US20050239141A1 - Modified antibody variable domains - Google Patents

Modified antibody variable domains Download PDF

Info

Publication number
US20050239141A1
US20050239141A1 US11/133,775 US13377505A US2005239141A1 US 20050239141 A1 US20050239141 A1 US 20050239141A1 US 13377505 A US13377505 A US 13377505A US 2005239141 A1 US2005239141 A1 US 2005239141A1
Authority
US
United States
Prior art keywords
human
antibody
cells
amino acid
mouse
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/133,775
Inventor
Gary Studnicka
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.)
Individual
Original Assignee
Individual
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 Individual filed Critical Individual
Priority to US11/133,775 priority Critical patent/US20050239141A1/en
Publication of US20050239141A1 publication Critical patent/US20050239141A1/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
    • C07K16/461Igs containing Ig-regions, -domains or -residues form different species
    • C07K16/467Igs with modifications in the FR-residues only
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39516Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum from serum, plasma
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/12Keratolytics, e.g. wart or anti-corn preparations
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/04Drugs for disorders of the muscular or neuromuscular system for myasthenia gravis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/14Drugs for disorders of the endocrine system of the thyroid hormones, e.g. T3, T4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure
    • 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
    • C07K16/2866Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for cytokines, lymphokines, interferons
    • 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
    • C07K16/46Hybrid immunoglobulins
    • C07K16/461Igs containing Ig-regions, -domains or -residues form different species
    • C07K16/464Igs containing CDR-residues from one specie grafted between FR-residues from another
    • C07K16/465Igs containing CDR-residues from one specie grafted between FR-residues from another with additional modified FR-residues
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6854Immunoglobulins
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16BBIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
    • G16B15/00ICT specially adapted for analysing two-dimensional or three-dimensional molecular structures, e.g. structural or functional relations or structure alignment
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16BBIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
    • G16B15/00ICT specially adapted for analysing two-dimensional or three-dimensional molecular structures, e.g. structural or functional relations or structure alignment
    • G16B15/20Protein or domain folding
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16BBIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
    • G16B20/00ICT specially adapted for functional genomics or proteomics, e.g. genotype-phenotype associations
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16BBIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
    • G16B20/00ICT specially adapted for functional genomics or proteomics, e.g. genotype-phenotype associations
    • G16B20/30Detection of binding sites or motifs
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16BBIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
    • G16B30/00ICT specially adapted for sequence analysis involving nucleotides or amino acids
    • G16B30/10Sequence alignment; Homology search
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/74Inducing cell proliferation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16BBIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
    • G16B30/00ICT specially adapted for sequence analysis involving nucleotides or amino acids

Definitions

  • the present invention relates, in general, to methods for preparing a modified antibody variable domain by determining the amino acid residues of the antibody variable domain which may be modified without diminishing the native affinity of the domain for antigen while reducing its immunogenicity with respect to a heterologous species; to methods of preparation of and use of antibody variable domains having modifications at the identified residues which are useful for administration to heterologous species; and to the variable regions so modified. More particularly, the invention relates to the preparation of modified mouse antibody variable domains, which are modified for administration to humans, the resulting antibody variable domains themselves, and the use of such “humanized” antibodies in the treatment of diseases in humans.
  • mouse monoclonal antibodies in human therapy is problematic for three reasons. First, an immune response against the mouse antibodies is mounted in the human body. Second, the antibodies have a reduced half-life in the human circulatory system. Third, the mouse antibody effector domains may not efficiently trigger the human immune system.
  • CDRs murine hypervariable complementarity determining regions
  • This technique is known as “CDR grafting”. See, e.g., Jones et al., Nature, 321, 522-525 (1986); Junghans et al., supra.
  • Protein structure analysis may be used to “add back” murine residues, again by genetic engineering, to first generation variable regions generated by CDR grafting in order to restore lost antigen binding capability.
  • Queen et al., Proc. Natl. Acad. Sci. USA, 86, 10029-10033 (1989); Co, et al., Proc. Natl. Acad. Sci. USA, 88, 2869-2873 (1991) describe versions of this method.
  • the foregoing three methods are techniques to “humanize” mouse monoclonal antibodies.
  • CD System Clusters of Differentation System
  • the CD System represents standard nomenclature for molecular markers of leukocyte cell differentation molecules. See Leukocyte Typing III White Cell Differentiation Antigens (Michael, ed. Oxford Press 1987), which is incorporated herein by reference.
  • pan T cell markers are those markers which occur on T Cells generally and are not specific to any particular T cell subset(s).
  • Pan T Cell markers include CD2, CD3, CD5, CD6, and CD7.
  • the CD5 cluster antigen for example, is one of the pan T markers present on about 85-100% of the human mature T lymphocytes and a majority of human thymocytes. CD5 is also present on a subset, about 20%, of B cells.
  • CD5 marker is found in McMichael and Gotch, in Leukocyte Typing III White Cell Differentiation Antigens (Michael, ed. Oxford Press 1987).
  • the CD5 molecule has also been described in the literature as reactive with immunoglobulins. See, e.g., Kernan et al., J. Immunol., 33:137-146 (1984), which is incorporated herein by reference.
  • the present invention provides methods for preparing a modified antibody variable domain useful for administration to humans by determining the amino acids of a subject antibody variable domain which may be modified without diminishing the native affinity of the domain for antigen while reducing its immunogenicity with respect to a heterologous species.
  • subject antibody variable domain refers to the antibody upon which determinations are made.
  • the method includes the following steps: determining the amino acid sequence of a subject light chain and a subject heavy chain of a subject antibody variable domain to be modified; aligning by homology the subject light and heavy chains with a plurality of human light and heavy chain amino acid sequences; identifying the amino acids in the subject light and heavy chain sequences which are least likely to diminish the native affinity of the subject variable domain for antigen while, at the same time, reducing its immunogenicity by selecting each amino acid which is not in an interface region of the subject antibody variable domain and which is not in a complementarity-determining region or in an antigen-binding region of the subject antibody variable domain, but which amino acid is in a position exposed to a solvent containing the antibody; changing each residue identified above which aligns with a highly or a moderately conserved residue in the plurality of human light and heavy chain amino acid sequences if said identified amino acid is different from the amino acid in the plurality.
  • FIGS. 1A and 1B Another group of sequences, such as those in FIGS. 1A and 1B may be used to determine an alignment from which the skilled artisan may determine appropriate changes to make.
  • the present invention provides a further method wherein the plurality of human light and heavy chain amino acid seqeunces is selected from the human consensus sequences in FIGS. 5A and 5B .
  • human engineering according to the above methods may be used to treat various diseases against which monoclonal antibodies generally may be effective.
  • humanized antibodies possess the additional advantage of reducing the immunogenic response in the treated patient.
  • the present invention also discloses products and pharmaceutical compositions useful in the treatment of a myriad human diseases.
  • products prepared by the foregoing methods include a modified H65 mouse monoclonal variable domain. Additionally, DNA sequences encoding the modified H65 variable domain are provided.
  • Modified antibody variable domains which are products of the methods of the present invention may be used, inter alia, as components of various immunoglobulin molecules such as Fab, Fab′, and F(ab′) 2 domains, single chain antibodies, and Fv or single variable domains.
  • Immunoglobulin molecules comprising modified variable domains according to the invention are particularly suited for therapeutic administration to human by themselves or, for example, as components of immunoconjugates such as those described in co-pending, co-owned U.S. patent application Ser. No. 07/787,567 filed on Nov. 4, 1991.
  • the present invention also provides methods for treatment of autoimmune diseases, wherein animal models are predictive of the efficacy of treatment in humans.
  • the present invention includes pharmaceutical compositions containing the humanized antibodies according to the invention are disclosed.
  • FIGS. 1A and 1B are alignments of the amino acid sequences of the light and heavy chains, respectively, of four antibody variable domains [HYH (HYHEL-10 Fab-lysosyme complex), MCPC (IgA Fab MCPC603-phosphocholine complex), NEWM (Ig Fab′ NEW) and KOL (IgG1 KOL)] by criteria of sequence and structural homology;
  • HYH HYHEL-10 Fab-lysosyme complex
  • MCPC IgA Fab MCPC603-phosphocholine complex
  • NEWM Ig Fab′ NEW
  • KOL IgG1 KOL
  • FIG. 2 is a schematic depiction of the structural relationships between the amino acid residues of the light chain of the variable domain
  • FIG. 3 is a schematic depiction of the structural relationships between the amino acid residues of the heavy chain of the variable domain
  • FIG. 4 is a schematic representation of an antibody variable domain
  • FIGS. 5A and 5B are alignments of the consensus amino acid sequences for the subgroups of light [hK1 (human kappa light chain subgroup 1), hK3 (human kappa light chain subgroup 3), hK2 (human kappa light chain subgroup 2), hL1 (human lambda light chain subgroup 1), hL2 (human lambda light chain subgroup 2), HL3 (human lambda light chain subgroup 3), hL6 (human lambda light chain subgroup 6), hK4 (human kappa light chain subgroup 4), hL4 (human lambda light chain subgroup 4) and hL5 (human lambda light chain subgroup 5] and heavy chains [hH3 (human heavy chain subgroup 3), hH1 (human heavy chain subgroup 1) and hH2 (human heavy chain subgroup 2)], respectively, of human antibody variable domains;
  • FIGS. 6A and 6B are alignments of human light chain consensus sequence hK1 with the actual (h65) and low-risk modified (prop) light chain sequences of the H65 mouse monoclonal antibody variable domain and of human heavy chain consensus sequence hH3 with the actual (h65) and modified (prop) heavy chain sequences of the H65 mouse monoclonal antibody variable domain, respectively;
  • FIGS. 7A and 7B are listings of the nucleotide sequences of the oligonucleotides utilized in the construction of the genes encoding modified V/J-regions of the light and heavy chains of the H65 mouse monoclonal antibody variable domain;
  • FIGS. 8A and 8B are listings of the nucleotide sequences of the genes encoding modified V/J-regions of the heavy and light chains, respectively, of the H65 mouse monoclonal antibody variable domain;
  • FIG. 9 is a graph of the results of a competitive binding assay showing that the H65 antibody variable domain modified by a method according to the present invention retains the antigen-binding capability of the natural H65 antibody variable region;
  • FIGS. 10A and 10B are alignments of human light chain consensus hK1 and heavy chain consensus hH1 with the light and heavy chain sequences, respectively, of the variable domain of human antibody EU, human antibody TAC, murine antibody TAC modified according to the present invention (prop) and murine antibody TAC modified according to a different method (Que);
  • FIG. 11 is a graph of He3 IgG binding to CD5 found on Molt-4M, demonstrating that such binding is similar to that of cH65 IgG;
  • FIG. 12 is a graph showing the effects of anti-Lyt-1 administration on the severity of collagen-induced arthritis in DBA/1J mice;
  • FIGS. 13A and 13B are depictions of human T cell recovery in spleen and blood, respectively from PBMC/SCID mice following treatment with H65 MoAb;
  • FIGS. 14A and 14B are schematic depictions of human T cell recovery in spleen and blood, respectively from PBMC/SCID mice following treatment with H65-based F(ab′) 2 fragment;
  • FIG. 15 is a graph of the effects of OX19 MoAb on the severity of DR BB rat collagen-induced arthritis.
  • FIGS. 16A and 16B are alignments of human light chain consensus sequence hK1 with the actual (h65) and low and moderate risk modified (prop) light chain sequences of the H65 mouse monoclonal antibody variable domain and of human heavy chain consensus sequence hH3 with the actual (h65) and modified (prop) heavy chain sequences of the H65 mouse monoclonal antibody variable domain, respectively.
  • Methods according to the present invention include: (1) identification of the amino acid residues of an antibody variable domain which may be modified without diminishing the native affinity of the domain for antigen while reducing its immunogenicity with respect to a heterologous species; (2) the preparation of antibody variable domains having modifications at the identified residues which are useful for administration to heterologous species; and (3) use of the humanized antibodies of the invention in the treatment of autoimmune diseases in humans.
  • the methods of the invention are based on a model of the antibody variable domain described herein which predicts the involvement of each amino acid in the structure of the domain.
  • the methods described herein introduce human residues into the variable domain of an antibody only in positions which are not critical for antigen-binding activity and which are likely to be exposed to immunogenicity-stimulating factors.
  • the present methods are designed to retain sufficient natural internal structure of the variable domain so that the antigen-binding capacity of the modified domain is not diminished in comparison to the natural domain.
  • FIGS. 1A and 1B provide the sequences of the four antibody variable domains which have been crystallized.
  • the amino acid sequences of the light and heavy chains of HYH (SEQ ID Nos. 1 and 5, respectively), MCPC (SEQ ID Nos. 2 and 6, respectively), NEWM (SEQ ID Nos. 3 and 7, respectively) and KOL (SEQ ID Nos. 4 and 8, respectively) are shown, wherein the exclamation points “!” in the MCPC light chain sequence at position 30 ⁇ , the MCPC heavy chain sequence at positions 52 ⁇ and 98 ⁇ , the NEWM light chain at position 30 ⁇ , the KOL light chain at position 93 ⁇ , and the KOL heavy chain sequence at position 98 ⁇ , stand for the amino acid sequences NSGNQK (SEQ ID No.
  • FIGS. 2 and 3 comprise depictions of the structure of the light and heavy chains, respectively, wherein each chain is displayed “unfolded” into a flattened beta sheet structure so that interactions among the residues are easier to visualize.
  • the strands of folded polypeptide chains are represented as thick vertical lines, connected by eight beta-turn loops. Three of the loops are identified as antigen-binding loops or CDRs, one is accessory to the loops, and the remaining four at the “bottom” of the variable domain are not involved in antigen binding.
  • the amino and carboxy termini of the variable domain are symbolized by small black dots at the ends of the polypeptide chains.
  • Each amino acid position is represented as either a circle, a triangle, or a square.
  • the covalent disulfide bond between the two cysteines at positions 23 and 88 in the light chain and the covalent disulfide bond between positions 22 and 92 in the heavy chain are each shown as a thick horizontal line. All of the residues in each chain are shown on the map, including antigen-binding residues and framework residues.
  • the amino acid positions are numbered according to Kabat et al., Sequences of Proteins of Immunological Interest , Fourth Edition, U.S.
  • Solid slanted lines (either single or double) connecting pairs of residues which are adjacent in three-dimensional space but not in linear sequence, represent one or two hydrogen bonds between the mutually aligned amino nitrogens and carbonyl oxygens in the backbones of the residues.
  • variable domain The basic structure of the antibody variable domain is strongly conserved.
  • the variable domain is composed of a light chain (or subunit) and a heavy chain (or subunit), which are structurally homologous to each other and which are related by a pseudo-two-fold axis of rotational symmetry.
  • the variable domain is functionally distinct from the constant domain, being connected only by two highly flexible chains and pivoting on both “ball-and-socket” joints formed by five amino acids in the heavy and light chains.
  • Each amino acid chain folds back on itself repeatedly to create nine distinct strands.
  • the various strands in each layer are extensively hydrogen-bonded to each other.
  • the two beta-sheet layers within the subunit are held together by a single covalent disulfide bond and by numerous internal hydrophobic interactions.
  • the sequences involved in bonding the strands of the subunits together are called “framework” sequences.
  • Amino acid sidechains exist in various different chemical environments within the subunits. Some residues are exposed to the solvent on the outer accessible surface while other residues are buried in hydrophobic interactions within a subunit.
  • Much of the immunoglobulin variable domain is constructed from antiparallel beta pleated sheets which create amphipathic surfaces, such that the “inside” surface is hydrophobic and the “outside” surface is hydrophilic. The outside is exposed to solvent, and therefore is also exposed to the humoral environment when the domain is in the circulatory system of an animal. Amino acid sidechains which are completely exposed to the solvent and which do not physically interact with other residues in the variable domain are likely to be immunogenic and are unlikely to have any structural importance within the immunoglobulin molecule.
  • a highly schematic representation of the variable domain is shown in FIG. 4 , wherein thick lines represent peptide bonds and shaded circles denote amino acid side chains.
  • the two subunits of antibody variable domains adhere to each other via a hydrophobic interface region which extends along the inside beta-sheet layer from the border of the variable domain with the constant domain to the antigen-binding loops.
  • Amino acid side chains from both subunits interact to form a three-layered “herringbone” structure.
  • Some of these interfacial residues are components of the antigen-binding loops, and thus have a direct effect upon binding affinity. Every residue in the interface is structurally important because the conformation of the binding regions is strongly influenced by changes in the conformation of the interface.
  • the foregoing data and information on the structure of antibody variable domains aids in a determination of whether a particular amino acid of any variable domain is likely to influence antigen binding or immunogenicity.
  • the determination for each amino acid position is represented by a pair of symbols (e.g., + and +, in the lines labelled “bind” and “bury”, respectively) in FIGS. 1A, 1B , (and also in FIGS. 5A, 5B , 6 A, 6 B, 10 A and 10 B).
  • the first symbol relates to antigen binding
  • the second symbol relates to immunogenicity and framework structure. Tables 1, 2 and 3, below, set out the significance of the symbols and possible pairings.
  • FIGS. 5A and 5B thus set out and align the consensus sequences (derived from Kabat et al., supra) of the subgroups of light (hK1, SEQ ID NO: 13; hK3, SEQ ID NO: 14; hK2, SEQ ID NO: 15; hL1 SEQ ID NO: 16; hL2, SEQ ID NO: 17; hL3, SEQ ID NO: 18; hL6, SEQ ID NO: 19; hK4, SEQ ID NO: 20; hL4, SEQ ID NO: 21; and hL5, SEQ ID NO: 22) and heavy chains (hH3, SEQ ID NO: 23; hH1, SEQ ID NO: 24; and hH2, SEQ ID NO: 25) of antibody variable domains with the pairings representing the structural characteristics of each amino acid position, wherein the consensus sequences for the hL
  • upper case amino acid designations indicate that the amino acid is present at that location in about 90% to about 100% of the known human sequences (excluding small incomplete fragments) of that subgroup (i.e., is “highly conserved”); whereas lower case amino acid designations indicate that the amino acid is present at that location in about 50% to about 90% of the known human sequences in that subgroup (i.e., is “moderately conserved”).
  • a lower case “x” denotes conservation in less than about 50% of the known sequences in that subgroup (i.e., a “poorly conserved” position).
  • FIGS. 5A and 5B The information presented in FIGS. 5A and 5B on the relationship of a particular amino acid in a sequence of an antibody variable domain to the structure and antigen-binding capacity of the domain is sufficient to determine whether an amino acid is modifiable. Additional structural studies, such as those on which FIGS. 5A and 5B are based, are not required.
  • FIGS. 5A and 5B may be used to prepare, for example, a modified mouse antibody variable domain that retains the affinity of the natural domain for antigen while exhibiting reduced immunogenicity in humans by the following steps.
  • the amino acid sequences of both the light chain and the heavy chain from the mouse variable domain are first determined by techniques known in the art (e.g., by Edman degradation or by sequencing of a cDNA encoding the variable domain).
  • the consensus sequences set out in FIGS. 5A and 5B for human antibody variable regions are examined to identify both a light chain consensus and a heavy chain consensus sequence that are the most homologous to the particular mouse subunit sequences that are to be modified.
  • the mouse sequences are aligned to the consensus human sequences based on homology either by sight or by using a commercially available computer program such as the PCGENE package (Intelligenetics, Mountain View, Calif.).
  • FIGS. 5A and 5B are then used again to identify all of the “low risk” or “moderate risk” positions at which the mouse sequence differs significantly from the chosen human consensus.
  • the mouse amino acid residues at these low risk and moderate risk positions are candidates for modification. If the human consensus is strongly conserved at a given low risk or moderate risk position, the human residue may be substituted for the corresponding mouse residue. If the human consensus is poorly conserved at a given low risk or moderate risk position, the mouse residue is retained at that position.
  • the mouse residue is normally replaced with a human residue, unless the mouse residue occurs at that position in at least one of the sequences (e.g., in Kabat et al., supra) on which the human consensus sequence is based. If the mouse residue does occur at that position in a human sequence then the mouse residue may be retained.
  • the mouse sequence does not contain a proline at a location where the human consensus sequence has one, it is likely that substituting a proline in the mouse sequence would affect proper conformation of the sequence, therefore the mouse residue is preferably retained. Where a proline at a particular position involving proline is changed from mouse to human, such a change is considered to be at least moderate risk even if that position would otherwise be low risk.
  • insertions and deletions in a mouse sequence are normally preserved intact. If the mouse sequence has an alteration in the length and spacing of the variable region backbone, it is likely that the alteration is necessary to provide a surface for proper folding of the antigen-binding loops. The alteration is preferably retained in a modified version of the sequence.
  • the side chains in the interface region are buried deep within the structure, so they are unlikely to elicit a therapeutic immunogenic response in a heterologous species.
  • DNAs encoding the complete variable domain are synthesized [via oligonucleotide synthesis as described, for example, in Sinha et al., Nucleic Acids Res., 12, 4539-4557 (1984)], assembled [via PCR as described, for example in Innis, Ed., PCR Protocols, Academic Press (1990) and also in Better et al. J. Biol. Chem. 267, 16712-16118 (1992)], cloned and expressed [via standard procedures as described, for example, in Ausubel et al., Eds., Current Protocols in Molecular Biology , John Wiley & Sons, New York (1989) and also in Robinson et al., Hum. Antibod.
  • Hybridomas 2, 84-93 (1991)]
  • specific antigen binding activity via competition assay as described, for example, in Harlow et al., Eds., Antibodies: A Laboratory Manual, Chapter 14, Cold Spring Harbor Laboratory, Cold Spring Harbor (1988) and Munson et al., Anal. Biochem., 107, 220-239 (1980)].
  • An immunoglobulin such as an anti-T-cell immunoglobulin may be conjugated to a cytotoxic molecule.
  • the cytotoxic molecule to which the immunoglobulin is conjugated may be any of a number of toxins such as lectin A or a ricin A chain.
  • the above-referenced '297 application also describes use of an anti-CD5 antibody conjugated to a ricin A chain providing an anti-T-cell immunotoxin.
  • autoimmune diseases may be characterized, inter alia, by abnormal immunological regulation which results in excessive B Cell activity and diminished, enhanced, or inappropriate T Cell activity. Such altered T cell activity may result in excessive production of autoantibodies.
  • the autoimmune diseases are complex and diverse in their manifestations, they possess the common feature of an impaired immune system.
  • Therapeutic depletion of circulating T cells through the administration of an anti-pan T cell immunoglobulin improves the clinical course and prognosis of patients with autoimmune disease.
  • the additional depletion of CD5 B cells may have a further beneficial effect since CD5 B cells have been implicated in some autoimmune diseases.
  • an anti-CD5 monoclonal antibody is presented as an example of a preferred embodiment of the invention.
  • An example of an anti-pan T cell immunoglobulin is an CD5 antibody which is primarily reactive with a surface antigen of mature T cells, but is also reactive with 10-20% of mature B cells.
  • Clinical data obtained using the anti-pan T cell immunoglobulin in models of autoimmune diseases in non-human animals are predictive of the effects of using such immunoglobulins as therapy against human autoimmune diseases.
  • an immunoglobulin such as an antibody
  • an immunoglobulin is “reactive” with or “binds to” an antigen if it interacts with the antigen forms an antigen-immunoglobulin complex.
  • the antigen is generally a unique surface protein or marker.
  • a most preferred marker is the CD5 antigen cluster.
  • the anti-pan T cell immunoglobulin may be obtained from a number of sources. It is reactive with most mature T cells or with both T cells and subsets of other lymphoid cells, such as B cells or natural killer (NK) cells.
  • the immunoglobulin may be synthetic or recombinant, including genetically-engineered immunoglobulins such as chimeric immunoglobulins, humanized antibodies, hybrid antibodies, or derivatives of any of these.
  • Chimeric immunoglobulins, antibodies or peptides are comprised of fused portions from different species as a product of chimeric DNA.
  • Chimeric DNA is recombinant DNA containing genetic material from more than one mammalian species.
  • Chimeric immunoglobulins include one portion having an amino acid sequence derived from, or homologous to, a corresponding sequence in an immunoglobulin, antibody or peptide derived from a first gene source while the remaining segment of the chain(s) is homologous to corresponding sequences of another gene source.
  • a chimeric antibody peptide may comprise an antibody heavy chain with a murine variable region and a human constant region. The two gene sources will typically involve two species, but will occasionally involve different sources from one species.
  • Chimeric immunoglobulins, antibodies or peptides are typically produced using recombinant molecular and/or cellular techniques.
  • chimeric antibodies have variable regions of both light and heavy chains that mimic the variable regions of antibodies derived from one mammalian species, while the constant portions are homologous to the sequences in antibodies derived from a second, different mammalian species.
  • chimeric antibody is any antibody in which either or both of the heavy or light chains are composed of combinations of sequences mimicking the sequences in antibodies of different sources regardless of whether these sources are differing classes, differing antigen responses, or differing species of origin, and whether or not the fusion point is at the variable/constant boundary.
  • humanized refers to an immunoglobulin wherein the constant regions have at least about 80% or greater homology to human immunoglobulin, and wherein some of the nonhuman (i.e. murine) variable region amino acid residues may be modified to contain amino acid residues of human origin.
  • Humanized antibodies may be referred to as “reshaped” antibodies.
  • Manipulation of the complementarity-determining regions (CDRs) is one means of manufacturing humanized antibodies. See, e.g., Jones, et al. Replacing the Complementarity—Determining Regions in a Human Antibody With Those From a Mouse, Nature 321:522-525 (1988); Riechmann, et al. Reshaping Human Antibodies For Therapy, Nature 332, 323-327 (1988).
  • CDRs complementarity-determining regions
  • immunoglobulins of the present invention are monoclonal antibodies (hereinafter referred to as “MoAbs”) of the IgM or IgG isotype of murine, human or other mammalian origin. Most preferably, the MoAb is reactive with the CD5 antigen found on both T and B cells. MoAbs of other animal species may be prepared using analogous non-human mammalian markers.
  • MoAbs A variety of methods for producing MoAbs are known in the art. See, e.g., Goding, Monoclonal Antibodies; Principles and practice (2d ed., Academic Press 1986), which is incorporated herein by reference. Less preferred forms of immunoglobulins may be produced by methods well-known to those skilled in the art, such as by chromatographic purification of polyclonal sera to produce substantially monospecific antibody populations.
  • Monoclonal antibodies specifically directed against human CD5 antigen may be obtained by using combinations of immunogens and screening antigens which have only there human CD5 antigen in common or bay a screening assay designed to be specific for only anti-CD5 monoclonals.
  • production of monoclonal antibodies directed against CD5 may be accomplished by 1) immunization with human T cells expressing the CD5 antigen followed by screening of the resultant hybridomas for reactivity against a non-human cell line transfected with human CD5 (constructed in a manner similar to that described in Nishimura, et al., Eur. J.
  • a preferred monoclonal antibody for use in this invention is produced by hybridoma cell line XMMLY-H65 (H65) deposited with the American Type Culture Collection in Rockville, Md. (A.T.C.C.) and given the Accession No. HB9286.
  • a preferred antibody is prepared as disclosed herein using the humanized forms of the murine H65 antibody.
  • DNA sequences which encode a human MoAb or portions thereof which specifically bind to the human T cell may be isolated by screening a DNA library from human B cells according to the general protocols outlined by Huse et al., Science 246:1275-1281 (1989), Marks, et al., J. Mol. Biol. 222:581-597 (1991) which are incorporated herein by reference, and then cloning and amplifying the sequences which encode the antibody (or binding fragment) of the desired specificity.
  • immunoglobulins specifically described herein other “substantially homologous” modified immunoglobulins can be readily designed and manufactured utilizing various recombinant DNA techniques known to those skilled in the art. Modifications of the immunoglobulin genes may be readily accomplished by a variety of well-known techniques, such as site-directed mutagenesis. See, Gillman and Smith, Gene 8:81-97 (1979); Roberts, et al., Nature 328:731-734 (1987), both of which are incorporated herein by reference. Also, modifications which affect the binding affinity of the antibody may be selected using the general protocol outlined by McCafferty, et al., Nature 348:552-554 (1990), which is incorporated herein by reference.
  • an immunoglobulin, antibody, or peptide is specific for a T cell if it binds or is capable of binding T cells as determined by standard antibody-antigen or ligand-receptor assays.
  • assays include competitive assays, immunocytochemistry assays, saturation assays, or standard immunoassays such as ELISA, RIA and flow cytometric assays.
  • This definition of specificity also applies to single heavy and/or light chains, CDRS, fusion proteins, or fragments of heavy and/or light chains, which bind T cells alone or are capable of binding T cells if properly incorporated into immunoglobulin conformation with complementary variable regions and constant regions as appropriate.
  • the ability of an immunoglobulin, antibody, or peptide fragment to bind an antigen is determined by detecting the ability of the immunoglobulin, antibody, or peptide to compete with the binding of a compound known to bind the antigen.
  • Numerous types of competitive assays are known and are discussed herein.
  • assays which measure binding of a test compound in the absence of an inhibitor may also be used.
  • the ability of a molecule or other compound to bind T cells can be detected by labelling the molecule of interest directly, or it may be unlabelled and detected indirectly using various sandwich assay formats.
  • Numerous types of binding assays such as competitive binding assays are known. See, e.g., U.S. Pat. Nos. 3,376,110, 4,016,043; Harlow and Lane, Antibodies: A Laboratory Manual , Cold Spring Harbor Publications, N.Y. (1988), which are incorporated herein by reference.
  • Assays for measuring binding of a test compound to one component alone rather than using a competition assay are also available.
  • immunoglobulins may be used to identify the presence of a T cell marker.
  • Standard procedures for monoclonal antibody assays, such as ELISA, may be used see, Harlow and Lane, supra).
  • signal producing systems See U.S. Pat. No. 4,391,904, which is incorporated herein by reference.
  • Humanized antibodies of the present invention may be administered to patients having a disease having targetable cellular markers.
  • diseases include, but are not limited to, autoimmune diseases such as lupus (including systemic lupus erythematosus and lupus nephritis), scleroderma diseases (including lichen sclerosis, morphea and lichen planus), rheumatoid arthritis and the spondylarthropathies, thyroiditis, pemphigus vulgaris, diabetes mellitus type 1, progressive systemic sclerosis, aplastic anemia, myasthenia gravis, myositis including polymyositis and dermatomyositis, Sjogren's disease, collagen vascular disease, polyarteritis, inflammatory bowel disease (including Crohn's disease and ulcerative colitis), multiple sclerosis, psoriasis and primary biliary cirrhosis; diseases caused by viral infections; diseases caused by fungal infections; diseases caused
  • Immunoglobulins, antibodies or peptides according to the invention may be administered to a patient either singly or in a cocktail containing two or more antibodies, other therapeutic agents, compositions, or the like, including, but not limited to, immunosuppressive agents, potentiators and side-effect relieving agents.
  • immunosuppressive agents useful in suppressing allergic or other undesired reactions of a host.
  • Immunosuppressive agents include prednisone, prednisolone, dexamethasone, cyclophosphamide, cyclosporine, 6-mercaptopurine, methotrexate, azathioprine, and gamma globulin.
  • anti-pan T cell immunoglobulins may be formulated into various preparations such as injectable and topical forms.
  • Parenteral formulations are preferred for use in the invention, most preferred is intramuscular (i.m.) or intravenous (i.v.) administration.
  • the formulations containing therapeutically effective amounts of anti-pan T cell antibodies are either sterile liquid solutions, liquid suspensions or lyophilized versions and optionally contain stabilizers or excipients.
  • Lyophilized compositions are reconstituted with suitable diluents, e.g., water for injection, saline, 0.3% glycine and the like, at a level of from about 0.01 mg/kg of host body weight to about 10 mg/kg or more of host body weight.
  • the pharmaceutical compositions containing anti-pan T cell immunoglobulins are administered in a therapeutically effective dose in a range of from about 0.01 mg/kg to about 5 mg/kg body weight of the treated animal.
  • a preferred dose range of the anti-pan T cell antibody is from about 0.05 mg/kg to about 2 mg/kg body weight of the treated animal.
  • the immunoglobulin dose is administered over either a single day or several days by daily intravenous infusion. For example, for a patient weighing 70 kg, about 0.7 mg to about 700 mg per day is a preferred dose. A more preferred dose is from about 3.5 mg to about 140 mg per day.
  • Anti-pan T cell immunoglobulin may be administered systemically by injection intramuscularly, subcutaneously, intrathecally, intraperitoneally, into vascular spaces, or into joints (e.g., intraarticular injection at a dosage of greater than about 1 ⁇ g/cc joint fluid/day).
  • the dose will be dependent upon the properties of the anti-pan T cell immunoglobulin employed, e.g., its activity and biological half-life, the concentration of anti-pan T cell antibody in the formulation, the site and rate of dosage, the clinical tolerance of the patient involved, the autoimmune disease afflicting the patient and the like as is well within the knowledge of the skilled artisan.
  • the anti-pan T cell immunoglobulin of the present invention may be administered in solution.
  • the pH of the solution should be in the range of about pH 5.0 to about 9.5, preferably pH 6.5 to 7.5.
  • the anti-pan T cell immunoglobulin or derivatives thereof should be in a solution having a pharmaceutically acceptable buffer, such as phosphate, tris (hydroxymethyl) aminomethane-HCl, or citrate and the like. Buffer concentrations should be in the range from about 1 to about 100 mM.
  • a solution containing anti-pan T cell immunoglobulin may also contain a salt, such as sodium chloride or potassium chloride in a concentration from about 50 to about 150 mM.
  • a stabilizing agent such as albumin, a globulin, a detergent, a gelatin, a protamine, or a salt of protamine may also be included and may be added to a solution containing anti-pan T cell immunoglobulin or to the composition from which the solution is prepared systemic administration of anti-pan T cell immunoglobulin is typically made every two to three days or once a week if a chimeric or humanized form is used. Alternatively, daily administration is useful. Usually administration is by either intramuscular injection or intravascular infusion.
  • anti-pan T cell immunoglobulin is formulated into topical preparations for local therapy by including a therapeutically effective concentration of anti-pan T cell immunoglobulin in a dermatological vehicle.
  • Topical preparations may be useful to treat skin lesions such as psoriasis and dermatitis associated with lupus.
  • the amount of anti-pan T cell immunoglobulin to be administered, and the anti-pan T cell immunoglobulin concentration in the topical formulations will depend upon the vehicle selected, the clinical condition of the patient, the systemic toxicity and the stability of the anti-pan T cell immunoglobulin in the formulation.
  • the physician will necessarily employ the appropriate preparation containing the appropriate concentration of anti-pan T cell immunoglobulin in the formulation, as well as the amount of formulation administered depending upon clinical experience with the patient in question or with similar patients.
  • the concentration of anti-pan T cell immunoglobulin for topical formulations is in the range from about 0.1 mg/ml to about 25 mg/ml. Typically, the concentration of anti-pan T cell immunoglobulin for topical formulations is in the range from about 1 mg/ml to about 20 mg/ml.
  • Solid dispersions of anti-pan T cell immunoglobulin as well as solubilized preparations may be used. Thus, the precise concentration to be used in the vehicle may be subject to modest experimental manipulation in order to optimize the therapeutic response. Greater than about 10 mg of anti-pan T cell immunoglobulin/100 grams of vehicle may be useful with 1% w/w hydrogel vehicles in the treatment of skin inflammation. Suitable vehicles, in addition to gels, are oil-in-water or water-in-oil emulsions using mineral oils, petrolatum, and the like.
  • Anti-pan T cell immunoglobulin may be optionally administered topically by the use of a transdermal therapeutic system (Barry, Dermatological Formulations , p. 181 (1983)). While such topical delivery systems have been designed largely for transdermal administration of low molecular weight drugs, by definition they are capable of percutaneous delivery. They may be readily adapted to administration of anti-pan T cell immunoglobulin or derivatives thereof and associated therapeutic proteins by appropriate selection of the rate-controlling microporous membrane.
  • Preparations of anti-pan T cell immunoglobulin either for systemic or local delivery may be employed and may contain excipients as described above for parenteral administration and other excipients used in a topical preparation such as cosolvents, surfactants, oils, humectants, emollients, preservatives, stabilizers and antioxidants.
  • Any pharmacologically acceptable buffer may be used, e.g., tris or phosphate buffers.
  • Anti-pan T cell immunoglobulin may also be administered via microspheres, liposomes or other microparticulate delivery systems placed in certain tissues including blood.
  • Anti-pan T cell immunoglobulin may also be administered by aerosol to achieve localized delivery to the lungs. This is accomplished by preparing an aqueous aerosol or liposomal preparation. A nonaqueous (e.g., fluorocarbon propellent) suspension may be used. Sonic nebulizers preferably are used in preparing aerosols. Sonic nebulizers minimize exposing the anti-pan T cell antibody or derivatives thereof to shear, which can result in degradation of anti-pan T cell immunoglobulin.
  • an aqueous aerosol is made by formulating an aqueous solution or suspension of anti-pan T cell immunoglobulin together with conventional pharmaceutically acceptable carriers and stabilizers.
  • the carriers and stabilizers will vary depending upon the requirements for the particular anti-pan T cell immunoglobulin, but typically include nonionic surfactants (Tweens, Pluronics, or polyethylene glycol), innocuous proteins such as serum albumin, sorbitan esters, oleic acid, lecithin, amino acids such as glycine, buffers, salts, sugars, or sugar alcohols.
  • the formulations are sterile. Aerosols generally may be prepared from isotonic solutions.
  • a method of the present invention was utilized to prepare modified antibody variable domains by identifying low risk residues in a mouse monoclonal antibody variable domain, designated H65, which may be modified without diminishing the native affinity of the domain for antigen while still reducing its immunogenicity with respect to humans.
  • the light and heavy chains of the variable domain of H65 were determined to most closely resemble the consensus sequences of subgroup 1 (“hK1”) of the human kappa chains and subgroup 3 (“hH3”) of the human heavy chains, respectively.
  • the H65 V/J-segments of the light and heavy chain sequences are aligned with the two human subgroup consensus sequences in FIGS. 6A and 6B .
  • the H65 sequences are also contained in SEQ ID Nos. 26 and 28.
  • upper and lower case letters denote the degree of conservation at any given position.
  • an “A” indicates that alanine is present at that position in about 90% to about 100% of the known human sequences of that subgroup (excluding small, incomplete fragments); whereas an “a” indicates that alanine is present only about 50% to about 90% of the time at that position in known human sequences of that subgroup.
  • a lower case “x” indicates conservation of the amino acid at that position less than about 50% of the time.
  • the line labelled “bind” shows which residues directly affect ( ⁇ ) or do not directly affect (+) antigen binding of CDR loops.
  • a “0” indicates a residue of intermediate significance in terms of antigen binding or placement of the residue, respectively.
  • FIGS. 6A and 6B reveal that the mouse H65 sequences differ from the human consensus sequences with which they are aligned at a total of 94 positions. Sixty-nine of these differences occur at moderate-risk (15 positions) or high risk (54 positions) positions suggesting that the mouse residue at that position may be important for the function of the antibody.
  • the “M/H” line of FIG. 6 specifically indicates which positions differ between the two pairs of aligned sequences.
  • mice residue aligns with a human consensus amino acid which is highly conserved. Therefore, the mouse residue at that position is identified as one to be changed to the conserved human residue.
  • the mouse residue aligns with a human consensus amino acid which is moderately conserved. However, since the mouse residue is found at that position in other actual sequences of human antibodies (in Kabat's sequences of Proteins of Immunoglobulin Interest), the positions are identified as ones to be kept “mouse.” At seven low risk positions (designated “h”), the mouse residue aligns with a human consensus amino acid which is moderately conserved but the mouse residue is not found at that position in an actual human antibody sequence in the Kabat book. Therefore, those positions are identified as ones to be changed to “human.”
  • mouse residue aligns with a human consensus amino acid which is poorly conserved. Therefore, that position is identified as one to be kept “mouse.”
  • the “prop” lines of FIG. 6 set out the sequences of the light and heavy chains of the H65 antibody variable domain in which the residues identified by the methods of the present invention as those which may be modified without diminishing the native affinity of the H65 variable domain for CD5 are changed to human residues.
  • the “prop” lines of FIGS. 6A and 6B set out the amino acid sequences of humanized light (SEQ ID NO: 27) and heavy chains (SEQ ID NO: 29) of the H65 antibody variable domain.
  • polynucleotides encoding the native mouse leader sequences were included as part of the humanized genes.
  • Each gene, heavy or light, was assembled from six overlapping oligonucleotides and amplified by PCR. Each oligonucleotide was synthesized with a Cyclone Model 8400 DNA Synthesizer (Milligen/Biosearch, Burlington, Mass.). Restriction sites were introduced into the amplified DNA segments for cloning into the final expression vectors for antibody genes (heavy or light).
  • a SalI restriction site was introduced into each V-region upstream of the initiation codon, ATG.
  • a BstEII restriction site was introduced into the 3′-end of the heavy chain J-region, while a HindIII site was introduced into the 3′-end of the light chain J-region.
  • the humanized V- and J-segments of the heavy chain were assembled from six oligonucleotides, HUH-G1, HUH-G2, HUH-G3, HUH-G4, HUH-G5, and HUH-G6, the sequences of which are contained in FIG. 7B and in SEQ ID Nos. 36 to 41, respectively.
  • the oligonucleotides were amplified with PCR primers H65G-2S and H65-G2 (SEQ ID Nos. 42 and 43, respectively). Oligonucleotides greater than 50 bp in length were purified on a 15% polyacrylamide gel in the presence of 25% urea.
  • DNA strand extension and DNA amplification was accomplished with a Taq polymerase and the GeneAmp Kit used according to the manufacturer's instructions (Perkin-Elmer Cetus, Germany). Oligonucleotides containing the synthetic humanized antibody gene were mixed in pairs (HUH-G1+HUH-G2, HUH-G3+HUH-G4, and HUH-G5+HUH-G6) in 100 ⁇ l reactions with 1 ⁇ g of each DNA, 2.5 U Taq polymerase, 50 mM KCl, 10 mm TRIS-CL pa 8.3, 1.5 mM MgCl 2 , and 200 uM each dNTP.
  • the tube was incubated in a Coy TempCycler for 1 minute at 94° C., 2 minutes at 55° C. and 20 minutes at 72° C.
  • a portion of each reaction product (40 ⁇ l) was mixed in pairs (HUH-G1,2+HUH-G3,4; HUH-G3,4+HUH-G5,6), 2.5 U Taq was added and the tubes were re-incubated at 94° C. for 1 minute, 55° C. for 2 minutes and 72° C. for 20 minutes.
  • the heavy chain gene was then assembled by mixing an equal amount of the HUH-G1,2,3,4 reaction product with the HUH-G3,4,5,6 reaction product and bringing the volume to 100 ⁇ l of 2.5 U Taq, 50 mM KCl, 10 mM TRIS-CL pH 8.3, 1.5 mM MgCl 2 , 200 uM each dNTP, and 0.5 ⁇ g of each amplification primer H65G-2S and H65-G2.
  • the reaction was overlaid with mineral oil, and the cycle profile used for amplification was: denaturation 94° C. for 1 minute, annealing 55° C. for 2 minutes, and primer extension at 72° C. for 3 minutes. Primer extension was carried out for 30 cycles.
  • the DNA sequence of the assembled V/J-region is contained in FIG. 8A and in SEQ ID NO: 46.
  • the assembled V/J-region was cut with SalI and BstEII, purified by electrophoresis on an agarose gel, and assembled into a heavy chain expression vector, pING4612, which is similar to that described for heavy chain expression in Robinson et al., Hum. Antib. Hybridomas, 2, 84 (1991) and described in detail in co-pending, co-owned U.S. patent application Ser. No. 07/659,409 filed on Sep. 6, 1989, which is incorporated herein by reference.
  • the humanized V- and J-segments of the light chain were also assembled from six oligonucleotides, $H65K-1, HUH-K1, HUH-K2, HUH-K3, HUH-K4 and HUH-K5, the sequences of which are contained in FIG. 7 and in SEQ ID NOs. 30 to 35, respectively.
  • the oligonucleotides were amplified with PCR primers H65K-2S and JK1-HindIII (SEQ ID NOs. 44 and 45, respectively).
  • Oligonucleotides containing the synthetic humanized antibody gene were mixed in pairs ($H65K-K1+HUH-K1, HUH-K2+HUH-K3, and HUH-K4+HUH-K5) and incubated as described above for the heavy chain. A portion of each reaction product (40 ⁇ l) was mixed in pairs ($H65K-1/HUH-K1+HUH-K2,3; HUH-K2,3+HUH-K4,5) and treated as above.
  • the light chain gene was then assembled by amplifying the full length gene with PCR primers H65K-2S and JK1-HindIII as outlined above for the heavy chain.
  • the DNA sequence of the assembled V/J-region is contained in FIG. 8B and in SEQ ID NO. 47.
  • the assembled V/J-region was cut with SalI and HindIII, purified by electrophoresis on an agarose gel, and assembled into a light chain antibody expression vector, pING4614 similar to those described for light chain expression in Robinson et al., supra. and in U.S. patent application Ser. No. 07/659,409, supra.
  • Expression vectors containing the humanized H65 light chain and heavy chain sequences under the control of the Abelson Leukemia virus LTR promoter (described in Robinson et al., supra, and in U.S. patent application Ser. No. 07/659,409, supra) and 3′ untranslated regions from human gamma-1 (for heavy chain) and mouse kappa (for light chain) were transfected by lipofection into a CHO-K1 strain which expresses the SV40 T antigen.
  • the CHO-K1 cells were also transfected with chimeric H65 light chain and heavy chain (each consisting of unmodified mouse V/J-segments fused to a human C-segment) in expression vectors similar to those described above. Following incubation, the supernatants were collected and tested by ELISA for the presence of secreted IgG. All of the supernatants contained about 0.03-0.06 ⁇ g/ml IgG.
  • the H65 antibody modified according to the methods of the present invention was tested to determine whether it retained native affinity for antigen. Its binding capability was compared to that of a chimeric H65 IgG antibody (consisting of the chimeric H65 light chain and heavy chain described in Example 2) which has the same affinity for CD5 as unmodified H65 mouse antibody.
  • the humanized H65 (hH65) and chimeric H65 IgG (cH65) from transient transfections described above were concentrated from 4 ml to a final volume of 100 ⁇ l by centrifugation using a Centricon 30 (Amicon, Amicon Division of W.R. Grace and Co., Beverley, Mass.) at 4° C. Both hH65 and cH65 concentrates were then washed once with 1.0 ml of phosphate buffered saline (PBS), pH 7.2 and reconcentrated to approximately 100 ⁇ l. As a control, HB-CHO culture media alone (CM) or media supplemented with purified cH65 (CM+cH65) was concentrated in a similar manner. The final concentrations of hH65 and cH65 were determined by ELISA (anti-human Kappa pre-coat, peroxidase-labelled anti-human gamma for detection) using chimeric IgG as a standard.
  • ELISA anti
  • CH65 IgG 20 ⁇ g of purified CH65 IgG was iodinated (1 mCi of Na 125 I, Amersham, Arlington Heights, Ill.) using lactoperoxidase beads (Enzymobeads, BioRad Laboratories, Richmond, Calif.) in PBS. Iodination was allowed to proceed for 45 minutes at 23° C.
  • 125 I-cH65 IgG was purified from unbound 125 I by gel filtration using a Sephadex G-25-80 column. Concentration and specific activity was determined by measuring the TCA-precipitated counts before and after purification.
  • Molt4-M cells which express CD5 on their surface, were plated into 96 well V-bottom plates at a density of 3 ⁇ 10 5 cells per well and pelleted by centrifugation. The medium was decanted, and 100 ⁇ l of purified cH65 IgG at final concentrations from 200 nM to 0.0017 nM (diluted in 3-fold steps) in “BHD” [DMEM (Dulbecco's Modified Eagle's Medium)+It BSA+10 mM Hepes, pH 7.2] (BHD) was added to each well, followed by 100 ⁇ l of 125 I-cH65 IgG (final concentration 0.1 nM) in BHD.
  • BHD DMEM (Dulbecco's Modified Eagle's Medium)+It BSA+10 mM Hepes, pH 7.2
  • the method of the present invention for preparing modified antibody variable domains by identifying modifiable amino acids was applied to the anti-TAC antibody variable domain sequence [SEQ ID Nos. 49 (light chain) and 53 (heavy chain)] and the resulting modified sequence is compared to the humanized anti-TAC antibody sequence [SEQ ID Nos. 51 (light chain) and 55 (heavy chain)] described in Queen et al., supra.
  • FIGS. 10A and 10B The results are shown in FIGS. 10A and 10B .
  • the sequence modified according to the present invention [SEQ ID Nos. 50 (light chain) and 54 (heavy chain)] is shown on the lines labelled “prop,” and the Queen humanized sequence is shown on lines labelled “Que.” Modifications to the Queen humanized sequence were based on the human EU antibody sequence [SEQ ID Nos. 48 (light chain) and 52 (heavy chain)].
  • the comparison reveals many differences between the proposed sequence generated by the methods of the present invention and the Queen humanized sequence.
  • the differences which are the most likely to affect binding activity of their humanized antibody are positions 4 (L vs. M), 15 (P vs. V), 36 (F vs. Y), 47 (W vs. L), 71 (Y vs. F), and 80 (A vs. P) in the light chain, as well as position 69 (L vs. I) in the heavy chain.
  • the moderate or high risk mutations may therefore be divided into four groups (consisting of F 1 and F 3 in the light subunit and F 1 and F 3 in the heavy subunit).
  • Four different constructs may be made, each containing higher-risk “human” mutations in only one framework region with the other three frameworks left completely “mouse,” and assayed for activity. This technique avoids the dilemma raised by other humanization methods in which all higher-risk changes are made at once, making it difficult to determine which of the many amino acid changes is responsible for affecting antigen-binding activity.
  • the creation of antibodies according to the invention which possess moderate risk changes are described below.
  • FIGS. 16A and 16B The human consensus sequences in which moderate risk residues are converted from mouse residues to human residues are represented in FIGS. 16A and 16B as lines labelled hxl (i.e., subgroup 1 of the human kappa chain) and hH3 (i.e., subgroup 3 of the human heavy chain). Symbols in this Figure, for conservation and for risk are used in accordance with FIGS. 6A and 6B .
  • a dot (.) represents a residue which may be mutated from “mouse” to “human” at moderate risk. There are 29 such moderate risk positions.
  • the mouse residue matches the human consensus residue more than 50% of the time at 131 positions (102 positions match 90%-100% and 29 positions match 50% to 90%). These positions were not changed.
  • the lines labelled M/H in FIGS. 16A and 16B indicate the 91 positions which differed significantly between the mouse and human sequences (i.e., where the human sequences have the mouse residue less than 50% of the time).
  • Moderate risk positions designated m in the M/H line, were kept “mouse”; whereas those designated H or h were changed to human.
  • the 25 low risk positions which were already human-like or which were previously humanized (as described supra in Example 1) are designated “ ⁇ circumflex over ( ) ⁇ ” in the M/H line.
  • the 54 high risk positions in which the mouse and human residues did not match are designated M and are kept “mouse”.
  • mouse residue aligns with a human consensus amino acid which is moderately conserved.
  • the positions are identified as ones to be kept “mouse.” Although there are no such positions in this particular sequence, such positions may occur in other antibodies.
  • mice residue aligns with a human consensus amino acid which is moderately conserved but the mouse residue is not found at that position in an actual human antibody sequence in Kabat, et al. Sequences of Proteins of Immunoglobulin Interest , supra. Therefore, that position is identified as ones to be changed to “human.”
  • mouse residue aligns with a human consensus amino acid which is poorly conserved. Therefore, that position is identified as one to be kept “mouse.”
  • the humanized H65 heavy chain containing the moderate risk residues was assembled by a strategy similar to that for the low risk residues.
  • the moderate-risk expression vector was assembled from intermediate vectors.
  • the six oligonucleotide sequences (oligos), disclosed in FIG. 7B and labelled HUH-G11, HUH-G12, HUH-G3, HUH-G4, HUH-G5, and HUH-G6 (the sequences of HUH-G11 and HUH-G12 are set out in SEQ ID Nos. 56 and 57) were assembled by PCR.
  • Oligonucleotides containing the synthetic humanized antibody gene were mixed in pairs (HUH-GIl+HUH-G12, HUH-G3+HUH-G4, and HUH-G5+HUH-G6) in a 100 ⁇ l reaction with 1 ⁇ g of each DNA and filled in as described above. A portion of each reaction product was mixed in pairs (HUH-G11, 12+HUH-G3, 4; HUH-G3, 4+HUH-G5, 6), 2.5 U Taq was added and samples were reincubated as described above.
  • the in-J-region was assembled by mixing equal amounts of the HUH-Gil, 12, 3, 4 reaction product with the HUH-G3, 4, 5, 6 product, followed by PCR with 0.5 ug of primers H65G-2S and H65-G2 as described above.
  • the reaction product was cut with SalI and BstEII and cloned into the expression vector, similar to that described for heavy chain in Robinson et al., Hum. Antibod. Hybridomas 2:84 (1991), generating pING4617. That plasmid was sequenced with Sequenase (USB, Cleveland), revealing that two residues were altered (a G-A at position 288 and a A-T at position 312, numbered from the beginning of the leader sequence). The correct variable region was restored by substitution of this region from pING4612, generating the expected V-region sequence in pING4619.
  • An intermediate vector containing the other moderate-risk changes was constructed by PCR assembly of the oligos HUH-G13, HUH-G14, HUH-G15, and HUH-G16 ( FIG. 7A and SEQ ID Nos: 58-61).
  • Oligos HUH-G13+HUH-G14 and HUH-G15+HUH-G16 were mixed and filled in with Vent polymerase (New England Biotabs) in a reaction containing 10 mM KCl, 20 mM TRIS pH 8.8, 10 mM (NH 4 ) 2 SO 2 , 2 mM MgSO 4 , 0.1% Triton X-100, 100 ng/ml BSA, 200 uM of each dNTP, and 2 units of Vent polymerase in a total volume of 100 ⁇ l.
  • the reaction mix was incubated at 94° C. for 1 minute, followed by 2 minutes at 50° C. and 20 minutes at 72° C.
  • reaction products (40 ⁇ l) were mixed and amplified with the oligonucleotides H65-G13 and H65-G2 with Vent polymerase in the same reaction buffer and amplified for 25 cycles with denaturation at 94° C. for 1 minute, annealing at 50° C. for 2 minutes and polymerization at 72° C. for 3 minutes.
  • the reaction product was treated with T4 polymerase and then digested with AccI.
  • the 274 base pair (bp) fragment was purified on an agarose gel and ligated along with the 141 bp SalI to AccI fragment from pING4619 into pUC18 cut with SalI and SmaI to generate pING4620.
  • pING4620 contains the entire signal sequence, V-region, and J-region of the moderate-risk H65 heavy chain.
  • the final expression vector for the moderate-risk H65 heavy chain, pING4621 was assembled by cloning the SalI to BstEII fragment from pING4620 into the same expression vector described above.
  • the moderate-risk humanized V- and J-segments of the light chain were assembled from six oligonucleotides, $H65K-1, HUH-K7, HUH-K6, HUH-K8, HUH-K4 and HUH-K5.
  • the sequences of HUH-K7, HUH-K6 and HUH-K5 are set out in SEQ ID Nos. 62-64 and FIGS. 7 and 7 A, respectively.
  • the oligonucleotides were amplified with PCR primers H65K-2S and JK1-HindIII.
  • Oligonucleotides containing the synthetic humanized antibody gene were mixed in pairs ($H65-K1+HUH-K7, HUH-K6+HUH-K4+HUH-K5) and incubated with Vent polymerase as described for the moderate-risk heavy chain. A portion of each reaction product (40 ul) was mixed in pairs ($H65H-K1/HUH-K7+HUH-K6, 8; HUH-K6, 8+HUH-K4, 5) and filled in as above.
  • the light chain gene was then assembled by amplifying the full length gene with the PCR primers H65K-2S and JK1-HindIII with Vent polymerase for 25 cycles as outlined above. The assembled V/J region was cut with SaLI and HindIII, purified by electrophoresis on an agarose gel, and assembled into a light chain antibody expression vector, pING4630.
  • the cell line Sp2/0 (American Type Culture Collection #CRL1581) was grown in Dulbecco's Modified Eagle Medium plus 4.5 g/l glucose (DMEM, Gibco) plus 10% fetal bovine serum. Media were supplemented with glutamine/penicillin/streptomycin (Irvine Scientific, Irvine, Calif.).
  • the electroporation method of Potter, H., et al., Proc. Natl. Acad. Sci., USA, 81:7161 (1984) was used. After transfection, cells were allowed to recover in complete DMEM for 24-48 hours, and then seeded at 10,000 to 50,000 cells per well in 96-well culture plates in the presence of selective medium. Histidinol (Sigma) selection was at 1.71 ug/ml, and mycophenolic acid (Calbiochem) was at 6 ug/ml plus 0.25 mg/ml xanthine (Sigma). The electroporation technique gave a transfection frequency of 1-10 ⁇ 10 ⁇ 5 for the Sp2/0 cells.
  • the He3 light chain expression plasmid pING4630 was linearized by digestion with PvuI restriction endonuclease and transfected into Sp2/0 cells, giving mycophenolic acid—resistant clones which were screened for light chain synthesis.
  • the best 4 light chain—producing transfectants after outgrowth were pooled into 2 groups of 2 transfectants/pool and each pool was transfected with the He3 heavy chain expression plasmid, pING4621, that had been linearized with PvuI.
  • Sp2/0-4630+4621 cells are grown in culture medium HB101 (Hana Biologics)+1% Fetal Bovine Serum, supplemented with 10 mM HEPES, 1 ⁇ Glutamine-Pen-Strep (Irvine Scientific #9316).
  • the spent medium is centrifuged at about 5,000 ⁇ g for 20 minutes.
  • the antibody level is measured by ELISA.
  • Approximately 200 ml of cell culture supernatant is loaded onto a 2 ml Protein A-column (Sigma Chemicals), equiliberated with PBS (buffer 0.15 M NaCl, 5 mM sodium phosphate, 1 mM potassium phosphate, buffer pH 7.2).
  • the He3 antibody is eluted with a step pH gradient (pH 5.5, 4.5 and 2.5).
  • a fraction containing He3 antibody (9% yield) but not bovine antibody is neutralized with 1 M Tris pH 8.5, and then concentrated 10-fold by Centrium 30 (Amicon)-diluted 10-fold with PBS, reconcentrated 10-fold by Centricon 30, diluted 10-fold with PBS, and finally reconcentrated 10-fold.
  • the antibody was stored in 0.25 ml aliquots at ⁇ 20° C.
  • the affinity of He3 for CD5 was determined using Molt-4M cells, which express CD5 on their surface and I 125 -labeled chimeric H65 IgG in a competitive binding assay.
  • cH65 IgG chimeric H65 IgG
  • 20 ⁇ g of chimeric H65 IgG was iodinated by exposure to 100 ⁇ l lactoperoxidase-glucose oxidase immobilized beads (Enzymobeads, BioRad), 100 ⁇ l of PBS, 1.0 mCi I 125 (Amersham, IMS30), 50 ⁇ l of 55 mM b-D-glucose for 45 minutes at 23° C.
  • the reaction was quenched by the addition of 20 ⁇ l of 105 mM sodium metabisulfite and 120 mM potassium iodine followed by centrifugation for 1 minute to pellet the beads.
  • 125 -cH65 IgG was purified by gel filtration using 7 mls of sephadex G25, using PBS (137 mM NaCl, 1.47 mM KH 2 PO 4 , 8.1 mM Na 2 HPO 4 , 2.68 mM KCl at pH 7.2-7.4) plus 0.1% BSA. 125 I-cH65 IgG recovery and specific activity were determined by TCA precipitation.
  • the murine monoclonal antibody produced by cell line XMMLY-H65 (MoAbH65) is reactive with the human CD5 antigen.
  • the cell line XMMLY-H65 was deposited with the American Type Culture Collection, 12301 Parklawn Drive, Rockville, Md., 20852 and designated Accession No. EB9286.
  • MoAb H65 was produced after immunization of BALB/c mice with the human T-cell line HSB-2 originally isolated from a patient with T-cell acute lymphocytic leukemia. Adams, et al. Can. Res. 28:1121 (1968).
  • the murine myeloma cell line P3 7 NS/1-Ag-1-4 of Kohler et al. Ernr. J. Immunol. 6:292 (1976) was fused with spleen cells from an immunized mouse by the technique of Galfre et al., Nature 266:550 (1977).
  • the H65 antibody-producing hybrid cell line was cloned twice by limiting dilution and was grown as ascites tumors in BALB/c mice.
  • MoAb H65 was purified from mouse ascites by a modification of the method of Ey et al. Immunochem. 15:429 (1978). In brief, the thawed mouse ascites was filtered to remove lipid-like materials and was diluted with 2 to 3 volumes of 0.14 M NaPO 4 , pH 8.0, before application onto an immobilized protein A-Sepharose column of appropriate size. The unbound materials were removed from the column by washing with 0.14 M NaPO 4 , pH 8.0, until no further change in absorbance at 280 nm was seen. A series of column washes with 0.1 M sodium citrate (pH 6.0, pH 5.0, pH 4.0, and pH 3.0) were then performed to elute bound antibody.
  • Peak fractions were pooled, adjusted to pH 7.0 with saturated Tris base, and concentrated by using a cell stirred with Amicon YM10 membrane (Amicon, Lexington, N.Y.). An antibody solution was then dialyzed against phosphate-buffered saline (PBS), pH 7.0, and was stored frozen at ⁇ 70° C.
  • PBS phosphate-buffered saline
  • MoAb H65 is of the IgG 1 subclass, as determined by double diffusion in agar with the use of subclass-specific antisera (Miles-Yeda, Ltd. Rehovot, Israel). The serologic characteristics of this antibody and the biochemical characteristics of the gp67 (i.e., CD5) antigen were examined during the First International Workshop on Human Leukocyte Differentiation Antigens (Paris, 1982). MoAb H65 (workshop number: T34), and nine other MoAbs were found to have the same serologic pattern and to immunoprecipiate the gp67 antigen. Knowles, in Reinherz, et al., Leukocyte Typing II, 2: 259-288 (Springer-Verlag, 1986).
  • MoAb H65 has been shown to block the binding of FITC-conjugated anti-Leu-1 (Becton Dickson, Mountain View, Calif.) on gp67+cells indicating that both antibodies recognize the same epitope on the gp67 molecule or determinants that are located in such a configuration as to result in blocking by steric hindrance.
  • CIA Collagen-induced arthritis
  • MEC major histocompatibility complex
  • CIA Native, heterologous Type II collagen emulsified with complete Freund's adjuvant induces an arthritis-like autoimmune reaction in DBA/IJ mice after a single intradermal tail injection.
  • the mice were obtained from Jackson Laboratories, Bar Harbor, Me. Initially, the arthritis is noticeable as a slight swelling of one or more digits in the fourth week post-immunization.
  • the chronic phase of CIA continually worsens over the ensuing 8 weeks as the arthritis progresses from the digits into the remaining peripheral articulating joints and eventually ends with ankylosis of the involved joints.
  • the histopathology of CIA is characterized by lymphocyte infiltration of the joint space, synovial MHC class II expression and pannus formation. Not all joints are involved on every mouse, so there is a spectrum of arthritic severity. In a group of ten or more mice, the overall arthritic severity develops in a linear fashion over the course of 10-12 weeks.
  • the CIA model was used to test the potential efficacy of a monoclonal antibody directed against the pan-T cell surface antigen, Lyt-1, the murine equivalent of CD5.
  • the antibody was administered to the mice before the immunization with Type II collagen. Normal DBA/I mice were also treated with a single 0.4 mg/kg i.v. injection of anti-Lyt-1 and were sacrificed after 72 hours for FACS analysis and for in vitro proliferation assays on spleen and lymph node cells. Any efficacy of this antibody would indicate a beneficial T cell-directed approach in rheumatoid arthritis via the CD5 surface antigen.
  • Antibody 53-7.313 is a rat IgG 2a monoclonal antibody (ATCC Accession No. TIB 104) reactive with all alleles of the mouse lymphocyte differentiation antigen, Lyt-1.
  • the IND1 antibody is a mouse IgG 1 , anti-human melanoma antibody used as a negative control (Xoma Corp., Berkeley, Calif.). All other antibodies were obtained from Pharmingen Inc. (San Diego, Calif.) as direct conjugates for quantitation on a Becton-Dickinson FACScan instrument.
  • mice Male DBA/IJ mice, age 6-8 weeks, were administered a single intravenous dose of either phosphate buffered saline, IND1 or anti-Lyt-1 via the tail vein at 0.4 mg/kg in 0.1 ml of phosphate buffered saline. Mice were sacrificed for analysis three days after dosing. Single cell suspensions of spleens and peripheral lymph nodes were prepared by standard procedures and 1 ⁇ 10 6 cells were stained with the respective antibodies for fluorescence activated cell sorter (FACS) analysis. Proliferation assays were also performed to provide a second measure of T cell depletion.
  • FACS fluorescence activated cell sorter
  • Cells (1 ⁇ 10 5 /well) were stimulated with Concanavalin A, Interleukin-2 (IL-2), IL-2 and H57.597 (a pan ⁇ , ⁇ T cell receptor antibody) or the Staphylococcal enterotoxins A and B.
  • Cells were cultured for a total of 72 hours and proliferation was quantitated by the addition of 3 H-methylthymidine for the last 24 hours. After 72 hours, the cells were harvested with an Inotech INB-384 harvesting and counting system, which collects the cells onto glass fiber filters with subsequent gas proportional beta particle detection. Results are generally expressed as the mean of triplicate wells ⁇ SEM in Tables 5 and 6.
  • LNC lymph node cells
  • SPC spleen cells
  • mice Male DBA/IJ mice, age 6-8 weeks, were administered the antibodies 53-7.313 (anti-Lyt-1), IND1 (anti-melanoma) or phosphate buffered saline (PBS) in two intravenous (0.4 mg/kg) doses 48 hours apart starting four days prior to immunization with 100 ⁇ g of bovine type II collagen emulsified with an equal volume of Fraund's complete adjuvant to a final injection volume of 100 ⁇ l. Each dose group was comprised of ten mice.
  • An individual mouse could have a maximum arthritic severity score of 8. Mice were monitored until day 80 after collagen immunization and then were sacrificed by cervical dislocation. Results are expressed as the mean arthritic score for each dose group.
  • FIG. 12 The changes in arthritic score during the course of the study are shown in FIG. 12 .
  • the overall conclusion in FIG. 12 is that administration of the anti-Lyt-1 antibody prior to collagen immunization caused a significant decrease in the resulting severity of arthritis.
  • the anti-Lyt-1 treatment group began to show ameliorated arthritic symptoms at Day 48 and never developed arthritis to the same extent as the other two groups. The onset of arthritis was not significantly delayed by the anti-Lyt-1 treatment.
  • the intravenous administration of a rat monoclonal antibody reactive to the mouse equivalent of CD5, Lyt-1 is able to significantly decrease T lymphocytes in the spleen and in peripheral lymph nodes after a single 0.4 mg/kg dose.
  • This T cell decrease is the probable mechanism for the significant (p ⁇ 0.01) decrease in arthritic severity seen with the same anti-Lyt-1 dose prior to type II collagen immunization.
  • mice Severe combined immunodeficient mice maintain human lymphoid cells for several months following transplantation of human peripheral blood mononuclear cells (PBMC).
  • PBMC/SCID mice Such chimeric mice, referred to as PBMC/SCID mice, have functional human cells, as shown by the presence of human Ig in their serum.
  • PBMC/SCID mice maintain human T cells in tissues such as spleen and blood.
  • Human T cells present in PBMC/SCID mice are predominantly of a mature phenotype and express T cell antigens, including CD3, CD5, CD7, and CD4 or CD8.
  • most T cells appear to be activated memory cells, as judged by the expression of HLA-DR and CD45RO.
  • T cells appear to be functional since (a) they may provide help to B cells to produce anti-tetanus toxoid antibodies, (b) they produce soluble interleukin-2 receptor (sIL-2R) which may be detected in plasma, and (c) they proliferate in response to mitogenic anti-human CD3 monoclonal antibodies supplemented with IL-2 in vitro.
  • sIL-2R soluble interleukin-2 receptor
  • PBMC/SCID mice offer an in vivo model system in which to evaluate the efficacy of anti-human T cell drugs, such as H65 MoAb, a mouse IgG1 directed against human CD5.
  • anti-human T cell drugs such as H65 MoAb, a mouse IgG1 directed against human CD5.
  • SCID mice were obtained from Taconic, Germantown, N.Y., and at 6 to 7 weeks of age were injected with 200 mg/kg cyclophosphamide intraperitoneally (i.p.) to ensure engraftment of human PBMC.
  • cyclophosphamide intraperitoneally i.p.
  • mice were bled from the retro-orbital sinus and levels of human immunoglobulin (Ig) and human sIL-2R in plasma were quantified using sandwich ELISAs. Mice with low or undetectable levels of these human proteins were eliminated from the study and the remainder were divided into the various treatment groups (6 per group).
  • the mice were then administered H65 MoAb (0.2 or 0.02 mg/kg/day), H65-based F(ab′) 2 fragment (2 mg/kg/day) or vehicle (buffer) intravenously (i.v.) for 10 consecutive daily injections one day after the last injection, the mice were bled and spleens were collected. Single cell suspensions of blood cells and splenocytes were prepared by standard methods. Recovered cells were then assayed for human T cell surface markers using flow cytometry.
  • FITC— or PE-conjugated Abs Becton-Dickinson, Mountain View, Calif.: HLe-1-FITC (anti-CD45), Leu-2-FITC•(anti-CD5), and Leu-3-PE (anti-CD4). Samples were analyzed on a FACScan using log amplifiers. Regions to quantify positive cells were set based on staining of cells obtained from naive SCID mice. The absolute numbers of human antigen-positive cells recovered from SCID tissues were determined by multiplying the percent positive cells by the total number of cells recovered from each tissue sample. The total number of leukocytes in blood was calculated using a theoretical blood volume of 1.4 ml/mouse. Statistical comparisons between treatment groups were made using the Mann-Whitney U test.
  • FIG. 13 The number of human T cells (CD4 plus CD8 cells) recovered from spleens and blood of PBMC/SCID mice following treatment with H65 MoAb or vehicle (control) is shown in FIG. 13 .
  • Significantly lower numbers of T cells were recovered from spleens and blood of mice treated with either 0.2 or 0.02 mg/kg/day H65 MoAb as compared to vehicle-treated mice.
  • treatment with 2 mg/kg/day of an H65-based F(ab′) 2 fragment did not significantly deplete human T cells from spleens or blood, even though a 10 to 100-fold higher dose was used ( FIG. 14 ).
  • Collagen-induced arthritis (CIA) in the diabetes-resistant Biobreeding (DR BB) rat is a particularly relevant animal model of human rheumatoid arthritis, in that the DR BB rat RT1.D ⁇ gene encodes a nucleotide sequence homologous to the human HLA-DR ⁇ gene reported to be associated with rheumatoid arthritis susceptibility.
  • DR BB rats are administered a single intradermal tail injection of heterologous Type II collagen emulsified with incomplete Freund's adjuvant. Development of the arthritis is considerably faster than in the DBA/1J CIA model. Onset of clinical signs occurs 1.5 to 2 weeks after collagen immunization, with peak swelling observed a few days after onset.
  • Incidence is generally quite high (>85% of animals immunized).
  • the swelling is generally severe, involves the entire footpad and ankle joint, and is restricted to the hindlimbs.
  • Histopathological examination has revealed that the arthritis begins as a proliferative synovitis with pannus formation at the joint margins that is followed by a bidirectional erosion of both the outer (unmineralized) and inner (mineralized) layers of cartilage.
  • This experiment uses the DR BB CIA rat model to assess the efficacy of a monoclonal antibody (MoAb), OX19 directed against the equivalent of the CD5 antigen in the rat.
  • the antibody was administered to the rats prior to immunization with Type II collagen.
  • Normal Sprague-Dawley rats were also treated with a single 0.5 mg/kg i.v. injection and were sacrificed after 3 hours for evaluation of MoAb binding to T cells, or after 2 days for quantitation of T cells in lymphoid tissues using flow cytometry.
  • OXI9 MoAb is a mouse IgG1 directed against the equivalent of rat CD5 antigen present on rat T cells.
  • OXl9 hybridoma is available from the European Collection of Animal Cell Cultures (ECACC) and has ECACC No. 84112012.
  • H65 MoAb a mouse IgG1 reactive against human CD5, was used as an isotype matched negative control.
  • Fluorescein-conjugated antibodies directed against surface antigens on rat pan-T cells (W3/13), CD4 cells (W3/25) and CD5 cells (OX8) were obtained from Accurate Chemical and Scientific Corporation, Westbury, N.Y. for flow cytometric quantitation of T cells in rat lymphoid tissues.
  • Phycoerythrin-conjugated goat anti-mouse IgG1 (Caltag Laboratories, South San Francisco, Calif.) was used to detect OX19 MoAb bound to rat T cells in a two-color analysis.
  • T cells were depleted from the blood at 3 hours after OX19 MoAb administration. Almost all of the T cells that remained in the blood, and most of those present in the spleen and lymph nodes in the OX19 MoAb-treated rat also stained positively for surface-bound mouse IgG1, indicating that the dose of OX19 MoAb used was sufficient to saturate most of the T cells in these major lymphoid organs. These results provide doses useful in therapeutic applications.
  • RA rheumatoid arthritis
  • Anti-CD5 antibody prepared as described above is administered to patients at doses of about 0.005 to 2.0 mg/kg/day for a period of 1-5 days, preferably 1-2 days. Alternatively, the dose may be given every 2-30 TABLE 8 Effect of OX19 MoAb Treatment on Arthritis Incidence Total Total arthritics Arthritics Score of “2” Score of “2” (1 or both (Both (1 or both (Both TREATMENT limbs) limbs) limbs) Limbs) PBS/Tween 7/8 (88%) 7/8 (88%) 7/8 (88%) 5/8 (63%) Control MoAb 7/8 (88%) 4/8 (50%) 6/8 (75%) 4/8 (50%) OX19 MoAb 0/8 (0%) 0/8 (0%) 0/8 (0%) 0/8 (0%) days instead of daily if chimeric and humanized MoAbs are used due to their increased half-life. To determine optimum dose and schedule, patients are treated at each dose and schedule in a dose escalating regimen. Patients are monitored
  • SLE Systemic Lupus Erythematosus
  • SLE Systemic Lupus Erythematosus
  • Some of the more frequent manifestations include fatigue, anemia, fever, rashes, photosensitivity, alopecia, arthritis, pericarditis, pleurisy, vasculitis, nephritis and central nervous system disease.
  • Revised Criteria for Classification of SLE a person is said to have SLE for purposes of clinical studies if any four or more of the aforementioned specified criteria are present, serially or simultaneously, during any interval of observation.
  • Anti-CD5 antibody prepared as described above is administered to patients at doses of about 0.005 to 2.0 mg/kg/day for a period of 1-5 days, preferably 1-2 days. Alternatively, the dose may be given every 2-30 days instead of daily if chimeric and humanized MoAbs are used due to their increased half-life. To determine optimum dose and schedule, patients are treated at each dose and schedule in a dose escalating regimen.
  • Psoriasis is a disease of autoimmune etiology which Classically appears as plaques over the elbows and knees, although other areas of the skin are frequently afflicted. Abnormalities of the nails and the joints are also frequently observed. Particularly inflammatory joint disease can occur in an occasionally erosive and severe form.
  • Anti-CD5 antibody prepared as described above is administered to patients at doses of about 0.005 to 2.0 mg/kg/day for a period of 1-5 days, preferably 1-2 days. Alternatively, the dose may be given every 2-30 days instead of daily if chimeric and humanized MoAbs are used due to their increased half-life. To determine optimum dose and schedule, patients are treated at each dose and schedule in a dose escalating regimen.
  • Clinical observation includes evaluation of the patient's overall status as well as special attention to the psoriatic plaques. Additionally, monitoring of laboratory parameters such as white blood count and differential are recommended. Symptoms which may indicate poor tolerance to therapy or complications include nausea, vomiting, fatigue, rash, fever, chills and syncope. Any unexplained depletion in white blood cells other than lymphocytes is an indication to discontinue therapy.
  • differential analysis of lymphocytes is carried out. That is, analysis of the total number of T cells and B cells should be determined.
  • Type I diabetes There are two major types of diabetes. Type I has classically been associated with a requirement for exogenous insulin. Type I typically occurs before the age of 40 and is associated with an absence of insulin secretion. The pancreas of patients with long-term Type I insulin-dependent diabetes are devoid of pancreatic islet cells. There is a large body of evidence that the etiology of Type I insulin-dependent diabetes (IDDM) is autoimmune.
  • IDDM insulin-dependent diabetes
  • Anti-CD5 antibody prepared as described above is administered to patients at doses of about 0.005 to 2.0 mg/kg/day for a period of 1-5 days, preferably 1-2 days. Alternatively, the dose may be given every 2-30 days instead of daily if chimeric and humanized MoAbs are used due to their increased half-life. To determine optimum dose and schedule, patients are treated at each dose and schedule in a dose escalating regimen.
  • Clinical symptoms indicating poor tolerance to therapy or complications include fatigue, vomiting, rash, fever, chills, and syncope.
  • Laboratory evaluation included white blood cell counts with differential analysis daily and blood glucose levels at least twice a day.
  • patients may be selected for prophylactic treatment. This treatment follows the dose and schedule noted above for treatment of clinical insulin dependent diabetes.

Abstract

Methods are described for identifying the amino acid residues of an antibody variable domain which may be modified without diminishing the native affinity of the domain for antigen while reducing its immunogenicity with respect to a hereterologous species and for preparing so modified antibody variable domains which are useful for administration to heterologous species. Antibody variable regions prepared by the methods of the invention are also described.

Description

    FIELD OF THE INVENTION
  • The present invention relates, in general, to methods for preparing a modified antibody variable domain by determining the amino acid residues of the antibody variable domain which may be modified without diminishing the native affinity of the domain for antigen while reducing its immunogenicity with respect to a heterologous species; to methods of preparation of and use of antibody variable domains having modifications at the identified residues which are useful for administration to heterologous species; and to the variable regions so modified. More particularly, the invention relates to the preparation of modified mouse antibody variable domains, which are modified for administration to humans, the resulting antibody variable domains themselves, and the use of such “humanized” antibodies in the treatment of diseases in humans.
  • BACKGROUND
  • Application of unmodified mouse monoclonal antibodies in human therapy is problematic for three reasons. First, an immune response against the mouse antibodies is mounted in the human body. Second, the antibodies have a reduced half-life in the human circulatory system. Third, the mouse antibody effector domains may not efficiently trigger the human immune system.
  • There are three methods which have attempted to eliminate the foregoing problems. Junghans et al., Cancer Res., 50, 1495-1502 (1990) and other publications describe the utilization of genetic engineering techniques to link DNA encoding murine variable regions to DNA encoding human constant regions, creating constructs which when expressed generate a hybrid mouse/human antibody.
  • Also by genetic engineering techniques, the genetic information from murine hypervariable complementarity determining regions (CDRs) may be inserted in place of the DNA encoding the CDRs of a human monoclonal antibody to generate a construct encoding a human antibody with murine CDRs. This technique is known as “CDR grafting”. See, e.g., Jones et al., Nature, 321, 522-525 (1986); Junghans et al., supra.
  • Protein structure analysis may be used to “add back” murine residues, again by genetic engineering, to first generation variable regions generated by CDR grafting in order to restore lost antigen binding capability. Queen et al., Proc. Natl. Acad. Sci. USA, 86, 10029-10033 (1989); Co, et al., Proc. Natl. Acad. Sci. USA, 88, 2869-2873 (1991) describe versions of this method. The foregoing three methods are techniques to “humanize” mouse monoclonal antibodies.
  • As a result of the humanization of mouse monoclonal antibodies, specific binding activity of the resulting humanized antibodies may be diminished or even completely abolished. For example, the binding affinity of the modified antibody described in Queen et al., supra, is reported to be reduced three-fold; in Co et al., supra, is reported to be reduced two-fold; and in Jones et al., supra, is reported to be reduced two- to three-fold. Other reports describe order-of-magnitude reductions in binding affinity. See, e.g., Tempest et al., Bio/Technology, 9, 266-271 (1991); Verhoeyen et al., Science, 239, 1534-1536 (1988).
  • A system for differentiating between the various subsets of T Cells, based upon cell surface antigens, is the Clusters of Differentation System (hereinafter referred to as the “CD System”). The CD System represents standard nomenclature for molecular markers of leukocyte cell differentation molecules. See Leukocyte Typing III White Cell Differentiation Antigens (Michael, ed. Oxford Press 1987), which is incorporated herein by reference.
  • So-called “pan T cell” markers (or antigens) are those markers which occur on T Cells generally and are not specific to any particular T cell subset(s). Pan T Cell markers include CD2, CD3, CD5, CD6, and CD7. The CD5 cluster antigen, for example, is one of the pan T markers present on about 85-100% of the human mature T lymphocytes and a majority of human thymocytes. CD5 is also present on a subset, about 20%, of B cells. Extensive studies using flow cytometry, immunoperoxidase staining, and red cell lysis have demonstrated that this antigen is not normally present on hematopoietic progenitor cells or on any other normal adult or fetal human tissue with the exception of the aforementioned subpopulation of B cells.
  • Further information regarding the CD5 marker is found in McMichael and Gotch, in Leukocyte Typing III White Cell Differentiation Antigens (Michael, ed. Oxford Press 1987). The CD5 molecule has also been described in the literature as reactive with immunoglobulins. See, e.g., Kernan et al., J. Immunol., 33:137-146 (1984), which is incorporated herein by reference.
  • There are reports of attempted treatment of rheumatoid arthritis patients with monoclonal antibodies against CD4. See Horneff, et al. Arthritis and Rheumatism 34:2, 129-140 (February 1991); Goldberg, et al., Arthritis and Rheumatism, Abstract D115, 33:S153 (September. 1990); Goldberg, Journal of Autoimmunity, 4:617-630 (1991); Choy, et al. Scand. J. Immunol. 36:291-298 (1992).
  • There are reports of attempted treatment of autoimmune disease, particularly rheumatoid arthritis, with an anti-CD5 monoclonal antibody. See Kirkham, et al., British Journal of Rheumatology 30:459-463 (1991); Kirkham, et al., British Journal of Rheumatology 30:88 (1991); Kirkham, et al., Journal of Rheumatology 19:1348-1352 (1992). There is also a report of an attempt to treat multiple sclerosis with an anti-T12 antibody. Hafler, et al., Neurology 36:777-784 (1986).
  • As demonstrated by the foregoing, there exists a need in the art for a method of preparing antibody variable domains by identification of residues in mouse monoclonal variable region domains which may be modified without diminishing the native affinity of the domains for antigen while reducing their immunogenicity with respect to a heterologous species for use in the treatment of diseases.
  • SUMMARY OF THE INVENTION
  • The present invention provides methods for preparing a modified antibody variable domain useful for administration to humans by determining the amino acids of a subject antibody variable domain which may be modified without diminishing the native affinity of the domain for antigen while reducing its immunogenicity with respect to a heterologous species. As used herein, the term “subject antibody variable domain” refers to the antibody upon which determinations are made. The method includes the following steps: determining the amino acid sequence of a subject light chain and a subject heavy chain of a subject antibody variable domain to be modified; aligning by homology the subject light and heavy chains with a plurality of human light and heavy chain amino acid sequences; identifying the amino acids in the subject light and heavy chain sequences which are least likely to diminish the native affinity of the subject variable domain for antigen while, at the same time, reducing its immunogenicity by selecting each amino acid which is not in an interface region of the subject antibody variable domain and which is not in a complementarity-determining region or in an antigen-binding region of the subject antibody variable domain, but which amino acid is in a position exposed to a solvent containing the antibody; changing each residue identified above which aligns with a highly or a moderately conserved residue in the plurality of human light and heavy chain amino acid sequences if said identified amino acid is different from the amino acid in the plurality.
  • Another group of sequences, such as those in FIGS. 1A and 1B may be used to determine an alignment from which the skilled artisan may determine appropriate changes to make.
  • The present invention provides a further method wherein the plurality of human light and heavy chain amino acid seqeunces is selected from the human consensus sequences in FIGS. 5A and 5B.
  • In general, human engineering according to the above methods may be used to treat various diseases against which monoclonal antibodies generally may be effective. However, humanized antibodies possess the additional advantage of reducing the immunogenic response in the treated patient.
  • The present invention also discloses products and pharmaceutical compositions useful in the treatment of a myriad human diseases. In particular, products prepared by the foregoing methods include a modified H65 mouse monoclonal variable domain. Additionally, DNA sequences encoding the modified H65 variable domain are provided.
  • Modified antibody variable domains which are products of the methods of the present invention may be used, inter alia, as components of various immunoglobulin molecules such as Fab, Fab′, and F(ab′)2 domains, single chain antibodies, and Fv or single variable domains.
  • Immunoglobulin molecules comprising modified variable domains according to the invention are particularly suited for therapeutic administration to human by themselves or, for example, as components of immunoconjugates such as those described in co-pending, co-owned U.S. patent application Ser. No. 07/787,567 filed on Nov. 4, 1991.
  • The present invention also provides methods for treatment of autoimmune diseases, wherein animal models are predictive of the efficacy of treatment in humans. Finally, the present invention includes pharmaceutical compositions containing the humanized antibodies according to the invention are disclosed.
  • BRIEF DESCRIPTION OF THE DRAWING
  • FIGS. 1A and 1B are alignments of the amino acid sequences of the light and heavy chains, respectively, of four antibody variable domains [HYH (HYHEL-10 Fab-lysosyme complex), MCPC (IgA Fab MCPC603-phosphocholine complex), NEWM (Ig Fab′ NEW) and KOL (IgG1 KOL)] by criteria of sequence and structural homology;
  • FIG. 2 is a schematic depiction of the structural relationships between the amino acid residues of the light chain of the variable domain;
  • FIG. 3 is a schematic depiction of the structural relationships between the amino acid residues of the heavy chain of the variable domain;
  • FIG. 4 is a schematic representation of an antibody variable domain;
  • FIGS. 5A and 5B are alignments of the consensus amino acid sequences for the subgroups of light [hK1 (human kappa light chain subgroup 1), hK3 (human kappa light chain subgroup 3), hK2 (human kappa light chain subgroup 2), hL1 (human lambda light chain subgroup 1), hL2 (human lambda light chain subgroup 2), HL3 (human lambda light chain subgroup 3), hL6 (human lambda light chain subgroup 6), hK4 (human kappa light chain subgroup 4), hL4 (human lambda light chain subgroup 4) and hL5 (human lambda light chain subgroup 5] and heavy chains [hH3 (human heavy chain subgroup 3), hH1 (human heavy chain subgroup 1) and hH2 (human heavy chain subgroup 2)], respectively, of human antibody variable domains;
  • FIGS. 6A and 6B are alignments of human light chain consensus sequence hK1 with the actual (h65) and low-risk modified (prop) light chain sequences of the H65 mouse monoclonal antibody variable domain and of human heavy chain consensus sequence hH3 with the actual (h65) and modified (prop) heavy chain sequences of the H65 mouse monoclonal antibody variable domain, respectively;
  • FIGS. 7A and 7B are listings of the nucleotide sequences of the oligonucleotides utilized in the construction of the genes encoding modified V/J-regions of the light and heavy chains of the H65 mouse monoclonal antibody variable domain;
  • FIGS. 8A and 8B are listings of the nucleotide sequences of the genes encoding modified V/J-regions of the heavy and light chains, respectively, of the H65 mouse monoclonal antibody variable domain;
  • FIG. 9 is a graph of the results of a competitive binding assay showing that the H65 antibody variable domain modified by a method according to the present invention retains the antigen-binding capability of the natural H65 antibody variable region;
  • FIGS. 10A and 10B are alignments of human light chain consensus hK1 and heavy chain consensus hH1 with the light and heavy chain sequences, respectively, of the variable domain of human antibody EU, human antibody TAC, murine antibody TAC modified according to the present invention (prop) and murine antibody TAC modified according to a different method (Que);
  • FIG. 11 is a graph of He3 IgG binding to CD5 found on Molt-4M, demonstrating that such binding is similar to that of cH65 IgG;
  • FIG. 12 is a graph showing the effects of anti-Lyt-1 administration on the severity of collagen-induced arthritis in DBA/1J mice;
  • FIGS. 13A and 13B are depictions of human T cell recovery in spleen and blood, respectively from PBMC/SCID mice following treatment with H65 MoAb;
  • FIGS. 14A and 14B are schematic depictions of human T cell recovery in spleen and blood, respectively from PBMC/SCID mice following treatment with H65-based F(ab′)2 fragment;
  • FIG. 15 is a graph of the effects of OX19 MoAb on the severity of DR BB rat collagen-induced arthritis; and
  • FIGS. 16A and 16B are alignments of human light chain consensus sequence hK1 with the actual (h65) and low and moderate risk modified (prop) light chain sequences of the H65 mouse monoclonal antibody variable domain and of human heavy chain consensus sequence hH3 with the actual (h65) and modified (prop) heavy chain sequences of the H65 mouse monoclonal antibody variable domain, respectively.
  • DETAILED DESCRIPTION
  • Methods according to the present invention include: (1) identification of the amino acid residues of an antibody variable domain which may be modified without diminishing the native affinity of the domain for antigen while reducing its immunogenicity with respect to a heterologous species; (2) the preparation of antibody variable domains having modifications at the identified residues which are useful for administration to heterologous species; and (3) use of the humanized antibodies of the invention in the treatment of autoimmune diseases in humans. The methods of the invention are based on a model of the antibody variable domain described herein which predicts the involvement of each amino acid in the structure of the domain.
  • Unlike other methods for humanization of antibodies, which advocate replacement of the entire classical antibody framework regions with those from a human antibody, the methods described herein introduce human residues into the variable domain of an antibody only in positions which are not critical for antigen-binding activity and which are likely to be exposed to immunogenicity-stimulating factors. The present methods are designed to retain sufficient natural internal structure of the variable domain so that the antigen-binding capacity of the modified domain is not diminished in comparison to the natural domain.
  • Data obtained from the analysis of amino acid sequences of antibody variable regions using the MacImdad (Molecular Applications Group, Stanford, California) three-dimensional molecular modeling program, in conjunction with data obtained from previous theoretical studies of hypervariable region structure, and data obtained from the crystal structures of the HYH (HYHEL-10 Fab-lysosyme complex, Brookhaven structure “3HFM”), MCPC (IgA Fab MCPC603-phosphocholine complex, Brookhaven structure “2MCP”), NEWM (Ig Fab′ NEW, Brookhaven structure “3FAB”) and KOL (IgG1 KOL, Brookhaven structure “21G2”) antibody variable domains from the Brookhaven database (Brookhaven National Laboratory, Upton, N.Y.), are utilized to develop the antibody variable domain model.
  • FIGS. 1A and 1B provide the sequences of the four antibody variable domains which have been crystallized. The amino acid sequences of the light and heavy chains of HYH (SEQ ID Nos. 1 and 5, respectively), MCPC (SEQ ID Nos. 2 and 6, respectively), NEWM (SEQ ID Nos. 3 and 7, respectively) and KOL (SEQ ID Nos. 4 and 8, respectively) are shown, wherein the exclamation points “!” in the MCPC light chain sequence at position 30×, the MCPC heavy chain sequence at positions 52× and 98×, the NEWM light chain at position 30×, the KOL light chain at position 93×, and the KOL heavy chain sequence at position 98×, stand for the amino acid sequences NSGNQK (SEQ ID No. 9), NKG (SEQ ID No 10), GST (SEQ ID No 11), AG, SL and HGFCSSASC (SEQ ID No 12), respectively which are variations in the length of hypervariable loop sequences among the various antibodies. FIGS. 2 and 3 comprise depictions of the structure of the light and heavy chains, respectively, wherein each chain is displayed “unfolded” into a flattened beta sheet structure so that interactions among the residues are easier to visualize. The strands of folded polypeptide chains are represented as thick vertical lines, connected by eight beta-turn loops. Three of the loops are identified as antigen-binding loops or CDRs, one is accessory to the loops, and the remaining four at the “bottom” of the variable domain are not involved in antigen binding. The amino and carboxy termini of the variable domain are symbolized by small black dots at the ends of the polypeptide chains. Each amino acid position is represented as either a circle, a triangle, or a square. The covalent disulfide bond between the two cysteines at positions 23 and 88 in the light chain and the covalent disulfide bond between positions 22 and 92 in the heavy chain are each shown as a thick horizontal line. All of the residues in each chain are shown on the map, including antigen-binding residues and framework residues. The amino acid positions are numbered according to Kabat et al., Sequences of Proteins of Immunological Interest, Fourth Edition, U.S. Department of Health and Human Services, Public Health Service, National Institutes of Health (1987), with the exception of those designated with a lower-case “x”, which are variations in length of hypervariable loops which Kabat has numbered as “a,b,c,d . . . . ”. Solid slanted lines (either single or double) connecting pairs of residues which are adjacent in three-dimensional space but not in linear sequence, represent one or two hydrogen bonds between the mutually aligned amino nitrogens and carbonyl oxygens in the backbones of the residues.
  • The analysis of each amino acid position to determine whether the position influences antigen binding and/or is immunogenic was based upon the information in FIGS. 1A, 1B, 2 and 3, as well as the additional variable region structural information in the following paragraphs.
  • The basic structure of the antibody variable domain is strongly conserved. The variable domain is composed of a light chain (or subunit) and a heavy chain (or subunit), which are structurally homologous to each other and which are related by a pseudo-two-fold axis of rotational symmetry. At the “top” of the variable domain, the region farthest away from the constant domain, there are six antigen-binding loops which are built upon a larger structural framework region. The variable domain is functionally distinct from the constant domain, being connected only by two highly flexible chains and pivoting on both “ball-and-socket” joints formed by five amino acids in the heavy and light chains.
  • Each subunit, light or heavy, resembles a “sandwich” structure, composed of two layers of antiparallel beta pleated sheets with a propeller twist in three-dimensional space. Each amino acid chain folds back on itself repeatedly to create nine distinct strands. Three-and-one-half of these strands form the “outside” beta-sheet layer of each subunit and the other five-and-one-half form the “inside” layer. The various strands in each layer are extensively hydrogen-bonded to each other. The two beta-sheet layers within the subunit are held together by a single covalent disulfide bond and by numerous internal hydrophobic interactions. The sequences involved in bonding the strands of the subunits together are called “framework” sequences.
  • Certain amino acids, either in antigen-binding sequences or in framework sequences, do not actually bind antigen but are critical for determining the spatial conformation of those residues which do bind. Each antigen-binding loop requires a properly formed “platform” of buried residues, which provides a surface upon which the loop folds. One or more of the loop residues often will be buried in the platform as an “anchor” which restricts the conformational entropy of the loop and which determines the precise orientation of antigen-contacting sidechains. Thus, the shapes of the residues which make up the platform contribute to the ultimate shape of the antigen-binding loop and its affinity for specific antigens.
  • Amino acid sidechains exist in various different chemical environments within the subunits. Some residues are exposed to the solvent on the outer accessible surface while other residues are buried in hydrophobic interactions within a subunit. Much of the immunoglobulin variable domain is constructed from antiparallel beta pleated sheets which create amphipathic surfaces, such that the “inside” surface is hydrophobic and the “outside” surface is hydrophilic. The outside is exposed to solvent, and therefore is also exposed to the humoral environment when the domain is in the circulatory system of an animal. Amino acid sidechains which are completely exposed to the solvent and which do not physically interact with other residues in the variable domain are likely to be immunogenic and are unlikely to have any structural importance within the immunoglobulin molecule. A highly schematic representation of the variable domain is shown in FIG. 4, wherein thick lines represent peptide bonds and shaded circles denote amino acid side chains.
  • The two subunits of antibody variable domains adhere to each other via a hydrophobic interface region which extends along the inside beta-sheet layer from the border of the variable domain with the constant domain to the antigen-binding loops. Amino acid side chains from both subunits interact to form a three-layered “herringbone” structure. Some of these interfacial residues are components of the antigen-binding loops, and thus have a direct effect upon binding affinity. Every residue in the interface is structurally important because the conformation of the binding regions is strongly influenced by changes in the conformation of the interface.
  • The foregoing data and information on the structure of antibody variable domains aids in a determination of whether a particular amino acid of any variable domain is likely to influence antigen binding or immunogenicity. The determination for each amino acid position is represented by a pair of symbols (e.g., + and +, in the lines labelled “bind” and “bury”, respectively) in FIGS. 1A, 1B, (and also in FIGS. 5A, 5B, 6A, 6B, 10A and 10B). In each of these pairs, the first symbol relates to antigen binding, while the second symbol relates to immunogenicity and framework structure. Tables 1, 2 and 3, below, set out the significance of the symbols and possible pairings.
    TABLE 1
    FIRST SYMBOL IN PAIR (LIGAND BINDING)
    + Little or no direct influence on antigen-
    binding loops, low risk if substituted
    Indirectly involved in antigen-binding loop
    structure, moderate risk if changed
    Directly involved in antigen-binding loop
    conformation or antigen contact, great risk if
    modified
  • Table 2
  • SECOND SYMBOL IN PAIR (IMMUNOGENICITY and STRUCTURE)
    + Highly accessible to the solvent, high
    immunogenicity, low risk if substituted
    Partially buried, moderate immunogenicity,
    moderate risk if altered
    Completely buried in subunit's hydrophobic
    core, low immunogenicity, high risk if changed
    = Completely buried in the interface between
    subunits, low immunogenicity, high risk if
    modified.
  • TABLE 3
    SIGNIFICANCE OF PAIRS
    ++ Low risk Highly accessible to the
    solvent and high
    immunogenicity, but
    little or no effect on
    specific antigen binding
    ∘+, +∘, ∘∘ Moderate risk Slight immunogenicity or
    indirect involvement with
    antigen binding
    any − or = High risk Buried within the subunit
    core/interface or
    strongly involved in
    antigen binding, but little
    no immunogenic potential
  • The pairings set out in the Figures indicate that making mouse-to-human modifications at positions which have a pair of low risk symbols (++) (i.e., a symbol in the “bind” line and a symbol in the “bury” line corresponding to one position) results in a major reduction in therapeutic immunogenicity with little chance of affecting binding affinity. At the opposite end of the spectrum, modifying positions which have a pair of high risk symbols (−−) may degrade or abolish binding activity with little or no actual reduction in therapeutic immunogenicity. There are 73 low risk positions in the variable domain (38 in the light chain and 35 in the heavy chain) which are indicated by circles in the lines labelled “risk” in FIGS. 1A, 1B, 5A, 5B, 6A, 6B, 10A and 10B. There are 29 moderate risk positions in the variable domain (12 in the light chain and 17 in the heavy chain) as indicated by the triangles in the lines labelled “risk” in FIGS. 1A, 1B, 5A, 5B, 6A, 6B, 10A, and 10B.
  • The results of the above analysis may be applied to consensus sequences for the different subgroups of antibody variable domains because the structural characteristics they represent are highly conserved, even among various species. FIGS. 5A and 5B thus set out and align the consensus sequences (derived from Kabat et al., supra) of the subgroups of light (hK1, SEQ ID NO: 13; hK3, SEQ ID NO: 14; hK2, SEQ ID NO: 15; hL1 SEQ ID NO: 16; hL2, SEQ ID NO: 17; hL3, SEQ ID NO: 18; hL6, SEQ ID NO: 19; hK4, SEQ ID NO: 20; hL4, SEQ ID NO: 21; and hL5, SEQ ID NO: 22) and heavy chains (hH3, SEQ ID NO: 23; hH1, SEQ ID NO: 24; and hH2, SEQ ID NO: 25) of antibody variable domains with the pairings representing the structural characteristics of each amino acid position, wherein the consensus sequences for the hL6, hK4, hL4, hL5 and hH2 subgroups were derived from less than twenty actual light or heavy chain sequences.
  • In the consensus sequences set out in FIGS. 5A and 5B, upper case amino acid designations indicate that the amino acid is present at that location in about 90% to about 100% of the known human sequences (excluding small incomplete fragments) of that subgroup (i.e., is “highly conserved”); whereas lower case amino acid designations indicate that the amino acid is present at that location in about 50% to about 90% of the known human sequences in that subgroup (i.e., is “moderately conserved”). A lower case “x” denotes conservation in less than about 50% of the known sequences in that subgroup (i.e., a “poorly conserved” position).
  • The information presented in FIGS. 5A and 5B on the relationship of a particular amino acid in a sequence of an antibody variable domain to the structure and antigen-binding capacity of the domain is sufficient to determine whether an amino acid is modifiable. Additional structural studies, such as those on which FIGS. 5A and 5B are based, are not required.
  • Thus, according to the present invention, FIGS. 5A and 5B may be used to prepare, for example, a modified mouse antibody variable domain that retains the affinity of the natural domain for antigen while exhibiting reduced immunogenicity in humans by the following steps. The amino acid sequences of both the light chain and the heavy chain from the mouse variable domain are first determined by techniques known in the art (e.g., by Edman degradation or by sequencing of a cDNA encoding the variable domain). Next, the consensus sequences set out in FIGS. 5A and 5B for human antibody variable regions are examined to identify both a light chain consensus and a heavy chain consensus sequence that are the most homologous to the particular mouse subunit sequences that are to be modified. The mouse sequences are aligned to the consensus human sequences based on homology either by sight or by using a commercially available computer program such as the PCGENE package (Intelligenetics, Mountain View, Calif.).
  • FIGS. 5A and 5B are then used again to identify all of the “low risk” or “moderate risk” positions at which the mouse sequence differs significantly from the chosen human consensus. The mouse amino acid residues at these low risk and moderate risk positions are candidates for modification. If the human consensus is strongly conserved at a given low risk or moderate risk position, the human residue may be substituted for the corresponding mouse residue. If the human consensus is poorly conserved at a given low risk or moderate risk position, the mouse residue is retained at that position. If the human consensus is moderately conserved at a specific position, the mouse residue is normally replaced with a human residue, unless the mouse residue occurs at that position in at least one of the sequences (e.g., in Kabat et al., supra) on which the human consensus sequence is based. If the mouse residue does occur at that position in a human sequence then the mouse residue may be retained.
  • Other criteria may be important to the determination of which identified residues of a variable region are to be modified. For example, since the side chain of proline is connected to both its α-carbon and its peptide nitrogen, free rotation is restricted around the carbon-nitrogen bond (the Ramachandran φ angle). Therefore, wherever there is a proline in a sequence, the shape of the backbone is distorted and that distortion can influence other residues involved in antigen binding. The presence or absence of a proline residue at any point in the amino acid sequence is a structurally important feature. If the mouse sequence contains a proline at a certain location, it is likely that its presence is necessary for a proper backbone and framework conformation and proline is perferably retained. If the mouse sequence does not contain a proline at a location where the human consensus sequence has one, it is likely that substituting a proline in the mouse sequence would affect proper conformation of the sequence, therefore the mouse residue is preferably retained. Where a proline at a particular position involving proline is changed from mouse to human, such a change is considered to be at least moderate risk even if that position would otherwise be low risk.
  • Similarly, insertions and deletions in a mouse sequence, relative to a human consensus framework, are normally preserved intact. If the mouse sequence has an alteration in the length and spacing of the variable region backbone, it is likely that the alteration is necessary to provide a surface for proper folding of the antigen-binding loops. The alteration is preferably retained in a modified version of the sequence.
  • Residues participating in the interface between the light and heavy chains of a variable domain are also preferably left intact in a modified version. They are all designated high risk, with = symbols on the “bury” lines in FIGS. 1, 5, 6, 10. The side chains in the interface region are buried deep within the structure, so they are unlikely to elicit a therapeutic immunogenic response in a heterologous species.
  • Once a modified sequence has been designed, DNAs encoding the complete variable domain are synthesized [via oligonucleotide synthesis as described, for example, in Sinha et al., Nucleic Acids Res., 12, 4539-4557 (1984)], assembled [via PCR as described, for example in Innis, Ed., PCR Protocols, Academic Press (1990) and also in Better et al. J. Biol. Chem. 267, 16712-16118 (1992)], cloned and expressed [via standard procedures as described, for example, in Ausubel et al., Eds., Current Protocols in Molecular Biology, John Wiley & Sons, New York (1989) and also in Robinson et al., Hum. Antibod. Hybridomas, 2, 84-93 (1991)], and finally tested for specific antigen binding activity [via competition assay as described, for example, in Harlow et al., Eds., Antibodies: A Laboratory Manual, Chapter 14, Cold Spring Harbor Laboratory, Cold Spring Harbor (1988) and Munson et al., Anal. Biochem., 107, 220-239 (1980)].
  • Treatment of certain autoimmune diseases with immunotoxin conjugates is described in co-pending, commonly assigned U.S. patent application Ser. No. 07/759,297 filed Sep. 13, 1991, and Bernhard, et al., “Materials Comprising and Methods of Preparation and Use for Ribosome-Inactivating Proteins”, a United States patent application filed Dec. 9, 1992, which are incorporated herein by reference. An immunoglobulin such as an anti-T-cell immunoglobulin may be conjugated to a cytotoxic molecule. The cytotoxic molecule to which the immunoglobulin is conjugated may be any of a number of toxins such as lectin A or a ricin A chain. The above-referenced '297 application also describes use of an anti-CD5 antibody conjugated to a ricin A chain providing an anti-T-cell immunotoxin.
  • A general description of various autoimmune diseases is found in The Autoimmune Diseases (Rose & Mackey, eds 1985). Autoimmune diseases may be characterized, inter alia, by abnormal immunological regulation which results in excessive B Cell activity and diminished, enhanced, or inappropriate T Cell activity. Such altered T cell activity may result in excessive production of autoantibodies. Although the autoimmune diseases are complex and diverse in their manifestations, they possess the common feature of an impaired immune system. Therapeutic depletion of circulating T cells through the administration of an anti-pan T cell immunoglobulin improves the clinical course and prognosis of patients with autoimmune disease. For anti-CD5 antibody therapy, the additional depletion of CD5 B cells may have a further beneficial effect since CD5 B cells have been implicated in some autoimmune diseases.
  • Once prepared, humanized antibodies are then useful in the treatment of autoimmune disease. In this regard, an anti-CD5 monoclonal antibody is presented as an example of a preferred embodiment of the invention. An example of an anti-pan T cell immunoglobulin is an CD5 antibody which is primarily reactive with a surface antigen of mature T cells, but is also reactive with 10-20% of mature B cells. Clinical data obtained using the anti-pan T cell immunoglobulin in models of autoimmune diseases in non-human animals are predictive of the effects of using such immunoglobulins as therapy against human autoimmune diseases.
  • For the purpose of the present invention, an immunoglobulin, such as an antibody, is “reactive” with or “binds to” an antigen if it interacts with the antigen forms an antigen-immunoglobulin complex. The antigen is generally a unique surface protein or marker. A most preferred marker is the CD5 antigen cluster.
  • The anti-pan T cell immunoglobulin may be obtained from a number of sources. It is reactive with most mature T cells or with both T cells and subsets of other lymphoid cells, such as B cells or natural killer (NK) cells. The immunoglobulin may be synthetic or recombinant, including genetically-engineered immunoglobulins such as chimeric immunoglobulins, humanized antibodies, hybrid antibodies, or derivatives of any of these.
  • Chimeric immunoglobulins, antibodies or peptides are comprised of fused portions from different species as a product of chimeric DNA. Chimeric DNA is recombinant DNA containing genetic material from more than one mammalian species. Chimeric immunoglobulins include one portion having an amino acid sequence derived from, or homologous to, a corresponding sequence in an immunoglobulin, antibody or peptide derived from a first gene source while the remaining segment of the chain(s) is homologous to corresponding sequences of another gene source. For example, a chimeric antibody peptide may comprise an antibody heavy chain with a murine variable region and a human constant region. The two gene sources will typically involve two species, but will occasionally involve different sources from one species.
  • Chimeric immunoglobulins, antibodies or peptides are typically produced using recombinant molecular and/or cellular techniques. Typically, chimeric antibodies have variable regions of both light and heavy chains that mimic the variable regions of antibodies derived from one mammalian species, while the constant portions are homologous to the sequences in antibodies derived from a second, different mammalian species.
  • The definition of chimeric antibody, however, is not limited to this example. A chimeric antibody is any antibody in which either or both of the heavy or light chains are composed of combinations of sequences mimicking the sequences in antibodies of different sources regardless of whether these sources are differing classes, differing antigen responses, or differing species of origin, and whether or not the fusion point is at the variable/constant boundary.
  • The terms “humanized,” “human-like” or “human-engineered” refers to an immunoglobulin wherein the constant regions have at least about 80% or greater homology to human immunoglobulin, and wherein some of the nonhuman (i.e. murine) variable region amino acid residues may be modified to contain amino acid residues of human origin.
  • Humanized antibodies may be referred to as “reshaped” antibodies. Manipulation of the complementarity-determining regions (CDRs) is one means of manufacturing humanized antibodies. See, e.g., Jones, et al. Replacing the Complementarity—Determining Regions in a Human Antibody With Those From a Mouse, Nature 321:522-525 (1988); Riechmann, et al. Reshaping Human Antibodies For Therapy, Nature 332, 323-327 (1988). For a review article concerning chimeric and humanized antibodies, see Winter and Milstein, Man-Made Antibodies, Nature 349, 293-299 (1991).
  • Preferably, immunoglobulins of the present invention are monoclonal antibodies (hereinafter referred to as “MoAbs”) of the IgM or IgG isotype of murine, human or other mammalian origin. Most preferably, the MoAb is reactive with the CD5 antigen found on both T and B cells. MoAbs of other animal species may be prepared using analogous non-human mammalian markers.
  • A variety of methods for producing MoAbs are known in the art. See, e.g., Goding, Monoclonal Antibodies; Principles and practice (2d ed., Academic Press 1986), which is incorporated herein by reference. Less preferred forms of immunoglobulins may be produced by methods well-known to those skilled in the art, such as by chromatographic purification of polyclonal sera to produce substantially monospecific antibody populations.
  • Monoclonal antibodies specifically directed against human CD5 antigen may be obtained by using combinations of immunogens and screening antigens which have only there human CD5 antigen in common or bay a screening assay designed to be specific for only anti-CD5 monoclonals. For example, production of monoclonal antibodies directed against CD5 may be accomplished by 1) immunization with human T cells expressing the CD5 antigen followed by screening of the resultant hybridomas for reactivity against a non-human cell line transfected with human CD5 (constructed in a manner similar to that described in Nishimura, et al., Eur. J. Immunol., 18:747-753 (1988)); 2) immunization with a non-human cell line transfected with human CD5 followed by screening of the resultant hybridomas for reactivity against a human T cell line expressing the CD5 antigen; 3) immunization with human or non-human cell lines expressing human CD5 followed by screening of the resultant hybridomas for ability to block reactivity of existing anti-CD5 monoclonals with a human T cell line; 4) immunization with human or non-human cell lines expressing human CD5 followed by screening of the resultant hybridomas for reactivity with purified native or recombinant CD5 antigen; or 5) immunization with a recombinant derivative of the human CD5 antigen followed by screening of the resultant hybridomas for reactivity against a human T cell line expressing CD5.
  • A preferred monoclonal antibody for use in this invention is produced by hybridoma cell line XMMLY-H65 (H65) deposited with the American Type Culture Collection in Rockville, Md. (A.T.C.C.) and given the Accession No. HB9286. A preferred antibody is prepared as disclosed herein using the humanized forms of the murine H65 antibody.
  • The generation of human MoAbs to a human antigen is also known in the art. See, e.g., Koda and Glassy, Hum. Antibod. Hybridomas, 1(1) 15-22 (1990). Generation of such MoAbs may be difficult with conventional techniques. Thus, it may be desirable to modify the antigen binding regions of the non-human antibodies, e.g., the F(ab′)z or hypervariable regions, to human constant regions (Fc) or framework regions by recombinant DNA techniques to produce substantially human molecules using general modification methods described in, for example, U.S. Pat. No. 4,816,397; and EP publications 173,494 and 239,400, which are incorporated herein by reference.
  • Alternatively, one may isolate DNA sequences which encode a human MoAb or portions thereof which specifically bind to the human T cell by screening a DNA library from human B cells according to the general protocols outlined by Huse et al., Science 246:1275-1281 (1989), Marks, et al., J. Mol. Biol. 222:581-597 (1991) which are incorporated herein by reference, and then cloning and amplifying the sequences which encode the antibody (or binding fragment) of the desired specificity.
  • In addition to the immunoglobulins specifically described herein, other “substantially homologous” modified immunoglobulins can be readily designed and manufactured utilizing various recombinant DNA techniques known to those skilled in the art. Modifications of the immunoglobulin genes may be readily accomplished by a variety of well-known techniques, such as site-directed mutagenesis. See, Gillman and Smith, Gene 8:81-97 (1979); Roberts, et al., Nature 328:731-734 (1987), both of which are incorporated herein by reference. Also, modifications which affect the binding affinity of the antibody may be selected using the general protocol outlined by McCafferty, et al., Nature 348:552-554 (1990), which is incorporated herein by reference.
  • In the present invention, an immunoglobulin, antibody, or peptide is specific for a T cell if it binds or is capable of binding T cells as determined by standard antibody-antigen or ligand-receptor assays. Examples of such assays include competitive assays, immunocytochemistry assays, saturation assays, or standard immunoassays such as ELISA, RIA and flow cytometric assays. This definition of specificity also applies to single heavy and/or light chains, CDRS, fusion proteins, or fragments of heavy and/or light chains, which bind T cells alone or are capable of binding T cells if properly incorporated into immunoglobulin conformation with complementary variable regions and constant regions as appropriate.
  • In some competition assays, the ability of an immunoglobulin, antibody, or peptide fragment to bind an antigen is determined by detecting the ability of the immunoglobulin, antibody, or peptide to compete with the binding of a compound known to bind the antigen. Numerous types of competitive assays are known and are discussed herein. Alternatively, assays which measure binding of a test compound in the absence of an inhibitor may also be used. For instance, the ability of a molecule or other compound to bind T cells can be detected by labelling the molecule of interest directly, or it may be unlabelled and detected indirectly using various sandwich assay formats. Numerous types of binding assays such as competitive binding assays are known. See, e.g., U.S. Pat. Nos. 3,376,110, 4,016,043; Harlow and Lane, Antibodies: A Laboratory Manual, Cold Spring Harbor Publications, N.Y. (1988), which are incorporated herein by reference.
  • Assays for measuring binding of a test compound to one component alone rather than using a competition assay are also available. For instance, immunoglobulins may be used to identify the presence of a T cell marker. Standard procedures for monoclonal antibody assays, such as ELISA, may be used see, Harlow and Lane, supra). For a review of various signal producing systems which may be used, see U.S. Pat. No. 4,391,904, which is incorporated herein by reference.
  • Other assay formats may involve the detection of the presence or absence of various physiological or chemical changes that result from an antigen-antibody interaction. See Receptor-Effector Coupling—A Practical Approach, (Hulme, ed., IRL Press, Oxford 1990), which is incorporated herein by reference.
  • Humanized antibodies of the present invention may be administered to patients having a disease having targetable cellular markers. Such disease include, but are not limited to, autoimmune diseases such as lupus (including systemic lupus erythematosus and lupus nephritis), scleroderma diseases (including lichen sclerosis, morphea and lichen planus), rheumatoid arthritis and the spondylarthropathies, thyroiditis, pemphigus vulgaris, diabetes mellitus type 1, progressive systemic sclerosis, aplastic anemia, myasthenia gravis, myositis including polymyositis and dermatomyositis, Sjogren's disease, collagen vascular disease, polyarteritis, inflammatory bowel disease (including Crohn's disease and ulcerative colitis), multiple sclerosis, psoriasis and primary biliary cirrhosis; diseases caused by viral infections; diseases caused by fungal infections; diseases caused by parasites; and the like.
  • Immunoglobulins, antibodies or peptides according to the invention may be administered to a patient either singly or in a cocktail containing two or more antibodies, other therapeutic agents, compositions, or the like, including, but not limited to, immunosuppressive agents, potentiators and side-effect relieving agents. Of particular interest are immunosuppressive agents useful in suppressing allergic or other undesired reactions of a host. Immunosuppressive agents include prednisone, prednisolone, dexamethasone, cyclophosphamide, cyclosporine, 6-mercaptopurine, methotrexate, azathioprine, and gamma globulin. All of these agents are administered in generally accepted efficacious dose ranges such as those disclosed in the Physician's Desk Reference, 41st Ed. (1987). In addition to immunosuppressive agents, other compounds such as an angiogenesis inhibitor may be administered with the anti-pan T immunoglobin. See Peacock, et al., Arthritis and Rheum. 35 (Suppl.), Abstract, for ACR meeting No. B141 (September 1992).
  • In a preferred embodiment of the present invention, anti-pan T cell immunoglobulins may be formulated into various preparations such as injectable and topical forms. Parenteral formulations are preferred for use in the invention, most preferred is intramuscular (i.m.) or intravenous (i.v.) administration. The formulations containing therapeutically effective amounts of anti-pan T cell antibodies are either sterile liquid solutions, liquid suspensions or lyophilized versions and optionally contain stabilizers or excipients. Lyophilized compositions are reconstituted with suitable diluents, e.g., water for injection, saline, 0.3% glycine and the like, at a level of from about 0.01 mg/kg of host body weight to about 10 mg/kg or more of host body weight.
  • Typically, the pharmaceutical compositions containing anti-pan T cell immunoglobulins are administered in a therapeutically effective dose in a range of from about 0.01 mg/kg to about 5 mg/kg body weight of the treated animal. A preferred dose range of the anti-pan T cell antibody is from about 0.05 mg/kg to about 2 mg/kg body weight of the treated animal. The immunoglobulin dose is administered over either a single day or several days by daily intravenous infusion. For example, for a patient weighing 70 kg, about 0.7 mg to about 700 mg per day is a preferred dose. A more preferred dose is from about 3.5 mg to about 140 mg per day.
  • Anti-pan T cell immunoglobulin may be administered systemically by injection intramuscularly, subcutaneously, intrathecally, intraperitoneally, into vascular spaces, or into joints (e.g., intraarticular injection at a dosage of greater than about 1 μg/cc joint fluid/day). The dose will be dependent upon the properties of the anti-pan T cell immunoglobulin employed, e.g., its activity and biological half-life, the concentration of anti-pan T cell antibody in the formulation, the site and rate of dosage, the clinical tolerance of the patient involved, the autoimmune disease afflicting the patient and the like as is well within the knowledge of the skilled artisan.
  • The anti-pan T cell immunoglobulin of the present invention may be administered in solution. The pH of the solution should be in the range of about pH 5.0 to about 9.5, preferably pH 6.5 to 7.5. The anti-pan T cell immunoglobulin or derivatives thereof should be in a solution having a pharmaceutically acceptable buffer, such as phosphate, tris (hydroxymethyl) aminomethane-HCl, or citrate and the like. Buffer concentrations should be in the range from about 1 to about 100 mM. A solution containing anti-pan T cell immunoglobulin may also contain a salt, such as sodium chloride or potassium chloride in a concentration from about 50 to about 150 mM. An effective amount of a stabilizing agent such as albumin, a globulin, a detergent, a gelatin, a protamine, or a salt of protamine may also be included and may be added to a solution containing anti-pan T cell immunoglobulin or to the composition from which the solution is prepared systemic administration of anti-pan T cell immunoglobulin is typically made every two to three days or once a week if a chimeric or humanized form is used. Alternatively, daily administration is useful. Usually administration is by either intramuscular injection or intravascular infusion.
  • Alternatively, anti-pan T cell immunoglobulin is formulated into topical preparations for local therapy by including a therapeutically effective concentration of anti-pan T cell immunoglobulin in a dermatological vehicle. Topical preparations may be useful to treat skin lesions such as psoriasis and dermatitis associated with lupus. The amount of anti-pan T cell immunoglobulin to be administered, and the anti-pan T cell immunoglobulin concentration in the topical formulations, will depend upon the vehicle selected, the clinical condition of the patient, the systemic toxicity and the stability of the anti-pan T cell immunoglobulin in the formulation. Thus, the physician will necessarily employ the appropriate preparation containing the appropriate concentration of anti-pan T cell immunoglobulin in the formulation, as well as the amount of formulation administered depending upon clinical experience with the patient in question or with similar patients.
  • The concentration of anti-pan T cell immunoglobulin for topical formulations is in the range from about 0.1 mg/ml to about 25 mg/ml. Typically, the concentration of anti-pan T cell immunoglobulin for topical formulations is in the range from about 1 mg/ml to about 20 mg/ml. Solid dispersions of anti-pan T cell immunoglobulin as well as solubilized preparations may be used. Thus, the precise concentration to be used in the vehicle may be subject to modest experimental manipulation in order to optimize the therapeutic response. Greater than about 10 mg of anti-pan T cell immunoglobulin/100 grams of vehicle may be useful with 1% w/w hydrogel vehicles in the treatment of skin inflammation. Suitable vehicles, in addition to gels, are oil-in-water or water-in-oil emulsions using mineral oils, petrolatum, and the like.
  • Anti-pan T cell immunoglobulin may be optionally administered topically by the use of a transdermal therapeutic system (Barry, Dermatological Formulations, p. 181 (1983)). While such topical delivery systems have been designed largely for transdermal administration of low molecular weight drugs, by definition they are capable of percutaneous delivery. They may be readily adapted to administration of anti-pan T cell immunoglobulin or derivatives thereof and associated therapeutic proteins by appropriate selection of the rate-controlling microporous membrane.
  • Preparations of anti-pan T cell immunoglobulin either for systemic or local delivery may be employed and may contain excipients as described above for parenteral administration and other excipients used in a topical preparation such as cosolvents, surfactants, oils, humectants, emollients, preservatives, stabilizers and antioxidants. Any pharmacologically acceptable buffer may be used, e.g., tris or phosphate buffers.
  • Administration may also be intranasal or by other nonparenteral routes. Anti-pan T cell immunoglobulin may also be administered via microspheres, liposomes or other microparticulate delivery systems placed in certain tissues including blood.
  • Anti-pan T cell immunoglobulin may also be administered by aerosol to achieve localized delivery to the lungs. This is accomplished by preparing an aqueous aerosol or liposomal preparation. A nonaqueous (e.g., fluorocarbon propellent) suspension may be used. Sonic nebulizers preferably are used in preparing aerosols. Sonic nebulizers minimize exposing the anti-pan T cell antibody or derivatives thereof to shear, which can result in degradation of anti-pan T cell immunoglobulin.
  • Ordinarily, an aqueous aerosol is made by formulating an aqueous solution or suspension of anti-pan T cell immunoglobulin together with conventional pharmaceutically acceptable carriers and stabilizers. The carriers and stabilizers will vary depending upon the requirements for the particular anti-pan T cell immunoglobulin, but typically include nonionic surfactants (Tweens, Pluronics, or polyethylene glycol), innocuous proteins such as serum albumin, sorbitan esters, oleic acid, lecithin, amino acids such as glycine, buffers, salts, sugars, or sugar alcohols. The formulations are sterile. Aerosols generally may be prepared from isotonic solutions.
  • Each of the foregoing methods are illustrated by way of the following examples, which are not to be construed as limiting the invention. All references cited herein are incorporated by reference.
  • EXAMPLES Example 1
  • A. Identification of Low Risk Residues in a Mouse Variable Domain
  • A method of the present invention was utilized to prepare modified antibody variable domains by identifying low risk residues in a mouse monoclonal antibody variable domain, designated H65, which may be modified without diminishing the native affinity of the domain for antigen while still reducing its immunogenicity with respect to humans.
  • The light and heavy chains of the variable domain of H65 were determined to most closely resemble the consensus sequences of subgroup 1 (“hK1”) of the human kappa chains and subgroup 3 (“hH3”) of the human heavy chains, respectively. The H65 V/J-segments of the light and heavy chain sequences are aligned with the two human subgroup consensus sequences in FIGS. 6A and 6B. The H65 sequences are also contained in SEQ ID Nos. 26 and 28.
  • In FIGS. 6A and 6B, upper and lower case letters denote the degree of conservation at any given position. For example, an “A” indicates that alanine is present at that position in about 90% to about 100% of the known human sequences of that subgroup (excluding small, incomplete fragments); whereas an “a” indicates that alanine is present only about 50% to about 90% of the time at that position in known human sequences of that subgroup. A lower case “x” indicates conservation of the amino acid at that position less than about 50% of the time.
  • The line labelled “bind” shows which residues directly affect (−) or do not directly affect (+) antigen binding of CDR loops. The “bury” line indicates exposed (+), buried (−), or interfacial (=) residues. On either the “bind” or “bury” line, a “0” indicates a residue of intermediate significance in terms of antigen binding or placement of the residue, respectively.
  • FIGS. 6A and 6B reveal that the mouse H65 sequences differ from the human consensus sequences with which they are aligned at a total of 94 positions. Sixty-nine of these differences occur at moderate-risk (15 positions) or high risk (54 positions) positions suggesting that the mouse residue at that position may be important for the function of the antibody. The “M/H” line of FIG. 6 specifically indicates which positions differ between the two pairs of aligned sequences. Based on the considerations of the level of risk and the degree of conservation of the human residue at each position presented in the foregoing paragraphs, those residues in the H65 sequences designated M or m in the M/H line are identified as residues to be kept “mouse” in a humanized sequence, while those designated H or h are identified as residues to be changed to “human.”
  • Twenty-five differences occur at low risk positions at which the mouse and human sequences differ. At thirteen of those positions (designated “H” on the M/H lines of FIG. 6) the mouse residue aligns with a human consensus amino acid which is highly conserved. Therefore, the mouse residue at that position is identified as one to be changed to the conserved human residue.
  • At four low risk positions (designated “m”) in which the mouse and the human sequences differ, the mouse residue aligns with a human consensus amino acid which is moderately conserved. However, since the mouse residue is found at that position in other actual sequences of human antibodies (in Kabat's sequences of Proteins of Immunoglobulin Interest), the positions are identified as ones to be kept “mouse.” At seven low risk positions (designated “h”), the mouse residue aligns with a human consensus amino acid which is moderately conserved but the mouse residue is not found at that position in an actual human antibody sequence in the Kabat book. Therefore, those positions are identified as ones to be changed to “human.”
  • At one low risk position (designated “m”) in which the mouse and human sequences differ, the mouse residue aligns with a human consensus amino acid which is poorly conserved. Therefore, that position is identified as one to be kept “mouse.”
  • The “prop” lines of FIG. 6 set out the sequences of the light and heavy chains of the H65 antibody variable domain in which the residues identified by the methods of the present invention as those which may be modified without diminishing the native affinity of the H65 variable domain for CD5 are changed to human residues. Thus, the “prop” lines of FIGS. 6A and 6B set out the amino acid sequences of humanized light (SEQ ID NO: 27) and heavy chains (SEQ ID NO: 29) of the H65 antibody variable domain.
  • Example 2
  • A. Synthesis of H65 V/J Segments of Light and Heavy Chain
  • Based on the low risk humanized amino acid sequences of the V/J-segments of the light and heavy chains of the H65 antibody variable domain described in Example 1, synthetic genes for heavy and light chain V/J-segments of H65 were synthesized. The humanized amino acid sequences were reverse-translated with the PCGENE package (Intelligenetics, Mountain View, Calif.). Amino acid codons for each position were chosen which were identical to the mouse codon at positions where the mouse amino acid residue was maintained, or which matched as closely as possible a codon in a native antibody gene based on those gene sequences published in Kabat et al, supra. For expression of humanized whole antibody in mammalian cells, polynucleotides encoding the native mouse leader sequences were included as part of the humanized genes. Each gene, heavy or light, was assembled from six overlapping oligonucleotides and amplified by PCR. Each oligonucleotide was synthesized with a Cyclone Model 8400 DNA Synthesizer (Milligen/Biosearch, Burlington, Mass.). Restriction sites were introduced into the amplified DNA segments for cloning into the final expression vectors for antibody genes (heavy or light). A SalI restriction site was introduced into each V-region upstream of the initiation codon, ATG. A BstEII restriction site was introduced into the 3′-end of the heavy chain J-region, while a HindIII site was introduced into the 3′-end of the light chain J-region.
  • B. Construction of the Gene Encoding the Humanized H65 Heavy Chain Variable Region
  • The humanized V- and J-segments of the heavy chain were assembled from six oligonucleotides, HUH-G1, HUH-G2, HUH-G3, HUH-G4, HUH-G5, and HUH-G6, the sequences of which are contained in FIG. 7B and in SEQ ID Nos. 36 to 41, respectively. The oligonucleotides were amplified with PCR primers H65G-2S and H65-G2 (SEQ ID Nos. 42 and 43, respectively). Oligonucleotides greater than 50 bp in length were purified on a 15% polyacrylamide gel in the presence of 25% urea. DNA strand extension and DNA amplification was accomplished with a Taq polymerase and the GeneAmp Kit used according to the manufacturer's instructions (Perkin-Elmer Cetus, Germany). Oligonucleotides containing the synthetic humanized antibody gene were mixed in pairs (HUH-G1+HUH-G2, HUH-G3+HUH-G4, and HUH-G5+HUH-G6) in 100 μl reactions with 1 μg of each DNA, 2.5 U Taq polymerase, 50 mM KCl, 10 mm TRIS-CL pa 8.3, 1.5 mM MgCl2, and 200 uM each dNTP. The tube was incubated in a Coy TempCycler for 1 minute at 94° C., 2 minutes at 55° C. and 20 minutes at 72° C. A portion of each reaction product (40 μl) was mixed in pairs (HUH-G1,2+HUH-G3,4; HUH-G3,4+HUH-G5,6), 2.5 U Taq was added and the tubes were re-incubated at 94° C. for 1 minute, 55° C. for 2 minutes and 72° C. for 20 minutes. The heavy chain gene was then assembled by mixing an equal amount of the HUH-G1,2,3,4 reaction product with the HUH-G3,4,5,6 reaction product and bringing the volume to 100 μl of 2.5 U Taq, 50 mM KCl, 10 mM TRIS-CL pH 8.3, 1.5 mM MgCl2, 200 uM each dNTP, and 0.5 μg of each amplification primer H65G-2S and H65-G2. The reaction was overlaid with mineral oil, and the cycle profile used for amplification was: denaturation 94° C. for 1 minute, annealing 55° C. for 2 minutes, and primer extension at 72° C. for 3 minutes. Primer extension was carried out for 30 cycles. The DNA sequence of the assembled V/J-region is contained in FIG. 8A and in SEQ ID NO: 46. The assembled V/J-region was cut with SalI and BstEII, purified by electrophoresis on an agarose gel, and assembled into a heavy chain expression vector, pING4612, which is similar to that described for heavy chain expression in Robinson et al., Hum. Antib. Hybridomas, 2, 84 (1991) and described in detail in co-pending, co-owned U.S. patent application Ser. No. 07/659,409 filed on Sep. 6, 1989, which is incorporated herein by reference.
  • C. Construction of the Gene Encoding the Humanized H65 Light Chain Variable Region
  • The humanized V- and J-segments of the light chain were also assembled from six oligonucleotides, $H65K-1, HUH-K1, HUH-K2, HUH-K3, HUH-K4 and HUH-K5, the sequences of which are contained in FIG. 7 and in SEQ ID NOs. 30 to 35, respectively. The oligonucleotides were amplified with PCR primers H65K-2S and JK1-HindIII (SEQ ID NOs. 44 and 45, respectively). Oligonucleotides containing the synthetic humanized antibody gene were mixed in pairs ($H65K-K1+HUH-K1, HUH-K2+HUH-K3, and HUH-K4+HUH-K5) and incubated as described above for the heavy chain. A portion of each reaction product (40 μl) was mixed in pairs ($H65K-1/HUH-K1+HUH-K2,3; HUH-K2,3+HUH-K4,5) and treated as above. The light chain gene was then assembled by amplifying the full length gene with PCR primers H65K-2S and JK1-HindIII as outlined above for the heavy chain. The DNA sequence of the assembled V/J-region is contained in FIG. 8B and in SEQ ID NO. 47. The assembled V/J-region was cut with SalI and HindIII, purified by electrophoresis on an agarose gel, and assembled into a light chain antibody expression vector, pING4614 similar to those described for light chain expression in Robinson et al., supra. and in U.S. patent application Ser. No. 07/659,409, supra.
  • D. Transient Expression of Humanized H65
  • Expression vectors containing the humanized H65 light chain and heavy chain sequences under the control of the Abelson Leukemia virus LTR promoter (described in Robinson et al., supra, and in U.S. patent application Ser. No. 07/659,409, supra) and 3′ untranslated regions from human gamma-1 (for heavy chain) and mouse kappa (for light chain) were transfected by lipofection into a CHO-K1 strain which expresses the SV40 T antigen. Following treatment with lipofection reagent (Bethesda Research Labs, Gaithersburg, Md.) plus DNA for 5 hours at 37° C., Ham's F12 media containing fetal bovine serum (FBS, final FBS conc.=10%) was added and the cells were incubated for an additional 48 hours. Following this incubation period, the FBS-supplemented media was removed and replaced with serum-free media (EB-CHO) (Irvine Scientific, Irvine, Calif.) and the cells were incubated for an additional 7 days. As a control, the CHO-K1 cells were also transfected with chimeric H65 light chain and heavy chain (each consisting of unmodified mouse V/J-segments fused to a human C-segment) in expression vectors similar to those described above. Following incubation, the supernatants were collected and tested by ELISA for the presence of secreted IgG. All of the supernatants contained about 0.03-0.06 μg/ml IgG.
  • Example 3
  • The H65 antibody modified according to the methods of the present invention was tested to determine whether it retained native affinity for antigen. Its binding capability was compared to that of a chimeric H65 IgG antibody (consisting of the chimeric H65 light chain and heavy chain described in Example 2) which has the same affinity for CD5 as unmodified H65 mouse antibody.
  • A. Preparation of Humanized and Chimeric H65 IaG for Competition Binding
  • The humanized H65 (hH65) and chimeric H65 IgG (cH65) from transient transfections described above were concentrated from 4 ml to a final volume of 100 μl by centrifugation using a Centricon 30 (Amicon, Amicon Division of W.R. Grace and Co., Beverley, Mass.) at 4° C. Both hH65 and cH65 concentrates were then washed once with 1.0 ml of phosphate buffered saline (PBS), pH 7.2 and reconcentrated to approximately 100 μl. As a control, HB-CHO culture media alone (CM) or media supplemented with purified cH65 (CM+cH65) was concentrated in a similar manner. The final concentrations of hH65 and cH65 were determined by ELISA (anti-human Kappa pre-coat, peroxidase-labelled anti-human gamma for detection) using chimeric IgG as a standard.
  • B. Radiolabelling of cH65 IaG
  • 20 μg of purified CH65 IgG was iodinated (1 mCi of Na125I, Amersham, Arlington Heights, Ill.) using lactoperoxidase beads (Enzymobeads, BioRad Laboratories, Richmond, Calif.) in PBS. Iodination was allowed to proceed for 45 minutes at 23° C. 125I-cH65 IgG was purified from unbound 125I by gel filtration using a Sephadex G-25-80 column. Concentration and specific activity was determined by measuring the TCA-precipitated counts before and after purification.
  • C. Competitive Binding of hH65 for cH65 IaG
  • Molt4-M cells, which express CD5 on their surface, were plated into 96 well V-bottom plates at a density of 3×105 cells per well and pelleted by centrifugation. The medium was decanted, and 100 μl of purified cH65 IgG at final concentrations from 200 nM to 0.0017 nM (diluted in 3-fold steps) in “BHD” [DMEM (Dulbecco's Modified Eagle's Medium)+It BSA+10 mM Hepes, pH 7.2] (BHD) was added to each well, followed by 100 μl of 125I-cH65 IgG (final concentration 0.1 nM) in BHD. For single point determinations, 50-100 μl of the Centricon® concentrates were added to the wells as follows: hH65 (final concentration=0.54 nM), cH65 (final concentration=0.22 nM), CM+purified cH65 IgG (final concentration=30 nM) and CM alone. These were followed by addition of 125I-cH65 IgG (final concentration=0.1 nM). Binding was allowed to proceed for 5 hours at 4° C. At the end of 5 hours, binding was terminated by three washes with ice cold BHD using centrifugation to pellet cells. Radioactivity was determined by solubilizing bound 125I-cH65 IgG with 1N NaOH and counting in a Beckman Gamma 8000 (Beckman Instruments, Fullerton, Calif.).
  • Purified cH65 IgG effectively displaced 125I-cH65 IgG binding with an ED50 of approximately 1.0 nM as shown in FIG. 9, wherein open circles indicate cH65, shaded squares indicate hH65 and shaded triangles indicate CM+purified cH65. The hH65 was as effective in displacing 125I-cH65 IgG as were purified cH65 and CM+purified cH65 IgG, at their respective concentrations. No competition was observed with CM as expected. These results demonstrate that the low-risk changes made in the course of modification of hH65 did not diminish the binding affinity of this antibody for the CD5 antigen.
  • Example 4
  • The method of the present invention for preparing modified antibody variable domains by identifying modifiable amino acids was applied to the anti-TAC antibody variable domain sequence [SEQ ID Nos. 49 (light chain) and 53 (heavy chain)] and the resulting modified sequence is compared to the humanized anti-TAC antibody sequence [SEQ ID Nos. 51 (light chain) and 55 (heavy chain)] described in Queen et al., supra.
  • The results are shown in FIGS. 10A and 10B. The sequence modified according to the present invention [SEQ ID Nos. 50 (light chain) and 54 (heavy chain)] is shown on the lines labelled “prop,” and the Queen humanized sequence is shown on lines labelled “Que.” Modifications to the Queen humanized sequence were based on the human EU antibody sequence [SEQ ID Nos. 48 (light chain) and 52 (heavy chain)]. The comparison reveals many differences between the proposed sequence generated by the methods of the present invention and the Queen humanized sequence. The differences which are the most likely to affect binding activity of their humanized antibody are positions 4 (L vs. M), 15 (P vs. V), 36 (F vs. Y), 47 (W vs. L), 71 (Y vs. F), and 80 (A vs. P) in the light chain, as well as position 69 (L vs. I) in the heavy chain.
  • Example 5
  • Active Modified Antibodies May Be Evolved Toward Human
  • If it is desirable to humanize an antibody variable domain beyond the changes identified above, further, higher-risk changes may be made to evolve the domain.
  • Higher-risk residues may be changed in a round of mutagenesis subsequent to the low risk changes, in smaller groups, so that deleterious mutations may be identified quickly and corrected before binding activity is abolished. (Low risk changes can be made all at once, with little fear of abolishing activity.)
  • For example, because in the three-dimensional model of each subunit, framework 1 and framework 3 (F1 and F3 in FIGS. 2 and 3) form semi-independent loops on the surface of the subunit, the moderate or high risk mutations may therefore be divided into four groups (consisting of F1 and F3 in the light subunit and F1 and F3 in the heavy subunit). Four different constructs may be made, each containing higher-risk “human” mutations in only one framework region with the other three frameworks left completely “mouse,” and assayed for activity. This technique avoids the dilemma raised by other humanization methods in which all higher-risk changes are made at once, making it difficult to determine which of the many amino acid changes is responsible for affecting antigen-binding activity. The creation of antibodies according to the invention which possess moderate risk changes are described below.
  • Example 6
  • Identification of Moderate Risk Residues in Mouse Variable Domain
  • The human consensus sequences in which moderate risk residues are converted from mouse residues to human residues are represented in FIGS. 16A and 16B as lines labelled hxl (i.e., subgroup 1 of the human kappa chain) and hH3 (i.e., subgroup 3 of the human heavy chain). Symbols in this Figure, for conservation and for risk are used in accordance with FIGS. 6A and 6B.
  • In the line labelled “mod”, a dot (.) represents a residue which may be mutated from “mouse” to “human” at moderate risk. There are 29 such moderate risk positions.
  • The mouse residue matches the human consensus residue more than 50% of the time at 131 positions (102 positions match 90%-100% and 29 positions match 50% to 90%). These positions were not changed.
  • The lines labelled M/H in FIGS. 16A and 16B indicate the 91 positions which differed significantly between the mouse and human sequences (i.e., where the human sequences have the mouse residue less than 50% of the time). Moderate risk positions, designated m in the M/H line, were kept “mouse”; whereas those designated H or h were changed to human. The 25 low risk positions which were already human-like or which were previously humanized (as described supra in Example 1) are designated “{circumflex over ( )}” in the M/H line. Finally, the 54 high risk positions in which the mouse and human residues did not match are designated M and are kept “mouse”.
  • Fifteen differences occur at moderate-risk positions at which the mouse and human sequences differ. At ten of those positions (designated “H” on the M/H lines of FIG. 6) the mouse residue aligns with a human consensus amino acid which is highly conserved. Therefore, the mouse residue at that position is identified as one to be changed to the conserved human residue.
  • At moderate risk positions (designated “m”) in which the mouse and the human sequences differ, the mouse residue aligns with a human consensus amino acid which is moderately conserved. However, since the mouse residue is found at that position in other actual sequences of human antibodies (in Kabat's sequences of Proteins of Immunoglobulin Interest, the positions are identified as ones to be kept “mouse.” Although there are no such positions in this particular sequence, such positions may occur in other antibodies.
  • At four moderate risk positions (designated “h”), the mouse residue aligns with a human consensus amino acid which is moderately conserved but the mouse residue is not found at that position in an actual human antibody sequence in Kabat, et al. Sequences of Proteins of Immunoglobulin Interest, supra. Therefore, that position is identified as ones to be changed to “human.”
  • At one moderate risk position (designated “m”) in which the mouse and human sequences differ, the mouse residue aligns with a human consensus amino acid which is poorly conserved. Therefore, that position is identified as one to be kept “mouse.”
  • The humanized H65 heavy chain containing the moderate risk residues was assembled by a strategy similar to that for the low risk residues. The moderate-risk expression vector was assembled from intermediate vectors. The six oligonucleotide sequences (oligos), disclosed in FIG. 7B and labelled HUH-G11, HUH-G12, HUH-G3, HUH-G4, HUH-G5, and HUH-G6 (the sequences of HUH-G11 and HUH-G12 are set out in SEQ ID Nos. 56 and 57) were assembled by PCR. Oligonucleotides containing the synthetic humanized antibody gene were mixed in pairs (HUH-GIl+HUH-G12, HUH-G3+HUH-G4, and HUH-G5+HUH-G6) in a 100 μl reaction with 1 μg of each DNA and filled in as described above. A portion of each reaction product was mixed in pairs (HUH-G11, 12+HUH-G3, 4; HUH-G3, 4+HUH-G5, 6), 2.5 U Taq was added and samples were reincubated as described above. The in-J-region was assembled by mixing equal amounts of the HUH-Gil, 12, 3, 4 reaction product with the HUH-G3, 4, 5, 6 product, followed by PCR with 0.5 ug of primers H65G-2S and H65-G2 as described above. The reaction product was cut with SalI and BstEII and cloned into the expression vector, similar to that described for heavy chain in Robinson et al., Hum. Antibod. Hybridomas 2:84 (1991), generating pING4617. That plasmid was sequenced with Sequenase (USB, Cleveland), revealing that two residues were altered (a G-A at position 288 and a A-T at position 312, numbered from the beginning of the leader sequence). The correct variable region was restored by substitution of this region from pING4612, generating the expected V-region sequence in pING4619.
  • An intermediate vector containing the other moderate-risk changes was constructed by PCR assembly of the oligos HUH-G13, HUH-G14, HUH-G15, and HUH-G16 (FIG. 7A and SEQ ID Nos: 58-61). Oligos HUH-G13+HUH-G14 and HUH-G15+HUH-G16 were mixed and filled in with Vent polymerase (New England Biotabs) in a reaction containing 10 mM KCl, 20 mM TRIS pH 8.8, 10 mM (NH4)2SO2, 2 mM MgSO4, 0.1% Triton X-100, 100 ng/ml BSA, 200 uM of each dNTP, and 2 units of Vent polymerase in a total volume of 100 μl. The reaction mix was incubated at 94° C. for 1 minute, followed by 2 minutes at 50° C. and 20 minutes at 72° C. The reaction products (40 μl) were mixed and amplified with the oligonucleotides H65-G13 and H65-G2 with Vent polymerase in the same reaction buffer and amplified for 25 cycles with denaturation at 94° C. for 1 minute, annealing at 50° C. for 2 minutes and polymerization at 72° C. for 3 minutes. The reaction product was treated with T4 polymerase and then digested with AccI. The 274 base pair (bp) fragment was purified on an agarose gel and ligated along with the 141 bp SalI to AccI fragment from pING4619 into pUC18 cut with SalI and SmaI to generate pING4620. pING4620 contains the entire signal sequence, V-region, and J-region of the moderate-risk H65 heavy chain.
  • The final expression vector for the moderate-risk H65 heavy chain, pING4621, was assembled by cloning the SalI to BstEII fragment from pING4620 into the same expression vector described above.
  • Example 7
  • A. Assembly of Moderate-Risk Light Chain
  • The moderate-risk humanized V- and J-segments of the light chain were assembled from six oligonucleotides, $H65K-1, HUH-K7, HUH-K6, HUH-K8, HUH-K4 and HUH-K5. The sequences of HUH-K7, HUH-K6 and HUH-K5 are set out in SEQ ID Nos. 62-64 and FIGS. 7 and 7A, respectively. The oligonucleotides were amplified with PCR primers H65K-2S and JK1-HindIII. Oligonucleotides containing the synthetic humanized antibody gene were mixed in pairs ($H65-K1+HUH-K7, HUH-K6+HUH-K4+HUH-K5) and incubated with Vent polymerase as described for the moderate-risk heavy chain. A portion of each reaction product (40 ul) was mixed in pairs ($H65H-K1/HUH-K7+HUH-K6, 8; HUH-K6, 8+HUH-K4, 5) and filled in as above. The light chain gene was then assembled by amplifying the full length gene with the PCR primers H65K-2S and JK1-HindIII with Vent polymerase for 25 cycles as outlined above. The assembled V/J region was cut with SaLI and HindIII, purified by electrophoresis on an agarose gel, and assembled into a light chain antibody expression vector, pING4630.
  • B. Stable Transfection of Mouse Lymphoid Cells for the Production of He3 Antibody
  • The cell line Sp2/0 (American Type Culture Collection #CRL1581) was grown in Dulbecco's Modified Eagle Medium plus 4.5 g/l glucose (DMEM, Gibco) plus 10% fetal bovine serum. Media were supplemented with glutamine/penicillin/streptomycin (Irvine Scientific, Irvine, Calif.).
  • The electroporation method of Potter, H., et al., Proc. Natl. Acad. Sci., USA, 81:7161 (1984) was used. After transfection, cells were allowed to recover in complete DMEM for 24-48 hours, and then seeded at 10,000 to 50,000 cells per well in 96-well culture plates in the presence of selective medium. Histidinol (Sigma) selection was at 1.71 ug/ml, and mycophenolic acid (Calbiochem) was at 6 ug/ml plus 0.25 mg/ml xanthine (Sigma). The electroporation technique gave a transfection frequency of 1-10×10−5 for the Sp2/0 cells.
  • The He3 light chain expression plasmid pING4630 was linearized by digestion with PvuI restriction endonuclease and transfected into Sp2/0 cells, giving mycophenolic acid—resistant clones which were screened for light chain synthesis. The best 4 light chain—producing transfectants after outgrowth were pooled into 2 groups of 2 transfectants/pool and each pool was transfected with the He3 heavy chain expression plasmid, pING4621, that had been linearized with PvuI. After selection with histidinol, the clone producing the most light plus heavy chain, Sp2/0-4630+4621 Clone C1718, secreted antibody at approximately 22 μg/ul in the presence of 10−7 in dexamethasone in an overgrown culture in a T25 flask. This transfectoma has been deposited with the American Type Culture Collection, 1230 Parklawn Drive, Rockville, Md., 20852 on Dec. 1, 1992 as ATCC HB 11206.
  • C. Purification of He3 Antibody Secreted in Tissue Culture
  • Sp2/0-4630+4621 cells are grown in culture medium HB101 (Hana Biologics)+1% Fetal Bovine Serum, supplemented with 10 mM HEPES, 1×Glutamine-Pen-Strep (Irvine Scientific #9316). The spent medium is centrifuged at about 5,000×g for 20 minutes. The antibody level is measured by ELISA. Approximately 200 ml of cell culture supernatant is loaded onto a 2 ml Protein A-column (Sigma Chemicals), equiliberated with PBS (buffer 0.15 M NaCl, 5 mM sodium phosphate, 1 mM potassium phosphate, buffer pH 7.2). The He3 antibody is eluted with a step pH gradient (pH 5.5, 4.5 and 2.5). A fraction containing He3 antibody (9% yield) but not bovine antibody, is neutralized with 1 M Tris pH 8.5, and then concentrated 10-fold by Centrium 30 (Amicon)-diluted 10-fold with PBS, reconcentrated 10-fold by Centricon 30, diluted 10-fold with PBS, and finally reconcentrated 10-fold. The antibody was stored in 0.25 ml aliquots at −20° C.
  • D. Affinity Measurements of He3 IgG for CD5
  • The affinity of He3 for CD5 was determined using Molt-4M cells, which express CD5 on their surface and I125-labeled chimeric H65 IgG in a competitive binding assay.
  • For this assay, 20 μg of chimeric H65 IgG (cH65 IgG) was iodinated by exposure to 100 μl lactoperoxidase-glucose oxidase immobilized beads (Enzymobeads, BioRad), 100 μl of PBS, 1.0 mCi I125 (Amersham, IMS30), 50 μl of 55 mM b-D-glucose for 45 minutes at 23° C. The reaction was quenched by the addition of 20 μl of 105 mM sodium metabisulfite and 120 mM potassium iodine followed by centrifugation for 1 minute to pellet the beads. 125-cH65 IgG was purified by gel filtration using 7 mls of sephadex G25, using PBS (137 mM NaCl, 1.47 mM KH2PO4, 8.1 mM Na2HPO4, 2.68 mM KCl at pH 7.2-7.4) plus 0.1% BSA. 125I-cH65 IgG recovery and specific activity were determined by TCA precipitation.
  • Competitive binding was performed as follows: 100 μl of Molt-4M cells were washed two times in ice cold DHB binding buffer (Dubellco's modified Eagle's medium (Gibco, 320-1965PJ), 1.0% BSA and 10 mM Hepes at pH 7.2.-7.4). Cells were resuspended in the same buffer, plated into 96 v-bottomed wells (Costar) at 3×105 cells per well and pelleted at 4° C. by centrifugation for 5 min at 1,000 rpm using a Beckman J S 4.2 rotor; 50 μl of 2×—concentrated 0.1 nM 125I-cH65 IgG in DHB was then added to each well and competed with 50 μl of 2×—concentrated cH65 IgG or humanized antibody in DHB at final antibody concentrations from 100 nM to 0.0017 nM. Humanized antibody was obtained from culture supernatants of Sp2/0 clone C1718 which expresses He3 IgG. The concentration of the antibody in the supernatants was established by ELISA using a chimeric antibody as a standard. Binding was allowed to proceed at 4° C. for 5 hrs and was terminated by washing cells three times with 200 μl of DHB binding buffer by centrifugation for 5 min at 1,000 rpm. All buffers and operations were at 4° C. Radioactivity was determined by solubilizing cells in 100 μl of 1.0 M NaOH and counting in a Cobra II auto gamma counter (Packard). Data from binding experiments were analyzed by the weighted nonlinear least squares curve fitting program, MacLigand, a Macintosh version of the computer program “Ligand” from Munson, Analyt. Biochem., 107:220 (1980). Objective statistical criteria (F, test, extra sum squares principle) were used to evaluate goodness of fit and for discriminating between models. Nonspecific binding was treated as a parameter subject to error and was fitted simultaneously with other parameters.
  • The results of the competition binding assay are provided in FIG. 11. These results demonstrate that the moderate-risk changes made in He3 IgG result in an antibody with a higher affinity than the chimeric mouse-human form of this antibody (cH65) for its target CD5. In this particular case, moderate risk changes appear to increase affinity slightly, but a decrease may be expected in most cases.
  • Example 8
  • Preparation of XMMLY-H65 Anti-pan T Cell Immunoglobulin
  • The murine monoclonal antibody produced by cell line XMMLY-H65 (MoAbH65) is reactive with the human CD5 antigen. The cell line XMMLY-H65 was deposited with the American Type Culture Collection, 12301 Parklawn Drive, Rockville, Md., 20852 and designated Accession No. EB9286.
  • MoAb H65 was produced after immunization of BALB/c mice with the human T-cell line HSB-2 originally isolated from a patient with T-cell acute lymphocytic leukemia. Adams, et al. Can. Res. 28:1121 (1968). The murine myeloma cell line P3 7 NS/1-Ag-1-4 of Kohler et al. Ernr. J. Immunol. 6:292 (1976) was fused with spleen cells from an immunized mouse by the technique of Galfre et al., Nature 266:550 (1977). One of the resulting hybrid colonies was found to secrete a MoAb that recognizes a pan-T-lymphocyte antigen with a molecular weight of 67 kD, expressed on approximately 95% of peripheral T-lymphocytes [Knowles, Leukocyte Typing II, 1, (E. Reinherz, et al. eds., Springer Verlag (1986)]. This antigen is not present on the surface of any other hematopoietic cells, and the antibody itself has been tested for binding to a large range of normal human tissues and found to be negative for all cells except for T-lymphocytes and a subpopulation of B lymphocytes.
  • The H65 antibody-producing hybrid cell line was cloned twice by limiting dilution and was grown as ascites tumors in BALB/c mice.
  • MoAb H65 was purified from mouse ascites by a modification of the method of Ey et al. Immunochem. 15:429 (1978). In brief, the thawed mouse ascites was filtered to remove lipid-like materials and was diluted with 2 to 3 volumes of 0.14 M NaPO4, pH 8.0, before application onto an immobilized protein A-Sepharose column of appropriate size. The unbound materials were removed from the column by washing with 0.14 M NaPO4, pH 8.0, until no further change in absorbance at 280 nm was seen. A series of column washes with 0.1 M sodium citrate (pH 6.0, pH 5.0, pH 4.0, and pH 3.0) were then performed to elute bound antibody.
  • Peak fractions were pooled, adjusted to pH 7.0 with saturated Tris base, and concentrated by using a cell stirred with Amicon YM10 membrane (Amicon, Lexington, N.Y.). An antibody solution was then dialyzed against phosphate-buffered saline (PBS), pH 7.0, and was stored frozen at −70° C.
  • MoAb H65 is of the IgG1 subclass, as determined by double diffusion in agar with the use of subclass-specific antisera (Miles-Yeda, Ltd. Rehovot, Israel). The serologic characteristics of this antibody and the biochemical characteristics of the gp67 (i.e., CD5) antigen were examined during the First International Workshop on Human Leukocyte Differentiation Antigens (Paris, 1982). MoAb H65 (workshop number: T34), and nine other MoAbs were found to have the same serologic pattern and to immunoprecipiate the gp67 antigen. Knowles, in Reinherz, et al., Leukocyte Typing II, 2: 259-288 (Springer-Verlag, 1986). In other studies, MoAb H65 has been shown to block the binding of FITC-conjugated anti-Leu-1 (Becton Dickson, Mountain View, Calif.) on gp67+cells indicating that both antibodies recognize the same epitope on the gp67 molecule or determinants that are located in such a configuration as to result in blocking by steric hindrance.
  • Example 9
  • The Use of Lyt-1 In The Prophylactic Treatment of Collagen Induced Arthritis in DBA/IJ Mice
  • Collagen-induced arthritis (CIA) is a widely utilized model of human rheumatoid arthritis. CIA is characterized by a chronic polyarticular arthritis which can be induced in rodents and in primates by immunization with homologous or heterologous, native Type II collagen. The resulting arthritis resembles rheumatoid arthritis because there are similar histopathologic sequelae, cellular and humoral immune responses and restricted association with specific major histocompatibility complex (MEC) haplotypes.
  • Native, heterologous Type II collagen emulsified with complete Freund's adjuvant induces an arthritis-like autoimmune reaction in DBA/IJ mice after a single intradermal tail injection. The mice were obtained from Jackson Laboratories, Bar Harbor, Me. Initially, the arthritis is noticeable as a slight swelling of one or more digits in the fourth week post-immunization. The chronic phase of CIA continually worsens over the ensuing 8 weeks as the arthritis progresses from the digits into the remaining peripheral articulating joints and eventually ends with ankylosis of the involved joints. The histopathology of CIA is characterized by lymphocyte infiltration of the joint space, synovial MHC class II expression and pannus formation. Not all joints are involved on every mouse, so there is a spectrum of arthritic severity. In a group of ten or more mice, the overall arthritic severity develops in a linear fashion over the course of 10-12 weeks.
  • The CIA model was used to test the potential efficacy of a monoclonal antibody directed against the pan-T cell surface antigen, Lyt-1, the murine equivalent of CD5. The antibody was administered to the mice before the immunization with Type II collagen. Normal DBA/I mice were also treated with a single 0.4 mg/kg i.v. injection of anti-Lyt-1 and were sacrificed after 72 hours for FACS analysis and for in vitro proliferation assays on spleen and lymph node cells. Any efficacy of this antibody would indicate a beneficial T cell-directed approach in rheumatoid arthritis via the CD5 surface antigen.
  • Effects of anti-Lyt-1 on DBA/IJ Spleen Cells and Peripheral Lymph Nodes.
  • Antibody 53-7.313 is a rat IgG2a monoclonal antibody (ATCC Accession No. TIB 104) reactive with all alleles of the mouse lymphocyte differentiation antigen, Lyt-1. The IND1 antibody is a mouse IgG1, anti-human melanoma antibody used as a negative control (Xoma Corp., Berkeley, Calif.). All other antibodies were obtained from Pharmingen Inc. (San Diego, Calif.) as direct conjugates for quantitation on a Becton-Dickinson FACScan instrument.
  • Male DBA/IJ mice, age 6-8 weeks, were administered a single intravenous dose of either phosphate buffered saline, IND1 or anti-Lyt-1 via the tail vein at 0.4 mg/kg in 0.1 ml of phosphate buffered saline. Mice were sacrificed for analysis three days after dosing. Single cell suspensions of spleens and peripheral lymph nodes were prepared by standard procedures and 1×106 cells were stained with the respective antibodies for fluorescence activated cell sorter (FACS) analysis. Proliferation assays were also performed to provide a second measure of T cell depletion. Cells (1×105/well) were stimulated with Concanavalin A, Interleukin-2 (IL-2), IL-2 and H57.597 (a pan α,β T cell receptor antibody) or the Staphylococcal enterotoxins A and B. Cells were cultured for a total of 72 hours and proliferation was quantitated by the addition of 3H-methylthymidine for the last 24 hours. After 72 hours, the cells were harvested with an Inotech INB-384 harvesting and counting system, which collects the cells onto glass fiber filters with subsequent gas proportional beta particle detection. Results are generally expressed as the mean of triplicate wells±SEM in Tables 5 and 6.
  • A. FACS Analysis of Lymph Node and Spleen Cells
  • FACS analysis of lymph node cells (LNC) and spleen cells (SPC) from each treatment group (n=3/group) were analyzed for percent expression of α,β T cell receptor, CD3, CD4, CD5, and CD8. The results are presented in Table 4.
  • In Table 4, statistical significance was determined by Analysis of Variance followed by Duncan's New Multiple Range post-hoc test. These data indicate that administration of anti-Lyt-1 antibody results in a significant depletion of peripheral T lymphocytes at the 72 hour time point. The results could not be explained by residual circulating antibody as other T cell markers (CD3, etc.) are also depleted to a similar extent.
  • B. Effects of anti-Lyt-1 Administration on Proliferation Analysis
  • In vitro proliferation assays were performed on mice from each treatment group (n=3/group) in response to Concanavalin A, IL-2, IL-2+H57, Staphylococcal enterotoxin A and B (SEA and SEB). The results are presented in Table 5.
  • Overall, these data indicate that there is an observable and functional depletion of DBA/IJ T peripheral lymphocytes 72 hours after a single (0.4 mg/kg) intravenous dose of anti-Lyt-1 antibody.
  • C. Effects of anti-Lyt-1 on Collagen-induced Arthritisin DBA/IJ Mice.
  • A. Materials and Methods
  • Male DBA/IJ mice, age 6-8 weeks, were administered the antibodies 53-7.313 (anti-Lyt-1), IND1 (anti-melanoma) or phosphate buffered saline (PBS) in two intravenous (0.4 mg/kg) doses 48 hours apart starting four days prior to immunization with 100 μg of bovine type II collagen emulsified with an equal volume of Fraund's complete adjuvant to a final injection volume of 100 μl. Each dose group was comprised of ten mice. Mice were monitored weekly starting on Day 21
    TABLE 4
    FACS Analysis of anti-Lyt-1 Treated DBA/1J Mice
    CELL
    TREATMENT TYPE α, βTCR CD3 CD4 CD8 CD5
    PBS LNC 80.2 ± 2.2% 79.8 ± 1.6% 58.7 ± 1.4% 19.4 ± 2.6% 80.0 ± 0.6%
    IND1 LNC 82.5 ± 1.3% 82.6 ± 1.9% 60.9 ± 2.0% 21.1 ± 1.5% 78.5 ± 1.2%
    αLyt-1 LNC *62.7 ± 5.8%  *62.4 ± 1.0%  *42.0 ± 1.9%  21.1 ± 0.2% *56.0 ± 2.6% 
    PBS SPC 18.0 ± 2.8% 25.0 ± 0.1% 16.5 ± 2.1% 4.10 ± 0.5% 23.1 ± 0.1%
    IND1 SPC 19.3 ± 1.6% 22.8 ± 1.4% 13.9 ± 0.8% 4.20 ± 0.3% 20.8 ± 1.5%
    αLyt-1 SPC 14.0 ± 0.3% *13.8 ± 0.4%  *8.07 ± 0.3%  *2.40 ± 0.1%  *11.0 ± 0.1% 
  • TABLE 5
    Proliferation Analysis of anti-Lyt-1 Treated DBA/1J mice.
    TREATMENT Concanavalin A IL-2 IL-2 + H57 SEA SEB
    IND1 26547 ± 3501 1181 ± 234 11341 ± 1663 12324 + 1968 8747 ± 2025
    αLyt-1 *11561 ± 4375  *593 ± 274 *4090 ± 2383 *5568 ± 2576 *1138 ± 350 

    after immunization. Individual mice were scored for arthritic severity by grading each paw on a scale from 0 to 2. A score of 1 indicated swelling in up to two toes and a score of 2 indicated swelling in more than two toes up to total paw involvement and ankylosis of the large joint in the later time points. An individual mouse could have a maximum arthritic severity score of 8. Mice were monitored until day 80 after collagen immunization and then were sacrificed by cervical dislocation. Results are expressed as the mean arthritic score for each dose group.
  • The changes in arthritic score during the course of the study are shown in FIG. 12. The overall conclusion in FIG. 12 is that administration of the anti-Lyt-1 antibody prior to collagen immunization caused a significant decrease in the resulting severity of arthritis. In all of the treatment groups, the appearance of visible symptoms initiated at approximately 30 days after immunization and progressed linearly until the end of the study. The anti-Lyt-1 treatment group began to show ameliorated arthritic symptoms at Day 48 and never developed arthritis to the same extent as the other two groups. The onset of arthritis was not significantly delayed by the anti-Lyt-1 treatment.
  • Statistical significance was determined by a Repeated Measures Analysis of Variance with one between subjects variable (antibody treatment). A Repeated Measures Analysis was necessary as each mouse was continually monitored for the duration of the study. Thus, the arthritic scores for consecutive days cannot be considered as independent observations contributing to the overall degrees of freedom in the F test for significant differences among groups. A Repeated Measures Analysis uses the degrees of freedom from the number of individuals per group instead of the number of observations. A typical between subjects Analysis of Variance may be inappropriate and may indicate false significance among the treatment groups. A comparison of means in the Treatment by Day after Immunization was done to determine the significance of anti-Lyt-1 treatment relative to PBS and IND1 control groups.
  • In conclusion, the intravenous administration of a rat monoclonal antibody reactive to the mouse equivalent of CD5, Lyt-1, is able to significantly decrease T lymphocytes in the spleen and in peripheral lymph nodes after a single 0.4 mg/kg dose. This T cell decrease is the probable mechanism for the significant (p<0.01) decrease in arthritic severity seen with the same anti-Lyt-1 dose prior to type II collagen immunization.
  • Example 10
  • Depletion of Human T Cells From SCID Mice by Treatment With H65 MoAb
  • Severe combined immunodeficient (CB.17 scid/scid; SCID) mice maintain human lymphoid cells for several months following transplantation of human peripheral blood mononuclear cells (PBMC). Such chimeric mice, referred to as PBMC/SCID mice, have functional human cells, as shown by the presence of human Ig in their serum. PBMC/SCID mice maintain human T cells in tissues such as spleen and blood. Human T cells present in PBMC/SCID mice are predominantly of a mature phenotype and express T cell antigens, including CD3, CD5, CD7, and CD4 or CD8. In addition, most T cells appear to be activated memory cells, as judged by the expression of HLA-DR and CD45RO. These engrafted T cells appear to be functional since (a) they may provide help to B cells to produce anti-tetanus toxoid antibodies, (b) they produce soluble interleukin-2 receptor (sIL-2R) which may be detected in plasma, and (c) they proliferate in response to mitogenic anti-human CD3 monoclonal antibodies supplemented with IL-2 in vitro.
  • Because of the presence of human T and B cells, PBMC/SCID mice offer an in vivo model system in which to evaluate the efficacy of anti-human T cell drugs, such as H65 MoAb, a mouse IgG1 directed against human CD5.
  • The SCID mice were obtained from Taconic, Germantown, N.Y., and at 6 to 7 weeks of age were injected with 200 mg/kg cyclophosphamide intraperitoneally (i.p.) to ensure engraftment of human PBMC. Two days later, 25 to 40×106 human PBMC, isolated by Ficoll-Hypaque density gradient centrifugation from lymphapheresis samples obtained from normal donors (HemaCare Corporation, Sherman Oaks, Calif.), were injected i.p.
  • At 2 to 3 weeks after PBMC injection, the mice were bled from the retro-orbital sinus and levels of human immunoglobulin (Ig) and human sIL-2R in plasma were quantified using sandwich ELISAs. Mice with low or undetectable levels of these human proteins were eliminated from the study and the remainder were divided into the various treatment groups (6 per group). The mice were then administered H65 MoAb (0.2 or 0.02 mg/kg/day), H65-based F(ab′)2 fragment (2 mg/kg/day) or vehicle (buffer) intravenously (i.v.) for 10 consecutive daily injections one day after the last injection, the mice were bled and spleens were collected. Single cell suspensions of blood cells and splenocytes were prepared by standard methods. Recovered cells were then assayed for human T cell surface markers using flow cytometry.
  • Two to five hundred thousand cells were stained with the following FITC— or PE-conjugated Abs (Becton-Dickinson, Mountain View, Calif.): HLe-1-FITC (anti-CD45), Leu-2-FITC•(anti-CD5), and Leu-3-PE (anti-CD4). Samples were analyzed on a FACScan using log amplifiers. Regions to quantify positive cells were set based on staining of cells obtained from naive SCID mice. The absolute numbers of human antigen-positive cells recovered from SCID tissues were determined by multiplying the percent positive cells by the total number of cells recovered from each tissue sample. The total number of leukocytes in blood was calculated using a theoretical blood volume of 1.4 ml/mouse. Statistical comparisons between treatment groups were made using the Mann-Whitney U test.
  • The number of human T cells (CD4 plus CD8 cells) recovered from spleens and blood of PBMC/SCID mice following treatment with H65 MoAb or vehicle (control) is shown in FIG. 13. Significantly lower numbers of T cells were recovered from spleens and blood of mice treated with either 0.2 or 0.02 mg/kg/day H65 MoAb as compared to vehicle-treated mice. In contrast, treatment with 2 mg/kg/day of an H65-based F(ab′)2 fragment did not significantly deplete human T cells from spleens or blood, even though a 10 to 100-fold higher dose was used (FIG. 14).
  • These results indicate that an anti-human CD5 MoAb depletes human T cells in an experimental animal model. The ability of this MoAb to deplete human T cells from SCID mice is apparently dependent on the Fc portion of the MoAb, as an F(ab′)2 fragment was ineffective.
  • Example 11
  • The Use of OX19 Monoclonal Antibody In The Prophylactic Treatment of Collagen Induced Arthritis in Diabetes-Resistant BB Rats
  • Collagen-induced arthritis (CIA) in the diabetes-resistant Biobreeding (DR BB) rat is a particularly relevant animal model of human rheumatoid arthritis, in that the DR BB rat RT1.Dβ gene encodes a nucleotide sequence homologous to the human HLA-DRβ gene reported to be associated with rheumatoid arthritis susceptibility. In this model, DR BB rats are administered a single intradermal tail injection of heterologous Type II collagen emulsified with incomplete Freund's adjuvant. Development of the arthritis is considerably faster than in the DBA/1J CIA model. Onset of clinical signs occurs 1.5 to 2 weeks after collagen immunization, with peak swelling observed a few days after onset. Incidence is generally quite high (>85% of animals immunized). The swelling is generally severe, involves the entire footpad and ankle joint, and is restricted to the hindlimbs. Histopathological examination has revealed that the arthritis begins as a proliferative synovitis with pannus formation at the joint margins that is followed by a bidirectional erosion of both the outer (unmineralized) and inner (mineralized) layers of cartilage.
  • This experiment uses the DR BB CIA rat model to assess the efficacy of a monoclonal antibody (MoAb), OX19 directed against the equivalent of the CD5 antigen in the rat. The antibody was administered to the rats prior to immunization with Type II collagen. Normal Sprague-Dawley rats were also treated with a single 0.5 mg/kg i.v. injection and were sacrificed after 3 hours for evaluation of MoAb binding to T cells, or after 2 days for quantitation of T cells in lymphoid tissues using flow cytometry.
  • A. Effects of OX19 MoAb on T Cells In Lymphoid Tissues of Normal Spraoue-Dawley Rats
  • OXI9 MoAb is a mouse IgG1 directed against the equivalent of rat CD5 antigen present on rat T cells. OXl9 hybridoma is available from the European Collection of Animal Cell Cultures (ECACC) and has ECACC No. 84112012. H65 MoAb, a mouse IgG1 reactive against human CD5, was used as an isotype matched negative control. Fluorescein-conjugated antibodies directed against surface antigens on rat pan-T cells (W3/13), CD4 cells (W3/25) and CD5 cells (OX8) were obtained from Accurate Chemical and Scientific Corporation, Westbury, N.Y. for flow cytometric quantitation of T cells in rat lymphoid tissues. Phycoerythrin-conjugated goat anti-mouse IgG1 (Caltag Laboratories, South San Francisco, Calif.) was used to detect OX19 MoAb bound to rat T cells in a two-color analysis.
  • Male Sprague-Dawley rats (Simonsen Laboratories, Gilroy, Calif.), 100 to 150 grams, were administered a single i.v. bolus injection of OX19 MoAb (0.5 mg/kg) or control MoAb (0.5 mg/kg) in phosphate buffered saline containing 0.1% Tween 80 (PBS/Tween). Animals were sacrificed at 3 hours (binding experiment) or 2 days (depletion experiment) after dosing. Single cell suspensions of blood, spleens and lymph nodes were prepared by standard procedures and 1×106 cells were stained with appropriate antibodies for FACS analysis.
  • A. Binding of OX19 MoAb to Rat T Cells In Vivo.
  • Blood, spleen and lymph node cells from one animal in each treatment group were analyzed for percentage of CD4 and CD8 T cells, and percentage of CD4 and CD8 T cells that also stained positively for surface-bound mouse IgG1. The results are presented in Table 6. T cells were depleted from the blood at 3 hours after OX19 MoAb administration. Almost all of the T cells that remained in the blood, and most of those present in the spleen and lymph nodes in the OX19 MoAb-treated rat also stained positively for surface-bound mouse IgG1, indicating that the dose of OX19 MoAb used was sufficient to saturate most of the T cells in these major lymphoid organs. These results provide doses useful in therapeutic applications.
  • B. Effect of OX19 MoAb Treatment on T Cell Subpopulations in Rat Lymphoid Tissues.
  • Blood, spleen and lymph node cells from two animals in each treatment group were analyzed for percentage of pan-T, CD4 and CD8 cells. The results are presented in Table 7 as the mean of the two animals. OX19 MoAb treatment resulted in depletion of T cells from all tissues examined as compared to treatment with the control MoAb. These results also provide appropriate doses to be used in therapeutic applications using antibodies according to the invention.
  • Example 12
  • Effect of OX19 MoAb Treatment on Development of Collagen-Induced Arthritis in DR BB Rats
  • Male DR BB/Wor rats (obtained from the University of Massachusetts breeding facility; 8 per treatment group), age 6 weeks, were administered i.v. injections of OX19 MoAb (0.5 mg/kg), control MoAb (0.5 mg/kg) or buffer (PBS/Tween) on day 7 and day 4 prior to immunization at the base of the tail on day 0 with 0.3 mg of bovine Type II collagen emulsified in 0.15 ml
    TABLE 6
    Bind of OX19 MoAb to Rat T Cells In Vivo.
    % Positive Cell
    Tissue Treatment CD4 CD4/mIgG1* CD8 CDB/mIgG1*
    Blood H65 MoAb 47.0 6.7 11.1 5.7
    OX19 8.7 96.2 4.1 70.2
    Spleen H65 MoAb 23.1 14.8 4.4 20.6
    OX19 MoAb 16.4 84.8 3.4 73.6
    Lymph H65 MoAb 66.9 4.2 7.4 6.5
    Node OX19 MoAb 54.7 96.2 7.3 96.8

    *The % of CD4 or CD8 cells that are also positive for mouse IgG1.
  • TABLE 7
    FACS Analysis of Tissues from OX19 MAb-Treated Rats.
    % Positive
    Cells
    Tissue Treatment Pan-T CD4 CD8
    Bl∘∘d H65 MoAB 61.8 50.4 12.0
    OX19 MoAb 47.0 37.3 8.8
    Spleen H65 MoAb 36.0 25.3 7.1
    OX19 MoAb 21.5 9.9 5.0
    Lymph Node H65 MoAb 74.5 62.7 13.1
    OX19 MoAb 33.8 24.9 4.3

    of incomplete Freund's adjuvant. Rats were scored daily for arthritis beginning 8 days after collagen immunization. Severity was graded on a scale from 0 to 2, with a score of 1 indicating moderate swelling and a score of 2 indicating severe swelling. An individual animal could have a maximum arthritic severity score of 4 if there was bilateral hindlimb involvement.
  • The changes in arthritic score during the course of the study are shown in FIG. 15 and the arthritic incidence for each treatment group is presented in Table 8.
  • Control (buffer and control MoAb-treated) rats developed severe, predominantly bilateral hindlimb arthritis between days 10 and 14 with high incidence (88% for both groups). Treatment with OX19 MoAb completely prevented development of arthritis (0% incidence).
  • In conclusion, a 0.5 mg/kg intravenous dose of a mouse MoAb directed against the rat equivalent of CD5 was found to saturate and subsequently deplete T cells from lymphoid tissues of normal rats. This T cell depletion is the probable mechanism for the complete inhibition of arthritis development observed when the MoAb was administered prior to Type II collagen immunization in DR BB rats.
  • Example 13 Treatment of Rheumatoid Arthritis
  • Patients having rheumatoid arthritis (RA) are selected for treatment using an anti-pan T cell antibody of this invention.
  • Anti-CD5 antibody prepared as described above is administered to patients at doses of about 0.005 to 2.0 mg/kg/day for a period of 1-5 days, preferably 1-2 days. Alternatively, the dose may be given every 2-30
    TABLE 8
    Effect of OX19 MoAb Treatment on Arthritis Incidence
    Total Total
    arthritics Arthritics Score of “2” Score of “2”
    (1 or both (Both (1 or both (Both
    TREATMENT limbs) limbs) limbs) Limbs)
    PBS/Tween 7/8 (88%) 7/8 (88%) 7/8 (88%) 5/8 (63%)
    Control MoAb 7/8 (88%) 4/8 (50%) 6/8 (75%) 4/8 (50%)
    OX19 MoAb 0/8 (0%) 0/8 (0%) 0/8 (0%) 0/8 (0%)

    days instead of daily if chimeric and humanized MoAbs are used due to their increased half-life. To determine optimum dose and schedule, patients are treated at each dose and schedule in a dose escalating regimen. Patients are monitored using several indicia, including joint swelling and tenderness scores. Results are shown in FIG. 11.
  • Example 14 Treatment of SLE
  • Systemic Lupus Erythematosus (SLE) is a multisystemic disease characterized by inflammation and autoimmunity. Some of the more frequent manifestations include fatigue, anemia, fever, rashes, photosensitivity, alopecia, arthritis, pericarditis, pleurisy, vasculitis, nephritis and central nervous system disease. Under the Revised Criteria for Classification of SLE, a person is said to have SLE for purposes of clinical studies if any four or more of the aforementioned specified criteria are present, serially or simultaneously, during any interval of observation.
  • Anti-CD5 antibody prepared as described above is administered to patients at doses of about 0.005 to 2.0 mg/kg/day for a period of 1-5 days, preferably 1-2 days. Alternatively, the dose may be given every 2-30 days instead of daily if chimeric and humanized MoAbs are used due to their increased half-life. To determine optimum dose and schedule, patients are treated at each dose and schedule in a dose escalating regimen.
  • Example 15 Treatment of Psoriasis
  • Psoriasis is a disease of autoimmune etiology which Classically appears as plaques over the elbows and knees, although other areas of the skin are frequently afflicted. Abnormalities of the nails and the joints are also frequently observed. Particularly inflammatory joint disease can occur in an occasionally erosive and severe form.
  • Anti-CD5 antibody prepared as described above is administered to patients at doses of about 0.005 to 2.0 mg/kg/day for a period of 1-5 days, preferably 1-2 days. Alternatively, the dose may be given every 2-30 days instead of daily if chimeric and humanized MoAbs are used due to their increased half-life. To determine optimum dose and schedule, patients are treated at each dose and schedule in a dose escalating regimen.
  • Clinical observation includes evaluation of the patient's overall status as well as special attention to the psoriatic plaques. Additionally, monitoring of laboratory parameters such as white blood count and differential are recommended. Symptoms which may indicate poor tolerance to therapy or complications include nausea, vomiting, fatigue, rash, fever, chills and syncope. Any unexplained depletion in white blood cells other than lymphocytes is an indication to discontinue therapy. Preferably, differential analysis of lymphocytes is carried out. That is, analysis of the total number of T cells and B cells should be determined.
  • Example 16 Treatment of Type I Diabetes
  • There are two major types of diabetes. Type I has classically been associated with a requirement for exogenous insulin. Type I typically occurs before the age of 40 and is associated with an absence of insulin secretion. The pancreas of patients with long-term Type I insulin-dependent diabetes are devoid of pancreatic islet cells. There is a large body of evidence that the etiology of Type I insulin-dependent diabetes (IDDM) is autoimmune.
  • Patients are diagnosed as having IDDM based on the criteria established by the American Diabetes Association. Anti-CD5 antibody prepared as described above is administered to patients at doses of about 0.005 to 2.0 mg/kg/day for a period of 1-5 days, preferably 1-2 days. Alternatively, the dose may be given every 2-30 days instead of daily if chimeric and humanized MoAbs are used due to their increased half-life. To determine optimum dose and schedule, patients are treated at each dose and schedule in a dose escalating regimen.
  • During the study, the patients were monitored by clinical and laboratory parameters. Clinical symptoms indicating poor tolerance to therapy or complications include fatigue, vomiting, rash, fever, chills, and syncope. Laboratory evaluation included white blood cell counts with differential analysis daily and blood glucose levels at least twice a day.
  • Using diagnostic criteria predictive of the onset of Type I diabetes, patients may be selected for prophylactic treatment. This treatment follows the dose and schedule noted above for treatment of clinical insulin dependent diabetes.
  • While the invention has been described in terms of specific examples and preferred embodiments, is understood that variations and improvements will occur to those skilled in the art. Therefore, it is recognized that there are numerous variations and improvements which come within the scope of the invention as claimed.

Claims (5)

1. A method for determining the risk of changing an amino acid residue in an antibody variable domain, said method comprising
(a) aligning by computer the amino acid sequence of said variable domain with the amino acid sequence of an antibody light or heavy chain variable region or consensus sequence; and
(b) determining the risk assigned to changing said amino acid residue using the paired bind and bury lines as shown in FIG. 6A or 6B.
2. The method of claim 1, wherein said selected antibody variable region in part (b) is said light chain variable region.
3. The method of claim 1, wherein said selected antibody variable region in part (b) is said heavy chain variable region.
4. A method for determining the risk of changing an amino acid residue in an antibody variable region light chain sequence, said method comprising
(a) aligning by computer the amino acid sequence of said antibody variable domain light chain sequence with the amino acid sequence of an antibody light chain variable region sequence or consensus; and
(b) determining the risk assigned to changing said amino acid residue using the paired bind and bury lines as shown in FIG. 6A.
5. A method for determining the risk of changing an amino acid residue in an antibody variable region heavy chain sequence, said method comprising
(a) aligning by computer the amino acid sequence of said antibody variable domain heavy chain sequence with the amino acid sequence of an antibody heavy chain variable region sequence or consensus; and
(b) determining the risk assigned to changing said amino acid residue using the paired bind and bury lines as shown in FIG. 6B.
US11/133,775 1991-12-13 2005-05-19 Modified antibody variable domains Abandoned US20050239141A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/133,775 US20050239141A1 (en) 1991-12-13 2005-05-19 Modified antibody variable domains

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US80846491A 1991-12-13 1991-12-13
PCT/US1992/010906 WO1993011794A1 (en) 1991-12-13 1992-12-14 Methods and materials for preparation of modified antibody variable domains and therapeutic uses thereof
US08/107,669 US5766886A (en) 1991-12-13 1992-12-14 Modified antibody variable domains
US9798098A 1998-06-16 1998-06-16
US10/325,696 US20040005630A1 (en) 1991-12-13 2002-12-18 Methods and materials for preparation of modified antibody variable domains and therapeutic uses thereof
US11/133,775 US20050239141A1 (en) 1991-12-13 2005-05-19 Modified antibody variable domains

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US10/325,696 Continuation US20040005630A1 (en) 1991-12-13 2002-12-18 Methods and materials for preparation of modified antibody variable domains and therapeutic uses thereof

Publications (1)

Publication Number Publication Date
US20050239141A1 true US20050239141A1 (en) 2005-10-27

Family

ID=25198837

Family Applications (5)

Application Number Title Priority Date Filing Date
US08/107,669 Expired - Lifetime US5766886A (en) 1991-12-13 1992-12-14 Modified antibody variable domains
US08/477,531 Expired - Fee Related US5821123A (en) 1991-12-13 1995-06-07 Modified antibody variable domains
US08/472,788 Expired - Fee Related US5770196A (en) 1991-12-13 1995-06-07 Modified antibody variable domains and therapeutic uses thereof
US10/325,696 Abandoned US20040005630A1 (en) 1991-12-13 2002-12-18 Methods and materials for preparation of modified antibody variable domains and therapeutic uses thereof
US11/133,775 Abandoned US20050239141A1 (en) 1991-12-13 2005-05-19 Modified antibody variable domains

Family Applications Before (4)

Application Number Title Priority Date Filing Date
US08/107,669 Expired - Lifetime US5766886A (en) 1991-12-13 1992-12-14 Modified antibody variable domains
US08/477,531 Expired - Fee Related US5821123A (en) 1991-12-13 1995-06-07 Modified antibody variable domains
US08/472,788 Expired - Fee Related US5770196A (en) 1991-12-13 1995-06-07 Modified antibody variable domains and therapeutic uses thereof
US10/325,696 Abandoned US20040005630A1 (en) 1991-12-13 2002-12-18 Methods and materials for preparation of modified antibody variable domains and therapeutic uses thereof

Country Status (8)

Country Link
US (5) US5766886A (en)
EP (2) EP1291360A1 (en)
JP (3) JP4157160B2 (en)
AT (1) ATE249840T1 (en)
CA (2) CA2507749C (en)
DE (1) DE69233204T2 (en)
ES (1) ES2202310T3 (en)
WO (1) WO1993011794A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9511138B2 (en) 2011-12-23 2016-12-06 Laboratoire Francais Du Fractionnement Et Des Biotechnologies Pharmaceutical compositions comprising an antibody which binds the human anti-mullerian hormone receptor type II

Families Citing this family (720)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5618920A (en) 1985-11-01 1997-04-08 Xoma Corporation Modular assembly of antibody genes, antibodies prepared thereby and use
US7041871B1 (en) 1995-10-10 2006-05-09 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US7084260B1 (en) 1996-10-10 2006-08-01 Genpharm International, Inc. High affinity human antibodies and human antibodies against human antigens
US5770429A (en) * 1990-08-29 1998-06-23 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5874082A (en) * 1992-07-09 1999-02-23 Chiron Corporation Humanized anti-CD40 monoclonal antibodies and fragments capable of blocking B cell proliferation
US5397703A (en) 1992-07-09 1995-03-14 Cetus Oncology Corporation Method for generation of antibodies to cell surface molecules
US6774227B1 (en) * 1993-05-17 2004-08-10 Cytokine Pharmasciences, Inc. Therapeutic uses of factors which inhibit or neutralize MIF activity
CU22615A1 (en) * 1994-06-30 2000-02-10 Centro Inmunologia Molecular PROCEDURE FOR OBTAINING LESS IMMUNOGENIC MONOCLONAL ANTIBODIES. MONOCLONAL ANTIBODIES OBTAINED
DE4425115A1 (en) * 1994-07-15 1996-01-18 Boehringer Mannheim Gmbh Method for modifying the stability of antibodies
US6309636B1 (en) * 1995-09-14 2001-10-30 Cancer Research Institute Of Contra Costa Recombinant peptides derived from the Mc3 anti-BA46 antibody, methods of use thereof, and methods of humanizing antibody peptides
US6226620B1 (en) * 1996-06-11 2001-05-01 Yeong Kuang Oon Iterative problem solving technique
US20030109680A1 (en) * 2001-11-21 2003-06-12 Sunol Molecular Corporation Antibodies for inhibiting blood coagulation and methods of use thereof
US20060235209A9 (en) * 1997-03-10 2006-10-19 Jin-An Jiao Use of anti-tissue factor antibodies for treating thromboses
US5986065A (en) * 1997-03-10 1999-11-16 Sunol Molecular Corporation Antibodies for inhibiting blood coagulation and methods of use thereof
US7749498B2 (en) * 1997-03-10 2010-07-06 Genentech, Inc. Antibodies for inhibiting blood coagulation and methods of use thereof
CA2290485C (en) * 1997-05-21 2008-08-05 Biovation Limited Method for the production of non-immunogenic proteins
AU2444899A (en) * 1998-01-22 1999-08-09 Astrazeneca Ab Pharmaceutical formulation comprising an antibody and a citrate buffer
US6642011B2 (en) * 1998-04-15 2003-11-04 Genencor International, Inc. Human protease and use of such protease for pharmaceutical applications and for reducing the allergenicity of non-human proteins
US6686164B1 (en) 1998-10-30 2004-02-03 Novozymes A/S Low allergenic protein variants
US20030035798A1 (en) 2000-08-16 2003-02-20 Fang Fang Humanized antibodies
IT1306704B1 (en) * 1999-05-26 2001-10-02 Sirs Societa Italiana Per La R MONOCLONAL ANTIBODIES AND ITS SYNTHETIC OR BIOTECHNOLOGICAL DERIVATIVES ENABLE TO ACT AS ANTAGONIST MOLECULES FOR NGF.
US6686188B2 (en) * 2000-05-26 2004-02-03 Amersham Plc Polynucleotide encoding a human myosin-like polypeptide expressed predominantly in heart and muscle
US6656700B2 (en) 2000-05-26 2003-12-02 Amersham Plc Isoforms of human pregnancy-associated protein-E
KR100388942B1 (en) * 2000-07-08 2003-06-25 한화석유화학 주식회사 A Single Chain Variable Fragment of an Antibody for Protein Expressed on Cell Surface of Human Cortical Thymocyte and Leukemic Cell
UA81743C2 (en) 2000-08-07 2008-02-11 Центокор, Инк. HUMAN MONOCLONAL ANTIBODY WHICH SPECIFICALLY BINDS TUMOR NECROSIS FACTOR ALFA (TNFα), PHARMACEUTICAL MIXTURE CONTAINING THEREOF, AND METHOD FOR TREATING ARTHRITIS
US6902734B2 (en) 2000-08-07 2005-06-07 Centocor, Inc. Anti-IL-12 antibodies and compositions thereof
US7288390B2 (en) 2000-08-07 2007-10-30 Centocor, Inc. Anti-dual integrin antibodies, compositions, methods and uses
ATE327004T1 (en) 2000-10-02 2006-06-15 Chiron Corp METHOD FOR THE THERAPY OF B-CELL MALIGNITIES USING ANTAGONISTIC ANTIBODIES AGAINST CD40
US20020123474A1 (en) * 2000-10-04 2002-09-05 Shannon Mark E. Human GTP-Rho binding protein2
US20030049800A1 (en) * 2000-12-15 2003-03-13 Saravis Calvin A. Compositions and methods for producing antibodies to low molecular weight analytes
AU2002231736A1 (en) 2000-12-22 2002-07-08 Max-Planck-Gesellschaft Zur Forderung Der Wissenschaften E.V. Use of repulsive guidance molecule (rgm) and its modulators
JP2005522192A (en) 2001-07-19 2005-07-28 パーラン セラピューティクス, インコーポレイテッド Multimeric proteins and methods of making and using multimeric proteins
US20040078837A1 (en) * 2001-08-02 2004-04-22 Shannon Mark E. Four human zinc-finger-containing proteins: MDZ3, MDZ4, MDZ7 and MDZ12
US7658924B2 (en) * 2001-10-11 2010-02-09 Amgen Inc. Angiopoietin-2 specific binding agents
US7521053B2 (en) * 2001-10-11 2009-04-21 Amgen Inc. Angiopoietin-2 specific binding agents
AU2002346369B2 (en) 2001-11-14 2009-08-27 Eusa Pharma (Uk) Ltd Anti-IL-6 antibodies, compositions, methods and uses
US20050069549A1 (en) 2002-01-14 2005-03-31 William Herman Targeted ligands
US20080254027A1 (en) * 2002-03-01 2008-10-16 Bernett Matthew J Optimized CD5 antibodies and methods of using the same
ATE496134T1 (en) 2002-03-29 2011-02-15 Xoma Technology Ltd MULTIGENE VECTORS AND METHODS FOR INCREASED EXPRESSION OF RECOMBINANT POLYPEPTIDES
US20050152899A1 (en) * 2002-05-10 2005-07-14 Kinch Michael S. EphA2 agonistic monoclonal antibodies and methods of use thereof
WO2003094859A2 (en) * 2002-05-10 2003-11-20 Medimmune, Inc. Epha2 monoclonal antibodies and methods of use thereof
CA2485548A1 (en) * 2002-05-10 2004-02-19 Purdue Research Foundation Epha2 agonistic monoclonal antibodies and methods of use thereof
US7425618B2 (en) 2002-06-14 2008-09-16 Medimmune, Inc. Stabilized anti-respiratory syncytial virus (RSV) antibody formulations
WO2004016750A2 (en) 2002-08-14 2004-02-26 Macrogenics, Inc. FcϜRIIB-SPECIFIC ANTIBODIES AND METHODS OF USE THEREOF
HUE034378T2 (en) 2002-10-16 2018-02-28 Purdue Pharma Lp Antibodies that bind cell-associated CA 125/O722P and methods of use thereof
NZ563471A (en) 2002-11-08 2009-04-30 Ablynx Nv Camelidae antibodies against imminoglobulin E and use thereof for the treatment of allergic disorders
US20110091451A1 (en) * 2002-11-15 2011-04-21 Kavanaugh William M Methods for preventing and treating cancer metastasis and bone loss associated with cancer metastasis
DE10303974A1 (en) 2003-01-31 2004-08-05 Abbott Gmbh & Co. Kg Amyloid β (1-42) oligomers, process for their preparation and their use
US20050009097A1 (en) * 2003-03-31 2005-01-13 Better Marc D. Human engineered antibodies to Ep-CAM
WO2004092343A2 (en) * 2003-04-11 2004-10-28 Medimmune, Inc. Epha2, hypoproliferative cell disorders and epithelial and endothelial reconstitution
EP1617864A4 (en) * 2003-04-11 2006-06-21 Medimmune Inc Epha2 and non-neoplastic hyperproliferative cell disorders
KR20110094361A (en) 2003-04-11 2011-08-23 메디뮨 엘엘씨 Recombinant il-9 antibodies and uses thereof
US8613922B2 (en) 2003-04-24 2013-12-24 The University Of North Carolina At Chapel Hill Methods for inhibiting diabetic retinopathy with an antibody against integrin associated protein (IAP)
GB2401040A (en) * 2003-04-28 2004-11-03 Chugai Pharmaceutical Co Ltd Method for treating interleukin-6 related diseases
EP1639014B1 (en) 2003-06-13 2010-09-22 Biogen Idec MA Inc. Aglycosyl anti-cd154 (cd40 ligand) antibodies and uses thereof
ES2527102T3 (en) * 2003-06-19 2015-01-20 Genentech, Inc. Compositions and methods for the treatment of disorders related to coagulation
US7393534B2 (en) * 2003-07-15 2008-07-01 Barros Research Institute Compositions and methods for immunotherapy of cancer and infectious diseases
US20050058658A1 (en) * 2003-07-15 2005-03-17 Barros Research Institute Compositions and methods for immunotherapy of human immunodeficiency virus (HIV)
TW201319088A (en) 2003-07-18 2013-05-16 Amgen Inc Specific binding agents to hepatocyte growth factor
WO2005014795A2 (en) 2003-08-08 2005-02-17 Genenews Inc. Osteoarthritis biomarkers and uses thereof
CA2537055A1 (en) * 2003-08-22 2005-04-21 Medimmune, Inc. Humanization of antibodies
AU2003283136A1 (en) 2003-11-07 2005-05-26 Ablynx N.V. Camelidae single domain antibodies vhh directed against epidermal growth factor receptor and uses therefor
EP2311873B1 (en) 2004-01-07 2018-08-29 Novartis Vaccines and Diagnostics, Inc. M-csf-specific monoclonal antibody and uses thereof
ES2416344T3 (en) 2004-02-03 2013-07-31 Diadexus, Inc. Methods to determine the activity of Lp-PLA2.
EA010687B1 (en) * 2004-02-06 2008-10-30 Нимокс Корпорейшн Humanized antibody
US7625549B2 (en) * 2004-03-19 2009-12-01 Amgen Fremont Inc. Determining the risk of human anti-human antibodies in transgenic mice
US20050260679A1 (en) * 2004-03-19 2005-11-24 Sirid-Aimee Kellerman Reducing the risk of human anti-human antibodies through V gene manipulation
US6986264B1 (en) * 2004-07-15 2006-01-17 Carrier Corporation Economized dehumidification system
WO2006033702A2 (en) 2004-07-26 2006-03-30 Biogen Idec Ma Inc. Anti-cd154 antibodies
US7700720B2 (en) 2004-09-21 2010-04-20 Medimmune, Llc Antibodies against and methods for producing vaccines for respiratory syncytial virus
AU2005299355A1 (en) 2004-10-27 2006-05-04 Medimmune, Llc Modulation of antibody specificity by tailoring the affinity to cognate antigens
US20060134698A1 (en) * 2004-12-20 2006-06-22 Evanston Northwestern Healthcare Research Institute Methods for treating cardiac disease by modifying an N-terminal domain of troponin I
WO2006081171A1 (en) * 2005-01-24 2006-08-03 Amgen Inc. Humanized anti-amyloid antibody
US7402730B1 (en) 2005-02-03 2008-07-22 Lexicon Pharmaceuticals, Inc. Knockout animals manifesting hyperlipidemia
CN103497993A (en) 2005-02-07 2014-01-08 基因信息公司 Mild osteoarthritis biomarkers and uses thereof
WO2006089133A2 (en) 2005-02-15 2006-08-24 Duke University Anti-cd19 antibodies and uses in oncology
US20090142338A1 (en) * 2005-03-04 2009-06-04 Curedm, Inc. Methods and Compositions for Treating Type 1 and Type 2 Diabetes Mellitus and Related Conditions
JP2008531730A (en) * 2005-03-04 2008-08-14 キュアーディーエム、インク. Methods and pharmaceutical compositions for treating type I diabetes mellitus and other conditions
CA2601400A1 (en) 2005-03-19 2006-09-28 Medical Research Council Improvements in or relating to treatment and prevention of viral infections
ATE531800T1 (en) * 2005-03-25 2011-11-15 Ca Nat Research Council METHOD FOR ISOLATING SOLUBLE POLYPEPTIDES
WO2006110599A2 (en) 2005-04-07 2006-10-19 Novartis Vaccines And Diagnostics Inc. Cacna1e in cancer diagnosis, detection and treatment
JP2008535494A (en) 2005-04-07 2008-09-04 サグレシュ ディスカバリー, インコーポレイテッド Cancer-related gene (PRLR)
EP3479844B1 (en) 2005-04-15 2023-11-22 MacroGenics, Inc. Covalent diabodies and uses thereof
JO3058B1 (en) 2005-04-29 2017-03-15 Applied Molecular Evolution Inc Anti-IL-6 Antibodies,Compositions,Methods and uses
EP2221316A1 (en) 2005-05-05 2010-08-25 Duke University Anti-CD19 antibody therapy for autoimmune disease
BRPI0611069A2 (en) 2005-05-06 2010-11-09 Zymogenetics Inc monoclonal antibody, humanized antibody, antibody fragment, use of an antagonist, and hybridoma
DK2949668T3 (en) 2005-05-18 2019-11-11 Ablynx Nv IMPROVED NANOBODIES TM AGAINST TUMOR CANCER FACTOR-ALFA
WO2006128083A2 (en) * 2005-05-25 2006-11-30 Curedm, Inc. Human proislet peptide, derivatives and analogs thereof, and methods of using same
KR101502920B1 (en) * 2005-06-21 2015-03-17 조마 (유에스) 엘엘씨 IL-1β Binding antibodies and fragments thereof
KR20080025174A (en) 2005-06-23 2008-03-19 메디뮨 인코포레이티드 Antibody formulations having optimized aggregation and fragmentation profiles
EP1896073B1 (en) 2005-06-30 2013-03-06 Janssen Biotech, Inc. Anti-il-23 antibodies, compositions, methods and uses
EP1907421A4 (en) * 2005-06-30 2012-03-28 Abbott Lab Il-12/p40 binding proteins
EP1913028B1 (en) * 2005-07-28 2015-03-04 Novartis AG Use of antibody to m-csf
EP1913027B1 (en) 2005-07-28 2015-03-04 Novartis AG M-csf specific monoclonal antibody and uses thereof
JP5230420B2 (en) 2005-08-18 2013-07-10 ゲンマブ エー/エス Treatment with CD4 binding peptides and radiation
US7612181B2 (en) * 2005-08-19 2009-11-03 Abbott Laboratories Dual variable domain immunoglobulin and uses thereof
EP2500355A3 (en) 2005-08-19 2012-10-24 Abbott Laboratories Dual variable domain immunoglobulin and uses thereof
US20090215992A1 (en) * 2005-08-19 2009-08-27 Chengbin Wu Dual variable domain immunoglobulin and uses thereof
SG2014010029A (en) 2005-08-19 2014-08-28 Abbott Lab Dual variable domain immunoglobin and uses thereof
US8906864B2 (en) 2005-09-30 2014-12-09 AbbVie Deutschland GmbH & Co. KG Binding domains of proteins of the repulsive guidance molecule (RGM) protein family and functional fragments thereof, and their use
EP1945816B1 (en) 2005-10-21 2011-07-27 GeneNews Inc. Method and apparatus for correlating levels of biomarker products with disease
RS53270B2 (en) 2005-11-30 2018-05-31 Abbvie Deutschland Monoclonal antibodies against amyloid beta protein and uses thereof
AU2006319358B2 (en) 2005-11-30 2012-01-19 AbbVie Deutschland GmbH & Co. KG Anti-Abeta globulomer antibodies, antigen-binding moieties thereof, corresponding hybridomas, nucleic acids, vectors, host cells, methods of producing said antibodies, compositions comprising said antibodies, uses of said antibodies and methods of using said antibodies
RS53685B1 (en) 2005-12-29 2015-04-30 Janssen Biotech Inc. Human anti-il-23 antibodies, compositions, methods and uses
CA2636149A1 (en) * 2006-01-05 2007-07-19 Novartis Ag Methods for preventing and treating cancer metastasis and bone loss associated with cancer metastasis
EP2650306A1 (en) 2006-03-06 2013-10-16 Aeres Biomedical Limited Humanized Anti-CD22 antibodies and their use in treatment of oncology, transplantation and autoimmune disease
WO2007111661A2 (en) 2006-03-20 2007-10-04 Xoma Technology Ltd. Human antibodies specific for gastrin materials and methods
US7910798B2 (en) 2006-03-31 2011-03-22 Medarex, Inc. Transgenic animals expressing chimeric antibodies for use in preparing human antibodies
AU2007238636A1 (en) 2006-04-13 2007-10-25 Novartis Vaccines & Diagnostics, Inc. Methods of treating, diagnosing or detecting cancer
TWI395754B (en) 2006-04-24 2013-05-11 Amgen Inc Humanized c-kit antibody
WO2007143098A2 (en) * 2006-06-02 2007-12-13 Aveo Pharmaceuticals, Inc. Hepatocyte growth factor (hgf) binding proteins
ATE495195T1 (en) 2006-06-02 2011-01-15 Aveo Pharmaceuticals Inc HEPATOCYTE GROWTH FACTOR (HGF) BINDING PROTEI E
BRPI0713426A2 (en) 2006-06-14 2012-10-09 Macrogenics Inc methods of treating, slowing the progression, or ameliorating one or more symptoms of a disorder, and preventing or delaying the onset of a disorder
WO2008002933A2 (en) 2006-06-26 2008-01-03 Macrogenics, Inc. Combination of fcgammariib antibodies and cd20-specific antibodies and methods of use thereof
WO2008019199A2 (en) 2006-06-26 2008-02-14 Macrogenics, Inc. FCγRIIB-SPECIFIC ANTIBODIES AND METHODS OF USE THEREOF
ES2656359T3 (en) 2006-06-30 2018-02-26 Novo Nordisk A/S Anti-NKG2A antibodies and uses thereof
RU2009107707A (en) 2006-08-04 2010-09-10 Новартис АГ (CH) SPECIFIC TO EphB3 ANTIBODY AND ITS APPLICATION
EP3415532A1 (en) 2006-08-18 2018-12-19 XOMA Technology Ltd. Prlr-specific antibody and uses thereof
KR101314362B1 (en) 2006-08-28 2013-10-10 라 졸라 인스티튜트 포 앨러지 앤드 이뮤놀로지 Antagonistic human light-specific human monoclonal antibodies
CN101522713A (en) 2006-09-01 2009-09-02 津莫吉尼蒂克斯公司 Variable region sequences of IL-31 monoclonal antibodies and methods of use
MY188368A (en) 2006-09-08 2021-12-06 Abbott Lab Interleukin-13 binding proteins
US9492518B2 (en) 2006-10-04 2016-11-15 Dana-Farber Cancer Institute, Inc. Tumor immunity
US8785400B2 (en) * 2006-11-22 2014-07-22 Curedm Group Holdings, Llc Methods and compositions relating to islet cell neogenesis
US8455626B2 (en) 2006-11-30 2013-06-04 Abbott Laboratories Aβ conformer selective anti-aβ globulomer monoclonal antibodies
EP2687232A1 (en) 2006-12-06 2014-01-22 MedImmune, LLC Methods of treating systemic lupus erythematosus
BRPI0720437A2 (en) 2006-12-07 2014-01-07 Novartis Ag ANHPHONE ANTIBODIES AGAINST EPHB3
PT2121751T (en) 2006-12-08 2017-04-18 Lexicon Pharmaceuticals Inc Monoclonal antibodies against angptl3
WO2008077145A2 (en) 2006-12-20 2008-06-26 Xoma Technology Ltd. Treatment of il-1-beta related diseases
CA2675366A1 (en) * 2007-01-18 2008-07-24 University Of Southern California Gene polymorphisms predictive for dual tki therapy
EP2486928A1 (en) 2007-02-27 2012-08-15 Abbott GmbH & Co. KG Method for the treatment of amyloidoses
JP2010521180A (en) 2007-03-14 2010-06-24 ノバルティス アーゲー APCDD1 inhibitor for treating, diagnosing or detecting cancer
CN103214577B (en) 2007-03-22 2015-09-02 生物基因Ma公司 Specific binding CD154 comprises associated proteins of antibody, antibody derivatives and antibody fragment and uses thereof
KR101540823B1 (en) 2007-03-30 2015-07-30 메디뮨 엘엘씨 Antibody formulation
EP2164868B1 (en) 2007-05-04 2015-03-25 Technophage, Investigação E Desenvolvimento Em Biotecnologia, SA Engineered rabbit antibody variable domains and uses thereof
PL2068927T3 (en) 2007-05-14 2016-06-30 Medimmune Llc Methods of reducing eosinophil levels
US20090232801A1 (en) * 2007-05-30 2009-09-17 Abbot Laboratories Humanized Antibodies Which Bind To AB (1-42) Globulomer And Uses Thereof
PE20090329A1 (en) * 2007-05-30 2009-03-27 Abbott Lab HUMANIZED ANTIBODIES AGAINST GLOBULOMER AB (20-42) AND ITS USES
EP2158221B1 (en) 2007-06-21 2018-08-29 MacroGenics, Inc. Covalent diabodies and uses thereof
EP2615113A3 (en) 2007-08-23 2013-11-13 Amgen Inc. Antigen binding proteins to proprotein convertase subtilisin kexin type 9 (PCSK9)
JOP20080381B1 (en) 2007-08-23 2023-03-28 Amgen Inc Antigen Binding Proteins to Proprotein Convertase subtillisin Kexin type 9 (pcsk9)
PT2193142E (en) * 2007-08-30 2015-04-22 Curedm Group Holdings Llc Compositions and methods of using proislet peptides and analogs thereof
DK2915564T3 (en) 2007-09-28 2021-02-08 Alexion Pharma Inc Antidotes to factor XA inhibitors and methods for their use
EP2209810A1 (en) * 2007-10-15 2010-07-28 King's College London Viral therapeutic
CN101918579A (en) * 2007-10-22 2010-12-15 先灵公司 Fully human anti-VEGF antibodies and using method
US8663980B2 (en) * 2007-10-26 2014-03-04 Janssen Biotech, Inc. Vectors, host cells, and methods of production and uses
CA2703705A1 (en) 2007-11-05 2009-05-14 Medimmune, Llc Methods of treating scleroderma
EP2225275A4 (en) * 2007-11-28 2013-04-03 Medimmune Llc Protein formulation
ME03057B (en) 2007-12-07 2019-01-20 Zymogenetics Inc Humanized antibody molecules specific for il-31
DK2222706T4 (en) 2007-12-14 2016-11-21 Novo Nordisk As Antibodies that bind to NKG2D and its use
PL2391650T3 (en) 2007-12-20 2015-03-31 Xoma Us Llc Methods for the treatment of gout
US8414893B2 (en) 2007-12-21 2013-04-09 Amgen Inc. Anti-amyloid antibodies and uses thereof
EP2242843B1 (en) * 2007-12-31 2015-05-27 XOMA Technology Ltd. Methods and materials for targeted mutagenesis
ES2613963T3 (en) 2008-01-18 2017-05-29 Medimmune, Llc Cysteine manipulated antibodies for site specific conjugation
CA2712220A1 (en) 2008-01-24 2009-07-30 Novo Nordisk A/S Humanized anti-human nkg2a monoclonal antibody
EP2574628B1 (en) 2008-01-25 2015-05-20 Amgen Inc. Ferroportin antibodies and methods of use
PT2250279E (en) 2008-02-08 2016-06-03 Medimmune Llc Anti-ifnar1 antibodies with reduced fc ligand affinity
JO2913B1 (en) * 2008-02-20 2015-09-15 امجين إنك, Antibodies directed to angiopoietin-1 and angiopoietin-2 and uses thereof
US8962803B2 (en) 2008-02-29 2015-02-24 AbbVie Deutschland GmbH & Co. KG Antibodies against the RGM A protein and uses thereof
CA2720013C (en) * 2008-04-03 2016-02-16 Bart De Strooper Single domain antibodies capable of modulating bace activity
US9908943B2 (en) 2008-04-03 2018-03-06 Vib Vzw Single domain antibodies capable of modulating BACE activity
EP2282769A4 (en) 2008-04-29 2012-04-25 Abbott Lab Dual variable domain immunoglobulins and uses thereof
US20100260668A1 (en) * 2008-04-29 2010-10-14 Abbott Laboratories Dual Variable Domain Immunoglobulins and Uses Thereof
EP2294087B1 (en) 2008-05-01 2014-05-14 Amgen, Inc. Anti-hepcidin antibodies and methods of use
ES2579554T3 (en) 2008-05-09 2016-08-12 Abbvie Deutschland Gmbh & Co Kg Antibodies for the recipient of advanced glycation terminal products (RAGE) and uses thereof
CA2725666A1 (en) 2008-06-03 2009-12-10 Abbott Laboratories Dual variable domain immunoglobulins and uses thereof
RU2010153580A (en) 2008-06-03 2012-07-20 Эбботт Лэборетриз (Us) IMMUNOGLOBULINS WITH TWO VARIABLE DOMAINS AND THEIR APPLICATION
DK2293816T3 (en) * 2008-06-06 2013-02-04 Xoma Technology Ltd METHODS OF TREATING REUMATOID ARTHRITIS
JP6146949B2 (en) 2008-06-20 2017-06-21 ノバルティス アーゲー Immunoglobulin with reduced aggregation
JP5674654B2 (en) * 2008-07-08 2015-02-25 アッヴィ・インコーポレイテッド Prostaglandin E2 double variable domain immunoglobulin and use thereof
CN104829718A (en) 2008-07-08 2015-08-12 艾伯维公司 Prostaglandin E2 binding proteins and uses thereof
NZ590634A (en) 2008-08-05 2012-07-27 Novartis Ag Compositions and methods for antibodies targeting complement protein c5
EP2341936A4 (en) 2008-09-05 2012-07-25 Xoma Technology Ltd METHODS FOR TREATING OR PREVENTING IL-1ß RELATED DISEASES
MX2011005953A (en) * 2008-12-04 2011-08-17 Abbott Lab Dual variable domain immunoglobulins and uses thereof.
KR101940059B1 (en) 2008-12-19 2019-01-18 마크로제닉스, 인크. Covalent diabodies and uses thereof
EP2376122A2 (en) 2009-01-12 2011-10-19 Takeda Pharmaceutical Company Limited Agent for prophylaxis or treatment of cancer
WO2010082134A1 (en) 2009-01-14 2010-07-22 Iq Therapeutics Bv Combination antibodies for the treatment and prevention of disease caused by bacillus anthracis and related bacteria and their toxins
US20110165063A1 (en) * 2009-01-29 2011-07-07 Abbott Laboratories Il-1 binding proteins
UA102722C2 (en) * 2009-01-29 2013-08-12 Эббви Инк. Il-1 binding proteins
US8852608B2 (en) 2009-02-02 2014-10-07 Medimmune, Llc Antibodies against and methods for producing vaccines for respiratory syncytial virus
US8030026B2 (en) * 2009-02-24 2011-10-04 Abbott Laboratories Antibodies to troponin I and methods of use thereof
US8835610B2 (en) 2009-03-05 2014-09-16 Abbvie Inc. IL-17 binding proteins
EP3604510A1 (en) 2009-03-30 2020-02-05 Portola Pharmaceuticals, Inc. Antidotes for factor xa inhibitors and methods of using the same
US8834885B2 (en) 2009-06-04 2014-09-16 Novartis Ag Methods for identification of sites for IgG conjugation
US8580493B2 (en) 2009-06-08 2013-11-12 Vib Vzw Screening for compounds that modulate GPR3-mediated beta-arrestin signaling and amyloid beta peptide generation
JP5944822B2 (en) 2009-06-15 2016-07-05 フエー・イー・ベー・フエー・ゼツト・ウエー BACE1 inhibitory antibody
BRPI1015234A2 (en) 2009-06-22 2018-02-20 Medimmune Llc fc regions designed for site specific conjugation.
BR112012000032A2 (en) 2009-07-03 2016-03-15 Bionor Immuno As innovative therapeutic and diagnostic means
EP2453910B1 (en) 2009-07-15 2016-08-31 Portola Pharmaceuticals, Inc. Unit dose formulation of antidote for factor xa inhibitors for use in preventing bleeding
WO2011009090A1 (en) * 2009-07-16 2011-01-20 Xoma Technology Ltd. Antibodies to high molecular weight melanoma associated antigen
WO2011018421A1 (en) 2009-08-10 2011-02-17 Morphosys Ag Novel screening strategies for the identification of binders
WO2011020024A2 (en) 2009-08-13 2011-02-17 The Johns Hopkins University Methods of modulating immune function
PE20121647A1 (en) 2009-08-29 2012-12-31 Abbvie Inc THERAPEUTIC BINDING PROTEINS TO DLL4
TW201119673A (en) 2009-09-01 2011-06-16 Abbott Lab Dual variable domain immunoglobulins and uses thereof
AU2010298036B2 (en) 2009-09-25 2015-05-21 Xoma Technology Ltd. Screening methods
US8926976B2 (en) 2009-09-25 2015-01-06 Xoma Technology Ltd. Modulators
TW201119676A (en) 2009-10-15 2011-06-16 Abbott Lab Dual variable domain immunoglobulins and uses thereof
UY32979A (en) * 2009-10-28 2011-02-28 Abbott Lab IMMUNOGLOBULINS WITH DUAL VARIABLE DOMAIN AND USES OF THE SAME
WO2011053707A1 (en) 2009-10-31 2011-05-05 Abbott Laboratories Antibodies to receptor for advanced glycation end products (rage) and uses thereof
RU2673908C2 (en) * 2009-12-02 2018-12-03 Имэджинэб, Инк. J591 minibodies and cys-diabodies for targeted delivery of human prostate specific membrane antigen (psma) and methods for their use
US8790887B2 (en) 2009-12-04 2014-07-29 Vib Vzw Screening methods for compounds that modulate ARF-6 mediated endosomal redistribution
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
EP2332995A1 (en) 2009-12-10 2011-06-15 Bayer Schering Pharma Aktiengesellschaft Neutralizing prolactin receptor antibodies and their therapeutic use
SI2509409T1 (en) 2009-12-10 2016-12-30 Regeneron Pharmaceuticals, Inc. Mice that make heavy chain antibodies
SG181893A1 (en) 2009-12-22 2012-08-30 Hoffmann La Roche Sequence dependent aggregation
EP2528944B1 (en) 2010-01-29 2015-08-12 MorphoSys AG Rodent combinatorial antibody libraries
CA2789629A1 (en) 2010-02-10 2011-08-18 Immunogen, Inc. Cd20 antibodies and uses thereof
JP5778700B2 (en) 2010-02-24 2015-09-16 イミュノジェン, インコーポレイテッド Folate receptor 1 antibody and immunoconjugate and use thereof
SG10201501562VA (en) 2010-03-02 2015-04-29 Abbvie Inc Therapeutic dll4 binding proteins
CA2795043C (en) 2010-03-30 2019-04-23 Janssen Biotech, Inc. Humanized il-25 antibodies
RU2568051C2 (en) 2010-04-09 2015-11-10 Авео Фармасьютикалз, Инк. ANTI-ErbB3 ANTIBODIES
WO2011127580A1 (en) 2010-04-14 2011-10-20 National Research Council Of Canada Compositions and methods for brain delivery of analgesic peptides
MX336196B (en) 2010-04-15 2016-01-11 Abbvie Inc Amyloid-beta binding proteins.
MX337586B (en) 2010-05-07 2016-03-11 Xoma Us Llc Methods for the treatment of il-1î² related conditions.
HUE033063T2 (en) 2010-05-14 2017-11-28 Abbvie Inc Il-1 binding proteins
WO2011156617A2 (en) 2010-06-09 2011-12-15 Aveo Pharmaceuticals, Inc. Anti-egfr antibodies
WO2012003472A1 (en) 2010-07-02 2012-01-05 Aveo Pharmaceuticals, Inc. Anti-notch1 antibodies
WO2012006341A2 (en) 2010-07-06 2012-01-12 Aveo Pharmaceuticals, Inc. Anti-ron antibodies
US20120009196A1 (en) 2010-07-08 2012-01-12 Abbott Laboratories Monoclonal antibodies against hepatitis c virus core protein
UY33492A (en) 2010-07-09 2012-01-31 Abbott Lab IMMUNOGLOBULINS WITH DUAL VARIABLE DOMAIN AND USES OF THE SAME
SG187682A1 (en) 2010-08-02 2013-03-28 Macrogenics Inc Covalent diabodies and uses thereof
CA2807014A1 (en) 2010-08-03 2012-02-09 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
US8956859B1 (en) 2010-08-13 2015-02-17 Aviex Technologies Llc Compositions and methods for determining successful immunization by one or more vaccines
WO2012024187A1 (en) 2010-08-14 2012-02-23 Abbott Laboratories Amyloid-beta binding proteins
US9505829B2 (en) 2010-08-19 2016-11-29 Zoetis Belgium S.A. Anti-NGF antibodies and their use
CA2809433A1 (en) 2010-08-26 2012-03-01 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
US9066986B2 (en) 2010-10-01 2015-06-30 National Research Council Of Canada Anti-CEACAM6 antibodies and uses thereof
EP2632949B1 (en) 2010-10-25 2017-09-13 National Research Council of Canada Clostridium difficile-specific antibodies and uses thereof
WO2012075111A1 (en) 2010-11-30 2012-06-07 Novartis Ag Uses of anti-cd40 antibodies in combination therapy for b cell-related cancers
RU2627171C2 (en) 2010-12-21 2017-08-03 Эббви Инк. Il-1 alpha and beta bispecific immunoglobulins with double variable domains and their application
US20120275996A1 (en) 2010-12-21 2012-11-01 Abbott Laboratories IL-1 Binding Proteins
CA2822684A1 (en) 2010-12-23 2012-06-28 Intercell Austria Ag Oprf/i agents and their use in hospitalized and other patients
WO2012092539A2 (en) 2010-12-31 2012-07-05 Takeda Pharmaceutical Company Limited Antibodies to dll4 and uses thereof
DK2658970T3 (en) 2010-12-31 2017-01-16 Bioatla Llc Express humanization of antibodies
US20120171195A1 (en) 2011-01-03 2012-07-05 Ravindranath Mepur H Anti-hla-e antibodies, therapeutic immunomodulatory antibodies to human hla-e heavy chain, useful as ivig mimetics and methods of their use
US20120189633A1 (en) 2011-01-26 2012-07-26 Kolltan Pharmaceuticals, Inc. Anti-kit antibodies and uses thereof
WO2012110500A1 (en) 2011-02-15 2012-08-23 Vib Vzw Means and methods for improvement of synaptic dysfunction disorders
KR102001686B1 (en) 2011-04-07 2019-07-18 암젠 인크 Novel egfr binding proteins
EP2699264B1 (en) 2011-04-20 2018-03-14 Medlmmune, LLC Antibodies and other molecules that bind b7-h1 and pd-1
JOP20200043A1 (en) 2011-05-10 2017-06-16 Amgen Inc Methods of treating or preventing cholesterol related disorders
US9376495B2 (en) 2011-05-21 2016-06-28 Macrogenics, Inc. Deimmunized serum-binding domains and their use in extending serum half-life
EP2530089A1 (en) 2011-06-03 2012-12-05 Bayer Pharma Aktiengesellschaft Neutralising prolactin receptor antibody Mat3 and its therapeutical use
EP2714735B1 (en) 2011-06-03 2021-07-21 XOMA Technology Ltd. Antibodies specific for tgf-beta
DK3150632T3 (en) 2011-06-10 2020-04-27 Canada Minister Nat Defence ANTI-RICIN ANTIBODIES AND APPLICATIONS THEREOF
WO2012172495A1 (en) 2011-06-14 2012-12-20 Novartis Ag Compositions and methods for antibodies targeting tem8
MX2014000531A (en) 2011-07-13 2014-12-05 Abbvie Inc Methods and compositions for treating asthma using anti-il-13 antibodies.
US9238689B2 (en) 2011-07-15 2016-01-19 Morpho Sys AG Antibodies that are cross-reactive for macrophage migration inhibitory factor (MIF) and D-dopachrome tautomerase (D-DT)
EP2734546A1 (en) 2011-07-18 2014-05-28 Amgen Inc. Apelin antigen-binding proteins and uses thereof
AU2012296613B2 (en) 2011-08-15 2016-05-12 Amplimmune, Inc. Anti-B7-H4 antibodies and their uses
WO2013025479A1 (en) 2011-08-16 2013-02-21 Emory University Jaml specific binding agents, antibodies, and uses related thereto
EP2758422A1 (en) 2011-09-23 2014-07-30 Technophage, Investigação E Desenvolvimento Em Biotecnologia, SA Modified albumin-binding domains and uses thereof to improve pharmacokinetics
PE20142361A1 (en) 2011-09-30 2015-01-16 Dana Farber Cancer Inst Inc THERAPEUTIC PEPTIDES
ES2769786T3 (en) 2011-10-14 2020-06-29 Recordati Ag Antibodies and methods for diseases related to the Wnt pathway
AR088513A1 (en) 2011-10-24 2014-06-18 Abbvie Inc IMMUNO LINKERS AGAINST SCLEROSTINE
WO2013063419A2 (en) 2011-10-28 2013-05-02 The Trustees Of The University Of Pennsylvania A fully human, anti-mesothelin specific chimeric immune receptor for redirected mesothelin-expressing cell targeting
ES2861435T3 (en) 2011-11-03 2021-10-06 Univ Pennsylvania Specific compositions of isolated B7-H4 and methods of using them
US20140322216A1 (en) 2011-11-08 2014-10-30 The Trustees Of The University Of Pennsylvania Glypican-3-specific antibody and uses thereof
JP6180425B2 (en) 2011-11-23 2017-08-23 メディミューン,エルエルシー Binding molecules specific for HER3 and their use
US10118958B2 (en) 2011-12-14 2018-11-06 AbbVie Deutschland GmbH & Co. KG Composition and method for the diagnosis and treatment of iron-related disorders
CA2855570A1 (en) 2011-12-14 2013-06-20 AbbVie Deutschland GmbH & Co. KG Composition and method for the diagnosis and treatment of iron-related disorders
EP2794657B1 (en) 2011-12-19 2017-10-11 Xoma (Us) Llc Methods for treating acne
EP2794656B1 (en) 2011-12-21 2019-02-27 Novartis AG Compositions comprising antibodies targeting factor p and c5
EP2793947B1 (en) 2011-12-23 2021-02-03 Innate Pharma Enzymatic conjugation of polypeptides
ES2721882T3 (en) 2011-12-23 2019-08-06 Pfizer Constant regions of genetically engineered antibody for site-specific conjugation and procedures and uses thereof
BR112014015851A2 (en) 2011-12-30 2019-09-24 Abbvie Inc double specific binding proteins directed against il-13 and / or il-17
AU2013208007A1 (en) 2012-01-09 2014-07-31 The Scripps Research Institute Humanized antibodies with ultralong CDR3
JP6684490B2 (en) 2012-01-09 2020-04-22 ザ・スクリップス・リサーチ・インスティテュート Ultralong complementarity determining regions and uses thereof
US20130177574A1 (en) 2012-01-11 2013-07-11 Paul I. Terasaki Foundation Laboratory ANTI-HLA CLASS-Ib ANTIBODIES MIMIC IMMUNOREACTIVITY AND IMMUNOMODULATORY FUNCTIONS OF INTRAVENOUS IMMUNOGLOBULIN (IVIg) USEFUL AS THERAPEUTIC IVIg MIMETICS AND METHODS OF THEIR USE
US10800847B2 (en) 2012-01-11 2020-10-13 Dr. Mepur Ravindranath Anti-HLA class-IB antibodies mimic immunoreactivity and immunomodulatory functions of intravenous immunoglobulin (IVIG) useful as therapeutic IVIG mimetics and methods of their use
PT2804878T (en) 2012-01-20 2018-11-29 Genzyme Corp Anti-cxcr3 antibodies
ES2676725T3 (en) 2012-01-27 2018-07-24 AbbVie Deutschland GmbH & Co. KG Composition and method for the diagnosis and treatment of diseases associated with the degeneration of neurites
US20140170159A9 (en) 2012-03-08 2014-06-19 Ge Wei Conditionally active anti-epidermal growth factor receptor antibodies and methods of use thereof
EP3626254A1 (en) 2012-03-16 2020-03-25 University Health Network Soluble toso protein and its use in treating autoimmune disorders
US9592289B2 (en) 2012-03-26 2017-03-14 Sanofi Stable IgG4 based binding agent formulations
EA039663B1 (en) 2012-05-03 2022-02-24 Амген Инк. Use of an anti-pcsk9 antibody for lowering serum cholesterol ldl and treating cholesterol related disorders
JP6629069B2 (en) 2012-06-06 2020-01-15 ゾエティス・エルエルシー Canine anti-NGF antibody and method thereof
EP3967323A3 (en) 2012-06-06 2022-05-04 Bionor Immuno AS Hiv vaccine
NZ703724A (en) 2012-06-11 2017-06-30 Amgen Inc Dual receptor antagonistic antigen-binding proteins and uses thereof
US10656156B2 (en) 2012-07-05 2020-05-19 Mepur Ravindranath Diagnostic and therapeutic potential of HLA-E monospecific monoclonal IgG antibodies directed against tumor cell surface and soluble HLA-E
US9670276B2 (en) 2012-07-12 2017-06-06 Abbvie Inc. IL-1 binding proteins
US10132799B2 (en) 2012-07-13 2018-11-20 Innate Pharma Screening of conjugated antibodies
BR112015000638A2 (en) 2012-07-13 2017-08-08 Univ Pennsylvania isolated nucleic acid sequence, cell, methods for stimulating a cell-mediated immune response to a target cell or tissue population in a mammal, for providing antitumor immunity in a mammal, for treating a mammal having a disease, disorder or condition associated with high expression of a tumor antigen
NZ630363A (en) 2012-07-25 2018-09-28 Celldex Therapeutics Inc Anti-kit antibodies and uses thereof
KR102410763B1 (en) 2012-08-17 2022-06-20 모르포시스 아게 Complex-specific antibodies and antibody fragments and its use
CA3137438A1 (en) 2012-08-31 2014-03-06 Immunogen, Inc. Diagnostic assays and kits for detection of folate receptor 1
WO2014055442A2 (en) 2012-10-01 2014-04-10 The Trustees Of The University Of Pennsylvania Compositions and methods for targeting stromal cells for the treatment of cancer
US9598489B2 (en) 2012-10-05 2017-03-21 The Trustees Of The Univeristy Of Pennsylvania Human alpha-folate receptor chimeric antigen receptor
AU2013334451A1 (en) 2012-10-24 2015-06-04 National Research Council Of Canada Anti-Campylobacter jejuni antibodies and uses therefor
SG11201503412RA (en) 2012-11-01 2015-05-28 Abbvie Inc Anti-vegf/dll4 dual variable domain immunoglobulins and uses thereof
EP3564259A3 (en) 2012-11-09 2020-02-12 Innate Pharma Recognition tags for tgase-mediated conjugation
MX368067B (en) 2012-12-05 2019-09-18 Novartis Ag Compositions and methods for antibodies targeting epo.
US9383357B2 (en) 2012-12-07 2016-07-05 Northwestern University Biomarker for replicative senescence
UY35195A (en) 2012-12-18 2014-07-31 Novartis Ag COMPOSITIONS AND METHODS FOR PROLINED ACTION PROTEINS
CA2894879A1 (en) 2012-12-19 2014-06-26 Amplimmune, Inc. B7-h4 specific antibodies, and compositions and methods of use thereof
PL2935330T3 (en) 2012-12-19 2019-11-29 Aveo Pharmaceuticals Inc Anti-notch3 antibodies
EP2935324B1 (en) 2012-12-20 2019-10-23 MorphoSys AG Anti-staphylococcal antibodies
US9550986B2 (en) 2012-12-21 2017-01-24 Abbvie Inc. High-throughput antibody humanization
CA2896091C (en) 2012-12-21 2018-06-19 Amplimmune, Inc. Anti-h7cr antibodies
EA038645B1 (en) 2012-12-21 2021-09-28 Авео Фармасьютикалз, Инк. Anti-gdf15 antibodies
EP2951199A4 (en) 2013-01-31 2016-07-20 Univ Jefferson Fusion proteins for modulating regulatory and effector t cells
PT2953969T (en) 2013-02-08 2019-12-03 Novartis Ag Anti-il-17a antibodies and their use in treating autoimmune and inflammatory disorders
SG11201505896YA (en) 2013-02-20 2015-09-29 Novartis Ag Treatment of cancer using humanized anti-egfrviii chimeric antigen receptor
UY35340A (en) 2013-02-20 2014-09-30 Novartis Ag EFFECTIVE MARKING OF HUMAN LEUKEMIA USING CELLS DESIGNED WITH AN ANTIGEN CHEMERIC RECEIVER ANTI-CD123
BR112015023239A8 (en) 2013-03-14 2018-04-17 Abbott Lab hcv antibody-antigen combination assay and methods and compositions for use thereof
MX2015012824A (en) 2013-03-14 2016-06-24 Abbott Lab Hcv ns3 recombinant antigens and mutants thereof for improved antibody detection.
CN105283201B (en) 2013-03-14 2019-08-02 斯克利普斯研究所 Targeting agent antibody coupling matter and application thereof
JP6739329B2 (en) 2013-03-14 2020-08-12 アボット・ラボラトリーズAbbott Laboratories HCV core lipid binding domain monoclonal antibody
EA201591559A1 (en) 2013-03-15 2016-03-31 Дана-Фарбер Кэнсер Инститьют, Инк. THERAPEUTIC PEPTIDES
CN105324396A (en) 2013-03-15 2016-02-10 艾伯维公司 Dual specific binding proteins directed against il-1 beta and il-17
WO2014140300A1 (en) 2013-03-15 2014-09-18 Innate Pharma Solid phase tgase-mediated conjugation of antibodies
JP6566933B2 (en) 2013-03-15 2019-08-28 サッター ベイ ホスピタルズ FALZ for use as a target for therapy to treat cancer
US9745368B2 (en) 2013-03-15 2017-08-29 The Trustees Of The University Of Pennsylvania Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy
US9446105B2 (en) 2013-03-15 2016-09-20 The Trustees Of The University Of Pennsylvania Chimeric antigen receptor specific for folate receptor β
US9469686B2 (en) 2013-03-15 2016-10-18 Abbott Laboratories Anti-GP73 monoclonal antibodies and methods of obtaining the same
TWI654206B (en) 2013-03-16 2019-03-21 諾華公司 Treatment of cancer with a humanized anti-CD19 chimeric antigen receptor
US20160060347A1 (en) 2013-04-17 2016-03-03 Morphosys Ag Antibodies targeting specifically human cxcr2
CA2913312A1 (en) 2013-05-24 2014-11-27 Medimmune, Llc Anti-b7-h5 antibodies and their uses
SG11201509982UA (en) 2013-06-06 2016-04-28 Igenica Biotherapeutics Inc
JP6581572B2 (en) 2013-06-07 2019-09-25 デューク ユニバーシティ Complement factor H inhibitor
EP3010547B1 (en) 2013-06-20 2021-04-21 Innate Pharma Enzymatic conjugation of polypeptides
AR096601A1 (en) 2013-06-21 2016-01-20 Novartis Ag ANTIBODIES OF LEXINED OXIDATED LDL RECEIVER 1 AND METHODS OF USE
UY35620A (en) 2013-06-21 2015-01-30 Novartis Ag ANTIBODIES OF LEXINED OXIDATED LDL RECEIVER 1 AND METHODS OF USE
EP3010548A1 (en) 2013-06-21 2016-04-27 Innate Pharma Enzymatic conjugation of polypeptides
AU2013396206B2 (en) 2013-06-28 2019-11-14 Amgen Inc. Methods for treating homozygous familial hypercholesterolemia
WO2015010100A2 (en) 2013-07-18 2015-01-22 Fabrus, Inc. Humanized antibodies with ultralong complementarity determining regions
WO2015017146A2 (en) 2013-07-18 2015-02-05 Fabrus, Inc. Antibodies with ultralong complementarity determining regions
WO2015007337A1 (en) 2013-07-19 2015-01-22 Bionor Immuno As Method for the vaccination against hiv
IL293871A (en) 2013-08-30 2022-08-01 Immunogen Inc Antibodies and assays for detection of folate receptor 1
US9481729B2 (en) 2013-09-11 2016-11-01 The University Of Hong Kong Anti-HER2 and anti-IGF-IR BI-specific antibodies and uses thereof
SG11201601770YA (en) 2013-09-12 2016-04-28 Halozyme Inc Modified anti-epidermal growth factor receptor antibodies and methods of use thereof
US9321834B2 (en) 2013-12-05 2016-04-26 Leidos, Inc. Anti-malarial compositions
EP3077504B1 (en) 2013-12-06 2019-08-14 Dana-Farber Cancer Institute, Inc. Therapeutic peptides
US9045545B1 (en) 2014-07-15 2015-06-02 Kymab Limited Precision medicine by targeting PD-L1 variants for treatment of cancer
US9914769B2 (en) 2014-07-15 2018-03-13 Kymab Limited Precision medicine for cholesterol treatment
US8883157B1 (en) 2013-12-17 2014-11-11 Kymab Limited Targeting rare human PCSK9 variants for cholesterol treatment
US8986691B1 (en) 2014-07-15 2015-03-24 Kymab Limited Method of treating atopic dermatitis or asthma using antibody to IL4RA
US9034332B1 (en) 2014-07-15 2015-05-19 Kymab Limited Precision medicine by targeting rare human PCSK9 variants for cholesterol treatment
US9045548B1 (en) 2014-07-15 2015-06-02 Kymab Limited Precision Medicine by targeting rare human PCSK9 variants for cholesterol treatment
US8980273B1 (en) 2014-07-15 2015-03-17 Kymab Limited Method of treating atopic dermatitis or asthma using antibody to IL4RA
US9051378B1 (en) 2014-07-15 2015-06-09 Kymab Limited Targeting rare human PCSK9 variants for cholesterol treatment
US9023359B1 (en) 2014-07-15 2015-05-05 Kymab Limited Targeting rare human PCSK9 variants for cholesterol treatment
US8945560B1 (en) 2014-07-15 2015-02-03 Kymab Limited Method of treating rheumatoid arthritis using antibody to IL6R
US9017678B1 (en) 2014-07-15 2015-04-28 Kymab Limited Method of treating rheumatoid arthritis using antibody to IL6R
US9067998B1 (en) 2014-07-15 2015-06-30 Kymab Limited Targeting PD-1 variants for treatment of cancer
US8986694B1 (en) 2014-07-15 2015-03-24 Kymab Limited Targeting human nav1.7 variants for treatment of pain
US8992927B1 (en) 2014-07-15 2015-03-31 Kymab Limited Targeting human NAV1.7 variants for treatment of pain
EP3087101A4 (en) 2013-12-20 2017-12-06 Novartis AG Regulatable chimeric antigen receptor
CA2935378C (en) 2013-12-24 2023-04-18 Janssen Pharmaceutica Nv Anti-vista antibodies and fragments
US11028143B2 (en) 2014-01-21 2021-06-08 Novartis Ag Enhanced antigen presenting ability of RNA CAR T cells by co-introduction of costimulatory molecules
RS60593B1 (en) 2014-01-24 2020-08-31 Ngm Biopharmaceuticals Inc Antibodies binding beta klotho domain 2 and methods of use thereof
EP3102701A1 (en) 2014-02-07 2016-12-14 Novartis AG Impact of genetic factors on disease progression and response to anti-c5 antibody in geographic atrophy
GB201403775D0 (en) 2014-03-04 2014-04-16 Kymab Ltd Antibodies, uses & methods
EA035472B1 (en) 2014-03-06 2020-06-22 Нэшнл Рисеч Каунсил Оф Канада Insulin-like growth factor 1 receptor-specific antibodies and uses thereof
EA035517B1 (en) 2014-03-06 2020-06-29 Нэшнл Рисеч Каунсил Оф Канада Insulin-like growth factor 1 receptor-specific antibodies and uses thereof
AU2014385800B2 (en) 2014-03-06 2020-10-22 National Research Council Of Canada Insulin-like growth factor 1 receptor -specific antibodies and uses thereof
CA2941693A1 (en) 2014-03-07 2015-09-11 University Health Network Methods and compositions for detection of targets involved in cancer metastasis
CA3124243A1 (en) 2014-03-14 2015-09-17 Dana-Farber Cancer Institute, Inc. Vaccine compositions and methods for restoring nkg2d pathway function against cancers
WO2015142675A2 (en) 2014-03-15 2015-09-24 Novartis Ag Treatment of cancer using chimeric antigen receptor
WO2015142661A1 (en) 2014-03-15 2015-09-24 Novartis Ag Regulatable chimeric antigen receptor
KR102651707B1 (en) 2014-04-07 2024-03-29 노파르티스 아게 Treatment of cancer using anti-cd19 chimeric antigen receptor
RS60795B1 (en) 2014-04-08 2020-10-30 Boston Pharmaceuticals Inc Binding molecules specific for il-21 and uses thereof
BR112016022910A2 (en) 2014-04-11 2017-10-17 Medimmune Llc bispecific her2 antibodies
GB201406767D0 (en) 2014-04-15 2014-05-28 Cancer Rec Tech Ltd Humanized anti-Tn-MUC1 antibodies anf their conjugates
US9388239B2 (en) 2014-05-01 2016-07-12 Consejo Nacional De Investigation Cientifica Anti-human VEGF antibodies with unusually strong binding affinity to human VEGF-A and cross reactivity to human VEGF-B
AU2015264114A1 (en) 2014-05-21 2016-11-03 Dana-Farber Cancer Institute, Inc. Methods for treating cancer with anti BiP or anti MICA antibodies
NZ726513A (en) 2014-05-28 2023-07-28 Memorial Sloan Kettering Cancer Center Anti-gitr antibodies and methods of use thereof
JP6768527B2 (en) 2014-06-20 2020-10-14 アベオ ファーマシューティカルズ, インコーポレイテッド Treatment of chronic kidney disease and other renal dysfunctions with GDF15 regulators
WO2015196142A1 (en) 2014-06-20 2015-12-23 Aveo Pharmaceuticals, Inc. Treatment of congestive heart failure and other cardiac dysfunction using a gdf15 modulator
US20170291939A1 (en) 2014-06-25 2017-10-12 Novartis Ag Antibodies specific for il-17a fused to hyaluronan binding peptide tags
WO2015198240A2 (en) 2014-06-25 2015-12-30 Novartis Ag Compositions and methods for long acting proteins
US20170290876A1 (en) 2014-06-25 2017-10-12 Novartis Ag Compositions and methods for long acting proteins
US9139648B1 (en) 2014-07-15 2015-09-22 Kymab Limited Precision medicine by targeting human NAV1.9 variants for treatment of pain
US9150660B1 (en) 2014-07-15 2015-10-06 Kymab Limited Precision Medicine by targeting human NAV1.8 variants for treatment of pain
US9777061B2 (en) 2014-07-21 2017-10-03 Novartis Ag Treatment of cancer using a CD33 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
JP6721568B2 (en) 2014-07-29 2020-07-15 セレクティスCellectis ROR1 (NTRKR1)-specific chimeric antigen receptor for cancer immunotherapy
CA2956482A1 (en) 2014-07-31 2016-02-04 Cellectis Ror1 specific multi-chain chimeric antigen receptor
US20170209492A1 (en) 2014-07-31 2017-07-27 Novartis Ag Subset-optimized chimeric antigen receptor-containing t-cells
EP3194437B1 (en) 2014-08-07 2021-01-20 Novartis AG Angiopoietin-like 4 (angptl4) antibodies and methods of use
PE20170287A1 (en) 2014-08-07 2017-04-05 Novartis Ag ANTI-PROTEIN ANTIBODIES SIMILAR TO ANGIOPOYETIN 4 AND METHODS OF USE
JP6919118B2 (en) 2014-08-14 2021-08-18 ノバルティス アーゲー Treatment of cancer with GFRα-4 chimeric antigen receptor
BR112017003104A2 (en) 2014-08-19 2017-12-05 Novartis Ag cancer treatment using an anti-cd123 chimeric antigen receptor
ES2777305T3 (en) 2014-09-04 2020-08-04 Cellectis Trophoblastic glycoprotein-specific chimeric antigen receptors (5T4, TPBG) for cancer immunotherapy
WO2016043577A1 (en) 2014-09-16 2016-03-24 Academisch Medisch Centrum Ig-like molecules binding to bmp4
US10577417B2 (en) 2014-09-17 2020-03-03 Novartis Ag Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy
US10080790B2 (en) 2014-09-19 2018-09-25 The Regents Of The University Of Michigan Staphylococcus aureus materials and methods
US20170306008A1 (en) 2014-09-25 2017-10-26 Aveo Pharmaceuticals, Inc. Methods of reversing cachexia and prolonging survival comprising administering a gdf15 modulator and an anti-cancer agent
US10098973B2 (en) 2014-10-10 2018-10-16 National Research Council Of Canada Anti-tau antibody and uses thereof
CA2964948A1 (en) 2014-10-31 2016-05-06 The Trustees Of The University Of Pennsylvania Altering gene expression in modified t cells and uses thereof
MX2017005976A (en) 2014-11-10 2017-06-29 Medimmune Ltd Binding molecules specific for cd73 and uses thereof.
EP3789403A1 (en) 2014-11-11 2021-03-10 MedImmune Limited Therapeutic combinations comprising anti-cd73 antibodies and a2a receptor inhibitor and uses thereof
CA2967222C (en) 2014-11-12 2023-10-31 Rinat Neuroscience Corp. Inhibitory chimeric antigen receptors
US20180334490A1 (en) 2014-12-03 2018-11-22 Qilong H. Wu Methods for b cell preconditioning in car therapy
PT3333191T (en) 2014-12-11 2020-12-15 Pf Medicament Anti-c10orf54 antibodies and uses thereof
WO2016094881A2 (en) 2014-12-11 2016-06-16 Abbvie Inc. Lrp-8 binding proteins
UY36449A (en) 2014-12-19 2016-07-29 Novartis Ag COMPOSITIONS AND METHODS FOR ANTIBODIES DIRECTED TO BMP6
US10239942B2 (en) 2014-12-22 2019-03-26 Pd-1 Acquisition Group, Llc Anti-PD-1 antibodies
US10221248B2 (en) 2014-12-22 2019-03-05 The Rockefeller University Anti-MERTK agonistic antibodies and uses thereof
CN107530423B (en) 2015-01-14 2022-04-05 布里格姆及妇女医院股份有限公司 Treatment of cancer with anti-LAP monoclonal antibodies
RU2732925C2 (en) 2015-01-26 2020-09-24 Селлектис Mat-directed chimeric antigen receptor systems for sorting/depleting the engineered immune cells
US11161907B2 (en) 2015-02-02 2021-11-02 Novartis Ag Car-expressing cells against multiple tumor antigens and uses thereof
PL3265123T3 (en) 2015-03-03 2023-03-13 Kymab Limited Antibodies, uses & methods
EP3265491A1 (en) 2015-03-03 2018-01-10 Xoma (Us) Llc Treatment of post-prandial hyperinsulinemia and hypoglycemia after bariatric surgery
KR102618312B1 (en) 2015-03-17 2023-12-28 메모리얼 슬로안 케터링 캔서 센터 Anti-MUC16 antibody and use thereof
ES2820768T3 (en) 2015-04-03 2021-04-22 Xoma Technology Ltd Cancer treatment using TGF-beta and PD-1 inhibitors
SG10201912087SA (en) 2015-04-07 2020-02-27 Alector Llc Anti-sortilin antibodies and methods of use thereof
WO2016164580A1 (en) 2015-04-07 2016-10-13 Novartis Ag Combination of chimeric antigen receptor therapy and amino pyrimidine derivatives
IL254817B2 (en) 2015-04-08 2023-12-01 Novartis Ag Cd20 therapies, cd22 therapies, and combination therapies with a cd19 chimeric antigen receptor (car) - expressing cell
EP3286211A1 (en) 2015-04-23 2018-02-28 Novartis AG Treatment of cancer using chimeric antigen receptor and protein kinase a blocker
US20180140694A1 (en) 2015-05-04 2018-05-24 Bionor Immuno As Dosage regimen for hiv vaccine
AU2016256911B2 (en) 2015-05-07 2022-03-31 Agenus Inc. Anti-OX40 antibodies and methods of use thereof
AU2016264323B2 (en) 2015-05-18 2021-02-18 TCR2 Therapeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins
EP3298046A1 (en) 2015-05-20 2018-03-28 Cellectis Anti-gd3 specific chimeric antigen receptors for cancer immunotherapy
CN107849148B (en) 2015-05-21 2023-09-19 哈普恩治疗公司 Trispecific binding proteins and methods of use
HUE050750T2 (en) 2015-05-29 2021-01-28 Agenus Inc Anti-ctla-4 antibodies and methods of use thereof
PE20180193A1 (en) 2015-05-29 2018-01-26 Abbvie Inc ANTI-CD40 ANTIBODIES AND THEIR USES
EA201792661A1 (en) 2015-06-01 2018-05-31 Медиджин Иммьюнотерапиз Гмбх METHOD OF OBTAINING ANTIBODIES AGAINST T-CELL RECEPTOR
CA2987857A1 (en) 2015-06-01 2016-12-08 Medigene Immunotherapies Gmbh T cell receptor library
TN2017000417A1 (en) 2015-06-05 2019-01-16 Novartis Ag Antibodies targeting bone morphogenetic protein 9 (bmp9) and methods therefor
US11136390B2 (en) 2015-06-12 2021-10-05 Alector Llc Anti-CD33 antibodies and methods of use thereof
JP2018518491A (en) 2015-06-12 2018-07-12 アレクトル エルエルシー Anti-CD33 antibody and method of use thereof
TW201710286A (en) 2015-06-15 2017-03-16 艾伯維有限公司 Binding proteins against VEGF, PDGF, and/or their receptors
CN107922497B (en) 2015-06-24 2022-04-12 詹森药业有限公司 anti-VISTA antibodies and fragments
JOP20200312A1 (en) 2015-06-26 2017-06-16 Novartis Ag Factor xi antibodies and methods of use
JP7203497B2 (en) 2015-06-29 2023-01-13 イミュノジェン・インコーポレーテッド Anti-CD123 Antibodies, and Conjugates and Derivatives Thereof
WO2017011275A1 (en) 2015-07-10 2017-01-19 Nersissian Aram M Factor viii protein compositions and methods of treating hemophilia a
CA2993009A1 (en) 2015-07-31 2017-02-09 Research Institute At Nationwide Children's Hospital Peptides and antibodies for the removal of biofilms
CN115043944A (en) 2015-08-03 2022-09-13 诺华股份有限公司 Methods of treating FGF 21-associated disorders
EP3331562A2 (en) 2015-08-06 2018-06-13 Xoma (Us) Llc Antibody fragments against the insulin receptor and uses thereof to treat hypoglycemia
US11667691B2 (en) 2015-08-07 2023-06-06 Novartis Ag Treatment of cancer using chimeric CD3 receptor proteins
AU2016304764C1 (en) 2015-08-07 2023-06-01 Imaginab, Inc. Antigen binding constructs to target molecules
WO2017025323A1 (en) 2015-08-11 2017-02-16 Cellectis Cells for immunotherapy engineered for targeting cd38 antigen and for cd38 gene inactivation
WO2017040790A1 (en) 2015-09-01 2017-03-09 Agenus Inc. Anti-pd-1 antibodies and methods of use thereof
AU2016332725A1 (en) 2015-09-29 2018-03-22 Celgene Corporation PD-1 binding proteins and methods of use thereof
EP3365027B1 (en) 2015-10-14 2022-03-30 Research Institute at Nationwide Children's Hospital Hu specific antibodies and their use in inhibiting biofilm
KR20180085723A (en) 2015-10-28 2018-07-27 예일 유니버시티 Humanized anti-DKK2 antibodies and uses thereof
TWI726936B (en) 2015-11-10 2021-05-11 美商麥迪紐有限責任公司 Binding molecules specific for asct2 and uses thereof
US11052111B2 (en) 2015-12-08 2021-07-06 Chimera Bioengineering, Inc. Smart CAR devices and DE CAR polypeptides for treating disease and methods for enhancing immune responses
CN109069623A (en) 2015-12-18 2018-12-21 诺华股份有限公司 Target the antibody and its application method of CD32b
HUE053674T2 (en) 2015-12-23 2021-07-28 Medigene Immunotherapies Gmbh Novel generation of antigen-specific tcrs
CN109195988B (en) 2016-01-10 2023-12-01 尼奥克斯医疗有限公司 Methods and compositions for enhancing the efficacy of superantigen-mediated cancer immunotherapy
WO2017125897A1 (en) 2016-01-21 2017-07-27 Novartis Ag Multispecific molecules targeting cll-1
TWI756204B (en) 2016-02-12 2022-03-01 比利時商楊森製藥公司 Anti-vista antibodies and fragments, uses thereof, and methods of identifying same
KR20180118175A (en) 2016-03-04 2018-10-30 노파르티스 아게 Cells expressing multiple chimeric antigen receptor (CAR) molecules and their uses
JP6987072B2 (en) 2016-03-10 2021-12-22 アクセレロン ファーマ インコーポレーテッド Activin type 2 receptor binding protein and its use
WO2017161206A1 (en) 2016-03-16 2017-09-21 Halozyme, Inc. Conjugates containing conditionally active antibodies or antigen-binding fragments thereof, and methods of use
EP3432924A1 (en) 2016-03-23 2019-01-30 Novartis AG Cell secreted minibodies and uses thereof
US20190112380A1 (en) 2016-03-29 2019-04-18 University Of Southern California Chimeric antigen receptors targeting cancer
WO2017175058A1 (en) 2016-04-07 2017-10-12 Janssen Pharmaceutica Nv Anti-vista antibodies and fragments, uses thereof, and methods of identifying same
HRP20230457T1 (en) 2016-04-15 2023-07-21 Novartis Ag Compositions and methods for selective expression of chimeric antigen receptors
CN109071647B (en) 2016-04-27 2022-11-22 诺华股份有限公司 Anti-growth differentiation factor 15 antibody and application thereof
CA3021334C (en) 2016-04-27 2022-11-29 Abbvie Inc. Methods of treatment of diseases in which il-13 activity is detrimental using anti-il-13 antibodies
US11623958B2 (en) 2016-05-20 2023-04-11 Harpoon Therapeutics, Inc. Single chain variable fragment CD3 binding proteins
WO2017201488A1 (en) 2016-05-20 2017-11-23 Harpoon Therapeutics, Inc. Single domain serum albumin binding protein
TW201802121A (en) 2016-05-25 2018-01-16 諾華公司 Reversal binding agents for anti-factor XI/XIa antibodies and uses thereof
KR102366813B1 (en) 2016-05-27 2022-02-24 아게누스 인코포레이티드 Anti-TIM-3 Antibodies and Methods of Using Same
EP3464375A2 (en) 2016-06-02 2019-04-10 Novartis AG Therapeutic regimens for chimeric antigen receptor (car)- expressing cells
MA51586A (en) 2016-06-02 2019-04-10 Abbvie Inc GLUCOCORTICOID RECEPTOR AND IMMUNOCONJUGATE AGONIST
JP2019526529A (en) 2016-06-08 2019-09-19 アッヴィ・インコーポレイテッド Anti-B7-H3 antibody and antibody drug conjugate
CN109963870B (en) 2016-06-08 2023-07-28 艾伯维公司 anti-B7-H3 antibodies and antibody drug conjugates
JP2019521973A (en) 2016-06-08 2019-08-08 アッヴィ・インコーポレイテッド Anti-BH7-H3 antibody and antibody drug conjugate
GB201610198D0 (en) 2016-06-10 2016-07-27 Ucb Biopharma Sprl Anti-ige antibodies
CN110381988A (en) 2016-06-15 2019-10-25 诺华股份有限公司 Use the method for the inhibitor for treating disease of Bone Morphogenetic Protein 6 (BMP6)
WO2017218891A1 (en) 2016-06-17 2017-12-21 Life Technologies Corporation Site-specific crosslinking of antibodies
AU2017292184A1 (en) 2016-07-08 2019-02-07 Staten Biotechnology B.V. Anti-Apoc3 antibodies and methods of use thereof
CN110461315A (en) 2016-07-15 2019-11-15 诺华股份有限公司 Cytokines release syndrome is treated and prevented using with the Chimeric antigen receptor of kinase inhibitor combination
JP7178568B2 (en) 2016-07-18 2022-11-28 ナショナル リサーチ カウンシル オブ カナダ CAR immune cells targeting carcinoembryonic antigen-related cell adhesion molecule 6 for treating cancer
BR112019001570A2 (en) 2016-07-28 2019-07-09 Novartis Ag chimeric antigen receptor combination therapies and pd-1 inhibitors
BR112019002035A2 (en) 2016-08-01 2019-05-14 Novartis Ag cancer treatment using a chimeric antigen receptor in combination with an inhibitor of a m2 pro-macrophage molecule
EP3490600A1 (en) 2016-08-01 2019-06-05 Xoma (Us) Llc Parathyroid hormone receptor 1 (pth1r) antibodies and uses thereof
AU2017306432A1 (en) 2016-08-02 2019-03-21 TCR2 Therapeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins
US11878060B2 (en) 2016-08-07 2024-01-23 Novartis Ag mRNA-mediated immunization methods
CN109952317A (en) 2016-09-19 2019-06-28 细胞基因公司 Use the method for PD-1 binding protein treatment immune disorders
JP2019534859A (en) 2016-09-19 2019-12-05 セルジーン コーポレイション Method for treating vitiligo using PD-1 binding protein
EP3515478B1 (en) 2016-09-21 2024-02-28 Nextcure, Inc. Antibodies for siglec-15 and methods of use thereof
CA3038679A1 (en) 2016-09-28 2018-04-05 Xoma (Us) Llc Antibodies that bind interleukin-2 and uses thereof
CA3037961A1 (en) 2016-09-30 2018-04-05 Janssen Biotech, Inc. Safe and effective method of treating psoriasis with anti-il23 specific antibody
MX2019003473A (en) 2016-10-03 2019-10-15 Abbott Lab Improved methods of assessing uch-l1 status in patient samples.
WO2018067754A1 (en) 2016-10-04 2018-04-12 Fairbanks Pharmaceuticals, Inc. Anti-fstl3 antibodies and uses thereof
US10525083B2 (en) 2016-10-07 2020-01-07 Novartis Ag Nucleic acid molecules encoding chimeric antigen receptors comprising a CD20 binding domain
ES2875959T3 (en) 2016-10-07 2021-11-11 Tcr2 Therapeutics Inc Compositions and methods for T-cell receptor reprogramming using fusion proteins
IL265800B2 (en) 2016-10-11 2023-10-01 Agenus Inc Anti-lag-3 antibodies and methods of use thereof
US11779604B2 (en) 2016-11-03 2023-10-10 Kymab Limited Antibodies, combinations comprising antibodies, biomarkers, uses and methods
AU2017353939A1 (en) 2016-11-07 2019-06-06 Neuracle Science Co., Ltd. Anti-family with sequence similarity 19, member A5 antibodies and method of use thereof
KR20190078648A (en) 2016-11-16 2019-07-04 얀센 바이오테크 인코포레이티드 Methods for treating psoriasis with anti-IL23 specific antibodies
AU2017363311A1 (en) 2016-11-22 2019-06-13 TCR2 Therapeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins
AU2017363309A1 (en) 2016-11-23 2019-07-11 Bioverativ Therapeutics Inc. Mono- and bispecific antibodies binding to coagulation factor IX and coagulation factor X
EP3544628A4 (en) 2016-11-23 2020-11-18 Immunoah Therapeutics, Inc. 4-1bb binding proteins and uses thereof
JP7215997B2 (en) 2016-11-23 2023-01-31 ハープーン セラピューティクス,インク. Trispecific proteins targeting prostate specific membrane antigen (PSMA) and methods of use
WO2018098354A1 (en) 2016-11-23 2018-05-31 Harpoon Therapeutics, Inc. Prostate specific membrane antigen binding protein
MA50949B1 (en) 2016-12-07 2023-12-29 Memorial Sloan Kettering Cancer Center ANTI-CTLA-4 ANTIBODIES AND METHODS OF USE THEREOF
KR102603681B1 (en) 2016-12-07 2023-11-17 아게누스 인코포레이티드 Antibodies and methods of using them
JP7110199B2 (en) 2016-12-23 2022-08-01 ノバルティス アーゲー Methods of treatment with anti-factor XI/XIa antibodies
IL308980A (en) 2016-12-23 2024-01-01 Novartis Ag Factor xi antibodies and methods of use
AU2018206560A1 (en) 2017-01-04 2019-07-18 Research Institute At Nationwide Children's Hospital Antibody fragments for the treatment of biofilm-related disorders
WO2018129029A1 (en) 2017-01-04 2018-07-12 Immunogen, Inc. Met antibodies and immunoconjugates and uses thereof
CA3049105A1 (en) 2017-01-04 2018-07-12 Lauren O. Bakaletz Dnabii vaccines and antibodies with enhanced activity
WO2018127791A2 (en) 2017-01-06 2018-07-12 Biosion, Inc. Erbb2 antibodies and uses therefore
US11535662B2 (en) 2017-01-26 2022-12-27 Novartis Ag CD28 compositions and methods for chimeric antigen receptor therapy
EP3573658A4 (en) 2017-01-30 2021-07-21 Janssen Biotech, Inc. Anti-tnf antibodies, compositions, and methods for the treatment of active psoriatic arthritis
CN110582509A (en) 2017-01-31 2019-12-17 诺华股份有限公司 Treatment of cancer using chimeric T cell receptor proteins with multispecific properties
JP2020506947A (en) 2017-02-07 2020-03-05 ヤンセン バイオテツク,インコーポレーテツド Anti-TNF antibodies, compositions and methods for treating active ankylosing spondylitis
KR102572663B1 (en) 2017-02-08 2023-09-01 노파르티스 아게 FGF21 Mimetic Antibodies and Uses Thereof
US11266745B2 (en) 2017-02-08 2022-03-08 Imaginab, Inc. Extension sequences for diabodies
WO2018160754A2 (en) 2017-02-28 2018-09-07 Harpoon Therapeutics, Inc. Inducible monovalent antigen binding protein
AU2018236271B2 (en) 2017-03-15 2023-12-21 Research Institute At Nationwide Children's Hospital Composition and methods for disruption of bacterial biofilms without accompanying inflammation
CA3052513A1 (en) 2017-03-23 2018-09-27 Abbott Laboratories Methods for aiding in the diagnosis and determination of the extent of traumatic brain injury in a human subject using the early biomarker ubiquitin carboxy-terminal hydrolase l1
AU2018244276A1 (en) 2017-03-27 2019-10-17 Celgene Corporation Methods and compositions for reduction of immunogenicity
TW201841942A (en) 2017-04-13 2018-12-01 美商艾吉納斯公司 Anti-CD137 antibodies and methods of use thereof
JP7344797B2 (en) 2017-04-15 2023-09-14 アボット・ラボラトリーズ Methods to aid in hyperacute diagnosis and determination of traumatic brain injury in human subjects using early biomarkers
EP3612560A1 (en) 2017-04-21 2020-02-26 Staten Biotechnology B.V. Anti-apoc3 antibodies and methods of use thereof
WO2018196782A1 (en) 2017-04-27 2018-11-01 The University Of Hong Kong Use of hcn inhibitors for treatment of cancer
EP3615055A1 (en) 2017-04-28 2020-03-04 Novartis AG Cells expressing a bcma-targeting chimeric antigen receptor, and combination therapy with a gamma secretase inhibitor
CN110603449A (en) 2017-04-28 2019-12-20 雅培实验室 Method for determining traumatic brain injury using early biomarkers from at least two samples of the same human subject for aiding hyperacute diagnosis
MA50957A (en) 2017-05-01 2020-10-14 Agenus Inc ANTI-TIGIT ANTIBODIES AND THEIR METHODS OF USE
US10865238B1 (en) 2017-05-05 2020-12-15 Duke University Complement factor H antibodies
EP3621648A4 (en) 2017-05-12 2021-01-20 Harpoon Therapeutics, Inc. Msln targeting trispecific proteins and methods of use
JOP20190256A1 (en) 2017-05-12 2019-10-28 Icahn School Med Mount Sinai Newcastle disease viruses and uses thereof
KR102376863B1 (en) 2017-05-12 2022-03-21 하푼 테라퓨틱스, 인크. mesothelin binding protein
CN110651190A (en) 2017-05-25 2020-01-03 雅培实验室 Method for using early biomarkers to help determine whether to perform imaging on a human subject who has suffered or may have suffered a head injury
AU2018275235A1 (en) 2017-05-30 2019-10-31 Abbott Laboratories Methods for aiding in diagnosing and evaluating a mild traumatic brain injury in a human subject using cardiac troponin I and early biomarkers
WO2018224441A1 (en) 2017-06-05 2018-12-13 Numab Innovation Ag Novel anti-cd3 antibodies
EP3638295A1 (en) 2017-06-13 2020-04-22 TCR2 Therapeutics Inc. Compositions and methods for tcr reprogramming using fusion proteins
WO2018229715A1 (en) 2017-06-16 2018-12-20 Novartis Ag Compositions comprising anti-cd32b antibodies and methods of use thereof
WO2018229706A1 (en) 2017-06-16 2018-12-20 Novartis Ag Combination therapy for the treatment of cancer
WO2019005756A1 (en) 2017-06-28 2019-01-03 The Rockefeller University Anti-mertk agonistic antibody-drug conjugates and uses thereof
EP3649474A1 (en) 2017-07-03 2020-05-13 Abbott Laboratories Improved methods for measuring ubiquitin carboxy-terminal hydrolase l1 levels in blood
CN117050176A (en) 2017-07-31 2023-11-14 豪夫迈·罗氏有限公司 Humanization method based on three-dimensional structure
CN110662765B (en) 2017-08-03 2023-09-29 艾利妥 anti-CD 33 antibodies and methods of use thereof
EP3679070A1 (en) 2017-09-07 2020-07-15 Augusta University Research Institute, Inc. Antibodies to programmed cell death protein 1
TW201922780A (en) 2017-09-25 2019-06-16 美商健生生物科技公司 Safe and effective method of treating Lupus with anti-IL12/IL23 antibody
EP3692370A2 (en) 2017-10-04 2020-08-12 OPKO Pharmaceuticals, LLC Articles and methods directed to personalized therapy of cancer
CN111417720A (en) * 2017-10-05 2020-07-14 美国卫生和人力服务部 Methods for selectively amplifying cells expressing a TCR with a murine constant region
US11578133B2 (en) 2017-10-10 2023-02-14 Namab Therapeutics AG Antibodies targeting CD137 and methods of use thereof
EP3470428A1 (en) 2017-10-10 2019-04-17 Numab Innovation AG Antibodies targeting cd137 and methods of use thereof
KR20200063155A (en) 2017-10-10 2020-06-04 누맙 세러퓨틱스 아게 Multispecific antibodies
EP3470426A1 (en) 2017-10-10 2019-04-17 Numab Therapeutics AG Multispecific antibody
EP3694879A1 (en) 2017-10-10 2020-08-19 Numab Therapeutics AG Antibodies targeting pdl1 and methods of use thereof
EP3470429A1 (en) 2017-10-10 2019-04-17 Numab Innovation AG Antibodies targeting pdl1 and methods of use thereof
WO2019075090A1 (en) 2017-10-10 2019-04-18 Tilos Therapeutics, Inc. Anti-lap antibodies and uses thereof
EA202090739A1 (en) 2017-10-13 2020-09-07 Харпун Терапьютикс, Инк. PROTEINS BINDING ANTIGEN OF MATURING B-CELLS
CN111630070A (en) 2017-10-13 2020-09-04 哈普恩治疗公司 Trispecific proteins and methods of use
EP3700933A1 (en) 2017-10-25 2020-09-02 Novartis AG Antibodies targeting cd32b and methods of use thereof
BR112020008514A2 (en) 2017-10-31 2020-10-20 Staten Biotechnology B.V. anti-apoc3 antibodies and methods of using them
EP3713965A1 (en) 2017-11-22 2020-09-30 Novartis AG Reversal binding agents for anti-factor xi/xia antibodies and uses thereof
CN111670052A (en) * 2017-11-29 2020-09-15 美真达治疗公司 Compositions and methods for depleting CD5+ cells
WO2019112860A1 (en) 2017-12-09 2019-06-13 Abbott Laboratories Methods for aiding in diagnosing and evaluating a traumatic brain injury in a human subject using a combination of gfap and uch-l1
WO2019113525A2 (en) 2017-12-09 2019-06-13 Abbott Laboratories Methods for aiding in the diagnosis and evaluation of a subject who has sustained an orthopedic injury and that has or may have sustained an injury to the head, such as mild traumatic brain injury (tbi), using glial fibrillary acidic protein (gfap) and/or ubiquitin carboxy-terminal hydrolase l1 (uch-l1)
WO2019129054A1 (en) 2017-12-27 2019-07-04 信达生物制药(苏州)有限公司 Triabody, preparation method and use thereof
EP3732193A1 (en) 2017-12-29 2020-11-04 Alector LLC Anti-tmem106b antibodies and methods of use thereof
US11472874B2 (en) 2018-01-31 2022-10-18 Alector Llc Anti-MS4A4A antibodies and methods of use thereof
WO2019150309A1 (en) 2018-02-02 2019-08-08 Hammack Scott Modulators of gpr68 and uses thereof for treating and preventing diseases
CN111989117A (en) 2018-02-14 2020-11-24 维埃拉生物股份有限公司 Antibodies to the ligand of the mcdonald cat sarcoma (FMS) -like tyrosine kinase 3 receptor (FLT3L) and their use for the treatment of autoimmune and inflammatory diseases
WO2019169229A1 (en) 2018-03-01 2019-09-06 Nextcure, Inc. Klrg1 binding compositions and methods of use thereof
KR20200129125A (en) 2018-03-05 2020-11-17 얀센 바이오테크 인코포레이티드 How to treat Crohn's disease with anti-IL23 specific antibodies
WO2019177690A1 (en) 2018-03-12 2019-09-19 Zoetis Services Llc Anti-ngf antibodies and methods thereof
GB201804701D0 (en) 2018-03-23 2018-05-09 Gammadelta Therapeutics Ltd Lymphocytes expressing heterologous targeting constructs
WO2019184909A1 (en) 2018-03-27 2019-10-03 信达生物制药(苏州)有限公司 Novel antibody molecule, and preparation method and use thereof
WO2019210153A1 (en) 2018-04-27 2019-10-31 Novartis Ag Car t cell therapies with enhanced efficacy
SG11202010388SA (en) 2018-04-30 2020-11-27 Takeda Pharmaceuticals Co Cannabinoid receptor type 1 (cb1) binding proteins and uses thereof
US20210396739A1 (en) 2018-05-01 2021-12-23 Novartis Ag Biomarkers for evaluating car-t cells to predict clinical outcome
WO2019226973A1 (en) 2018-05-25 2019-11-28 Alector Llc Anti-sirpa antibodies and methods of use thereof
WO2019227003A1 (en) 2018-05-25 2019-11-28 Novartis Ag Combination therapy with chimeric antigen receptor (car) therapies
EP3802611A2 (en) 2018-06-01 2021-04-14 Novartis AG Binding molecules against bcma and uses thereof
US11884729B2 (en) 2018-06-29 2024-01-30 ApitBio, Inc Anti-L1CAM antibodies and uses thereof
US20210277113A1 (en) 2018-06-29 2021-09-09 Alector Llc Anti-SIRP-Beta1 Antibodies and Methods of Use Thereof
WO2020014306A1 (en) 2018-07-10 2020-01-16 Immunogen, Inc. Met antibodies and immunoconjugates and uses thereof
ES2940311T3 (en) 2018-07-13 2023-05-05 Alector Llc Anti-sortilin antibodies and methods of use thereof
WO2020016838A2 (en) 2018-07-18 2020-01-23 Janssen Biotech, Inc. Sustained response predictors after treatment with anti-il23 specific antibody
WO2020016459A1 (en) 2018-07-20 2020-01-23 Pierre Fabre Medicament Receptor for vista
WO2020041360A1 (en) 2018-08-21 2020-02-27 Quidel Corporation Dbpa antibodies and uses thereof
JP2021535140A (en) 2018-08-28 2021-12-16 アンブルックス, インコーポレイテッドAmbrx, Inc. Anti-CD3 antibody folic acid biocomplex and its use
US20210253666A1 (en) 2018-08-30 2021-08-19 TCR2 Therapeutics Inc. Compositions and methods for tcr reprogramming using fusion proteins
MX2021002299A (en) 2018-08-31 2021-04-28 Alector Llc Anti-cd33 antibodies and methods of use thereof.
WO2020044252A1 (en) 2018-08-31 2020-03-05 Novartis Ag Dosage regimes for anti-m-csf antibodies and uses thereof
WO2020056170A1 (en) 2018-09-12 2020-03-19 Fred Hutchinson Cancer Research Center Reducing cd33 expression to selectively protect therapeutic cells
AU2019342017A1 (en) 2018-09-18 2021-05-13 Merrimack Pharmaceuticals, Inc. Anti-TNFR2 antibodies and uses thereof
SI3883606T1 (en) 2018-09-24 2023-10-30 Janssen Biotech, Inc. Safe and effective method of treating ulcerative colitis with anti-il12/il23 antibody
SG11202103022WA (en) 2018-09-25 2021-04-29 Harpoon Therapeutics Inc Dll3 binding proteins and methods of use
EP3856779A1 (en) 2018-09-28 2021-08-04 Novartis AG Cd22 chimeric antigen receptor (car) therapies
EP3856773A1 (en) 2018-09-28 2021-08-04 Kyowa Kirin Co., Ltd. Il-36 antibodies and uses thereof
US20210347851A1 (en) 2018-09-28 2021-11-11 Novartis Ag Cd19 chimeric antigen receptor (car) and cd22 car combination therapies
JP2022504287A (en) 2018-10-03 2022-01-13 スターテン・バイオテクノロジー・ベー・フェー Antibodies specific for human and cynomolgus monkey APOC3, and methods of their use
CA3114925A1 (en) 2018-10-05 2020-04-09 Research Institute At Nationwide Children's Hospital Compositions and methods for enzymatic disruption of bacterial biofilms
EP3636320A1 (en) 2018-10-09 2020-04-15 Numab Therapeutics AG Antibodies targeting cd137 and methods of use thereof
TW202035457A (en) 2018-10-09 2020-10-01 瑞士商Numab治療公司 Antibodies targeting cd137 and methods of use thereof
WO2020076969A2 (en) 2018-10-10 2020-04-16 Tilos Therapeutics, Inc. Anti-lap antibody variants and uses thereof
WO2020092455A2 (en) 2018-10-29 2020-05-07 The Broad Institute, Inc. Car t cell transcriptional atlas
EP3883961A1 (en) 2018-11-20 2021-09-29 Takeda Vaccines, Inc. Novel anti-zika virus antibodies and uses thereof
US11548941B2 (en) 2018-11-20 2023-01-10 Janssen Biotech, Inc. Safe and effective method of treating psoriasis with anti-IL-23 specific antibody
WO2020118293A2 (en) 2018-12-07 2020-06-11 Georgia Tech Research Corporation Antibodies that bind to natively folded myocilin
EP3897722A4 (en) 2018-12-18 2022-09-14 Janssen Biotech, Inc. Safe and effective method of treating lupus with anti-il12/il23 antibody
WO2020128863A1 (en) 2018-12-19 2020-06-25 Novartis Ag Anti-tnf-alpha antibodies
JP2022514903A (en) 2018-12-20 2022-02-16 協和キリン株式会社 FN14 antibody and its use
US20220090125A1 (en) 2018-12-21 2022-03-24 Compass Therapeutics Llc Transgenic mouse expressing common human light chain
EP3911676A1 (en) 2019-01-15 2021-11-24 Janssen Biotech, Inc. Anti-tnf antibody compositions and methods for the treatment of juvenile idiopathic arthritis
KR20210118878A (en) 2019-01-23 2021-10-01 얀센 바이오테크 인코포레이티드 Anti-TNF antibody composition for use in a method of treating psoriatic arthritis
TWI756621B (en) 2019-01-25 2022-03-01 大陸商信達生物製藥(蘇州)有限公司 Novel bispecific antibody molecules and bispecific antibodies that simultaneously bind pd-l1 and lag-3
EP3689907A1 (en) 2019-01-31 2020-08-05 Numab Therapeutics AG Antibodies targeting il-17a and methods of use thereof
EP3917954A1 (en) 2019-01-31 2021-12-08 Numab Therapeutics AG Multispecific antibodies having specificity for tnfa and il-17a, antibodies targeting il-17a, and methods of use thereof
WO2020172553A1 (en) 2019-02-22 2020-08-27 Novartis Ag Combination therapies of egfrviii chimeric antigen receptors and pd-1 inhibitors
US11242407B2 (en) 2019-02-26 2022-02-08 Inspirna, Inc. High-affinity anti-MERTK antibodies and uses thereof
CN113874083A (en) 2019-03-01 2021-12-31 梅里麦克制药股份有限公司 anti-TNFR 2 antibodies and uses thereof
KR20210141998A (en) 2019-03-14 2021-11-23 얀센 바이오테크 인코포레이티드 Method of making anti-TNF antibody composition
MA55282A (en) 2019-03-14 2022-01-19 Janssen Biotech Inc MANUFACTURING METHODS FOR THE PRODUCTION OF ANTI-TNF ANTIBODY COMPOSITIONS
MA55283A (en) 2019-03-14 2022-01-19 Janssen Biotech Inc METHODS FOR PRODUCING ANTI-TNF ANTIBODY COMPOSITIONS
JP2022525145A (en) 2019-03-14 2022-05-11 ヤンセン バイオテツク,インコーポレーテツド A production method for producing an anti-IL12 / IL23 antibody composition.
EA202192459A1 (en) 2019-03-18 2021-11-25 Янссен Байотек, Инк. METHOD FOR TREATMENT OF PSORIASIS WITH ANTIBODY TO IL12 / IL23 IN CHILDREN
WO2020198731A2 (en) 2019-03-28 2020-10-01 Danisco Us Inc Engineered antibodies
US20230135930A1 (en) 2019-04-24 2023-05-04 Heidelberg Pharma Research Gmbh Amatoxin antibody-drug conjugates and uses thereof
MX2021013908A (en) 2019-05-15 2022-03-11 Neotx Therapeutics Ltd Cancer treatment.
WO2020236797A1 (en) 2019-05-21 2020-11-26 Novartis Ag Variant cd58 domains and uses thereof
CR20210576A (en) 2019-05-21 2021-12-15 Novartis Ag Cd19 binding molecules and uses thereof
KR20220012883A (en) 2019-05-23 2022-02-04 얀센 바이오테크 인코포레이티드 A method of treating inflammatory bowel disease with a combination therapy of IL-23 and an antibody against TNF alpha
EP3976648A1 (en) 2019-06-03 2022-04-06 Janssen Biotech, Inc. Anti-tnf antibody compositions, and methods for the treatment of psoriatic arthritis
CA3142580A1 (en) 2019-06-03 2020-12-10 Janssen Biotech, Inc. Anti-tnf antibodies, compositions, and methods for the treatment of active ankylosing spondylitis
EP3983441A1 (en) 2019-06-11 2022-04-20 Alector LLC Anti-sortilin antibodies for use in therapy
CA3138188A1 (en) 2019-07-02 2021-01-07 Fred Hutchinson Cancer Research Center Recombinant ad35 vectors and related gene therapy improvements
CA3145385A1 (en) 2019-07-08 2021-01-14 Steven D. Goodman Antibody compositions for disrupting biofilms
BR112022001733A2 (en) 2019-07-31 2022-06-28 Alector Llc ANTI-MS4A4A ANTIBODIES AND METHODS OF USE THEREOF
CN114867751A (en) 2019-08-12 2022-08-05 阿帕特夫研究和发展有限公司 4-1BB and OX40 binding proteins and related compositions and methods, anti-4-1 BB antibodies, anti-OX 40 antibodies
WO2021028752A1 (en) 2019-08-15 2021-02-18 Janssen Biotech, Inc. Anti-tfn antibodies for treating type i diabetes
WO2021035170A1 (en) 2019-08-21 2021-02-25 Precision Biosciences, Inc. Compositions and methods for tcr reprogramming using fusion proteins
WO2021042019A1 (en) 2019-08-30 2021-03-04 Agenus Inc. Anti-cd96 antibodies and methods of use thereof
CN115551594A (en) 2019-10-24 2022-12-30 米诺陶治疗公司 Cytokine-modified chimeric antibodies and methods of use thereof
EP3816185A1 (en) 2019-11-04 2021-05-05 Numab Therapeutics AG Multispecific antibody directed against pd-l1 and a tumor-associated antigen
US11897950B2 (en) 2019-12-06 2024-02-13 Augusta University Research Institute, Inc. Osteopontin monoclonal antibodies
CR20220329A (en) 2019-12-13 2022-11-23 Alector Llc Anti-mertk antibodies and methods of use thereof
EP4077376A2 (en) 2019-12-19 2022-10-26 Quidel Corporation Monoclonal antibody fusions
TW202140550A (en) 2020-01-29 2021-11-01 瑞士商諾華公司 Methods of treating an inflammatory or obstructive airway disease using anti-tslp antibody
WO2021155295A1 (en) 2020-01-31 2021-08-05 The Cleveland Clinic Foundation Anti-müllerian hormone receptor 2 antibodies and methods of use
IL295387A (en) 2020-02-05 2022-10-01 Larimar Therapeutics Inc Tat peptide binding proteins and uses thereof
US11180563B2 (en) 2020-02-21 2021-11-23 Harpoon Therapeutics, Inc. FLT3 binding proteins and methods of use
WO2021173889A1 (en) 2020-02-26 2021-09-02 Ambrx, Inc. Uses of anti-cd3 antibody folate bioconjugates
JP2023519962A (en) 2020-03-31 2023-05-15 アレクトル エルエルシー ANTI-MERTK ANTIBODY AND METHOD OF USE THEREOF
EP4132971A1 (en) 2020-04-09 2023-02-15 Merck Sharp & Dohme LLC Affinity matured anti-lap antibodies and uses thereof
CA3175523A1 (en) 2020-04-13 2021-10-21 Antti Virtanen Methods, complexes and kits for detecting or determining an amount of a .beta.-coronavirus antibody in a sample
EP3915580A1 (en) 2020-05-29 2021-12-01 Numab Therapeutics AG Multispecific antibody
AU2021288224A1 (en) 2020-06-11 2023-01-05 Novartis Ag ZBTB32 inhibitors and uses thereof
WO2022029494A1 (en) 2020-08-04 2022-02-10 Abbott Rapid Diagnostics International Unlimited Company Assays for detecting sars-cov-2
EP4193149A1 (en) 2020-08-04 2023-06-14 Abbott Laboratories Improved methods and kits for detecting sars-cov-2 protein in a sample
CA3193594A1 (en) 2020-09-11 2022-03-17 Medimmune Limited Therapeutic b7-h4 binding molecules
CA3194182A1 (en) 2020-09-12 2022-03-17 Medimmune Limited A scoring method for an anti-b7h4 antibody-drug conjugate therapy
US20240010750A1 (en) 2020-09-15 2024-01-11 Bayer Aktiengesellschaft Novel anti-a2ap antibodies and uses thereof
EP3988568A1 (en) 2020-10-21 2022-04-27 Numab Therapeutics AG Combination treatment
US20240025993A1 (en) 2020-11-06 2024-01-25 Novartis Ag Cd19 binding molecules and uses thereof
WO2022097061A1 (en) 2020-11-06 2022-05-12 Novartis Ag Anti-cd19 agent and b cell targeting agent combination therapy for treating b cell malignancies
EP4240765A2 (en) 2020-11-06 2023-09-13 Novartis AG Antibody fc variants
US20240033358A1 (en) 2020-11-13 2024-02-01 Novartis Ag Combination therapies with chimeric antigen receptor (car)-expressing cells
IL303328A (en) 2020-12-01 2023-07-01 Aptevo Res & Development Llc Heterodimeric psma and cd3-binding bispecific antibodies
US20220170948A1 (en) 2020-12-01 2022-06-02 Abbott Laboratories Use of one or more biomarkers to determine traumatic brain injury (tbi) in a human subject having received a head computerized tomography scan that is negative for a tbi
WO2023102384A1 (en) 2021-11-30 2023-06-08 Abbott Laboratories Use of one or more biomarkers to determine traumatic brain injury (tbi) in a subject having received a head computerized tomography scan that is negative for a tbi
JP2023553399A (en) 2020-12-02 2023-12-21 アレクトル エルエルシー How to use anti-Sortilin antibodies
TW202237638A (en) 2020-12-09 2022-10-01 日商武田藥品工業股份有限公司 Compositions of guanylyl cyclase c (gcc) antigen binding agents and methods of use thereof
TW202237639A (en) 2020-12-09 2022-10-01 日商武田藥品工業股份有限公司 Compositions of guanylyl cyclase c (gcc) antigen binding agents and methods of use thereof
EP4019546A1 (en) 2020-12-23 2022-06-29 Numab Therapeutics AG Antibody variable domains that bind il-31
EP4019547A1 (en) 2020-12-23 2022-06-29 Numab Therapeutics AG Multispecific antibodies having specificity for il-4r and il-31
EP4019090A1 (en) 2020-12-23 2022-06-29 Numab Therapeutics AG Antibody variable domains that bind il-4r
WO2022136693A1 (en) 2020-12-23 2022-06-30 Numab Therapeutics AG Antibody variable domains and antibodies having decreased immunogenicity
EP4267615A1 (en) 2020-12-23 2023-11-01 Numab Therapeutics AG Antibody variable domains that bind il-4r
EP4271998A1 (en) 2020-12-30 2023-11-08 Abbott Laboratories Methods for determining sars-cov-2 antigen and anti-sars-cov-2 antibody in a sample
EP4271482A2 (en) 2020-12-31 2023-11-08 Alamar Biosciences, Inc. Binder molecules with high affinity and/ or specificity and methods of making and use thereof
AR124681A1 (en) 2021-01-20 2023-04-26 Abbvie Inc ANTI-EGFR ANTIBODY-DRUG CONJUGATES
AU2022215847A1 (en) 2021-02-02 2023-08-10 Numab Therapeutics AG Multispecific antibodies having specificity for ror1 and cd3
WO2022174813A1 (en) 2021-02-19 2022-08-25 信达生物制药(苏州)有限公司 Anti-gprc5d×bcma×cd3 trispecific antibody and use thereof
CA3212729A1 (en) 2021-03-12 2022-09-15 Janssen Biotech, Inc. Safe and effective method of treating psoriatic arthritis with anti-il23 specific antibody
AU2022232007A1 (en) 2021-03-12 2023-10-26 Janssen Biotech, Inc. Method of treating psoriatic arthritis patients with inadequate response to tnf therapy with anti-il23 specific antibody
CN117203232A (en) 2021-03-17 2023-12-08 瑞塞普托斯有限责任公司 Methods of treating atopic dermatitis with anti-IL-13 antibodies
WO2022197947A1 (en) 2021-03-18 2022-09-22 Alector Llc Anti-tmem106b antibodies and methods of use thereof
WO2022195028A2 (en) 2021-03-18 2022-09-22 Medimmune Limited Therapeutic binding molecules
WO2022204274A1 (en) 2021-03-23 2022-09-29 Alector Llc Anti-tmem106b antibodies for treating and preventing coronavirus infections
JP2024512633A (en) 2021-03-30 2024-03-19 バイエル・アクチエンゲゼルシヤフト Anti-SEMA3A antibody and its uses
EP4314068A1 (en) 2021-04-02 2024-02-07 The Regents Of The University Of California Antibodies against cleaved cdcp1 and uses thereof
EP4330283A1 (en) 2021-04-26 2024-03-06 Millennium Pharmaceuticals, Inc. Anti-adgre2 antibodies and uses thereof
WO2022232321A1 (en) 2021-04-28 2022-11-03 Minotaur Therapeutics, Inc. Humanized chimeric bovine antibodies and methods of use
AU2022272307A1 (en) 2021-05-12 2023-11-16 Applied Biomedical Science Institute Methods of screening and expression of disulfide-bonded binding polypeptides
EP4337689A1 (en) 2021-05-12 2024-03-20 Applied Biomedical Science Institute Binding polypeptides against sars cov-2 and uses thereof
CA3216320A1 (en) 2021-05-18 2022-11-24 Abbott Laboratories Methods of evaluating brain injury in a pediatric subject
AU2022276189A1 (en) 2021-05-20 2024-01-18 Janssen Biotech, Inc. Method of treating inflammatory bowel disease with a combination therapy of antibodies to il-23 and tnf alpha
WO2022266034A1 (en) 2021-06-14 2022-12-22 Abbott Laboratories Methods of diagnosing or aiding in diagnosis of brain injury caused by acoustic energy, electromagnetic energy, an over pressurization wave, and/or blast wind
IL309349A (en) 2021-06-14 2024-02-01 argenx BV Anti-il-9 antibodies and methods of use thereof
WO2022266221A1 (en) 2021-06-16 2022-12-22 Alector Llc Monovalent anti-mertk antibodies and methods of use thereof
WO2022266223A1 (en) 2021-06-16 2022-12-22 Alector Llc Bispecific anti-mertk and anti-pdl1 antibodies and methods of use thereof
US20230174651A1 (en) 2021-06-23 2023-06-08 Janssen Biotech, Inc. Materials and methods for hinge regions in functional exogenous receptors
WO2023281462A1 (en) 2021-07-09 2023-01-12 Janssen Biotech, Inc. Manufacturing methods for producing anti-tnf antibody compositions
US20230038355A1 (en) 2021-07-09 2023-02-09 Janssen Biotech, Inc. Manufacturing Methods for Producing Anti-IL12/IL23 Antibody Compositions
KR20240032991A (en) 2021-07-09 2024-03-12 얀센 바이오테크 인코포레이티드 Manufacturing Methods for Producing Anti-TNF Antibody Compositions
US11807685B2 (en) 2021-08-05 2023-11-07 The Uab Research Foundation Anti-CD47 antibody and uses thereof
WO2023019200A1 (en) 2021-08-11 2023-02-16 Viela Bio, Inc. Inebilizumab and methods of using the same in the treatment or prevention of igg4-related disease
CA3229448A1 (en) 2021-08-23 2023-03-02 Immunitas Therapeutics, Inc. Anti-cd161 antibodies and uses thereof
CA3230038A1 (en) 2021-08-31 2023-03-09 Hongwei Zhang Methods and systems of diagnosing brain injury
CA3230117A1 (en) 2021-09-02 2023-03-09 Mark Trautwein Anti-cecam6 antibodies with reduced side-effects
AU2022354059A1 (en) 2021-09-30 2024-03-28 Abbott Laboratories Methods and systems of diagnosing brain injury
WO2023069919A1 (en) 2021-10-19 2023-04-27 Alector Llc Anti-cd300lb antibodies and methods of use thereof
WO2023073615A1 (en) 2021-10-29 2023-05-04 Janssen Biotech, Inc. Methods of treating crohn's disease with anti-il23 specific antibody
US20230151087A1 (en) 2021-11-15 2023-05-18 Janssen Biotech, Inc. Methods of Treating Crohn's Disease with Anti-IL23 Specific Antibody
US20230159633A1 (en) 2021-11-23 2023-05-25 Janssen Biotech, Inc. Method of Treating Ulcerative Colitis with Anti-IL23 Specific Antibody
WO2023114978A1 (en) 2021-12-17 2023-06-22 Abbott Laboratories Systems and methods for determining uch-l1, gfap, and other biomarkers in blood samples
US20230213536A1 (en) 2021-12-28 2023-07-06 Abbott Laboratories Use of biomarkers to determine sub-acute traumatic brain injury (tbi) in a subject having received a head computerized tomography (ct) scan that is negative for a tbi or no head ct scan
WO2023131901A1 (en) 2022-01-07 2023-07-13 Johnson & Johnson Enterprise Innovation Inc. Materials and methods of il-1beta binding proteins
WO2023150652A1 (en) 2022-02-04 2023-08-10 Abbott Laboratories Lateral flow methods, assays, and devices for detecting the presence or measuring the amount of ubiquitin carboxy-terminal hydrolase l1 and/or glial fibrillary acidic protein in a sample
TW202348252A (en) 2022-02-16 2023-12-16 英商梅迪繆思有限公司 Combination therapies for treatment of cancer with therapeutic binding molecules
WO2023170295A1 (en) 2022-03-11 2023-09-14 Janssen Pharmaceutica Nv Multispecific antibodies and uses thereof
WO2023170291A1 (en) 2022-03-11 2023-09-14 Janssen Pharmaceutica Nv Multispecific antibodies and uses thereof
TW202346355A (en) 2022-03-11 2023-12-01 比利時商健生藥品公司 Multispecific antibodies and uses thereof
EP4273162A1 (en) 2022-05-06 2023-11-08 Numab Therapeutics AG Antibody variable domains and antibodies having decreased immunogenicity
WO2023214047A1 (en) 2022-05-06 2023-11-09 Numab Therapeutics AG Antibody variable domains and antibodies having decreased immunogenicity
WO2023223265A1 (en) 2022-05-18 2023-11-23 Janssen Biotech, Inc. Method for evaluating and treating psoriatic arthritis with il23 antibody
WO2023240109A1 (en) 2022-06-07 2023-12-14 Regeneron Pharmaceuticals, Inc. Multispecific molecules for modulating t-cell activity, and uses thereof
WO2023240124A1 (en) 2022-06-07 2023-12-14 Regeneron Pharmaceuticals, Inc. Pseudotyped viral particles for targeting tcr-expressing cells
WO2024006876A1 (en) 2022-06-29 2024-01-04 Abbott Laboratories Magnetic point-of-care systems and assays for determining gfap in biological samples
WO2024015953A1 (en) 2022-07-15 2024-01-18 Danisco Us Inc. Methods for producing monoclonal antibodies
WO2024013727A1 (en) 2022-07-15 2024-01-18 Janssen Biotech, Inc. Material and methods for improved bioengineered pairing of antigen-binding variable regions
WO2024018426A1 (en) 2022-07-22 2024-01-25 Janssen Biotech, Inc. Enhanced transfer of genetic instructions to effector immune cells
WO2024026471A1 (en) 2022-07-29 2024-02-01 Alector Llc Cd98hc antigen-binding domains and uses therefor
WO2024026472A2 (en) 2022-07-29 2024-02-01 Alector Llc Transferrin receptor antigen-binding domains and uses therefor
WO2024026447A1 (en) 2022-07-29 2024-02-01 Alector Llc Anti-gpnmb antibodies and methods of use thereof
WO2024050524A1 (en) 2022-09-01 2024-03-07 University Of Georgia Research Foundation, Inc. Compositions and methods for directing apolipoprotein l1 to induce mammalian cell death
WO2024059708A1 (en) 2022-09-15 2024-03-21 Abbott Laboratories Biomarkers and methods for differentiating between mild and supermild traumatic brain injury
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

Family Cites Families (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4391904A (en) * 1979-12-26 1983-07-05 Syva Company Test strip kits in immunoassays and compositions therein
GB8308235D0 (en) * 1983-03-25 1983-05-05 Celltech Ltd Polypeptides
US4816567A (en) * 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
EP0173494A3 (en) * 1984-08-27 1987-11-25 The Board Of Trustees Of The Leland Stanford Junior University Chimeric receptors by dna splicing and expression
GB8422238D0 (en) * 1984-09-03 1984-10-10 Neuberger M S Chimeric proteins
US4888415A (en) * 1985-03-04 1989-12-19 Dana-Farber Cancer Institute, Inc. Gelonin immunotoxin
US5225539A (en) * 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
GB8607679D0 (en) * 1986-03-27 1986-04-30 Winter G P Recombinant dna product
NL8720442A (en) * 1986-08-18 1989-04-03 Clinical Technologies Ass DELIVERY SYSTEMS FOR PHARMACOLOGICAL AGENTS.
US4946778A (en) * 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
US4853871A (en) * 1987-04-06 1989-08-01 Genex Corporation Computer-based method for designing stablized proteins
CA1341235C (en) * 1987-07-24 2001-05-22 Randy R. Robinson Modular assembly of antibody genes, antibodies prepared thereby and use
GB8720833D0 (en) * 1987-09-04 1987-10-14 Celltech Ltd Recombinant dna product
WO1989006967A1 (en) * 1988-02-03 1989-08-10 Xoma Corporation Immunosupression with anti-pan t-cell immunotoxin compositions
JPH03503887A (en) * 1988-02-03 1991-08-29 エクソウマ コーポレーション Therapeutic use of anti-T cell immunotoxins for autoimmune diseases
AU631545B2 (en) * 1988-04-15 1992-12-03 Protein Design Labs, Inc. Il-2 receptor-specific chimeric antibodies
IL162181A (en) * 1988-12-28 2006-04-10 Pdl Biopharma Inc A method of producing humanized immunoglubulin, and polynucleotides encoding the same
US5530101A (en) * 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
US5101025A (en) * 1989-04-04 1992-03-31 Genelabs Incorporated Recombinant trichosanthin and coding sequence
US5614192A (en) * 1989-07-19 1997-03-25 Connective Therapeutics, Inc. T cell receptor peptides as therapeutics for immune-related disease
NZ236792A (en) * 1990-01-19 1993-05-26 Merck & Co Inc Recombinant human anti-cd18 antibody, murine 1b4 heavy or light chain variable region, dna, vectors, mammalian host and pharmaceutical compositions
GB9019553D0 (en) * 1990-09-07 1990-10-24 Unilever Plc Specific binding agents
US5858725A (en) * 1990-10-10 1999-01-12 Glaxo Wellcome Inc. Preparation of chimaeric antibodies using the recombinant PCR strategy
AU1582692A (en) * 1991-03-08 1992-10-06 Protein Design Labs, Inc. Recombinant double chain immunotoxins
DE69233482T2 (en) * 1991-05-17 2006-01-12 Merck & Co., Inc. Method for reducing the immunogenicity of antibody variable domains
DE69233254T2 (en) * 1991-06-14 2004-09-16 Genentech, Inc., South San Francisco Humanized Heregulin antibody
WO1992022324A1 (en) * 1991-06-14 1992-12-23 Xoma Corporation Microbially-produced antibody fragments and their conjugates
DE69227519T2 (en) * 1991-09-06 1999-06-02 Res Dev Foundation DNA SEQUENCES ENCODING THE POLYPEPTIDE GELONIN
US5639641A (en) * 1992-09-09 1997-06-17 Immunogen Inc. Resurfacing of rodent antibodies

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9511138B2 (en) 2011-12-23 2016-12-06 Laboratoire Francais Du Fractionnement Et Des Biotechnologies Pharmaceutical compositions comprising an antibody which binds the human anti-mullerian hormone receptor type II
US10179816B2 (en) 2011-12-23 2019-01-15 Labotatoire Francais Du Fractionnement Et Des Biotechnologies Pharmaceutical compositions comprising an antibody which binds the human anti-Müllerian hormone receptor type II

Also Published As

Publication number Publication date
EP0571613B1 (en) 2003-09-17
JPH06506362A (en) 1994-07-21
US5770196A (en) 1998-06-23
CA2103887A1 (en) 1993-06-14
JP2005160485A (en) 2005-06-23
EP0571613A4 (en) 1997-03-05
DE69233204T2 (en) 2004-07-15
US5821123A (en) 1998-10-13
WO1993011794A1 (en) 1993-06-24
JP4157160B2 (en) 2008-09-24
US20040005630A1 (en) 2004-01-08
ATE249840T1 (en) 2003-10-15
JP2008253267A (en) 2008-10-23
CA2103887C (en) 2005-08-30
CA2507749C (en) 2010-08-24
DE69233204D1 (en) 2003-10-23
ES2202310T3 (en) 2004-04-01
EP1291360A1 (en) 2003-03-12
EP0571613A1 (en) 1993-12-01
US5766886A (en) 1998-06-16
CA2507749A1 (en) 1993-06-24

Similar Documents

Publication Publication Date Title
US5766886A (en) Modified antibody variable domains
US5869619A (en) Modified antibody variable domains
US8153134B2 (en) Anti-MDL-1 antibodies
AU2004290044B2 (en) Interleukin-10 antibodies
CA2623109C (en) Nk cell-depleting antibodies for treating immunoproliferative disorders
EP0918797B1 (en) Humanized immunoglobulin reactive with alpha4beta7 integrin
US7666417B2 (en) Methods and compositions for treating autoimmune diseases or conditions
US20070122404A1 (en) Humanized immunoglobulin reactive with alpha4beta7 integrin
NZ227968A (en) Hybrid antibody and compositions
EP1708961B1 (en) Anti-ip-10 antibodies
US20030229207A1 (en) Modified antibody variable domains
MXPA06005210A (en) Interleukin-10 antibodies

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: EXPRESSLY ABANDONED -- DURING EXAMINATION