US20080262203A1 - Novel compounds - Google Patents

Novel compounds Download PDF

Info

Publication number
US20080262203A1
US20080262203A1 US12/103,761 US10376108A US2008262203A1 US 20080262203 A1 US20080262203 A1 US 20080262203A1 US 10376108 A US10376108 A US 10376108A US 2008262203 A1 US2008262203 A1 US 2008262203A1
Authority
US
United States
Prior art keywords
seq
antibody
human
gro
penta
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US12/103,761
Inventor
Stephanie Jane Clegg
Eric Dobrzynski
Jonathan H. Ellis
Volker Germaschewski
Alexis Paul Godillot
Zdenka Ludmila Jonak
Alan P. Lewis
John R. White
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.)
SmithKline Beecham Corp
Original Assignee
SmithKline Beecham Corp
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by SmithKline Beecham Corp filed Critical SmithKline Beecham Corp
Priority to US12/103,761 priority Critical patent/US20080262203A1/en
Assigned to SMITHKLINE BEECHAM CORPORATION reassignment SMITHKLINE BEECHAM CORPORATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: DOBRZYNSKI, ERIC, GODILLOT, ALEXIS PAUL, JONAK, ZDENKA LUDMILA, WHITE, JOHN R.
Assigned to SMITHKLINE BEECHAM CORPORATION reassignment SMITHKLINE BEECHAM CORPORATION RE-RECORD TO ADD THE NAMES OF THE PREVIOUSLY OMITTED CONVEYING PARTIES, PREVIOUSLY RECORDED ON REEL 021512 FRAME 0003. Assignors: LEWIS, ALAN P., CLEGG, STEPHANIE JANE, ELLIS, JONATHAN H., GERMASHEWSKI, VOLKER, DOBRZYNSKI, ERIC, GODILLOT, ALEX PAUL, JONAK, ZDENKA LUDMILA, WHITE, JOHN R.
Publication of US20080262203A1 publication Critical patent/US20080262203A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • 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
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/06Antigout agents, e.g. antihyperuricemic or uricosuric agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • 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]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/31Immunoglobulins specific features characterized by aspects of specificity or valency multispecific
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/34Identification of a linear epitope shorter than 20 amino acid residues or of a conformational epitope defined by amino acid residues
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding

Definitions

  • the present invention relates to an antibody which has multiple specificities.
  • the antibody of the present invention binds to (cross react with) human IL-8, Gro-alpha, Gro-beta, Gro-gamma, and ENA-78.
  • the present invention also concerns with methods of treating diseases or disorders characterised by elevated or unbalanced level of one or more of human IL-8, Gro-alpha, Gro-beta, Gro-gamma, and ENA-78, particularly COPD, osteoarthritis, rheumatoid arthritis, erosive arthritis, asthma, atherosclerosis, inflammatory bowel disease, psoriasis, transplant rejection, gout, cancer, acute lung injury, acute lung disease, sepsis, ARDS, peripheral artery disease, systemic sclerosis, neonatal respiratory distress syndrome, exacerbation of asthma and COPD, cystic fibrosis, diffuse panbronchiolitis, reperfusion injury, and/or endometriosis with said antibody.
  • chemokines could be involved in the development of diseases such as COPD, osteoarthritis, rheumatoid arthritis, erosive arthritis, asthma, atherosclerosis, inflammatory bowel disease, psoriasis, transplant rejection, gout, cancer, acute lung injury, acute lung disease, sepsis, ARDS, peripheral artery disease, systemic sclerosis, neonatal respiratory distress syndrome, exacerbation of asthma and COPD, cystic fibrosis, diffuse panbronchiolitis, reperfusion injury, or endometriosis.
  • diseases such as COPD, osteoarthritis, rheumatoid arthritis, erosive arthritis, asthma, atherosclerosis, inflammatory bowel disease, psoriasis, transplant rejection, gout, cancer, acute lung injury, acute lung disease, sepsis, ARDS, peripheral artery disease, systemic sclerosis, neonatal respiratory distress syndrome, exacerbation of asthma and COPD, cystic fibrosis, diffuse
  • CXC chemokines are known to stimulate neutrophil chemotaxis by engaging and activating the CXCR1 and/or CXCR2 receptors. Thus the inhibition of these chemokines could prevent inflammatory cells from infiltrating the lung tissue and thus prevent tissue damage.
  • the present invention is directed to inhibiting the activation of CXCR1 and CXCR2 receptors by using an antibody having the ability to bind to human IL-8, Gro-alpha, Gro-beta, Gro-gamma, and ENA-78, i.e. a penta-specific antibody.
  • the present invention relates to an antibody (immunoglobulin) which has multiple specificities contained within one immunoglobulin.
  • the antibody of the present invention binds to (cross react with) human IL-8, Gro-alpha, Gro-beta, Gro-gamma, and ENA-78.
  • the present invention also concerns with methods of treating diseases or disorders characterised by elevated or unbalanced level of one or more of human IL-8, Gro-alpha, Gro-beta, Gro-gamma, and ENA-78, particularly COPD, osteoarthritis, rheumatoid arthritis, erosive arthritis, asthma, atherosclerosis, inflammatory bowel disease, psoriasis, transplant rejection, gout, cancer, acute lung injury, acute lung disease, sepsis, ARDS, peripheral artery disease, systemic sclerosis, neonatal respiratory distress syndrome, exacerbation of asthma and COPD, cystic fibrosis, diffuse panbronchiolitis, reperfusion injury, and/or endometriosis with said antibody.
  • diseases or disorders characterised by elevated or unbalanced level of one or more of human IL-8, Gro-alpha, Gro-beta, Gro-gamma, and ENA-78, particularly COPD, osteoarthritis, rheumatoid arthritis
  • the present invention relates to an isolated antibody which has multiple specificity to (or cross reacts with) human IL-8, Gro-alpha, Gro-beta, Gro-gamma, and ENA-78; thus we define the antibody of the present invention as penta-specific (or pan-ELR) antibody.
  • the definition of antibody includes an antigen binding portion (or fragment) of the antibody such that the antigen binding portion (or fragment) binds to human IL-8, Gro-alpha, Gro-beta, Gro-gamma, and ENA-78.
  • the penta-specific antibody of the invention is preferably murine monoclonal, chimeric, human or humanized.
  • the penta-specific antibody of the present invention need not bind solely to human IL-8, Gro-alpha, Gro-beta, Gro-gamma, and ENA-78 antigens, but it may also to bind to other related proteins (such as human GCP-2, or may even bind to non-human orthologues of IL-8, Gro-alpha, Gro-beta, Gro-gamma, ENA-78, and GCP-2); in other words, the penta-specific antibody of the present invention minimally binds to human IL-8, Gro-alpha, Gro-beta, Gro-gamma, and ENA-78.
  • a penta-specific antibody of the present invention binds to each of human IL-8, Gro-alpha, Gro-beta, Gro-gamma, and ENA-78 with equilibrium constant, KD, values of less than 10 ⁇ 7 M, more preferably less than 10 ⁇ 8 M, and even more preferably less than 10 ⁇ 9 M as determined by surface plasmon resonance.
  • KD equilibrium constant
  • surface plasmon resonance measurement is conducted as described below.
  • the present invention comprises a method of decreasing the neutrophil chemotaxis through inhibition of CXCR1 and CXCR2 receptor activation by neutralizing human IL-8, Gro-alpha, Gro-beta, Gro-gamma, GCP-2 and ENA-78 with a penta-specific antibody of the present invention.
  • the present invention relates to a method of decreasing the neutrophil chemotaxis in a patient in need thereof by administering a penta-specific antibody of the present invention.
  • a penta-specific antibody binds within epitope of KELRCQCIKTYSKP (SEQ ID NO: 54) in human IL-8.
  • the penta-specific antibody of the present invention is generated by a method comprising the steps of using RIMMs (Kilpatrick, K. E., et al. Hybridoma. 1997: 16, 381) type protocol using a mixture (cocktail) of human IL-8, Gro-alpha, Gro-beta, Gro-gamma, and ENA-78 together with a set of five multiple antigenic peptides (MAPs) each MAP unit having one separate sequence from polypeptides of ID NOs: 49-53.
  • RIMMs Korean, K. E., et al. Hybridoma. 1997: 16, 381
  • a mixture (cocktail) of human IL-8, Gro-alpha, Gro-beta, Gro-gamma, and ENA-78 together with a set of five multiple antigenic peptides (MAPs) each MAP unit having one separate sequence from polypeptides of ID NOs: 49-53.
  • MAPs serve two functions within the immunization protocol.
  • MAPs allow for a selective multiple presentation of a known target amino acid sequence to the host immune system.
  • the increase in mass, due to multiple copies of the sequence linked via a core, such as, but not limited to lysine increases the immunogenicity of the sequence over that of individual peptides (Francis, J. P., et al., Immunology, 1991: 73; 249, Schott, M. E., et al., Cell. Immuno. 1996: 174: 199-209, Tam, J. P. Proc. Natl. Acad. Sci. 1988: 85; 5409-5413).
  • the MAPs used to generate this invention are comprised of multiple copies of the conserved target sequences (e.g. SEQ ID NOs: 49-53) found with and around the ELRCXC and GPHCA regions of target chemokines. Exemplary MAP set is depicted in FIG. 1 .
  • a penta-specific antibody of the present invention is generated by a method comprising the steps of:
  • a penta-specific antibody of the present invention is generated by a method comprising the steps of:
  • the present invention concerns a penta-specific antibody made by the foregoing process.
  • a penta-specific antibody has heavy and light chain variable regions encoded by nucleotide sequences comprising sequences of SEQ ID NO: 1 and SEQ ID NO:3, respectively, or conservative sequence modifications thereof.
  • a penta-specific antibody has heavy and light chain variable regions encoded by nucleotide sequences comprising sequences of SEQ ID NO:5 and SEQ ID NO:7, respectively, or conservative sequence modifications thereof.
  • a penta-specific antibody has heavy chain variable region encoded by a nucleotide sequence comprising sequence of SEQ ID NO:9, or conservative sequence modifications thereof.
  • a penta-specific antibody has heavy and light chain variable regions comprising the amino acid sequences of SEQ ID NO:2 and SEQ ID NO:4, respectively, or conservative sequence modifications thereof.
  • a penta-specific antibody has heavy and light chain variable regions comprising polypeptides which are at least 90%, 95%, 98% or 99% identical to the amino acid sequences of SEQ ID NO:2 and SEQ ID NO:4, respectively.
  • a penta-specific antibody has heavy and light chain variable regions comprising the amino acid sequences of SEQ ID NO:6 and SEQ ID NO:8, respectively, or conservative sequence modifications thereof.
  • a penta-specific antibody has heavy and light chain variable regions comprising polypeptides which are at least 90%, 95%, 98% or 99% identical to the amino acid sequences of SEQ ID NO:6 and SEQ ID NO:8, respectively.
  • a penta-specific antibody has heavy and light variable regions comprising the amino acid sequences of SEQ ID NOs:10 and 12, respectively, or conservative sequence modifications thereof.
  • a penta-specific antibody has heavy and light chain variable regions comprising polypeptide sequences which are at least 90%, 95%, 98% or 99% identical to the amino acid sequences of SEQ ID NOs:10 and 12, respectively.
  • a penta-specific antibody comprises at least one variable region selected from (i) the amino acid SEQ ID NO: 2, 4, 6, 8, 10, or 12; or (ii) an amino acid sequence which is at least 90%, 95%, 98% or 99% identical to any one of the amino acid sequences of (i) above.
  • a penta-specific antibody comprises CDR sequences of SEQ ID NOs: 13, 14, 15, 16, 17, and 18; or one or more of the CDR sequences can be conservative sequence modifications of the sequences SEQ ID NOs: 13, 14, 15, 16, 17, and 18.
  • the present invention relates to hybridoma or transfectoma which produces a penta-specific antibody which comprises CDR sequences of SEQ ID NOs: 13, 14, 15, 16, 17, and 18.
  • the present invention relates to a recombinant eukaryotic or prokaryotic cell which produces a penta-specific antibody which comprises CDR sequences of SEQ ID NOs: 13, 14, 15, 16, 17, and 18.
  • a penta-specific antibody comprises at least one CDR sequence selected from (i) SEQ ID NO: 13, 14, 15, 16, 17, or 18; or (ii) a conservative sequence modification of the sequences listed in (i).
  • a penta-specific antibody comprises a polypeptide of SEQ ID NO:15.
  • a penta-specific antibody comprises at least four CDR sequences selected from the group consisting of SEQ ID NOs: 13, 14, 15, 16, 17, and 18; or one or more of the CDR sequences can be conservative sequence modifications of the sequences listed in SEQ ID NOs: 13, 14, 15, 16, 17, and 18.
  • a penta-specific antibody comprises heavy and light chain variable regions which comprise the CDR amino acid sequences of SEQ ID NOs: 13, 14, and 15, and SEQ ID NOs: 16, 17, and 18, respectively.
  • a penta-specific antibody comprises CDR sequences of SEQ ID NOs: 19, 20, 21, 22, 23, and 24; or one or more the CDR sequences can be conservative sequence modifications of the sequences listed in SEQ ID NOs: 19, 20, 21, 22, 23, and 24.
  • the present invention relates to an hybridoma or transfectoma which produces a penta-specific antibody which comprises CDR sequences of SEQ ID NOs: 19, 20, 21, 22, 23, and 24.
  • the present invention relates to a recombinant eukaryotic or a prokaryotic cell which produces a penta-specific antibody which comprises CDR sequences of SEQ ID NOs: 19, 20, 21, 22, 23, and 24.
  • a penta-specific antibody comprises at least one CDR sequence selected from (i) SEQ ID NO: 19, 20, 21, 22, 23, or 24; or (ii) a conservative sequence modification of the sequences listed in (i).
  • a penta-specific antibody comprises a polypeptide of SEQ ID NO:21.
  • a penta-specific antibody comprises at least four CDR sequences selected from the group consisting of: SEQ ID NOs: 19, 20, 21, 22, 23, and 24; or one or more of the CDR sequences can be conservative sequence modifications of the sequences listed in SEQ ID NOs: 19, 20, 21, 22, 23, and 24.
  • a penta-specific antibody comprises heavy and light chain variable regions which comprise the CDR amino acid sequences of SEQ ID NOs: 19, 20, and 21, and SEQ ID NOs: 22, 23, and 24, respectively.
  • a penta-specific antibody comprises CDR sequences of SEQ ID NOs: 25, 26, 27, 28, 29, and 30; or one or more the CDR sequences can be conservative sequence modifications of the sequences listed in SEQ ID NOs: 25, 26, 27, 28, 29, and 30.
  • the present invention relates to an hybridoma or transfectoma which produces a penta-specific antibody which comprises CDR sequences of SEQ ID NOs: 25, 26, 27, 28, 29, and 30.
  • the present invention relates to a recombinant eukaryotic or prokaryotic cell which produces a penta-specific antibody which comprises CDR sequences of SEQ ID NOs: 25, 26, 27, 28, 29 and 30.
  • a penta-specific antibody comprises at least one CDR sequence selected from (i) SEQ ID NO: 25, 26, 27, 28, 29, or 30; or (ii) a conservative sequence modification of the sequences listed in (i).
  • a penta-specific antibody comprises a polypeptide of SEQ ID NO:27.
  • a penta-specific antibody comprises at least four CDR sequences selected from the group consisting of: SEQ ID NOs: 25, 26, 27, 28, 29, and 30; or one or more of the CDR sequences can be conservative modifications of the sequences listed in SEQ ID NOs: 25, 26, 27, 28, 29, and 30.
  • a penta-specific antibody comprises heavy and light variable chain regions which comprise the CDR amino acid sequences of SEQ ID NOs: 25, 26, and 27, and SEQ ID NOs: 28, 29 and 30, respectively.
  • the present invention concerns a hybridoma which produces a monoclonal antibody having heavy or light chain variable region encoded by nucleotide sequences comprising nucleotide sequences of SEQ ID NO:1, 5, or 9, or SEQ ID NO:3 or 7, respectively.
  • the present invention concerns a hybridoma which produces a monoclonal antibody having heavy or light chain variable region comprising the amino acid sequences of SEQ ID NO:2, 6, or 10, or SEQ ID NO: 4, 8, or 12, respectively, and conservative sequence modifications thereof.
  • the present invention relates to a recombinant eukaryotic or a prokaryotic host cell which produces a penta-specific antibody having heavy or light variable region which comprise the amino acid sequences of SEQ ID NO:2, 6, or 10, or SEQ ID NO:4, 8, or 12, respectively, and conservative sequence modifications thereof.
  • the present invention relates to an expression vector comprising nucleotide sequences encoding a variable heavy or light chain of a penta-specific antibody comprising the CDR sequences of SEQ ID NOs: 13, 14, and 15; or SEQ ID NOs: 16, 17, and 18, respectively.
  • the present invention relates to an expression vector comprising a nucleotide sequence encoding a CDR sequence of a penta-specific antibody selected from SEQ ID NO: 13, 14, 15, 16, 17, or 18.
  • the present invention relates to an expression vector comprising nucleotide sequences encoding at least four CDR sequences of a penta-specific antibody selected from the group consisting of SEQ ID NOs: 13, 14, 15, 16, 17, and 18.
  • the present invention relates to an expression vector comprising polynucleotide sequences of SEQ ID NOs: 31, 32, or 33.
  • the present invention relates to an expression vector comprising polynucleotide sequences of SEQ ID NOs: 34, 35, and 36.
  • the present invention relates to an expression vector comprising at least four polynucleotide sequences selected from the group of SEQ ID NOs: 31, 32, 33, 34, 35, and 36.
  • the present invention relates to an expression vector comprising nucleotide sequences encoding a variable heavy or light chain of a penta-specific antibody comprising the CDR sequences of SEQ ID NOs: 19, 20, and 21; or 22, 23, and 24, respectively.
  • the present invention relates to an expression vector comprising a nucleotide sequence encoding a CDR sequence of a penta-specific antibody selected from SEQ ID NO: 19, 20, 21, 22, 23, or 24.
  • the present invention relates to an expression vector comprising nucleotide sequences encoding at least four CDR sequences of a penta-specific antibody selected from the group consisting of SEQ ID NOs: 19, 20, 21, 22, 23, and 24.
  • the present invention relates to an expression vector comprising polynucleotide sequences of SEQ ID NOs: 37, 38, and 39.
  • the present invention relates to an expression vector comprising polynucleotide sequences of SEQ ID NOs: 40, 41, and 42.
  • the present invention relates to an expression vector comprising at least four polynucleotide sequences selected from the group of SEQ ID NOs: 37, 38, 39, 40, 41, and 42.
  • the present invention relates to an expression vector comprising nucleotide sequences encoding a variable heavy chain of a penta-specific antibody comprising the CDR sequences of SEQ ID NOs: 25, 26, and 27.
  • the present invention relates to an expression vector comprising a nucleotide sequence encoding a CDR sequence of a penta-specific antibody selected from SEQ ID NO: 25, 26, or 27.
  • the present invention relates to an expression vector comprising polynucleotide sequences of SEQ ID NOs: 43, 44, and 45.
  • a penta-specific antibody has heavy and light chains encoded by nucleotide sequences comprising sequences of SEQ ID NO:11, and SEQ ID NO:47, 59, 61, or 63, respectively.
  • a penta-specific antibody has heavy and light chains encoded by nucleotide sequences comprising sequences which are at least 90%, 95%, 98% or 99% identical to sequences of SEQ ID NO:11, and SEQ ID NO:47, 59, 61, or 63, respectively.
  • a penta-specific antibody has heavy and light chains comprising the amino acid sequences of SEQ ID NO:46, and SEQ ID NO:48, 60, 62, or 64, respectively.
  • a penta-specific antibody has heavy and light chains comprising the amino acid sequences of SEQ ID NO:46 and SEQ ID NO:48, respectively.
  • a penta-specific antibody has heavy and light chains comprising the amino acid sequences of SEQ ID NO:46 and SEQ ID NO:60, respectively.
  • a penta-specific antibody has heavy and light chains comprising the amino acid sequences of SEQ ID NO:46 and SEQ ID NO: 62, respectively.
  • a penta-specific antibody has heavy and light chains comprising the amino acid sequences of SEQ ID NO:46 and SEQ ID NO:64, respectively.
  • a penta-specific antibody has heavy and light chains comprising polypeptides which are at least 90%, 95%, 98% or 99% identical to the amino acid sequences of SEQ ID NO:46, and SEQ ID NO:48, 60, 62, or 64, respectively.
  • the present invention relates to a recombinant eukaryotic or a prokaryotic host cell which produces a penta-specific antibody having heavy or light chain comprising the amino acid sequence of SEQ ID NO: 46, or SEQ ID NO:48, 60, 62, or 64, respectively.
  • the present invention relates to a recombinant eukaryotic or a prokaryotic host cell which produces a penta-specific antibody having heavy and light chains comprising the amino acid sequences of SEQ ID NO: 46, and SEQ ID NO:48, 60, 62, or 64, respectively.
  • the present invention relates to a recombinant eukaryotic or a prokaryotic host cell which produces a penta-specific antibody having heavy or light chain comprising the amino acid sequence which is at least 90%, 95%, 98% or 99% identical to the amino acid sequence of SEQ ID NO: 46, or SEQ ID NO:48, 60, 62, or 64, respectively.
  • the present invention relates to a recombinant eukaryotic or a prokaryotic host cell which produces a penta-specific antibody having heavy and light chains comprising the amino acid sequences which are at least 90%, 95%, 98% or 99% identical to the amino acid sequences of SEQ ID NO: 46, and SEQ ID NO:48, 60, 62, or 64, respectively.
  • the present invention relates to an expression vector comprising polynucleotide sequences comprising sequences of SEQ ID NO:11, and SEQ ID NO:47, 59, 61, or 63, respectively.
  • the present invention relates to an expression vector comprising polynucleotide sequences comprising sequences which are at least 90%, 95%, 98% or 99% identical to sequences of SEQ ID NO:11, and SEQ ID NO:47, 59, 61, or 63, respectively.
  • the present invention relates to an expression vector comprising a polynucleotide sequence comprising a sequence of SEQ ID NO:11, 47, 59, 61, or 63.
  • the present invention relates to an expression vector comprising a polynucleotide sequence comprising a sequence which is at least 90%, 95%, 98% or 99% identical to a sequence of SEQ ID NO:11, 47, 59, 61, or 63, respectively.
  • the present invention relates to an expression vector comprising polynucleotide sequences which encode a penta-specific antibody comprising heavy and light chains comprising the amino acid sequences of SEQ ID NO:46, and SEQ ID NO:48, 60, 62, or 64, respectively.
  • the present invention relates to an expression vector comprising polynucleotide sequences which encode a penta-specific antibody comprising heavy and light chains comprising the amino acid sequences which are at least 90%, 95%, 98% or 99% identical to the amino acid sequences of SEQ ID NO:46, and SEQ ID NO:48, 60, 62, or 64, respectively.
  • the present invention relates to an expression vector comprising a polynucleotide sequence which encode a polypeptide comprising the amino acid sequence of SEQ ID NO:46, 48, 60, 62, or 64.
  • the present invention relates to an expression vector comprising a polynucleotide sequence which encode a polypeptide comprising an amino acid sequence which is at least 90%, 95%, 98% or 99% identical to the amino acid sequences of SEQ ID NO:46, 48, 60, 62, or 64.
  • the present invention relates to a process for producing a penta-specific antibody (immunoglobulin) in a single host cell, comprising the steps of:
  • the present invention relates to a process for producing a penta-specific antibody (immunoglobulin) in a single host cell, comprising the steps of:
  • the present invention relates to a process for producing a penta-specific antibody (immunoglobulin) in a single host cell, comprising the steps of:
  • the present invention relates to an antibody that fully or partially blocks the binding of any one of the aforementioned penta-specific antibody to human IL-8, Gro-alpha, Gro-beta, Gro-gamma, ENA-78, and GCP-2 in an immunoassay, such as ELISA assay.
  • partial blocking occurs when the antibody blocks the binding of the penta-specific antibody by more than 10%, 20%, 40% or 50%.
  • the present invention relates to an antibody that competes with the binding of any of the aforementioned penta-specific antibody to human IL-8, Gro-alpha, Gro-beta, Gro-gamma, ENA-78, and GCP-2.
  • the present invention relates to an antibody that fully or partially blocks the binding of any one of the aforementioned penta-specific antibody to epitope of KELRCQCIKTYSKP (SEQ ID NO: 54) in human IL-8 in an immunoassay, such as ELISA assay.
  • partial blocking occurs when the antibody blocks the binding of the penta-specific antibody by more than 10%, 20%, 40% or 50%.
  • the present invention relates to an antibody that competes with the binding of any one of the aforementioned penta-specific antibody to epitope of KELRCQCIKTYSKP (SEQ ID NO: 54) in human IL-8.
  • the present invention relates to a composition
  • a composition comprising an aforementioned penta-specific antibody and a pharmaceutically acceptable carrier.
  • the present invention relates to a method of treating or preventing in a mammal COPD, osteoarthritis, rheumatoid arthritis, erosive arthritis, asthma, atherosclerosis, inflammatory bowel disease, psoriasis, transplant rejection, gout, cancer, acute lung injury, acute lung disease, sepsis, ARDS, peripheral artery disease, systemic sclerosis, neonatal respiratory distress syndrome, exacerbation of asthma and COPD, cystic fibrosis, diffuse panbronchiolitis, reperfusion injury, and/or endometriosis comprising administering an effective amount of an aforementioned penta-specific antibody to said mammal.
  • the present invention relates to an aforementioned penta-specific antibody for use in the treatment of diseases or disorders characterised by elevated or unbalanced level of one or more of human IL-8, Gro-alpha, Gro-beta, Gro-gamma, GCP-2 and ENA-78, particularly COPD, osteoarthritis, rheumatoid arthritis, erosive arthritis, asthma, atherosclerosis, inflammatory bowel disease, psoriasis, transplant rejection, gout, cancer, acute lung injury, acute lung disease, sepsis, ARDS, peripheral artery disease, systemic sclerosis, neonatal respiratory distress syndrome, exacerbation of asthma and COPD, cystic fibrosis, diffuse panbronchiolitis, reperfusion injury, or endometriosis.
  • diseases or disorders characterised by elevated or unbalanced level of one or more of human IL-8, Gro-alpha, Gro-beta, Gro-gamma, GCP-2 and ENA-78, particularly COPD,
  • the present invention relates to an aforementioned penta-specific antibody for use in preventing and/or treating COPD, osteoarthritis, rheumatoid arthritis, erosive arthritis, asthma, atherosclerosis, inflammatory bowel disease, psoriasis, transplant rejection, gout, cancer, acute lung injury, acute lung disease, sepsis, ARDS, peripheral artery disease, systemic sclerosis, neonatal respiratory distress syndrome, exacerbation of asthma and COPD, cystic fibrosis, diffuse panbronchiolitis, reperfusion injury, and/or endometriosis in a mammal.
  • the present invention relates to use of an aforementioned penta-specific antibody in the manufacture of a medicament for use in preventing and/or treating COPD, osteoarthritis, rheumatoid arthritis, erosive arthritis, asthma, atherosclerosis, inflammatory bowel disease, psoriasis, transplant rejection, gout, cancer, acute lung injury, acute lung disease, sepsis, ARDS, peripheral artery disease, systemic sclerosis, neonatal respiratory distress syndrome, exacerbation of asthma and COPD, cystic fibrosis, diffuse panbronchiolitis, reperfusion injury, and/or endometriosis in a mammal.
  • the present invention relates to use of an aforementioned penta-specific antibody in the manufacture of a medicament for preventing and/or treating COPD, osteoarthritis, rheumatoid arthritis, erosive arthritis, asthma, atherosclerosis, inflammatory bowel disease, psoriasis, transplant rejection, gout, cancer, acute lung injury, acute lung disease, sepsis, ARDS, peripheral artery disease, systemic sclerosis, neonatal respiratory distress syndrome, exacerbation of asthma and COPD, cystic fibrosis, diffuse panbronchiolitis, reperfusion injury, and/or endometriosis in a mammal.
  • above mammal is human.
  • FIG. 1 depicts an exemplary set of MAPs to generate a penta-specific antibody. For avoidance of doubt, five MAP peptide units are depicted. Each unit contains one identical amino acid sequence selected from linear peptides of SEQ ID NO: 49-53.
  • FIG. 3 shows hematological analysis of total circulating neutrophils (cell count).
  • IV injection of 1 (open squares, ⁇ ) and 10 mg/kg (closed circles, ⁇ ) of the Chimera Antibody (HcLc) prevented inhaled LPS stimulation of circulating neutrophils.
  • Both 1 and 10 mg/kg treatments are significantly reduced compared to the vehicle NHP ( ⁇ ) and 10 mg/kg treatment was also significantly reduced compared to the 1 mg/kg dose (*).
  • FIG. 4 shows inhibition human IL-8 stimulated neutrophil activation (increased CD11b surface expression).
  • Humanized construct H0L10 and H0M0 are more effective at inhibiting hIL-8 stimulated CD11b surface expression.
  • FIG. 5A shows mouse 656.35 mAb binding to target human chemokines.
  • FIG. 5B shows the Chimera Antibody (HcLc) mAb binding to human target chemokines.
  • FIG. 5C shows humanized mAbs binding to human target chemokines.
  • an “isolated penta-specific antibody” or simply “penta-specific antibody”, as used herein, is intended to refer to an antibody that binds to and therefore cross reacts with human IL-8, Gro-alpha, Gro-beta, Gro-gamma, and ENA-78, and is substantially free of other antibodies having different antigenic specificities, and furthermore, is a single composition of matter.
  • the penta-specific antibody of the present invention need not bind solely to human IL-8, Gro-alpha, Gro-beta, Gro-gamma, and ENA-78 antigens, but it may also happen to bind to other related proteins, such as human GCP-2, or may even bind to non-human orthologues of IL-8, Gro-alpha, Gro-beta, Gro-gamma, ENA-78, and GCP-2; in other words the penta-specific antibody of the present invention minimally binds to human IL-8, Gro-alpha, Gro-beta, Gro-gamma, and ENA-78.
  • an isolated penta-specific antibody is substantially free of other cellular material and/or chemicals.
  • a penta-specific antibody also includes antigen binding fragments and/or derivatives having the binding characteristics of their “parent” penta-specific antibody.
  • the penta-specific antibody of the present invention may comprise two identical heavy chains and two identical light chains, forming the typical, bilaterally symmetric immunoglobulin molecule comprised of two heterodimers which are each comprised of a heavy chain and a light chain. Accordingly, the penta-specific antibody of the present invention may comprise two copies of the same antigen binding domain formed by the association of one heavy chain with one light chain.
  • the penta-specific antibody of the present invention though having only one kind of binding domain is still able to bind to and therefore cross react with human IL-8, Gro-alpha, Gro-beta, Gro-gamma, and ENA-78.
  • antibody is also referred to as “immunoglobulin”.
  • an antibody that cross reacts with means the antibody binds not only to one antigen but binds to other antigens as well.
  • Neutralizing is intended to refer to a partial or full inhibition of at least one biological activities of human IL-8, Gro-alpha, Gro-beta, Gro-gamma, GCP-2, or ENA-78.
  • one of the biological activities of human IL-8, Gro-alpha, Gro-beta, Gro-gamma, GCP-2, or ENA-78 is its ability to induce neutrophil chemotaxis.
  • surface plasmon resonance refers to an optical phenomenon that allows for the analysis of real-time biospecific interactions by detection of alterations in protein concentrations within a biosensor matrix, for example using the BIAcore system (Pharmacia Biosensor AB, Uppsala, Sweden and Piscataway, N.J.).
  • BIAcore Phharmacia Biosensor AB, Uppsala, Sweden and Piscataway, N.J.
  • epitopes means a protein determinant capable of specific binding to an antibody. Epitopes usually consist of chemically active surface groupings of molecules such as amino acids or sugar side chains and usually have specific three dimensional structural characteristics, as well as specific charge characteristics.
  • a “monoclonal antibody” or mAb (as opposed to polyclonal antibody) as used herein is intended to refer to a preparation of antibody molecules of single molecular composition.
  • a murine derived monoclonal antibody can be prepared by hybridoma technology, such as the standard Kohler and Milstein hybridoma methodology.
  • Antibody-producing cells can be obtained from the subject and used to prepare monoclonal antibodies by standard techniques, such as the hybridoma technique originally described by Kohler and Milstein (1975, Nature 256:495-497) (see also, Brown et al. (1981) J. Immunol 127:539-46; Brown et al. (1980) J Biol Chem 255:4980-83; Yeh et al. (1976) PNAS 76:2927-31; and Yeh et al. (1982) Int. J. Cancer 29:269-75).
  • the technology for producing monoclonal antibody hybridomas is well known (see generally R. H.
  • transfectoma includes recombinant eukaryotic host cell expressing the antibody, such as CHO cells, NS/0 cells, HEK293 cells, plant cells, or fungi, including yeast cells.
  • specific binding refers to antibody binding to a predetermined antigen.
  • the antibody binds with an equilibrium constant, KD, corresponding to about 1 ⁇ 10 ⁇ 7 M or less, and binds to the predetermined antigen with an affinity corresponding to a KD that is at least two orders of magnitude lower than its affinity for binding to a non-specific antigen (e.g., BSA, casein) other than the predetermined antigen or a closely-related antigen.
  • KD equilibrium constant
  • the term “kd” (sec-1), as used herein, is intended to refer to the dissociation rate constant of a particular antibody-antigen interaction.
  • ka (M ⁇ sec-1), as used herein, is intended to refer to the association rate constant of a particular antibody-antigen interaction.
  • KD (M), as used herein, is intended to refer to the equilibrium constant of a particular antibody-antigen interaction and is obtained by dividing the kd by the ka.
  • Constant sequence modifications for nucleotide and amino acid sequence modifications means changes which do not significantly affect or alter the binding characteristics of the antibody encoded by the nucleotide sequence or containing the amino acid sequence. Such conservative sequence modifications include nucleotide and amino acid substitutions, additions and deletions. Modifications can be introduced into the sequences by standard techniques known in the art, such as site-directed mutagenesis and PCR-mediated mutagenesis. Conservative amino acid substitutions include ones in which the amino acid residue is replaced with an amino acid residue having a similar side chain. Families of amino acid residues having similar side chains have been defined in the art.
  • amino acids with basic side chains e.g., lysine, arginine, histidine
  • acidic side chains e.g., aspartic acid, glutamic acid
  • uncharged polar side chains e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine, tryptophan
  • nonpolar side chains e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine
  • beta-branched side chains e.g., threonine, valine, isoleucine
  • aromatic side chains e.g., tyrosine, phenylalanine, tryptophan, histidine
  • a predicted nonessential amino acid residue in an antibody for which sequence is specifically disclosed is preferably replaced with another amino acid residue from the same side chain family.
  • the penta-specific antibody of the present invention includes all the conservative sequence modifications of the specifically disclosed amino acid sequences.
  • the present invention also encompasses “derivatives” of the amino acid sequences as specifically disclosed, wherein one or more of the amino acid residues have been derivatized, e.g., by acylation or glycosylation, without significantly affecting or altering the binding characteristics of the antibody containing the amino acid sequences.
  • nucleic acids For nucleic acids, the term “substantial identity” indicates that two nucleic acids, or designated sequences thereof, when optimally aligned and compared, are identical, with appropriate nucleotide insertions or deletions, in at least about 80% of the nucleotides, usually at least about 90% to 95%, and more preferably at least about 98% to 99.5% of the nucleotides. Alternatively, substantial identity when the segments will hybridize under selective hybridization conditions, to the complement of the strand.
  • identity indicates the degree of identity between two nucleic acid or amino acid sequences when optimally aligned and compared with appropriate insertions or deletions. Alternatively, substantial identity exists when the DNA segments will hybridize under selective hybridization conditions, to the complement of the strand.
  • the comparison of sequences and determination of percent identity between two sequences can be accomplished using a mathematical algorithm, as described in the non-limiting examples below.
  • the percent identity between two nucleotide or polypeptide sequences can be determined using the GAP program in the GCG software package (available at http://www.gcg.com), using a NWSgapdna.CMP matrix and a gap weight of 40, 50, 60, 70, or 80 and a length weight of 1, 2, 3, 4, 5, or 6.
  • the percent identity between two nucleotide or amino acid sequences can also be determined using the algorithm of E. Meyers and W. Miller (Comput. Appl. Biosci., 4:11-17 (1988)) which has been incorporated into the ALIGN program (version 2.0), using a PAM120 weight residue table, a gap length penalty of 12 and a gap penalty of 4.
  • the percent identity between two amino acid sequences can be determined using the Needleman and Wunsch (J. Mol. Biol. 48:444-453 (1970)) algorithm which has been incorporated into the GAP program in the GCG software package (available at http://www.gcg.com), using either a Blossum 62 matrix or a PAM250 matrix, and a gap weight of 16, 14, 12, 10, 8, 6, or 4 and a length weight of 1, 2, 3, 4, 5, or 6.
  • a nucleic acid is “operably linked” when it is placed into a functional relationship with another nucleic acid sequence.
  • a promoter or enhancer is operably linked to a coding sequence if it affects the transcription of the sequence.
  • operably linked means that the DNA sequences being linked are contiguous and, where necessary to join two protein coding regions, contiguous and in reading frame.
  • operably linked indicates that the sequences are capable of effecting switch recombination.
  • vector is intended to refer to a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked.
  • plasmid refers to a circular double stranded DNA loop into which additional DNA segments may be ligated.
  • viral vector Another type of vector is a viral vector, wherein additional DNA segments may be ligated into the viral genome.
  • Certain vectors are capable of autonomous replication in a host cell into which they are introduced (e.g., bacterial vectors having a bacterial origin of replication and episomal mammalian vectors).
  • vectors e.g., non-episomal mammalian vectors
  • vectors can be integrated into the genome of a host cell upon introduction into the host cell, and thereby are replicated along with the host genome.
  • certain vectors are capable of directing the expression of genes to which they are operatively linked.
  • Such vectors are referred to herein as “recombinant expression vectors” (or simply, “expression vectors”).
  • expression vectors of utility in recombinant DNA techniques are often in the form of plasmids.
  • plasmid and vector may be used interchangeably as the plasmid is the most commonly used form of vector.
  • the invention is intended to include such other forms of expression vectors, such as viral vectors (e.g., replication defective retroviruses, adenoviruses and adeno-associated viruses), which serve equivalent functions.
  • recombinant host cell (or simply “host cell”), as used herein, is intended to refer to a cell into which a recombinant expression vector has been introduced. It should be understood that such terms are intended to refer not only to the particular subject cell but to the progeny of such a cell. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term “host cell” as used herein.
  • Recombinant host cells include, for example, transfectomas, such as CHO cells, NS/0 cells, and lymphocytic cells.
  • the term “subject” includes any human or non-human animal.
  • non-human animal includes all vertebrates, e.g., mammals and non-mammals, such as non-human primates, sheep, dog, cow, chickens, amphibians, reptiles, etc.
  • Intact antibodies are usually heteromultimeric glycoproteins comprising at least two heavy and two light chains. Aside from IgM, intact antibodies are heterotetrameric glycoproteins of approximately 150 Kda, composed of two identical light (L) chains and two identical heavy (H) chains. Typically, each light chain is linked to a heavy chain by one covalent disulfide bond while the number of disulfide linkages between the heavy chains of different immunoglobulin isotypes varies. Each heavy and light chain also has intrachain disulfide bridges. Each heavy chain has at one end a variable domain (V H ) followed by a number of constant regions.
  • V H variable domain
  • Each light chain has a variable domain (V L ) and a constant region at its other end; the constant region of the light chain is aligned with the first constant region of the heavy chain and the light chain variable domain is aligned with the variable domain of the heavy chain.
  • V L variable domain
  • the light chains of antibodies from most vertebrate species can be assigned to one of two types called Kappa and Lambda based on the amino acid sequence of the constant region.
  • human antibodies can be assigned to five different classes, IgA, IgD, IgE, IgG and IgM.
  • IgG and IgA can be further subdivided into subclasses, IgG1, IgG2, IgG3 and IgG4; and IgA1 and IgA2.
  • Species variants exist with mouse and rat having at least IgG2a, IgG2b.
  • the variable domain of the antibody confers binding specificity upon the antibody with certain regions displaying particular variability called complementarity determining regions (CDRs).
  • CDRs complementarity determining regions
  • the more conserved portions of the variable region are called framework regions (FR).
  • the variable domains of intact heavy and light chains each comprise four FR connected by three CDRs.
  • the CDRs in each chain are held together in close proximity by the FR regions and with the CDRs from the other chain contribute to the formation of the antigen binding site of antibodies.
  • the constant regions are not directly involved in the binding of the antibody to the antigen but exhibit various effector functions such as participation in antibody dependent cell-mediated cytotoxicity (ADCC), phagocytosis via binding to Fc ⁇ receptor, half-life/clearance rate via neonatal Fc receptor (FcRn) and complement dependent cytotoxicity via the C1q component of the complement cascade.
  • ADCC antibody dependent cell-mediated cytotoxicity
  • FcRn neonatal Fc receptor
  • complement dependent cytotoxicity via the C1q component of the complement cascade.
  • the human IgG2 constant region lacks the ability to activate complement by the classical pathway or to mediate antibody-dependent cellular cytotoxicity.
  • the IgG4 constant region lacks the ability to activate complement by the classical pathway and mediates antibody-dependent cellular cytotoxicity only weakly. Antibodies essentially lacking these effector functions may be termed ‘non-lytic’ antibodies.
  • Human antibodies may be produced by a number of methods known to those of skill in the art. Human antibodies can be made by the hybridoma method using human myeloma or mouse-human heteromyeloma cells lines see Kozbor J. Immunol 133, 3001, (1984) and Brodeur, Monoclonal Antibody Production Techniques and Applications, pp 51-63 (Marcel Dekker Inc, 1987). Alternative methods include the use of phage libraries or transgenic mice both of which utilize human V region repertories (see Winter G, (1994), Annu. Rev. Immunol 12, 433-455, Green L L (1999), J. Immunol. methods 231, 11-23).
  • mice Several strains of transgenic mice are now available wherein their mouse immunoglobulin loci has been replaced with human immunoglobulin gene segments (see Tomizuka K, (2000) PNAS 97, 722-727; Fishwild D. M (1996) Nature Biotechnol. 14, 845-851, Mendez M J, 1997, Nature Genetics, 15, 146-156). Upon antigen challenge such mice are capable of producing a repertoire of human antibodies from which antibodies of interest can be selected.
  • TrimeraTM system (see Eren R et al, (1998) Immunology 93:154-161) where human lymphocytes are transplanted into irradiated mice
  • SAM Selected Lymphocyte Antibody System
  • SLAM Selected Lymphocyte Antibody System
  • human (or other species) lymphocytes are effectively put through a massive pooled in vitro antibody generation procedure followed by deconvulated, limiting dilution and selection procedure and the Xenomouse IITM (Abgenix Inc).
  • An alternative approach is available from Morphotek Inc using the MorphodomaTM technology.
  • Phage display technology can be used to produce human antibodies (and fragments thereof), see McCafferty; Nature, 348, 552-553 (1990) and Griffiths A D et al (1994) EMBO 13:3245-3260.
  • antibody V domain genes are cloned in frame into either a major or minor coat of protein gene of a filamentous bacteriophage such as M13 or fd and displayed (usually with the aid of a helper phage) as functional antibody fragments on the surface of the phage particle. Selections based on the functional properties of the antibody result in selection of the gene encoding the antibody exhibiting those properties.
  • the phage display technique can be used to select antigen specific antibodies from libraries made from human B cells taken from individuals afflicted with a disease or disorder described above or alternatively from unimmunized human donors (see Marks; J. Mol. Bio. 222, 581-597, 1991). Where an intact human antibody is desired comprising a Fc domain it is necessary to reclone the phage displayed derived fragment into a mammalian expression vectors comprising the desired constant regions and establishing stable expressing cell lines.
  • affinity maturation may be used to improve binding affinity wherein the affinity of the primary human antibody is improved by sequentially replacing the H and L chain V regions with naturally occurring variants and selecting on the basis of improved binding affinities.
  • Variants of this technique such as “epitope imprinting” are now also available see WO 93/06213. See also Waterhouse; Nucl. Acids Res 21, 2265-2266 (1993).
  • chimaeric antibodies which generally comprise a non-human (e.g. rodent such as mouse) variable domain fused to a human constant region.
  • Chimaeric antibodies are typically produced using recombinant DNA methods.
  • DNA encoding the antibodies e.g. cDNA
  • cDNA DNA encoding the antibodies
  • Hybridoma cells serve as a typical source of such DNA.
  • a chimaeric antibody comprising a V H domain having the sequence: SEQ ID No:2, 6, or 10 and a V L domain having the sequence: SEQ ID No: 4, 8, or 12 fused to a human constant region (which maybe of a IgG isotype e.g. IgG1).
  • the second approach involves the generation of humanised antibodies wherein the non-human content of the antibody is reduced by humanizing the variable regions.
  • Two techniques for humanisation have gained popularity.
  • the first is humanisation by CDR grafting.
  • CDRs build loops close to the antibody's N-terminus where they form a surface mounted in a scaffold provided by the framework regions.
  • Antigen-binding specificity of the antibody is mainly defined by the topography and by the chemical characteristics of its CDR surface. These features are in turn determined by the conformation of the individual CDRs, by the relative disposition of the CDRs, and by the nature and disposition of the side chains of the residues comprising the CDRs.
  • a large decrease in immunogenicity can be achieved by grafting only the CDRs of a non-human (e.g.
  • human V regions showing the greatest sequence homology (typically 60% or greater) to the non-human donor antibody maybe chosen from a database in order to provide the human framework (FR).
  • the selection of human FRs can be made either from human consensus or individual human antibodies. Where necessary key residues from the donor antibody are substituted into the human acceptor framework to preserve CDR conformations.
  • Computer modelling of the antibody maybe used to help identify such structurally important residues, see WO99/48523.
  • humanisation maybe achieved by a process of “veneering”.
  • a statistical analysis of unique human and murine immunoglobulin heavy and light chain variable regions revealed that the precise patterns of exposed residues are different in human and murine antibodies, and most individual surface positions have a strong preference for a small number of different residues (see Padlan E. A. et al; (1991) Mol. Immunol. 28, 489-498 and Pedersen J. T. et al (1994) J. Mol. Biol. 235; 959-973). Therefore it is possible to reduce the immunogenicity of a non-human Fv by replacing exposed residues in its framework regions that differ from those usually found in human antibodies.
  • therapeutic antibody which is an antigen binding fragment.
  • fragments may be functional antigen binding fragments of intact and/or humanised and/or chimaeric antibodies such as Fab, Fd, Fab′, F(ab′) 2 , Fv, ScFv fragments of the antibodies described supra. Fragments lacking the constant region lack the ability to activate complement by the classical pathway or to mediate antibody-dependent cellular cytotoxicity. Traditionally such fragments are produced by the proteolytic digestion of intact antibodies by e.g. papain digestion (see for example, WO 94/29348) but may be produced directly from recombinantly transformed host cells. For the production of ScFv, see Bird et al; (1988) Science, 242, 423-426.
  • antibody fragments may be produced using a variety of engineering techniques as described below.
  • Fv fragments appear to have lower interaction energy of their two chains than Fab fragments.
  • V H and V L domains have been linked with peptides (Bird et al, (1988) Science 242, 423-426, Huston et al, PNAS, 85, 5879-5883), disulphide bridges (Glockshuber et al, (1990) Biochemistry, 29, 1362-1367) and “knob in hole” mutations (Zhu et al (1997), Protein Sci., 6, 781-788).
  • ScFv fragments can be produced by methods well known to those skilled in the art see Whitlow et al (1991) Methods companion Methods Enzymol, 2, 97-105 and Huston et al (1993) Int. Rev. Immunol 10, 195-217.
  • ScFv may be produced in bacterial cells such as E. Coli but are more typically produced in eukaryotic cells.
  • One disadvantage of ScFv is the monovalency of the product, which precludes an increased avidity due to polyvalent binding, and their short half-life. Attempts to overcome these problems include bivalent (ScFv′) 2 produced from ScFV containing an additional C terminal cysteine by chemical coupling (Adams et al (1993) Can.
  • ScFv can be forced to form multimers by shortening the peptide linker to between 3 to 12 residues to form “diabodies”, see Holliger et al PNAS (1993), 90, 6444-6448.
  • ScFv-Sc-Fv tandems may also be produced by linking two ScFv units by a third peptide linker, see Kurucz et al (1995) J. Immol. 154, 4576-4582.
  • Bispecific diabodies can be produced through the noncovalent association of two single chain fusion products consisting of V H domain from one antibody connected by a short linker to the V L domain of another antibody, see Kipriyanov et al (1998), Int. J. Can 77, 763-772.
  • bispecific diabodies can be enhanced by the introduction of disulphide bridges or “knob in hole” mutations as described supra or by the formation of single chain diabodies (ScDb) wherein two hybrid ScFv fragments are connected through a peptide linker see Kontermann et al (1999) J. Immunol. Methods 226 179-188.
  • Tetravalent bispecific molecules are available by e.g. fusing a ScFv fragment to the CH3 domain of an IgG molecule or to a Fab fragment through the hinge region see Coloma et al (1997) Nature Biotechnol. 15, 159-163.
  • tetravalent bispecific molecules have been created by the fusion of bispecific single chain diabodies (see Alt et al, (1999) FEBS Lett 454, 90-94.
  • Smaller tetravalent bispecific molecules can also be formed by the dimerization of either ScFv-ScFv tandems with a linker containing a helix-loop-helix motif (DiBi miniantibodies, see Muller et al (1998) FEBS Lett 432, 45-49) or a single chain molecule comprising four antibody variable domains (V H and V L ) in an orientation preventing intramolecular pairing (tandem diabody, see Kipriyanov et al, (1999) J. Mol. Biol.
  • Bispecific F(ab′)2 fragments can be created by chemical coupling of Fab′ fragments or by heterodimerization through leucine zippers (see Shalaby et al, (1992) J. Exp. Med. 175, 217-225 and Kostelny et al (1992), J. Immunol. 148, 1547-1553). Also available are so-called domain antibodies based on isolated V H or V L domains (Domantis Ltd.), see U.S. Pat. No. 6,248,516; U.S. Pat. No. 6,291,158; U.S. Pat. No. 6,172,197.
  • Heteroconjugate antibodies are derivatives which also form an embodiment of the present invention.
  • Heteroconjugate antibodies are composed of two covalently joined antibodies formed using any convenient cross-linking methods. See U.S. Pat. No. 4,676,980.
  • Antibodies of the present invention may also incorporate any other modifications in the constant regions.
  • glycosylation of antibodies at conserved positions in their constant regions is known to have a profound effect on antibody function, particularly effector functioning such as those described above, see for example, Boyd et al (1996), Mol. Immunol. 32, 1311-1318.
  • Glycosylation variants of the therapeutic antibodies of the present invention wherein one or more carbohydrate moiety is added, substituted, deleted or modified are contemplated.
  • Introduction of an asparagine-X-serine or asparagine-X-threonine motif creates a potential site for enzymatic attachment of carbonhydrate moieties and may therefore be used to manipulate the glycosylation of an antibody.
  • the invention concerns a plurality of therapeutic antibodies (which maybe of the IgG isotype, e.g. IgG1) as described herein comprising a defined number (e.g. 7 or less, for example 5 or less such as two or a single) glycoform(s) of said antibodies.
  • a defined number e.g. 7 or less, for example 5 or less such as two or a single
  • Derivatives according to the invention also include therapeutic antibodies of the invention coupled to a non-proteinaeous polymer such as polyethylene glycol (PEG), polypropylene glycol or polyoxyalkylene.
  • PEG polyethylene glycol
  • PEG polypropylene glycol
  • polyoxyalkylene polyethylene glycol
  • Conjugation of proteins to PEG is an established technique for increasing half-life of proteins, as well as reducing antigenicity and immunogenicity of proteins.
  • the use of PEGylation with different molecular weights and styles (linear or branched) has been investigated with intact antibodies as well as Fab′ fragments, see Koumenis I. L. et al (2000) Int. J. Pharmaceut. 198:83-95.
  • a particular embodiment comprises an antigen-binding fragment of the invention without the effector functions of a) activation of complement by the classical pathway; and b) mediating antibody-dependent cellular cytotoxicity; (such as a Fab fragment or a scFv) coupled to PEG.
  • Fc ⁇ R Fc receptors
  • human constant regions which essentially lack the functions of a) activation of complement by the classical pathway; and b) mediating antibody-dependent cellular cytotoxicity include the IgG4 constant region, the IgG2 constant region and IgG1 constant regions containing specific mutations as for example mutations at positions 234, 235, 236, 237, 297, 318, 320 and/or 322 disclosed in EP0307434 (WO8807089), EP 0629 240 (WO9317105) and WO 2004/014953.
  • Fc ⁇ R Fc ⁇ R
  • Fc ⁇ RIIa Fc ⁇ RIIb
  • Fc ⁇ RIIIa neonatal FcRn.
  • One group of IgG1 residues reduced binding to all Fc ⁇ Rs when altered to alanine: Pro-238, Asp-265, Asp-270, Asn-297 and Pro-239. All are in the IgG CH2 domain and clustered near the hinge joining CH1 and CH2.
  • Fc ⁇ RI utilizes only the common set of IgG1 residues for binding
  • Fc ⁇ RII and Fc ⁇ RIII interact with distinct residues in addition to the common set.
  • Alteration of some residues reduced binding only to Fc ⁇ RII (e.g. Arg-292) or Fc ⁇ RIII (e.g. Glu-293).
  • Some variants showed improved binding to Fc ⁇ RII or Fc ⁇ RIII but did not affect binding to the other receptor (e.g. Ser-267Ala improved binding to Fc ⁇ RII but binding to Fc ⁇ RIII was unaffected).
  • Other variants exhibited improved binding to Fc ⁇ RII or Fc ⁇ RIII with reduction in binding to the other receptor (e.g.
  • the therapeutic antibody of the invention may incorporate any of the above constant region modifications.
  • Antibodies of the present invention may be produced in transgenic organisms such as goats (see Pollock et al (1999), J. Immunol. Methods 231:147-157), chickens (see Morrow K J J (2000) Genet. Eng. News 20:1-55), mice (see Pollock et al ibid) or plants (see Doran P M, (2000) Curr. Opinion Biotechnol. 11, 199-204, Ma J K-C (1998), Nat. Med. 4; 601-606, Baez J et al, BioPharm (2000) 13: 50-54, Stoger E et al; (2000) Plant Mol. Biol. 42:583-590). Antibodies may also be produced by chemical synthesis.
  • antibodies of the invention are typically produced using recombinant cell culturing technology well known to those skilled in the art.
  • a polynucleotide encoding the antibody is isolated and inserted into a replicable vector such as a plasmid for further cloning (amplification) or expression in a host cell.
  • a replicable vector such as a plasmid for further cloning (amplification) or expression in a host cell.
  • a glutamate synthetase system such as sold by Lonza Biologics
  • Polynucleotide encoding the antibody is readily isolated and sequenced using conventional procedures (e.g. oligonucleotide probes).
  • Vectors that may be used include plasmid, virus, phage, transposons, minichromsomes of which plasmids are a typical embodiment.
  • such vectors further include a signal sequence, origin of replication, one or more marker genes, an enhancer element, a promoter and transcription termination sequences operably linked to the light and/or heavy chain polynucleotide so as to facilitate expression.
  • Polynucleotide encoding the light and heavy chains may be inserted into separate vectors and introduced (e.g. by transformation, transfection, electroporation or transduction) into the same host cell concurrently or sequentially or, if desired both the heavy chain and light chain can be inserted into the same vector prior to such introduction.
  • Antibodies of the present invention maybe produced as a fusion protein with a heterologous signal sequence having a specific cleavage site at the N terminus of the mature protein.
  • the signal sequence should be recognised and processed by the host cell.
  • the signal sequence may be an alkaline phosphatase, penicillinase, or heat stable enterotoxin II leaders.
  • yeast secretion the signal sequences may be a yeast invertase leader, a factor leader or acid phosphatase leaders see e.g. WO90/13646.
  • viral secretory leaders such as herpes simplex gD signal and a native immunoglobulin signal sequence (such as human Ig heavy chain) are available.
  • the signal sequence is ligated in reading frame to polynucleotide encoding the antibody of the invention.
  • Origin of replications are well known in the art with pBR322 suitable for most gram-negative bacteria, 2 ⁇ plasmid for most yeast and various viral origins such as SV40, polyoma, adenovirus, VSV or BPV for most mammalian cells.
  • origin of replication component is not needed for integrated mammalian expression vectors, unless vector propagation is required in E. Coli .
  • the SV40 ori may be used since it contains the early promoter.
  • Typical selection genes encode proteins that (a) confer resistance to antibiotics or other toxins e.g. ampicillin, neomycin, methotrexate or tetracycline or (b) complement auxotrophic deficiencies or supply nutrients not available in the complex media or (c) combinations of both.
  • the selection scheme may involve arresting growth of the host cells that contain no vector or vectors. Cells, which have been successfully transformed with the genes encoding the therapeutic antibody of the present invention, survive due to e.g. drug resistance conferred by the co-delivered selection marker.
  • One example is the DHFR-selection system wherein transformants are generated in DHFR negative host strains (eg see Page and Sydenham 1991 Biotechnology 9: 64-68).
  • the DHFR gene is co-delivered with antibody polynucleotide sequences of the invention and DHFR positive cells then selected by nucleoside withdrawal.
  • the DHFR inhibitor methotrexate is also employed to select for transformants with DHFR gene amplification.
  • DHFR gene amplification results in concomitant amplification of the desired antibody sequences of interest.
  • CHO cells are a particularly useful cell line for this DHFR/methotrexate selection and methods of amplifying and selecting host cells using the DHFR system are well established in the art see Kaufman R. J. et al J. Mol. Biol.
  • Suitable promoters for expressing antibodies of the invention are operably linked to DNA/polynucleotide encoding the antibody.
  • Promoters for prokaryotic hosts include phoA promoter, Beta-lactamase and lactose promoter systems, alkaline phosphatase, tryptophan and hybrid promoters such as Tac.
  • Promoters suitable for expression in yeast cells include 3-phosphoglycerate kinase or other glycolytic enzymes e.g.
  • Inducible yeast promoters include alcohol dehydrogenase 2, isocytochrome C, acid phosphatase, metallothionein and enzymes responsible for nitrogen metabolism or maltose/galactose utilization, among others.
  • Promoters for expression in mammalian cell systems include RNA polymerase II promoters including viral promoters such as polyoma, fowlpox and adenoviruses (e.g. adenovirus 2), bovine papilloma virus, avian sarcoma virus, cytomegalovirus (in particular the immediate early gene promoter), retrovirus, hepatitis B virus, actin, rous sarcoma virus (RSV) promoter and the early or late Simian virus 40 and non-viral promoters such as EF-1alpha (Mizushima and Nagata Nucleic Acids Res 1990 18(17):5322, among others.
  • the choice of promoter may be based upon suitable compatibility with the host cell used for expression.
  • enhancer elements can included instead of or as well as those found located in the promoters described above.
  • suitable mammalian enhancer sequences include enhancer elements from globin, elastase, albumin, fetoprotein, metallothionine and insulin.
  • an enhancer element from a eukaroytic cell virus such as SV40 enhancer, cytomegalovirus early promoter enhancer, polyoma enhancer, baculoviral enhancer or murine IgG2a locus (see WO04/009823).
  • enhancers are typically located on the vector at a site upstream to the promoter, they can also be located elsewhere e.g. within the untranslated region or downstream of the polydenalytion signal.
  • the choice and positioning of enhancer may be based upon suitable compatibility with the host cell used for expression.
  • polyadenylation signals are operably linked to polynucleotide encoding the antibody of this invention. Such signals are typically placed 3′ of the open reading frame.
  • signals include those derived from growth hormones, elongation factor-1 alpha and viral (eg SV40) genes or retroviral long terminal repeats.
  • polydenylation/termination signals include those derived from the phosphoglycerate kinase (PGK) and the alcohol dehydrogenase 1 (ADH) genes.
  • PGK phosphoglycerate kinase
  • ADH alcohol dehydrogenase 1
  • polyadenylation signals are typically not required and it is instead usual to employ shorter and more defined terminator sequences. The choice of polyadenylation/termination sequences may be based upon suitable compatibility with the host cell used for expression.
  • chromatin remodelling elements introns and host-cell specific codon modification.
  • the codon usage of the antibody of this invention thereof can be modified to accommodate codon bias of the host cell such to augment transcript and/or product yield (eg Hoekema A et al Mol Cell Biol 1987 7(8):2914-24).
  • the choice of codons may be based upon suitable compatibility with the host cell used for expression.
  • Suitable host cells for cloning or expressing vectors encoding antibodies of the invention are, for example, prokaroytic, yeast or higher eukaryotic cells.
  • Suitable prokaryotic cells include eubacteria e.g. enterobacteriaceae such as Escherichia e.g. E. Coli (for example ATCC 31,446; 31,537; 27,325), Enterobacter, Erwinia, Klebsiella Proteus, Salmonella e.g. Salmonella typhimurium, Serratia e.g. Serratia marcescans and Shigella as well as Bacilli such as B. subtilis and B.
  • enterobacteriaceae such as Escherichia e.g. E. Coli (for example ATCC 31,446; 31,537; 27,325)
  • Enterobacter Erwinia
  • Klebsiella Proteus Salmonella e.g. Salmonella typhimurium
  • yeast host cells Saccharomyces cerevisiae, schizosaccharomyces pombe, Kluyveromyces (e.g. ATCC 16,045; 12,424; 24178; 56,500), yarrowia (EP402, 226), Pichia Pastoris (EP183, 070, see also Peng et al J. Biotechnol. 108 (2004) 185-192), Candida, Trichoderma reesia (EP244, 234), Penicillin, Tolypocladium and Aspergillus hosts such as A. nidulans and A. niger are also contemplated.
  • host cells of the present invention are vertebrate cells.
  • Suitable vertebrate host cells include mammalian cells such as COS-1 (ATCC No. CRL 1650) COS-7 (ATCC CRL 1651), human embryonic kidney line 293, PerC6 (Crucell), baby hamster kidney cells (BHK) (ATCC CRL. 1632), BHK570 (ATCC NO: CRL 10314), 293 (ATCC NO. CRL 1573), Chinese hamster ovary cells CHO (e.g.
  • CHO-K1 ATCC NO: CCL 61, DHFR-CHO cell line such as DG44 (see Urlaub et al, (1986) ibid), particularly those CHO cell lines adapted for suspension culture, mouse sertoli cells, monkey kidney cells, African green monkey kidney cells (ATCC CRL-1587), HELA cells, canine kidney cells (ATCC CCL 34), human lung cells (ATCC CCL 75), Hep G2 and myeloma or lymphoma cells e.g. NS0 (see U.S. Pat. No. 5,807,715), Sp2/0, Y0.
  • DG44 see Urlaub et al, (1986) ibid
  • a stably transformed host cell comprising a vector encoding a heavy chain and/or light chain of the therapeutic antibody as described herein.
  • host cells comprise a first vector encoding the light chain and a second vector encoding said heavy chain.
  • Such host cells may also be further engineered or adapted to modify quality, function and/or yield of the antibody of this invention.
  • Non-limiting examples include expression of specific modifying (eg glycosylation) enzymes and protein folding chaperones.
  • Host cells transformed with vectors encoding the therapeutic antibodies of the invention may be cultured by any method known to those skilled in the art.
  • Host cells may be cultured in spinner flasks, shake flasks, roller bottles or hollow fibre systems but it is preferred for large scale production that stirred tank reactors or bag reactors (eg Wave Biotech, Somerset, N.J. USA) are used particularly for suspension cultures.
  • stirred tank reactors or bag reactors eg Wave Biotech, Somerset, N.J. USA
  • the stirred tankers are adapted for aeration using e.g. spargers, baffles or low shear impellers.
  • For bubble columns and airlift reactors direct aeration with air or oxygen bubbles maybe used.
  • the host cells are cultured in a serum free culture media it is preferred that the media is supplemented with a cell protective agent such as pluronic F-68 to help prevent cell damage as a result of the aeration process.
  • a cell protective agent such as pluronic F-68 to help prevent cell damage as a result of the aeration process.
  • either microcarriers maybe used as growth substrates for anchorage dependent cell lines or the cells maybe adapted to suspension culture (which is typical).
  • the culturing of host cells, particularly vertebrate host cells may utilise a variety of operational modes such as batch, fed-batch, repeated batch processing (see Drapeau et al (1994) cytotechnology 15: 103-109), extended batch process or perfusion culture.
  • recombinantly transformed mammalian host cells may be cultured in serum-containing media such media comprising fetal calf serum (FCS), it is preferred that such host cells are cultured in synthetic serum-free media such as disclosed in Keen et al (1995) Cytotechnology 17:153-163, or commercially available media such as ProCHO-CDM or UltraCHOTM (Cambrex N.J., USA), supplemented where necessary with an energy source such as glucose and synthetic growth factors such as recombinant insulin.
  • FCS fetal calf serum
  • synthetic serum-free media such as disclosed in Keen et al (1995) Cytotechnology 17:153-163, or commercially available media such as ProCHO-CDM or UltraCHOTM (Cambrex N.J., USA), supplemented where necessary with an energy source such as glucose and synthetic growth factors such as recombinant insulin.
  • the serum-free culturing of host cells may require that those cells are adapted to grow in serum free conditions.
  • One adaptation approach is to culture such host cells in serum containing media and repeatedly exchange 80% of the culture medium for the serum-free media so that the host cells learn to adapt in serum free conditions (see e.g. Scharfenberg K et al (1995) in Animal Cell technology: Developments towards the 21 st century (Beuvery E. C. et al eds), pp 619-623, Kluwer Academic publishers).
  • Antibodies of the invention secreted into the media may be recovered and purified from the media using a variety of techniques to provide a degree of purification suitable for the intended use.
  • the use of therapeutic antibodies of the invention for the treatment of human patients typically mandates at least 95% purity as determined by reducing SDS-PAGE, more typically 98% or 99% purity, when compared to the culture media comprising the therapeutic antibodies.
  • cell debris from the culture media is typically removed using centrifugation followed by a clarification step of the supernatant using e.g. microfiltration, ultrafiltration and/or depth filtration.
  • the antibody can be harvested by microfiltration, ultrafiltration or depth filtration without prior centrifugation.
  • HA hydroxyapatite
  • affinity chromatography optionally involving an affinity tagging system such as polyhistidine
  • hydrophobic interaction chromatography HIC, see U.S. Pat. No. 5,429,746
  • HIC hydrophobic interaction chromatography
  • the antibodies of the invention following various clarification steps, are captured using Protein A or G affinity chromatography followed by further chromatography steps such as ion exchange and/or HA chromatography, anion or cation exchange, size exclusion chromatography and ammonium sulphate precipitation.
  • virus removal steps are also employed (e.g. nanofiltration using e.g. a DV-20 filter).
  • a purified (typically monoclonal) preparation comprising at least 10 mg/ml or greater e.g. 100 mg/ml or greater of the antibody of the invention is provided and therefore forms an embodiment of the invention. Concentration to 100 mg/ml or greater can be generated by ultracentrifugation. Suitably such preparations are substantially free of aggregated forms of antibodies of the invention.
  • Bacterial systems are particularly suited for the expression of antibody fragments. Such fragments are localised intracellularly or within the periplasma. Insoluble periplasmic proteins can be extracted and refolded to form active proteins according to methods known to those skilled in the art, see Sanchez et al (1999) J. Biotechnol. 72, 13-20 and Cupit P M et al (1999) Lett Appl Microbiol, 29, 273-277.
  • compositions for use in the treatment of human diseases and disorders may be incorporated into pharmaceutical compositions for use in the treatment of human diseases and disorders such as those outlined above.
  • compositions further comprise a pharmaceutically acceptable (i.e. inert) carrier as known and called for by acceptable pharmaceutical practice, see e.g. Remingtons Pharmaceutical Sciences, 16th ed, (1980), Mack Publishing Co.
  • pharmaceutically acceptable carriers include sterilised carrier such as saline, Ringers solution or dextrose solution, buffered with suitable buffers to a pH within a range of 5 to 8.
  • Pharmaceutical compositions for injection e.g.
  • compositions by intravenous, intraperitoneal, intradermal, subcutaneous, intramuscular or intraportal) or continuous infusion are suitably free of visible particulate matter and may comprise from 0.1 mg to 10 g of therapeutic antibody, typically between 5 mg and 25 mg of antibody. Methods for the preparation of such pharmaceutical compositions are well known to those skilled in the art. In one embodiment, pharmaceutical compositions comprise from 0.1 mg to 10 g of therapeutic antibodies of the invention in unit dosage form, optionally together with instructions for use. Pharmaceutical compositions of the invention may be lyophilised (freeze dried) for reconstitution prior to administration according to methods well known or apparent to those skilled in the art.
  • a chelator of copper such as citrate (e.g. sodium citrate) or EDTA or histidine may be added to the pharmaceutical composition to reduce the degree of copper-mediated degradation of antibodies of this isotype, see EP0612251.
  • Effective doses and treatment regimes for administering the antibody of the invention are generally determined empirically and are dependent on factors such as the age, weight and health status of the patient and disease or disorder to be treated. Such factors are within the purview of the attending physician. Guidance in selecting appropriate doses may be found in e.g. Smith et al (1977) Antibodies in human diagnosis and therapy, Raven Press, New York but will in general be between 0.1 mg and 1 g.
  • the dosing regime for treating a human patient is 0.1 mg to 10 g of therapeutic antibody of the invention administered subcutaneously once per week or every two weeks, or by intravenous infusion every 1 or 2 months. Compositions of the present invention may also be used in prophylatically.
  • the present invention relates to an antibody which binds to human IL-8, Gro-alpha, Gro-beta, Gro-gamma, and ENA-78
  • the present invention also concerns methods of treating diseases or disorders characterised by elevated or unbalanced level of one or more of human IL-8, Gro-alpha, Gro-beta, Gro-gamma, and ENA-78, particularly, COPD, osteoarthritis, rheumatoid arthritis, erosive arthritis, asthma, atherosclerosis, inflammatory bowel disease, psoriasis, transplant rejection, gout, cancer, acute lung injury, acute lung disease, sepsis, ARDS, peripheral artery disease, systemic sclerosis, neonatal respiratory distress syndrome, exacerbation of asthma and COPD, cystic fibrosis, diffuse panbronchiolitis, reperfusion injury, and/or endometriosis with said antibody, pharmaceutical compositions comprising said antibody and methods of manufacture.
  • the present invention also relates to use of an antibody in the manufacture of a medicament for the treatment of diseases or disorders characterised by elevated or unbalanced level of one or more of human IL-8, Gro-alpha, Gro-beta, Gro-gamma, GCP-2 and ENA-78, particularly COPD, osteoarthritis, rheumatoid arthritis, erosive arthritis, asthma, atherosclerosis, inflammatory bowel disease, psoriasis, transplant rejection, gout, cancer, acute lung injury, acute lung disease, sepsis, ARDS, peripheral artery disease, systemic sclerosis, neonatal respiratory distress syndrome, exacerbation of asthma and COPD, cystic fibrosis, diffuse panbronchiolitis, reperfusion injury, or endometriosis.
  • the present invention has been described principally in relation to the treatment of human diseases or disorders, the present invention may also have applications in the treatment of similar diseases or disorders in non-human mammals.
  • pan-specific mAbs were generated using various mixtures of multiple antigenic peptides (MAPs) and/or intact target chemokines (IL-8, Gro- ⁇ , - ⁇ , - ⁇ , and ENA-78) mixed in complete or incomplete Freund's Adjuvant (cFA or iFA), following a modified Repetitive Immunization Multiple Sites (RIMMS) protocol in the SJL/JOrlCrl mouse strain.
  • MAPs multiple antigenic peptides
  • IL-8 intact target chemokines
  • cFA or iFA complete or incomplete Freund's Adjuvant
  • RIMMS Repetitive Immunization Multiple Sites
  • MAPs or multiple antigenic peptides serve two functions within the immunization protocol.
  • MAPs allow for a selective multiple presentation of a known target amino acid sequence.
  • mass due to multiple copies of the sequence linked, for example, via a lysine core, which also increases the immunogenicity of the sequence over that of individual peptides (Francis, J. P., et al., Immunology, 1991: 73; 249, Schott, M. E., et al., Cell. Immuno. 1996:174:199-209, Tam, J. P. Proc. Natl. Acad. Sci. 1988: 85; 5409-5413).
  • FIG. 1 is a schematic drawing of a set of MAPs having amino acid sequences SEQ ID NOs:49-53.
  • a linker in MAPs can be any linker other than lysines.
  • pan-specific mAbs Two different immunization protocols following the above time line produced pan-specific mAbs:
  • Pan-binding of the mAb to the target chemokines has been confirmed via a time-resolved fluorescence immunofluorescent assay (TRFIA).
  • each antigen human IL-8, Gro-alpha, Gro-beta, Gro-gamma, GCP-2, and ENA-78, and hIL-18 when used as negative control
  • each antigen human IL-8, Gro-alpha, Gro-beta, Gro-gamma, GCP-2, and ENA-78, and hIL-18 when used as negative control
  • Blocking solution is emptied and samples containing the mAb are added into each well and incubated for 40 to 60 minutes (this allows the mAb to bind to the target chemokines).
  • the plates are then washed again and a detection Ab is added to each well.
  • Detection reagents were as follows for 2 ⁇ 656.35, chimera (HcLc) and humanized MAbs respectively: biotinylated anti-mouse IgG, biotinylated anti-human IgG Fab2 reagent, and europium anti-human IgG.
  • the detection antibody is conjugated to either europium or biotin.
  • streptavidin-europium was added to biotin detected assays. Streptavidin-europium binds to the biotin allowing for detection.
  • FIG. 5A shows mouse 656.35 mAb binding to target human chemokines.
  • FIG. 5B shows the Chimera Antibody (HcLc) mAb binding to human target chemokines.
  • FIG. 5C shows humanized mAbs binding to human target chemokines.
  • a microtiter plate based calcium mobilization assay FLIPR (Fluorometric Imaging Plate Reader, Molecular Devices, Sunnyvale Calif., [Schroeder, 1996]), was used for the functional characterization of the neutralizing effect of antibodies on ELR+ chemokine induced [Ca 2+ ]i-mobilization in CHO-K1 cells transfected with and stably expressing hCXCR2 and G ⁇ 16.
  • the dye containing media was then aspirated off the cells and replaced with identical media without Fluo-4 AM and with 0.1% Gelatin (BSA removed) and 2.5 mM probenecid.
  • Cells were incubated for 10 min. at 37° C. and then washed 3 times with KRH assay buffer [Krebs Ringer Henseleit (120 mM NaCl, 4.6 mM KCl, 1.03 mM KH 2 PO 4 , 25 mM NaHCO 3 , 1.0 mM CaCl 2 , 11 mM MgCl 2 , 11 mM Glucose, 20 mM HEPES (pH 7.4)) with 0.1% gelatin and 2.5 mM probenecid].
  • KRH assay buffer KRH assay buffer
  • the IC 50 was defined as the concentration of antibody required, during pre-treatment of 3 ⁇ EC 80 chemokine, to neutralize the CXCR2 mediated stimulatory effect of an EC 80 concentration of the ELR+ chemokine by 50%.
  • a secondary cellular response to 25 ⁇ M ATP was monitored to test cell viability [Sarau, 1999].
  • micro-Boyden apparatus consists of two small chambers one above and one below a 3 micron-porous membrane.
  • the lower chamber is loaded with the chemotaxic agent (i.e. IL-8) or a mixture of the chemokine with the penta-specific mAb.
  • the upper chamber contains purified non-activated human neutrophils. Once the chamber is assembled a concentration gradient is formed from the bottom well to the upper, stimulating the neutrophils to chemotax across the membrane.
  • the assay is expressed as a CI (chemotactic index) which is a ratio of the number of cells which chemotax in response to a stimulant over non-stimulated chemotaxic cell number.
  • CI chemotactic index
  • Pre-incubating the chemokine (i.e. IL-8) with the penta-specific mAb in the lower chamber dose dependently inhibited IL-8 stimulated neutrophil chemotaxis.
  • IL-8 at 10 nM achieved a CI of 3.6 ⁇ 0.8.
  • CD11b or Mac-1 mediates adhesion to substrates, aggregation and chemotaxis and is known to be up-regulated on the surface activated neutrophils (Molad, Y., J., et al., Clin. Immunol. Immunopathol. 1994: 71; 281-286). Briefly, non-activated human neutrophils are purified and ex vivo stimulated with either target chemokines (i.e. IL-8) or with chemokines pre-incubated with a chemokine penta-specific mAb. Data are presented as percent activation set to the maximal CD11b surface expression due to IL-8 stimulation.
  • target chemokines i.e. IL-8
  • rabbit anti mouse IgG-Fc (Biacore BR-1005-14) is immobilized on a CM5 chip by primary amine coupling in accordance with the manufactures instructions.
  • Supernatant or purified antibody from parental mouse mAb is captured on the anti-mouse IgG-Fc surface.
  • concentrations of each analyte IL8, Gro-band ENA-78
  • a separate capture event is used for each analyte injection. After each injection of analyte the surface is regenerated by injection
  • the present invention relates to a penta-specific antibody which binds within epitope of SEQ ID NO:54 of human IL-8.
  • NHP non-human primate
  • cynoIL-8 cynoIL-8
  • Selected NHP were then subjected to baseline sample collections (blood and bronchoalveolar lavage—BAL) five days prior to the first LPS challenge.
  • LPS challenged consisted of tranquilizing the NHP with a single IM injection of ketamine HCl ( ⁇ 10 mg/kg).
  • anesthesia is administered, via an intravenous infusion of propofol ( ⁇ 0.2 mg/kg/min, as necessary).
  • Anesthetized animals are placed on a circulating warm-water heating blanket and/or within a circulating warm-air blanket and an ophthalmic lubricant is administered to each eye.
  • the animals are then intubated and mechanically ventilated for challenge procedure.
  • a volume regulated Positive Pressure ventilator is used during the procedure (Stoelting Cat/Rabbit ventilator www.stoeltingco.com Cat #5019510).
  • Lipopolysaccharide (LPS) challenge is performed via aerosol inhalation using a DeVilbiss Ultraneb-99 ultrasonic nebulizer.
  • Aerosolized LPS is administered for 5 minutes at 100 ug/ml.
  • Heart rate, body temperature, and respiration rate are monitored and recorded during challenge procedures.
  • the initial challenge confirms that the NHP respond to LPS (increased neutrophil infiltration into the lungs and elevated cynoIL-8 levels).
  • Individual NHP responses were confirmed by sample collections at 6 and 24-hours post challenge.
  • the initial LPS challenged NHP were then randomly divided into two groups. Following a 4-week recovery all participating NHP were subjected to another baseline sample (blood and BAL). Four days post baseline measurements the subject groups received IV injection of either a vehicle or an injection of 1 mg/kg of the Chimera Antibody. The next day the NHP were challenged again with inhaled LPS and samples for analysis were collected 6 and 24-hour post challenge. Following an additional recovery period base line samples were collected from the vehicle NHP, 4 days later these NHP were treated with a single IV bolus injection of the Chimera Antibody at 10 mg/kg. The next day the animals were exposed to an LPS challenge and samples were collected for analysis at 6 and 24 hours.
  • the LPS inhalation is an acute inflammatory model which stimulates an elevation of chemotactic chemokines such at IL-8, which lead to increased infiltration of neutrophils into the lungs.
  • the primary efficacy parameter for the Chimera Antibody treatment is inhibition of infiltrating neutrophils into the lungs of NHP challenged with LPS. Neutrophil infiltration in this acute inflammatory model occurs during the first 24 hours following LPS challenge at which point other inflammatory processes are becoming more prominent.
  • a Protein A capture method was used to generate kinetics for the humanised and chimeric constructs as follows:
  • Protein A is immobilised on a CM5 chip by primary amine coupling in accordance with the manufactures instructions.
  • Purified humanised or chimeric antibody is captured on the Protein A surface.
  • capture defined concentrations of each analyte IL8, Gro- ⁇ and ENA-78
  • a separate capture event is used for each analyte injection.
  • an injection of analyte the surface is regenerated by injection of a mild acidic solution, which removes the captured antibody but does not significantly affect the capability of the Protein A to perform another capture event.
  • An injection of buffer is also injected over the antibody captured surface and this is used for double referencing. The data is analysed using the analysis software inherent to the machine, using the 1:1 model of binding.
  • a microtiter plate based calcium mobilization assay FLIPR (Fluorometric Imaging Plate Reader, Molecular Devices, Sunnyvale Calif., [Schroeder, 1996]), was used for the functional characterization of the neutralizing effect of antibodies on ELR+ chemokine induced [Ca 2+ ]i-mobilization in CHO-K1 cells transfected with and stably expressing hCXCR2 and G ⁇ 16.
  • the dye containing media was then aspirated off the cells and replaced with identical media without Fluo-4 AM and with 0.1% Gelatin (BSA removed) and 2.5 mM probenecid.
  • Cells were incubated for 10 min. at 37° C. and then washed 3 times with KRH assay buffer [Krebs Ringer Henseleit (120 mM NaCl, 4.6 mM KCl, 1.03 mM KH 2 PO 4 , 25 mM NaHCO 3 , 1.0 mM CaCl 2 , 1.1 mM MgCl 2 , 11 mM Glucose, 20 mM HEPES (pH 7.4)) with 0.1% gelatin and 2.5 mM probenecid].
  • KRH assay buffer KRH assay buffer
  • the IC 50 was defined as the concentration of antibody required, during pre-treatment of 3 ⁇ EC 80 chemokine, to neutralize the CXCR2 mediated stimulatory effect of an EC 80 concentration of the ELR+ chemokine by 50%.
  • a secondary cellular response to 25 ⁇ M ATP was monitored to test cell viability [Sarau, 1999].
  • FLIPR a new instrument for accurate, high throughput optical screening. J. Biomol. Screen. 1996:1-75. Sarau H M, Ames R S, Chambers J, Ellis C, Elshourbagy N, Foley J J et al. Identification, molecular cloning, expression, and characterization of a cysteinyl leukotrien receptor. Mol Pharmacol. 1999; 56:657-663.
  • the forward primer for RT-PCR was a mixture of degenerate primers specific for murine immunoglobulin gene leader-sequences and the reverse primer was specific for the antibody constant regions, in this case isotype IgG2a/ ⁇ . Primers were designed according to the strategy described by Jones and Bendig (Bio/Technology 9:88, 1991). RT-PCR was carried out in duplicate for both V-region sequences to enable subsequent verification of the correct V-region sequences.
  • V-region products generated by the RT-PCR were cloned (Invitrogen TA Cloning Kit) and sequence data obtained. This process was not successful for 81.1 light variable domain.
  • the amino acid sequence shown below (Seq ID NO: 12) was therefore generated by protein sequencing the light chain isolated on a SDS-polyacrylamide gel run under reducing conditions. Complementarity Determining Regions (CDRs) are underlined.
  • CDRs Complementarity Determining Regions
  • VYGMN SEQ ID NO: 25 NFDPYFSVTSYNQKFQD SEQ ID NO: 26 GSWETIFAY SEQ ID NO: 27 Polynucleotide sequences for heavy chain CDRs (SEQ ID NO: 43, 44, 45) for 81.1

Abstract

The present invention relates to an antibody which has multiple specificities. In particular the antibody of the present invention binds to (cross react with) human IL-8, Gro-alpha, Gro-beta, Gro-gamma, and ENA-78.

Description

    CROSS-REFERENCE TO RELATED APPLICATION
  • This application claims priority to U.S. provisional application 60/912,229 filed Apr. 17, 2007 and 61/044,132 filed Apr. 11, 2008, which are incorporated by reference in their entirety.
  • FIELD OF THE INVENTION
  • The present invention relates to an antibody which has multiple specificities. In particular the antibody of the present invention binds to (cross react with) human IL-8, Gro-alpha, Gro-beta, Gro-gamma, and ENA-78. The present invention also concerns with methods of treating diseases or disorders characterised by elevated or unbalanced level of one or more of human IL-8, Gro-alpha, Gro-beta, Gro-gamma, and ENA-78, particularly COPD, osteoarthritis, rheumatoid arthritis, erosive arthritis, asthma, atherosclerosis, inflammatory bowel disease, psoriasis, transplant rejection, gout, cancer, acute lung injury, acute lung disease, sepsis, ARDS, peripheral artery disease, systemic sclerosis, neonatal respiratory distress syndrome, exacerbation of asthma and COPD, cystic fibrosis, diffuse panbronchiolitis, reperfusion injury, and/or endometriosis with said antibody.
  • BACKGROUND OF THE INVENTION
  • Published data and reports indicate that the members of the ELRCXC subfamily of CXCL chemokines are elevated in a number of diseases. There are a total of 16 CXCL family members. The chemokines are reported to be up-regulated in a number of inflammatory diseases, including COPD, in which CXCL1-3, 5, and 8, also known as Gro-1-α, -β, -γ (Haskill, S., et al. Proc. Natl. Acad. Sci., 1990: 87, 7732-7736), ENA-78 (Wang, D. and Richmond, A., Cytokine Reference. Oppenheim, J. J. and Feldman, M. ed., Academic Press, London, 1023-1027, Powerm C. A. et al. Gene., 1994: 151, 333-334), and IL-8 (Iizasa, H. and Matsushima, K., Cytokine Reference. Oppenheim, J. J. and Feldman, M. ed., Academic Press, London, 1061-1067, Matsushima, K. et al., J. Exp. Med. 1988: 167, 1883-1893) respectively (Am. J. Respir. Crit Care Med., 163: 349-355, 2001, Am. J. Respir. Crit Care Med., 168: 968-975, 2003, Thorax, 57: 590-595, 2002). It has be postulated that prolonged and elevated expression of these chemokines could be involved in the development of diseases such as COPD, osteoarthritis, rheumatoid arthritis, erosive arthritis, asthma, atherosclerosis, inflammatory bowel disease, psoriasis, transplant rejection, gout, cancer, acute lung injury, acute lung disease, sepsis, ARDS, peripheral artery disease, systemic sclerosis, neonatal respiratory distress syndrome, exacerbation of asthma and COPD, cystic fibrosis, diffuse panbronchiolitis, reperfusion injury, or endometriosis. These CXC chemokines are known to stimulate neutrophil chemotaxis by engaging and activating the CXCR1 and/or CXCR2 receptors. Thus the inhibition of these chemokines could prevent inflammatory cells from infiltrating the lung tissue and thus prevent tissue damage. The present invention is directed to inhibiting the activation of CXCR1 and CXCR2 receptors by using an antibody having the ability to bind to human IL-8, Gro-alpha, Gro-beta, Gro-gamma, and ENA-78, i.e. a penta-specific antibody.
  • SUMMARY OF THE INVENTION
  • The present invention relates to an antibody (immunoglobulin) which has multiple specificities contained within one immunoglobulin. In particular the antibody of the present invention binds to (cross react with) human IL-8, Gro-alpha, Gro-beta, Gro-gamma, and ENA-78. The present invention also concerns with methods of treating diseases or disorders characterised by elevated or unbalanced level of one or more of human IL-8, Gro-alpha, Gro-beta, Gro-gamma, and ENA-78, particularly COPD, osteoarthritis, rheumatoid arthritis, erosive arthritis, asthma, atherosclerosis, inflammatory bowel disease, psoriasis, transplant rejection, gout, cancer, acute lung injury, acute lung disease, sepsis, ARDS, peripheral artery disease, systemic sclerosis, neonatal respiratory distress syndrome, exacerbation of asthma and COPD, cystic fibrosis, diffuse panbronchiolitis, reperfusion injury, and/or endometriosis with said antibody.
  • In one aspect, the present invention relates to an isolated antibody which has multiple specificity to (or cross reacts with) human IL-8, Gro-alpha, Gro-beta, Gro-gamma, and ENA-78; thus we define the antibody of the present invention as penta-specific (or pan-ELR) antibody. The definition of antibody includes an antigen binding portion (or fragment) of the antibody such that the antigen binding portion (or fragment) binds to human IL-8, Gro-alpha, Gro-beta, Gro-gamma, and ENA-78. The penta-specific antibody of the invention is preferably murine monoclonal, chimeric, human or humanized. For avoidance of doubt, the penta-specific antibody of the present invention need not bind solely to human IL-8, Gro-alpha, Gro-beta, Gro-gamma, and ENA-78 antigens, but it may also to bind to other related proteins (such as human GCP-2, or may even bind to non-human orthologues of IL-8, Gro-alpha, Gro-beta, Gro-gamma, ENA-78, and GCP-2); in other words, the penta-specific antibody of the present invention minimally binds to human IL-8, Gro-alpha, Gro-beta, Gro-gamma, and ENA-78.
  • Preferably, a penta-specific antibody of the present invention binds to each of human IL-8, Gro-alpha, Gro-beta, Gro-gamma, and ENA-78 with equilibrium constant, KD, values of less than 10−7 M, more preferably less than 10−8 M, and even more preferably less than 10−9 M as determined by surface plasmon resonance. Typically surface plasmon resonance measurement is conducted as described below.
  • In one embodiment the present invention comprises a method of decreasing the neutrophil chemotaxis through inhibition of CXCR1 and CXCR2 receptor activation by neutralizing human IL-8, Gro-alpha, Gro-beta, Gro-gamma, GCP-2 and ENA-78 with a penta-specific antibody of the present invention.
  • In one embodiment the present invention relates to a method of decreasing the neutrophil chemotaxis in a patient in need thereof by administering a penta-specific antibody of the present invention.
  • In one embodiment, a penta-specific antibody binds within epitope of KELRCQCIKTYSKP (SEQ ID NO: 54) in human IL-8.
  • In one embodiment, the penta-specific antibody of the present invention is generated by a method comprising the steps of using RIMMs (Kilpatrick, K. E., et al. Hybridoma. 1997: 16, 381) type protocol using a mixture (cocktail) of human IL-8, Gro-alpha, Gro-beta, Gro-gamma, and ENA-78 together with a set of five multiple antigenic peptides (MAPs) each MAP unit having one separate sequence from polypeptides of ID NOs: 49-53.
  • LATELRSQSLQTLQG SEQ ID NO: 49
    SAKELRSQSIKTYSK SEQ ID NO: 50
    LRELRSVSLQTTQG SEQ ID NO: 51
    SPGPHSAQTEVIAT SEQ ID NO: 52
    ESGPHSANTEIIVK SEQ ID NO: 53
  • Without being bound by theory, MAPs serve two functions within the immunization protocol. First, MAPs allow for a selective multiple presentation of a known target amino acid sequence to the host immune system. Secondly, the increase in mass, due to multiple copies of the sequence linked via a core, such as, but not limited to lysine, increases the immunogenicity of the sequence over that of individual peptides (Francis, J. P., et al., Immunology, 1991: 73; 249, Schott, M. E., et al., Cell. Immuno. 1996: 174: 199-209, Tam, J. P. Proc. Natl. Acad. Sci. 1988: 85; 5409-5413).
  • The MAPs used to generate this invention are comprised of multiple copies of the conserved target sequences (e.g. SEQ ID NOs: 49-53) found with and around the ELRCXC and GPHCA regions of target chemokines. Exemplary MAP set is depicted in FIG. 1.
  • In one embodiment, a penta-specific antibody of the present invention is generated by a method comprising the steps of:
      • a. injecting into a mouse a mixture of human IL-8, Gro-alpha, Gro-beta, Gro-gamma, and ENA-78 in complete Freund's adjuvant (cFA);
      • b. injecting into the mouse a mixture of human IL-8, Gro-alpha, Gro-beta, Gro-gamma, and ENA-78 in incomplete Freund's adjuvant (iFA); and
      • c. injecting into the mouse a mixture of human IL-8, Gro-alpha, Gro-beta, Gro-gamma, and ENA-78, and a set of five multiple antigenic peptides (MAPs), each MAP unit having one separate sequence from polypeptides of ID NOs: 49-53 in incomplete Freund's adjuvant;
      • d. isolating B cells from the mouse;
      • e. fusing the B cells with myeloma cells to form immortal hybridoma cells that secrete the desired penta-specific antibody; and
      • f. isolating the penta-specific antibody from the culture supernatant of the hybridoma.
      • If desired, one can optionally inject into the mouse a mixture of human IL-8, Gro-alpha, Gro-beta, Gro-gamma, and ENA-78, and a set of MAPs comprising amino acid sequences of SEQ ID NOs: 49-53 in PBS between steps c and d.
  • In another embodiment, a penta-specific antibody of the present invention is generated by a method comprising the steps of:
      • a. injecting into a mouse a set of five multiple antigenic peptides (MAPs) each MAP unit having one separate sequence from polypeptides of ID NOs: 49-53 (hereinafter also referred to as the MAP set) in complete Freund's adjuvant;
      • b. injecting into the mouse the MAP set in incomplete Freund adjuvant;
      • c. injecting into the mouse a mixture of all human IL-8, Gro-alpha, Gro-beta, Gro-gamma, and ENA-78, and the MAP set in incomplete Freund's adjuvant;
      • d. isolating B cells from the mouse; and
      • e. fusing the B cells with myeloma cells to form immortal hybridoma cells that secrete the desired penta-specific antibody; and
      • f. isolating the penta-specific antibody from the culture supernatant of the hybridoma.
      • If desired, one can optionally inject into the mouse a mixture of human IL-8, Gro-alpha, Gro-beta, Gro-gamma, and ENA-78, and a set of MAPs having SEQ ID NOs: 49-53 in PBS between steps c and d.
  • In another embodiment, the present invention concerns a penta-specific antibody made by the foregoing process.
  • In one embodiment, a penta-specific antibody has heavy and light chain variable regions encoded by nucleotide sequences comprising sequences of SEQ ID NO: 1 and SEQ ID NO:3, respectively, or conservative sequence modifications thereof.
  • In one embodiment, a penta-specific antibody has heavy and light chain variable regions encoded by nucleotide sequences comprising sequences of SEQ ID NO:5 and SEQ ID NO:7, respectively, or conservative sequence modifications thereof.
  • In one embodiment, a penta-specific antibody has heavy chain variable region encoded by a nucleotide sequence comprising sequence of SEQ ID NO:9, or conservative sequence modifications thereof.
  • In one embodiment, a penta-specific antibody has heavy and light chain variable regions comprising the amino acid sequences of SEQ ID NO:2 and SEQ ID NO:4, respectively, or conservative sequence modifications thereof.
  • In one embodiment, a penta-specific antibody has heavy and light chain variable regions comprising polypeptides which are at least 90%, 95%, 98% or 99% identical to the amino acid sequences of SEQ ID NO:2 and SEQ ID NO:4, respectively.
  • In one embodiment, a penta-specific antibody has heavy and light chain variable regions comprising the amino acid sequences of SEQ ID NO:6 and SEQ ID NO:8, respectively, or conservative sequence modifications thereof.
  • In one embodiment, a penta-specific antibody has heavy and light chain variable regions comprising polypeptides which are at least 90%, 95%, 98% or 99% identical to the amino acid sequences of SEQ ID NO:6 and SEQ ID NO:8, respectively.
  • In one embodiment, a penta-specific antibody has heavy and light variable regions comprising the amino acid sequences of SEQ ID NOs:10 and 12, respectively, or conservative sequence modifications thereof.
  • In one embodiment, a penta-specific antibody has heavy and light chain variable regions comprising polypeptide sequences which are at least 90%, 95%, 98% or 99% identical to the amino acid sequences of SEQ ID NOs:10 and 12, respectively.
  • In one embodiment, a penta-specific antibody comprises at least one variable region selected from (i) the amino acid SEQ ID NO: 2, 4, 6, 8, 10, or 12; or (ii) an amino acid sequence which is at least 90%, 95%, 98% or 99% identical to any one of the amino acid sequences of (i) above.
  • In one embodiment, a penta-specific antibody comprises CDR sequences of SEQ ID NOs: 13, 14, 15, 16, 17, and 18; or one or more of the CDR sequences can be conservative sequence modifications of the sequences SEQ ID NOs: 13, 14, 15, 16, 17, and 18.
  • In one embodiment, the present invention relates to hybridoma or transfectoma which produces a penta-specific antibody which comprises CDR sequences of SEQ ID NOs: 13, 14, 15, 16, 17, and 18.
    In one embodiment, the present invention relates to a recombinant eukaryotic or prokaryotic cell which produces a penta-specific antibody which comprises CDR sequences of SEQ ID NOs: 13, 14, 15, 16, 17, and 18.
  • In one embodiment, a penta-specific antibody comprises at least one CDR sequence selected from (i) SEQ ID NO: 13, 14, 15, 16, 17, or 18; or (ii) a conservative sequence modification of the sequences listed in (i).
  • In one embodiment, a penta-specific antibody comprises a polypeptide of SEQ ID NO:15.
  • In one embodiment, a penta-specific antibody comprises at least four CDR sequences selected from the group consisting of SEQ ID NOs: 13, 14, 15, 16, 17, and 18; or one or more of the CDR sequences can be conservative sequence modifications of the sequences listed in SEQ ID NOs: 13, 14, 15, 16, 17, and 18.
  • In one embodiment, a penta-specific antibody comprises heavy and light chain variable regions which comprise the CDR amino acid sequences of SEQ ID NOs: 13, 14, and 15, and SEQ ID NOs: 16, 17, and 18, respectively.
  • In one embodiment, a penta-specific antibody comprises CDR sequences of SEQ ID NOs: 19, 20, 21, 22, 23, and 24; or one or more the CDR sequences can be conservative sequence modifications of the sequences listed in SEQ ID NOs: 19, 20, 21, 22, 23, and 24.
  • In one embodiment, the present invention relates to an hybridoma or transfectoma which produces a penta-specific antibody which comprises CDR sequences of SEQ ID NOs: 19, 20, 21, 22, 23, and 24.
  • In one embodiment, the present invention relates to a recombinant eukaryotic or a prokaryotic cell which produces a penta-specific antibody which comprises CDR sequences of SEQ ID NOs: 19, 20, 21, 22, 23, and 24.
  • In one embodiment, a penta-specific antibody comprises at least one CDR sequence selected from (i) SEQ ID NO: 19, 20, 21, 22, 23, or 24; or (ii) a conservative sequence modification of the sequences listed in (i).
  • In one embodiment, a penta-specific antibody comprises a polypeptide of SEQ ID NO:21.
  • In one embodiment, a penta-specific antibody comprises at least four CDR sequences selected from the group consisting of: SEQ ID NOs: 19, 20, 21, 22, 23, and 24; or one or more of the CDR sequences can be conservative sequence modifications of the sequences listed in SEQ ID NOs: 19, 20, 21, 22, 23, and 24.
  • In one embodiment, a penta-specific antibody comprises heavy and light chain variable regions which comprise the CDR amino acid sequences of SEQ ID NOs: 19, 20, and 21, and SEQ ID NOs: 22, 23, and 24, respectively.
  • In one embodiment, a penta-specific antibody comprises CDR sequences of SEQ ID NOs: 25, 26, 27, 28, 29, and 30; or one or more the CDR sequences can be conservative sequence modifications of the sequences listed in SEQ ID NOs: 25, 26, 27, 28, 29, and 30.
  • In one embodiment, the present invention relates to an hybridoma or transfectoma which produces a penta-specific antibody which comprises CDR sequences of SEQ ID NOs: 25, 26, 27, 28, 29, and 30.
  • In one embodiment, the present invention relates to a recombinant eukaryotic or prokaryotic cell which produces a penta-specific antibody which comprises CDR sequences of SEQ ID NOs: 25, 26, 27, 28, 29 and 30.
  • In one embodiment, a penta-specific antibody comprises at least one CDR sequence selected from (i) SEQ ID NO: 25, 26, 27, 28, 29, or 30; or (ii) a conservative sequence modification of the sequences listed in (i).
  • In one embodiment, a penta-specific antibody comprises a polypeptide of SEQ ID NO:27.
  • In one embodiment, a penta-specific antibody comprises at least four CDR sequences selected from the group consisting of: SEQ ID NOs: 25, 26, 27, 28, 29, and 30; or one or more of the CDR sequences can be conservative modifications of the sequences listed in SEQ ID NOs: 25, 26, 27, 28, 29, and 30.
  • In one embodiment, a penta-specific antibody comprises heavy and light variable chain regions which comprise the CDR amino acid sequences of SEQ ID NOs: 25, 26, and 27, and SEQ ID NOs: 28, 29 and 30, respectively.
  • In one embodiment, the present invention concerns a hybridoma which produces a monoclonal antibody having heavy or light chain variable region encoded by nucleotide sequences comprising nucleotide sequences of SEQ ID NO:1, 5, or 9, or SEQ ID NO:3 or 7, respectively.
  • In one embodiment, the present invention concerns a hybridoma which produces a monoclonal antibody having heavy or light chain variable region comprising the amino acid sequences of SEQ ID NO:2, 6, or 10, or SEQ ID NO: 4, 8, or 12, respectively, and conservative sequence modifications thereof.
  • In one embodiment, the present invention relates to a recombinant eukaryotic or a prokaryotic host cell which produces a penta-specific antibody having heavy or light variable region which comprise the amino acid sequences of SEQ ID NO:2, 6, or 10, or SEQ ID NO:4, 8, or 12, respectively, and conservative sequence modifications thereof.
  • In one embodiment, the present invention relates to an expression vector comprising nucleotide sequences encoding a variable heavy or light chain of a penta-specific antibody comprising the CDR sequences of SEQ ID NOs: 13, 14, and 15; or SEQ ID NOs: 16, 17, and 18, respectively.
  • In one embodiment, the present invention relates to an expression vector comprising a nucleotide sequence encoding a CDR sequence of a penta-specific antibody selected from SEQ ID NO: 13, 14, 15, 16, 17, or 18.
  • In one embodiment, the present invention relates to an expression vector comprising nucleotide sequences encoding at least four CDR sequences of a penta-specific antibody selected from the group consisting of SEQ ID NOs: 13, 14, 15, 16, 17, and 18.
  • In one embodiment, the present invention relates to an expression vector comprising polynucleotide sequences of SEQ ID NOs: 31, 32, or 33.
  • In one embodiment, the present invention relates to an expression vector comprising polynucleotide sequences of SEQ ID NOs: 34, 35, and 36.
  • In one embodiment the present invention relates to an expression vector comprising at least four polynucleotide sequences selected from the group of SEQ ID NOs: 31, 32, 33, 34, 35, and 36.
  • In one embodiment, the present invention relates to an expression vector comprising nucleotide sequences encoding a variable heavy or light chain of a penta-specific antibody comprising the CDR sequences of SEQ ID NOs: 19, 20, and 21; or 22, 23, and 24, respectively.
  • In one embodiment, the present invention relates to an expression vector comprising a nucleotide sequence encoding a CDR sequence of a penta-specific antibody selected from SEQ ID NO: 19, 20, 21, 22, 23, or 24.
  • In one embodiment, the present invention relates to an expression vector comprising nucleotide sequences encoding at least four CDR sequences of a penta-specific antibody selected from the group consisting of SEQ ID NOs: 19, 20, 21, 22, 23, and 24.
  • In one embodiment, the present invention relates to an expression vector comprising polynucleotide sequences of SEQ ID NOs: 37, 38, and 39.
  • In one embodiment, the present invention relates to an expression vector comprising polynucleotide sequences of SEQ ID NOs: 40, 41, and 42.
  • In one embodiment the present invention relates to an expression vector comprising at least four polynucleotide sequences selected from the group of SEQ ID NOs: 37, 38, 39, 40, 41, and 42.
  • In one embodiment, the present invention relates to an expression vector comprising nucleotide sequences encoding a variable heavy chain of a penta-specific antibody comprising the CDR sequences of SEQ ID NOs: 25, 26, and 27.
  • In one embodiment, the present invention relates to an expression vector comprising a nucleotide sequence encoding a CDR sequence of a penta-specific antibody selected from SEQ ID NO: 25, 26, or 27.
  • In one embodiment, the present invention relates to an expression vector comprising polynucleotide sequences of SEQ ID NOs: 43, 44, and 45.
  • In one embodiment, a penta-specific antibody has heavy and light chains encoded by nucleotide sequences comprising sequences of SEQ ID NO:11, and SEQ ID NO:47, 59, 61, or 63, respectively.
  • In one embodiment, a penta-specific antibody has heavy and light chains encoded by nucleotide sequences comprising sequences which are at least 90%, 95%, 98% or 99% identical to sequences of SEQ ID NO:11, and SEQ ID NO:47, 59, 61, or 63, respectively.
  • In one embodiment, a penta-specific antibody has heavy and light chains comprising the amino acid sequences of SEQ ID NO:46, and SEQ ID NO:48, 60, 62, or 64, respectively.
  • In one embodiment, a penta-specific antibody has heavy and light chains comprising the amino acid sequences of SEQ ID NO:46 and SEQ ID NO:48, respectively.
  • In one embodiment, a penta-specific antibody has heavy and light chains comprising the amino acid sequences of SEQ ID NO:46 and SEQ ID NO:60, respectively.
  • In one embodiment, a penta-specific antibody has heavy and light chains comprising the amino acid sequences of SEQ ID NO:46 and SEQ ID NO: 62, respectively.
  • In one embodiment, a penta-specific antibody has heavy and light chains comprising the amino acid sequences of SEQ ID NO:46 and SEQ ID NO:64, respectively.
  • In one embodiment, a penta-specific antibody has heavy and light chains comprising polypeptides which are at least 90%, 95%, 98% or 99% identical to the amino acid sequences of SEQ ID NO:46, and SEQ ID NO:48, 60, 62, or 64, respectively.
  • In one embodiment, the present invention relates to a recombinant eukaryotic or a prokaryotic host cell which produces a penta-specific antibody having heavy or light chain comprising the amino acid sequence of SEQ ID NO: 46, or SEQ ID NO:48, 60, 62, or 64, respectively.
  • In one embodiment, the present invention relates to a recombinant eukaryotic or a prokaryotic host cell which produces a penta-specific antibody having heavy and light chains comprising the amino acid sequences of SEQ ID NO: 46, and SEQ ID NO:48, 60, 62, or 64, respectively.
  • In one embodiment, the present invention relates to a recombinant eukaryotic or a prokaryotic host cell which produces a penta-specific antibody having heavy or light chain comprising the amino acid sequence which is at least 90%, 95%, 98% or 99% identical to the amino acid sequence of SEQ ID NO: 46, or SEQ ID NO:48, 60, 62, or 64, respectively.
  • In one embodiment, the present invention relates to a recombinant eukaryotic or a prokaryotic host cell which produces a penta-specific antibody having heavy and light chains comprising the amino acid sequences which are at least 90%, 95%, 98% or 99% identical to the amino acid sequences of SEQ ID NO: 46, and SEQ ID NO:48, 60, 62, or 64, respectively.
  • In one embodiment, the present invention relates to an expression vector comprising polynucleotide sequences comprising sequences of SEQ ID NO:11, and SEQ ID NO:47, 59, 61, or 63, respectively.
  • In one embodiment, the present invention relates to an expression vector comprising polynucleotide sequences comprising sequences which are at least 90%, 95%, 98% or 99% identical to sequences of SEQ ID NO:11, and SEQ ID NO:47, 59, 61, or 63, respectively.
  • In one embodiment, the present invention relates to an expression vector comprising a polynucleotide sequence comprising a sequence of SEQ ID NO:11, 47, 59, 61, or 63.
  • In one embodiment, the present invention relates to an expression vector comprising a polynucleotide sequence comprising a sequence which is at least 90%, 95%, 98% or 99% identical to a sequence of SEQ ID NO:11, 47, 59, 61, or 63, respectively.
  • In one embodiment, the present invention relates to an expression vector comprising polynucleotide sequences which encode a penta-specific antibody comprising heavy and light chains comprising the amino acid sequences of SEQ ID NO:46, and SEQ ID NO:48, 60, 62, or 64, respectively.
  • In one embodiment, the present invention relates to an expression vector comprising polynucleotide sequences which encode a penta-specific antibody comprising heavy and light chains comprising the amino acid sequences which are at least 90%, 95%, 98% or 99% identical to the amino acid sequences of SEQ ID NO:46, and SEQ ID NO:48, 60, 62, or 64, respectively.
  • In one embodiment, the present invention relates to an expression vector comprising a polynucleotide sequence which encode a polypeptide comprising the amino acid sequence of SEQ ID NO:46, 48, 60, 62, or 64.
  • In one embodiment, the present invention relates to an expression vector comprising a polynucleotide sequence which encode a polypeptide comprising an amino acid sequence which is at least 90%, 95%, 98% or 99% identical to the amino acid sequences of SEQ ID NO:46, 48, 60, 62, or 64.
  • In one embodiment the present invention relates to a process for producing a penta-specific antibody (immunoglobulin) in a single host cell, comprising the steps of:
      • (i) transforming said single host cell with a first DNA sequence encoding a heavy chain comprising polypeptide of SEQ ID NO: 46; and a second DNA sequence encoding a light chain comprising a polypeptide of SEQ ID NO: 48, 60, 62, or 64; and
      • (ii) expressing said first DNA sequence and said second DNA sequence so that said immunoglobulin heavy and light chains are produced as separate molecules in said transformed single host cell;
      • furthermore, this process can be carried out such that said first and second DNA sequences are present in different vectors or said first and second DNA sequences are present in a single vector.
  • In one embodiment the present invention relates to a process for producing a penta-specific antibody (immunoglobulin) in a single host cell, comprising the steps of:
      • (i) transforming said single host cell with a first DNA sequence encoding at least the variable domain of the immunoglobulin heavy chain comprising CDR domains of SEQ ID NOs: 13, 14, and 15; and a second DNA sequence encoding at least the variable domain of the immunoglobulin light chain comprising CDR domains of SEQ ID NOs: 16, 17, and 18; and
      • (ii) expressing said first DNA sequence and said second DNA sequence so that said immunoglobulin heavy and light chains are produced as separate molecules in said transformed single host cell;
        furthermore, this process can be carried out such that said first and second DNA sequences are present in different vectors or said first and second DNA sequences are present in a single vector.
  • In one embodiment the present invention relates to a process for producing a penta-specific antibody (immunoglobulin) in a single host cell, comprising the steps of:
      • (i) transforming said single host cell with a first DNA sequence encoding at least the variable domain of the immunoglobulin heavy chain comprising CDR domains of SEQ ID NOs: 19, 20, and 21; and a second DNA sequence encoding at least the variable domain of the immunoglobulin light chain comprising CDR domains of SEQ ID NOs: 22, 23, and 24; and
      • (ii) expressing said first DNA sequence and said second DNA sequence so that said immunoglobulin heavy and light chains are produced as separate molecules in said transformed single host cell;
        this process can be carried out such that said first and second DNA sequences are present in different vectors or said first and second DNA sequences are present in a single vector.
  • In one embodiment the present invention relates to an antibody that fully or partially blocks the binding of any one of the aforementioned penta-specific antibody to human IL-8, Gro-alpha, Gro-beta, Gro-gamma, ENA-78, and GCP-2 in an immunoassay, such as ELISA assay. In one embodiment, partial blocking occurs when the antibody blocks the binding of the penta-specific antibody by more than 10%, 20%, 40% or 50%.
  • In one embodiment the present invention relates to an antibody that competes with the binding of any of the aforementioned penta-specific antibody to human IL-8, Gro-alpha, Gro-beta, Gro-gamma, ENA-78, and GCP-2.
  • In one embodiment the present invention relates to an antibody that fully or partially blocks the binding of any one of the aforementioned penta-specific antibody to epitope of KELRCQCIKTYSKP (SEQ ID NO: 54) in human IL-8 in an immunoassay, such as ELISA assay. In one embodiment, partial blocking occurs when the antibody blocks the binding of the penta-specific antibody by more than 10%, 20%, 40% or 50%.
  • In one embodiment the present invention relates to an antibody that competes with the binding of any one of the aforementioned penta-specific antibody to epitope of KELRCQCIKTYSKP (SEQ ID NO: 54) in human IL-8.
  • In one embodiment, the present invention relates to a composition comprising an aforementioned penta-specific antibody and a pharmaceutically acceptable carrier.
  • In one embodiment, the present invention relates to a method of treating or preventing in a mammal COPD, osteoarthritis, rheumatoid arthritis, erosive arthritis, asthma, atherosclerosis, inflammatory bowel disease, psoriasis, transplant rejection, gout, cancer, acute lung injury, acute lung disease, sepsis, ARDS, peripheral artery disease, systemic sclerosis, neonatal respiratory distress syndrome, exacerbation of asthma and COPD, cystic fibrosis, diffuse panbronchiolitis, reperfusion injury, and/or endometriosis comprising administering an effective amount of an aforementioned penta-specific antibody to said mammal.
  • In one embodiment the present invention relates to an aforementioned penta-specific antibody for use in the treatment of diseases or disorders characterised by elevated or unbalanced level of one or more of human IL-8, Gro-alpha, Gro-beta, Gro-gamma, GCP-2 and ENA-78, particularly COPD, osteoarthritis, rheumatoid arthritis, erosive arthritis, asthma, atherosclerosis, inflammatory bowel disease, psoriasis, transplant rejection, gout, cancer, acute lung injury, acute lung disease, sepsis, ARDS, peripheral artery disease, systemic sclerosis, neonatal respiratory distress syndrome, exacerbation of asthma and COPD, cystic fibrosis, diffuse panbronchiolitis, reperfusion injury, or endometriosis.
  • In one aspect, the present invention relates to an aforementioned penta-specific antibody for use in preventing and/or treating COPD, osteoarthritis, rheumatoid arthritis, erosive arthritis, asthma, atherosclerosis, inflammatory bowel disease, psoriasis, transplant rejection, gout, cancer, acute lung injury, acute lung disease, sepsis, ARDS, peripheral artery disease, systemic sclerosis, neonatal respiratory distress syndrome, exacerbation of asthma and COPD, cystic fibrosis, diffuse panbronchiolitis, reperfusion injury, and/or endometriosis in a mammal.
  • In one aspect, the present invention relates to use of an aforementioned penta-specific antibody in the manufacture of a medicament for use in preventing and/or treating COPD, osteoarthritis, rheumatoid arthritis, erosive arthritis, asthma, atherosclerosis, inflammatory bowel disease, psoriasis, transplant rejection, gout, cancer, acute lung injury, acute lung disease, sepsis, ARDS, peripheral artery disease, systemic sclerosis, neonatal respiratory distress syndrome, exacerbation of asthma and COPD, cystic fibrosis, diffuse panbronchiolitis, reperfusion injury, and/or endometriosis in a mammal.
  • In one aspect, the present invention relates to use of an aforementioned penta-specific antibody in the manufacture of a medicament for preventing and/or treating COPD, osteoarthritis, rheumatoid arthritis, erosive arthritis, asthma, atherosclerosis, inflammatory bowel disease, psoriasis, transplant rejection, gout, cancer, acute lung injury, acute lung disease, sepsis, ARDS, peripheral artery disease, systemic sclerosis, neonatal respiratory distress syndrome, exacerbation of asthma and COPD, cystic fibrosis, diffuse panbronchiolitis, reperfusion injury, and/or endometriosis in a mammal.
  • In one embodiment, above mammal is human.
  • DESCRIPTION OF FIGURES
  • FIG. 1 depicts an exemplary set of MAPs to generate a penta-specific antibody. For avoidance of doubt, five MAP peptide units are depicted. Each unit contains one identical amino acid sequence selected from linear peptides of SEQ ID NO: 49-53.
  • FIG. 2 shows flow cytometric data comparing neutrophils present in BAL samples. Sessions 1, 2 and 3 are presented. The black solid bars show the baselines 5 day pre-LPS challenge. The grey solid bars depict the BAL data 6-hours post LPS challenge. The Chimera Antibody (HcLc) treatment significantly and dose dependently inhibited neutrophil infiltration. The grey hatched bars represent 24-hour data. (n=6, *p≦0.05, †p≦0.01, error bars represent standard deviation).
  • FIG. 3 shows hematological analysis of total circulating neutrophils (cell count). IV injection of 1 (open squares, □) and 10 mg/kg (closed circles, ) of the Chimera Antibody (HcLc) prevented inhaled LPS stimulation of circulating neutrophils. Both 1 and 10 mg/kg treatments are significantly reduced compared to the vehicle NHP (†) and 10 mg/kg treatment was also significantly reduced compared to the 1 mg/kg dose (*). Data are represented as total count per ul of blood (n=6, *p≦0.05 vs. 1 mg/kg, †p≦0.01 vs. vehicle, error bars represent standard deviation).
  • FIG. 4 shows inhibition human IL-8 stimulated neutrophil activation (increased CD11b surface expression). Various concentrations of the humanized penta-specific antibodies were pre-incubated with 10 nM hIL-8 prior to addition to purified human neutrophils. Data are expressed as mean values of 4 different donors (n=4). Bars represent standard error. All samples are compared to purified neutrophils stimulated with hIL-8 only (no mAb prior to addition to purified neutrophils). Humanized construct H0L10 and H0M0 are more effective at inhibiting hIL-8 stimulated CD11b surface expression.
  • FIG. 5A shows mouse 656.35 mAb binding to target human chemokines.
  • FIG. 5B shows the Chimera Antibody (HcLc) mAb binding to human target chemokines.
  • FIG. 5C shows humanized mAbs binding to human target chemokines.
  • DETAILED DESCRIPTION OF THE INVENTION
  • An “isolated penta-specific antibody” or simply “penta-specific antibody”, as used herein, is intended to refer to an antibody that binds to and therefore cross reacts with human IL-8, Gro-alpha, Gro-beta, Gro-gamma, and ENA-78, and is substantially free of other antibodies having different antigenic specificities, and furthermore, is a single composition of matter. For avoidance of doubt, the penta-specific antibody of the present invention need not bind solely to human IL-8, Gro-alpha, Gro-beta, Gro-gamma, and ENA-78 antigens, but it may also happen to bind to other related proteins, such as human GCP-2, or may even bind to non-human orthologues of IL-8, Gro-alpha, Gro-beta, Gro-gamma, ENA-78, and GCP-2; in other words the penta-specific antibody of the present invention minimally binds to human IL-8, Gro-alpha, Gro-beta, Gro-gamma, and ENA-78. Moreover, an isolated penta-specific antibody is substantially free of other cellular material and/or chemicals. A penta-specific antibody, as used in this context, also includes antigen binding fragments and/or derivatives having the binding characteristics of their “parent” penta-specific antibody. The penta-specific antibody of the present invention may comprise two identical heavy chains and two identical light chains, forming the typical, bilaterally symmetric immunoglobulin molecule comprised of two heterodimers which are each comprised of a heavy chain and a light chain. Accordingly, the penta-specific antibody of the present invention may comprise two copies of the same antigen binding domain formed by the association of one heavy chain with one light chain. The penta-specific antibody of the present invention, though having only one kind of binding domain is still able to bind to and therefore cross react with human IL-8, Gro-alpha, Gro-beta, Gro-gamma, and ENA-78.
  • As used herein, “antibody” is also referred to as “immunoglobulin”.
  • As used herein, “an antibody that cross reacts with” means the antibody binds not only to one antigen but binds to other antigens as well.
  • “Neutralizing,” as used herein is intended to refer to a partial or full inhibition of at least one biological activities of human IL-8, Gro-alpha, Gro-beta, Gro-gamma, GCP-2, or ENA-78. For example, one of the biological activities of human IL-8, Gro-alpha, Gro-beta, Gro-gamma, GCP-2, or ENA-78 is its ability to induce neutrophil chemotaxis.
  • One way of measuring the binding kinetics of an antibody is by surface plasmon resonance. The term “surface plasmon resonance”, as used herein, refers to an optical phenomenon that allows for the analysis of real-time biospecific interactions by detection of alterations in protein concentrations within a biosensor matrix, for example using the BIAcore system (Pharmacia Biosensor AB, Uppsala, Sweden and Piscataway, N.J.). For further descriptions, see Jonsson, U., et al. (1993) Ann. Biol. Clin. 51:19-26; Jonsson, U., et al. (1991) Biotechniques 11:620-627; Johnsson, B., et al. (1995) J. Mol. Recognit. 8:125-131; and Johnnson, B., et al. (1991) Anal. Biochem. 198:268-277. The term “epitope” means a protein determinant capable of specific binding to an antibody. Epitopes usually consist of chemically active surface groupings of molecules such as amino acids or sugar side chains and usually have specific three dimensional structural characteristics, as well as specific charge characteristics.
  • A “monoclonal antibody” or mAb (as opposed to polyclonal antibody) as used herein is intended to refer to a preparation of antibody molecules of single molecular composition. For example, a murine derived monoclonal antibody (mouse monoclonal antibody) can be prepared by hybridoma technology, such as the standard Kohler and Milstein hybridoma methodology.
  • Antibody-producing cells can be obtained from the subject and used to prepare monoclonal antibodies by standard techniques, such as the hybridoma technique originally described by Kohler and Milstein (1975, Nature 256:495-497) (see also, Brown et al. (1981) J. Immunol 127:539-46; Brown et al. (1980) J Biol Chem 255:4980-83; Yeh et al. (1976) PNAS 76:2927-31; and Yeh et al. (1982) Int. J. Cancer 29:269-75). The technology for producing monoclonal antibody hybridomas is well known (see generally R. H. Kenneth, in Monoclonal Antibodies: A New Dimension In Biological Analyses, Plenum Publishing Corp., New York, N.Y. (1980); E. A. Lerner (1981) Yale J. Biol. Med., 54:387-402; M. L. Gefter et al. (1977) Somatic Cell Genet., 3:231-36).
  • The term “transfectoma”, as used herein, includes recombinant eukaryotic host cell expressing the antibody, such as CHO cells, NS/0 cells, HEK293 cells, plant cells, or fungi, including yeast cells.
  • As used herein, “specific” binding refers to antibody binding to a predetermined antigen. Typically, the antibody binds with an equilibrium constant, KD, corresponding to about 1×10−7 M or less, and binds to the predetermined antigen with an affinity corresponding to a KD that is at least two orders of magnitude lower than its affinity for binding to a non-specific antigen (e.g., BSA, casein) other than the predetermined antigen or a closely-related antigen. The phrases “an antibody recognizing an antigen” and “an antibody specific for an antigen” are used interchangeably herein with the term “an antibody which binds specifically to an antigen”.
  • As used herein, the term “kd” (sec-1), as used herein, is intended to refer to the dissociation rate constant of a particular antibody-antigen interaction.
  • The term “ka” (M×sec-1), as used herein, is intended to refer to the association rate constant of a particular antibody-antigen interaction.
  • The term “KD” (M), as used herein, is intended to refer to the equilibrium constant of a particular antibody-antigen interaction and is obtained by dividing the kd by the ka.
  • “Conservative sequence modifications” for nucleotide and amino acid sequence modifications means changes which do not significantly affect or alter the binding characteristics of the antibody encoded by the nucleotide sequence or containing the amino acid sequence. Such conservative sequence modifications include nucleotide and amino acid substitutions, additions and deletions. Modifications can be introduced into the sequences by standard techniques known in the art, such as site-directed mutagenesis and PCR-mediated mutagenesis. Conservative amino acid substitutions include ones in which the amino acid residue is replaced with an amino acid residue having a similar side chain. Families of amino acid residues having similar side chains have been defined in the art. These families include amino acids with basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine, tryptophan), nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine), beta-branched side chains (e.g., threonine, valine, isoleucine) and aromatic side chains (e.g., tyrosine, phenylalanine, tryptophan, histidine). Thus, a predicted nonessential amino acid residue in an antibody for which sequence is specifically disclosed is preferably replaced with another amino acid residue from the same side chain family. Thus in one aspect, the penta-specific antibody of the present invention includes all the conservative sequence modifications of the specifically disclosed amino acid sequences.
  • The present invention also encompasses “derivatives” of the amino acid sequences as specifically disclosed, wherein one or more of the amino acid residues have been derivatized, e.g., by acylation or glycosylation, without significantly affecting or altering the binding characteristics of the antibody containing the amino acid sequences.
  • For nucleic acids, the term “substantial identity” indicates that two nucleic acids, or designated sequences thereof, when optimally aligned and compared, are identical, with appropriate nucleotide insertions or deletions, in at least about 80% of the nucleotides, usually at least about 90% to 95%, and more preferably at least about 98% to 99.5% of the nucleotides. Alternatively, substantial identity when the segments will hybridize under selective hybridization conditions, to the complement of the strand.
  • For nucleotide and amino acid sequences, the term “identity” indicates the degree of identity between two nucleic acid or amino acid sequences when optimally aligned and compared with appropriate insertions or deletions. Alternatively, substantial identity exists when the DNA segments will hybridize under selective hybridization conditions, to the complement of the strand.
  • The percent identity between two sequences is a function of the number of identical positions shared by the sequences (i.e., % identity=# of identical positions/total # of positions times 100), taking into account the number of gaps, and the length of each gap, which need to be introduced for optimal alignment of the two sequences. The comparison of sequences and determination of percent identity between two sequences can be accomplished using a mathematical algorithm, as described in the non-limiting examples below.
  • The percent identity between two nucleotide or polypeptide sequences can be determined using the GAP program in the GCG software package (available at http://www.gcg.com), using a NWSgapdna.CMP matrix and a gap weight of 40, 50, 60, 70, or 80 and a length weight of 1, 2, 3, 4, 5, or 6. The percent identity between two nucleotide or amino acid sequences can also be determined using the algorithm of E. Meyers and W. Miller (Comput. Appl. Biosci., 4:11-17 (1988)) which has been incorporated into the ALIGN program (version 2.0), using a PAM120 weight residue table, a gap length penalty of 12 and a gap penalty of 4. In addition, the percent identity between two amino acid sequences can be determined using the Needleman and Wunsch (J. Mol. Biol. 48:444-453 (1970)) algorithm which has been incorporated into the GAP program in the GCG software package (available at http://www.gcg.com), using either a Blossum 62 matrix or a PAM250 matrix, and a gap weight of 16, 14, 12, 10, 8, 6, or 4 and a length weight of 1, 2, 3, 4, 5, or 6.
  • A nucleic acid is “operably linked” when it is placed into a functional relationship with another nucleic acid sequence. For instance, a promoter or enhancer is operably linked to a coding sequence if it affects the transcription of the sequence. With respect to transcription of regulatory sequences, operably linked means that the DNA sequences being linked are contiguous and, where necessary to join two protein coding regions, contiguous and in reading frame. For switch sequences, operably linked indicates that the sequences are capable of effecting switch recombination.
  • The term “vector,” as used herein, is intended to refer to a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked. One type of vector is a “plasmid”, which refers to a circular double stranded DNA loop into which additional DNA segments may be ligated. Another type of vector is a viral vector, wherein additional DNA segments may be ligated into the viral genome. Certain vectors are capable of autonomous replication in a host cell into which they are introduced (e.g., bacterial vectors having a bacterial origin of replication and episomal mammalian vectors). Other vectors (e.g., non-episomal mammalian vectors) can be integrated into the genome of a host cell upon introduction into the host cell, and thereby are replicated along with the host genome. Moreover, certain vectors are capable of directing the expression of genes to which they are operatively linked. Such vectors are referred to herein as “recombinant expression vectors” (or simply, “expression vectors”). In general, expression vectors of utility in recombinant DNA techniques are often in the form of plasmids. In the present specification, “plasmid” and “vector” may be used interchangeably as the plasmid is the most commonly used form of vector. However, the invention is intended to include such other forms of expression vectors, such as viral vectors (e.g., replication defective retroviruses, adenoviruses and adeno-associated viruses), which serve equivalent functions.
  • The term “recombinant host cell” (or simply “host cell”), as used herein, is intended to refer to a cell into which a recombinant expression vector has been introduced. It should be understood that such terms are intended to refer not only to the particular subject cell but to the progeny of such a cell. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term “host cell” as used herein. Recombinant host cells include, for example, transfectomas, such as CHO cells, NS/0 cells, and lymphocytic cells.
  • As used herein, the term “subject” includes any human or non-human animal. The term “non-human animal” includes all vertebrates, e.g., mammals and non-mammals, such as non-human primates, sheep, dog, cow, chickens, amphibians, reptiles, etc.
  • 1. Antibody Structures Intact Antibodies
  • Intact antibodies are usually heteromultimeric glycoproteins comprising at least two heavy and two light chains. Aside from IgM, intact antibodies are heterotetrameric glycoproteins of approximately 150 Kda, composed of two identical light (L) chains and two identical heavy (H) chains. Typically, each light chain is linked to a heavy chain by one covalent disulfide bond while the number of disulfide linkages between the heavy chains of different immunoglobulin isotypes varies. Each heavy and light chain also has intrachain disulfide bridges. Each heavy chain has at one end a variable domain (VH) followed by a number of constant regions. Each light chain has a variable domain (VL) and a constant region at its other end; the constant region of the light chain is aligned with the first constant region of the heavy chain and the light chain variable domain is aligned with the variable domain of the heavy chain. The light chains of antibodies from most vertebrate species can be assigned to one of two types called Kappa and Lambda based on the amino acid sequence of the constant region. Depending on the amino acid sequence of the constant region of their heavy chains, human antibodies can be assigned to five different classes, IgA, IgD, IgE, IgG and IgM. IgG and IgA can be further subdivided into subclasses, IgG1, IgG2, IgG3 and IgG4; and IgA1 and IgA2. Species variants exist with mouse and rat having at least IgG2a, IgG2b. The variable domain of the antibody confers binding specificity upon the antibody with certain regions displaying particular variability called complementarity determining regions (CDRs). The more conserved portions of the variable region are called framework regions (FR). The variable domains of intact heavy and light chains each comprise four FR connected by three CDRs. The CDRs in each chain are held together in close proximity by the FR regions and with the CDRs from the other chain contribute to the formation of the antigen binding site of antibodies. The constant regions are not directly involved in the binding of the antibody to the antigen but exhibit various effector functions such as participation in antibody dependent cell-mediated cytotoxicity (ADCC), phagocytosis via binding to Fcγ receptor, half-life/clearance rate via neonatal Fc receptor (FcRn) and complement dependent cytotoxicity via the C1q component of the complement cascade. The human IgG2 constant region lacks the ability to activate complement by the classical pathway or to mediate antibody-dependent cellular cytotoxicity. The IgG4 constant region lacks the ability to activate complement by the classical pathway and mediates antibody-dependent cellular cytotoxicity only weakly. Antibodies essentially lacking these effector functions may be termed ‘non-lytic’ antibodies.
  • Human Antibodies
  • Human antibodies may be produced by a number of methods known to those of skill in the art. Human antibodies can be made by the hybridoma method using human myeloma or mouse-human heteromyeloma cells lines see Kozbor J. Immunol 133, 3001, (1984) and Brodeur, Monoclonal Antibody Production Techniques and Applications, pp 51-63 (Marcel Dekker Inc, 1987). Alternative methods include the use of phage libraries or transgenic mice both of which utilize human V region repertories (see Winter G, (1994), Annu. Rev. Immunol 12, 433-455, Green L L (1999), J. Immunol. methods 231, 11-23).
  • Several strains of transgenic mice are now available wherein their mouse immunoglobulin loci has been replaced with human immunoglobulin gene segments (see Tomizuka K, (2000) PNAS 97, 722-727; Fishwild D. M (1996) Nature Biotechnol. 14, 845-851, Mendez M J, 1997, Nature Genetics, 15, 146-156). Upon antigen challenge such mice are capable of producing a repertoire of human antibodies from which antibodies of interest can be selected.
  • Of particular note is the Trimera™ system (see Eren R et al, (1998) Immunology 93:154-161) where human lymphocytes are transplanted into irradiated mice, the Selected Lymphocyte Antibody System (SLAM, see Babcook et al, PNAS (1996) 93:7843-7848) where human (or other species) lymphocytes are effectively put through a massive pooled in vitro antibody generation procedure followed by deconvulated, limiting dilution and selection procedure and the Xenomouse II™ (Abgenix Inc). An alternative approach is available from Morphotek Inc using the Morphodoma™ technology.
  • Phage display technology can be used to produce human antibodies (and fragments thereof), see McCafferty; Nature, 348, 552-553 (1990) and Griffiths A D et al (1994) EMBO 13:3245-3260. According to this technique antibody V domain genes are cloned in frame into either a major or minor coat of protein gene of a filamentous bacteriophage such as M13 or fd and displayed (usually with the aid of a helper phage) as functional antibody fragments on the surface of the phage particle. Selections based on the functional properties of the antibody result in selection of the gene encoding the antibody exhibiting those properties. The phage display technique can be used to select antigen specific antibodies from libraries made from human B cells taken from individuals afflicted with a disease or disorder described above or alternatively from unimmunized human donors (see Marks; J. Mol. Bio. 222, 581-597, 1991). Where an intact human antibody is desired comprising a Fc domain it is necessary to reclone the phage displayed derived fragment into a mammalian expression vectors comprising the desired constant regions and establishing stable expressing cell lines.
  • The technique of affinity maturation (Marks; Bio/technol 10, 779-783 (1992)) may be used to improve binding affinity wherein the affinity of the primary human antibody is improved by sequentially replacing the H and L chain V regions with naturally occurring variants and selecting on the basis of improved binding affinities. Variants of this technique such as “epitope imprinting” are now also available see WO 93/06213. See also Waterhouse; Nucl. Acids Res 21, 2265-2266 (1993).
  • Chimaeric and Humanised Antibodies
  • The use of intact non-human antibodies in the treatment of human diseases or disorders carries with it the now well established problems of potential immunogenicity especially upon repeated administration of the antibody that is the immune system of the patient may recognise the non-human intact antibody as non-self and mount a neutralising response. In addition to developing fully human antibodies (see above) various techniques have been developed over the years to overcome these problems and generally involve reducing the composition of non-human amino acid sequences in the intact therapeutic antibody whilst retaining the relative ease in obtaining non-human antibodies from an immunised animal e.g. mouse, rat or rabbit. Broadly two approaches have been used to achieve this. The first are chimaeric antibodies, which generally comprise a non-human (e.g. rodent such as mouse) variable domain fused to a human constant region. Because the antigen-binding site of an antibody is localised within the variable regions the chimaeric antibody retains its binding affinity for the antigen but acquires the effector functions of the human constant region and are therefore able to perform effector functions such as described supra. Chimaeric antibodies are typically produced using recombinant DNA methods. DNA encoding the antibodies (e.g. cDNA) is isolated and sequenced using conventional procedures (e.g. by using oligonucleotide probes that are capable of binding specifically to genes encoding the H and L chain variable regions of the antibody of the invention. Hybridoma cells serve as a typical source of such DNA. Once isolated, the DNA is placed into expression vectors which are then transfected into host cells such as E. Coli, COS cells, CHO cells or myeloma cells that do not otherwise produce immunoglobulin protein to obtain synthesis of the antibody. The DNA may be modified by substituting the coding sequence for human L and H chains for the corresponding non-human (e.g. murine) H and L constant regions see e.g. Morrison; PNAS 81, 6851 (1984). Thus another embodiment of the invention there is provided a chimaeric antibody comprising a VH domain having the sequence: SEQ ID No:2, 6, or 10 and a VL domain having the sequence: SEQ ID No: 4, 8, or 12 fused to a human constant region (which maybe of a IgG isotype e.g. IgG1).
  • The second approach involves the generation of humanised antibodies wherein the non-human content of the antibody is reduced by humanizing the variable regions. Two techniques for humanisation have gained popularity. The first is humanisation by CDR grafting. CDRs build loops close to the antibody's N-terminus where they form a surface mounted in a scaffold provided by the framework regions. Antigen-binding specificity of the antibody is mainly defined by the topography and by the chemical characteristics of its CDR surface. These features are in turn determined by the conformation of the individual CDRs, by the relative disposition of the CDRs, and by the nature and disposition of the side chains of the residues comprising the CDRs. A large decrease in immunogenicity can be achieved by grafting only the CDRs of a non-human (e.g. murine) antibodies (“donor” antibodies) onto a suitable human framework (“acceptor framework”) and constant regions (see Jones et al (1986) Nature 321, 522-525 and Verhoeyen M et al (1988) Science 239, 1534-1536). However, CDR grafting per se may not result in the complete retention of antigen-binding properties and it is frequently found that some framework residues of the donor antibody need to be preserved (sometimes referred to as “backmutations”) in the humanised molecule if significant antigen-binding affinity is to be recovered (see Queen C et al (1989) PNAS 86, 10,029-10,033, Co, M et al (1991) Nature 351, 501-502). In this case, human V regions showing the greatest sequence homology (typically 60% or greater) to the non-human donor antibody maybe chosen from a database in order to provide the human framework (FR). The selection of human FRs can be made either from human consensus or individual human antibodies. Where necessary key residues from the donor antibody are substituted into the human acceptor framework to preserve CDR conformations. Computer modelling of the antibody maybe used to help identify such structurally important residues, see WO99/48523.
  • Alternatively, humanisation maybe achieved by a process of “veneering”. A statistical analysis of unique human and murine immunoglobulin heavy and light chain variable regions revealed that the precise patterns of exposed residues are different in human and murine antibodies, and most individual surface positions have a strong preference for a small number of different residues (see Padlan E. A. et al; (1991) Mol. Immunol. 28, 489-498 and Pedersen J. T. et al (1994) J. Mol. Biol. 235; 959-973). Therefore it is possible to reduce the immunogenicity of a non-human Fv by replacing exposed residues in its framework regions that differ from those usually found in human antibodies. Because protein antigenicity can be correlated with surface accessibility, replacement of the surface residues may be sufficient to render the mouse variable region “invisible” to the human immune system (see also Mark G. E. et al (1994) in Handbook of Experimental Pharmacology vol. 113: The pharmacology of monoclonal Antibodies, Springer-Verlag, pp 105-134). This procedure of humanisation is referred to as “veneering” because only the surface of the antibody is altered, the supporting residues remain undisturbed. A further alternative approach is set out in WO04/006955. Further alternative approaches include that set out in WO04/006955 and the procedure of Humaneering™ (Kalobios) which makes use of bacterial expression systems and produces antibodies that are close to human germline in sequence (Alfenito-M Advancing Protein Therapeutics January 2007, San Diego, Calif.). Another, approach to humanisation involves selecting human acceptor frameworks on the basis of structural similarity of the human CDR regions to those of the donor mouse antibody CDR regions rather than on homology between other regions of the antibody such as framework regions. This process is also known as Superhumanisation™ (Evogenix Inc.; Hwang et al (2005) Methods 36:35-42).
  • It will be apparent to those skilled in the art that the term “derived” is intended to define not only the source in the sense of it being the physical origin for the material but also to define material which is structurally identical to the material but which does not originate from the reference source. Thus “residues found in the donor antibody” need not necessarily have been purified from the donor antibody.
  • Antibody Fragments
  • In certain embodiments of the invention there is provided therapeutic antibody which is an antigen binding fragment. Such fragments may be functional antigen binding fragments of intact and/or humanised and/or chimaeric antibodies such as Fab, Fd, Fab′, F(ab′)2, Fv, ScFv fragments of the antibodies described supra. Fragments lacking the constant region lack the ability to activate complement by the classical pathway or to mediate antibody-dependent cellular cytotoxicity. Traditionally such fragments are produced by the proteolytic digestion of intact antibodies by e.g. papain digestion (see for example, WO 94/29348) but may be produced directly from recombinantly transformed host cells. For the production of ScFv, see Bird et al; (1988) Science, 242, 423-426. In addition, antibody fragments may be produced using a variety of engineering techniques as described below.
  • Fv fragments appear to have lower interaction energy of their two chains than Fab fragments. To stabilise the association of the VH and VL domains, they have been linked with peptides (Bird et al, (1988) Science 242, 423-426, Huston et al, PNAS, 85, 5879-5883), disulphide bridges (Glockshuber et al, (1990) Biochemistry, 29, 1362-1367) and “knob in hole” mutations (Zhu et al (1997), Protein Sci., 6, 781-788). ScFv fragments can be produced by methods well known to those skilled in the art see Whitlow et al (1991) Methods companion Methods Enzymol, 2, 97-105 and Huston et al (1993) Int. Rev. Immunol 10, 195-217. ScFv may be produced in bacterial cells such as E. Coli but are more typically produced in eukaryotic cells. One disadvantage of ScFv is the monovalency of the product, which precludes an increased avidity due to polyvalent binding, and their short half-life. Attempts to overcome these problems include bivalent (ScFv′)2 produced from ScFV containing an additional C terminal cysteine by chemical coupling (Adams et al (1993) Can. Res 53, 4026-4034 and McCartney et al (1995) Protein Eng. 8, 301-314) or by spontaneous site-specific dimerization of ScFv containing an unpaired C terminal cysteine residue (see Kipriyanov et al (1995) Cell. Biophys 26, 187-204). Alternatively, ScFv can be forced to form multimers by shortening the peptide linker to between 3 to 12 residues to form “diabodies”, see Holliger et al PNAS (1993), 90, 6444-6448. Reducing the linker still further can result in ScFV trimers (“triabodies”, see Kortt et al (1997) Protein Eng, 10, 423-433) and tetramers (“tetrabodies”, see Le Gall et al (1999) FEBS Lett, 453, 164-168). Construction of bivalent ScFV molecules can also be achieved by genetic fusion with protein dimerizing motifs to form “miniantibodies” (see Pack et al (1992) Biochemistry 31, 1579-1584) and “minibodies” (see Hu et al (1996), Cancer Res. 56, 3055-3061). ScFv-Sc-Fv tandems ((ScFV)2) may also be produced by linking two ScFv units by a third peptide linker, see Kurucz et al (1995) J. Immol. 154, 4576-4582. Bispecific diabodies can be produced through the noncovalent association of two single chain fusion products consisting of VH domain from one antibody connected by a short linker to the VL domain of another antibody, see Kipriyanov et al (1998), Int. J. Can 77, 763-772. The stability of such bispecific diabodies can be enhanced by the introduction of disulphide bridges or “knob in hole” mutations as described supra or by the formation of single chain diabodies (ScDb) wherein two hybrid ScFv fragments are connected through a peptide linker see Kontermann et al (1999) J. Immunol. Methods 226 179-188. Tetravalent bispecific molecules are available by e.g. fusing a ScFv fragment to the CH3 domain of an IgG molecule or to a Fab fragment through the hinge region see Coloma et al (1997) Nature Biotechnol. 15, 159-163. Alternatively, tetravalent bispecific molecules have been created by the fusion of bispecific single chain diabodies (see Alt et al, (1999) FEBS Lett 454, 90-94. Smaller tetravalent bispecific molecules can also be formed by the dimerization of either ScFv-ScFv tandems with a linker containing a helix-loop-helix motif (DiBi miniantibodies, see Muller et al (1998) FEBS Lett 432, 45-49) or a single chain molecule comprising four antibody variable domains (VH and VL) in an orientation preventing intramolecular pairing (tandem diabody, see Kipriyanov et al, (1999) J. Mol. Biol. 293, 41-56). Bispecific F(ab′)2 fragments can be created by chemical coupling of Fab′ fragments or by heterodimerization through leucine zippers (see Shalaby et al, (1992) J. Exp. Med. 175, 217-225 and Kostelny et al (1992), J. Immunol. 148, 1547-1553). Also available are so-called domain antibodies based on isolated VH or VL domains (Domantis Ltd.), see U.S. Pat. No. 6,248,516; U.S. Pat. No. 6,291,158; U.S. Pat. No. 6,172,197.
  • Heteroconjugate Antibodies
  • Heteroconjugate antibodies are derivatives which also form an embodiment of the present invention. Heteroconjugate antibodies are composed of two covalently joined antibodies formed using any convenient cross-linking methods. See U.S. Pat. No. 4,676,980.
  • Other Modifications.
  • Antibodies of the present invention may also incorporate any other modifications in the constant regions. For example glycosylation of antibodies at conserved positions in their constant regions is known to have a profound effect on antibody function, particularly effector functioning such as those described above, see for example, Boyd et al (1996), Mol. Immunol. 32, 1311-1318. Glycosylation variants of the therapeutic antibodies of the present invention wherein one or more carbohydrate moiety is added, substituted, deleted or modified are contemplated. Introduction of an asparagine-X-serine or asparagine-X-threonine motif creates a potential site for enzymatic attachment of carbonhydrate moieties and may therefore be used to manipulate the glycosylation of an antibody. In Raju et al (2001) Biochemistry 40, 8868-8876 the terminal sialyation of a TNFR-IgG immunoadhesin was increased through a process of regalactosylation and/or resialylation using beta-1,4-galactosyltransferace and/or alpha, 2,3 sialyltransferase. Increasing the terminal sialylation is believed to increase the half-life of the immunoglobulin. Antibodies, in common with most glycoproteins, are typically produced in nature as a mixture of glycoforms. This mixture is particularly apparent when antibodies are produced in eukaryotic, particularly mammalian cells. A variety of methods have been developed to manufacture defined glycoforms, see Zhang et al Science (2004), 303, 371, Sears et al, Science, (2001) 291, 2344, Wacker et al (2002) Science, 298 1790, Davis et al (2002) Chem. Rev. 102, 579, Hang et al (2001) Acc. Chem. Res 34, 727. Thus the invention concerns a plurality of therapeutic antibodies (which maybe of the IgG isotype, e.g. IgG1) as described herein comprising a defined number (e.g. 7 or less, for example 5 or less such as two or a single) glycoform(s) of said antibodies.
  • Derivatives according to the invention also include therapeutic antibodies of the invention coupled to a non-proteinaeous polymer such as polyethylene glycol (PEG), polypropylene glycol or polyoxyalkylene. Conjugation of proteins to PEG is an established technique for increasing half-life of proteins, as well as reducing antigenicity and immunogenicity of proteins. The use of PEGylation with different molecular weights and styles (linear or branched) has been investigated with intact antibodies as well as Fab′ fragments, see Koumenis I. L. et al (2000) Int. J. Pharmaceut. 198:83-95. A particular embodiment comprises an antigen-binding fragment of the invention without the effector functions of a) activation of complement by the classical pathway; and b) mediating antibody-dependent cellular cytotoxicity; (such as a Fab fragment or a scFv) coupled to PEG.
  • The interaction between the Fc region of an antibody and various Fc receptors (FcγR) is believed to mediate the effector functions of the antibody which include antibody-dependent cellular cytotoxicity (ADCC), fixation of complement, phagocytosis and half-life/clearance of the antibody. Various modifications to the Fc region of antibodies of the invention may be carried out depending on the desired effector property. In particular, human constant regions which essentially lack the functions of a) activation of complement by the classical pathway; and b) mediating antibody-dependent cellular cytotoxicity include the IgG4 constant region, the IgG2 constant region and IgG1 constant regions containing specific mutations as for example mutations at positions 234, 235, 236, 237, 297, 318, 320 and/or 322 disclosed in EP0307434 (WO8807089), EP 0629 240 (WO9317105) and WO 2004/014953. Mutations at residues 235 or 237 within the CH2 domain of the heavy chain constant region (Kabat numbering; EU Index system) have separately been described to reduce binding to FcγRI, FcγRII and FcγRIII binding and therefore reduce antibody-dependent cellular cytotoxicity (ADCC) (Duncan et al. Nature 1988, 332; 563-564; Lund et al. J. Immunol. 1991, 147; 2657-2662; Chappel et al. PNAS 1991, 88; 9036-9040; Burton and Woof, Adv. Immunol. 1992, 51; 1-84; Morgan et al., Immunology 1995, 86; 319-324; Hezareh et al., J. Virol. 2001, 75 (24); 12161-12168). Further, some reports have also described involvement of some of these residues in recruiting or mediating complement dependent cytotoxicity (CDC) (Morgan et al., 1995; Xu et al., Cell. Immunol. 2000; 200:16-26; Hezareh et al., J. Virol. 2001, 75 (24); 12161-12168). Residues 235 and 237 have therefore both been mutated to alanine residues (Brett et al. Immunology 1997, 91; 346-353; Bartholomew et al. Immunology 1995, 85; 41-48; and WO9958679) to reduce both complement mediated and FcγR-mediated effects. Antibodies comprising these constant regions may be termed ‘non-lytic’ antibodies.
  • One may incorporate a salvage receptor binding epitope into the antibody to increase serum half life see U.S. Pat. No. 5,739,277.
  • There are five currently recognised human Fcγ receptors, FcγR (I), FcγRIIa, FcγRIIb, FcγRIIIa and neonatal FcRn. Shields et al, (2001) J. Biol. Chem 276, 6591-6604 demonstrated that a common set of IgG1 residues is involved in binding all FcγRs, while FcγRII and FcγRIII utilize distinct sites outside of this common set. One group of IgG1 residues reduced binding to all FcγRs when altered to alanine: Pro-238, Asp-265, Asp-270, Asn-297 and Pro-239. All are in the IgG CH2 domain and clustered near the hinge joining CH1 and CH2. While FcγRI utilizes only the common set of IgG1 residues for binding, FcγRII and FcγRIII interact with distinct residues in addition to the common set. Alteration of some residues reduced binding only to FcγRII (e.g. Arg-292) or FcγRIII (e.g. Glu-293). Some variants showed improved binding to FcγRII or FcγRIII but did not affect binding to the other receptor (e.g. Ser-267Ala improved binding to FcγRII but binding to FcγRIII was unaffected). Other variants exhibited improved binding to FcγRII or FcγRIII with reduction in binding to the other receptor (e.g. Ser-298Ala improved binding to FcγRIII and reduced binding to FcγRII). For FcγRIIIa, the best binding IgG1 variants had combined alanine substitutions at Ser-298, Glu-333 and Lys-334. The neonatal FcRn receptor is believed to be involved in protecting IgG molecules from degradation and thus enhancing serum half life and the transcytosis across tissues (see Junghans R. P (1997) Immunol. Res 16. 29-57 and Ghetie et al (2000) Annu. Rev. Immunol. 18, 739-766). Human IgG1 residues determined to interact directly with human FcRn includes Ile253, Ser254, Lys288, Thr307, Gln311, Asn434 and His435.
  • The therapeutic antibody of the invention may incorporate any of the above constant region modifications.
  • 2. Production Methods
  • Antibodies of the present invention may be produced in transgenic organisms such as goats (see Pollock et al (1999), J. Immunol. Methods 231:147-157), chickens (see Morrow K J J (2000) Genet. Eng. News 20:1-55), mice (see Pollock et al ibid) or plants (see Doran P M, (2000) Curr. Opinion Biotechnol. 11, 199-204, Ma J K-C (1998), Nat. Med. 4; 601-606, Baez J et al, BioPharm (2000) 13: 50-54, Stoger E et al; (2000) Plant Mol. Biol. 42:583-590). Antibodies may also be produced by chemical synthesis. However, antibodies of the invention are typically produced using recombinant cell culturing technology well known to those skilled in the art. A polynucleotide encoding the antibody is isolated and inserted into a replicable vector such as a plasmid for further cloning (amplification) or expression in a host cell. One useful expression system is a glutamate synthetase system (such as sold by Lonza Biologics), particularly where the host cell is CHO or NS0 (see below). Polynucleotide encoding the antibody is readily isolated and sequenced using conventional procedures (e.g. oligonucleotide probes). Vectors that may be used include plasmid, virus, phage, transposons, minichromsomes of which plasmids are a typical embodiment. Generally such vectors further include a signal sequence, origin of replication, one or more marker genes, an enhancer element, a promoter and transcription termination sequences operably linked to the light and/or heavy chain polynucleotide so as to facilitate expression. Polynucleotide encoding the light and heavy chains may be inserted into separate vectors and introduced (e.g. by transformation, transfection, electroporation or transduction) into the same host cell concurrently or sequentially or, if desired both the heavy chain and light chain can be inserted into the same vector prior to such introduction.
  • It will be immediately apparent to those skilled in the art that due to the redundancy of the genetic code, alternative polynucleotides to those disclosed herein are also available that will encode the polypeptides of the invention.
  • Signal Sequences
  • Antibodies of the present invention maybe produced as a fusion protein with a heterologous signal sequence having a specific cleavage site at the N terminus of the mature protein. The signal sequence should be recognised and processed by the host cell. For prokaryotic host cells, the signal sequence may be an alkaline phosphatase, penicillinase, or heat stable enterotoxin II leaders. For yeast secretion the signal sequences may be a yeast invertase leader, a factor leader or acid phosphatase leaders see e.g. WO90/13646. In mammalian cell systems, viral secretory leaders such as herpes simplex gD signal and a native immunoglobulin signal sequence (such as human Ig heavy chain) are available. Typically the signal sequence is ligated in reading frame to polynucleotide encoding the antibody of the invention.
  • Origin of Replication
  • Origin of replications are well known in the art with pBR322 suitable for most gram-negative bacteria, 2μ plasmid for most yeast and various viral origins such as SV40, polyoma, adenovirus, VSV or BPV for most mammalian cells. Generally the origin of replication component is not needed for integrated mammalian expression vectors, unless vector propagation is required in E. Coli. However the SV40 ori may be used since it contains the early promoter.
  • Selection Marker
  • Typical selection genes encode proteins that (a) confer resistance to antibiotics or other toxins e.g. ampicillin, neomycin, methotrexate or tetracycline or (b) complement auxotrophic deficiencies or supply nutrients not available in the complex media or (c) combinations of both. The selection scheme may involve arresting growth of the host cells that contain no vector or vectors. Cells, which have been successfully transformed with the genes encoding the therapeutic antibody of the present invention, survive due to e.g. drug resistance conferred by the co-delivered selection marker. One example is the DHFR-selection system wherein transformants are generated in DHFR negative host strains (eg see Page and Sydenham 1991 Biotechnology 9: 64-68). In this system the DHFR gene is co-delivered with antibody polynucleotide sequences of the invention and DHFR positive cells then selected by nucleoside withdrawal. If required, the DHFR inhibitor methotrexate is also employed to select for transformants with DHFR gene amplification. By operably linking DHFR gene to the antibody coding sequences of the invention or functional derivatives thereof, DHFR gene amplification results in concomitant amplification of the desired antibody sequences of interest. CHO cells are a particularly useful cell line for this DHFR/methotrexate selection and methods of amplifying and selecting host cells using the DHFR system are well established in the art see Kaufman R. J. et al J. Mol. Biol. (1982) 159, 601-621, for review, see Werner R G, Noe W, Kopp K, Schluter M, “Appropriate mammalian expression systems for biopharmaceuticals”, Arzneimittel-Forschung. 48(8):870-80, 1998 Aug. A further example is the glutamate synthetase expression system (Lonza Biologics). A suitable selection gene for use in yeast is the trp1 gene; see Stinchcomb et al Nature 282, 38, 1979.
  • Promoters
  • Suitable promoters for expressing antibodies of the invention are operably linked to DNA/polynucleotide encoding the antibody. Promoters for prokaryotic hosts include phoA promoter, Beta-lactamase and lactose promoter systems, alkaline phosphatase, tryptophan and hybrid promoters such as Tac. Promoters suitable for expression in yeast cells include 3-phosphoglycerate kinase or other glycolytic enzymes e.g. enolase, glyceralderhyde 3 phosphate dehydrogenase, hexokinase, pyruvate decarboxylase, phosphofructokinase, glucose 6 phosphate isomerase, 3-phosphoglycerate mutase and glucokinase, among others. Inducible yeast promoters include alcohol dehydrogenase 2, isocytochrome C, acid phosphatase, metallothionein and enzymes responsible for nitrogen metabolism or maltose/galactose utilization, among others. Promoters for expression in mammalian cell systems include RNA polymerase II promoters including viral promoters such as polyoma, fowlpox and adenoviruses (e.g. adenovirus 2), bovine papilloma virus, avian sarcoma virus, cytomegalovirus (in particular the immediate early gene promoter), retrovirus, hepatitis B virus, actin, rous sarcoma virus (RSV) promoter and the early or late Simian virus 40 and non-viral promoters such as EF-1alpha (Mizushima and Nagata Nucleic Acids Res 1990 18(17):5322, among others. The choice of promoter may be based upon suitable compatibility with the host cell used for expression.
  • Enhancer Element
  • Where appropriate, e.g. for expression in higher eukaroytics, additional enhancer elements can included instead of or as well as those found located in the promoters described above. Suitable mammalian enhancer sequences include enhancer elements from globin, elastase, albumin, fetoprotein, metallothionine and insulin. Alternatively, one may use an enhancer element from a eukaroytic cell virus such as SV40 enhancer, cytomegalovirus early promoter enhancer, polyoma enhancer, baculoviral enhancer or murine IgG2a locus (see WO04/009823). Whilst such enhancers are typically located on the vector at a site upstream to the promoter, they can also be located elsewhere e.g. within the untranslated region or downstream of the polydenalytion signal. The choice and positioning of enhancer may be based upon suitable compatibility with the host cell used for expression.
  • Polyadenylation/Termination
  • In eukaryotic systems, polyadenylation signals are operably linked to polynucleotide encoding the antibody of this invention. Such signals are typically placed 3′ of the open reading frame. In mammalian systems, non-limiting example signals include those derived from growth hormones, elongation factor-1 alpha and viral (eg SV40) genes or retroviral long terminal repeats. In yeast systems non-limiting examples of polydenylation/termination signals include those derived from the phosphoglycerate kinase (PGK) and the alcohol dehydrogenase 1 (ADH) genes. In prokaryotic system polyadenylation signals are typically not required and it is instead usual to employ shorter and more defined terminator sequences. The choice of polyadenylation/termination sequences may be based upon suitable compatibility with the host cell used for expression.
  • Other Methods/Elements for Enhanced Yields
  • In addition to the above, other features that can be employed to enhance yields include chromatin remodelling elements, introns and host-cell specific codon modification. The codon usage of the antibody of this invention thereof can be modified to accommodate codon bias of the host cell such to augment transcript and/or product yield (eg Hoekema A et al Mol Cell Biol 1987 7(8):2914-24). The choice of codons may be based upon suitable compatibility with the host cell used for expression.
  • Host Cells
  • Suitable host cells for cloning or expressing vectors encoding antibodies of the invention are, for example, prokaroytic, yeast or higher eukaryotic cells. Suitable prokaryotic cells include eubacteria e.g. enterobacteriaceae such as Escherichia e.g. E. Coli (for example ATCC 31,446; 31,537; 27,325), Enterobacter, Erwinia, Klebsiella Proteus, Salmonella e.g. Salmonella typhimurium, Serratia e.g. Serratia marcescans and Shigella as well as Bacilli such as B. subtilis and B. licheniformis (see DD 266 710), Pseudomonas such as P. aeruginosa and Streptomyces. Of the yeast host cells, Saccharomyces cerevisiae, schizosaccharomyces pombe, Kluyveromyces (e.g. ATCC 16,045; 12,424; 24178; 56,500), yarrowia (EP402, 226), Pichia Pastoris (EP183, 070, see also Peng et al J. Biotechnol. 108 (2004) 185-192), Candida, Trichoderma reesia (EP244, 234), Penicillin, Tolypocladium and Aspergillus hosts such as A. nidulans and A. niger are also contemplated.
  • Although Prokaryotic and yeast host cells are specifically contemplated by the invention, typically however, host cells of the present invention are vertebrate cells. Suitable vertebrate host cells include mammalian cells such as COS-1 (ATCC No. CRL 1650) COS-7 (ATCC CRL 1651), human embryonic kidney line 293, PerC6 (Crucell), baby hamster kidney cells (BHK) (ATCC CRL. 1632), BHK570 (ATCC NO: CRL 10314), 293 (ATCC NO. CRL 1573), Chinese hamster ovary cells CHO (e.g. CHO-K1, ATCC NO: CCL 61, DHFR-CHO cell line such as DG44 (see Urlaub et al, (1986) ibid), particularly those CHO cell lines adapted for suspension culture, mouse sertoli cells, monkey kidney cells, African green monkey kidney cells (ATCC CRL-1587), HELA cells, canine kidney cells (ATCC CCL 34), human lung cells (ATCC CCL 75), Hep G2 and myeloma or lymphoma cells e.g. NS0 (see U.S. Pat. No. 5,807,715), Sp2/0, Y0.
  • Thus in one embodiment of the invention there is provided a stably transformed host cell comprising a vector encoding a heavy chain and/or light chain of the therapeutic antibody as described herein. Typically such host cells comprise a first vector encoding the light chain and a second vector encoding said heavy chain.
  • Such host cells may also be further engineered or adapted to modify quality, function and/or yield of the antibody of this invention. Non-limiting examples include expression of specific modifying (eg glycosylation) enzymes and protein folding chaperones.
  • Cell Culturing Methods.
  • Host cells transformed with vectors encoding the therapeutic antibodies of the invention may be cultured by any method known to those skilled in the art. Host cells may be cultured in spinner flasks, shake flasks, roller bottles or hollow fibre systems but it is preferred for large scale production that stirred tank reactors or bag reactors (eg Wave Biotech, Somerset, N.J. USA) are used particularly for suspension cultures. Typically the stirred tankers are adapted for aeration using e.g. spargers, baffles or low shear impellers. For bubble columns and airlift reactors direct aeration with air or oxygen bubbles maybe used. Where the host cells are cultured in a serum free culture media it is preferred that the media is supplemented with a cell protective agent such as pluronic F-68 to help prevent cell damage as a result of the aeration process. Depending on the host cell characteristics, either microcarriers maybe used as growth substrates for anchorage dependent cell lines or the cells maybe adapted to suspension culture (which is typical). The culturing of host cells, particularly vertebrate host cells may utilise a variety of operational modes such as batch, fed-batch, repeated batch processing (see Drapeau et al (1994) cytotechnology 15: 103-109), extended batch process or perfusion culture. Although recombinantly transformed mammalian host cells may be cultured in serum-containing media such media comprising fetal calf serum (FCS), it is preferred that such host cells are cultured in synthetic serum-free media such as disclosed in Keen et al (1995) Cytotechnology 17:153-163, or commercially available media such as ProCHO-CDM or UltraCHO™ (Cambrex N.J., USA), supplemented where necessary with an energy source such as glucose and synthetic growth factors such as recombinant insulin. The serum-free culturing of host cells may require that those cells are adapted to grow in serum free conditions. One adaptation approach is to culture such host cells in serum containing media and repeatedly exchange 80% of the culture medium for the serum-free media so that the host cells learn to adapt in serum free conditions (see e.g. Scharfenberg K et al (1995) in Animal Cell technology: Developments towards the 21st century (Beuvery E. C. et al eds), pp 619-623, Kluwer Academic publishers).
  • Antibodies of the invention secreted into the media may be recovered and purified from the media using a variety of techniques to provide a degree of purification suitable for the intended use. For example the use of therapeutic antibodies of the invention for the treatment of human patients typically mandates at least 95% purity as determined by reducing SDS-PAGE, more typically 98% or 99% purity, when compared to the culture media comprising the therapeutic antibodies. In the first instance, cell debris from the culture media is typically removed using centrifugation followed by a clarification step of the supernatant using e.g. microfiltration, ultrafiltration and/or depth filtration. Alternatively, the antibody can be harvested by microfiltration, ultrafiltration or depth filtration without prior centrifugation. A variety of other techniques such as dialysis and gel electrophoresis and chromatographic techniques such as hydroxyapatite (HA), affinity chromatography (optionally involving an affinity tagging system such as polyhistidine) and/or hydrophobic interaction chromatography (HIC, see U.S. Pat. No. 5,429,746) are available. In one embodiment, the antibodies of the invention, following various clarification steps, are captured using Protein A or G affinity chromatography followed by further chromatography steps such as ion exchange and/or HA chromatography, anion or cation exchange, size exclusion chromatography and ammonium sulphate precipitation. Typically, various virus removal steps are also employed (e.g. nanofiltration using e.g. a DV-20 filter). Following these various steps, a purified (typically monoclonal) preparation comprising at least 10 mg/ml or greater e.g. 100 mg/ml or greater of the antibody of the invention is provided and therefore forms an embodiment of the invention. Concentration to 100 mg/ml or greater can be generated by ultracentrifugation. Suitably such preparations are substantially free of aggregated forms of antibodies of the invention.
  • Bacterial systems are particularly suited for the expression of antibody fragments. Such fragments are localised intracellularly or within the periplasma. Insoluble periplasmic proteins can be extracted and refolded to form active proteins according to methods known to those skilled in the art, see Sanchez et al (1999) J. Biotechnol. 72, 13-20 and Cupit P M et al (1999) Lett Appl Microbiol, 29, 273-277.
  • 3. Pharmaceutical Compositions and Mode of Administration
  • Purified preparations of antibodies of the invention as described supra, may be incorporated into pharmaceutical compositions for use in the treatment of human diseases and disorders such as those outlined above. Typically such compositions further comprise a pharmaceutically acceptable (i.e. inert) carrier as known and called for by acceptable pharmaceutical practice, see e.g. Remingtons Pharmaceutical Sciences, 16th ed, (1980), Mack Publishing Co. Examples of such carriers include sterilised carrier such as saline, Ringers solution or dextrose solution, buffered with suitable buffers to a pH within a range of 5 to 8. Pharmaceutical compositions for injection (e.g. by intravenous, intraperitoneal, intradermal, subcutaneous, intramuscular or intraportal) or continuous infusion are suitably free of visible particulate matter and may comprise from 0.1 mg to 10 g of therapeutic antibody, typically between 5 mg and 25 mg of antibody. Methods for the preparation of such pharmaceutical compositions are well known to those skilled in the art. In one embodiment, pharmaceutical compositions comprise from 0.1 mg to 10 g of therapeutic antibodies of the invention in unit dosage form, optionally together with instructions for use. Pharmaceutical compositions of the invention may be lyophilised (freeze dried) for reconstitution prior to administration according to methods well known or apparent to those skilled in the art. Where embodiments of the invention comprise antibodies of the invention with an IgG1 isotype, a chelator of copper such as citrate (e.g. sodium citrate) or EDTA or histidine may be added to the pharmaceutical composition to reduce the degree of copper-mediated degradation of antibodies of this isotype, see EP0612251.
  • Effective doses and treatment regimes for administering the antibody of the invention are generally determined empirically and are dependent on factors such as the age, weight and health status of the patient and disease or disorder to be treated. Such factors are within the purview of the attending physician. Guidance in selecting appropriate doses may be found in e.g. Smith et al (1977) Antibodies in human diagnosis and therapy, Raven Press, New York but will in general be between 0.1 mg and 1 g. In one embodiment, the dosing regime for treating a human patient is 0.1 mg to 10 g of therapeutic antibody of the invention administered subcutaneously once per week or every two weeks, or by intravenous infusion every 1 or 2 months. Compositions of the present invention may also be used in prophylatically.
  • 4. Clinical Uses.
  • The present invention relates to an antibody which binds to human IL-8, Gro-alpha, Gro-beta, Gro-gamma, and ENA-78 The present invention also concerns methods of treating diseases or disorders characterised by elevated or unbalanced level of one or more of human IL-8, Gro-alpha, Gro-beta, Gro-gamma, and ENA-78, particularly, COPD, osteoarthritis, rheumatoid arthritis, erosive arthritis, asthma, atherosclerosis, inflammatory bowel disease, psoriasis, transplant rejection, gout, cancer, acute lung injury, acute lung disease, sepsis, ARDS, peripheral artery disease, systemic sclerosis, neonatal respiratory distress syndrome, exacerbation of asthma and COPD, cystic fibrosis, diffuse panbronchiolitis, reperfusion injury, and/or endometriosis with said antibody, pharmaceutical compositions comprising said antibody and methods of manufacture.
  • The present invention also relates to use of an antibody in the manufacture of a medicament for the treatment of diseases or disorders characterised by elevated or unbalanced level of one or more of human IL-8, Gro-alpha, Gro-beta, Gro-gamma, GCP-2 and ENA-78, particularly COPD, osteoarthritis, rheumatoid arthritis, erosive arthritis, asthma, atherosclerosis, inflammatory bowel disease, psoriasis, transplant rejection, gout, cancer, acute lung injury, acute lung disease, sepsis, ARDS, peripheral artery disease, systemic sclerosis, neonatal respiratory distress syndrome, exacerbation of asthma and COPD, cystic fibrosis, diffuse panbronchiolitis, reperfusion injury, or endometriosis. Although the present invention has been described principally in relation to the treatment of human diseases or disorders, the present invention may also have applications in the treatment of similar diseases or disorders in non-human mammals.
  • SPECIFIC EMBODIMENTS Example 1 Generation of Mouse Monoclonal Antibody 656.35, 197.2, and 81.1
  • Generation of a Penta-Specific mAb Against the Target Chemokines Human IL-8, Gro-Alpha, Gro-Beta, Gro-Gamma, and ENA-78).
  • Multiple methods and schemes were used to immunize mice in an attempt to generate pan-specific mAbs. The generation of pan-specific mAbs were generated using various mixtures of multiple antigenic peptides (MAPs) and/or intact target chemokines (IL-8, Gro-α, -β, -γ, and ENA-78) mixed in complete or incomplete Freund's Adjuvant (cFA or iFA), following a modified Repetitive Immunization Multiple Sites (RIMMS) protocol in the SJL/JOrlCrl mouse strain.
  • MAPs or multiple antigenic peptides serve two functions within the immunization protocol. First, MAPs allow for a selective multiple presentation of a known target amino acid sequence. Secondly, there is an increase in mass, due to multiple copies of the sequence linked, for example, via a lysine core, which also increases the immunogenicity of the sequence over that of individual peptides (Francis, J. P., et al., Immunology, 1991: 73; 249, Schott, M. E., et al., Cell. Immuno. 1996:174:199-209, Tam, J. P. Proc. Natl. Acad. Sci. 1988: 85; 5409-5413). FIG. 1 is a schematic drawing of a set of MAPs having amino acid sequences SEQ ID NOs:49-53. A linker in MAPs can be any linker other than lysines.
  • General Immunization Time Line:
  • Two different immunization protocols following the above time line produced pan-specific mAbs:
      • 1. Initial immunization (day 0) consists of multiple subcutaneous injections (hind quarters, back and neck) of all target chemokines mixed in cFA (10 μg each). The following 4 boosts consisted of a mixture of all the target chemokines mixed in iFA (10 μg each). The fifth and all subsequent boosts consisted of a cocktail of all the target chemokines and all 5 linear MAPs (10 μg each) in iFA. The final boost 3 days prior to sacrifice and fusion consisted of all the target chemokines and linear MAPs in PBS and was delivered via an intraperitoneal (IP) injection.
      • 2. Initial immunization (day 0) consists of multiple subcutaneous injections (hind quarters, back and neck) of all five linear MAPs mixed in cFA (10 μg each). The following 4 boosts consisted of a mixture of all five linear MAPs in iFA (10 μg each). The fifth and all subsequent boosts consisted of a cocktail of all the all 5 linear MAPs and all target chemokines (10 μg each) in iFA. The final boost 3 days prior to sacrifice and fusion consisted of all the all 5 linear MAPs and all target chemokines (10 μg each) in PBS and was delivered via an intraperitoneal (IP) injection.
    Example 2
  • Pan-binding of the mAb to the target chemokines (human IL-8, Gro-alpha, Gro-beta, Gro-gamma, GCP-2, and ENA-78) has been confirmed via a time-resolved fluorescence immunofluorescent assay (TRFIA).
  • Briefly, each antigen (human IL-8, Gro-alpha, Gro-beta, Gro-gamma, GCP-2, and ENA-78, and hIL-18 when used as negative control) is individually coated onto a 96-well immunofluorescent plate. The plates are then washed and blocked with a commercially available blocking solution. Blocking solution is emptied and samples containing the mAb are added into each well and incubated for 40 to 60 minutes (this allows the mAb to bind to the target chemokines). The plates are then washed again and a detection Ab is added to each well. Detection reagents were as follows for 2×656.35, chimera (HcLc) and humanized MAbs respectively: biotinylated anti-mouse IgG, biotinylated anti-human IgG Fab2 reagent, and europium anti-human IgG. The detection antibody is conjugated to either europium or biotin. Following another round of washing, to remove unbound detection Ab, a solution of streptavidin-europium was added to biotin detected assays. Streptavidin-europium binds to the biotin allowing for detection. After a final wash to remove unbound streptavidin-europium, or in the case of the humanized MAb assay after the europium conjugate secondary is washed, each well is filled with a chelating detergent solution to activate the europium producing fluorescence. The greater the fluorescence recorded is directly proportionate to the amount of detection Ab present in the well which is directly linked and proportionate to the amount of the chemokine binding mAb.
    FIG. 5A shows mouse 656.35 mAb binding to target human chemokines.
    FIG. 5B shows the Chimera Antibody (HcLc) mAb binding to human target chemokines. FIG. 5C shows humanized mAbs binding to human target chemokines.
  • Example 3 Functional Pan-Inhibition by Murine mAb was Confirmed Using a Variety of Methods: CXCR2 Mediated Ca2+ Mobilization, Human Neutrophil Chemotaxis, and Human Neutrophil Activation (CD11b Surface Expression)
  • A microtiter plate based calcium mobilization assay, FLIPR (Fluorometric Imaging Plate Reader, Molecular Devices, Sunnyvale Calif., [Schroeder, 1996]), was used for the functional characterization of the neutralizing effect of antibodies on ELR+ chemokine induced [Ca2+]i-mobilization in CHO-K1 cells transfected with and stably expressing hCXCR2 and Gα16.
  • On the day prior to assay, cells were plated in 96 well, blackwall, clear bottom plates (Packard View) at a concentration of 40000 cells per well. After 18 to 24 hours, media was aspirated off cells and replaced with 100 μl load media containing Eagles Minimal Essential Medium (EMEM) with Earl's salts and L-Glutamine, 0.1% BSA (Serologicals Corporation), 4 μM Fluo-4-acetoxymethyl ester fluorescent indicator dye (Fluo-4 AM) and 2.5 mM probenecid. Cells were incubated in this dye containing media for 1 hour at 37° C. The dye containing media was then aspirated off the cells and replaced with identical media without Fluo-4 AM and with 0.1% Gelatin (BSA removed) and 2.5 mM probenecid. Cells were incubated for 10 min. at 37° C. and then washed 3 times with KRH assay buffer [Krebs Ringer Henseleit (120 mM NaCl, 4.6 mM KCl, 1.03 mM KH2PO4, 25 mM NaHCO3, 1.0 mM CaCl2, 11 mM MgCl2, 11 mM Glucose, 20 mM HEPES (pH 7.4)) with 0.1% gelatin and 2.5 mM probenecid]. After the final buffer wash, 100 μl KRH assay buffer with 0.1% gelatin and 2.5 mM probenecid was added to cells and warmed to 37° C. for 10 min. before being placed in FLIPR where dye loaded cells were exposed to excitation light (488 nm) from a 6 watt Argon Laser. [Ca2+]i-mobilization was monitored as a result of an increase in 516 nm emission intensity of Fluo-4 when bound to Ca2+. Change in emission intensity is directly related to cytosolic calcium levels, [Ca2+]i. After monitoring baseline for 10 sec., 50 μl of 3×ELR+ chemokine, which had been pre-incubated with a concentration range of antibody, was added to the cell plate and data collected every sec. for 1 min., followed by an additional half min. of recording in 3 sec. intervals. Maximal cellular response above basal reading was exported for plotting in GraphPad Prism (v4.03).
  • The IC50 was defined as the concentration of antibody required, during pre-treatment of 3×EC80 chemokine, to neutralize the CXCR2 mediated stimulatory effect of an EC80 concentration of the ELR+ chemokine by 50%. A secondary cellular response to 25 μM ATP was monitored to test cell viability [Sarau, 1999].
  • TABLE
    Inhibition of human ELR+ chemokine induced calcium flux (geometrically
    meaned IC50, ug/ml) by three murine mAb. (n = 3 to 6)
    3 nM hIL-8 10 nM hGROα 6 nM hGROβ 30 nM hENA-78 100 nM hGCP-2
    murine IC50 IC50 IC50 IC50 IC50
    mAB (ug/ml) 95% C. I. (ug/ml) 95% C. I. (ug/ml) 95% C. I. (ug/ml) 95% C. I. (ug/ml) 95% C. I.
    656.35 0.18 0.14-0.25 1.43 0.60-3.42 1.06 0.78-1.42 2.09 0.71-6.14 9.58 1.17-78.57
    81.1 67.65  18.80-243.50 0.94 0.37-2.38 n.d. N/A 1.84 0.69-4.93 9.11 1.44-57.61
    197.2 2.19 0.61-7.88 2.24  0.39-12.98 n.d. N/A 13.86  1.71-112.49 >157 N/A
    (n.d. = not determined, N/A = not available)

    Schroeder K S, Neagle, B D. FLIPR: a new instrument for accurate, high throughput optical screening. J. Biomol. Screen. 1996:1-75.
    Sarau H M, Ames R S, Chambers J, Ellis C, Elshourbagy N, Foley J J et al. Identification, molecular cloning, expression, and characterization of a cysteinyl leukotrien receptor. Mol Pharmacol. 1999; 56:657-663.
  • Inhibition of IL-8 stimulated human neutrophil chemotaxis was also demonstrated using a micro-Boyden chamber. The micro-Boyden apparatus consists of two small chambers one above and one below a 3 micron-porous membrane. The lower chamber is loaded with the chemotaxic agent (i.e. IL-8) or a mixture of the chemokine with the penta-specific mAb. The upper chamber contains purified non-activated human neutrophils. Once the chamber is assembled a concentration gradient is formed from the bottom well to the upper, stimulating the neutrophils to chemotax across the membrane. The assay is expressed as a CI (chemotactic index) which is a ratio of the number of cells which chemotax in response to a stimulant over non-stimulated chemotaxic cell number. Pre-incubating the chemokine (i.e. IL-8) with the penta-specific mAb in the lower chamber, dose dependently inhibited IL-8 stimulated neutrophil chemotaxis. IL-8 at 10 nM achieved a CI of 3.6±0.8. Pre-incubation of IL-8 with increasing concentrations of the penta-specific mAb (656.35) dose dependently inhibited human neutrophil chemotaxis, significance was reached at CI of 2.2±0.6 at 1 μg/ml. Due to the amounts and sensitivity of the assay the other chemokines were unable to be examined (Gro-α, -β, -γ, and ENA-78).
  • Inhibition of purified human neutrophil activation via monitoring surface expression of CD11b. CD11b or Mac-1 mediates adhesion to substrates, aggregation and chemotaxis and is known to be up-regulated on the surface activated neutrophils (Molad, Y., J., et al., Clin. Immunol. Immunopathol. 1994: 71; 281-286). Briefly, non-activated human neutrophils are purified and ex vivo stimulated with either target chemokines (i.e. IL-8) or with chemokines pre-incubated with a chemokine penta-specific mAb. Data are presented as percent activation set to the maximal CD11b surface expression due to IL-8 stimulation. Pre-incubation of IL-8 with the 656.35 mAb dose dependently inhibited increased levels of surface expression of CD11b and thus indicates inhibition of neutrophil activation (79.9%±3.7, 48.5%±7.2, 28.7%±3.2, and 31.6%±3.4, at 0.01, 0.1, 1, 10 and 50 μg/ml respectively).
  • Example 4 Penta-Specific mAbs Association/Dissociation Values for Each Target Chemokine (human IL-8, Gro-Beta, and ENA-78) Methods for Biacore Analysis.
  • For experiment designated KL, rabbit anti mouse IgG-Fc (Biacore BR-1005-14) is immobilized on a CM5 chip by primary amine coupling in accordance with the manufactures instructions.
  • For experiment designated BE, purified antibodies directly immobilized to the chip by amine coupling was used.
  • Supernatant or purified antibody from parental mouse mAb is captured on the anti-mouse IgG-Fc surface. Defined concentrations of each analyte (IL8, Gro-band ENA-78) are passed over the immobilized or captured antibody surface, a separate capture event is used for each analyte injection. After each injection of analyte the surface is regenerated by injection
  • of a mild acidic solution, which removes the captured antibody but does not significantly affect the capability anti mouse IgG-Fc surface to perform another capture event nor does it affect the directly immobilized antibodies. An injection of buffer is also injected over the antibody captured surface and this is used for double referencing. The data is analyzed using the analysis software inherent to the machine, using the 1:1 model of binding.
  • IL-8 Gro-B ENA-78
    mAb kd ka KD kd ka KD kd ka KD
    KL 656.35 1.48E−03 1.53E+06 9.64E−10 2.01E−03 3.82E+06 5.26E−10 1.87E−03 1.07E+06 1.74E−09
    BE 6.87E−04 8.19E+06 8.39E−11 1.46E−03 1.32E+07 1.11E−10 7.96E−04 2.94E+06 2.71E−10
    KL 81.1 9.32E−03 1.95E+06 4.80E−09 3.12E−04 2.76E+06 1.13E−10 3.76E−04 6.11E+05 6.16E−10
    BE 1.08E−01 1.95E+06 5.55E−08 4.89E−04 5.44E+06 8.98E−11 3.94E−06 1.65E+05 2.39E−11
    KL 197.2 3.69E−03 6.27E+05 5.89E−09 9.93E−04 1.55E+05 6.39E−10 2.69E−03 2.99E+05 9.00E−09
    BE 4.99E−04 7.23E+05 6.90E−10 4.80E−08 1.04E+06 4.62E−14 3.02E−03 3.01E+05 1.01E−08
  • Example 5 Epitope Mapping
  • 656.35 mAb was epitope mapped and found to bind within KELRCQCIKTYSKP (SEQ ID NO: 54) in human IL-8. Thus in another embodiment, the present invention relates to a penta-specific antibody which binds within epitope of SEQ ID NO:54 of human IL-8.
  • Example 6 Efficacy Study of a Chimera Antibody Made from 656.35 Having Heavy Chain Sequence of SEQ ID NO: 56, and Light Chain Sequence of SEQ ID NO:58. (This Antibody Will be Referred to as the Chimera Antibody)
  • In Vivo Studies:
  • An inhaled LPS acute lung inflammatory model was used to examine the ability of the a penta-specific antibody to inhibit infiltration of neutrophils into the lungs of non-human primates (NHPs—cynomolgus). Briefly, NHP (non-human primate) were pre-screened for health and responsiveness to exogenously added cynomolgus IL-8 (cynoIL-8). Selected NHP were then subjected to baseline sample collections (blood and bronchoalveolar lavage—BAL) five days prior to the first LPS challenge. LPS challenged consisted of tranquilizing the NHP with a single IM injection of ketamine HCl (˜10 mg/kg). Once the NHP is sedated, anesthesia is administered, via an intravenous infusion of propofol (˜0.2 mg/kg/min, as necessary). Anesthetized animals are placed on a circulating warm-water heating blanket and/or within a circulating warm-air blanket and an ophthalmic lubricant is administered to each eye. The animals are then intubated and mechanically ventilated for challenge procedure. A volume regulated Positive Pressure ventilator is used during the procedure (Stoelting Cat/Rabbit ventilator www.stoeltingco.com Cat #5019510). Lipopolysaccharide (LPS) challenge is performed via aerosol inhalation using a DeVilbiss Ultraneb-99 ultrasonic nebulizer. Aerosolized LPS is administered for 5 minutes at 100 ug/ml. Heart rate, body temperature, and respiration rate are monitored and recorded during challenge procedures. The initial challenge confirms that the NHP respond to LPS (increased neutrophil infiltration into the lungs and elevated cynoIL-8 levels). Individual NHP responses were confirmed by sample collections at 6 and 24-hours post challenge.
  • The initial LPS challenged NHP were then randomly divided into two groups. Following a 4-week recovery all participating NHP were subjected to another baseline sample (blood and BAL). Four days post baseline measurements the subject groups received IV injection of either a vehicle or an injection of 1 mg/kg of the Chimera Antibody. The next day the NHP were challenged again with inhaled LPS and samples for analysis were collected 6 and 24-hour post challenge. Following an additional recovery period base line samples were collected from the vehicle NHP, 4 days later these NHP were treated with a single IV bolus injection of the Chimera Antibody at 10 mg/kg. The next day the animals were exposed to an LPS challenge and samples were collected for analysis at 6 and 24 hours.
  • The LPS inhalation is an acute inflammatory model which stimulates an elevation of chemotactic chemokines such at IL-8, which lead to increased infiltration of neutrophils into the lungs. The primary efficacy parameter for the Chimera Antibody treatment is inhibition of infiltrating neutrophils into the lungs of NHP challenged with LPS. Neutrophil infiltration in this acute inflammatory model occurs during the first 24 hours following LPS challenge at which point other inflammatory processes are becoming more prominent.
  • Pretreatment of with the Chimera Antibody significantly and dose dependently inhibited neutrophil infiltration into lungs of LPS challenged NHP. Treatment with the Chimera Antibody also prevented elevation of circulating neutrophils, while not affecting the actual neutrophil function, (i.e. the cells ability to phagocytos). Treatment also did not greatly affect other cell types such as macrophages/monocytes in the lung or circulation. See FIG. 2.
  • Example 7 Further Studies on Humanized mAbs
  • Multiple humanized penta-specific antibody constructs have been produced, four of which have been shown to bind tightly to, and inhibit all target chemokines. (For each sequence, see Sequence Information below). This analysis consisted of affinity measurements (BiaCore), calcium mobilization assays (in vitro functional assay, FLIPR), and CD11b human and NHP neutrophil stimulation assay (ex vivo functional assay, flow cytometry).
  • BiaCore
  • A Protein A capture method was used to generate kinetics for the humanised and chimeric constructs as follows:
  • Protein A is immobilised on a CM5 chip by primary amine coupling in accordance with the manufactures instructions. Purified humanised or chimeric antibody is captured on the Protein A surface. After capture defined concentrations of each analyte (IL8, Gro-β and ENA-78) are passed over the antibody captured surface, a separate capture event is used for each analyte injection. After each injection of analyte the surface is regenerated by injection of a mild acidic solution, which removes the captured antibody but does not significantly affect the capability of the Protein A to perform another capture event. An injection of buffer is also injected over the antibody captured surface and this is used for double referencing. The data is analysed using the analysis software inherent to the machine, using the 1:1 model of binding.
  • Construct ka (1/Ms) kd(1/s) KD(M)
    Measurements with Gro-beta
    HcLc 7.39E+06 8.16E−04 1.11E−10
    HcLc 2.11E+07 0.0011  5.21E−11
    HcLc 1.59E+07 8.88E−04 5.57E−11
    HcLc 1.19E+07 6.16E−04 5.17E−11
    HcLc 1.43E+07 5.43E−04 3.79E−11
    HcLc 1.33E+07 6.76E−04 5.09E−11
    HcLc 1.51E+07 6.73E−04 4.45E−11
    HcLc 1.49E+07 6.44E−04 4.33E−11
    HcLc 2.54E+07 0.001163 4.57E−11
    HcLc 1.81E+07 0.001058 5.85E−11
    HcLc 2.04E+07 8.54E−04 4.18E−11
    HcLc 1.82E+07 5.06E−04 2.78E−11
    H0L7 6.99E+06 9.66E−04 1.38E−10
    H0L7 1.76E+07 0.001305 7.40E−11
    H0L7 1.63E+07 0.001039 6.39E−11
    H0L7 1.14E+07 0.005616 4.93E−10
    H0L7 1.20E+07 0.004398 3.66E−10
    H0L7 1.42E+07 9.57E−04 6.73E−11
    H0L7 1.76E+07 0.00118 6.72E−11
    H0L7 1.63E+07 0.001389 8.52E−11
    H0L7 1.12E+07 0.008212 7.32E−10
    H0L7 2.63E+07 0.001546 5.88E−11
    H0L7 2.10E+07 9.28E−04 4.41E−11
    H0L8 1.28E+07 0.005462 4.27E−10
    H0L8 1.99E+07 0.01065 5.36E−10
    H0L8 1.19E+07 0.007727 6.52E−10
    H0L8 1.18E+07 0.007799 6.60E−10
    H0L8 1.61E+07 0.006903 4.29E−10
    H0L8 1.85E+07 0.004586 2.48E−10
    H0L10 1.23E+07 0.001395 1.13E−10
    H0L10 1.45E+07 0.001261 8.68E−11
    H0L10 1.44E+07 0.001477 1.03E−10
    H0L10 1.28E+07 0.001394 1.09E−10
    H0L10 1.98E+07 0.00144 7.28E−11
    H0L10 1.94E+07 0.00109 5.62E−11
    H0M0 1.20E+07 0.001394 1.17E−10
    H0M0 1.13E+07 0.007071 6.24E−10
    H0M0 1.58E+07 0.001708 1.08E−10
    H0M0 1.43E+07 0.001829 1.28E−10
    H0M0 1.03E+07 0.009588 9.34E−10
    H0M0 1.85E+07 0.001728 9.35E−11
    H0M0 1.81E+07 0.001252 6.91E−11
    Measurements with IL8
    HcLc 3.77E+06 2.34E−04 6.22E−11
    HcLc 1.03E+07 2.87E−04 2.78E−11
    HcLc 8.20E+06 2.76E−04 3.37E−11
    HcLc 1.04E+07 2.68E−04 2.58E−11
    HcLc 9.44E+06 2.53E−04 2.68E−11
    HcLc 9.97E+06 2.30E−04 2.31E−11
    H0L7 2.74E+06 2.84E−04 1.04E−10
    H0L7 8.99E+06 4.00E−04 4.45E−11
    H0L7 7.64E+06 2.84E−04 3.71E−11
    H0L7 5.98E+06 0.001167 1.95E−10
    H0L7 8.74E+06 3.52E−04 4.03E−11
    H0L7 7.51E+06 3.63E−04 4.83E−11
    H0L7 9.10E+06 3.89E−04 4.28E−11
    H0L8 5.90E+06 0.00111 1.88E−10
    H0L8 7.89E+06 0.001126 1.43E−10
    H0L8 6.80E+06 0.001193 1.75E−10
    H0L8 7.30E+06 0.001112 1.52E−10
    H0L10 6.86E+06 4.57E−04 6.66E−11
    H0L10 7.20E+06 3.67E−04 5.10E−11
    H0L10 7.21E+06 3.96E−04 5.48E−11
    H0L10 7.43E+06 3.73E−04 5.02E−11
    H0M0 7.02E+06 3.98E−04 5.67E−11
    H0M0 6.40E+06 0.001467 2.29E−10
    H0M0 7.96E+06 4.09E−04 5.14E−11
    H0M0 7.97E+06 4.60E−04 5.77E−11
    H0M0 7.50E+06 4.18E−04 5.57E−11
    Measurements with ENA78
    HcLc 3.67E+06 1.70E−04 4.63E−11
    HcLc 5.81E+06 1.71E−04 2.94E−11
    HcLc 5.11E+06 1.84E−04 3.60E−11
    HcLc 5.28E+06 1.54E−04 2.91E−11
    HcLc 4.87E+06 1.56E−04 3.20E−11
    HcLc 4.78E+06 1.23E−04 2.58E−11
    H0L7 2.95E+06 2.04E−04 6.93E−11
    H0L7 5.28E+06 2.97E−04 5.63E−11
    H0L7 4.59E+06 2.17E−04 4.74E−11
    H0L7 3.03E+06 3.33E−04 1.10E−10
    H0L7 5.11E+06 2.58E−04 5.04E−11
    H0L7 4.76E+06 3.07E−04 6.45E−11
    H0L7 4.91E+06 2.77E−04 5.63E−11
    H0L8 2.88E+06 2.88E−04 1.00E−10
    H0L8 2.82E+06 2.66E−04 9.41E−11
    H0L8 2.95E+06 2.69E−04 9.12E−11
    H0L8 3.29E+06 2.48E−04 7.55E−11
    H0L10 4.07E+06 3.66E−04 8.99E−11
    H0L10 4.72E+06 3.40E−04 7.22E−11
    H0L10 4.20E+06 3.12E−04 7.42E−11
    H0L10 4.38E+06 2.68E−04 6.13E−11
    H0M0 3.91E+06 3.09E−04 7.89E−11
    H0M0 2.97E+06 3.49E−04 1.17E−10
    H0M0 4.39E+06 3.16E−04 7.20E−11
    H0M0 4.18E+06 3.34E−04 7.99E−11
    H0M0 4.52E+06 2.94E−04 6.51E−11
    Affinity (KD) is determined by examining the association rate (ka) and disassociation rate (kd) of proteins.
    HcLc refers to chimera antibody made from 656.35 having heavy chain sequence of SEQ ID NO: 56, and light chain sequence of SEQ ID NO: 58.
  • Functional Assay: Calcium Mobilization (FLIPR) Using CHO-K1 CXCR2 (w/G α16)
  • A microtiter plate based calcium mobilization assay, FLIPR (Fluorometric Imaging Plate Reader, Molecular Devices, Sunnyvale Calif., [Schroeder, 1996]), was used for the functional characterization of the neutralizing effect of antibodies on ELR+ chemokine induced [Ca2+]i-mobilization in CHO-K1 cells transfected with and stably expressing hCXCR2 and Gα16.
  • On the day prior to assay, cells were plated in 96 well, blackwall, clear bottom plates (Packard View) at a concentration of 40000 cells per well. After 18 to 24 hours, media was aspirated off cells and replaced with 100 μl load media containing Eagles Minimal Essential Medium (EMEM) with Earl's salts and L-Glutamine, 0.1% BSA (Serologicals Corporation), 4 μM Fluo-4-acetoxymethyl ester fluorescent indicator dye (Fluo-4 AM) and 2.5 mM probenecid. Cells were incubated in this dye containing media for 1 hour at 37° C. The dye containing media was then aspirated off the cells and replaced with identical media without Fluo-4 AM and with 0.1% Gelatin (BSA removed) and 2.5 mM probenecid. Cells were incubated for 10 min. at 37° C. and then washed 3 times with KRH assay buffer [Krebs Ringer Henseleit (120 mM NaCl, 4.6 mM KCl, 1.03 mM KH2PO4, 25 mM NaHCO3, 1.0 mM CaCl2, 1.1 mM MgCl2, 11 mM Glucose, 20 mM HEPES (pH 7.4)) with 0.1% gelatin and 2.5 mM probenecid]. After the final buffer wash, 100 μl KRH assay buffer with 0.1% gelatin and 2.5 mM probenecid was added to cells and warmed to 37° C. for 10 min. before being placed in FLIPR where dye loaded cells were exposed to excitation light (488 nm) from a 6 watt Argon Laser. [Ca2+]i-mobilization was monitored as a result of an increase in 516 nm emission intensity of Fluo-4 when bound to Ca2+. Change in emission intensity is directly related to cytosolic calcium levels, [Ca2+]i. After monitoring baseline for 10 sec., 50 μl of 3×ELR+ chemokine, which had been pre-incubated with a concentration range of antibody, was added to the cell plate and data collected every sec. for 1 min., followed by an additional half min. of recording in 3 sec. intervals. Maximal cellular response above basal reading was exported for plotting in GraphPad Prism (v4.03).
  • The IC50 was defined as the concentration of antibody required, during pre-treatment of 3×EC80 chemokine, to neutralize the CXCR2 mediated stimulatory effect of an EC80 concentration of the ELR+ chemokine by 50%. A secondary cellular response to 25 μM ATP was monitored to test cell viability [Sarau, 1999].
  • TABLE
    Inhibition of human ELR+ chemokine induced calcium flux (geometrically
    meaned IC50, ug/ml) by four humanized mAb constructs. (n = 3)
    Pre-treated 3X H0L7 H0L8 H0L10 H0M0
    EC80 ELR+ IC50 IC50 IC50 IC50
    Chemokine (ug/ml) 95% C.I. (ug/ml) 95% C.I. (ug/ml) 95% C.I. (ug/ml) 95% C.I.
    3 nM hIL8 0.30 0.12-0.72 0.83 0.70-0.98 0.19 0.15-0.22 0.19 0.15-0.23
    10 nM hGROa 1.48 0.98-2.23 3.52 2.74-4.53 1.09 0.58-2.06 1.11 0.67-1.83
    6 nM hGROb 1.04 0.19-5.58 7.93  4.20-14.97 0.66 0.24-1.86 0.99 0.35-2.76
    30 nM hENA-78 2.48 0.78-7.91 2.42 1.27-4.63 1.88 0.83-4.26 1.75 1.33-2.29
    100 nM hGCP-2 11.43  8.19-15.96 40.38 38.45-42.41 11.54  6.30-21.17 13.03  7.20-23.56

    Schroeder K S, Neagle, B D. FLIPR: a new instrument for accurate, high throughput optical screening. J. Biomol. Screen. 1996:1-75.
    Sarau H M, Ames R S, Chambers J, Ellis C, Elshourbagy N, Foley J J et al. Identification, molecular cloning, expression, and characterization of a cysteinyl leukotrien receptor. Mol Pharmacol. 1999; 56:657-663.
  • Ex Vivo CD11b Human Neutrophil Stimulation Assay
  • Flow cytometry was used to examine the humanized penta-specific antibodies' ability to prevent chemokine induced human purified neutrophil activation, by following physical cellular changes (size and shape—granulation) and by examining surface activation markers (increased CD11b surface expression). The neutrophil control stimulant FMLP was used to confirm the purified human neutrophils ability to activate. See FIG. 4.
  • Sequence Information
  • Total RNA was extracted from 656.35, 197.2 and 81.1 hybridoma cells, heavy and light variable domain cDNA sequence was then generated by reverse transcription and polymerase chain reaction (RT-PCR). The forward primer for RT-PCR was a mixture of degenerate primers specific for murine immunoglobulin gene leader-sequences and the reverse primer was specific for the antibody constant regions, in this case isotype IgG2a/κ. Primers were designed according to the strategy described by Jones and Bendig (Bio/Technology 9:88, 1991). RT-PCR was carried out in duplicate for both V-region sequences to enable subsequent verification of the correct V-region sequences. The V-region products generated by the RT-PCR were cloned (Invitrogen TA Cloning Kit) and sequence data obtained. This process was not successful for 81.1 light variable domain. Thus, the amino acid sequence shown below (Seq ID NO: 12) was therefore generated by protein sequencing the light chain isolated on a SDS-polyacrylamide gel run under reducing conditions.
    Complementarity Determining Regions (CDRs) are underlined.
    Polynucleotide sequences for heavy and light variable regions (SEQ ID NO: 1 and 3, respectively) for 656.35
  • SEQ ID NO: 1
    CAGGTCCAGTTGCAGCAGTCTGGAGCTGAACTGGTAAGGCCTGGGACTTC
    AGTGACGATATCCTGTAAGGCTTCTGGCTACACCTTCACTAACTACTGGA
    TAGTTTGGGTCAAACAGAGGCCTGGACATGGACTTGAGTGGATTGGAGAT
    CTTTACTCTGGAGGTGGTTATACTTTCTACAGTGAAAATTTCAAGGGGAA
    GGCCACACTGACTGCAGACACATCCTCCAGCACTGCCTACATGCACCTCA
    TTAGCCTGACATCTGAGGACTCTGCTGTCTATTTCTGTGCAAGATCGGGT
    TACGACAGAACCTGGTTTGCTCACTGGGGCCAAGGGTCTCTGGTCACTGT
    CTCTGCA
    SEQ ID NO: 3
    GACATCAAGATGACCCAGTCTCCATCCTCCATGTCTGCATCGCTGGGAGA
    GAGAGTCACTATCACTTGTCAGGCGAGTCAGGACATTGAAAGCTATTTAA
    GCTGGTATCAGCAGAAACCATGGAAATCTCCTAAGACCCTGATCTATTAC
    GCTACAAGGTTGGCAGATGGGGTCCCATCAAGATTCAGTGGCAGTGGATC
    TGGTCAAGATTATTCTCTAACCATCAGCAGCCTGGAGTCTGACGATACAG
    CAACTTATTACTGTCTACAACATGGTGAGAGCCCTCCCACGTTCGGTGCT
    GGGACCAAGCTGGAGCTGAAACGG

    Polypeptide sequences for heavy and light variable regions (SEQ ID NO: 2 and 4, respectively) for 656.35
  • SEQ ID NO: 2
    QVQLQQSGAELVRPGTSVTISCKASGYTFTNYWIVWVKQRPGHGLEWIGD
    LYSGGGYTFYSENFKGKATLTADTSSSTAYMHLISLTSEDSAVYFCARSG
    YDRTWFAHWGQGSLVTVSA
    SEQ ID NO: 4
    DIKMTQSPSSMSASLGERVTITCQASQDIESYLSWYQQKPWKSPKTLIYY
    ATRLADGVPSRFSGSGSGQDYSLTISSLESDDTATYYCLQHGESPPTFGA
    GTKLELKR

    Polypeptide sequences for heavy chain CDRs (SEQ ID NO: 13, 14, 15, respectively) for 656.35
  • NYWIV SEQ ID NO: 13
    DLYSGGGYTFYSENFKG SEQ ID NO: 14
    SGYDRTWFAH SEQ ID NO: 15

    Polypeptide sequences for light chain CDRs (SEQ ID NO: 16, 17, 18) for 656.35
  • QASQDIESYLS SEQ ID NO: 16
    YATRLAD SEQ ID NO: 17
    LQHGESPPT SEQ ID NO: 18

    Polynucleotide sequences for heavy chain CDRs (SEQ ID NO: 31, 32, 33) for 656.35
  • SEQ ID NO: 31
    AACTACTGGATAGTT
    SEQ ID NO: 32
    GATCTTTACTCTGGAGGTGGTTATACTTTCTACAGTGAAAATTTCAAGG
    GG
    SEQ ID NO: 33
    TCGGGTTACGACAGAACCTGGTTTGCTCAC

    Polynucleotide sequences for light chain CDRs (SEQ ID NO:34, 35, 36) for 656.35
  • CAGGCGAGTCAGGACATTGAAAGCTATTTAAGC SEQ ID NO: 34
    TACGCTACAAGGTTGGCAGAT SEQ ID NO: 35
    CTACAACATGGTGAGAGCCCTCCCACG SEQ ID NO: 36

    Polynucleotide sequences for heavy and light variable regions (SEQ ID NO: 5 and 7, respectively) for 197.2
  • SEQ ID NO: 5
    GAGTTCCAGCTGCAGCAGTCTGGACCTGAGCTGGTGAAGCCTGGCGCTTC
    AGTGAAGATATCCTGCAAGGCTTCTGGTTACTCATTCACTGACTACAACA
    TGAACTGGGTGAAGCAGAGCAATGGAAAGAGCCTTGAGTGGATTGGAGTA
    ATTAATCCTAAGTATGGTACTACTAGTTACAATCAGAAGTTCAAGGGCAA
    GGCCACGTTGACTGTAGACCAATCCTCCAACACAGCCTACATGCAGCTCA
    GCAGCCTGACATCTGAGGACTCTGCAGTCTATCACTGTGCAAGAGGAATG
    GGACTCCTCTTTGGTATGGACTACTGGGGCCAAGGAACCTCTGTCACCGT
    CTCCTCA
    SEQ ID NO: 7
    GACATTGTGATGACACAGTCTCCATCCTCCCTGAGTGTGTCAGCAGGAGA
    GAAGGTCACTATGAGCTGCAAGTCCAGTCAGAGTCTGTTAAACAGTGGAA
    ATCARAAGAACTACTTGGCCTGGTACCAGCAGAAACCAGGGCAGCCTCCT
    AAACTGTTGATCTACGGGGCATCCACTAGGAAATCTGGGGTCCCTGATCG
    CTTCACAGGCAGTGGATCTGGAACCGATTTCACTCTTACCATCAGCAGTG
    TGCAGGCTGAAGACCTGGCAGTTTATTACTGTCAGAATGATCATAGTTTT
    CCGTGCACGTTCGGAGGGGGGACCAAGCTGGAAATAAAACGG

    Polypeptide sequences for heavy and light variable regions (SEQ ID NO: 6 and 8, respectively) for 197.2
  • SEQ ID NO: 6
    EFQLQQSGPELVKPGASVKISCKASGYSFTDYNMNWVKQSNGKSLEWIGV
    INPKYGTTSYNQKFKGKATLTVDQSSNTAYMQLSSLTSEDSAVYHCARGM
    GLLFGMDYWGQGTSVTVSS
    SEQ ID NO: 8
    DIVMTQSPSSLSVSAGEKVTMSCKSSQSLLNSGNQKNYLAWYQQKPGQPP
    KLLIYGASTRKSGVPDRFTGSGSGTDFTLTISSVQAEDLAVYYCQNDHSF
    PCTFGGGTKLEIKR

    Polypeptide sequences for heavy chain CDRs (SEQ ID NO: 19, 20, 21, respectively) for 197.2
  • DYNMN SEQ ID NO: 19
    VINPKYGTTSYNQKFKG SEQ ID NO: 20
    GMGLLFGMDY SEQ ID NO: 21

    Polypeptide sequences for light chain CDRs (SEQ ID NO: 22, 23, 24) for 197.2
  • KSSQSLLNSGNQKNYLA SEQ ID NO: 22
    GASTRKS SEQ ID NO: 23
    QNDHSFPCT SEQ ID NO: 24

    Polynucleotide sequences for heavy chain CDRs (SEQ ID NO: 37, 38, 39) for 197.2
  • SEQ ID NO: 37
    GACTACAACATGAAC
    SEQ ID NO: 38
    GTAATTAATCCTAAGTATGGTACTACTAGTTACAATCAGAAGTTCAAG
    GGC
    SEQ ID NO: 39
    GGAATGGGACTCCTCTTTGGTATGGACTAC

    Polynucleotide sequences for light chain CDRs (SEQ ID NO:40, 41, 42) for 197.2
  • SEQ ID NO: 40
    AAGTCCAGTCAGAGTCTGTTAAACAGTGGAAATCAAAAGAACTACTTG
    GCC
    SEQ ID NO: 41
    GGGGCATCCACTAGGAAATCT
    SEQ ID NO: 42
    CAGAATGATCATAGTTTTCCGTGCACG

    Polynucleotide sequence for heavy variable region (SEQ ID NO: 9) for 81.1
  • SEQ ID NO: 9
    GAGGTCCAGCTGCAGCAGTCTGGACCTGAACTGGAGAAGCCTGGCGCTTC
    AGTGAAGATATCCTGCAAGGCTTCTGGTTACTCTTTCACTGTCTACGGCA
    TGAACTGGGTGAGACAGAGCAATGGAAAGAGCCTTGAATGGATTGGAAAT
    TTTGATCCTTACTTTAGTGTCACTTCCTACAACCAGAAGTTCCAGGACAA
    GGCCACATTGACTGTAGACAAATCCTCCAGCACAGCCTACATGCAGCTCA
    AGAACCTCACATCTGAAGACTCTGCAGTCTATTTCTGTGCAAGAGGGAGC
    TGGGAAACCATTTTTGCTTACTGGGGCCAAGGGACTCTGGTCACTGTCTC
    TGCA

    Polypeptide sequence for heavy variable region (SEQ ID NO: 10) for 81.1
  • SEQ ID NO: 10
    EVQLQQSGPELEKPGASVKISCKASGYSFTVYGMNWVRQSNGKSLEWIGN
    FDPYFSVTSYNQKFQDKATLTVDKSSSTAYMQLKNLTSEDSAVYFCARGS
    WETIFAYWGQGTLVTVSA

    Polypeptide sequences for heavy chain CDRs (SEQ ID NO: 25, 26, 27, respectively) for 81.1
  • VYGMN SEQ ID NO: 25
    NFDPYFSVTSYNQKFQD SEQ ID NO: 26
    GSWETIFAY SEQ ID NO: 27

    Polynucleotide sequences for heavy chain CDRs (SEQ ID NO: 43, 44, 45) for 81.1
  • SEQ ID NO: 43
    GTCTACGGCATGAAC
    SEQ ID NO: 44
    AATTTTGATCCTTACTTTAGTGTCACTTCCTACAACCAGAAGTTCCAGG
    AC
    SEQ ID NO: 45
    GGGAGCTGGGAAACCATTTTTGCTTAC

    Polypeptide sequence for light variable region (SEQ ID NO: 12) for 81.1
  • QAVVTQESALTTSPGETVTLTCRSSTGAVTTSNYANWVQEKPDHLFTGLI
    GGTNNRAPGVPARFSGSLIGDKAALTITGAQTEDEAIYFCALWYSNHVFG
    GGTKLTVLQPK

    Polypeptide sequences for light chain CDRs (SEQ ID NO: 28, 29, and 30, respectively) for 81.1
  • RSSTGAVTTSNYAN SEQ ID NO:28
    GTNNRAP SEQ ID NO:29
    ALWYSNHV SEQ ID NO:30

    A chimera* polynucleotide sequence (variable heavy region+codon optimised IgG1)
  • SEQ ID NO: 55
    CAGGTCCAGTTGCAGCAGTCTGGAGCTGAACTGGTAAGGCCTGGGACTTC
    AGTGACGATATCCTGTAAGGCTTCTGGCTACACCTTCACTAACTACTGGA
    TAGTTTGGGTCAAACAGAGGCCTGGACATGGACTTGAGTGGATTGGAGAT
    CTTTACTCTGGAGGTGGTTATACTTTCTACAGTGAAAATTTCAAGGGGAA
    GGCCACACTGACTGCAGACACATCCTCCAGCACTGCCTACATGCACCTCA
    TTAGCCTGACATCTGAGGACTCTGCTGTCTATTTCTGTGCAAGATCGGGT
    TACGACAGAACCTGGTTTGCTCACTGGGGCCAAGGGTCACTAGTGACCGT
    GTCCAGCGCCAGCACCAAGGGCCCCAGCGTGTTCCCCCTGGCCCCCAGCA
    GCAAGAGCACCAGCGGCGGCACAGCCGCCCTGGGCTGCCTGGTGAAGGAC
    TACTTCCCCGAACCGGTGACCGTGTCCTGGAACAGCGGAGCCCTGACCAG
    CGGCGTGCACACCTTCCCCGCCGTGCTGCAGAGCAGCGGCCTGTACAGCC
    TGAGCAGCGTGGTGACCGTGCCCAGCAGCAGCCTGGGCACCCAGACCTAC
    ATCTGTAACGTGAACCACAAGCCCAGCAACACCAAGGTGGACAAGAAGG
    TGGAGCCCAAGAGCTGTGACAAGACCCACACCTGCCCCCCCTGCCCTGCC
    CCCGAGCTGCTGGGAGGCCCCAGCGTGTTCCTGTTCCCCCCCAAGCCTAA
    GGACACCCTGATGATCAGCAGAACCCCCGAGGTGACCTGTGTGGTGGTGG
    ATGTGAGCCACGAGGACCCTGAGGTGAAGTTCAACTGGTACGTGGACGGC
    GTGGAGGTGCACAATGCCAAGACCAAGCCCAGGGAGGAGCAGTACAACA
    GCACCTACCGGGTGGTGTCCGTGCTGACCGTGCTGCACCAGGATTGGCTG
    AACGGCAAGGAGTACAAGTGTAAGGTGTCCAACAAGGCCCTGCCTGCCCC
    TATCGAGAAAACCATCAGCAAGGCCAAGGGCCAGCCCAGAGAGCCCCAG
    GTGTACACCCTGCCCCCTAGCAGAGATGAGCTGACCAAGAACCAGGTGTC
    CCTGACCTGCCTGGTGAAGGGCTTCTACCCCAGCGACATCGCCGTGGAGT
    GGGAGAGCAACGGCCAGCCCGAGAACAACTACAAGACCACCCCCCCTGT
    GCTGGACAGCGATGGCAGCTTCTTCCTGTACAGCAAGCTGACCGTGGACA
    AGAGCAGATGGCAGCAGGGCAACGTGTTCAGCTGCTCCGTGATGCACGA
    GGCCCTGCACAATCACTACACCCAGAAGAGCCTGAGCCTGTCCCCTGGCA
    AG

    chimera* polypeptide sequence (variable heavy region+codon optimised IgG1)
  • SEQ ID NO: 56
    QVQLQQSGAELVRPGTSVTISCKASGYTFTNYWIVWVKQRPGHGLEWIGD
    LYSGGGYTFYSENFKGKATLTADTSSSTAYMHLISLTSEDSAVYFCARSG
    YDRTWFAHWGQGSLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKD
    YFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTY
    ICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPK
    DTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNS
    TYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQV
    YTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVL
    DSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK

    A chimera* polynucleotide sequence (variable light region+codon optimised human cK)
  • SEQ ID NO: 57
    GACATCAAGATGACCCAGTCTCCATCCTCCATGTCTGCATCGCTGGGAGA
    GAGAGTCACTATCACTTGTCAGGCGAGTCAGGACATTGAAAGCTATTTAA
    GCTGGTATCAGCAGAAACCATGGAAATCTCCTAAGACCCTGATCTATTAC
    GCTACAAGGTTGGCAGATGGGGTCCCATCAAGATTCAGTGGCAGTGGATC
    TGGTCAAGATTATTCTCTAACCATCAGCAGCCTGGAGTCTGACGATACAG
    CAACTTATTACTGTCTACAACATGGTGAGAGCCCTCCCACGTTCGGTGCT
    GGGACCAAGCTGGAGCTGAAACGTACGGTGGCCGCCCCCAGCGTGTTCAT
    CTTCCCCCCCAGCGATGAGCAGCTGAAGAGCGGCACCGCCAGCGTGGTGT
    GTCTGCTGAACAACTTCTACCCCCGGGAGGCCAAGGTGCAGTGGAAGGTG
    GACAATGCCCTGCAGAGCGGCAACAGCCAGGAGAGCGTGACCGAGCAGGA
    CAGCAAGGACTCCACCTACAGCCTGAGCAGCACCCTGACCCTGAGCAAGG
    CCGACTACGAGAAGCACAAGGTGTACGCCTGTGAGGTGACCCACCAGGG
    CCTGTCCAGCCCCGTGACCAAGAGCTTCAACCGGGGCGAGTGC

    Chimera* polypeptide sequence (variable light region+codon optimised human cK)
  • SEQ ID NO: 58
    DIKMTQSPSSMSASLGERVTITCQASQDIESYLSWYQQKPWKSPKTLIYY
    ATRLADGVPSRFSGSGSGQDYSLTISSLESDDTATYYCLQHGESPPTFGA
    GTKLELKRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKV
    DNALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQG
    LSSPVTKSFNRGEC

    Chimera* refers to chimeric antibody made from murine 656.35 antibody
  • H0 DNA sequence of mature heavy chain
    SEQ ID NO: 11
    CAGGTGCAGCTCGTGCAGAGCGGCGCCGAAGTGAAGAAGCCCGGGGCCAG
    CGTGAAGGTGAGCTGCAAGGCCAGCGGCTACACCTTCACCAACTACTGGA
    TCGTGTGGGTCAGGCAGGCCCCCGGCCAGGGACTGGAGTGGATGGGCGAC
    CTGTATAGCGGCGGCGGCTACACCTTCTACAGCGAGAACTTCAAGGGCAG
    GGTGACCATGACCAGGGACACCAGCACCAGCACCGTGTACATGGAGCTGA
    GCAGCCTGAGGAGCGAGGACACCGCCGTGTACTACTGCGCCAGGAGCGGC
    TACGACAGGACTTGGTTTGCTCACTGGGGCCAGGGCACACTAGTGACCGT
    GTCCAGCGCCAGCACCAAGGGCCCCAGCGTGTTCCCCCTGGCCCCCAGCA
    GCAAGAGCACCAGCGGCGGCACAGCCGCCCTGGGCTGCCTGGTGAAGGAC
    TACTTCCCCGAACCGGTGACCGTGTCCTGGAACAGCGGAGCCCTGACCAG
    CGGCGTGCACACCTTCCCCGCCGTGCTGCAGAGCAGCGGCCTGTACAGCC
    TGAGCAGCGTGGTGACCGTGCCCAGCAGCAGCCTGGGCACCCAGACCTAC
    ATCTGTAACGTGAACCACAAGCCCAGCAACACCAAGGTGGACAAGAAGGT
    GGAGCCCAAGAGCTGTGACAAGACCCACACCTGCCCCCCCTGCCCTGCCC
    CCGAGCTGCTGGGAGGCCCCAGCGTGTTCCTGTTCCCCCCCAAGCCTAAG
    GACACCCTGATGATCAGCAGAACCCCCGAGGTGACCTGTGTGGTGGTGGA
    TGTGAGCCACGAGGACCCTGAGGTGAAGTTCAACTGGTACGTGGACGGCG
    TGGAGGTGCACAATGCCAAGACCAAGCCCAGGGAGGAGCAGTACAACAGC
    ACCTACCGGGTGGTGTCCGTGCTGACCGTGCTGCACCAGGATTGGCTGAA
    CGGCAAGGAGTACAAGTGTAAGGTGTCCAACAAGGCCCTGCCTGCCCCTA
    TCGAGAAAACCATCAGCAAGGCCAAGGGCCAGCCCAGAGAGCCCCAGGTG
    TACACCCTGCCCCCTAGCAGAGATGAGCTGACCAAGAACCAGGTGTCCCT
    GACCTGCCTGGTGAAGGGCTTCTACCCCAGCGACATCGCCGTGGAGTGGG
    AGAGCAACGGCCAGCCCGAGAACAACTACAAGACCACCCCCCCTGTGCTG
    GACAGCGATGGCAGCTTCTTCCTGTACAGCAAGCTGACCGTGGACAAGAG
    CAGATGGCAGCAGGGCAACGTGTTCAGCTGCTCCGTGATGCACGAGGCCC
    TGCACAATCACTACACCCAGAAGAGCCTGAGCCTGTCCCCTGGCAAG
    H0 protein sequence of mature heavy chain
    SEQ ID NO: 46
    QVQLVQSGAEVKKPGASVKVSCKASGYTFTNYWIVWVRQAPGQGLEWMGD
    LYSGGGYTFYSENFKGRVTMTRDTSTSTVYMELSSLRSEDTAVYYCARSG
    YDRTWFAHWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKD
    YFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTY
    ICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPK
    DTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNS
    TYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQV
    YTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVL
    DSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK
    L7 DNA sequence of mature light chain
    SEQ ID NO: 47
    GATATCCAGATGACCCAGAGCCCTAGCTCCCTCAGCGCATCAGTCGGCGA
    CAGAGTGACAATCACCTGCCAGGCATCCCAGGACATCGAGTCTTACCTGA
    GCTGGTACCAGCAGAAGCCCGGAAAGGCCCCAAAGCTCCTGATCTACTAC
    GCCACTCGGCTGGCAgacGGCGTGCCTAGCAGGTTCTCCGGCTCAGGGTC
    TGGGACAGACTTCACCCTGACCATCAGCTCACTGCAGCCCGAGGATTTCG
    CCACCTACTACTGTCTGCAGCACGGAGAGAGCCCCCCAACCTTTGGCCAG
    GGAACCAAGCTGGAGATCaagCGTACGGTGGCCGCCCCCAGCGTGTTCAT
    CTTCCCCCCCAGCGATGAGCAGCTGAAGAGCGGCACCGCCAGCGTGGTGT
    GTCTGCTGAACAACTTCTACCCCCGGGAGGCCAAGGTGCAGTGGAAGGTG
    GACAATGCCCTGCAGAGCGGCAACAGCCAGGAGAGCGTGACCGAGCAGGA
    CAGCAAGGACTCCACCTACAGCCTGAGCAGCACCCTGACCCTGAGCAAGG
    CCGACTACGAGAAGCACAAGGTGTACGCCTGTGAGGTGACCCACCAGGGC
    CTGTCCAGCCCCGTGACCAAGAGCTTCAACCGGGGCGAGTGC
    L7 protein sequence of mature light chain
    SEQ ID NO: 48
    DIQMTQSPSSLSASVGDRVTITCQASQDIESYLSWYQQKPGKAPKLLIYY
    ATRLADGVPSRFSGSGSGTDFTLTISSLQPEDFATYYCLQHGESPPTFGQ
    GTKLEIKRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKV
    DNALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQG
    LSSPVTKSFNRGEC
    L8 DNA sequence of mature light chain
    SEQ ID NO: 59
    GATATCCAGATGACCCAGAGCCCTAGCTCCCTCAGCGCATCAGTCGGCGA
    CAGAGTGACAATCACCTGCCAGGCATCCCAGGACATCGAGTCTTACCTGA
    GCTGGTACCAGCAGAAGCCCGGAAAGGCCCCAAAGCTCCTGATCTACTAC
    GCCACTCGGCTGGCAgacGGCGTGCCTAGCAGGTTCTCCGGCTCAGGGTC
    TGGGACAGACTTCACCttcACCATCAGCTCACTGCAGCCCGAGGATatcG
    CCACCTACTACTGTCTGCAGCACGGAGAGAGCCCCCCAACCTTTGGCCAG
    GGAACCAAGCTGGAGATCaagCGTACGGTGGCCGCCCCCAGCGTGTTCAT
    CTTCCCCCCCAGCGATGAGCAGCTGAAGAGCGGCACCGCCAGCGTGGTGT
    GTCTGCTGAACAACTTCTACCCCCGGGAGGCCAAGGTGCAGTGGAAGGTG
    GACAATGCCCTGCAGAGCGGCAACAGCCAGGAGAGCGTGACCGAGCAGGA
    CAGCAAGGACTCCACCTACAGCCTGAGCAGCACCCTGACCCTGAGCAAGG
    CCGACTACGAGAAGCACAAGGTGTACGCCTGTGAGGTGACCCACCAGGGC
    CTGTCCAGCCCCGTGACCAAGAGCTTCAACCGGGGCGAGTGC
    L8 protein sequence of mature light chain
    SEQ ID NO: 60
    DIQMTQSPSSLSASVGDRVTITCQASQDIESYLSWYQQKPGKAPKLLIYY
    ATRLADGVPSRFSGSGSGTDFTFTISSLQPEDIATYYCLQHGESPPTFGQ
    GTKLEIKRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKV
    DNALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQG
    LSSPVTKSFNRGEC
    L10 DNA sequence of mature light chain
    SEQ ID NO: 61
    GATATCCAGATGACCCAGAGCCCTAGCTCCCTCAGCGCATCAGTCGGCGA
    CAGAGTGACAATCACCTGCCAGGCATCCCAGGACATCGAGTCTTACCTGA
    GCTGGTACCAGCAGAAGCCCGGAAAGGCCCCAAAGCTCCTGATCTACTAC
    GCCACTCGGCTGGCAgacGGCGTGCCTAGCAGGTTCTCCGGCTCAGGGTC
    TGGGcagGACtacACCCTGACCATCAGCTCACTGCAGCCCGAGGATTTCG
    CCACCTACTACTGTCTGCAGCACGGAGAGAGCCCCCCAACCTTTGGCCAG
    GGAACCAAGCTGGAGATCAAGCGTACGGTGGCCGCCCCCAGCGTGTTCAT
    CTTCCCCCCCAGCGATGAGCAGCTGAAGAGCGGCACCGCCAGCGTGGTGT
    GTCTGCTGAACAACTTCTACCCCCGGGAGGCCAAGGTGCAGTGGAAGGTG
    GACAATGCCCTGCAGAGCGGCAACAGCCAGGAGAGCGTGACCGAGCAGGA
    CAGCAAGGACTCCACCTACAGCCTGAGCAGCACCCTGACCCTGAGCAAGG
    CCGACTACGAGAAGCACAAGGTGTACGCCTGTGAGGTGACCCACCAGGGC
    CTGTCCAGCCCCGTGACCAAGAGCTTCAACCGGGGCGAGTGC
    L10 protein sequence of mature light chain
    SEQ ID NO: 62
    DIQMTQSPSSLSASVGDRVTITCQASQDIESYLSWYQQKPGKAPKLLIYY
    ATRLADGVPSRFSGSGSGQDYTLTISSLQPEDFATYYCLQHGESPPTFGQ
    GTKLEIKRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKV
    DNALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQG
    LSSPVTKSFNRGEC
    M0 DNA sequence of mature light chain
    SEQ ID NO:63
    GAGATCGTGCTGACCCAGTCTCCCGCCACCCTGTCACTGTCTCCCGGCGA
    AAGGGCAACCCTGAGCTGCCAGGCCAGCCAGGACATCGAGAGCTACCTGA
    GCTGGTACCAGCAGAAGCCCGGCCAGGCCCCCAGGCTGCTGATCTACTAC
    GCCACCAGGCTGGCCGACGGCATTCCCGCCAGGTTCAGCGGAAGCGGCAG
    CGGCACCGACTTCACTCTGACCATCAGCAGCCTGGAGCCCGAGGACTTCG
    CCGTGTACTACTGCCTGCAGCACGGCGAGAGCCCTCCCACCTTCGGCCAG
    GGCACCAAGCTCGAGATCAAGCGTACGGTGGCCGCCCCCAGCGTGTTCAT
    CTTCCCCCCCAGCGATGAGCAGCTGAAGAGCGGCACCGCCAGCGTGGTGT
    GTCTGCTGAACAACTTCTACCCCCGGGAGGCCAAGGTGCAGTGGAAGGTG
    GACAATGCCCTGCAGAGCGGCAACAGCCAGGAGAGCGTGACCGAGCAGGA
    CAGCAAGGACTCCACCTACAGCCTGAGCAGCACCCTGACCCTGAGCAAGG
    CCGACTACGAGAAGCACAAGGTGTACGCCTGTGAGGTGACCCACCAGGGC
    CTGTCCAGCCCCGTGACCAAGAGCTTCAACCGGGGCGAGTGC
    M0 protein sequence of mature light chain
    SEQ ID NO: 64
    EIVLTQSPATLSLSPGERATLSCQASQDIESYLSWYQQKPGQAPRLLIYY
    ATRLADGIPARFSGSGSGTDFTLTISSLEPEDFAVYYCLQHGESPPTFGQ
    GTKLEIKRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKV
    DNALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQG
    LSSPVTKSFNRGEC

Claims (4)

1. An antibody comprising heavy and light chains comprising the amino acid sequences of SEQ ID NO:46 and SEQ ID NO:48, respectively.
2. An antibody comprising heavy and light chains comprising the amino acid sequences of SEQ ID NO:46 and SEQ ID NO: 62, respectively.
3. An antibody comprising heavy and light chains comprising the amino acid sequences of SEQ ID NO:46 and SEQ ID NO: 60, respectively.
4. An antibody comprising heavy and light chains comprising the amino acid sequences of SEQ ID NO:46 and SEQ ID NO: 64, respectively
US12/103,761 2007-04-17 2008-04-16 Novel compounds Abandoned US20080262203A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/103,761 US20080262203A1 (en) 2007-04-17 2008-04-16 Novel compounds

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US91222907P 2007-04-17 2007-04-17
US4413208P 2008-04-11 2008-04-11
US12/103,761 US20080262203A1 (en) 2007-04-17 2008-04-16 Novel compounds

Publications (1)

Publication Number Publication Date
US20080262203A1 true US20080262203A1 (en) 2008-10-23

Family

ID=39872910

Family Applications (3)

Application Number Title Priority Date Filing Date
US12/103,761 Abandoned US20080262203A1 (en) 2007-04-17 2008-04-16 Novel compounds
US12/596,262 Expired - Fee Related US8496932B2 (en) 2007-04-17 2008-04-16 Compounds
US13/927,352 Expired - Fee Related US8828384B2 (en) 2007-04-17 2013-06-26 Compounds

Family Applications After (2)

Application Number Title Priority Date Filing Date
US12/596,262 Expired - Fee Related US8496932B2 (en) 2007-04-17 2008-04-16 Compounds
US13/927,352 Expired - Fee Related US8828384B2 (en) 2007-04-17 2013-06-26 Compounds

Country Status (30)

Country Link
US (3) US20080262203A1 (en)
EP (1) EP2146745B1 (en)
JP (1) JP5441885B2 (en)
KR (1) KR101560841B1 (en)
CN (1) CN101687035B (en)
AU (1) AU2008243049B2 (en)
BR (1) BRPI0810250A8 (en)
CA (1) CA2684626C (en)
CL (1) CL2008001071A1 (en)
CO (1) CO6251323A2 (en)
CR (1) CR11116A (en)
CY (1) CY1116671T1 (en)
DK (1) DK2146745T3 (en)
DO (1) DOP2009000236A (en)
EA (1) EA024621B1 (en)
ES (1) ES2547475T3 (en)
HK (1) HK1136970A1 (en)
HR (1) HRP20150937T1 (en)
HU (1) HUE027584T2 (en)
IL (1) IL201440A (en)
MA (1) MA31310B1 (en)
MX (1) MX2009011153A (en)
MY (1) MY150525A (en)
NZ (1) NZ580239A (en)
PE (1) PE20090302A1 (en)
PL (1) PL2146745T3 (en)
PT (1) PT2146745E (en)
SI (1) SI2146745T1 (en)
TW (1) TWI473815B (en)
WO (1) WO2008130969A2 (en)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011071883A1 (en) * 2009-12-07 2011-06-16 Decimmune Therapeutics, Inc. Anti-inflammatory antibodies and uses therefor
EP2534256A1 (en) * 2010-02-09 2012-12-19 GlaxoSmithKline LLC Novel uses
WO2013166099A1 (en) * 2012-05-01 2013-11-07 Glaxosmithkline Llc Novel antibodies
EP2796875A1 (en) * 2013-04-23 2014-10-29 Albert-Ludwigs-Universität Freiburg Inhibitors of proteins specific for the secretome of a chondrocyte for use in the treatment of breast cancer metastasis
US9243059B2 (en) 2013-03-12 2016-01-26 Decimmune Therapeutics, Inc. Humanized anti-N2 antibodies and methods of treating ischemia-reperfusion injury
WO2019182867A1 (en) * 2018-03-23 2019-09-26 Board Of Regents, The University Of Texas System Dual specificity antibodies to human pd-l1 and pd-l2 and methods of use therefor
US20220080044A1 (en) * 2020-09-14 2022-03-17 National Yang-Ming University Method for preventing or treating peripheral arterial occlusive disease
US11525002B2 (en) 2017-10-11 2022-12-13 Board Of Regents, The University Of Texas System Human PD-L1 antibodies and methods of use therefor

Families Citing this family (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11046784B2 (en) 2006-03-31 2021-06-29 Chugai Seiyaku Kabushiki Kaisha Methods for controlling blood pharmacokinetics of antibodies
US9096651B2 (en) 2007-09-26 2015-08-04 Chugai Seiyaku Kabushiki Kaisha Method of modifying isoelectric point of antibody via amino acid substitution in CDR
LT2708559T (en) 2008-04-11 2018-06-11 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding to two or more antigen molecules repeatedly
TWI812066B (en) 2010-11-30 2023-08-11 日商中外製藥股份有限公司 Antibody having calcium-dependent antigen-binding ability
TW202237660A (en) 2012-08-24 2022-10-01 日商中外製藥股份有限公司 Fcγriib-specific fc region variant
WO2014030750A1 (en) 2012-08-24 2014-02-27 中外製薬株式会社 MOUSE FcγRII-SPECIFIC Fc ANTIBODY
US9290570B2 (en) 2013-03-15 2016-03-22 Eli Lilly And Company Pan-ELR+ CXC Chemokine Antibodies
JO3580B1 (en) 2013-03-15 2020-07-05 Lilly Co Eli Pan-ELR+ CXC CHEMOKINE ANTIBODIES
AU2014250434B2 (en) 2013-04-02 2019-08-08 Chugai Seiyaku Kabushiki Kaisha Fc region variant
SG11201509284VA (en) * 2013-05-17 2015-12-30 Centre Nat Rech Scient Anti-cxcl1, cxcl7 and cxcl8 antibodies and their applications
KR102650420B1 (en) 2014-12-19 2024-03-21 추가이 세이야쿠 가부시키가이샤 Anti-myostatin antibodies, polypeptides containing variant fc regions, and methods of use
SG10201907215QA (en) 2015-02-05 2019-09-27 Chugai Pharmaceutical Co Ltd Antibodies Comprising An Ion Concentration Dependent Antigen-Binding Domain, Fc Region Variants, Il-8-Binding Antibodies, And Uses Therof
WO2017110981A1 (en) 2015-12-25 2017-06-29 Chugai Seiyaku Kabushiki Kaisha Anti-myostatin antibodies and methods of use
CN116271014A (en) 2016-08-05 2023-06-23 中外制药株式会社 Compositions for preventing or treating IL-8 related diseases
SG10201607778XA (en) 2016-09-16 2018-04-27 Chugai Pharmaceutical Co Ltd Anti-Dengue Virus Antibodies, Polypeptides Containing Variant Fc Regions, And Methods Of Use
EP3684811A2 (en) * 2017-08-17 2020-07-29 Massachusetts Institute of Technology Multiple specificity binders of cxc chemokines and uses thereof
CN112119090B (en) 2018-03-15 2023-01-13 中外制药株式会社 Anti-dengue virus antibodies cross-reactive to Zika virus and methods of use
MX2022006132A (en) * 2019-12-05 2022-06-17 Seagen Inc Anti-avb6 antibodies and antibody-drug conjugates.

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070161089A1 (en) * 2005-11-08 2007-07-12 Genentech, Inc. Method of Producing Pan-Specific Antibodies

Family Cites Families (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IE913192A1 (en) 1990-09-12 1992-02-25 Scripps Research Inst Active site of interleukin-8: polypeptide analogs and¹antibodies
US6448379B1 (en) * 1993-09-14 2002-09-10 Chiron Corporation IL8 inhibitors
DK0833911T3 (en) * 1995-06-07 2004-08-30 Ortho Mcneil Pharm Inc CDR grafted anti-tissue factor antibodies and methods for their use
GB0020685D0 (en) * 2000-08-22 2000-10-11 Novartis Ag Organic compounds
AR030753A1 (en) * 2000-09-25 2003-09-03 Smithkline Beecham Corp USE OF MODULATORS-PROTEIN OF IL-8, GROALFA, GROBETA, GROGAMMA, NAP-2, AND ENA-78, TO TREAT VIRIC INFECTIONS
TR200602095T2 (en) * 2000-10-13 2007-02-21 Biogen Idec Ma Inc. Humanized Anti-LT-Beta-R antibodies
DE60234933D1 (en) 2001-04-26 2010-02-11 Dako Denmark As PAN-SPECIFIC MONOCLONAL ANTIBODY OF THE GLYCOSILIZED HEMOGLOBIN
US20040022726A1 (en) * 2002-06-03 2004-02-05 Goldenberg David M. Methods and compositions for intravesical therapy of bladder cancer
ES2373947T3 (en) * 2002-12-16 2012-02-10 Genmab A/S HUMAN MONOCLONAL ANTIBODIES AGAINST INTERLEUCINE 8 (IL-8).
AU2003303543A1 (en) * 2002-12-26 2004-07-29 Chugai Seiyaku Kabushiki Kaisha Agonist antibody against heteroreceptor
EP1610818A4 (en) * 2004-03-03 2007-09-19 Millennium Pharm Inc Modified antibodies to prostate-specific membrane antigen and uses thereof
WO2006074399A2 (en) * 2005-01-05 2006-07-13 Biogen Idec Ma Inc. Multispecific binding molecules comprising connecting peptides
CN101282993A (en) * 2005-06-02 2008-10-08 阿斯利康公司 Antibodies directed to cd20 and uses thereof
ES2452115T3 (en) * 2005-06-17 2014-03-31 Imclone Llc An anti-PDGFRalpha antibody for use in the treatment of metastatic bone cancer
GB0519398D0 (en) * 2005-09-23 2005-11-02 Antisoma Plc Biological materials and uses thereof
US8454960B2 (en) * 2008-01-03 2013-06-04 The Scripps Research Institute Multispecific antibody targeting and multivalency through modular recognition domains

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070161089A1 (en) * 2005-11-08 2007-07-12 Genentech, Inc. Method of Producing Pan-Specific Antibodies

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8324352B2 (en) 2009-12-07 2012-12-04 Decimmune Therapeutics, Inc. Anti-inflammatory antibodies and uses therefor
WO2011071883A1 (en) * 2009-12-07 2011-06-16 Decimmune Therapeutics, Inc. Anti-inflammatory antibodies and uses therefor
EP2534256A1 (en) * 2010-02-09 2012-12-19 GlaxoSmithKline LLC Novel uses
EP2534256A4 (en) * 2010-02-09 2013-07-24 Glaxosmithkline Llc Novel uses
EP2844290A4 (en) * 2012-05-01 2015-12-16 Glaxosmithkline Llc Novel antibodies
WO2013166099A1 (en) * 2012-05-01 2013-11-07 Glaxosmithkline Llc Novel antibodies
US9605058B2 (en) 2012-05-01 2017-03-28 Glaxosmithkline Llc Antibodies against the CXC-ELR family of chemokines
US9409977B2 (en) 2013-03-12 2016-08-09 Decimmune Therapeutics, Inc. Humanized, anti-N2 antibodies
US9243059B2 (en) 2013-03-12 2016-01-26 Decimmune Therapeutics, Inc. Humanized anti-N2 antibodies and methods of treating ischemia-reperfusion injury
WO2014173967A1 (en) * 2013-04-23 2014-10-30 Albert-Ludwigs-Universität Freiburg Inhibitors of proteins specific for the secretome of a chondrocyte for use in the treatment of breast cancer metastasis
EP2796875A1 (en) * 2013-04-23 2014-10-29 Albert-Ludwigs-Universität Freiburg Inhibitors of proteins specific for the secretome of a chondrocyte for use in the treatment of breast cancer metastasis
US10495642B2 (en) 2013-04-23 2019-12-03 Albert-Ludwigs-Universität Freiburg Inhibitors of proteins specific for the secretome of a chondrocyte for use in the treatment of breast cancer metastasis
US11525002B2 (en) 2017-10-11 2022-12-13 Board Of Regents, The University Of Texas System Human PD-L1 antibodies and methods of use therefor
WO2019182867A1 (en) * 2018-03-23 2019-09-26 Board Of Regents, The University Of Texas System Dual specificity antibodies to human pd-l1 and pd-l2 and methods of use therefor
US20220080044A1 (en) * 2020-09-14 2022-03-17 National Yang-Ming University Method for preventing or treating peripheral arterial occlusive disease

Also Published As

Publication number Publication date
US20110229475A1 (en) 2011-09-22
ES2547475T3 (en) 2015-10-06
WO2008130969A2 (en) 2008-10-30
AU2008243049B2 (en) 2011-04-21
CO6251323A2 (en) 2011-02-21
PT2146745E (en) 2015-10-14
CA2684626A1 (en) 2008-10-30
CL2008001071A1 (en) 2009-05-22
SI2146745T1 (en) 2015-10-30
US8828384B2 (en) 2014-09-09
EA024621B1 (en) 2016-10-31
AU2008243049A1 (en) 2008-10-30
KR20100020946A (en) 2010-02-23
MX2009011153A (en) 2009-10-30
PE20090302A1 (en) 2009-04-03
NZ580239A (en) 2012-07-27
CA2684626C (en) 2014-11-04
HRP20150937T1 (en) 2015-10-09
HUE027584T2 (en) 2016-11-28
HK1136970A1 (en) 2010-07-16
CR11116A (en) 2010-03-22
CN101687035B (en) 2013-06-12
DOP2009000236A (en) 2009-11-15
PL2146745T3 (en) 2015-11-30
IL201440A0 (en) 2010-06-16
EP2146745B1 (en) 2015-06-17
TW200906855A (en) 2009-02-16
KR101560841B1 (en) 2015-11-11
JP5441885B2 (en) 2014-03-12
DK2146745T3 (en) 2015-09-14
BRPI0810250A2 (en) 2014-10-14
EA200970954A1 (en) 2010-08-30
MA31310B1 (en) 2010-04-01
IL201440A (en) 2016-08-31
US20140010807A1 (en) 2014-01-09
CN101687035A (en) 2010-03-31
JP2010524469A (en) 2010-07-22
US8496932B2 (en) 2013-07-30
EP2146745A2 (en) 2010-01-27
MY150525A (en) 2014-01-30
WO2008130969A3 (en) 2008-12-18
CY1116671T1 (en) 2017-03-15
TWI473815B (en) 2015-02-21
EP2146745A4 (en) 2011-11-30
BRPI0810250A8 (en) 2017-02-21

Similar Documents

Publication Publication Date Title
US8828384B2 (en) Compounds
US20110105724A1 (en) Novel compounds
US20120315267A1 (en) Novel uses
JP4809828B2 (en) Immunoglobulin
US9193784B2 (en) Antibodies
US8940303B2 (en) CD127 binding proteins
CN113825765A (en) Method for producing anti-IL 12/IL23 antibody composition
JP7104463B2 (en) Trispecific antibodies against IL-17A, IL-17F, and other pro-inflammatory molecules
CA3139508A1 (en) Materials and methods for modulating t cell mediated immunity
CN117916260A (en) Methods of manufacture for preparing anti-IL 12/IL23 antibody compositions
KR20240034215A (en) Manufacturing Methods for Producing Anti-IL12/IL23 Antibody Compositions
JP2023514567A (en) Safe and effective methods of treating ulcerative colitis with anti-IL12/IL23 antibodies

Legal Events

Date Code Title Description
AS Assignment

Owner name: SMITHKLINE BEECHAM CORPORATION, PENNSYLVANIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:DOBRZYNSKI, ERIC;GODILLOT, ALEXIS PAUL;JONAK, ZDENKA LUDMILA;AND OTHERS;REEL/FRAME:021512/0003

Effective date: 20080630

AS Assignment

Owner name: SMITHKLINE BEECHAM CORPORATION, PENNSYLVANIA

Free format text: RE-RECORD TO ADD THE NAMES OF THE PREVIOUSLY OMITTED CONVEYING PARTIES, PREVIOUSLY RECORDED ON REEL 021512 FRAME 0003.;ASSIGNORS:DOBRZYNSKI, ERIC;GODILLOT, ALEX PAUL;JONAK, ZDENKA LUDMILA;AND OTHERS;REEL/FRAME:021553/0624;SIGNING DATES FROM 20080630 TO 20080703

STCB Information on status: application discontinuation

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