2 (en) 2018-02-13
AU2010234031B2 (en) 2015-10-01
CY1116376T1 (en) 2017-02-08
CN102369214B (en) 2019-04-12
CA2757931A1 (en) 2010-10-14
JP5616428B2 (en) 2014-10-29
MX2011010168A (en) 2011-10-11
ES2537100T3 (en) 2015-06-02
AU2010234031A8 (en) 2012-06-28
US20200062826A1 (en) 2020-02-27
WO2010115589A8 (en) 2011-10-27
EP2417156A1 (en) 2012-02-15
BRPI1010297A2 (en) 2017-06-06
DK2417156T3 (en) 2015-03-02
SI2417156T1 (en) 2015-06-30
AR076193A1 (en) 2011-05-26
IL214884A (en) 2015-09-24
CN102369214A (en) 2012-03-07
CA2757931C (en) 2019-03-26
KR101456326B1 (en) 2014-11-12
HRP20150444T1 (en) 2015-05-22
US11993642B2 (en) 2024-05-28
AU2010234031A1 (en) 2011-09-08
JP2014193181A (en) 2014-10-09
TW201039850A (en) 2010-11-16
US20180282399A1 (en) 2018-10-04
US20130022601A1 (en) 2013-01-24
IL214884A0 (en) 2011-11-30
EP2417156B1 (en) 2015-02-11
JP2012522527A (en) 2012-09-27
WO2010115589A1 (en) 2010-10-14
SG175077A1 (en) 2011-11-28
KR20110130460A (en) 2011-12-05

Similar Documents

Publication Publication Date Title
US11993642B2 (en) Trivalent, bispecific antibodies
EP2414391B1 (en) Multispecific antibodies comprising full length antibodies and single chain fab fragments
US9879095B2 (en) Bispecific antibodies comprising a disulfide stabilized-Fv fragment
US20130273054A1 (en) Bispecific Anti ErbB2/Anti cMet Antibodies
US20140135482A1 (en) Bispecific Anti ErbB3 / Anti cMet Antibodies
MX2011013615A (en) Bispecific antigen binding proteins.
SE et al. G0) Priority Data:• HN, HR, HU, ID, IL, IN, IS, JP, KE, KG, KM, KN, KP

Legal Events

Date Code Title Description
AS Assignment

Owner name: F. HOFFMANN-LA ROCHE AG, SWITZERLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:BRINKMANN, ULRICH;CROASDALE, REBECCA;HOFFMANN, EIKE;AND OTHERS;SIGNING DATES FROM 20100412 TO 20100414;REEL/FRAME:024629/0461

Owner name: ROCHE GLYCART AG, SWITZERLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:UMANA, PABLO;MOESSNER, EKKEHARD;REEL/FRAME:024629/0438

Effective date: 20100414

Owner name: F. HOFFMANN-LA ROCHE AG, SWITZERLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:KLEIN, CHRISTIAN;REEL/FRAME:024642/0041

Effective date: 20100414

Owner name: ROCHE GLYCART AG, SWITZERLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:F. HOFFMANN-LA ROCHE AG;REEL/FRAME:024629/0451

Effective date: 20100414

STCB Information on status: application discontinuation

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