US20020004587A1 - Multivalent antibodies and uses therefor - Google Patents

Multivalent antibodies and uses therefor Download PDF

Info

Publication number
US20020004587A1
US20020004587A1 US09/813,341 US81334101A US2002004587A1 US 20020004587 A1 US20020004587 A1 US 20020004587A1 US 81334101 A US81334101 A US 81334101A US 2002004587 A1 US2002004587 A1 US 2002004587A1
Authority
US
United States
Prior art keywords
antibody
receptor
antigen
cell
cells
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
US09/813,341
Other languages
English (en)
Inventor
Kathy Miller
Leonard Presta
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Genentech Inc
Original Assignee
Genentech Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Genentech Inc filed Critical Genentech Inc
Priority to US09/813,341 priority Critical patent/US20020004587A1/en
Assigned to GENENTECH, INC. reassignment GENENTECH, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: MILLER, KATHY L., PRESTA, LEONARD G.
Publication of US20020004587A1 publication Critical patent/US20020004587A1/en
Priority to US11/218,821 priority patent/US20060025576A1/en
Priority to US11/535,031 priority patent/US20080299120A1/en
Priority to US12/939,117 priority patent/US20110110852A1/en
Priority to US13/470,189 priority patent/US8722859B2/en
Priority to US14/229,683 priority patent/US9493579B2/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/46Hybrid immunoglobulins
    • C07K16/468Immunoglobulins having two or more different antigen binding sites, e.g. multifunctional antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6875Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody being a hybrid immunoglobulin
    • A61K47/6879Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody being a hybrid immunoglobulin the immunoglobulin having two or more different antigen-binding sites, e.g. bispecific or multispecific immunoglobulin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2878Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30, CD40, CD95
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2896Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against molecules with a "CD"-designation, not provided for elsewhere
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/32Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against translation products of oncogenes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/55Fab or Fab'
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/64Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising a combination of variable region and constant region components
    • 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
    • C07K2317/77Internalization into the cell
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide

Definitions

  • the present invention concerns engineered antibodies, with three or more functional antigen binding sites, and uses, such as therapeutic uses, for such engineered antibodies.
  • Naturally occurring antibodies comprise two heavy chains linked together by disulfide bonds and two light chains, one light chain being linked to each of the heavy chains by disulfide bonds.
  • Each heavy chain has at one end a variable domain (VH) followed by a number of constant domains (three or four constant domains, CH1, CH2, CH3 and CH4, depending on the antibody class).
  • Each light chain has a variable domain (VL) at one end and a constant domain (CL) at its other end; the constant domain of the light chain is aligned with the first constant domain of the heavy chain, and the light chain variable domain is aligned with the variable domain of the heavy chain. See FIG. 1 herein.
  • variable domains are not involved directly in binding the antibody to an antigen, but are involved in various effector functions, such as participation of the antibody in antibody-dependent cell-mediated cytotoxicity (ADCC) and complement dependent cytotoxicity (CDC).
  • the variable domains of each pair of light and heavy chains are involved directly in binding the antibody to the antigen.
  • the variable domains of naturally occurring light and heavy chains have the same general structure; each comprising four framework regions (FRs), whose sequences are somewhat conserved, connected by three complementarity determining regions (CDRs) (see Kabat et al., Sequences of Proteins of Immunological Interest , National Institutes of Health, Bethesda, Md., (1991)).
  • the four FRs largely adopt a beta-sheet conformation and the CDRs form loops connecting, and in some cases forming part of, the beta-sheet structure.
  • the CDRs in each chain are held in close proximity by the FRs and, with the CDRs from the other chain, contribute to the formation of the antigen binding site.
  • FIGS. 2 A-E herein depict the structures of the five major naturally occurring immunoglobulin isotypes.
  • IgG, IgD and IgE immunoglobulins possess only two antigen binding sites.
  • IgA and IgM are capable of forming polymeric structures with higher valencies.
  • IgM is secreted by plasma cells as a pentamer in which five monomer units are held together by disulfide bonds linking their carboxyl-terminal (C ⁇ 4/C ⁇ 4) domains and C ⁇ 3/C ⁇ 3 domains.
  • the five monomer subunits are arranged with their Fc regions in the center of the pentamer and the 10 antigen-binding sites on the periphery of the molecule.
  • Each pentamer contains an additional Fc-linked polypeptide called the J (joining) chain, which is disulfide-bonded to the carboxyl-terminal cysteine residue of 2 of the 10 ⁇ chains.
  • the J chain appears to be required for polymerization of the monomers to form pentameric IgM; it is added just before secretion of the pentamer.
  • An IgM molecule can bind 10 small hapten molecules; however, because of steric hindrance, only 5 molecules of larger antigens can be bound simultaneously.
  • the increased valency of pentameric IgM increases its capacity to bind such multi-dimensional antigens as viral particles and red blood cells (RBCs).
  • IgA exists primarily as a monomer, although polymeric forms such as dimers, trimers, and even tetramers are sometimes seen.
  • the IgA of external secretions consists of a dimer or tetramer, a J-chain polypeptide, and a polypeptide chain called secretory component.
  • the art has attempted to overcome this problem by constructing “chimeric” antibodies in which an animal antigen binding variable domain is coupled to a human constant domain (Cabilly et al., U.S. Pat. No. 4,816,567; Morrison et al., Proc. Natl. Acad. Sci. USA 81:6851-6855 (1984); Boulianne et al., Nature 312:643-646 (1984); and Neuberger et al., Nature 314:268-270 (1985)).
  • the isotype of the human constant domain may be selected to tailor the chimeric antibody for participation in ADCC and CDC (see e.g. Brüggemann et al., J. Exp. Med.
  • such chimeric antibodies contain about one third rodent (or other non-human species) sequence and thus are capable of eliciting a significant anti-globulin response in humans.
  • rodent or other non-human species
  • OKT3 much of the resulting anti-globulin response is directed against the variable region rather than the constant region
  • a chimeric/humanized antibody instead of a chimeric/humanized antibody, one may treat a patient with a human antibody in order to avoid human antibodies raised against a murine antibody (known as the “HAMA response”).
  • HAMA response human antibodies raised against a murine antibody
  • Several technologies are available for generating human antibodies.
  • Human antibodies may be selected using phage display technology. Phage display has been adapted to select human antibodies from an unimmunized donor (Marks et al. J. Mol. Biol. 222:581-597 (1991)). According to this approach, PCR is used to amplify variable domain genes from mRNA prepared from human peripheral blood lymphocytes (PBLs). Primers are used such that DNA from both IgG and IgM heavy chains and both ⁇ and ⁇ chains is amplified. These genes are then randomly combined and expressed as single chain Fv (scFv) fused to the gene III coat protein of M13 phage. Human antibodies against an antigen of interest may then be identified by rounds of growth and selection by binding to that antigen (e.g. to the immobilized antigen). See Griffiths et al. EMBO J. 12:725-734 (1993).
  • “Synthetic” phage-antibody repertoires have also been built from cloned human VH-gene segments.
  • a repertoire (2 ⁇ 10 7 clones) was first constructed using a short H3 loop of five or eight random residues with each of 49 segments, and combined with a fixed light chain (Hoogenboom et al. J. Mol. Biol. 227:381-388 (1992)).
  • H3 loops of different lengths, up to 12 residues, a single library was created from which a range of more than 20 binding specificities could be selected (Winter et al. Ann. Rev. Immuno. 12:433-55 (1994)).
  • the affinity of low affinity “primary” phage-antibodies may be improved by using phage display technology.
  • One approach is to use a chain-shuffling strategy in which the VH domain is held constant and then recombined with the original library of VL genes and tighter binders selected by binding to immobilized antigen. This cycle is repeated by fixing the new VL domain and recombining with the original VH library (Marks et al. Bio/Technology 10:779-783 (1992)).
  • point mutations in the primary antibody may be introduced using error-prone PCR and higher affinity binders selected by using phage display. Gram et al. PNAS ( USA ) 89: 3576-3580 (1992).
  • Severe combined immune deficient (SCID) mice lack the ability to produce their own immunoglobulins due to a defect in the recombinase gene.
  • SCID mice Several groups have reconstituted a functional humoral immune system in these mice by transfer of human peripheral blood lymphocytes (PBLs). These hu-PBL-SCID mice can be used to raise human antibodies upon immunization with antigen. Duchosal et al. Nature 355:258-262 (1992).
  • mice are turned off and then yeast artificial chromosomes (YACs) engineered with large DNA sequences containing human heavy- and light-chain genes are introduced into the mice.
  • yeast artificial chromosomes YACs
  • Such “XenoMice” are able to produce human antibodies upon immunization with an antigen of interest. See U.S. Pat. No. 5,434,340; US Patent No. 5,591,699; US Patent No. 5,569,825; US Patent No. 5,545,806; and US Patent No. 5,545,807.
  • Human monoclonal antibodies may also be generated by immortalizing a human B lymphocyte producing an antibody of interest.
  • the ethical issues surrounding immunizing humans in order to generate activated human B lymphocytes can be avoided by immunizing human lymphocytes in vitro.
  • human PBLs Boerner et al. Proc. Natl. Acad. Sci. USA 85:3995-4000 (1988)
  • human splenocytes Boerner et al. J. Immunol. 147, 86-95 (1991)
  • Improvements in human hybridoma technology have been achieved by using a mouse-human heterohybrid as the fusion partner (Boerner et al.).
  • Antibodies have been modified in order to increase their antigen-binding valency. For instance, Ghetie et al. homodimerized tumor-reactive monoclonal antibodies (anti-CD19, anti-CD20, anti-CD21, anti-CD22 and anti-HER2 antibodies) by chemically introducing a thioether bond between a pair of IgGs using two heterobifunctional crosslinkers. Ghetie et al., PNAS ( USA ) 94:7509-7514 (1997); and WO 99/02567. Wolff et al.
  • the homodimeric IgG was said to have similar avidity to the parent IgG, but apparently showed an improved ability to internalize and retain radioisotope in target leukemia cells, and was more potent at complement-mediated leukemia cell killing and antibody-dependent cellular cytotoxicity using human effectors.
  • Coloma and Morrison Nature Biotech. 15: 159-163 describe a tetravalent bispecific antibody which was engineered by fusing DNA encoding a single chain anti-dansyl antibody Fv (scFv) after the C terminus (CH3-scFv) or after the hinge (Hinge-scFv) of an IgG3 anti-dansyl antibody. See, also, WO95/09917. Smith and Morrison engineered three versions of mu-like IgG3 by engineering either (1) Cys414 of an IgM heavy chain or (2) Cys575 of an IgM heavy chain, or both (1) and (2), into the IgG3 heavy chain gene.
  • U.S. Pat. No. 5,641,870 (Rinderknecht et al.) describes a bivalent, linear F(ab′) 2 fragment comprising tandem repeats of a heavy chain fragment (VH-CH1-VH-CH1) cosecreted with a light chain. The C-terminus of CH1 was joined directed to the N-terminus of VH without any extraneous linking protein sequences.
  • Other publications on antibody variants include WO 00/06605; U.S. Pat. No. 5,591,828; US Patent No. 5,959,083; US Patent No. 6,027,725; WO98/58965; WO94/13804; Tutt et al. J. Immunol.
  • the ErbB receptor tyrosine kinases are important mediators of cell growth, differentiation and survival.
  • the receptor family includes at least four distinct members including Epidermal Growth Factor Receptor (EGFR or ErbB1), HER2 (ErbB2 or p185 neu ), HER3 (ErbB3) and HER4 (ErbB4 or tyro2).
  • EGFR Epidermal Growth Factor Receptor
  • HER2 ErbB2 or p185 neu
  • HER3 ErbB3
  • HER4 ErbB4 or tyro2
  • EGFR encoded by the erbB1 gene
  • increased expression of EGFR has been observed in breast, bladder, lung, head, neck and stomach cancer, as well as glioblastomas.
  • Increased EGFR receptor expression is often associated with increased production of the EGFR ligand, Transforming Growth Factor alpha (TGF-alpha), by the same tumor cells resulting in receptor activation by an autocrine stimulatory pathway.
  • TGF-alpha Transforming Growth Factor alpha
  • Monoclonal antibodies directed against the EGFR or its ligands, TGF-alpha and EGF have been evaluated as therapeutic agents in the treatment of such malignancies. See, e.g., Baselga and Mendelsohn., supra; Masui et al. Cancer Research 44:1002-1007 (1984); and Wu et al. J. Clin. Invest. 95:1897-1905 (1995).
  • the second member of the ErbB family, p185 neu was originally identified as the product of the transforming gene from neuroblastomas of chemically treated rats.
  • the activated form of the neu proto-oncogene results from a point mutation (valine to glutamic acid) in the transmembrane region of the encoded protein.
  • Amplification of the human homolog of neu is observed in breast and ovarian cancers and correlates with a poor prognosis (Slamon et al., Science, 235:177-182 (1987); Slamon et al., Science, 244:707-712 (1989); and U.S. Pat No. 4,968,603).
  • HER2 (frequently but not uniformly due to gene amplification) has also been observed in other carcinomas including carcinomas of the stomach, endometrium, salivary gland, lung, kidney, colon, thyroid, pancreas and bladder.
  • Hudziak et al., Mol. Cell. Biol. 9(3):1165-1172 (1989) describe the generation of a panel of anti-HER2 antibodies which were characterized using the human breast tumor cell line SKBR3. Relative cell proliferation of the SKBR3 cells following exposure to the antibodies was determined by crystal violet staining of the monolayers after 72 hours. Using this assay, maximum inhibition was obtained with the antibody called 4D5 which inhibited cellular proliferation by 56%. Other antibodies in the panel reduced cellular proliferation to a lesser extent in this assay. The antibody 4D5 was further found to sensitize HER2-overexpressing breast tumor cell lines to the cytotoxic effects of TNF-alpha. See, also, U.S. Pat. No.
  • a recombinant humanized IgG1 version of the murine anti-HER2 antibody 4D5 (rhuMAb HER2 or HERCEPTIN®; commercially available from Genentech, Inc., South San Francisco) is clinically active in patients with HER2-overexpressing metastatic breast cancers that have received extensive prior anti-cancer therapy (Baselga et al., J. Clin. Oncol. 14:737-744 (1996)).
  • HERCEPTIN® received marketing approval from the Food and Drug Administration Sep. 25, 1998 for the treatment of patients with metastatic breast cancer whose tumors overexpress the HER2 protein.
  • the ErbB receptors are generally found in various combinations in cells and heterodimerization is thought to increase the diversity of cellular responses to a variety of ErbB ligands (Earp et al. Breast Cancer Research and Treatment 35: 115-132(1995)).
  • EGFR is bound by six different ligands; Epidermal Growth Factor (EGF), Transforming Growth Factor alpha (TGF-alpha), amphiregulin, Heparin Binding Epidermal Growth Factor (HB-EGF), betacellulin and epiregulin (Groenen et al. Growth Factors 11:235-257 (1994)).
  • a family of heregulin proteins resulting from alternative splicing of a single gene are ligands for HER3 and HER4.
  • the heregulin family includes alpha, beta and gamma heregulins (Holmes et al., Science, 256:1205-1210 (1992); U.S. Pat. No. 5,641,869; and Schaefer et al. Oncogene 15:1385-1394 (1997)); neu differentiation factors (NDFs), glial growth factors (GGFs); acetylcholine receptor inducing activity (ARIA); and sensory and motor neuron derived factor (SMDF).
  • NDFs neu differentiation factors
  • GGFs glial growth factors
  • ARIA acetylcholine receptor inducing activity
  • SMDF sensory and motor neuron derived factor
  • EGF and TGF-alpha do not bind HER2, EGF stimulates EGFR and HER2 to form a heterodimer, which activates EGFR and results in transphosphorylation of HER2 in the heterodimer. Dimerization and/or transphosphorylation appears to activate the HER2 tyrosine kinase. See Earp et al., supra.
  • HER3 is co-expressed with HER2
  • an active signaling complex is formed and antibodies directed against HER2 are capable of disrupting this complex (Sliwkowski et al., J. Biol. Chem., 269(20):14661-14665 (1994)).
  • HER3 for heregulin (HRG) is increased to a higher affinity state when co-expressed with HER2.
  • HRG heregulin
  • HER4 like HER3, forms an active signaling complex with HER2 (Carraway and Cantley, Cell 78:5-8 (1994)).
  • TNF-alpha Tumor Necrosis Factor-alpha
  • TNF-beta Tumor Necrosis Factor-beta
  • LT-alpha Lymphotoxin-alpha
  • CD30 ligand CD27 ligand
  • CD40 ligand OX-40 ligand
  • 4-1 BB ligand Apo-1 ligand (also referred to as Fas ligand or CD95 ligand)
  • Apo-2 ligand also referred to as TRAIL
  • Apo-3 ligand also referred to as TWEAK
  • osteoprotegerin OPG
  • APRIL RANK ligand
  • TRANCE TALL-1
  • BlyS Tumor Necrosis Factor
  • TNF-alpha, TNF-beta, CD30 ligand, 4-1BB ligand, Apo-1 ligand, Apo-2 ligand (Apo2L/TRAIL) and Apo-3 ligand (TWEAK) have been reported to be involved in apoptotic cell death.
  • Both TNF-alpha and TNF-beta have been reported to induce apoptotic death in susceptible tumor cells (Schmid et al., Proc. Natl. Acad. Sci., 83:1881 (1986); Dealtry et al., Eur. J. Immunol., 17:689 (1987)).
  • TNF-alpha is involved in post-stimulation apoptosis of CD8-positive T cells (Zheng et al., Nature, 377:348-351 (1995)).
  • CD30 ligand may be involved in deletion of self-reactive T cells in the thymus (Amakawa et al., Cold Spring Harbor Laboratory Symposium on Programmed Cell Death , Abstr. No. 10, (1995)).
  • CD40 ligand activates many functions of B cells, including proliferation, immunoglobulin secretion, and survival (Renshaw et al., J. Exp. Med., 180:1889 (1994)).
  • TNF family cytokine TALL-1 (BlyS)
  • BlyS TNF family cytokine
  • Apo-1 ligand is also reported to induce post-stimulation apoptosis in CD4-positive T lymphocytes and in B lymphocytes, and may be involved in the elimination of activated lymphocytes when their function is no longer needed (Krammer et al., supra; Nagata et al., supra).
  • Agonist mouse monoclonal antibodies specifically binding to the Apo-1 receptor have been reported to exhibit cell killing activity that is comparable to or similar to that of TNF-alpha (Yonehara et al., J. Exp. Med., 169:1747-1756 (1989)).
  • TNF family cytokines Induction of various cellular responses mediated by such TNF family cytokines is believed to be initiated by their binding to specific cell receptors.
  • TNF receptors Two distinct TNF receptors of approximately 55-kDa (TNFR1) and 75-kDa (TNFR2) were identified (Hohman et al., J. Biol. Chem., 264:14927-14934 (1989); Brockhaus et al., Proc. Natl. Acad. Sci., 87:3127-3131 (1990); EP 417,563, published Mar.
  • Those TNFRs were found to share the typical structure of cell surface receptors including extracellular, transmembrane and intracellular regions.
  • TNFR1 and TNFR2 The extracellular portion of type 1 and type 2 TNFRs (TNFR1 and TNFR2) contains a repetitive amino acid sequence pattern of four cysteine-rich domains (CRDs) designated 1 through 4, starting from the NH 2 -terminus.
  • CCDs cysteine-rich domains
  • CRDs CRDs
  • NGFR nerve growth factor receptor
  • CD40 B cell antigen CD40
  • OX40 T cell antigen OX40
  • Fas antigen Yonehara et al., supra and Itoh et al., Cell, 66:233-243 (1991)
  • CRDs are also found in the soluble TNFR (sTNFR)-like T2 proteins of the Shope and myxoma poxviruses (Upton et al., Virology, 160:20-29 (1987); Smith et al., Biochem. Biophys. Res. Commun., 176:335 (1991); Upton et al., Virology, 184:370 (1991)).
  • sTNFR soluble TNFR
  • Optimal alignment of these sequences indicates that the positions of the cysteine residues are well conserved.
  • These receptors are sometimes collectively referred to as members of the TNF/NGF receptor superfamily.
  • TNF family ligands identified to date are type 11 transmembrane proteins, whose C-terminus is extracellular.
  • most receptors in the TNF receptor (TNFR) family identified to date are type I transmembrane proteins.
  • ECD extracellular domain
  • TNF family cytokines including TNF-alpha, Apo-1 ligand and CD40 ligand, are cleaved proteolytically at the cell surface; the resulting protein in each case typically forms a homotrimeric molecule that functions as a soluble cytokine.
  • TNF receptor family proteins are also usually cleaved proteolytically to release soluble receptor ECDs that can function as inhibitors of the cognate cytokines.
  • TACI Transmembrane Activator and CAML-Interactor
  • Apo-3 has also been referred to by other investigators as DR3, wsl-1, TRAMP, and LARD (Chinnaiyan et al., Science, 274:990 (1996); Kitson et al., Nature, 384:372 (1996); Bodmer et al., Immunity, 6:79 (1997); Screaton et al., Proc. Natl. Acad. Sci., 94:4615-4619 (1997)).
  • Pan et al. have disclosed another TNF receptor family member referred to as “DR4” (Pan et al., Science, 276:111-113 (1997); see also WO98/32856 published Jul. 30, 1998).
  • the DR4 was reported to contain a cytoplasmic death domain capable of engaging the cell suicide apparatus.
  • Pan et al. disclose that DR4 is believed to be a receptor for the ligand known as Apo2L/TRAIL.
  • DR5 is reported to contain a cytoplasmic death domain and be capable of signaling apoptosis.
  • the crystal structure of the complex formed between Apo2L/TRAIL and DR5 is described in Hymowitz et al., Molecular Cell, 4:563-571 (1999).
  • DR6 death domain-containing receptor 6
  • DR6 is believed to contain a putative leucine-zipper sequence that overlaps with a proline-rich motif in the cytoplasmic region.
  • the proline-rich motif resembles sequences that bind to src-homology-3 domains, which are found in many intracellular signal-transducing molecules.
  • DcR1 also referred to as TRID, LIT or TRAIL-R3
  • TRID TRID
  • TRAIL-R3 TRID, LIT
  • McFarlane et al. J. Biol. Chem., 272:25417-25420 (1997); Schneider et al., FEBS Letters, 416:329-334 (1997); Degii-Esposti et al., J. Exp.
  • TNFR family include CAR1, HVEM, GITR, ZTNFR-5, NTR-1, and TNFL1 (Brojatsch et al., Cell, 87:845-855 (1996); Montgomery et al., Cell, 87:427-436 (1996); Marsters et al., J. Biol. Chem., 272:14029-14032 (1997); Nocentini et al., Proc. Natl. Acad. Sci. USA 94:6216-6221 (1997); Emery et al., J. Biol. Chem., 273:14363-14367 (1998); WO99/04001 published Jan. 28, 1999; WO99/07738 published Feb. 18, 1999; WO99/33980 published Jul. 8, 1999).
  • NF- ⁇ B is the prototype of a family of dimeric transcription factors whose subunits contain conserved Rel regions (Verma et al., Genes Develop., 9:2723-2735 (1996); Baldwin, Ann. Rev. Immunol., 14:649-681 (1996)).
  • NF- ⁇ B In its latent form, NF- ⁇ B is complexed with members of the I- ⁇ B inhibitor family; upon inactivation of the I- ⁇ B in response to certain stimuli, released NF- ⁇ B translocates to the nucleus where it binds to specific DNA sequences and activates gene transcription.
  • the TNFR members identified to date either include or lack an intracellular death domain region.
  • Some TNFR molecules lacking a death domain, such as TNFR2, CD40, HVEM, and GITR are capable of modulating NF- ⁇ B activity. (see, e.g., Lotz et al., J. Leukocyte Biol., 60:1-7 (1996)).
  • Lymphocytes are one of many types of white blood cells produced in the bone marrow during the process of hematopoiesis. There are two major populations of lymphocytes: B lymphocytes (B cells) and T lymphocytes (T cells). The lymphocytes of particular interest herein are B cells.
  • B cells mature within the bone marrow and leave the marrow expressing an antigen-binding antibody on their cell surface.
  • a naive B cell first encounters the antigen for which its membrane-bound antibody is specific, the cell begins to divide rapidly and its progeny differentiate into memory B cells and effector cells called “plasma cells”.
  • Memory B cells have a longer life span and continue to express membrane-bound antibody with the same specificity as the original parent cell.
  • Plasma cells do not produce membrane-bound antibody but instead produce the antibody in a form that can be secreted. Secreted antibodies are the major effector molecule of humoral immunity.
  • the CD20 antigen also called human B-lymphocyte-restricted differentiation antigen, Bp35
  • Bp35 human B-lymphocyte-restricted differentiation antigen
  • CD20 regulates an early step(s) in the activation process for cell cycle initiation and differentiation (Tedder et al., supra) and possibly functions as a calcium ion channel (Tedder et al., J. Cell. Biochem. 14D:195 (1990)).
  • this antigen can serve as a candidate for “targeting” of such lymphomas.
  • targeting can be generalized as follows: antibodies specific to the CD20 surface antigen of B cells are administered to a patient. These anti-CD20 antibodies specifically bind to the CD20 antigen of (ostensibly) both normal and malignant B cells; the antibody bound to the CD20 surface antigen may lead to the destruction and depletion of neoplastic B cells. Additionally, chemical agents or radioactive labels having the potential to destroy the tumor can be conjugated to the anti-CD20 antibody such that the agent is specifically “delivered” to the neoplastic B cells. Irrespective of the approach, a primary goal is to destroy the tumor; the specific approach can be determined by the particular anti-CD20 antibody which is utilized and, thus, the available approaches to targeting the CD20 antigen can vary considerably.
  • CD19 is another antigen that is expressed on the surface of cells of the B lineage. Like CD20, CD19 is found on cells throughout differentiation of the lineage from the stem cell stage up to a point just prior to terminal differentiation into plasma cells (Nadler, L. Lymphocyte Typing II 2: 3-37 and Appendix, Renling et al. eds. (1986) by Springer Verlag). Unlike CD20 however, antibody binding to CD19 causes internalization of the CD19 antigen. CD19 antigen is identified by the HD237-CD19 antibody (also called the “B4” antibody) (Kiesel et al. Leukemia Research II, 12: 1119 (1987)), among others.
  • HD237-CD19 antibody also called the “B4” antibody
  • the CD19 antigen is present on 4-8% of peripheral blood mononuclear cells and on greater than 90% of B cells isolated from peripheral blood, spleen, lymph node or tonsil. CD19 is not detected on peripheral blood T cells, monocytes or granulocytes.
  • Virtually all non-T cell acute lymphoblastic leukemias (ALL), B cell chronic lymphocytic leukemias (CLL) and B cell lymphomas express CD19 detectable by the antibody B4 (Nadler et al. J. Immunol. 131:244 (1983); and Nadler et al. in Progress in Hematology Vol. XII pp. 187-206. Brown, E. ed. (1981) by Grune & Stratton, Inc).
  • the rituximab (RITUXAN®) antibody is a genetically engineered chimeric murine/human monoclonal antibody directed against the CD20 antigen.
  • Rituximab is the antibody called “C2B8” in U.S. Pat. No. 5,736,137 issued Apr. 7, 1998 (Anderson et al.).
  • RITUXAN® is indicated for the treatment of patients with relapsed or refractory low-grade or follicular, CD20 positive, B cell non-Hodgkin's lymphoma. In vitro mechanism of action studies have demonstrated that RITUXAN® binds human complement and lyses lymphoid B cell lines through CDC (Reff et al.
  • RITUXAN® has been shown to have anti-proliferative effects in tritiated thymidine incorporation assays and to induce apoptosis directly, while other anti-CD19 and CD20 antibodies do not (Maloney et al., Blood 88(10):637a (1996)). Synergy between RITUXAN® and chemotherapies and toxins has also been observed experimentally.
  • RITUXAN® sensitizes drug-resistant human B cell lymphoma cell lines to the cytotoxic effects of doxorubicin, CDDP, VP-16, diphtheria toxin and ricin (Demidem et al. Cancer Chemotherapy & Radiopharmaceuticals 12(3):177-186 (1997)).
  • doxorubicin doxorubicin
  • CDDP CDDP
  • VP-16 diphtheria toxin
  • ricin diphtheria toxin
  • ricin diphtheria toxin
  • ricin diphtheria toxin and ricin
  • the present invention provides multivalent antibodies (e.g. tetravalent antibodies) with three or more antigen binding sites, which can be readily produced by recombinant expression of nucleic acid encoding the polypeptide chains of the antibody.
  • the multivalent antibody comprises a dimerization domain and three or more antigen binding sites.
  • the preferred dimerization domain comprises (or consists of) an Fc region or a hinge region.
  • the invention provides an isolated antibody comprising a dimerization domain and three or more antigen binding sites amino-terminal thereto.
  • the invention further provides an isolated antibody comprising an Fc region and three or more antigen binding sites amino-terminal to the Fc region.
  • the preferred multivalent antibody herein comprises (or consists of) three to about eight, but preferably four, antigen binding sites (which are generally all “functional”, as hereindefined). In one embodiment, the multivalent antibody comprises five or more (e.g. up to about eight) antigen binding sites.
  • the multivalent antibody herein is preferably not a native sequence IgA or IgM, and may lack an Fc region or have only one Fc region.
  • the multivalent antibody comprises at least one polypeptide chain (and preferably two polypeptide chains), wherein the polypeptide chain(s) comprise two or more variable domains.
  • the polypeptide chain(s) may comprise VD1-(X1) n -VD2-(X2) n -Fc, wherein VD1 is a first variable domain, VD2 is a second variable domain, Fc is one polypeptide chain of an Fc region, X1 and X2 represent an amino acid or polypeptide, and n is 0 or 1.
  • the polypeptide chain(s) may comprise: VH-CH1-flexible linker-VH-CH1-Fc region chain; VH-CH1-VH-CH1-Fc region chain; VL-CL-flexible linker-VL-CL-Fc region chain; or VL-CL-VL-CL-Fc region chain.
  • the flexible linker may comprise a peptide such as gly-ser, gly-ser-gly-ser (SEQ ID NO:10), ala-ser, or gly-gly-gly-ser (SEQ ID NO:11).
  • the multivalent antibody herein preferably further comprises at least two (and preferably four) light chain variable domain polypeptides.
  • the multivalent antibody herein may, for instance, comprise from about two to about eight light chain variable domain polypeptides.
  • the light chain variable domain polypeptides contemplated here comprise a light chain variable domain and, optionally, further comprise a CL domain.
  • the multivalent antibodies herein have properties which are desirable, among other things, from a therapeutic standpoint.
  • the multivalent antibody may (1) be internalized (and/or catabolized) faster than a bivalent antibody by a cell expressing an antigen to which the antibodies bind; (2) be an agonist antibody; and/or (3) induce cell death and/or apoptosis of a cell expressing an antigen which the multivalent antibody is capable of binding to.
  • the “parent antibody” which provides at least one antigen binding specificity of the multivalent antibody may be one which is internalized (and/or catabolized) by a cell expressing an antigen to which the antibody binds; and/or may be an agonist, cell-death-inducing, and/or apoptosis-inducing antibody, and the multivalent form of the antibody as described herein may display improvement(s) in one or more of these properties.
  • the parent antibody may lack any one or more of these properties, but may be endowed with them when constructed as a multivalent antibody as hereindescribed.
  • the three or more antigen binding sites of the multivalent antibodies herein may all bind the same antigen; or may bind two or more (e.g. from two to about three) different antigens.
  • the multivalent antibody may bind (1) a cell surface protein expressed (or overexpressed) by tumor cells, e.g. Epidermal Growth Factor Receptor (EGFR), HER2 receptor, HER3 receptor, HER4 receptor, or DcR3; (2) a receptor in the Tumor Necrosis Factor (TNF) receptor superfamily (e.g. an Apo2L receptor, such as DR4, DR5, DcR1 or DcR2); and/or (3) a B cell surface antigen (such as CD19, CD20, CD22 or CD40).
  • EGFR Epidermal Growth Factor Receptor
  • HER2 receptor HER3 receptor
  • HER4 receptor HER4 receptor
  • DcR3 DcR3
  • TNF Tumor Necrosis Factor
  • B cell surface antigen such as CD19, CD20, CD22 or CD40.
  • all of the functional antigen binding sites of the multivalent antibody bind the same antigen as listed above (e.g. all four antigen binding sites of
  • the invention also provides immunoconjugates comprising the multivalent antibody conjugated with a cytotoxic agent.
  • the cytotoxic agent here may be one which is active in killing cells once internalized.
  • the invention additionally pertains to a polypeptide chain comprising VD1-(X1) n -VD2 (X2) n -Fc, wherein VD1 is a first variable domain, VD2 is a second variable domain, Fc is one polypeptide chain of an Fc region, X1 and X2 represent an amino acid or polypeptide, and n is 0 or 1.
  • the polypeptide chain may comprise VH-CH1-flexible linker-VH-CH1-Fc region chain; VH-CH1-VH-CH1-Fc region chain; VL-CL-flexible linker-VL-CL-Fc region chain; or VL-CL-VL-CL-Fc region chain.
  • the polypeptide chain comprises VH-CH1-flexible linker-VH-CH1-dimerization domain; VH-CH1-VH-CH1-dimerization domain; VL-CL-flexible linker-VL-CL-dimerization domain; or VL-CL-VL-CL-dimerization domain.
  • the polypeptide chain may comprise VH-CH1-flexible linker-VH-CH1-hinge region; VH-CH1-VH-CH1-hinge region.
  • the invention additionally provides an antibody comprising one or more (preferably two) of such polypeptide chains.
  • the antibody preferably further comprises at least two (and preferably four) light chain or heavy chain variable domain polypeptides, e.g., where the light chain variable domain polypeptides comprise VL-CL and the heavy chain variable domain polypeptides comprise VH-CH1.
  • the invention further provides a polypeptide chain comprising three or more heavy chain or light chain variable domains, wherein each of the variable domains is able to combine with three or more light chain or heavy chain variable domain polypeptides to form three or more antigen binding sites, each directed against the same antigen.
  • the invention also provides an isolated antibody comprising the polypeptide chain.
  • the polypeptide chain comprises three or more heavy chain variable domains
  • the antibody preferably further comprises three or more light chain variable domain polypeptides which can combine with the heavy chain variable domains to form the three or more antigen binding sites. Examples of such antibodies are shown in FIG. 23D (with three antigen binding sites) and FIG. 23E (with four antigen binding sites).
  • the invention provides a polypeptide chain comprising the formula: (a) VL-CL-flexible linker-VL-CL-flexible linker-VL-CL; (b) VH-CH1-flexible linker-VH-CH1-flexible linker-VH-CH1; (c) (VL-CL) n , wherein n is three or more; or (d) (VH-CH1) n , wherein n is three or more.
  • the invention further provides: isolated nucleic acid encoding the multivalent antibody or polypeptide chain; a vector comprising nucleic acid encoding the multimeric antibody or polypeptide chain, optionally, operably linked to control sequences recognized by a host cell transformed with the vector; a host cell comprising (e.g. transformed with) nucleic acid encoding the multimeric antibody or polypeptide chain; a method for producing the multivalent antibody or polypeptide chain comprising culturing the host cell so that the nucleic acid is expressed and, optionally, recovering the multivalent antibody or polypeptide chain from the host cell culture (e.g. from the host cell culture medium).
  • a vector comprising nucleic acid encoding the multimeric antibody or polypeptide chain, optionally, operably linked to control sequences recognized by a host cell transformed with the vector
  • a host cell comprising (e.g. transformed with) nucleic acid encoding the multimeric antibody or polypeptide chain
  • a method for producing the multivalent antibody or polypeptide chain compris
  • Nucleic acids encoding (1) the heavy chain variable domains and (2) the light chain variable domains of the multivalent antibody are preferrably co-expressed by a host cell transformed with both (1) and (2).
  • Nucleic acids (1) and (2) may be present in the same, or different, vectors.
  • the invention provides a method for determining the presence of an antigen of interest comprising exposing a sample suspected of containing the antigen to the multivalent antibody and determining binding of the multivalent antibody to the sample. Both in vitro and in vivo diagnostic methods are provided.
  • the invention provides a method of treating a mammal suffering from, or predisposed to, a disease or disorder, comprising administering to the mammal a therapeutically effective amount of a multivalent antibody as disclosed herein, or of a composition comprising the multivalent antibody and a pharmaceutically acceptable carrier.
  • the disorder to be treated herein may be cancer, in which case the method may further comprise administering a therapeutically effective amount of a cytotoxic agent to the mammal.
  • the present invention further relates to a method of inducing apoptosis of a cancer cell comprising exposing the cell to a multivalent antibody as described herein, wherein the multivalent antibody binds a receptor in the Tumor Necrosis Factor (TNF) receptor superfamily.
  • the method may involve killing a B cell by exposing the B cell to a multivalent antibody that binds a B cell surface antigen.
  • the method may relate to killing a cell which expresses (or overexpresses) an ErbB receptor comprising exposing the cell to an antibody that binds the ErbB receptor.
  • FIG. 1 is a schematic representation of a native IgG and digestion thereof with (1) papain to generate two Fab fragments and an Fc region or (2) pepsin to generate a F(ab′) 2 fragment and multiple small fragments. Disulfide bonds are represented by lines between CH1 and CL domains and the two CH2 domains. V is variable domain; C is constant domain; L stands for light chain and H stands for heavy chain.
  • FIGS. 2 A-E depict the structures of the five major naturally occurring immunoglobulin isotypes; IgG (FIG. 2A), IgD (FIG. 2B), IgE (FIG. 2C), IgA dimer (FIG. 2D), and IgM pentamer (FIG. 2E).
  • FIG. 3 depicts alignments of native sequence IgG Fc regions.
  • Native sequence human IgG Fc region sequences humIgG1 (non-A and A allotypes) (SEQ ID NOs: 1 and 2, respectively), humIgG2 (SEQ ID NO:3), humIgG3 (SEQ ID NO:4) and humIgG4 (SEQ ID NO:5), are shown.
  • the human IgG1 sequence is the non-A allotype, and differences between this sequence and the A allotype (at positions 356 and 358; EU numbering system) are shown below the human IgG1 sequence.
  • murIgG1 SEQ ID NO:6
  • murIgG2A SEQ ID NO:7
  • murIgG2B SEQ ID NO:8
  • murIgG3 SEQ ID NO:9
  • FIGS. 4 A-B depict schematically tetravalent antibodies according to the present invention.
  • the four antigen binding Fabs are numbered (1 and 2 for each arm of the tetravalent antibody) and X represents a dimerization domain.
  • the dimerization domain of the tetravalent antibody is an Fc region.
  • FIG. 5 shows the construct used for expression of a tetravalent anti-HER2 antibody (OctHER2) in Example 1.
  • FIGS. 6 A-C illustrate binding of OctHER2 (FIG. 6A); bivalent IgG1 rhuMAb 4D5-8 expressed by 293 cells (FIG. 6B); and vialed HERCEPTIN® (expressed by Chinese hamster ovary (CHO) cells) (FIG. 6C) to HER2 extracellular domain (ECD) as determined using an enzyme-linked immunosorbent assay (ELISA).
  • FIGS. 6A-C illustrate binding of OctHER2 (FIG. 6A); bivalent IgG1 rhuMAb 4D5-8 expressed by 293 cells (FIG. 6B); and vialed HERCEPTIN® (expressed by Chinese hamster ovary (CHO) cells) (FIG. 6C) to HER2 extracellular domain (ECD) as determined using an enzyme-linked immunosorbent assay (ELISA).
  • ELISA enzyme-linked immunosorbent assay
  • FIG. 7 depicts ultracentrifugation analysis of binding of OctHER2 to HER2 ECD. Average molecular weights (theoretical or experimentally determined) versus molar ratio of OctHER2 to HER2 ECD are shown. Theoretical calculated average molecular weights assuming tetravalent antibody has four fully functional binding sites are shown in circles; theoretical calculated average molecular weights assuming tetravalent antibody has three fully functional binding sites are shown in squares; and triangles represent experimentally determined molecular weights.
  • FIGS. 8 A-D depict the growth inhibitory activity of HERCEPTIN® compared to OctHER2 using SKBR3 (3+ HER2 overexpressing) (FIG. 8A), MDA 361 (2+ HER2 overexpressing) (FIG. 8B), BT474 (3+ HER2 overexpressing) (FIG. 8C) and MCF7 (0+ HER2 expressing) (FIG. 8D) cell lines.
  • FIG. 9 depicts the effect of flexible linkers on the growth inhibitory activity of tetravalent anti-HER2 antibodies with respect to MDA 231 cells (1+ HER2 overexpressing) or SKBR3 cells (3+ HER2 overexpressing).
  • FIGS. 10 A-B compare the rate of OctHER2 internalization/catabolism (FIG. 10A) to that of HERCEPTIN® (FIG. 10B), in relation to both MDA 453 (2+ HER2 overexpressing) and SKBR3 (3+ HER2 overexpressing) cell lines.
  • FIGS. 11 A-I are electron microscopy photographs showing internalization of OctHER2.
  • FIGS. 11 G-I show internalization at time 0 hours (FIG. 11G) and 5 hours (FIGS. 11H and 11I).
  • FIGS. 12 A-E depict apoptosis induced by an anti-DR5 tetravalent antibody (16E2 Octopus), an anti-DR5 bivalent IgG antibody (16E2 IgG), and Apo2L/TRAIL (Apo2L) on cancer cell lines: COLO 205 (FIG. 12A), SK-MES-1 (FIG. 12B), HCT116 (FIG. 12C), and HOP 92 (FIG. 12D), compared to a non-cancer control cell line, HUMEC (FIG. 12E).
  • an anti-DR5 tetravalent antibody (16E2 Octopus
  • an anti-DR5 bivalent IgG antibody (16E2 IgG
  • Apo2L/TRAIL Apo2L/TRAIL
  • FIGS. 13 A-D are histology slides stained to detect apoptotic cells. Tumor tissues from mice treated with 16E2 Octopus or Apo2L/TRAIL were fixed in 10% formalin and then embedded into parafilm and sectioned onto slides which were then stained with hematoxylin and eosin and visualized under a 400 ⁇ magnification. The effect of 16E2 Octopus at 6 and 24 hours is shown in FIGS. 13A and B, respectively; control-treated cells are shown in FIG. 13C; and Apo2L/TRAIL-treated cells are shown in FIG. 13D.
  • FIG. 14 represents the in vivo activity of Apo2L/TRAIL (60 mg/kg, 5 ⁇ /week), 3H3 bivalent IgG (5 mg/kg given days 0, 3, 5 and 9), 16E2 bivalent IgG (16E2) (5 mg/kg given days 0, 3, 5 and 9), and 16E2 Octopus (5 mg/kg given days 0, 3, 5 and 9) with respect to COLO 205 tumors in athymic nude mice.
  • FIG. 15 represents an alamarBlue in vitro assay confirming the apoptotic activity of the material used in the mouse studies (Apo2L/TRAIL and 16E2 Octopus) as compared to an Apo2L standard positive control.
  • the anti-IgE antibody (E25) used as a negative control in the mouse studies was confirmed to have no apoptotic activity.
  • FIG. 16 represents the results of a crystal violet apoptosis assay comparing anti-DR5 3H3 Octopus to various batches of the anti-DR5 16E2 Octopus.
  • FIGS. 17 A-B reveal the results of the alamarBlue apoptosis assay with respect to Apo2L/TRAIL (WO97/25428), anti-DR5 3H3 Octopus antibody, anti-DR5 16E2 Octopus antibody, and Apo2L/TRAIL with a FLAG epitope-tag cross linked by an anti-FLAG antibody (WO97/25428), with respect to SK-MES-1 (FIG. 17A) and Jurkat (FIG. 17B) cells in the presence of 5% fetal bovine serum (FBS).
  • Apo2L/TRAIL WO97/25428
  • anti-DR5 3H3 Octopus antibody anti-DR5 16E2 Octopus antibody
  • Apo2L/TRAIL with a FLAG epitope-tag cross linked by an anti-FLAG antibody WO97/25428
  • SK-MES-1 FIG. 17A
  • Jurkat FIG. 17B
  • FIGS. 18 A-C depict dose response curves that show the effect of the anti-DR5 16E2 Octopus (upper graphs) compared to Apo2L/TRAIL (lower graphs) on the growth of leukemia, non-small cell lung cancer, colon cancer, central nervous system (CNS) cancer, melanoma, ovarian cancer, renal cancer, prostate cancer and breast cancer human tumor cell lines at 2 days.
  • Results are from the National Cancer Institute Developmental Therapeutics Program. All samples were tested at 5 concentrations, starting at 1% of the stock solution (16E2 Octopus stock 0.2 mg/ml) and 4 ⁇ 0.5 log dilutions.
  • FIGS. 19 A-C depict dose response curves that show the effect of the anti-DR5 16E2 Octopus (upper graphs) compared to Apo2L/TRAIL (lower graphs) on the growth of leukemia, non-small cell lung cancer, colon cancer, central nervous system (CNS) cancer, melanoma, ovarian cancer, renal cancer, prostate cancer and breast cancer human tumor cell lines at 6 days.
  • Results are from the National Cancer Institute Developmental Therapeutics Program. All samples were tested at 5 concentrations, starting at 1% of the stock solution (16E2 Octopus stock 0.2 mg/ml) and 4 ⁇ 0.5 log dilutions.
  • FIGS. 20 A-B present a quantitative summary of the 2 day in vitro results from the National Cancer Institute Developmental Therapeutics Program comparing the anti-DR5 16E2 Octopus (FIG. 20A) to Apo2L/TRAIL (FIG. 20B) analyzing growth inhibition (GI50), stasis (TGI), and toxicity (LC50).
  • FIGS. 21 A-B present a quantitative summary of the 6 day in vitro results from the National Cancer Institute Developmental Therapeutics Program comparing the anti-DR5 16E2 Octopus (FIG. 21A) to Apo2L/TRAIL (FIG. 21B) analyzing growth inhibition (GI50), stasis (TGI), and toxicity (LC50).
  • FIG. 22 depicts apoptosis of Wil-2 cells by the anti-CD20 antibody RITUXAN®, RITUXAN® cross-linked with anti-human IgG (RITUXAN®-IgG) and a tetravalent anti-CD20 antibody (OctCD20).
  • FIGS. 23 A-E are cartoons depicting the full-length Octopus/tetravalent antibody (FIG. 23B), the Octopus F(ab)′ 2 (FIG. 23C), POPoct-3 Fab (FIG. 23D) and POPoct-4 Fab (FIG. 23E) in comparison to the native IgG (FIG. 23A).
  • a representative coomassie stained Tris-Glycine gel of anti-CD20 (C2B8) Octopus proteins compares the sizes of the intact antibodies in non-reducing conditions (FIG. 23F), and of the heavy chains in reducing conditions, under which disulfide bonds are disrupted resulting in separation of the heavy and light chains (FIG. 23G).
  • FIG. 24 depicts the construction of the Octopus F(ab′) 2 backbone. Any VH/CH1 region can be substituted into the F(ab′) 2 backbone via the BamHI, NheI and BspEI restriction enzyme sites.
  • FIG. 25 depicts the construction of the POPoct-3 heavy chain.
  • FIG. 26 depicts the construction of the POPoct-4 heavy chain.
  • FIG. 27 depicts the activity of multivalent anti-HER2 antibodies in cytostasis assays using BT474 cells.
  • FIGS. 28 A-B depict the activity of multivalent anti-HER2 antibodies in cytostasis assays using SKBR3 cells.
  • FIGS. 29 A-B show internalization capability of multivalent anti-HER2 antibodies in SKBR3 cells (FIG. 29A) and BT474 cells (FIG. 29B).
  • FIGS. 30 A-B reveal apoptosis of COL0205 cells by multivalent anti-DR5 antibodies
  • FIGS. 31 A-B demonstrate signalling of multivalent anti-DR5 antibodies through the caspase pathway.
  • FIG. 32 compares apoptosis induced by IgG cross-linked RITUXAN®(RITUXAN-IgG) and IgG cross-linked OctCD20 (OctCD20-IgG).
  • FIG. 33 shows apoptosis of WIL2 cells by multivalent anti-CD20 antibodies, the IF5 anti-CD20 antibody (Clark et al. PNAS ( USA ) 82: 1766-1770 (1985)) and IgG cross-linked IF5 antibody (IF5 +IgG-X).
  • FIG. 34 depicts homotypic cell adhesion in WIL2S cells induced by IF5 anti-CD20 antibody, IgG cross-linked IF5 antibody and POPoct-3 CD20.
  • FIG. 35 reflects RITUXAN® or OctCD20 internalization/catabolism on DB, WIL2 and Ramos B-cell lymphoma lines.
  • the numbering of the residues in an immunoglobulin heavy chain is that of the EU index as in Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md. (1991), expressly incorporated herein by reference.
  • the “EU index as in Kabat” refers to the residue numbering of the human IgG1 EU antibody.
  • An “ErbB receptor” is a receptor protein tyrosine kinase which belongs to the ErbB receptor family and includes EGFR, HER2, ErbB3 and ErbB4 receptors as well as TEGFR (U.S. Pat. No. 5,708,156) and other members of this family to be identified in the future.
  • the ErbB receptor will generally comprise an extracellular domain, which may bind an ErbB ligand; a lipophilic transmembrane domain; a conserved intracellular tyrosine kinase domain; and a carboxyl-terminal signaling domain harboring several tyrosine residues which can be phosphorylated.
  • the ErbB receptor may be a native sequence ErbB receptor or an amino acid sequence variant thereof.
  • the ErbB receptor is native sequence human ErbB receptor.
  • ErbB ligand is meant a polypeptide which binds to and/or activates an ErbB receptor.
  • the ErbB ligand of particular interest herein is a native sequence human ErbB ligand such as Epidermal Growth Factor (EGF) (Savage et al., J. Biol. Chem.
  • TGF-alpha Tansforming Growth Factor alpha
  • amphiregulin also known as schwanoma or keratinocyte autocrine growth factor
  • betacellulin Greek et al., Science 259:1604-1607 (1993); and Sasada et al. Biochem. Biophys. Res. Commun.
  • HB-EGF heparin-binding epidermal growth factor
  • epiregulin Toyoda et al., J. Biol. Chem. 270:7495-7500 (1995); and Komurasaki et al. Oncogene 15:2841-2848 (1997)), a heregulin (see below); neuregulin-2 (NRG-2) (Carraway et al., Nature 387:512-516 (1997)); neuregulin-3 (NRG-3) (Zhang et al., Proc. Natl. Acad. Sci.
  • ErbB ligands which bind EGFR include EGF, TGF-alpha, amphiregulin, betacellulin, HB-EGF and epiregulin.
  • ErbB ligands which bind HER3 include heregulins.
  • ErbB ligands capable of binding HER4 include betacellulin, epiregulin, HB-EGF, NRG-2, NRG-3 and heregulins.
  • Heregulin when used herein refers to a polypeptide comprising an amino acid sequence encoded by the heregulin gene product as disclosed in U.S. Pat. No. 5,641,869 or Marchionni et al., Nature, 362:312-318 (1993), and biologically active variants of such polypeptides.
  • heregulins include heregulin-alpha heregulin-beta1, heregulin-beta2 and heregulin-beta3 (Holmes et al., Science, 256:1205-1210 (1992); and U.S. Pat. No. 5,641,869); neu differentiation factor (NDF) (Peles et al.
  • GGFs glial growth factors
  • SMDF motor neuron derived factor
  • An “ErbB hetero-oligomer” herein is a noncovalently associated oligomer comprising at least two different ErbB receptors. Such complexes may form when a cell expressing two or more ErbB receptors is exposed to an ErbB ligand and can be isolated by immunoprecipitation and analyzed by SDS-PAGE as described in Sliwkowski et al., J. Biol. Chem., 269(20):14661-14665 (1994), for example. Examples of such ErbB hetero-oligomers include EGFR-HER2, HER2-HER3 and HER3-HER4 complexes.
  • the ErbB hetero-oligomer may comprise two or more HER2 receptors combined with a different ErbB receptor, such as HER3, HER4 or EGFR.
  • Other proteins such as a cytokine receptor subunit (e.g. gp130), may be included in the hetero-oligomer.
  • ErbB1 refers to native sequence EGFR as disclosed, for example, in Carpenter et al. Ann. Rev. Biochem. 56:881-914 (1987), including variants thereof (e.g. a deletion mutant EGFR as in Humphrey et al. PNAS ( USA ) 87:4207-4211 (1990)).
  • erbB1 refers to the gene encoding the EGFR protein product.
  • antibodies which bind to EGFR include MAb 579 (ATCC CRL HB 8506), MAb 455 (ATCC CRL HB8507), MAb 225 (ATCC CRL 8508), MAb 528 (ATCC CRL 8509) (see, U.S. Pat. No. 4,943, 533, Mendelsohn et al.) and variants thereof, such as chimerized 225 (C225) and reshaped human 225 (H225) (see, WO 96/40210, Imclone Systems Inc.).
  • ErbB2 and HER2 are used interchangeably herein and refer to native sequence human HER2 protein described, for example, in Semba et al., PNAS ( USA ) 82:6497-6501 (1985) and Yamamoto et al., Nature 319:230-234 (1986) (Genebank accession number X03363), and variants thereof.
  • the term erbB2 refers to the gene encoding human HER2 and neu refers to the gene encoding rat p185neu.
  • Preferred HER2 is native sequence human HER2.
  • antibodies which bind HER2 include MAbs 4D5 (ATCC CRL 10463), 2C4 (ATCC HB-12697), 7F3 (ATCC HB-12216), and 7C2 (ATCC HB 12215) (see, U.S. Pat. No. 5,772,997; WO98/77797; and U.S. Pat. No. 5,840,525, expressly incorporated herein by reference).
  • Humanized anti-HER2 antibodies include huMAb4D5-1, huMAb4D5-2, huMAb4D5-3, huMAb4D5-4, huMAb4D5-5, huMAb4D5-6, huMAb4D5-7 and huMAb4D5-8 (HERCEPTIN®) as described in Table 3 of U.S. Pat. No. 5,821,337 expressly incorporated herein by reference; humanized 520C9 (WO93/21319). Human anti-HER2 antibodies are described in U.S. Pat. No. 5,772,997 issued Jun. 30, 1998 and WO 97/00271 published Jan. 3, 1997.
  • ErbB3 and “HER3” refer to the receptor polypeptide as disclosed, for example, in U.S. Pat. Nos. 5,183,884 and 5,480,968 as well as Kraus et al., PNAS ( USA ) 86:9193-9197 (1989), including variants thereof.
  • Examples of antibodies which bind HER3 are described in U.S. Pat. No. 5,968,511 (Akita and Sliwkowski), e.g. the 8B8 antibody (ATCC HB 12070) or a humanized variant thereof.
  • ErbB4 and HER4 herein refer to the receptor polypeptide as disclosed, for example, in EP Pat Appln No 599,274; Plowman et al., Proc. Natl. Acad. Sci. USA, 90:1746-1750 (1993); and Plowman et al., Nature, 366:473-475 (1993), including variants thereof such as the HER4 isoforms disclosed in WO 99/19488.
  • a “B cell surface marker” herein is an antigen expressed on the surface of a B cell which can be targeted with an antibody which binds thereto.
  • Exemplary B cell surface markers include the CD10, CD19, CD20, CD21, CD22, CD23, CD24, CD37, CD40, CD53, CD72, CD73, CD74, CDw75, CDw76, CD77, CDw78, CD79a, CD79b, CD80, CD81, CD82, CD83, CDw84, CD85 and CD86 leukocyte surface markers.
  • the B cell surface marker of particular interest is preferentially expressed on B cells compared to other non-B cell tissues of a mammal and may be expressed on both precursor B cells and mature B cells.
  • the marker is one, like CD20 or CD19, which is found on B cells throughout differentiation of the lineage from the stem cell stage up to a point just prior to terminal differentiation into plasma cells.
  • the preferred B cell surface markers herein are CD19, CD20, CD22 and CD40.
  • the “CD20” antigen is an about 35 kDa, non-glycosylated phosphoprotein found on the surface of greater than 90% of B cells from peripheral blood or lymphoid organs. CD20 is expressed during early pre-B cell development and remains until plasma cell differentiation. CD20 is present on both normal B cells as well as malignant B cells. Other names for CD20 in the literature include “B-lymphocyte-restricted antigen” and “Bp35”. The CD20 antigen is described in Clark et al. PNAS ( USA ) 82:1766 (1985), for example. Examples of antibodies which bind the CD20 antigen include: “C2B8” which is now called “rituximab” (“RITUXAN®)”) (U.S.
  • the “CD19” antigen refers to the about 90 kDa antigen identified, for example, by the HD237-CD19 or B4 antibody (Kiesel et al. Leukemia Research II, 12: 1119 (1987)). Like CD20, CD19 is found on cells throughout differentiation of the lineage from the stem cell stage up to a point just prior to terminal differentiation into plasma cells. Binding of an antibody to CD19 may cause internalization of the CD19 antigen. Examples of antibodies which bind the CD19 antigen include the anti-CD19 antibodies in Hekman et al. Cancer Immunol. Immunother. 32:364-372 (1991) and Vlasveld et al., Cancer Immunol Immunother. 40:37-47 (1995); and the B4 antibody in Kiesel et al. Leukemia Research II, 12: 1119(1987).
  • the “CD22” antigen has a molecular weight of about 140,000 kD. CD22 is expressed in the cytoplasm of early pre-B and progenitor cells, appears on the surface of only mature B cells and on the majority of non-Hodgkin's lymphoma (NHL) cells, and is then lost during terminal differentiation prior to the plasma cell stage from both the surface and cytoplasm.
  • An example of an anti-CD22 antibody is the LL2 antibody described in Juweid et al., Cancer Research 55:5899-5907 (1995), including chimeric/humanized variants thereof.
  • the “CD40” antigen is a cell surface phosphorylated glycoprotein that is expressed on a variety of cell types, including B cells, B cell malignancies, follicular dendritic cells, basal epithelial cells and carcinomas.
  • CD40 binds CD40 ligand (CD40L). Aside from being a B cell surface antigen, CD40 is also a member of the TNF receptor superfamily. Examples of antibodies that bind CD40 include those which (1) block CD40/CD40L interaction and have anti-neoplastic properties (Armitage et al., U.S. Pat. No. 5,674,492); (2) antagonize signaling through CD40 (deBoer et al., U.S. Pat. No.
  • TNF receptor superfamily refers to receptor polypeptides bound by cytokines in the TNF family. Generally, these receptors are Type I transmembrane receptors with one or more cysteine rich repeat sequences in their extracellular domain.
  • the TNF receptor superfamily may be further subdivided into (1) death receptors; (2) decoy receptors; and (3) signaling receptors that lack death domains.
  • the “death receptors” contain in their cytoplasmic or intracellular region a “death domain”, i.e., a region or sequence which acts to transduce signals in the cell which can result in apoptosis or in induction of certain genes.
  • the “decoy receptors” lack a functional death domain and are incapable of transducing signals which result in apoptosis.
  • cytokines in the TNF gene family include Tumor Necrosis Factor-alpha (TNF-alpha), Tumor Necrosis Factor-beta (TNF-beta or lymphotoxin), CD30 ligand, CD27 ligand, CD40 ligand, OX-40 ligand, 4-1 BB ligand, Apo-1 ligand (also referred to as Fas ligand or CD95 ligand), Apo-2 ligand (also referred to as TRAIL), Apo-3 ligand (also referred to as TWEAK), osteoprotegerin (OPG), APRIL, RANK ligand (also referred to as TRANCE), and TALL-1 (also referred to as BlyS, BAFF or THANK).
  • TNF-alpha Tumor Necrosis Factor-alpha
  • TNF receptor superfamily examples include: type 1 Tumor Necrosis Factor Receptor (TNFR1), type 2 Tumor Necrosis Factor Receptor (TNFR2), p75 Nerve Growth Factor receptor (NGFR), the B cell surface antigen CD40, the T cell antigen OX-40, Apo-1 receptor (also called Fas or CD95), Apo-3 receptor (also called DR3, swl-1, TRAMP and LARD), the receptor called “Transmembrane Activator and CAML-Interactor” or “TACI”, BCMA protein, DR4, DR5 (alternatively referred to as Apo-2; TRAIL-R2, TR6, Tango-63, hAPO8, TRICK2 or KILLER), DR6, DcR1 (also referred to as TRID, LIT or TRAIL-R3), DcR2 (also called TRAIL-R4 or TRUNDD), OPG, DcR3 (also called TR6 or M68), CAR1, HVE
  • Apo-2 ligand or “Apo2L” refer to the Apo2L polypeptides disclosed in WO97/25428, published Jul. 17, 1997 and expressly incorporated herein by reference. For purposes of the present application, these terms also refer to the polypeptides referred to as TRAIL disclosed in WO97/01633, published Jan. 16, 1997 and U.S. Pat. No. 5,763,223, issued Jun. 9, 1998 and expressly incorporated herein by reference.
  • An “Apo2L receptor” is a polypeptide to which Apo2L can specifically bind.
  • the term “Apo2L receptor” when used herein encompasses native sequence Apo2L receptors and variants thereof. These terms encompass Apo2L receptor from a variety of mammals, including humans.
  • the Apo2L receptor may be isolated from a variety of sources, such as from human tissue types or from another source, or prepared by recombinant or synthetic methods. Examples of “native sequence” Apo2L receptors include Apo-2 polypeptide or DR5 (WO98/51793, expressly incorporated herein by reference), native sequence DR4 as described in Pan et al.
  • anti-DR5 antibodies include 3F11.39.
  • anti-DR4 antibodies include 4E7.24.3 (ATCC HB-12454) and 4H6.17.8 (ATCC HB-12455) (see, WO 99/37684, expressly incorporated herein by reference).
  • DcR3 Native sequence “DcR3” is described in WO99/14330, expressly incorporated herein by reference. That patent publication describes the following mAbs directed against DcR3: 4C4.1.4 (ATCC HB-12573); 5C4.14.7 (ATCC HB-12574); 11C5.2.8 (ATCC HB-12572); 8D3.1.5 (ATCC HB-12571); and 4B7.1.1 (ATCC HB-12575).
  • a “native sequence” polypeptide comprises a polypeptide having the same amino acid sequence as a polypeptide derived from nature.
  • a native sequence polypeptide can have the amino acid sequence of naturally-occurring polypeptide from any mammal.
  • Such native sequence polypeptide can be isolated from nature or can be produced by recombinant or synthetic means.
  • the term “native sequence” polypeptide specifically encompasses naturally-occurring truncated or secreted forms of the polypeptide (e.g., an extracellular domain sequence), naturally-occurring variant forms (e.g., alternatively spliced forms) and naturally-occurring allelic variants of the polypeptide.
  • a polypeptide “variant” means a biologically active polypeptide having at least about 80% amino acid sequence identity with the native sequence polypeptide.
  • variants include, for instance, polypeptides wherein one or more amino acid residues are added, or deleted, at the N- or C-terminus of the polypeptide.
  • a variant will have at least about 80% amino acid sequence identity, more preferably at least about 90% amino acid sequence identity, and even more preferably at least about 95% amino acid sequence identity with the native sequence polypeptide.
  • Apoptosis refers to programmed cell death. Physiological events often indicative of the occurrence of apoptosis include: fragmentation of DNA, cell shrinkage, dilation of endoplasmic reticulum, cell fragmentation, and/or formation of membrane vesicles (called apoptotic bodies). Various methods are available for evaluating the cellular events associated with apoptosis. For example, phosphatidyl serine (PS) translocation can be measured by annexin V binding; DNA fragmentation can be evaluated through DNA laddering or propidium-iodine staining; and nuclear/chromatin condensation along with DNA fragmentation can be evaluated by any increase in hypodiploid cells.
  • PS phosphatidyl serine
  • antibody is used in the broadest sense and includes monoclonal antibodies (including full length or intact monoclonal antibodies), polyclonal antibodies, multivalent antibodies, multispecific antibodies (e.g., bispecific antibodies), and antibody fragments (see below) so long as they exhibit the desired biological activity.
  • multivalent antibody is used throughout this specification to denote an antibody comprising three or more antigen binding sites.
  • the multivalent antibody is preferably engineered to have the three or more antigen binding sites and is generally not a native sequence IgM or IgA antibody.
  • Antibody fragments comprise only a portion of an intact antibody, generally including an antigen binding site of the intact antibody and thus retaining the ability to bind antigen.
  • Examples of antibody fragments encompassed by the present definition include: (i) the Fab fragment, having VL, CL, VH and CH1 domains; (ii) the Fab′ fragment, which is a Fab fragment having one or more cysteine residues at the C-terminus of the CHI domain; (iii) the Fd fragment having VH and CH1 domains; (iv) the Fd′ fragment having VH and CH1 domains and one or more cysteine residues at the C-terminus of the CH1 domain; (v) the Fv fragment having the VL and VH domains of a single arm of an antibody; (vi) the dAb fragment (Ward et al., Nature 341, 544-546 (1989)) which consists of a VH domain; (vii) isolated CDR regions; (viii) F(ab′) 2 fragment
  • the term “monoclonal antibody” as used herein refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally occurring mutations that may be present in minor amounts. Monoclonal antibodies are highly specific, being directed against a single antigen. Furthermore, in contrast to polyclonal antibody preparations that typically include different antibodies directed against different determinants (epitopes), each monoclonal antibody is directed against a single determinant on the antigen.
  • the modifier “monoclonal” is not to be construed as requiring production of the antibody by any particular method.
  • the monoclonal antibodies to be used in accordance with the present invention may be made by the hybridoma method first described by Kohler et al., Nature 256:495 (1975), or may be made by recombinant DNA methods (see, e.g., U.S. Pat. No. 4,816,567).
  • the “monoclonal antibodies” may also be isolated from phage antibody libraries using the techniques described in Clackson et al., Nature 352:624-628 (1991) or Marks et al., J. Mol. Biol. 222:581-597 (1991), for example.
  • the monoclonal antibodies herein specifically include “chimeric” antibodies in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is identical with or homologous to corresponding sequences in antibodies derived from another species or belonging to another antibody class or subclass, as well as fragments of such antibodies, so long as they exhibit the desired biological activity (U.S. Pat. No. 4,816,567; and Morrison et al., Proc. Natl. Acad. Sci. USA 81:6851-6855 (1984)).
  • “Humanized” forms of non-human (e.g., murine) antibodies are chimeric antibodies that contain minimal sequence derived from non-human immunoglobulin.
  • humanized antibodies are human immunoglobulins (recipient antibody) in which residues from a hypervariable region of the recipient are replaced by residues from a hypervariable region of a non-human species (donor antibody) such as mouse, rat, rabbit or nonhuman primate having the desired specificity, affinity, and capacity.
  • donor antibody such as mouse, rat, rabbit or nonhuman primate having the desired specificity, affinity, and capacity.
  • framework region (FR) residues of the human immunoglobulin are replaced by corresponding non-human residues.
  • humanized antibodies may comprise residues that are not found in the recipient antibody or in the donor antibody. These modifications are made to further refine antibody performance.
  • the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the hypervariable loops correspond to those of a non-human immunoglobulin and all or substantially all of the FRs are those of a human immunoglobulin sequence.
  • the humanized antibody optionally will also comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin.
  • Fc immunoglobulin constant region
  • a “human antibody” is one which possesses an amino acid sequence which corresponds to that of an antibody produced by a human and/or has been made using any of the techniques for making human antibodies as disclosed herein. This definition of a human antibody specifically excludes a humanized antibody comprising non-human antigen-binding residues.
  • Human antibodies can be produced using various techniques known in the art. In one embodiment, the human antibody is selected from a phage library, where that phage library expresses human antibodies (Vaughan et al., Nature Biotechnology 14:309-314 (1996): Sheets et al. PNAS ( USA ) 95:6157-6162 (1998)); Hoogenboom and Winter, J. Mol.
  • Human antibodies can also be made by introducing human immunoglobulin loci into transgenic animals, e.g., mice in which the endogenous immunoglobulin genes have been partially or completely inactivated. Upon challenge, human antibody production is observed, which closely resembles that seen in humans in all respects, including gene rearrangement, assembly, and antibody repertoire. This approach is described, for example, in U.S. Pat. Nos.
  • the human antibody may be prepared via immortalization of human B lymphocytes producing an antibody directed against a target antigen (such B lymphocytes may be recovered from an individual or may have been immunized in vitro). See, e.g., Cole et al., Monoclonal Antibodies and Cancer Therapy , Alan R. Liss, p. 77 (1985); Boerner et al., J. Immunol., 147 (1):86-95 (1991); and U.S. Pat No. 5,750,373.
  • variable refers to the fact that certain portions of the variable domains differ extensively in sequence among antibodies and are used in the binding and specificity of each particular antibody for its particular antigen. However, the variability is not evenly distributed throughout the variable domains of antibodies. It is concentrated in three segments called hypervariable regions both in the light chain and the heavy chain variable domains. The more highly conserved portions of variable domains are called the framework regions (FRs).
  • the variable domains of native heavy and light chains each comprise four FRs, largely adopting a beta-sheet configuration, connected by three hypervariable regions, which form loops connecting, and in some cases forming part of, the beta-sheet structure.
  • the hypervariable regions in each chain are held together in close proximity by the FRs and, with the hypervariable regions from the other chain, contribute to the formation of the antigen-binding site of antibodies (see Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md. (1991)).
  • the constant domains are not involved directly in binding an antibody to an antigen, but exhibit various effector functions, such as participation of the antibody in antibody-dependent cell-mediated cytotoxicity (ADCC).
  • hypervariable region when used herein refers to the amino acid residues of an antibody which are responsible for antigen-binding.
  • the hypervariable region generally comprises amino acid residues from a “complementarity determining region” or “CDR” (e.g. residues 24-34 (L1), 50-56 (L2) and 89-97 (L3) in the light chain variable domain and 31-35 (H1), 50-65 (H2) and 95-102 (H3) in the heavy chain variable domain; Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md.
  • CDR complementarity determining region
  • “hypervariable loop” e.g. residues 26-32 (L1), 50-52 (L2) and 91-96 (L3) in the light chain variable domain and 26-32 (H1), 53-55 (H2) and 96-101 (H3) in the heavy chain variable domain; Chothia and Lesk J. Mol. Biol. 196:901-917 (1987)).
  • “Framework Region” or “FR” residues are those variable domain residues other than the hypervariable region residues as herein defined.
  • intact antibodies can be assigned to different “classes”. There are five major classes of intact antibodies: IgA, IgD, IgE, IgG, and IgM, and several of these may be further divided into “subclasses” (isotypes), e.g., IgG1 (including non-A and A allotypes), IgG2, IgG3, IgG4, IgA, and IgA2.
  • the heavy-chain constant domains that correspond to the different classes of antibodies are called ⁇ , ⁇ , ⁇ , ⁇ and ⁇ , respectively.
  • the subunit structures and three-dimensional configurations of different classes of immunoglobulins are well known.
  • the light chains of antibodies from any vertebrate species can be assigned to one of two clearly distinct types, called kappa ( ⁇ ) and lambda ( ⁇ ), based on the amino acid sequences of their constant domains.
  • the term “Fc region” is used to define the C-terminal region of an immunoglobulin heavy chain which may be generated by papain digestion of an intact antibody.
  • the Fc region may be a native sequence Fc region or a variant Fc region.
  • the human IgG heavy chain Fc region is usually defined to stretch from an amino acid residue at about position Cys226, or from about position Pro230, to the carboxyl-terminus of the Fc region.
  • the Fc region of an immunoglobulin generally comprises two constant domains, a CH2 domain and a CH3 domain, and optionally comprises a CH4 domain.
  • Fc region chain herein is meant one of the two polypeptide chains of an Fc region.
  • the “CH2 domain” of a human IgG Fc region usually extends from an amino acid residue at about position 231 to an amino acid residue at about position 340.
  • the CH2 domain is unique in that it is not closely paired with another domain. Rather, two N-linked branched carbohydrate chains are interposed between the two CH2 domains of an intact native IgG molecule. It has been speculated that the carbohydrate may provide a substitute for the domain-domain pairing and help stabilize the CH2 domain.
  • the CH2 domain herein may be a native sequence CH2 domain or variant CH2 domain.
  • the “CH3 domain” comprises the stretch of residues C-terminal to a CH2 domain in an Fc region (i.e. from an amino acid residue at about position 341 to an amino acid residue at about position 447 of an IgG).
  • the CH3 region herein may be a native sequence CH3 domain or a variant CH3 domain (e.g. a CH3 domain with an introduced “protroberance” in one chain thereof and a corresponding introduced “cavity” in the other chain thereof; see U.S. Pat. No. 5,821,333, expressly incorporated herein by reference).
  • Such variant CH3 domains may be used to make multispecific (e.g. bispecific) antibodies as herein described.
  • “Hinge region” is generally defined as stretching from about Glu216, or about Cys226, to about Pro230 of human IgG1 (Burton, Molec. Immunol. 22:161-206 (1985)). Hinge regions of other IgG isotypes may be aligned with the IgG1 sequence by placing the first and last cysteine residues forming inter-heavy chain S-S bonds in the same positions.
  • the hinge region herein may be a native sequence hinge region or a variant hinge region.
  • the two polypeptide chains of a variant hinge region generally retain at least one cysteine residue per polypeptide chain, so that the two polypeptide chains of the variant hinge region can form a disulfide bond between the two chains.
  • the preferred hinge region herein is a native sequence human hinge region, e.g. a native sequence human IgG1 hinge region.
  • a “functional Fc region” possesses at least one “effector function” of a native sequence Fc region.
  • effector functions include C1q binding; complement dependent cytotoxicity (CDC); Fc receptor binding; antibody-dependent cell-mediated cytotoxicity (ADCC); phagocytosis; down regulation of cell surface receptors (e.g. B cell receptor; BCR), etc.
  • Such effector functions generally require the Fc region to be combined with a binding domain (e.g. an antibody variable domain) and can be assessed using various assays known in the art for evaluating such antibody effector functions.
  • a “native sequence Fc region” comprises an amino acid sequence identical to the amino acid sequence of an Fc region found in nature.
  • FIG. 3 provides amino acid sequences of native sequence human and murine IgG Fc regions.
  • a “variant Fc region” comprises an amino acid sequence which differs from that of a native sequence Fc region by virtue of at least one amino acid modification.
  • the variant Fc region has at least one amino acid substitution compared to a native sequence Fc region or to the Fc region of a parent polypeptide, e.g. from about one to about ten amino acid substitutions, and preferably from about one to about five amino acid substitutions in a native sequence Fc region or in the Fc region of the parent polypeptide.
  • the variant Fc region herein will preferably possess at least about 80% sequence identity with a native sequence Fc region and/or with an Fc region of a parent polypeptide, and most preferably at least about 90% sequence identity therewith, more preferably at least about 95% sequence identity therewith.
  • Antibody-dependent cell-mediated cytotoxicity and “ADCC” refer to a cell-mediated reaction in which nonspecific cytotoxic cells that express Fc receptors (FcRs) (e.g. Natural Killer (NK) cells, neutrophils, and macrophages) recognize bound antibody on a target cell and subsequently cause lysis of the target cell.
  • FcRs Fc receptors
  • FcR expression on hematopoietic cells is summarized in Table 3 on page 464 of Ravetch and Kinet, Annu. Rev. Immunol 9:457-92 (1991).
  • ADCC activity of a molecule of interest may be assessed in vitro, such as that described in U.S. Pat. No. 5,500,362 or 5,821,337.
  • Useful effector cells for such assays include peripheral blood mononuclear cells (PBMC) and Natural Killer (NK) cells.
  • PBMC peripheral blood mononuclear cells
  • NK Natural Killer
  • ADCC activity of the molecule of interest may be assessed in vivo, e.g., in a animal model such as that disclosed in Clynes et al. PNAS ( USA ) 95:652-656 (1998).
  • Human effector cells are leukocytes which express one or more FcRs and perform effector functions. Preferably, the cells express at least Fc ⁇ RIll and perform ADCC effector function. Examples of human leukocytes which mediate ADCC include peripheral blood mononuclear cells (PBMC), natural killer (NK) cells, monocytes, cytotoxic T cells and neutrophils; with PBMCs and NK cells being preferred.
  • PBMC peripheral blood mononuclear cells
  • NK natural killer cells
  • monocytes monocytes
  • cytotoxic T cells and neutrophils cytotoxic T cells and neutrophils
  • the effector cells may be isolated from a native source thereof, e.g. from blood or PBMCs as described herein.
  • Fc receptor and “FcR” are used to describe a receptor that binds to the Fc region of an antibody.
  • the preferred FcR is a native sequence human FcR.
  • a preferred FcR is one which binds an IgG antibody (a gamma receptor) and includes receptors of the Fc ⁇ RI, Fc ⁇ RII, and Fc ⁇ RIII subclasses, including allelic variants and alternatively spliced forms of these receptors.
  • Fc ⁇ RII receptors include Fc ⁇ RIIA (an “activating receptor”) and Fc ⁇ RIIB (an “inhibiting receptor”), which have similar amino acid sequences that differ primarily in the cytoplasmic domains thereof.
  • Activating receptor Fc ⁇ RIIA contains an immunoreceptor tyrosine-based activation motif (ITAM) in its cytoplasmic domain.
  • Inhibiting receptor Fc ⁇ RIIB contains an immunoreceptor tyrosine-based inhibition motif (ITIM) in its cytoplasmic domain (reviewed in Da ⁇ ron, Annu. Rev. Immunol 15:203-234 (1997)).
  • FcRs are reviewed in Ravetch and Kinet, Annu. Rev. Immunol 9:457-92 (1991); Capel et al., Immunomethods 4:25-34 (1994); and de Haas et al., J. Lab. Clin. Med. 126:330-41 (1995).
  • FcR FcR
  • FcRn neonatal receptor
  • “Complement dependent cytotoxicity” and “CDC” refer to the lysing of a target in the presence of complement.
  • the complement activation pathway is initiated by the binding of the first component of the complement system (C1q) to a molecule (e.g. an antibody) complexed with a cognate antigen.
  • a CDC assay e.g. as described in Gazzano-Santoro et al., J. Immunol. Methods 202:163 (1996), may be performed.
  • An “affinity matured” antibody is one with one or more alterations in one or more CDRs thereof which result an improvement in the affinity of the antibody for antigen, compared to a parent antibody which does not possess those alteration(s).
  • Preferred affinity matured antibodies will have nanomolar or even picomolar affinities for the target antigen.
  • Affinity matured antibodies are produced by procedures known in the art. Marks et al. Bio/Technology 10:779-783 (1992) describes affinity maturation by VH and VL domain shuffling. Random mutagenesis of CDR and/or framework residues is described by: Barbas et al. Proc Nat Acad. Sci, USA 91:3809-3813 (1994); Schier et al.
  • Percent (%) amino acid sequence identity herein is defined as the percentage of amino acid residues in a candidate sequence that are identical with the amino acid residues in a selected sequence, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity, and not considering any conservative substitutions as part of the sequence identity. Alignment for purposes of determining percent amino acid sequence identity can be achieved in various ways that are within the skill in the art, for instance, using publicly available computer software such as BLAST, BLAST-2, ALIGN, ALIGN-2 or Megalign (DNASTAR) software. Those skilled in the art can determine appropriate parameters for measuring alignment, including any algorithms needed to achieve maximal alignment over the full-length of the sequences being compared.
  • % amino acid sequence identity values are obtained as described below by using the sequence comparison computer program ALIGN-2.
  • the ALIGN-2 sequence comparison computer program was authored by Genentech, Inc. has been filed with user documentation in the U.S. Copyright Office, Washington D.C., 20559, where it is registered under U.S. Copyright Registration No. TXU510087, and is publicly available through Genentech, Inc., South San Francisco, Calif.
  • the ALIGN-2 program should be compiled for use on a UNIX operating system, preferably digital UNIX V4.0D. All sequence comparison parameters are set by the ALIGN-2 program and do not vary.
  • % amino acid sequence identity of a given amino acid sequence A to, with, or against a given amino acid sequence B is calculated as follows:
  • a “polypeptide chain” is a polypeptide wherein each of the domains thereof is joined to other domain(s) by peptide bond(s), as opposed to non-covalent interactions or disulfide bonds.
  • a “flexible linker” herein refers to a peptide comprising two or more amino acid residues joined by peptide bond(s), and provides more rotational freedom for two polypeptides (such as two Fd regions) linked thereby. Such rotational freedom allows two or more antigen binding sites joined by the flexible linker to each access target antigen(s) more efficiently.
  • suitable flexible linker peptide sequences include gly-ser, gly-ser-gly-ser (SEQ ID NO:10), ala-ser, and gly-gly-gly-ser (SEQ ID NO:11).
  • the flexible linker comprises 2 to about 10 amino acid residues, and most preferably four or less residues.
  • a “dimerization domain” is formed by the association of at least two amino acid residues (generally cysteine residues) or of at least two peptides or polypeptides (which may have the same, or different, amino acid sequences).
  • the peptides or polypeptides may interact with each other through covalent and/or non-covalent association(s).
  • dimerization domains herein include an Fc region; a hinge region; a CH3 domain; a CH4 domain; a CH1-CL pair; an “interface” with an engineered “knob” and/or “protruberance” as described in U.S. Pat. No. 5,821,333, expressly incorporated herein by reference; a leucine zipper (e.g.
  • a jun/fos leucine zipper see Kostelney et al., J. Immunol., 148: 1547-1553 (1992); or a yeast GCN4 leucine zipper); an isoleucine zipper; a receptor dimer pair (e.g., interleukin-8 receptor (IL-8R); and integrin heterodimers such as LFA-1 and GPIIIb/IIIa), or the dimerization region(s) thereof; dimeric ligand polypeptides (e.g. nerve growth factor (NGF), neurotrophin-3 (NT-3), interleukin-8 (IL-8), vascular endothelial growth factor (VEGF), and brain-derived neurotrophic factor (BDNF); see Arakawa et al. J.
  • NGF nerve growth factor
  • NT-3 neurotrophin-3
  • IL-8 interleukin-8
  • VEGF vascular endothelial growth factor
  • BDNF brain-derived neurotrophic factor
  • dimerization region(s) thereof a pair of cysteine residues able to form a disulfide bond; a pair of peptides or polypeptides, each comprising at least one cysteine residue (e.g. from about one, two or three to about ten cysteine residues) such that disulfide bond(s) can form between the peptides or polypeptides (hereinafter “a synthetic hinge”); and antibody variable domains.
  • the most preferred dimerization domain herein is an Fc region or a hinge region.
  • “Naturally occurring amino acid residues” may be selected from the group consisting of: alanine (Ala); arginine (Arg); asparagine (Asn); aspartic acid (Asp); cysteine (Cys); glutamine (Gin); glutamic acid (Glu); glycine (Gly); histidine (His); isoleucine (Ile): leucine (Leu); lysine (Lys); methionine (Met); phenylalanine (Phe); proline (Pro); serine (Ser); threonine (Thr); tryptophan (Trp); tyrosine (Tyr); and valine (Val).
  • non-naturally occurring amino acid residue refers to a residue, other than those naturally occurring amino acid residues listed above, which is able to covalently bind adjacent amino acid residues(s) in a polypeptide chain.
  • non-naturally occurring amino acid residues include norleucine, ornithine, norvaline, homoserine and other amino acid residue analogues such as those described in ElIman et al. Meth. Enzym. 202:301-336 (1991).
  • the procedures of Noren et al. Science 244:182 (1989) and Ellman et al., supra can be used. Briefly, these procedures involve chemically activating a suppressor tRNA with a non-naturally occurring amino acid residue followed by in vitro transcription and translation of the RNA.
  • An “isolated” polypeptide is one that has been identified and separated and/or recovered from a component of its natural environment. Contaminant components of its natural environment are materials that would interfere with diagnostic or therapeutic uses for the polypeptide, and may include enzymes, hormones, and other proteinaceous or nonproteinaceous solutes.
  • the polypeptide will be purified (1) to greater than 95% by weight of polypeptide as determined by the Lowry method, and most preferably more than 99% by weight, (2) to a degree sufficient to obtain at least 15 residues of N-terminal or internal amino acid sequence by use of a spinning cup sequenator, or (3) to homogeneity by SDS-PAGE under reducing or nonreducing conditions using Coomassie blue or, preferably, silver stain.
  • Isolated polypeptide includes the polypeptide in situ within recombinant cells since at least one component of the polypeptide's natural environment will not be present. Ordinarily, however, isolated polypeptide will be prepared by at least one purification step.
  • a “functional antigen binding site” of an antibody is one which is capable of binding a target antigen.
  • the antigen binding affinity of the antigen binding site is not necessarily as strong as the parent antibody from which the antigen binding site is derived, but the ability to bind antigen must be measurable using any one of a variety of methods known for evaluating antibody binding to an antigen.
  • the antigen binding affinity of each of the antigen binding sites of a multivalent antibody herein need not be quantitatively the same.
  • the number of functional antigen binding sites can be evaluated using ultracentrifugation analysis as described in Example 2 below.
  • ligand activation of a receptor is meant signal transduction (e.g. for a tyrosine kinase receptor, that caused by an intracellular kinase domain of a tyrosine kinase receptor phosphorylating tyrosine residues in the receptor or a substrate polypeptide) mediated by ligand binding to the receptor (or a receptor complex comprising the receptor of interest).
  • an ErbB receptor In the case of an ErbB receptor, generally, this will involve binding of an ErbB ligand to an ErbB hetero-oligomer which activates a kinase domain of one or more of the ErbB receptors in the hetero-oligomer and thereby results in phosphorylation of tyrosine residues in one or more of the ErbB receptors and/or phosphorylation of tyrosine residues in additional substrate polypeptides(s).
  • An antibody which “blocks” ligand activation of an receptor is one which reduces or prevents such activation as hereinabove defined. Such blocking can occur by any means, e.g. by interfering with: ligand binding to the receptor, receptor complex formation, tyrosine kinase activity of a tyrosine kinase receptor in a receptor complex and/or phosphorylation of tyrosine kinase residue(s) in or by the receptor.
  • antibodies which block ligand activation of an ErbB receptor include monoclonal antibodies 2C4 and 7F3 (which block HRG activation of HER2/HER3 and HER2/HER4 heterooligomers; and EGF, TGF-beta or amphiregulin activation of an EGFR/HER2 hetero-oligomer); and L26, L96 and L288 antibodies (Klapper et al. Oncogene 14:2099-2109 (1997)), which block EGF and NDF binding to T47D cells which express EGFR, HER2, HER3 and HER4.
  • An antibody having a “biological characteristic” of a designated antibody is one which possesses one or more of the biological characteristics of that antibody which distinguish it from other antibodies that bind to the same antigen.
  • a “growth inhibitory agent” when used herein refers to a compound or composition which inhibits growth of a cell in vitro and/or in vivo.
  • the growth inhibitory agent may be one which significantly reduces the percentage of cells in S phase.
  • growth inhibitory agents include agents that block cell cycle progression (at a place other than S phase), such as agents that induce G1 arrest and M-phase arrest.
  • Classical M-phase blockers include the vincas (vincristine and vinblastine), TAXOL®, and topo 11 inhibitors such as doxorubicin, epirubicin, daunorubicin, etoposide, and bleomycin.
  • DNA alkylating agents such as tamoxifen, prednisone, dacarbazine, mechlorethamine, cisplatin, methotrexate, 5-fluorouracil, and ara-C. Further information can be found in The Molecular Basis of Cancer , Mendelsohn and Israel, eds., Chapter 1, entitled “Cell cycle regulation, oncogenes, and antineoplastic drugs” by Murakami et al. (WB Saunders: Philadelphia, 1995), especially p. 13.
  • growth inhibitory anti-HER2 antibodies are those which bind to HER2 and inhibit the growth of cancer cells overexpressing HER2.
  • Preferred growth inhibitory anti-HER2 antibodies inhibit growth of SKBR3 breast tumor cells in cell culture by greater than 20%, and preferably greater than 50% (e.g. from about 50% to about 100%) at an antibody concentration of about 0.5 to 30 ⁇ g/ml, where the growth inhibition is determined six days after exposure of the SKBR3 cells to the antibody (see U.S. Pat. No. 5,677,171 issued Oct. 14, 1997).
  • An antibody which “induces cell death” is one which causes a viable cell to become nonviable.
  • the cell is generally one which expresses the antigen to which the antibody binds, especially where the cell overexpresses the antigen.
  • the cell is a cancer cell, e.g. a breast, ovarian, stomach, endometrial, salivary gland, lung, kidney, colon, thyroid, pancreatic or bladder cell.
  • the cell may be a SKBR3, BT474, Calu 3, MDA-MB-453, MDA-MB-361 or SKOV3 cell.
  • Cell death in vitro may be determined in the absence of complement and immune effector cells to distinguish cell death induced by antibody dependent cell-mediated cytotoxicity (ADCC) or complement dependent cytotoxicity (CDC).
  • ADCC antibody dependent cell-mediated cytotoxicity
  • CDC complement dependent cytotoxicity
  • the assay for cell death may be performed using heat inactivated serum (ie. in the absence of complement) and in the absence of immune effector cells.
  • heat inactivated serum ie. in the absence of complement
  • immune effector cells ie. in the absence of immune effector cells.
  • loss of membrane integrity as evaluated by uptake of propidium iodide (PI), trypan blue (see Moore et al. Cytotechnology 17:1-11 (1995)) or 7AAD can be assessed relative to untreated cells.
  • PI propidium iodide
  • trypan blue see Moore et al. Cytotechnology 17:1-11 (1995)
  • 7AAD can be assessed relative to untreated cells.
  • An antibody which “induces apoptosis” is one which induces programmed cell death as determined by binding of annexin V, fragmentation of DNA, cell shrinkage, dilation of endoplasmic reticulum, cell fragmentation, and/or formation of membrane vesicles (called apoptotic bodies).
  • the cell is one which expresses the antigen to which the antibody binds and may be one which overexpresses the antigen.
  • the cell may be a tumor cell, e.g. a breast, ovarian, stomach, endometrial, salivary gland, lung, kidney, colon, thyroid, pancreatic or bladder cell.
  • the cell may be a SKBR3, BT474, Calu 3 cell, MDA-MB-453, MDA-MB-361 or SKOV3 cell.
  • Various methods are available for evaluating the cellular events associated with apoptosis. For example, phosphatidyl serine (PS) translocation can be measured by annexin binding; DNA fragmentation can be evaluated through DNA laddering as disclosed in the example herein; and nuclear/chromatin condensation along with DNA fragmentation can be evaluated by any increase in hypodiploid cells.
  • PS phosphatidyl serine
  • the antibody which induces apoptosis is one which results in about 2 to 50 fold, preferably about 5 to 50 fold, and most preferably about 10 to 50 fold, induction of annexin binding relative to untreated cell in an annexin binding assay using cells expressing the antigen to which the antibody binds.
  • antibodies which induce apoptosis include the anti-HER2 monoclonal antibodies 7F3 (ATCC HB-12216), and 7C2 (ATCC HB 12215), including humanized and/or affinity matured variants thereof; the anti-DR5 antibodies 3F11.39.7 (ATCC HB-12456); 3H3.14.5 (ATCC HB-12534); 3D5.1.10 (ATCC HB-12536); and 3H3.14.5 (ATCC HB-12534), including humanized and/or affinity matured variants thereof; the human anti-DR5 receptor antibodies 16E2 and 20E6, including affinity matured variants thereof (WO98/51793, expressly incorporated herein by reference); the anti-DR4 antibodies 4E7.24.3 (ATCC HB-12454); 4H6.17.8 (ATCC HB-12455); 1H5.25.9 (ATCC HB-12695); 4G7.18.8 (ATCC PTA-99); and 5G11.17.1 (ATCC HB-12694), including humanized
  • an “agonist antibody” is an antibody which binds to and activates a receptor. Generally, the receptor activation capability of the agonist antibody will be at least qualitatively similar (and may be essentially quantitatively similar) to a native agonist ligand of the receptor.
  • An example of an agonist antibody is one which binds to a receptor in the TNF receptor superfamily and induces apoptosis of cells expressing the TNF receptor. Assays for determining induction of apoptosis are described in WO98/51793 and WO99/37684, both of which are expressly incorporated herein by reference.
  • a “disorder” is any condition that would benefit from treatment with the antibody. This includes chronic and acute disorders or diseases including those pathological conditions which predispose the mammal to the disorder in question.
  • disorders to be treated herein include benign and malignant tumors; leukemias and lymphoid malignancies; neuronal, glial, astrocytal, hypothalamic and other glandular, macrophagal, epithelial, stromal and blastocoelic disorders; and inflammatory, angiogenic and immunologic disorders.
  • the term “therapeutically effective amount” refers to an amount of a drug effective to treat a disease or disorder in a mammal.
  • the therapeutically effective amount of the drug may reduce the number of cancer cells; reduce the tumor size; inhibit (i.e., slow to some extent and preferably stop) cancer cell infiltration into peripheral organs; inhibit (i.e., slow to some extent and preferably stop) tumor metastasis; inhibit, to some extent, tumor growth; and/or relieve to some extent one or more of the symptoms associated with the disorder.
  • the drug may prevent growth and/or kill existing cancer cells, it may be cytostatic and/or cytotoxic.
  • efficacy in vivo can, for example, be measured by assessing the time to disease progression (TTP) and/or determining the response rates (RR).
  • Treatment refers to both therapeutic treatment and prophylactic or preventative measures. Those in need of treatment include those already with the disorder as well as those in which the disorder is to be prevented.
  • cancer and “cancerous” refer to or describe the physiological condition in mammals that is typically characterized by unregulated cell growth.
  • Examples of cancer include but are not limited to, carcinoma, lymphoma, blastoma, sarcoma, and leukemia.
  • cancers include squamous cell cancer, small-cell lung cancer, non-small cell lung cancer, adenocarcinoma of the lung, squamous carcinoma of the lung, cancer of the peritoneum, hepatocellular cancer, gastrointestinal cancer, pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, hepatoma, breast cancer, colon cancer, colorectal cancer, endometrial or uterine carcinoma, salivary gland carcinoma, kidney cancer, liver cancer, prostate cancer, vulval cancer, thyroid cancer, hepatic carcinoma and various types of head and neck cancer.
  • autoimmune disease herein is a non-malignant disease or disorder arising from and directed against an individual's own tissues.
  • autoimmune diseases or disorders include, but are not limited to, inflammatory responses such as inflammatory skin diseases including psoriasis and dermatitis (e.g.
  • atopic dermatitis atopic dermatitis
  • systemic scleroderma and sclerosis responses associated with inflammatory bowel disease (such as Crohn's disease and ulcerative colitis); respiratory distress syndrome (including adult respiratory distress syndrome; ARDS); dermatitis; meningitis; encephalitis; uveitis; colitis; glomerulonephritis; allergic conditions such as eczema and asthma and other conditions involving infiltration of T cells and chronic inflammatory responses; atherosclerosis; leukocyte adhesion deficiency; rheumatoid arthritis; systemic lupus erythematosus (SLE); diabetes mellitus (e.g.
  • Type I diabetes mellitus or insulin dependent diabetes mellitis multiple sclerosis; Reynaud's syndrome; autoimmune thyroiditis; allergic encephalomyelitis; Sjorgen's syndrome; juvenile onset diabetes; and immune responses associated with acute and delayed hypersensitivity mediated by cytokines and T-lymphocytes typically found in tuberculosis, sarcoidosis, polymyositis, granulomatosis and vasculitis; pernicious anemia (Addison's disease); diseases involving leukocyte diapedesis; central nervous system (CNS) inflammatory disorder; multiple organ injury syndrome; hemolytic anemia (including, but not limited to cryoglobinemia or Coombs positive anemia); myasthenia gravis; antigen-antibody complex mediated diseases; anti-glomerular basement membrane disease; antiphospholipid syndrome; allergic neuritis; Graves' disease; Lambert-Eaton myasthenic syndrome; pemphigoid bullous; pemphigus; autoimmune polyen
  • foreign antigen is meant a molecule or molecules which is/are not endogenous or native to a mammal which is exposed to it.
  • the foreign antigen may elicit an immune response, e.g. a humoral and/or T cell mediated response in the mammal. Generally, the foreign antigen will provoke the production of antibodies thereagainst.
  • immunogenic therapeutic agents e.g. proteins such as antibodies, particularly antibodies comprising non-human amino acid residues (e.g.
  • rodent, chimeric/humanized, and primatized antibodies include toxins (optionally conjugated to a targeting molecule such as an antibody, wherein the targeting molecule may also be immunogenic); gene therapy viral vectors, such as retroviruses and adenoviruses; grafts; infectious agents (e.g. bacteria and virus); alloantigens (i.e. an antigen that occurs in some, but not in other members of the same species) such as differences in blood types, human lymphocyte antigens (HLA), platelet antigens, antigens expressed on transplanted organs, blood components, pregnancy (Rh), and hemophilic factors (e.g. Factor VIII and Factor IX).
  • HLA human lymphocyte antigens
  • platelet antigens antigens expressed on transplanted organs
  • blood components blood components
  • pregnancy (Rh) pregnancy
  • hemophilic factors e.g. Factor VIII and Factor IX
  • blocking an immune response” to a foreign antigen is meant reducing or preventing at least one immune-mediated response resulting from exposure to a foreign antigen.
  • a foreign antigen For example, one may dampen a humoral response to the foreign antigen, i.e., by preventing or reducing the production of antibodies directed against the antigen in the mammal.
  • graft refers to biological material derived from a donor for transplantation into a recipient. Grafts include such diverse material as, for example, isolated cells such as islet cells; tissue such as the amniotic membrane of a newborn, bone marrow, hematopoietic precursor cells, and ocular tissue, such as corneal tissue; and organs such as skin, heart, liver, spleen, pancreas, thyroid lobe, lung, kidney, tubular organs (e.g., intestine, blood vessels, or esophagus), etc. The tubular organs can be used to replace damaged portions of esophagus, blood vessels, or bile duct.
  • isolated cells such as islet cells
  • tissue such as the amniotic membrane of a newborn, bone marrow, hematopoietic precursor cells, and ocular tissue, such as corneal tissue
  • organs such as skin, heart, liver, spleen, pancreas, thyroid lobe, lung, kidney, tubular
  • the skin grafts can be used not only for burns, but also as a dressing to damaged intestine or to close certain defects such as diaphragmatic hernia.
  • the graft is derived from any mammalian source, including human, whether from cadavers or living donors.
  • the graft is bone marrow or an organ such as heart and the donor of the graft and the host are matched for HLA class 11 antigens.
  • mammalian host refers to any compatible transplant recipient.
  • compatible is meant a mammalian host that will accept the donated graft.
  • the host is human. If both the donor of the graft and the host are human, they are preferably matched for HLA class II antigens so as to improve histocompatibility.
  • the term “donor” as used herein refers to the mammalian species, dead or alive, from which the graft is derived.
  • the donor is human.
  • Human donors are preferably volunteer blood-related donors that are normal on physical examination and of the same major ABO blood group, because crossing major blood group barriers possibly prejudices survival of the allograft. It is, however, possible to transplant, for example, a kidney of a type O donor into an A, B or AB recipient.
  • transplant refers to the insertion of a graft into a host, whether the transplantation is syngeneic (where the donor and recipient are genetically identical), allogeneic (where the donor and recipient are of different genetic origins but of the same species), or xenogeneic (where the donor and recipient are from different species).
  • the host is human and the graft is an isograft, derived from a human of the same or different genetic origins.
  • the graft is derived from a species different from that into which it is transplanted, such as a baboon heart transplanted into a human recipient host, and including animals from phylogenically widely separated species, for example, a pig heart valve, or animal beta islet cells or neuronal cells transplanted into a human host.
  • a species different from that into which it is transplanted such as a baboon heart transplanted into a human recipient host, and including animals from phylogenically widely separated species, for example, a pig heart valve, or animal beta islet cells or neuronal cells transplanted into a human host.
  • the expression “desensitizing a mammal awaiting transplantation” refers to reducing or abolishing allergic sensitivity or reactivity to a transplant, prior to administration of the transplant to the mammal. This may be achieved by any mechanism, such as a reduction in anti-donor antibodies in the desensitized mammal, e.g. where such anti-donor antibodies are directed against human lymphocyte antigen (HLA).
  • HLA human lymphocyte antigen
  • cytotoxic agent refers to a substance that inhibits or prevents the function of cells and/or causes destruction of cells.
  • the term is intended to include radioactive isotopes (e.g. At 211 , I 131 , I 125 , Y 90 , Re 186 , Re 188 , Sm 153 , Bi 212 , P 32 and radioactive isotopes of Lu), chemotherapeutic agents, and toxins such as small molecule toxins or enzymatically active toxins of bacterial, fungal, plant or animal origin, including fragments and/or variants thereof.
  • radioactive isotopes e.g. At 211 , I 131 , I 125 , Y 90 , Re 186 , Re 188 , Sm 153 , Bi 212 , P 32 and radioactive isotopes of Lu
  • chemotherapeutic agents e.g. At 211 , I 131 , I 125 , Y 90 , Re 186
  • a “chemotherapeutic agent” is a chemical compound useful in the treatment of cancer.
  • examples of chemotherapeutic agents include alkylating agents such as thiotepa and cyclosphosphamide (CYTOXAN TM); alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines including altretamine, triethylenemelamine, trietylenephosphoramide, triethylenethiophosphaoramide and trimethylolomelamine; acetogenins (especially bullatacin and bullatacinone); a camptothecin (including the synthetic analogue topotecan); bryostatin; callystatin; CC-1065 (including its adozelesin, carzelesin and bizelesin synthetic analogues);
  • calicheamicin especially calicheamicin ⁇ 1 I and calicheamicin ⁇ 1 I , see, e.g., Agnew Chem Intl. Ed. Engl. 33:183-186 (1994); dynemicin, including dynemicin A; an esperamicin; as well as neocarzinostatin chromophore and related chromoprotein enediyne antiobiotic chromomophores), aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, carabicin, carminomycin, carzinophilin, chromomycins, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-L-norleucine, doxorubicin (including morpholino-doxorubicin, cyanomorpholino-doxorubicin, 2-
  • paclitaxel TAXOL®, Bristol-Myers Squibb Oncology, Princeton, N.J.
  • doxetaxel TAXOTERE®, Rhône-Poulenc Rorer, Antony, France
  • chlorambucil gemcitabine
  • 6-thioguanine mercaptopurine
  • methotrexate platinum analogs such as cisplatin and carboplatin; vinblastine; platinum; etoposide (VP-16); ifosfamide; mitomycin C; mitoxantrone; vincristine; vinorelbine; navelbine; novantrone; teniposide; daunomycin; aminopterin; xeloda; ibandronate; CPT-11; topoisomerase inhibitor RFS 2000; difluoromethylornithine (DMFO); retinoic acid; capecitabine; and pharmaceutically acceptable salts, acids or derivatives of any of the above.
  • DMFO diflu
  • anti-hormonal agents that act to regulate or inhibit hormone action on tumors
  • anti-estrogens including for example tamoxifen, raloxifene, aromatase inhibiting 4(5)-imidazoles, 4-hydroxytamoxifen, trioxifene, keoxifene, LY117018, onapristone, and toremifene (Fareston); and anti-androgens such as flutamide, nilutamide, bicalutamide, leuprolide, and goserelin; and pharmaceutically acceptable salts, acids or derivatives of any of the above.
  • cytokine is a generic term for proteins released by one cell population which act on another cell as intercellular mediators.
  • cytokines are lymphokines, monokines, and traditional polypeptide hormones. Included among the cytokines are growth hormone such as human growth hormone, N-methionyl human growth hormone, and bovine growth hormone; parathyroid hormone; thyroxine; insulin; proinsulin; relaxin; prorelaxin; glycoprotein hormones such as follicle stimulating hormone (FSH), thyroid stimulating hormone (TSH), and luteinizing hormone (LH); hepatic growth factor; fibroblast growth factor; prolactin; placental lactogen; tumor necrosis factor-alpha and -beta; mullerian-inhibiting substance; mouse gonadotropin-associated peptide; inhibin; activin; vascular endothelial growth factor; integrin; thrombopoietin (TPO); nerve growth factors such as NGF-al
  • prodrug refers to a precursor or derivative form of a pharmaceutically active substance that is less cytotoxic to tumor cells compared to the parent drug and is capable of being enzymatically activated or converted into the more active parent form. See, e.g., Wilman, “Prodrugs in Cancer Chemotherapy” Biochemical Society Transactions, 14, pp. 375-382, 615th Meeting Harbor (1986) and Stella et al., “Prodrugs: A Chemical Approach to Targeted Drug Delivery,” Directed Drug Delivery , Borchardt et al., (ed.), pp. 247-267, Humana Press (1985).
  • the prodrugs of this invention include, but are not limited to, phosphate-containing prodrugs, thiophosphate-containing prodrugs, sulfate-containing prodrugs, peptide-containing prodrugs, D-amino acid-modified prodrugs, glycosylated prodrugs, beta-lactam-containing prodrugs, optionally substituted phenoxyacetamide-containing prodrugs or optionally substituted phenylacetamide-containing prodrugs, 5-fluorocytosine and other 5-fluorouridine prodrugs which can be converted into the more active cytotoxic free drug.
  • cytotoxic drugs that can be derivatized into a prodrug form for use in this invention include, but are not limited to, those chemotherapeutic agents described above.
  • angiogenic factor is a growth factor which stimulates the development of blood vessels.
  • the preferred angiogenic factor herein is Vascular Endothelial Growth Factor (VEGF).
  • label when used herein refers to a detectable compound or composition which is conjugated directly or indirectly to the polypeptide.
  • the label may be itself be detectable (e.g., radioisotope labels or fluorescent labels) or, in the case of an enzymatic label, may catalyze chemical alteration of a substrate compound or composition which is detectable.
  • An “isolated” nucleic acid molecule is a nucleic acid molecule that is identified and separated from at least one contaminant nucleic acid molecule with which it is ordinarily associated in the natural source of the polypeptide nucleic acid.
  • An isolated nucleic acid molecule is other than in the form or setting in which it is found in nature. Isolated nucleic acid molecules therefore are distinguished from the nucleic acid molecule as it exists in natural cells.
  • an isolated nucleic acid molecule includes a nucleic acid molecule contained in cells that ordinarily express the polypeptide where, for example, the nucleic acid molecule is in a chromosomal location different from that of natural cells.
  • control sequences refers to DNA sequences necessary for the expression of an operably linked coding sequence in a particular host organism.
  • the control sequences that are suitable for prokaryotes include a promoter, optionally an operator sequence, and a ribosome binding site.
  • Eukaryotic cells are known to utilize promoters, polyadenylation signals, and enhancers.
  • Nucleic acid is “operably linked” when it is placed into a functional relationship with another nucleic acid sequence.
  • DNA for a presequence or secretory leader is operably linked to DNA for a polypeptide if it is expressed as a preprotein that participates in the secretion of the polypeptide;
  • a promoter or enhancer is operably linked to a coding sequence if it affects the transcription of the sequence; or
  • a ribosome binding site is operably linked to a coding sequence if it is positioned so as to facilitate translation.
  • “operably linked” means that the DNA sequences being linked are contiguous, and, in the case of a secretory leader, contiguous and in reading phase. However, enhancers do not have to be contiguous. Linking is accomplished by ligation at convenient restriction sites. If such sites do not exist, the synthetic oligonucleotide adaptors or linkers are used in accordance with conventional practice.
  • the expressions “cell,” “cell line,” and “cell culture” are used interchangeably and all such designations include progeny.
  • the words “transformants” and “transformed cells” include the primary subject cell and cultures derived therefrom without regard for the number of transfers. It is also understood that all progeny may not be precisely identical in DNA content, due to deliberate or inadvertent mutations. Mutant progeny that have the same function or biological activity as screened for in the originally transformed cell are included. Where distinct designations are intended, it will be clear from the context.
  • the invention herein relates to a method for making a multivalent antibody.
  • Various techniques for generating the “parent” or “starting” antibody from which the variable domain(s) of the multivalent antibody may be derived will be described later in this application.
  • the multivalent antibody of particular interest herein is one which comprises at least three (and preferably four, or more, e.g. four or five to about eight) antigen binding sites. Generally, all of the antigen binding sites are “functional” as defined hereinabove.
  • the multivalent antibody does not exist in nature and is not a native sequence IgM or IgA antibody.
  • the multivalent antibody herein is preferably not produced in vitro by chemically cross-linking a pair antibodies (e.g. as in Ghetie et al. (1997), supra or Wolff et al. (1993), supra).
  • the present application also provides multivalent antibodies which do not require introduced cysteine residue(s) in a parent antibody in order to make the multivalent antibody via disulfide bond(s) between a pair of Fc regions (e.g. as in Shopes et al. (1992), supra or Caron et al. (1992), supra).
  • the multivalent antibody comprises a first polypeptide chain comprising at least two heavy chain (or light chain) variable domains and a second polypeptide chain comprising at least two heavy chain (or light chain) variable domains.
  • the first polypeptide chain comprises two heavy chain variable domains and the second polypeptide chain also comprises two heavy chain variable domains, which can be combined with corresponding light chain variable domains (at least two for each polypeptide chain) to generate four (or more) antigen binding sites.
  • the multivalent antibody comprises a dimerization domain which combines (1) two (or more) antigen binding sites with (2) one, two (or more) antigen binding sites.
  • dimerization domains are contemplated herein, but the preferred dimerization domain is an Fc region or a hinge region.
  • the multivalent antibody comprises an Fc region (e.g. a native sequence or variant Fc region)
  • the Fc region is preferably “functional” as defined hereinabove and thus is capable of performing one or more antibody effector functions, such as ADCC or CDC.
  • the multivalent antibody has only one Fc region or lacks an Fc region.
  • the multivalent antibody comprises an Fc region
  • the three or more antigen binding sites are provided amino terminal to the Fc region (rather than at the carboxy terminus of the Fc region as in Coloma and Morrison, (1997) supra).
  • This may be achieved by providing a first polypeptide chain represented by the formula VD1-X1-VD2-X2-Fc, wherein (1) VD1 is a first heavy or light chain variable domain (preferably a heavy chain variable domain), (2) VD2 is a second heavy or light chain variable domain (preferably a heavy chain variable domain), (3) Fc comprises one chain of an Fc region, and (4) X1 and X2 represent an optional intervening amino acid or polypeptide.
  • X1 and X2 comprise, or consist of, a CH1 domain (where VD1 or VD2 is a heavy chain variable domain) or a CL domain (where VD1 or VD2 is a light chain variable domain).
  • X1 further comprises a flexible linker which is generally C-terminal to VD1 (or C-terminal to CH1 or CL, if present).
  • the flexible linker may comprise a peptide such as gly-ser, gly-ser-gly-ser (SEQ ID NO:10), ala-ser or gly-gly-gly-ser (SEQ ID NO:11).
  • the multivalent antibody of particular interest herein comprises three or more (e.g. four or five to about eight) Fab polypeptides, each capable of binding antigen.
  • the Fab fragments are preferably provided amino terminal to the Fc region (where the multivalent antibody has an Fc region).
  • two or more Fd fragments may be fused to the amino terminus of one chain of an Fc region.
  • the polypeptide chain thus engineered may be combined with (1) another polypeptide chain formed by two or more Fd fragments fused to the amino terminus of the other chain of the Fc region, as well as (2) complementary VL domains (e.g. four or more VL domains which each, optionally, are fused to a CL domain).
  • the antibody comprises a flexible linker between the two or more Fd fragments.
  • the multivalent antibody may, for example, comprise a pair of polypeptide chains with the formula (1) VH-CH1-flexible linker-VH-CH1-Fc chain, or (2) VH-CH1-VH-CH1-Fc chain (i.e. where there is no flexible linker between the two Fd fragments).
  • the three or more functional antigen binding sites of the multivalent antibody herein are each preferably formed by a heavy and light chain variable domain.
  • two or more heavy chain variable domains are fused together (optionally with intervening amino acid residue(s) as noted above)
  • two or more complementary light chain variable domain-containing polypeptides are combined with the heavy chain variable domains (for instance by co-expressing the fusion protein and the light chain variable domain polypeptide(s) in the same host cell).
  • the antibody comprises four, or five, or more (e.g. up to about eight) light chain variable domain polypeptides, which each, optionally, comprise a CL domain.
  • the antibody with three or more (e.g. three to about ten, but preferably three or four) antigen binding sites may comprise a polypeptide chain comprising three or more (e.g. three to about ten, but preferably three or four) heavy chain or light chain variable domains, wherein each of the variable domains is combined with, or associated with, three or more (e.g. three to about ten, but preferably three or four) light chain or heavy chain variable domain polypeptides in such a way as to form the antigen binding sites.
  • the polypeptide chain comprises three or more heavy chain variable domains, it is combined or associated with three or more corresponding light chain variable domain polypeptides (e.g. with VL-CL polypeptides).
  • the polypeptide chain comprises three or more light chain variable domains
  • it is combined or associated with three or more corresponding heavy chain variable domain polypeptides (e.g. with VH-CH1 polypeptides).
  • each of the three or more antigen binding sites is directed against the same antigen.
  • antigens bound by such antibodies include (1) a receptor in the Tumor Necrosis Factor (TNF) receptor superfamily (such receptors may be ‘trimeric receptors’, hence the antibody need only include only three antigen binding sites as desired) such as DR4 and DR5; (2) a B cell surface antigen such as CD20; (3) an ErbB receptor exemplified by the HER2 receptor; or (4) a cell surface protein expressed by tumor cells.
  • TNF Tumor Necrosis Factor
  • the polypeptide chain may comprise three (or four) heavy chain variable domains which are able to combine with three (or four) light chain variable domain polypeptides (preferably VL-CL polypeptides) to generate three (or four) antigen binding sites directed against the same antigen.
  • VL-CL polypeptides preferably VL-CL polypeptides
  • the multivalent antibody may also comprise a polypeptide chain comprising the formula: (a) VL-CL-flexible linker-VL-CL-flexible linker-VL-CL;
  • the polypeptide may comprise three to about eight VL-CL polypeptides joined by flexlible linkers.
  • the polypeptide may comprise three to about eight VH-CH1 polypeptides joined by flexible linkers.
  • (c) (VL-CL) n wherein n is three or more (e.g. three to about eight, but preferably three or four); or
  • (d) (VH-CH1) n wherein n is three or more (e.g. three to about eight, but preferably three or four).
  • the polypeptide chain comprises the formula: (a) VH-CH1-flexible linker-VH-CH1-flexible linker-VH-CH1; (b) VH-CH1-flexible linker-VH-CH1-flexible linker-VH-CH1-flexible linker-VH-CH1; or (c) (VH-CH1) n , wherein n is three or four.
  • the multivalent antibodies herein have desirable properties particularly for in vivo therapy and diagnosis.
  • the multivalent antibody may be internalized and catabolized by a cell expressing an antigen, to which the antibody binds, faster than a bivalent antibody.
  • the invention provides an immunoconjugate comprising the multivalent antibody conjugated with a cytotoxic agent (e.g. one which is active in killing cells once internalized).
  • a cytotoxic agent e.g. one which is active in killing cells once internalized.
  • cytotoxic agents for generating an immunoconjugate are described herein, but the preferred cytotoxic agent is a radioactive isotope, a maytansinoid or a calecheamicin.
  • the multivalent antibody, and/or a parent antibody from which at least one of the multivalent antibody's antigen binding specificities is derived may have certain properties.
  • the multivalent antibody and/or parent antibody may (1) be an agonist antibody (e.g. where an antigen bound by the antibody is a receptor in the TNF receptor family or a B cell surface antigen); (2) induce apoptosis (for instance, where an antigen bound by the antibody is an ErbB receptor or a receptor in the TNF receptor superfamily); (3) bind a cell surface protein (such as a B cell surface antigen or an ErbB receptor) expressed on tumor cells; (4) bind a cell surface protein (e.g. Epidermal Growth Factor Receptor (EGFR), HER2 receptor, ErbB3 receptor, ErbB4 receptor, or DcR3 receptor) overexpressed by tumor cells; and/or (5) be a growth inhibitory antibody.
  • EGFR Epidermal Growth Factor Receptor
  • HER2 receptor ErbB3 receptor
  • the multivalent antibody herein may have specificity for only one antigen, or more than one antigens (e.g. from two to about three antigens).
  • the three or more functional antigen binding sites of the multivalent antibody may all bind the same antigen (preferably the same epitope on that antigen, in which case the multivalent antibody would be considered to be “monospecific”).
  • This application also provides “multispecific” antibodies.
  • the three or more functional antigen binding sites may bind two or more (e.g. from two to about three) different antigens or epitopes.
  • a multivalent antibody directed against a receptor antigen can be engineered which, surprisingly, has agonistic and/or apoptosis-inducing capability which is quantitatively similar to that of the native ligand.
  • quantitatively similar here is meant that the multivalent antibody has an activity in an assay which determines agonistic and/or apoptosis-inducing activity, within about ten fold, and preferably within about five fold of the agonistic and/or apoptosis-inducing activity of the native ligand.
  • the antibody with agonistic and/or apoptosis-inducing activity may be one with specificity for a receptor in the TNF receptor superfamily, e.g.
  • an Apo2L receptor such as DR4, DR5, DcR1 and DcR2 (preferably DR4 or DR5), in which case the activity of the antibody in an apoptosis assay such as those described in Example 3 below is within about ten fold, e.g. within about five fold, of the activity of Apo2L in that assay.
  • the multivalent antibody herein may, in one embodiment of the invention, bind a B cell surface antigen.
  • Preferred B cell surface antigens include CD19, CD20, CD22 and CD40, and most preferably CD20.
  • multivalent antibodies herein are contemplated and described in more detail below. Where the multivalent antibody possesses one or more functional Fc regions, it is anticipated to have the ability to mediate effector functions (such as ADCC and CDC) and have a longer half-life than multivalent antibodies lacking an Fc region. Such multivalent antibodies may be used where killing of cells, such as tumor or cancer cells, is desired. Other forms of the multivalent antibodies herein which lack a Fc region may be desirable where a shorter half-life is desired (e.g. for treating cardiovascular or inflammatory diseases or disorders, or where the antibody is conjugated with a cytotoxic agent); where internalization of the antibody is desired (e.g.
  • an immunoconjugate comprising the antibody and a cytotoxic agent
  • a cytotoxic agent for improved penetration of a solid tumor
  • expression of the multivalent antibody in a non-mammalian host cell e.g. a prokaryotic host cell such as an E. coli host cell
  • a non-mammalian host cell e.g. a prokaryotic host cell such as an E. coli host cell
  • Such forms of the antibody may comprise a multivalent antibody including a dimerization domain, wherein the dimerization domain comprises an antibody hinge region fused to a leucine zipper domain (the leucine zipper domain facilitates association of the polypeptides which form the dimerization domain, but may be subsequently proteolytically removed prior to administration to a patient) (see FIG. 23C); a multivalent antibody with three antigen binding sites such as that shown in FIG. 23D; or a multivalent antibody with four antigen binding sites such as that depicted in FIG. 23E.
  • the multivalent antibody herein is directed against, or binds specifically to, one or more target antigen(s).
  • at least one of the antigens bound by the multivalent antibody is a biologically important polypeptide and administration of the antibody to a mammal suffering from a disease or disorder can result in a therapeutic benefit in that mammal.
  • antibodies directed against nonpolypeptide antigens are also contemplated.
  • the antigen is a polypeptide, it may be a transmembrane molecule (e.g. receptor) or ligand such as a growth factor.
  • exemplary antigens include molecules such as renin; a growth hormone, including human growth hormone and bovine growth hormone; growth hormone releasing factor; parathyroid hormone; thyroid stimulating hormone; lipoproteins; alpha-1-antitrypsin; insulin A-chain; insulin B-chain; proinsulin; follicle stimulating hormone; calcitonin; luteinizing hormone; glucagon; clotting factors such as factor VIIIC, factor IX, tissue factor (TF), and von Willebrands factor; anti-clotting factors such as Protein C; atrial natriuretic factor; lung surfactant; a plasminogen activator, such as urokinase or human urine or tissue-type plasminogen activator (t-PA); bombesin; thrombin; hemopoietic growth factor; tumor necrosis factor-alpha
  • Preferred molecular targets for antibodies encompassed by the present invention include leukocyte surface markers or CD proteins such as CD1 a-c, CD2, CD2R, CD3, CD4, CD5, CD6, CD7, CD8, CD9, CD10, CD11a, CD11b, CD11c, CDw12, CD13, CD14, CD15, CD15s, CD16, CD16b, CDw17, CD18, CD19, CD20, CD21, CD22, CD23, CD24, CD25, CD26, CD27, CD28, CD29, CD30, CD31, CD32, CD33, CD34, CD35, CD36, CD37, CD38, CD39, CD40, C41, CD42a-d, CD43, CD44, CD44R, CD45, CD45A, CD45B, CD450, CD46-CD48, CD49a-f, CD50, CD51, CD52, CD53-CD59, CDw60, CD61, CD62E, CD62L, CD62P, CD63, CD64
  • anti-CD11a, anti-CD18 or anti-CD11b antibodies growth factors such as VEGF; tissue factor (TF); alpha interferon ( ⁇ -IFN); an interleukin, such as IL-8; IgE; blood group antigens; flk2/flt3 receptor; obesity (OB) receptor; c-mpl receptor; CTLA-4; protein C etc.
  • growth factors such as VEGF; tissue factor (TF); alpha interferon ( ⁇ -IFN); an interleukin, such as IL-8; IgE; blood group antigens; flk2/flt3 receptor; obesity (OB) receptor; c-mpl receptor; CTLA-4; protein C etc.
  • Soluble antigens or fragments thereof, optionally conjugated to other molecules, can be used as immunogens for generating antibodies.
  • immunogens for transmembrane molecules, such as receptors, fragments of these (e.g. the extracellular domain of a receptor) can be used as the immunogen.
  • transmembrane molecules such as receptors
  • fragments of these e.g. the extracellular domain of a receptor
  • cells expressing the transmembrane molecule can be used as the immunogen.
  • Such cells can be derived from a natural source (e.g. cancer cell lines) or may be cells which have been transformed by recombinant techniques to express the transmembrane molecule.
  • Other antigens and forms thereof useful for preparing antibodies will be apparent to those in the art.
  • Preferred target antigens for the multivalent antibodies herein include (1) ErbB receptors, including EGFR, HER2, HER3 and HER4; (2) receptors in the TNF receptor superfamily, e.g. Apo2L receptors, such as DR4, DR5, DcR1 and DcR2; (3) B cell surface antigens, especially CD19, CD20, CD22 and CD40; (4) antigens expressed by tumor cells; (5) antigens overexpressed by tumor cells (e.g. ErbB receptors; DcR3 receptors); (6) receptors activated by multimeric (e.g. dimeric or trimeric) ligands (e.g. receptors in the TNF receptor superfamily; VEGF receptors, etc.).
  • three or more (e.g. four to about eight) of the antigen binding sites of the multivalent antibody may all be directed against the same antigenic determinant or epitope on one of the above antigens.
  • the present application also provides multispecific antibodies, i.e., antibodies that have binding specificities for at least two different epitopes or antigenic determinants.
  • Multispecific antibodies e.g. bispecific antibodies; BsAbs
  • BsAbs bispecific antibodies
  • Bispecific antibodies have been very useful in probing the functional properties of cell surface molecules and in defining the ability of the different Fc receptors to mediate cytotoxicity (Fanger et al., Crit. Rev. Immunol 12:101-124 (1992)). Nolan et al., Biochem. Biophys. Acta. 1040:1-11 (1990) describe other diagnostic applications for BsAbs.
  • BsAbs can be constructed to immobilize enzymes for use in enzyme immunoassays.
  • one arm of the BsAb can be designed to bind to a specific epitope on the enzyme so that binding does not cause enzyme inhibition, the other arm of the BsAb binds to the immobilizing matrix ensuring a high enzyme density at the desired site.
  • diagnostic BsAbs include the rabbit anti-IgG/anti-ferritin BsAb described by Hammerling et al., J. Exp. Med. 128:1461-1473 (1968) which was used to locate surface antigens.
  • BsAbs having binding specificities for Horse Radish Peroxidase (HRP) as well as a hormone have also been developed.
  • Another potential immunochemical application for BsAbs involves their use in two-site immunoassays. For example, two BsAbs are produced binding to two separate epitopes on the analyte protein—one BsAb binds the complex to an insoluble matrix, the other binds an indicator enzyme (see Nolan et al., supra).
  • Multispecific antibodies can also be used for in vitro or in vivo immunodiagnosis of various diseases such as cancer (Songsivilai et al., Clin. Exp. Immunol. 79:315 (1990)).
  • one arm of the BsAb can bind a tumor associated antigen and the other arm can bind a detectable marker such as a chelator which tightly binds a radionuclide.
  • a detectable marker such as a chelator which tightly binds a radionuclide.
  • Multispecific antibodies can also be used for human therapy in redirected cytotoxicity by providing one arm which binds a target (e.g. pathogen or tumor cell) and another arm which binds a cytotoxic trigger molecule, such as the T-cell receptor or an Fc gamma receptor. Accordingly, multispecific antibodies can be used to direct a patient's cellular immune defense mechanisms specifically to the tumor cell or infectious agent. Using this strategy, it has been demonstrated that bispecific antibodies which bind to the Fc gamma RIII (i.e. CD16) can mediate tumor cell killing by natural killer (NK) cell/large granular lymphocyte (LGL) cells in vitro and are effective in preventing tumor growth in vivo.
  • NK natural killer
  • LGL large granular lymphocyte
  • the bispecific antibodies link the CD3 complex on T cells to a tumor-associated antigen.
  • a fully humanized F(ab′) 2 BsAb consisting of anti-CD3 linked to anti-p185 HER2 has been used to target T cells to kill tumor cells overexpressing the HER2 receptor.
  • Bispecific antibodies have been tested in several early phase clinical trials with encouraging results. In one trial, 12 patients with lung, ovarian or breast cancer were treated with infusions of activated T-lymphocytes targeted with an anti-CD3/anti-tumor (MOC31) bispecific antibody.
  • Multispecific antibodies may also be used as fibrinolytic agents or vaccine adjuvants. Furthermore, these antibodies may be used in the treatment of infectious diseases (e.g. for targeting of effector cells to virally infected cells such as HIV or influenza virus or protozoa such as Toxoplasma gondii), used to deliver immunotoxins to tumor cells, or target immune complexes to cell surface receptors (see Fanger et al., supra).
  • infectious diseases e.g. for targeting of effector cells to virally infected cells such as HIV or influenza virus or protozoa such as Toxoplasma gondii
  • the multispecific antibody may bind two or more different epitopes on an antigen of interest.
  • the multispecfic antibody may have specificity for (1) an antigen expressed by a target cell (e.g. where the target cell is a tumor cell) and (2) a triggering molecule on a leukocyte, such as a T-cell receptor molecule (e.g. CD2 or CD3), or Fc receptors for IgG (Fc gamma R), such as Fc gamma RI (CD64), Fc gamma RII (CD32) and Fc gamma RIII (CD16) so as to focus cellular defense mechanisms to the antigen-expressing cell.
  • a target cell e.g. where the target cell is a tumor cell
  • a triggering molecule on a leukocyte such as a T-cell receptor molecule (e.g. CD2 or CD3), or Fc receptors for IgG (Fc gamma R), such as Fc gamma RI (CD
  • Multispecific antibodies may also be used to localize cytotoxic agents to cells which express the target antigen. These antibodies possess an target antigen-binding arm and an arm which binds the cytotoxic agent (e.g. saporin, interferon-alpha, vinca alkaloid, ricin A chain, methotrexate or radioactive isotope hapten).
  • cytotoxic agent e.g. saporin, interferon-alpha, vinca alkaloid, ricin A chain, methotrexate or radioactive isotope hapten.
  • WO 96/16673 describes a bispecific anti-HER2/anti-Fc gamma RIII antibody and U.S. Pat. No. 5,837,234 discloses a bispecific anti-HER2/anti-Fc gamma RI antibody. A bispecific anti-HER2/Fc alpha antibody is shown in WO98/02463. U.S. Pat. No. 5,821,337 teaches a bispecific anti-HER2/anti-CD3 antibody.
  • a “parent” or “starting” antibody with variable domains directed against an antigen may be prepared using various methodologies for making antibodies, such as those described hereinbelow.
  • the sequences of the variable domains of the starting or parent antibody may be used in the design of the multivalent antibody herein.
  • Polyclonal antibodies are preferably raised in animals by multiple subcutaneous (sc) or intraperitoneal (ip) injections of the relevant antigen and an adjuvant. It may be useful to conjugate the relevant antigen to a protein that is immunogenic in the species to be immunized, e.g., keyhole limpet hemocyanin, serum albumin, bovine thyroglobulin, or soybean trypsin inhibitor using a bifunctional or derivatizing agent, for example, maleimidobenzoyl sulfosuccinimide ester (conjugation through cysteine residues), N-hydroxysuccinimide (through lysine residues), glutaraldehyde, succinic anhydride, SOCI 2 , or R 1 N ⁇ C ⁇ NR, where R and R 1 are different alkyl groups.
  • a protein that is immunogenic in the species to be immunized e.g., keyhole limpet hemocyanin, serum albumin, bovine thyrog
  • Animals are immunized against the antigen, immunogenic conjugates, or derivatives by combining, e.g., 100 ⁇ g or 5 ⁇ g of the protein or conjugate (for rabbits or mice, respectively) with 3 volumes of Freund's complete adjuvant and injecting the solution intradermally at multiple sites.
  • the animals are boosted with 1 ⁇ 5 to ⁇ fraction (1/10) ⁇ the original amount of peptide or conjugate in Freund's complete adjuvant by subcutaneous injection at multiple sites.
  • Seven to 14 days later the animals are bled and the serum is assayed for antibody titer. Animals are boosted until the titer plateaus.
  • the animal is boosted with the conjugate of the same antigen, but conjugated to a different protein and/or through a different cross-linking reagent.
  • Conjugates also can be made in recombinant cell culture as protein fusions.
  • aggregating agents such as alum are suitably used to enhance the immune response.
  • Monoclonal antibodies are obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally occurring mutations that may be present in minor amounts.
  • the modifier “monoclonal” indicates the character of the antibody as not being a mixture of discrete antibodies.
  • the monoclonal antibodies may be made using the hybridoma method first described by Kohler et al., Nature, 256:495 (1975), or may be made by recombinant DNA methods (U.S. Pat. No. 4,816,567).
  • lymphocytes that produce or are capable of producing antibodies that will specifically bind to the protein used for immunization.
  • lymphocytes may be immunized in vitro. Lymphocytes then are fused with myeloma cells using a suitable fusing agent, such as polyethylene glycol, to form a hybridoma cell (Goding, Monoclonal Antibodies: Principles and Practice , pp.59-103 (Academic Press, 1986)).
  • the hybridoma cells thus prepared are seeded and grown in a suitable culture medium that preferably contains one or more substances that inhibit the growth or survival of the unfused, parental myeloma cells.
  • a suitable culture medium that preferably contains one or more substances that inhibit the growth or survival of the unfused, parental myeloma cells.
  • the culture medium for the hybridomas typically will include hypoxanthine, aminopterin, and thymidine (HAT medium), which substances prevent the growth of HGPRT-deficient cells.
  • Preferred myeloma cells are those that fuse efficiently, support stable high-level production of antibody by the selected antibody-producing cells, and are sensitive to a medium such as HAT medium.
  • preferred myeloma cell lines are murine myeloma lines, such as those derived from MOPC-21 and MPC-11 mouse tumors available from the Salk Institute Cell Distribution Center, San Diego, Calif. USA, and SP-2 or X63-Ag8-653 cells available from the American Type Culture Collection, Rockville, Md. USA.
  • Human myeloma and mouse-human heteromyeloma cell lines also have been described for the production of human monoclonal antibodies (Kozbor, J. Immunol., 133:3001 (1984); and Brodeur et al., Monoclonal Antibody Production Techniques and Applications , pp. 51-63 (Marcel Dekker, Inc., New York, 1987)).
  • Culture medium in which hybridoma cells are growing is assayed for production of monoclonal antibodies directed against the antigen.
  • the binding specificity of monoclonal antibodies produced by hybridoma cells is determined by immunoprecipitation or by an in vitro binding assay, such as radioimmunoassay (RIA) or enzyme-linked immunoabsorbent assay (ELISA).
  • RIA radioimmunoassay
  • ELISA enzyme-linked immunoabsorbent assay
  • the binding affinity of the monoclonal antibody can, for example, be determined by the Scatchard analysis of Munson et al., Anal. Biochem., 107:220 (1980).
  • the clones may be subcloned by limiting dilution procedures and grown by standard methods (Goding, Monoclonal Antibodies: Principles and Practice , pp.59-103 (Academic Press, 1986)). Suitable culture media for this purpose include, for example, D-MEM or RPMI-1640 medium.
  • the hybridoma cells may be grown in vivo as ascites tumors in an animal.
  • the monoclonal antibodies secreted by the subclones are suitably separated from the culture medium, ascites fluid, or serum by conventional antibody purification procedures such as, for example, protein A-Sepharose, hydroxylapatite chromatography, gel electrophoresis, dialysis, or affinity chromatography.
  • DNA encoding the monoclonal antibodies is readily isolated and sequenced using conventional procedures (e.g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of murine antibodies).
  • the hybridoma cells serve as a preferred source of such DNA.
  • the DNA may be placed into expression vectors, which are then transfected into host cells such as E. coli cells, simian COS cells, Chinese Hamster Ovary (CHO) cells, or myeloma cells that do not otherwise produce antibody protein, to obtain the synthesis of monoclonal antibodies in the recombinant host cells.
  • monoclonal antibodies or antibody fragments can be isolated from antibody phage libraries generated using the techniques described in McCafferty et al., Nature, 348:552-554 (1990). Clackson et al., Nature, 352:624-628 (1991) and Marks et al., J. Mol. Biol., 222:581-597 (1991) describe the isolation of murine and human antibodies, respectively, using phage libraries.
  • the DNA also may be modified, for example, by substituting the coding sequence for human heavy chain and light chain constant domains in place of the homologous murine sequences (U.S. Pat. No. 4,816,567; and Morrison, et al., Proc. Natl Acad. Sci. USA, 81:6851 (1984)), or by covalently joining to the immunoglobulin coding sequence all or part of the coding sequence for a non-immunoglobulin polypeptide.
  • non-immunoglobulin polypeptides are substituted for the constant domains of an antibody, or they are substituted for the variable domains of one antigen-combining site of an antibody to create a chimeric bivalent antibody comprising one antigen-combining site having specificity for an antigen and another antigen-combining site having specificity for a different antigen.
  • Human monoclonal antibodies may be made via an adaptation of the hybridoma method first described by Kohler and Milstein by using human B lymphocytes as the fusion partner.
  • Human B lymphocytes producing an antibody of interest may, for example, be isolated from a human individual, after obtaining informed consent.
  • the individual may be producing antibodies against an autoantigen as occurs with certain disorders such as systemic lupus erythematosus (Shoenfeld et al. J. Clin. Invest 70:205 (1982)), immune-mediated thrombocytopenic purpura (ITP) (Nugent et al. Blood 70(1):16-22 (1987)), or cancer.
  • lymphocytes may be immunized in vitro.
  • a lysomotrophic agent e.g. L-leucine-O-methyl ester, L-glutamic acid dimethly ester or L-leucyl-L-leucine-O-methyl ester
  • adjuvants such as 8-mercaptoguanosine and cytokines
  • the B lymphocytes recovered from the subject or immunized in vitro are then generally immortalized in order to generate a human monoclonal antibody.
  • Techniques for immortalizing the B lymphocyte include, but are not limited to: (a) fusion of the human B lymphocyte with human, murine myelomas or mouse-human heteromyeloma cells; (b) viral transformation (e.g. with an Epstein-Barr virus; see Nugent et al., supra, for example); (c) fusion with a lymphoblastoid cell line; or (d) fusion with lymphoma cells.
  • Lymphocytes may be fused with myeloma cells using a suitable fusing agent, such as polyethylene glycol, to form a hybridoma cell (Goding, Monoclonal Antibodies: Principles and Practice , pp.59-103 (Academic Press, 1986)).
  • a suitable fusing agent such as polyethylene glycol
  • the hybridoma cells thus prepared are seeded and grown in a suitable culture medium that preferably contains one or more substances that inhibit the growth or survival of the unfused, parental myeloma cells.
  • the culture medium for the hybridomas typically will include hypoxanthine, aminopterin, and thymidine (HAT medium), which substances prevent the growth of HGPRT-deficient cells.
  • HGPRT medium hypoxanthine, aminopterin, and thymidine
  • Suitable human myeloma and mouse-human heteromyeloma cell lines have been described (Kozbor, J. Immunol., 133:3001 (1984); Brodeur et al., Monoclonal Antibody Production Techniques and Applications , pp. 51-63 (Marcel Dekker, Inc., New York, 1987)).
  • Culture medium in which hybridoma cells are growing is assayed for production of monoclonal antibodies directed against the antigen.
  • the binding specificity of monoclonal antibodies produced by hybridoma cells is determined by immunoprecipitation or by an in vitro binding assay, such as radioimmunoassay (RIA) or enzyme-linked immunoabsorbent assay (ELISA).
  • RIA radioimmunoassay
  • ELISA enzyme-linked immunoabsorbent assay
  • the clones may be subcloned by limiting dilution procedures and grown by standard methods (Goding, Monoclonal Antibodies: Principles and Practice , pp.59-103 (Academic Press, 1986)).
  • Suitable culture media for this purpose include, for example, D-MEM or RPMI-1640 medium.
  • the monoclonal antibodies secreted by the subclones are suitably separated from the culture medium, ascites fluid, or serum by conventional immunoglobulin purification procedures such as, for example, protein A chromatography, gel electrophoresis, dialysis, or affinity chromatography.
  • Human antibodies may also be generated using a non-human host, such as a mouse, which is capable of producing human antibodies.
  • a non-human host such as a mouse
  • transgenic mice are now available that are capable, upon immunization, of producing a full repertoire of human antibodies in the absence of endogenous immunoglobulin production.
  • J H antibody heavy-chain joining region
  • the human antibody may be selected from a human antibody phage display library.
  • the preparation of libraries of antibodies or fragments thereof is well known in the art and any of the known methods may be used to construct a family of transformation vectors which may be introduced into host cells.
  • Libraries of antibody light and heavy chains in phage (Huse et al., Science, 246:1275 (1989)) or of fusion proteins in phage or phagemid can be prepared according to known procedures. See, for example, Vaughan et al., Nature Biotechnology 14:309-314 (1996); Barbas et al., Proc. Natl. Acad. Sci., USA, 88:7978-7982 (1991); Marks et al., J.
  • a humanized antibody has one or more amino acid residues introduced into it from a source which is non-human. These non-human amino acid residues are often referred to as “import” residues, which are typically taken from an “import” variable domain. Humanization can be essentially performed following the method of Winter and co-workers (Jones et al., Nature, 321:522-525 (1986); Riechmann et al., Nature, 332:323-327 (1988); Verhoeyen et al., Science, 239:1534-1536 (1988)), by substituting hypervariable region sequences for the corresponding sequences of a human antibody.
  • humanized antibodies are chimeric antibodies (U.S. Pat. No. 4,816,567) wherein substantially less than an intact human variable domain has been substituted by the corresponding sequence from a non-human species.
  • humanized antibodies are typically human antibodies in which some hypervariable region residues and possibly some FR residues are substituted by residues from analogous sites in rodent antibodies.
  • variable domains both light and heavy
  • sequence of the variable domain of a rodent antibody is screened against the entire library of known human variable-domain sequences.
  • the human sequence which is closest to that of the rodent is then accepted as the human framework for the humanized antibody (Sims et al., J. Immunol., 151:2296 (1993); Chothia et al., J. Mol. Biol., 196:901 (1987)).
  • Another method uses a particular framework derived from the consensus sequence of all human antibodies of a particular subgroup of light or heavy chains.
  • the same framework may be used for several different humanized antibodies (Carter et al., Proc. Natl. Acad. Sci. USA, 89:4285 (1992); Presta et al., J. Immunol., 151:2623 (1993)).
  • humanized antibodies are prepared by a process of analysis of the parental sequences and various conceptual humanized products using three-dimensional models of the parental and humanized sequences.
  • Three-dimensional immunoglobulin models are commonly available and are familiar to those skilled in the art.
  • Computer programs are available which illustrate and display probable three-dimensional conformational structures of selected candidate immunoglobulin sequences. Inspection of these displays permits analysis of the likely role of the residues in the functioning of the candidate immunoglobulin sequence, i.e., the analysis of residues that influence the ability of the candidate immunoglobulin to bind its antigen.
  • FR residues can be selected and combined from the recipient and import sequences so that the desired antibody characteristic, such as increased affinity for the target antigen(s), is achieved.
  • the hypervariable region residues are directly and most substantially involved in influencing antigen binding.
  • F(ab′) 2 fragments can be isolated directly from recombinant host cell culture.
  • the antibody of choice is a single chain Fv fragment (scFv). See WO 93/16185; U.S. Pat. No. 5,571,894; and U.S. Pat. No. 5,587,458.
  • the antibody fragment may also be a “linear antibody”, e.g., as described in U.S. Pat. No. 5,641,870 for example. Such linear antibody fragments may be monospecific or bispecific.
  • Amino acid sequence modification(s) of the antibodies described herein are contemplated. For example, it may be desirable to improve the binding affinity and/or other biological properties of the antibody.
  • Amino acid sequence variants of the antibody are prepared by introducing appropriate nucleotide changes into the antibody nucleic acid, or by peptide synthesis. Such modifications include, for example, deletions from, and/or insertions into and/or substitutions of, residues within the amino acid sequences of the antibody. Any combination of deletion, insertion, and substitution is made to arrive at the final construct, provided that the final construct possesses the desired characteristics.
  • the amino acid changes also may alter post-translational processes of the antibody, such as changing the number or position of glycosylation sites. Such alterations may be made to the parent antibody and/or multivalent antibody and/or may be introduced in the multivalent antibody amino acid sequence at the time that sequence is made.
  • a useful method for identification of certain residues or regions of the antibody that are preferred locations for mutagenesis is called “alanine scanning mutagenesis” as described by Cunningham and Wells Science, 244:1081-1085 (1989).
  • a residue or group of target residues are identified (e.g., charged residues such as arg, asp, his, lys, and glu) and replaced by a neutral or negatively charged amino acid (most preferably alanine or polyalanine) to affect the interaction of the amino acids with antigen.
  • Those amino acid locations demonstrating functional sensitivity to the substitutions then are refined by introducing further or other variants at, or for, the sites of substitution.
  • the site for introducing an amino acid sequence variation is predetermined, the nature of the mutation per se need not be predetermined. For example, to analyze the performance of a mutation at a given site, ala scanning or random mutagenesis is conducted at the target codon or region and the expressed multivalent antibodies are screened for the desired activity.
  • Amino acid sequence insertions include amino- and/or carboxyl-terminal fusions ranging in length from one residue to polypeptides containing a hundred or more residues, as well as intrasequence insertions of single or multiple amino acid residues.
  • terminal insertions include an antibody with an N-terminal methionyl residue or the antibody fused to a cytotoxic polypeptide.
  • Other insertional variants of the antibody molecule include the fusion to the N- or C-terminus of the antibody to an enzyme (e.g. for ADEPT) or a polypeptide which increases the serum half-life of the antibody.
  • variants Another type of variant is an amino acid substitution variant. These variants have at least one amino acid residue in the antibody molecule replaced by a different residue.
  • the sites of greatest interest for substitutional mutagenesis include the hypervariable regions, but FR alterations are also contemplated.
  • Conservative substitutions are shown in Table 1 under the heading of “preferred substitutions”. If such substitutions result in a change in biological activity, then more substantial changes, denominated “exemplary substitutions” in Table 1, or as further described below in reference to amino acid classes, may be introduced and the products screened.
  • Substantial modifications in the biological properties of the antibody are accomplished by selecting substitutions that differ significantly in their effect on maintaining (a) the structure of the polypeptide backbone in the area of the substitution, for example, as a sheet or helical conformation, (b) the charge or hydrophobicity of the molecule at the target site, or (c) the bulk of the side chain.
  • Naturally occurring residues are divided into groups based on common side-chain properties:
  • Non-conservative substitutions will entail exchanging a member of one of these classes for another class.
  • cysteine residue not involved in maintaining the proper conformation of the antibody also may be substituted, generally with serine, to improve the oxidative stability of the molecule and prevent aberrant crosslinking. Conversely, cysteine bond(s) may be added to the antibody to improve its stability.
  • a particularly preferred type of substitutional variant involves substituting one or more hypervariable region residues of a parent antibody (e.g. a humanized or human antibody).
  • a parent antibody e.g. a humanized or human antibody
  • the resulting variant(s) selected for further development will have improved biological properties relative to the parent antibody from which they are generated.
  • a convenient way for generating such substitutional variants involves affinity maturation using phage display. Briefly, several hypervariable region sites (e.g. 6-7 sites) are mutated to generate all possible amino substitutions at each site.
  • the multivalent antibodies thus generated are displayed in a monovalent fashion from filamentous phage particles as fusions to the gene IlIl product of M13 packaged within each particle. The phage-displayed variants are then screened for their biological activity (e.g.
  • alanine scanning mutagenesis can be performed to identify hypervariable region residues contributing significantly to antigen binding.
  • Another type of amino acid variant of the antibody alters the original glycosylation pattern of the antibody. By altering is meant deleting one or more carbohydrate moieties found in the antibody, and/or adding one or more glycosylation sites that are not present in the antibody.
  • N-linked refers to the attachment of the carbohydrate moiety to the side chain of an asparagine residue.
  • the tripeptide sequences asparagine-X-serine and asparagine-X-threonine, where X is any amino acid except proline, are the recognition sequences for enzymatic attachment of the carbohydrate moiety to the asparagine side chain.
  • X is any amino acid except proline
  • O-linked glycosylation refers to the attachment of one of the sugars N-aceylgalactosamine, galactose, or xylose to a hydroxyamino acid, most commonly serine or threonine, although 5-hydroxyproline or 5-hydroxylysine may also be used.
  • Addition of glycosylation sites to the antibody is conveniently accomplished by altering the amino acid sequence such that it contains one or more of the above-described tripeptide sequences (for N-linked glycosylation sites).
  • the alteration may also be made by the addition of, or substitution by, one or more serine or threonine residues to the sequence of the original antibody (for O-linked glycosylation sites).
  • Nucleic acid molecules encoding amino acid sequence variants of the antibody are prepared by a variety of methods known in the art. These methods include, but are not limited to, isolation from a natural source (in the case of naturally occurring amino acid sequence variants) or preparation by oligonucleotide-mediated (or site-directed) mutagenesis, PCR mutagenesis, and cassette mutagenesis of an earlier prepared variant or a non-variant version of the antibody.
  • the antibody of the invention may be desirable to modify the antibody of the invention with respect to effector function, e.g. so as to enhance antigen-dependent cell-mediated cyotoxicity (ADCC) and/or complement dependent cytotoxicity (CDC) of the antibody.
  • ADCC antigen-dependent cell-mediated cyotoxicity
  • CDC complement dependent cytotoxicity
  • This may be achieved by introducing one or more amino acid modifications in an Fc region of the antibody, thereby generating a variant Fc region.
  • the Fc region variant may comprise a human Fc region sequence (e.g., a human IgG1, IgG2, IgG3 or IgG4 Fc region) comprising an amino acid modification (e.g. a substitution) at one or more amino acid positions.
  • the variant Fc region may mediate antibody-dependent cell-mediated cytotoxicity (ADCC) in the presence of human effector cells more effectively, or bind an Fc gamma receptor (Fc ⁇ R) with better affinity, than a native sequence Fc region.
  • ADCC antibody-dependent cell-mediated cytotoxicity
  • Such Fc region variants may comprise an amino acid modification at any one or more of positions 256, 290, 298, 312, 326, 330, 333, 334, 360, 378 or 430 of the Fc region, wherein the numbering of the residues in the Fc region is that of the EU index as in Kabat.
  • the Fc region variant with reduced binding to an Fc ⁇ R may comprise an amino acid modification at any one or more of amino acid positions 238, 239, 248, 249, 252, 254, 265, 268, 269, 270, 272, 278, 289, 292, 293, 294, 295, 296, 298, 301, 303, 322, 324, 327, 329, 333, 335, 338, 340, 373, 376, 382, 388, 389, 414, 416, 419, 434, 435, 437, 438 or 439 of the Fc region, wherein the numbering of the residues in the Fc region is that of the EU index as in Kabat.
  • the Fc region variant may display reduced binding to an Fc ⁇ RI and comprise an amino acid modification at any one or more of amino acid positions 238, 265, 269, 270, 327 or 329 of the Fc region, wherein the numbering of the residues in the Fc region is that of the EU index as in Kabat.
  • the Fc region variant may display reduced binding to an Fc ⁇ RII and comprise an amino acid modification at any one or more of amino acid positions 238, 265, 269, 270, 292, 294, 295, 298, 303, 324, 327, 329, 333, 335, 338, 373, 376, 414, 416, 419, 435, 438 or 439 of the Fc region, wherein the numbering of the residues in the Fc region is that of the EU index as in Kabat.
  • the Fc region variant of interest may display reduced binding to an Fc ⁇ RIII and comprise an amino acid modification at one or more of amino acid positions 238, 239, 248, 249, 252, 254, 265, 268, 269, 270, 272, 278, 289, 293, 294, 295, 296, 301, 303, 322, 327, 329, 338, 340, 373, 376, 382, 388, 389, 416, 434, 435 or 437 of the Fc region, wherein the numbering of the residues in the Fc region is that of the EU index as in Kabat.
  • the Fc region variant displays improved binding to the Fc ⁇ R and comprises an amino acid modification at any one or more of amino acid positions 255, 256, 258, 267, 268,272,276,280,283, 285,286,290, 298, 301, 305, 307,309, 312,315, 320, 322,326, 330, 331, 333, 334, 337, 340, 360, 378, 398 or 430 of the Fc region, wherein the numbering of the residues in the Fc region is that of the EU index as in Kabat.
  • the Fc region variant may display increased binding to an Fc ⁇ RIII and, optionally, may further display decreased binding to an Fc ⁇ RII.
  • An exemplary such variant comprises amino acid modification(s) at position(s) 298 and/or 333 of the Fc region, wherein the numbering of the residues in the Fc region is that of the EU index as in Kabat.
  • the Fc region variant may display increased binding to an Fc ⁇ RII and comprise an amino acid modification at any one or more of amino acid positions 255, 256, 258, 267, 268, 272, 276, 280, 283, 285, 286, 290, 301, 305, 307, 309, 312, 315, 320, 322, 326, 330, 331, 337, 340, 378, 398 or 430 of the Fc region, wherein the numbering of the residues in the Fc region is that of the EU index as in Kabat.
  • Such Fc region variants with increased binding to an Fc ⁇ RII may optionally further display decreased binding to an Fc ⁇ RIII and may, for example, comprise an amino acid modification at any one or more of amino acid positions 268, 272, 298, 301, 322 or 340 of the Fc region, wherein the numbering of the residues in the Fc region is that of the EU index as in Kabat.
  • the variant Fc region may alternatively or additionally have altered neonatal Fc receptor (FcRn) binding affinity.
  • Such variant Fc regions may comprise an amino acid modification at any one or more of amino acid positions 238, 252, 253, 254, 255, 256, 265, 272, 286, 288, 303, 305, 307, 309, 311, 312, 317, 340, 356, 360, 362, 376, 378, 380, 382, 386, 388, 400, 413, 415, 424, 433, 434, 435, 436, 439 or 447 of the Fc region, wherein the numbering of the residues in the Fc region is that of the EU index as in Kabat.
  • Fc region variants with reduced binding to an FcRn may comprise an amino acid modification at any one or more of amino acid positions 252, 253, 254, 255, 288, 309, 386, 388, 400, 415, 433, 435, 436, 439 or 447 of the Fc region, wherein the numbering of the residues in the Fc region is that of the EU index as in Kabat.
  • the above-mentioned Fc region variants may, alternatively, display increased binding to FcRn and comprise an amino acid modification at any one or more of amino acid positions 238, 256, 265, 272, 286, 303, 305, 307, 311, 312, 317, 340, 356, 360, 362, 376, 378, 380, 382, 413, 424 or 434 of the Fc region, wherein the numbering of the residues in the Fc region is that of the EU index as in Kabat.
  • Fc region variants with altered (i.e. improved or diminished) C1q binding and/or Complement Dependent Cytotoxicity (CDC) are described in WO99/51642.
  • Such variants may comprise an amino acid substitution at one or more of amino acid positions 270, 322, 326, 327, 329, 331, 333 or 334 of the Fc region. See, also, Duncan & Winter Nature 322:738-40 (1988); U.S. Pat. No. 5,648,260; U.S. Pat. No. 5,624,821; and WO94/29351 concerning Fc region variants.
  • a salvage receptor binding epitope into the antibody (especially an antibody fragment) as described in U.S. Pat. No. 5,739,277, for example.
  • the term “salvage receptor binding epitope” refers to an epitope of the Fc region of an IgG molecule (e.g., IgG1, IgG 2 , IgG 3 , or IgG 4 ) that is responsible for increasing the in vivo serum half-life of the IgG molecule.
  • the invention also pertains to immunoconjugates comprising an antibody conjugated to a cytotoxic agent such as a chemotherapeutic agent, toxin (e.g. a small molecule toxin or an enzymatically active toxin of bacterial, fungal, plant or animal origin, including fragments and/or variants thereof), or a radioactive isotope (i.e., a radioconjugate).
  • a cytotoxic agent such as a chemotherapeutic agent, toxin (e.g. a small molecule toxin or an enzymatically active toxin of bacterial, fungal, plant or animal origin, including fragments and/or variants thereof), or a radioactive isotope (i.e., a radioconjugate).
  • Conjugates of an antibody and one or more small molecule toxins such as a calicheamicin, a maytansine (U.S. Pat. No. 5,208,020), a trichothene, and CC1065 are also contemplated herein.
  • the antibody is conjugated to one or more maytansine molecules (e.g. about 1 to about 10 maytansine molecules per antibody molecule).
  • Maytansine may, for example, be converted to May-SS-Me which may be reduced to May-SH3 and reacted with modified antibody (Chari et al. Cancer Research 52: 127-131 (1992)) to generate a maytansinoid-antibody immunoconjugate.
  • Another immunoconjugate of interest comprises an antibody conjugated to one or more calicheamicin molecules.
  • the calicheamicin family of antibiotics are capable of producing double-stranded DNA breaks at sub-picomolar concentrations.
  • Structural analogues of calicheamicin which may be used include, but are not limited to, ⁇ 1 I , ⁇ 2 I , ⁇ 3 I , N-acetyl- ⁇ 1 I , PSAG and ⁇ 1 I (Hinman et al. Cancer Research 53: 3336-3342 (1993) and Lode et al. Cancer Research 58: 2925-2928 (1998)). See, also, U.S. Pat. Nos. 5,714,586; 5,712,374; 5,264,586; and 5,773,001 expressly incorporated herein by reference.
  • Enzymatically active toxins and fragments thereof which can be used include diphtheria A chain, nonbinding active fragments of diphtheria toxin, exotoxin A chain (from Pseudomonas aeruginosa ), ricin A chain, abrin A chain, modeccin A chain, alpha-sarcin, Aleurites fordii proteins, dianthin proteins, Phytolaca americana proteins (PAPI, PAPII, and PAP-S), momordica charantia inhibitor, curcin, crotin, sapaonaria officinalis inhibitor, gelonin, mitogellin, restrictocin, phenomycin, enomycin and the tricothecenes. See, for example, WO 93/21232 published Oct. 28, 1993.
  • the present invention further contemplates an immunoconjugate formed between an antibody and a compound with nucleolytic activity (e.g. a ribonuclease or a DNA endonuclease such as a deoxyribonuclease; DNase).
  • a compound with nucleolytic activity e.g. a ribonuclease or a DNA endonuclease such as a deoxyribonuclease; DNase.
  • radioactive isotopes are available for the production of radioconjugated antibodies. Examples include At 211 , I 131 , I 125 , Y 90 , Re 186 , Re 188 , Sm 153 , Bi 212 , P 32 and radioactive isotopes of Lu.
  • Conjugates of the antibody and cytotoxic agent may be made using a variety of bifunctional protein coupling agents such as N-succinimidyl-3-(2-pyridyldithiol) propionate (SPDP), succinimidyl-4-(N-maleimidomethyl) cyclohexane-1-carboxylate, iminothiolane (IT), bifunctional derivatives of imidoesters (such as dimethyl adipimidate HCL), active esters (such as disuccinimidyl suberate), aldehydes (such as glutareldehyde), bis-azido compounds (such as bis (p-azidobenzoyl) hexanediamine), bis-diazonium derivatives (such as bis-(p-diazoniumbenzoyl)-ethylenediamine), diisocyanates (such as tolyene 2,6-diisocyanate), and bis-active fluorine compounds (such as 1,5), N-
  • a ricin immunotoxin can be prepared as described in Vitetta et al. Science 238: 1098 (1987).
  • Carbon-14-labeled 1-isothiocyanatobenzyl-3-methyldiethylene triaminepentaacetic acid (MX-DTPA) is an exemplary chelating agent for conjugation of radionucleotide to the antibody. See WO94/11026.
  • the linker may be a “cleavable linker” facilitating release of the cytotoxic drug in the cell.
  • an acid-labile linker, peptidase-sensitive linker, dimethyl linker or disulfide-containing linker (Chari et al. Cancer Research 52: 127-131 (1992)) may be used.
  • a fusion protein comprising the antibody and cytotoxic agent may be made, e.g. by recombinant techniques or peptide synthesis.
  • the antibody may be conjugated to a “receptor” (such streptavidin) for utilization in tumor pretargeting wherein the antibody-receptor conjugate is administered to the patient, followed by removal of unbound conjugate from the circulation using a clearing agent and then administration of a “ligand” (e.g. avidin) which is conjugated to a cytotoxic agent (e.g. a radionucleotide).
  • a “receptor” such streptavidin
  • a ligand e.g. avidin
  • cytotoxic agent e.g. a radionucleotide
  • the antibodies of the present invention may also be used in ADEPT by conjugating the antibody to a prodrug-activating enzyme which converts a prodrug (e.g. a peptidyl chemotherapeutic agent, see WO81/01145) to an active anti-cancer drug.
  • a prodrug e.g. a peptidyl chemotherapeutic agent, see WO81/01145
  • WO 88/07378 and U.S. Pat. No. 4,975,278 See, for example, WO 88/07378 and U.S. Pat. No. 4,975,278.
  • the enzyme component of the immunoconjugate useful for ADEPT includes any enzyme capable of acting on a prodrug in such a way so as to covert it into its more active, cytotoxic form.
  • Enzymes that are useful in the method of this invention include, but are not limited to, alkaline phosphatase useful for converting phosphate-containing prodrugs into free drugs; arylsulfatase useful for converting sulfate-containing prodrugs into free drugs; cytosine deaminase useful for converting non-toxic 5-fluorocytosine into the anti-cancer drug, 5-fluorouracil; proteases, such as serratia protease, thermolysin, subtilisin, carboxypeptidases and cathepsins (such as cathepsins B and L), that are useful for converting peptide-containing prodrugs into free drugs; D-alanylcarboxypeptidases, useful for converting prodrugs that contain D-amino acid substituents; carbohydrate-cleaving enzymes such as beta-galactosidase and neuraminidase useful for converting glycosylated prodrugs into free drugs;
  • antibodies with enzymatic activity can be used to convert the prodrugs of the invention into free active drugs (see, e.g., Massey, Nature 328: 457-458 (1987)).
  • Antibody-abzyme conjugates can be prepared as described herein for delivery of the abzyme to a tumor cell population.
  • the enzymes of this invention can be covalently bound to the antibodies by techniques well known in the art such as the use of the heterobifunctional crosslinking reagents discussed above.
  • fusion proteins comprising at least the antigen binding region of an antibody of the invention linked to at least a functionally active portion of an enzyme of the invention can be constructed using recombinant DNA techniques well known in the art (see, e.g., Neuberger et al., Nature, 312: 604-608 (1984).
  • the antibody may be linked to one of a variety of nonproteinaceous polymers, e.g., polyethylene glycol, polypropylene glycol, polyoxyalkylenes, or copolymers of polyethylene glycol and polypropylene glycol.
  • nonproteinaceous polymers e.g., polyethylene glycol, polypropylene glycol, polyoxyalkylenes, or copolymers of polyethylene glycol and polypropylene glycol.
  • the antibody also may be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization (for example, hydroxymethylcellulose or gelatin-microcapsules and poly-(methylmethacylate) microcapsules, respectively), in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules), or in macroemulsions.
  • colloidal drug delivery systems for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules
  • the antibodies disclosed herein may also be formulated as immunoliposomes.
  • Liposomes containing the antibody are prepared by methods known in the art, such as described in Epstein et al., Proc. Natl. Acad. Sci. USA, 82:3688 (1985); Hwang et al., Proc. Natl Acad. Sci. USA, 77:4030 (1980); U.S. Pat. Nos. 4,485,045 and 4,544,545; and WO97/38731 published Oct. 23, 1997. Liposomes with enhanced circulation time are disclosed in U.S. Pat. No. 5,013,556.
  • Particularly useful liposomes can be generated by the reverse phase evaporation method with a lipid composition comprising phosphatidylcholine, cholesterol and PEG-derivatized phosphatidylethanolamine (PEG-PE). Liposomes are extruded through filters of defined pore size to yield liposomes with the desired diameter.
  • Fab′ fragments of the antibody of the present invention can be conjugated to the liposomes as described in Martin et al. J. Biol. Chem. 257: 286-288 (1982) via a disulfide interchange reaction. A chemotherapeutic agent is optionally contained within the liposome. See Gabizon et al. J. National Cancer Inst. 81(19)1484 (1989).
  • the invention also provides isolated nucleic acid encoding an antibody as disclosed herein, vectors and host cells comprising the nucleic acid, and recombinant techniques for the production of the antibody.
  • the nucleic acid encoding it is isolated and inserted into a replicable vector for further cloning (amplification of the DNA) or for expression.
  • DNA encoding the antibody is readily isolated and sequenced using conventional procedures (e.g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the antibody).
  • Many vectors are available.
  • the vector components generally include, but are not limited to, one or more of the following: a signal sequence, an origin of replication, one or more marker genes, an enhancer element, a promoter, and a transcription termination sequence.
  • the multivalent antibody of this invention may be produced recombinantly not only directly, but also as a fusion polypeptide with a heterologous polypeptide, which is preferably a signal sequence or other polypeptide having a specific cleavage site at the N-terminus of the mature protein or polypeptide.
  • a heterologous polypeptide which is preferably a signal sequence or other polypeptide having a specific cleavage site at the N-terminus of the mature protein or polypeptide.
  • the heterologous signal sequence selected preferably is one that is recognized and processed (i.e., cleaved by a signal peptidase) by the host cell.
  • the signal sequence is substituted by a prokaryotic signal sequence selected, for example, from the group of the alkaline phosphatase, penicillinase, Ipp, or heat-stable enterotoxin 11 leaders.
  • a prokaryotic signal sequence selected, for example, from the group of the alkaline phosphatase, penicillinase, Ipp, or heat-stable enterotoxin 11 leaders.
  • yeast secretion the native signal sequence may be substituted by, e.g., the yeast invertase leader, a factor leader (including Saccharomyces and Kluyveromyces ⁇ -factor leaders), or acid phosphatase leader, the C. albicans glucoamylase leader, or the signal described in WO 90/13646.
  • mammalian signal sequences as well as viral secretory leaders for example, the herpes simplex gD signal, are available.
  • the DNA for such precursor region is ligated in reading frame to DNA encoding the multivalent antibody.
  • Both expression and cloning vectors contain a nucleic acid sequence that enables the vector to replicate in one or more selected host cells.
  • this sequence is one that enables the vector to replicate independently of the host chromosomal DNA, and includes origins of replication or autonomously replicating sequences.
  • origins of replication or autonomously replicating sequences are well known for a variety of bacteria, yeast, and viruses.
  • the origin of replication from the plasmid pBR322 is suitable for most Gram-negative bacteria, the 2 ⁇ plasmid origin is suitable for yeast, and various viral origins (SV40, polyoma, adenovirus, VSV or BPV) are useful for cloning vectors in mammalian cells.
  • the origin of replication component is not needed for mammalian expression vectors (the SV40 origin may typically be used only because it contains the early promoter).
  • Expression and cloning vectors may contain a selection gene, also termed a selectable marker.
  • Typical selection genes encode proteins that (a) confer resistance to antibiotics or other toxins, e.g., ampicillin, neomycin, methotrexate, or tetracycline, (b) complement auxotrophic deficiencies, or (c) supply critical nutrients not available from complex media, e.g., the gene encoding D-alanine racemase for Bacilli.
  • One example of a selection scheme utilizes a drug to arrest growth of a host cell. Those cells that are successfully transformed with a heterologous gene produce a protein conferring drug resistance and thus survive the selection regimen. Examples of such dominant selection use the drugs neomycin, mycophenolic acid and hygromycin.
  • Suitable selectable markers for mammalian cells are those that enable the identification of cells competent to take up the multivalent antibody nucleic acid, such as DHFR, thymidine kinase, metallothionein-I and -II, preferably primate metallothionein genes, adenosine deaminase, ornithine decarboxylase, etc.
  • cells transformed with the DHFR selection gene are first identified by culturing all of the transformants in a culture medium that contains methotrexate (Mtx), a competitive antagonist of DHFR.
  • Mtx methotrexate
  • An appropriate host cell when wild-type DHFR is employed is the Chinese hamster ovary (CHO) cell line deficient in DHFR activity.
  • host cells transformed or co-transformed with DNA sequences encoding multivalent antibody, wild-type DHFR protein, and another selectable marker such as aminoglycoside 3′-phosphotransferase (APH) can be selected by cell growth in medium containing a selection agent for the selectable marker such as an aminoglycosidic antibiotic, e.g., kanamycin, neomycin, or G418. See U.S. Pat. No. 4,965,199.
  • APH aminoglycoside 3′-phosphotransferase
  • a suitable selection gene for use in yeast is the trp1 gene present in the yeast plasmid YRp7 (Stinchcomb et al., Nature, 282:39 (1979)).
  • the trp1 gene provides a selection marker for a mutant strain of yeast lacking the ability to grow in tryptophan, for example, ATCC No. 44076 or PEP4-1. Jones, Genetics, 85:12 (1977).
  • the presence of the trpl lesion in the yeast host cell genome then provides an effective environment for detecting transformation by growth in the absence of tryptophan.
  • Leu2-deficient yeast strains (ATCC 20,622 or 38,626) are complemented by known plasmids bearing the Leu2 gene.
  • vectors derived from the 1.6 ⁇ m circular plasmid pKD1 can be used for transformation of Kluyveromyces yeasts.
  • an expression system for large-scale production of recombinant calf chymosin was reported for K. lactis . Van den Berg, Bio/Technology, 8:135 (1990).
  • Stable multi-copy expression vectors for secretion of mature recombinant human serum albumin by industrial strains of Kluyveromyces have also been disclosed. Fleer et al., BiolTechnology, 9:968-975 (1991).
  • Expression and cloning vectors usually contain a promoter that is recognized by the host organism and is operably linked to the multivalent antibody nucleic acid.
  • Promoters suitable for use with prokaryotic hosts include the phoA promoter, ⁇ -lactamase and lactose promoter systems, alkaline phosphatase, a tryptophan (trp) promoter system, and hybrid promoters such as the tac promoter.
  • phoA promoter phoA promoter
  • ⁇ -lactamase and lactose promoter systems alkaline phosphatase
  • trp tryptophan
  • hybrid promoters such as the tac promoter.
  • Other known bacterial promoters are suitable. Promoters for use in bacterial systems also will contain a Shine-Dalgarno (S.D.) sequence operably linked to the DNA encoding the multivalent antibody.
  • S.D. Shine-Dalgarno
  • Promoter sequences are known for eukaryotes. Virtually all eukaryotic genes have an AT-rich region located approximately 25 to 30 bases upstream from the site where transcription is initiated. Another sequence found 70 to 80 bases upstream from the start of transcription of many genes is a CNCAAT region where N may be any nucleotide. At the 3′ end of most eukaryotic genes is an AATAAA sequence that may be the signal for addition of the poly A tail to the 3′ end of the coding sequence. All of these sequences are suitably inserted into eukaryotic expression vectors.
  • suitable promoting sequences for use with yeast hosts include the promoters for 3-phosphoglycerate kinase or other glycolytic enzymes, such as enolase, glyceraldehyde-3-phosphate dehydrogenase, hexokinase, pyruvate decarboxylase, phosphofructokinase, glucose-6-phosphate isomerase, 3-phosphoglycerate mutase, pyruvate kinase, triosephosphate isomerase, phosphoglucose isomerase, and glucokinase.
  • 3-phosphoglycerate kinase or other glycolytic enzymes such as enolase, glyceraldehyde-3-phosphate dehydrogenase, hexokinase, pyruvate decarboxylase, phosphofructokinase, glucose-6-phosphate isomerase, 3-phosphoglycerate mutase, pyruvate
  • yeast promoters which are inducible promoters having the additional advantage of transcription controlled by growth conditions, are the promoter regions for alcohol dehydrogenase 2, isocytochrome C, acid phosphatase, degradative enzymes associated with nitrogen metabolism, metallothionein, glyceraldehyde-3-phosphate dehydrogenase, and enzymes responsible for maltose and galactose utilization.
  • Suitable vectors and promoters for use in yeast expression are further described in EP 73,657.
  • Yeast enhancers also are advantageously used with yeast promoters.
  • Multivalent antibody transcription from vectors in mammalian host cells is controlled, for example, by promoters obtained from the genomes of viruses such as polyoma virus, fowlpox virus, adenovirus (such as Adenovirus 2), bovine papilloma virus, avian sarcoma virus, cytomegalovirus, a retrovirus, hepatitis-B virus and most preferably Simian Virus 40 (SV40), from heterologous mammalian promoters, e.g., the actin promoter or an immunoglobulin promoter, from heat-shock promoters, provided such promoters are compatible with the host cell systems.
  • viruses such as polyoma virus, fowlpox virus, adenovirus (such as Adenovirus 2), bovine papilloma virus, avian sarcoma virus, cytomegalovirus, a retrovirus, hepatitis-B virus and most preferably Simian Virus 40 (SV
  • the early and late promoters of the SV40 virus are conveniently obtained as an SV40 restriction fragment that also contains the SV40 viral origin of replication.
  • the immediate early promoter of the human cytomegalovirus is conveniently obtained as a HindIII E restriction fragment.
  • a system for expressing DNA in mammalian hosts using the bovine papilloma virus as a vector is disclosed in U.S. Pat. No. 4,419,446. A modification of this system is described in U.S. Pat. No. 4,601,978.
  • Enhancer sequences are now known from mammalian genes (globin, elastase, albumin, ⁇ -fetoprotein, and insulin). Typically, however, one will use an enhancer from a eukaryotic cell virus. Examples include the SV40 enhancer on the late side of the replication origin (bp 100-270), the cytomegalovirus early promoter enhancer, the polyoma enhancer on the late side of the replication origin, and adenovirus enhancers.
  • the enhancer may be spliced into the vector at a position 5′ or 3′ to the multivalent antibody-encoding sequence, but is preferably located at a site 5′ from the promoter.
  • Expression vectors used in eukaryotic host cells will also contain sequences necessary for the termination of transcription and for stabilizing the mRNA. Such sequences are commonly available from the 5′ and, occasionally 3′, untranslated regions of eukaryotic or viral DNAs or cDNAs. These regions contain nucleotide segments transcribed as polyadenylated fragments in the untranslated portion of the mRNA encoding the multivalent antibody.
  • One useful transcription termination component is the bovine growth hormone polyadenylation region. See WO94/11026 and the expression vector disclosed therein.
  • Suitable host cells for cloning or expressing the DNA in the vectors herein are the prokaryote, yeast, or higher eukaryote cells described above.
  • Suitable prokaryotes for this purpose include eubacteria, such as Gram-negative or Gram-positive organisms, for example, Enterobacteriaceae such as Escherichia, e.g., E. coli , Enterobacter, Erwinia, Klebsiella, Proteus, Salmonella, e.g., Salmonella typhimurium , Serratia, e.g., Serratia marcescans , and Shigella, as well as Bacilli such as B. subtilis and B.
  • Enterobacteriaceae such as Escherichia, e.g., E. coli , Enterobacter, Erwinia, Klebsiella, Proteus
  • Salmonella e.g., Salmonella typhimurium
  • Serratia e
  • E. coli cloning host is E. coli 294 (ATCC 31,446), although other strains such as E. coli B, E. coli X1776 (ATCC 31,537), and E. coli W3110 (ATCC 27,325) are suitable. These examples are illustrative rather than limiting.
  • eukaryotic microbes such as filamentous fungi or yeast are suitable cloning or expression hosts for multivalent antibody-encoding vectors.
  • Saccharomyces cerevisiae or common baker's yeast, is the most commonly used among lower eukaryotic host microorganisms.
  • a number of other genera, species, and strains are commonly available and useful herein, such as Schizosaccharomyces pombe ; Kluyveromyces hosts such as, e.g., K. lactis, K. fragilis (ATCC 12,424), K. bulgaricus (ATCC 16,045), K. wickeramii (ATCC 24,178), K.
  • watii ATCC 56,500
  • K. drosophilarum ATCC 36,906
  • K. thermotolerans K. marxianus
  • yarrowia EP 402,226
  • Pichia pastoris EP 183,070
  • Candida Trichoderma reesia
  • Neurospora crassa Schwanniomyces such as Schwanniomyces occidentalis
  • filamentous fungi such as, e.g., Neurospora, Penicillium, Tolypocladium, and Aspergillus hosts such as A. nidulans and A. niger.
  • Suitable host cells for the expression of glycosylated multivalent antibody are derived from multicellular organisms.
  • invertebrate cells include plant and insect cells.
  • Numerous baculoviral strains and variants and corresponding permissive insect host cells from hosts such as Spodoptera frugiperda (caterpillar), Aedes aegypti (mosquito), Aedes albopictus (mosquito), Drosophila melanogaster (fruitfly), and Bombyx mori have been identified.
  • a variety of viral strains for transfection are publicly available, e.g., the L-1 variant of Autographa califormica NPV and the Bm-5 strain of Bombyx mori NPV, and such viruses may be used as the virus herein according to the present invention, particularly for transfection of Spodoptera frugiperda cells.
  • Plant cell cultures of cotton, corn, potato, soybean, petunia, tomato, and tobacco can also be utilized as hosts.
  • vertebrate cells have been greatest in vertebrate cells, and propagation of vertebrate cells in culture (tissue culture) has become a routine procedure.
  • useful mammalian host cell lines are monkey kidney CV1 line transformed by SV40 (COS-7, ATCC CRL 1651); human embryonic kidney line (293 or 293 cells subcloned for growth in suspension culture, Graham et al., J. Gen Virol. 36:59 (1977)); baby hamster kidney cells (BHK, ATCC CCL 10); Chinese hamster ovary cells/-DHFR (CHO, Urlaub et al., Proc. Natl. Acad. Sci. USA 77:4216 (1980)); mouse sertoli cells (TM4, Mather, Biol. Reprod.
  • monkey kidney cells (CV1 ATCC CCL 70); African green monkey kidney cells (VERO-76, ATCC CRL-1587); human cervical carcinoma cells (HELA, ATCC CCL 2); canine kidney cells (MDCK, ATCC CCL 34); buffalo rat liver cells (BRL 3A, ATCC CRL 1442); human lung cells (WI 38, ATCC CCL 75); human liver cells (Hep G2, HB 8065); mouse mammary tumor (MMT 060562, ATCC CCL51); TRI cells (Mather et al., Annals N.Y. Acad. Sci.
  • MRC 5 cells MRC 5 cells; FS4 cells; a human hepatoma line (Hep G2); and myeloma or lymphoma cells (e.g. YO, J558L, P3 and NSO cells) (see U.S. Pat. No. 5,807,715).
  • Hep G2 human hepatoma line
  • myeloma or lymphoma cells e.g. YO, J558L, P3 and NSO cells
  • Host cells are transformed with the above-described expression or cloning vectors for multivalent antibody production and cultured in conventional nutrient media modified as appropriate for inducing promoters, selecting transformants, or amplifying the genes encoding the desired sequences.
  • the host cells used to produce the multivalent antibody of this invention may be cultured in a variety of media.
  • Commercially available media such as Ham's F10 (Sigma), Minimal Essential Medium ((MEM), (Sigma), RPMI-1640 (Sigma), and Dulbecco's Modified Eagle's Medium ((DMEM), Sigma) are suitable for culturing the host cells.
  • any of these media may be supplemented as necessary with hormones and/or other growth factors (such as insulin, transferrin, or epidermal growth factor), salts (such as sodium chloride, calcium, magnesium, and phosphate), buffers (such as HEPES), nucleotides (such as adenosine and thymidine), antibiotics (such as GENTAMYCINTM drug), trace elements (defined as inorganic compounds usually present at final concentrations in the micromolar range), and glucose or an equivalent energy source. Any other necessary supplements may also be included at appropriate concentrations that would be known to those skilled in the art.
  • the culture conditions such as temperature, pH, and the like, are those previously used with the host cell selected for expression, and will be apparent to the ordinarily skilled artisan.
  • the multivalent antibody can be produced intracellularly, in the periplasmic space, or directly secreted into the medium. If the multivalent antibody is produced intracellularly, as a first step, the particulate debris, either host cells or lysed fragments, is removed, for example, by centrifugation or ultrafiltration. Carter et al., Bio/Technology 10:163-167 (1992) describe a procedure for isolating antibodies which are secreted to the periplasmic space of E. col. Briefly, cell paste is thawed in the presence of sodium acetate (pH 3.5), EDTA, and phenylmethylsulfonylfluoride (PMSF) over about 30 min.
  • sodium acetate pH 3.5
  • EDTA EDTA
  • PMSF phenylmethylsulfonylfluoride
  • Cell debris can be removed by centrifugation.
  • supernatants from such expression systems are generally first concentrated using a commercially available protein concentration filter, for example, an Amicon or Millipore Pellicon ultrafiltration unit.
  • a protease inhibitor such as PMSF may be included in any of the foregoing steps to inhibit proteolysis and antibiotics may be included to prevent the growth of adventitious contaminants.
  • the multivalent antibody composition prepared from the cells can be purified using, for example, hydroxylapatite chromatography, gel electrophoresis, dialysis, and affinity chromatography, with affinity chromatography being the preferred purification technique.
  • affinity chromatography is the preferred purification technique.
  • the suitability of protein A as an affinity ligand depends on the species and isotype of any immunoglobulin Fc region that is present in the multivalent antibody.
  • Protein A can be used to purify antibodies that are based on human ⁇ 1, ⁇ 2, or ⁇ 4 heavy chains (Lindmark et al., J. Immunol. Meth. 62:1-13 (1983)). Protein G is recommended for all mouse isotypes and for human ⁇ 3 (Guss et al., EMBO J.
  • the matrix to which the affinity ligand is attached is most often agarose, but other matrices are available. Mechanically stable matrices such as controlled pore glass or poly(styrenedivinyl)benzene allow for faster flow rates and shorter processing times than can be achieved with agarose.
  • the Bakerbond ABXTM resin J. T. Baker, Phillipsburg, N.J. is useful for purification.
  • Therapeutic formulations of the multivalent antibody are prepared for storage by mixing the multivalent antibody having the desired degree of purity with optional physiologically acceptable carriers, excipients or stabilizers ( Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980)), in the form of aqueous solutions, lyophilized or other dried formulations.
  • Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine,
  • the formulation herein may also contain more than one active compound as necessary for the particular indication being treated, preferably those with complementary activities that do not adversely affect each other.
  • active compounds are provided in Section G below entitled “In Vivo Uses for the Multivalent Antibody”. Such molecules are suitably present in combination in amounts that are effective for the purpose intended.
  • the active ingredients may also be entrapped in microcapsule prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsule and poly-(methylmethacylate) microcapsule, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) or in macroemulsions.
  • colloidal drug delivery systems for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules
  • formulations to be used for in vivo administration must be sterile. This is readily accomplished by filtration through sterile filtration membranes.
  • sustained-release preparations may be prepared. Suitable examples of sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing the multivalent antibody, which matrices are in the form of shaped articles, e.g., films, or microcapsule. Examples of sustained-release matrices include polyesters, hydrogels (for example, poly(2-hydroxyethylmethacrylate), or poly(vinylalcohol)), polylactides (U.S. Pat. No.
  • copolymers of L-glutamic acid and y ethyl-L-glutamate non-degradable ethylene-vinyl acetate
  • degradable lactic acid-glycolic acid copolymers such as the LUPRON DEPOTM (injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate)
  • poly-D-( ⁇ )-3-hydroxybutyric acid While polymers such as ethylene-vinyl acetate and lactic acid-glycolic acid enable release of molecules for over 100 days, certain hydrogels release proteins for shorter time periods.
  • encapsulated antibodies When encapsulated antibodies remain in the body for a long time, they may denature or aggregate as a result of exposure to moisture at 37° C., resulting in a loss of biological activity and possible changes in immunogenicity. Rational strategies can be devised for stabilization depending on the mechanism involved. For example, if the aggregation mechanism is discovered to be intermolecular S-S bond formation through thio-disulfide interchange, stabilization may be achieved by modifying sulfhydryl residues, lyophilizing from acidic solutions, controlling moisture content, using appropriate additives, and developing specific polymer matrix compositions.
  • the multivalent antibody of the invention may be used as an affinity purification agent.
  • the multivalent antibody is immobilized on a solid phase such a Sephadex resin or filter paper, using methods well known in the art.
  • the immobilized multivalent antibody is contacted with a sample containing the antigen to be purified, and thereafter the support is washed with a suitable solvent that will remove substantially all the material in the sample except the antigen to be purified, which is bound to the immobilized multivalent antibody. Finally, the support is washed with another suitable solvent, such as glycine buffer, pH 5.0, that will release the antigen from the multivalent antibody.
  • the multivalent antibody may also be useful in diagnostic assays, e.g., for detecting expression of an antigen of interest in specific cells, tissues, or serum.
  • the multivalent antibody typically will be labeled with a detectable moiety.
  • a detectable moiety Numerous labels are available which can be generally grouped into the following categories:
  • Radioisotopes such as 35 S, 14 C, 125 I, 3 H, and 131 I.
  • the multivalent antibody can be labeled with the radioisotope using the techniques described in Current Protocols in Immunology , Volumes 1 and 2, Coligen et al., Ed. Wiley-Interscience, New York, N.Y., Pubs. (1991) for example and radioactivity can be measured using scintillation counting.
  • Fluorescent labels such as rare earth chelates (europium chelates) or fluorescein and its derivatives, rhodamine and its derivatives, dansyl, Lissamine, phycoerythrin and Texas Red are available.
  • the fluorescent labels can be conjugated to the multivalent antibody using the techniques disclosed in Current Protocols in Immunology , supra, for example. Fluorescence can be quantified using a fluorimeter.
  • the enzyme generally catalyzes a chemical alteration of the chromogenic substrate that can be measured using various techniques. For example, the enzyme may catalyze a color change in a substrate, which can be measured spectrophotometrically. Alternatively, the enzyme may alter the fluorescence or chemiluminescence of the substrate. Techniques for quantifying a change in fluorescence are described above.
  • the chemiluminescent substrate becomes electronically excited by a chemical reaction and may then emit light which can be measured (using a chemiluminometer, for example) or donates energy to a fluorescent acceptor.
  • enzymatic labels include luciferases (e.g., firefly luciferase and bacterial luciferase; U.S. Pat. No. 4,737,456), luciferin, 2,3-dihydrophthalazinediones, malate dehydrogenase, urease, peroxidase such as horseradish peroxidase (HRPO), alkaline phosphatase, ⁇ -galactosidase, glucoamylase, lysozyme, saccharide oxidases (e.g., glucose oxidase, galactose oxidase, and glucose-6-phosphate dehydrogenase), heterocyclic oxidases (such as uricase and xanthine oxidase), lactoperoxidase, microperoxidase, and the like.
  • luciferases e.g., firefly luciferase and bacterial lucifera
  • enzyme-substrate combinations include, for example:
  • HRPO Horseradish peroxidase
  • HPO horseradish peroxidase
  • OPD orthophenylene diamine
  • TMB 3,3′,5,5′-tetramethyl benzidine hydrochloride
  • ⁇ -D-galactosidase ( ⁇ -D-Gal) with a chromogenic substrate (e.g., p-nitrophenyl- ⁇ -D-galactosidase) or fluorogenic substrate 4-methylumbelliferyl- ⁇ -D-galactosidase.
  • a chromogenic substrate e.g., p-nitrophenyl- ⁇ -D-galactosidase
  • fluorogenic substrate 4-methylumbelliferyl- ⁇ -D-galactosidase
  • the label is indirectly conjugated with the multivalent antibody.
  • the multivalent antibody can be conjugated with biotin and any of the three broad categories of labels mentioned above can be conjugated with avidin, or vice versa. Biotin binds selectively to avidin and thus, the label can be conjugated with the multivalent antibody in this indirect manner.
  • the multivalent antibody is conjugated with a small hapten (e.g., digoxin) and one of the different types of labels mentioned above is conjugated with an anti-hapten multivalent antibody (e.g., anti-digoxin antibody).
  • a small hapten e.g., digoxin
  • an anti-hapten multivalent antibody e.g., anti-digoxin antibody
  • the multivalent antibody need not be labeled, and the presence thereof can be detected using a labeled antibody which binds to the multivalent antibody.
  • the multivalent antibody of the present invention may be employed in any known assay method, such as competitive binding assays, direct and indirect sandwich assays, and immunoprecipitation assays. Zola, Monoclonal Antibodies: A Manual of Techniques , pp.147-158 (CRC Press, Inc. 1987).
  • the multivalent antibody may also be used for in vivo diagnostic assays.
  • the multivalent antibody is labeled with a radionuclide (such as 111 In, 99 Tc, 14 C, 131 I, 125 I, 3 H, 32 P or 35S) so that the antigen or cells expressing it can be localized using immunoscintiography.
  • a radionuclide such as 111 In, 99 Tc, 14 C, 131 I, 125 I, 3 H, 32 P or 35S
  • the multivalent antibody of the present invention may be used to treat a mammal e.g. a patient suffering from, or predisposed to, a disease or disorder who could benefit from administration of the multivalent antibody.
  • conditions to be treated therewith include benign or malignant tumors; leukemias and lymphoid malignancies; other disorders such as neuronal, glial, astrocytal, hypothalamic, glandular, macrophagal, epithelial, stromal, blastocoelic, inflammatory, angiogenic and immunologic disorders.
  • the disease or disorder to be treated with the antibody that binds an ErbB receptor is cancer.
  • cancers to be treated herein include, but are not limited to, carcinoma, lymphoma, blastoma, sarcoma, and leukemia or lymphoid malignancies. More particular examples of such cancers include squamous cell cancer, lung cancer including small-cell lung cancer, non-small cell lung cancer, adenocarcinoma of the lung and squamous carcinoma of the lung, cancer of the peritoneum, hepatocellular cancer, gastric or stomach cancer including gastrointestinal cancer, pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, hepatoma, breast cancer, colon cancer, colorectal cancer, endometrial or uterine carcinoma, salivary gland carcinoma, kidney or renal cancer, liver cancer, prostate cancer, vulval cancer, thyroid cancer, hepatic carcinoma as well as head and neck cancer.
  • lung cancer including small-cell lung cancer, non-small cell lung cancer, adenocarcinoma of the lung and squamous carcinoma of
  • the cancer will generally comprise cells that express an antigen bound by the antibody, such that the antibody is able to bind to the cancer.
  • the cancer may be characterized by overexpression of the antigen (e.g. overexpression of an ErbB receptor).
  • overexpression may be analyzed by IHC, e.g. using the HERCEPTEST® (Dako) where the antigen is HER2.
  • HERCEPTEST® Dako
  • parrafin embedded tissue sections from a tumor biopsy may be subjected to the IHC assay and accorded a HER2 protein staining intensity criteria as follows:
  • Those tumors with 0 or 1 + scores for HER2 overexpression assessment may be characterized as not overexpressing HER2, whereas those tumors with 2+ or 3+ scores may be characterized as overexpressing HER2.
  • FISH assays such as the INFORMTM (sold by Ventana, Ariz.) or PATHVISIONTM (Vysis, Ill.) may be carried out on formalin-fixed, paraffin-embedded tumor tissue to determine the extent (if any) of antigen overexpression by the tumor.
  • the cancer will be one which expresses (and may overexpress) an ErbB receptor selected from the group consisting of EGFR, ErbB3 and ErbB4.
  • Examples of cancers which may express/overexpress EGFR, ErbB3 or ErbB4 include squamous cell cancer, lung cancer including small-cell lung cancer, non-small cell lung cancer, adenocarcinoma of the lung and squamous carcinoma of the lung, cancer of the peritoneum, hepatocellular cancer, gastric or stomach cancer including gastrointestinal cancer, pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, hepatoma, breast cancer, colon cancer, colorectal cancer, endometrial or uterine carcinoma, salivary gland carcinoma, kidney or renal cancer, liver cancer, prostate cancer, vulval cancer, thyroid cancer, hepatic carcinoma as well as head and neck cancer as well as glioblastomas.
  • the cancer to be treated herein may be one characterized by excessive activation of an ErbB receptor, e.g. EGFR. Such excessive activation may be attributable to overexpression or increased production of the ErbB receptor or an ErbB ligand.
  • a diagnostic or prognostic assay will be performed to determine whether the patient's cancer is characterized by excessive activation of an ErbB receptor. For example, ErbB gene amplification and/or overexpression of an ErbB receptor in the cancer may be determined.
  • Assays for determining such amplification/overexpression are available in the art and include the IHC, FISH and shed antigen assays described above.
  • levels of an ErbB ligand, such as TGF-alpha, in or associated with the tumor may be determined according to known procedures. Such assays may detect protein and/or nucleic acid encoding it in the sample to be tested. In one embodiment, ErbB ligand levels in the tumor may be determined using immunohistochemistry (IHC); see, for example, Scher et al., Clin. Cancer Research 1:545-550 (1995). Alternatively, or additionally, one may evaluate levels of ErbB ligand-encoding nucleic acid in the sample to be tested; e.g. via fluorescent in situ hybridization or FISH, southern blotting, or polymerase chain reaction (PCR) techniques.
  • IHC immunohistochemistry
  • ErbB receptor or ErbB ligand overexpression or amplification may be evaluated using an in vivo diagnostic assay, e.g. by administering a molecule (such as an antibody) which binds the molecule to be detected and is tagged with a detectable label (e.g. a radioactive isotope) and externally scanning the patient for localization of the label.
  • a detectable label e.g. a radioactive isotope
  • the antibody may be used to treat a B cell lymphoma (including low gradeffollicular non-Hodkin's lymphoma (NHL); small lymphocytic (SL) NHL; intermediate gradeffollicular NHL; intermediate grade diffuse NHL; high grade immunoblastic NHL; high grade lymphoblastic NHL; high grade small non-cleaved cell NHL; bulky disease NHL; mantle cell lymphoma; AIDS-related lymphoma; and Waldenstrom's Macroglobulinemia); chronic lymphocytic leukemia (CLL); acute lymphoblastic leukemia (ALL); Hairy cell leukemia; and chronic myeloblastic leukemia; and post-transplant lymphoproliferative disorder (PTLD).
  • B cell lymphoma including low gradeffollicular non-Hodkin's lymphoma (NHL); small lymphocytic (SL) NHL; intermediate gradeffollicular NHL; intermediate grade diffuse NHL; high grade immunoblastic NHL; high grade lymphoblastic NHL; high grade
  • the antibody e.g. the anti-B cell surface antigen antibody
  • autoimmune diseases or disorders include, but are not limited to, inflammatory responses such as inflammatory skin diseases including psoriasis and dermatitis (e.g.
  • atopic dermatitis atopic dermatitis
  • systemic scleroderma and sclerosis responses associated with inflammatory bowel disease (such as Crohn's disease and ulcerative colitis); respiratory distress syndrome (including adult respiratory distress syndrome; ARDS); dermatitis; meningitis; encephalitis; uveitis; colitis; glomerulonephritis; allergic conditions such as eczema and asthma and other conditions involving infiltration of T cells and chronic inflammatory responses; atherosclerosis; leukocyte adhesion deficiency; rheumatoid arthritis; systemic lupus erythematosus (SLE); diabetes mellitus (e.g.
  • Type I diabetes mellitus or insulin dependent diabetes mellitis multiple sclerosis; Reynaud's syndrome; autoimmune thyroiditis; allergic encephalomyelitis; Sjorgen's syndrome; juvenile onset diabetes; and immune responses associated with acute and delayed hypersensitivity mediated by cytokines and T-lymphocytes typically found in tuberculosis, sarcoidosis, polymyositis, granulomatosis and vasculitis; pernicious anemia (Addison's disease); diseases involving leukocyte diapedesis; central nervous system (CNS) inflammatory disorder; multiple organ injury syndrome; hemolytic anemia (including, but not limited to cryoglobinemia or Coombs positive anemia); myasthenia gravis; antigen-antibody complex mediated diseases; anti-glomerular basement membrane disease; antiphospholipid syndrome; allergic neuritis; Graves' disease; Lambert-Eaton myasthenic syndrome; pemphigoid bullous; pemphigus; autoimmune polyen
  • Antibodies directed against B cell surface antigens may also be used to block an immune response to a foreign antigen.
  • foreign antigen here is meant a molecule or molecules which is/are not endogenous or native to a mammal which is exposed to it.
  • the foreign antigen may elicit an immune response, e.g. a humoral and/or T cell mediated response in the mammal. Generally, the foreign antigen will provoke the production of antibodies thereagainst.
  • foreign antigens contemplated herein include immunogenic therapeutic agents, e.g. proteins such as antibodies, particularly antibodies comprising non-human amino acid residues (e.g.
  • rodent, chimeric/humanized, and primatized antibodies include toxins (optionally conjugated to a targeting molecule such as an antibody, wherein the targeting molecule may also be immunogenic); gene therapy viral vectors, such as retroviruses and adenoviruses; grafts; infectious agents (e.g. bacteria and virus); alloantigens (i.e. an antigen that occurs in some, but not in other members of the same species) such as differences in blood types, human lymphocyte antigens (HLA), platelet antigens, antigens expressed on transplanted organs, blood components, pregnancy (Rh), and hemophilic factors (e.g. Factor VIII and Factor IX).
  • HLA human lymphocyte antigens
  • platelet antigens antigens expressed on transplanted organs
  • blood components blood components
  • pregnancy (Rh) pregnancy
  • hemophilic factors e.g. Factor VIII and Factor IX
  • the anti-B cell surface antigen antibody may also be used to desenzitize a mammal awaiting transplantation.
  • Antibodies directed against a receptor in the TNF receptor superfamily may be employed to activate or stimulate apoptosis in cancer cells.
  • an immunoconjugate comprising the antibody conjugated with a cytotoxic agent is administered to the patient.
  • the immunoconjugate and/or antigen to which it is bound is/are internalized by the cell, resulting in increased therapeutic efficacy of the immunoconjugate in killing the cancer cell to which it binds.
  • the cytotoxic agent targets or interferes with nucleic acid in the cancer cell. Examples of such cytotoxic agents include any of the chemotherapeutic agents noted herein (such as a maytansinoid or a calicheamicin), a radioactive isotope, or a ribonuclease or a DNA endonuclease.
  • the multivalent antibody may also be used for ADEPT.
  • the present application contemplates combining the multivalent antibody (or immunoconjugate thereof) with one or more other therapeutic agent(s), especially for treating cancer.
  • the multivalent antibody may be co-administered with another multivalent antibody (or multivalent antibodies), a monovalent or bivalent antibody (or antibodies), chemotherapeutic agent(s) (including cocktails of chemotherapeutic agents), other cytotoxic agent(s), anti-angiogenic agent(s), cytokines, and/or growth inhibitory agent(s).
  • the multivalent antibody induces apoptosis
  • it may be particularly desirable to combine the multivalent antibody with one or more other therapeutic agent(s) which also induce apoptosis.
  • pro-apoptotic antibodies e.g.
  • bivalent or multivalent antibodies directed against B cell surface antigens may be combined with (1) pro-apoptotic antibodies (e.g. bivalent or multivalent antibodies directed against a receptor in the TNF receptor superfamily, such as anti-DR4 or anti-DR5 antibodies) or (2) with cytokines in the TNF family of cytokines (e.g. Apo2L).
  • pro-apoptotic antibodies e.g. bivalent or multivalent antibodies directed against a receptor in the TNF receptor superfamily, such as anti-DR4 or anti-DR5 antibodies
  • cytokines in the TNF family of cytokines e.g. Apo2L
  • anti-ErbB antibodies e.g. HERCEPTIN® anti-HER2 antibody
  • the patient may receive combined radiation therapy (e.g.
  • Such combined therapies noted above include combined administration (where the two or more agents are included in the same or separate formulations), and separate administration, in which case, administration of the multivalent antibody can occur prior to, and/or following, administration of the adjunct therapy or therapies.
  • the multivalent antibody (and adjunct therapeutic agent) is/are administered by any suitable means, including parenteral, subcutaneous, intraperitoneal, intrapulmonary, and intranasal, and, if desired for local treatment, intralesional administration.
  • Parenteral infusions include intramuscular, intravenous, intraarterial, intraperitoneal, or subcutaneous administration.
  • the multivalent antibody is suitably administered by pulse infusion, particularly with declining doses of the multivalent antibody.
  • the dosing is given by injections, most preferably intravenous or subcutaneous injections, depending in part on whether the administration is brief or chronic.
  • the present application contemplates administration of the antibody by gene therapy.
  • administration of nucleic acid encoding the antibody is encompassed by the expression “administering a therapeutically effective amount of an antibody”. See, for example, WO96/07321 published Mar. 14, 1996 concerning the use of gene therapy to generate intracellular antibodies.
  • nucleic acid (optionally contained in a vector) into the patient's cells; in vivo and ex vivo.
  • in vivo delivery the nucleic acid is injected directly into the patient, usually at the site where the antibody is required.
  • ex vivo treatment the patient's cells are removed, the nucleic acid is introduced into these isolated cells and the modified cells are administered to the patient either directly or, for example, encapsulated within porous membranes which are implanted into the patient (see, e.g. U.S. Pat. Nos. 4,892,538 and 5,283,187).
  • porous membranes which are implanted into the patient
  • the techniques vary depending upon whether the nucleic acid is transferred into cultured cells in vitro, or in vivo in the cells of the intended host.
  • Techniques suitable for the transfer of nucleic acid into mammalian cells in vitro include the use of liposomes, electroporation, microinjection, cell fusion, DEAE-dextran, the calcium phosphate precipitation method, etc.
  • a commonly used vector for ex vivo delivery of the gene is a retrovirus.
  • the currently preferred in vivo nucleic acid transfer techniques include transfection with viral vectors (such as adenovirus, Herpes simplex I virus, or adeno-associated virus) and lipid-based systems (useful lipids for lipid-mediated transfer of the gene are DOTMA, DOPE and DC-Chol, for example).
  • viral vectors such as adenovirus, Herpes simplex I virus, or adeno-associated virus
  • lipid-based systems useful lipids for lipid-mediated transfer of the gene are DOTMA, DOPE and DC-Chol, for example.
  • an agent that targets the target cells such as an antibody specific for a cell surface membrane protein or the target cell, a ligand for a receptor on the target cell, etc.
  • proteins which bind to a cell surface membrane protein associated with endocytosis may be used for targeting and/or to facilitate uptake, e.g.
  • capsid proteins or fragments thereof tropic for a particular cell type antibodies for proteins which undergo internalization in cycling, and proteins that target intracellular localization and enhance intracellular half-life.
  • the technique of receptor-mediated endocytosis is described, for example, by Wu et al., J. Biol. Chem. 262:4429-4432 (1987); and Wagner et al., Proc. Natl. Acad. Sci. USA 87:3410-3414 (1990).
  • Wu et al. J. Biol. Chem. 262:4429-4432 (1987); and Wagner et al., Proc. Natl. Acad. Sci. USA 87:3410-3414 (1990).
  • the appropriate dosage of multivalent antibody will depend on the type of disease to be treated, the severity and course of the disease, whether the multivalent antibody is administered for preventive or therapeutic purposes, previous therapy, the patient's clinical history and response to the multivalent antibody, and the discretion of the attending physician.
  • the multivalent antibody is suitably administered to the patient at one time or over a series of treatments.
  • ⁇ g/kg to 15 mg/kg (e.g., 0.1-20 mg/kg) of multivalent antibody is an initial candidate dosage for administration to the patient, whether, for example, by one or more separate administrations, or by continuous infusion.
  • a typical daily dosage might range from about 1 ⁇ g/kg to 100 mg/kg or more, depending on the factors mentioned above.
  • the treatment is sustained until a desired suppression of disease symptoms occurs.
  • other dosage regimens may be useful. The progress of this therapy is easily monitored by conventional techniques and assays.
  • the multivalent antibody composition will be formulated, dosed, and administered in a fashion consistent with good medical practice. Factors for consideration in this context include the particular disorder being treated, the particular mammal being treated, the clinical condition of the individual patient, the cause of the disorder, the site of delivery of the agent, the method of administration, the scheduling of administration, and other factors known to medical practitioners.
  • the “therapeutically effective amount” of the multivalent antibody to be administered will be governed by such considerations, and is the minimum amount necessary to prevent, ameliorate, or treat a disease or disorder.
  • the multivalent antibody need not be, but is optionally formulated with one or more agents currently used to prevent or treat the disorder in question.
  • the effective amount of such other agents depends on the amount of multivalent antibody present in the formulation, the type of disorder or treatment, and other factors discussed above. These are generally used in the same dosages and with administration routes as used hereinbefore or about from 1 to 99% of the heretofore employed dosages.
  • an article of manufacture containing materials useful for the treatment of the disorders described above comprises a container and a label or package insert on or associated with the container.
  • Suitable containers include, for example, bottles, vials, syringes, etc.
  • the containers may be formed from a variety of materials such as glass or plastic.
  • the container holds a composition which is effective for treating the condition and may have a sterile access port (for example the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle).
  • At least one active agent in the composition is a multivalent antibody.
  • the label or package insert indicates that the composition is used for treating the condition of choice, such as cancer.
  • the article of manufacture may comprise (a) a first container with a composition contained therein, wherein the composition comprises a multivalent antibody; and (b) a second container with a composition contained therein, wherein the composition comprises a further cytotoxic agent.
  • the article of manufacture in this embodiment of the invention may further comprises a package insert indicating that the first and second antibody compositions can be used to treat cancer.
  • the article of manufacture may further comprise a second (or third) container comprising a pharmaceutically-acceptable buffer, such as bacteriostatic water for injection (BWFI), phosphate-buffered saline, Ringer's solution and dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, and syringes.
  • BWFI bacteriostatic water for injection
  • phosphate-buffered saline such as bacteriostatic water for injection (BWFI), phosphate-buffered saline,
  • FIG. 5 The construct used to generate a tetravalent anti-HER2 antibody, called an “Octopus antibody” (OctHER2), is illustrated in FIG. 5 herein.
  • the backbone of this Octopus antibody is the recombinant, humanized monoclonal antibody 4D5 variant 8 (rhuMAb 4D5-8) (U.S. Pat. No. 5,821,337, Carter et al., expressly incorporated herein by reference).
  • the heavy chain of rhuMAb 4D5-8 was subcloned into the pRK5 vector (EP 307,247, published Mar. 15, 1989).
  • the VH-CH1 region of the heavy chain was removed by mutagenesis, and three unique restriction sites (BamHI; NheI; BspEI) were inserted. These sites were incorporated into PCR primers designed to amplify the VH-CH1 region from different antibodies. The resulting fragments were subcloned into the vector to create the Octopus heavy chain. Co-expression of the Octopus heavy chain with the appropriate light chain in a pRK5 vector in mammalian cell transfections results in the completed Octopus antibody (FIG. 4).
  • Octopus constructs containing flexible linkers inserted between the tandem Fd regions were also engineered. Through mutagenesis, DNA encoding either “gly-ser” (flex 1 linker) or “gly-ser-gly-ser” (SEQ ID NO:10) (flex 2 linker) was inserted between the DNA encoding the VH-CH1 regions of the heavy chain.
  • OctHER2 was expressed in transiently transfected 293 cells (Graham et al., J. Gen. Virol. 36:59-72 (1977)) and purified over a Protein A sepharose column.
  • the complete antibody is approximately 245 kDa, as compared to the 150 kDa molecular weight of the parent antibody.
  • the Octopus heavy chain is 75 kDa (without carbohydrate), and the light chain is 30 kDa.
  • FIGS. 6 A-C The ELISA results for OctHER2, bivalent human IgG1 anti-HER2 antibody rhuMAb 4D5-8 expressed by 293 cells, or bivalent anti-HER2 antibody HERCEPTIN®) (commercially available from Genentech, Inc., South San Francisco, USA), are shown in FIGS. 6 A-C.
  • OctHER2 binds the HER2 ECD similar to HERCEPTIN® when analyzed in an ELISA assay.
  • the rhuMAb 4D5-8 expressed by 293 cells binds identically to the vialed HERCEPTIN® (produced by Chinese Hamster Ovary (CHO) cells), indicating that 293 cells do not substantially alter the antigen binding capability of the antibodies.
  • OctHER2 was compared to HERCEPTIN® in functional assays measuring growth inhibition of HER2 overexpressing tumor cell lines.
  • the flexible linker Octopus constructs (OctHER2flexl, OctHER2flex2) inhibited cell growth more effectively than HERCEPTIN®).
  • the antibody was radioiodinated, and incubated for varying times with the cells. This was followed by measurements of the amount of intact, unbound antibody in the supernatant, the amount bound to the cell surface, the amount internalized, and finally, the amount catabolized and degraded.
  • DR5 a member of the TNF receptor superfamily that binds the trimeric Apo2L/TRAIL (Apo2L). After Apo2L receptors bind Apo2L and are clustered, death domains in the cytosolic region of the receptors induce caspases to trigger cellular apoptosis.
  • anti-DR5 Octopus constructs Two versions of anti-DR5 Octopus constructs were made: one from 16E2, an anti-DR5 cloned from a single-chain human Fv phage library (see WO98/51793, expressly incorporated herein by reference); the second anti-DR5 Octopus antibody was made from Mab 3H3.14.5 (the “3H3” antibody; ATCC HB-12534, WO99/64461), a murine anti-DR5 MAb that induces apoptosis when it is crosslinked. Since anti-Death receptor monoclonal antibodies may require crosslinking to trigger apoptosis, they are candidates for the Octopus antibody construct.
  • the anti-DR5 Octopus antibodies were prepared by replacing the variable domains of the OctHER2 construct described above with the VL and VH domains from 16E2 or 3H3.
  • the anti-DR5 Octopus antibodies were analyzed in apoptosis assays using either crystal violet or alamarBlue staining. Briefly, serial dilutions of the Octopus antibody or Apo2L were added to the media of plated cells which were then allowed to continue growing for 24 hours. After this time, the media was either removed and the cells were stained with crystal violet, or alamarBlue was added to the media and incubated briefly with the cells. Crystal violet stains the cells, whereas alamarBlue detects metabolic activity in the culture media, thus these dyes allow for measurement of cells that survive treatment. Staining by both colorimetric dyes, crystal violet and alamarBlue, was quantitated by spectrophotometry.
  • the 16E2 Octopus surprisingly, induces apoptosis with comparable potency to Apo2L in lung (SK-MES-1; HOP 92) and colon (HCT116; COLO 205) tumor cell lines, however does not cause apoptosis on normal control cell line (HUMEC).
  • HUMEC normal control cell line
  • the anti-DR5 16E2 Octopus was also effective in vivo in inducing apoptosis and shrinking a colon tumor, human COL0205, in athymic nude mice. As shown in FIGS. 13 A-D, histology slides of tumor tissues stained with hematoxylin and eosin from mice treated with the 16E2 Octopus or Apo2L induced similar levels of apoptotic cells.
  • mice also demonstrated significant decrease in tumor volume, similar to that measured for the Apo2L and two bivalent anti-DR5 mAbs, 16E2 and 3H3, as shown in FIG. 14. Mice that did not receive any anti-DR5 antibodies or Apo2L (Vehicle) showed dramatic increase in their tumor volume due to uncontrolled growth.
  • FIG. 16 demonstrates that another anti-DR5 Octopus, 3H3 Octopus, is capable of inducing apoptosis similar to the 16E2 Octopus. Additionally, FIG. 16 shows that the apoptotic activity of the Octopus antibody is not lot dependent, as several 16E2 Octopus antibodies prepared on different dates retain similar function.
  • the apoptotic activity of both the 16E2 and 3H3 Octopus antibodies is better than Apo2L on a lung tumor cell line, SK-MES-1 (FIG. 17A), and a T cell tumor line, Jurkat (FIG. 17B).
  • the anti-DR5 Octopus antibodies may be more effective at clustering DR5 on the tumor cell surface than Apo2L.
  • FIGS. 18 A-C depict the 2-day dose response curves showing the effects of the 16E2 Octopus and Apo2L on the growth of several human leukemia, non-small cell lung cancer, colon cancer, central nervous system (CNS) cancer, melanoma, ovarian cancer, renal cancer, prostate cancer and breast cancer tumor cell lines, while FIGS. 19 A-C show dose response curves from the 6 day screens.
  • FIGS. 20A and B 2-day results
  • FIGS. 21A and B 6-day results
  • 16E2 Octopus may be effective against more types of cancer than previously observed.
  • RITUXAN® alone does not trigger much apoptosis of a non-Hodgkins lymphoma B cell line, Wil-2, unless it is crosslinked with anti-human IgG (RITUXAN®-IgG).
  • OctCD20 is capable of inducing apoptosis in Wil-2 cells independent of crosslinking.
  • the level of apoptosis observed with OctCD20 is lower than that of crosslinked RITUXAN®, however, suggesting that the apoptotic activity of OctCD20 could be improved, perhaps through the use of the flexible linkers.
  • FIG. 23C Versions of the Octopus antibodies of Example 2 (anti-HER2), Example 3 (anti-DR5) and Example 4 (anti-CD20) with an antibody hing region dimerization domain (designated “Octopus F(ab′) 2 ” herein) were engineered.
  • the anti-HER2 Octopus F(ab′) 2 construct was engineered by replacing the Fc region of the heavy chain cDNA with sequence encoding a leucine-zipper motif which, when expressed as protein, dimerizes to effectively join the Octopus Fab arms (FIG. 23C).
  • the octopus F(ab′) 2 can maintain the leucine zipper motif, or that motif can, e.g., be proteolytically removed as desired.
  • PCR was used to amplify the duplicate VH/CH1 domains and to insert a restriction site onto the end of the Octopus heavy chain cDNA (Notl) to permit in-frame subcloning into a vector (VG15) containing a leucine-zipper motif. PCR was again utilized to add another restriction site downstream of the heavy chain termination codon (Xhol) to allow subcloning into the pRK vector for expression in mammalian cells.
  • the VH/CH1 domains of anti-DR5 Mab16E2 and anti-CD20 Mab C2B8 were substituted into the Oct F(ab)′ 2 heavy chain backbone using the unique restriction sites BamHI, NheI, and BspEI.
  • POPoctopus antibodies were created by linking together Fab domains in tandem repeats to form linear Fab multimers.
  • POPoct-3 contains three linked Fab domains (FIG. 23D), while “POPoct-4” has four Fab repeats (FIG. 23E).
  • Anti-HER2 (rhuMab 4D5), anti-DR5 (16E2), and anti-CD20 (C2B8) POPoct-3 constructs were generated, as were anti-HER2 (rhuMab 4D5) and anti-DR5 (16E2) POPoct-4 constructs.
  • POPoct-3 antibodies were engineered both with and without flex 1 linkers.
  • FIG. 25 depicts the construction of the POPoct-3 heavy chain cDNA.
  • PCR was used to amplify the VH/CH1 domain adding a 5′-BspEI site and a 3′-Notl site. This sequence was digested and along with BamHI/BspEI digested Octopus heavy chain, ligated into a pRK vector to yield an Octopus heavy chain containing sequence for three VH/CH1 domains.
  • the BspEI site encodes for a serine and a glycine residue.
  • the different Octopus heavy chains were transiently cotransfected with the appropriate light chain cDNAs into 293 mammalian cells to express antibodies containing either three Fab domains (POPoct-3 Fab) or four Fab domains (full-length Octopus; Octopus F(ab)′ 2 ; POPoct-4 Fab). While native IgG Mabs and full-length Octopus antibodies were purified over Protein A sepharose, Octopus F(ab)′ 2 and POPoct-3 and-4 were purified over Protein G sepharose columns.
  • the Octopus F(ab)′ 2 is approximately 200 kDa (FIG. 23F, lane 4), smaller than the 240 kDa of the full-length Octopus antibody (FIG. 23F, lane 3), but larger than the 150 kDa native IgG Mab (FIG. 23F, lanes 1 and 2).
  • POPoct-3 is slightly smaller than native IgG Mab
  • POPoct-4 is slightly larger at 190 kDa.
  • the heavy chain of the Octopus F(ab)′ 2 (FIG. 23G, lane 4) is approximately the same size as the native IgG Mab heavy chain (FIG.
  • the POPoct-3 heavy chain (FIG. 23G, lane 5) is similar in size to the full-length Octopus heavy chain (FIG. 23G, lane 3), while at approximately 97 kDa the POPoct-4 has the largest heavy chain
  • POPoct-3HER2 was compared to OctHER and HERCEPTIN® in internalization assays on two 3+ HER2 over-expressing tumor cell lines, SKBR3 and BT474, to assess its candidacy for applications in immunotoxin therapies. Although structurally different than the full-length OctHER2 antibody, POPoct-3HER2 was internalized and catabolized identically to OctHER2 by both cell lines (FIGS. 29A and B) and at twice the rate of HERCEPTIN®.
  • OctDR5flex-1>OctDR5 POPoct-4
  • DR5 POPoct-3flex-1
  • DR5 POPoct-3DR5>OctDR5F(ab)′ 2 >16E2 MAb.
  • OctDR5flex-1 showed increased potency compared to OctDR5, especially at lower concentrations (FIG. 30A), indicating that flexibility between the Fab arms improves efficacy.
  • POPoct-3flex-1 DR5 induced equivalent levels of apoptosis as OctHER (FIG. 30A) and showed similar efficacy to POPoct16-3 and POPoct16-4 (FIG. 30B).
  • Apo2L binds to the death receptors and triggers cellular apoptosis through the caspase signaling pathway.
  • the anti-DR5 Octopus antibodies were shown to induce apoptosis through the same signaling pathway as Apo2L.
  • Oct16E2 triggered similar levels of apoptosis as APO2L on the lung tumor cell line SK-MES-1 (FIG. 31A, dashed lines), but after the addition of ZVAD, an inhibitor of caspase 3 and 9, cellular apoptosis triggered by both Apo2L and Oct16E2 was inhibited (FIG. 31B solid lines).
  • FIG. 31B BJAB cells, a B-cell lymphoma line that expresses DR5, was incubated at two different concentrations of two anti-DR5 Octopus antibodies, Oct16E2 and Oct3H3, for varying times. Purification of the antibody-DR5 complexes was followed Western blot analysis to identify the signaling molecules that copurified with the complexes. As with Apo2L, the signaling molecules caspase 8 and FADD associated with DR5 after the receptor was bound by both Oct16E2 and Oct3H3 (FIG. 31B).
  • RITUXAN® did not efficiently trigger apoptosis in vitro on the B-cell lymphoma cell line WIL-2 unless first crosslinked by anti-IgG antibody.
  • OctCD20 was capable of inducing apoptosis of WIL-2 cells independent of crosslinking, at levels higher than RITUXAN® alone, yet slightly lower than anti-IgG-crosslinked RITUXAN®.
  • When crosslinked with anti-IgG antibody OctCD20 induced more apoptosis of the WIL-2 cells than crosslinked RITUXAN® (FIG. 32). Since one potential explanation for the efficacy of RITUXAN® in vivo is that the antibody is being crosslinked by either complement or Fc ⁇ R bearing cells, this observation suggests that OctCD20 will be even more efficacious in vivo.
  • OctCD20 F(ab)′ 2 , POPoct-3CD20 and POPoct-3CD20flex-1 were tested at various concentrations in apoptosis assays with WIL-2 cells, and the optimal doses are shown in the maximum response curves in FIG. 33.
  • the Octopus antibodies were compared to the anti-CD20 antibody 1 F5 (Clark et al. supra), which functions similar to RITUXAN® in that it does not induce apoptosis unless crosslinked with anti-IgG antibody.
  • Octopus antibodies tested induced either similar (OctCD20 F(ab)′ 2 ) or higher (POPoct-3CD20, POPoct-3CD20flex-1) levels of apoptosis than crosslinked IF5 anti-CD20. Additionally, the Octopus antibodies were efficacious at considerably lower concentrations than the crosslinked anti-CD20.
  • Apoptosis induction by the various anti-CD20 antibodies was further assessed using blood from a patient with chronic lymphocytic leukemia (CLL). PBL's were separated out using dextran sedimentation, washed and plated in serum-free lymphocyte medium treated overnight with no sample, 1F5 (20 ⁇ g/ml), 1F5+ cross-linking mouse anti-IgG (100 ⁇ g/ml), OctCD20 F(ab′) 2 at approx 0.5 or 1.0 ⁇ g/ml and POPoct-3 CD20 at 0.5 ⁇ g/ml.
  • CLL chronic lymphocytic leukemia
  • OctCD20 was also evaluated as a candidate for immunotoxin therapy in internalization assays on three B-cell lymphoma lines, DB, WIL-2, and Ramos, and compared to RITUXAN®. As shown in FIG. 35, twice as much OctCD20 was internalized by the cells as compared to RITUXAN®, which was not internalized by the cells at appreciable levels. The higher avidity that would be expected for the multivalent antibodies due to the increased number of binding sites is evident in the fact that more OctCD20 remains bound to the cell surface of the cells over time as compared to RITUXAN®.
US09/813,341 2000-04-11 2001-03-20 Multivalent antibodies and uses therefor Abandoned US20020004587A1 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
US09/813,341 US20020004587A1 (en) 2000-04-11 2001-03-20 Multivalent antibodies and uses therefor
US11/218,821 US20060025576A1 (en) 2000-04-11 2005-09-02 Multivalent antibodies and uses therefor
US11/535,031 US20080299120A1 (en) 2000-04-11 2006-09-25 Multivalent antibodies and uses therefor
US12/939,117 US20110110852A1 (en) 2000-04-11 2010-11-03 Multivalent Antibodies and Uses Therefor
US13/470,189 US8722859B2 (en) 2000-04-11 2012-05-11 Multivalent antibodies and uses therefor
US14/229,683 US9493579B2 (en) 2000-04-11 2014-03-28 Multivalent antibodies and uses therefor

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US19581900P 2000-04-11 2000-04-11
US09/813,341 US20020004587A1 (en) 2000-04-11 2001-03-20 Multivalent antibodies and uses therefor

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US11/218,821 Continuation US20060025576A1 (en) 2000-04-11 2005-09-02 Multivalent antibodies and uses therefor

Publications (1)

Publication Number Publication Date
US20020004587A1 true US20020004587A1 (en) 2002-01-10

Family

ID=22722944

Family Applications (6)

Application Number Title Priority Date Filing Date
US09/813,341 Abandoned US20020004587A1 (en) 2000-04-11 2001-03-20 Multivalent antibodies and uses therefor
US11/218,821 Abandoned US20060025576A1 (en) 2000-04-11 2005-09-02 Multivalent antibodies and uses therefor
US11/535,031 Abandoned US20080299120A1 (en) 2000-04-11 2006-09-25 Multivalent antibodies and uses therefor
US12/939,117 Abandoned US20110110852A1 (en) 2000-04-11 2010-11-03 Multivalent Antibodies and Uses Therefor
US13/470,189 Expired - Fee Related US8722859B2 (en) 2000-04-11 2012-05-11 Multivalent antibodies and uses therefor
US14/229,683 Expired - Lifetime US9493579B2 (en) 2000-04-11 2014-03-28 Multivalent antibodies and uses therefor

Family Applications After (5)

Application Number Title Priority Date Filing Date
US11/218,821 Abandoned US20060025576A1 (en) 2000-04-11 2005-09-02 Multivalent antibodies and uses therefor
US11/535,031 Abandoned US20080299120A1 (en) 2000-04-11 2006-09-25 Multivalent antibodies and uses therefor
US12/939,117 Abandoned US20110110852A1 (en) 2000-04-11 2010-11-03 Multivalent Antibodies and Uses Therefor
US13/470,189 Expired - Fee Related US8722859B2 (en) 2000-04-11 2012-05-11 Multivalent antibodies and uses therefor
US14/229,683 Expired - Lifetime US9493579B2 (en) 2000-04-11 2014-03-28 Multivalent antibodies and uses therefor

Country Status (22)

Country Link
US (6) US20020004587A1 (el)
EP (2) EP2857516B1 (el)
JP (1) JP2003531588A (el)
KR (1) KR20020093029A (el)
CN (2) CN101289511A (el)
AU (2) AU4761601A (el)
BR (1) BR0110610A (el)
CA (1) CA2403425C (el)
CY (1) CY1119291T1 (el)
DK (2) DK1272647T3 (el)
ES (2) ES2637801T3 (el)
HK (1) HK1208705A1 (el)
HU (1) HUP0300369A2 (el)
IL (3) IL151853A0 (el)
LT (1) LT2857516T (el)
MX (1) MXPA02010011A (el)
NZ (1) NZ521540A (el)
PL (1) PL357939A1 (el)
PT (1) PT2857516T (el)
SI (1) SI2857516T1 (el)
WO (1) WO2001077342A1 (el)
ZA (1) ZA200207589B (el)

Cited By (374)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020058029A1 (en) * 2000-09-18 2002-05-16 Nabil Hanna Combination therapy for treatment of autoimmune diseases using B cell depleting/immunoregulatory antibody combination
US20030091561A1 (en) * 2001-06-13 2003-05-15 Genmab A/S Human monoclonal antibodies to epidermal growth factor receptor (EGFR)
US20030118592A1 (en) * 2001-01-17 2003-06-26 Genecraft, Inc. Binding domain-immunoglobulin fusion proteins
US20030180296A1 (en) * 2001-12-20 2003-09-25 Theodora Salcedo Antibodies that immunospecifically bind to trail receptors
US20030194403A1 (en) * 2001-06-13 2003-10-16 Genmab, Inc. Human monoclonal antibodies to epidermal growth factor receptor (EGFR)
US20030219433A1 (en) * 2002-02-14 2003-11-27 Immunomedics, Inc. Anti-CD20 antibodies and fusion proteins thereof and methods of use
US20030219476A1 (en) * 2000-10-16 2003-11-27 Neopharm, Inc. Liposomal formulation of mitoxantrone
US20030228309A1 (en) * 2000-11-08 2003-12-11 Theodora Salcedo Antibodies that immunospecifically bind to TRAIL receptors
US20040002587A1 (en) * 2002-02-20 2004-01-01 Watkins Jeffry D. Fc region variants
WO2004039329A2 (en) * 2002-10-31 2004-05-13 Biogen Idec Ma Inc. Treatment of immunological renal disorders by lymphotoxin pathway inhibitors
US20040132101A1 (en) * 2002-09-27 2004-07-08 Xencor Optimized Fc variants and methods for their generation
WO2004011611A3 (en) * 2002-07-25 2004-08-12 Genentech Inc Taci antibodies and uses thereof
US20040202658A1 (en) * 2003-04-09 2004-10-14 Genentech, Inc. Therapy of autoimmune disease in a patient with an inadequate response to TNF-alpha inhibitor
WO2004029207A3 (en) * 2002-09-27 2004-10-28 Xencor Inc Optimized fc variants and methods for their generation
US20040219643A1 (en) * 2001-06-28 2004-11-04 Greg Winter Dual-specific ligand
US20050032130A1 (en) * 2003-07-29 2005-02-10 Genentech, Inc. Neutralizing antibody assay and uses therefor
US20050053602A1 (en) * 2003-08-29 2005-03-10 Genentech, Inc. Therapy of ocular disorders
US20050064514A1 (en) * 2003-01-09 2005-03-24 Macrogenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
US20050069552A1 (en) * 2003-07-28 2005-03-31 Bleck Gregory T. Fusion antibodies
US20050070689A1 (en) * 2001-08-03 2005-03-31 Genentech, Inc. Taci and br3 polypeptides and uses thereof
US20050095243A1 (en) * 2003-06-05 2005-05-05 Genentech, Inc. Combination therapy for B cell disorders
US20050100548A1 (en) * 2001-07-24 2005-05-12 Biogen Idec Ma Inc. BAFF, inhibitors thereof and their use in the modulation of B-cell response
WO2004101756A3 (en) * 2003-05-09 2005-06-09 Diadexus Inc Ovr110 antibody compositions and methods of use
US20050129616A1 (en) * 2001-05-25 2005-06-16 Human Genome Sciences, Inc. Antibodies that immunospecifically bind to TRAIL receptors
US20050163775A1 (en) * 2003-06-05 2005-07-28 Genentech, Inc. Combination therapy for B cell disorders
US20050186206A1 (en) * 2003-12-19 2005-08-25 Genentech, Inc. Detection of CD20 in therapy of autoimmune diseases
US20050191297A1 (en) * 2003-12-19 2005-09-01 Genentech, Inc. Detection of CD20 in transplant rejection
US20050202028A1 (en) * 2001-01-17 2005-09-15 Trubion Pharmaceuticals, Inc. Binding domain-immunoglobulin fusion proteins
US20050214209A1 (en) * 2001-05-25 2005-09-29 Human Genome Sciences, Inc. Antibodies that immunospecifically bind to TRAIL receptors
US20050214286A1 (en) * 2004-01-27 2005-09-29 University Of Southern California Polymer-bound antibody cancer therapeutic agent
US20050244411A1 (en) * 1999-01-25 2005-11-03 Biogen Idec Ma Inc. BAFF, inhibitors thereof and their use in the modulation of B-cell response and treatment of autoimmune disorders
US20050244403A1 (en) * 2004-03-24 2005-11-03 Xencor, Inc. Immunoglobulin variants outside the Fc region
US20050249723A1 (en) * 2003-12-22 2005-11-10 Xencor, Inc. Fc polypeptides with novel Fc ligand binding sites
US20050271658A1 (en) * 2004-05-05 2005-12-08 Genentech, Inc. Preventing autoimmune disease
US20050271663A1 (en) * 2001-06-28 2005-12-08 Domantis Limited Compositions and methods for treating inflammatory disorders
US20060024295A1 (en) * 2004-06-04 2006-02-02 Genentech, Inc. Method for treating lupus
US20060034835A1 (en) * 2002-12-16 2006-02-16 Genentech, Inc. Immunoglobulin variants and uses thereof
WO2006017574A1 (en) * 2004-08-03 2006-02-16 Mayo Foundation For Medical Education And Research Improving treatments
US20060051345A1 (en) * 2004-06-04 2006-03-09 Genentech, Inc. Method for treating multiple sclerosis
US20060062787A1 (en) * 2004-07-22 2006-03-23 Genentech, Inc. Method for treating Sjogren's syndrome
US20060062786A1 (en) * 2000-11-08 2006-03-23 Human Genome Sciences, Inc. Antibodies that immunospecifically bind to TRAIL receptors
US20060074225A1 (en) * 2004-09-14 2006-04-06 Xencor, Inc. Monomeric immunoglobulin Fc domains
US20060073146A1 (en) * 2000-02-16 2006-04-06 Genentech, Inc. Uses of agonists and antagonists to modulate activity of TNF-related molecules
WO2006041680A2 (en) 2004-10-05 2006-04-20 Genentech, Inc. Method for treating vasculitis
US20060106203A1 (en) * 2002-06-28 2006-05-18 Domantis Limited Ligand
US20060121042A1 (en) * 2004-10-27 2006-06-08 Medimmune, Inc. Modulation of antibody specificity by tailoring the affinity to cognate antigens
US20060173170A1 (en) * 2004-11-12 2006-08-03 Xencor, Inc. Fc variants with altered binding to FcRn
US20060188495A1 (en) * 2005-01-13 2006-08-24 Genentech, Inc. Treatment method
US20060204505A1 (en) * 2005-03-08 2006-09-14 Sliwkowski Mark X Methods for identifying tumors responsive to treatment with HER dimerization inhibitors (HDIs)
US20060235208A1 (en) * 2002-09-27 2006-10-19 Xencor, Inc. Fc variants with optimized properties
US20060233797A1 (en) * 2005-04-15 2006-10-19 Genentech, Inc. Treatment of inflammatory bowel disease (IBD)
US20060246004A1 (en) * 2005-02-07 2006-11-02 Genentech, Inc. Antibody variants and uses thereof
US20060257406A1 (en) * 2002-12-27 2006-11-16 Domantis Limited Ligand
US20060263349A1 (en) * 2005-05-20 2006-11-23 Genentech, Inc. Pretreatment of a biological sample from an autoimmune disease subject
US20060263355A1 (en) * 2005-02-28 2006-11-23 Joanne Quan Treatment of bone disorders
US20060270837A1 (en) * 2001-05-25 2006-11-30 Human Genome Sciences, Inc. Antibodies that immunospecifically bind to TRAIL receptors
US20060275282A1 (en) * 2005-01-12 2006-12-07 Xencor, Inc. Antibodies and Fc fusion proteins with altered immunogenicity
US20070003546A1 (en) * 2002-03-01 2007-01-04 Xencor, Inc. Optimized Fc variants and methods for their generation
US20070036799A1 (en) * 2005-08-10 2007-02-15 Macrogenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
US20070059793A1 (en) * 2003-03-19 2007-03-15 Biogen Idec Ma Inc. Nogo receptor binding protein
US20070059306A1 (en) * 2005-07-25 2007-03-15 Trubion Pharmaceuticals, Inc. B-cell reduction using CD37-specific and CD20-specific binding molecules
US20070071675A1 (en) * 2005-08-19 2007-03-29 Chengbin Wu Dual variable domain immunoglobulin and uses thereof
WO2006105338A3 (en) * 2005-03-31 2007-03-29 Xencor Inc Fc VARIANTS WITH OPTIMIZED PROPERTIES
US20070135620A1 (en) * 2004-11-12 2007-06-14 Xencor, Inc. Fc variants with altered binding to FcRn
US20070148171A1 (en) * 2002-09-27 2007-06-28 Xencor, Inc. Optimized anti-CD30 antibodies
EP1810035A2 (en) * 2004-11-10 2007-07-25 Magrogenics, Inc. Engineering fc antibody regions to confer effector function
US20070212733A1 (en) * 2005-11-23 2007-09-13 Genentech, Inc. Methods and compositions related to B cell assays
WO2007025044A3 (en) * 2005-08-24 2007-09-20 Bristol Myers Squibb Co Biomarkers and methods for determining sensitivity to epidermal growth factor receptor modulators
US20070219133A1 (en) * 2002-03-01 2007-09-20 Xencor, Inc. CD52 OPTIMIZED Fc VARIANTS AND METHODS FOR THEIR GENERATION
US20070237767A1 (en) * 2003-03-03 2007-10-11 Xencor, Inc. Fc Variants Having Decreased Affinity for FcyRllla
US20070275460A1 (en) * 2003-03-03 2007-11-29 Xencor.Inc. Fc Variants With Optimized Fc Receptor Binding Properties
WO2008011081A2 (en) 2006-07-19 2008-01-24 The Trustees Of The University Of Pennsylvania Wsx-1/p28 as a target for anti-inflammatory responses
US20080044429A1 (en) * 2006-06-26 2008-02-21 Macrogenics, Inc. Fc.gamma.RIIB-Specific Antibodies and Methods of Use Thereof
US20080057056A1 (en) * 2003-03-03 2008-03-06 Xencor, Inc. Fc Variants with Increased Affinity for FcyRIIC
US7348003B2 (en) 2001-05-25 2008-03-25 Human Genome Sciences, Inc. Methods of treating cancer using antibodies that immunospecifically bind to TRAIL receptors
US20080075719A1 (en) * 2004-04-16 2008-03-27 Genentech, Inc. Method for Augmenting B Cell Depletion
US20080112961A1 (en) * 2006-10-09 2008-05-15 Macrogenics, Inc. Identification and Engineering of Antibodies with Variant Fc Regions and Methods of Using Same
US20080131435A1 (en) * 2003-01-09 2008-06-05 Macrogenics, Inc. Identification and Engineering of Antibodies With Variant Fc Regions and Methods of Using Same
US20080138349A1 (en) * 2006-12-08 2008-06-12 Macrogenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
US20080206867A1 (en) * 2005-10-03 2008-08-28 Desjarlais John R Fc variants with optimized Fc receptor binding properties
US20080226659A1 (en) * 2000-03-16 2008-09-18 Sharon Erickson Methods of treatment using anti-erbb antibody-maytansinoid conjugates
US20080254027A1 (en) * 2002-03-01 2008-10-16 Bernett Matthew J Optimized CD5 antibodies and methods of using the same
US20080260731A1 (en) * 2002-03-01 2008-10-23 Bernett Matthew J Optimized antibodies that target cd19
US20080267965A1 (en) * 2002-02-21 2008-10-30 Kalled Susan L Use of Bcma as an Immunoregulatory Agent
US20080267976A1 (en) * 2005-10-06 2008-10-30 Gregory Alan Lazar Optimized Anti-Cd30 Antibodies
US20080279850A1 (en) * 2005-07-25 2008-11-13 Trubion Pharmaceuticals, Inc. B-Cell Reduction Using CD37-Specific and CD20-Specific Binding Molecules
US20080313379A1 (en) * 2007-06-15 2008-12-18 United Memories, Inc. Multiple bus charge sharing
US20090017027A1 (en) * 2002-08-14 2009-01-15 Macrogenics, Inc. FcGammaRIIB Specific Antibodies and Methods of Use Thereof
US20090017023A1 (en) * 2002-08-14 2009-01-15 Macrogenics, Inc. FcGammaRIIB Specific Antibodies and Methods of Use Thereof
US20090028872A1 (en) * 2005-09-26 2009-01-29 Jonathan Alexander Terret Human monoclonal antibodies to cd70
US20090041770A1 (en) * 2004-11-12 2009-02-12 Chamberlain Aaron Keith Fc VARIANTS WITH ALTERED BINDING TO FcRn
US20090053240A1 (en) * 2004-10-21 2009-02-26 Xencor, Inc. Novel Immunoglobulin Insertions, Deletions and Substitutions
US20090076251A1 (en) * 2002-08-14 2009-03-19 Macrogenics, Inc. FcGammaRIIB Specific Antibodies and Methods of Use Thereof
US20090074771A1 (en) * 2002-08-14 2009-03-19 Macrogenics, Inc. FcGammaRIIB Specific Antibodies and Methods of Use Thereof
US20090092610A1 (en) * 2002-08-14 2009-04-09 Macrogenics, Inc. FcGammaRIIB Specific Antibodies and Methods of Use Thereof
US20090098124A1 (en) * 2006-03-10 2009-04-16 Macrogenics, Inc. Identification and engineering of antibodies with variant heavy chains and methods of using same
US20090155255A1 (en) * 2007-09-27 2009-06-18 Biogen Idec Ma Inc. Cd23 binding molecules and methods of use thereof
US20090155283A1 (en) * 2005-12-01 2009-06-18 Drew Philip D Noncompetitive Domain Antibody Formats That Bind Interleukin 1 Receptor Type 1
US20090163699A1 (en) * 2004-11-12 2009-06-25 Chamberlain Aaron Keith Fc VARIANTS WITH ALTERED BINDING TO FcRn
EP2075256A2 (en) 2002-01-14 2009-07-01 William Herman Multispecific binding molecules
EP2077281A1 (en) 2008-01-02 2009-07-08 Bergen Teknologioverforing AS Anti-CD20 antibodies or fragments thereof for the treatment of chronic fatigue syndrome
US20090186034A1 (en) * 2006-12-19 2009-07-23 Genetech, Inc. Gene expression markers for inflammatory bowel disease
US20090191195A1 (en) * 2006-06-26 2009-07-30 Macrogenics, Inc. Combination of FcgammaRIIB-Specific Antibodies and CD20-Specific Antibodies and Methods of Use Thereof
US20090196879A1 (en) * 2008-01-02 2009-08-06 Olav Mella B-cell depleting agents, like anti-CD20 antibodies or fragments thereof for the treatment of chronic fatigue syndrome
US20090202537A1 (en) * 2004-05-10 2009-08-13 Macrogenics, Inc. FcGammaRIIB Specific Antibodies and Methods of Use Thereof
US20090215992A1 (en) * 2005-08-19 2009-08-27 Chengbin Wu Dual variable domain immunoglobulin and uses thereof
US20090226429A1 (en) * 2001-05-25 2009-09-10 Human Genome Sciences, Inc. Antibodies That Immunospecifically Bind to TRAIL Receptors
US20090259026A1 (en) * 2002-06-28 2009-10-15 Ian Tomlinson Ligand
US20090274692A1 (en) * 2008-04-11 2009-11-05 Trubion Pharmaceuticals, Inc. Cd37 immunotherapeutic and combination with bifunctional chemotherapeutic thereof
US20090304693A1 (en) * 2008-06-03 2009-12-10 Abbott Laboratories Dual Variable Domain Immunoglobulins and Uses Thereof
US20090311253A1 (en) * 2008-06-03 2009-12-17 Abbott Laboratories Dual Variable Domain Immunoglobulins and Uses Thereof
US20100003252A1 (en) * 1999-07-12 2010-01-07 Genentech, Inc. Blocking immune response to a graft
US20100015131A1 (en) * 2008-07-09 2010-01-21 Biogen Idec Ma Inc. Composition Comprising Antibodies to LINGO or Fragments Thereof
US20100034738A1 (en) * 2008-07-21 2010-02-11 Immunomedics, Inc. Structural Variants of Antibodies for Improved Therapeutic Characteristics
US20100040541A1 (en) * 2002-02-14 2010-02-18 Immunomedics, Inc. Structural Variants of Antibodies for Improved Therapeutic Characteristics
US7696320B2 (en) 2004-08-24 2010-04-13 Domantis Limited Ligands that have binding specificity for VEGF and/or EGFR and methods of use therefor
US20100092475A1 (en) * 2007-03-15 2010-04-15 Terrance Grant Johns Treatment method using egfr antibodies and src inhibitors and related formulations
US20100104557A1 (en) * 2006-09-18 2010-04-29 Xencor, Inc. Optimized Antibodies that Target HM1.24
US20100111979A1 (en) * 2003-06-27 2010-05-06 Amgen Fremont Inc. Antibodies directed to the deletion mutants of epidermal growth factor receptor and uses thereof
US20100150950A1 (en) * 2006-12-14 2010-06-17 Medarex, Inc. Human antibodies that bind cd70 and uses thereof
US20100158903A1 (en) * 2008-09-16 2010-06-24 Craig Smith Methods for treating progressive multiple sclerosis
US20100166751A1 (en) * 2002-09-04 2010-07-01 Biopolymer Engineering (D/B/A Biothera) Cancer therapy using whole glucan particles and antibodies
US20100166744A1 (en) * 2007-01-25 2010-07-01 Wong Kwok-Kin Use of anti-egfr antibodies in treatment of egfr mutant mediated disease
WO2010075249A2 (en) 2008-12-22 2010-07-01 Genentech, Inc. A method for treating rheumatoid arthritis with b-cell antagonists
US20100233079A1 (en) * 2008-12-04 2010-09-16 Abbott Laboratories Dual Variable Domain Immunoglobulins and Uses Thereof
US20100233689A1 (en) * 2007-03-22 2010-09-16 The General Hospital Corporation Pyrazoloanthrone and derivatives thereof for the treatment of cancer expressing 'mullerian inhibiting substance' type ii receptor (misrii) and of excess androgen states
US20100239582A1 (en) * 2007-09-26 2010-09-23 Ucb Pharma S.A. Dual Specificity Antibody Fusions
EP2233149A1 (en) 2007-10-16 2010-09-29 ZymoGenetics, Inc. Combination of BLYS inhibition and anti-CD20 agents for treatment of autoimmune disease
US20100260668A1 (en) * 2008-04-29 2010-10-14 Abbott Laboratories Dual Variable Domain Immunoglobulins and Uses Thereof
US20100266542A1 (en) * 2004-03-23 2010-10-21 Biogen Idec Ma Inc. Receptor coupling agents and therapeutic uses thereof
US7820161B1 (en) 1999-05-07 2010-10-26 Biogen Idec, Inc. Treatment of autoimmune diseases
US20100272723A1 (en) * 2006-08-14 2010-10-28 Xencor, Inc. Optimized Antibodies that Target CD19
US20100279932A1 (en) * 2003-07-26 2010-11-04 Trubion Pharmaceuticals, Inc. Binding constructs and methods for use thereof
US20100291103A1 (en) * 2007-06-06 2010-11-18 Domantis Limited Polypeptides, antibody variable domains and antagonists
US20100297121A1 (en) * 2007-10-11 2010-11-25 Biogen Idec Ma Inc. Methods for Treating Pressure Induced Optic Neuropathy, Preventing Neuronal Degeneration and Promoting Neuronal Cell Survival Via Administration of LINGO-1 Antagonists and TrkB Agonists
EP2261367A2 (en) 2007-11-29 2010-12-15 Genentech, Inc. Gene expression markers for inflammatory bowel disease
US20100322939A1 (en) * 2007-06-21 2010-12-23 Genmab A/S Novel methods for treating egfr-associated tumors
US20110008345A1 (en) * 2007-11-30 2011-01-13 Claire Ashman Antigen-binding constructs
WO2011011339A1 (en) 2009-07-20 2011-01-27 Genentech, Inc. Gene expression markers for crohn's disease
US20110033483A1 (en) * 2006-06-12 2011-02-10 Trubion Pharmaceuticals Inc. Single-chain multivalent binding proteins with effector function
WO2011019619A1 (en) 2009-08-11 2011-02-17 Genentech, Inc. Production of proteins in glutamine-free cell culture media
US20110044980A1 (en) * 2009-07-29 2011-02-24 Abbott Laboratories Dual Variable Domain Immunoglobulins and Uses Thereof
WO2011022264A1 (en) 2009-08-15 2011-02-24 Genentech, Inc. Anti-angiogenesis therapy for the treatment of previously treated breast cancer
US20110076232A1 (en) * 2009-09-29 2011-03-31 Ludwig Institute For Cancer Research Specific binding proteins and uses thereof
US7923011B2 (en) 2006-10-12 2011-04-12 Genentech, Inc. Antibodies to lymphotoxin-alpha
US20110097323A1 (en) * 2008-04-02 2011-04-28 Macrogenics, Inc. Her2/neu-Specific Antibodies and Methods of Using Same
US20110123553A1 (en) * 2007-11-08 2011-05-26 Biogen Idec Ma Inc. Use of LINGO-4 Antagonists in the Treatment of Conditions Involving Demyelination
US20110142836A1 (en) * 2009-01-02 2011-06-16 Olav Mella B-cell depleting agents for the treatment of chronic fatigue syndrome
US7981418B2 (en) 2007-03-02 2011-07-19 Genentech, Inc. Predicting response to a HER inhibitor
US20110184152A1 (en) * 2008-09-26 2011-07-28 Ucb Pharma S.A. Biological Products
WO2011100403A1 (en) 2010-02-10 2011-08-18 Immunogen, Inc Cd20 antibodies and uses thereof
WO2011106300A2 (en) 2010-02-23 2011-09-01 Genentech, Inc. Anti-angiogenesis therapy for the treatment of ovarian cancer
US20110212094A1 (en) * 2009-10-28 2011-09-01 Abbott Laboratories Dual variable domain immunoglobulins and uses thereof
EP2371388A2 (en) 2004-10-20 2011-10-05 Genentech, Inc. Antibody formulations
WO2011133886A2 (en) 2010-04-23 2011-10-27 Genentech, Inc. Production of heteromultimeric proteins
WO2011146568A1 (en) 2010-05-19 2011-11-24 Genentech, Inc. Predicting response to a her inhibitor
WO2011153243A2 (en) 2010-06-02 2011-12-08 Genentech, Inc. Anti-angiogenesis therapy for treating gastric cancer
US8084582B2 (en) 2003-03-03 2011-12-27 Xencor, Inc. Optimized anti-CD20 monoclonal antibodies having Fc variants
EP2399605A1 (en) 2005-02-23 2011-12-28 Genentech, Inc. Extending time to disease progression or survival in cancer patients
WO2012012750A1 (en) 2010-07-23 2012-01-26 Trustees Of Boston University ANTI-DEsupR INHIBITORS AS THERAPEUTICS FOR INHIBITION OF PATHOLOGICAL ANGIOGENESIS AND TUMOR CELL INVASIVENESS AND FOR MOLECULAR IMAGING AND TARGETED DELIVERY
EP2415483A1 (en) 2005-07-25 2012-02-08 Emergent Product Development Seattle, LLC Single dose use of cd20-specific binding molecules
US8128926B2 (en) 2007-01-09 2012-03-06 Biogen Idec Ma Inc. Sp35 antibodies and uses thereof
WO2012064627A2 (en) 2010-11-08 2012-05-18 Genentech, Inc. Subcutaneously administered anti-il-6 receptor antibody
US8188231B2 (en) 2002-09-27 2012-05-29 Xencor, Inc. Optimized FC variants
WO2012071436A1 (en) 2010-11-24 2012-05-31 Genentech, Inc. Method of treating autoimmune inflammatory disorders using il-23r loss-of-function mutants
WO2012075037A1 (en) 2010-11-30 2012-06-07 Genentech, Inc. Low affinity blood brain barrier receptor antibodies and uses therefor
US8216579B2 (en) 2006-05-26 2012-07-10 Macrogenics, Inc. Humanized FcγRIIB-specific antibodies and methods of use thereof
WO2012106587A1 (en) 2011-02-04 2012-08-09 Genentech, Inc. Fc VARIANTS AND METHODS FOR THEIR PRODUCTION
US20120309943A1 (en) * 2010-02-16 2012-12-06 Yoichi Kumada Antibody-immobilized carrier, method of producing antibody-immobilized carrier, and use of said antibody-immobilized carrier
US8349332B2 (en) 2005-04-06 2013-01-08 Ibc Pharmaceuticals, Inc. Multiple signaling pathways induced by hexavalent, monospecific and bispecific antibodies for enhanced toxicity to B-cell lymphomas and other diseases
EP2543384A2 (en) 2005-12-02 2013-01-09 Biogen Idec MA Inc. Treatment of conditions involving demyelination
US20130060011A1 (en) * 2011-08-23 2013-03-07 Peter Bruenker Fc-free antibodies comprising two fab fragments and methods of use
US20130078249A1 (en) * 2011-08-23 2013-03-28 Oliver Ast Bispecific t cell activating antigen binding molecules
WO2013055958A1 (en) 2011-10-11 2013-04-18 Genentech, Inc. Improved assembly of bispecific antibodies
EP2586788A1 (en) 2007-07-09 2013-05-01 Genentech, Inc. Prevention of disulfide bond reduction during recombinant production of polypeptides
EP2592156A2 (en) 2007-06-08 2013-05-15 Genentech, Inc. Gene expression markers of tumor resistance to HER2 inhibitor treatment
US8475794B2 (en) 2005-04-06 2013-07-02 Ibc Pharmaceuticals, Inc. Combination therapy with anti-CD74 antibodies provides enhanced toxicity to malignancies, Autoimmune disease and other diseases
WO2013101771A2 (en) 2011-12-30 2013-07-04 Genentech, Inc. Compositions and method for treating autoimmune diseases
WO2013112467A1 (en) 2012-01-23 2013-08-01 Trustees Of Boston University DEspR ANTAGONISTS AND AGONISTS AS THERAPEUTICS
US8530627B2 (en) 2002-08-14 2013-09-10 Macrogenics, Inc. FcγRIIB specific antibodies and methods of use thereof
US8546543B2 (en) 2004-11-12 2013-10-01 Xencor, Inc. Fc variants that extend antibody half-life
US8551476B2 (en) 2005-07-08 2013-10-08 Biogen Idec Ma Inc. SP35 antibodies and uses thereof
US20130287780A1 (en) * 2008-01-22 2013-10-31 Multimerics Aps Products and methods to prevent infections
US8586714B2 (en) 2009-09-01 2013-11-19 Abbvie, Inc. Dual variable domain immunoglobulins and uses thereof
WO2013177062A2 (en) 2012-05-21 2013-11-28 Genentech, Inc. Methods for improving safety of blood-brain barrier transport
WO2014033074A1 (en) 2012-08-29 2014-03-06 F. Hoffmann-La Roche Ag Blood brain barrier shuttle
US8716450B2 (en) 2009-10-15 2014-05-06 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
US8735546B2 (en) 2010-08-03 2014-05-27 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
EP2752189A1 (en) 2008-11-22 2014-07-09 F. Hoffmann-La Roche AG Use of anti-vegf antibody in combination with chemotherapy for treating breast cancer
US20140242079A1 (en) * 2013-02-26 2014-08-28 Roche Glycart Ag Bispecific t cell activating antigen binding molecules
US8822645B2 (en) 2008-07-08 2014-09-02 Abbvie Inc. Prostaglandin E2 dual variable domain immunoglobulins and uses thereof
US8945553B2 (en) 2012-05-22 2015-02-03 Bristol-Myers Squibb Company Bispecific antibodies to IL-23 and IL-17A/F
WO2015023596A1 (en) 2013-08-12 2015-02-19 Genentech, Inc. Compositions and method for treating complement-associated conditions
US8987418B2 (en) 2013-03-15 2015-03-24 Abbvie Inc. Dual specific binding proteins directed against IL-1β and/or IL-17
US9029508B2 (en) 2008-04-29 2015-05-12 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
US9045551B2 (en) 2012-11-01 2015-06-02 Abbvie Inc. Anti-DLL4/VEGF dual variable domain immunoglobulin and uses thereof
US9046513B2 (en) 2010-08-26 2015-06-02 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
US9051373B2 (en) 2003-05-02 2015-06-09 Xencor, Inc. Optimized Fc variants
US9068992B2 (en) 2004-06-24 2015-06-30 Biogen Ma Inc. Screening methods for identifying Sp35 antagonists
US9072798B2 (en) 2009-02-18 2015-07-07 Ludwig Institute For Cancer Research Ltd. Specific binding proteins and uses thereof
WO2015103072A1 (en) * 2013-12-30 2015-07-09 Epimab Biotherapeutics Fabs-in-tandem immunoglobulin and uses thereof
US9085622B2 (en) 2010-09-03 2015-07-21 Glaxosmithkline Intellectual Property Development Limited Antigen binding proteins
US9120870B2 (en) 2011-12-30 2015-09-01 Abbvie Inc. Dual specific binding proteins directed against IL-13 and IL-17
WO2015138920A1 (en) 2014-03-14 2015-09-17 Novartis Ag Antibody molecules to lag-3 and uses thereof
WO2015142675A2 (en) 2014-03-15 2015-09-24 Novartis Ag Treatment of cancer using chimeric antigen receptor
WO2015171822A1 (en) 2014-05-06 2015-11-12 Genentech, Inc. Production of heteromultimeric proteins using mammalian cells
WO2015175375A1 (en) 2014-05-13 2015-11-19 Short Jay M Conditionally active biological proteins
WO2016014565A2 (en) 2014-07-21 2016-01-28 Novartis Ag Treatment of cancer using humanized anti-bcma chimeric antigen receptor
WO2016014553A1 (en) 2014-07-21 2016-01-28 Novartis Ag Sortase synthesized chimeric antigen receptors
WO2016014530A1 (en) 2014-07-21 2016-01-28 Novartis Ag Combinations of low, immune enhancing. doses of mtor inhibitors and cars
WO2016025880A1 (en) 2014-08-14 2016-02-18 Novartis Ag Treatment of cancer using gfr alpha-4 chimeric antigen receptor
WO2016033331A1 (en) 2014-08-28 2016-03-03 Bioatla, Llc Conditionally active chimeric antigen receptors for modified t-cells
US9283276B2 (en) 2007-08-14 2016-03-15 Ludwig Institute For Cancer Research Ltd. Monoclonal antibody 175 targeting the EGF receptor and derivatives and uses thereof
WO2016044605A1 (en) 2014-09-17 2016-03-24 Beatty, Gregory Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy
WO2016061142A1 (en) 2014-10-14 2016-04-21 Novartis Ag Antibody molecules to pd-l1 and uses thereof
US20160130347A1 (en) * 2012-10-08 2016-05-12 Roche Glycart Ag Fc-free antibodies comprising two Fab-fragments and methods of use
JP2016514098A (ja) * 2013-02-26 2016-05-19 ロシュ グリクアート アーゲー 二重特異性t細胞活性化抗原結合分子
WO2016081643A1 (en) 2014-11-19 2016-05-26 Genentech, Inc. Anti-transferrin receptor antibodies and methods of use
WO2016090034A2 (en) 2014-12-03 2016-06-09 Novartis Ag Methods for b cell preconditioning in car therapy
WO2016115274A1 (en) * 2015-01-14 2016-07-21 Compass Therapeutics Llc Multispecific immunomodulatory antigen-binding constructs
WO2016123329A2 (en) 2015-01-28 2016-08-04 Genentech, Inc. Gene expression markers and treatment of multiple sclerosis
WO2016164731A2 (en) 2015-04-08 2016-10-13 Novartis Ag Cd20 therapies, cd22 therapies, and combination therapies with a cd19 chimeric antigen receptor (car) - expressing cell
US9475881B2 (en) 2010-01-19 2016-10-25 Xencor, Inc. Antibody variants with enhanced complement activity
WO2016172583A1 (en) 2015-04-23 2016-10-27 Novartis Ag Treatment of cancer using chimeric antigen receptor and protein kinase a blocker
WO2016196381A1 (en) 2015-05-29 2016-12-08 Genentech, Inc. Pd-l1 promoter methylation in cancer
WO2017019894A1 (en) 2015-07-29 2017-02-02 Novartis Ag Combination therapies comprising antibody molecules to lag-3
WO2017019897A1 (en) 2015-07-29 2017-02-02 Novartis Ag Combination therapies comprising antibody molecules to tim-3
US9562102B2 (en) 2001-05-11 2017-02-07 Ludwig Institute For Cancer Research Specific binding proteins and uses thereof
WO2017055542A1 (en) 2015-10-02 2017-04-06 F. Hoffmann-La Roche Ag Bispecific anti-human cd20/human transferrin receptor 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
WO2017055540A1 (en) 2015-10-02 2017-04-06 F. Hoffmann-La Roche Ag Bispecific anti-human a-beta/human transferrin receptor antibodies and methods of use
WO2017062682A2 (en) 2015-10-06 2017-04-13 Genentech, Inc. Method for treating multiple sclerosis
WO2017106810A2 (en) 2015-12-17 2017-06-22 Novartis Ag Combination of c-met inhibitor with antibody molecule to pd-1 and uses thereof
WO2017106656A1 (en) 2015-12-17 2017-06-22 Novartis Ag Antibody molecules to pd-1 and uses thereof
WO2017124002A1 (en) * 2016-01-13 2017-07-20 Compass Therapeutics Llc Multispecific immunomodulatory antigen-binding constructs
WO2017123537A1 (en) 2016-01-12 2017-07-20 Bioatla, Llc Diagnostics using conditionally active antibodies
US9714282B2 (en) 2003-09-26 2017-07-25 Xencor, Inc. Optimized Fc variants and methods for their generation
WO2017125897A1 (en) 2016-01-21 2017-07-27 Novartis Ag Multispecific molecules targeting cll-1
US9732162B2 (en) 2007-10-15 2017-08-15 Sanofi Antibodies that bind IL-4 and/or IL-13 and their uses
WO2017149515A1 (en) 2016-03-04 2017-09-08 Novartis Ag Cells expressing multiple chimeric antigen receptor (car) molecules and uses therefore
US9771417B2 (en) 2014-08-07 2017-09-26 Novartis Ag Angiopoietin-like 4 antibodies and methods of use
WO2017165683A1 (en) 2016-03-23 2017-09-28 Novartis Ag Cell secreted minibodies and uses thereof
WO2017181119A2 (en) 2016-04-15 2017-10-19 Novartis Ag Compositions and methods for selective protein expression
US9796780B2 (en) 2012-05-14 2017-10-24 Biogen Ma Inc. LINGO-2 antagonists for treatment of conditions involving motor neurons
US20170306053A1 (en) * 2014-11-21 2017-10-26 Astellas Pharma Inc. Novel bispecific antibody format
WO2017210617A2 (en) 2016-06-02 2017-12-07 Porter, David, L. Therapeutic regimens for chimeric antigen receptor (car)- expressing cells
US9840554B2 (en) 2015-06-15 2017-12-12 Abbvie Inc. Antibodies against platelet-derived growth factor (PDGF)
WO2018013918A2 (en) 2016-07-15 2018-01-18 Novartis Ag Treatment and prevention of cytokine release syndrome using a chimeric antigen receptor in combination with a kinase inhibitor
WO2018023025A1 (en) 2016-07-28 2018-02-01 Novartis Ag Combination therapies of chimeric antigen receptors adn pd-1 inhibitors
WO2018026819A2 (en) 2016-08-01 2018-02-08 Novartis Ag Treatment of cancer using a chimeric antigen receptor in combination with an inhibitor of a pro-m2 macrophage molecule
US9914776B2 (en) 2014-08-04 2018-03-13 Hoffmann-La Roche Inc. Bispecific T cell activating antigen binding molecules
WO2018067992A1 (en) 2016-10-07 2018-04-12 Novartis Ag Chimeric antigen receptors for the treatment of cancer
US9988443B2 (en) 2014-08-07 2018-06-05 Novartis Ag Angiopoetin-like 4 (ANGPTL4) antibodies and methods of use
WO2018118780A1 (en) 2016-12-19 2018-06-28 Calico Biolabs, Inc. Monovalent and divalent binding proteins
WO2018129007A1 (en) 2017-01-03 2018-07-12 Bioatla Llc Protein therapeutics for treatment of senescent cells
WO2018140725A1 (en) 2017-01-26 2018-08-02 Novartis Ag Cd28 compositions and methods for chimeric antigen receptor therapy
WO2018160731A1 (en) 2017-02-28 2018-09-07 Novartis Ag Shp inhibitor compositions and uses for chimeric antigen receptor therapy
WO2018162517A1 (en) 2017-03-10 2018-09-13 F. Hoffmann-La Roche Ag Method for producing multispecific antibodies
US10093733B2 (en) 2014-12-11 2018-10-09 Abbvie Inc. LRP-8 binding dual variable domain immunoglobulin proteins
WO2018201051A1 (en) 2017-04-28 2018-11-01 Novartis Ag Bcma-targeting agent, and combination therapy with a gamma secretase inhibitor
WO2018201056A1 (en) 2017-04-28 2018-11-01 Novartis Ag Cells expressing a bcma-targeting chimeric antigen receptor, and combination therapy with a gamma secretase inhibitor
WO2018197533A1 (en) 2017-04-28 2018-11-01 F. Hoffmann-La Roche Ag Antibody selection method
EP3401335A1 (en) 2008-01-30 2018-11-14 Genentech, Inc. Composition comprising antibody that binds to domain ii of her2 and acidic variants thereof
WO2018237157A1 (en) 2017-06-22 2018-12-27 Novartis Ag CD73 BINDING ANTIBODY MOLECULES AND USES THEREOF
WO2019006007A1 (en) 2017-06-27 2019-01-03 Novartis Ag POSOLOGICAL REGIMES FOR ANTI-TIM3 ANTIBODIES AND USES THEREOF
WO2019018730A1 (en) 2017-07-20 2019-01-24 Novartis Ag DOSAGE REGIMES FOR ANTI-LAG3 ANTIBODIES AND USES THEREOF
WO2019077092A1 (en) 2017-10-20 2019-04-25 F. Hoffmann-La Roche Ag METHOD FOR GENERATING MULTISPECIFIC ANTIBODIES FROM MONOSPECIFIC ANTIBODIES
WO2019086362A1 (en) 2017-10-30 2019-05-09 F. Hoffmann-La Roche Ag Method for in vivo generation of multispecific antibodies from monospecific antibodies
WO2019099838A1 (en) 2017-11-16 2019-05-23 Novartis Ag Combination therapies
EP3514179A1 (en) 2014-01-24 2019-07-24 Dana-Farber Cancer Institute, Inc. Antibody molecules to pd-1 and uses thereof
WO2019152660A1 (en) 2018-01-31 2019-08-08 Novartis Ag Combination therapy using a chimeric antigen receptor
US10435467B2 (en) 2015-01-08 2019-10-08 Biogen Ma Inc. LINGO-1 antagonists and uses for treatment of demyelinating disorders
WO2019200229A1 (en) 2018-04-13 2019-10-17 Novartis Ag Dosage regimens for anti-pd-l1 antibodies and uses thereof
US10450379B2 (en) 2005-11-15 2019-10-22 Genetech, Inc. Method for treating joint damage
WO2019210153A1 (en) 2018-04-27 2019-10-31 Novartis Ag Car t cell therapies with enhanced efficacy
WO2019219721A1 (en) 2018-05-18 2019-11-21 F. Hoffmann-La Roche Ag Targeted intracellular delivery of large nucleic acids
WO2019226617A1 (en) 2018-05-21 2019-11-28 Compass Therapeutics Llc Compositions and methods for enhancing the killing of target cells by nk cells
WO2019226658A1 (en) 2018-05-21 2019-11-28 Compass Therapeutics Llc Multispecific antigen-binding compositions and methods of use
WO2019227003A1 (en) 2018-05-25 2019-11-28 Novartis Ag Combination therapy with chimeric antigen receptor (car) therapies
WO2019232244A2 (en) 2018-05-31 2019-12-05 Novartis Ag Antibody molecules to cd73 and uses thereof
US10501552B2 (en) 2015-01-26 2019-12-10 Macrogenics, Inc. Multivalent molecules comprising DR5-binding domains
WO2019241426A1 (en) 2018-06-13 2019-12-19 Novartis Ag Bcma chimeric antigen receptors and uses thereof
WO2019246293A2 (en) 2018-06-19 2019-12-26 Atarga, Llc Antibody molecules to complement component 5 and uses thereof
US10526413B2 (en) 2015-10-02 2020-01-07 Hoffmann-La Roche Inc. Bispecific antibodies specific for OX40
EP3594240A1 (en) 2013-05-20 2020-01-15 F. Hoffmann-La Roche AG Anti-transferrin receptor antibodies and methods of use
WO2020012337A1 (en) 2018-07-10 2020-01-16 Novartis Ag 3-(5-amino-1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and their use in the treatment of i karos family zinc finger 2 (ikzf2)-dependent diseases
WO2020021465A1 (en) 2018-07-25 2020-01-30 Advanced Accelerator Applications (Italy) S.R.L. Method of treatment of neuroendocrine tumors
US10563194B2 (en) 2015-02-24 2020-02-18 BioAlta, LLC Conditionally active biological proteins
US10596257B2 (en) 2016-01-08 2020-03-24 Hoffmann-La Roche Inc. Methods of treating CEA-positive cancers using PD-1 axis binding antagonists and anti-CEA/anti-CD3 bispecific antibodies
WO2020084032A1 (en) 2018-10-25 2020-04-30 F. Hoffmann-La Roche Ag Modification of antibody fcrn binding
WO2020084034A1 (en) 2018-10-26 2020-04-30 F. Hoffmann-La Roche Ag Multispecific antibody screening method using recombinase mediated cassette exchange
EP3653641A1 (en) 2004-02-19 2020-05-20 Genentech, Inc. Cdr-repaired antibodies
EP3660042A1 (en) 2014-07-31 2020-06-03 Novartis AG Subset-optimized chimeric antigen receptor-containing t-cells
US10689447B2 (en) 2011-02-04 2020-06-23 Genentech, Inc. Fc variants and methods for their production
WO2020128898A1 (en) 2018-12-20 2020-06-25 Novartis Ag Pharmaceutical combinations
WO2020132230A2 (en) 2018-12-20 2020-06-25 Genentech, Inc. Modified antibody fcs and methods of use
WO2020128972A1 (en) 2018-12-20 2020-06-25 Novartis Ag Dosing regimen and pharmaceutical combination comprising 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives
US10697972B2 (en) 2016-01-12 2020-06-30 Bioatla, Llc Diagnostics using conditionally active antibodies
WO2020165868A1 (en) 2019-02-15 2020-08-20 Perkinelmer Cellular Technologies Germany Gmbh Low-power microscope-objective pre-scan and high-power microscope-objective scan in x,y and z-direction for imaging objects such as cells using a microscope
WO2020165834A1 (en) 2019-02-15 2020-08-20 Novartis Ag Substituted 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
WO2020165833A1 (en) 2019-02-15 2020-08-20 Novartis Ag 3-(1-oxo-5-(piperidin-4-yl)isoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
EP3698807A1 (en) 2005-01-21 2020-08-26 Genentech, Inc. Fixed dosing of her antibodies
WO2020172553A1 (en) 2019-02-22 2020-08-27 Novartis Ag Combination therapies of egfrviii chimeric antigen receptors and pd-1 inhibitors
US10766967B2 (en) 2015-10-02 2020-09-08 Hoffmann-La Roche Inc. Bispecific T cell activating antigen binding molecules
EP3712171A1 (en) 2014-08-19 2020-09-23 Novartis AG Treatment of cancer using a cd123 chimeric antigen receptor
WO2020205523A1 (en) 2019-03-29 2020-10-08 Atarga, Llc Anti fgf23 antibody
EP3722316A1 (en) 2014-07-21 2020-10-14 Novartis AG Treatment of cancer using a cd33 chimeric antigen receptor
US10844135B2 (en) 2003-10-10 2020-11-24 Immunogen, Inc. Method of targeting specific cell populations using cell-binding agent maytansinoid conjugates linked via a non-cleavable linker, said conjugates and methods of making said
WO2020254355A1 (en) 2019-06-19 2020-12-24 F. Hoffmann-La Roche Ag Method for the generation of a bivalent, bispecific antibody expressing cell by targeted integration of multiple expression cassettes in a defined organization
WO2020254351A1 (en) 2019-06-19 2020-12-24 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
WO2020254356A1 (en) 2019-06-19 2020-12-24 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
WO2020254352A1 (en) 2019-06-19 2020-12-24 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
WO2020254357A1 (en) 2019-06-19 2020-12-24 F. Hoffmann-La Roche Ag Method for the generation of a protein expressing cell by targeted integration using cre mrna
WO2020260327A1 (en) 2019-06-26 2020-12-30 F. Hoffmann-La Roche Ag Mammalian cell lines with sirt-1 gene knockout
WO2021079195A1 (en) 2019-10-21 2021-04-29 Novartis Ag Tim-3 inhibitors and uses thereof
WO2021079188A1 (en) 2019-10-21 2021-04-29 Novartis Ag Combination therapies with venetoclax and tim-3 inhibitors
US11008389B2 (en) 2011-03-16 2021-05-18 Sanofi Uses of a dual V region antibody-like protein
US11013801B2 (en) 2015-12-09 2021-05-25 Hoffmann-La Roche Inc. Treatment method
WO2021108661A2 (en) 2019-11-26 2021-06-03 Novartis Ag Chimeric antigen receptors and uses thereof
WO2021123996A1 (en) 2019-12-20 2021-06-24 Novartis Ag Uses of anti-tgf-beta antibodies and checkpoint inhibitors for the treatment of proliferative diseases
US11046769B2 (en) 2018-11-13 2021-06-29 Compass Therapeutics Llc Multispecific binding constructs against checkpoint molecules and uses thereof
WO2021146636A1 (en) 2020-01-17 2021-07-22 Becton, Dickinson And Company Methods and compositions for single cell secretomics
WO2021144657A1 (en) 2020-01-17 2021-07-22 Novartis Ag Combination comprising a tim-3 inhibitor and a hypomethylating agent for use in treating myelodysplastic syndrome or chronic myelomonocytic leukemia
WO2021173995A2 (en) 2020-02-27 2021-09-02 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
US11111288B2 (en) 2014-08-28 2021-09-07 Bioatla, Inc. Conditionally active chimeric antigen receptors for modified t-cells
WO2021194861A1 (en) 2020-03-23 2021-09-30 Genentech, Inc. Biomarkers for predicting response to il-6 antagonist in covid-19 pneumonia
WO2021194860A1 (en) 2020-03-23 2021-09-30 Genentech, Inc. Tocilizumab and remdesivir combination therapy for covid-19 pneumonia
WO2021194865A1 (en) 2020-03-23 2021-09-30 Genentech, Inc. Method for treating pneumonia, including covid-19 pneumonia, with an il6 antagonist
WO2021204743A1 (en) 2020-04-08 2021-10-14 F. Hoffmann-La Roche Ag Large molecule unspecific clearance assay
US11186636B2 (en) 2017-04-21 2021-11-30 Amgen Inc. Anti-human TREM2 antibodies and uses thereof
WO2021260528A1 (en) 2020-06-23 2021-12-30 Novartis Ag Dosing regimen comprising 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives
WO2022013787A1 (en) 2020-07-16 2022-01-20 Novartis Ag Anti-betacellulin antibodies, fragments thereof, and multi-specific binding molecules
WO2022026592A2 (en) 2020-07-28 2022-02-03 Celltas Bio, Inc. Antibody molecules to coronavirus and uses thereof
WO2022029573A1 (en) 2020-08-03 2022-02-10 Novartis Ag Heteroaryl substituted 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
WO2022043558A1 (en) 2020-08-31 2022-03-03 Advanced Accelerator Applications International Sa Method of treating psma-expressing cancers
WO2022043557A1 (en) 2020-08-31 2022-03-03 Advanced Accelerator Applications International Sa Method of treating psma-expressing cancers
WO2022060412A1 (en) 2020-09-17 2022-03-24 Genentech, Inc. Results of empacta: a randomized, double-blind, placebo-controlled, multicenter study to evaluate the efficacy and safety of tocilizumab in hospitalized patients with covid-19 pneumonia
WO2022063877A1 (en) 2020-09-24 2022-03-31 F. Hoffmann-La Roche Ag Mammalian cell lines with gene knockout
WO2022097065A2 (en) 2020-11-06 2022-05-12 Novartis Ag ANTIBODY Fc VARIANTS
WO2022104061A1 (en) 2020-11-13 2022-05-19 Novartis Ag Combination therapies with chimeric antigen receptor (car)-expressing cells
US11352426B2 (en) 2015-09-21 2022-06-07 Aptevo Research And Development Llc CD3 binding polypeptides
WO2022136140A1 (en) 2020-12-22 2022-06-30 F. Hoffmann-La Roche Ag Oligonucleotides targeting xbp1
US11401348B2 (en) 2009-09-02 2022-08-02 Xencor, Inc. Heterodimeric Fc variants
WO2022162569A1 (en) 2021-01-29 2022-08-04 Novartis Ag Dosage regimes for anti-cd73 and anti-entpd2 antibodies and uses thereof
US11421028B2 (en) 2016-02-06 2022-08-23 Epimab Biotherapeutics, Inc. Fabs-in-tandem immunoglobulin and uses thereof
US11447558B2 (en) 2017-01-03 2022-09-20 Hoffmann-La Roche Inc. Bispecific antigen binding molecules comprising anti-4-1BB clone 20H4.9
WO2022215011A1 (en) 2021-04-07 2022-10-13 Novartis Ag USES OF ANTI-TGFβ ANTIBODIES AND OTHER THERAPEUTIC AGENTS FOR THE TREATMENT OF PROLIFERATIVE DISEASES
WO2022214565A1 (en) 2021-04-09 2022-10-13 F. Hoffmann-La Roche Ag Process for selecting cell clones expressing a heterologous polypeptide
US11472876B2 (en) 2015-11-02 2022-10-18 Bioatla, Inc. Conditionally active polypeptides
US11505616B2 (en) * 2016-03-25 2022-11-22 Biomunex Pharmaceuticals Binding molecules to CD38 and PD-L1
WO2022243846A1 (en) 2021-05-18 2022-11-24 Novartis Ag Combination therapies
WO2022261417A1 (en) 2021-06-11 2022-12-15 Genentech, Inc. Method for treating chronic obstructive pulmonary disease with an st2 antagonist
US11560437B2 (en) 2017-03-27 2023-01-24 Biomunex Pharmaceuticals Stable multispecific antibodies
US11584793B2 (en) 2015-06-24 2023-02-21 Hoffmann-La Roche Inc. Anti-transferrin receptor antibodies with tailored affinity
WO2023044483A2 (en) 2021-09-20 2023-03-23 Voyager Therapeutics, Inc. Compositions and methods for the treatment of her2 positive cancer
US11634502B2 (en) 2013-03-15 2023-04-25 Amgen Inc. Heterodimeric bispecific antibodies
US11639397B2 (en) 2011-08-23 2023-05-02 Roche Glycart Ag Bispecific antibodies specific for T-cell activating antigens and a tumor antigen and methods of use
WO2023092004A1 (en) 2021-11-17 2023-05-25 Voyager Therapeutics, Inc. Compositions and methods for the treatment of tau-related disorders
WO2023117325A1 (en) 2021-12-21 2023-06-29 F. Hoffmann-La Roche Ag Method for the determination of hydrolytic activity
US11718679B2 (en) 2017-10-31 2023-08-08 Compass Therapeutics Llc CD137 antibodies and PD-1 antagonists and uses thereof
WO2023150778A1 (en) 2022-02-07 2023-08-10 Visterra, Inc. Anti-idiotype antibody molecules and uses thereof
US11739160B2 (en) 2018-12-24 2023-08-29 Sanofi PseudoFab-based multispecific binding proteins
US11752207B2 (en) 2017-07-11 2023-09-12 Compass Therapeutics Llc Agonist antibodies that bind human CD137 and uses thereof
WO2023202967A1 (en) 2022-04-19 2023-10-26 F. Hoffmann-La Roche Ag Improved production cells
US11814409B2 (en) 2012-02-15 2023-11-14 Hoffmann-La Roche Inc. Fc-receptor based affinity chromatography
WO2023220695A2 (en) 2022-05-13 2023-11-16 Voyager Therapeutics, Inc. Compositions and methods for the treatment of her2 positive cancer
US11820830B2 (en) 2004-07-20 2023-11-21 Xencor, Inc. Optimized Fc variants
WO2023232961A1 (en) 2022-06-03 2023-12-07 F. Hoffmann-La Roche Ag Improved production cells
US11851497B2 (en) 2017-11-20 2023-12-26 Compass Therapeutics Llc CD137 antibodies and tumor antigen-targeting antibodies and uses thereof
US11879011B2 (en) 2016-05-13 2024-01-23 Bioatla, Inc. Anti-ROR2 antibodies, antibody fragments, their immunoconjucates and uses thereof
WO2024030976A2 (en) 2022-08-03 2024-02-08 Voyager Therapeutics, Inc. Compositions and methods for crossing the blood brain barrier
EP4324518A2 (en) 2014-01-31 2024-02-21 Novartis AG Antibody molecules to tim-3 and uses thereof
US11932685B2 (en) 2007-10-31 2024-03-19 Xencor, Inc. Fc variants with altered binding to FcRn
WO2024079069A1 (en) 2022-10-12 2024-04-18 F. Hoffmann-La Roche Ag Method for classifying cells

Families Citing this family (713)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6136311A (en) 1996-05-06 2000-10-24 Cornell Research Foundation, Inc. Treatment and diagnosis of cancer
US7833529B1 (en) 1999-01-07 2010-11-16 Zymogenetics, Inc. Methods for inhibiting B lymphocyte proliferation with soluble ztnf4 receptor
US7696325B2 (en) 1999-03-10 2010-04-13 Chugai Seiyaku Kabushiki Kaisha Polypeptide inducing apoptosis
IL151853A0 (en) 2000-04-11 2003-04-10 Genentech Inc Multivalent antibodies and uses therefor
US7279160B2 (en) 2000-05-02 2007-10-09 The Uab Research Foundation Combinations of DR5 antibodies and other therapeutic agents
US7476383B2 (en) 2000-05-02 2009-01-13 The Uab Research Foundation Antibody selective for DR4 and uses thereof
TWI318983B (en) 2000-05-02 2010-01-01 Uab Research Foundation An antibody selective for a tumor necrosis factor-related apoptosis-inducing ligand receptor and uses thereof
KR20030055274A (ko) 2000-10-20 2003-07-02 츄가이 세이야꾸 가부시키가이샤 저분자화 트롬보포에틴 아고니스트 항체
ES2390425T3 (es) 2000-12-22 2012-11-12 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. Uso de moléculas de orientación repulsivas (RGM) y sus moduladores
JP2004533997A (ja) 2001-02-20 2004-11-11 ザイモジェネティクス,インコーポレイティド Bcma及びtaciの両者を結合する抗体
PL403488A1 (pl) 2001-05-24 2013-07-08 Zymogenetics, Inc. Zastosowanie bialka fuzyjnego TACI-immunoglobulina, bialko fuzyjne, czasteczka kwasu nukleinowego kodujaca bialko fuzyjne, kompozycja farmaceutyczna i sposób wytwarzania bialka fuzyjnego TACI-immunoglobulina
WO2003011909A1 (en) * 2001-07-31 2003-02-13 University Of Southampton Binding agents with differential activity
WO2003038043A2 (en) * 2001-11-01 2003-05-08 Uab Research Foundation Combinations of dr5 antibody and other therapeutic agents
US20030149246A1 (en) * 2002-02-01 2003-08-07 Russell John C. Macromolecular conjugates and processes for preparing the same
US9028822B2 (en) 2002-06-28 2015-05-12 Domantis Limited Antagonists against TNFR1 and methods of use therefor
CA2494105C (en) * 2002-07-31 2013-04-02 Seattle Genetics, Inc. Drug conjugates and their use for treating cancer, an autoimmune disease or an infectious disease
TWI229650B (en) * 2002-11-19 2005-03-21 Sharp Kk Substrate accommodating tray
AU2003299984A1 (en) * 2002-12-20 2004-07-22 Biogen Idec Ma Inc. Multivalent lymphotoxin beta receptor agonists and therapeutic uses thereof
JP2004279086A (ja) 2003-03-13 2004-10-07 Konica Minolta Holdings Inc 放射線画像変換パネル及び放射線画像変換パネルの製造方法
CA2529027C (en) * 2003-06-13 2013-09-10 Immunomedics, Inc. D-amino acid peptides
AU2004253835B2 (en) 2003-07-04 2009-01-29 Affibody Ab Polypeptides having binding affinity for HER2
ATE490787T1 (de) * 2003-07-15 2010-12-15 Chugai Pharmaceutical Co Ltd Produktion von igm durch transformierte zellen und verfahren zur quantifizierung dieser igm- produktion
WO2005081687A2 (en) * 2003-09-30 2005-09-09 Centocor, Inc. Human hinge core mimetibodies, compositions, methods and uses
US20070212346A1 (en) 2003-10-09 2007-09-13 Tomoyuki Igawa Highly Concentrated Stabilized Igm Solution
EP1710255A4 (en) * 2003-12-12 2008-09-24 Chugai Pharmaceutical Co Ltd MODIFIED ANTIBODIES RECOGNIZING A TRIMER OR LARGER RECEPTOR
EP1784427A2 (en) 2004-06-01 2007-05-16 Domantis Limited Bispecific fusion antibodies with enhanced serum half-life
MXPA06014065A (es) 2004-06-01 2007-01-31 Genentech Inc Conjugados de droga-anticuerpo y metodos.
WO2006019447A1 (en) * 2004-07-15 2006-02-23 Xencor, Inc. Optimized fc variants
JP5562521B2 (ja) 2005-02-02 2014-07-30 ザ ユーエービー リサーチ ファンデーション アポトーシス誘導デスレセプターアゴニストに対する抵抗性を低減することに関する薬剤及び方法
TW200722518A (en) 2005-03-31 2007-06-16 Chugai Pharmaceutical Co Ltd Sc(fv)2 structural isomers
KR20080021614A (ko) * 2005-05-26 2008-03-07 쉐링 코포레이션 인터페론-IgG 융합체
AU2006256041B2 (en) 2005-06-10 2012-03-29 Chugai Seiyaku Kabushiki Kaisha Stabilizer for protein preparation comprising meglumine and use thereof
JP5085322B2 (ja) 2005-06-10 2012-11-28 中外製薬株式会社 sc(Fv)2を含有する医薬組成物
CN101262876A (zh) 2005-08-09 2008-09-10 酶遗传学股份有限公司 用taci-ig融合分子治疗b细胞恶性肿瘤的方法
US8808696B2 (en) 2005-08-09 2014-08-19 Ares Trading S.A. Methods for the treatment and prevention of abnormal cell proliferation using TACI-fusion molecules
AU2012205249B2 (en) * 2005-08-19 2014-03-20 Abbvie Inc. Dual variable domain immunoglobin and uses thereof
AU2014203217B2 (en) * 2005-08-19 2017-03-30 Abbvie Inc. Dual variable domain immunoglobin and uses thereof
JP2012228248A (ja) * 2005-08-19 2012-11-22 Abbott Lab 二重可変ドメイン免疫グロブリン及びその使用
BRPI0615026A8 (pt) 2005-08-19 2018-03-06 Abbott Lab imunoglobulina de domínio variável duplo e seus usos
EP2500359A3 (en) 2005-08-19 2012-10-17 Abbott Laboratories Dual variable domain immunoglobulin and uses thereof
EP1928905B1 (de) 2005-09-30 2015-04-15 AbbVie Deutschland GmbH & Co KG Bindungsdomänen von proteinen der repulsive guidance molecule (rgm) proteinfamilie und funktionale fragmente davon sowie deren verwendung
SG10201405107YA (en) * 2006-02-23 2014-10-30 Viacyte Inc Compositions and methods useful for culturing differentiable cells
JP2009531324A (ja) 2006-03-20 2009-09-03 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア 癌標的化のための操作された抗前立腺幹細胞抗原(psca)抗体
UA98462C2 (ru) 2006-05-15 2012-05-25 Арес Трейдинг С.А. Способы лечения аутоиммунных заболеваний с использованием слитой молекулы taci-ig
JP2010516625A (ja) 2007-01-24 2010-05-20 インサート セラピューティクス, インコーポレイテッド 制御された薬物送達のためのテザー基を有するポリマー−薬物コンジュゲート
CA2694488A1 (en) 2007-07-31 2009-02-05 Medimmune, Llc Multispecific epitope binding proteins and uses thereof
US8940298B2 (en) 2007-09-04 2015-01-27 The Regents Of The University Of California High affinity anti-prostate stem cell antigen (PSCA) antibodies for cancer targeting and detection
EP2033971A1 (de) * 2007-09-06 2009-03-11 Abbott GmbH & Co. KG Bone Morphogenetic Protein (BMP)-bindende Domänen von Proteinen der Repulsive Guidance Molecule (RGM) Proteinfamilie und funktionale Fragmente davon sowie deren Verwendung
EP3415529B1 (en) 2007-09-26 2020-11-04 Chugai Seiyaku Kabushiki Kaisha Modified antibody constant region
JP5951929B2 (ja) * 2007-10-03 2016-07-13 コーネル ユニヴァーシティー Psma抗体を用いる増殖性障害の治療
AU2008328779B2 (en) 2007-11-27 2014-06-05 Ablynx N.V. Amino acid sequences directed against HER2 and polypeptides comprising the same for the treatment of cancers and/or tumors
US20090162359A1 (en) 2007-12-21 2009-06-25 Christian Klein Bivalent, bispecific antibodies
US9266967B2 (en) 2007-12-21 2016-02-23 Hoffmann-La Roche, Inc. Bivalent, bispecific antibodies
US8557243B2 (en) 2008-01-03 2013-10-15 The Scripps Research Institute EFGR antibodies comprising modular recognition domains
US8574577B2 (en) 2008-01-03 2013-11-05 The Scripps Research Institute VEGF antibodies comprising modular recognition domains
DK2769991T3 (en) * 2008-01-03 2018-12-10 Scripps Research Inst ANTIBODY TARGETING WITH A MODULAR RECOGNITION AREA
US8557242B2 (en) 2008-01-03 2013-10-15 The Scripps Research Institute ERBB2 antibodies comprising modular recognition domains
US8454960B2 (en) 2008-01-03 2013-06-04 The Scripps Research Institute Multispecific antibody targeting and multivalency through modular recognition domains
US8962803B2 (en) 2008-02-29 2015-02-24 AbbVie Deutschland GmbH & Co. KG Antibodies against the RGM A protein and uses thereof
BRPI0906099A2 (pt) * 2008-03-06 2015-07-21 Genentech Inc "método de tratamento do câncer em um indivíduo"
WO2009120905A2 (en) * 2008-03-26 2009-10-01 Cellerant Therapeutics, Inc. Immunoglobulin and/or toll-like receptor proteins associated with myelogenous haematological proliferative disorders and uses thereof
BRPI0906261A2 (pt) * 2008-03-31 2015-07-07 Genentech Inc "métodos de diagnóstico de um subtipo de asma em uma amostra de paciente, usos de um agente terapêutico e kits de diagnóstico de um subtipo de asma em uma amostra de paciente"
ES2828627T3 (es) * 2008-04-25 2021-05-27 Kyowa Kirin Co Ltd Anticuerpo multivalente estable
US8268314B2 (en) 2008-10-08 2012-09-18 Hoffmann-La Roche Inc. Bispecific anti-VEGF/anti-ANG-2 antibodies
AU2009308707A1 (en) 2008-10-31 2010-05-06 Biogen Idec Ma Inc. LIGHT targeting molecules and uses thereof
WO2010096486A1 (en) 2009-02-17 2010-08-26 Cornell Research Foundation, Inc. Methods and kits for diagnosis of cancer and prediction of therapeutic value
WO2010102251A2 (en) 2009-03-05 2010-09-10 Abbott Laboratories Il-17 binding proteins
CN101830986A (zh) * 2009-03-13 2010-09-15 北京表源生物技术有限公司 一种融合蛋白多聚体
TW201544123A (zh) * 2009-03-20 2015-12-01 Genentech Inc 抗-her抗體
NZ594343A (en) 2009-03-25 2013-10-25 Genentech Inc Novel anti-alpha5beta1 antibodies and uses thereof
WO2010112194A1 (en) 2009-04-02 2010-10-07 F. Hoffmann-La Roche Ag Antigen-binding polypeptides and multispecific antibodies comprising them
PE20120591A1 (es) 2009-04-02 2012-05-23 Roche Glycart Ag Anticuerpos multiespecificos que comprenden anticuerpos de longitud completa y fragmentos fab de cadena sencilla
CA2757669A1 (en) 2009-04-07 2010-10-14 Roche Glycart Ag Bispecific anti-erbb-1/anti-c-met antibodies
MX2011010166A (es) 2009-04-07 2011-10-11 Roche Glycart Ag Anticuerpos biespecificos anti-erbb-3/anti-c-met.
AU2010234031B2 (en) 2009-04-07 2015-10-01 Roche Glycart Ag Trivalent, bispecific antibodies
TW201100543A (en) 2009-05-27 2011-01-01 Hoffmann La Roche 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
ES2536996T3 (es) 2009-07-06 2015-06-01 F. Hoffmann-La Roche Ag Anticuerpos biespecíficos de unión a digoxigenina
CN105131112A (zh) 2009-08-29 2015-12-09 Abbvie公司 治疗用dll4结合蛋白
US20120283415A1 (en) * 2009-09-10 2012-11-08 Ucb Pharma S.A. Multivalent Antibodies
MX2012003396A (es) 2009-09-16 2012-04-10 Genentech Inc Complejos de proteina conteniendo helice super-enrollada y/o enlazador tether y usos de los mismos.
US20120201823A1 (en) * 2009-10-14 2012-08-09 Schering Corporation April antagonists and methods of use
CA3040276A1 (en) 2009-12-02 2011-06-09 Imaginab, Inc. J591 minibodies and cys-diabodies for targeting human prostate specific membrane antigen (psma) and methods for their use
CA2780069C (en) * 2009-12-08 2018-07-17 Abbott Gmbh & Co. Kg Monoclonal antibodies against the rgm a protein for use in the treatment of retinal nerve fiber layer degeneration
US9120855B2 (en) * 2010-02-10 2015-09-01 Novartis Ag Biologic compounds directed against death receptor 5
US9556249B2 (en) 2010-02-18 2017-01-31 Genentech, Inc. Neuregulin antagonists and use thereof in treating cancer
MX348312B (es) 2010-03-02 2017-06-06 Abbvie Inc Proteínas terapéuticas de enlace dll4.
AR080795A1 (es) 2010-03-24 2012-05-09 Genentech Inc Anticuerpos anti-lrp6 (proteina relacionada con el receptor ldl tipo 6)
US20150231215A1 (en) 2012-06-22 2015-08-20 Randolph J. Noelle VISTA Antagonist and Methods of Use
EP2552947A4 (en) 2010-03-26 2013-11-13 Dartmouth College VISTA REGULATORY T CELL MEDIATOR PROTEIN, VISTA BINDING ACTIVE SUBSTANCES AND USE THEREOF
TWI426920B (zh) 2010-03-26 2014-02-21 Hoffmann La Roche 雙專一性、雙價抗-vegf/抗-ang-2抗體
US10745467B2 (en) 2010-03-26 2020-08-18 The Trustees Of Dartmouth College VISTA-Ig for treatment of autoimmune, allergic and inflammatory disorders
TW201138821A (en) 2010-03-26 2011-11-16 Roche Glycart Ag Bispecific antibodies
SG185366A1 (en) 2010-05-04 2012-12-28 Merrimack Pharmaceuticals Inc Antibodies against epidermal growth factor receptor (egfr) and uses thereof
MY161302A (en) 2010-05-14 2017-04-14 Abbvie Inc IL-1 binding proteins
WO2011147834A1 (en) 2010-05-26 2011-12-01 Roche Glycart Ag Antibodies against cd19 and uses thereof
BR112012027995A2 (pt) 2010-06-18 2017-01-10 Genentech Inc anticorpo e ácido nucleíco isolado, célula hospedeira, método de produção de um anticorpo, imunoconjugado, formulação farmacêutica, uso do anticorpo, método de tratamento de um indivíduo com câncer, de um indivíduo possuíndo um distúrbio imune, de inibição da angiogênese e para inibir a ativação constitutiva de axl
WO2011161119A1 (en) 2010-06-22 2011-12-29 F. Hoffmann-La Roche Ag Antibodies against insulin-like growth factor i receptor and uses thereof
WO2011161189A1 (en) 2010-06-24 2011-12-29 F. Hoffmann-La Roche Ag Anti-hepsin antibodies and methods of use
US8741288B2 (en) * 2010-07-07 2014-06-03 Chang Gung Medical Foundation, Linkou Branch Protein markers for detecting liver cancer and method for identifying the markers thereof
WO2012006500A2 (en) 2010-07-08 2012-01-12 Abbott Laboratories Monoclonal antibodies against hepatitis c virus core protein
UY33492A (es) 2010-07-09 2012-01-31 Abbott Lab Inmunoglobulinas con dominio variable dual y usos de las mismas
CN103097418A (zh) 2010-07-09 2013-05-08 霍夫曼-拉罗奇有限公司 抗神经毡蛋白抗体及使用方法
US20120100166A1 (en) 2010-07-15 2012-04-26 Zyngenia, Inc. Ang-2 Binding Complexes and Uses Thereof
CA2804246A1 (en) 2010-07-19 2012-01-26 F. Hoffmann-La Roche Ag Method to identify a patient with an increased likelihood of responding to an anti-cancer therapy
BR112013001431A2 (pt) 2010-07-19 2016-05-31 Hoffmann La Roche método de identificação de pacientes com maior probabilidade de reação a terapia anticâncer
WO2012010582A1 (en) 2010-07-21 2012-01-26 Roche Glycart Ag Anti-cxcr5 antibodies and methods of use
CN103153341B (zh) 2010-08-03 2015-05-27 霍夫曼-拉罗奇有限公司 慢性淋巴细胞性白血病(cll)生物标志物
BR112013002532A2 (pt) 2010-08-05 2016-05-31 Hoffmann La Roche proteína de fusão de citocina anti-viral do anticorpo anti-mhc
MX2013001336A (es) 2010-08-13 2013-03-08 Roche Glycart Ag Anticuerpos ani-tenascina c a2 y metodos de utilizacion.
CA2806021C (en) 2010-08-13 2019-05-21 Roche Glycart Ag Anti-fap antibodies and methods of use
KR20130100125A (ko) 2010-08-13 2013-09-09 제넨테크, 인크. 질환의 치료를 위한, IL-1β 및 IL-18에 대한 항체
EP2609112B1 (en) 2010-08-24 2017-11-22 Roche Glycart AG Activatable bispecific antibodies
RU2013110875A (ru) 2010-08-24 2014-09-27 Ф.Хоффманн-Ля Рош Аг БИСПЕЦИФИЧЕСКИЕ АНТИТЕЛА, СОДЕРЖАЩИЕ СТАБИЛИЗИРОВАННЫЙ ДИСУЛЬФИДОМ ФРАГМЕНТ Fv
RU2013110874A (ru) 2010-08-25 2014-09-27 Ф.Хоффманн-Ля Рош Аг Антитела против il-18r1 и их применения
WO2012031027A1 (en) 2010-08-31 2012-03-08 Genentech, Inc. Biomarkers and methods of treatment
DK2635604T3 (en) * 2010-11-01 2017-02-27 Symphogen As PAN-HER-ANTIBODY COMPOSITION
TW201300417A (zh) 2010-11-10 2013-01-01 Genentech Inc 用於神經疾病免疫療法之方法及組合物
EP2646468B1 (en) 2010-12-01 2018-07-25 AlderBio Holdings LLC Anti-ngf compositions and use thereof
US9884909B2 (en) 2010-12-01 2018-02-06 Alderbio Holdings Llc Anti-NGF compositions and use thereof
US11214610B2 (en) 2010-12-01 2022-01-04 H. Lundbeck A/S High-purity production of multi-subunit proteins such as antibodies in transformed microbes such as Pichia pastoris
US9539324B2 (en) 2010-12-01 2017-01-10 Alderbio Holdings, Llc Methods of preventing inflammation and treating pain using anti-NGF compositions
US9078878B2 (en) 2010-12-01 2015-07-14 Alderbio Holdings Llc Anti-NGF antibodies that selectively inhibit the association of NGF with TrkA, without affecting the association of NGF with p75
US9067988B2 (en) 2010-12-01 2015-06-30 Alderbio Holdings Llc Methods of preventing or treating pain using anti-NGF antibodies
CN107312796A (zh) 2010-12-15 2017-11-03 大学共同利用机关法人情报·系统研究机构 蛋白质的生产方法
NZ609493A (en) 2010-12-16 2015-11-27 Genentech Inc Diagnosis and treatments relating to th2 inhibition
AU2011349502B2 (en) * 2010-12-20 2016-12-22 The Rockefeller University Modulating agonistic TNFR antibodies
EA028744B1 (ru) 2010-12-20 2017-12-29 Дженентек, Инк. Антитела против мезотелина и иммуноконъюгаты
US8853365B2 (en) 2010-12-21 2014-10-07 Abbvie Inc. Dual variable domain immunnoglobulins and uses thereof
US20120195910A1 (en) 2010-12-22 2012-08-02 Genentech, Inc. Anti-pcsk9 antibodies and methods of use
WO2012085111A1 (en) 2010-12-23 2012-06-28 F. Hoffmann-La Roche Ag Polypeptide-polynucleotide-complex and its use in targeted effector moiety delivery
RU2607038C2 (ru) 2011-02-28 2017-01-10 Ф. Хоффманн-Ля Рош Аг Антигенсвязывающие белки
CA2824824A1 (en) 2011-02-28 2012-09-07 F. Hoffmann-La Roche Ag Monovalent antigen binding proteins
JP2014509588A (ja) 2011-03-01 2014-04-21 アムジエン・インコーポレーテツド 二特異性結合剤
WO2012119989A2 (en) 2011-03-04 2012-09-13 Oryzon Genomics, S.A. Methods and antibodies for the diagnosis and treatment of cancer
TWI803876B (zh) * 2011-03-28 2023-06-01 法商賽諾菲公司 具有交叉結合區定向之雙重可變區類抗體結合蛋白
KR101614195B1 (ko) 2011-03-29 2016-04-20 로슈 글리카트 아게 항체 Fc 변이체
RU2013143358A (ru) 2011-04-07 2015-05-20 Дженентек, Инк. Анти-fgfr4 антитела и способы их применения
GB201106395D0 (en) 2011-04-14 2011-06-01 Hubrecht Inst Compounds
RU2013150331A (ru) 2011-04-20 2015-05-27 Рош Гликарт Аг СПОСОБ И УСТРОЙСТВА ДЛЯ рН-ЗАВИСИМОГО ПРОХОЖДЕНИЯ ГЕМАТОЭНЦЕФАЛИЧЕСКОГО БАРЬЕРА
WO2012155019A1 (en) 2011-05-12 2012-11-15 Genentech, Inc. Multiple reaction monitoring lc-ms/ms method to detect therapeutic antibodies in animal samples using framework signature pepides
DK2710035T3 (en) 2011-05-16 2017-06-19 Hoffmann La Roche FGFR1 agonists and methods of use
EP2714738B1 (en) 2011-05-24 2018-10-10 Zyngenia, Inc. Multivalent and monovalent multispecific complexes and their uses
US8691231B2 (en) 2011-06-03 2014-04-08 Merrimack Pharmaceuticals, Inc. Methods of treatment of tumors expressing predominantly high affinity EGFR ligands or tumors expressing predominantly low affinity EGFR ligands with monoclonal and oligoclonal anti-EGFR antibodies
BR112013032235A2 (pt) 2011-06-15 2016-11-22 Hoffmann La Roche anticorpos do receptor de epo anti-humano e métodos de uso
WO2013003680A1 (en) 2011-06-30 2013-01-03 Genentech, Inc. Anti-c-met antibody formulations
EP2543680A1 (en) * 2011-07-07 2013-01-09 Centre National de la Recherche Scientifique Multispecific mutated antibody Fab fragments
US20140363438A1 (en) 2011-08-17 2014-12-11 Genentech, Inc. Neuregulin antibodies and uses thereof
LT2748202T (lt) * 2011-08-23 2018-09-25 Roche Glycart Ag Bispecifinės antigeną surišančios molekulės
WO2013026832A1 (en) 2011-08-23 2013-02-28 Roche Glycart Ag Anti-mcsp antibodies
US20130058947A1 (en) 2011-09-02 2013-03-07 Stem Centrx, Inc Novel Modulators and Methods of Use
WO2013036543A2 (en) * 2011-09-10 2013-03-14 H. Lee Moffitt Cancer Center And Research Institute, Inc. Molecular imaging of cancer cells in vivo
CN103930781A (zh) 2011-09-15 2014-07-16 霍夫曼-拉罗奇有限公司 促进分化的方法
MX2014002990A (es) 2011-09-19 2014-05-21 Genentech Inc Tratamientos combinados que comprenden antagonistas de c-met y antagonistas de b-raf.
US20130089562A1 (en) 2011-10-05 2013-04-11 Genenthech, Inc. Methods of treating liver conditions using notch2 antagonists
JP6053688B2 (ja) * 2011-10-07 2016-12-27 国立大学法人三重大学 キメラ抗原受容体
JP6532678B2 (ja) 2011-10-14 2019-06-19 ジェネンテック, インコーポレイテッド 抗HtrA1抗体及び使用方法
CA2850836A1 (en) 2011-10-15 2013-04-18 Genentech, Inc. Methods of using scd1 antagonists
WO2013059531A1 (en) 2011-10-20 2013-04-25 Genentech, Inc. Anti-gcgr antibodies and uses thereof
CN104203978A (zh) 2011-10-24 2014-12-10 艾伯维股份有限公司 针对硬化蛋白的免疫结合剂
PE20142245A1 (es) 2011-10-24 2015-01-22 Abbvie Inc Inmunoligantes biespecificos dirigidos contra tnf e il-17
JP6251682B2 (ja) 2011-10-28 2017-12-20 ジェネンテック, インコーポレイテッド メラノーマ治療の治療の組み合わせ及び方法
CN103906533A (zh) * 2011-11-07 2014-07-02 米迪缪尼有限公司 多特异性和多价结合蛋白及其用途
KR102038310B1 (ko) * 2011-11-16 2019-11-01 베링거 인겔하임 인터내셔날 게엠베하 항 il-36r 항체
US20130129718A1 (en) 2011-11-21 2013-05-23 Genentech, Inc. Purification of anti-c-met antibodies
WO2013083497A1 (en) 2011-12-06 2013-06-13 F. Hoffmann-La Roche Ag Antibody formulation
SG11201403445YA (en) 2011-12-22 2014-07-30 Hoffmann La Roche Full length antibody display system for eukaryotic cells and its use
WO2013096346A1 (en) * 2011-12-22 2013-06-27 Development Center For Biotechnology Bispecific t-cell activator antibody
ES2930215T3 (es) 2011-12-22 2022-12-07 Hoffmann La Roche Combinaciones de elementos de vector de expresión, procedimientos de generación de células de producción novedosos y su uso para la producción recombinante de polipéptidos
RU2628703C2 (ru) 2011-12-22 2017-08-21 Ф. Хоффманн-Ля Рош Аг Строение экспрессионного вектора, новые способы получения клеток-продуцентов и их применение для рекомбинантного получения полипептидов
WO2013096791A1 (en) 2011-12-23 2013-06-27 Genentech, Inc. Process for making high concentration protein formulations
US20130171059A1 (en) 2011-12-30 2013-07-04 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
CN104066449B (zh) 2012-01-18 2018-04-27 霍夫曼-拉罗奇有限公司 抗lrp5抗体及使用方法
US20130183294A1 (en) 2012-01-18 2013-07-18 Genentech, Inc. Methods of using fgf19 modulators
CN107880124B (zh) 2012-01-27 2021-08-13 艾伯维德国有限责任两合公司 用于诊断和治疗与神经突变性相关的疾病的组合物和方法
KR102091297B1 (ko) 2012-02-03 2020-03-20 에프. 호프만-라 로슈 아게 항원-형질감염된 t 세포와 함께 사용되는 이중특이적 항체 분자 및 의약에서의 이들의 용도
EP2812357B1 (en) 2012-02-10 2020-11-04 F.Hoffmann-La Roche Ag Single-chain antibodies and other heteromultimers
AU2013216753B2 (en) 2012-02-11 2017-09-21 Genentech, Inc. R-spondin translocations and methods using the same
US9815909B2 (en) * 2012-03-13 2017-11-14 Novimmune S.A. Readily isolated bispecific antibodies with native immunoglobulin format
WO2013139956A1 (en) 2012-03-22 2013-09-26 Thrombogenics Nv Antibodies abrogating cell binding to lactadherin
WO2013148315A1 (en) 2012-03-27 2013-10-03 Genentech, Inc. Diagnosis and treatments relating to her3 inhibitors
AR090549A1 (es) 2012-03-30 2014-11-19 Genentech Inc Anticuerpos anti-lgr5 e inmunoconjugados
WO2013150043A1 (en) 2012-04-05 2013-10-10 F. Hoffmann-La Roche Ag Bispecific antibodies against human tweak and human il17 and uses thereof
RU2014140116A (ru) 2012-04-24 2016-06-10 Тромбодженикс Н.В. Антитела против pdgf-c
WO2013165940A1 (en) 2012-05-01 2013-11-07 Genentech, Inc. Anti-pmel17 antibodies and immunoconjugates
WO2013170191A1 (en) 2012-05-11 2013-11-14 Genentech, Inc. Methods of using antagonists of nad biosynthesis from nicotinamide
AU2013266340A1 (en) * 2012-05-22 2014-12-04 Shire Human Genetic Therapies, Inc. Anti-CCL2 antibodies for treatment of scleroderma
CN104335047B (zh) 2012-05-23 2017-08-15 弗·哈夫曼-拉罗切有限公司 治疗剂的选择方法
KR20150013188A (ko) 2012-05-24 2015-02-04 에프. 호프만-라 로슈 아게 다중특이적 항체
CN104364266A (zh) 2012-06-15 2015-02-18 霍夫曼-拉罗奇有限公司 抗-pcsk9抗体,制剂,剂量给药,和使用方法
US9890215B2 (en) 2012-06-22 2018-02-13 King's College London Vista modulators for diagnosis and treatment of cancer
KR20150030744A (ko) 2012-06-27 2015-03-20 에프. 호프만-라 로슈 아게 표적에 특이적으로 결합하는 하나 이상의 결합 단위를 포함하는 항체 Fc-영역 접합체의 제조 방법 및 그의 용도
KR20150023889A (ko) 2012-06-27 2015-03-05 에프. 호프만-라 로슈 아게 2 개 이상의 상이한 결합 단위를 함유하는 맞춤-제작된 고도로 선별적이고 다중-특이적인 표적화 단위의 선별 및 제조 방법 및 이의 용도
CA2871112C (en) 2012-07-04 2020-05-12 F. Hoffmann-La Roche Ag Covalently linked antigen-antibody conjugates
BR112014030844A2 (pt) 2012-07-04 2019-10-15 Hoffmann La Roche anticorpo anti-biotina humanizado, formulação farmacêutica e uso do anticorpo
JP6324376B2 (ja) 2012-07-04 2018-05-16 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft 抗テオフィリン抗体および使用方法
KR20150030693A (ko) 2012-07-05 2015-03-20 제넨테크, 인크. 발현 및 분비 시스템
SG11201500087VA (en) 2012-07-09 2015-02-27 Genentech Inc Immunoconjugates comprising anti-cd22 antibodies
TR201802376T4 (tr) 2012-07-09 2018-03-21 Genentech Inc Anti-CD79b antikorları içeren immünokonjugatlar.
CN104540526A (zh) 2012-07-09 2015-04-22 基因泰克公司 包含抗cd79b抗体的免疫偶联物
EP2869851A1 (en) 2012-07-09 2015-05-13 Genentech, Inc. Immunoconjugates comprising anti-cd22 antibodies
TW201402608A (zh) 2012-07-12 2014-01-16 Abbvie Inc Il-1結合蛋白質
AU2013289971A1 (en) 2012-07-13 2015-01-22 The Trustees Of The University Of Pennsylvania Enhancing activity of CAR T cells by co-introducing a bispecific antibody
KR101774121B1 (ko) 2012-07-13 2017-09-01 로슈 글리카트 아게 이중특이적 항-vegf/항-ang-2 항체 및 안 혈관 질환의 치료에 있어서 이의 용도
US20150203591A1 (en) * 2012-08-02 2015-07-23 Regeneron Pharmaceuticals, Inc. Mutivalent antigen-binding proteins
CN109793893B (zh) 2012-09-07 2023-05-26 达特茅斯大学理事会 用于诊断和治疗癌症的vista调节剂
WO2014055493A1 (en) 2012-10-02 2014-04-10 Cerulean Pharma Inc. Methods and systems for polymer precipitation and generation of particles
CA2890207A1 (en) 2012-11-05 2014-05-08 Foundation Medicine, Inc. Novel ntrk1 fusion molecules and uses thereof
EP2917243B1 (en) 2012-11-08 2018-03-14 F.Hoffmann-La Roche Ag Her3 antigen binding proteins binding to the beta-hairpin of her3
EP3461501A1 (en) 2012-11-13 2019-04-03 F. Hoffmann-La Roche AG Anti-hemagglutinin antibodies and methods of use
AU2013355324A1 (en) 2012-12-04 2015-05-21 Abbvie Inc. Blood-brain barrier (BBB) penetrating dual specific binding proteins
WO2014106001A2 (en) 2012-12-28 2014-07-03 Abbvie, Inc. Dual specific binding proteins having a receptor sequence
US9458244B2 (en) 2012-12-28 2016-10-04 Abbvie Inc. Single chain multivalent binding protein compositions and methods
WO2014106015A2 (en) 2012-12-28 2014-07-03 Abbvie, Inc. Multivalent binding protein compositions
EP3939614A1 (en) 2013-01-18 2022-01-19 Foundation Medicine, Inc. Methods of treating cholangiocarcinoma
WO2014116596A1 (en) 2013-01-22 2014-07-31 Abbvie, Inc. Methods for optimizing domain stability of binding proteins
WO2014116749A1 (en) 2013-01-23 2014-07-31 Genentech, Inc. Anti-hcv antibodies and methods of using thereof
US10047163B2 (en) 2013-02-08 2018-08-14 Abbvie Stemcentrx Llc Multispecific constructs
CA2900097A1 (en) 2013-02-22 2014-08-28 F. Hoffmann-La Roche Ag Methods of treating cancer and preventing drug resistance
CA2896259A1 (en) 2013-02-26 2014-09-04 Roche Glycart Ag Anti-mcsp antibodies
CA2902263A1 (en) 2013-03-06 2014-09-12 Genentech, Inc. Methods of treating and preventing cancer drug resistance
US10450354B2 (en) 2013-03-12 2019-10-22 Molecular Templates, Inc. CD20-binding immunotoxins for inducing cellular internalization and methods using same
BR112015022576A2 (pt) 2013-03-14 2017-10-24 Genentech Inc produto farmacêutico e seu uso, kit e método para tratar uma disfunção hiperproliferativa
US9562099B2 (en) 2013-03-14 2017-02-07 Genentech, Inc. Anti-B7-H4 antibodies and immunoconjugates
PE20151672A1 (es) 2013-03-14 2015-11-27 Genentech Inc Anticuerpos e inmunoconjugados anti-b7-h4
EP2968565A2 (en) 2013-03-14 2016-01-20 Genentech, Inc. Methods of treating cancer and preventing cancer drug resistance
MX2015012872A (es) 2013-03-15 2016-02-03 Ac Immune Sa Anticuerpos anti-tau y metodos de uso.
MX2015013163A (es) 2013-03-15 2016-04-04 Zyngenia Inc Complejos multiespecificos multivalente y monovalentes y sus usos.
SG11201507423YA (en) 2013-03-15 2015-10-29 Abbvie Inc DUAL SPECIFIC BINDING PROTEINS DIRECTED AGAINST TNFα
MA38495A1 (fr) 2013-03-15 2017-08-31 Genentech Inc Compositions et méthodes pour le diagnostic et le traitement de cancers hépatiques
SG11201507333XA (en) 2013-03-15 2015-10-29 Genentech Inc Biomarkers and methods of treating pd-1 and pd-l1 related conditions
MX2015011899A (es) 2013-03-15 2016-05-05 Genentech Inc Metodos para el tratamiento de cáncer y prevención de resistencia a los fármacos para el cáncer.
JP2016517441A (ja) 2013-03-15 2016-06-16 ジェネンテック, インコーポレイテッド 抗CRTh2抗体及び使用方法
UA118028C2 (uk) 2013-04-03 2018-11-12 Рош Глікарт Аг Біспецифічне антитіло, специфічне щодо fap і dr5, антитіло, специфічне щодо dr5, і спосіб їх застосування
EA201501063A1 (ru) 2013-04-29 2016-05-31 Ф. Хоффманн-Ля Рош Аг СВЯЗЫВАЮЩИЕСЯ С ЧЕЛОВЕЧЕСКИМ FcRn МОДИФИЦИРОВАННЫЕ АНТИТЕЛА И СПОСОБЫ ИХ ПРИМЕНЕНИЯ
EP3878866A1 (en) 2013-04-29 2021-09-15 F. Hoffmann-La Roche AG Fc-receptor binding modified asymmetric antibodies and methods of use
JP2016528168A (ja) 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 FcRn結合が無効となった抗IGF−1R抗体及び血管性眼疾患の処置におけるそれらの使用
CN104341504B (zh) 2013-08-06 2017-10-24 百奥泰生物科技(广州)有限公司 双特异性抗体
WO2015025054A1 (en) 2013-08-22 2015-02-26 Medizinische Universität Wien Dye-specific antibodies for prestained molecular weight markers and methods producing the same
WO2015042108A1 (en) 2013-09-17 2015-03-26 Genentech, Inc. Methods of using anti-lgr5 antibodies
EP3049442A4 (en) 2013-09-26 2017-06-28 Costim Pharmaceuticals Inc. Methods for treating hematologic cancers
EP3052524A2 (en) 2013-10-06 2016-08-10 Abbvie Inc. Dual specific binding proteins directed against immune cell receptors and tlr signaling autoantigens
CA2922912A1 (en) 2013-10-11 2015-04-16 F. Hoffmann-La Roche Ag Multispecific domain exchanged common variable light chain antibodies
JP2016537965A (ja) 2013-10-11 2016-12-08 ジェネンテック, インコーポレイテッド Nsp4阻害剤及び使用方法
KR20160070136A (ko) 2013-10-18 2016-06-17 제넨테크, 인크. 항-rspo2 및/또는 항-rspo3 항체 및 그의 용도
JP6715767B2 (ja) 2013-10-23 2020-07-01 ジェネンテック, インコーポレイテッド 好酸球性疾患の診断及び治療方法
US10519247B2 (en) 2013-11-01 2019-12-31 Board Of Regents,The University Of Texas System Targeting HER2 and HER3 with bispecific antibodies in cancerous cells
PT3071597T (pt) 2013-11-21 2020-10-08 Hoffmann La Roche Anticorpos anti-alfa-sinucleína e métodos de utilização
EP3077418A2 (en) 2013-12-02 2016-10-12 AbbVie Inc. Compositions and methods for treating osteoarthritis
MA39095A1 (fr) 2013-12-13 2018-08-31 Genentech Inc Anticorps et immunoconjugués anti-cd33
SI3083689T1 (sl) 2013-12-17 2020-10-30 Genentech, Inc. Anti-CD3 protitelesa in načini uporabe
PT3083680T (pt) 2013-12-20 2020-03-17 Hoffmann La Roche Anticorpos humanizados anti-tau(ps422) e métodos de utilização
TWI670283B (zh) 2013-12-23 2019-09-01 美商建南德克公司 抗體及使用方法
TW201609805A (zh) 2013-12-23 2016-03-16 美國禮來大藥廠 結合egfr及met之多功能抗體
PL3712174T3 (pl) 2013-12-24 2022-07-04 Janssen Pharmaceutica Nv Przeciwciała i fragmenty anty-vista
US11014987B2 (en) 2013-12-24 2021-05-25 Janssen Pharmaceutics Nv Anti-vista antibodies and fragments, uses thereof, and methods of identifying same
BR112016014945A2 (pt) 2014-01-03 2018-01-23 F. Hoffmann-La Roche Ag conjugado, formulação farmacêutica e uso
CN105899540B (zh) 2014-01-03 2020-02-07 豪夫迈·罗氏有限公司 双特异性抗-半抗原/抗-血脑屏障受体的抗体、其复合物及它们作为血脑屏障穿梭物的应用
KR102278979B1 (ko) 2014-01-03 2021-07-19 에프. 호프만-라 로슈 아게 공유적으로 연결된 헬리카-항-헬리카 항체 접합체 및 그의 용도
CA2932547C (en) 2014-01-06 2023-05-23 F. Hoffmann-La Roche Ag Monovalent blood brain barrier shuttle modules
MA39171A1 (fr) 2014-01-10 2017-07-31 Glaxosmithkline Intellectual Property (No 2) Ltd Dérivés d'hydroxy formamide et leur utilisation
KR20160107190A (ko) 2014-01-15 2016-09-13 에프. 호프만-라 로슈 아게 변형된 FcRn-결합 및 유지된 단백질 A-결합 성질을 갖는 Fc-영역 변이체
WO2015112909A1 (en) 2014-01-24 2015-07-30 Genentech, Inc. Methods of using anti-steap1 antibodies and immunoconjugates
KR20230028808A (ko) 2014-01-27 2023-03-02 몰레큘러 템플레이츠, 인코퍼레이션. 포유류에 적용하기 위한 탈면역된 시가 독소 a 서브유닛 작동체 폴리펩티드
KR20210121288A (ko) 2014-02-08 2021-10-07 제넨테크, 인크. 알츠하이머 질환을 치료하는 방법
BR112016018205A8 (pt) 2014-02-08 2018-04-17 Genentech Inc métodos de tratamento de mal de alzheimer, de seleção de pacientes, de identificação de pacientes, de previsão, de otimização da eficácia terapêutica, kit, uso de agente e uso in vitro de agente
TW201902515A (zh) 2014-02-12 2019-01-16 美商建南德克公司 抗jagged1抗體及使用方法
EP3104882B1 (en) 2014-02-14 2019-06-05 Centrose, Llc Extracellular targeted drug conjugates
UA117608C2 (uk) 2014-02-21 2018-08-27 Дженентек, Інк. Спосіб лікування еозинофільного захворювання у пацієнта шляхом застосування біспецифічного анти-il-13/il-17 антитіла
US11142584B2 (en) 2014-03-11 2021-10-12 Molecular Templates, Inc. CD20-binding proteins comprising Shiga toxin A subunit effector regions for inducing cellular internalization and methods using same
MA39746A (fr) 2014-03-14 2021-04-28 Hoffmann La Roche Compositions de sécrétion de polypeptides hétérologues et procédés associés
US20170107294A1 (en) 2014-03-21 2017-04-20 Nordlandssykehuset Hf Anti-cd14 antibodies and uses thereof
JP2017516458A (ja) 2014-03-24 2017-06-22 ジェネンテック, インコーポレイテッド c−met拮抗剤による癌治療及びc−met拮抗剤のHGF発現との相関
US10308712B2 (en) 2014-03-27 2019-06-04 Bird Rock Bio, Inc. Antibodies that bind human cannabinoid 1 (CB1) receptor
DK3126394T3 (da) 2014-03-31 2020-01-13 Hoffmann La Roche Anti-OX40-antistoffer og fremgangsmåder til anvendelse
AU2015241038A1 (en) 2014-03-31 2016-10-13 Genentech, Inc. Combination therapy comprising anti-angiogenesis agents and OX40 binding agonists
CN106164288A (zh) 2014-04-02 2016-11-23 豪夫迈·罗氏有限公司 检测多特异性抗体轻链错配的方法
UA117289C2 (uk) 2014-04-02 2018-07-10 Ф. Хоффманн-Ля Рош Аг Мультиспецифічне антитіло
AU2015240595B2 (en) 2014-04-03 2020-02-27 Igm Biosciences, Inc. Modified J-chain
US11485790B2 (en) 2014-04-07 2022-11-01 Chugai Seiyaku Kabushiki Kaisha Immunoactivating antigen-binding molecule
BR112016024319B1 (pt) 2014-04-18 2024-01-23 Acceleron Pharma Inc USO DE UMA COMPOSIÇÃO COMPREENDENDO UM ANTAGONISTA DE ActRII PARA A FABRICAÇÃO DE UM MEDICAMENTO PARA TRATAR OU PREVINIR UMA COMPLICAÇÃO DE ANEMIA FALCIFORME
WO2015164615A1 (en) 2014-04-24 2015-10-29 University Of Oslo Anti-gluten antibodies and uses thereof
MA39599A (fr) 2014-05-14 2016-10-05 Merrimack Pharmaceuticals Inc Dosage et administration d'agents thérapeutiques anti-egfr
CA2946662A1 (en) 2014-05-22 2015-11-26 Genentech, Inc. Anti-gpc3 antibodies and immunoconjugates
KR20170005016A (ko) 2014-05-23 2017-01-11 제넨테크, 인크. MiT 바이오마커 및 그의 사용 방법
CN108064242B (zh) 2014-05-28 2022-10-21 阿吉纳斯公司 抗gitr抗体和其使用方法
CN106604934A (zh) 2014-06-11 2017-04-26 分子模板公司 耐受蛋白酶切割的志贺毒素a亚基效应子多肽和包含其的细胞靶向分子
EP3155015A1 (en) 2014-06-11 2017-04-19 F. Hoffmann-La Roche AG Anti-lgr5 antibodies and uses thereof
MX2016016310A (es) 2014-06-11 2017-10-20 A Green Kathy Uso de agonistas y antagonistas vista para suprimir o aumentar la inmunidad humoral.
AU2015274277B2 (en) 2014-06-13 2021-03-18 Acceleron Pharma, Inc. Methods and compositions for treating ulcers
WO2015191986A1 (en) 2014-06-13 2015-12-17 Genentech, Inc. Methods of treating and preventing cancer drug resistance
GB201411320D0 (en) 2014-06-25 2014-08-06 Ucb Biopharma Sprl Antibody construct
AR100978A1 (es) 2014-06-26 2016-11-16 Hoffmann La Roche LANZADERAS CEREBRALES DE ANTICUERPO HUMANIZADO ANTI-Tau(pS422) Y USOS DE LAS MISMAS
RU2705299C2 (ru) 2014-06-26 2019-11-06 Ф. Хоффманн-Ля Рош Аг Антитела против 5-бром-2'-дезоксиуридина и способы применения
TW201623329A (zh) 2014-06-30 2016-07-01 亞佛瑞司股份有限公司 針對骨調素截斷變異體的疫苗及單株抗體暨其用途
ES2785551T3 (es) 2014-06-30 2020-10-07 Glykos Finland Oy Derivado de sacárido de una carga útil tóxica y sus conjugados con anticuerpos
SG11201609707WA (en) 2014-07-01 2017-01-27 Pfizer Bispecific heterodimeric diabodies and uses thereof
US9212225B1 (en) 2014-07-01 2015-12-15 Amphivena Therapeutics, Inc. Bispecific CD33 and CD3 binding proteins
SI3164492T1 (sl) 2014-07-03 2020-02-28 F. Hoffmann-La Roche Ag Sistem polipeptidne ekspresije
EP3166966A1 (en) 2014-07-10 2017-05-17 Affiris AG Substances and methods for the use in prevention and/or treatment in huntington's disease
BR112017000130A2 (pt) 2014-07-11 2018-01-09 Genentech Inc método para atenuar a toxicidade associada à inibição da via de notch e método de tratamento do câncer
MY188940A (en) 2014-07-11 2022-01-13 Ventana Med Syst Inc Anti-pd-l1 antibodies and diagnostic uses thereof
RU2702553C2 (ru) * 2014-07-15 2019-10-08 Астеллас Фарма Инк. Новое антитело против tie-2 человека
EA039598B9 (ru) 2014-09-03 2022-03-10 Бёрингер Ингельхайм Интернациональ Гмбх Соединение, нацеленное на ил-23а и фно-альфа, и его применение
SI3191135T1 (sl) 2014-09-12 2021-01-29 Genentech, Inc. Anti-HER2 protitelesa in imunokonjugati
PE20170670A1 (es) 2014-09-12 2017-06-06 Genentech Inc Anticuerpos anti-cll-1 e inmunoconjugados
CA2957354A1 (en) 2014-09-12 2016-03-17 Genentech, Inc. Cysteine engineered antibodies and conjugates
WO2016040724A1 (en) 2014-09-12 2016-03-17 Genentech, Inc. Anti-b7-h4 antibodies and immunoconjugates
US11344620B2 (en) 2014-09-13 2022-05-31 Novartis Ag Combination therapies
WO2016044396A1 (en) 2014-09-17 2016-03-24 Genentech, Inc. Immunoconjugates comprising anti-her2 antibodies and pyrrolobenzodiazepines
LT3262071T (lt) 2014-09-23 2020-06-25 F. Hoffmann-La Roche Ag Anti-cd79b imunokonjugatų naudojimo būdai
CN107074938A (zh) 2014-10-16 2017-08-18 豪夫迈·罗氏有限公司 抗‑α‑突触核蛋白抗体和使用方法
BR112017009151A2 (pt) 2014-11-03 2018-03-06 Genentech, Inc. ensaios para detectar subgrupos imunológicos de célula t e métodos de uso dos mesmos
AU2015343339A1 (en) 2014-11-03 2017-06-15 Genentech, Inc. Methods and biomarkers for predicting efficacy and evaluation of an OX40 agonist treatment
AR102417A1 (es) 2014-11-05 2017-03-01 Lilly Co Eli Anticuerpos biespecíficos anti-tnf- / anti-il-23
WO2016073157A1 (en) 2014-11-06 2016-05-12 Genentech, Inc. Anti-ang2 antibodies and methods of use thereof
RU2714116C2 (ru) 2014-11-06 2020-02-11 Ф. Хоффманн-Ля Рош Аг ВАРИАНТЫ Fc-ОБЛАСТИ С МОДИФИЦИРОВАННЫМ СВЯЗЫВАНИЕМ FcRn И СПОСОБЫ ИХ ПРИМЕНЕНИЯ
AR102522A1 (es) 2014-11-06 2017-03-08 Hoffmann La Roche Variantes de región fc con propiedades modificadas de unión a fcrn y proteína a
EP3552488A1 (en) 2014-11-10 2019-10-16 F. Hoffmann-La Roche AG Animal model for nephropathy and agents for treating the same
TWI705976B (zh) 2014-11-10 2020-10-01 美商建南德克公司 抗介白素-33抗體及其用途
NZ767672A (en) * 2014-11-14 2024-02-23 Hoffmann La Roche Antigen binding molecules comprising a tnf family ligand trimer
WO2016081640A1 (en) 2014-11-19 2016-05-26 Genentech, Inc. Anti-transferrin receptor / anti-bace1 multispecific antibodies and methods of use
EP3221364B1 (en) 2014-11-19 2020-12-16 Genentech, Inc. Antibodies against bace1 and use thereof for neural disease immunotherapy
BR112017010324A2 (pt) 2014-11-20 2018-05-15 F. Hoffmann-La Roche Ag método para tratar ou retardar a progressão de um câncer em um indivíduo, moléculas, métodos para aumentar a função imune em um indivíduo e para selecionar um paciente para tratamento, kits, composição farmacêutica e usos de uma combinação de uma molécula
EP3227341A1 (en) 2014-12-02 2017-10-11 CeMM - Forschungszentrum für Molekulare Medizin GmbH Anti-mutant calreticulin antibodies and their use in the diagnosis and therapy of myeloid malignancies
MA41119A (fr) 2014-12-03 2017-10-10 Acceleron Pharma Inc Méthodes de traitement de syndromes myélodysplasiques et d'anémie sidéroblastique
EP3227332B1 (en) 2014-12-03 2019-11-06 F.Hoffmann-La Roche Ag Multispecific antibodies
AR102918A1 (es) 2014-12-05 2017-04-05 Genentech Inc Anticuerpos anti-cd79b y métodos de uso
MX2017007136A (es) 2014-12-05 2017-12-04 Immunext Inc Identificacion de vsig8 como el receptor vista putativo y su uso para producir moduladores vista/vsig8.
WO2016094566A2 (en) 2014-12-10 2016-06-16 Genentech, Inc. Blood brain barrier receptor antibodies and methods of use
US9993535B2 (en) 2014-12-18 2018-06-12 Siwa Corporation Method and composition for treating sarcopenia
US10358502B2 (en) 2014-12-18 2019-07-23 Siwa Corporation Product and method for treating sarcopenia
BR112017011170A2 (pt) 2014-12-18 2018-02-27 Hoffmann La Roche método para determinar a citotoxicidade dependente do complemento de uma composição
AU2015365167B2 (en) 2014-12-19 2021-07-29 Chugai Seiyaku Kabushiki Kaisha Anti-C5 antibodies and methods of use
NZ732922A (en) 2015-01-20 2022-07-29 Igm Biosciences Inc Tumor necrosis factor (tnf) superfamily receptor binding molecules and uses thereof
US20180016327A1 (en) 2015-01-22 2018-01-18 Chugai Seiyaku Kabushiki Kaisha A Combination of Two or More Anti-C5 Antibodies and Methods of Use
WO2016122738A1 (en) 2015-01-31 2016-08-04 The Trustees Of The University Of Pennsylvania Compositions and methods for t cell delivery of therapeutic molecules
EA201791754A1 (ru) 2015-02-05 2019-01-31 Чугаи Сейяку Кабусики Кайся АНТИТЕЛА, СОДЕРЖАЩИЕ ЗАВИСЯЩИЙ ОТ КОНЦЕНТРАЦИИ ИОНОВ АНТИГЕНСВЯЗЫВАЮЩИЙ ДОМЕН, ВАРИАНТЫ Fc-ОБЛАСТИ, IL-8-СВЯЗЫВАЮЩИЕ АНТИТЕЛА И ИХ ПРИМЕНЕНИЯ
EP3795594A1 (en) 2015-02-05 2021-03-24 Molecular Templates, Inc. Multivalent cd20-binding molecules comprising shiga toxin a subunit effector regions and enriched compositions thereof
KR20170138410A (ko) 2015-02-23 2017-12-15 시걸 테라퓨틱스 에스에이에스 비-천연 세마포린 3 및 이의 의학적 용도
US10787520B2 (en) 2015-03-04 2020-09-29 Igm Biosciences, Inc. Multimeric bispecific binding molecules specific for CD20 and CD3
CA2978256A1 (en) 2015-03-06 2016-09-15 Genentech, Inc. Ultrapurified dsba and dsbc and methods of making and using the same
WO2016149276A1 (en) 2015-03-16 2016-09-22 Genentech, Inc. Methods of detecting and quantifying il-13 and uses in diagnosing and treating th2-associated diseases
WO2016146833A1 (en) 2015-03-19 2016-09-22 F. Hoffmann-La Roche Ag Biomarkers for nad(+)-diphthamide adp ribosyltransferase resistance
EP3828199A1 (en) 2015-04-06 2021-06-02 Acceleron Pharma Inc. Alk7: actriib heteromultimers and uses thereof
MA41919A (fr) 2015-04-06 2018-02-13 Acceleron Pharma Inc Hétéromultimères alk4:actriib et leurs utilisations
WO2016164480A1 (en) 2015-04-07 2016-10-13 Genentech, Inc. Antigen binding complex having agonistic activity and methods of use
RS62546B1 (sr) 2015-04-07 2021-12-31 Alector Llc Antitela protiv sortilina i postupci za njihovu upotrebu
EP3283523A1 (en) 2015-04-17 2018-02-21 Elsalys Biotech Anti-tyro3 antibodies and uses thereof
WO2016172551A2 (en) 2015-04-24 2016-10-27 Genentech, Inc. Methods of identifying bacteria comprising binding polypeptides
CA2984794A1 (en) 2015-05-07 2016-11-10 Agenus Inc. Anti-ox40 antibodies and methods of use thereof
US10570185B2 (en) 2015-05-11 2020-02-25 Northwestern University Method to detect autoantibody reactivity for deamidated insulin autoantigen in diabetes
CN107592812A (zh) 2015-05-11 2018-01-16 豪夫迈·罗氏有限公司 治疗狼疮性肾炎的组合物和方法
HUE051815T2 (hu) 2015-05-12 2021-03-29 Hoffmann La Roche Terápiás és diagnosztikai eljárások rákra
CN107771182A (zh) 2015-05-29 2018-03-06 豪夫迈·罗氏有限公司 人源化抗埃博拉病毒糖蛋白抗体和使用方法
CA2984003A1 (en) 2015-05-29 2016-12-08 Genentech, Inc. Therapeutic and diagnostic methods for cancer
EP3302555A4 (en) 2015-05-29 2018-07-11 Amphivena Therapeutics, Inc. Methods of using bispecific cd33 and cd3 binding proteins
PL3303373T3 (pl) 2015-05-30 2020-09-21 Molecular Templates, Inc. Deimmunizowane rusztowania podjednostki a toksyny shiga oraz zawierające je cząsteczki nakierowane na komórki
CN107810012A (zh) 2015-06-02 2018-03-16 豪夫迈·罗氏有限公司 使用抗il‑34抗体治疗神经疾病的组合物和方法
EP3302527B1 (en) 2015-06-03 2020-01-08 The Medical College of Wisconsin, Inc. An engineered ccl20 locked dimer polypeptide
US11571462B2 (en) 2015-06-03 2023-02-07 The Medical College Of Wisconsin, Inc. Engineered CCL20 locked dimer polypeptide
TWI827405B (zh) 2015-06-05 2023-12-21 美商建南德克公司 抗-tau抗體及使用方法
WO2016200835A1 (en) 2015-06-08 2016-12-15 Genentech, Inc. Methods of treating cancer using anti-ox40 antibodies and pd-1 axis binding antagonists
KR20180011839A (ko) 2015-06-08 2018-02-02 제넨테크, 인크. 항-ox40 항체를 이용한 암의 치료 방법
US10017577B2 (en) 2015-06-15 2018-07-10 Genentech, Inc. Antibodies and immunoconjugates
WO2016204966A1 (en) 2015-06-16 2016-12-22 Genentech, Inc. Anti-cd3 antibodies and methods of use
WO2016205200A1 (en) 2015-06-16 2016-12-22 Genentech, Inc. Anti-cll-1 antibodies and methods of use
MX2017016169A (es) 2015-06-17 2018-08-15 Genentech Inc Anticuerpos anti-her2 y metodos de uso.
ES2809728T3 (es) 2015-06-24 2021-03-05 Hoffmann La Roche Anticuerpos anti-tau(pS422) humanizados y procedimientos de uso
EP3313882B1 (en) 2015-06-24 2020-03-11 Janssen Pharmaceutica NV Anti-vista antibodies and fragments
CN107810420B (zh) 2015-06-25 2020-08-14 豪夫迈·罗氏有限公司 用于测定抗体或配体结合和功能的基于细胞的测定
AU2016288461B2 (en) 2015-06-29 2021-10-07 Ventana Medical Systems, Inc. Materials and methods for performing histochemical assays for human pro-epiregulin and amphiregulin
US20170029520A1 (en) 2015-06-29 2017-02-02 Genentech, Inc. Compositions and methods for use in organ transplantation
EP3112381A1 (en) * 2015-07-01 2017-01-04 FONDAZIONE IRCCS Istituto Nazionale dei Tumori Bispecific antibodies for use in cancer immunotherapy
IL257030B2 (en) 2015-07-23 2023-03-01 Inhibrx Inc Multivalent and multispecific cleavage proteins that bind to gitr, preparations containing them and their uses
JP7320350B2 (ja) 2015-08-04 2023-08-03 アクセルロン ファーマ インコーポレイテッド 骨髄増殖性障害を処置するための方法
CN108136012B (zh) 2015-08-07 2022-10-04 伊麦吉纳博公司 靶向分子的抗原结合构建体
CN105384825B (zh) 2015-08-11 2018-06-01 南京传奇生物科技有限公司 一种基于单域抗体的双特异性嵌合抗原受体及其应用
EP3340995A4 (en) 2015-08-28 2019-04-03 The Trustees Of The University Of Pennsylvania METHODS AND COMPOSITIONS FOR CELLS EXPRESSING A CHIMERIC INTRACELLULAR SIGNALING MOLECULE
US11649435B2 (en) 2015-08-28 2023-05-16 The Trustees Of The University Of Pennsylvania Methods and compositions for cells expressing a chimeric intracellular signaling molecule
CN113372443A (zh) 2015-09-18 2021-09-10 中外制药株式会社 Il-8-结合抗体及其应用
WO2017050729A1 (en) 2015-09-22 2017-03-30 Spring Bioscience Corporation Anti-ox40 antibodies and diagnostic uses thereof
MY191756A (en) 2015-09-23 2022-07-14 Genentech Inc Optimized variants of anti-vegf antibodies
RU2757135C2 (ru) 2015-09-24 2021-10-11 АБВИТРО ЭлЭлСи Композиции антител к вич и способы их применения
CN113956358A (zh) 2015-09-25 2022-01-21 豪夫迈·罗氏有限公司 抗tigit抗体和使用方法
WO2017059387A1 (en) 2015-09-30 2017-04-06 Igm Biosciences, Inc. Binding molecules with modified j-chain
AU2016329197B2 (en) 2015-09-30 2021-01-21 Igm Biosciences, Inc. Binding molecules with modified J-chain
WO2017058771A1 (en) 2015-09-30 2017-04-06 Bird Rock Bio, Inc. Antibodies that bind human cannabinoid 1 (cb1) recetor
AR106188A1 (es) 2015-10-01 2017-12-20 Hoffmann La Roche Anticuerpos anti-cd19 humano humanizados y métodos de utilización
MA43345A (fr) 2015-10-02 2018-08-08 Hoffmann La Roche Conjugués anticorps-médicaments de pyrrolobenzodiazépine et méthodes d'utilisation
EP3356404B1 (en) 2015-10-02 2021-08-18 F. Hoffmann-La Roche AG Anti-pd1 antibodies and methods of use
EP3150637A1 (en) 2015-10-02 2017-04-05 F. Hoffmann-La Roche AG Multispecific antibodies
EP3150636A1 (en) 2015-10-02 2017-04-05 F. Hoffmann-La Roche AG Tetravalent multispecific antibodies
WO2017060144A1 (en) * 2015-10-07 2017-04-13 F. Hoffmann-La Roche Ag Bispecific antibodies with tetravalency for a costimulatory tnf receptor
MA43354A (fr) 2015-10-16 2018-08-22 Genentech Inc Conjugués médicamenteux à pont disulfure encombré
MA45326A (fr) 2015-10-20 2018-08-29 Genentech Inc Conjugués calichéamicine-anticorps-médicament et procédés d'utilisation
SG10201913247XA (en) 2015-10-23 2020-02-27 Eureka Therapeutics Inc Antibody/t-cell receptor chimeric constructs and uses thereof
MX2018005036A (es) 2015-10-29 2018-08-01 Hoffmann La Roche Anticuerpos y metodos de uso de anti-regiones de fragmentos cristalizables (fc) variantes.
EP3184547A1 (en) 2015-10-29 2017-06-28 F. Hoffmann-La Roche AG Anti-tpbg antibodies and methods of use
RS61871B1 (sr) 2015-10-30 2021-06-30 Hoffmann La Roche Anti-htra1 antitela i postupci za njihovu primenu
US10407510B2 (en) 2015-10-30 2019-09-10 Genentech, Inc. Anti-factor D antibodies and conjugates
EP3371217A1 (en) 2015-11-08 2018-09-12 H. Hoffnabb-La Roche Ag Methods of screening for multispecific antibodies
WO2017085693A1 (en) 2015-11-19 2017-05-26 AbbVie Deutschland GmbH & Co. KG Reporter gene assay methods for identifying and analyzing multi-specific binding proteins
CN106729743B (zh) 2015-11-23 2021-09-21 四川科伦博泰生物医药股份有限公司 抗ErbB2抗体-药物偶联物及其组合物、制备方法和应用
US10550170B2 (en) 2015-11-23 2020-02-04 Acceleron Pharma Inc. Methods for treating vascular eye disorders with actrii antagonists
JP7089470B2 (ja) 2015-12-02 2022-06-22 アジェナス インコーポレイテッド 抗体およびその使用方法
EP3383431A4 (en) * 2015-12-02 2019-08-28 Agenus Inc. ANTI-GITR ANTIBODIES AND METHOD OF USE THEREOF
EP3178848A1 (en) 2015-12-09 2017-06-14 F. Hoffmann-La Roche AG Type ii anti-cd20 antibody for reducing formation of anti-drug antibodies
PE20240365A1 (es) 2015-12-18 2024-03-04 Chugai Pharmaceutical Co Ltd Anticuerpos anti-c5 y metodos de uso
EP3405489A1 (en) 2016-01-20 2018-11-28 Genentech, Inc. High dose treatments for alzheimer's disease
TWI756204B (zh) 2016-02-12 2022-03-01 比利時商楊森製藥公司 抗vista抗體及片段、其用途及鑑定其之方法
ES2912064T3 (es) 2016-02-19 2022-05-24 Siwa Corp Método y composición para tratar el cáncer, destruir las células cancerosas metastásicas y evitar la metástasis del cáncer usando anticuerpo para productos finales de glicación avanzada (AGE)
MX2018010361A (es) 2016-02-29 2019-07-08 Genentech Inc Métodos terapéuticos y de diagnóstico para el cáncer.
EP3216458A1 (en) 2016-03-07 2017-09-13 Max-Planck-Gesellschaft zur Förderung der Wissenschaften e.V. Modified vascular endothelial growth factor a (vegf-a) and its medical use
US11291721B2 (en) 2016-03-21 2022-04-05 Marengo Therapeutics, Inc. Multispecific and multifunctional molecules and uses thereof
JP6943872B2 (ja) 2016-03-25 2021-10-06 ジェネンテック, インコーポレイテッド 多重全抗体及び抗体複合体化薬物定量化アッセイ
EP3865511A1 (en) 2016-04-14 2021-08-18 F. Hoffmann-La Roche AG Anti-rspo3 antibodies and methods of use
WO2017180842A1 (en) 2016-04-15 2017-10-19 Bioatla, Llc Anti-axl antibodies, antibody fragments and their immunoconjugates and uses thereof
CN117562992A (zh) 2016-04-15 2024-02-20 伊穆奈克斯特股份有限公司 抗人vista抗体及其用途
WO2017181116A1 (en) 2016-04-15 2017-10-19 Siwa Corporation Anti-age antibodies for treating neurodegenerative disorders
EP3443120A2 (en) 2016-04-15 2019-02-20 H. Hoffnabb-La Roche Ag Methods for monitoring and treating cancer
CN109154613A (zh) 2016-04-15 2019-01-04 豪夫迈·罗氏有限公司 用于监测和治疗癌症的方法
CN109071640B (zh) 2016-05-11 2022-10-18 豪夫迈·罗氏有限公司 经修饰抗生腱蛋白抗体及使用方法
KR20190007026A (ko) 2016-05-17 2019-01-21 제넨테크, 인크. 면역요법에서의 진단 및 사용을 위한 기질 유전자 특질
JP2019522633A (ja) 2016-05-20 2019-08-15 ジェネンテック, インコーポレイテッド Protac抗体コンジュゲート及び使用方法
AU2017271588B2 (en) 2016-05-27 2022-01-20 Agenus Inc. Anti-TIM-3 antibodies and methods of use thereof
EP3465221B1 (en) 2016-05-27 2020-07-22 H. Hoffnabb-La Roche Ag Bioanalytical method for the characterization of site-specific antibody-drug conjugates
CA3059010A1 (en) 2016-06-02 2018-12-06 F. Hoffmann-La Roche Ag Type ii anti-cd20 antibody and anti-cd20/cd3 bispecific antibody for treatment of cancer
EP3252078A1 (en) 2016-06-02 2017-12-06 F. Hoffmann-La Roche AG Type ii anti-cd20 antibody and anti-cd20/cd3 bispecific antibody for treatment of cancer
EP3464280B1 (en) 2016-06-06 2021-10-06 F. Hoffmann-La Roche AG Silvestrol antibody-drug conjugates and methods of use
CA3026518A1 (en) 2016-06-17 2017-12-21 Genentech, Inc. Purification of multispecific antibodies
KR20190053835A (ko) 2016-06-21 2019-05-20 테네오바이오, 인코포레이티드 Cd3 결합 항체
JP2019518763A (ja) 2016-06-23 2019-07-04 シワ コーポレーション 様々な疾患及び障害の治療において使用するためのワクチン
CN116143918A (zh) 2016-06-24 2023-05-23 豪夫迈·罗氏有限公司 抗聚泛素多特异性抗体
EP3496739B1 (en) 2016-07-15 2021-04-28 Acceleron Pharma Inc. Compositions comprising actriia polypeptides for use in treating pulmonary hypertension
WO2018014260A1 (en) 2016-07-20 2018-01-25 Nanjing Legend Biotech Co., Ltd. Multispecific antigen binding proteins and methods of use thereof
JP7264580B2 (ja) 2016-07-27 2023-04-25 アクセルロン ファーマ インコーポレイテッド 骨髄線維症を処置するための方法および組成物
CA3032146A1 (en) 2016-08-03 2018-02-08 Bio-Techne Corporation Identification of vsig3/vista as a novel immune checkpoint and use thereof for immunotherapy
KR20230079499A (ko) 2016-08-05 2023-06-07 추가이 세이야쿠 가부시키가이샤 Il-8 관련 질환의 치료용 또는 예방용 조성물
KR20230044038A (ko) 2016-08-09 2023-03-31 키맵 리미티드 항-icos 항체
EP3496763A1 (en) 2016-08-11 2019-06-19 Genentech, Inc. Pyrrolobenzodiazepine prodrugs and antibody conjugates thereof
ES2963385T3 (es) * 2016-08-16 2024-03-26 Epimab Biotherapeutics Inc Anticuerpos biespecíficos Fab en tándem asimétricos monovalentes
WO2018038046A1 (ja) 2016-08-22 2018-03-01 中外製薬株式会社 ヒトgpc3ポリペプチドを発現する遺伝子改変非ヒト動物
UA126384C2 (uk) 2016-09-14 2022-09-28 Тенеобіо, Інк. Антитіло, яке зв'язує cd3
SG10201607778XA (en) 2016-09-16 2018-04-27 Chugai Pharmaceutical Co Ltd Anti-Dengue Virus Antibodies, Polypeptides Containing Variant Fc Regions, And Methods Of Use
JP6976315B2 (ja) 2016-09-19 2021-12-08 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft 補体因子に基づくアフィニティークロマトグラフィー
RU2752785C2 (ru) 2016-09-23 2021-08-04 Дженентек, Инк. Применение антагонистов il-13 для лечения атопического дерматита
WO2018065501A1 (en) 2016-10-05 2018-04-12 F. Hoffmann-La Roche Ag Methods for preparing antibody drug conjugates
US11976111B2 (en) 2016-10-05 2024-05-07 Acceleron Pharma Inc. ActRIIa and ALK4 polypeptides for treating kidney fibrosis, inflammation and injury
JP2019535237A (ja) 2016-10-06 2019-12-12 ジェネンテック, インコーポレイテッド がんのための治療方法及び診断方法
WO2018068201A1 (en) 2016-10-11 2018-04-19 Nanjing Legend Biotech Co., Ltd. Single-domain antibodies and variants thereof against ctla-4
BR112019006876A2 (pt) 2016-10-13 2019-06-25 Symphogen As anticorpos e composições anti-lag-3
CN110267678A (zh) 2016-10-29 2019-09-20 霍夫曼-拉罗奇有限公司 抗mic抗体和使用方法
MA46770A (fr) 2016-11-09 2019-09-18 Agenus Inc Anticorps anti-ox40, anticorps anti-gitr, et leurs procédés d'utilisation
WO2018093821A1 (en) 2016-11-15 2018-05-24 Genentech, Inc. Dosing for treatment with anti-cd20/anti-cd3 bispecific antibodies
TW201829463A (zh) 2016-11-18 2018-08-16 瑞士商赫孚孟拉羅股份公司 抗hla-g抗體及其用途
JOP20190100A1 (ar) 2016-11-19 2019-05-01 Potenza Therapeutics Inc بروتينات ربط مولد ضد مضاد لـ gitr وطرق استخدامها
JP7227146B2 (ja) 2016-11-23 2023-02-21 バイオベラティブ セラピューティクス インコーポレイテッド 凝固第ix因子および凝固第x因子に結合する二重特異性抗体
EP3608333A1 (en) 2016-12-07 2020-02-12 Molecular Templates, Inc. Shiga toxin a subunit effector polypeptides, shiga toxin effector scaffolds, and cell-targeting molecules for site-specific conjugation
JP7193457B2 (ja) 2016-12-07 2022-12-20 ジェネンテック, インコーポレイテッド 抗tau抗体及び使用方法
KR20240035636A (ko) 2016-12-07 2024-03-15 제넨테크, 인크. 항-타우 항체 및 이의 이용 방법
JP7141407B2 (ja) 2016-12-13 2022-09-22 エフ.ホフマン-ラ ロシュ アーゲー 腫瘍試料における標的抗原の存在を決定する方法
JP7125400B2 (ja) 2016-12-19 2022-08-24 エフ・ホフマン-ラ・ロシュ・アクチェンゲゼルシャフト 標的化4-1bb(cd137)アゴニストとの併用療法
PL3559034T3 (pl) 2016-12-20 2021-04-19 F. Hoffmann-La Roche Ag Terapia skojarzona dwuswoistymi przeciwciałami anty-CD20/anty-CD3 i agonistami 4-1BB (CD137)
WO2018114877A1 (en) 2016-12-21 2018-06-28 F. Hoffmann-La Roche Ag In vitro glycoengineering of antibodies
JP7215999B2 (ja) 2016-12-21 2023-01-31 エフ.ホフマン-ラ ロシュ アーゲー 抗体またはリガンドの結合および機能を決定するためのアッセイ法
KR102390246B1 (ko) 2016-12-21 2022-04-22 에프. 호프만-라 로슈 아게 항체의 시험관내 글리코조작에 있어서의 효소의 재사용
CA3045970A1 (en) 2016-12-21 2018-06-28 F. Hoffmann-La Roche Ag Method for in vitro glycoengineering of antibodies
CA3048174A1 (en) 2016-12-22 2018-06-28 Daiichi Sankyo Company, Limited Anti-cd3 antibody and molecules comprising the antibody
BR112019014147A2 (pt) * 2017-01-09 2020-02-11 Biomunex Pharmaceuticals Ligante polipeptídico para a preparação de anticorpos multiespecíficos
MX2019008840A (es) 2017-01-25 2019-09-09 Molecular Templates Inc Moleculas con direccion hacia las celulas que comprenden efectores de la sub-unidad a de la toxina shiga desinmunizados y epitopos de las celulas t cd8+.
CA3049806A1 (en) 2017-01-30 2018-08-02 Alexion Pharmaceuticals, Inc. Monovalent anti-properdin antibodies and antibody fragments
US11266745B2 (en) 2017-02-08 2022-03-08 Imaginab, Inc. Extension sequences for diabodies
KR102362006B1 (ko) 2017-02-10 2022-02-14 제넨테크, 인크. 항-트립타제 항체, 이의 조성물, 및 그의 용도
US20200291089A1 (en) 2017-02-16 2020-09-17 Elstar Therapeutics, Inc. Multifunctional molecules comprising a trimeric ligand and uses thereof
ES2953595T3 (es) 2017-03-01 2023-11-14 Hoffmann La Roche Procedimientos diagnósticos y terapéuticos para el cáncer
US11578136B2 (en) * 2017-03-16 2023-02-14 Innate Pharma Compositions and methods for treating cancer
WO2018175752A1 (en) 2017-03-22 2018-09-27 Genentech, Inc. Optimized antibody compositions for treatment of ocular disorders
SG11201908547VA (en) 2017-03-22 2019-10-30 Genentech Inc Hydrogel cross-linked hyaluronic acid prodrug compositions and methods
CN110494446A (zh) 2017-03-28 2019-11-22 基因泰克公司 治疗神经退行性疾病的方法
MA49289A (fr) 2017-04-03 2020-02-12 Hoffmann La Roche Anticorps se liant à steap-1
WO2018184964A1 (en) 2017-04-03 2018-10-11 F. Hoffmann-La Roche Ag Immunoconjugates of an anti-pd-1 antibody with a mutant il-2 or with il-15
PE20191463A1 (es) 2017-04-05 2019-10-16 Hoffmann La Roche Anticuerpos biespecificos de union especifica pd1 y lag3
TWI707871B (zh) 2017-04-05 2020-10-21 瑞士商赫孚孟拉羅股份公司 抗lag3抗體
CA3058960C (en) 2017-04-05 2023-08-29 Symphogen A/S Combination therapies targeting pd-1, tim-3, and lag-3
EP3619235A1 (en) 2017-04-11 2020-03-11 Inhibrx, Inc. Multispecific polypeptide constructs having constrained cd3 binding and methods of using the same
EP3609923A1 (en) 2017-04-13 2020-02-19 Siwa Corporation Humanized monoclonal advanced glycation end-product antibody
US20190078160A1 (en) 2017-04-21 2019-03-14 Genentech, Inc. Use of klk5 antagonists for treatment of a disease
IL310182A (en) 2017-04-26 2024-03-01 Eureka Therapeutics Inc ׂ A Delaware Corp Structures of chimeric antibody/T-cell receptor and uses thereof
EP3630836A1 (en) 2017-05-31 2020-04-08 Elstar Therapeutics, Inc. Multispecific molecules that bind to myeloproliferative leukemia (mpl) protein and uses thereof
EP3642237A2 (en) 2017-06-20 2020-04-29 Teneobio, Inc. Anti-bcma heavy chain-only antibodies
JP7369113B2 (ja) 2017-07-20 2023-10-25 アプティーボ リサーチ アンド デベロップメント エルエルシー 癌胎児性抗原結合タンパク質、関連する化合物および方法
US11674962B2 (en) 2017-07-21 2023-06-13 Genentech, Inc. Therapeutic and diagnostic methods for cancer
WO2019028292A1 (en) 2017-08-03 2019-02-07 Alector Llc ANTI-TREM2 ANTIBODIES AND METHODS OF USE
WO2019035938A1 (en) 2017-08-16 2019-02-21 Elstar Therapeutics, Inc. MULTISPECIFIC MOLECULES BINDING TO BCMA AND USES THEREOF
US11814432B2 (en) 2017-09-20 2023-11-14 The University Of British Columbia Anti-HLA-A2 antibodies, related chimeric antigen receptors, and uses thereof
CA3075450A1 (en) 2017-09-22 2019-03-28 F. Hoffmann-La Roche Ag Multivalent mono- or bispecific recombinant antibodies for analytic purpose
MA50667A (fr) 2017-09-29 2020-08-05 Chugai Pharmaceutical Co Ltd Molécule multispécifique de liaison à l'antigène ayant une activité de substitution de la fonction de cofacteur du facteur viii de coagulation sanguine (fviii), et formulation pharmaceutique contenant ladite molécule en tant que principe actif
JP2020536967A (ja) 2017-10-12 2020-12-17 イミュノウェイク インコーポレイテッド Vegfr−抗体軽鎖融合タンパク質
CN111315781A (zh) 2017-11-01 2020-06-19 豪夫迈·罗氏有限公司 用靶向性ox40激动剂的组合疗法
TW201923089A (zh) 2017-11-06 2019-06-16 美商建南德克公司 癌症之診斷及治療方法
JP7311425B2 (ja) 2017-11-08 2023-07-19 協和キリン株式会社 CD40とEpCAMに結合するバイスペシフィック抗体
US11952422B2 (en) 2017-12-05 2024-04-09 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule comprising altered antibody variable region binding CD3 and CD137
SG11202005557TA (en) 2017-12-12 2020-07-29 Macrogenics Inc Bispecific cd 16-binding molecules and their use in the treatment of disease
US11629189B2 (en) 2017-12-19 2023-04-18 Kymab Limited Bispecific antibody for ICOS and PD-L1
BR112020011469A2 (pt) 2017-12-21 2020-11-24 F. Hoffmann-La Roche Ag anticorpos, molécula de ligação ao antígeno biespecífica, um ou mais polinucleotídeos isolados, um ou mais vetores, célula hospedeira, método para produzir um anticorpo, composição farmacêutica, uso do anticorpo, método de tratamento de uma doença e invenção
WO2019126472A1 (en) 2017-12-22 2019-06-27 Genentech, Inc. Use of pilra binding agents for treatment of a disease
JP7074859B2 (ja) 2017-12-22 2022-05-24 エフ.ホフマン-ラ ロシュ アーゲー 疎水性相互作用クロマトグラフィーによる軽鎖誤対合抗体変種の枯渇の方法
US11518801B1 (en) 2017-12-22 2022-12-06 Siwa Corporation Methods and compositions for treating diabetes and diabetic complications
SG11202005880XA (en) 2017-12-22 2020-07-29 Teneobio Inc Heavy chain antibodies binding to cd22
CN111542543B (zh) 2017-12-28 2023-12-22 南京传奇生物科技有限公司 针对pd-l1的抗体及其变体
CN111699198B (zh) 2017-12-28 2023-09-05 南京传奇生物科技有限公司 针对tigit的单域抗体和其变体
CN111479588A (zh) 2017-12-29 2020-07-31 豪夫迈·罗氏有限公司 用于改善抗vegf抗体的vegf受体阻断选择性的方法
JP2021508498A (ja) 2017-12-29 2021-03-11 アレクター リミテッド ライアビリティ カンパニー 抗tmem106b抗体及びその使用方法
EP3740507A4 (en) 2018-01-15 2022-08-24 Nanjing Legend Biotech Co., Ltd. SINGLE DOMAIN ANTIBODIES AND VARIANTS THEREOF AGAINST PD-1
EP3746476A1 (en) 2018-01-31 2020-12-09 Alector LLC Anti-ms4a4a antibodies and methods of use thereof
JP2021511782A (ja) 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 安定化された免疫グロブリンドメイン
WO2019149716A1 (en) 2018-01-31 2019-08-08 F. Hoffmann-La Roche Ag Bispecific antibodies comprising an antigen-binding site binding to lag3
WO2019152810A1 (en) 2018-02-02 2019-08-08 Bio-Techne Corporation Compounds that modulate the interaction of vista and vsig3 and methods of making and using
US11866498B2 (en) 2018-02-08 2024-01-09 Genentech, Inc. Bispecific antigen-binding molecules and methods of use
KR102417088B1 (ko) 2018-02-09 2022-07-07 제넨테크, 인크. 비만 세포 매개 염증성 질환에 대한 치료 및 진단 방법
TWI829667B (zh) 2018-02-09 2024-01-21 瑞士商赫孚孟拉羅股份公司 結合gprc5d之抗體
AU2019231791B2 (en) 2018-03-09 2022-08-11 Agenus Inc. Anti-CD73 antibodies and methods of use thereof
EP3765501A1 (en) 2018-03-13 2021-01-20 F. Hoffmann-La Roche AG Combination therapy with targeted 4-1bb (cd137) agonists
WO2019175071A1 (en) 2018-03-13 2019-09-19 F. Hoffmann-La Roche Ag Therapeutic combination of 4-1 bb agonists with anti-cd20 antibodies
US20200040103A1 (en) 2018-03-14 2020-02-06 Genentech, Inc. Anti-klk5 antibodies and methods of use
WO2019178362A1 (en) 2018-03-14 2019-09-19 Elstar Therapeutics, Inc. Multifunctional molecules that bind to calreticulin and uses thereof
WO2019178364A2 (en) 2018-03-14 2019-09-19 Elstar Therapeutics, Inc. Multifunctional molecules and uses thereof
CA3093729A1 (en) 2018-03-15 2019-09-19 Chugai Seiyaku Kabushiki Kaisha Anti-dengue virus antibodies having cross-reactivity to zika virus and methods of use
AU2019245243A1 (en) 2018-03-29 2020-09-03 Genentech, Inc Modulating lactogenic activity in mammalian cells
KR20200138720A (ko) 2018-03-30 2020-12-10 난징 레전드 바이오테크 씨오., 엘티디. Lag-3에 대한 단일-도메인 항체 및 이의 용도
WO2019190327A2 (en) * 2018-03-30 2019-10-03 Merus N.V. Multivalent antibody
TW202011029A (zh) 2018-04-04 2020-03-16 美商建南德克公司 偵測及定量fgf21之方法
CN110612117B (zh) 2018-04-17 2024-04-12 分子模板公司 包含去免疫化的志贺毒素a亚基支架的her2靶向分子
AR114789A1 (es) 2018-04-18 2020-10-14 Hoffmann La Roche Anticuerpos anti-hla-g y uso de los mismos
AR115052A1 (es) 2018-04-18 2020-11-25 Hoffmann La Roche Anticuerpos multiespecíficos y utilización de los mismos
BR112020023844A2 (pt) 2018-05-25 2021-04-13 Alector Llc Anticorpos, métodos de tratamento do câncer e de produção de um anticorpo, ácido nucleico, vetor, células hospedeiras e composição farmacêutica
CN110551216B (zh) * 2018-05-31 2022-11-18 信达生物制药(苏州)有限公司 多价抗ox40抗体及其用途
EP3805400A4 (en) 2018-06-04 2022-06-29 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule showing changed half-life in cytoplasm
WO2020004492A1 (ja) 2018-06-26 2020-01-02 協和キリン株式会社 Cell Adhesion Molecule3に結合する抗体
CN112424357A (zh) 2018-06-26 2021-02-26 协和麒麟株式会社 与硫酸软骨素蛋白聚糖5结合的抗体
CA3099176A1 (en) 2018-06-29 2020-01-02 Alector Llc Anti-sirp-beta1 antibodies and methods of use thereof
EP3820905A1 (en) * 2018-07-03 2021-05-19 NGM Biopharmaceuticals, Inc. Bispecific antibodies
CA3105448A1 (en) 2018-07-03 2020-01-09 Elstar Therapeutics, Inc. Anti-tcr antibody molecules and uses thereof
TWI809147B (zh) 2018-07-13 2023-07-21 美商阿列克特有限責任公司 抗分揀蛋白抗體及其使用方法
CN112584865A (zh) 2018-07-20 2021-03-30 表面肿瘤学公司 抗cd112r组合物和方法
EP3831854A4 (en) 2018-08-03 2022-05-04 Chugai Seiyaku Kabushiki Kaisha ANTIGEN-BINDING MOLECULE WITH TWO LINKED ANTIGEN-BINDING DOMAINS
MA50586A (fr) 2018-08-09 2020-09-16 Regeneron Pharma Procédés d'évaluation de l'affinité de liaison d'une variante d'anticorps au récepteur fc néonatal
CN112839960A (zh) 2018-08-10 2021-05-25 中外制药株式会社 抗cd137抗原结合分子及其应用
GB201814281D0 (en) 2018-09-03 2018-10-17 Femtogenix Ltd Cytotoxic agents
WO2020054979A1 (ko) * 2018-09-12 2020-03-19 아주대학교산학협력단 Cd83 억제제를 유효성분으로 포함하는 베체트병의 예방 또는 치료용 조성물
EP3853611A1 (en) 2018-09-19 2021-07-28 F. Hoffmann-La Roche AG Therapeutic and diagnostic methods for bladder cancer
AU2019361983A1 (en) 2018-10-18 2021-05-20 Genentech, Inc. Diagnostic and therapeutic methods for sarcomatoid kidney cancer
WO2020086858A1 (en) 2018-10-24 2020-04-30 Genentech, Inc. Conjugated chemical inducers of degradation and methods of use
US20220170097A1 (en) 2018-10-29 2022-06-02 The Broad Institute, Inc. Car t cell transcriptional atlas
AU2018451747A1 (en) 2018-12-06 2021-06-17 F. Hoffmann-La Roche Ag Combination therapy of diffuse large B-cell lymphoma comprising an anti-CD79b immunoconjugates, an alkylating agent and an anti-CD20 antibody
WO2020123275A1 (en) 2018-12-10 2020-06-18 Genentech, Inc. Photocrosslinking peptides for site specific conjugation to fc-containing proteins
BR112021010374A2 (pt) 2018-12-21 2021-08-24 23Andme, Inc. Anticorpos anti-il-36 e métodos de uso dos mesmos
AU2019410075B2 (en) 2018-12-21 2022-08-25 F. Hoffmann-La Roche Ag Antibodies binding to CD3
EP3674319A1 (en) * 2018-12-24 2020-07-01 Sanofi Pseudofab-based multispecific binding proteins
AU2019412561A1 (en) 2018-12-24 2021-08-12 Sanofi Multispecific binding proteins with mutant Fab domains
JP2022515473A (ja) 2018-12-28 2022-02-18 エフ.ホフマン-ラ ロシュ アーゲー 免疫応答が増幅された患者における治療的使用のためのペプチド-mhc-i-抗体融合タンパク質
CN111378044B (zh) 2018-12-28 2022-07-15 长春金赛药业有限责任公司 抗体融合蛋白、制备方法及其应用
CN111378045B (zh) 2018-12-28 2022-08-02 长春金赛药业有限责任公司 二价双特异性抗体及其制备方法、编码基因、宿主细胞、组合物
AU2019413278A1 (en) 2018-12-28 2021-07-15 Kyowa Kirin Co., Ltd. Bispecific antibody binding to TfR
CA3124688A1 (en) * 2018-12-31 2020-07-09 Merus N.V. Truncated multivalent multimers
JPWO2020153467A1 (ja) 2019-01-24 2021-12-02 中外製薬株式会社 新規がん抗原及びそれらの抗原に対する抗体
GB201901197D0 (en) 2019-01-29 2019-03-20 Femtogenix Ltd G-A Crosslinking cytotoxic agents
SG11202108955QA (en) 2019-02-21 2021-09-29 Marengo Therapeutics Inc Antibody molecules that bind to nkp30 and uses thereof
WO2020172601A1 (en) 2019-02-21 2020-08-27 Elstar Therapeutics, Inc. Multifunctional molecules that bind to calreticulin and uses thereof
WO2020172571A1 (en) 2019-02-21 2020-08-27 Elstar Therapeutics, Inc. Multifunctional molecules that bind to t cell related cancer cells and uses thereof
CA3131014A1 (en) 2019-02-21 2020-08-27 Andreas Loew Anti-tcr antibody molecules and uses thereof
JP2022522662A (ja) 2019-02-21 2022-04-20 マレンゴ・セラピューティクス,インコーポレーテッド T細胞に結合する多機能性分子および自己免疫障害を処置するためのその使用
TW202100141A (zh) 2019-03-08 2021-01-01 德商百靈佳殷格翰國際股份有限公司 抗il-36r抗體調配物
AU2020238811A1 (en) 2019-03-08 2021-07-22 Genentech, Inc. Methods for detecting and quantifying membrane-associated proteins on extracellular vesicles
TW202043291A (zh) 2019-04-19 2020-12-01 美商建南德克公司 抗mertk抗體及使用方法
CA3138045C (en) 2019-05-14 2024-02-20 Genentech, Inc. Methods of using anti-cd79b immunoconjugates to treat follicular lymphoma
JP7082245B2 (ja) 2019-06-10 2022-06-07 中外製薬株式会社 サイトカイン阻害剤と組み合わせて使用するための抗t細胞抗原結合分子
TW202112817A (zh) 2019-06-11 2021-04-01 美商阿列克特有限責任公司 抗揀選蛋白抗體之使用方法
AR119746A1 (es) 2019-06-14 2022-01-05 Teneobio Inc Anticuerpos multiespecíficos de cadena pesada que se unen a cd22 y cd3
AR119393A1 (es) 2019-07-15 2021-12-15 Hoffmann La Roche Anticuerpos que se unen a nkg2d
CN114174338A (zh) 2019-07-31 2022-03-11 豪夫迈·罗氏有限公司 与gprc5d结合的抗体
MX2022001156A (es) 2019-07-31 2022-02-22 Hoffmann La Roche Anticuerpos que se fijan a gprc5d.
JP2022542964A (ja) 2019-07-31 2022-10-07 アレクトル エルエルシー 抗ms4a4a抗体、及びその使用方法
TW202118512A (zh) 2019-09-12 2021-05-16 美商建南德克公司 治療狼瘡性腎炎之組成物及方法
BR112022004972A2 (pt) 2019-09-18 2022-06-28 Genentech Inc Anticorpos, ácidos nucleicos isolados, células hospedeiras isoladas, métodos de produção de um anticorpo, de produção de um anticorpo biespecífico e de tratamento de um indivíduo, anticorpos biespecíficos, composição farmacêutica, anticorpo, anticorpo biespecífico ou composição farmacêutica, combinação de um anticorpo, uso do anticorpo, uso de uma combinação do anticorpo e métodos para reduzir a inflamação e para melhorar a descamação e/ou de erupção cutânea e combinação para uso
TW202126699A (zh) 2019-09-20 2021-07-16 美商建南德克公司 用於抗類胰蛋白酶抗體之投藥
US20220281997A1 (en) 2019-09-27 2022-09-08 Nanjing GenScript Biotech Co., Ltd. Anti-VHH Domain Antibodies and Use Thereof
WO2021076196A1 (en) 2019-10-18 2021-04-22 Genentech, Inc. Methods of using anti-cd79b immunoconjugates to treat diffuse large b-cell lymphoma
CA3152314A1 (en) 2019-11-15 2021-05-20 Andrea ALLMENDINGER Prevention of visible particle formation in aqueous protein solutions
MX2022007158A (es) 2019-12-13 2022-07-11 Genentech Inc Anticuerpos anti-ly6g6d y metodos de uso.
KR20220113791A (ko) 2019-12-18 2022-08-16 에프. 호프만-라 로슈 아게 이중특이적 항-ccl2 항체
JP7296467B2 (ja) 2019-12-18 2023-06-22 エフ・ホフマン-ラ・ロシュ・アクチェンゲゼルシャフト Hla-a2/mage-a4に結合する抗体
KR20220118527A (ko) 2019-12-23 2022-08-25 제넨테크, 인크. 아포지질단백질 l1-특이적 항체 및 이용 방법
AU2019479791B2 (en) 2019-12-27 2024-05-02 Chugai Seiyaku Kabushiki Kaisha Anti-CTLA-4 antibody and use thereof
WO2021138407A2 (en) 2020-01-03 2021-07-08 Marengo Therapeutics, Inc. Multifunctional molecules that bind to cd33 and uses thereof
CN110818795B (zh) 2020-01-10 2020-04-24 上海复宏汉霖生物技术股份有限公司 抗tigit抗体和使用方法
JP2023510382A (ja) 2020-01-15 2023-03-13 エフ. ホフマン-ラ ロシュ アーゲー 組換えタンパク質製造プロセスから不純物を減少させる方法
TW202144395A (zh) 2020-02-12 2021-12-01 日商中外製藥股份有限公司 用於癌症之治療的抗cd137抗原結合分子
CR20220461A (es) 2020-03-13 2022-10-21 Genentech Inc Anticuerpos anti-interleucina-33 y usos de estos
IL296427A (en) 2020-03-19 2022-11-01 Genentech Inc Isoform-selective anti-tgf antibodies and methods of use
CN115867649A (zh) 2020-03-24 2023-03-28 基因泰克公司 Tie2结合剂及其使用方法
KR20220156912A (ko) 2020-03-25 2022-11-28 일라이 릴리 앤드 캄파니 다중특이적 결합 단백질 및 그의 개발 방법
JP2023518841A (ja) 2020-03-26 2023-05-08 ジェネンテック, インコーポレイテッド 宿主細胞タンパク質が減少した修飾哺乳動物細胞
TWI799824B (zh) 2020-03-31 2023-04-21 日商中外製藥股份有限公司 Dll3靶向之多特異性抗原結合分子及其用途
EP4127724A1 (en) 2020-04-03 2023-02-08 Genentech, Inc. Therapeutic and diagnostic methods for cancer
IL297060A (en) 2020-04-09 2022-12-01 Univ Muenchen Tech Targeted administration of an inhibitor of mir-21 to macrophages for the treatment of pulmonary fibrosis
CN115843312A (zh) 2020-04-24 2023-03-24 马伦戈治疗公司 结合至t细胞相关癌细胞的多功能性分子及其用途
CA3175530A1 (en) 2020-04-24 2021-10-28 Genentech, Inc. Methods of using anti-cd79b immunoconjugates
JP2023523450A (ja) 2020-04-28 2023-06-05 ジェネンテック, インコーポレイテッド 非小細胞肺がん免疫療法のための方法及び組成物
CN113563473A (zh) * 2020-04-29 2021-10-29 三生国健药业(上海)股份有限公司 四价双特异性抗体、其制备方法和用途
TW202200212A (zh) 2020-05-03 2022-01-01 中國大陸商聯寧(蘇州)生物製藥有限公司 包含抗-trop-2抗體之抗體藥物結合物
WO2021228917A1 (en) 2020-05-15 2021-11-18 F. Hoffmann-La Roche Ag Prevention of visible particle formation in parenteral protein solutions
WO2021233853A1 (en) 2020-05-19 2021-11-25 F. Hoffmann-La Roche Ag The use of chelators for the prevention of visible particle formation in parenteral protein solutions
TW202204396A (zh) 2020-06-08 2022-02-01 瑞士商赫孚孟拉羅股份公司 抗hbv抗體及使用方法
EP4165415A1 (en) 2020-06-12 2023-04-19 Genentech, Inc. Methods and compositions for cancer immunotherapy
CR20220628A (es) 2020-06-19 2023-01-24 Hoffmann La Roche Anticuerpos que se unen a cd3
KR20230152789A (ko) 2020-06-19 2023-11-03 추가이 세이야쿠 가부시키가이샤 혈관신생 저해제와 조합하여 사용하기 위한 항t세포 항원 결합 분자
WO2021255146A1 (en) 2020-06-19 2021-12-23 F. Hoffmann-La Roche Ag Antibodies binding to cd3 and cea
EP4168447A1 (en) 2020-06-19 2023-04-26 F. Hoffmann-La Roche AG Antibodies binding to cd3 and cd19
CR20220639A (es) 2020-06-19 2023-02-17 Hoffmann La Roche Anticuerpos que se unen a cd3 y folr1
CA3184747A1 (en) 2020-06-24 2021-12-30 Genentech, Inc. Apoptosis resistant cell lines
CN116133689A (zh) 2020-07-07 2023-05-16 豪夫迈·罗氏有限公司 作为治疗性蛋白质制剂的稳定剂的替代表面活性剂
CN115803010A (zh) 2020-07-09 2023-03-14 豪夫迈·罗氏有限公司 浓缩的蛋白质组合物、它们的制备及其用途
CN115812149A (zh) 2020-07-13 2023-03-17 基因泰克公司 用于预测多肽免疫原性的基于细胞的方法
TW202216780A (zh) 2020-07-17 2022-05-01 美商建南德克公司 抗notch2抗體及其使用方法
CA3188656A1 (en) 2020-07-17 2022-01-20 Simurx, Inc. Chimeric myd88 receptors for redirecting immunosuppressive signaling and related compositions and methods
CA3188649A1 (en) 2020-07-21 2022-01-27 Genentech, Inc. Antibody-conjugated chemical inducers of degradation of brm and methods thereof
GB2597532A (en) 2020-07-28 2022-02-02 Femtogenix Ltd Cytotoxic compounds
MX2023001120A (es) 2020-07-31 2023-02-22 Chugai Pharmaceutical Co Ltd Composicion farmaceutica que comprende una celula que expresa un receptor quimerico.
WO2022031948A1 (en) 2020-08-07 2022-02-10 Genentech, Inc. T cell-based methods for predicting polypeptide immunogenicity
WO2022047046A1 (en) 2020-08-26 2022-03-03 Marengo Therapeutics, Inc. Methods of detecting trbc1 or trbc2
KR20230074144A (ko) 2020-08-26 2023-05-26 마렝고 테라퓨틱스, 인크. NKp30에 결합하는 항체 분자 및 이의 용도
WO2022046920A2 (en) 2020-08-26 2022-03-03 Marengo Therapeutics, Inc. Multifunctional molecules that bind to calreticulin and uses thereof
WO2022047222A2 (en) 2020-08-28 2022-03-03 Genentech, Inc. Crispr/cas9 multiplex knockout of host cell proteins
KR20230073196A (ko) 2020-09-21 2023-05-25 제넨테크, 인크. 다중특이성 항체의 정제
EP4225443A1 (en) 2020-10-05 2023-08-16 Genentech, Inc. Dosing for treatment with anti-fcrh5/anti-cd3 bispecific antibodies
WO2022086957A1 (en) 2020-10-20 2022-04-28 Genentech, Inc. Peg-conjugated anti-mertk antibodies and methods of use
WO2022084210A1 (en) 2020-10-20 2022-04-28 F. Hoffmann-La Roche Ag Combination therapy of pd-1 axis binding antagonists and lrrk2 inhitibors
AU2021392039A1 (en) 2020-12-02 2023-06-29 Alector Llc Methods of use of anti-sortilin antibodies
KR20220082775A (ko) 2020-12-10 2022-06-17 주식회사 유틸렉스 항-pd-1 항체 및 이의 용도
JP7326584B2 (ja) 2020-12-17 2023-08-15 エフ・ホフマン-ラ・ロシュ・アクチェンゲゼルシャフト 抗hla-g抗体及びその使用
CN114716548A (zh) 2021-01-05 2022-07-08 (株)爱恩德生物 抗-fgfr3抗体及其用途
WO2022162587A1 (en) 2021-01-27 2022-08-04 Centre Hospitalier Universitaire Vaudois (C.H.U.V.) Anti-sars-cov-2 antibodies and use thereof in the treatment of sars-cov-2 infection
CA3209479A1 (en) 2021-02-03 2022-08-11 Mozart Therapeutics, Inc. Binding agents and methods of using the same
WO2022169872A1 (en) 2021-02-03 2022-08-11 Genentech, Inc. Multispecific binding protein degrader platform and methods of use
CN117440832A (zh) 2021-03-03 2024-01-23 索伦托药业有限公司 包括抗bcma抗体的抗体-药物缀合物
AR125074A1 (es) 2021-03-12 2023-06-07 Genentech Inc Anticuerpos anti-klk7, anticuerpos anti-klk5, anticuerpos multiespecíficos anti-klk5 / klk7 y métodos de uso
CA3213599A1 (en) 2021-03-15 2022-09-22 Genentech, Inc. Compositions and methods of treating lupus nephritis
CN116981696A (zh) 2021-03-18 2023-10-31 艾莱克特有限责任公司 抗tmem106b抗体及其使用方法
WO2022197877A1 (en) 2021-03-19 2022-09-22 Genentech, Inc. Methods and compositions for time delayed bio-orthogonal release of cytotoxic agents
JP2024511610A (ja) 2021-03-23 2024-03-14 アレクトル エルエルシー コロナウイルス感染の治療及び予防のための抗tmem106b抗体
BR112023019205A2 (pt) 2021-03-26 2023-10-24 Janssen Biotech Inc Anticorpos humanizados contra tau de filamento helicoidal pareado e usos dos mesmos
AU2022255506A1 (en) 2021-04-08 2023-11-09 Marengo Therapeutics, Inc. Multifunctional molecules binding to tcr and uses thereof
TW202304524A (zh) 2021-04-10 2023-02-01 美商普方生物製藥美國公司 Folr1結合劑、其結合物及使用方法
AR125344A1 (es) 2021-04-15 2023-07-05 Chugai Pharmaceutical Co Ltd Anticuerpo anti-c1s
JP2024514222A (ja) 2021-04-19 2024-03-28 ジェネンテック, インコーポレイテッド 改変された哺乳動物細胞
JP2024516631A (ja) 2021-04-23 2024-04-16 プロファウンドバイオ ユーエス カンパニー 抗cd70抗体、そのコンジュゲートおよびこれを使用する方法
IL307821A (en) 2021-04-30 2023-12-01 Hoffmann La Roche Dosage for treatment with BISPIFIC ANTI-CD20/ANTI-CD3 antibody
CA3213632A1 (en) 2021-04-30 2022-11-03 F. Hoffmann-La Roche Ag Dosing for combination treatment with anti-cd20/anti-cd3 bispecific antibody and anti-cd79b antibody drug conjugate
AU2022273063A1 (en) 2021-05-12 2023-11-23 Genentech, Inc. Methods of using anti-cd79b immunoconjugates to treat diffuse large b-cell lymphoma
TW202306993A (zh) 2021-05-14 2023-02-16 美商建南德克公司 Trem2之促效劑
KR20240010469A (ko) 2021-05-21 2024-01-23 제넨테크, 인크. 관심 재조합 생성물의 생성을 위한 변형된 세포
CN113214393B (zh) * 2021-05-25 2022-11-18 深圳市新产业生物医学工程股份有限公司 Il-6抗体或其抗原结合片段及包含其的检测试剂盒
CN113278071B (zh) 2021-05-27 2021-12-21 江苏荃信生物医药股份有限公司 抗人干扰素α受体1单克隆抗体及其应用
CN117480184A (zh) 2021-06-04 2024-01-30 中外制药株式会社 抗ddr2抗体及其用途
TW202313045A (zh) 2021-06-09 2023-04-01 瑞士商赫孚孟拉羅股份公司 用於癌症治療之組合療法
WO2022263638A1 (en) 2021-06-17 2022-12-22 Centre Hospitalier Universitaire Vaudois (C.H.U.V.) Anti-sars-cov-2 antibodies and use thereof in the treatment of sars-cov-2 infection
CN117500829A (zh) 2021-06-18 2024-02-02 豪夫迈·罗氏有限公司 双特异性抗ccl2抗体
IL308633A (en) 2021-06-25 2024-01-01 Chugai Pharmaceutical Co Ltd Use of anti-CTLA-4 antibodies
CA3221833A1 (en) 2021-06-25 2022-12-29 Chugai Seiyaku Kabushiki Kaisha Anti-ctla-4 antibody
TW202320857A (zh) 2021-07-06 2023-06-01 美商普方生物製藥美國公司 連接子、藥物連接子及其結合物及其使用方法
US20230197278A1 (en) 2021-07-13 2023-06-22 Genentech, Inc. Multi-variate model for predicting cytokine release syndrome
KR20240036570A (ko) 2021-07-22 2024-03-20 에프. 호프만-라 로슈 아게 이종이량체 Fc 도메인 항체
WO2023004386A1 (en) 2021-07-22 2023-01-26 Genentech, Inc. Brain targeting compositions and methods of use thereof
CN117794953A (zh) 2021-08-03 2024-03-29 豪夫迈·罗氏有限公司 双特异性抗体及使用方法
CN117897409A (zh) 2021-08-13 2024-04-16 基因泰克公司 抗类胰蛋白酶抗体的给药
KR20240049296A (ko) 2021-08-27 2024-04-16 제넨테크, 인크. 타우 병증을 치료하는 방법
TW202325727A (zh) 2021-08-30 2023-07-01 美商建南德克公司 抗聚泛素多特異性抗體
CN113683694B (zh) 2021-09-03 2022-05-13 江苏荃信生物医药股份有限公司 一种抗人tslp单克隆抗体及其应用
CN113603775B (zh) 2021-09-03 2022-05-20 江苏荃信生物医药股份有限公司 抗人白介素-33单克隆抗体及其应用
JPWO2023058723A1 (el) 2021-10-08 2023-04-13
CA3234162A1 (en) 2021-10-15 2023-04-20 Michele Fiscella Antibodies and methods of using thereof
WO2023076876A1 (en) 2021-10-26 2023-05-04 Mozart Therapeutics, Inc. Modulation of immune responses to viral vectors
WO2023086807A1 (en) 2021-11-10 2023-05-19 Genentech, Inc. Anti-interleukin-33 antibodies and uses thereof
AR127887A1 (es) 2021-12-10 2024-03-06 Hoffmann La Roche Anticuerpos que se unen a cd3 y plap
WO2023129974A1 (en) 2021-12-29 2023-07-06 Bristol-Myers Squibb Company Generation of landing pad cell lines
US20230322958A1 (en) 2022-01-19 2023-10-12 Genentech, Inc. Anti-Notch2 Antibodies and Conjugates and Methods of Use
AR128330A1 (es) 2022-01-26 2024-04-17 Genentech Inc Inductores químicos de degradación conjugados con anticuerpo y métodos de estos
AR128331A1 (es) 2022-01-26 2024-04-17 Genentech Inc Inductores químicos de degradación conjugados con anticuerpos y métodos de estos
WO2023173026A1 (en) 2022-03-10 2023-09-14 Sorrento Therapeutics, Inc. Antibody-drug conjugates and uses thereof
TW202400636A (zh) 2022-03-11 2024-01-01 比利時商健生藥品公司 多特異性抗體及其用途(一)
TW202345899A (zh) 2022-03-11 2023-12-01 比利時商健生藥品公司 多特異性抗體及其用途(三)
WO2023170290A1 (en) 2022-03-11 2023-09-14 Janssen Pharmaceutica Nv Multispecific antibodies and uses thereof
WO2023180353A1 (en) 2022-03-23 2023-09-28 F. Hoffmann-La Roche Ag Combination treatment of an anti-cd20/anti-cd3 bispecific antibody and chemotherapy
WO2023191816A1 (en) 2022-04-01 2023-10-05 Genentech, Inc. Dosing for treatment with anti-fcrh5/anti-cd3 bispecific antibodies
JP2024517042A (ja) 2022-04-13 2024-04-19 エフ・ホフマン-ラ・ロシュ・アクチェンゲゼルシャフト 抗cd20/抗cd3二重特異性抗体の薬学的組成物及び使用方法
US11958906B2 (en) 2022-04-13 2024-04-16 Genentech, Inc. Pharmaceutical compositions of mosunetuzumab and methods of use
WO2023215737A1 (en) 2022-05-03 2023-11-09 Genentech, Inc. Anti-ly6e antibodies, immunoconjugates, and uses thereof
WO2023219613A1 (en) 2022-05-11 2023-11-16 Genentech, Inc. Dosing for treatment with anti-fcrh5/anti-cd3 bispecific antibodies
WO2024015897A1 (en) 2022-07-13 2024-01-18 Genentech, Inc. Dosing for treatment with anti-fcrh5/anti-cd3 bispecific antibodies
WO2024020432A1 (en) 2022-07-19 2024-01-25 Genentech, Inc. Dosing for treatment with anti-fcrh5/anti-cd3 bispecific antibodies
WO2024020564A1 (en) 2022-07-22 2024-01-25 Genentech, Inc. Anti-steap1 antigen-binding molecules and uses thereof
WO2024020579A1 (en) 2022-07-22 2024-01-25 Bristol-Myers Squibb Company Antibodies binding to human pad4 and uses thereof
WO2024030956A2 (en) 2022-08-03 2024-02-08 Mozart Therapeutics, Inc. Cd39-specific binding agents and methods of using the same
WO2024049949A1 (en) 2022-09-01 2024-03-07 Genentech, Inc. Therapeutic and diagnostic methods for bladder cancer
WO2024068996A1 (en) 2022-09-30 2024-04-04 Centre Hospitalier Universitaire Vaudois (C.H.U.V.) Anti-sars-cov-2 antibodies and use thereof in the treatment of sars-cov-2 infection
WO2024079010A1 (en) 2022-10-10 2024-04-18 F. Hoffmann-La Roche Ag Combination therapy of a gprc5d tcb and cd38 antibodies
WO2024079009A1 (en) 2022-10-10 2024-04-18 F. Hoffmann-La Roche Ag Combination therapy of a gprc5d tcb and proteasome inhibitors
WO2024079015A1 (en) 2022-10-10 2024-04-18 F. Hoffmann-La Roche Ag Combination therapy of a gprc5d tcb and imids
WO2024086796A1 (en) 2022-10-20 2024-04-25 Alector Llc Anti-ms4a4a antibodies with amyloid-beta therapies
WO2024091991A1 (en) 2022-10-25 2024-05-02 Genentech, Inc. Therapeutic and diagnostic methods for multiple myeloma

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6214984B1 (en) * 1995-04-20 2001-04-10 Genentech, Inc. Isolated nucleic acid encoding, and methods for preparing, antibody fragments
US6670453B2 (en) * 1997-10-27 2003-12-30 Unilever Patent Holdings B.V. Multivalent antigen-binding proteins

Family Cites Families (162)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3773919A (en) 1969-10-23 1973-11-20 Du Pont Polylactide-drug mixtures
USRE30985E (en) 1978-01-01 1982-06-29 Serum-free cell culture media
US4275149A (en) 1978-11-24 1981-06-23 Syva Company Macromolecular environment control in specific receptor assays
US4318980A (en) 1978-04-10 1982-03-09 Miles Laboratories, Inc. Heterogenous specific binding assay employing a cycling reactant as label
WO1981001145A1 (en) 1979-10-18 1981-04-30 Univ Illinois Hydrolytic enzyme-activatible pro-drugs
US4419446A (en) 1980-12-31 1983-12-06 The United States Of America As Represented By The Department Of Health And Human Services Recombinant DNA process utilizing a papilloma virus DNA as a vector
US4485045A (en) 1981-07-06 1984-11-27 Research Corporation Synthetic phosphatidyl cholines useful in forming liposomes
NZ201705A (en) 1981-08-31 1986-03-14 Genentech Inc Recombinant dna method for production of hepatitis b surface antigen in yeast
US4601978A (en) 1982-11-24 1986-07-22 The Regents Of The University Of California Mammalian metallothionein promoter system
US4560655A (en) 1982-12-16 1985-12-24 Immunex Corporation Serum-free cell culture medium and process for making same
US4657866A (en) 1982-12-21 1987-04-14 Sudhir Kumar Serum-free, synthetic, completely chemically defined tissue culture media
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
DD266710A3 (de) 1983-06-06 1989-04-12 Ve Forschungszentrum Biotechnologie Verfahren zur biotechnischen Herstellung van alkalischer Phosphatase
US4544545A (en) 1983-06-20 1985-10-01 Trustees University Of Massachusetts Liposomes containing modified cholesterol for organ targeting
US4767704A (en) 1983-10-07 1988-08-30 Columbia University In The City Of New York Protein-free culture medium
US4943533A (en) 1984-03-01 1990-07-24 The Regents Of The University Of California Hybrid cell lines that produce monoclonal antibodies to epidermal growth factor receptor
US4965199A (en) 1984-04-20 1990-10-23 Genentech, Inc. Preparation of functional human factor VIII in mammalian cells using methotrexate based selection
US5807715A (en) 1984-08-27 1998-09-15 The Board Of Trustees Of The Leland Stanford Junior University Methods and transformed mammalian lymphocyte cells for producing functional antigen-binding protein including chimeric immunoglobulin
US4879231A (en) 1984-10-30 1989-11-07 Phillips Petroleum Company Transformation of yeasts of the genus pichia
US4737456A (en) 1985-05-09 1988-04-12 Syntex (U.S.A.) Inc. Reducing interference in ligand-receptor binding assays
GB8516415D0 (en) 1985-06-28 1985-07-31 Celltech Ltd Culture of animal cells
US5091178A (en) 1986-02-21 1992-02-25 Oncogen Tumor therapy with biologically active anti-tumor antibodies
US4927762A (en) 1986-04-01 1990-05-22 Cell Enterprises, Inc. Cell culture medium with antioxidant
GB8610600D0 (en) 1986-04-30 1986-06-04 Novo Industri As Transformation of trichoderma
US5182368A (en) 1986-06-13 1993-01-26 Ledbetter Jeffrey A Ligands and methods for augmenting B-cell proliferation
US6121424A (en) * 1991-11-25 2000-09-19 Enzon, Inc. Multivalent antigen-binding proteins
US5567610A (en) 1986-09-04 1996-10-22 Bioinvent International Ab Method of producing human monoclonal antibodies and kit therefor
US4968603A (en) 1986-12-31 1990-11-06 The Regents Of The University Of California Determination of status in neoplastic disease
IL85035A0 (en) 1987-01-08 1988-06-30 Int Genetic Eng Polynucleotide molecule,a chimeric antibody with specificity for human b cell surface antigen,a process for the preparation and methods utilizing the same
US4816467A (en) * 1987-01-09 1989-03-28 Farmitalia Carlo Erba S.R.L Heteroaryl 3-oxo-propanenitrile derivatives, pharmaceutical compositions and use
GB8705477D0 (en) 1987-03-09 1987-04-15 Carlton Med Prod Drug delivery systems
WO1988007089A1 (en) 1987-03-18 1988-09-22 Medical Research Council Altered antibodies
US4975278A (en) 1988-02-26 1990-12-04 Bristol-Myers Company Antibody-enzyme conjugates in combination with prodrugs for the delivery of cytotoxic agents to tumor cells
IL87737A (en) 1987-09-11 1993-08-18 Genentech Inc Method for culturing polypeptide factor dependent vertebrate recombinant cells
US5283187A (en) 1987-11-17 1994-02-01 Brown University Research Foundation Cell culture-containing tubular capsule produced by co-extrusion
US4892538A (en) 1987-11-17 1990-01-09 Brown University Research Foundation In vivo delivery of neurotransmitters by implanted, encapsulated cells
US5824311A (en) * 1987-11-30 1998-10-20 Trustees Of The University Of Pennsylvania Treatment of tumors with monoclonal antibodies against oncogene antigens
WO1989006692A1 (en) 1988-01-12 1989-07-27 Genentech, Inc. Method of treating tumor cells by inhibiting growth factor receptor function
US5223409A (en) 1988-09-02 1993-06-29 Protein Engineering Corp. Directed evolution of novel binding proteins
DE68925971T2 (de) 1988-09-23 1996-09-05 Cetus Oncology Corp Zellenzuchtmedium für erhöhtes zellenwachstum, zur erhöhung der langlebigkeit und expression der produkte
GB8823869D0 (en) 1988-10-12 1988-11-16 Medical Res Council Production of antibodies
US5175384A (en) 1988-12-05 1992-12-29 Genpharm International Transgenic mice depleted in mature t-cells and methods for making transgenic mice
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
FR2646437B1 (fr) 1989-04-28 1991-08-30 Transgene Sa Nouvelles sequences d'adn, leur application en tant que sequence codant pour un peptide signal pour la secretion de proteines matures par des levures recombinantes, cassettes d'expression, levures transformees et procede de preparation de proteines correspondant
ATE155813T1 (de) 1989-05-19 1997-08-15 Genentech Inc Her2 extrazellulare domäne
EP0402226A1 (en) 1989-06-06 1990-12-12 Institut National De La Recherche Agronomique Transformation vectors for yeast yarrowia
DE3920358A1 (de) 1989-06-22 1991-01-17 Behringwerke Ag Bispezifische und oligospezifische, mono- und oligovalente antikoerperkonstrukte, ihre herstellung und verwendung
DK0417563T3 (da) 1989-09-12 2000-11-06 Hoffmann La Roche TNF-bindende proteiner
DE68926248T2 (de) * 1989-09-29 1996-12-19 Oncogene Science Inc p100 "neu" menschlisches Protein and Verwendung dieses Proteins zum Nachweis von preneoplasmatischen- oder neoplasmatischen beim Menschen
US5013556A (en) 1989-10-20 1991-05-07 Liposome Technology, Inc. Liposomes with enhanced circulation time
US5208020A (en) 1989-10-25 1993-05-04 Immunogen Inc. Cytotoxic agents comprising maytansinoids and their therapeutic use
US5183884A (en) 1989-12-01 1993-02-02 United States Of America Dna segment encoding a gene for a receptor related to the epidermal growth factor receptor
US5229275A (en) 1990-04-26 1993-07-20 Akzo N.V. In-vitro method for producing antigen-specific human monoclonal antibodies
US5427908A (en) 1990-05-01 1995-06-27 Affymax Technologies N.V. Recombinant library screening methods
US5723286A (en) 1990-06-20 1998-03-03 Affymax Technologies N.V. Peptide library and screening systems
WO1991019515A1 (en) 1990-06-21 1991-12-26 The Board Of Trustees Of The Leland Stanford Junior University Oligomeric immunoglobulin constant domain mutant with enhanced complement-mediated cytolytic activity
US5545806A (en) 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
US5625126A (en) 1990-08-29 1997-04-29 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
US5661016A (en) 1990-08-29 1997-08-26 Genpharm International Inc. Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US5633425A (en) 1990-08-29 1997-05-27 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
KR100272077B1 (ko) 1990-08-29 2000-11-15 젠팜인터내셔날,인코포레이티드 이종 항체를 생산할 수 있는 전이유전자를 가진 인간이외의 동물
IL99553A0 (en) 1990-09-28 1992-08-18 Ixsys Inc Compositions containing oligonucleotides linked to expression elements,a kit for the preparation of vectors useful for the expression of a diverse population of random peptides and methods utilizing the same
US5122469A (en) 1990-10-03 1992-06-16 Genentech, Inc. Method for culturing Chinese hamster ovary cells to improve production of recombinant proteins
ATE164395T1 (de) 1990-12-03 1998-04-15 Genentech Inc Verfahren zur anreicherung von proteinvarianten mit geänderten bindungseigenschaften
US5571894A (en) 1991-02-05 1996-11-05 Ciba-Geigy Corporation Recombinant antibodies specific for a growth factor receptor
IL101943A0 (en) 1991-05-24 1992-12-30 Genentech Inc Structure,production and use of heregulin
DE4118120A1 (de) * 1991-06-03 1992-12-10 Behringwerke Ag Tetravalente bispezifische rezeptoren, ihre herstellung und verwendung
GB9112536D0 (en) 1991-06-11 1991-07-31 Celltech Ltd Chemical compounds
ATE255131T1 (de) 1991-06-14 2003-12-15 Genentech Inc Humanisierter heregulin antikörper
US5264586A (en) 1991-07-17 1993-11-23 The Scripps Research Institute Analogs of calicheamicin gamma1I, method of making and using the same
US5587458A (en) 1991-10-07 1996-12-24 Aronex Pharmaceuticals, Inc. Anti-erbB-2 antibodies, combinations thereof, and therapeutic and diagnostic uses thereof
WO1994000136A1 (en) 1992-06-30 1994-01-06 Oncologix, Inc. A COMBINATION OF ANTI-erbB-2 MONOCLONAL ANTIBODIES AND METHOD OF USING
US5270170A (en) 1991-10-16 1993-12-14 Affymax Technologies N.V. Peptide library and screening method
EP1136556B1 (en) 1991-11-25 2005-06-08 Enzon, Inc. Method of producing multivalent antigen-binding proteins
WO1993016185A2 (en) 1992-02-06 1993-08-19 Creative Biomolecules, Inc. Biosynthetic binding protein for cancer marker
CA2128862C (en) 1992-02-11 2008-05-20 Jonathan G. Seidman Homogenotization of gene-targeting events
AU4025193A (en) 1992-04-08 1993-11-18 Cetus Oncology Corporation Humanized C-erbB-2 specific antibodies
ZA932522B (en) 1992-04-10 1993-12-20 Res Dev Foundation Immunotoxins directed against c-erbB-2(HER/neu) related surface antigens
CA2134773A1 (en) 1992-06-04 1993-12-09 Robert J. Debs Methods and compositions for in vivo gene therapy
US5397703A (en) 1992-07-09 1995-03-14 Cetus Oncology Corporation Method for generation of antibodies to cell surface molecules
US5736137A (en) 1992-11-13 1998-04-07 Idec Pharmaceuticals Corporation Therapeutic application of chimeric and radiolabeled antibodies to human B lymphocyte restricted differentiation antigen for treatment of B cell lymphoma
PL174494B1 (pl) 1992-11-13 1998-08-31 Idec Pharma Corp Kompozycja farmaceutyczna do leczenia chłoniaka z limfocytów B i sposób wytwarzania kompozycji farmaceutycznej do leczenia chłoniaka z limfocytów B
CA2103323A1 (en) 1992-11-24 1994-05-25 Gregory D. Plowman Her4 human receptor tyrosine kinase
DK0672142T3 (da) 1992-12-04 2001-06-18 Medical Res Council Multivalente og multispecifikke bindingsproteiner samt fremstilling og anvendelse af disse
JP3312357B2 (ja) 1992-12-11 2002-08-05 ザ ダウ ケミカル カンパニー 多価の一本鎖抗体
US5861156A (en) 1993-01-08 1999-01-19 Creative Biomolecules Methods of delivering agents to target cells
CA2115811A1 (en) 1993-02-17 1994-08-18 Claus Krebber A method for in vivo selection of ligand-binding proteins
US5591699A (en) 1993-02-24 1997-01-07 E. I. Du Pont De Nemours And Company Particle transport fluids thickened with acetylate free xanthan heteropolysaccharide biopolymer plus guar gum
WO1994022478A1 (en) 1993-03-30 1994-10-13 The Trustees Of The University Of Pennsylvania PREVENTION OF TUMORS WITH MONOCLONAL ANTIBODIES AGAINST $i(NEU)
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
US5595721A (en) 1993-09-16 1997-01-21 Coulter Pharmaceutical, Inc. Radioimmunotherapy of lymphoma using anti-CD20
WO1995009917A1 (en) 1993-10-07 1995-04-13 The Regents Of The University Of California Genetically engineered bispecific tetravalent antibodies
SE9304060D0 (sv) 1993-12-06 1993-12-06 Bioinvent Int Ab Sätt att selektera specifika bakteriofager
DE69433820T2 (de) 1993-12-23 2005-06-23 Immunex Corp., Seattle Verwendung von löslichen oligomerischen cd40 liganden oder monoklonalen antikörpern zur herstellung eines arzneimitells zur vorbeugung oder behandlung von neoplastischen krankheiten
US5773001A (en) 1994-06-03 1998-06-30 American Cyanamid Company Conjugates of methyltrithio antitumor agents and intermediates for their synthesis
US5516637A (en) 1994-06-10 1996-05-14 Dade International Inc. Method involving display of protein binding pairs on the surface of bacterial pili and bacteriophage
US5910486A (en) 1994-09-06 1999-06-08 Uab Research Foundation Methods for modulating protein function in cells using, intracellular antibody homologues
US6214388B1 (en) 1994-11-09 2001-04-10 The Regents Of The University Of California Immunoliposomes that optimize internalization into target cells
JPH10511085A (ja) 1994-12-02 1998-10-27 カイロン コーポレイション 二重特異性抗体を用いる免疫応答を促進する方法
US5731168A (en) 1995-03-01 1998-03-24 Genentech, Inc. Method for making heteromultimeric polypeptides
US5739277A (en) 1995-04-14 1998-04-14 Genentech Inc. Altered polypeptides with increased half-life
WO1996033206A1 (en) 1995-04-17 1996-10-24 University Of Utah Research Foundation Conotoxin peptides
US5702892A (en) 1995-05-09 1997-12-30 The United States Of America As Represented By The Department Of Health And Human Services Phage-display of immunoglobulin heavy chain libraries
AU6267896A (en) 1995-06-07 1996-12-30 Imclone Systems Incorporated Antibody and antibody fragments for inhibiting the growth oftumors
US5874409A (en) * 1995-06-07 1999-02-23 La Jolla Pharmaceutical Company APL immunoreactive peptides, conjugates thereof and methods of treatment for APL antibody-mediated pathologies
US5712374A (en) 1995-06-07 1998-01-27 American Cyanamid Company Method for the preparation of substantiallly monomeric calicheamicin derivative/carrier conjugates
US5837234A (en) 1995-06-07 1998-11-17 Cytotherapeutics, Inc. Bioartificial organ containing cells encapsulated in a permselective polyether suflfone membrane
US5714586A (en) 1995-06-07 1998-02-03 American Cyanamid Company Methods for the preparation of monomeric calicheamicin derivative/carrier conjugates
EP2258726A1 (en) 1995-06-14 2010-12-08 The Regents of the University of California High affinity human antibodies to c-erbB-2
NZ311982A (en) 1995-06-29 1999-08-30 Immunex Corp Tnf related apoptosis inducing ligand (trail), a method for producing them and associated antibodies
PT859841E (pt) 1995-08-18 2002-11-29 Morphosys Ag Bibliotecas de proteinas/ (poli) peptidos
WO1997014719A1 (en) 1995-10-16 1997-04-24 Unilever N.V. A bifunctional or bivalent antibody fragment analogue
US5783186A (en) 1995-12-05 1998-07-21 Amgen Inc. Antibody-induced apoptosis
US6030945A (en) 1996-01-09 2000-02-29 Genentech, Inc. Apo-2 ligand
ATE278958T1 (de) 1996-03-20 2004-10-15 Dyax Corp Reinigung des gewebe plasminogenaktivators (tpa)
US5968511A (en) 1996-03-27 1999-10-19 Genentech, Inc. ErbB3 antibodies
WO1997038102A1 (en) * 1996-04-04 1997-10-16 Unilever Plc Multivalent and multispecific antigen-binding protein
AU3135097A (en) 1996-05-22 1997-12-09 Johns Hopkins University, The Methods of detection utilizing modified bacteriophage
US5708156A (en) 1996-05-31 1998-01-13 Ilekis; John V. Epidermal growth factor receptor-like gene product and its uses
CA2257873A1 (en) 1996-06-10 1997-12-18 The Scripps Research Institute Use of substrate subtraction libraries to distinguish enzyme specificities
US5922845A (en) 1996-07-11 1999-07-13 Medarex, Inc. Therapeutic multispecific compounds comprised of anti-Fcα receptor antibodies
WO1998005344A1 (en) 1996-08-05 1998-02-12 Brigham And Women's Hospital, Inc. Bacteriophage-mediated gene therapy
DE69718341T2 (de) 1996-10-08 2003-10-30 Bisys B V U Verfahren und mittel zur auswahl von peptiden und proteinen mit spezifischer affinität zu einem zielmolekül
IL129354A0 (en) 1996-10-18 2000-02-17 Genentech Inc Anti-ErbB2 antibodies
AU731553B2 (en) 1996-10-25 2001-04-05 Human Genome Sciences, Inc. Neutrokine alpha
DE122010000048I1 (de) 1996-12-23 2011-05-05 Immunex Corp Ligand für rezeptor aktivator of nf-kappa b, ligand ist mitglied der tnf superfamilie
AU6250098A (en) 1997-01-28 1998-08-18 Human Genome Sciences, Inc. Death domain containing receptor 4 (dr4: death receptor 4), member of the tnf-receptor superfamily and binding to trail (ap02-l)
US6072047A (en) 1997-02-13 2000-06-06 Immunex Corporation Receptor that binds trail
CA2282410A1 (en) * 1997-03-03 1998-10-01 Bristol-Myers Squibb Company Monoclonal antibodies to human cd6
US5969102A (en) 1997-03-03 1999-10-19 St. Jude Children's Research Hospital Lymphocyte surface receptor that binds CAML, nucleic acids encoding the same and methods of use thereof
US20010010924A1 (en) 1997-03-14 2001-08-02 Keith Charles Deen Tumor necrosis factor related receptor, tr6 polynecleotides
ES2281126T3 (es) 1997-03-17 2007-09-16 Human Genome Sciences, Inc. Receptor 5 que contiene un dominio de muerte.
US6268411B1 (en) * 1997-09-11 2001-07-31 The Johns Hopkins University Use of multivalent chimeric peptide-loaded, MHC/ig molecules to detect, activate or suppress antigen-specific T cell-dependent immune responses
WO1998046643A1 (en) 1997-04-16 1998-10-22 Millennium Biotherapeutics, Inc. TUMOR NECROSIS FACTOR RECEPTOR RELATED PROTEINS TANGO-63d AND TANGO-63e
RO128635A2 (ro) 1997-04-16 2013-07-30 Amgen Inc. Anticorp sau fragment al acestuia care se leagă la opgbp şi utilizarea acestuia, utilizarea unei forme solubile de odar, şi metodă de identificare a unui compus care descreşte activitatea opgbp
ATE370232T1 (de) 1997-05-15 2007-09-15 Genentech Inc Anti-apo-2 antikörper
AU7830398A (en) 1997-06-11 1998-12-30 Human Genome Sciences, Inc. Human tumor necrosis factor receptor tr9
WO1998058965A2 (en) 1997-06-20 1998-12-30 Innogenetics N.V. B7-binding molecules for treating immune diseases
AU8296098A (en) 1997-07-08 1999-02-08 Board Of Regents, The University Of Texas System Compositions and methods for homoconjugates of antibodies which induce growth arrest or apoptosis of tumor cells
AU8400398A (en) 1997-07-11 1999-02-08 Trustees Of The University Of Pennsylvania, The Nucleic acid encoding a novel chemotherapy-induced protein, and methods of use
AU9013998A (en) 1997-07-21 1999-02-10 Zymogenetics Inc. Tumor necrosis factor receptor ztnfr-5
WO1999006587A2 (en) 1997-08-01 1999-02-11 Morphosys Ag Novel method and phage for the identification of nucleic acid sequences encoding members of a multimeric (poly)peptide complex
JP2001512667A (ja) 1997-08-06 2001-08-28 リジェネロン・ファーマシューティカルズ・インコーポレイテッド ヒトオーファンレセプターntr−1
EP1003864A1 (en) 1997-08-15 2000-05-31 Idun Pharmaceuticals, Inc. Trail receptors, nucleic acids encoding the same, and methods of use thereof
AU9376498A (en) 1997-09-05 1999-03-22 University Of Washington Tumor necrosis factor family receptors and ligands, encoding nucleic acids and related binding agents
CA2303225C (en) 1997-09-18 2015-03-31 Genentech, Inc. Dna30942 polypeptide, a tnfr homolog
AU9805398A (en) 1997-10-15 1999-05-03 Children's Medical Center Corporation Novel human egf receptors and use thereof
AU2093499A (en) 1997-12-30 1999-07-19 Chiron Corporation Members of tnf and tnfr families
WO1999037791A1 (en) * 1998-01-23 1999-07-29 Vlaams Interuniversitair Instituut Voor Biotechnologie Multipurpose antibody derivatives
EP1053256B9 (en) 1998-01-26 2012-01-18 Genentech, Inc. Antibodies to death receptor 4 (dr4) and uses thereof
CA2323757C (en) 1998-04-02 2011-08-02 Genentech, Inc. Antibody variants and fragments thereof
DE19819846B4 (de) 1998-05-05 2016-11-24 Deutsches Krebsforschungszentrum Stiftung des öffentlichen Rechts Multivalente Antikörper-Konstrukte
WO1999060846A1 (en) * 1998-05-26 1999-12-02 Innogenetics N.V. Method for expanding primate b cells selectively in immunocompromised mice and producing large numbers of antigen-specific b lymphocytes for the production of primate monoclonal antibodies
JP2002517223A (ja) 1998-06-12 2002-06-18 ジェネンテック・インコーポレーテッド モノクローナル抗体、交叉反応性抗体およびそれを生成する方法
ATE251181T1 (de) 1998-07-28 2003-10-15 Micromet Ag Heterominikörper
US6824780B1 (en) * 1999-10-29 2004-11-30 Genentech, Inc. Anti-tumor antibody compositions and methods of use
IL151853A0 (en) * 2000-04-11 2003-04-10 Genentech Inc Multivalent antibodies and uses therefor
CA2417185A1 (en) 2000-07-25 2002-01-31 Shui-On Leung Multivalent target binding protein
SI1517921T1 (sl) 2002-06-28 2006-10-31 Domantis Ltd Dvojno-specificni ligandi z zvisano serumsko razpolovno dobo
US20060263367A1 (en) * 2005-05-23 2006-11-23 Fey Georg H Bispecific antibody devoid of Fc region and method of treatment using same
US20130149308A1 (en) * 2011-08-12 2013-06-13 Genentech, Inc. Antibodies to il-1beta and il-18, for treatment of disease

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6214984B1 (en) * 1995-04-20 2001-04-10 Genentech, Inc. Isolated nucleic acid encoding, and methods for preparing, antibody fragments
US20020002271A1 (en) * 1995-04-20 2002-01-03 Genentech, Inc. Antibody purification
US6670453B2 (en) * 1997-10-27 2003-12-30 Unilever Patent Holdings B.V. Multivalent antigen-binding proteins

Cited By (691)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9545086B2 (en) 1999-01-25 2017-01-17 Biogen Ma Inc. BAFF, inhibitors thereof and their use in the modulation of B-cell response and treatment of autoimmune disorders
US20050244411A1 (en) * 1999-01-25 2005-11-03 Biogen Idec Ma Inc. BAFF, inhibitors thereof and their use in the modulation of B-cell response and treatment of autoimmune disorders
US20090110676A1 (en) * 1999-01-25 2009-04-30 Biogen Idec Ma Inc. BAFF, Inhibitors Thereof and Their Use in the Modulation of B-Cell Response and Treatment of Autoimmune Disorders
US20110008337A1 (en) * 1999-05-07 2011-01-13 Genetech, Inc. Treatment of Autoimmune Diseases
US7820161B1 (en) 1999-05-07 2010-10-26 Biogen Idec, Inc. Treatment of autoimmune diseases
US8545843B2 (en) 1999-05-07 2013-10-01 Genentech, Inc. Treatment of vasculitis
US20110008250A1 (en) * 1999-05-07 2011-01-13 Genentech, Inc. Treatment of Autoimmune Diseases
US9993550B2 (en) 1999-05-07 2018-06-12 Genentech, Inc. Treatment of pemphigus
US20110008336A1 (en) * 1999-05-07 2011-01-13 Genentech, Inc. Treatment of Autoimmune Diseases
US20110008338A1 (en) * 1999-05-07 2011-01-13 Genentech, Inc. Treatment of Autoimmune Diseases
US20100003252A1 (en) * 1999-07-12 2010-01-07 Genentech, Inc. Blocking immune response to a graft
US20060073146A1 (en) * 2000-02-16 2006-04-06 Genentech, Inc. Uses of agonists and antagonists to modulate activity of TNF-related molecules
US8337856B2 (en) 2000-03-16 2012-12-25 Immunogen, Inc. Methods of treatment using anti-ERBB antibody-maytansinoid conjugates
US20080226659A1 (en) * 2000-03-16 2008-09-18 Sharon Erickson Methods of treatment using anti-erbb antibody-maytansinoid conjugates
US20020058029A1 (en) * 2000-09-18 2002-05-16 Nabil Hanna Combination therapy for treatment of autoimmune diseases using B cell depleting/immunoregulatory antibody combination
US20030219476A1 (en) * 2000-10-16 2003-11-27 Neopharm, Inc. Liposomal formulation of mitoxantrone
US20030228309A1 (en) * 2000-11-08 2003-12-11 Theodora Salcedo Antibodies that immunospecifically bind to TRAIL receptors
US20060062786A1 (en) * 2000-11-08 2006-03-23 Human Genome Sciences, Inc. Antibodies that immunospecifically bind to TRAIL receptors
US20110091461A1 (en) * 2001-01-17 2011-04-21 Trubion Pharmaceuticals, Inc. Binding domain-immunoglobulin fusion proteins
US20110223164A1 (en) * 2001-01-17 2011-09-15 Emergent Product Development Seattle, Llc Binding domain-immunoglobulin fusion proteins
US8106161B2 (en) 2001-01-17 2012-01-31 Emergent Product Development Seattle, Llc Binding domain-immunoglobulin fusion proteins
US9005612B2 (en) 2001-01-17 2015-04-14 Emergent Product Development Seattle, Llc Binding domain-immunoglobulin fusion proteins
US8147835B2 (en) 2001-01-17 2012-04-03 Emergent Product Development Seattle, Llc Binding domain-immunoglobulin fusion proteins
US20100034820A1 (en) * 2001-01-17 2010-02-11 Trubion Pharmaceuticals, Inc. Binding domain-immunoglobulin fusion proteins
US8188237B2 (en) 2001-01-17 2012-05-29 Emergent Product Development Seattle, Llc Binding domain-immunoglobulin fusion proteins
US20050202028A1 (en) * 2001-01-17 2005-09-15 Trubion Pharmaceuticals, Inc. Binding domain-immunoglobulin fusion proteins
US7754208B2 (en) * 2001-01-17 2010-07-13 Trubion Pharmaceuticals, Inc. Binding domain-immunoglobulin fusion proteins
US8853366B2 (en) 2001-01-17 2014-10-07 Emergent Product Development Seattle, Llc Binding domain-immunoglobulin fusion proteins
US8197810B2 (en) 2001-01-17 2012-06-12 Emergent Product Development Seattle, Llc Binding domain-immunoglobulin fusion proteins
US20110105729A1 (en) * 2001-01-17 2011-05-05 Trubion Pharmaceuticals, Inc. Binding domain-immunoglobulin fusion proteins
US20030118592A1 (en) * 2001-01-17 2003-06-26 Genecraft, Inc. Binding domain-immunoglobulin fusion proteins
US9562102B2 (en) 2001-05-11 2017-02-07 Ludwig Institute For Cancer Research Specific binding proteins and uses thereof
US20050129616A1 (en) * 2001-05-25 2005-06-16 Human Genome Sciences, Inc. Antibodies that immunospecifically bind to TRAIL receptors
US7348003B2 (en) 2001-05-25 2008-03-25 Human Genome Sciences, Inc. Methods of treating cancer using antibodies that immunospecifically bind to TRAIL receptors
US20060270837A1 (en) * 2001-05-25 2006-11-30 Human Genome Sciences, Inc. Antibodies that immunospecifically bind to TRAIL receptors
US20090226429A1 (en) * 2001-05-25 2009-09-10 Human Genome Sciences, Inc. Antibodies That Immunospecifically Bind to TRAIL Receptors
US20050214209A1 (en) * 2001-05-25 2005-09-29 Human Genome Sciences, Inc. Antibodies that immunospecifically bind to TRAIL receptors
US7361341B2 (en) 2001-05-25 2008-04-22 Human Genome Sciences, Inc. Methods of treating cancer using antibodies that immunospecifically bind to trail receptors
US9458236B2 (en) 2001-06-13 2016-10-04 Genmab A/S Human monoclonal antibodies to epidermal growth factor receptor (EGFR)
US7247301B2 (en) 2001-06-13 2007-07-24 Genmab A/S Human monoclonal antibodies to epidermal growth factor receptor (EGFR)
US8586041B2 (en) 2001-06-13 2013-11-19 Genmab A/S Human monoclonal antibodies to epidermal growth factor receptor (EGFR)
US20030091561A1 (en) * 2001-06-13 2003-05-15 Genmab A/S Human monoclonal antibodies to epidermal growth factor receptor (EGFR)
US20030194403A1 (en) * 2001-06-13 2003-10-16 Genmab, Inc. Human monoclonal antibodies to epidermal growth factor receptor (EGFR)
US7595378B2 (en) 2001-06-13 2009-09-29 Genmab A/S Human monoclonal antibodies to epidermal growth factor receptor (EGFR)
US20040219643A1 (en) * 2001-06-28 2004-11-04 Greg Winter Dual-specific ligand
US20070093651A1 (en) * 2001-06-28 2007-04-26 Domantis Limited Ligand
US20050271663A1 (en) * 2001-06-28 2005-12-08 Domantis Limited Compositions and methods for treating inflammatory disorders
US20050100548A1 (en) * 2001-07-24 2005-05-12 Biogen Idec Ma Inc. BAFF, inhibitors thereof and their use in the modulation of B-cell response
US20060079457A1 (en) * 2001-07-24 2006-04-13 Biogen Idec Ma Inc. BAFF, inhibitors thereof and their use in the modulation of B-cell response
US20050070689A1 (en) * 2001-08-03 2005-03-31 Genentech, Inc. Taci and br3 polypeptides and uses thereof
US20030180296A1 (en) * 2001-12-20 2003-09-25 Theodora Salcedo Antibodies that immunospecifically bind to trail receptors
EP2075256A2 (en) 2002-01-14 2009-07-01 William Herman Multispecific binding molecules
US7435803B2 (en) 2002-02-14 2008-10-14 Immunomedics, Inc. Anti-CD20 antibodies and fusion proteins thereof and methods of use
US20100040541A1 (en) * 2002-02-14 2010-02-18 Immunomedics, Inc. Structural Variants of Antibodies for Improved Therapeutic Characteristics
US20030219433A1 (en) * 2002-02-14 2003-11-27 Immunomedics, Inc. Anti-CD20 antibodies and fusion proteins thereof and methods of use
US8057793B2 (en) 2002-02-14 2011-11-15 Immunomedics, Inc. Anti-CD20 antibodies and fusion proteins thereof and methods of use
US7151164B2 (en) * 2002-02-14 2006-12-19 Immunomedics, Inc. Anti-CD20 antibodies and fusion proteins thereof and methods of use
US20090155253A1 (en) * 2002-02-14 2009-06-18 Immunomedics, Inc. Anti-cd20 antibodies and fusion proteins therof and methods of use
US8287864B2 (en) 2002-02-14 2012-10-16 Immunomedics, Inc. Structural variants of antibodies for improved therapeutic characteristics
US20040002587A1 (en) * 2002-02-20 2004-01-01 Watkins Jeffry D. Fc region variants
US20060153838A1 (en) * 2002-02-20 2006-07-13 Watkins Jeffry D Fc region variants
US20080267965A1 (en) * 2002-02-21 2008-10-30 Kalled Susan L Use of Bcma as an Immunoregulatory Agent
US20110177093A1 (en) * 2002-02-21 2011-07-21 Biogen, Inc. Use of bcma as an immunoregulatory agent
US8734791B2 (en) 2002-03-01 2014-05-27 Xencor, Inc. Optimized fc variants and methods for their generation
US20070219133A1 (en) * 2002-03-01 2007-09-20 Xencor, Inc. CD52 OPTIMIZED Fc VARIANTS AND METHODS FOR THEIR GENERATION
US20090068175A1 (en) * 2002-03-01 2009-03-12 Xencor, Inc. Optimized FC Variants and Methods for Their Generation
US7662925B2 (en) 2002-03-01 2010-02-16 Xencor, Inc. Optimized Fc variants and methods for their generation
US20080181890A1 (en) * 2002-03-01 2008-07-31 Xencor, Inc. Optimized Fc Variants and Methods for Their Generation
US20090142340A1 (en) * 2002-03-01 2009-06-04 Xencor, Inc. Optimized Fc Variants and Methods for Their Generation
US20070160597A1 (en) * 2002-03-01 2007-07-12 Xencor, Inc. Optimized Fc variants and methods for their generation
US20070003546A1 (en) * 2002-03-01 2007-01-04 Xencor, Inc. Optimized Fc variants and methods for their generation
US8124731B2 (en) 2002-03-01 2012-02-28 Xencor, Inc. Optimized Fc variants and methods for their generation
US8093357B2 (en) 2002-03-01 2012-01-10 Xencor, Inc. Optimized Fc variants and methods for their generation
US20080254027A1 (en) * 2002-03-01 2008-10-16 Bernett Matthew J Optimized CD5 antibodies and methods of using the same
US7317091B2 (en) 2002-03-01 2008-01-08 Xencor, Inc. Optimized Fc variants
US20070224189A1 (en) * 2002-03-01 2007-09-27 Xencor, Inc. CD20 OPTIMIZED Fc VARIANTS AND METHODS FOR THEIR GENERATION
US20080260731A1 (en) * 2002-03-01 2008-10-23 Bernett Matthew J Optimized antibodies that target cd19
US20090259026A1 (en) * 2002-06-28 2009-10-15 Ian Tomlinson Ligand
US9321832B2 (en) 2002-06-28 2016-04-26 Domantis Limited Ligand
US20060106203A1 (en) * 2002-06-28 2006-05-18 Domantis Limited Ligand
US20080171036A1 (en) * 2002-07-25 2008-07-17 Anan Chuntharapai Taci antibodies and uses thereof
EP1551877A4 (en) * 2002-07-25 2006-01-18 Genentech Inc TACI ANTIBODIES AND THEIR USE
US20050043516A1 (en) * 2002-07-25 2005-02-24 Genentech, Inc. TACI antibodies and uses thereof
WO2004011611A3 (en) * 2002-07-25 2004-08-12 Genentech Inc Taci antibodies and uses thereof
EP1551877A2 (en) * 2002-07-25 2005-07-13 Genentech, Inc. Taci antibodies and uses thereof
US8193318B2 (en) 2002-08-14 2012-06-05 Macrogenics, Inc. FcγRIIB specific antibodies and methods of use thereof
US8946387B2 (en) 2002-08-14 2015-02-03 Macrogenics, Inc. FcγRIIB specific antibodies and methods of use thereof
US20090092610A1 (en) * 2002-08-14 2009-04-09 Macrogenics, Inc. FcGammaRIIB Specific Antibodies and Methods of Use Thereof
US20090074771A1 (en) * 2002-08-14 2009-03-19 Macrogenics, Inc. FcGammaRIIB Specific Antibodies and Methods of Use Thereof
US20090076251A1 (en) * 2002-08-14 2009-03-19 Macrogenics, Inc. FcGammaRIIB Specific Antibodies and Methods of Use Thereof
US8968730B2 (en) 2002-08-14 2015-03-03 Macrogenics Inc. FcγRIIB specific antibodies and methods of use thereof
US20090017023A1 (en) * 2002-08-14 2009-01-15 Macrogenics, Inc. FcGammaRIIB Specific Antibodies and Methods of Use Thereof
US20090017027A1 (en) * 2002-08-14 2009-01-15 Macrogenics, Inc. FcGammaRIIB Specific Antibodies and Methods of Use Thereof
US8530627B2 (en) 2002-08-14 2013-09-10 Macrogenics, Inc. FcγRIIB specific antibodies and methods of use thereof
US8044180B2 (en) 2002-08-14 2011-10-25 Macrogenics, Inc. FcγRIIB specific antibodies and methods of use thereof
US20100166751A1 (en) * 2002-09-04 2010-07-01 Biopolymer Engineering (D/B/A Biothera) Cancer therapy using whole glucan particles and antibodies
JP2006512407A (ja) * 2002-09-27 2006-04-13 ゼンコー・インコーポレイテッド 最適化Fc変異体およびそれらの生成方法
US8809503B2 (en) 2002-09-27 2014-08-19 Xencor, Inc. Optimized Fc variants and methods for their generation
US8735547B2 (en) 2002-09-27 2014-05-27 Xencor, Inc. Optimized Fc Variants
US10183999B2 (en) 2002-09-27 2019-01-22 Xencor, Inc. Optimized Fc variants and methods for their generation
US20090081208A1 (en) * 2002-09-27 2009-03-26 Xencor, Inc. Optimized Fc variants and methods for their generation
US8188231B2 (en) 2002-09-27 2012-05-29 Xencor, Inc. Optimized FC variants
US20040132101A1 (en) * 2002-09-27 2004-07-08 Xencor Optimized Fc variants and methods for their generation
US20090092599A1 (en) * 2002-09-27 2009-04-09 Xencor, Inc. Optimized Fc variants and methods for their generation
US8753628B2 (en) 2002-09-27 2014-06-17 Xencor, Inc. Optimized Fc variants
US10184000B2 (en) 2002-09-27 2019-01-22 Xencor, Inc. Optimized Fc variants and methods for their generation
US8383109B2 (en) 2002-09-27 2013-02-26 Xencor, Inc. Optimized Fc variants and methods for their generation
US8753629B2 (en) 2002-09-27 2014-06-17 Xencor, Inc. Optimized Fc variants
AU2003279719B8 (en) * 2002-09-27 2009-01-08 Xencor Inc. Optimized Fc variants and methods for their generation
AU2003279719B2 (en) * 2002-09-27 2008-12-11 Xencor Inc. Optimized Fc variants and methods for their generation
WO2004029207A3 (en) * 2002-09-27 2004-10-28 Xencor Inc Optimized fc variants and methods for their generation
US8093359B2 (en) 2002-09-27 2012-01-10 Xencor, Inc. Optimized Fc variants and methods for their generation
US9193798B2 (en) 2002-09-27 2015-11-24 Xencor, Inc. Optimized Fc variants and methods for their generation
US9353187B2 (en) 2002-09-27 2016-05-31 Xencor, Inc. Optimized FC variants and methods for their generation
US8802823B2 (en) 2002-09-27 2014-08-12 Xencor, Inc. Optimized Fc variants
US8858937B2 (en) 2002-09-27 2014-10-14 Xencor, Inc. Optimized Fc variants and methods for their generation
US8039592B2 (en) 2002-09-27 2011-10-18 Xencor, Inc. Optimized Fc variants and methods for their generation
US20070148171A1 (en) * 2002-09-27 2007-06-28 Xencor, Inc. Optimized anti-CD30 antibodies
US20060235208A1 (en) * 2002-09-27 2006-10-19 Xencor, Inc. Fc variants with optimized properties
WO2004039329A3 (en) * 2002-10-31 2004-07-15 Biogen Inc Treatment of immunological renal disorders by lymphotoxin pathway inhibitors
WO2004039329A2 (en) * 2002-10-31 2004-05-13 Biogen Idec Ma Inc. Treatment of immunological renal disorders by lymphotoxin pathway inhibitors
US20060034835A1 (en) * 2002-12-16 2006-02-16 Genentech, Inc. Immunoglobulin variants and uses thereof
US8562992B2 (en) 2002-12-16 2013-10-22 Genentech, Inc. Immunoglobulin variants and uses thereof
US7799900B2 (en) 2002-12-16 2010-09-21 Genentech, Inc. Immunoglobulin variants and uses thereof
US20090155257A1 (en) * 2002-12-16 2009-06-18 Genentech, Inc. Immunoglobulin variants and uses thereof
US20060257406A1 (en) * 2002-12-27 2006-11-16 Domantis Limited Ligand
US9028815B2 (en) 2003-01-09 2015-05-12 Macrogenics, Inc. Identification and engineering of antibodies with variant FC regions and methods of using same
US8192737B2 (en) 2003-01-09 2012-06-05 Macrogenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
US20080131435A1 (en) * 2003-01-09 2008-06-05 Macrogenics, Inc. Identification and Engineering of Antibodies With Variant Fc Regions and Methods of Using Same
US8951517B2 (en) 2003-01-09 2015-02-10 Macrogenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
US20080138344A1 (en) * 2003-01-09 2008-06-12 Macrogenics, Inc. Identification and Engineering of Antibodies with Variant Fc Regions and Methods of Using Same
US7960512B2 (en) 2003-01-09 2011-06-14 Macrogenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
US20050064514A1 (en) * 2003-01-09 2005-03-24 Macrogenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
US8003774B2 (en) 2003-01-09 2011-08-23 Macrogenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
US20070141052A1 (en) * 2003-02-20 2007-06-21 Watkins Jeffry D Fc region variants
WO2004074455A3 (en) * 2003-02-20 2005-02-17 Applied Molecular Evolution Fc REGION VARIANTS
US20070237765A1 (en) * 2003-03-03 2007-10-11 Xencor, Inc. Fc Variants Having Increased Affinity for FcyRl
US20070248603A1 (en) * 2003-03-03 2007-10-25 Xencor, Inc. Fc Variants with Increased Affinity for FcyRlla
US20070238665A1 (en) * 2003-03-03 2007-10-11 Xencor, Inc. Fc Variants Having Decreased Affinity for FcyRIIc
US20070275460A1 (en) * 2003-03-03 2007-11-29 Xencor.Inc. Fc Variants With Optimized Fc Receptor Binding Properties
US20070237766A1 (en) * 2003-03-03 2007-10-11 Xencor, Inc. Fc Variants Having Increased Affinity for FcyRllla
US10113001B2 (en) 2003-03-03 2018-10-30 Xencor, Inc. Fc variants with increased affinity for FcyRIIc
US20110021755A1 (en) * 2003-03-03 2011-01-27 Xencor, Inc. Optimized Fc Variants
US9657106B2 (en) 2003-03-03 2017-05-23 Xencor, Inc. Optimized Fc variants
US9663582B2 (en) 2003-03-03 2017-05-30 Xencor, Inc. Optimized Fc variants
US8735545B2 (en) 2003-03-03 2014-05-27 Xencor, Inc. Fc variants having increased affinity for fcyrllc
US20080057056A1 (en) * 2003-03-03 2008-03-06 Xencor, Inc. Fc Variants with Increased Affinity for FcyRIIC
US20090010920A1 (en) * 2003-03-03 2009-01-08 Xencor, Inc. Fc Variants Having Decreased Affinity for FcyRIIb
US20070248602A1 (en) * 2003-03-03 2007-10-25 Xencor, Inc. Fc Variants Having Increased Affinity for FcyRllc
US20070243188A1 (en) * 2003-03-03 2007-10-18 Xencor, Inc. Fc Variants Having Decreased Affinity for FcyRlla
US8084582B2 (en) 2003-03-03 2011-12-27 Xencor, Inc. Optimized anti-CD20 monoclonal antibodies having Fc variants
US20070237767A1 (en) * 2003-03-03 2007-10-11 Xencor, Inc. Fc Variants Having Decreased Affinity for FcyRllla
US8388955B2 (en) 2003-03-03 2013-03-05 Xencor, Inc. Fc variants
US10584176B2 (en) 2003-03-03 2020-03-10 Xencor, Inc. Fc variants with increased affinity for FcγRIIc
US8765662B2 (en) 2003-03-19 2014-07-01 Biogen Idec Ma Inc. NOGO receptor binding protein
US20070059793A1 (en) * 2003-03-19 2007-03-15 Biogen Idec Ma Inc. Nogo receptor binding protein
US8932821B2 (en) 2003-03-19 2015-01-13 Biogen Idec Ma Inc. NOGO receptor binding protein
US7785829B2 (en) 2003-03-19 2010-08-31 Biogen Idec Ma, Inc. Nogo receptor binding protein
US20080213280A1 (en) * 2003-04-09 2008-09-04 Genentech, Inc. Therapy of autoimmune disease in a patient with an inadequate response to a tnf-alpha inhibitor
US7708994B2 (en) 2003-04-09 2010-05-04 Genentech, Inc. Therapy of autoimmune disease in a patient with an inadequate response to a TNF-α inhibitor
WO2004091657A2 (en) 2003-04-09 2004-10-28 Genentech, Inc. Therapy of autoimmune disease in a patient with an inadequate response to a tnf-alpha inhibitor
US20040202658A1 (en) * 2003-04-09 2004-10-14 Genentech, Inc. Therapy of autoimmune disease in a patient with an inadequate response to TNF-alpha inhibitor
US20060240008A1 (en) * 2003-04-09 2006-10-26 Genentech, Inc. Therapy of autoimmune disease in a patient with an inadequate response to a TNF-alpha inhibitor
US7976838B2 (en) 2003-04-09 2011-07-12 Genentech, Inc. Therapy of autoimmune disease in a patient with an inadequate response to a TNF-α inhibitor
EP2062916A2 (en) 2003-04-09 2009-05-27 Genentech, Inc. Therapy of autoimmune disease in a patient with an inadequate response to a TNF-Alpha inhibitor
US9051373B2 (en) 2003-05-02 2015-06-09 Xencor, Inc. Optimized Fc variants
WO2004101756A3 (en) * 2003-05-09 2005-06-09 Diadexus Inc Ovr110 antibody compositions and methods of use
US20070178101A1 (en) * 2003-05-09 2007-08-02 Glenn Pilkington Ovr110 antibody compositions and methods of use
US7619068B2 (en) 2003-05-09 2009-11-17 Diadexus, Inc. Ovr110 antibody compositions and methods of use
EP2272868A2 (en) 2003-06-05 2011-01-12 Genentech, Inc. Combination therapy for B cell disorders
US20050095243A1 (en) * 2003-06-05 2005-05-05 Genentech, Inc. Combination therapy for B cell disorders
US20100143352A1 (en) * 2003-06-05 2010-06-10 Genentech, Inc. Combination therapy for b cell disorders
US20050163775A1 (en) * 2003-06-05 2005-07-28 Genentech, Inc. Combination therapy for B cell disorders
US11492411B2 (en) 2003-06-27 2022-11-08 Amgen Fremont Inc. Antibodies directed to the deletion mutants of epidermal growth factor receptor and uses thereof
US10118968B2 (en) * 2003-06-27 2018-11-06 Amgen Fremont Inc. Antibodies directed to the deletion mutants of epidermal growth factor receptor and uses thereof
US10508153B2 (en) 2003-06-27 2019-12-17 Amgen Fremont Inc. Antibodies directed to the deletion mutants of epidermal growth factor receptor and uses thereof
US20100111979A1 (en) * 2003-06-27 2010-05-06 Amgen Fremont Inc. Antibodies directed to the deletion mutants of epidermal growth factor receptor and uses thereof
US20100279932A1 (en) * 2003-07-26 2010-11-04 Trubion Pharmaceuticals, Inc. Binding constructs and methods for use thereof
US20050069552A1 (en) * 2003-07-28 2005-03-31 Bleck Gregory T. Fusion antibodies
US20050032130A1 (en) * 2003-07-29 2005-02-10 Genentech, Inc. Neutralizing antibody assay and uses therefor
US20090136492A1 (en) * 2003-08-29 2009-05-28 Genentech, Inc. Therapy of ocular disorders
US20050053602A1 (en) * 2003-08-29 2005-03-10 Genentech, Inc. Therapy of ocular disorders
US9714282B2 (en) 2003-09-26 2017-07-25 Xencor, Inc. Optimized Fc variants and methods for their generation
US10844135B2 (en) 2003-10-10 2020-11-24 Immunogen, Inc. Method of targeting specific cell populations using cell-binding agent maytansinoid conjugates linked via a non-cleavable linker, said conjugates and methods of making said
US20050186206A1 (en) * 2003-12-19 2005-08-25 Genentech, Inc. Detection of CD20 in therapy of autoimmune diseases
US20050191297A1 (en) * 2003-12-19 2005-09-01 Genentech, Inc. Detection of CD20 in transplant rejection
US20100093979A1 (en) * 2003-12-22 2010-04-15 Gregory Alan Lazar Fc Polypeptides With Novel Fc Ligand Binding Sites
US20050249723A1 (en) * 2003-12-22 2005-11-10 Xencor, Inc. Fc polypeptides with novel Fc ligand binding sites
WO2005072479A3 (en) * 2004-01-27 2006-04-13 Univ Southern California Polymer-bound antibody concer therapeutic agent
US20050214286A1 (en) * 2004-01-27 2005-09-29 University Of Southern California Polymer-bound antibody cancer therapeutic agent
EP3653641A1 (en) 2004-02-19 2020-05-20 Genentech, Inc. Cdr-repaired antibodies
US20100266542A1 (en) * 2004-03-23 2010-10-21 Biogen Idec Ma Inc. Receptor coupling agents and therapeutic uses thereof
US7947271B2 (en) 2004-03-23 2011-05-24 Biogen Idec Ma Inc. Methods of decreasing tumor volume and reducing tumor burden using TNF-receptor-coupling agents
US20110064727A9 (en) * 2004-03-24 2011-03-17 Xencor, Inc. Immunoglobulin Variants Outside the Fc Region
US20050244403A1 (en) * 2004-03-24 2005-11-03 Xencor, Inc. Immunoglobulin variants outside the Fc region
US7276585B2 (en) 2004-03-24 2007-10-02 Xencor, Inc. Immunoglobulin variants outside the Fc region
US20080248028A1 (en) * 2004-03-24 2008-10-09 Xencor, Inc. Immunoglobulin Variants Outside the Fc Region
US20080075719A1 (en) * 2004-04-16 2008-03-27 Genentech, Inc. Method for Augmenting B Cell Depletion
US20050271658A1 (en) * 2004-05-05 2005-12-08 Genentech, Inc. Preventing autoimmune disease
US8133982B2 (en) 2004-05-10 2012-03-13 Macrogenics, Inc. FcγRIIB specific antibodies and methods of use thereof
US20090202537A1 (en) * 2004-05-10 2009-08-13 Macrogenics, Inc. FcGammaRIIB Specific Antibodies and Methods of Use Thereof
US8784808B2 (en) 2004-05-10 2014-07-22 Macrogenics, Inc. FcγRIIB specific antibodies and methods of use thereof
EP3130349A1 (en) 2004-06-04 2017-02-15 Genentech, Inc. Method for treating multiple sclerosis
US20060024295A1 (en) * 2004-06-04 2006-02-02 Genentech, Inc. Method for treating lupus
US20100303810A1 (en) * 2004-06-04 2010-12-02 Genentech, Inc. Method for treating lupus
US20060051345A1 (en) * 2004-06-04 2006-03-09 Genentech, Inc. Method for treating multiple sclerosis
US20100233121A1 (en) * 2004-06-04 2010-09-16 Genentech, Inc. Method for treating multiple sclerosis
US9068992B2 (en) 2004-06-24 2015-06-30 Biogen Ma Inc. Screening methods for identifying Sp35 antagonists
US11820830B2 (en) 2004-07-20 2023-11-21 Xencor, Inc. Optimized Fc variants
US20060062787A1 (en) * 2004-07-22 2006-03-23 Genentech, Inc. Method for treating Sjogren's syndrome
WO2006017574A1 (en) * 2004-08-03 2006-02-16 Mayo Foundation For Medical Education And Research Improving treatments
US20070286855A1 (en) * 2004-08-03 2007-12-13 Mayo Foundation For Medical Education And Research Improving treatments
US7696320B2 (en) 2004-08-24 2010-04-13 Domantis Limited Ligands that have binding specificity for VEGF and/or EGFR and methods of use therefor
US20060074225A1 (en) * 2004-09-14 2006-04-06 Xencor, Inc. Monomeric immunoglobulin Fc domains
WO2006041680A2 (en) 2004-10-05 2006-04-20 Genentech, Inc. Method for treating vasculitis
US20060110387A1 (en) * 2004-10-05 2006-05-25 Genentech, Inc. Method for treating vasculitis
US20070025987A1 (en) * 2004-10-05 2007-02-01 Genentech, Inc. Method for Treating Vasculitis
EP3498294A1 (en) 2004-10-20 2019-06-19 Genentech, Inc. Antibody formulations
EP2371388A2 (en) 2004-10-20 2011-10-05 Genentech, Inc. Antibody formulations
US20090053240A1 (en) * 2004-10-21 2009-02-26 Xencor, Inc. Novel Immunoglobulin Insertions, Deletions and Substitutions
US8101720B2 (en) 2004-10-21 2012-01-24 Xencor, Inc. Immunoglobulin insertions, deletions and substitutions
US20060121042A1 (en) * 2004-10-27 2006-06-08 Medimmune, Inc. Modulation of antibody specificity by tailoring the affinity to cognate antigens
US20100196362A1 (en) * 2004-11-10 2010-08-05 Macrogenics, Inc. Engineering Fc Antibody Regions to Confer Effector Function
EP1810035A4 (en) * 2004-11-10 2010-03-17 Macrogenics Inc EFFECTOR FUNCTION OBTAINED BY CREATION BY BIOLOGICAL GENE OF FC ANTIBODY REGIONS
US8216574B2 (en) 2004-11-10 2012-07-10 Macrogenics, Inc. Engineering Fc antibody regions to confer effector function
AU2005335714B2 (en) * 2004-11-10 2012-07-26 Macrogenics, Inc. Engineering Fc antibody regions to confer effector function
EP1810035A2 (en) * 2004-11-10 2007-07-25 Magrogenics, Inc. Engineering fc antibody regions to confer effector function
US10336818B2 (en) 2004-11-12 2019-07-02 Xencor, Inc. Fc variants with altered binding to FcRn
US8802820B2 (en) 2004-11-12 2014-08-12 Xencor, Inc. Fc variants with altered binding to FcRn
US20090041770A1 (en) * 2004-11-12 2009-02-12 Chamberlain Aaron Keith Fc VARIANTS WITH ALTERED BINDING TO FcRn
US8318907B2 (en) 2004-11-12 2012-11-27 Xencor, Inc. Fc variants with altered binding to FcRn
US8883973B2 (en) 2004-11-12 2014-11-11 Xencor, Inc. Fc variants with altered binding to FcRn
US20070135620A1 (en) * 2004-11-12 2007-06-14 Xencor, Inc. Fc variants with altered binding to FcRn
US20060173170A1 (en) * 2004-11-12 2006-08-03 Xencor, Inc. Fc variants with altered binding to FcRn
US8338574B2 (en) 2004-11-12 2012-12-25 Xencor, Inc. FC variants with altered binding to FCRN
US8367805B2 (en) 2004-11-12 2013-02-05 Xencor, Inc. Fc variants with altered binding to FcRn
US20090163699A1 (en) * 2004-11-12 2009-06-25 Chamberlain Aaron Keith Fc VARIANTS WITH ALTERED BINDING TO FcRn
US20100234575A1 (en) * 2004-11-12 2010-09-16 Xencor, Inc. Fc variants with altered binding to fcrn
US9803023B2 (en) 2004-11-12 2017-10-31 Xencor, Inc. Fc variants with altered binding to FcRn
US8852586B2 (en) 2004-11-12 2014-10-07 Xencor, Inc. Fc variants with altered binding to FcRn
US20100234573A1 (en) * 2004-11-12 2010-09-16 Xencor, Inc. Fc Variants with altered binding to FcRn
US8324351B2 (en) 2004-11-12 2012-12-04 Xencor, Inc. Fc variants with altered binding to FcRn
US8546543B2 (en) 2004-11-12 2013-10-01 Xencor, Inc. Fc variants that extend antibody half-life
US9200079B2 (en) 2004-11-12 2015-12-01 Xencor, Inc. Fc variants with altered binding to FcRn
US20100204454A1 (en) * 2004-11-12 2010-08-12 Xencor, Inc. Fc Variants with altered binding to FcRn
US11198739B2 (en) 2004-11-12 2021-12-14 Xencor, Inc. Fc variants with altered binding to FcRn
US20100234572A1 (en) * 2004-11-12 2010-09-16 Xencor, Inc. Fc Variants with altered binding to FcRn
US20060275282A1 (en) * 2005-01-12 2006-12-07 Xencor, Inc. Antibodies and Fc fusion proteins with altered immunogenicity
US20060188495A1 (en) * 2005-01-13 2006-08-24 Genentech, Inc. Treatment method
US20080299117A1 (en) * 2005-01-13 2008-12-04 Barron Hal V Treatment Method
US20080095771A1 (en) * 2005-01-13 2008-04-24 Genentech, Inc. Treatment Method
EP3698807A1 (en) 2005-01-21 2020-08-26 Genentech, Inc. Fixed dosing of her antibodies
US20060246004A1 (en) * 2005-02-07 2006-11-02 Genentech, Inc. Antibody variants and uses thereof
EP2399605A1 (en) 2005-02-23 2011-12-28 Genentech, Inc. Extending time to disease progression or survival in cancer patients
US20060263355A1 (en) * 2005-02-28 2006-11-23 Joanne Quan Treatment of bone disorders
US20060204505A1 (en) * 2005-03-08 2006-09-14 Sliwkowski Mark X Methods for identifying tumors responsive to treatment with HER dimerization inhibitors (HDIs)
WO2006105338A3 (en) * 2005-03-31 2007-03-29 Xencor Inc Fc VARIANTS WITH OPTIMIZED PROPERTIES
US9737617B2 (en) 2005-04-06 2017-08-22 Ibc Pharmaceuticals, Inc. Multiple signaling pathways induced by hexavalent, monospecific and bispecific antibodies for enhanced toxicity to B-cell lymphomas and other diseases
US8906378B2 (en) 2005-04-06 2014-12-09 Ibc Pharmaceuticals, Inc. Combination therapy with anti-CD74 antibodies provides enhanced toxicity to malignancies, autoimmune disease and other diseases
US9359443B2 (en) 2005-04-06 2016-06-07 Ibc Pharmaceuticals, Inc. Combination therapy with anti-CD74 and anti-CD20 antibodies provides enhanced toxicity to B-cell diseases
US8349332B2 (en) 2005-04-06 2013-01-08 Ibc Pharmaceuticals, Inc. Multiple signaling pathways induced by hexavalent, monospecific and bispecific antibodies for enhanced toxicity to B-cell lymphomas and other diseases
US8871216B2 (en) 2005-04-06 2014-10-28 Ibc Pharmaceuticals, Inc. Multiple signaling pathways induced by hexvalent, monospecific and bispecific antibodies for enhanced toxicity to B-cell lymphomas and other diseases
US8475794B2 (en) 2005-04-06 2013-07-02 Ibc Pharmaceuticals, Inc. Combination therapy with anti-CD74 antibodies provides enhanced toxicity to malignancies, Autoimmune disease and other diseases
US20060233797A1 (en) * 2005-04-15 2006-10-19 Genentech, Inc. Treatment of inflammatory bowel disease (IBD)
US7601335B2 (en) 2005-05-20 2009-10-13 Genentech, Inc. Pretreatment of a biological sample from an autoimmune disease subject
US20100015055A1 (en) * 2005-05-20 2010-01-21 Genentech, Inc. Pretreatment of a biological sample from an autoimmune disease subject
US20060263349A1 (en) * 2005-05-20 2006-11-23 Genentech, Inc. Pretreatment of a biological sample from an autoimmune disease subject
US8551476B2 (en) 2005-07-08 2013-10-08 Biogen Idec Ma Inc. SP35 antibodies and uses thereof
US9066984B2 (en) 2005-07-08 2015-06-30 Biogen Ma Inc. Sp35 antibodies and uses thereof
US20080279850A1 (en) * 2005-07-25 2008-11-13 Trubion Pharmaceuticals, Inc. B-Cell Reduction Using CD37-Specific and CD20-Specific Binding Molecules
EP2415483A1 (en) 2005-07-25 2012-02-08 Emergent Product Development Seattle, LLC Single dose use of cd20-specific binding molecules
EP2586798A2 (en) 2005-07-25 2013-05-01 Emergent Product Development Seattle, LLC B-cell reduction using CD37-specific and CD20-specific binding molecules
EP2298815A1 (en) 2005-07-25 2011-03-23 Trubion Pharmaceuticals, Inc. B-cell reduction using CD37-specific and CD20-specific binding molecules
US20090214539A1 (en) * 2005-07-25 2009-08-27 Trubion Pharmaceuticals, Inc. B-cell reduction using cd37-specific and cd20-specific binding molecules
US20070059306A1 (en) * 2005-07-25 2007-03-15 Trubion Pharmaceuticals, Inc. B-cell reduction using CD37-specific and CD20-specific binding molecules
US10143748B2 (en) 2005-07-25 2018-12-04 Aptevo Research And Development Llc B-cell reduction using CD37-specific and CD20-specific binding molecules
EP2295080A2 (en) 2005-07-25 2011-03-16 Trubion Pharmaceuticals, Inc. B-cell reduction using CD37-specific and CD20-specific binding molecules
US10307481B2 (en) 2005-07-25 2019-06-04 Aptevo Research And Development Llc CD37 immunotherapeutics and uses thereof
US8697071B2 (en) 2005-08-10 2014-04-15 Macrogenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
US20070036799A1 (en) * 2005-08-10 2007-02-15 Macrogenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
US8217147B2 (en) 2005-08-10 2012-07-10 Macrogenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
US20070071675A1 (en) * 2005-08-19 2007-03-29 Chengbin Wu Dual variable domain immunoglobulin and uses thereof
US20090215992A1 (en) * 2005-08-19 2009-08-27 Chengbin Wu Dual variable domain immunoglobulin and uses thereof
US20100047239A1 (en) * 2005-08-19 2010-02-25 Abbott Laboratories Dual variable domain immunoglobulin and uses thereof
US8258268B2 (en) 2005-08-19 2012-09-04 Abbott Laboratories Dual variable domain immunoglobulin and uses thereof
US7612181B2 (en) 2005-08-19 2009-11-03 Abbott Laboratories Dual variable domain immunoglobulin and uses thereof
US8129114B2 (en) 2005-08-24 2012-03-06 Bristol-Myers Squibb Company Biomarkers and methods for determining sensitivity to epidermal growth factor receptor modulators
WO2007025044A3 (en) * 2005-08-24 2007-09-20 Bristol Myers Squibb Co Biomarkers and methods for determining sensitivity to epidermal growth factor receptor modulators
US20100221754A1 (en) * 2005-08-24 2010-09-02 Bristol-Myers Squibb Company Biomarkers and methods for determining sensitivity to epidermal growth factor receptor modulators
US20090028872A1 (en) * 2005-09-26 2009-01-29 Jonathan Alexander Terret Human monoclonal antibodies to cd70
US8124738B2 (en) 2005-09-26 2012-02-28 Medarex, Inc. Human monoclonal antibodies to CD70
US20080206867A1 (en) * 2005-10-03 2008-08-28 Desjarlais John R Fc variants with optimized Fc receptor binding properties
US9040041B2 (en) 2005-10-03 2015-05-26 Xencor, Inc. Modified FC molecules
US20100249382A1 (en) * 2005-10-03 2010-09-30 Xencor, Inc. MODIFIED Fc MOLECULES
US7973136B2 (en) 2005-10-06 2011-07-05 Xencor, Inc. Optimized anti-CD30 antibodies
US9574006B2 (en) 2005-10-06 2017-02-21 Xencor, Inc. Optimized anti-CD30 antibodies
US20080267976A1 (en) * 2005-10-06 2008-10-30 Gregory Alan Lazar Optimized Anti-Cd30 Antibodies
US10450379B2 (en) 2005-11-15 2019-10-22 Genetech, Inc. Method for treating joint damage
US10654940B2 (en) 2005-11-15 2020-05-19 Genentech, Inc. Method for treating joint damage
US20070212733A1 (en) * 2005-11-23 2007-09-13 Genentech, Inc. Methods and compositions related to B cell assays
US9726673B2 (en) 2005-11-23 2017-08-08 Genentech, Inc. Methods and compositions related to B cell assays
US20090155283A1 (en) * 2005-12-01 2009-06-18 Drew Philip D Noncompetitive Domain Antibody Formats That Bind Interleukin 1 Receptor Type 1
EP2543384A2 (en) 2005-12-02 2013-01-09 Biogen Idec MA Inc. Treatment of conditions involving demyelination
US20090098124A1 (en) * 2006-03-10 2009-04-16 Macrogenics, Inc. Identification and engineering of antibodies with variant heavy chains and methods of using same
US8216579B2 (en) 2006-05-26 2012-07-10 Macrogenics, Inc. Humanized FcγRIIB-specific antibodies and methods of use thereof
US8409577B2 (en) 2006-06-12 2013-04-02 Emergent Product Development Seattle, Llc Single chain multivalent binding proteins with effector function
US20110033483A1 (en) * 2006-06-12 2011-02-10 Trubion Pharmaceuticals Inc. Single-chain multivalent binding proteins with effector function
US9737599B2 (en) 2006-06-26 2017-08-22 Macrogenics, Inc. Combination of FcγRIIB-specific antibodies and CD20-specific antibodies and methods of use thereof
US8778339B2 (en) 2006-06-26 2014-07-15 Macrogenics, Inc. Combination of FcγRIIB-specific antibodies and CD20-specific antibodies and methods of use thereof
US20090191195A1 (en) * 2006-06-26 2009-07-30 Macrogenics, Inc. Combination of FcgammaRIIB-Specific Antibodies and CD20-Specific Antibodies and Methods of Use Thereof
US11098125B2 (en) 2006-06-26 2021-08-24 Macrogenics, Inc. FcγRIIB-specific antibodies and methods of use thereof
US20080044429A1 (en) * 2006-06-26 2008-02-21 Macrogenics, Inc. Fc.gamma.RIIB-Specific Antibodies and Methods of Use Thereof
US10100116B2 (en) 2006-06-26 2018-10-16 Macrogenics, Inc. FcγRIIB-specific antibodies and methods of use thereof
US8785599B2 (en) 2006-06-26 2014-07-22 Macrogenics, Inc. FcγRIIB—specific antibodies and methods of use thereof
WO2008011081A2 (en) 2006-07-19 2008-01-24 The Trustees Of The University Of Pennsylvania Wsx-1/p28 as a target for anti-inflammatory responses
US11618788B2 (en) 2006-08-14 2023-04-04 Xencor, Inc. Optimized antibodies that target CD19
US8524867B2 (en) 2006-08-14 2013-09-03 Xencor, Inc. Optimized antibodies that target CD19
US10626182B2 (en) 2006-08-14 2020-04-21 Xencor, Inc. Optimized antibodies that target CD19
US20100272723A1 (en) * 2006-08-14 2010-10-28 Xencor, Inc. Optimized Antibodies that Target CD19
US9803020B2 (en) 2006-08-14 2017-10-31 Xencor, Inc. Optimized antibodies that target CD19
WO2008024188A3 (en) * 2006-08-18 2008-09-04 Abbott Lab Dual variable domain immunoglobulin and uses thereof
US8394374B2 (en) 2006-09-18 2013-03-12 Xencor, Inc. Optimized antibodies that target HM1.24
US9040042B2 (en) 2006-09-18 2015-05-26 Xencor, Inc. Optimized antibodies that target HM1.24
US20100104557A1 (en) * 2006-09-18 2010-04-29 Xencor, Inc. Optimized Antibodies that Target HM1.24
US20080112961A1 (en) * 2006-10-09 2008-05-15 Macrogenics, Inc. Identification and Engineering of Antibodies with Variant Fc Regions and Methods of Using Same
US8541552B2 (en) 2006-10-12 2013-09-24 Genetech, Inc. Antibodies to lymphotoxin-α
US8642740B2 (en) 2006-10-12 2014-02-04 Genentech, Inc. Antibodies to lymphotoxin-alpha
US20110150865A1 (en) * 2006-10-12 2011-06-23 Genentech, Inc. Antibodies to lymphotoxin-alpha
US7923011B2 (en) 2006-10-12 2011-04-12 Genentech, Inc. Antibodies to lymphotoxin-alpha
US8216807B2 (en) 2006-10-12 2012-07-10 Genentech, Inc. Antibodies to lymphotoxin-α
US20110208673A1 (en) * 2006-10-12 2011-08-25 Genentech, Inc. Antibodies to lymphotoxin-alpha
US10711069B2 (en) 2006-12-08 2020-07-14 Macrogenics, Inc. Methods for the treatment of disease using immunoglobulins having Fc regions with altered affinities for FcγRactivating and FcγRinhibiting
US9708408B2 (en) 2006-12-08 2017-07-18 Macrogenics, Inc. Methods for the treatment of disease using immunoglobulins having Fc Regions with altered affinities for FcγRactivating and FcγRinhibiting
US8652466B2 (en) 2006-12-08 2014-02-18 Macrogenics, Inc. Methods for the treatment of disease using immunoglobulins having Fc regions with altered affinities for FcγRactivating and FcγRinhibiting
US20080138349A1 (en) * 2006-12-08 2008-06-12 Macrogenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
US11787871B2 (en) 2006-12-08 2023-10-17 Macrogenics, Inc. Methods for the treatment of disease using immunoglobulins having fc regions with altered affinities for FcgammaRactivating and FegammaRinhibiting
US20100150950A1 (en) * 2006-12-14 2010-06-17 Medarex, Inc. Human antibodies that bind cd70 and uses thereof
US20090186034A1 (en) * 2006-12-19 2009-07-23 Genetech, Inc. Gene expression markers for inflammatory bowel disease
US8128926B2 (en) 2007-01-09 2012-03-06 Biogen Idec Ma Inc. Sp35 antibodies and uses thereof
US8609407B2 (en) 2007-01-09 2013-12-17 Biogen Idec Ma Inc. Sp35 antibodies and uses thereof
US20100166744A1 (en) * 2007-01-25 2010-07-01 Wong Kwok-Kin Use of anti-egfr antibodies in treatment of egfr mutant mediated disease
US9090693B2 (en) 2007-01-25 2015-07-28 Dana-Farber Cancer Institute Use of anti-EGFR antibodies in treatment of EGFR mutant mediated disease
US8940302B2 (en) 2007-03-02 2015-01-27 Genentech, Inc. Predicting response to a HER inhibitor
US7981418B2 (en) 2007-03-02 2011-07-19 Genentech, Inc. Predicting response to a HER inhibitor
EP2899541A1 (en) 2007-03-02 2015-07-29 Genentech, Inc. Predicting response to a HER dimerisation inhbitor based on low HER3 expression
US9023356B2 (en) 2007-03-15 2015-05-05 Ludwig Institute For Cancer Research Ltd Treatment method using EGFR antibodies and SRC inhibitors and related formulations
US20100092475A1 (en) * 2007-03-15 2010-04-15 Terrance Grant Johns Treatment method using egfr antibodies and src inhibitors and related formulations
US20100233689A1 (en) * 2007-03-22 2010-09-16 The General Hospital Corporation Pyrazoloanthrone and derivatives thereof for the treatment of cancer expressing 'mullerian inhibiting substance' type ii receptor (misrii) and of excess androgen states
US9260759B2 (en) 2007-03-22 2016-02-16 The General Hospital Corporation Pyrazoloanthrone and derivatives thereof for the treatment of cancers expressing MISRII
US8877186B2 (en) 2007-06-06 2014-11-04 Domantis Limited Polypeptides, antibody variable domains and antagonists
US20100291103A1 (en) * 2007-06-06 2010-11-18 Domantis Limited Polypeptides, antibody variable domains and antagonists
EP2592156A2 (en) 2007-06-08 2013-05-15 Genentech, Inc. Gene expression markers of tumor resistance to HER2 inhibitor treatment
US20080313379A1 (en) * 2007-06-15 2008-12-18 United Memories, Inc. Multiple bus charge sharing
US20100322939A1 (en) * 2007-06-21 2010-12-23 Genmab A/S Novel methods for treating egfr-associated tumors
EP4219522A2 (en) 2007-07-09 2023-08-02 Genentech, Inc. Prevention of disulfide bond reduction during recombinant production of polypeptides
EP4365189A2 (en) 2007-07-09 2024-05-08 Genentech, Inc. Prevention of disulfide bond reduction during recombinant production of polypeptides
EP3327026A1 (en) 2007-07-09 2018-05-30 Genentech, Inc. Prevention of disulfide bond reduction during recombinant production of polypeptides
EP2586788A1 (en) 2007-07-09 2013-05-01 Genentech, Inc. Prevention of disulfide bond reduction during recombinant production of polypeptides
EP4245766A2 (en) 2007-07-09 2023-09-20 Genentech, Inc. Prevention of disulfide bond reduction during recombinant production of polypeptides
EP4335863A2 (en) 2007-07-09 2024-03-13 Genentech, Inc. Prevention of disulfide bond reduction during recombinant production of polypeptides
US9283276B2 (en) 2007-08-14 2016-03-15 Ludwig Institute For Cancer Research Ltd. Monoclonal antibody 175 targeting the EGF receptor and derivatives and uses thereof
US20100239582A1 (en) * 2007-09-26 2010-09-23 Ucb Pharma S.A. Dual Specificity Antibody Fusions
US10100130B2 (en) 2007-09-26 2018-10-16 Ucb Biopharma Sprl Dual specificity antibody fusions
US8629246B2 (en) 2007-09-26 2014-01-14 Ucb Pharma S.A. Dual specificity antibody fusions
US9309327B2 (en) 2007-09-26 2016-04-12 Ucb Pharma S.A. Dual specificity antibody fusions
US11427650B2 (en) 2007-09-26 2022-08-30 UCB Biopharma SRL Dual specificity antibody fusions
US9828438B2 (en) 2007-09-26 2017-11-28 Ucb Pharma S.A. Dual specificity antibody fusions
US20090155255A1 (en) * 2007-09-27 2009-06-18 Biogen Idec Ma Inc. Cd23 binding molecules and methods of use thereof
US20100297121A1 (en) * 2007-10-11 2010-11-25 Biogen Idec Ma Inc. Methods for Treating Pressure Induced Optic Neuropathy, Preventing Neuronal Degeneration and Promoting Neuronal Cell Survival Via Administration of LINGO-1 Antagonists and TrkB Agonists
US9738728B2 (en) 2007-10-15 2017-08-22 Sanofi Antibodies that bind IL-4 and/or IL-13 and their uses
US11453727B2 (en) 2007-10-15 2022-09-27 Sanofi Antibodies that bind IL-4 and/or IL-13 and their uses
US10759871B2 (en) 2007-10-15 2020-09-01 Sanofi Antibodies that bind IL-4 and/or IL-13 and their uses
US9732162B2 (en) 2007-10-15 2017-08-15 Sanofi Antibodies that bind IL-4 and/or IL-13 and their uses
EP2233149A1 (en) 2007-10-16 2010-09-29 ZymoGenetics, Inc. Combination of BLYS inhibition and anti-CD20 agents for treatment of autoimmune disease
US11932685B2 (en) 2007-10-31 2024-03-19 Xencor, Inc. Fc variants with altered binding to FcRn
US20110123553A1 (en) * 2007-11-08 2011-05-26 Biogen Idec Ma Inc. Use of LINGO-4 Antagonists in the Treatment of Conditions Involving Demyelination
EP2261367A2 (en) 2007-11-29 2010-12-15 Genentech, Inc. Gene expression markers for inflammatory bowel disease
US20110008345A1 (en) * 2007-11-30 2011-01-13 Claire Ashman Antigen-binding constructs
EP2077281A1 (en) 2008-01-02 2009-07-08 Bergen Teknologioverforing AS Anti-CD20 antibodies or fragments thereof for the treatment of chronic fatigue syndrome
US20090196879A1 (en) * 2008-01-02 2009-08-06 Olav Mella B-cell depleting agents, like anti-CD20 antibodies or fragments thereof for the treatment of chronic fatigue syndrome
US7914785B2 (en) 2008-01-02 2011-03-29 Bergen Teknologieverforing As B-cell depleting agents, like anti-CD20 antibodies or fragments thereof for the treatment of chronic fatigue syndrome
US20130287780A1 (en) * 2008-01-22 2013-10-31 Multimerics Aps Products and methods to prevent infections
US9259440B2 (en) * 2008-01-22 2016-02-16 Multimerics Aps Methods for passive immunization
EP3401335A1 (en) 2008-01-30 2018-11-14 Genentech, Inc. Composition comprising antibody that binds to domain ii of her2 and acidic variants thereof
EP4119583A1 (en) 2008-01-30 2023-01-18 Genentech, Inc. Composition comprising antibody that binds to domain ii of her2 and acidic variants thereof
US10131713B2 (en) 2008-04-02 2018-11-20 Macrogenics, Inc. HER2/neu-specific antibodies and methods of using same
US8802093B2 (en) 2008-04-02 2014-08-12 Macrogenics, Inc. HER2/neu-specific antibodies and methods of using same
US11028183B2 (en) 2008-04-02 2021-06-08 Macrogenics, Inc. HER2/neu-specific antibodies and methods of using same
US9469692B2 (en) 2008-04-02 2016-10-18 Macrogenics, Inc. HER2/neu-specific antibodies and methods of using same
US20110097323A1 (en) * 2008-04-02 2011-04-28 Macrogenics, Inc. Her2/neu-Specific Antibodies and Methods of Using Same
US9243069B2 (en) 2008-04-02 2016-01-26 Macrogenics, Inc. HER2/neu-specific antibodies and methods of using the same
US9101609B2 (en) 2008-04-11 2015-08-11 Emergent Product Development Seattle, Llc CD37 immunotherapeutic and combination with bifunctional chemotherapeutic thereof
US20090274692A1 (en) * 2008-04-11 2009-11-05 Trubion Pharmaceuticals, Inc. Cd37 immunotherapeutic and combination with bifunctional chemotherapeutic thereof
US8333966B2 (en) 2008-04-11 2012-12-18 Emergent Product Development Seattle, Llc CD37 immunotherapeutics and uses thereof
US20100260668A1 (en) * 2008-04-29 2010-10-14 Abbott Laboratories Dual Variable Domain Immunoglobulins and Uses Thereof
US9029508B2 (en) 2008-04-29 2015-05-12 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
US9109026B2 (en) 2008-06-03 2015-08-18 Abbvie, Inc. Dual variable domain immunoglobulins and uses thereof
US20090304693A1 (en) * 2008-06-03 2009-12-10 Abbott Laboratories Dual Variable Domain Immunoglobulins and Uses Thereof
US9035027B2 (en) 2008-06-03 2015-05-19 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
US20090311253A1 (en) * 2008-06-03 2009-12-17 Abbott Laboratories Dual Variable Domain Immunoglobulins and Uses Thereof
US8822645B2 (en) 2008-07-08 2014-09-02 Abbvie Inc. Prostaglandin E2 dual variable domain immunoglobulins and uses thereof
US9745375B2 (en) 2008-07-09 2017-08-29 Biogen Ma Inc. Compositions comprising antibodies to LINGO or fragments thereof
US8058406B2 (en) 2008-07-09 2011-11-15 Biogen Idec Ma Inc. Composition comprising antibodies to LINGO or fragments thereof
US20100015131A1 (en) * 2008-07-09 2010-01-21 Biogen Idec Ma Inc. Composition Comprising Antibodies to LINGO or Fragments Thereof
US8425910B2 (en) 2008-07-09 2013-04-23 Biogen Idec Ma Inc. Composition comprising antibodies to LINGO or fragments thereof
US20110236304A1 (en) * 2008-07-21 2011-09-29 Immunomedics, Inc. Structural variants of antibodies for improved therapeutic characteristics
US7919273B2 (en) 2008-07-21 2011-04-05 Immunomedics, Inc. Structural variants of antibodies for improved therapeutic characteristics
US20100034738A1 (en) * 2008-07-21 2010-02-11 Immunomedics, Inc. Structural Variants of Antibodies for Improved Therapeutic Characteristics
EP3095463A2 (en) 2008-09-16 2016-11-23 F. Hoffmann-La Roche AG Methods for treating progressive multiple sclerosis
US9683047B2 (en) 2008-09-16 2017-06-20 Genentech, Inc. Methods for treating progressive multiple sclerosis
US9994642B2 (en) 2008-09-16 2018-06-12 Genentech, Inc. Methods for treating progressive multiple sclerosis
EP3747464A1 (en) 2008-09-16 2020-12-09 F. Hoffmann-La Roche AG Methods for treating progessive multiple sclerosis using an anti-cd20 antibody
EP4364800A2 (en) 2008-09-16 2024-05-08 F. Hoffmann-La Roche AG Methods for treating progressive multiple sclerosis
US20100158903A1 (en) * 2008-09-16 2010-06-24 Craig Smith Methods for treating progressive multiple sclerosis
US10407513B2 (en) 2008-09-26 2019-09-10 Ucb Biopharma Sprl Biological products
US20110184152A1 (en) * 2008-09-26 2011-07-28 Ucb Pharma S.A. Biological Products
EP2752189A1 (en) 2008-11-22 2014-07-09 F. Hoffmann-La Roche AG Use of anti-vegf antibody in combination with chemotherapy for treating breast cancer
EP3178478A1 (en) 2008-11-22 2017-06-14 F. Hoffmann-La Roche AG Use of anti-vegf antibody in combination with chemotherapy for treating breast cancer
US20100233079A1 (en) * 2008-12-04 2010-09-16 Abbott Laboratories Dual Variable Domain Immunoglobulins and Uses Thereof
WO2010075249A2 (en) 2008-12-22 2010-07-01 Genentech, Inc. A method for treating rheumatoid arthritis with b-cell antagonists
US20110142836A1 (en) * 2009-01-02 2011-06-16 Olav Mella B-cell depleting agents for the treatment of chronic fatigue syndrome
US9072798B2 (en) 2009-02-18 2015-07-07 Ludwig Institute For Cancer Research Ltd. Specific binding proteins and uses thereof
EP2584049A2 (en) 2009-07-20 2013-04-24 Genentech, Inc. Gene expression markers for Crohn's disease
WO2011011339A1 (en) 2009-07-20 2011-01-27 Genentech, Inc. Gene expression markers for crohn's disease
EP2757160A2 (en) 2009-07-20 2014-07-23 Genentech, Inc. Gene expression markers for Crohn's disease
US20110044980A1 (en) * 2009-07-29 2011-02-24 Abbott Laboratories Dual Variable Domain Immunoglobulins and Uses Thereof
WO2011019619A1 (en) 2009-08-11 2011-02-17 Genentech, Inc. Production of proteins in glutamine-free cell culture media
US9714293B2 (en) 2009-08-11 2017-07-25 Genentech, Inc. Production of proteins in glutamine-free cell culture media
US8512983B2 (en) 2009-08-11 2013-08-20 Martin Gawlitzek Production of proteins in glutamine-free cell culture media
US10982003B2 (en) 2009-08-11 2021-04-20 Genentech, Inc. Production of proteins in glutamine-free cell culture media
EP3760712A1 (en) 2009-08-11 2021-01-06 F. Hoffmann-La Roche AG Production of proteins in glutamine-free cell culture media
WO2011022264A1 (en) 2009-08-15 2011-02-24 Genentech, Inc. Anti-angiogenesis therapy for the treatment of previously treated breast cancer
EP3090758A1 (en) 2009-08-15 2016-11-09 F. Hoffmann-La Roche AG Anti-angiogenesis therapy for the treatment of previously treated breast cancer
US8586714B2 (en) 2009-09-01 2013-11-19 Abbvie, Inc. Dual variable domain immunoglobulins and uses thereof
US11401348B2 (en) 2009-09-02 2022-08-02 Xencor, Inc. Heterodimeric Fc variants
US20110076232A1 (en) * 2009-09-29 2011-03-31 Ludwig Institute For Cancer Research Specific binding proteins and uses thereof
US8716450B2 (en) 2009-10-15 2014-05-06 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
US20110212094A1 (en) * 2009-10-28 2011-09-01 Abbott Laboratories Dual variable domain immunoglobulins and uses thereof
US8722855B2 (en) 2009-10-28 2014-05-13 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
US9475881B2 (en) 2010-01-19 2016-10-25 Xencor, Inc. Antibody variants with enhanced complement activity
WO2011100403A1 (en) 2010-02-10 2011-08-18 Immunogen, Inc Cd20 antibodies and uses thereof
US8883985B2 (en) * 2010-02-16 2014-11-11 National University Corporation Kyoto Institute Of Technology Antibody-immobilized carrier, method of producing antibody-immobilized carrier, and use of said antibody-immobilized carrier
US20120309943A1 (en) * 2010-02-16 2012-12-06 Yoichi Kumada Antibody-immobilized carrier, method of producing antibody-immobilized carrier, and use of said antibody-immobilized carrier
US9598501B2 (en) 2010-02-16 2017-03-21 National University Corporation Kyoto Institute Of Technology Antibody-immobilized carrier, method of producing antibody-immobilized carrier, and use of said antibody-immobilized carrier
EP3696194A1 (en) 2010-02-23 2020-08-19 F. Hoffmann-La Roche AG Anti-angiogenesis therapy for the treatment of ovarian cancer
WO2011106300A2 (en) 2010-02-23 2011-09-01 Genentech, Inc. Anti-angiogenesis therapy for the treatment of ovarian cancer
EP3064509A2 (en) 2010-02-23 2016-09-07 F. Hoffmann-La Roche AG Anti-angiogenesis therapy for the treatment of ovarian cancer
US9637557B2 (en) 2010-04-23 2017-05-02 Genentech, Inc. Production of heteromultimeric proteins
WO2011133886A2 (en) 2010-04-23 2011-10-27 Genentech, Inc. Production of heteromultimeric proteins
WO2011146568A1 (en) 2010-05-19 2011-11-24 Genentech, Inc. Predicting response to a her inhibitor
WO2011153243A2 (en) 2010-06-02 2011-12-08 Genentech, Inc. Anti-angiogenesis therapy for treating gastric cancer
WO2012012750A1 (en) 2010-07-23 2012-01-26 Trustees Of Boston University ANTI-DEsupR INHIBITORS AS THERAPEUTICS FOR INHIBITION OF PATHOLOGICAL ANGIOGENESIS AND TUMOR CELL INVASIVENESS AND FOR MOLECULAR IMAGING AND TARGETED DELIVERY
EP3696195A1 (en) 2010-07-23 2020-08-19 Trustees of Boston University Anti-despr inhibitors as therapeutics for inhibition of pathological angiogenesis and tumor cell invasiveness and for molecular imaging and targeted delivery
US9493560B2 (en) 2010-08-03 2016-11-15 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
US8735546B2 (en) 2010-08-03 2014-05-27 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
US9046513B2 (en) 2010-08-26 2015-06-02 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
US9085622B2 (en) 2010-09-03 2015-07-21 Glaxosmithkline Intellectual Property Development Limited Antigen binding proteins
EP3351559A2 (en) 2010-11-08 2018-07-25 F. Hoffmann-La Roche AG Subcutaneously administered anti-il-6 receptor antibody
EP2787007A2 (en) 2010-11-08 2014-10-08 F. Hoffmann-La Roche AG Subcutaneously administered ANTI-IL-6 receptor antibody
WO2012064627A2 (en) 2010-11-08 2012-05-18 Genentech, Inc. Subcutaneously administered anti-il-6 receptor antibody
EP4029881A1 (en) 2010-11-08 2022-07-20 F. Hoffmann-La Roche AG Subcutaneously administered anti-il-6 receptor antibody
WO2012071436A1 (en) 2010-11-24 2012-05-31 Genentech, Inc. Method of treating autoimmune inflammatory disorders using il-23r loss-of-function mutants
EP3208282A1 (en) 2010-11-30 2017-08-23 F. Hoffmann-La Roche AG Low affinity anti transferrin receptor and their use to transfer therapeutic scfv across the blood brain barrier
WO2012075037A1 (en) 2010-11-30 2012-06-07 Genentech, Inc. Low affinity blood brain barrier receptor antibodies and uses therefor
US11912773B2 (en) 2011-02-04 2024-02-27 Genentech, Inc. Fc variants and methods for their production
US10689447B2 (en) 2011-02-04 2020-06-23 Genentech, Inc. Fc variants and methods for their production
WO2012106587A1 (en) 2011-02-04 2012-08-09 Genentech, Inc. Fc VARIANTS AND METHODS FOR THEIR PRODUCTION
US11008389B2 (en) 2011-03-16 2021-05-18 Sanofi Uses of a dual V region antibody-like protein
US20130060011A1 (en) * 2011-08-23 2013-03-07 Peter Bruenker Fc-free antibodies comprising two fab fragments and methods of use
US20130078249A1 (en) * 2011-08-23 2013-03-28 Oliver Ast Bispecific t cell activating antigen binding molecules
US11639397B2 (en) 2011-08-23 2023-05-02 Roche Glycart Ag Bispecific antibodies specific for T-cell activating antigens and a tumor antigen and methods of use
US10626189B2 (en) 2011-10-11 2020-04-21 Genentech, Inc. Assembly of bispecific antibodies
US11725065B2 (en) 2011-10-11 2023-08-15 Genentech, Inc. Assembly of bispecific antibodies
US9862778B2 (en) 2011-10-11 2018-01-09 Genentech, Inc. Assembly of bispecific antibodies
EP3418306A1 (en) 2011-10-11 2018-12-26 F. Hoffmann-La Roche AG Improved assembly of bispecific antibodies
WO2013055958A1 (en) 2011-10-11 2013-04-18 Genentech, Inc. Improved assembly of bispecific antibodies
US9120870B2 (en) 2011-12-30 2015-09-01 Abbvie Inc. Dual specific binding proteins directed against IL-13 and IL-17
WO2013101771A2 (en) 2011-12-30 2013-07-04 Genentech, Inc. Compositions and method for treating autoimmune diseases
WO2013112467A1 (en) 2012-01-23 2013-08-01 Trustees Of Boston University DEspR ANTAGONISTS AND AGONISTS AS THERAPEUTICS
US11814409B2 (en) 2012-02-15 2023-11-14 Hoffmann-La Roche Inc. Fc-receptor based affinity chromatography
US9796780B2 (en) 2012-05-14 2017-10-24 Biogen Ma Inc. LINGO-2 antagonists for treatment of conditions involving motor neurons
WO2013177062A2 (en) 2012-05-21 2013-11-28 Genentech, Inc. Methods for improving safety of blood-brain barrier transport
EP3594239A1 (en) 2012-05-21 2020-01-15 F. Hoffmann-La Roche AG Methods for improving safety of blood-brain barrier transport
US11254740B2 (en) 2012-05-22 2022-02-22 Bristol-Myers Squibb Company Methods of treatment using IL-23p19 monoclonal antibodies
US9783606B2 (en) 2012-05-22 2017-10-10 Bristol-Myers Squibb Company Polynucleotides encoding IL-17/IL-23 bispecific, IL-17A/F and IL-23p19 antibodies
US8945553B2 (en) 2012-05-22 2015-02-03 Bristol-Myers Squibb Company Bispecific antibodies to IL-23 and IL-17A/F
US10358489B2 (en) 2012-05-22 2019-07-23 Bristol-Myers Squibb Company IL-23p19 monoclonal antibodies
US10358490B2 (en) 2012-05-22 2019-07-23 Bristol-Myers Squibb Company Methods of treating inflammation with IL-17A/F and IL-23P19 bispecific antibodies
US9708401B2 (en) 2012-05-22 2017-07-18 Bristol-Myers Squibb Company IL-17A/F cross-reactive monoclonal antibodies and methods of using the same
WO2014033074A1 (en) 2012-08-29 2014-03-06 F. Hoffmann-La Roche Ag Blood brain barrier shuttle
US20160130347A1 (en) * 2012-10-08 2016-05-12 Roche Glycart Ag Fc-free antibodies comprising two Fab-fragments and methods of use
US10087250B2 (en) * 2012-10-08 2018-10-02 Roche Glycart Ag Fc-free antibodies comprising two fab-fragments and methods of use
US9045551B2 (en) 2012-11-01 2015-06-02 Abbvie Inc. Anti-DLL4/VEGF dual variable domain immunoglobulin and uses thereof
US9163093B2 (en) 2012-11-01 2015-10-20 Abbvie Inc. Anti-DLL4/VEGF dual variable domain immunoglobulin and uses thereof
US9944720B2 (en) 2012-11-01 2018-04-17 Abbvie Inc. Anti-DLL4/VEGF dual variable domain immunoglobulin and uses thereof
US20140242079A1 (en) * 2013-02-26 2014-08-28 Roche Glycart Ag Bispecific t cell activating antigen binding molecules
JP2019135233A (ja) * 2013-02-26 2019-08-15 ロシュ グリクアート アーゲー 二重特異性t細胞活性化抗原結合分子
JP7057801B2 (ja) 2013-02-26 2022-04-20 ロシュ グリクアート アーゲー 二重特異性t細胞活性化抗原結合分子
US11459404B2 (en) 2013-02-26 2022-10-04 Roche Glycart Ag Bispecific T cell activating antigen binding molecules
JP2016514098A (ja) * 2013-02-26 2016-05-19 ロシュ グリクアート アーゲー 二重特異性t細胞活性化抗原結合分子
US10781258B2 (en) 2013-02-26 2020-09-22 Roche Glycart Ag Bispecific T cell activating antigen binding molecules
US10781257B2 (en) 2013-02-26 2020-09-22 Roche GlyeArt AG Bispecific T cell activating antigen binding molecules
US10155815B2 (en) * 2013-02-26 2018-12-18 Roche Glycart Ag Bispecific T cell activating antigen binding molecules
JP2020202829A (ja) * 2013-02-26 2020-12-24 ロシュ グリクアート アーゲー 二重特異性t細胞活性化抗原結合分子
US11634502B2 (en) 2013-03-15 2023-04-25 Amgen Inc. Heterodimeric bispecific antibodies
US8987418B2 (en) 2013-03-15 2015-03-24 Abbvie Inc. Dual specific binding proteins directed against IL-1β and/or IL-17
US9062108B2 (en) 2013-03-15 2015-06-23 Abbvie Inc. Dual specific binding proteins directed against IL-1 and/or IL-17
EP4324480A2 (en) 2013-05-20 2024-02-21 F. Hoffmann-La Roche AG Anti-transferrin receptor antibodies and methods of use
EP3594240A1 (en) 2013-05-20 2020-01-15 F. Hoffmann-La Roche AG Anti-transferrin receptor antibodies and methods of use
WO2015023596A1 (en) 2013-08-12 2015-02-19 Genentech, Inc. Compositions and method for treating complement-associated conditions
US10519251B2 (en) * 2013-12-30 2019-12-31 Epimab Biotherapeutics, Inc. Fabs-in-tandem immunoglobulin and uses thereof
US20160289341A1 (en) * 2013-12-30 2016-10-06 Epimab Biotherapeutics Inc. Fabs-in-tandem immunoglobulin and uses thereof
US10266608B2 (en) 2013-12-30 2019-04-23 Epimab Biotherapeutics, Inc. Fabs-in-tandem immunoglobulin and uses thereof
WO2015103072A1 (en) * 2013-12-30 2015-07-09 Epimab Biotherapeutics Fabs-in-tandem immunoglobulin and uses thereof
EP3514179A1 (en) 2014-01-24 2019-07-24 Dana-Farber Cancer Institute, Inc. Antibody molecules to pd-1 and uses thereof
EP4324518A2 (en) 2014-01-31 2024-02-21 Novartis AG Antibody molecules to tim-3 and uses thereof
WO2015138920A1 (en) 2014-03-14 2015-09-17 Novartis Ag Antibody molecules to lag-3 and uses thereof
EP3660050A1 (en) 2014-03-14 2020-06-03 Novartis AG Antibody molecules to lag-3 and uses thereof
WO2015142675A2 (en) 2014-03-15 2015-09-24 Novartis Ag Treatment of cancer using chimeric antigen receptor
WO2015171822A1 (en) 2014-05-06 2015-11-12 Genentech, Inc. Production of heteromultimeric proteins using mammalian cells
EP4306544A2 (en) 2014-05-06 2024-01-17 F. Hoffmann-La Roche AG Production of heteromultimeric proteins using mammalian cells
WO2015175375A1 (en) 2014-05-13 2015-11-19 Short Jay M Conditionally active biological proteins
WO2016014530A1 (en) 2014-07-21 2016-01-28 Novartis Ag Combinations of low, immune enhancing. doses of mtor inhibitors and cars
WO2016014553A1 (en) 2014-07-21 2016-01-28 Novartis Ag Sortase synthesized chimeric antigen receptors
EP3722316A1 (en) 2014-07-21 2020-10-14 Novartis AG Treatment of cancer using a cd33 chimeric antigen receptor
WO2016014565A2 (en) 2014-07-21 2016-01-28 Novartis Ag Treatment of cancer using humanized anti-bcma chimeric antigen receptor
EP4205749A1 (en) 2014-07-31 2023-07-05 Novartis AG Subset-optimized chimeric antigen receptor-containing cells
EP3660042A1 (en) 2014-07-31 2020-06-03 Novartis AG Subset-optimized chimeric antigen receptor-containing t-cells
US10611841B2 (en) 2014-08-04 2020-04-07 Hoffmann-La Roche Inc. Bispecific T cell activating antigen binding molecules
US9914776B2 (en) 2014-08-04 2018-03-13 Hoffmann-La Roche Inc. Bispecific T cell activating antigen binding molecules
US10611840B2 (en) 2014-08-04 2020-04-07 Hoffman-La Roche Inc. Bispecific T cell activating antigen binding molecules
US11117965B2 (en) 2014-08-04 2021-09-14 Hoffmann-La Roche Inc. Bispecific T cell activating antigen binding molecules
US9771417B2 (en) 2014-08-07 2017-09-26 Novartis Ag Angiopoietin-like 4 antibodies and methods of use
US9988443B2 (en) 2014-08-07 2018-06-05 Novartis Ag Angiopoetin-like 4 (ANGPTL4) antibodies and methods of use
US10577411B2 (en) 2014-08-07 2020-03-03 Novartis Ag Angiopoietin-like 4 antibodies and methods of use
WO2016025880A1 (en) 2014-08-14 2016-02-18 Novartis Ag Treatment of cancer using gfr alpha-4 chimeric antigen receptor
EP3712171A1 (en) 2014-08-19 2020-09-23 Novartis AG Treatment of cancer using a cd123 chimeric antigen receptor
US11111288B2 (en) 2014-08-28 2021-09-07 Bioatla, Inc. Conditionally active chimeric antigen receptors for modified t-cells
US11584927B2 (en) 2014-08-28 2023-02-21 Bioatla, Inc. Conditionally active chimeric antigen receptors for modified T-cells
EP4074735A1 (en) 2014-08-28 2022-10-19 BioAtla, Inc. Conditionally active chimeric antigen receptors for modified t-cells
WO2016033331A1 (en) 2014-08-28 2016-03-03 Bioatla, Llc Conditionally active chimeric antigen receptors for modified t-cells
WO2016044605A1 (en) 2014-09-17 2016-03-24 Beatty, Gregory Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy
EP3967709A1 (en) 2014-09-17 2022-03-16 Novartis AG Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy
WO2016061142A1 (en) 2014-10-14 2016-04-21 Novartis Ag Antibody molecules to pd-l1 and uses thereof
EP4245376A2 (en) 2014-10-14 2023-09-20 Novartis AG Antibody molecules to pd-l1 and uses thereof
WO2016081643A1 (en) 2014-11-19 2016-05-26 Genentech, Inc. Anti-transferrin receptor antibodies and methods of use
US10927185B2 (en) * 2014-11-21 2021-02-23 Astellas Pharma Inc. Bispecific antibody format
US20170306053A1 (en) * 2014-11-21 2017-10-26 Astellas Pharma Inc. Novel bispecific antibody format
WO2016090034A2 (en) 2014-12-03 2016-06-09 Novartis Ag Methods for b cell preconditioning in car therapy
US10093733B2 (en) 2014-12-11 2018-10-09 Abbvie Inc. LRP-8 binding dual variable domain immunoglobulin proteins
US10435467B2 (en) 2015-01-08 2019-10-08 Biogen Ma Inc. LINGO-1 antagonists and uses for treatment of demyelinating disorders
WO2016115274A1 (en) * 2015-01-14 2016-07-21 Compass Therapeutics Llc Multispecific immunomodulatory antigen-binding constructs
US10501552B2 (en) 2015-01-26 2019-12-10 Macrogenics, Inc. Multivalent molecules comprising DR5-binding domains
WO2016123329A2 (en) 2015-01-28 2016-08-04 Genentech, Inc. Gene expression markers and treatment of multiple sclerosis
US11236391B2 (en) 2015-01-28 2022-02-01 Genentech, Inc. Gene expression markers and treatment of multiple sclerosis
US10563194B2 (en) 2015-02-24 2020-02-18 BioAlta, LLC Conditionally active biological proteins
US11254932B2 (en) 2015-02-24 2022-02-22 Bioatla, Inc. Conditionally active biological proteins
EP4056588A1 (en) 2015-04-08 2022-09-14 Novartis AG Cd20 therapies, cd22 therapies, and combination therapies with a cd19 chimeric antigen receptor (car)- expressing cell
WO2016164731A2 (en) 2015-04-08 2016-10-13 Novartis Ag Cd20 therapies, cd22 therapies, and combination therapies with a cd19 chimeric antigen receptor (car) - expressing cell
WO2016172583A1 (en) 2015-04-23 2016-10-27 Novartis Ag Treatment of cancer using chimeric antigen receptor and protein kinase a blocker
EP3763827A1 (en) 2015-05-29 2021-01-13 F. Hoffmann-La Roche AG Pd-l1 promoter methylation in cancer
US11254987B2 (en) 2015-05-29 2022-02-22 Genentech, Inc. PD-L1 promoter methylation in cancer
WO2016196381A1 (en) 2015-05-29 2016-12-08 Genentech, Inc. Pd-l1 promoter methylation in cancer
US9840554B2 (en) 2015-06-15 2017-12-12 Abbvie Inc. Antibodies against platelet-derived growth factor (PDGF)
US11584793B2 (en) 2015-06-24 2023-02-21 Hoffmann-La Roche Inc. Anti-transferrin receptor antibodies with tailored affinity
WO2017019894A1 (en) 2015-07-29 2017-02-02 Novartis Ag Combination therapies comprising antibody molecules to lag-3
EP3964528A1 (en) 2015-07-29 2022-03-09 Novartis AG Combination therapies comprising antibody molecules to lag-3
EP3878465A1 (en) 2015-07-29 2021-09-15 Novartis AG Combination therapies comprising antibody molecules to tim-3
WO2017019897A1 (en) 2015-07-29 2017-02-02 Novartis Ag Combination therapies comprising antibody molecules to tim-3
US11352426B2 (en) 2015-09-21 2022-06-07 Aptevo Research And Development Llc CD3 binding polypeptides
US10941205B2 (en) 2015-10-02 2021-03-09 Hoffmann-La Roche Inc. Bispecific anti-human A-beta/human transferrin receptor antibodies and methods of use
US10526413B2 (en) 2015-10-02 2020-01-07 Hoffmann-La Roche Inc. Bispecific antibodies specific for OX40
US10766967B2 (en) 2015-10-02 2020-09-08 Hoffmann-La Roche Inc. Bispecific T cell activating antigen binding molecules
US11603411B2 (en) 2015-10-02 2023-03-14 Hoffmann-La Roche Inc. Bispecific anti-human CD20/human transferrin receptor antibodies and methods of use
WO2017055542A1 (en) 2015-10-02 2017-04-06 F. Hoffmann-La Roche Ag Bispecific anti-human cd20/human transferrin receptor 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
WO2017055540A1 (en) 2015-10-02 2017-04-06 F. Hoffmann-La Roche Ag Bispecific anti-human a-beta/human transferrin receptor antibodies and methods of use
WO2017062682A2 (en) 2015-10-06 2017-04-13 Genentech, Inc. Method for treating multiple sclerosis
US11472876B2 (en) 2015-11-02 2022-10-18 Bioatla, Inc. Conditionally active polypeptides
US11013801B2 (en) 2015-12-09 2021-05-25 Hoffmann-La Roche Inc. Treatment method
WO2017106810A2 (en) 2015-12-17 2017-06-22 Novartis Ag Combination of c-met inhibitor with antibody molecule to pd-1 and uses thereof
WO2017106656A1 (en) 2015-12-17 2017-06-22 Novartis Ag Antibody molecules to pd-1 and uses thereof
US10596257B2 (en) 2016-01-08 2020-03-24 Hoffmann-La Roche Inc. Methods of treating CEA-positive cancers using PD-1 axis binding antagonists and anti-CEA/anti-CD3 bispecific antibodies
WO2017123537A1 (en) 2016-01-12 2017-07-20 Bioatla, Llc Diagnostics using conditionally active antibodies
US10697972B2 (en) 2016-01-12 2020-06-30 Bioatla, Llc Diagnostics using conditionally active antibodies
WO2017124002A1 (en) * 2016-01-13 2017-07-20 Compass Therapeutics Llc Multispecific immunomodulatory antigen-binding constructs
CN109562162A (zh) * 2016-01-13 2019-04-02 指南针制药有限责任公司 多特异性免疫调节性抗原结合构建体
EP3851457A1 (en) 2016-01-21 2021-07-21 Novartis AG Multispecific molecules targeting cll-1
WO2017125897A1 (en) 2016-01-21 2017-07-27 Novartis Ag Multispecific molecules targeting cll-1
US11421028B2 (en) 2016-02-06 2022-08-23 Epimab Biotherapeutics, Inc. Fabs-in-tandem immunoglobulin and uses thereof
WO2017149515A1 (en) 2016-03-04 2017-09-08 Novartis Ag Cells expressing multiple chimeric antigen receptor (car) molecules and uses therefore
WO2017165683A1 (en) 2016-03-23 2017-09-28 Novartis Ag Cell secreted minibodies and uses thereof
US11505616B2 (en) * 2016-03-25 2022-11-22 Biomunex Pharmaceuticals Binding molecules to CD38 and PD-L1
WO2017181119A2 (en) 2016-04-15 2017-10-19 Novartis Ag Compositions and methods for selective protein expression
EP4219721A2 (en) 2016-04-15 2023-08-02 Novartis AG Compositions and methods for selective protein expression
US11879011B2 (en) 2016-05-13 2024-01-23 Bioatla, Inc. Anti-ROR2 antibodies, antibody fragments, their immunoconjucates and uses thereof
WO2017210617A2 (en) 2016-06-02 2017-12-07 Porter, David, L. Therapeutic regimens for chimeric antigen receptor (car)- expressing cells
WO2018013918A2 (en) 2016-07-15 2018-01-18 Novartis Ag Treatment and prevention of cytokine release syndrome using a chimeric antigen receptor in combination with a kinase inhibitor
WO2018023025A1 (en) 2016-07-28 2018-02-01 Novartis Ag Combination therapies of chimeric antigen receptors adn pd-1 inhibitors
WO2018026819A2 (en) 2016-08-01 2018-02-08 Novartis Ag Treatment of cancer using a chimeric antigen receptor in combination with an inhibitor of a pro-m2 macrophage molecule
WO2018067992A1 (en) 2016-10-07 2018-04-12 Novartis Ag Chimeric antigen receptors for the treatment of cancer
WO2018118780A1 (en) 2016-12-19 2018-06-28 Calico Biolabs, Inc. Monovalent and divalent binding proteins
WO2018129007A1 (en) 2017-01-03 2018-07-12 Bioatla Llc Protein therapeutics for treatment of senescent cells
US11447558B2 (en) 2017-01-03 2022-09-20 Hoffmann-La Roche Inc. Bispecific antigen binding molecules comprising anti-4-1BB clone 20H4.9
WO2018140725A1 (en) 2017-01-26 2018-08-02 Novartis Ag Cd28 compositions and methods for chimeric antigen receptor therapy
EP4043485A1 (en) 2017-01-26 2022-08-17 Novartis AG Cd28 compositions and methods for chimeric antigen receptor therapy
WO2018160731A1 (en) 2017-02-28 2018-09-07 Novartis Ag Shp inhibitor compositions and uses for chimeric antigen receptor therapy
WO2018162517A1 (en) 2017-03-10 2018-09-13 F. Hoffmann-La Roche Ag Method for producing multispecific antibodies
US11560437B2 (en) 2017-03-27 2023-01-24 Biomunex Pharmaceuticals Stable multispecific antibodies
US11186636B2 (en) 2017-04-21 2021-11-30 Amgen Inc. Anti-human TREM2 antibodies and uses thereof
WO2018201051A1 (en) 2017-04-28 2018-11-01 Novartis Ag Bcma-targeting agent, and combination therapy with a gamma secretase inhibitor
WO2018197533A1 (en) 2017-04-28 2018-11-01 F. Hoffmann-La Roche Ag Antibody selection method
US11668722B2 (en) 2017-04-28 2023-06-06 Hoffmann-La Roche Inc. Antibody selection method
WO2018201056A1 (en) 2017-04-28 2018-11-01 Novartis Ag Cells expressing a bcma-targeting chimeric antigen receptor, and combination therapy with a gamma secretase inhibitor
WO2018237157A1 (en) 2017-06-22 2018-12-27 Novartis Ag CD73 BINDING ANTIBODY MOLECULES AND USES THEREOF
WO2019006007A1 (en) 2017-06-27 2019-01-03 Novartis Ag POSOLOGICAL REGIMES FOR ANTI-TIM3 ANTIBODIES AND USES THEREOF
US11752207B2 (en) 2017-07-11 2023-09-12 Compass Therapeutics Llc Agonist antibodies that bind human CD137 and uses thereof
WO2019018730A1 (en) 2017-07-20 2019-01-24 Novartis Ag DOSAGE REGIMES FOR ANTI-LAG3 ANTIBODIES AND USES THEREOF
WO2019077092A1 (en) 2017-10-20 2019-04-25 F. Hoffmann-La Roche Ag METHOD FOR GENERATING MULTISPECIFIC ANTIBODIES FROM MONOSPECIFIC ANTIBODIES
WO2019086362A1 (en) 2017-10-30 2019-05-09 F. Hoffmann-La Roche Ag Method for in vivo generation of multispecific antibodies from monospecific antibodies
US11718679B2 (en) 2017-10-31 2023-08-08 Compass Therapeutics Llc CD137 antibodies and PD-1 antagonists and uses thereof
WO2019099838A1 (en) 2017-11-16 2019-05-23 Novartis Ag Combination therapies
US11851497B2 (en) 2017-11-20 2023-12-26 Compass Therapeutics Llc CD137 antibodies and tumor antigen-targeting antibodies and uses thereof
WO2019152660A1 (en) 2018-01-31 2019-08-08 Novartis Ag Combination therapy using a chimeric antigen receptor
WO2019200229A1 (en) 2018-04-13 2019-10-17 Novartis Ag Dosage regimens for anti-pd-l1 antibodies and uses thereof
WO2019210153A1 (en) 2018-04-27 2019-10-31 Novartis Ag Car t cell therapies with enhanced efficacy
WO2019219721A1 (en) 2018-05-18 2019-11-21 F. Hoffmann-La Roche Ag Targeted intracellular delivery of large nucleic acids
WO2019226617A1 (en) 2018-05-21 2019-11-28 Compass Therapeutics Llc Compositions and methods for enhancing the killing of target cells by nk cells
WO2019226658A1 (en) 2018-05-21 2019-11-28 Compass Therapeutics Llc Multispecific antigen-binding compositions and methods of use
WO2019227003A1 (en) 2018-05-25 2019-11-28 Novartis Ag Combination therapy with chimeric antigen receptor (car) therapies
WO2019232244A2 (en) 2018-05-31 2019-12-05 Novartis Ag Antibody molecules to cd73 and uses thereof
WO2019241426A1 (en) 2018-06-13 2019-12-19 Novartis Ag Bcma chimeric antigen receptors and uses thereof
WO2019246293A2 (en) 2018-06-19 2019-12-26 Atarga, Llc Antibody molecules to complement component 5 and uses thereof
WO2020012337A1 (en) 2018-07-10 2020-01-16 Novartis Ag 3-(5-amino-1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and their use in the treatment of i karos family zinc finger 2 (ikzf2)-dependent diseases
WO2020021465A1 (en) 2018-07-25 2020-01-30 Advanced Accelerator Applications (Italy) S.R.L. Method of treatment of neuroendocrine tumors
WO2020084032A1 (en) 2018-10-25 2020-04-30 F. Hoffmann-La Roche Ag Modification of antibody fcrn binding
WO2020084034A1 (en) 2018-10-26 2020-04-30 F. Hoffmann-La Roche Ag Multispecific antibody screening method using recombinase mediated cassette exchange
US11970538B2 (en) 2018-11-13 2024-04-30 Compass Therapeutics Llc Multispecific binding constructs against checkpoint molecules and uses thereof
US11046769B2 (en) 2018-11-13 2021-06-29 Compass Therapeutics Llc Multispecific binding constructs against checkpoint molecules and uses thereof
WO2020128898A1 (en) 2018-12-20 2020-06-25 Novartis Ag Pharmaceutical combinations
WO2020132230A2 (en) 2018-12-20 2020-06-25 Genentech, Inc. Modified antibody fcs and methods of use
WO2020128972A1 (en) 2018-12-20 2020-06-25 Novartis Ag Dosing regimen and pharmaceutical combination comprising 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives
US11739160B2 (en) 2018-12-24 2023-08-29 Sanofi PseudoFab-based multispecific binding proteins
WO2020165868A1 (en) 2019-02-15 2020-08-20 Perkinelmer Cellular Technologies Germany Gmbh Low-power microscope-objective pre-scan and high-power microscope-objective scan in x,y and z-direction for imaging objects such as cells using a microscope
WO2020165834A1 (en) 2019-02-15 2020-08-20 Novartis Ag Substituted 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
WO2020165833A1 (en) 2019-02-15 2020-08-20 Novartis Ag 3-(1-oxo-5-(piperidin-4-yl)isoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
WO2020172553A1 (en) 2019-02-22 2020-08-27 Novartis Ag Combination therapies of egfrviii chimeric antigen receptors and pd-1 inhibitors
WO2020205523A1 (en) 2019-03-29 2020-10-08 Atarga, Llc Anti fgf23 antibody
WO2020254351A1 (en) 2019-06-19 2020-12-24 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
WO2020254357A1 (en) 2019-06-19 2020-12-24 F. Hoffmann-La Roche Ag Method for the generation of a protein expressing cell by targeted integration using cre mrna
WO2020254352A1 (en) 2019-06-19 2020-12-24 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
WO2020254355A1 (en) 2019-06-19 2020-12-24 F. Hoffmann-La Roche Ag Method for the generation of a bivalent, bispecific antibody expressing cell by targeted integration of multiple expression cassettes in a defined organization
WO2020254356A1 (en) 2019-06-19 2020-12-24 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
WO2020260327A1 (en) 2019-06-26 2020-12-30 F. Hoffmann-La Roche Ag Mammalian cell lines with sirt-1 gene knockout
WO2021079195A1 (en) 2019-10-21 2021-04-29 Novartis Ag Tim-3 inhibitors and uses thereof
WO2021079188A1 (en) 2019-10-21 2021-04-29 Novartis Ag Combination therapies with venetoclax and tim-3 inhibitors
WO2021108661A2 (en) 2019-11-26 2021-06-03 Novartis Ag Chimeric antigen receptors and uses thereof
WO2021123902A1 (en) 2019-12-20 2021-06-24 Novartis Ag Combination of anti tim-3 antibody mbg453 and anti tgf-beta antibody nis793, with or without decitabine or the anti pd-1 antibody spartalizumab, for treating myelofibrosis and myelodysplastic syndrome
WO2021123996A1 (en) 2019-12-20 2021-06-24 Novartis Ag Uses of anti-tgf-beta antibodies and checkpoint inhibitors for the treatment of proliferative diseases
WO2021146636A1 (en) 2020-01-17 2021-07-22 Becton, Dickinson And Company Methods and compositions for single cell secretomics
WO2021144657A1 (en) 2020-01-17 2021-07-22 Novartis Ag Combination comprising a tim-3 inhibitor and a hypomethylating agent for use in treating myelodysplastic syndrome or chronic myelomonocytic leukemia
WO2021173995A2 (en) 2020-02-27 2021-09-02 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
WO2021194860A1 (en) 2020-03-23 2021-09-30 Genentech, Inc. Tocilizumab and remdesivir combination therapy for covid-19 pneumonia
WO2021194861A1 (en) 2020-03-23 2021-09-30 Genentech, Inc. Biomarkers for predicting response to il-6 antagonist in covid-19 pneumonia
WO2021194865A1 (en) 2020-03-23 2021-09-30 Genentech, Inc. Method for treating pneumonia, including covid-19 pneumonia, with an il6 antagonist
WO2021204743A1 (en) 2020-04-08 2021-10-14 F. Hoffmann-La Roche Ag Large molecule unspecific clearance assay
WO2021260528A1 (en) 2020-06-23 2021-12-30 Novartis Ag Dosing regimen comprising 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives
WO2022013787A1 (en) 2020-07-16 2022-01-20 Novartis Ag Anti-betacellulin antibodies, fragments thereof, and multi-specific binding molecules
WO2022026592A2 (en) 2020-07-28 2022-02-03 Celltas Bio, Inc. Antibody molecules to coronavirus and uses thereof
WO2022029573A1 (en) 2020-08-03 2022-02-10 Novartis Ag Heteroaryl substituted 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
WO2022043557A1 (en) 2020-08-31 2022-03-03 Advanced Accelerator Applications International Sa Method of treating psma-expressing cancers
WO2022043558A1 (en) 2020-08-31 2022-03-03 Advanced Accelerator Applications International Sa Method of treating psma-expressing cancers
WO2022060412A1 (en) 2020-09-17 2022-03-24 Genentech, Inc. Results of empacta: a randomized, double-blind, placebo-controlled, multicenter study to evaluate the efficacy and safety of tocilizumab in hospitalized patients with covid-19 pneumonia
WO2022063877A1 (en) 2020-09-24 2022-03-31 F. Hoffmann-La Roche Ag Mammalian cell lines with gene knockout
WO2022097065A2 (en) 2020-11-06 2022-05-12 Novartis Ag ANTIBODY Fc VARIANTS
WO2022104061A1 (en) 2020-11-13 2022-05-19 Novartis Ag Combination therapies with chimeric antigen receptor (car)-expressing cells
WO2022136140A1 (en) 2020-12-22 2022-06-30 F. Hoffmann-La Roche Ag Oligonucleotides targeting xbp1
WO2022162569A1 (en) 2021-01-29 2022-08-04 Novartis Ag Dosage regimes for anti-cd73 and anti-entpd2 antibodies and uses thereof
WO2022215011A1 (en) 2021-04-07 2022-10-13 Novartis Ag USES OF ANTI-TGFβ ANTIBODIES AND OTHER THERAPEUTIC AGENTS FOR THE TREATMENT OF PROLIFERATIVE DISEASES
WO2022214565A1 (en) 2021-04-09 2022-10-13 F. Hoffmann-La Roche Ag Process for selecting cell clones expressing a heterologous polypeptide
WO2022243846A1 (en) 2021-05-18 2022-11-24 Novartis Ag Combination therapies
WO2022261417A1 (en) 2021-06-11 2022-12-15 Genentech, Inc. Method for treating chronic obstructive pulmonary disease with an st2 antagonist
WO2023044483A2 (en) 2021-09-20 2023-03-23 Voyager Therapeutics, Inc. Compositions and methods for the treatment of her2 positive cancer
WO2023092004A1 (en) 2021-11-17 2023-05-25 Voyager Therapeutics, Inc. Compositions and methods for the treatment of tau-related disorders
WO2023117325A1 (en) 2021-12-21 2023-06-29 F. Hoffmann-La Roche Ag Method for the determination of hydrolytic activity
WO2023150778A1 (en) 2022-02-07 2023-08-10 Visterra, Inc. Anti-idiotype antibody molecules and uses thereof
WO2023202967A1 (en) 2022-04-19 2023-10-26 F. Hoffmann-La Roche Ag Improved production cells
WO2023220695A2 (en) 2022-05-13 2023-11-16 Voyager Therapeutics, Inc. Compositions and methods for the treatment of her2 positive cancer
WO2023232961A1 (en) 2022-06-03 2023-12-07 F. Hoffmann-La Roche Ag Improved production cells
WO2024030976A2 (en) 2022-08-03 2024-02-08 Voyager Therapeutics, Inc. Compositions and methods for crossing the blood brain barrier
WO2024079069A1 (en) 2022-10-12 2024-04-18 F. Hoffmann-La Roche Ag Method for classifying cells

Also Published As

Publication number Publication date
HK1208705A1 (en) 2016-03-11
CN100390288C (zh) 2008-05-28
BR0110610A (pt) 2003-04-29
EP1272647B1 (en) 2014-11-12
AU2001247616B2 (en) 2007-06-14
DK2857516T3 (en) 2017-08-07
EP1272647A1 (en) 2003-01-08
IL151853A (en) 2009-09-01
US20060025576A1 (en) 2006-02-02
CN101289511A (zh) 2008-10-22
LT2857516T (lt) 2017-09-11
PT2857516T (pt) 2017-08-28
CA2403425C (en) 2013-08-27
CA2403425A1 (en) 2001-10-18
EP2857516A1 (en) 2015-04-08
IL151853A0 (en) 2003-04-10
US20140322221A1 (en) 2014-10-30
SI2857516T1 (sl) 2017-09-29
US20080299120A1 (en) 2008-12-04
CN1464908A (zh) 2003-12-31
MXPA02010011A (es) 2003-04-25
PL357939A1 (en) 2004-08-09
HUP0300369A2 (hu) 2003-06-28
US20120238728A1 (en) 2012-09-20
ZA200207589B (en) 2003-09-22
US9493579B2 (en) 2016-11-15
US20110110852A1 (en) 2011-05-12
JP2003531588A (ja) 2003-10-28
ES2637801T3 (es) 2017-10-17
AU4761601A (en) 2001-10-23
DK1272647T3 (en) 2014-12-15
WO2001077342A1 (en) 2001-10-18
EP2857516B1 (en) 2017-06-14
KR20020093029A (ko) 2002-12-12
NZ521540A (en) 2004-09-24
US8722859B2 (en) 2014-05-13
IL190741A0 (en) 2008-11-03
CY1119291T1 (el) 2018-02-14
ES2528794T3 (es) 2015-02-12

Similar Documents

Publication Publication Date Title
US9493579B2 (en) Multivalent antibodies and uses therefor
AU2001247616A1 (en) Multivalent antibodies and uses therefor
AU2002250352B2 (en) Combination therapy
AU765003B2 (en) Apo-2 ligand-anti-her-2 antibody synergism
JP2018058906A (ja) B細胞表面マーカーに結合するアンタゴニストを用いた自己免疫疾患の治療
JP2007504138A (ja) 眼疾患の治療法
JP2008507473A (ja) 移植片拒絶におけるcd20の検出
AU2007216733A1 (en) Multivalent antibodies and uses therefor
NZ533374A (en) Multivalent antibodies and uses therefor
EP1941905A1 (en) APO-2 Ligand-anti-her-2 antibody synergism

Legal Events

Date Code Title Description
AS Assignment

Owner name: GENENTECH, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:MILLER, KATHY L.;PRESTA, LEONARD G.;REEL/FRAME:011645/0698

Effective date: 20010405

STCB Information on status: application discontinuation

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