US20060040363A1 - Human antibodies derived from immunized xenomice - Google Patents

Human antibodies derived from immunized xenomice Download PDF

Info

Publication number
US20060040363A1
US20060040363A1 US10/656,623 US65662303A US2006040363A1 US 20060040363 A1 US20060040363 A1 US 20060040363A1 US 65662303 A US65662303 A US 65662303A US 2006040363 A1 US2006040363 A1 US 2006040363A1
Authority
US
United States
Prior art keywords
human
antibody
leukocyte marker
immunoglobulin
analog
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
US10/656,623
Inventor
Raju Kucherlapati
Aya Jakobovits
Sue Klapholz
Daniel Brenner
Daniel Capon
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
Priority claimed from US08/031,801 external-priority patent/US6673986B1/en
Application filed by Individual filed Critical Individual
Priority to US10/656,623 priority Critical patent/US20060040363A1/en
Priority to US10/978,290 priority patent/US20050241006A1/en
Publication of US20060040363A1 publication Critical patent/US20060040363A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • C12N15/90Stable introduction of foreign DNA into chromosome
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/12Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria
    • C07K16/1267Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria from Gram-positive bacteria
    • C07K16/1282Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria from Gram-positive bacteria from Clostridium (G)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/22Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against growth factors ; against growth regulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
    • C07K16/241Tumor Necrosis Factors
    • 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
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
    • C07K16/244Interleukins [IL]
    • 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
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
    • C07K16/244Interleukins [IL]
    • C07K16/248IL-6
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2812Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against CD4
    • 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
    • C07K16/2851Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the lectin superfamily, e.g. CD23, CD72
    • C07K16/2854Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the lectin superfamily, e.g. CD23, CD72 against selectins, e.g. CD62
    • 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
    • C07K16/2875Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the NGF/TNF superfamily, e.g. CD70, CD95L, CD153, CD154
    • 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
    • C07K16/462Igs containing a variable region (Fv) from one specie and a constant region (Fc) from another
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/8509Vectors or expression systems specially adapted for eukaryotic hosts for animal cells for producing genetically modified animals, e.g. transgenic
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2207/00Modified animals
    • A01K2207/15Humanized animals
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/05Animals comprising random inserted nucleic acids (transgenic)
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/07Animals genetically altered by homologous recombination
    • A01K2217/072Animals genetically altered by homologous recombination maintaining or altering function, i.e. knock in
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2267/00Animals characterised by purpose
    • A01K2267/01Animal expressing industrially exogenous proteins
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2267/00Animals characterised by purpose
    • A01K2267/03Animal model, e.g. for test or diseases
    • A01K2267/035Animal model for multifactorial diseases
    • A01K2267/0381Animal model for diseases of the hematopoietic system
    • 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
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2840/00Vectors comprising a special translation-regulating system
    • C12N2840/20Vectors comprising a special translation-regulating system translation of more than one cistron
    • C12N2840/203Vectors comprising a special translation-regulating system translation of more than one cistron having an IRES

Definitions

  • the invention relates to the field of immunology, and in particular to the production of antibodies. More specifically, it concerns producing such antibodies by a process which includes the step of immunizing a transgenic animal with an antigen to which antibodies are desired.
  • the transgenic animal has been modified so as to produce human, as opposed to endogenous antibodies.
  • Antibodies with various immunospecificities are desirable for therapeutic and diagnostic use.
  • Those antibodies intended for human therapeutic and in vivo diagnostic use have been problematic because prior art sources for such antibodies resulted in immunoglobulins bearing the characteristic structures of antibodies produced by nonhuman hosts. Such antibodies tend to be immunogenic when used in humans.
  • the invention is directed to methods to produce human antibodies by a process wherein at least one step of the process includes immunizing a transgenic nonhuman animal with the desired antigen.
  • the modified animal fails to produce endogenous antibodies, but instead produces B-cells which secrete immunoglobulins with fully human variable regions.
  • the antibodies produced include fully human antibodies and can be obtained from the animal directly, or from immortalized B-cells derived from the animal. Alternatively, the genes encoding the immunoglobulins with human variable regions can be recovered and expressed to obtain the antibodies directly or modified to obtain analogs of antibodies such as, for example, single chain F V molecules.
  • the invention is directed to a method to produce an immunoglobulin with a fully human variable region to a specific antigen or to produce an analog of said immunoglobulin by a process which comprises immunizing a nonhuman animal with the antigen under conditions that stimulate an immune response.
  • the nonhuman animal is characterized by being substantially incapable of producing endogenous heavy or light immunoglobulin chain, but capable of producing immunoglobulins either with both human variable regions and constant regions or with fully human variable regions or both.
  • the animal produces B cells which secrete immunoglobulins, with at least variable regions that are fully human, specific for the antigen.
  • the human immunoglobulin of desired specificity can be directly recovered from the animal, for example, from the serum, or primary B cells can be obtained from the animal and immortalized.
  • the immortalized B cells can be used directly as the source of human antibodies or, alternatively, the genes encoding the antibodies can be prepared from the immortalized B cells or from primary B cells of the blood or lymphoid tissue (spleen, tonsils, lymph nodes, bone marrow) of the immunized animal and expressed in recombinant hosts, with or without modification, to produce the immunoglobulin or its analogs.
  • the genes encoding the repertoire of immunoglobulins produced by the immunized animal can be used to generate a library of immunoglobulins to permit screening for those variable regions which provide the desired affinity. Clones from the library which have the desired characteristics can then be used as a source of nucleotide sequences encoding the desired variable regions for further manipulation to generate antibodies or analogs with these characteristics using standard recombinant techniques.
  • the invention relates to an immortalized nonhuman B cell line derived from the above described animal.
  • the invention is directed to a recombinant host cell which is modified to contain the gene encoding either the human immunoglobulin with the desired specificity, or an analog thereof which exhibits the same specificity.
  • the invention is directed to antibodies or antibody analogs prepared by the above described methods and to recombinant materials for their production.
  • the invention is directed to antibodies with fully human variable regions, including fully human antibodies which are immunospecific with respect to particular antigens set forth herein and to analogs which are similarly immunospecific, as well as to the recombinant materials useful in the production of these antibodies.
  • FIG. 1 shows the serum titers of anti-IL-6 antibodies from a xenomouse immunized with human IL-6 and which antibodies contain human kappa light chains and/or human ⁇ heavy chains.
  • FIG. 2 shows the serum titers of anti-IL-8 antibodies from a xenomouse immunized with human IL-8 and which antibodies contain human kappa light chains and/or human ⁇ heavy chains.
  • FIG. 3 shows the serum titers of anti-TNF ⁇ antibodies from a xenomouse immunized with human TNF- ⁇ and which antibodies contain human kappa light chains and/or human ⁇ heavy chains.
  • FIG. 4 shows the serum titers of anti-CD4 antibodies from a xenomouse immunized with human CD4 and which antibodies contain human kappa light chains and/or human ⁇ heavy chains.
  • FIG. 5 shows the serum titers of a xenomouse immunized with 300.19 cells expressing L-selectin at their surface. In the ELISA assay used, these antibodies are detectable only if they carry human ⁇ constant region heavy chains.
  • FIG. 6 shows the serum titers of a xenomouse immunized with 300.19 cells expressing L-selectin at their surface. In the ELISA assay used, these antibodies are detectable only if they carry human kappa light chains.
  • FIG. 7 shows the serum titers of a xenomouse immunized with 300.19 cells expressing L-selectin.
  • these antibodies are detectable if they carry human kappa light chain and/or murine y constant regions.
  • FIG. 8 shows a FACS analysis of human neutrophils coupled to sera from a xenomouse (A195-2) immunized with human L-selectin and labeled with an antibody immunoreactive with murine heavy chain ⁇ constant region.
  • FIG. 9 shows a FACS analysis of human neutrophils incubated with serum from a xenomouse (A195-2) immunized with human L-selectin and labeled with an antibody immunoreactive with human light chain kappa region.
  • FIG. 10 is a diagram of a plasmid used to transfect mammalian cells to effect the production of the human protein gp39.
  • FIG. 11 represents the serum titration curve of mice immunized with CHO cells expressing human gp39.
  • the antibodies detected in this ELISA must be immunoreactive with gp39 and contain human heavy chain ⁇ constant regions or human kappa light chains.
  • FIG. 12 shows the results of a FACS analysis of antibodies from a xenomouse (labeled A247-4) immunized with human gp39 reacted with activated human T cells.
  • FIG. 12A shows the separation of human activated T cells into CD4+ and CD4 ⁇ populations.
  • Panel B shows the results of a FACS analysis of the activated CD4+ T cells with antibodies from the xenomouse immunized with gp39 which contain murine heavy chain ⁇ constant regions;
  • panel C shows the corresponding results with respect to CD4 ⁇ populations.
  • FIG. 13 is a titration curve with respect to monoclonal antibodies secreted by the hybridoma clone D5.1.
  • This clone is obtained from a xenomouse immunized with tetanus toxin C (TTC) and contains human kappa light chain and human ⁇ constant region in the heavy chain.
  • TTC tetanus toxin C
  • FIG. 14 is a titration curve with respect to the hybridoma supernatant from clone K4.1.
  • This hybridoma clone is obtained from a xenomouse immunized with TTC and contains human kappa light chain and heavy chain having the murine ⁇ constant region.
  • FIG. 15 shows binding curves for various concentrations of the K4.1 monoclonal antibody in a determination of the affinity of the monoclonal with its antigen in a BIAcore instrument.
  • FIG. 16 shows the complete nucleotide sequence of the heavy chain from the antibody secreted by K4.1.
  • FIG. 17 shows the complete nucleotide sequence of the light chain from the antibody secreted by K4.1.
  • FIG. 18 shows the complete nucleotide sequence of the heavy chain from the antibody secreted by D5.1.
  • FIG. 19 shows the complete nucleotide sequence of the light chain from the antibody secreted by D5.1.
  • the methods of the invention include administering an antigen for which human forms of immunospecific reagents are desired to a transgenic nonhuman animal which has been modified genetically so as to be capable of producing human, but not endogenous, antibodies.
  • the animal has been modified to disable the endogenous heavy and/or light chain loci in its genome, so that these endogenous loci are incapable of the rearrangement required to generate genes encoding immunoglobulins in response to an antigen.
  • the animal will have been provided, stably, in its genome, at least one human heavy chain locus and at least one human light chain locus so that in response to an administered antigen, the human loci can rearrange to provide genes encoding human immunoglobulins immunospecific for the antigen.
  • the first step is administration of the antigen.
  • Techniques for such administration are conventional and involve suitable immunization protocols and formulations which will depend on the nature of the antigen per se. It may be necessary to provide the antigen with a carrier to enhance its immunogenicity and/or to include formulations which contain adjuvants and/or to administer multiple injections, and the like. Such techniques are standard and optimization of them will depend on the characteristics of the particular antigen for which immunospecific reagents are desired.
  • immunospecific reagents includes immunoglobulins and their analogs.
  • analogs has a specific meaning in this context. It refers to moieties that contain the fully human portions of the immunoglobulin which account for its immunospecificity. In particular, variable regions including the complementarity determining regions (CDRs) are required, along with sufficient portions of the framework regions (FRs) to result in the appropriate three dimensional conformation.
  • Typical immunospecific analogs of antibodies include F (ab′) 2 , F ab′ , and F ab regions. Modified forms of the variable regions to obtain, for example, single chain F V analogs with the appropriate immunospecificity are known. A review of such F V construction is found, for example, in Tibtech (1991) 9:________. The construction of antibody analogs with multiple immunospecificities is also possible by coupling the human variable regions derived from antibodies with varying specificities.
  • variable regions with fully human characteristics can also be coupled to a variety of additional substances which can provide toxicity, biological functionality, alternative binding specificities and the like.
  • the moieties including the fully human variable regions produced by the methods of the invention include single-chain fusion proteins, molecules coupled by covalent methods other than those involving peptide linkages, and aggregated molecules. Examples of analogs which include variable regions coupled to additional molecules covalently or noncovalently include those in the following nonlimiting illustrative list. Traunecker, A. et al. Int J Cancer Supp (1992) Supp 7:51-52 describe the bispecific reagent janusin in which the F V region directed to CD3 is coupled to soluble CD4 or to other ligands such as OVCA and IL-7.
  • the fully human variable regions produced by the method of the invention can be constructed into F V molecules and coupled to alternative ligands such as those illustrated in the cited article.
  • Higgins, P. J. et al. J Infect Disease (1992) 166:198-202 describe a heteroconjugate antibody composed of OKT3 cross-linked to an antibody directed to a specific sequence in the V3 region or GP120.
  • Such heteroconjugate antibodies can also be constructed using at least the human variable regions contained in the immunoglobulins produced by the invention methods. Additional examples of bispecific antibodies include those described by Fanger, M. W. et al. Cancer Treat Res (1993) 68:181-194 and by Fanger, M. W. et al.
  • Conjugates that are immunotoxins including conventional antibodies have been widely described in the art.
  • the toxins may be coupled to the antibodies by conventional coupling techniques or immunotoxins containing protein toxin portions can be produced as fusion proteins.
  • the analogs of the present invention can be used in a corresponding way to obtain such immunotoxins.
  • Illustrative of such immunotoxins are those described by Byrs, B. S. et al. Seminars Cell Biol (1991) 2:59-70 and by Fanger, M. W. et al. Immunol Today (1991) 12:51-54.
  • the genes encoding the immunoglobulins produced by the transgenic animals of the invention can be retrieved and the nucleotide sequences encoding the fully human variable region can be manipulated according to known techniques to provide a variety of analogs such as those described above.
  • the immunoglobulins themselves containing the human variable regions can be modified using standard coupling techniques to provide conjugates retaining immunospecificity and fully human characteristics in the immunospecific region.
  • immunoglobulin “analogs” refers to moieties which contain those portions of the antibodies of the invention which retain their human characteristics and their immunospecificity. These will retain sufficient human variable region to provide the desired specificity.
  • all of the methods of the invention include administering the appropriate antigen to the transgenic animal.
  • the recovery or production of the antibodies themselves can be achieved in various ways.
  • the polyclonal antibodies produced by the animal and secreted into the bloodstream can be recovered using known techniques. Purified forms of these antibodies can, of course, be readily prepared by standard purification techniques, preferably including affinity chromatography with respect to the particular antigen, or even with respect to the particular epitope of the antigen for which specificity is desired. In any case, in order to monitor the success of immunization, the antibody levels with respect to the antigen in serum will be monitored using standard techniques such as ELISA, RIA and the like.
  • a portion of the polyclonal antiserum obtained may include an endogenous heavy chain constant region derived from the host, even though the variable regions are fully human.
  • use of the polyclonal antiserum directly would be inappropriate.
  • the presence of these chimeras which is believed to result from in vivo isotype switching as described by Gerstein et al. Cell (1990) 63:537, is not problematic, in view of conventional purification and modification methods and in view of the availability of alternative methods to recover fully human antibodies, if desired, described in the following paragraphs.
  • the polyclonal antiserum could be subjected to suitable separation techniques to provide compositions containing only fully human immunoglobulins. Portions of the serum which display characteristics of the host species can be removed, for example, using affinity reagents with the appropriate anti species immunoglobulins or immunospecific portions thereof. Furthermore, for applications where only the variable regions of the antibodies are required, treating the polyclonal antiserum with suitable reagents so as to generate F ab , F ab′ , or F (ab′) 2 portions results in compositions containing fully human characteristics. Such fragments are sufficient for use, for example, in immunodiagnostic procedures involving coupling the immunospecific portions of immunoglobulins to detecting reagents such as radioisotopes. Thus, for some applications, the polyclonal antiserum can be treated to provide compositions with the desired characteristics including compositions consisting essentially of fully human antibodies and compositions including immunoglobulin analogs wherein the immunospecific portion is fully human.
  • immunoglobulins and analogs with desired characteristics can be generated from immortalized B cells derived from the transgenic animals used in the method of the invention or from the rearranged genes provided by these animals in response to immunization. It will be apparent that hybridomas derived from the B cells of the immunized animal can be screened so as to choose only those secreting fully human antibodies and that the genetic material can be recovered from the hybridomas or from lymphocytes in spleen, blood, or lymph nodes of the immunized animal and manipulated using conventional techniques to replace any endogenous constant region with a human one or to produce a desired analog.
  • the B cells can be obtained, typically from the spleen, but also, if desired, from the peripheral blood lymphocytes or lymph nodes and immortalized using any of a variety of techniques, most commonly using the fusion methods described by Kohler and Milstein.
  • the resulting hybridomas (or otherwise immortalized B cells) can then be cultured as single colonies and screened for secretion of antibodies of the desired specificity.
  • the screen can also include a determination of the fully human character of the antibody. For example, as described in the examples below, a sandwich ELISA wherein the monoclonal in the hybridoma supernatant is bound both to antigen and to an antihuman constant region can be employed.
  • hybridomas that secrete antibodies which are immunoreactive with antispecies antibodies directed to the species of the immunized animal can be discarded.
  • the desired antibodies can be recovered, again using conventional techniques. They can be prepared in quantity by culturing the immortalized B cells using conventional methods, either in vitro, or in vivo to produce ascites fluid. Purification of the resulting monoclonal antibody preparations is less burdensome than in the case of serum since each immortalized colony will secrete only a single type of antibody. In any event, standard purification techniques to isolate the antibody from other proteins in the culture medium can be employed.
  • the immortalized cells can be used as a source of rearranged heavy chain and light chain loci for subsequent expression and/or genetic manipulation. Isolation of genes from such antibody-producing cells is straightforward since high levels of the appropriate mRNAs are available for production of a cDNA library.
  • the recovered rearranged loci can be manipulated as desired. For example, the constant region can be exchanged for that of a different isotype or that of a human antibody, as described above, or eliminated altogether.
  • the variable regions can be linked to encode single chain F V regions. Multiple F V regions can be linked to confer binding ability to more than one target or chimeric heavy and light chain combinations can be employed.
  • the coding sequences including those that encode, at a minimum, the variable regions of the human heavy and light chain can be inserted into expression systems contained on vectors which can be transfected into standard recombinant host cells.
  • host cells As described below, a variety of such host cells may be used; for efficient processing, however, mammalian cells are preferred. Typical mammalian cell lines useful for this purpose include CHO cells, 293 cells, or NSO-GS cells.
  • the production of the antibody or analog is then undertaken by culturing the modified recombinant host under culture conditions appropriate for the growth of the host cells and the expression of the coding sequences.
  • the antibodies are then recovered from the culture.
  • the expression systems are preferably designed to include signal peptides so that the resulting antibodies are secreted into the medium; however, intracellular production is also possible.
  • Phage display libraries may also be constructed using previously manipulated nucleotide sequences and screened in similar fashion. In general, the cDNAs encoding heavy and light chain are independently supplied or are linked to form F V analogs for production in the phage library.
  • the phage library is thus screened for the antibodies with highest affinity for the antigen and the genetic material recovered from the appropriate clone. Further rounds of screening can increase the affinity of the original antibody isolated.
  • the manipulations described above for recombinant production of the antibody or modification to form a desired analog can then be employed.
  • the modified or unmodified rearranged loci are manipulated using standard recombinant techniques by constructing expression systems operable in a desired host cell, such as, typically, a Chinese hamster ovary cell, and the desired immunoglobulin or analog is produced using standard recombinant expression techniques, and recovered and purified using conventional methods.
  • a desired host cell such as, typically, a Chinese hamster ovary cell
  • the desired immunoglobulin or analog is produced using standard recombinant expression techniques, and recovered and purified using conventional methods.
  • compositions of the invention will have utilities similar to those ascribable to nonhuman antibodies directed against the same antigen. Such utilities include, for example, use as a affinity ligands for purification, as reagents in immunoassays, as components of immunoconjugates, and as therapeutic agents for appropriate indications.
  • antibodies or their analogs with fully human characteristics.
  • These reagents avoid the undesired immune responses engendered by antibodies or analogs which have characteristics marking them as originating from non-human species.
  • Other attempts to “humanize” antibodies do not result in reagents with fully human characteristics.
  • chimeric antibodies with murine variable regions and human constant regions are easily prepared, but, of course, retain murine characteristics in the variable regions.
  • Even the much more difficult procedure of “humanizing” the variable regions by manipulating the genes encoding the amino acid sequences that form the framework regions does not provide the desired result since the CDRs, typically of nonhuman origin, cannot be manipulated without destroying immunospecificity.
  • the methods of the present invention provide, for the first time, immunoglobulins that are fully human or analogs which contain immunospecific regions with fully human characteristics.
  • immunoglobulins and analogs are those immunospecific with respect to human IL-6, human IL-8, human TNF ⁇ , human CD4, human L-selectin, and human gp39.
  • Human antibodies against IL-8 are particularly useful in preventing tumor metastasis and inflammatory states such as asthma.
  • Antibodies and analogs immunoreactive with human TNF ⁇ and human IL-6 are useful in treating cachexia and septic shock as well as autoimmune disease.
  • Antibodies and analogs immunoreactive with gp39 or with L-selectin are also effective in treating or preventing autoimmune disease.
  • anti-gp39 is helpful in treating graft versus host disease, in preventing organ transplant rejection, and in treating glomerulonephritis.
  • Antibodies and analogs against L-selectin are useful in treating ischemia associated with reperfusion injury.
  • Typical autoimmune diseases which can be treated using the above-mentioned antibodies and analogs include systemic lupus erythematosus, rheumatoid arthritis, psoriasis, Sjogren's, scleroderma, mixed connective tissue disease, dermatomyositis, polymyositis, Reiter's syndrome, Behcet's disease, Type 1 diabetes, Hashimoto's thyroiditis, Grave's disease, multiple sclerosis, myasthenia gravis and pemphigus.
  • mice designated “xenomice”
  • xenomice mice, designated “xenomice”
  • the sera of the immunized xenomice (or the supernatants from immortalized B cells) were titrated for antigen specific human antibodies in each case using a standard ELISA format. In this format, the antigen used for immunization was immobilized onto wells of microtiter plates. The plates were washed and blocked and the sera (or supernatants) were added as serial dilutions for 1-2 hours of incubation.
  • bound antibody having human characteristics was detected by adding the appropriate antispecies Ig (typically antihuman kappa or antihuman ⁇ ) conjugated to horseradish peroxidase (HRP) for one hour.
  • HRP horseradish peroxidase
  • the bound antibodies were tested for murine characteristics using antimurine antibodies, typically antimurine ⁇ .
  • OPD chromogenic reagent o-phenylene diamine
  • the antigen was coated using plate coating buffer (0.1 M carbonate buffer, pH 9.6); the assay blocking buffer used was 0.5% BSA, 0.1% Tween 20 and 0.01% Thimerosal in PBS; the substrate buffer used in color development was citric acid 7.14 g/l: dibasic sodium phosphate 17.96 g/l; the developing solution (made immediately before use) was 10 ml substrate buffer, 10 mg OPD, plus 5 ml hydrogen peroxide; the stop solution (used to stop color development) was 2 M sulfuric acid. The wash solution was 0.05% Tween 20 in PBS.
  • mice Three to 5 xenomice aged 8-20 weeks were age-matched and immunized intraperitoneally with 50 ⁇ g human IL-6 emulsified in complete Freund's adjuvant for primary immunization and in incomplete Freund's adjuvant for subsequent injections. The mice received 6 injections 2-3 weeks apart. Serum titers were determined after the second dose and following each dose thereafter. Bleeds were performed 6-7 days after injections from the retrobulbar plexus. The blood was allowed to clot at room temperature for about 2 hours and then incubated at 4° C. for at least 2 hours before separating and collecting the sera.
  • ELISAs were conducted as described above by applying 100 ⁇ l per well of recombinant human IL-6 at 2 mg/ml in coating buffer. Plates were then incubated at 4° C. overnight or at 37° C. for 2 hours and then washed three times in washing buffer. Addition of 100 ⁇ l/well blocking buffer was followed by incubation at room temperature for 2 hours, and an additional 3 washes.
  • Immunization and serum preparation were as described in Example 1 as except that human recombinant IL-8 was used as an immunogen.
  • ELISA assays were performed with respect to the recovered serum, also exactly as described in Example 1, except that the ELISA plates were initially coated using 100 ⁇ l/well of recombinant human IL-8 at 0.5 mg/ml in the coating buffer.
  • the results obtained for various serum dilutions from xenomouse A260-5 after 6 injections are shown in FIG. 2 .
  • Human anti-IL-8 reactivity was again shown at serum dilutions having concentrations higher than that represented by a 1:1,000 dilution.
  • Example 1 Immunization and serum preparation were conducted as described in Example 1 except that human recombinant TNF ⁇ was substituted for human IL-6.
  • ELISAs were conducted as described in Example 1 except that the initial coating of the ELISA plate employed 100 ⁇ l/well recombinant human TNF ⁇ at 1 mg/ml in coating buffer.
  • the dilution curves for serum from xenomouse A210-8 after 6 injections obtained are shown in FIG. 3 . Again significant titers of human anti-TNF ⁇ reactivity were shown.
  • the human CD4 antigen was prepared as a surface protein on transfected recombinant cells as follows: Human CD4 zeta (F15 LTR) as described in Roberts, et al., Blood (1994) 84:2878 was introduced into the rat basophil leukemic cell line RBL-2H3, described by Callan, M., et al., Proc Natl Acad Sci USA (1993) 90:10454 using the kat high efficiency transduction system described by Finer, et al., Blood (1994) 83:43.
  • RBL-2H3 cells at 10 6 cells per well were cultured in 750 ml DMEM low +20% FBS (Gibco) and 16 ⁇ g/ml polybrene with an equal volume of proviral supernatant for 2 hours at 37° C., 5% CO 2 .
  • One ml of medium was removed and 750 ⁇ l of infection medium and retroviral supernatant were added to each well and the cultures incubated overnight.
  • the cells were washed and expanded in DMEM low +10% FBS until sufficient cells were available for sorting.
  • the CD4-zeta transduced RBL-2H3 cells were sorted using the FACSTAR plus (Becton Dickinson). The cells were stained for human CD4 with a mouse antihuman CD4-PE antibody and the top 2-3% expressing cells were selected.
  • Example 1 Immunizations were conducted as described in Example 1 using 10 ⁇ 10 6 cells per mouse except that the primary injection was subcutaneous at the base of the neck. The mice received 6 injections 2-3 weeks apart. Serum was prepared and analyzed by ELISA as described in Example 1 except that the initial coating of the ELISA plate utilized 100 ⁇ l per well of recombinant soluble CD4 at 2 mg/ml of coating buffer. The titration curve for serum from xenomouse A207-1 after 6 injections is shown in FIG. 4 . Titers of human anti-CD4 reactivity were shown at concentrations representing greater than those at 1:1,000 dilution.
  • the antigen was prepared as a surface displayed protein in C51 cells, a high expressing clone derived by transfecting the mouse pre-B cell 300.19 with LAM-1 cDNA (LAM-1 is the gene encoding L-selectin) (Tedder, et al., J Immunol (1990) 144:532) or with similarly transfected CHO cells.
  • LAM-1 is the gene encoding L-selectin
  • the transfected cells were sorted using fluorescent activated cell sorting using anti-Leu-8 antibody as label.
  • the C51 and the transfected CHO cells were grown in DME 4.5 g/l glucose with 10% FCS and 1 mg/ml G418 in 100 mm dishes.
  • Negative control cells, 3T3-P317 (transfected with gag/pol/env genes of Moloney virus) were grown in the same medium without G418.
  • Sera were collected as described in Example 1 and analyzed by ELISA in a protocol similar to that set forth in Example 1.
  • the transfected cells were plated into 96 well plates and cell monolayers grown for 1-2 days depending on cell number and used for ELISA when confluent.
  • the cells were fixed by first washing with cold 1 ⁇ PBS and then fixing solution (5% glacial acetic acid, 95% ethanol) was added.
  • the plates were incubated at ⁇ 25° C. for 5 minutes and can be stored at this temperature if sealed with plate sealers.
  • the ELISA is begun by bringing the plates to room temperature, flicking to remove fixing solution and washing 5 times with DMEM medium containing 10% FCS at 200 ⁇ l per well.
  • Positive control wells contained murine IgG1 to human L-selectin.
  • ELISAs were also performed using as the immobilized antigen a fusion protein consisting of the extracellular domain of human L-selectin fused to the constant domain of human IgG 1 (Guo, et al., Cell Immunol (1994) 154:202).
  • the L-selectin fusion protein was made by transient transfection of human 293 cells using calcium phosphate transfection (Wigler, M., Cell (1979) 16:777). Serum preparation was performed as described in Example 1.
  • ELISAs were conducted essentially as in Example 1, except that the initial coating of the ELISA plate employed 100 ⁇ l transfected 293 cell culture supernatant containing the L-selectin-Ig fusion protein. Detection employed HRP-mouse antihuman kappa and HRP-goat antimouse IgG.
  • FIG. 7 shows the results from xenomouse A195-2; antibodies specific for L-selectin having human kappa light chains and/or murine heavy chain ⁇ regions are present in the serum.
  • the antisera obtained from the immunized xenomice were also tested for staining of human neutrophils.
  • Human neutrophils were prepared as follows: peripheral blood was collected from normal volunteers with 100 units/ml heparin. About 3.5 ml blood was layered over an equal volume of One-step Polymorph Gradient (Accurate Chemical, Westbury, N.Y.) and spun for 30 minutes at 450 ⁇ G at 20° C. The neutrophil fraction was removed and washed twice in DPBS/2% FBS.
  • the neutrophils were then stained with either:
  • the stained, washed neutrophils were analyzed by FACS.
  • the results for antiserum from xenomouse A195-2 are shown in FIGS. 8 and 9 .
  • these antibodies containing human variable regions are readily convertible to fully human antibodies.
  • the cDNAs encoding them can be obtained.
  • amplifying the genes encoding human V regions using primers containing restriction enzyme recognition sites and cloning them into plasmids containing the coding sequences for human constant regions as described by Queen, et al., Proc Natl Acad Sci (1989) 86:10029 genes encoding the fully human antibodies can be obtained for recombinant production.
  • gp39 (the ligand for CD40) is expressed on activated human CD4+ T cells.
  • the sera of xenomice immunized with recombinant gp39 according to this example contained antibodies immunospecific for gp39 with fully human variable regions; the sera contained fully human IgM antibodies and chimeric IgG antibodies containing human variable regions and murine constant heavy chain y region.
  • the antigen consisted of stable transfectants of 300.19 cells or of CHO cells expressing gp39 cDNA cloned into the mammalian expression vector P1K1.HUgp39/IRES NEO as shown in FIG. 10 .
  • CHO cells were split 1:10 prior to transfection in DMEM 4.5 g/l glucose, 10% FBS, 2 mM glutamine, MEM, NEAA supplemented with additional glycine, hypoxanthine and thymidine.
  • the cells were cotransfected with the gp39 vector at 9 ⁇ g/10 cm plate (6 ⁇ 10 5 cells) and the DHFR expressing vector pSV2DHFRs (Subranani et al.
  • mice grouped as described in Example 1 were immunized with 300.19 cells expressing gp39 using a primary immunization subcutaneously at the base of the neck and with secondary intraperitoneal injections every 2-3 weeks.
  • Sera were harvested as described in Example 1 for the ELISA assay.
  • the ELISA procedure was conducted substantially as set forth in Example 1; the microtiter plates were coated with CHOD-gp39 cells grown in a 100 mm dish in DMEM, 4.5 g/l glucose, 10% FCS, 4 mM glutamine, and nonessential amino acid (NEAA) solution for MEM (100 ⁇ ).
  • the cells were trypsinized and plated into 96-well filtration plates at 10 5 cells/200 ⁇ l well and incubated at 37° C. overnight.
  • the positive controls were mouse antihuman gp39; negative controls were antisera from mice immunized with an antigen other than gp39. 50 ⁇ l of sample were used for each assay. The remainder of the assay is as described in Example 1.
  • the dilution curves for the sera obtained after 4 injections from mice immunized with gp39 expressed on CHO cells are shown in FIG. 11 .
  • the sera contained antihuman gp39 immunospecificity which is detectable with human kappa and human ⁇ HRP-coupled antibodies.
  • PBMC peripheral blood was collected from normal volunteers with the addition of 100 unit/ml heparin.
  • PBMC peripheral blood was collected from normal volunteers with the addition of 100 unit/ml heparin.
  • PBMC peripheral blood was collected from normal volunteers with the addition of 100 unit/ml heparin.
  • PBMC peripheral blood was collected from normal volunteers with the addition of 100 unit/ml heparin.
  • PBMC peripheral blood was collected from normal volunteers with the addition of 100 unit/ml heparin.
  • PBMC were isolated over Ficoll gradient and activated with 3 ⁇ g/ml PHA, 1 ⁇ g/ml PMA in IMDM plus 10% FBS plus 25 ⁇ M 2-mercaptoethanol for 4 hours.
  • the PBMC were stained with mouse Mab against human CD4 labeled with FITC to permit separation of activated human T cells from unactivated cells.
  • the activated CD4+ and CD4 ⁇ T cells were then analyzed by FACS using staining with either:
  • the detecting antibody in the FACS analysis was goat antimouse IgG (PE). The results are shown in FIG. 12 .
  • CD4+ (R2) and CD4 ⁇ (R3) cells were separated prior to FACS analysis.
  • Panel B shows the results for CD4+ cells and shows that sera from mice immunized with gp39 (labeled A247-4 in the figure) reacted with these activated PBMC;
  • panel C shows that these sera did not react with CD4 ⁇ cells.
  • These antibodies carried immune heavy chain ⁇ constant regions.
  • the results of panels B and C confirm that the TNF-injected xenomouse did not make antibodies against activated human T cells.
  • the antibodies prepared in this example were secreted by hybridomas obtained by immortalizing B cells from xenomice immunized with tetanus toxin.
  • the immunization protocol was similar to that set forth in Example 1 using 50 ⁇ g tetanus toxin emulsified in complete Freund's adjuvant for intraperitoneal primary immunization followed by subsequent intraperitoneal injections with antigen incorporated into incomplete Freund's adjuvant. The mice received a total of 4 injections 2-3 weeks apart.
  • anti-TTC antitetanus toxinC
  • the spleen cells were fused with myeloma cells P3 ⁇ 63-Ag8.653 as described by Galfre, G. and Milstein, C. Methods in Enzymology (1981) 73:3-46.
  • the cells were resuspended in DMEM, 15% FCS, containing HAT supplemented with glutamine, pen/strep for culture at 37° C. and 10% CO 2 .
  • the cells were plated in microtiter trays and maintained in HAT-supplemented medium for two weeks before transfer to HT-supplemented media.
  • Supernatants from wells containing hybridomas were collected for a primary screen using an ELISA.
  • the ELISA was conducted as described in Example 1 wherein the antigen coating consisted of 100 ⁇ l/well of tetanus toxin C (TTC) protein at 2 mg/ml in coating buffer, followed by incubation at 4° C. overnight or at 37° C. for two hours.
  • TTC tetanus toxin C
  • HRP-conjugated goat antimouse IgG at 1/2000 was used in addition to HRP mouse antihuman IgM as described in Example 1.
  • Two hybridomas that secreted anti-TTC according to the ELISA assay, clone D5.1 and clone K4.1 were used for further analysis.
  • clone D5.1 secretes fully human anti-TTC which is detectable using HRP-conjugated antihuman ⁇ and HRP-conjugated antihuman kappa.
  • FIG. 14 shows that clone K4.1 secretes anti-TTC which is immunoreactive with antimurine ⁇ and antihuman kappa HRP-conjugated antibodies.
  • clone K4.1 provides anti-TTC with human variable regions and a murine constant heavy chain ⁇ region.
  • the antibodies secreted by D5.1 and K4.1 did not immunoreact in ELISAs using TNF ⁇ , IL-6, or IL-8 as immobilized antigen under conditions where positive controls (sera from xenomice immunized with TNF ⁇ , IL-6 and IL-8 respectively) showed positive ELISA results.
  • TTC TTC antigen
  • the surface was activated with 35 ⁇ l of equal volumes 0.1 M NHS and 0.1 M EDC injected across the surface followed by 30 ⁇ l of TTC fragment at 100 ⁇ g/ml in 10 mM sodium acetate buffer pH 5.0. The surface was blocked by injecting 35 ⁇ l 1 M ethanolamine and washed to remove noncovalently bound TCC using 5 ⁇ l 0.1 M HCl. The entire immobilization procedure was conducted with a continuous flow of buffer at 5 ⁇ l/min. This results in about 7500-8500 response units (RU) of TTC per chip. (1000 RU corresponds to about 1 ng of protein per mm2.)
  • the procedure utilizes 15 ⁇ l rather than 30 ⁇ l of TTC, resulting in chips containing 550-950 RU.
  • Chips could be regenerated after use in single determinations by injecting 10 ⁇ l formal or MgCl 2 .
  • the chips are used to determine binding affinities by determining k a and k b (the association and dissociation rate constants) for the antibody with respect to the immobilized TTC.
  • the association rate constant is measured over six minutes at a flow rate of 5 ⁇ l/min. at different concentrations of K4.1 Mab in the range of 2.16 nm-69.33 nm.
  • the dissociation rate constant is measured at a constant buffer flow rate of 5 ⁇ l/min after completion of the antibody injection.
  • the raw data are graphed in FIG. 15 and the calculated results are shown in Table 1. TABLE 1 Kinetic Constants of K4.1 Measured Using the BIAcore on Two Different Surface Association Dissociation Binding Dis Immobilized K4.1 conc.
  • the K4.1 antibody has a binding constant for TTC somewhat larger than 10 10 l/mol.
  • the complete nucleotide sequence of the cDNAs encoding the heavy and light chains of the K4.1 and D5.1 monoclonals were determined as shown in FIGS. 16-19 .
  • PolyA mRNA was isolated from about 10 6 hybridoma cells and used to generate cDNA using random hexamers as primers. Portions of the product were amplified by PCR using the appropriate primers.
  • Both cell lines were known to provide human kappa light chains; for PCR amplification of light chain encoding cDNA, the primers used were HKP1 (5′-CTCTGTGACACTCTCCTGGGAGTT-3′) for priming from the constant region terminus and two oligos, used in equal amounts to prime from the variable segments: B3 (5′-CCACCATCAACTGCAAGTCCAGCCA-3′) and B2/B1 (5′-GAAACGACACTCACGCAGTCTCCAGC-3′).
  • the primers were MG-24Vi for the human variable regions: 5′-CAGGTGCAGCTGGAGCAGTCiGG-3′ which, with inosine as shown recognizes the human variable regions V H1-2 , V H1-3 , V H4 and V H6 , and from the constant region MG-25 i.e., 5′-GCACACCGCTGGACAGGGATCCAiAGTTTC-3′, which, containing inosine as shown recognizes murine ⁇ 1, ⁇ 2A, ⁇ 2B, and ⁇ 3.
  • MG-24VI was used to prime from the variable and ⁇ P1 (5′-TTTTCTTTGTTGCCGTTGGGGTGC-3′) was used to prime from the constant region terminus.
  • FIG. 16 representing the heavy chain of the Mab secreted by K4.1
  • the sequence shows the presence of the human variable segment VH6, the human diversity region DN1, and the human joining segment JH4 linked to the murine ⁇ 1 constant region.
  • VH6 the human variable segment
  • DN1 the human diversity region
  • JH4 the human joining segment
  • FIG. 17 which shows the light chain of the K4.1 antibody
  • analysis shows the presence of the human kappa variable region B3 and joining region JK4. Eight nucleotides are missing from B3 at the V K -J K junction and four mutations were found in the variable region. Five nongermline nucleotide additions were present at the V K -J K junction.
  • FIG. 18 which sets forth the sequence for the heavy chain of the antibody secreted by clone D5.1, this shows the heavy chain is comprised of the human variable fragment VH6, the human diversity region DN1 and the human joining segment JH4 linked to the human ⁇ constant region.
  • VH6 the human variable fragment
  • DN1 the human diversity region
  • JH4 the human joining segment
  • FIG. 19 which presents the light chain of the antibody secreted by D5.1, the human kappa variable region B3 and human kappa joining region JK3 are shown. There are nine base-pair differences from the germline sequences, three falling within CDR1.

Abstract

Antibodies with fully human variable regions against a specific antigen can be prepared by administering the antigen to a transgenic animal which has been modified to produce such antibodies in response to antigenic challenge, but whose endogenous loci have been disabled. Various subsequent manipulations can be performed to obtain either antibodies per se or analogs thereof.

Description

    TECHNICAL FIELD
  • The invention relates to the field of immunology, and in particular to the production of antibodies. More specifically, it concerns producing such antibodies by a process which includes the step of immunizing a transgenic animal with an antigen to which antibodies are desired. The transgenic animal has been modified so as to produce human, as opposed to endogenous antibodies.
  • BACKGROUND ART
  • PCT application WO 94/02602, published 3 Feb. 1994 and incorporated herein by reference, describes in detail the production of transgenic nonhuman animals which are modified so as to produce antibodies with fully human variable regions rather than endogenous antibodies in response to antigenic challenge. Briefly, the endogenous loci encoding the light and heavy immunoglobulin chains are incapacitated in the transgenic hosts and loci encoding human heavy and light chain proteins are inserted into the genome. In general, the animal which provides all the desired modifications is obtained by cross-breeding intermediate animals containing fewer than the full complement of modifications. The preferred embodiment of the nonhuman animal described in the specification is a mouse. Thus., mice, specifically, are described which, when administered immunogens, produce antibodies with human variable regions, including fully human antibodies, rather than murine antibodies that are immunospecific for these antigens.
  • The availability of such transgenic animals makes possible new approaches to the production of fully human antibodies. Antibodies with various immunospecificities are desirable for therapeutic and diagnostic use. Those antibodies intended for human therapeutic and in vivo diagnostic use, in particular, have been problematic because prior art sources for such antibodies resulted in immunoglobulins bearing the characteristic structures of antibodies produced by nonhuman hosts. Such antibodies tend to be immunogenic when used in humans.
  • The availability of the nonhuman, immunogen-responsive transgenic animals described in the above-referenced WO 94/02602 make possible convenient production of human antibodies without the necessity of employing human hosts.
  • DISCLOSURE OF THE INVENTION
  • The invention is directed to methods to produce human antibodies by a process wherein at least one step of the process includes immunizing a transgenic nonhuman animal with the desired antigen. The modified animal fails to produce endogenous antibodies, but instead produces B-cells which secrete immunoglobulins with fully human variable regions. The antibodies produced include fully human antibodies and can be obtained from the animal directly, or from immortalized B-cells derived from the animal. Alternatively, the genes encoding the immunoglobulins with human variable regions can be recovered and expressed to obtain the antibodies directly or modified to obtain analogs of antibodies such as, for example, single chain FV molecules.
  • Thus, in one aspect, the invention is directed to a method to produce an immunoglobulin with a fully human variable region to a specific antigen or to produce an analog of said immunoglobulin by a process which comprises immunizing a nonhuman animal with the antigen under conditions that stimulate an immune response. The nonhuman animal is characterized by being substantially incapable of producing endogenous heavy or light immunoglobulin chain, but capable of producing immunoglobulins either with both human variable regions and constant regions or with fully human variable regions or both. In the resulting immune response, the animal produces B cells which secrete immunoglobulins, with at least variable regions that are fully human, specific for the antigen. The human immunoglobulin of desired specificity can be directly recovered from the animal, for example, from the serum, or primary B cells can be obtained from the animal and immortalized. The immortalized B cells can be used directly as the source of human antibodies or, alternatively, the genes encoding the antibodies can be prepared from the immortalized B cells or from primary B cells of the blood or lymphoid tissue (spleen, tonsils, lymph nodes, bone marrow) of the immunized animal and expressed in recombinant hosts, with or without modification, to produce the immunoglobulin or its analogs. In addition, the genes encoding the repertoire of immunoglobulins produced by the immunized animal can be used to generate a library of immunoglobulins to permit screening for those variable regions which provide the desired affinity. Clones from the library which have the desired characteristics can then be used as a source of nucleotide sequences encoding the desired variable regions for further manipulation to generate antibodies or analogs with these characteristics using standard recombinant techniques.
  • In another aspect, the invention relates to an immortalized nonhuman B cell line derived from the above described animal. In still another aspect, the invention is directed to a recombinant host cell which is modified to contain the gene encoding either the human immunoglobulin with the desired specificity, or an analog thereof which exhibits the same specificity.
  • In still other aspects, the invention is directed to antibodies or antibody analogs prepared by the above described methods and to recombinant materials for their production.
  • In still other aspects, the invention is directed to antibodies with fully human variable regions, including fully human antibodies which are immunospecific with respect to particular antigens set forth herein and to analogs which are similarly immunospecific, as well as to the recombinant materials useful in the production of these antibodies.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 shows the serum titers of anti-IL-6 antibodies from a xenomouse immunized with human IL-6 and which antibodies contain human kappa light chains and/or human μ heavy chains.
  • FIG. 2 shows the serum titers of anti-IL-8 antibodies from a xenomouse immunized with human IL-8 and which antibodies contain human kappa light chains and/or human μ heavy chains.
  • FIG. 3 shows the serum titers of anti-TNFα antibodies from a xenomouse immunized with human TNF-α and which antibodies contain human kappa light chains and/or human μ heavy chains.
  • FIG. 4 shows the serum titers of anti-CD4 antibodies from a xenomouse immunized with human CD4 and which antibodies contain human kappa light chains and/or human μ heavy chains.
  • FIG. 5 shows the serum titers of a xenomouse immunized with 300.19 cells expressing L-selectin at their surface. In the ELISA assay used, these antibodies are detectable only if they carry human μconstant region heavy chains.
  • FIG. 6 shows the serum titers of a xenomouse immunized with 300.19 cells expressing L-selectin at their surface. In the ELISA assay used, these antibodies are detectable only if they carry human kappa light chains.
  • FIG. 7 shows the serum titers of a xenomouse immunized with 300.19 cells expressing L-selectin. In this ELISA, these antibodies are detectable if they carry human kappa light chain and/or murine y constant regions.
  • FIG. 8 shows a FACS analysis of human neutrophils coupled to sera from a xenomouse (A195-2) immunized with human L-selectin and labeled with an antibody immunoreactive with murine heavy chain γ constant region.
  • FIG. 9 shows a FACS analysis of human neutrophils incubated with serum from a xenomouse (A195-2) immunized with human L-selectin and labeled with an antibody immunoreactive with human light chain kappa region.
  • FIG. 10 is a diagram of a plasmid used to transfect mammalian cells to effect the production of the human protein gp39.
  • FIG. 11 represents the serum titration curve of mice immunized with CHO cells expressing human gp39. The antibodies detected in this ELISA must be immunoreactive with gp39 and contain human heavy chain μ constant regions or human kappa light chains.
  • FIG. 12 shows the results of a FACS analysis of antibodies from a xenomouse (labeled A247-4) immunized with human gp39 reacted with activated human T cells. FIG. 12A shows the separation of human activated T cells into CD4+ and CD4− populations. Panel B shows the results of a FACS analysis of the activated CD4+ T cells with antibodies from the xenomouse immunized with gp39 which contain murine heavy chain γ constant regions; panel C shows the corresponding results with respect to CD4− populations.
  • FIG. 13 is a titration curve with respect to monoclonal antibodies secreted by the hybridoma clone D5.1. This clone is obtained from a xenomouse immunized with tetanus toxin C (TTC) and contains human kappa light chain and human μ constant region in the heavy chain.
  • FIG. 14 is a titration curve with respect to the hybridoma supernatant from clone K4.1. This hybridoma clone is obtained from a xenomouse immunized with TTC and contains human kappa light chain and heavy chain having the murine γ constant region.
  • FIG. 15 shows binding curves for various concentrations of the K4.1 monoclonal antibody in a determination of the affinity of the monoclonal with its antigen in a BIAcore instrument.
  • FIG. 16 shows the complete nucleotide sequence of the heavy chain from the antibody secreted by K4.1.
  • FIG. 17 shows the complete nucleotide sequence of the light chain from the antibody secreted by K4.1.
  • FIG. 18 shows the complete nucleotide sequence of the heavy chain from the antibody secreted by D5.1.
  • FIG. 19 shows the complete nucleotide sequence of the light chain from the antibody secreted by D5.1.
  • MODES OF CARRYING OUT THE INVENTION
  • In general, the methods of the invention include administering an antigen for which human forms of immunospecific reagents are desired to a transgenic nonhuman animal which has been modified genetically so as to be capable of producing human, but not endogenous, antibodies. Typically, the animal has been modified to disable the endogenous heavy and/or light chain loci in its genome, so that these endogenous loci are incapable of the rearrangement required to generate genes encoding immunoglobulins in response to an antigen. In addition, the animal will have been provided, stably, in its genome, at least one human heavy chain locus and at least one human light chain locus so that in response to an administered antigen, the human loci can rearrange to provide genes encoding human immunoglobulins immunospecific for the antigen.
  • The details for constructing such an animal useful in the method of the invention are provided in the PCT application WO 94/02602 referenced above.
  • For production of the desired antibodies, the first step is administration of the antigen. Techniques for such administration are conventional and involve suitable immunization protocols and formulations which will depend on the nature of the antigen per se. It may be necessary to provide the antigen with a carrier to enhance its immunogenicity and/or to include formulations which contain adjuvants and/or to administer multiple injections, and the like. Such techniques are standard and optimization of them will depend on the characteristics of the particular antigen for which immunospecific reagents are desired.
  • As used herein, the term “immunospecific reagents” includes immunoglobulins and their analogs. The term “analogs” has a specific meaning in this context. It refers to moieties that contain the fully human portions of the immunoglobulin which account for its immunospecificity. In particular, variable regions including the complementarity determining regions (CDRs) are required, along with sufficient portions of the framework regions (FRs) to result in the appropriate three dimensional conformation. Typical immunospecific analogs of antibodies include F(ab′) 2 , Fab′, and Fab regions. Modified forms of the variable regions to obtain, for example, single chain FV analogs with the appropriate immunospecificity are known. A review of such FV construction is found, for example, in Tibtech (1991) 9:______. The construction of antibody analogs with multiple immunospecificities is also possible by coupling the human variable regions derived from antibodies with varying specificities.
  • The variable regions with fully human characteristics can also be coupled to a variety of additional substances which can provide toxicity, biological functionality, alternative binding specificities and the like. The moieties including the fully human variable regions produced by the methods of the invention include single-chain fusion proteins, molecules coupled by covalent methods other than those involving peptide linkages, and aggregated molecules. Examples of analogs which include variable regions coupled to additional molecules covalently or noncovalently include those in the following nonlimiting illustrative list. Traunecker, A. et al. Int J Cancer Supp (1992) Supp 7:51-52 describe the bispecific reagent janusin in which the FV region directed to CD3 is coupled to soluble CD4 or to other ligands such as OVCA and IL-7. Similarly, the fully human variable regions produced by the method of the invention can be constructed into FV molecules and coupled to alternative ligands such as those illustrated in the cited article. Higgins, P. J. et al. J Infect Disease (1992) 166:198-202 describe a heteroconjugate antibody composed of OKT3 cross-linked to an antibody directed to a specific sequence in the V3 region or GP120. Such heteroconjugate antibodies can also be constructed using at least the human variable regions contained in the immunoglobulins produced by the invention methods. Additional examples of bispecific antibodies include those described by Fanger, M. W. et al. Cancer Treat Res (1993) 68:181-194 and by Fanger, M. W. et al. Crit Rev Immunol (1992) 12:101-124. Conjugates that are immunotoxins including conventional antibodies have been widely described in the art. The toxins may be coupled to the antibodies by conventional coupling techniques or immunotoxins containing protein toxin portions can be produced as fusion proteins. The analogs of the present invention can be used in a corresponding way to obtain such immunotoxins. Illustrative of such immunotoxins are those described by Byrs, B. S. et al. Seminars Cell Biol (1991) 2:59-70 and by Fanger, M. W. et al. Immunol Today (1991) 12:51-54.
  • In short, the genes encoding the immunoglobulins produced by the transgenic animals of the invention can be retrieved and the nucleotide sequences encoding the fully human variable region can be manipulated according to known techniques to provide a variety of analogs such as those described above. In addition, the immunoglobulins themselves containing the human variable regions can be modified using standard coupling techniques to provide conjugates retaining immunospecificity and fully human characteristics in the immunospecific region.
  • Thus, immunoglobulin “analogs” refers to moieties which contain those portions of the antibodies of the invention which retain their human characteristics and their immunospecificity. These will retain sufficient human variable region to provide the desired specificity.
  • As stated above, all of the methods of the invention include administering the appropriate antigen to the transgenic animal. The recovery or production of the antibodies themselves can be achieved in various ways.
  • First, and most straightforward, the polyclonal antibodies produced by the animal and secreted into the bloodstream can be recovered using known techniques. Purified forms of these antibodies can, of course, be readily prepared by standard purification techniques, preferably including affinity chromatography with respect to the particular antigen, or even with respect to the particular epitope of the antigen for which specificity is desired. In any case, in order to monitor the success of immunization, the antibody levels with respect to the antigen in serum will be monitored using standard techniques such as ELISA, RIA and the like.
  • It will be noted, from the examples below, that a portion of the polyclonal antiserum obtained may include an endogenous heavy chain constant region derived from the host, even though the variable regions are fully human. Under these circumstances, to the extent that an application requires fully human antibodies, use of the polyclonal antiserum directly would be inappropriate. However, the presence of these chimeras, which is believed to result from in vivo isotype switching as described by Gerstein et al. Cell (1990) 63:537, is not problematic, in view of conventional purification and modification methods and in view of the availability of alternative methods to recover fully human antibodies, if desired, described in the following paragraphs.
  • First, and most simply, the polyclonal antiserum could be subjected to suitable separation techniques to provide compositions containing only fully human immunoglobulins. Portions of the serum which display characteristics of the host species can be removed, for example, using affinity reagents with the appropriate anti species immunoglobulins or immunospecific portions thereof. Furthermore, for applications where only the variable regions of the antibodies are required, treating the polyclonal antiserum with suitable reagents so as to generate Fab, Fab′, or F(ab′) 2 portions results in compositions containing fully human characteristics. Such fragments are sufficient for use, for example, in immunodiagnostic procedures involving coupling the immunospecific portions of immunoglobulins to detecting reagents such as radioisotopes. Thus, for some applications, the polyclonal antiserum can be treated to provide compositions with the desired characteristics including compositions consisting essentially of fully human antibodies and compositions including immunoglobulin analogs wherein the immunospecific portion is fully human.
  • Alternatively, immunoglobulins and analogs with desired characteristics can be generated from immortalized B cells derived from the transgenic animals used in the method of the invention or from the rearranged genes provided by these animals in response to immunization. It will be apparent that hybridomas derived from the B cells of the immunized animal can be screened so as to choose only those secreting fully human antibodies and that the genetic material can be recovered from the hybridomas or from lymphocytes in spleen, blood, or lymph nodes of the immunized animal and manipulated using conventional techniques to replace any endogenous constant region with a human one or to produce a desired analog.
  • Thus, as an alternative to harvesting the antibodies directly from the animal, the B cells can be obtained, typically from the spleen, but also, if desired, from the peripheral blood lymphocytes or lymph nodes and immortalized using any of a variety of techniques, most commonly using the fusion methods described by Kohler and Milstein. The resulting hybridomas (or otherwise immortalized B cells) can then be cultured as single colonies and screened for secretion of antibodies of the desired specificity. As described above, the screen can also include a determination of the fully human character of the antibody. For example, as described in the examples below, a sandwich ELISA wherein the monoclonal in the hybridoma supernatant is bound both to antigen and to an antihuman constant region can be employed. Conversely, hybridomas that secrete antibodies which are immunoreactive with antispecies antibodies directed to the species of the immunized animal can be discarded. After the appropriate hybridomas are selected, the desired antibodies can be recovered, again using conventional techniques. They can be prepared in quantity by culturing the immortalized B cells using conventional methods, either in vitro, or in vivo to produce ascites fluid. Purification of the resulting monoclonal antibody preparations is less burdensome than in the case of serum since each immortalized colony will secrete only a single type of antibody. In any event, standard purification techniques to isolate the antibody from other proteins in the culture medium can be employed.
  • As an alternative to obtaining human immunoglobulins directly from the culture of immortalized B cells derived from the animal, the immortalized cells can be used as a source of rearranged heavy chain and light chain loci for subsequent expression and/or genetic manipulation. Isolation of genes from such antibody-producing cells is straightforward since high levels of the appropriate mRNAs are available for production of a cDNA library. The recovered rearranged loci can be manipulated as desired. For example, the constant region can be exchanged for that of a different isotype or that of a human antibody, as described above, or eliminated altogether. The variable regions can be linked to encode single chain FV regions. Multiple FV regions can be linked to confer binding ability to more than one target or chimeric heavy and light chain combinations can be employed. Once the genetic material is available, design of analogs as described above which retain their ability to bind the desired target, as well as their human characteristics, is straightforward.
  • Once the appropriate genetic material is obtained and, if desired, modified to encode an analog, the coding sequences including those that encode, at a minimum, the variable regions of the human heavy and light chain can be inserted into expression systems contained on vectors which can be transfected into standard recombinant host cells. As described below, a variety of such host cells may be used; for efficient processing, however, mammalian cells are preferred. Typical mammalian cell lines useful for this purpose include CHO cells, 293 cells, or NSO-GS cells.
  • The production of the antibody or analog is then undertaken by culturing the modified recombinant host under culture conditions appropriate for the growth of the host cells and the expression of the coding sequences. The antibodies are then recovered from the culture. The expression systems are preferably designed to include signal peptides so that the resulting antibodies are secreted into the medium; however, intracellular production is also possible.
  • In addition to deliberate design of modified forms of the immunoglobulin genes to produce analogs, advantage can be taken of phage display techniques to provide libraries containing a repertoire of antibodies with varying affinities for the desired antigen. For production of such repertoires, it is unnecessary to immortalize the B cells from the immunized animal; rather the primary B cells can be used directly as a source of DNA. The mixture of cDNAs obtained from B cells, e.g., derived from spleens, is used to prepare an expression library, for example, a phage display library transfected into E. coli. The resulting cells are tested for immunoreactivity to the desired antigen. Techniques for the identification of high affinity human antibodies from such libraries are described by Griffiths, A. D., et al., EMBO J (1994) 13:3245-3260; by Nissim, A., et al. ibid,. 692-698, and by Griffiths, A. D., et al., ibid, 725-734. Ultimately, clones from the library are identified which produce binding affinities of a desired magnitude for the antigen, and the DNA encoding the product responsible for such binding is recovered and manipulated for standard recombinant expression. Phage display libraries may also be constructed using previously manipulated nucleotide sequences and screened in similar fashion. In general, the cDNAs encoding heavy and light chain are independently supplied or are linked to form FV analogs for production in the phage library.
  • The phage library is thus screened for the antibodies with highest affinity for the antigen and the genetic material recovered from the appropriate clone. Further rounds of screening can increase the affinity of the original antibody isolated. The manipulations described above for recombinant production of the antibody or modification to form a desired analog can then be employed.
  • As above, the modified or unmodified rearranged loci are manipulated using standard recombinant techniques by constructing expression systems operable in a desired host cell, such as, typically, a Chinese hamster ovary cell, and the desired immunoglobulin or analog is produced using standard recombinant expression techniques, and recovered and purified using conventional methods.
  • The application of the foregoing processes to antibody production has enabled the preparation of human immunospecific reagents with respect to antigens for which human antibodies have not heretofore been available. The immunoglobulins that result from the above-described methods and the analogs made possible thereby, provide novel compositions for use in analysis, diagnosis, research, and therapy. The particular use will, of course, depend on the immunoglobulin or analog prepared. In general, the compositions of the invention will have utilities similar to those ascribable to nonhuman antibodies directed against the same antigen. Such utilities include, for example, use as a affinity ligands for purification, as reagents in immunoassays, as components of immunoconjugates, and as therapeutic agents for appropriate indications.
  • Particularly in the case of therapeutic agents or diagnostic agents for use in vivo, it is highly advantageous to employ antibodies or their analogs with fully human characteristics. These reagents avoid the undesired immune responses engendered by antibodies or analogs which have characteristics marking them as originating from non-human species. Other attempts to “humanize” antibodies do not result in reagents with fully human characteristics. For example, chimeric antibodies with murine variable regions and human constant regions are easily prepared, but, of course, retain murine characteristics in the variable regions. Even the much more difficult procedure of “humanizing” the variable regions by manipulating the genes encoding the amino acid sequences that form the framework regions does not provide the desired result since the CDRs, typically of nonhuman origin, cannot be manipulated without destroying immunospecificity. Thus, the methods of the present invention provide, for the first time, immunoglobulins that are fully human or analogs which contain immunospecific regions with fully human characteristics.
  • There are large numbers of antigens for which human antibodies and their human analogs would be made available by the methods of the invention. These include the following as a nonlimiting set:
      • leukocyte markers, such as CD2, CD3, CD4, CD5, CD6, CD7, CD8, CD11a,b,c, CD13, CD14, CD18, CD19, CD20, CD22. CD23. CD27 and its liqand, CD28 and its ligands B7.1, B7.2, B7.3, CD29 and its ligand, CD30 and its ligand, CD40 and its ligand gp39, CD44, CD45 and isoforms, CDw52 (Campath antigen), CD56, CD58, CD69, CD72, CTLA-4, LFA-1 and TCR
      • histocompatibility antigens, such as MHC class I or II, the Lewis Y antigens, SLex, SLey, SLea, and SLeb;
      • integrins, such as VLA-1, VLA-2, VLA-3, VLA-4, VLA-5, VLA-6, and LFA-1;
      • adhesion molecules, such as Mac-1 and p150, 95;
      • selectins, such as L-selectin, P-selectin, and E-selectin and their counterreceptors VCAM-1, ICAM-1, ICAM-2, and LFA-3;
      • interleukins, such as IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-10, IL-ll, IL-12, IL-13, IL-14, and IL-15;
      • interleukin receptors, such as IL-1R, IL-2R, IL-4R, IL-5R, IL-6R, IL-7R, IL-8R, IL-10R, IL-11R, IL-12R, IL-13R, IL-14R, and IL-15R;
      • chemokines, such as PF4, RANTES, MIPla, MCP1, NAP-2, Groα, Groβ, and IL-8;
      • growth factors, such as TNFalpha, TGFbeta, TSH, VEGF/VPF, PTHrP, EGF family, FGF, PDGF family, endothelin, and gastrin releasing peptide (GRP);
      • growth factor receptors, such as TNFalphaR, RGFbetaR, TSHR, VEGFR/VPFR, FGFR, EGFR, PTHrPR, PDGFR family, EPO-R, GCSF-R and other hematopoietic receptors;
      • interferon receptors, such as IFNαR, IFNβR, and IFNγR;
      • Igs and their receptors, such as IgE, FceRI, and FCeR11;
      • tumor antigens, such as her2-neu, mucin, CEA and endosialin;
      • allergens, such as house dust mite antigen, lol p1 (grass) antigens, and urushiol;
      • viral proteins, such as CMV glycoproteins B, H, and gCIII, HIV-1 envelope glycoproteins, RSV envelope glycoproteins, HSV envelope glycoproteins, EBV envelope glycoproteins, VZV envelope glycoproteins, HPV envelope glycoproteins, Hepatitis family surface antigens;
      • toxins, such as pseudomonas endotoxin and osteopontin/uropontin, snake venom, and bee venom;
      • blood factors, such as complement C3b, complement C5a, complement C5b-9, Rh factor, fibrinogen, fibrin, and myelin associated growth inhibitor;
      • enzymes, such as cholesterol ester transfer protein, membrane bound matrix metalloproteases, and glutamic acid decarboxylase (GAD); and
      • miscellaneous antigens including ganglioside GD3, ganglioside GM2, LMP1, LMP2, eosinophil major basic protein, eosinophil cationic protein, pANCA, Amadori protein, Type IV collagen, glycated lipids, γ-interferon, A7, P-glycoprotein and Fas (AFO-1) and oxidized-LDL.
  • Particularly preferred immunoglobulins and analogs are those immunospecific with respect to human IL-6, human IL-8, human TNFα, human CD4, human L-selectin, and human gp39. Human antibodies against IL-8 are particularly useful in preventing tumor metastasis and inflammatory states such as asthma. Antibodies and analogs immunoreactive with human TNFα and human IL-6 are useful in treating cachexia and septic shock as well as autoimmune disease. Antibodies and analogs immunoreactive with gp39 or with L-selectin are also effective in treating or preventing autoimmune disease. In addition, anti-gp39 is helpful in treating graft versus host disease, in preventing organ transplant rejection, and in treating glomerulonephritis. Antibodies and analogs against L-selectin are useful in treating ischemia associated with reperfusion injury.
  • Typical autoimmune diseases which can be treated using the above-mentioned antibodies and analogs include systemic lupus erythematosus, rheumatoid arthritis, psoriasis, Sjogren's, scleroderma, mixed connective tissue disease, dermatomyositis, polymyositis, Reiter's syndrome, Behcet's disease, Type 1 diabetes, Hashimoto's thyroiditis, Grave's disease, multiple sclerosis, myasthenia gravis and pemphigus.
  • The examples below are intended to illustrate but not to limit the invention.
  • In these examples, mice, designated “xenomice”, are used for initial immunizations. A detailed description of such xenomice is found in the above referenced PCT application WO 94/02602. Immunization protocols appropriate to each antigen-are described in the specific examples below. The sera of the immunized xenomice (or the supernatants from immortalized B cells) were titrated for antigen specific human antibodies in each case using a standard ELISA format. In this format, the antigen used for immunization was immobilized onto wells of microtiter plates. The plates were washed and blocked and the sera (or supernatants) were added as serial dilutions for 1-2 hours of incubation. After washing bound antibody having human characteristics was detected by adding the appropriate antispecies Ig (typically antihuman kappa or antihuman μ) conjugated to horseradish peroxidase (HRP) for one hour. In some cases, the bound antibodies were tested for murine characteristics using antimurine antibodies, typically antimurine γ. After again washing, the chromogenic reagent o-phenylene diamine (OPD) substrate and hydrogen peroxide were added and the plates were read 30 minutes later at 492=m using a microplate reader.
  • Unless otherwise noted, the antigen was coated using plate coating buffer (0.1 M carbonate buffer, pH 9.6); the assay blocking buffer used was 0.5% BSA, 0.1% Tween 20 and 0.01% Thimerosal in PBS; the substrate buffer used in color development was citric acid 7.14 g/l: dibasic sodium phosphate 17.96 g/l; the developing solution (made immediately before use) was 10 ml substrate buffer, 10 mg OPD, plus 5 ml hydrogen peroxide; the stop solution (used to stop color development) was 2 M sulfuric acid. The wash solution was 0.05% Tween 20 in PBS.
  • EXAMPLE 1 Human Antibodies Against Human IL-6
  • Three to 5 xenomice aged 8-20 weeks were age-matched and immunized intraperitoneally with 50 μg human IL-6 emulsified in complete Freund's adjuvant for primary immunization and in incomplete Freund's adjuvant for subsequent injections. The mice received 6 injections 2-3 weeks apart. Serum titers were determined after the second dose and following each dose thereafter. Bleeds were performed 6-7 days after injections from the retrobulbar plexus. The blood was allowed to clot at room temperature for about 2 hours and then incubated at 4° C. for at least 2 hours before separating and collecting the sera.
  • ELISAs were conducted as described above by applying 100 μl per well of recombinant human IL-6 at 2 mg/ml in coating buffer. Plates were then incubated at 4° C. overnight or at 37° C. for 2 hours and then washed three times in washing buffer. Addition of 100 μl/well blocking buffer was followed by incubation at room temperature for 2 hours, and an additional 3 washes.
  • Then, 50 μl/well of diluted serum samples (and positive and negative controls) were added to the plates. Plates were then incubated at room temperature for 2 hours and again washed 3 times.
  • After washing, 100 μl per well of either HRP-mouse antihuman IgM at 1/2,000 or HRP-mouse antihuman kappa at 1/2,000, diluted in blocking buffer were added. After a 1 hour incubation at room temperature, the plates were washed 3 times and developed with OPD substrate for 10-25 minutes. 50 μl/well of stop solution were then added and the results read on an ELISA plate reader at 492 nm. The dilution curves resulting from the titration of serum from xenomouse A40-7 after 6 injections are shown in FIG. 1. The data in FIG. 1 show production of anti-IL-6 immunoreactive with antihuman kappa and antihuman μ detectable at serum dilutions above 1:1,000.
  • EXAMPLE 2 Human Antibodies Against Human IL-8
  • Immunization and serum preparation were as described in Example 1 as except that human recombinant IL-8 was used as an immunogen.
  • ELISA assays were performed with respect to the recovered serum, also exactly as described in Example 1, except that the ELISA plates were initially coated using 100 μl/well of recombinant human IL-8 at 0.5 mg/ml in the coating buffer. The results obtained for various serum dilutions from xenomouse A260-5 after 6 injections are shown in FIG. 2. Human anti-IL-8 reactivity was again shown at serum dilutions having concentrations higher than that represented by a 1:1,000 dilution.
  • EXAMPLE 3 Human Antibodies Against Human TNFα
  • Immunization and serum preparation were conducted as described in Example 1 except that human recombinant TNFα was substituted for human IL-6. ELISAs were conducted as described in Example 1 except that the initial coating of the ELISA plate employed 100 μl/well recombinant human TNFα at 1 mg/ml in coating buffer.
  • The dilution curves for serum from xenomouse A210-8 after 6 injections obtained are shown in FIG. 3. Again significant titers of human anti-TNFα reactivity were shown.
  • EXAMPLE 4 Human Antibodies Against Human CD4
  • The human CD4 antigen was prepared as a surface protein on transfected recombinant cells as follows: Human CD4 zeta (F15 LTR) as described in Roberts, et al., Blood (1994) 84:2878 was introduced into the rat basophil leukemic cell line RBL-2H3, described by Callan, M., et al., Proc Natl Acad Sci USA (1993) 90:10454 using the kat high efficiency transduction system described by Finer, et al., Blood (1994) 83:43. Briefly, RBL-2H3 cells at 106 cells per well were cultured in 750 ml DMEMlow+20% FBS (Gibco) and 16 μg/ml polybrene with an equal volume of proviral supernatant for 2 hours at 37° C., 5% CO2. One ml of medium was removed and 750 μl of infection medium and retroviral supernatant were added to each well and the cultures incubated overnight. The cells were washed and expanded in DMEMlow+10% FBS until sufficient cells were available for sorting. The CD4-zeta transduced RBL-2H3 cells were sorted using the FACSTAR plus (Becton Dickinson). The cells were stained for human CD4 with a mouse antihuman CD4-PE antibody and the top 2-3% expressing cells were selected.
  • Immunizations were conducted as described in Example 1 using 10×106 cells per mouse except that the primary injection was subcutaneous at the base of the neck. The mice received 6 injections 2-3 weeks apart. Serum was prepared and analyzed by ELISA as described in Example 1 except that the initial coating of the ELISA plate utilized 100 μl per well of recombinant soluble CD4 at 2 mg/ml of coating buffer. The titration curve for serum from xenomouse A207-1 after 6 injections is shown in FIG. 4. Titers of human anti-CD4 reactivity were shown at concentrations representing greater than those at 1:1,000 dilution.
  • EXAMPLE 5 Human Antibodies Against Human L-selectin
  • The antigen was prepared as a surface displayed protein in C51 cells, a high expressing clone derived by transfecting the mouse pre-B cell 300.19 with LAM-1 cDNA (LAM-1 is the gene encoding L-selectin) (Tedder, et al., J Immunol (1990) 144:532) or with similarly transfected CHO cells. The transfected cells were sorted using fluorescent activated cell sorting using anti-Leu-8 antibody as label.
  • The C51 and the transfected CHO cells were grown in DME 4.5 g/l glucose with 10% FCS and 1 mg/ml G418 in 100 mm dishes. Negative control cells, 3T3-P317 (transfected with gag/pol/env genes of Moloney virus) were grown in the same medium without G418.
  • Primary immunization was by injection subcutaneously at the base of the neck; subsequent injections were intraperitoneal. 70-100 million C51 or transfected CHO cells were used per injection for a total of five injections 2-3 weeks apart.
  • Sera were collected as described in Example 1 and analyzed by ELISA in a protocol similar to that set forth in Example 1.
  • For the ELISA, the transfected cells were plated into 96 well plates and cell monolayers grown for 1-2 days depending on cell number and used for ELISA when confluent. The cells were fixed by first washing with cold 1×PBS and then fixing solution (5% glacial acetic acid, 95% ethanol) was added. The plates were incubated at −25° C. for 5 minutes and can be stored at this temperature if sealed with plate sealers.
  • The ELISA is begun by bringing the plates to room temperature, flicking to remove fixing solution and washing 5 times with DMEM medium containing 10% FCS at 200 μl per well.
  • The wells were treated with various serum dilutions or with positive or negative controls. Positive control wells contained murine IgG1 to human L-selectin.
  • The wells were incubated for 45 minutes and monolayer integrity was checked under a microscope. The wells were then incubated with either HRP-antimouse IgG (1/1000) or with the antihuman kappa or antihuman μ conjugates described in Example 1. The plates were then washed with 1% BSA/PBS and again with PBS and monolayer integrity was checked. The plates were developed, stopped, and read as described above. The results for serum from xenomouse A303-3 are shown in FIGS. 5 and 6; human antibodies both to L-selectin and control 3T3 cells were obtained. However, the serum titers are higher for the L-selectin-expressing cells as compared to parental 3T3 cells. These results show that xenomouse A303-3 produces antibodies specific for L-selectin with human μ heavy chain regions and/or human kappa light chains.
  • ELISAs were also performed using as the immobilized antigen a fusion protein consisting of the extracellular domain of human L-selectin fused to the constant domain of human IgG1 (Guo, et al., Cell Immunol (1994) 154:202). The L-selectin fusion protein was made by transient transfection of human 293 cells using calcium phosphate transfection (Wigler, M., Cell (1979) 16:777). Serum preparation was performed as described in Example 1. ELISAs were conducted essentially as in Example 1, except that the initial coating of the ELISA plate employed 100 μl transfected 293 cell culture supernatant containing the L-selectin-Ig fusion protein. Detection employed HRP-mouse antihuman kappa and HRP-goat antimouse IgG.
  • FIG. 7 shows the results from xenomouse A195-2; antibodies specific for L-selectin having human kappa light chains and/or murine heavy chain γ regions are present in the serum.
  • The antisera obtained from the immunized xenomice were also tested for staining of human neutrophils. Human neutrophils were prepared as follows: peripheral blood was collected from normal volunteers with 100 units/ml heparin. About 3.5 ml blood was layered over an equal volume of One-step Polymorph Gradient (Accurate Chemical, Westbury, N.Y.) and spun for 30 minutes at 450×G at 20° C. The neutrophil fraction was removed and washed twice in DPBS/2% FBS.
  • The neutrophils were then stained with either:
      • (1) antiserum from xenomouse A195-2 immunized with C51 cells (expressing L-selectin);
      • (2) as a positive control, mouse monoclonal antibody LAM1-3 (against L-selectin); and
      • (3) as negative control, antiserum from a xenomouse immunized with cells expressing human gp39.
  • The stained, washed neutrophils were analyzed by FACS. The results for antiserum from xenomouse A195-2 are shown in FIGS. 8 and 9.
  • These results show the presence of antibodies in immunized xenomouse serum which contain fully human variable regions immunoreactive with L-selectin. The negative control antiserum from mice immunized with gp39 does not contain antibodies reactive against human neutrophils. Serum from A195-2 (immunized with L-selectin-expressing cells) contains antibodies binding to human neutrophils detectable with a goat antimouse IgG (FIG. 8), which immunoreacts with heavy chain protein composed of fully human variable regions and mouse γ constant regions. Staining with anti L-selectin xenomouse antisera detected with a mouse monoclonal antibody against human kappa is shown in FIG. 9, showing the presence of fully human kappa light chain.
  • As explained above, these antibodies containing human variable regions are readily convertible to fully human antibodies. For example, using hybridomas secreting these antibodies, the cDNAs encoding them can be obtained. By amplifying the genes encoding human V regions using primers containing restriction enzyme recognition sites and cloning them into plasmids containing the coding sequences for human constant regions as described by Queen, et al., Proc Natl Acad Sci (1989) 86:10029, genes encoding the fully human antibodies can be obtained for recombinant production.
  • EXAMPLE 6 Human Antibodies Against Human g039
  • gp39 (the ligand for CD40) is expressed on activated human CD4+ T cells. The sera of xenomice immunized with recombinant gp39 according to this example contained antibodies immunospecific for gp39 with fully human variable regions; the sera contained fully human IgM antibodies and chimeric IgG antibodies containing human variable regions and murine constant heavy chain y region.
  • The antigen consisted of stable transfectants of 300.19 cells or of CHO cells expressing gp39 cDNA cloned into the mammalian expression vector P1K1.HUgp39/IRES NEO as shown in FIG. 10. CHO cells were split 1:10 prior to transfection in DMEM 4.5 g/l glucose, 10% FBS, 2 mM glutamine, MEM, NEAA supplemented with additional glycine, hypoxanthine and thymidine. The cells were cotransfected with the gp39 vector at 9 μg/10 cm plate (6×105 cells) and the DHFR expressing vector pSV2DHFRs (Subranani et al. Mol Cell Biol (1981) 9:854) at 1 μg/10 cm plate using calcium phosphate transfection. 24 hours later the cells were split 1:10 into the original medium containing G418 at 0.6 mg/ml. Cells producing gp39 were sorted by FACS using an anti-gp39 antibody.
  • Mice grouped as described in Example 1 were immunized with 300.19 cells expressing gp39 using a primary immunization subcutaneously at the base of the neck and with secondary intraperitoneal injections every 2-3 weeks. Sera were harvested as described in Example 1 for the ELISA assay. The ELISA procedure was conducted substantially as set forth in Example 1; the microtiter plates were coated with CHOD-gp39 cells grown in a 100 mm dish in DMEM, 4.5 g/l glucose, 10% FCS, 4 mM glutamine, and nonessential amino acid (NEAA) solution for MEM (100×). On the day preceding the ELISA assay, the cells were trypsinized and plated into 96-well filtration plates at 105 cells/200 μl well and incubated at 37° C. overnight. The positive controls were mouse antihuman gp39; negative controls were antisera from mice immunized with an antigen other than gp39. 50 μl of sample were used for each assay. The remainder of the assay is as described in Example 1.
  • The dilution curves for the sera obtained after 4 injections from mice immunized with gp39 expressed on CHO cells are shown in FIG. 11. As shown, the sera contained antihuman gp39 immunospecificity which is detectable with human kappa and human μ HRP-coupled antibodies.
  • In addition, the sera were tested for their ability to react with activated human T cells included in PBMC using FACS analysis. To prepare the PBMC, human peripheral blood was collected from normal volunteers with the addition of 100 unit/ml heparin. PBMC were isolated over Ficoll gradient and activated with 3 μg/ml PHA, 1 μg/ml PMA in IMDM plus 10% FBS plus 25 μM 2-mercaptoethanol for 4 hours. After washing, the PBMC were stained with mouse Mab against human CD4 labeled with FITC to permit separation of activated human T cells from unactivated cells.
  • The activated CD4+ and CD4−T cells were then analyzed by FACS using staining with either:
      • 1) antiserum from a Xenomouse immunized with 300.19 cells producing gp39;
      • 2) a positive control mouse Mab directed against a-CD40L (human gp39); and
      • 3) a negative control antiserum from a Xenomouse immunized with TNF.
  • The detecting antibody in the FACS analysis was goat antimouse IgG (PE). The results are shown in FIG. 12.
  • As shown in FIG. 12A, CD4+ (R2) and CD4− (R3) cells were separated prior to FACS analysis. Panel B shows the results for CD4+ cells and shows that sera from mice immunized with gp39 (labeled A247-4 in the figure) reacted with these activated PBMC; panel C shows that these sera did not react with CD4− cells. These antibodies carried immune heavy chain γ constant regions. The results of panels B and C confirm that the TNF-injected xenomouse did not make antibodies against activated human T cells.
  • EXAMPLE 7 Preparation of High-Affinity Human Mabs Against Tetanus Toxin
  • The antibodies prepared in this example were secreted by hybridomas obtained by immortalizing B cells from xenomice immunized with tetanus toxin. The immunization protocol was similar to that set forth in Example 1 using 50 μg tetanus toxin emulsified in complete Freund's adjuvant for intraperitoneal primary immunization followed by subsequent intraperitoneal injections with antigen incorporated into incomplete Freund's adjuvant. The mice received a total of 4 injections 2-3 weeks apart.
  • After acceptable serum titers of antitetanus toxinC (anti-TTC) were obtained, a final dose of antigen in PBS was given 4 days before the animals were sacrificed and the spleens harvested for fusion.
  • The spleen cells were fused with myeloma cells P3×63-Ag8.653 as described by Galfre, G. and Milstein, C. Methods in Enzymology (1981) 73:3-46.
  • After fusion the cells were resuspended in DMEM, 15% FCS, containing HAT supplemented with glutamine, pen/strep for culture at 37° C. and 10% CO2. The cells were plated in microtiter trays and maintained in HAT-supplemented medium for two weeks before transfer to HT-supplemented media. Supernatants from wells containing hybridomas were collected for a primary screen using an ELISA.
  • The ELISA was conducted as described in Example 1 wherein the antigen coating consisted of 100 μl/well of tetanus toxin C (TTC) protein at 2 mg/ml in coating buffer, followed by incubation at 4° C. overnight or at 37° C. for two hours. In the primary ELISA, HRP-conjugated goat antimouse IgG at 1/2000 was used in addition to HRP mouse antihuman IgM as described in Example 1. Two hybridomas that secreted anti-TTC according to the ELISA assay, clone D5.1 and clone K4.1 were used for further analysis.
  • As shown in FIG. 13, clone D5.1 secretes fully human anti-TTC which is detectable using HRP-conjugated antihuman μ and HRP-conjugated antihuman kappa. FIG. 14 shows that clone K4.1 secretes anti-TTC which is immunoreactive with antimurine γ and antihuman kappa HRP-conjugated antibodies. Thus, clone K4.1 provides anti-TTC with human variable regions and a murine constant heavy chain γ region.
  • The antibodies secreted by D5.1 and K4.1 did not immunoreact in ELISAs using TNFα, IL-6, or IL-8 as immobilized antigen under conditions where positive controls (sera from xenomice immunized with TNFα, IL-6 and IL-8 respectively) showed positive ELISA results.
  • The affinity of the monoclonal antibodies secreted by K4.1 for TTC antigen was determined using commercially available reagents and instrumentation. BIAcore Instrument, CM5 sensor chips, surfactant P20 and the amine coupling kit were purchased from Pharmacia Biosensor (Piscataway, N.J.). TTC was immobilized at two levels of antigen density on the surface of the sensor chips according to the manufacturer's instructions. Briefly, after washing and equilibrating the instrument with buffer containing surfactant, the surfaces were activated and the TCC was immobilized.
  • For high antigen density, the surface was activated with 35 μl of equal volumes 0.1 M NHS and 0.1 M EDC injected across the surface followed by 30 μl of TTC fragment at 100 μg/ml in 10 mM sodium acetate buffer pH 5.0. The surface was blocked by injecting 35 μl 1 M ethanolamine and washed to remove noncovalently bound TCC using 5 μl 0.1 M HCl. The entire immobilization procedure was conducted with a continuous flow of buffer at 5 μl/min. This results in about 7500-8500 response units (RU) of TTC per chip. (1000 RU corresponds to about 1 ng of protein per mm2.)
  • For chips with low antigen density, the procedure utilizes 15 μl rather than 30 μl of TTC, resulting in chips containing 550-950 RU.
  • Chips could be regenerated after use in single determinations by injecting 10 μl formal or MgCl2.
  • The chips are used to determine binding affinities by determining ka and kb (the association and dissociation rate constants) for the antibody with respect to the immobilized TTC. The association rate constant is measured over six minutes at a flow rate of 5 μl/min. at different concentrations of K4.1 Mab in the range of 2.16 nm-69.33 nm. The dissociation rate constant is measured at a constant buffer flow rate of 5 μl/min after completion of the antibody injection. The raw data are graphed in FIG. 15 and the calculated results are shown in Table 1.
    TABLE 1
    Kinetic Constants of K4.1 Measured Using the BIAcore on Two Different Surface
    Figure US20060040363A1-20060223-P00899
    Association Dissociation Binding Dis
    Figure US20060040363A1-20060223-P00899
    Immobilized K4.1 conc. rate rate constant c
    Figure US20060040363A1-20060223-P00899
    tetanus toxinC range nM ka(105 M−1s−1) kd(105s−1) KA(M−1) = ka/kd KD
    Figure US20060040363A1-20060223-P00899
    5931 RU 4.3-34.7 6.47 ± 1.05 4.02 ± 1.42 1.6 × 1010 0.6
    Figure US20060040363A1-20060223-P00899
     868 RU 4.3-34.7 7.19 ± 2.18 2.02 ± 1.01 3.5 × 1010 0.2
    Figure US20060040363A1-20060223-P00899
  • As shown, the K4.1 antibody has a binding constant for TTC somewhat larger than 1010 l/mol.
  • The complete nucleotide sequence of the cDNAs encoding the heavy and light chains of the K4.1 and D5.1 monoclonals were determined as shown in FIGS. 16-19. PolyA mRNA was isolated from about 106 hybridoma cells and used to generate cDNA using random hexamers as primers. Portions of the product were amplified by PCR using the appropriate primers.
  • Both cell lines were known to provide human kappa light chains; for PCR amplification of light chain encoding cDNA, the primers used were HKP1 (5′-CTCTGTGACACTCTCCTGGGAGTT-3′) for priming from the constant region terminus and two oligos, used in equal amounts to prime from the variable segments: B3 (5′-CCACCATCAACTGCAAGTCCAGCCA-3′) and B2/B1 (5′-GAAACGACACTCACGCAGTCTCCAGC-3′).
  • For amplification of the heavy chain from K4.1 (which contains the murine γ1 constant region), the primers were MG-24Vi for the human variable regions: 5′-CAGGTGCAGCTGGAGCAGTCiGG-3′ which, with inosine as shown recognizes the human variable regions VH1-2, VH1-3, VH4 and VH6, and from the constant region MG-25 i.e., 5′-GCACACCGCTGGACAGGGATCCAiAGTTTC-3′, which, containing inosine as shown recognizes murine γ1, γ2A, γ2B, and γ3.
  • For amplification of the heavy chain of the antibody derived from D5.1 (which contains the human μ constant region), MG-24VI was used to prime from the variable and μP1 (5′-TTTTCTTTGTTGCCGTTGGGGTGC-3′) was used to prime from the constant region terminus.
  • Turning first to the results shown in FIG. 16 representing the heavy chain of the Mab secreted by K4.1, the sequence shows the presence of the human variable segment VH6, the human diversity region DN1, and the human joining segment JH4 linked to the murine γ1 constant region. Nine base-pair mutations from the published germline sequence were present in the variable region, two of them within CDR2. One mutation was observed in the D segment. Three nongermline nucleotide additions were present in the DH-JH junction.
  • Referring to FIG. 17 which shows the light chain of the K4.1 antibody, analysis shows the presence of the human kappa variable region B3 and joining region JK4. Eight nucleotides are missing from B3 at the VK-JK junction and four mutations were found in the variable region. Five nongermline nucleotide additions were present at the VK-JK junction.
  • Referring now to FIG. 18 which sets forth the sequence for the heavy chain of the antibody secreted by clone D5.1, this shows the heavy chain is comprised of the human variable fragment VH6, the human diversity region DN1 and the human joining segment JH4 linked to the human μ constant region. There were two base-pair mutations from the germline sequence in the variable region, neither within the CDRs. Two additional mutations were in the D segment and six nongermline nucleotide additions were present at the DH-JH junction.
  • Finally, referring to FIG. 19 which presents the light chain of the antibody secreted by D5.1, the human kappa variable region B3 and human kappa joining region JK3 are shown. There are nine base-pair differences from the germline sequences, three falling within CDR1.

Claims (99)

1. A method to produce an immunoglobulin with fully human variable region or an analog thereof, specific for a desired antigen, which method comprises:
administering said antigen or an immunogenic portion thereof to a nonhuman animal under conditions to stimulate an immune response, whereby said animal produces B cells that secrete immunoglobulin specific for said antigen; wherein said nonhuman animal is characterized by being substantially incapable of producing endogenous heavy or light immunoglobulin chain variable regions, but capable of producing human immunoglobulin variable regions; and
recovering said immunoglobulin or analog.
2. (canceled)
3. The method of claim 1 wherein said recovering step comprises immortalizing B cells from said animal, screening the resulting immortalized cells for the secretion of said immunoglobulin, and
1) recovering immunoglobulin secreted by said immortalized B cells, or
2) recovering the genes encoding at least the variable region of said immunoglobulin from the immortalized B cells, and optionally modifying said genes;
expressing said genes or modified forms thereof to produce immunoglobulin or analog; and
recovering said immunoglobulin or analog.
4. The method of claim 1 wherein said recovering step comprises
recovering genes encoding at least the variable region of immunoglobulins from the primary B cells of the animal;
generating a library of said genes expressing the variable regions;
screening the library for a variable region with desired affinity for the antigen;
recovering the genes encoding said variable regions;
expressing said recovered genes to produce an immunoglobulin or analog containing said variable region and recovering said immunoglobulin or analog.
5. A recombinant DNA molecule comprising a nucleotide sequence encoding the immunoglobulin or analog produced by the method of claim 1.
6. (canceled)
7. A cell or cell line modified to contain the DNA molecule of claim 5.
8. A method to produce an immunoglobulin with fully human variable region or an analog thereof which method comprises culturing the cells of claim 7 under conditions whereby said encoding nucleotide sequence is expressed to produce said immunoglobulin or analog; and recovering said immunoglobulin or analog.
9. A DNA molecule comprising a nucleotide sequence corresponding to the gene or modified gene prepared by the method of claim 3.
10. (canceled)
11. A cell or cell line modified to contain the DNA molecule of claim 9.
12. A method to produce an immunoglobulin with fully human variable regions or an analog thereof which method comprises culturing the cells of claim 11 under conditions whereby said encoding nucleotide sequence is expressed to produce said immunoglobulin or analog; and recovering said immunoglobulin or analog.
13. A DNA molecule which comprises a nucleotide sequence encoding a variable region with desired affinity prepared according to the method of claim 4.
14. (canceled)
15. A cell or cell line modified to contain the DNA molecule of claim 13.
16. A method to produce an immunoglobulin with fully human variable region or an analog thereof which method comprises culturing the cells of claim 15 under conditions whereby said encoding nucleotide sequence is expressed to produce said immunoglobulin or analog; and recovering said immunoglobulin or analog.
17. An immortalized B cell which secretes an immunoglobulin to a desired antigen with a fully human variable region prepared according to claim 3.
18. A method to produce an immunoglobulin or analog which comprises culturing the cells of claim 17 and recovering said immunoglobulin or analog.
19. An immunoglobulin with fully human variable region or analog produced by the method of claim 1.
20. The immunoglobulin or analog of claim 19 wherein the desired antigen is selected from the group consisting of
the leukocyte markers, CD2, CD3, CD4, CD5, CD6, CD7, CD8, CD11a,b,c, CD13, CD14, CD18, CD19, CD20, CD22, CD23, CD27 and its ligand, CD28 and its ligands B7.1, B7.2, B7.3, CD29 and its ligand, CD30 and its ligand, CD40 and its ligand gp39, CD44, CD45 and isoforms, CDw52 (Campath antigen), CD56, CD58, CD69, CD72, CTLA-4, LFA-1 and TCR;
the histocompatibility antigens, MHC class I or II, the Lewis Y antigens, SLex, SLey, SLea, and SLeb;
the integrins, VLA-1, VLA-2, VLA-3, VLA-4, VLA-5, VLA-6, and LFA-1;
the adhesion molecules, Mac-1 and p150, 95;
the selectins, L-selectin, P-selectin, and E-selectin and their counterreceptors VCAM-1, ICAM-1, ICAM-2, and LFA-3;
the interleukins, IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-10, IL-11, IL-12, IL-13, IL-14, and IL-15;
interleukin receptors, IL-1R, IL-2R, IL-4R, IL-5R, IL-6R, IL-7R, IL-8R, IL-10R, IL-11R, IL-12R, IL-13R, IL-14R, and IL-15R;
chemokines, PF4, RANTES, MIP1α, MCP1, NAP-2, Groα, Groβ, and IL-8;
growth factors, TNFalpha, TGFbeta, TSH, VEGF/VPF, PTHrP, EGF family, FGF, PDGF family, endothelin, and gastrin releasing peptide (GRP);
growth factor receptors, TNFalphaR, RGFbetaR, TSHR, VEGFR/VPFR, FGFR, EGFR, PTHrPR, PDGFR family, EPO-R, GCSF-R and other hematopoietic receptors;
interferon receptors, IFNαR, IFNβR, and IFNγR;
Igs and their receptors, IgE, FceRI, and FCeRII;
tumor antigens, her2-neu, mucin, CEA and endosialin;
the allergens, house dust mite antigen, lol p1 (grass) antigens, and urushiol;
the viral proteins, CMV glycoproteins B, H, and gCIII, HIV-1 envelope glycoproteins, RSV envelope glycoproteins, HSV envelope glycoproteins, EBV envelope glycoproteins, VZV envelope glycoproteins, HPV envelope glycoproteins, Hepatitis family surface antigens;
the toxins, pseudomonas endotoxin and osteopontin/uropontin, snake venom, and bee venom;
the blood factors, complement C3b, complement C5a, complement C5b-9, Rh factor, fibrinogen, fibrin, and myelin associated growth inhibitor;
the enzymes, cholesterol ester transfer protein, membrane bound matrix metalloproteases, and Glutamic acid decarboxylase (GAD); and
the miscellaneous antigens ganglioside GD3, ganglioside GM2, LMP1, LMP2, eosinophil major basic protein, eosinophil cationic protein, pANCA, Amadori protein, Type IV collagen, glycated lipids, γ-interferon, A7, P-glycoprotein and Fas (AFO-1) and oxidized-LDL.
21. A DNA molecule comprising a nucleotide sequence that encodes the immunoglobulin or analog of claim 19.
22. (canceled)
23. A cell or cell line modified to contain the DNA molecule of claim 21.
24. A method to produce an immunoglobulin or analog specific for the antigen selected from the group consisting of leukocyte markers, CD2, CD3, CD4, CD5, CD6, CD7, CD8, CD11a,b,c, CD13, CD14, CD18, CD19, CD20, CD22, CD23, CD27 and its ligand, CD28 and its ligands B7.1, B7.2, B7.3, CD29 and its ligand, CD30 and its ligand, CD40 and its ligand gp39, CD44, CD45 and isoforms, CDw52 (Campath antigen), CD56, CD58, CD69, CD72, CTLA-4, LFA-1 and TCR
histocompatibility antigens, MHC class I or II, the Lewis Y antigens, SLex, SLey, SLea, and SLeb;
integrins, VLA-1, VLA-2, VLA-3, VLA-4, VLA-5, VLA-6, and LFA-1;
adhesion molecules, Mac-1 and p150, 95;
selectins, L-selectin, P-selectin, and E-selectin and their counterreceptors VCAM-1, ICAM-1, ICAM-2, and LFA-3;
interleukins, IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-10, IL-ll, IL-12, IL-13, IL-14, and IL-15;
interleukin receptors, IL-1R, IL-2R, IL-4R, IL-5R, IL-6R, IL-7R, IL-8R, IL-10R, IL-11R, IL-12R, IL-13R, IL-14R, and IL-15R;
chemokines, PF4, RANTES, MIP1α, MCP1, NAP-2, Groα, Groβ, and IL-8;
growth factors, TNFalpha, TGFbeta, TSH, VEGF/VPF, PTHrP, EGF family, FGF, PDGF family, endothelin, and gastrin releasing peptide (GRP);
growth factor receptors, TNFalphaR, RGFbetaR, TSHR, VEGFR/VPFR, FGFR, EGFR, PTHrPR, PDGFR family, EPO-R, GCSF-R and other hematopoietic receptors;
integrin receptors, IFNαR, IFNβR, and IFNγR;
Igs and their receptors, IgE, FceRI, and FCeR11;
tumor antigens, her2-neu, mucin, CEA and endosialin;
allergens, house dust mite antigen, lol p1 (grass) antigens, and urushiol;
viral proteins, CMV glycoproteins B, H, and gCIII, HIV-1 envelope glycoproteins, RSV envelope glycoproteins, HSV envelope glycoproteins, EBV envelope glycoproteins, VZV envelope glycoproteins, HPV envelope glycoproteins, Hepatitis family surface antigens;
toxins, pseudomonas endotoxin and osteopontin/uropontin, snake venom, and bee venom;
blood factors, complement C3b, complement C5a, complement C5b-9, Rh factor, fibrinogen, fibrin, and myelin associated growth inhibitor;
enzymes, cholesterol ester transfer protein, membrane bound matrix metalloproteases, and glutamic acid decarboxylase (GAD); and
miscellaneous antigens, ganglioside GD3, ganglioside GM2, LMP1, LMP2, eosinophil major basic protein, eosinophil cationic protein, pANCA, Amadori protein, Type IV dollagen, glycated lipids, γ-interferon, A7, P-glycoprotein and Fas (AFO-1) and oxidized-LDL
which method comprises culturing the cell or cell line of claim 23 under conditions wherein said nucleotide sequence is expressed to produce said immunoglobulin or analog; and
recovering the immunoglobulin or analog.
25. (canceled)
26. A antibody containing a fully human variable region or analog thereof which is specifically immunoreactive with an antigen selected from the group consisting of human IL-6, human IL-8, human TNFα, human CD4, human L-selectin, human gp39 and tetanus toxin C (TTC).
27. (canceled)
28. (canceled)
29. (canceled)
30. (canceled)
31. (canceled)
32. (canceled)
33. (canceled)
34. The analog of claim 26 which is a single chain Fv.
35. A recombinant DNA molecule encoding the antibody or analog of claim 26.
36. A recombinant DNA molecule which comprises an expression system for the production of the antibody or analog of claim 26 which expression system comprises a nucleotide sequence encoding said antibody or analog operably linked to control sequences capable of effecting its expression.
37. A recombinant host cell which is modified to contain the DNA molecule of claim 36.
38. A method to produce an antibody or analog immunospecific for an antigen selected from the group consisting of human IL-6, human IL-8, human TNFα, human CD4, human L-selectin, human gp39 and tetanus toxin C (TTC), which method comprises culturing the cells of claim 37 under conditions wherein said coding sequence is expressed; and recovering the antibody or analog produced.
39. Use of the antibody or analog of claim 27, 29, 31 or 32 for treating an autoimmune disease in a mammal.
40. (canceled)
41. (canceled)
42. (canceled)
43. (canceled)
44. (canceled)
45. (canceled)
46. An isolated human antibody or an antigen-binding fragment thereof that specifically binds a leukocyte marker selected from the group consisting of CD2, CD3, CD4, CD5, CD6, CD7, CD8, CD11a, b, c, CD13, CD14, CD18, CD19, CD20, CD22, CD23, CD27, CD28, CD29, CD30, CD40, CD44, CD45 and isoforms, Cdw52 (Campath antigen), CD56, CD58, CD69, CD72, CTLA-4, LFA-1, and TCR,
wherein said antibody or fragment modulates the activity of said leukocyte marker.
47. The antibody or fragment according to claim 46 wherein the leukocyte marker is CD4.
48. The antibody or fragment according to claim 46 herein the leukocyte marker is CD8.
49. The antibody or fragment according to claim 46 wherein the leukocyte marker is CD28.
50. The antibody or fragment according to claim 46 wherein the leukocyte marker is CD40.
51. The antibody or fragment according to claim 46 wherein the leukocyte marker is CD45.
52. The antibody or fragment according to claim 46 wherein the leukocyte marker is TCR.
53. The antibody according to any one of claims 46-52, wherein the antibody is monoclonal.
54. The fragment according to any one of claims 46-52, wherein the fragment comprises an scFv, Fab, Fab′, or F(ab′)2 fragment.
55. The antibody according to any one of claims 46-52, wherein the antibody is detectably labeled.
56. The antibody according to any one of claims 46-52, wherein the leukocyte marker is human.
57. The antibody according to any one of claims 46-52, wherein the antibody decreases activity of the leukocyte marker.
58. The antibody according to any one of claims 46-52, wherein the antibody comprises lambda light chain sequence.
59. The antibody according to any one of claims 46-52, wherein the antibody increases an activity of the leukocyte marker.
60. The antibody according to any one of claims 46-52, further comprising a pharmaceutical formulation.
61. A host cell that expresses the antibody according to any one of claims 46-52.
62. A nucleic acid that encodes the antibody according to any one of claims 46-52.
63. A host cell comprising the nucleic acid of claim 62.
64. A method of producing an isolated human antibody that specifically binds and modulates the activity of a leukocyte marker selected from the group consisting of CD2, CD3, CD4, CD5, CD6, CD7, CD8, CD11a, b, c, CD13, CD14, CD18, CD19, CD20, CD22, CD23, CD27, CD28, CD29, CD30, CD40, CD44, CD45 and isoforms, Cdw52 (Campath antigen), CD56, CD58, CD69, CD72, CTLA-4, LFA-1, and TCR comprising:
(a) administering the leukocyte marker or an immunogenic fragment thereof to a mouse capable of expressing human immunoglobulin;
(b) screening the administered mouse for expression of a human antibody that specifically binds to the leukocyte marker;
(c) selecting a mouse that produces a human antibody that specifically binds to the leukocyte marker;
(d) isolating an antibody from the mouse that produces a human antibody that specifically binds to the leukocyte marker; and
(e) determining whether the antibody modulates an activity of the leukocyte marker thereby producing a human antibody that specifically binds to the leukocyte marker and modulates an activity of the leukocyte marker.
65. A method of producing an isolated human antibody that specifically binds to and modulates the activity of a leukocyte marker selected from the group consisting of CD2, CD3, CD4, CD5, CD6, CD7, CD8, CD11a, b, c, CD13, CD14, CD18, CD19, CD20, CD22, CD23, CD27, CD28, CD29, CD30, CD40, CD44, CD45 and isoforms, Cdw52 (Campath antigen), CD56, CD58, CD69, CD72, CTLA-4, LFA-1, and TCR comprising:
(a) administering the leukocyte marker or an immunogenic fragment thereof to a mouse capable of expressing human immunoglobulin;
(b) isolating spleen cells from the mouse that produces a human antibody that specifically binds to the leukocyte marker;
(c) fusing the spleen cells with a myeloma cell to produce a hybridoma; and
(d) screening the hybridoma for expression of a human antibody that specifically binds to and modulates an activity of the leukocyte marker thereby producing a human monoclonal antibody that specifically binds to and modulates an activity of the leukocyte marker.
66. The method according to claim 64 or 65 wherein the leukocyte marker is CD4.
67. The method according to claim 64 or 65 wherein the leukocyte marker is CD8.
68. The method according to claim 64 or 65 wherein the leukocyte marker is CD28.
69. The method according to claim 64 or 65 wherein the leukocyte marker is CD40.
70. The method according to claim 64 or 65 wherein the leukocyte marker is CD45.
71. The method according to claim 64 or 65 wherein the leukocyte marker is TCR.
72. A monoclonal antibody isolated from a hybridoma produced by the method of claim 65.
73. A method for modulating an activity of a leukocyte marker selected from the group consisting of CD2, CD3, CD4, CD5, CD6, CD7, CD8, CD11a, b, c, CD13, CD14, CD18, CD19, CD20, CD22, CD23, CD27, CD28, CD29, CD30, CD40, CD44, CD45 and isoforms, Cdw52 (Campath antigen), CD56, CD58, CD69, CD72, CTLA-4, LFA-1, and TCR comprising contacting a cell that expresses the leukocyte marker with a modulating amount of the antibody of claim 46.
74. The method according to claim 73 wherein the leukocyte marker is CD4.
75. The method according to claim 73 wherein the leukocyte marker is CD8.
76. The method according to claim 73 wherein the leukocyte marker is CD28.
77. The method according to claim 73 wherein the leukocyte marker is CD40.
78. The method according to claim 73 wherein the leukocyte marker is CD45.
79. The method according to claim 73 wherein the leukocyte marker is TCR.
80. The method of claim 73, wherein the leukocyte marker is human.
81. The method of claim 73, wherein the activity is increased.
82. The method of claim 73, wherein the activity is decreased.
83. A method of increasing an activity of a leukocyte marker selected from the group consisting of CD2, CD3, CD4, CD5, CD6, CD7, CD8, CD11a, b, c, CD13, CD14, CD18, CD19, CD20, CD22, CD23, CD27, CD28, CD29, CD30, CD40, CD44, CD45 and isoforms, Cdw52 (Campath antigen), CD56, CD58, CD69, CD72, CTLA-4, LFA-1, and TCR in a subject comprising administering to the subject an amount of a human antibody that increases an activity of the leukocyte marker.
84. A method of decreasing an activity of a leukocyte marker selected from the group consisting of CD2, CD3, CD4, CD5, CD6, CD7, CD8, CD11a, b, c, CD13, CD14, CD18, CD19, CD20, CD22, CD23, CD27, CD28, CD29, CD30, CD40, CD44, CD45 and isoforms, Cdw52 (Campath antigen), CD56, CD58, CD69, CD72, CTLA-4, LFA-1, and TCR in a subject comprising administering to the subject an amount of a human antibody that decreases an activity of the leukocyte marker.
85. A method of detecting the presence of a leukocyte marker selected from the group consisting of CD2, CD3, CD4, CD5, CD6, CD7, CD8, CD11a, b, c, CD13, CD14, CD18, CD19, CD20, CD22, CD23, CD27, CD28, CD29, CD30, CD40, CD44, CD45 and isoforms, Cdw52 (Campath antigen), CD56, CD58, CD69, CD72, CTLA-4, LFA-1, and TCR in a sample or a cell, comprising contacting a sample having or suspected of having the leukocyte marker, or a cell expressing or suspected of expressing the leukocyte marker, with the antibody of claim 1, and detecting the presence of the leukocyte marker in the sample or cell.
86. The method according to claim 83, 84 or 85, wherein the leukocyte marker is CD4.
87. The method according to claim 83, 84 or 85, wherein the leukocyte marker is CD8.
88. The method according to claim 83, 84 or 85, wherein the leukocyte marker is CD28.
89. The method according to claim 83, 84 or 85, wherein the leukocyte marker is CD40.
90. The method according to claim 83, 84 or 85, wherein the leukocyte marker is CD45.
91. The method according to claim 83, 84 or 85, wherein the leukocyte marker is TCR.
92. The method of claim 85, wherein the cell is in a subject.
93. A method of detecting the presence of a disorder associated with increased or decreased expression of a a leukocyte marker selected from the group consisting of CD2, CD3, CD4, CD5, CD6, CD7, CD8, CD11a, b, c, CD13, CD14, CD18, CD19, CD20, CD22, CD23, CD27, CD28, CD29, CD30, CD40, CD44, CD45 and isoforms, Cdw52 (Campath antigen), CD56, CD58, CD69, CD72, CTLA-4, LFA-1, and TCR in a human, comprising contacting a sample having or suspected of having the leukocyte marker or a cell expressing or suspected of expressing the leukocyte marker, wherein the sample or cell is from or present in the human, with the human antibody of claim 46 and detecting the presence of increased or decreased expression of the leukocte marker in the sample or cell relative to a control thereby detecting the presence of a disorder associated with increased or decreased expression of the leukocyte marker in the human.
94. The method according to claim 93, wherein the leukocyte marker is CD4.
95. The method according to claim 93, wherein the leukocyte marker is CD8.
96. The method according to claim 93, wherein the leukocyte marker is CD28.
97. The method according to claim 93, wherein the leukocyte marker is CD40.
98. The method according to claim 93, wherein the leukocyte marker is CD45.
99. The method according to claim 93, wherein the leukocyte marker is TCR.
US10/656,623 1990-01-12 2003-09-04 Human antibodies derived from immunized xenomice Abandoned US20060040363A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US10/656,623 US20060040363A1 (en) 1990-01-12 2003-09-04 Human antibodies derived from immunized xenomice
US10/978,290 US20050241006A1 (en) 1995-04-27 2004-10-29 Human antibodies derived from immunized xenomice

Applications Claiming Priority (9)

Application Number Priority Date Filing Date Title
US46600890A 1990-01-12 1990-01-12
US61051590A 1990-11-08 1990-11-08
US91929792A 1992-07-24 1992-07-24
US08/031,801 US6673986B1 (en) 1990-01-12 1993-03-15 Generation of xenogeneic antibodies
US11284893A 1993-08-27 1993-08-27
US23414594A 1994-04-28 1994-04-28
US43093895A 1995-04-27 1995-04-27
US08/923,138 US6657103B1 (en) 1990-01-12 1997-09-04 Human antibodies derived from immunized xenomice
US10/656,623 US20060040363A1 (en) 1990-01-12 2003-09-04 Human antibodies derived from immunized xenomice

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US08/923,138 Continuation US6657103B1 (en) 1990-01-12 1997-09-04 Human antibodies derived from immunized xenomice

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US10/978,290 Continuation US20050241006A1 (en) 1995-04-27 2004-10-29 Human antibodies derived from immunized xenomice

Publications (1)

Publication Number Publication Date
US20060040363A1 true US20060040363A1 (en) 2006-02-23

Family

ID=29554595

Family Applications (2)

Application Number Title Priority Date Filing Date
US08/923,138 Expired - Fee Related US6657103B1 (en) 1990-01-12 1997-09-04 Human antibodies derived from immunized xenomice
US10/656,623 Abandoned US20060040363A1 (en) 1990-01-12 2003-09-04 Human antibodies derived from immunized xenomice

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US08/923,138 Expired - Fee Related US6657103B1 (en) 1990-01-12 1997-09-04 Human antibodies derived from immunized xenomice

Country Status (1)

Country Link
US (2) US6657103B1 (en)

Cited By (38)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070269868A1 (en) * 2006-05-19 2007-11-22 Carvalho Jensen Anne E Culture method for obtaining a clonal population of antigen-specific B cells
US20090291089A1 (en) * 2007-05-21 2009-11-26 Smith Jeffrey T L Antagonists of IL-6 to prevent or treat Thrombosis
US20090291085A1 (en) * 2007-02-16 2009-11-26 Merrimack Pharmaceuticals, Inc. Antibodies against erbb3 and uses thereof
WO2010054403A1 (en) 2008-11-10 2010-05-14 Alexion Pharmaceuticals, Inc. Methods and compositions for treating complement-associated disorders
WO2010056898A2 (en) * 2008-11-12 2010-05-20 University Of Maryland, Baltimore Rapid expression cloning of human monoclonal antibodies from memory b cells
US20100150829A1 (en) * 2008-11-25 2010-06-17 Leon Garcia-Martinez Antibodies to IL-6 and use thereof
US20100290993A1 (en) * 2007-05-21 2010-11-18 Leon Garcia-Martinez Antibodies to IL-6 and use thereof
US20110027291A1 (en) * 2008-08-15 2011-02-03 Merrimack Pharmaceuticals, Inc. Methods, systems and products for predicting response of tumor cells to a therapeutic agent and treating a patient according to the predicted response
WO2011085343A1 (en) 2010-01-11 2011-07-14 Alexion Pharmaceuticals, Inc Biomarkers of immunomodulatory effects in humans treated with anti-cd200 antibodies
US20110217303A1 (en) * 2007-05-21 2011-09-08 Smith Jeffrey T L Antagonists of il-6 to prevent or treat cachexia, weakness, fatigue, and/or fever
US20110236378A1 (en) * 2008-09-30 2011-09-29 Ablexis, Llc Non-human mammals for the production of chimeric antibodies
WO2011137395A1 (en) 2010-04-30 2011-11-03 Rother Russell P Anti-c5a antibodies and methods for using the antibodies
WO2012020096A1 (en) 2010-08-13 2012-02-16 Medimmune Limited Monomeric polypeptides comprising variant fc regions and methods of use
WO2012022734A2 (en) 2010-08-16 2012-02-23 Medimmune Limited Anti-icam-1 antibodies and methods of use
EP2463305A1 (en) 2006-01-12 2012-06-13 Alexion Pharmaceuticals, Inc. Antibodies to OX-2/CD200 and uses thereof
WO2012106634A1 (en) 2011-02-03 2012-08-09 Alexion Pharmaceuticals, Inc. Use of an anti-cd200 antibody for prolonging the survival of allografts
US20140017238A1 (en) 2001-02-16 2014-01-16 Regeneron Pharmaceuticals, Inc. Methods of Modifying Eukaryotic Cells
US8895001B2 (en) 2010-03-11 2014-11-25 Merrimack Pharmaceuticals, Inc. Use of ErbB3 inhibitors in the treatment of triple negative and basal-like breast cancers
US8992920B2 (en) 2008-11-25 2015-03-31 Alderbio Holdings Llc Anti-IL-6 antibodies for the treatment of arthritis
US8992908B2 (en) 2010-11-23 2015-03-31 Alderbio Holdings Llc Anti-IL-6 antibodies for the treatment of oral mucositis
WO2015050959A1 (en) 2013-10-01 2015-04-09 Yale University Anti-kit antibodies and methods of use thereof
US9085615B2 (en) 2008-11-25 2015-07-21 Alderbio Holdings Llc Antibodies to IL-6 to inhibit or treat inflammation
US9187560B2 (en) 2008-11-25 2015-11-17 Alderbio Holdings Llc Antagonists of IL-6 to treat cachexia, weakness, fatigue, and/or fever
WO2015175874A2 (en) 2014-05-16 2015-11-19 Medimmune, Llc Molecules with altered neonate fc receptor binding having enhanced therapeutic and diagnostic properties
US9212223B2 (en) 2008-11-25 2015-12-15 Alderbio Holdings Llc Antagonists of IL-6 to prevent or treat thrombosis
US9241990B2 (en) 2007-05-21 2016-01-26 Alderbio Holdings Llc Antagonists of IL-6 to raise albumin and/or lower CRIP
US9265825B2 (en) 2008-11-25 2016-02-23 Alderbio Holdings Llc Antagonists of IL-6 to raise albumin and/or lower CRP
US9468676B2 (en) 2009-11-24 2016-10-18 Alderbio Holdings Llc Antagonists of IL-6 to prevent or treat thrombosis
US9580491B2 (en) 2010-03-31 2017-02-28 Ablexis, Llc Genetic engineering of non-human animals for the production of chimeric antibodies
US9688761B2 (en) 2013-12-27 2017-06-27 Merrimack Pharmaceuticals, Inc. Biomarker profiles for predicting outcomes of cancer therapy with ERBB3 inhibitors and/or chemotherapies
US9701747B2 (en) 2007-05-21 2017-07-11 Alderbio Holdings Llc Method of improving patient survivability and quality of life by anti-IL-6 antibody administration
US9725509B2 (en) 2007-05-21 2017-08-08 Alderbio Holdings Llc Expression vectors containing isolated nucleic acids encoding anti-human IL-6 antibody
US9775921B2 (en) 2009-11-24 2017-10-03 Alderbio Holdings Llc Subcutaneously administrable composition containing anti-IL-6 antibody
US9815890B2 (en) 2010-06-22 2017-11-14 The Regents Of The University Of Colorado, A Body Corporate Antibodies to the C3d fragment of complement component 3
US9834603B2 (en) 2007-05-21 2017-12-05 Alderbio Holdings Llc Antibodies to IL-6 and use thereof
US10184006B2 (en) 2015-06-04 2019-01-22 Merrimack Pharmaceuticals, Inc. Biomarkers for predicting outcomes of cancer therapy with ErbB3 inhibitors
US10344299B2 (en) 2000-10-31 2019-07-09 Regeneron Pharmaceuticals, Inc. Compositions and methods for modifying cells
EP3524626A1 (en) 2007-03-22 2019-08-14 Biogen MA Inc. Binding proteins, including antibodies, antibody derivatives and antibody fragments, that specifically bind cd154 and uses thereof

Families Citing this family (88)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050241006A1 (en) * 1995-04-27 2005-10-27 Abgenix, Inc. Human antibodies derived from immunized xenomice
US20050287630A1 (en) * 1995-04-27 2005-12-29 Abgenix, Inc. Human antibodies derived from immunized xenomice
US20020173629A1 (en) * 1997-05-05 2002-11-21 Aya Jakobovits Human monoclonal antibodies to epidermal growth factor receptor
US6946129B1 (en) 1999-06-08 2005-09-20 Seattle Genetics, Inc. Recombinant anti-CD40 antibody and uses thereof
EP1307557A2 (en) * 2000-08-02 2003-05-07 The Johns Hopkins University Endothelial cell expression patterns
NZ569856A (en) 2001-01-05 2010-03-26 Pfizer Antibodies to insulin-like growth factor 1 receptor
US7357928B2 (en) * 2002-04-08 2008-04-15 University Of Louisville Research Foundation, Inc. Method for the diagnosis and prognosis of malignant diseases
AU2003225009A1 (en) * 2002-04-22 2003-11-03 Dyax Corporation Antibodies specific for mucin polypeptide
AU2003239505A1 (en) * 2002-05-20 2003-12-12 Amgen Fremont Inc. Treatment of renal carcinoma using antibodies against the egfr
DE60331310D1 (en) * 2002-06-26 2010-04-01 Univ Louisville Res Found PROCEDURE FOR APOPTOSE DETECTION
EP1596813A4 (en) * 2003-01-31 2008-02-20 Five Prime Therapeutics Inc Lung-expressed polypeptides
KR20070086866A (en) * 2003-02-13 2007-08-27 화이자 프로덕츠 인크. Uses of anti-insulin-like growth factor ⅰ receptor antibodies
BRPI0409383A (en) * 2003-04-14 2006-04-18 Univ Texas methods for ex vivo hybridoma free production of polyclonal and monoclonal antibodies and generation of immortalized cell populations
DK1656391T3 (en) * 2003-08-13 2011-01-03 Pfizer Prod Inc Modified human IGF-1R antibodies
US8114969B2 (en) * 2003-10-01 2012-02-14 Isis Innovation, Ltd. Immunotoxin derived from a recombinant human autoantibody and method of using thereof
EP1697417A2 (en) * 2003-11-07 2006-09-06 Curagen Corporation Antibodies against secretory leukocyte protease inhibitor
EP1753871B1 (en) 2004-05-28 2015-07-15 Agensys, Inc. Antibodies and related molecules that bind to psca proteins
EP2322217A3 (en) * 2004-07-16 2011-09-28 Pfizer Products Inc. Combination treatment for non-hematologic malignancies using an anti-IGF-1R antibody
TWI374935B (en) 2004-08-27 2012-10-21 Pfizer Ireland Pharmaceuticals Production of α-abeta
TWI384069B (en) 2004-08-27 2013-02-01 Pfizer Ireland Pharmaceuticals Production of polypeptides
US7300773B2 (en) * 2004-08-27 2007-11-27 Wyeth Research Ireland Limited Production of TNFR-Ig
JP5651285B2 (en) 2005-02-15 2015-01-07 デューク ユニバーシティ Anti-CD19 antibodies and use in oncology
JP4324636B2 (en) 2005-03-31 2009-09-02 アジェンシス,インコーポレイテッド Antibodies and related molecules that bind to 161P2F10B protein
EP2221316A1 (en) 2005-05-05 2010-08-25 Duke University Anti-CD19 antibody therapy for autoimmune disease
JP2009529915A (en) 2006-03-20 2009-08-27 ゾーマ テクノロジー リミテッド Human antibodies and methods specific for gastrin substances
AU2007240614B2 (en) 2006-04-20 2013-09-12 The Henry M. Jackson Foundation For The Advancement Of Military Medicine Incorporated Methods and compositions based on Shiga toxin type 1 protein
MX2009006709A (en) 2006-12-20 2009-07-02 Xoma Technology Ltd Methods for the treatment of il-1ã¿ related diseases.
NZ581396A (en) 2007-06-01 2012-07-27 Omt Inc Compositions and methods for inhibiting endogenous immunoglobulin genes and producing transgenic human idiotype antibodies
MX2010006823A (en) 2007-12-20 2010-09-30 Xoma Technology Ltd Methods for the treatment of gout.
DK2564695T3 (en) 2009-07-08 2015-05-26 Kymab Ltd Animal models and therapeutic molecules
US20120204278A1 (en) 2009-07-08 2012-08-09 Kymab Limited Animal models and therapeutic molecules
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
WO2011062997A2 (en) 2009-11-17 2011-05-26 Musc Foundation For Research Development Human monoclonal antibodies to human nucleolin
US9796788B2 (en) 2010-02-08 2017-10-24 Regeneron Pharmaceuticals, Inc. Mice expressing a limited immunoglobulin light chain repertoire
CA2789230C (en) 2010-02-08 2019-04-02 Agensys, Inc. Antibody drug conjugates (adc) that bind to 161p2f10b proteins
NZ619512A (en) 2010-02-08 2014-10-31 Regeneron Pharma Common light chain mouse
US20130045492A1 (en) 2010-02-08 2013-02-21 Regeneron Pharmaceuticals, Inc. Methods For Making Fully Human Bispecific Antibodies Using A Common Light Chain
CN105753979B (en) 2010-08-02 2021-05-07 瑞泽恩制药公司 Mice that make binding proteins comprising VL domains
CA2807607A1 (en) 2010-08-20 2012-02-23 Wyeth Llc Cell culture of growth factor-free adapted cells
ES2680624T3 (en) 2010-09-29 2018-09-10 Agensys, Inc. Antibody and drug (CAF) conjugates that bind to 191P4D12 proteins
RS53880B1 (en) 2011-02-25 2015-08-31 Regeneron Pharmaceuticals Inc. Adam6 mice
US9452219B2 (en) 2011-06-02 2016-09-27 University Of Louisville Research Foundation, Inc. Anti-nucleolin agent-conjugated nanoparticles
EA032790B1 (en) 2011-06-03 2019-07-31 Ксома Текнолоджи Лтд. Antibodies specific for tgf-beta
LT3572517T (en) 2011-08-05 2021-04-26 Regeneron Pharmaceuticals, Inc. Humanized universal light chain mice
EP3839049A3 (en) 2011-09-19 2021-10-20 Kymab Limited Antibodies, variable domains & chains tailored for human use
SI2627773T1 (en) 2011-10-17 2017-12-29 Regeneron Pharmaceuticals, Inc., Restricted immunoglobulin heavy chain mice
ES2784146T3 (en) 2011-10-21 2020-09-22 Pfizer Addition of iron to improve cell culture
EP3050900A1 (en) 2011-12-19 2016-08-03 Xoma (Us) Llc Methods for treating acne
JP6327750B2 (en) 2011-12-20 2018-05-23 リジェネロン・ファーマシューティカルズ・インコーポレイテッドRegeneron Pharmaceuticals, Inc. Humanized light chain mouse
GB2502127A (en) 2012-05-17 2013-11-20 Kymab Ltd Multivalent antibodies and in vivo methods for their production
US10251377B2 (en) 2012-03-28 2019-04-09 Kymab Limited Transgenic non-human vertebrate for the expression of class-switched, fully human, antibodies
WO2013187953A1 (en) * 2012-06-12 2013-12-19 Regeneron Pharmaceuticals, Inc. Humanized non-human animals with restricted immunoglobulin heavy chain loci
SG10201701424QA (en) 2012-08-23 2017-04-27 Agensys Inc Antibody drug conjugates (adc) that bind to 158p1d7 proteins
AR092745A1 (en) 2012-10-01 2015-04-29 Univ Pennsylvania COMPOSITIONS THAT INCLUDE AN ANTI-FAP UNION DOMAIN AND METHODS TO MAKE WHITE IN STROMAL CELLS FOR THE TREATMENT OF CANCER
RS57582B1 (en) 2013-02-20 2018-11-30 Regeneron Pharma Non-human animals with modified immunoglobulin heavy chain sequences
US9788534B2 (en) 2013-03-18 2017-10-17 Kymab Limited Animal models and therapeutic molecules
US11707056B2 (en) 2013-05-02 2023-07-25 Kymab Limited Animals, repertoires and methods
US9783593B2 (en) 2013-05-02 2017-10-10 Kymab Limited Antibodies, variable domains and chains tailored for human use
US9925273B2 (en) 2013-08-01 2018-03-27 Agensys, Inc. Antibody drug conjugates (ADC) that bind to CD37 proteins
WO2015049517A2 (en) 2013-10-01 2015-04-09 Kymab Limited Animal models and therapeutic molecules
WO2015140708A1 (en) 2014-03-19 2015-09-24 Pfizer Inc. Method of cell culture
WO2015143414A2 (en) 2014-03-21 2015-09-24 Regeneron Pharmaceuticals, Inc. Non-human animals that make single domain binding proteins
CA2941514A1 (en) 2014-03-21 2015-09-24 Regeneron Pharmaceuticals, Inc. Vl antigen binding proteins exhibiting distinct binding characteristics
EP3265491A1 (en) 2015-03-03 2018-01-10 Xoma (Us) Llc Treatment of post-prandial hyperinsulinemia and hypoglycemia after bariatric surgery
TWI719967B (en) 2015-03-09 2021-03-01 美商艾澤西公司 Antibody drug conjugates (adc) that bind to flt3 proteins
EP3271403A1 (en) 2015-03-19 2018-01-24 Regeneron Pharmaceuticals, Inc. Non-human animals that select for light chain variable regions that bind antigen
ES2820768T3 (en) 2015-04-03 2021-04-22 Xoma Technology Ltd Cancer treatment using TGF-beta and PD-1 inhibitors
WO2016179394A1 (en) 2015-05-05 2016-11-10 Malik Mohammad Tariq Anti-nucleolin agent-conjugated nanoparticles as radio-sensitizers and mri and/or x-ray contrast agents
CN108136006A (en) 2015-08-06 2018-06-08 佐马美国有限公司 The purposes of antibody fragment and its treatment hypoglycemia for insulin receptor
US10738334B2 (en) 2015-09-23 2020-08-11 Pfizer Inc. Cells having reduced BCAT1
KR102168981B1 (en) 2016-04-05 2020-10-22 화이자 인코포레이티드 Cell culture process
CN109803680A (en) 2016-08-01 2019-05-24 佐马美国有限公司 (PTH1R) antibody of parathyroid hormone receptor 1 and its purposes
CA3038679A1 (en) 2016-09-28 2018-04-05 Xoma (Us) Llc Antibodies that bind interleukin-2 and uses thereof
US20200056190A1 (en) 2017-03-16 2020-02-20 Pfizer Inc. Tyrosine prototrophy
EP3681989A1 (en) 2017-09-15 2020-07-22 Bristol-Myers Squibb Company Online biomass capacitance monitoring during large scale production of polypeptides of interest
EP3733699A4 (en) * 2017-12-29 2022-06-08 Zhuhai Trinomab Biotechnology Co., Ltd. Completely humanized monoclonal neutralizing antibody for tetanus toxin and application thereof
CN111902160A (en) 2018-03-16 2020-11-06 百时美施贵宝公司 Metabolic enzyme activity and disulfide bond reduction during protein production
WO2020059847A1 (en) 2018-09-21 2020-03-26 国立大学法人 東京医科歯科大学 Human monoclonal antibody binding specifically to human hmgb1, and pharmaceutical composition for treating or preventing alzheimer's disease containing said human monoclonal antibody
US20220073942A1 (en) 2018-12-06 2022-03-10 Pfizer Inc. Cells with reduced inhibitor production and methods of use thereof
AU2020258384A1 (en) 2019-04-15 2021-11-04 Qwixel Therapeutics Llc Fusion protein composition(s) comprising targeted masked type I interferons (IFNA and IFNB) and an antibody against tumor antigen, for use in the treatment of cancer
WO2021001458A1 (en) 2019-07-01 2021-01-07 Tonix Pharma Holdings Limited Anti-cd154 antibodies and uses thereof
US20230165967A1 (en) 2019-10-04 2023-06-01 TAE Life Sciences Antibody Compositions Comprising Fc Mutations and Site-Specific Conjugation Properties for use in Treating Cancer, Immunological Disorders, and Methods Thereof
WO2022124247A1 (en) 2020-12-09 2022-06-16 国立大学法人 東京医科歯科大学 Agent for preventing or treating frontotemporal lobar degeneration
JP2024503625A (en) 2021-01-06 2024-01-26 トニックス ファーマ リミテッド Method of inducing immune tolerance with a modified anti-CD154 antibody
CA3221878A1 (en) 2021-06-18 2022-12-22 David Stover Fusion protein composition(s) comprising masked type i interferons (ifna and ifnb) for use in the treatment of cancer and methods thereof
WO2023079058A1 (en) 2021-11-05 2023-05-11 Yokogawa Insilico Biotechnology Gmbh Cell culture with reduced production of lactate
WO2023148598A1 (en) 2022-02-02 2023-08-10 Pfizer Inc. Cysteine prototrophy

Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4816397A (en) * 1983-03-25 1989-03-28 Celltech, Limited Multichain polypeptides or proteins and processes for their production
US4950599A (en) * 1987-01-29 1990-08-21 Wolf Bertling Method for exchanging homologous DNA sequences in a cell using polyoma encapsulated DNA fragments
US4959313A (en) * 1987-06-22 1990-09-25 The Jackson Laboratory Cellular enhancer for expressing genes in undifferentiated stem cells
US5204244A (en) * 1987-10-27 1993-04-20 Oncogen Production of chimeric antibodies by homologous recombination
US5545807A (en) * 1988-10-12 1996-08-13 The Babraham Institute Production of antibodies from transgenic animals
US5545806A (en) * 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
US5559012A (en) * 1993-07-23 1996-09-24 Immunotech Therapeutic, IL-6 antibody kits, and process for their preparation
US5569824A (en) * 1991-01-04 1996-10-29 Baylor College Of Medicine Transgenic mice containing a disrupted p53 gene
US5569825A (en) * 1990-08-29 1996-10-29 Genpharm International Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US5591669A (en) * 1988-12-05 1997-01-07 Genpharm International, Inc. Transgenic mice depleted in a mature lymphocytic cell-type
US6150584A (en) * 1990-01-12 2000-11-21 Abgenix, Inc. Human antibodies derived from immunized xenomice

Family Cites Families (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2637613B1 (en) 1987-06-19 1991-09-27 Transgene Sa PROCESS FOR THE PREPARATION OF STABLE CELL LINES FOR THE PRODUCTION OF DETERMINED PROTEINS FROM TRANSGENIC ANIMALS; TUMOR CELL LINES AND PROTEINS OBTAINED
EP0322240B1 (en) 1987-12-23 1995-03-01 The Board Of Trustees Of The Leland Stanford Junior University Chimeric immunocompromised mammals and their use
WO1991000906A1 (en) 1989-07-12 1991-01-24 Genetics Institute, Inc. Chimeric and transgenic animals capable of producing human antibodies
EP1690935A3 (en) 1990-01-12 2008-07-30 Abgenix, Inc. Generation of xenogeneic antibodies
JP3008208B2 (en) 1990-06-01 2000-02-14 武田薬品工業株式会社 Novel hybridoma, its production method and production method of bioactive substance
GB9119338D0 (en) 1991-09-10 1991-10-23 Inst Of Animal Physiology And Control of gene expression
CA2135313A1 (en) 1992-06-18 1994-01-06 Theodore Choi Methods for producing transgenic non-human animals harboring a yeast artificial chromosome
AU675661B2 (en) 1992-07-24 1997-02-13 Abgenix, Inc. Generation of xenogeneic antibodies
US5827690A (en) 1993-12-20 1998-10-27 Genzyme Transgenics Corporatiion Transgenic production of antibodies in milk
ATE390933T1 (en) 1995-04-27 2008-04-15 Amgen Fremont Inc HUMAN ANTIBODIES AGAINST IL-8 DERIVED FROM IMMUNIZED XENOMICES

Patent Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4816397A (en) * 1983-03-25 1989-03-28 Celltech, Limited Multichain polypeptides or proteins and processes for their production
US4950599A (en) * 1987-01-29 1990-08-21 Wolf Bertling Method for exchanging homologous DNA sequences in a cell using polyoma encapsulated DNA fragments
US4959313A (en) * 1987-06-22 1990-09-25 The Jackson Laboratory Cellular enhancer for expressing genes in undifferentiated stem cells
US5204244A (en) * 1987-10-27 1993-04-20 Oncogen Production of chimeric antibodies by homologous recombination
US5545807A (en) * 1988-10-12 1996-08-13 The Babraham Institute Production of antibodies from transgenic animals
US5591669A (en) * 1988-12-05 1997-01-07 Genpharm International, Inc. Transgenic mice depleted in a mature lymphocytic cell-type
US6150584A (en) * 1990-01-12 2000-11-21 Abgenix, Inc. Human antibodies derived from immunized xenomice
US5545806A (en) * 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
US5569825A (en) * 1990-08-29 1996-10-29 Genpharm International Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US5569824A (en) * 1991-01-04 1996-10-29 Baylor College Of Medicine Transgenic mice containing a disrupted p53 gene
US5559012A (en) * 1993-07-23 1996-09-24 Immunotech Therapeutic, IL-6 antibody kits, and process for their preparation

Cited By (134)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9708635B2 (en) 2000-10-31 2017-07-18 Regeneron Pharmaceuticals, Inc. Methods of making a nucleic acid encoding a human variable region
US10227625B2 (en) 2000-10-31 2019-03-12 Regeneron Pharmaceuticals, Inc. Methods of modifying eukaryotic cells
US9376699B2 (en) 2000-10-31 2016-06-28 Regeneron Pharmaceuticals, Inc. Methods of producing hybrid antibodies
US10344299B2 (en) 2000-10-31 2019-07-09 Regeneron Pharmaceuticals, Inc. Compositions and methods for modifying cells
US10584364B2 (en) 2000-12-07 2020-03-10 Rgeneron Pharmaceuticals, Inc. Mice that produce hybrid antibodies
US9388446B2 (en) 2001-02-16 2016-07-12 Regeneron Pharmaceuticals, Inc. Methods of producing hybrid antibodies
US10526630B2 (en) 2001-02-16 2020-01-07 Regeneron Pharmaceuticals, Inc. Genetically modified mice that produce hybrid antibodies
US10378038B2 (en) 2001-02-16 2019-08-13 Regeneron Pharmaceuticals, Inc. Mice that produce hybrid antibodies
US20140017238A1 (en) 2001-02-16 2014-01-16 Regeneron Pharmaceuticals, Inc. Methods of Modifying Eukaryotic Cells
US10378040B2 (en) 2001-02-16 2019-08-13 Regeneron Pharmaceuticals, Inc. Mice that produce hybrid antibodies
US10640800B2 (en) 2001-02-16 2020-05-05 Regeneron Pharmaceuticals, Inc. Mice that produce hybrid antibodies
US10378037B2 (en) 2001-02-16 2019-08-13 Regeneron Pharmaceuticals, Inc. Methods of making a nucleic acid encoding a human variable region
US9353394B2 (en) 2001-02-16 2016-05-31 Regeneron Pharmaceuticals, Inc. Methods of producing hybrid antibodies
US10378039B2 (en) 2001-02-16 2019-08-13 Regeneron Pharmaceuticals, Inc. Mouse embryonic stem cells comprising a hybrid heavy chain immunoglobulin locus
US9528136B2 (en) 2001-02-16 2016-12-27 Regeneron Pharmaceuticals, Inc. Methods of modifying eukaryotic cells
US9371553B2 (en) 2001-02-16 2016-06-21 Regeneron Pharmaceuticals, Inc. Genetically modified mice that produce hybrid antibodies
EP2463305A1 (en) 2006-01-12 2012-06-13 Alexion Pharmaceuticals, Inc. Antibodies to OX-2/CD200 and uses thereof
US20070269868A1 (en) * 2006-05-19 2007-11-22 Carvalho Jensen Anne E Culture method for obtaining a clonal population of antigen-specific B cells
WO2008045140A1 (en) * 2006-05-19 2008-04-17 Alder Biopharmaceuticals, Inc. Culture method for obtaining a clonal population of antigen-specific b cells
US8961966B2 (en) 2007-02-16 2015-02-24 Merrimack Pharmaceuticals, Inc. Antibodies against ERBB3 and uses thereof
US20090291085A1 (en) * 2007-02-16 2009-11-26 Merrimack Pharmaceuticals, Inc. Antibodies against erbb3 and uses thereof
US9487588B2 (en) 2007-02-16 2016-11-08 Merrimack Pharmaceuticals, Inc. Antibodies against the ectodomain of ERBB3 and uses thereof
US20110123523A1 (en) * 2007-02-16 2011-05-26 Merrimack Pharmaceuticals, Inc. Antibodies against erbb3 and uses thereof
US7846440B2 (en) 2007-02-16 2010-12-07 Merrimack Pharmaceuticals, Inc. Antibodies against ErbB3 and uses thereof
US20100266584A1 (en) * 2007-02-16 2010-10-21 Merrimack Pharmaceuticals, Inc. Antibodies against the ectodomain of erbb3 and uses thereof
US8691225B2 (en) 2007-02-16 2014-04-08 Merrimack Pharmaceuticals, Inc. Antibodies against the ectodomain of ErbB3 and uses thereof
EP3524626A1 (en) 2007-03-22 2019-08-14 Biogen MA Inc. Binding proteins, including antibodies, antibody derivatives and antibody fragments, that specifically bind cd154 and uses thereof
US10800841B2 (en) 2007-05-21 2020-10-13 Vitaeris, Inc. Methods of treating autoimmunity using specific anti-IL-6 antibodies
US9758579B2 (en) 2007-05-21 2017-09-12 Alder Bioholdings, Llc Nucleic acids encoding anti-IL-6 antibodies of defined epitopic specificity
US10787507B2 (en) 2007-05-21 2020-09-29 Vitaeris Inc. Antagonists of IL-6 to prevent or treat thrombosis
US10040851B2 (en) 2007-05-21 2018-08-07 Alderbio Holdings Llc Antagonists to IL-6 to raise albumin and/or lower CRP
US20090291089A1 (en) * 2007-05-21 2009-11-26 Smith Jeffrey T L Antagonists of IL-6 to prevent or treat Thrombosis
US10759853B2 (en) 2007-05-21 2020-09-01 Alderbio Holdings Llc Antibodies to IL-6 and use thereof
US8999330B2 (en) 2007-05-21 2015-04-07 Alderbio Holdings Llc Antagonists of IL-6 to prevent or treat thrombosis
US10233239B2 (en) 2007-05-21 2019-03-19 Alderbio Holdings Llc Isolated host cells expressing anti-IL-6 antibodies
US9926370B2 (en) 2007-05-21 2018-03-27 Alderbio Holdings Llc Antagonists of IL-6 to prevent or treat thrombosis
US9884912B2 (en) 2007-05-21 2018-02-06 Alderbio Holdings Llc Antibodies to IL-6 and use thereof
US9834603B2 (en) 2007-05-21 2017-12-05 Alderbio Holdings Llc Antibodies to IL-6 and use thereof
US10344086B2 (en) 2007-05-21 2019-07-09 Alderbio Holdings Llc Antibodies to IL-6 and use thereof
US9771421B2 (en) 2007-05-21 2017-09-26 Alderbio Holdings Llc Treating anemia in chronic IL-6 associated diseases using anti-IL-6 antibodies
US9725509B2 (en) 2007-05-21 2017-08-08 Alderbio Holdings Llc Expression vectors containing isolated nucleic acids encoding anti-human IL-6 antibody
US20100290993A1 (en) * 2007-05-21 2010-11-18 Leon Garcia-Martinez Antibodies to IL-6 and use thereof
US8252286B2 (en) 2007-05-21 2012-08-28 Alderbio Holdings Llc Antagonists of IL-6 to prevent or treat thrombosis
US9241990B2 (en) 2007-05-21 2016-01-26 Alderbio Holdings Llc Antagonists of IL-6 to raise albumin and/or lower CRIP
US9701747B2 (en) 2007-05-21 2017-07-11 Alderbio Holdings Llc Method of improving patient survivability and quality of life by anti-IL-6 antibody administration
US9546213B2 (en) 2007-05-21 2017-01-17 Alderbio Holdings Llc Antagonists of IL-6 to prevent or treat cachexia, weakness, fatigue, and/or fever
US11827700B2 (en) 2007-05-21 2023-11-28 Vitaeris Inc. Treatment or prevention of diseases and disorders associated with cells that express IL-6 with Anti-IL-6 antibodies
US20110217303A1 (en) * 2007-05-21 2011-09-08 Smith Jeffrey T L Antagonists of il-6 to prevent or treat cachexia, weakness, fatigue, and/or fever
US10160804B2 (en) 2007-05-21 2018-12-25 Alderbio Holdings Llc Antagonists of IL-6 to prevent or treat cachexia, weakness, fatigue, and/or fever
US10913794B2 (en) 2007-05-21 2021-02-09 Vitaeris Inc. Antibodies to IL-6 and use thereof
US8623592B2 (en) 2008-08-15 2014-01-07 Merrimack Pharmaceuticals, Inc. Methods and systems for predicting response of cells to a therapeutic agent
US20110027291A1 (en) * 2008-08-15 2011-02-03 Merrimack Pharmaceuticals, Inc. Methods, systems and products for predicting response of tumor cells to a therapeutic agent and treating a patient according to the predicted response
US20110159513A1 (en) * 2008-08-15 2011-06-30 Merrimack Pharmaceuticals, Inc. Methods and systems for predicting response of cells to a therapeutic agent
US10561123B2 (en) 2008-09-30 2020-02-18 Ablexis, Llc Non-human mammals for the production of chimeric antibodies
US10575504B2 (en) 2008-09-30 2020-03-03 Ablexis, Llc Non-human mammals for the production of chimeric antibodies
US10492476B2 (en) 2008-09-30 2019-12-03 Ablexis, Llc Non-human mammals for the production of chimeric antibodies
US10638736B2 (en) 2008-09-30 2020-05-05 Ablexis, Llc Non-human mammals for the production of chimeric antibodies
US9346873B2 (en) 2008-09-30 2016-05-24 Ablexis, Llc Non-human mammals for the production of chimeric antibodies
US10555506B2 (en) 2008-09-30 2020-02-11 Ablexis, Llc Non-human mammals for the production of chimeric antibodies
US20110236378A1 (en) * 2008-09-30 2011-09-29 Ablexis, Llc Non-human mammals for the production of chimeric antibodies
EP3121197A1 (en) 2008-11-10 2017-01-25 Alexion Pharmaceuticals, Inc. Methods and compositions for treating complement-associated disorders
EP3974448A1 (en) 2008-11-10 2022-03-30 Alexion Pharmaceuticals, Inc. Methods and compositions for treating complementassociated disorders
EP3101031A1 (en) 2008-11-10 2016-12-07 Alexion Pharmaceuticals, Inc. Methods and compositions for treating complement-associated disorders
EP2894166A1 (en) 2008-11-10 2015-07-15 Alexion Pharmaceuticals, Inc. Methods and compositions for treating complement-associated disorders
EP2894165A1 (en) 2008-11-10 2015-07-15 Alexion Pharmaceuticals, Inc. Methods and compositions for treating complement-associated disorders
WO2010054403A1 (en) 2008-11-10 2010-05-14 Alexion Pharmaceuticals, Inc. Methods and compositions for treating complement-associated disorders
WO2010056898A2 (en) * 2008-11-12 2010-05-20 University Of Maryland, Baltimore Rapid expression cloning of human monoclonal antibodies from memory b cells
US8840890B2 (en) 2008-11-12 2014-09-23 University Of Maryland, Baltimore Rapid expression cloning of human monoclonal antibodies from memory B cells
WO2010056898A3 (en) * 2008-11-12 2010-10-07 University Of Maryland, Baltimore Rapid expression cloning of human monoclonal antibodies from memory b cells
US20110223615A1 (en) * 2008-11-12 2011-09-15 University Of Maryland, Baltimore Rapid Expression Cloning of Human Monoclonal Antibodies from Memory B Cells
US9879074B2 (en) 2008-11-25 2018-01-30 Alderbio Holdings Llc Antibodies to IL-6 and use thereof
US10858424B2 (en) 2008-11-25 2020-12-08 Alderbio Holdings Llc Anti-IL-6 antibodies for the treatment of arthritis
US9187560B2 (en) 2008-11-25 2015-11-17 Alderbio Holdings Llc Antagonists of IL-6 to treat cachexia, weakness, fatigue, and/or fever
US10787511B2 (en) 2008-11-25 2020-09-29 Vitaeris Inc. Antagonists of IL-6 to raise albumin and/or lower CRP
US9085615B2 (en) 2008-11-25 2015-07-21 Alderbio Holdings Llc Antibodies to IL-6 to inhibit or treat inflammation
US20100150829A1 (en) * 2008-11-25 2010-06-17 Leon Garcia-Martinez Antibodies to IL-6 and use thereof
US9212223B2 (en) 2008-11-25 2015-12-15 Alderbio Holdings Llc Antagonists of IL-6 to prevent or treat thrombosis
US9765138B2 (en) 2008-11-25 2017-09-19 Alderbio Holdings Llc Isolated anti-IL-6 antibodies
US9265825B2 (en) 2008-11-25 2016-02-23 Alderbio Holdings Llc Antagonists of IL-6 to raise albumin and/or lower CRP
US10640560B2 (en) 2008-11-25 2020-05-05 Alderbio Holdings Llc Antagonists of IL-6 to prevent or treat cachexia, weakness, fatigue, and /or fever
US10117955B2 (en) 2008-11-25 2018-11-06 Alderbio Holdings Llc Methods of aiding in the diagnosis of diseases using anti-IL-6 antibodies
US8992920B2 (en) 2008-11-25 2015-03-31 Alderbio Holdings Llc Anti-IL-6 antibodies for the treatment of arthritis
US9994635B2 (en) 2008-11-25 2018-06-12 Alderbio Holdings Llc Antagonists of IL-6 to raise albumin and/or lower CRP
US9452227B2 (en) 2008-11-25 2016-09-27 Alderbio Holdings Llc Methods of treating or diagnosing conditions associated with elevated IL-6 using anti-IL-6 antibodies or fragments
US10053506B2 (en) 2008-11-25 2018-08-21 Alderbio Holdings Llc Antagonists of IL-6 to prevent or treat cachexia, weakness, fatigue, and/or fever
US10391169B2 (en) 2009-07-28 2019-08-27 Alderbio Holdings Llc Method of treating allergic asthma with antibodies to IL-6
US10471143B2 (en) 2009-11-24 2019-11-12 Alderbio Holdings Llc Antagonists of IL-6 to raise albumin and/or lower CRP
US9717793B2 (en) 2009-11-24 2017-08-01 Alderbio Holdings Llc Method of improving patient survivability and quality of life by administering an anti-IL-6 antibody
US9468676B2 (en) 2009-11-24 2016-10-18 Alderbio Holdings Llc Antagonists of IL-6 to prevent or treat thrombosis
US9821057B2 (en) 2009-11-24 2017-11-21 Alderbio Holdings Llc Anti-IL-6 antibody for use in the treatment of cachexia
US9775921B2 (en) 2009-11-24 2017-10-03 Alderbio Holdings Llc Subcutaneously administrable composition containing anti-IL-6 antibody
US9724410B2 (en) 2009-11-24 2017-08-08 Alderbio Holdings Llc Anti-IL-6 antibodies or fragments thereof to treat or inhibit cachexia, associated with chemotherapy toxicity
WO2011085343A1 (en) 2010-01-11 2011-07-14 Alexion Pharmaceuticals, Inc Biomarkers of immunomodulatory effects in humans treated with anti-cd200 antibodies
US8895001B2 (en) 2010-03-11 2014-11-25 Merrimack Pharmaceuticals, Inc. Use of ErbB3 inhibitors in the treatment of triple negative and basal-like breast cancers
US9518130B2 (en) 2010-03-11 2016-12-13 Merrimack Pharmaceuticals, Inc. Use of ERBB3 inhibitors in the treatment of triple negative and basal-like breast cancers
US10604587B2 (en) 2010-03-31 2020-03-31 Ablexis, Llc Genetic engineering of non-human animals for the production of chimeric antibodies
US11220555B2 (en) 2010-03-31 2022-01-11 Ablexis, Llc Genetic engineering of non-human animals for the production of chimeric antibodies
US9580491B2 (en) 2010-03-31 2017-02-28 Ablexis, Llc Genetic engineering of non-human animals for the production of chimeric antibodies
US10836832B2 (en) 2010-03-31 2020-11-17 Ablexis, Llc Genetic engineering of non-human animals for the production of chimeric antibodies
US10829564B2 (en) 2010-03-31 2020-11-10 Ablexis, Llc Genetic engineering of non-human animals for the production of chimeric antibodies
US11104743B2 (en) 2010-03-31 2021-08-31 Ablexis, Llc Genetic engineering of non-human animals for the production of chimeric antibodies
US10494445B2 (en) 2010-03-31 2019-12-03 Ablexis, Llc Genetic engineering of non-human animals for the production of chimeric antibodies
US10618977B2 (en) 2010-03-31 2020-04-14 Ablexis, Llc Genetic engineering of non-human animals for the production of chimeric antibodies
US11242409B2 (en) 2010-03-31 2022-02-08 Ablexis, Llc Genetic engineering of non-human animals for the production of chimeric antibodies
US10526420B2 (en) 2010-03-31 2020-01-07 Ablexis, Llc Genetic engineering of non-human animals for the production of chimeric antibodies
US10662255B2 (en) 2010-03-31 2020-05-26 Ablexis, Llc Genetic engineering of non-human animals for the production of chimeric antibodies
US11352444B2 (en) 2010-03-31 2022-06-07 Ablexis, Llc Genetic engineering of non-human animals for the production of chimeric antibodies
US10626188B2 (en) 2010-03-31 2020-04-21 Ablexis, Llc Genetic engineering of non-human animals for the production of chimeric antibodies
US11104744B2 (en) 2010-03-31 2021-08-31 Ablexis, Llc Genetic engineering of non-human animals for the production of chimeric antibodies
US9221901B2 (en) 2010-04-30 2015-12-29 Alexion Pharmaceuticals, Inc. Methods of treating complement-associated disorders with anti-C5a antibodies
US11407821B2 (en) 2010-04-30 2022-08-09 Alexion Pharmaceuticals, Inc. Anti-C5A antibodies
US9309310B2 (en) 2010-04-30 2016-04-12 Alexion Pharmaceuticals, Inc. Nucleic acids encoding anti-C5a antibodies
US9011852B2 (en) 2010-04-30 2015-04-21 Alexion Pharmaceuticals, Inc. Anti-C5a antibodies
WO2011137395A1 (en) 2010-04-30 2011-11-03 Rother Russell P Anti-c5a antibodies and methods for using the antibodies
US9371378B1 (en) 2010-04-30 2016-06-21 Alexion Pharmaceuticals, Inc. Anti-C5a antibodies
US9434784B1 (en) 2010-04-30 2016-09-06 Alexion Pharmaceuticals, Inc. Nucleic acids encodng anti-C5A antibodies
EP2824111A2 (en) 2010-04-30 2015-01-14 Alexion Pharmaceuticals, Inc. Anti-C5A Antibodies and Methods for Using the Antibodies
US9469690B2 (en) 2010-04-30 2016-10-18 Alexion Pharmaceuticals, Inc. Methods of treating complement-associated disorders with anti-C5a antibodies
US9963503B2 (en) 2010-04-30 2018-05-08 Alexion Pharmaceuticals, Inc. Methods of producing anti-C5a antibodies
US10450370B2 (en) 2010-04-30 2019-10-22 Alexion Pharmaceuticals, Inc. Anti-C5a antibodies
USRE49339E1 (en) 2010-06-22 2022-12-20 The Regents Of The University Of Colorado, A Body Corporate Antibodies to the C3D fragment of complement component 3
US9815890B2 (en) 2010-06-22 2017-11-14 The Regents Of The University Of Colorado, A Body Corporate Antibodies to the C3d fragment of complement component 3
WO2012020096A1 (en) 2010-08-13 2012-02-16 Medimmune Limited Monomeric polypeptides comprising variant fc regions and methods of use
WO2012022734A2 (en) 2010-08-16 2012-02-23 Medimmune Limited Anti-icam-1 antibodies and methods of use
US9957321B2 (en) 2010-11-23 2018-05-01 Alderbio Holdings Llc Anti-IL-6 antibodies for the treatment of oral mucositis
US8992908B2 (en) 2010-11-23 2015-03-31 Alderbio Holdings Llc Anti-IL-6 antibodies for the treatment of oral mucositis
US9304134B2 (en) 2010-11-23 2016-04-05 Alderbio Holdings Llc Anti-IL-6 antibodies for the treatment of anemia
WO2012106634A1 (en) 2011-02-03 2012-08-09 Alexion Pharmaceuticals, Inc. Use of an anti-cd200 antibody for prolonging the survival of allografts
WO2015050959A1 (en) 2013-10-01 2015-04-09 Yale University Anti-kit antibodies and methods of use thereof
US9688761B2 (en) 2013-12-27 2017-06-27 Merrimack Pharmaceuticals, Inc. Biomarker profiles for predicting outcomes of cancer therapy with ERBB3 inhibitors and/or chemotherapies
US10273304B2 (en) 2013-12-27 2019-04-30 Merrimack Pharmaceuticals, Inc. Biomarker profiles for predicting outcomes of cancer therapy with ERBB3 inhibitors and/or chemotherapies
EP3888690A2 (en) 2014-05-16 2021-10-06 MedImmune, LLC Molecules with altered neonate fc receptor binding having enhanced therapeutic and diagnostic properties
WO2015175874A2 (en) 2014-05-16 2015-11-19 Medimmune, Llc Molecules with altered neonate fc receptor binding having enhanced therapeutic and diagnostic properties
US10184006B2 (en) 2015-06-04 2019-01-22 Merrimack Pharmaceuticals, Inc. Biomarkers for predicting outcomes of cancer therapy with ErbB3 inhibitors

Also Published As

Publication number Publication date
US6657103B1 (en) 2003-12-02

Similar Documents

Publication Publication Date Title
US6075181A (en) Human antibodies derived from immunized xenomice
US20060040363A1 (en) Human antibodies derived from immunized xenomice
CA2219361C (en) Human antibodies derived from immunized xenomice
AU2466895A (en) Human antibodies derived from immunized xenomice
US20050054055A1 (en) Human antibodies derived from immunized xenomice
US6150584A (en) Human antibodies derived from immunized xenomice
KR19990008197A (en) Human Antibodies from Immunized Genomous
US20050287630A1 (en) Human antibodies derived from immunized xenomice
US20050241006A1 (en) Human antibodies derived from immunized xenomice
AU2006200868B2 (en) Human Antibodies Derived From Immunized Xenomice
AU5336100A (en) Human antibodies derived from immunized xenomice
AU2008202860B9 (en) Human Antibodies Derived From Immunized Xenomice
AU2003227322B2 (en) Human Antibodies Derived From Immunized Xenomice

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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