WO2006013107A1 - Il-17 antagonistic antibodies - Google Patents

Il-17 antagonistic antibodies Download PDF

Info

Publication number
WO2006013107A1
WO2006013107A1 PCT/EP2005/008470 EP2005008470W WO2006013107A1 WO 2006013107 A1 WO2006013107 A1 WO 2006013107A1 EP 2005008470 W EP2005008470 W EP 2005008470W WO 2006013107 A1 WO2006013107 A1 WO 2006013107A1
Authority
WO
WIPO (PCT)
Prior art keywords
amino acid
seq
acid sequence
binding molecule
cdr3
Prior art date
Application number
PCT/EP2005/008470
Other languages
French (fr)
Inventor
Franco E. Di Padova
Hermann Gram
Hans Hofstetter
Margit Jeschke
Jean-Michel Rondeau
Wim Van Den Berg
Original Assignee
Novartis Ag
Novartis Pharma Gmbh
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=32982602&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2006013107(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority to CA2573586A priority Critical patent/CA2573586C/en
Priority to AU2005268857A priority patent/AU2005268857C1/en
Priority to BR122018075556-3A priority patent/BR122018075556B1/en
Priority to BR122017009404-1A priority patent/BR122017009404B1/en
Priority to SI200531390T priority patent/SI1776142T1/en
Priority to US11/658,344 priority patent/US7807155B2/en
Priority to PL05770286T priority patent/PL1776142T4/en
Priority to JP2007524286A priority patent/JP4682200B2/en
Priority to NZ552658A priority patent/NZ552658A/en
Priority to MX2007001338A priority patent/MX2007001338A/en
Priority to EP05770286.2A priority patent/EP1776142B9/en
Priority to EP18166164.6A priority patent/EP3409288A1/en
Priority to AT05770286T priority patent/ATE517924T1/en
Priority to CN2005800265694A priority patent/CN101001645B/en
Priority to ES05770286.2T priority patent/ES2367440T7/en
Priority to BRPI0513078A priority patent/BRPI0513078C1/en
Priority to DK05770286.2T priority patent/DK1776142T6/en
Application filed by Novartis Ag, Novartis Pharma Gmbh filed Critical Novartis Ag
Publication of WO2006013107A1 publication Critical patent/WO2006013107A1/en
Priority to IL180717A priority patent/IL180717A/en
Priority to TNP2007000034A priority patent/TNSN07034A1/en
Priority to NO20070985A priority patent/NO336279B1/en
Priority to HK07109203.7A priority patent/HK1101277A1/en
Priority to US12/707,934 priority patent/US8119131B2/en
Priority to AU2010201689A priority patent/AU2010201689B2/en
Priority to HRP20110758TT priority patent/HRP20110758T4/en
Priority to US13/349,689 priority patent/US8617552B2/en
Priority to US14/085,074 priority patent/US20140079719A1/en
Priority to NO20150065A priority patent/NO337129B1/en
Priority to NO20150064A priority patent/NO337286B1/en
Priority to US14/625,073 priority patent/US9765140B2/en
Priority to FR15C0048C priority patent/FR15C0048I2/en
Priority to NL300749C priority patent/NL300749I2/en
Priority to LTPA2015029C priority patent/LTC1776142I2/en
Priority to CY2015030C priority patent/CY2015030I2/en
Priority to NO2015023C priority patent/NO2015023I1/en
Priority to NO20151787A priority patent/NO341384B1/en
Priority to NO2016017C priority patent/NO2016017I1/en
Priority to US15/674,970 priority patent/US10344084B2/en
Priority to NO20171697A priority patent/NO20171697A1/en
Priority to NO2018007C priority patent/NO2018007I1/en
Priority to US16/412,543 priority patent/US20190270804A1/en
Priority to US17/859,653 priority patent/US20230235038A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • 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
    • 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
    • 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
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/55Fab or Fab'
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value

Definitions

  • This invention relates to an IL- 17 binding molecule, in particular an antibody to human IL- 17, more preferably a human antibody to human EL- 17 (also named IL- 17A) and to the use of such antibodies in the treatment of IL-17 mediated diseases and disorders.
  • an IL- 17 binding molecule in particular an antibody to human IL- 17, more preferably a human antibody to human EL- 17 (also named IL- 17A) and to the use of such antibodies in the treatment of IL-17 mediated diseases and disorders.
  • IL- 17 a T-cell derived cytokine present e.g. in rheumatoid arthritis (RA), acts as a pro- inflammatory cytokine, particularly in conjunction with EL-I and TNF- ⁇ (Chabaud M & Miossec P (1999) Arthritis Rheum 42, 963-970; Awane M et al (1999) J. Immunol 162, 5337-5344).
  • IL- 17 induces MMP production and downregulates TIMP (Jovanovic DV et al (2001) J. Rheumatol.
  • IL-17 blockage of IL-I and IL-17 has a synergistic effect on inflammation and bone destruction in vivo (Chabaud M & Miossec (2001) Arthritis Rheum 44, 1293-1303). Inappropriate or excessive production of IL-17 is associated with the pathology of various diseases and disorders, such as rheumatoid arthritis (W ⁇ towski et al., 2004 Cell MoI Life Sci 61:567-579), osteoarthritis, loosening of bone implants, acute transplant rejection (Antonysamy et al.
  • the invention provides an DL- 17 binding molecule which comprises an antigen binding site comprising at least one immunoglobulin heavy chain variable domain (V H ) which comprises in sequence hypervariable regions CDRl, CDR2 and CDR3, said CDRl having the amino acid sequence SEQ DD NO: 1 (N-Y-W-M-N), said CDR2 having the amino acid sequence SEQ ID NO: 2 (A-I-N-Q-D-G-S-E-K-Y-Y-V-G-S-V-K-G), and said CDR3 having the amino acid sequence SEQ ID NO: 3 (D-Y-Y-D-I-L-T-D-Y-Y-I-H-Y-W-Y-F-D-L); or direct CDR equivalents thereof.
  • V H immunoglobulin heavy chain variable domain
  • the invention also provides an IL- 17 binding molecule comprising at least one immunoglobulin light chain variable domain (VL) which comprises in sequence hypervariable regions CDRl', CDR2' and CDR3', said CDRl' having the amino acid sequence SEQ ID NO: 4 (R-A-S-Q-S-V-S-S-S-Y-L-A), said CDR2' having the amino acid sequence SEQ ID NO: 5 (G-A-S-S-R-A-T) and said CDR3' having the amino acid sequence SEQ ID NO: 6 (Q- Q-Y-G-S-S-P-C-T) or direct CDR' equivalents thereof.
  • VL immunoglobulin light chain variable domain
  • the invention provides an IL- 17 binding molecule which comprises an antigen binding site comprising at least one immunoglobulin heavy chain variable domain (VH) which comprises in sequence hypervariable regions CDRl -x, CDR2- x and CDR3-X, said CDRl-x having the amino acid sequence SEQ ID NO: 11 (G-F-T-F-S-N-Y- W-M-N), said CDR2-X having the amino acid sequence SEQ ID NO: 12 (A-I-N-Q-D-G-S-E-K- Y-Y), and said CDR3-X having the amino acid sequence SEQ ID NO: 13 (C-V-R-D-Y-Y-D-I-L- T-D-Y-Y-I-H-Y-W-Y-F-D-L-W-G); or direct CDR-x equivalents thereof.
  • VH immunoglobulin heavy chain variable domain
  • the invention also provides an IL- 17 binding molecule comprising both heavy (V H ) and light chain (V L ) variable domains; said IL- 17 binding molecule comprises at least one antigen binding site comprising: a) an immunoglobulin heavy chain variable domain (V H ) which comprises in sequence hypervariable regions CDRl, CDR2 and CDR3, said CDRl having the amino acid sequence SEQ ID NO:1, said CDR2 having the amino acid sequence SEQ ID NO:2, and said CDR3 having the amino acid sequence SEQ ID NO:3 or direct CDR equivalents thereof; and b) an immunoglobulin light chain variable domain (VL) which comprises in sequence hypervariable regions CDRl', CDR2' and CDR3', said CDRl' having the amino acid sequence SEQ ID NO:4, said CDR2' having the amino acid sequence SEQ ID NO:5, and said CDR3' having the amino acid sequence SEQ ID NO: 6 or direct CDR' equivalents thereof.
  • V H immunoglobulin heavy chain variable domain
  • the invention also provides an IL- 17 binding molecule comprising both heavy (VH) and light chain (V L ) variable domains; said IL- 17 binding molecule comprises at least one antigen binding site comprising: a) an immunoglobulin heavy chain variable domain (VH) which comprises in sequence hypervariable regions CDRl -x, CDR2-X and CDR3-X, said CDRl -x having the amino acid sequence SEQ ID NO:11, said CDR2-X having the amino acid sequence SEQ ID NO: 12, and said CDR3-X having the amino acid sequence SEQ ID NO: 13 or direct CDR- x equivalents thereof; and b) an immunoglobulin light chain variable domain (V L ) which comprises in sequence hypervariable regions CDRl', CDR2' and CDR3', said CDRl' having the amino acid sequence SEQ ID NO:4, said CDR2' having the amino acid sequence SEQ ID NO:5, and said CDR3' having the amino acid sequence SEQ ID NO: 6 or direct CDR'
  • any polypeptide chain is herein described as having an amino acid sequence starting at the N-terminal extremity and ending at the C-terminal extremity.
  • the antigen binding site comprises both the V H and V L domains, these may be located on the same polypeptide molecule or, preferably, each domain may be on a different chain, the V H domain being part of an immunoglobulin heavy chain or fragment thereof and the V L being part of an immunoglobulin light chain or fragment thereof.
  • EL- 17 binding molecule any molecule capable of binding to the IL- 17 antigen either alone or associated with other molecules.
  • the binding reaction may be shown by standard methods (qualitative assays) including, for example, a binding assay, competition assay or a bioassay for determining the inhibition of DL-17 binding to its receptor or any kind of binding assays, with reference to a negative control test in which an antibody of unrelated specificity but of the same isotype, e.g. an anti-CD25 antibody, is used (see also Example 1).
  • antigen binding molecules include antibodies as produced by B-cells or hybridomas and chimeric, CDR-grafted or human antibodies or any fragment thereof, e.g. F(ab') 2 and Fab fragments, as well as single chain or single domain antibodies.
  • a single chain antibody consists of the variable domains of the heavy and light chains of an antibody covalently bound by a peptide linker usually consisting of from 10 to 30 amino acids, preferably from 15 to 25 amino acids. Therefore, such a structure does not include the constant part of the heavy and light chains and it is believed that the small peptide spacer should be less antigenic than a whole constant part.
  • chimeric antibody is meant an antibody in which the constant regions of heavy or light chains or both are of human origin while the variable domains of both heavy and light chains are of non-human (e.g. murine) origin or of human origin but derived from a different human antibody.
  • CDR-grafted antibody is meant an antibody in which the hypervariable regions (CDRs) are derived from a donor antibody, such as a non-human (e.g. murine) antibody or a different human antibody, while all or substantially all the other parts of the immunoglobulin e.g. the constant regions and the highly conserved parts of the variable domains, i.e. the framework regions, are derived from an acceptor antibody, e.g. an antibody of human origin.
  • a CDR-grafted antibody may however contain a few amino acids of the donor sequence in the framework regions, for instance in the parts of the framework regions adjacent to the hypervariable regions.
  • human antibody is meant an antibody in which the constant and variable regions of both the heavy and light chains are all of human origin, or substantially identical to sequences of human origin, not necessarily from the same antibody and includes antibodies produced by mice in which the murine immunoglobulin variable and constant part genes have been replaced by their human counterparts, e.g. as described in general terms in EP 0546073 Bl, USP 5545806, USP 5569825, USP 5625126, USP 5633425, USP 5661016, USP 5770429, EP 0 438474 Bl and EP 0463151 Bl.
  • Particularly preferred EL- 17 binding molecules of the invention are human antibodies, especially the AIN457 antibody as hereinafter described in Examples 1 and 2.
  • the constant region domains preferably also comprise suitable human constant region domains, for instance as described in "Sequences of Proteins of Immunological Interest", Kabat E.A. et al, US Department of Health and Human Services, Public Health Service, National Institute of Health. Hypervariable regions may be associated with any kind of framework regions, though preferably are of human origin.
  • the preferred heavy chain framework is a human heavy chain framework, for instance that of the AIN457 antibody. It consists in sequence e.g. of FRl (amino acid 1 to 30 of SEQ ID NO: 8), FR2 (amino acid 36 to 49 of SEQ ID NO: 8), FR3 (amino acid 67 to 98 of SEQ ID NO: 8) and FR4 (amino acid 117 to 127 of SEQ ID NO: 8) regions.
  • another preferred heavy chain framework consists in sequence of FRl -x (amino acid 1 to 25 of SEQ ID NO: 8), FR2-X (amino acid 36 to 49 of SEQ ID NO: 8), FR3-x (amino acid 61 to 95 of SEQ ID NO: 8) and FR4 (amino acid 119 to 127 of SEQ ID NO: 8) regions.
  • the light chain framework consists, in sequence, of FRl' (amino acid 1 to 23 of SEQ ID NO: 10), FR2' (amino acid 36 to 50 of SEQ ID NO: 10), FR3' (amino acid 58 to 89 of SEQ ID NO: 10) and FR4' (amino acid 99 to 109 of SEQ ID NO: 10) regions.
  • the invention also provides an IL- 17 binding molecule which comprises at least one antigen binding site comprising either a first domain having an amino acid sequence substantially identical to that shown in SEQ ID NO: 8 starting with the amino acid at position 1 and ending with the amino acid at position 127 or a first domain as described above and a second domain having an amino acid sequence substantially identical to that shown in SEQ ID NO: 10, starting with the amino acid at position 1 and ending with the amino acid at position 109.
  • Monoclonal antibodies raised against a protein naturally found in all humans are typically developed in a non-human system e.g. in mice, and as such are typically non-human proteins.
  • a xenogenic antibody as produced by a hybridoma when administered to humans, elicits an undesirable immune response which is predominantly mediated by the constant part of the xenogenic immunoglobulin.
  • a more preferred IL- 17 binding molecule of the invention is selected from a human anti IL- 17 antibody which comprises at least a) an immunoglobulin heavy chain or fragment thereof which comprises (i) a variable domain comprising in sequence the hypervariable regions CDRl, CDR2 and CDR3 or direct CDR equivalents thereof and (ii) the constant part or fragment thereof of a human heavy chain; said CDRl having the amino acid sequence SEQ DD NO: 1, said CDR2 having the amino acid sequence SEQ ID NO: 2, and said CDR3 having the amino acid sequence SEQ ID NO: 3; and b) an immunoglobulin light chain or fragment thereof which comprises (i) a variable domain comprising in sequence the hypervariable regions and optionally also the CDRl', CDR2', and CDR3' hypervariable regions or direct CDR' equivalents thereof and (ii) the constant part or fragment thereof of a human light chain, said CDRl ' having the amino acid sequence SEQ DD NO: 4, said C
  • an IL- 17 binding molecule of the invention may be selected from a single chain binding molecule which comprises an antigen binding site comprising a) a first domain comprising in sequence the hypervariable regions CDRl, CDR2 and CDR3 or direct CDR equivalents thereof, said CDRl having the amino acid sequence SEQ ID NO: 1, said CDR2 having the amino acid sequence SEQ ID NO: 2, and said CDR3 having the amino acid sequence SEQ ID NO: 3; and b) a second domain comprising the hypervariable regions CDRl', CDR2' and CDR3' or direct CDR' equivalents thereof, said CDRl' having the amino acid sequence SEQ ID NO: 4, said CDR2' having the amino acid sequence SEQ ID NO: 5, and said CDR3' having the amino acid sequence SEQ ID NO: 6; and c ) a peptide linker which is bound either to the N-terminal extremity of the first domain and to the C-terminal extremity of the second domain or to the C-terminal extremity of
  • directly CDR equivalents thereof are meant IL- 17 binding molecules comprising in sequence the hypervariable regions CDRl;, CDR2i ; and CDR3j, (instead of CDRl , CDR2, and CDR3), wherein (i) the hypervariable region CDRIj differs by 3, preferably 2, more preferably 1 amino acid(s) from the hypervariable region CDRl as shown in SEQ ID NO: 1; and
  • the hypervariable region CDR2j differs by 3, preferably 2, more preferably 1 amino acid(s) from the hypervariable region CDR2 as shown in SEQ ID NO: 2;
  • the hypervariable region CDR3j differs by 3, preferably 2, more preferably 1 amino acid(s) from the hypervariable region CDR3 as shown in SEQ ID NO: 3;
  • direct CDR-x equivalents thereof are meant IL- 17 binding molecules comprising in sequence the hypervariable regions CDRlj-x, CDR2j-x, and CDR3i-x, (instead of CDRl -x, CDR2-X, and CDR3-x), wherein
  • the hypervariable region CDRl ,-x differs by 3, preferably 2, more preferably 1 amino acid(s) from the hypervariable region CDRl -x as shown in SEQ ID NO: 11 ;
  • the hypervariable region CDR2j-x differs by 3, preferably 2, more preferably 1 amino acid(s) from the hypervariable region CDR2-X as shown in SEQ ID NO: 12;
  • the hypervariable region CDR3;-x differs by 3, preferably 2, more preferably 1 amino acid(s) from the hypervariable region CDR3-X as shown in SEQ ID NO: 13;
  • direct CDR' equivalents thereof is meant a domain comprising in sequence the hypervariable regions CDRl 'j, CDR2'i, and CDR3'j, wherein (i) the hypervariable region CDRl 'i differs by 3, preferably 2, more preferably 1 amino acid(s) from the hypervariable region CDRl' as shown in SEQ ID NO: 4; and
  • the hypervariable region CDR2'j differs by 3, preferably 2, more preferably 1 amino acid(s) from the hypervariable region CDR2' as shown in SEQ ID NO: 5;
  • the hypervariable region CDR3'i differs by 3, preferably 2, more preferably 1 amino acid(s) from the hypervariable region CDR3' as shown in SEQ ID NO: 6;
  • an IL- 17 binding molecule of the invention may be an IL- 17 binding molecule which comprises at least one antigen binding site comprising at least one immunoglobulin heavy chain variable domain (V H ) which comprises in sequence a) hypervariable regions CDRl (SEQ ID NO: 1), CDR2 (SEQ ID NO: 2) and CDR3 (SEQ ID NO: 3); or b) hypervariable regions CDRIi, CDR2j, CDR3j, said hypervariable region CDRl; differs by 3, preferably 2, more preferably 1 amino acid(s) from the hypervariable region of CDRl as shown in SEQ ID NO: 1, said hypervariable region CDR2j differs by 3, preferably 2, more preferably 1 amino acid(s) from the hypervariable region of CDR2 as shown in SEQ ID NO: 2; and said hypervariable region CDR3j differs by 3, preferably 2, more preferably 1 amino acid(s) from the hypervariable region of CDR3 as shown in SEQ ID NO: 3; and said binding
  • an IL- 17 binding molecule of the invention may be an IL- 17 binding molecule which comprises at least one antigen binding site comprising at least one immunoglobulin heavy chain variable domain (V H ) which comprises in sequence a) hypervariable regions CDRl -x (SEQ ID NO: 11), CDR2-X (SEQ ID NO: 12) and CDR3-X (SEQ ID NO: 13); or b) hypervariable regions CDRlj-x, CDR2j-x, CDR3i-x, said hypervariable region CDRlj-x differs by 3, preferably 2, more preferably 1 amino acid(s) from the hypervariable region of CDRl-x as shown in SEQ ID NO: 11, said hypervariable region CDR2j-x differs by 3, preferably
  • an IL-17 binding molecule of the invention may be a IL-17 binding molecule comprising both heavy (V H ) and light chain (V L ) variable domains and said IL-17 binding molecule comprises at least one antigen binding site comprising: a) an immunoglobulin heavy chain variable domain (VH) which comprises in sequence hypervariable regions CDRl (SEQ ID NO: 1), CDR2 (SEQ ID NO: 2) and CDR3 (SEQ ID NO: 3); and an immunoglobulin light chain variable domain (V L ) which comprises in sequence hypervariable regions CDRl' (SEQ ID NO: 4), CDR2' (SEQ ID NO: 5) and CDR3' (SEQ ID NO: 6); or b) an immunoglobulin heavy chain variable domain (VH) which comprises in sequence hypervariable regions CDRIj, CDR2j, and CDR3j, said hypervariable region hypervariable regions CDRIj, CDR2j, CDR3j, said hypervariable region CDRIj differs by 3,
  • an IL-17 binding molecule of the invention may be a IL-17 binding molecule comprising both heavy (VH) and light chain (V L ) variable domains and said IL-17 binding molecule comprises at least one antigen binding site comprising: a) an immunoglobulin heavy chain variable domain (VH) which comprises in sequence hypervariable regions CDRl-x (SEQ ID NO: 11), CDR2-X (SEQ ID NO: 12) and CDR3-X (SEQ ID NO: 13); and an immunoglobulin light chain variable domain (V L ) which comprises in sequence hypervariable regions CDRl ' (SEQ ID NO: 4), CDR2' (SEQ ID NO: 5) and CDR3' (SEQ ID NO: 6); or b) an immunoglobulin heavy chain variable domain (VH) which comprises in sequence hypervariable regions CDRl ; -x, CDR2j-x, and CDR3j-x, said hypervariable region hypervariable regions CDRl 1 -X, CDR2i
  • IL- 17 binding molecules of the invention typically have IC 50 S for the inhibition of human IL- 17 on IL-6 production induced by human IL- 17 in human dermal fibroblasts which are within +/- ⁇ 5, i.e.
  • the assay used may be an assay of competitive inhibition of binding of IL- 17 by soluble IL- 17 receptors (e.g. the human IL- 17 R/Fc constructs of Example 1) and the IL- 17 binding molecules of the invention.
  • the human IL- 17 antibody comprises at least a) one heavy chain which comprises a variable domain having an amino acid sequence substantially identical to that shown in SEQ ID NO: 8 starting with the amino acid at position 1 and ending with the amino acid at position 127 and the constant part of a human heavy chain; and b) one light chain which comprises a variable domain having an amino acid sequence substantially identical to that shown in SEQ ID NO: 10 starting with the amino acid at position 1 and ending with the amino acid at position 109 and the constant part of a human light chain.
  • the constant part of a human heavy chain may be of the j ⁇ , ⁇ 2 , ⁇ 3 , ⁇ 4 , ⁇ , Oc 1 , Oc 2 , ⁇ or ⁇ type, preferably of the ⁇ type, more preferably of the Y 1 type, whereas the constant part of a human light chain may be of the K or ⁇ type (which includes the ⁇ ⁇ , ⁇ 2 and ⁇ 3 subtypes) but is preferably of the K type.
  • the amino acid sequences of all these constant parts are given in Kabat et al (supra).
  • Conjugates of the binding molecules of the invention e. g. enzyme or toxin or radioisotope conjugates, are also included within the scope of the invention.
  • Polypeptide if not otherwise specified herein, includes any peptide or protein comprising amino acids joined to each other by peptide bonds, having an amino acid sequence starting at the N-terminal extremity and ending at the C-terminal extremity.
  • the polypeptide of the present invention is a monoclonal antibody, more preferred is a chimeric (also called V-grafted) or humanised (also called CDR-grafted) monoclonal antibody, most preferred a fully human antibody obtainable e.g. by the technology exemplified in Example 1.
  • the humanised (CDR- grafted) or fully human monoclonal antibody may or may not include further mutations introduced into the framework (FR) sequences of the acceptor antibody.
  • a functional derivative of a polypeptide as used herein includes a molecule having a qualitative biological activity in common with a polypeptide to the present invention, i.e. having the ability to bind to the human IL-17.
  • a functional derivative includes fragments and peptide analogs of a polypeptide according to the present invention. Fragments comprise regions within the sequence of a polypeptide according to the present invention, e.g. of a specified sequence.
  • the term "derivative" is used to define amino acid sequence variants, and covalent modifications of a polypeptide according to the present invention, e.g. of a specified sequence.
  • the functional derivatives of a polypeptide according to the present invention e.g. of a specified sequence, e.g.
  • the hypervariable region of the light and the heavy chain preferably have at least about 65%, more preferably at least about 75%, even more preferably at least about 85%, most preferably at least about 95, 96, 97, 98, 99% overall sequence homology with the amino acid sequence of a polypeptide according to the present invention, e.g. of a specified sequence, and substantially retain the ability to bind the human IL-17 or e.g. neutralize IL-6 production of IL-17 induced human dermal fibroblasts.
  • covalent modification includes modifications of a polypeptide according to the present invention, e.g. of a specified sequence; or a fragment thereof with an organic proteinaceous or non-proteinaceous derivatizing agent, fusions to heterologous polypeptide sequences, and post-translational modifications.
  • Covalent modified polypeptides e.g. of a specified sequence, still have the ability to bind the human IL-17 or e.g. neutralize IL-6 production of IL-17 induced human dermal fibroblasts by crosslinking.
  • Covalent modifications are traditionally introduced by reacting targeted amino acid residues with an organic derivatizing agent that is capable of reacting with selected sides or terminal residues, or by harnessing mechanisms of post-translational modifications that function in selected recombinant host cells.
  • Certain post-translational modifications are the result of the action of recombinant host cells on the expressed polypeptide. Glutaminyl and asparaginyl residues are frequently post- translationally deamidated to the corresponding glutamyl and aspartyl residues. Alternatively, these residues are deaminated under mildly acidic conditions.
  • Covalent modifications include fusion proteins comprising a polypeptide according to the present invention, e.g. of a specified sequence and their amino acid sequence variants, such as immunoadhesins, and N-terminal fusions to heterologous signal sequences.
  • “Homology” with respect to a native polypeptide and its functional derivative is defined herein as the percentage of amino acid residues in the candidate sequence that are identical with the residues of a corresponding native polypeptide, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent homology, and not considering any conservative substitutions as part of the sequence identity. Neither N- or C-terminal extensions nor insertions shall be construed as reducing identity or homology. Methods and computer programs for the alignment are well known.
  • amino acid(s) refer to all naturally occurring L- ⁇ -amino acids, e.g. and including D-amino acids.
  • the amino acids are identified by either the well known single-letter or three-letter designations.
  • amino acid sequence variant refers to molecules with some differences in their amino acid sequences as compared to a polypeptide according to the present invention, e.g. of a specified sequence.
  • Amino acid sequence variants of a polypeptide according to the present invention, e.g. of a specified sequence still have the ability to bind the human IL-17 or e.g. neutralize IL-6 production of IL-17 induced human dermal fibroblasts.
  • Substitutional variants are those that have at least one amino acid residue removed and a different amino acid inserted in its place at the same position in a polypeptide according to the present invention, e.g. of a specified sequence.
  • Insertional variants are those with one or more amino acids inserted immediately adjacent to an amino acid at a particular position in a polypeptide according to the present invention, e.g. of a specified sequence. Immediately adjacent to an amino acid means connected to either the ⁇ -carboxy or ⁇ -amino functional group of the amino acid.
  • Deletional variants are those with one or more amino acids in a polypeptide according to the present invention, e.g. of a specified sequence, removed. Ordinarily, deletional variants will have one or two amino acids deleted in a particular region of the molecule.
  • An IL-17 binding molecule of the invention maybe produced by recombinant DNA techniques.
  • one or more DNA molecules encoding the binding molecule must be constructed, placed under appropriate control sequences and transferred into a suitable host organism for expression.
  • the invention provides a DNA molecule encoding an IL- 17 binding molecule as described above.
  • the invention provides a DNA construct comprising a DNA molecule which is substantially homologous to SEQ ID NO: 7 or SEQ ID NO: 9.
  • the invention provides a DNA construct comprising two DNA molecules of which one is substantially homologous to SEQ ID NO: 7 or is a direct DNAH equivalent thereof and the other substantially homologous to SEQ ID NO: 9, or is a direct DNAL equivalent thereof.
  • a method for constructing a variable domain gene is for example described in EPA 239 400 and may be briefly summarized as follows: A gene encoding a variable domain of a MAb of whatever specificity is cloned. The DNA segments encoding the framework and hypervariable regions are determined and the DNA segments encoding the hypervariable regions are removed so that the DNA segments encoding the framework regions are fused together with suitable restriction sites at the junctions. The restriction sites may be generated at the appropriate positions by mutagenesis of the DNA molecule by standard procedures.
  • Double stranded synthetic CDR cassettes are prepared by DNA synthesis according to the sequences encoding for SEQ ID NO: 1 (CDRl), SEQ ID NO: 2 (CDR2), SEQ ID NO: 3 (CDR3), SEQ ID NO: 4 (CDRl '), SEQ ID NO: 5 (CDR2'), SEQ ID NO: 6 (CDR6'), SEQ ID NO: 11 (CDRl-x), SEQ ID NO: 12 (CDR2-x), SEQ ID NO: 13 (CDR3-x) v
  • These cassettes are provided with sticky ends so that they can be ligated at the junctions of the framework
  • PCT application WO 90/07861 gives full instructions for the production of an antibody by recombinant DNA techniques given only written information as to the nucleotide sequence of the gene.
  • the method comprises the synthesis of a number of oligonucleotides, their amplification by the PCR method, and their splicing to give the desired DNA sequence.
  • Expression vectors comprising a suitable promoter or genes encoding heavy and light chain constant parts are publicly available. Thus, once a DNA molecule of the invention is prepared it may be conveniently transferred in an appropriate expression vector. DNA molecules encoding single chain antibodies may also be prepared by standard methods, for example, as described in WO 88/1649.
  • the term "direct DNA H equivalents thereof is meant to stand for a first DNA construct encoding a heavy chain or fragment thereof of an IL- 17 binding molecule of the invention and comprises: a) a first part which encodes a variable domain comprising alternatively framework and hypervariable regions, said hypervariable regions being in sequence CDRIj, CDR2, and CDR3j, said CDRIi is at least 50% homologous, preferably at least 60, 70, 80, 85, or 90% homologous, more preferably at least 95% homologous to the hypervariable region CDRl as shown in SEQ ID NO: 1, said CDR2i is at least 50% homologous, preferably at least 60, 70, 80, 85, or 90% homologous, more preferably at least 95% homologous to the hypervariable region CDR2 as shown in SEQ ID NO: 2, and CDR3j is at least 50% homologous, preferably at least 60, 70, 80, 85, or 90% homologous, more preferably at least at least
  • the term "direct DNAH-X equivalents thereof is meant to stand for a first alternative DNA construct encoding a heavy chain or fragment thereof of an IL- 17 binding molecule of the invention and comprises: a) a first part which encodes a variable domain comprising alternatively framework and hypervariable regions, said hypervariable regions being in sequence CDRlj-x, CDR2i-x and CDR3i-x, said CDRl 1 -X is at least 50% homologous, preferably at least 60, 70, 80, 85, or 90% homologous, more preferably at least 95% homologous to the hypervariable region CDRl as shown in SEQ ID NO: 11, said CDR2j-x is at least 50% homologous, preferably at least 60, 70, 80, 85, or 90% homologous, more preferably at least 95% homologous to the hypervariable region CDR2 as shown in SEQ ID NO: 12, and CDR3i-x is at least 50% homologous, preferably at least 60, 70, 80, 85, or
  • these DNA constructs encode a variable domain comprising alternatively framework and hypervariable regions, said hypervariable regions being in sequence CDRl, CDR2 and CDR3, said CDRl having the amino acid sequence SEQ ID NO: 1, said CDR2 having the amino acid sequence SEQ ID NO: 2, and said CDR3 having the amino acid sequence SEQ ID NO: 3. More preferably, these DNA constructs encode a variable domain comprising alternatively framework and hypervariable regions, said hypervariable regions being in sequence CDRl-x, CDR2-X and CDR3-X, said CDRl-x having the amino acid sequence SEQ ID NO: 11, said CDR2-X having the amino acid sequence SEQ ID NO: 12, and said CDR3-X having the amino acid sequence SEQ ID NO: 13.
  • this first part encodes a variable domain having an amino acid sequence substantially identical to the amino acid sequence as shown in SEQ ID NO: 8 starting with the amino acid at position 1 and ending with the amino acid at position 127. More preferably the first part has the nucleotide sequence as shown in SEQ ID NO: 7 starting with the nucleotide at position 1 and ending with the nucleotide at position 381. Also preferably, the second part encodes the constant part of a human heavy chain, more preferably the constant part of the human ⁇ l chain. This second part may be a DNA fragment of genomic origin (comprising introns) or a cDNA fragment (without introns).
  • the term "direct DNA L equivalents thereof is meant to stand for a second DNA construct encoding a light chain or fragment thereof of an IL- 17 binding molecule of the invention and comprises: a) a first part which encodes a variable domain comprising alternatively framework and hypervariable regions; said hypervariable regions being CDR3j' and optionally CDRl;' and CDR2j', said CDRl;' is at least 50% homologous, preferably at least 60, 70, 80, 85, or 90% homologous, more preferably at least 95% homologous to the hypervariable region CDRl ' as shown in SEQ ID NO: 4, said CDR2j' is at least 50% homologous, preferably at least 60, 70, 80, 85, or 90% homologous, more preferably at least 95% homologous to the hypervariable region CDR2' as shown in SEQ ID NO: 5, and said CDR3i' is at least 50% homologous, preferably at least 60, 70, 80, 85, or 90% homologous, more
  • this second DNA construct encodes a variable domain comprising alternatively framework and hypervariable regions, said hypervariable regions being in sequence CDRl', CDR2' and CDR3', said CDRl ' having the amino acid sequence SEQ ID NO: 4, said CDR2' having the amino acid sequence SEQ ID NO: 5, and said CDR3' having the amino acid sequence SEQ ID NO: 6. More preferably, this first part of the second DNA construct encodes a variable domain having an amino acid sequence substantially identical to the amino acid sequence as shown in SEQ ID NO: 10 starting with the amino acid at position 1 and ending with the amino acid at position 109.
  • the first part has the nucleotide sequence as shown in SEQ ID NO: 9 starting with the nucleotide at position 1 and ending with the nucleotide at position 327.
  • the second part encodes the constant part of a human light chain, more preferably the constant part of the human K
  • the first and second DNA construct will be used together, but may be also used separately.
  • the invention also includes IL- 17 binding molecules in which one or more of the amino acid residues of CDRl, CDR2, CDR3, CDRl-x, CDR2-X, CDR3-X, CDRl ' , CDR2' or CDR3' or the frameworks, typically only a few (e.g. 1-4), are changed; for instance by mutation e.g. site directed mutagenesis of the corresponding DNA sequences.
  • the invention includes the DNA sequences coding for such changed IL- 17 binding molecules.
  • Li particular the invention includes IL- 17 binding molecules in which one or more residues of CDRl' or CDR2' have been changed from the residues shown in SEQ ID NO: 4 (for CDRl ') and SEQ ID NO: 5 (for CDR2').
  • the first and second parts may be separated by an intron, and, an enhancer may be conveniently located in the intron between the first and second parts.
  • an enhancer which is transcribed but not translated, may assist in efficient transcription.
  • the first and second DNA constructs comprise the enhancer of a heavy chain gene advantageously of human origin.
  • Each of the DNA constructs are placed under the control of suitable control sequences, in particular under the control of a suitable promoter.
  • suitable control sequences in particular under the control of a suitable promoter.
  • Any kind of promoter may be used, provided that it is adapted to the host organism in which the DNA constructs will be transferred for expression.
  • the desired antibody may be produced in a cell culture or in a transgenic animal.
  • a suitable transgenic animal may be obtained according to standard methods which include micro injecting into eggs the first and second DNA constructs placed under suitable control sequences transferring the so prepared eggs into appropriate pseudo-pregnant females and selecting a descendant expressing the desired antibody.
  • the DNA constructs When the antibody chains are produced in a cell culture, the DNA constructs must first be inserted into either a single expression vector or into two separate but compatible expression vectors, the latter possibility being preferred.
  • the invention also provides an expression vector able to replicate in a prokaryotic or eukaryotic cell line which comprises at least one of the DNA constructs above described.
  • Each expression vector containing a DNA construct is then transferred into a suitable host organism.
  • the DNA constructs are separately inserted on two expression vectors, they may be transferred separately, i.e. one type of vector per cell, or co-transferred, this latter possibility being preferred.
  • a suitable host organism may be a bacterium, a yeast or a mammalian cell line, this latter being preferred. More preferably, the mammalian cell line is of lymphoid origin, e.g. a myeloma, hybridoma or a normal immortalised B-cell, which conveniently does not express any endogenous antibody heavy or light chain.
  • the JL- 17 binding molecule coding sequence is integrated into the host cell DNA within a locus which permits or favours high level expression of the JL-Il binding molecule.
  • Cells in which the JL- 17 binding molecule coding sequence is integrated into such favourable loci maybe identified and selected on the basis of the levels of the IL- 17 binding molecule which they express.
  • Any suitable selectable marker maybe used for preparation of host cells containing the IL- 17 binding molecule coding sequence; for instance, a dhfr gene/methotrexate or equivalent selection system may be used.
  • Alternative systems for expression of the IL-17 binding molecules of the invention include GS-based amplification/selection systems, such as those described in EP 0256055 B, EP 0323997 B and European patent application 89303964.4.
  • a process for the product of an IL- 17 binding molecule which comprises (i) culturing an organism which is transformed with an expression vector as defined above and (ii) recovering the IL- 17 binding molecule from the culture.
  • an antibody is "capable of inhibiting the binding of IL-17 as AIN457” if the antibody is capable of inhibiting the binding of IL-17 to its receptor substantially to the same extent as the AIN457 antibody, wherein "to the same extent” has meaning as defined above.
  • the AIN457 antibody has binding affinity for IL-17 which is higher than affinities previously reported for anti-IL-17 antibodies, in particular to any anti human IL-17 antibodies.
  • AIN457 has a dissociation equilibrium constant K D for binding to IL-17 of about 0.188 ⁇ 0.036 nM (determined by BIAcore, e.g. as shown in Example 2). This high binding affinity makes the AIN457 antibody particularly suitable for therapeutic applications.
  • 11-17 mediated disease encompasses all diseases and medical conditions in which IL-17 plays a role, whether directly or indirectly, in the disease or medical condition, including the causation, development, progress, persistence or pathology of the disease or condition.
  • treatment refers to both prophylactic or preventative treatment as well as curative or disease modifying treatment, including treatment of patient at risk of contracting the disease or suspected to have contracted the disease as well as patients who are ill or have been diagnosed as suffering from a disease or medical condition, and includes suppression of clinical relapse.
  • the Antibodies of the Invention are human antibodies, most preferably the AIN457 antibody or direct equivalents thereof.
  • the Antibodies of the Invention block the effects of IL- 17 on its target cells and thus are indicated for use in the treatment of IL- 17 mediated diseases and disorders. These and other pharmacological activities of the Antibodies of the Invention may be demonstrated in standard test methods for example as described below:
  • human dermal fibroblasts are stimulated with recombinant IL- 17 in the presence of various concentrations of Antibody of the Invention or human IL- 17 receptor with Fc part.
  • the chimeric anti-CD25 antibody Simulect ® (basiliximab) is used as a negative control.
  • Supernatant is taken after 16 h stimulation and assayed for IL-6 by ELISA.
  • Antibodies of the Invention typically have IC 50 S for inhibition of IL-6 production (in the presence 1 nM human IL- 17) of about 50 nM or less (e.g. from about 0.01 to about 50 nM) when tested as above, i.e.
  • the Antibodies of the Invention have an ICs 0 for inhibition of IL-6 production as defined above of about 20 nM or less, more preferably of about 10 nM or less, more preferably of about 5 nM or less, more preferably of about 2 nM or less, more preferably of about 1 nM or less.
  • Antibodies of the Invention potently block the effects of IL-17. Accordingly, the Antibodies of the Invention have pharmaceutical utility as follows:
  • Antibodies of the Invention are useful for the prophylaxis and treatment of IL-17 mediated diseases or medical conditions, e.g. inflammatory conditions, allergies and allergic conditions, hypersensitivity reactions, autoimmune diseases, severe infections, and organ or tissue transplant rejection.
  • diseases or medical conditions e.g. inflammatory conditions, allergies and allergic conditions, hypersensitivity reactions, autoimmune diseases, severe infections, and organ or tissue transplant rejection.
  • Antibodies of the Invention maybe use for the treatment of recipients of heart, lung, combined heart-lung, liver, kidney, pancreatic, skin or corneal transplants, including allograft rejection or xenograft rejection, and for the prevention of graft- versus-host disease, such as following bone marrow transplant, and organ transplant associated arteriosclerosis.
  • Antibodies of the Invention are particularly useful for the treatment, prevention, or amelioration of autoimmune disease and of inflammatory conditions, in particular inflammatory conditions with an aetiology including an autoimmune component such as arthritis (for example rheumatoid arthritis, arthritis chronica progrediente and arthritis deformans) and rheumatic diseases, including inflammatory conditions and rheumatic diseases involving bone loss, inflammatory pain, spondyloarthropathies including ankolsing spondylitis, Reiter syndrome, reactive arthritis, psoriatic arthritis, and enterophathis arthritis,, hypersensitivity (including both airways hypersensitivity and dermal hypersensitivity) and allergies.
  • arthritis for example rheumatoid arthritis, arthritis chronica progrediente and arthritis deformans
  • rheumatic diseases including inflammatory conditions and rheumatic diseases involving bone loss, inflammatory pain, spondyloarthropathies including ankolsing spondylitis, Reiter syndrome, reactive arthritis, p
  • autoimmune haematological disorders including e.g. hemolytic anaemia, aplastic anaemia, pure red cell anaemia and idiopathic thrombocytopenia
  • systemic lupus erythematosus inflammatory muscle disorders, polychondritis, sclerodoma, Wegener granulomatosis, dermatomyositis, chronic active hepatitis, myasthenia gravis, psoriasis, Steven- Johnson syndrome, idiopathic sprue, autoimmune inflammatory bowel disease (including e.g.
  • ulcerative colitis Crohn's disease and Irritable Bowel Syndrome
  • endocrine ophthalmopathy Graves disease, sarcoidosis, multiple sclerosis, primary biliary cirrhosis, juvenile diabetes (diabetes mellitus type T), uveitis (anterior and posterior), keratoconjunctivitis sicca and vernal keratoconjunctivitis, interstitial lung fibrosis, psoriatic arthritis and glomerulonephritis (with and without nephrotic syndrome, e.g.
  • idiopathic nephrotic syndrome or minimal change nephropathy including idiopathic nephrotic syndrome or minimal change nephropathy), tumors, multiple sclerosis, inflammatory disease of skin and cornea, myositis, loosening of bone implants, metabolic disorders, such as atherosclerosis, diabetes, and dislipidemia.
  • Antibodies of the Invention are also useful for the treatment, prevention, or amelioration of asthma, bronchitis, pneumoconiosis, pulmonary emphysema, and other obstructive or inflammatory diseases of the airways.
  • Antibodies of the Invention are useful for treating undesirable acute and hyperacute inflammatory reactions which are mediated by EL-17or involve IL- 17 production, or the promotion of TNF release by DL-17, e.g.
  • septic shock e.g., endotoxic shock and adult respiratory distress syndrome
  • meningitis e.g., meningitis
  • severe burns e.g., burns
  • cachexia or wasting syndrome associated with morbid TNF release consequent to infection, cancer, or organ dysfunction, especially AIDS -related cachexia, e.g., associated with or consequential to HTV infection.
  • Antibodies of the Invention are particularly useful for treating diseases of bone metabolism including osteoarthritis, osteoporosis and other inflammatory arthritides, and bone loss in general, including age-related bone loss, and in particular periodontal disease.
  • the appropriate dosage will, of course, vary depending upon, for example, the particular Antibody of the Invention to be employed, the host, the mode of administration and the nature and severity of the condition being treated. However, in prophylactic use, satisfactory results are generally indicated to be obtained at dosages from about 0.05 mg to about 10 mg per kilogram body weight more usually from about 0.1 mg to about 5 mg per kilogram body weight.
  • the frequency of dosing for prophylactic uses will normally be in the range from about once per week up to about once every 3 months, more usually in the range from about once every 2 weeks up to about once every 10 weeks, e.g. once every 4 to 8 weeks.
  • Antibody of the Invention is conveniently administered parenterally, intravenously, e.g. into the antecubital or other peripheral vein, intramuscularly, or subcutaneously.
  • a prophylactic treatment typically comprises administering the Antibody of the Invention once per month to once every 2 to 3 months, or less frequently.
  • the Antibodies of the Invention may be administered as the sole active ingredient or in conjunction with, e.g. as an adjuvant to or in combination to, other drugs e.g. immunosuppressive or immunomodulating agents or other anti-inflammatory agents, e.g. for the treatment or prevention of diseases mentioned above.
  • other drugs e.g. immunosuppressive or immunomodulating agents or other anti-inflammatory agents, e.g. for the treatment or prevention of diseases mentioned above.
  • the Antibodies of the Invention maybe used in combination with DMARD, e.g.
  • Gold salts sulphasalazine, antimalarias, methotrexate, D-penicillamine, azathioprine, mycophenolic acid, cyclosporine A, tacrolimus, sirolimus, minocycline, leflunomide, glucocorticoids; a calcineurin inhibitor, e.g. cyclosporin A or FK 506; a modulator of lymphocyte recirculation, e.g. FTY720 and FTY720 analogs; a mTOR inhibitor, e.g.
  • rapamycin 40-O-(2-hydroxyethyl)-rapamycin, CCI779, ABT578, AP23573 or TAFA-93; an ascomycin having immuno-suppressive properties, e.g. ABT-281, ASM981, etc.; corticosteroids; cyclo-phos-phamide; azathioprene; methotrexate; leflunomide; mizoribine; mycophenolic acid; myco-pheno-late mofetil; 15-deoxyspergualine or an immunosuppressive homologue, analogue or derivative thereof; immunosuppressive monoclonal antibodies, e.g., monoclonal antibodies to leukocyte receptors, e.g., MHC, CD2, CD3, CD4, CD7, CD8, CD25, CD28, CD40.
  • immunosuppressive monoclonal antibodies e.g., monoclonal antibodies to leukocyte receptors, e.g., MHC, CD2, CD3, CD4, CD
  • CD45, CD58, CD80, CD86 or their ligands other immunomodulatory compounds, e.g. a recombinant binding molecule having at least a portion of the extracellular domain of CTLA4 or a mutant thereof, e.g. an at least extracellular portion of CTLA4 or a mutant thereof joined to a non-CTLA4 protein sequence, e.g. CTLA4Ig (for ex. designated ATCC 68629) or a mutant thereof, e.g. LEA29Y; adhesion molecule inhibitors, e.g. LFA-I antagonists, ICAM-I or -3 antagonists, VCAM-4 antagonists or VLA-4 antagonists; or a chemotherapeutic agent, e.g.
  • other immunomodulatory compounds e.g. a recombinant binding molecule having at least a portion of the extracellular domain of CTLA4 or a mutant thereof, e.g. an at least extracellular portion of CTLA4 or a mutant thereof joined to a non-
  • anti TNF agents e.g. monoclonal antibodies to TNF, e.g. infliximab, adalimumab, CDP870, or receptor constructs to TNF-RI or TNF-RII, e.g. Etanercept, PEG-TNF-RI; blockers of proinflammatory cytokines, . IL-I blockers, e.g. Anakinra or IL-I trap, AAL160, ACZ 885, IL-6 blockers; chemokmes blockers, e.g inhibitors or activators of proteases, e.g. metalloproteases, anti-IL-15 antibodies, anti-IL-6 antibodies, anti-CD20 antibodies, NSAIDs, such as aspirin or an anti-infectious agent (list not limited to the agent mentioned).
  • anti TNF agents e.g. monoclonal antibodies to TNF, e.g. infliximab, adalimumab, CDP870, or receptor construct
  • a method as defined above comprising co-administration, e.g. concomitantly or in sequence, of a therapeutically effective amount of an IL-17 binding molecule, e.g. an Antibody of the Invention, and at least one second drug substance, said second drug substance being a immuno-suppressive / immunomodulatory, anti-inflammatory chemotherapeutic or anti- infectious drug, e.g. as indicated above.
  • a therapeutically effective amount of an IL-17 binding molecule e.g. an Antibody of the Invention
  • at least one second drug substance said second drug substance being a immuno-suppressive / immunomodulatory, anti-inflammatory chemotherapeutic or anti- infectious drug, e.g. as indicated above.
  • a therapeutic combination e.g. a kit, comprising of a therapeutically effective amount of a) an IL-17 binding molecule, e.g. an Antibody of the Invention, and b) at least one second substance selected from a immuno-suppressive / immunomodulatory, anti-inflammatory chemotherapeutic or anti-infectious drug, e.g. as indicated above.
  • the kit may comprise instructions for its administration.
  • Antibodies of the Invention are administered in conjunction with other immuno-suppressive / immunomodulatory, anti-inflammatory chemotherapeutic or anti- infectious therapy
  • dosages of the co-administered combination compound will of course vary depending on the type of co-drug employed, e.g. whether it is a DMARD, anti-TNF. IL-I blocker or others, on the specific drug employed, on the condition being treated and so forth.
  • compositions of the invention maybe manufactured in conventional manner.
  • a composition according to the invention is preferably provided in lyophilized form.
  • a suitable aqueous carrier for example sterile water for injection or sterile buffered physiological saline.
  • a suitable aqueous carrier for example sterile water for injection or sterile buffered physiological saline.
  • human serum albumin or the patient's own heparinised blood into the saline at the time of formulation.
  • the formulation is given subcutaneous. The presence of an excess of such physiologically inert protein prevents loss of antibody by adsorption onto the walls of the container and tubing used with the infusion solution.
  • albumin a suitable concentration is from 0.5 to 4.5% by weight of the saline solution.
  • Other formulations comprise liquid or lyophilized formulation.
  • mice engineered to express the human IgG/ ⁇ repertoire instead of the murine immunoglobulin repertoire are used to generate antibodies to human IL- 17.
  • B cells from these mice are immortalized by standard hybridoma technology and murine hybridoma cells are obtained which secrete the human IgGl /K antibody AIN457.
  • Example 1 Generation of the hybridoma, purification of the antibodies, selection of AIN457 antibody
  • Recombinant huIL-17 is either produced in E. coli in inclusion bodies and refolded by conventional techniques (produced in house carrier free (E. coli; Novartis Pharma, batch BM-E3141/98) or bought (carrier free, E.
  • HEK.EBNA Recombinant huIL-17, carrier free (IL- 17 APP-C6 from transfected HEK/EBNA cells; Novartis Pharma, batch En.E-3382/82; 0.28 mg/ml; recombinant huIL-17, carrier free (IL-17 APP-C4 from transfected HEK/EBNA cells; Novartis Pharma, batch En.E-3382/83; 0.29 mg/ml)).
  • the latter form features a C-terminal 4 amino acids extension for rapid purification from culture supernatants by immunoaff ⁇ nity chromatography.
  • culture supernatants are loaded on a column of appropriate size of a specific immobilized anti-tag antibody coupled to CNBr activated Sepharose 4B at a density of 10 mg/ml resin following the manufacturer's instructions (Pharmacia).
  • bound huIL-17 is eluted with 100 mM glycine, pH 2.7 and immediately neutralized with diluted NaOH.
  • mice 27340 The genetically engineered mouse 27340 (female; MEDAREX Inc, Annandale, NJ) in which the murine immunoglobulin variable and constant part genes are functionally replaced by their human counterparts (Genotype Tg code 221100-TgH (CMD)++;TgN(Hco7)11952+; TgH(JKD)++; TgN(KCO5)9272+ (see also Sherie L. Morrison, 1994, Nature, Vol. 368, p. 812-813; Nils Lonberg et al., 1994, Nature, Vol. 368, p. 856-859) is immunized following the scheme reported in table 1. Table 1. Immunization schedule Day Date Immunogen Dose and route of immunization
  • H ⁇ X-17 (BM-E3141/98) coupled to KLH 25 ⁇ g of each s.c. in two mixed 1:1 with huJJL-17 (BM-E3141/98) in spots; total volume/mouse Gerbu adjuvant with adjuvant 100 ⁇ l
  • HuJL-17 (BM-E3141/98) coupled to KLH 20 ⁇ g of each s.c. in two (4 th boost) mixed 1:1 with huJL-17 (En.E-3382/83) spots; total volume/mouse coupled to KLH in Gerbu adjuvant with adjuvant 100 ⁇ l
  • HuIL- 17 (En.E-3382/83) coupled to KLH 10 ⁇ g/mouse i.p.
  • Sera samples are obtained 35 and 99 days after the start of the immunization protocol, for measuring levels of anti-huIL17 antibody by enzyme-linked immunosorbent assay (ELISA).
  • ELISA enzyme-linked immunosorbent assay
  • mice 27340 is killed by CO 2 inhalation.
  • Total spleen cells (1 x 10 8 ) are fused with PAI-O cells (5 x 10 7 cells) using PEG 4000.
  • Fused cells are plated out in 720 wells (1 ml/well), containing a feeder layer of mouse peritoneal cells (Balb/c mice), in HAT medium (RPMI 1640 containing 2 g/1 Sodium Bicarbonate, 5 x 10 "5 M j8-Mercaptoethanol, 10 "4 M Hypoxanthine, 1.6 x 10 '5 M Thymidine, 4 x 10 '7 M Aminopterin, 10% heat inactivated FCS and 50 ⁇ g/ml Gentamycin).
  • HAT medium is exchanged with HT medium i.e. HAT medium without Aminopterin. Screening starts on day 10, and lasts two weeks. Of the initial 720 wells plated, 684 wells (95%) are positive for hybridoma growth. Supernatants are collected and screened for huIL-17 reactive MAb in ELISA using both the E. coli and the HEKTEBNA derived huIL-17. Fifty-two primary wells score positive for the presence of anti- huIL-17 antibodies. Twenty-eight hybridomas are cloned and the remaining ones are frozen. Cloning is done, in 4 x 96 well microtiter plates, in HT medium and a feeder layer of mouse peritoneal cells.
  • Hybridomas are plated at 0.5 cell/100 ⁇ l per well. Wells are screened microscopically for growth, and positive ones are fed 100 ⁇ l of HT medium. The following day, supernatants are tested for antibody production in a huIL-17 specific ELISA. Upon cloning, the majority of the cloned hybridomas retain the capacity to secrete anti-huIL-17 specific Monoclonal Antibody (MAb).
  • MAb Monoclonal Antibody
  • the selected clones are transferred in serum free medium (5 ml) into 25 cm 2 TC (TC: tissue culture) flasks. Hybridomas are progressively expanded in serum free medium to 75 cm 2 TC flasks and roller flasks. All the different anti-hu- IL- 17 MAb including NVP-AM457-NX (340-110-28 i.e. mouse number-hybridoma number- clone number) are purified by Protein A affinity chromatography. Culture supernatants are adjusted to pH 7.3 and loaded on a column of appropriate size of Protein A Sepharose 4 fast flow (Pharmacia).
  • Human dermal fibroblasts are cultured in FBM supplemented with 2% FCS, insulin (5 ⁇ g/ml) huFGF-basic (0.1 ⁇ g/ml) and Gentamycin (50 ⁇ g/ml). The fibroblasts are detached from plastic using a Trypsin/EDTA solution. Fibroblasts are distributed into 96 well microtiter plates at a density of 1x10 4 cells/well in FBM supplemented with 1% FCS. Fibroblasts are allowed to adhere to the plates overnight.
  • FBM FBM supplemented with 1% FCS, huBL-17 (different concentrations ranging from 30 to 500 ng/ml) and hybridoma supernatants (1/5 final dilution) or purified antibodies are added to a final volume of 200 ⁇ l.
  • Culture supernatants are collected after an incubation of 24 h and huIL-6 production is measured by ELISA.
  • ELISA for detection of anti-huIL- 17 antibodies ELISA microtiter plates are coated with recombinant huIL-17 (100 ⁇ l/well at 3 ⁇ g/ml; batch BM-E3141/98 or En.E-3382/82) in PBS 0.02% NaN 3 and incubated overnight at room temperature. The following day, microtiter plates are blocked with 300 ⁇ l of PBS/ 2% BSA/ 0.02% NaN 3 for 2 h at 37 0 C. Plates are then washed 4 times with PBS/ 0.05% Tween 20/ 0.02% NaN 3 .
  • Serum dilutions of mouse 27340 (final dilution range at day 35: 1/100 to 1/3200; final dilution range at day 99: 1/200 to 1/12800; 100 ⁇ l/well) or culture supernatants of hybridomas (final dilution 1:3; 100 ⁇ l/well) are added. After an overnight incubation at room temperature, plates are washed 4 times with PBS/ 0.05% Tween 20/ 0.02% NaN 3 . A biotin-conjugated mouse anti-hu-IgG, Fc fragment specific antibody is added at a final dilution of 1/20000 (100 ⁇ l/well). Samples are left to react for 4 h at room temperature.
  • alkaline phosphatase-conjugated streptavidin is added at a final dilution of 1/8000 (100 ⁇ l/well). After 40 minutes at room temperature, plates are washed again 4 times and the substrate (p-nitrophenylphosphate in diethylamino buffer pH 9.8; 150 ⁇ l/well) is added. Plates are read after 30 or 45 min depending on the development of the reaction in a microtiter reader (Bio-Rad) using filters of 405 and 490 nm.
  • ELISA for detection of antibody isotype For revealing the isotype of the MAb, culture supernatants (100 ⁇ l; final dilution 1/5) are added to the wells of microtiter plates coated with huIL-17 (see above), and incubated overnight at room temperature. After washing (4 times), 100 ⁇ l/well of biotin-conjugated mouse MAbs anti-human IgGl (final dilution 1/1000), IgG2 (final dilution 1/1000), IgG3 (final dilution 1/1000) IgG4 (final dilution 1/2000) or anti human K light chain (final dilution 1/1000) are added for 4 h at room temperature.
  • a biotin- conjugated rat anti-mouse ⁇ l and ⁇ 2 light chain specific MAb is used (final dilution 1/1000). This is followed as previously described by washing and addition of alkaline phosphatase- conjugated streptavidin (100 ⁇ l; final dilution 1/8000). After washing (4 times) the substrate (p- nitrophenylphosphate in diethylamino buffer; 100 ⁇ l) is added. Plates are read after 30 or 45 min depending on development of reaction, in a microtiter reader (Bio-Rad) using filters of 405 and 490 nm.
  • ELISA for detection of huIL-6 production ELISA microtiter plates are coated with an anti-huIL- 6 mouse MAb (MAB206 from R&D system; 100 ⁇ l/well at 4 ⁇ g/ml) in PBS 0.02% NaN 3 and incubated overnight at room temperature. The following day, microtiter plates were blocked with 300 ⁇ l of PBS/ 2% BSA/ 0.02% NaN 3 for 2 h at 37 0 C. Plates were then washed 4 times with PBS/ 0.05% Tween 20/ 0.02% NaN 3 . Culture supernatants of human dermal fibroblasts (final dilution 1:3; 100 ⁇ l/well) were added.
  • huIL-6 100 ⁇ l/well is titrated from 400 pg/ml to 3.1 pg/ml in 1:2 dilution steps. After an overnight incubation at room temperature, plates are washed 4 times with PBS/ 0.05% Tween 20/ 0.02% NaN 3 . A biotin- conjugated goat anti-huIL-6 antibody (BAP206; R&D Systems) is added (25 ng/ml; 100 ⁇ l/well). Samples are left to react for 4 h at room temperature. After washing (4 times), alkaline phosphatase-conjugated streptavidin is added at a final dilution of 1/8000 (100 ⁇ l/well).
  • Microtiter plates were coated with huIL-17 (3 ⁇ g/ml) from E. coli (BM-E3141/98) or HEK/EBNA cells (En.E-3382/82).
  • mice 27340 The serum of mouse 27340 is analyzed in ELISA for the presence of anti-huIL-17 antibodies on days 35 and 99 on two different preparations of huIL-17 (Table 2). Results show that serum titers of mouse 27340 increase about fourfold between day 35 and day 99 and that both huIL-17 preparations are recognized.
  • binding in ELISA ofhybridoma supernatants 684 supernatants are tested in ELISA for the presence of anti-huIL-17 antibodies, using two preparations of recombinant huIL-17, the former from E coli (BM-E3141/98) the latter from HEK/EBNA cells (En.E-3382/82). Fifty-two supernatants score positive for the presence of anti-huIL-17 antibodies (Table 3). Preferential binding to one or the other preparation of huIL-17 is observed in a few cases. The 28 hybridomas that are subsequently cloned are underlined. Table 3. ELISA reactivity of culture supernatants
  • Microtiter plates are coated with recombinant huDL-17 (3 ⁇ g/ml) from HEK/EBNA cells (En.E- 3382/82).
  • Culture supematants are tested for inhibition of huIL-6 production by human dermal fibroblasts stimulated with recombinant huIL-17. As shown in Table 5, the majority of the culture supematants show inhibitory activity. Table 5. Inhibition of IL-6 production induced by huEL-17 in human dermal fibroblasts by culture supernatants
  • Neutralizing activity ofAIN45 Selection of clone 110-28 for the production of development candidate AIN457 (preferred embodiment of the invention) is based on neutralizing activity and affinity measurement on BIACORE 2000 of the purified antibodies (see below Example 2).
  • Example 2 AIN457 binds with very high affinity to recombinant human IL-17 (huIL-17); the K D is 122 ⁇ 22 pM (BIAcore) and neutralizes human IL-6 production induced by huIL- 17 in human dermal fibroblast; IC50 is 2.1 ⁇ 0.1 nM at a concentration of 1.87nM huIL-17
  • Reagents General laboratory reagents are purchased from Merck or Sigma and are of the highest purity grade available; the sources of specialty reagents are detailed below.
  • Proteins Monoclonal antibodies are generated by immunizing MEDAREX transgenic mice with recombinant human IL-17, and then following the standard procedure for producing cell lines, from which the secreted material could be purified by Protein A Sepharose chromatography [essentially as described in Example 1). AIN457 is stored as a sterile-filtered solution in 50 mM Na-citrate, pH 7.0, 140 mM NaCl at 4°C .
  • the recombinant human AIN457 (batch KB03303A) is obtained in sterile stock solution of either 20 mM Na-citrate/40 mM phosphate buffer, pH 7, 150 mM NaCl or 20 mM acetic acid pH 5.5 adjusted with IM Tris-base. Concentrations are usually in the range of 2 mg/ml and diluted to a final concentration of 5 ⁇ g/ml into BIA buffer (20 mM HEPES, pH 7,4, 150 mM NaCl, 0,05% v/v Tween-20) for the Biacore experiments.
  • Recombinant human IL-17 is produced in-house; batch En/E 3882/83; 0.29 mg/ml.
  • ATN457 is captured by an anti- human Fc ⁇ antibody (Jackson Immunochemicals; Cat.No. 109-005-098) that is previously immobilized onto a CM-5 BIAcore sensor chip (Research grade). Covalent binding of Fc ⁇ capture antibody is done with the 'Amine coupling kit' provided by BIAcore (BIAcore, Cat.No. BR-1000-50). Typically, 3000 RUs of capture antibody are attached to the activated dextran surface with a 30 ⁇ g/ml anti Fc ⁇ antibody solution in 10 mM Ac buffer, pH 4.5 at a flow rate of 5 ⁇ l/min which lead to approximately 250 RUs of AIN457 immobilization.
  • IL-17 As a guideline, 1000 RUs correspond to a mass transfer of 1 ng/mm 2 .
  • IL-17 (Section 3.2; Table 4)
  • AIN457 antibody is coupled directly to the chip surface without capture antibody. The results are compared to the protocol described in Table 9 (see below).
  • the equilibrium dissociation constant K D allows some judgement about the stability of complexes, once formed in vivo.
  • Table 3 shows the summary of data obtained when the curves of 2 experiments are fitted to the Langmuir model using the BIAevaluation 3.0 software.
  • the antibody is, of course, bivalent, the binding can be treated as a 1:1 event, with individual antibody binding sites displayed at the surface that become occupied by monomelic IL-17 molecules. This experiment shows both, the extremely fast association as well as the very slow dissociation kinetics of the antibody-chemokine complex.
  • AIN457 abrogates the IL-17-dependent secretion of huIL-6 by human dermal fibroblasts.
  • the potency is comparable to that of huIL-17R/Fc and superior to that of a commercially available mouse anti-huIL-17 MAb. It is interesting to note that a more complete inhibition is observed with AIN457 than with TL-ITRfFc.
  • Amino-terminal amino acid sequences of V ⁇ and VH regions The first 48 amino-acid residues of the heavy and the light chain for two anti ⁇ IL-17A antibodies, clone 110-7 (see table 4) and 110- 28 (see table 4), are determined by Edman degradation. The amino-acid sequence is identical for both clones.
  • the GeneBank is searched by blast analysis and the most homologous DNA sequence found is used to design the cloning primers.
  • RNA is prepared from 2x10 7 hybridoma cells (clone 110-7, clone 110-28) with the RNeasy Midi Kit according to the vendor's protocol (Quiagen Hilden Germany). Total RNA is eluted in 200 ⁇ l RNase-free water and stored at -80 0 C. The first strand cDNA synthesis is carried out with M-MLV reverse transcriptase (Promega, Madison, WT), oligo-dT primer, PCR nucleotide mix (dNTPs) and RNAsin inhibitor (Roche, Mannheim).
  • M-MLV reverse transcriptase Promega, Madison, WT
  • dNTPs PCR nucleotide mix
  • RNAsin inhibitor Roche, Mannheim.
  • RNA Five ⁇ g of total RNA is mixed with 1 ⁇ l oligo-dT primer (0.5 ⁇ g/ ⁇ l), and RNase- free water is added to a final volume of 36 ⁇ l. The mixture is incubated at 70°C for 10 minutes and then stored on ice. While on ice, the following reagents are added: lO ⁇ l 5x RT buffer, 2 ⁇ l dNTPs (1OmM each), 2 ⁇ l RNasin and l ⁇ l M-MLV reverse transcriptase. The reaction is carried out at 42°C for lhour.
  • the PCR reaction is assembled using 4 ⁇ l of cDNA template, 2 ⁇ l of each primer at 1 O ⁇ M each (see below and Tables 10 and 11 for overview) 20 ⁇ l of 2xQiamix (containing Buffer, dNTP's, TAQPolymerase) and l ⁇ l of Pwo DNA polymerase in a total volume of 40 ⁇ l.
  • the PCR conditions are set for 35 cycles of 94 0 C for 15 seconds, 55°C for 20 seconds and 72°C for 30 seconds.
  • the PCR product is subcloned into the pCR4-TOPO-Zero (Stragagene, La Jolla, Ca.) cloning vector.
  • the cDNA encoding the heavy chain is amplified using the primer pairs MV416 (SEQ ID NO: 15)/#265 (SEQ ID NO: 16) and MV418 (SEQ ID NO: 17) /#265 (SEQ ID NO: 16).
  • the primers cover the nucleotide sequences corresponding to the following amino acid positions of the heavy chain: MV416 position -19/-13 (signal peptide); MV418 position +1/+7; #265 position +2531+259. Position +1 is the first amino acid of the mature protein.
  • the cDNA encoding the light chain is amplified using the primer pairs MV417 (SEQ ID NO: 18)/#223 (SEQ ID NO: 19) and MV419 (SEQ ID NO: 20)/#223 (SEQ ID NO: 19).
  • the primers cover the nucleotide sequences corresponding to the following amino-acid positions of the light chain: MV417 position -20/-14 (signal peptide); MV419 position +1/+7; #223 position +210/+215. This approach permitted to make two independent PCR amplifications for each immunoglobulin chain, resulting in two independently established DNA sequences.
  • the cloned PCR products coding for the heavy and light chain from two hybridomas are characterized by DNA sequencing. Five and six independent sequences are used to assemble the light and heavy chain sequences . .
  • the light chain cDNAs are all identical and covere the entire coding sequence (amino acid position -20 to +215).
  • the heavy chain cDNAs had 2 different mismatches in one cDNA each. These are excluded from the final sequence, which extends from the initiation codon to the end of the hinge region after the constant domain 1 (amino acid position -19 to +238).
  • the sequences for both hybridomas are identical.
  • the cDNA obtained from hybridoma 110-28 is selected and used for all further expression work.
  • SEQ ID NO: 7 (cDNA of heavy chain of AIN457), SEQ ID NO: 8 (amino acid sequence of heavy chain of ATN457), SEQ ID NO: 9 (cDNA of light chain of AIN457) and SEQ E) NO: 10 (amino acid sequence of AIN457) show the DNA sequence coding for the light and heavy chain of AIN457, along with the protein sequence and the position of the primers used for PCR amplification and DNA sequencing. The DNA sequences have been registered in PlasNova, accession number NPL003689 for the heavy chain, and accession number NPL003690 for the light chain.
  • amino acid sequence found by cDNA cloning is identical to that previously obtained by Edman degradation of the purified immunoglobulin heavy and light chains, indicating that the correct cDNA has been cloned.
  • amino-acid sequence coding for the variable domain is bold and underlined.
  • the oligonucleotide primers used for cloning are indicated (underlined).
  • amino-acid sequence coding for the variable domain is bold and underlined.
  • the oligonucleotide primers used for cloning and sequencing are indicated.
  • CDR' s Complementarily-Determining Regions
  • a single crystal of AIN457 Fab is cross-linked with glutaraldehyde using the method of Lusty (J. Appl. Cryst. (1999) 32, 106-112) and then transferred to a solution containing 2.0M Li 2 SO 4 , 2% PEG 400, and 0.1M Na MES pH 6.5.
  • the crystal is subsequently mounted in a cryo-loop and flash-frozen for data collection at 95K. 180 diffraction images corresponding to l.Odeg oscillation each are recorded.
  • the diffraction data are processed with the HKL program suite.
  • the structure is determined to 2.5A resolution by molecular replacement.
  • the structure is then refined by torsion angle dynamics and energy minimization using the program CNX.
  • Two AIN457 Fab molecules are present in the asymmetric unit of the crystal, with the H-CDR3 loop of both Fab molecules involved in protein-protein contacts to the H-CDR3 loop of symmetry-related Fabs.
  • the two Fab molecules show different elbow angles but have otherwise essentially identical CDR loop conformations (see Table 12 for amino acid sequence of the CDR loops).
  • the H-CDRl loop adopts the expected Hl :1 canonical structure, while the conformation of the H-CDR2 loop matches that of canonical structure H2:3A.
  • the H-CDR3 loop of the ATN457 antibody is exceptionally long, comprising 18 residues between Kabat positions 94 (Arg H98) and 101 (Asp Hl 15).
  • the head of the H-CDR3 loop has the structure of a long, twisted beta-hairpin with a second beta-bulge at its base and a type F beta-turn at its apex.
  • a striking feature of the AIN457 H-CDR3 loop is its very high content in aromatic residues: 6 tyrosines, 2 tryptophanes, 1 phenylalanine.
  • the antigen-combining site of AIN457 possesses 11 tyrosines in total.
  • the conformations of the L-CDRl and L-CDR2 loops correspond to canonical structures Ll :6 and L2:l, respectively.
  • the L-CDR3 loop is short (6 residues) and shows the commonly observed canonical structure L3:l, with a cis-proline at its tip (Pro L96), a glutamine at Kabat position 90 (GIn L91) and a threonine at Kabat position 97 (Thr L98).
  • Cys L97 a cysteine residue after the cis-proline.
  • the side-chain of Cys L97 is at the bottom of a shallow depression located at the V L -V H interface and lined by residues Trp Hl 12, Trp H47 and Tyr L92.
  • Table 1 Amino acid sequences of the hypervariable regions of the AIN457 monoclonal antibodies, based on the Kabat definition and as determined by the X-ray analysis, using the approach of Chothia and coworkers. Amino acid highlighted in bold are part of the CDR loops, while those shown in plain style are part of the antibody framework.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Rheumatology (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Microbiology (AREA)
  • Ophthalmology & Optometry (AREA)
  • Dermatology (AREA)
  • Pain & Pain Management (AREA)
  • Mycology (AREA)
  • Epidemiology (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

An IL-17 binding molecule, in particular an antibody to human IL-17, more preferably a human antibody to IL-17 is provided, wherein the hypervariable regions of the heavy and light chains have amino acid sequences as defined, for use in the treatment of an IL-17 mediated disease or disorder, e.g. rheumatoid arthritis.

Description

IL-17 antagonistic antibodies
This invention relates to an IL- 17 binding molecule, in particular an antibody to human IL- 17, more preferably a human antibody to human EL- 17 (also named IL- 17A) and to the use of such antibodies in the treatment of IL-17 mediated diseases and disorders.
IL- 17, a T-cell derived cytokine present e.g. in rheumatoid arthritis (RA), acts as a pro- inflammatory cytokine, particularly in conjunction with EL-I and TNF-α (Chabaud M & Miossec P (1999) Arthritis Rheum 42, 963-970; Awane M et al (1999) J. Immunol 162, 5337-5344). IL- 17 induces MMP production and downregulates TIMP (Jovanovic DV et al (2001) J. Rheumatol. 28, 712-718), and blockage of IL-I and IL-17 has a synergistic effect on inflammation and bone destruction in vivo (Chabaud M & Miossec (2001) Arthritis Rheum 44, 1293-1303). Inappropriate or excessive production of IL-17 is associated with the pathology of various diseases and disorders, such as rheumatoid arthritis (Wϊtowski et al., 2004 Cell MoI Life Sci 61:567-579), osteoarthritis, loosening of bone implants, acute transplant rejection (Antonysamy et al.5 1999, J Immunol 162, 577-584; van Kooten et al., 1998, J Am Soc Nephrol 9, 1526-1534), septicemia, septic or endotoxic shock, allergies, asthma (Molet et al., 2001, J Allergy Clin Immunol 108, 430-438), bone loss, psoriasis (Teunissen et al., 1998, J Invest Dermatol 111, 645- 649), ischemia, systemic sclerosis (Kurasawa et al., 2000, Arthritis Rheum 43, 2455-2463), stroke, and other inflammatory disorders. Antibodies to IL-17 have been proposed for use in the treatment of IL-17 mediated diseases and disorders; see for instance, WO 95/18826 and the discussion in the introduction thereof.
We have now prepared improved antibodies to human DL- 17 suitable for use in the treatment of DL- 17 mediated diseases and disorders.
Accordingly the invention provides an DL- 17 binding molecule which comprises an antigen binding site comprising at least one immunoglobulin heavy chain variable domain (VH) which comprises in sequence hypervariable regions CDRl, CDR2 and CDR3, said CDRl having the amino acid sequence SEQ DD NO: 1 (N-Y-W-M-N), said CDR2 having the amino acid sequence SEQ ID NO: 2 (A-I-N-Q-D-G-S-E-K-Y-Y-V-G-S-V-K-G), and said CDR3 having the amino acid sequence SEQ ID NO: 3 (D-Y-Y-D-I-L-T-D-Y-Y-I-H-Y-W-Y-F-D-L); or direct CDR equivalents thereof.
Accordingly the invention also provides an IL- 17 binding molecule comprising at least one immunoglobulin light chain variable domain (VL) which comprises in sequence hypervariable regions CDRl', CDR2' and CDR3', said CDRl' having the amino acid sequence SEQ ID NO: 4 (R-A-S-Q-S-V-S-S-S-Y-L-A), said CDR2' having the amino acid sequence SEQ ID NO: 5 (G-A-S-S-R-A-T) and said CDR3' having the amino acid sequence SEQ ID NO: 6 (Q- Q-Y-G-S-S-P-C-T) or direct CDR' equivalents thereof.
In another embodiment of the invention, the invention provides an IL- 17 binding molecule which comprises an antigen binding site comprising at least one immunoglobulin heavy chain variable domain (VH) which comprises in sequence hypervariable regions CDRl -x, CDR2- x and CDR3-X, said CDRl-x having the amino acid sequence SEQ ID NO: 11 (G-F-T-F-S-N-Y- W-M-N), said CDR2-X having the amino acid sequence SEQ ID NO: 12 (A-I-N-Q-D-G-S-E-K- Y-Y), and said CDR3-X having the amino acid sequence SEQ ID NO: 13 (C-V-R-D-Y-Y-D-I-L- T-D-Y-Y-I-H-Y-W-Y-F-D-L-W-G); or direct CDR-x equivalents thereof.
Furthermore, the invention also provides an IL- 17 binding molecule comprising both heavy (VH) and light chain (VL) variable domains; said IL- 17 binding molecule comprises at least one antigen binding site comprising: a) an immunoglobulin heavy chain variable domain (VH) which comprises in sequence hypervariable regions CDRl, CDR2 and CDR3, said CDRl having the amino acid sequence SEQ ID NO:1, said CDR2 having the amino acid sequence SEQ ID NO:2, and said CDR3 having the amino acid sequence SEQ ID NO:3 or direct CDR equivalents thereof; and b) an immunoglobulin light chain variable domain (VL) which comprises in sequence hypervariable regions CDRl', CDR2' and CDR3', said CDRl' having the amino acid sequence SEQ ID NO:4, said CDR2' having the amino acid sequence SEQ ID NO:5, and said CDR3' having the amino acid sequence SEQ ID NO: 6 or direct CDR' equivalents thereof. Moreover, the invention also provides an IL- 17 binding molecule comprising both heavy (VH) and light chain (VL) variable domains; said IL- 17 binding molecule comprises at least one antigen binding site comprising: a) an immunoglobulin heavy chain variable domain (VH) which comprises in sequence hypervariable regions CDRl -x, CDR2-X and CDR3-X, said CDRl -x having the amino acid sequence SEQ ID NO:11, said CDR2-X having the amino acid sequence SEQ ID NO: 12, and said CDR3-X having the amino acid sequence SEQ ID NO: 13 or direct CDR- x equivalents thereof; and b) an immunoglobulin light chain variable domain (VL) which comprises in sequence hypervariable regions CDRl', CDR2' and CDR3', said CDRl' having the amino acid sequence SEQ ID NO:4, said CDR2' having the amino acid sequence SEQ ID NO:5, and said CDR3' having the amino acid sequence SEQ ID NO: 6 or direct CDR' equivalents thereof.
Unless otherwise indicated, any polypeptide chain is herein described as having an amino acid sequence starting at the N-terminal extremity and ending at the C-terminal extremity. When the antigen binding site comprises both the VH and VL domains, these may be located on the same polypeptide molecule or, preferably, each domain may be on a different chain, the VH domain being part of an immunoglobulin heavy chain or fragment thereof and the VL being part of an immunoglobulin light chain or fragment thereof.
By "EL- 17 binding molecule" is meant any molecule capable of binding to the IL- 17 antigen either alone or associated with other molecules. The binding reaction may be shown by standard methods (qualitative assays) including, for example, a binding assay, competition assay or a bioassay for determining the inhibition of DL-17 binding to its receptor or any kind of binding assays, with reference to a negative control test in which an antibody of unrelated specificity but of the same isotype, e.g. an anti-CD25 antibody, is used (see also Example 1).
Examples of antigen binding molecules include antibodies as produced by B-cells or hybridomas and chimeric, CDR-grafted or human antibodies or any fragment thereof, e.g. F(ab')2 and Fab fragments, as well as single chain or single domain antibodies. A single chain antibody consists of the variable domains of the heavy and light chains of an antibody covalently bound by a peptide linker usually consisting of from 10 to 30 amino acids, preferably from 15 to 25 amino acids. Therefore, such a structure does not include the constant part of the heavy and light chains and it is believed that the small peptide spacer should be less antigenic than a whole constant part. By "chimeric antibody" is meant an antibody in which the constant regions of heavy or light chains or both are of human origin while the variable domains of both heavy and light chains are of non-human (e.g. murine) origin or of human origin but derived from a different human antibody. By "CDR-grafted antibody" is meant an antibody in which the hypervariable regions (CDRs) are derived from a donor antibody, such as a non-human (e.g. murine) antibody or a different human antibody, while all or substantially all the other parts of the immunoglobulin e.g. the constant regions and the highly conserved parts of the variable domains, i.e. the framework regions, are derived from an acceptor antibody, e.g. an antibody of human origin. A CDR-grafted antibody may however contain a few amino acids of the donor sequence in the framework regions, for instance in the parts of the framework regions adjacent to the hypervariable regions. By "human antibody" is meant an antibody in which the constant and variable regions of both the heavy and light chains are all of human origin, or substantially identical to sequences of human origin, not necessarily from the same antibody and includes antibodies produced by mice in which the murine immunoglobulin variable and constant part genes have been replaced by their human counterparts, e.g. as described in general terms in EP 0546073 Bl, USP 5545806, USP 5569825, USP 5625126, USP 5633425, USP 5661016, USP 5770429, EP 0 438474 Bl and EP 0463151 Bl.
Particularly preferred EL- 17 binding molecules of the invention are human antibodies, especially the AIN457 antibody as hereinafter described in Examples 1 and 2.
Thus in preferred chimeric antibodies the variable domains of both heavy and light chains are of human origin, for instance those of the AIN457 antibody which are shown in SEQ E) NO: 10 (= variable domain of light chain, i.e. amino acid 1 to 109 of SEQ DD NO: 10) and SEQ ID NO: 8 (= variable domain of heavy chain, i.e. amino acid 1 to 127 of SEQ TD NO: 8). The constant region domains preferably also comprise suitable human constant region domains, for instance as described in "Sequences of Proteins of Immunological Interest", Kabat E.A. et al, US Department of Health and Human Services, Public Health Service, National Institute of Health. Hypervariable regions may be associated with any kind of framework regions, though preferably are of human origin. Suitable framework regions are described in Kabat E. A. et al, ibid. The preferred heavy chain framework is a human heavy chain framework, for instance that of the AIN457 antibody. It consists in sequence e.g. of FRl (amino acid 1 to 30 of SEQ ID NO: 8), FR2 (amino acid 36 to 49 of SEQ ID NO: 8), FR3 (amino acid 67 to 98 of SEQ ID NO: 8) and FR4 (amino acid 117 to 127 of SEQ ID NO: 8) regions. Taking into consideration the determined hypervariable regions of AIN457 by X-ray analysis, another preferred heavy chain framework consists in sequence of FRl -x (amino acid 1 to 25 of SEQ ID NO: 8), FR2-X (amino acid 36 to 49 of SEQ ID NO: 8), FR3-x (amino acid 61 to 95 of SEQ ID NO: 8) and FR4 (amino acid 119 to 127 of SEQ ID NO: 8) regions. In a similar manner, the light chain framework consists, in sequence, of FRl' (amino acid 1 to 23 of SEQ ID NO: 10), FR2' (amino acid 36 to 50 of SEQ ID NO: 10), FR3' (amino acid 58 to 89 of SEQ ID NO: 10) and FR4' (amino acid 99 to 109 of SEQ ID NO: 10) regions.
Accordingly, the invention also provides an IL- 17 binding molecule which comprises at least one antigen binding site comprising either a first domain having an amino acid sequence substantially identical to that shown in SEQ ID NO: 8 starting with the amino acid at position 1 and ending with the amino acid at position 127 or a first domain as described above and a second domain having an amino acid sequence substantially identical to that shown in SEQ ID NO: 10, starting with the amino acid at position 1 and ending with the amino acid at position 109.
Monoclonal antibodies raised against a protein naturally found in all humans are typically developed in a non-human system e.g. in mice, and as such are typically non-human proteins. As a direct consequence of this, a xenogenic antibody as produced by a hybridoma, when administered to humans, elicits an undesirable immune response which is predominantly mediated by the constant part of the xenogenic immunoglobulin. This clearly limits the use of such antibodies as they cannot be administered over a prolonged period of time. Therefore it is particularly preferred to use single chain, single domain, chimeric, CDR-grafted, or especially human antibodies which are not likely to elicit a substantial allogenic response when administered to humans.
In view of the foregoing, a more preferred IL- 17 binding molecule of the invention is selected from a human anti IL- 17 antibody which comprises at least a) an immunoglobulin heavy chain or fragment thereof which comprises (i) a variable domain comprising in sequence the hypervariable regions CDRl, CDR2 and CDR3 or direct CDR equivalents thereof and (ii) the constant part or fragment thereof of a human heavy chain; said CDRl having the amino acid sequence SEQ DD NO: 1, said CDR2 having the amino acid sequence SEQ ID NO: 2, and said CDR3 having the amino acid sequence SEQ ID NO: 3; and b) an immunoglobulin light chain or fragment thereof which comprises (i) a variable domain comprising in sequence the hypervariable regions and optionally also the CDRl', CDR2', and CDR3' hypervariable regions or direct CDR' equivalents thereof and (ii) the constant part or fragment thereof of a human light chain, said CDRl ' having the amino acid sequence SEQ DD NO: 4, said CDR2' having the amino acid sequence SEQ ID NO: 5, and said CDR3' having the amino acid sequence SEQ ID NO: 6.
Alternatively, an IL- 17 binding molecule of the invention may be selected from a single chain binding molecule which comprises an antigen binding site comprising a) a first domain comprising in sequence the hypervariable regions CDRl, CDR2 and CDR3 or direct CDR equivalents thereof, said CDRl having the amino acid sequence SEQ ID NO: 1, said CDR2 having the amino acid sequence SEQ ID NO: 2, and said CDR3 having the amino acid sequence SEQ ID NO: 3; and b) a second domain comprising the hypervariable regions CDRl', CDR2' and CDR3' or direct CDR' equivalents thereof, said CDRl' having the amino acid sequence SEQ ID NO: 4, said CDR2' having the amino acid sequence SEQ ID NO: 5, and said CDR3' having the amino acid sequence SEQ ID NO: 6; and c ) a peptide linker which is bound either to the N-terminal extremity of the first domain and to the C-terminal extremity of the second domain or to the C-terminal extremity of the first domain and to the N-terminal extremity of the second domain.
As it is well known, minor changes in an amino acid sequence such as deletion, addition or substitution of one, a few or even several amino acids may lead to an allelic form of the original protein which has substantially identical properties.
Thus, by the term "direct CDR equivalents thereof are meant IL- 17 binding molecules comprising in sequence the hypervariable regions CDRl;, CDR2i; and CDR3j, (instead of CDRl, CDR2, and CDR3), wherein (i) the hypervariable region CDRIj differs by 3, preferably 2, more preferably 1 amino acid(s) from the hypervariable region CDRl as shown in SEQ ID NO: 1; and
(ii) the hypervariable region CDR2j differs by 3, preferably 2, more preferably 1 amino acid(s) from the hypervariable region CDR2 as shown in SEQ ID NO: 2; and
(iii) the hypervariable region CDR3j differs by 3, preferably 2, more preferably 1 amino acid(s) from the hypervariable region CDR3 as shown in SEQ ID NO: 3; and
(iv) such a molecule comprising in sequence the hypervariable regions CDRl ^ CDR2i, and CDR3j is capable of inhibiting the activity of 1 nM (= 30ng/ml) human IL-17 at a concentration of 50 nM, preferably 2OnM, more preferably 10 nM, more preferably 5 nM of said molecule by 50%, said inhibitory activity is measured on IL-6 production induced by hu-IL-17 in human dermal fibroblasts.
Similarly, by the term "direct CDR-x equivalents thereof are meant IL- 17 binding molecules comprising in sequence the hypervariable regions CDRlj-x, CDR2j-x, and CDR3i-x, (instead of CDRl -x, CDR2-X, and CDR3-x), wherein
(v) the hypervariable region CDRl ,-x differs by 3, preferably 2, more preferably 1 amino acid(s) from the hypervariable region CDRl -x as shown in SEQ ID NO: 11 ; and
(vi) the hypervariable region CDR2j-x differs by 3, preferably 2, more preferably 1 amino acid(s) from the hypervariable region CDR2-X as shown in SEQ ID NO: 12; and
(vii) the hypervariable region CDR3;-x differs by 3, preferably 2, more preferably 1 amino acid(s) from the hypervariable region CDR3-X as shown in SEQ ID NO: 13; and
(viii) such a molecule comprising in sequence the hypervariable regions CDRl j-x, CDR2j-x; and CDR3j-x is capable of inhibiting the activity of 1 nM (= 30ng/ml) human IL-17 at a concentration of 50 nM, preferably 2OnM, more preferably 10 nM, more preferably 5 nM of said molecule by 50%, said inhibitory activity is measured on IL-6 production induced by hu-IL-17 in human dermal fibroblasts.
Similarly, by the term "direct CDR' equivalents thereof is meant a domain comprising in sequence the hypervariable regions CDRl 'j, CDR2'i, and CDR3'j, wherein (i) the hypervariable region CDRl 'i differs by 3, preferably 2, more preferably 1 amino acid(s) from the hypervariable region CDRl' as shown in SEQ ID NO: 4; and
(ii) the hypervariable region CDR2'j differs by 3, preferably 2, more preferably 1 amino acid(s) from the hypervariable region CDR2' as shown in SEQ ID NO: 5; and
(iii) the hypervariable region CDR3'i differs by 3, preferably 2, more preferably 1 amino acid(s) from the hypervariable region CDR3' as shown in SEQ ID NO: 6; and
(iv) such a molecule comprising in sequence the hypervariable regions CDRl \ CDR2'i; and CDR3'j is capable of inhibiting the activity of 1 nM (= 30ng/ml) human IL- 17 at a concentration of 50 nM, preferably 2OnM, more preferably 10 nM, more preferably 5 nM of said molecule by 50%, said inhibitory activity is measured on EL-6 production induced by hu-IL-17 in human dermal fibroblasts.
Alternatively, an IL- 17 binding molecule of the invention may be an IL- 17 binding molecule which comprises at least one antigen binding site comprising at least one immunoglobulin heavy chain variable domain (VH) which comprises in sequence a) hypervariable regions CDRl (SEQ ID NO: 1), CDR2 (SEQ ID NO: 2) and CDR3 (SEQ ID NO: 3); or b) hypervariable regions CDRIi, CDR2j, CDR3j, said hypervariable region CDRl; differs by 3, preferably 2, more preferably 1 amino acid(s) from the hypervariable region of CDRl as shown in SEQ ID NO: 1, said hypervariable region CDR2j differs by 3, preferably 2, more preferably 1 amino acid(s) from the hypervariable region of CDR2 as shown in SEQ ID NO: 2; and said hypervariable region CDR3j differs by 3, preferably 2, more preferably 1 amino acid(s) from the hypervariable region of CDR3 as shown in SEQ ID NO: 3; and said binding IL- 17 molecule comprising in sequence the hypervariable regions CDRlx, CDR2Xj and CDR3X is capable of inhibiting the activity of 1 nM (= 30ng/ml) human IL- 17 at a concentration of 50 nM, preferably 2OnM, more preferably 10 nM, more preferably 5 nM of said molecule by 50%, said inhibitory activity is measured on IL-6 production induced by hu-IL-17 in human dermal fibroblasts.
Similarly, an IL- 17 binding molecule of the invention may be an IL- 17 binding molecule which comprises at least one antigen binding site comprising at least one immunoglobulin heavy chain variable domain (VH) which comprises in sequence a) hypervariable regions CDRl -x (SEQ ID NO: 11), CDR2-X (SEQ ID NO: 12) and CDR3-X (SEQ ID NO: 13); or b) hypervariable regions CDRlj-x, CDR2j-x, CDR3i-x, said hypervariable region CDRlj-x differs by 3, preferably 2, more preferably 1 amino acid(s) from the hypervariable region of CDRl-x as shown in SEQ ID NO: 11, said hypervariable region CDR2j-x differs by 3, preferably
2, more preferably 1 amino acid(s) from the hypervariable region of CDR2-X as shown in SEQ ID NO: 12; and said hypervariable region CDR3j-x differs by 3, preferably 2, more preferably 1 amino acid(s) from the hypervariable region of CDR3-X as shown in SEQ ID NO: 13; and said binding IL-17 molecule comprising in sequence the hypervariable regions CDRlj-x, CDR2;- x, and CDR3j-x is capable of inhibiting the activity of 1 nM (= 30ng/ml) human IL-17 at a concentration of 50 nM, preferably 2OnM, more preferably 10 nM, more preferably 5 nM of said molecule by 50%, said inhibitory activity is measured on IL-6 production induced by hu-IL-17 in human dermal fibroblasts.
Similarly, an IL-17 binding molecule of the invention may be an IL-17 binding molecule which comprises at least one antigen binding site comprising at least one immunoglobulin light chain variable domain (VL) which comprises in sequence a) hypervariable regions CDR' 1 (SEQ ID NO: 4), CDR'2 (SEQ ID NO: 5) and CDR'3 (SEQ E) NO: 6); or b) hypervariable regions CDRl 'j, CDR2'j, CDR3';, said hypervariable region CDR' I1- differs by
3, preferably 2, more preferably 1 amino acid(s) from the hypervariable region of CDR' 1 as shown in SEQ ID NO: 4, said hypervariable region CDR' 2\ differs by 3, preferably 2, more preferably 1 amino acid(s) from the hypervariable region of CDR'2 as shown in SEQ ID NO: 5; and said hypervariable region CDR'3j differs by 3, preferably 2, more preferably 1 amino acid(s) from the hypervariable region of CDR'3 as shown in SEQ ID NO: 6; and said binding EL-17 molecule comprises in sequence the hypervariable regions CDR' Ii1 CDR'2i, and CDR'3j is capable of inhibiting the activity of 1 nM (= 30ng/ml) human IL-17 at a concentration of 50 nM, preferably 2OnM, more preferably 10 nM, more preferably 5 nM of said molecule by 50%, said inhibitory activity is measured on IL-6 production induced by hu-IL-17 in human dermal fibroblasts.
Alternatively, an IL-17 binding molecule of the invention may be a IL-17 binding molecule comprising both heavy (VH) and light chain (VL) variable domains and said IL-17 binding molecule comprises at least one antigen binding site comprising: a) an immunoglobulin heavy chain variable domain (VH) which comprises in sequence hypervariable regions CDRl (SEQ ID NO: 1), CDR2 (SEQ ID NO: 2) and CDR3 (SEQ ID NO: 3); and an immunoglobulin light chain variable domain (VL) which comprises in sequence hypervariable regions CDRl' (SEQ ID NO: 4), CDR2' (SEQ ID NO: 5) and CDR3' (SEQ ID NO: 6); or b) an immunoglobulin heavy chain variable domain (VH) which comprises in sequence hypervariable regions CDRIj, CDR2j, and CDR3j, said hypervariable region hypervariable regions CDRIj, CDR2j, CDR3j, said hypervariable region CDRIj differs by 3, preferably 2, more preferably 1 amino acid(s) from the hypervariable region of CDRl as shown in SEQ ID NO: 1, said hypervariable region CDR2j differs by 3, preferably 2, more preferably 1 amino acid(s) from the hypervariable region of CDR2 as shown in SEQ ID NO: 2; and said hypervariable region CDR3j differs by 3, preferably 2, more preferably 1 amino acid(s) from the hypervariable region of CDR3 as shown in SEQ ID NO: 3; and an immunoglobulin light chain variable domain (VL) which comprises in sequence hypervariable regions CDRl 'i5 CDR2'j, CDR3'j, said hypervariable region CDR' Ij differs by 3, preferably 2, more preferably 1 amino acid(s) from the hypervariable region of CDR' 1 as shown in SEQ ID NO: 4, said hypervariable region CDR'2j differs by 3, preferably 2, more preferably 1 amino acid(s) from the hypervariable region of CDR'2 as shown in SEQ ID NO: 5; and said hypervariable region CDR' 3 j differs by 3, preferably 2, more preferably 1 amino acid(s) from the hypervariable region of CDR'3 as shown in SEQ ID NO: 6; and said binding IL- 17 molecule defined in b) comprises in sequence the hypervariable regions CDRIj, CDR2j, CDR3j, CDR' Ij, CDR'2j, and CDR'3j is capable of inhibiting the activity of 1 nM (= 30ng/ml) human IL-17 at a concentration of 50 nM, preferably 2OnM, more preferably 10 nM, more preferably 5 nM of said molecule by 50%, said inhibitory activity is measured on TL-6 production induced by hu-IL-17 in human dermal fibroblasts.
Alternatively, an IL-17 binding molecule of the invention may be a IL-17 binding molecule comprising both heavy (VH) and light chain (VL) variable domains and said IL-17 binding molecule comprises at least one antigen binding site comprising: a) an immunoglobulin heavy chain variable domain (VH) which comprises in sequence hypervariable regions CDRl-x (SEQ ID NO: 11), CDR2-X (SEQ ID NO: 12) and CDR3-X (SEQ ID NO: 13); and an immunoglobulin light chain variable domain (VL) which comprises in sequence hypervariable regions CDRl ' (SEQ ID NO: 4), CDR2' (SEQ ID NO: 5) and CDR3' (SEQ ID NO: 6); or b) an immunoglobulin heavy chain variable domain (VH) which comprises in sequence hypervariable regions CDRl ;-x, CDR2j-x, and CDR3j-x, said hypervariable region hypervariable regions CDRl1-X, CDR2i-x, CDR3j-x, said hypervariable region CDRl j-x differs by 3, preferably 2, more preferably 1 amino acid(s) from the hypervariable region of CDRl -x as shown in SEQ DD NO: 11, said hypervariable region CDR2j-x differs by 3, preferably 2, more preferably 1 amino acid(s) from the hypervariable region of CDR2-X as shown in SEQ ID NO: 12; and said hypervariable region CDR3;-x differs by 3, preferably 2, more preferably 1 amino acid(s) from the hypervariable region of CDR3-X as shown in SEQ ID NO: 13; and an immunoglobulin light chain variable domain (VL) which comprises in sequence hypervariable regions CDRl 'i, CDR2'i5 CDR3'i5 said hypervariable region CDR'lj differs by 3, preferably 2, more preferably 1 amino acid(s) from the hypervariable region of CDR' 1 as shown in SEQ ID NO: 4, said hypervariable region CDR'2; differs by 3, preferably 2, more preferably 1 amino acid(s) from the hypervariable region of CDR'2 as shown in SEQ ID NO: 5; and said hypervariable region CDR'3{ differs by 3, preferably 2, more preferably 1 amino acid(s) from the hypervariable region of CDR' 3 as shown in SEQ ID NO: 6; and said binding IL- 17 molecule defined in b) comprises in sequence the hypervariable regions CDRIi, CDR2i,CDR3i, CDR' Ij, CDR'2i, and CDR'3j is capable of inhibiting the activity of 1 nM (= 30ng/ml) human IL- 17 at a concentration of 50 nM, preferably 2OnM, more preferably 10 nM, more preferably 5 nM of said molecule by 50%, said inhibitory activity is measured on IL-6 production induced by hu-EL-17 in human dermal fibroblasts.
The inhibition of the binding of IL- 17 to its receptor may be conveniently tested in various assays including such assays are described hereinafter in the text. By the term "to the same extent" is meant that the reference and the equivalent molecules exhibit, on a statistical basis, essentially identical IL- 17 inhibitory activity in one of the assays referred to herein (see Example 1). For example, IL- 17 binding molecules of the invention typically have IC50S for the inhibition of human IL- 17 on IL-6 production induced by human IL- 17 in human dermal fibroblasts which are within +/-χ5, i.e. below 10 nM, more preferably 9, 8, 7, 6, 5, 4, 3 or 2 nM of that of, preferably substantially the same as, the IC50 of the corresponding reference molecule when assayed as described in Example 1. Alternatively, the assay used may be an assay of competitive inhibition of binding of IL- 17 by soluble IL- 17 receptors (e.g. the human IL- 17 R/Fc constructs of Example 1) and the IL- 17 binding molecules of the invention.
Most preferably, the human IL- 17 antibody comprises at least a) one heavy chain which comprises a variable domain having an amino acid sequence substantially identical to that shown in SEQ ID NO: 8 starting with the amino acid at position 1 and ending with the amino acid at position 127 and the constant part of a human heavy chain; and b) one light chain which comprises a variable domain having an amino acid sequence substantially identical to that shown in SEQ ID NO: 10 starting with the amino acid at position 1 and ending with the amino acid at position 109 and the constant part of a human light chain.
The constant part of a human heavy chain may be of the j\, γ2, γ3, γ4, μ, Oc1 , Oc2, δ or ε type, preferably of the γ type, more preferably of the Y1 type, whereas the constant part of a human light chain may be of the K or λ type (which includes the λ\, λ2 and λ3 subtypes) but is preferably of the K type. The amino acid sequences of all these constant parts are given in Kabat et al (supra).
Conjugates of the binding molecules of the invention, e. g. enzyme or toxin or radioisotope conjugates, are also included within the scope of the invention.
"Polypeptide", if not otherwise specified herein, includes any peptide or protein comprising amino acids joined to each other by peptide bonds, having an amino acid sequence starting at the N-terminal extremity and ending at the C-terminal extremity. Preferably the polypeptide of the present invention is a monoclonal antibody, more preferred is a chimeric (also called V-grafted) or humanised (also called CDR-grafted) monoclonal antibody, most preferred a fully human antibody obtainable e.g. by the technology exemplified in Example 1. The humanised (CDR- grafted) or fully human monoclonal antibody may or may not include further mutations introduced into the framework (FR) sequences of the acceptor antibody.
A functional derivative of a polypeptide as used herein includes a molecule having a qualitative biological activity in common with a polypeptide to the present invention, i.e. having the ability to bind to the human IL-17. A functional derivative includes fragments and peptide analogs of a polypeptide according to the present invention. Fragments comprise regions within the sequence of a polypeptide according to the present invention, e.g. of a specified sequence. The term "derivative" is used to define amino acid sequence variants, and covalent modifications of a polypeptide according to the present invention, e.g. of a specified sequence. The functional derivatives of a polypeptide according to the present invention, e.g. of a specified sequence, e.g. of the hypervariable region of the light and the heavy chain, preferably have at least about 65%, more preferably at least about 75%, even more preferably at least about 85%, most preferably at least about 95, 96, 97, 98, 99% overall sequence homology with the amino acid sequence of a polypeptide according to the present invention, e.g. of a specified sequence, and substantially retain the ability to bind the human IL-17 or e.g. neutralize IL-6 production of IL-17 induced human dermal fibroblasts.
The term "covalent modification" includes modifications of a polypeptide according to the present invention, e.g. of a specified sequence; or a fragment thereof with an organic proteinaceous or non-proteinaceous derivatizing agent, fusions to heterologous polypeptide sequences, and post-translational modifications. Covalent modified polypeptides, e.g. of a specified sequence, still have the ability to bind the human IL-17 or e.g. neutralize IL-6 production of IL-17 induced human dermal fibroblasts by crosslinking. Covalent modifications are traditionally introduced by reacting targeted amino acid residues with an organic derivatizing agent that is capable of reacting with selected sides or terminal residues, or by harnessing mechanisms of post-translational modifications that function in selected recombinant host cells. Certain post-translational modifications are the result of the action of recombinant host cells on the expressed polypeptide. Glutaminyl and asparaginyl residues are frequently post- translationally deamidated to the corresponding glutamyl and aspartyl residues. Alternatively, these residues are deaminated under mildly acidic conditions. Other post-translational modifications include hydroxylation of proline and lysine, phosphorylation of hydroxyl groups of seryl, tyrosine or threonyl residues, methylation of the α-amino groups of lysine, arginine, and histidine side chains, see e.g. T. E. Creighton, Proteins: Structure and Molecular Properties, W. H. Freeman δc Co., San Francisco, pp. 79-86 (1983). Covalent modifications e.g. include fusion proteins comprising a polypeptide according to the present invention, e.g. of a specified sequence and their amino acid sequence variants, such as immunoadhesins, and N-terminal fusions to heterologous signal sequences.
"Homology" with respect to a native polypeptide and its functional derivative is defined herein as the percentage of amino acid residues in the candidate sequence that are identical with the residues of a corresponding native polypeptide, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent homology, and not considering any conservative substitutions as part of the sequence identity. Neither N- or C-terminal extensions nor insertions shall be construed as reducing identity or homology. Methods and computer programs for the alignment are well known.
"Amino acid(s)" refer to all naturally occurring L-α-amino acids, e.g. and including D-amino acids. The amino acids are identified by either the well known single-letter or three-letter designations.
The term "amino acid sequence variant" refers to molecules with some differences in their amino acid sequences as compared to a polypeptide according to the present invention, e.g. of a specified sequence. Amino acid sequence variants of a polypeptide according to the present invention, e.g. of a specified sequence, still have the ability to bind the human IL-17 or e.g. neutralize IL-6 production of IL-17 induced human dermal fibroblasts. Substitutional variants are those that have at least one amino acid residue removed and a different amino acid inserted in its place at the same position in a polypeptide according to the present invention, e.g. of a specified sequence. These substitutions may be single, where only one amino acid in the molecule has been substituted, or they may be multiple, where two or more amino acids have been substituted in the same molecule. Insertional variants are those with one or more amino acids inserted immediately adjacent to an amino acid at a particular position in a polypeptide according to the present invention, e.g. of a specified sequence. Immediately adjacent to an amino acid means connected to either the α-carboxy or α-amino functional group of the amino acid. Deletional variants are those with one or more amino acids in a polypeptide according to the present invention, e.g. of a specified sequence, removed. Ordinarily, deletional variants will have one or two amino acids deleted in a particular region of the molecule.
An IL-17 binding molecule of the invention maybe produced by recombinant DNA techniques. In view of this, one or more DNA molecules encoding the binding molecule must be constructed, placed under appropriate control sequences and transferred into a suitable host organism for expression.
In a very general manner, there are accordingly provided
(i) DNA molecules encoding a single domain IL-17 binding molecule of the invention, a single chain IL-17 binding molecule of the invention, an IL-17 binding molecule comprising a heavy and light chain as defined herein, or fragments of a IL- 17 binding molecule of the invention; and
(ii) the use of the DNA molecules of the invention for the production of a IL- 17 binding molecule of the invention by recombinant means.
Accordingly, the invention provides a DNA molecule encoding an IL- 17 binding molecule as described above.
Furthermore, the invention provides a DNA construct comprising a DNA molecule which is substantially homologous to SEQ ID NO: 7 or SEQ ID NO: 9.
Furthermore, the invention provides a DNA construct comprising two DNA molecules of which one is substantially homologous to SEQ ID NO: 7 or is a direct DNAH equivalent thereof and the other substantially homologous to SEQ ID NO: 9, or is a direct DNAL equivalent thereof.
The present state of the art is such that the skilled worker in the art is able to synthesize the DNA molecules of the invention given the information provided herein i.e. the amino acid sequences of the hypervariable regions and the DNA sequences coding for them. A method for constructing a variable domain gene is for example described in EPA 239 400 and may be briefly summarized as follows: A gene encoding a variable domain of a MAb of whatever specificity is cloned. The DNA segments encoding the framework and hypervariable regions are determined and the DNA segments encoding the hypervariable regions are removed so that the DNA segments encoding the framework regions are fused together with suitable restriction sites at the junctions. The restriction sites may be generated at the appropriate positions by mutagenesis of the DNA molecule by standard procedures. Double stranded synthetic CDR cassettes are prepared by DNA synthesis according to the sequences encoding for SEQ ID NO: 1 (CDRl), SEQ ID NO: 2 (CDR2), SEQ ID NO: 3 (CDR3), SEQ ID NO: 4 (CDRl '), SEQ ID NO: 5 (CDR2'), SEQ ID NO: 6 (CDR6'), SEQ ID NO: 11 (CDRl-x), SEQ ID NO: 12 (CDR2-x), SEQ ID NO: 13 (CDR3-x)v These cassettes are provided with sticky ends so that they can be ligated at the junctions of the framework
Furthermore, it is not necessary to have access to the mRNA from a producing hybridoma cell line in order to obtain a DNA construct coding for the IL- 17 binding molecules of the invention. Thus PCT application WO 90/07861 gives full instructions for the production of an antibody by recombinant DNA techniques given only written information as to the nucleotide sequence of the gene. The method comprises the synthesis of a number of oligonucleotides, their amplification by the PCR method, and their splicing to give the desired DNA sequence.
Expression vectors comprising a suitable promoter or genes encoding heavy and light chain constant parts are publicly available. Thus, once a DNA molecule of the invention is prepared it may be conveniently transferred in an appropriate expression vector. DNA molecules encoding single chain antibodies may also be prepared by standard methods, for example, as described in WO 88/1649.
Li analogy to the case for CDR equivalents, the term "direct DNAH equivalents thereof is meant to stand for a first DNA construct encoding a heavy chain or fragment thereof of an IL- 17 binding molecule of the invention and comprises: a) a first part which encodes a variable domain comprising alternatively framework and hypervariable regions, said hypervariable regions being in sequence CDRIj, CDR2, and CDR3j, said CDRIi is at least 50% homologous, preferably at least 60, 70, 80, 85, or 90% homologous, more preferably at least 95% homologous to the hypervariable region CDRl as shown in SEQ ID NO: 1, said CDR2i is at least 50% homologous, preferably at least 60, 70, 80, 85, or 90% homologous, more preferably at least 95% homologous to the hypervariable region CDR2 as shown in SEQ ID NO: 2, and CDR3j is at least 50% homologous, preferably at least 60, 70, 80, 85, or 90% homologous, more preferably at least 95% homologous to the hypervariable region CDR3 as shown in SEQ ID NO: 3; this first part starting with a codon encoding the first amino acid of the variable domain and ending with a codon encoding the last amino acid of the variable domain; and b) a second part encoding a heavy chain constant part or fragment thereof which starts with a codon encoding the first amino acid of the constant part of the heavy chain and ends with a codon encoding the last amino acid of the constant part or fragment thereof, followed by a stop codon; and c) said DNA construct encoding for a polypeptide which is capable either alone or in combination with another polypeptide of inhibiting the activity of 1 nM (= 30ng/ml) human IL- 17 at a concentration of 50 nM, preferably 2OnM, more preferably 10 nM, more preferably 5 nM of said molecule by 50%, said inhibitory activity is measured on JL-6 production induced by hu- IL- 17 in human dermal fibroblasts.
Similarly, the term "direct DNAH-X equivalents thereof is meant to stand for a first alternative DNA construct encoding a heavy chain or fragment thereof of an IL- 17 binding molecule of the invention and comprises: a) a first part which encodes a variable domain comprising alternatively framework and hypervariable regions, said hypervariable regions being in sequence CDRlj-x, CDR2i-x and CDR3i-x, said CDRl1-X is at least 50% homologous, preferably at least 60, 70, 80, 85, or 90% homologous, more preferably at least 95% homologous to the hypervariable region CDRl as shown in SEQ ID NO: 11, said CDR2j-x is at least 50% homologous, preferably at least 60, 70, 80, 85, or 90% homologous, more preferably at least 95% homologous to the hypervariable region CDR2 as shown in SEQ ID NO: 12, and CDR3i-x is at least 50% homologous, preferably at least 60, 70, 80, 85, or 90% homologous, more preferably at least 95% homologous to the hypervariable region CDR3 as shown in SEQ ID NO: 13; this first part starting with a codon encoding the first amino acid of the variable domain and ending with a codon encoding the last amino acid of the variable domain; and b) a second part encoding a heavy chain constant part or fragment thereof which starts with a codon encoding the first amino acid of the constant part of the heavy chain and ends with a codon encoding the last amino acid of the constant part or fragment thereof, followed by a stop codon; and c) said DNA construct encoding for a polypeptide which is capable either alone or in combination with another polypeptide of inhibiting the activity of 1 nM (= 30ng/ml) human IL- 17 at a concentration of 50 nM, preferably 2OnM, more preferably 10 nM, more preferably 5 nM of said molecule by 50%, said inhibitory activity is measured on IL-6 production induced by hu- IL- 17 in human dermal fibroblasts.
Preferably, these DNA constructs encode a variable domain comprising alternatively framework and hypervariable regions, said hypervariable regions being in sequence CDRl, CDR2 and CDR3, said CDRl having the amino acid sequence SEQ ID NO: 1, said CDR2 having the amino acid sequence SEQ ID NO: 2, and said CDR3 having the amino acid sequence SEQ ID NO: 3. More preferably, these DNA constructs encode a variable domain comprising alternatively framework and hypervariable regions, said hypervariable regions being in sequence CDRl-x, CDR2-X and CDR3-X, said CDRl-x having the amino acid sequence SEQ ID NO: 11, said CDR2-X having the amino acid sequence SEQ ID NO: 12, and said CDR3-X having the amino acid sequence SEQ ID NO: 13. More preferably, this first part encodes a variable domain having an amino acid sequence substantially identical to the amino acid sequence as shown in SEQ ID NO: 8 starting with the amino acid at position 1 and ending with the amino acid at position 127. More preferably the first part has the nucleotide sequence as shown in SEQ ID NO: 7 starting with the nucleotide at position 1 and ending with the nucleotide at position 381. Also preferably, the second part encodes the constant part of a human heavy chain, more preferably the constant part of the human γl chain. This second part may be a DNA fragment of genomic origin (comprising introns) or a cDNA fragment (without introns).
Similarly, the term "direct DNAL equivalents thereof is meant to stand for a second DNA construct encoding a light chain or fragment thereof of an IL- 17 binding molecule of the invention and comprises: a) a first part which encodes a variable domain comprising alternatively framework and hypervariable regions; said hypervariable regions being CDR3j' and optionally CDRl;' and CDR2j', said CDRl;' is at least 50% homologous, preferably at least 60, 70, 80, 85, or 90% homologous, more preferably at least 95% homologous to the hypervariable region CDRl ' as shown in SEQ ID NO: 4, said CDR2j' is at least 50% homologous, preferably at least 60, 70, 80, 85, or 90% homologous, more preferably at least 95% homologous to the hypervariable region CDR2' as shown in SEQ ID NO: 5, and said CDR3i' is at least 50% homologous, preferably at least 60, 70, 80, 85, or 90% homologous, more preferably at least 95% homologous to the hypervariable region CDR3' as shown in SEQ ID NO: 6; this first part starting with a codon encoding the first amino acid of the variable domain and ending with a codon encoding the last amino acid of the variable domain; and b) a second part encoding a light chain constant part or fragment thereof which starts with a codon encoding the first amino acid of the constant part of the light chain and ends with a codon encoding the last amino acid of the constant part or fragment thereof followed by a stop codon; and c) said DNA construct encoding for a polypeptide which is capable either alone or in combination with another polypeptide of inhibiting the activity of 1 nM (= 30ng/ml) human IL- 17 at a concentration of 50 nM, preferably 2OnM, more preferably 10 nM, more preferably 5 nM of said molecule by 50%, said inhibitory activity is measured on IL-6 production induced by hu- IL- 17 in human dermal fibroblasts.
Preferably, this second DNA construct encodes a variable domain comprising alternatively framework and hypervariable regions, said hypervariable regions being in sequence CDRl', CDR2' and CDR3', said CDRl ' having the amino acid sequence SEQ ID NO: 4, said CDR2' having the amino acid sequence SEQ ID NO: 5, and said CDR3' having the amino acid sequence SEQ ID NO: 6. More preferably, this first part of the second DNA construct encodes a variable domain having an amino acid sequence substantially identical to the amino acid sequence as shown in SEQ ID NO: 10 starting with the amino acid at position 1 and ending with the amino acid at position 109. More preferably, the first part has the nucleotide sequence as shown in SEQ ID NO: 9 starting with the nucleotide at position 1 and ending with the nucleotide at position 327. Also preferably the second part encodes the constant part of a human light chain, more preferably the constant part of the human K
Preferably, the first and second DNA construct will be used together, but may be also used separately.
The invention also includes IL- 17 binding molecules in which one or more of the amino acid residues of CDRl, CDR2, CDR3, CDRl-x, CDR2-X, CDR3-X, CDRl', CDR2' or CDR3' or the frameworks, typically only a few (e.g. 1-4), are changed; for instance by mutation e.g. site directed mutagenesis of the corresponding DNA sequences. The invention includes the DNA sequences coding for such changed IL- 17 binding molecules. Li particular the invention includes IL- 17 binding molecules in which one or more residues of CDRl' or CDR2' have been changed from the residues shown in SEQ ID NO: 4 (for CDRl ') and SEQ ID NO: 5 (for CDR2').
In the first and second DNA constructs, the first and second parts may be separated by an intron, and, an enhancer may be conveniently located in the intron between the first and second parts. The presence of such an enhancer which is transcribed but not translated, may assist in efficient transcription. In particular embodiments the first and second DNA constructs comprise the enhancer of a heavy chain gene advantageously of human origin.
Each of the DNA constructs are placed under the control of suitable control sequences, in particular under the control of a suitable promoter. Any kind of promoter may be used, provided that it is adapted to the host organism in which the DNA constructs will be transferred for expression.
The desired antibody may be produced in a cell culture or in a transgenic animal. A suitable transgenic animal may be obtained according to standard methods which include micro injecting into eggs the first and second DNA constructs placed under suitable control sequences transferring the so prepared eggs into appropriate pseudo-pregnant females and selecting a descendant expressing the desired antibody.
When the antibody chains are produced in a cell culture, the DNA constructs must first be inserted into either a single expression vector or into two separate but compatible expression vectors, the latter possibility being preferred.
Accordingly, the invention also provides an expression vector able to replicate in a prokaryotic or eukaryotic cell line which comprises at least one of the DNA constructs above described.
Each expression vector containing a DNA construct is then transferred into a suitable host organism. When the DNA constructs are separately inserted on two expression vectors, they may be transferred separately, i.e. one type of vector per cell, or co-transferred, this latter possibility being preferred. A suitable host organism may be a bacterium, a yeast or a mammalian cell line, this latter being preferred. More preferably, the mammalian cell line is of lymphoid origin, e.g. a myeloma, hybridoma or a normal immortalised B-cell, which conveniently does not express any endogenous antibody heavy or light chain.
For expression in mammalian cells it is preferred that the JL- 17 binding molecule coding sequence is integrated into the host cell DNA within a locus which permits or favours high level expression of the JL-Il binding molecule. Cells in which the JL- 17 binding molecule coding sequence is integrated into such favourable loci maybe identified and selected on the basis of the levels of the IL- 17 binding molecule which they express. Any suitable selectable marker maybe used for preparation of host cells containing the IL- 17 binding molecule coding sequence; for instance, a dhfr gene/methotrexate or equivalent selection system may be used. Alternative systems for expression of the IL-17 binding molecules of the invention include GS-based amplification/selection systems, such as those described in EP 0256055 B, EP 0323997 B and European patent application 89303964.4.
In a further aspect of the invention there is provided a process for the product of an IL- 17 binding molecule which comprises (i) culturing an organism which is transformed with an expression vector as defined above and (ii) recovering the IL- 17 binding molecule from the culture.
For the purposes of the present description an antibody is "capable of inhibiting the binding of IL-17 as AIN457" if the antibody is capable of inhibiting the binding of IL-17 to its receptor substantially to the same extent as the AIN457 antibody, wherein "to the same extent" has meaning as defined above.
The AIN457 antibody has binding affinity for IL-17 which is higher than affinities previously reported for anti-IL-17 antibodies, in particular to any anti human IL-17 antibodies. Thus AIN457 has a dissociation equilibrium constant KD for binding to IL-17 of about 0.188 ± 0.036 nM (determined by BIAcore, e.g. as shown in Example 2). This high binding affinity makes the AIN457 antibody particularly suitable for therapeutic applications.
In the present description the phrase "11-17 mediated disease" encompasses all diseases and medical conditions in which IL-17 plays a role, whether directly or indirectly, in the disease or medical condition, including the causation, development, progress, persistence or pathology of the disease or condition.
In the present description the terms "treatment" or "treat" refer to both prophylactic or preventative treatment as well as curative or disease modifying treatment, including treatment of patient at risk of contracting the disease or suspected to have contracted the disease as well as patients who are ill or have been diagnosed as suffering from a disease or medical condition, and includes suppression of clinical relapse.
IL-17 binding molecules as defined above which have binding specificity for human IL- 17, in particular antibodies which are capable of inhibiting the binding of IL-17 to its receptor; and antibodies to IL-17 which are capable of inhibiting the activity of 1 nM (= 30ng/ml) human IL- 17 at a concentration of 50 nM, preferably 2OnM5 more preferably 10 nM, more preferably 5 nM of said molecule by 50%, said inhibitory activity is measured on IL-6 production induced by hu-IL-17 in human dermal fibroblasts, are herein referred to as Antibodies of the Invention.
Preferably the Antibodies of the Invention are human antibodies, most preferably the AIN457 antibody or direct equivalents thereof.
The Antibodies of the Invention block the effects of IL- 17 on its target cells and thus are indicated for use in the treatment of IL- 17 mediated diseases and disorders. These and other pharmacological activities of the Antibodies of the Invention may be demonstrated in standard test methods for example as described below:
Neutralization of IL-17 dependent production of interleukin-6 by primary human fibroblasts: The production of IL-6 in primary human (dermal) fibroblasts is dependent on IL- 17 (Hwang SY et al., (2004) Arthritis Res Ther; 6:R120-128.
In short, human dermal fibroblasts are stimulated with recombinant IL- 17 in the presence of various concentrations of Antibody of the Invention or human IL- 17 receptor with Fc part. The chimeric anti-CD25 antibody Simulect® (basiliximab) is used as a negative control. Supernatant is taken after 16 h stimulation and assayed for IL-6 by ELISA. Antibodies of the Invention typically have IC50S for inhibition of IL-6 production (in the presence 1 nM human IL- 17) of about 50 nM or less (e.g. from about 0.01 to about 50 nM) when tested as above, i.e. said inhibitory activity is measured on IL-6 production induced by hu-IL-17 in human dermal fibroblasts. Preferably, the Antibodies of the Invention have an ICs0 for inhibition of IL-6 production as defined above of about 20 nM or less, more preferably of about 10 nM or less, more preferably of about 5 nM or less, more preferably of about 2 nM or less, more preferably of about 1 nM or less.
As indicated in the above assay Antibodies of the Invention potently block the effects of IL-17. Accordingly, the Antibodies of the Invention have pharmaceutical utility as follows:
Antibodies of the Invention are useful for the prophylaxis and treatment of IL-17 mediated diseases or medical conditions, e.g. inflammatory conditions, allergies and allergic conditions, hypersensitivity reactions, autoimmune diseases, severe infections, and organ or tissue transplant rejection.
For example, Antibodies of the Invention maybe use for the treatment of recipients of heart, lung, combined heart-lung, liver, kidney, pancreatic, skin or corneal transplants, including allograft rejection or xenograft rejection, and for the prevention of graft- versus-host disease, such as following bone marrow transplant, and organ transplant associated arteriosclerosis.
Antibodies of the Invention are particularly useful for the treatment, prevention, or amelioration of autoimmune disease and of inflammatory conditions, in particular inflammatory conditions with an aetiology including an autoimmune component such as arthritis (for example rheumatoid arthritis, arthritis chronica progrediente and arthritis deformans) and rheumatic diseases, including inflammatory conditions and rheumatic diseases involving bone loss, inflammatory pain, spondyloarthropathies including ankolsing spondylitis, Reiter syndrome, reactive arthritis, psoriatic arthritis, and enterophathis arthritis,, hypersensitivity (including both airways hypersensitivity and dermal hypersensitivity) and allergies. Specific auto-immune diseases for which Antibodies of the Invention may be employed include autoimmune haematological disorders (including e.g. hemolytic anaemia, aplastic anaemia, pure red cell anaemia and idiopathic thrombocytopenia), systemic lupus erythematosus, inflammatory muscle disorders, polychondritis, sclerodoma, Wegener granulomatosis, dermatomyositis, chronic active hepatitis, myasthenia gravis, psoriasis, Steven- Johnson syndrome, idiopathic sprue, autoimmune inflammatory bowel disease (including e.g. ulcerative colitis, Crohn's disease and Irritable Bowel Syndrome), endocrine ophthalmopathy, Graves disease, sarcoidosis, multiple sclerosis, primary biliary cirrhosis, juvenile diabetes (diabetes mellitus type T), uveitis (anterior and posterior), keratoconjunctivitis sicca and vernal keratoconjunctivitis, interstitial lung fibrosis, psoriatic arthritis and glomerulonephritis (with and without nephrotic syndrome, e.g. including idiopathic nephrotic syndrome or minimal change nephropathy), tumors, multiple sclerosis, inflammatory disease of skin and cornea, myositis, loosening of bone implants, metabolic disorders, such as atherosclerosis, diabetes, and dislipidemia.
Antibodies of the Invention are also useful for the treatment, prevention, or amelioration of asthma, bronchitis, pneumoconiosis, pulmonary emphysema, and other obstructive or inflammatory diseases of the airways. Antibodies of the Invention are useful for treating undesirable acute and hyperacute inflammatory reactions which are mediated by EL-17or involve IL- 17 production, or the promotion of TNF release by DL-17, e.g. acute infections, for example septic shock (e.g., endotoxic shock and adult respiratory distress syndrome), meningitis, pneumonia; and severe burns; and for the treatment of cachexia or wasting syndrome associated with morbid TNF release, consequent to infection, cancer, or organ dysfunction, especially AIDS -related cachexia, e.g., associated with or consequential to HTV infection.
Antibodies of the Invention are particularly useful for treating diseases of bone metabolism including osteoarthritis, osteoporosis and other inflammatory arthritides, and bone loss in general, including age-related bone loss, and in particular periodontal disease.
For these indications, the appropriate dosage will, of course, vary depending upon, for example, the particular Antibody of the Invention to be employed, the host, the mode of administration and the nature and severity of the condition being treated. However, in prophylactic use, satisfactory results are generally indicated to be obtained at dosages from about 0.05 mg to about 10 mg per kilogram body weight more usually from about 0.1 mg to about 5 mg per kilogram body weight. The frequency of dosing for prophylactic uses will normally be in the range from about once per week up to about once every 3 months, more usually in the range from about once every 2 weeks up to about once every 10 weeks, e.g. once every 4 to 8 weeks. Antibody of the Invention is conveniently administered parenterally, intravenously, e.g. into the antecubital or other peripheral vein, intramuscularly, or subcutaneously. A prophylactic treatment typically comprises administering the Antibody of the Invention once per month to once every 2 to 3 months, or less frequently.
The Antibodies of the Invention may be administered as the sole active ingredient or in conjunction with, e.g. as an adjuvant to or in combination to, other drugs e.g. immunosuppressive or immunomodulating agents or other anti-inflammatory agents, e.g. for the treatment or prevention of diseases mentioned above. For example, the Antibodies of the Invention maybe used in combination with DMARD, e.g. Gold salts, sulphasalazine, antimalarias, methotrexate, D-penicillamine, azathioprine, mycophenolic acid, cyclosporine A, tacrolimus, sirolimus, minocycline, leflunomide, glucocorticoids; a calcineurin inhibitor, e.g. cyclosporin A or FK 506; a modulator of lymphocyte recirculation, e.g. FTY720 and FTY720 analogs; a mTOR inhibitor, e.g. rapamycin, 40-O-(2-hydroxyethyl)-rapamycin, CCI779, ABT578, AP23573 or TAFA-93; an ascomycin having immuno-suppressive properties, e.g. ABT-281, ASM981, etc.; corticosteroids; cyclo-phos-phamide; azathioprene; methotrexate; leflunomide; mizoribine; mycophenolic acid; myco-pheno-late mofetil; 15-deoxyspergualine or an immunosuppressive homologue, analogue or derivative thereof; immunosuppressive monoclonal antibodies, e.g., monoclonal antibodies to leukocyte receptors, e.g., MHC, CD2, CD3, CD4, CD7, CD8, CD25, CD28, CD40. CD45, CD58, CD80, CD86 or their ligands; other immunomodulatory compounds, e.g. a recombinant binding molecule having at least a portion of the extracellular domain of CTLA4 or a mutant thereof, e.g. an at least extracellular portion of CTLA4 or a mutant thereof joined to a non-CTLA4 protein sequence, e.g. CTLA4Ig (for ex. designated ATCC 68629) or a mutant thereof, e.g. LEA29Y; adhesion molecule inhibitors, e.g. LFA-I antagonists, ICAM-I or -3 antagonists, VCAM-4 antagonists or VLA-4 antagonists; or a chemotherapeutic agent, e.g. paclitaxel, gemcitabine, cisplatinum, doxorubicin or 5-fluorouracil; anti TNF agents, e.g. monoclonal antibodies to TNF, e.g. infliximab, adalimumab, CDP870, or receptor constructs to TNF-RI or TNF-RII, e.g. Etanercept, PEG-TNF-RI; blockers of proinflammatory cytokines, . IL-I blockers, e.g. Anakinra or IL-I trap, AAL160, ACZ 885, IL-6 blockers; chemokmes blockers, e.g inhibitors or activators of proteases, e.g. metalloproteases, anti-IL-15 antibodies, anti-IL-6 antibodies, anti-CD20 antibodies, NSAIDs, such as aspirin or an anti-infectious agent (list not limited to the agent mentioned).
In accordance with the foregoing the present invention provides in a yet further aspect:
A method as defined above comprising co-administration, e.g. concomitantly or in sequence, of a therapeutically effective amount of an IL-17 binding molecule, e.g. an Antibody of the Invention, and at least one second drug substance, said second drug substance being a immuno-suppressive / immunomodulatory, anti-inflammatory chemotherapeutic or anti- infectious drug, e.g. as indicated above.
Or, a therapeutic combination, e.g. a kit, comprising of a therapeutically effective amount of a) an IL-17 binding molecule, e.g. an Antibody of the Invention, and b) at least one second substance selected from a immuno-suppressive / immunomodulatory, anti-inflammatory chemotherapeutic or anti-infectious drug, e.g. as indicated above. The kit may comprise instructions for its administration.
Where the Antibodies of the Invention are administered in conjunction with other immuno-suppressive / immunomodulatory, anti-inflammatory chemotherapeutic or anti- infectious therapy, dosages of the co-administered combination compound will of course vary depending on the type of co-drug employed, e.g. whether it is a DMARD, anti-TNF. IL-I blocker or others, on the specific drug employed, on the condition being treated and so forth.
Pharmaceutical compositions of the invention maybe manufactured in conventional manner. A composition according to the invention is preferably provided in lyophilized form. For immediate administration it is dissolved in a suitable aqueous carrier, for example sterile water for injection or sterile buffered physiological saline. If it is considered desirable to make up a solution of larger volume for administration by infusion rather as a bolus injection, it is advantageous to incorporate human serum albumin or the patient's own heparinised blood into the saline at the time of formulation. Alternatively, the formulation is given subcutaneous. The presence of an excess of such physiologically inert protein prevents loss of antibody by adsorption onto the walls of the container and tubing used with the infusion solution. If albumin is used, a suitable concentration is from 0.5 to 4.5% by weight of the saline solution. Other formulations comprise liquid or lyophilized formulation.
The invention is further described by way of illustration in the following Examples.
EXAMPLES
Transgenic mice engineered to express the human IgG/κ repertoire instead of the murine immunoglobulin repertoire (Fishwild et al., 1996, Nat Biotechnol., 14, 845-851) are used to generate antibodies to human IL- 17. B cells from these mice are immortalized by standard hybridoma technology and murine hybridoma cells are obtained which secrete the human IgGl /K antibody AIN457.
Example 1: Generation of the hybridoma, purification of the antibodies, selection of AIN457 antibody
Production of recombinant human IL-I 7 (huIL-17): Recombinant huIL-17 is either produced in E. coli in inclusion bodies and refolded by conventional techniques (produced in house carrier free (E. coli; Novartis Pharma, batch BM-E3141/98) or bought (carrier free, E. coli; R&D Systems #317-IL/CF)) or as secreted and partially glycosylated protein in HEK.EBNA (Recombinant huIL-17, carrier free (IL- 17 APP-C6 from transfected HEK/EBNA cells; Novartis Pharma, batch En.E-3382/82; 0.28 mg/ml; recombinant huIL-17, carrier free (IL-17 APP-C4 from transfected HEK/EBNA cells; Novartis Pharma, batch En.E-3382/83; 0.29 mg/ml)). The latter form features a C-terminal 4 amino acids extension for rapid purification from culture supernatants by immunoaffϊnity chromatography. In this case, culture supernatants are loaded on a column of appropriate size of a specific immobilized anti-tag antibody coupled to CNBr activated Sepharose 4B at a density of 10 mg/ml resin following the manufacturer's instructions (Pharmacia). After base-line washing with PBS, bound huIL-17 is eluted with 100 mM glycine, pH 2.7 and immediately neutralized with diluted NaOH.
Coupling ofhuIL-17 to Keyhole Limpet Hemocyanin (KLH): HuEL-17 produced in either E. coli or HEK.EBNA is coupled to KLH pre-activated with an excess of the homobifunctional cross- linker Disuccinimidyl suberate (DSS). Briefly, 20 mg lyophilized Imject® Mariculture KLH (Pierce # 77600) are reconstituted with 2 ml H2O to give a 10 mg/ml solution containing Phosphate Buffered Saline (PBS), pH 7.2. To this solution 400 μl of 250 mM DSS in Dimethyl Sulfoxide (DMSO) are added and the mixture stirred for about 1 hr at room temperature (not all the reagent dissolved and some precipitate formed). After a brief centrifugation and filtration (0.45 μm) the solution is then desalted on Sephadex G25 fine (Pharmacia) in PBS (flow rate 2 ml/min) yielding about 11 mg activated KLH at 1.5 mg/ml (Bradford). 1 ml of the activated KLH (1.5 mg) is mixed with 1 ml of a 9 mg/ml solution in water of lyophilyzed E. coli derived huJL- 17 (batch BM-E3141/98). The solution remains clear and is incubated for 2 hrs at room temperature. The concentration of the resulting complex is 1.4 mg/ml (measured by Bradford). 1 ml of the activated KLH (1.5 mg) is mixed with 1 ml of HEK.EBNA huIL-17 (about 3 mg in water ; batch En.E-3382/83). The solution remains clear and is incubated for 2 hrs at room temperature. Concentration (Bradford) is 2.9 mg/ml.
Immunization: The genetically engineered mouse 27340 (female; MEDAREX Inc, Annandale, NJ) in which the murine immunoglobulin variable and constant part genes are functionally replaced by their human counterparts (Genotype Tg code 221100-TgH (CMD)++;TgN(Hco7)11952+; TgH(JKD)++; TgN(KCO5)9272+ (see also Sherie L. Morrison, 1994, Nature, Vol. 368, p. 812-813; Nils Lonberg et al., 1994, Nature, Vol. 368, p. 856-859) is immunized following the scheme reported in table 1. Table 1. Immunization schedule Day Date Immunogen Dose and route of immunization
0 07.06.01 HιύX-17 (BM-E3141/98) coupled to KLH 25 μg of each s.c. in two mixed 1:1 with huJJL-17 (BM-E3141/98) in spots; total volume/mouse Gerbu adjuvant with adjuvant 100 μl
14 21.06.01 HuJL-17 (BM-E3141/98) coupled to KLH 25 μg of each s.c. in two (1st boost) mixed 1:1 with huJL-17 (En.E-3382/83) spots; total volume/mouse coupled to KLH in Gerbu adjuvant with adjuvant 100 μl
28 05.07.01 HuJL-17 (BM-E3141/98) mixed 1:1 with 10 μg of each s.c. in two (2nd boost) huJL-17 (En.E-3382/83) in Gerbu adjuvant spots; total volume/mouse with adjuvant 100 μl
35 12.07.01 Sera collected for ELISA
42 19.07.01 HuJL-17 (BM-E3141/98) mixed 1:1 with 20 μg of each s.c. in two (3rd boost) huJL-17 (En.E-3382/83) coupled to KLH in spots; total volume/mouse
Gerbu adjuvant with adjuvant 100 μl
63 09.08.01 HuJL-17 (BM-E3141/98) coupled to KLH 20 μg of each s.c. in two (4th boost) mixed 1:1 with huJL-17 (En.E-3382/83) spots; total volume/mouse coupled to KLH in Gerbu adjuvant with adjuvant 100 μl
91 06.09.01 HuJL-17 (BM-E3141/98) mixed 1:1 with 20 μg of each s.c. in two (5th boost) huIL-17 (En.E-3382/83) in Gerbu adjuvant spots; total volume/mouse with adjuvant 100 μl 99 14.09.01 Sera collected for ELISA
117 02.10.01 HuIL-17 (En.E-3382/83) 10 μg/mouse i.v.
HuIL- 17 (En.E-3382/83) coupled to KLH 10 μg/mouse i.p.
118 03.10.01 HuEL-17 (En.E-3382/83) coupled to KLH 10 μg/mouse i.p.
119 04.10.01 HuIL-17 (En.E-3382/83) coupled to KLH 10 μg/mouse i.p.
120 05.10.01 fusion
Sera samples are obtained 35 and 99 days after the start of the immunization protocol, for measuring levels of anti-huIL17 antibody by enzyme-linked immunosorbent assay (ELISA).
Generation ofhybridomas: On day 120, mouse 27340 is killed by CO2 inhalation. Total spleen cells (1 x 108) are fused with PAI-O cells (5 x 107 cells) using PEG 4000. Fused cells are plated out in 720 wells (1 ml/well), containing a feeder layer of mouse peritoneal cells (Balb/c mice), in HAT medium (RPMI 1640 containing 2 g/1 Sodium Bicarbonate, 5 x 10"5 M j8-Mercaptoethanol, 10"4 M Hypoxanthine, 1.6 x 10'5 M Thymidine, 4 x 10'7 M Aminopterin, 10% heat inactivated FCS and 50 μg/ml Gentamycin). At day 14, HAT medium is exchanged with HT medium i.e. HAT medium without Aminopterin. Screening starts on day 10, and lasts two weeks. Of the initial 720 wells plated, 684 wells (95%) are positive for hybridoma growth. Supernatants are collected and screened for huIL-17 reactive MAb in ELISA using both the E. coli and the HEKTEBNA derived huIL-17. Fifty-two primary wells score positive for the presence of anti- huIL-17 antibodies. Twenty-eight hybridomas are cloned and the remaining ones are frozen. Cloning is done, in 4 x 96 well microtiter plates, in HT medium and a feeder layer of mouse peritoneal cells. Hybridomas are plated at 0.5 cell/100 μl per well. Wells are screened microscopically for growth, and positive ones are fed 100 μl of HT medium. The following day, supernatants are tested for antibody production in a huIL-17 specific ELISA. Upon cloning, the majority of the cloned hybridomas retain the capacity to secrete anti-huIL-17 specific Monoclonal Antibody (MAb).
Production and purification of antibody: The selected clones are transferred in serum free medium (5 ml) into 25 cm2 TC (TC: tissue culture) flasks. Hybridomas are progressively expanded in serum free medium to 75 cm2 TC flasks and roller flasks. All the different anti-hu- IL- 17 MAb including NVP-AM457-NX (340-110-28 i.e. mouse number-hybridoma number- clone number) are purified by Protein A affinity chromatography. Culture supernatants are adjusted to pH 7.3 and loaded on a column of appropriate size of Protein A Sepharose 4 fast flow (Pharmacia). After base-line washing with 100 mM phosphate buffer, pH 7.3 bound antibodies are eluted with 50 mM citrate, pH 2.7, 140 mM NaCl. The eluted fraction is immediately neutralized (pH 7.0) and sterile filtered. Protein concentration is determined by absorption at 280 nm using a factor of 1.35 Absorption Unit (AU)/mg.
Inhibitory activity ofanti-huIL-17 MAb on IL-6 production induced by huIL-17 in human dermal fibroblasts: Human dermal fibroblasts are cultured in FBM supplemented with 2% FCS, insulin (5 μg/ml) huFGF-basic (0.1 μg/ml) and Gentamycin (50 μg/ml). The fibroblasts are detached from plastic using a Trypsin/EDTA solution. Fibroblasts are distributed into 96 well microtiter plates at a density of 1x104 cells/well in FBM supplemented with 1% FCS. Fibroblasts are allowed to adhere to the plates overnight. The next morning medium is removed and fresh FBM supplemented with 1% FCS, huBL-17 (different concentrations ranging from 30 to 500 ng/ml) and hybridoma supernatants (1/5 final dilution) or purified antibodies are added to a final volume of 200 μl. Culture supernatants are collected after an incubation of 24 h and huIL-6 production is measured by ELISA.
ELISA for detection of anti-huIL- 17 antibodies: ELISA microtiter plates are coated with recombinant huIL-17 (100 μl/well at 3 μg/ml; batch BM-E3141/98 or En.E-3382/82) in PBS 0.02% NaN3 and incubated overnight at room temperature. The following day, microtiter plates are blocked with 300 μl of PBS/ 2% BSA/ 0.02% NaN3 for 2 h at 370C. Plates are then washed 4 times with PBS/ 0.05% Tween 20/ 0.02% NaN3. Serum dilutions of mouse 27340 (final dilution range at day 35: 1/100 to 1/3200; final dilution range at day 99: 1/200 to 1/12800; 100 μl/well) or culture supernatants of hybridomas (final dilution 1:3; 100 μl/well) are added. After an overnight incubation at room temperature, plates are washed 4 times with PBS/ 0.05% Tween 20/ 0.02% NaN3. A biotin-conjugated mouse anti-hu-IgG, Fc fragment specific antibody is added at a final dilution of 1/20000 (100 μl/well). Samples are left to react for 4 h at room temperature. After washing (4 times), alkaline phosphatase-conjugated streptavidin is added at a final dilution of 1/8000 (100 μl/well). After 40 minutes at room temperature, plates are washed again 4 times and the substrate (p-nitrophenylphosphate in diethylamino buffer pH 9.8; 150 μl/well) is added. Plates are read after 30 or 45 min depending on the development of the reaction in a microtiter reader (Bio-Rad) using filters of 405 and 490 nm.
ELISA for detection of antibody isotype: For revealing the isotype of the MAb, culture supernatants (100 μl; final dilution 1/5) are added to the wells of microtiter plates coated with huIL-17 (see above), and incubated overnight at room temperature. After washing (4 times), 100 μl/well of biotin-conjugated mouse MAbs anti-human IgGl (final dilution 1/1000), IgG2 (final dilution 1/1000), IgG3 (final dilution 1/1000) IgG4 (final dilution 1/2000) or anti human K light chain (final dilution 1/1000) are added for 4 h at room temperature. As a control a biotin- conjugated rat anti-mouse λl and λ2 light chain specific MAb is used (final dilution 1/1000). This is followed as previously described by washing and addition of alkaline phosphatase- conjugated streptavidin (100 μl; final dilution 1/8000). After washing (4 times) the substrate (p- nitrophenylphosphate in diethylamino buffer; 100 μl) is added. Plates are read after 30 or 45 min depending on development of reaction, in a microtiter reader (Bio-Rad) using filters of 405 and 490 nm.
ELISA for detection of huIL-6 production: ELISA microtiter plates are coated with an anti-huIL- 6 mouse MAb (MAB206 from R&D system; 100 μl/well at 4 μg/ml) in PBS 0.02% NaN3 and incubated overnight at room temperature. The following day, microtiter plates were blocked with 300 μl of PBS/ 2% BSA/ 0.02% NaN3 for 2 h at 370C. Plates were then washed 4 times with PBS/ 0.05% Tween 20/ 0.02% NaN3. Culture supernatants of human dermal fibroblasts (final dilution 1:3; 100 μl/well) were added. To establish a titration curve huIL-6 (100 μl/well) is titrated from 400 pg/ml to 3.1 pg/ml in 1:2 dilution steps. After an overnight incubation at room temperature, plates are washed 4 times with PBS/ 0.05% Tween 20/ 0.02% NaN3. A biotin- conjugated goat anti-huIL-6 antibody (BAP206; R&D Systems) is added (25 ng/ml; 100 μl/well). Samples are left to react for 4 h at room temperature. After washing (4 times), alkaline phosphatase-conjugated streptavidin is added at a final dilution of 1/8000 (100 μl/well). After 40 minutes at room temperature, plates are washed again 4 times and the substrate (p- nitrophenylphosphate in diethylamino buffer pH 9.8; 150 μl/well) is added. Plates are read after 30 min in a microtiter reader (Bio-Rad) using filters of 405 and 490 nm. Calculations: Values are reported as original O.D. values or as % inhibition calculated on the means of duplicate values. Additional data are reported as Means ± SEM. An huIL-6 standard curve was used to measure huIL-6 concentration in culture supernatants by using a cubic curve fit. Results
Serum titers of mouse 27340:
Table 2. Anti-huIL-17 serum titers (mouse 27340)
Serum dilution
O.D. values (Means ± SEM)
Day HuIL-17
1:100 1:200 1:400 1:800 1:1600 1:3200 1:6400 1:12800 batch*
1.795 1.524 1.167 0.854 0.615 0.378 E. coli
± 0.022 ± 0.006 ± 0.015 ± 0.013 ± 0.005 ± 0.032 35
2.180 1.875 1.577 1.313 1.031 0.728 HEKJEBNA
± 0.041 ± 0.005 ± 0.047 ± 0.016 ± 0.011 ± 0.003
2.130 1.913 1.635 1.494 1.125 0.810 0.559 E. coli
± 0.078 ± 0.075 ± 0.041 ± 0.066 ± 0.001 ± 0.070 ± 0.021 99
2.029 1.925 1.716 1.524 1.259 0.970 0.706 HEKJEBNA
± 0.005 ± 0.030 ± 0.012 ± 0.004 ± 0.018 ± 0.036 ± 0.002
* Microtiter plates were coated with huIL-17 (3 μg/ml) from E. coli (BM-E3141/98) or HEK/EBNA cells (En.E-3382/82).
The serum of mouse 27340 is analyzed in ELISA for the presence of anti-huIL-17 antibodies on days 35 and 99 on two different preparations of huIL-17 (Table 2). Results show that serum titers of mouse 27340 increase about fourfold between day 35 and day 99 and that both huIL-17 preparations are recognized.
Binding in ELISA ofhybridoma supernatants: 684 supernatants are tested in ELISA for the presence of anti-huIL-17 antibodies, using two preparations of recombinant huIL-17, the former from E coli (BM-E3141/98) the latter from HEK/EBNA cells (En.E-3382/82). Fifty-two supernatants score positive for the presence of anti-huIL-17 antibodies (Table 3). Preferential binding to one or the other preparation of huIL-17 is observed in a few cases. The 28 hybridomas that are subsequently cloned are underlined. Table 3. ELISA reactivity of culture supernatants
HuIL-17 batch* HuIL-17 batch
Hybridoma Hybridoma E. coli HEK/EBNA E. coli HEK/EBNA
(No) (No)
O.D. values O.D. values O.D. values O.D. values
1 1.935/1.830 ND 386 1.780/1.812 2.002/1.905
3 1.928/1.928 2.026/1.956 435 2.194/2.139 2.221/2.169
5 1.386/1.471 2.099/2.042 439 1.180/1.236 1.442/1.470
59 1.917/2.078 2.342/2.384 444 1.034/1.066 1.166/1.138
66 1.629/1.619 ND 450 2.060/2.209 2.079/2.237
104 2.650/2.716 2.439/2.366 477 1.392/1.348 1.515/1.524
106 1.329/1.371 1.362/1.465 496 2.131/2.078 2.569/2.798
110 2.355/2.363 2.425/2.497 504 1.755/1.559 ND
112 0.789/0.857 1.154/1.208 543 2.332/2.455 2.370/2.381
116 1.656/1.652 ND 544 1.145/1.196 1.187/1.201
128 1.244/1.669 0.714/0.695 548 0.728/0.750 0.891/0.909
142 1.192/1.322 0.847/0.810 552 0.824/0.811 0.969/0.943
173 1.899/2.108 1.966/2.023 557 2.241/2.326 2.347/2.483
182 0.948/0.903 0.874/0.866 564 0.628/0.675 0.808/0.820
190 2.249/2.084 2.150/2.139 566 1.092/1.068 1.239/1.152
196 1.406/1.305 1.797/1.752 577 1.018/0.928 1.226/1.206
216 1.120/1.146 1.114/1.128 597 0.781/0.821 1.117/1.121
234 1.890/1.990 ND 612 1.935/1.777 2.033/1.989
277 1.674/1.640 ND 622 2.121/2.230 2.592/2.277
285 0.678/0.789 0.735/0.784 627 1.000/1.077 1.203/1.209
298 2.475/2.677 2.340/2.358 649 1.335/1.389 1.311/1.337
305 1.721/1.789 0.602/0.634 658 1.218/1.297 1.415/1.437
319 1.111/1.073 1.223/1.202 674 1.112/1.087 1.134/1.127
328 1.738/1.762 1.869/1.835 686 1.447/1.549 1.730/1.646
343 2.478/2.702 2.302/2.448 705 1.899/1.803 1.870/1.872
373 1.200/1.194 1.212/1.233 720 2.249/2.420 2.383/2.385
*Plates are coated with recombinant huIL-17 (3 μg/ml) from E. coli (BM-E3141/98) or HEK/EBNA cells (En.E-3382/82). Supernatants are tested at the final dilution of 1A. Binding in ELISA of culture supematants of the hybridoma clones: The reactivity in ELISA of the supematants of the clones of the 11 hybridomas, which retained the best production of anti- huIL-17 MAb, is shown in Table 4. The clones, highlighted in bold, were selected for producing ~ 1 liter of supernatant in roller bottles for purification and analysis of the antibodies. With the exception of the clones derived from the hybridoma No 5, which produced a huIgG3κ antibody, all the other clones produced huIgGlκ MAb, as assessed by isotype specific monoclonal antibodies.
Table 4. ELISA reactivity of culture supematants for hu-IL-17.
Clone Supematants* Clone Supematants* Clone Supematants*
(No) O.D. values (No) O.D. values (No) O.D. values
3-2 2.198/1.940 106-1 1.244/1.306 543-4 1.003/0.913
3-20 1.909/1.939 106.2 1.203/1.138 543-16 0.795/0.717
3-21 1.873/1.812 106-3 1.176/1.166 557-6 0.879/0.940
5-18 1.240/1.168 110-7 1.535/1.393 557-36 0.980/0.925
5-22 1.340/1.396 110-28 1.376/1.370 557-37 1.104/1.109
5.29 1.316/1.354 305-21 1.484/1.518 622-2 0.923/0.894
5.31 1.227/1.302 305-38 1.669/1.858 622-5 1.070/1.032
5-40 1.364/1.543 343-1 1.351/1.375 622-6 0.980/0.953
104-2 1.385/1.299 439-80 2.506/2.543 658-2 0.744/0.744
104-4 1.085/1.044 450-13 1.568/1.610 658-6 0.769/0.772
104-9 1.488/1.304 450-23 1.658/1.667 658-16 0.741/0.758
104-11 1.670/1.380 543-1 1.074/0.991
Microtiter plates are coated with recombinant huDL-17 (3 μg/ml) from HEK/EBNA cells (En.E- 3382/82).
Neutralizing activity of culture supematants: Culture supematants are tested for inhibition of huIL-6 production by human dermal fibroblasts stimulated with recombinant huIL-17. As shown in Table 5, the majority of the culture supematants show inhibitory activity. Table 5. Inhibition of IL-6 production induced by huEL-17 in human dermal fibroblasts by culture supernatants
Inhibition of IL-6 production (%)
Clone Amount ofhuIL-17 used as stimulus (ng/ml)
(No) 62.5 125 250 500
3-20 86.3 75.0 33.1 23.2
5-40 23.3 41.4 20.3 19.0
104-11 47.7 48.5 22.2 16.3
106.1 61.6 19.8 5.7 9.8
110-28 99.8 92.5 88.6 61.3
305-38 47.2 47.1 36.6 23.7
343-1 96.8 102.4 90.5 66.4
450-23 51.7 48.5 47.5 26.6
543.4 -6.0 -12.0 -6.5 -7.1
622-2 34.0 23.2 20.3 18.4
658-16 34.4 27.7 12.7 18.8
Neutralizing activity ofAIN45: Selection of clone 110-28 for the production of development candidate AIN457 (preferred embodiment of the invention) is based on neutralizing activity and affinity measurement on BIACORE 2000 of the purified antibodies (see below Example 2).
Example 2: AIN457 binds with very high affinity to recombinant human IL-17 (huIL-17); the KD is 122 ± 22 pM (BIAcore) and neutralizes human IL-6 production induced by huIL- 17 in human dermal fibroblast; IC50 is 2.1±0.1 nM at a concentration of 1.87nM huIL-17
a) Methods
Reagents: General laboratory reagents are purchased from Merck or Sigma and are of the highest purity grade available; the sources of specialty reagents are detailed below.
Proteins: Monoclonal antibodies are generated by immunizing MEDAREX transgenic mice with recombinant human IL-17, and then following the standard procedure for producing cell lines, from which the secreted material could be purified by Protein A Sepharose chromatography [essentially as described in Example 1). AIN457 is stored as a sterile-filtered solution in 50 mM Na-citrate, pH 7.0, 140 mM NaCl at 4°C . The recombinant human AIN457 (batch KB03303A) is obtained in sterile stock solution of either 20 mM Na-citrate/40 mM phosphate buffer, pH 7, 150 mM NaCl or 20 mM acetic acid pH 5.5 adjusted with IM Tris-base. Concentrations are usually in the range of 2 mg/ml and diluted to a final concentration of 5 μg/ml into BIA buffer (20 mM HEPES, pH 7,4, 150 mM NaCl, 0,05% v/v Tween-20) for the Biacore experiments.
Recombinant human IL-17 is produced in-house; batch En/E 3882/83; 0.29 mg/ml.
BIAcore measurements
Determination of kinetic binding parameters and levels of crossreactivity are done by surface plasmon resonance measurements using the optical biosensor BIAcore 2000 (BIAcore AB, Upsalla, Sweden, see Lit. HS 1,2 for details). This technology allows the label-free determination the microscopic rate constants for binding (Ic0n) and dissociation (koff) of a ligand to a receptor. It is therefore especially suited for characterizing the antibody-antigen interactions. This technology complements and is in many respects superior to ELISA measurements (Van Regenmortel, Dev Biol (Basel). 2003;112:141-51.).Binding studies of recombinant IL-17 to the IL-17 antibody ATN457 are performed in two ways. In the standard protocol, ATN457 is captured by an anti- human Fcγ antibody (Jackson Immunochemicals; Cat.No. 109-005-098) that is previously immobilized onto a CM-5 BIAcore sensor chip (Research grade). Covalent binding of Fcγ capture antibody is done with the 'Amine coupling kit' provided by BIAcore (BIAcore, Cat.No. BR-1000-50). Typically, 3000 RUs of capture antibody are attached to the activated dextran surface with a 30μg/ml anti Fcγ antibody solution in 10 mM Ac buffer, pH 4.5 at a flow rate of 5 μl/min which lead to approximately 250 RUs of AIN457 immobilization. As a guideline, 1000 RUs correspond to a mass transfer of 1 ng/mm2. Alternatively, IL-17 (Section 3.2; Table 4), AIN457 antibody is coupled directly to the chip surface without capture antibody. The results are compared to the protocol described in Table 9 (see below).
b)Results
Binding kinetics of the IL-17/ AIN457 complex
The equilibrium dissociation constant KD allows some judgement about the stability of complexes, once formed in vivo. We have therefore determined kinetic constants for the binding of human IL-17 to the immobilized AIN457 antibody, and have derived the KD for the process from these data. Table 3 shows the summary of data obtained when the curves of 2 experiments are fitted to the Langmuir model using the BIAevaluation 3.0 software. Although the antibody is, of course, bivalent, the binding can be treated as a 1:1 event, with individual antibody binding sites displayed at the surface that become occupied by monomelic IL-17 molecules. This experiment shows both, the extremely fast association as well as the very slow dissociation kinetics of the antibody-chemokine complex. The best data fit is obtained when the sensorgrams are treated individually (rather than globally, as is suggested in the BIAevaluation.) Thus, after combining the titration series we obtain average values from 12 sensorgrams OfIc0n= (4.1 ± 0.1) xlO5 1/M s; koff= (3.8 ± 0.5) xlO"4 1/s; and for KD= 122 ± 22 pM.
Table 3. Kinetic constants for the 1:1 binding of rec human IL-17 to NVP-AIN457
Cone [nM] kon [I/Ms] koff [l/s] KD [M] Exp. IL-314
2 3.31E+05 3.36E-05 1.02E-10 Run l
4 1.28E+05 3.78E-05 2.95E-10
8 3.79E+05 1.86E-05 4.90E-11
12 3.60E+05 3.00E-05 8.33E-11
16 3.52E+05 5.70E-05 1.62E-10
20 3.52E+05 4.15E-05 1.18E-10
2 1.23E+06 1.97E-05 1.60E-11 Run 2
4 4.11E+05 1.20E-05 2.92E-11
8 3.78E+05 4.54E-05 1.20E-10
12 3.46E+05 5.13E-05 1.48E-10
16 3.17E+05 5.95E-05 1.88E-10
20 3.34E+05 5.01E-05 1.50E-10
Mean 4.10E+05 3.80E-05 1.22E-10 n=12
SEM 7.73E+04 4.51E-06 2.21E-11
Mean KD calculated from individual entries (vertically), rather than by applying the equation K0=Ic0J5Zk0n.
For the AIN457 produced in recombinant cells (KB03303A) affinity measurements are performed for the EL-17 cytokines from man, marmoset, rhesus and cynomolgous monkey, respectively. Experimental details of the Biacore measurements are the same as described above for MABllO-28 antibody. Two independent runs testing 6 IL-17 concentrations in each run are performed. Concentrations for human EL-17 are 2, 4, 8, 12, 16, 20 nM and 10, 20, 30, 40, 50, 60 nM for all other species. Complete data analysis yields n=12 individual measurements for each TL- 17 species. The KD as well as SEM is reported.
Table 4. Summary: Kinetic constants for the 1:1 binding of rec human, marmoset, rhesus and cynomolgous monkey IL-17 to NVP-AIN457 (KB03303A)
Species KD [M] SEM
Mean
Run 1+2
Human 0.227 nM +/- 0.03 nM
Marmoset 1.2 nM +/- 0.1 nM
Rhesus monkey 9 nM +/- I nM
Cynomolgous 6 nM +/- 0.7 nM monkey
A full set data of the BIAcore analysis for antibody KB03303A with Ic0n, koff and KD and the respective IL- 17 species are given below in tables 5 to 8.
Table 5. Kinetic constants for the 1:1 binding of rec human IL-17 to AIN457 (KB03303A)
Conc [nM] kon [l/Ms] koff [l/s] KD [M] Exp. IL-366/ IL-365
2 3.37E+05 6.43E-05 1.91E-10 Run l
4 2.59E+05 7.76E-05 2.99E-10
8 2.12E+05 5.21E-05 2.46E-10
12 2.18E+05 7.38E-05 3.38E-10
16 2.02E+05 7.15E-05 3.54E-10
20 1.92E+05 8.04E-05 4.20E-10
2 5.50E+05 7.01E-05 1.27E-10 Run 2
4 3.22E+05 3.30E-05 1.02E-10
8 2.85E+05 4.73E-05 1.66E-10
12 2.86E+05 4.84E-05 1.69E-10 16 2.61E+05 3.09E-05 1.18E-10
20 2.58E+05 4.90E-05 1.90E-10
Mean 2.82E+05 5.82E-05 2.27E-10 n=12
SEM 2.77E+04 4.91E-06 3.00E-11
Table 6. Kinetic constants for the 1 : 1 binding of rec marmoset IL-17 to AIN457
(KB03303A)
Cone [nM] kon [I/Ms] koff [l/s] KD [M] Exp. IL-366/
IL-365
1OnM 8.89E+04 7.96E-05 8.95E-10 Run 1
2OnM 1.11E+05 8.69E-05 7.82E-10
3OnM 9.82E+04 1.15E-04 1.17E-09
4OnM 9.92E+04 1.16E-04 1.17E-09
5OnM 9.81E+04 1.19E-04 1.21E-09
1OnM 8.83E+04 9.98E-05 1.13E-09 Run 2
2OnM 1.10E+05 1.28E-04 1.17E-09
3OnM 9.70E+04 1.52E-04 1.57E-09
4OnM 9.66E+04 1.31E-04 1.36E-09
5OnM 9.52E+04 1.59E-04 1.67E-09
Mean 9.83E+04 1.19E-04 1.21E-09 n=10
SEM 2.36E+03 8.09E-06 ±0.1
Table 7. Kinetic constants for the 1:1 binding of rec rhesus monkey IL-17 to AIN457 (KB03303A)
Cone [nM] kon [I/Ms] koff [l/s] KD [M] Exp. IL-366/
IL-365
10 1.70E+05 3.89E-04 2.28E-09 Run l
20 6.73E+04 4.94E-04 7.34E-09
30 5.86E+04 3.54E-04 6.04E-09
40 3.27E+04 4.05E-04 1.24E-08
50 4.05E+04 4.55E-04 1.12E-08
60 3.50E+04 4.60E-04 1.31E-08 10 5.47E+04 3.85E-04 7.04E-09 Run 2
20 4.62E+04 2.74E-04 5.93E-09
30 4.30E+04 3.51E-04 8.16E-09
40 3.76E+04 3.66E-04 9.74E-09
50 3.60E+04 4.32E-04 1.20E-08
60 3.44E+04 4.24E-04 1.23E-08
Mean 5.47E+04 3.99E-04 8.96E-09 n=12
SEM 1.09E+04 1.72E-05 9.70E-10
ble 8. Kinetic constants for the 1 :1 binding of rec cvnomolεous monkey IL-
AIN457 (KB03303A)
Conc [nM] kon [I/Ms] koff [l/s] KD [M] Exp. IL-366/ IL-365
5nM 3.27E+05 3.60E-04 1.10E-09 Run 1
1OnM 1.79E+05 4.02E-04 2.24E-09
15nM 1.03E+05 5.67E-04 5.50E-09
2OnM 1.10E+05 5.23E-04 4.75E-09
25nM 9.23E+04 5.78E-04 6.26E-09
3OnM 9.05E+04 7.14E-04 7.89E-09
5nM 7.18E+04 5.08E-04 7.08E-09 Run 2
1OnM 9.70E+04 6.69E-04 6.90E-09
15nM 1.03E+05 7.66E-04 7.41E-09
2OnM 1.02E+05 7.32E-04 7.17E-09
25nM 1.02E+05 7.47E-04 7.34E-09
3OnM 1.00E+05 8.34E-04 8.32E-09
Mean 1.23E+05 6.17E-04 6.00E-09 n=10
SEM 1.99E+04 4.34E-05 6.52E-10
Subsequently inhibitory activity of purified AIN457 (Batch En/E-10333/53; 0.54 mg/ml) on huDL-17 is evaluated. IC50 values are shown in Table 6. In these experiments, huDL-17R/Fc and a mouse anti-huIL-17 MAb are included as positive controls and Siniulect as negative control. Table 9. Neutralization of hu-IL-17 by the human anti-huIL-17 MAb AIN457 in comparison with IL-17R/Fc, and a mouse anti-huIL-17 MAb (R&D System).
AIN457 JLrITEUFc MAB 317
IC50 ± SEM IC50 ± SEM IC50 ± SEM
(n=3*) (n=3) (n=3)
Recombinant huIL-17 2.071 ± 0.116 nM 1.713 ± 0.305 nM 12.226 ± 2.050 nM
@ 1.87 nM (30 ng/ml)
*Means and SEM are calculated from three different and independent experiments.
In conclusion, AIN457 abrogates the IL-17-dependent secretion of huIL-6 by human dermal fibroblasts. The potency is comparable to that of huIL-17R/Fc and superior to that of a commercially available mouse anti-huIL-17 MAb. It is interesting to note that a more complete inhibition is observed with AIN457 than with TL-ITRfFc.
Example 3: Purity and partial amino acid sequences of heavy and light chain Amino acid sequencing
Amino-terminal amino acid sequences of V∑ and VH regions: The first 48 amino-acid residues of the heavy and the light chain for two anti~IL-17A antibodies, clone 110-7 (see table 4) and 110- 28 (see table 4), are determined by Edman degradation. The amino-acid sequence is identical for both clones. The GeneBank is searched by blast analysis and the most homologous DNA sequence found is used to design the cloning primers.
Molecular cloning of the VL and VH regions: Total RNA is prepared from 2x107 hybridoma cells (clone 110-7, clone 110-28) with the RNeasy Midi Kit according to the vendor's protocol (Quiagen Hilden Germany). Total RNA is eluted in 200μl RNase-free water and stored at -800C. The first strand cDNA synthesis is carried out with M-MLV reverse transcriptase (Promega, Madison, WT), oligo-dT primer, PCR nucleotide mix (dNTPs) and RNAsin inhibitor (Roche, Mannheim). Five μg of total RNA is mixed with 1 μl oligo-dT primer (0.5μg/μl), and RNase- free water is added to a final volume of 36 μl. The mixture is incubated at 70°C for 10 minutes and then stored on ice. While on ice, the following reagents are added: lOμl 5x RT buffer, 2μl dNTPs (1OmM each), 2 μl RNasin and lμl M-MLV reverse transcriptase. The reaction is carried out at 42°C for lhour. The PCR reaction is assembled using 4μl of cDNA template, 2μl of each primer at 1 OμM each (see below and Tables 10 and 11 for overview) 20μl of 2xQiamix (containing Buffer, dNTP's, TAQPolymerase) and lμl of Pwo DNA polymerase in a total volume of 40 μl. The PCR conditions are set for 35 cycles of 940C for 15 seconds, 55°C for 20 seconds and 72°C for 30 seconds. The PCR product is subcloned into the pCR4-TOPO-Zero (Stragagene, La Jolla, Ca.) cloning vector. Several clones are picked from each reaction and the nucleotide sequence determined by Solvias AG (Basel), using the primers MV432 (SEQ E) NO: 21), MV433 (SEQ ID NO: 22), MV434 (SEQ ID NO: 23), MV435 (SEQ ID NO: 14), and standard primers in the vector DNA.
The cDNA encoding the heavy chain is amplified using the primer pairs MV416 (SEQ ID NO: 15)/#265 (SEQ ID NO: 16) and MV418 (SEQ ID NO: 17) /#265 (SEQ ID NO: 16). The primers cover the nucleotide sequences corresponding to the following amino acid positions of the heavy chain: MV416 position -19/-13 (signal peptide); MV418 position +1/+7; #265 position +2531+259. Position +1 is the first amino acid of the mature protein.
The cDNA encoding the light chain is amplified using the primer pairs MV417 (SEQ ID NO: 18)/#223 (SEQ ID NO: 19) and MV419 (SEQ ID NO: 20)/#223 (SEQ ID NO: 19). The primers cover the nucleotide sequences corresponding to the following amino-acid positions of the light chain: MV417 position -20/-14 (signal peptide); MV419 position +1/+7; #223 position +210/+215. This approach permitted to make two independent PCR amplifications for each immunoglobulin chain, resulting in two independently established DNA sequences.
Results and Discussion
The cloned PCR products coding for the heavy and light chain from two hybridomas (110-7 and 110-28, see Table 4 above) are characterized by DNA sequencing. Five and six independent sequences are used to assemble the light and heavy chain sequences.. The light chain cDNAs are all identical and covere the entire coding sequence (amino acid position -20 to +215). The heavy chain cDNAs had 2 different mismatches in one cDNA each. These are excluded from the final sequence, which extends from the initiation codon to the end of the hinge region after the constant domain 1 (amino acid position -19 to +238). The sequences for both hybridomas are identical. The cDNA obtained from hybridoma 110-28 is selected and used for all further expression work. SEQ ID NO: 7 (cDNA of heavy chain of AIN457), SEQ ID NO: 8 (amino acid sequence of heavy chain of ATN457), SEQ ID NO: 9 (cDNA of light chain of AIN457) and SEQ E) NO: 10 (amino acid sequence of AIN457) show the DNA sequence coding for the light and heavy chain of AIN457, along with the protein sequence and the position of the primers used for PCR amplification and DNA sequencing. The DNA sequences have been registered in PlasNova, accession number NPL003689 for the heavy chain, and accession number NPL003690 for the light chain.
The amino acid sequence found by cDNA cloning is identical to that previously obtained by Edman degradation of the purified immunoglobulin heavy and light chains, indicating that the correct cDNA has been cloned.
Table 10: Nucleotide and amino-acid sequence of the light chain
The amino-acid sequence coding for the variable domain is bold and underlined. The oligonucleotide primers used for cloning are indicated (underlined).
MV417 ACCATGGAAACCCCAGCGGAGCTTCTCTTCCTCCTGCTACTCTGGCTCCCAGATACCACC
X + H + + + + go
TGGTACCTTTGGGGTCGCCTCGAAGAGAAGGAGGACGATGAGACCGAGGGTCTATGGTGG T M E T P A E L L F L L L L W L P D T T
MV419 GGAGAAATTGTGTTGACGCAGTCTCCAGGCACCCTGTCTTTGTCTCCAGGGGAAAGAGCC
61 + + + + + + 120
CCTCTTTAACACAACTGCGTCAGAGGTCCGTGGGACAGAAACAGAGGTCCCCTTTCTCGG
L T Q S G T L S G E R A
ACCCTCTCCTGCAGGGCCAGTCAGAGTGTTAGCAGCAGCTACTTAGCCTGGTACCAGCAG X2X ———— _^._—.__.._.___.{.--.--._______^__________-^~.________,f._________^. 3.80
TGGGAGAGGACGTCCCGGTCAGTCTCACAATCGTCGTCGATGAATCGGACCATGGTCGTC T L S C R A S Q S V S S S Y L A W Y Q Q
AAACCTGGCCAGGCTCCCAGGCTCCTCATCTATGGTGCATCCAGCAGGGCCACTGGCATC
181 + + + + + + 240
TTTGGACCGGTCCGAGGGTCCGAGGAGTAGATACCACGTAGGTCGTCCCGGTGACCGTAG
K P G Q A P R L L I Y G A S S R A T Q I
CCAGACAGGTTCAGTGGCAGTGGGTCTGGGACAGACTTCACTCTCACCATCAGCAGACTG
241 + + + + + + 300
GGTCTGTCCAAGTCACCGTCACCCAGACCCTGTCTGAAGTGAGAGTGGTAGTCGTCTGAC P D R F S G S G S G T D F T L T I S R L
GAGCCTGAAGATTTTGCAGTGTATTACTGTCAGCAGTATGGTAGCTCACCGTGCACCTTC
301 + + + + + + 360
CTCGGACTTCTAAAACGTCACATAATGACAGTCGTCATACCATCGAGTGGAGCGTGGAAG
E P E D F A V Y Y C Q Q Y G S S P C T F
GGCCAAGGGACACGACTGGAGATTAAACGAACTGTGGCTGCACCATCTGTCTTCATCTTC
361 + + + + + + 420
CCGGTTCCCTGTGCTGACCTCTAATTTGCTTGACACCGACGTGGTAGACAGAAGTAGAAG
G Q G T R L E I K R T V A A P S V F I F
CCGCCATCTGATGAGCAGTTGAAATCTGGAACTGCCTCTGTTGTGTGCCTGCTGAATAAC
421 + + + + + + 480
GGCGGTAGACTACTCGTCAACTTTAGACCTTGACGGAGACAACACACGGACGACTTATTG
P P S D E Q L K S G T A S V V C L L N N
TTCTATCCCAGAGAGGCCAAAGTACAGTGGAAGGTGGATAACGCCCTCCAATCGGGTAAC
481 + + + + + + 540
AAGATAGGGTCTCTCCGGTTTCATGTCACCTTCCACCTATTGCGGGAGGTTAGCCCATTG
F Y P R E A K V Q W K V D N A L Q S G N
TCCCAGGAGAGTGTCACAGAGCAGGACAGCAAGGACAGCACCTACAGCCTCAGCAGCACC
541 + + + + + + 600
AGGGTCCTCTCACAGTGTCTCGTCCTGTCGTTCCTGTCGTGGATGTCGGAGTCGTCGTGG
S Q E S V T E Q D S K D Ξ T Y S L S S T
CTGACGCTGAGCAAAGCAGACTACGAGAAACACAAAGTCTACGCCTGCGAAGTCACCCAT
601 + + + + + + 660
GACTGCGACTCGTTTCGTCTGATGCTCTTTGTGTTTCAGATGCGGACGCTTCAGTGGGTA
L T L S K A D Y E K H K V Y A C E V T H
CAGGGCCTGAGCTCGCCCGTCACAAAGAGCTTCAACAGGGGAGAGTGTTAG 661 + + + + +- 711 23 GTCCCGGACTCGAGCGGGCAGTGTTTCTCGAAGTTGTCCCCTCTCACAATC
Q G L S S P V T K S F N R G E C * Table 11: Nucleotide and amino-acid sequence of the heavy chain
The amino-acid sequence coding for the variable domain is bold and underlined. The oligonucleotide primers used for cloning and sequencing are indicated.
MV416 ACCATGGAATTGGGGCTGAGCTGGGTTTTCCTTGTTGCTATTTTAGAAGGTGTCCACTGT
1 + + + + + + 60
TGGTACCTTAACCCCGACTCGACCCAAAAGGAACAACGATAAAATCTTCCACAGGTGACA
T M E L G L S W V F L V A I L E G V H C
MV418 GAGGTGCAGTTGGTGGAGTCTGGGGGAGGCTTGGTCCAGCCTGGGGGGTCCCTGAGACTC g]_ + + + + + + 120
CTCCACGTCAACCACCTCAGACCCCCTCCGAACCAGGTCGGACCCCCCAGGGACTCTGAG
E V O L V E S G G G L V Q P G G S L R L
TCCTGTGCAGCCTCTGGATTCACCTTTAGTAACTATTGGATGAACTGGGTCCGCCAGGCT
121 + + + + + + 180
AGGACACGTCGGAGACCTAAGTGGAAATCATTGATAACCTACTTGACCCAGGCGGTCCGA
S C A A S G F T F S N Y W M N W V R Q A
CCAGGGAAAGGGCTGGAGTGGGTGGCCGCCATAAACCAAGATGGAAGTGAGAAATACTAT 181 + + + + + + 240
GGTCCCTTTCCCGACCTCACCCACCGGCGGTATTTGGTTCTACCTTCACTCTTTATGATA P G K G L E W V A A I N Q D G S E K Y Y
GTGGGCTCTGTGAAGGGCCGATTCACCATCTCCAGAGACAACGCCAAGAACTCACTGTAT
241 + H + + + + 300
CACCCGAGACACTTCCCGGCTAAGTGGTAGAGGTCTCTGTTGCGGTTCTTGAGTGACATA
V G S V K G R F T I S R D N A K N S L Y
MV432 CTGCAAATGAACAGCCTGAGAGTCGAGGACACGGCTGTGTATTACTGTGTGAGGGACTAT
301 + + + + + + 360
GACGTTTACTTGTCGGACTCTCAGCTCCTGTGCCGACACATAATGACACACTCCCTGATA
L Q M N S L R V E D T A V Y Y C V R D Y
TACGΆTATTTTGACCGATTATTACATCCACTATTGGTACTTCGATCTCTGGGGCCGTGGC
361 + + + + + + 420
ATGCTATAAAACTGGCTAATAATGTAGGTGATAACCATGAAGCTAGAGACCCCGGCACCG
Y D I L T D Y Y I H Y W Y F D L W G R G MV433 ACCCTGGTCACTGTCTCCTCAGCCTCCACCAAGGGCCCATCGGTCTTCCCCCTGGCACCC
421 + + + + + + 480
MV434 TGGGACCAGTGACAGAGGAGTCGGAGGTGGTTCCCGGGTAGCCAGAAGGGGGACCGTGGG
T L V T V S S A S T K G P S V F P L A P
TCCTCCAAGAGCACCTCTGGGGGCACAGCGGCCCTGGGCTGCCTGGTCAAGGACTACTTC
481 + + + + + + 540
AGGAGGTTCTCGTGGAGACCCCCGTGTCGCCGGGACCCGACGGACCAGTTCCTGATGAAG
S S K S T S G G T A A L G C L V K D Y F
CCCGAACCGGTGACGGTGTCGTGGAACTCAGGCGCCCTGACCAGCGGCGTGCACACCTTC
541 + + + + + + 600
GGGCTTGGCCACTGCCACAGCACCTTGAGTCCGCGGGACTGGTCGCCGCACGTGTGGAAG
P E P V T V S W N S G A L T S G V H T F
CCGGCTGTCCTACAGTCCTCAGGACTCTACTCCCTCAGCAGCGTGGTGACCGTGCCCTCC
601 + + + + + + 660
GGCCGACAGGATGTCAGGAGTCCTGAGATGAGGGAGTCGTCGCACCACTGGCACGGGAGG P A V L Q S S G L Y S L S S V V T V P S
AGCAGCTTGGGCACCCAGACCTACATCTGCAACGTGAATCACAAGCCCAGCAACACCAAG
661 + + +r + + + 720
MV435 TCGTCGAACCCGTGGGTCTGGATGTAGACGTTGCACTTAGTGTTCGGGTCGTTGTGGTTC
S S L G T Q T Y I C N V N H K P S N T K
GTGGACAAGAGAGTTGAGCCCAAATCTTGTGACAAAACTCACACATGCCCACCGTGCCCA 721 + + + + + + 780
#265 CACCTGTTCTCTCAACTCGGGTTTAGAACACTGTTTTGAGTGTGTACGGGTGGCACGGGT
V D K R V E P K S C D K T H T C P P C P
TAA
781 783
ATT
* Example 4: Three-dimensional structure of the Fab fragment of the anti human IL-17A monoclonal antibody AIN457
Ia order to determine the conformation of trie Complementarily-Determining Regions (CDR' s) and the structure of the antigen-binding site of AIN457, the Fab fragment is generated, crystallized and its X-ray structure is determined by protein crystallography.
Method: The Fab fragment of NVP-AIN457 is produced by papain cleavage from the whole antibody and purified by protein A chromatography followed by size-exclusion chromatography. The purified material is then concentrated by ultrafiltration to 20mg/ml in 1OmM Tris-HCl pH 7.4, 25mM NaCl, 5mM TCEP. Crystals are grown by the technique of vapor diffusion in hanging drops at 19°C, from 2.0M ammonium sulfate, 5% PEG 400, 0.1M Na MES pH 6.5. They are in space group P212121 with unit cell dimensions a=90.3A, b=106.7A, c=131.4A and 2 Fab molecules per asymmetric unit. Prior to X-ray data collection, a single crystal of AIN457 Fab is cross-linked with glutaraldehyde using the method of Lusty (J. Appl. Cryst. (1999) 32, 106-112) and then transferred to a solution containing 2.0M Li2SO4, 2% PEG 400, and 0.1M Na MES pH 6.5. The crystal is subsequently mounted in a cryo-loop and flash-frozen for data collection at 95K. 180 diffraction images corresponding to l.Odeg oscillation each are recorded. The diffraction data are processed with the HKL program suite. The structure is determined to 2.5A resolution by molecular replacement. The structure is then refined by torsion angle dynamics and energy minimization using the program CNX.
Results: Two AIN457 Fab molecules are present in the asymmetric unit of the crystal, with the H-CDR3 loop of both Fab molecules involved in protein-protein contacts to the H-CDR3 loop of symmetry-related Fabs. The two Fab molecules show different elbow angles but have otherwise essentially identical CDR loop conformations (see Table 12 for amino acid sequence of the CDR loops). The H-CDRl loop adopts the expected Hl :1 canonical structure, while the conformation of the H-CDR2 loop matches that of canonical structure H2:3A. The H-CDR3 loop of the ATN457 antibody is exceptionally long, comprising 18 residues between Kabat positions 94 (Arg H98) and 101 (Asp Hl 15). It shows the typical bulged torso structure stabilized by a salt bridge between the Arg side-chain in position 94 (Arg H98) and the Asp carboxylate group in position HlOl (Asp Hl 15), and by an H-bonded interaction between the side-chain of Trp Hl 17 and the main-chain carbonyl group of Phe Hl 14. The head of the H-CDR3 loop has the structure of a long, twisted beta-hairpin with a second beta-bulge at its base and a type F beta-turn at its apex. A striking feature of the AIN457 H-CDR3 loop is its very high content in aromatic residues: 6 tyrosines, 2 tryptophanes, 1 phenylalanine. Since all other CDR loops contribute 1 more tyrosine each, the antigen-combining site of AIN457 possesses 11 tyrosines in total. The conformations of the L-CDRl and L-CDR2 loops correspond to canonical structures Ll :6 and L2:l, respectively. In contrast to H-CDR3, the L-CDR3 loop is short (6 residues) and shows the commonly observed canonical structure L3:l, with a cis-proline at its tip (Pro L96), a glutamine at Kabat position 90 (GIn L91) and a threonine at Kabat position 97 (Thr L98). However, a very unusual feature of the AIN457 L-CDR3 loop is the presence of a cysteine residue after the cis-proline (Cys L97). The side-chain of Cys L97 is at the bottom of a shallow depression located at the VL-VH interface and lined by residues Trp Hl 12, Trp H47 and Tyr L92.
Table 12:
Figure imgf000049_0001
Figure imgf000050_0001
Table 1 : Amino acid sequences of the hypervariable regions of the AIN457 monoclonal antibodies, based on the Kabat definition and as determined by the X-ray analysis, using the approach of Chothia and coworkers. Amino acid highlighted in bold are part of the CDR loops, while those shown in plain style are part of the antibody framework.

Claims

1. An IL- 17 binding molecule which is capable of inhibiting the activity of 1 nM human TL-
17 at a concentration of less than 5 nM by 50%, said inhibitory activity is measured on IL-6 production induced by hu-IL-17 in human dermal fibroblasts.
2. An IL- 17 binding molecule comprising both heavy (VH) and light chain (VL) variable domains; said IL-17 binding molecule comprises at least one antigen binding site comprising: a) an immunoglobulin heavy chain variable domain (VH) which comprises in sequence hypervariable regions CDRl, CDR2 and CDR3, said CDRl having the amino acid sequence SEQ ID NO: I, said CDR2 having the amino acid sequence SEQ ID NO:2, and said CDR3 having the amino acid sequence SEQ ID NO:3 or direct CDR equivalents thereof; and b) an immunoglobulin light chain variable domain (VL) which comprises in sequence hypervariable regions CDRl', CDR2' and CDR3', said CDRl' having the amino acid sequence SEQ ID NO:4, said CDR2' having the amino acid sequence SEQ ID NO:5, and said CDR3' having the amino acid sequence SEQ ID NO:6 or direct CDR' equivalents thereof.
3. An IL- 17 binding molecule comprising both heavy (VH) and light chain (VL) variable domains; said IL- 17 binding molecule comprises at least one antigen binding site comprising: a) an immunoglobulin heavy chain variable domain (VH) which comprises in sequence hypervariable regions CDRl -x, CDR2-X and CDR3-X, said CDRl -x having the amino acid sequence SEQ ID NO: 11, said CDR2-X having the amino acid sequence SEQ ID NO: 12, and said CDR3-X having the amino acid sequence SEQ ID NO: 13 or direct CDR-x equivalents thereof; and c) an immunoglobulin light chain variable domain (VL) which comprises in sequence hypervariable regions CDRl', CDR2' and CDR3', said CDRl' having the amino acid sequence SEQ 3D NO:4, said CDR2' having the amino acid sequence SEQ ID NO:5, and said CDR3' having the amino acid sequence SEQ ID N0:6 or direct CDR' equivalents thereof.
4. The IL- 17 binding molecule according to claim 1, 2 or 3 which is a human antibody.
5. An IL- 17 binding molecule which comprises at least one antigen binding site comprising either a first domain having an amino acid sequence substantially homologous to that shown in SEQ ID NO: 8 starting with amino acid at position 1 and ending with amino acid at position 127 or a first domain as described above and a second domain having an amino acid sequence substantially homologous to that shown in SEQ ID NO: 10, starting with amino acid at position 1 and ending with amino acid at position 109.
6. A DNA construct encoding an IL- 17 binding molecule according to any of claims 1 to 5.
7. A DNA construct comprising a DNA molecule which is substantially homologous to either SEQ E) NO: 7 or a direct DNAH equivalent thereof ;or
SEQ E) NO: 9 or a direct DNAL equivalent thereof.
8. A DNA construct comprising 2 DNA molecules of which one is substantially homologous to SEQ ID NO: 7 or a direct DNAH equivalent thereof and the other substantially homologous to SEQ E) NO: 9 or a direct DNAL equivalent thereof.
9. An expression vector able to replicate in a prokaryotic or eukaryotic cell line which comprises at least one DNA construct according to claims 6, 7 or 8.
10. A process for the product of an E-- 17 binding molecule which comprises (i) culturing an organism which is transformed with an expression vector according to claim 9 and (ii) recovering the E,- 17 binding molecule from the culture.
11. The use of an IL- 17 binding molecule according to claims 1 to 5, for the manufacture of a medicament.
12. The use of an IL- 17 binding molecule according to claims 1 to 5, for the manufacture of a medicament for the treatment of an IL-17 mediated disease or disorder.
13. The use of an IL-17 binding molecule according to claims 1 to 5, for the treatment of osteoarthritis, rheumatoid arthritis osteoporosis and other inflammatory arthritides.
14. A method for the treatment of an IL-17 mediated disease or disorders in a patient in need thereof, which comprises administering to the patient an effective amount of an IL-17 binding molecule according to claims 1 to 5.
15. A pharmaceutical composition comprising an antibody to IL-17 according to claims 1 to 5, in combination with a pharmaceutically acceptable excipient, diluent or carrier.
PCT/EP2005/008470 2004-08-05 2005-08-04 Il-17 antagonistic antibodies WO2006013107A1 (en)

Priority Applications (41)

Application Number Priority Date Filing Date Title
AU2005268857A AU2005268857C1 (en) 2004-08-05 2005-08-04 IL-17 antagonistic antibodies
EP18166164.6A EP3409288A1 (en) 2004-08-05 2005-08-04 Il-17 antagonistic antibodies
EP05770286.2A EP1776142B9 (en) 2004-08-05 2005-08-04 Il-17 antagonistic antibodies
BR122017009404-1A BR122017009404B1 (en) 2004-08-05 2005-08-04 Uses of an anti-il-17 antibody
SI200531390T SI1776142T1 (en) 2004-08-05 2005-08-04 Il-17 antagonistic antibodies
US11/658,344 US7807155B2 (en) 2004-08-05 2005-08-04 IL-17 antagonistic antibodies
PL05770286T PL1776142T4 (en) 2004-08-05 2005-08-04 Il-17 antagonistic antibodies
JP2007524286A JP4682200B2 (en) 2004-08-05 2005-08-04 IL-17 antagonistic antibody
NZ552658A NZ552658A (en) 2004-08-05 2005-08-04 IL-17 antagonistic antibodies
AT05770286T ATE517924T1 (en) 2004-08-05 2005-08-04 ANTAGONISTIC ANTIBODIES AGAINST IL-17
BR122018075556-3A BR122018075556B1 (en) 2004-08-05 2005-08-04 DNA CONSTRUCTION ENCODING AN ANTI-IL-17 ANTIBODY AND EXPRESSION VECTOR ABLE TO REPLICATE IN A EUKARYOTIC CELL LINE
CA2573586A CA2573586C (en) 2004-08-05 2005-08-04 Il-17 antagonistic antibodies
MX2007001338A MX2007001338A (en) 2004-08-05 2005-08-04 Il-17 antagonistic antibodies.
CN2005800265694A CN101001645B (en) 2004-08-05 2005-08-04 Il-17 antagonistic antibodies
ES05770286.2T ES2367440T7 (en) 2004-08-05 2005-08-04 IL-17 Antagonist Antibodies
BRPI0513078A BRPI0513078C1 (en) 2004-08-05 2005-08-04 igg antibody that binds to human IL-17 and liquid composition
DK05770286.2T DK1776142T6 (en) 2004-08-05 2005-08-04 Antagonistic antibodies against IL-17th
IL180717A IL180717A (en) 2004-08-05 2007-01-15 Il-17 binding antibodies or fragments thereof, dna constructs encoding them, expression vectors comprising said dna constructs, processes for their preparation and use thereof in the preparation of medicaments
TNP2007000034A TNSN07034A1 (en) 2004-08-05 2007-02-02 Il - 17 antagonistic antibodies
NO20070985A NO336279B1 (en) 2004-08-05 2007-02-20 IL-17 binding antibody, pharmaceutical composition and kit comprising such antibodies, as well as DNA construction encoding such IL-17 binding antibody and expression vector.
HK07109203.7A HK1101277A1 (en) 2004-08-05 2007-08-23 Il-17 antagonistic antibodies il-17
US12/707,934 US8119131B2 (en) 2004-08-05 2010-02-18 IL-17 antagonistic antibodies
AU2010201689A AU2010201689B2 (en) 2004-08-05 2010-04-28 IL-17 antagonistic antibodies
HRP20110758TT HRP20110758T4 (en) 2004-08-05 2011-10-19 Il-17 antagonistic antibodies
US13/349,689 US8617552B2 (en) 2004-08-05 2012-01-13 IL-17 antibodies
US14/085,074 US20140079719A1 (en) 2004-08-05 2013-11-20 IL-17 Antagonistic Antibodies
NO20150065A NO337129B1 (en) 2004-08-05 2015-01-13 IL-17 binding antibody for use in the treatment of psoriatic arthritis
NO20150064A NO337286B1 (en) 2004-08-05 2015-01-13 IL-17 binding antibody for use in the treatment of ankylosing spondylitis
US14/625,073 US9765140B2 (en) 2004-08-05 2015-02-18 IL-17 antagonistic antibodies
FR15C0048C FR15C0048I2 (en) 2004-08-05 2015-07-03 IL-17 ANTAGONIST ANTIBODIES
NL300749C NL300749I2 (en) 2004-08-05 2015-07-09 Secukinumab
CY2015030C CY2015030I2 (en) 2004-08-05 2015-07-10 COMPETITIVE IL-17 ANTIBODIES
LTPA2015029C LTC1776142I2 (en) 2004-08-05 2015-07-10 IL-17 antagonist antibodies
NO2015023C NO2015023I1 (en) 2004-08-05 2015-10-26 Sekukinumab
NO20151787A NO341384B1 (en) 2004-08-05 2015-12-23 IL-17 binding antibody for use in the treatment of inflammatory arthritis and use of the IL-17 binding antibody for the preparation of drug for the treatment of ankylosing spondylitis.
NO2016017C NO2016017I1 (en) 2004-08-05 2016-08-23 Sekukinumab
US15/674,970 US10344084B2 (en) 2004-08-05 2017-08-11 IL-17 antagonistic antibodies
NO20171697A NO20171697A1 (en) 2004-08-05 2017-10-24 IL-17 Antagonistic Antibodies
NO2018007C NO2018007I1 (en) 2004-08-05 2018-02-14 sekukinumab
US16/412,543 US20190270804A1 (en) 2004-08-05 2019-05-15 Il-17 antagonistic antibodies
US17/859,653 US20230235038A1 (en) 2004-08-05 2022-07-07 Interleukin-17 (il-17) antagonistic antibodies

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GBGB0417487.6A GB0417487D0 (en) 2004-08-05 2004-08-05 Organic compound
GB0417487.6 2004-08-05

Related Child Applications (3)

Application Number Title Priority Date Filing Date
US11/658,344 A-371-Of-International US7807155B2 (en) 2004-08-05 2005-08-04 IL-17 antagonistic antibodies
US12/707,937 Continuation US8397539B2 (en) 2010-02-18 2010-02-18 Non-contact dancer mechanisms, web isolation apparatuses and methods for using the same
US12/707,934 Continuation US8119131B2 (en) 2004-08-05 2010-02-18 IL-17 antagonistic antibodies

Publications (1)

Publication Number Publication Date
WO2006013107A1 true WO2006013107A1 (en) 2006-02-09

Family

ID=32982602

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2005/008470 WO2006013107A1 (en) 2004-08-05 2005-08-04 Il-17 antagonistic antibodies

Country Status (39)

Country Link
US (8) US7807155B2 (en)
EP (5) EP1776142B9 (en)
JP (1) JP4682200B2 (en)
KR (2) KR20080029018A (en)
CN (1) CN101001645B (en)
AR (1) AR050200A1 (en)
AT (1) ATE517924T1 (en)
AU (2) AU2005268857C1 (en)
BR (3) BR122017009404B1 (en)
CA (1) CA2573586C (en)
CY (6) CY1111963T1 (en)
DK (4) DK2366405T3 (en)
EC (1) ECSP077198A (en)
ES (4) ES2677245T3 (en)
FR (1) FR15C0048I2 (en)
GB (1) GB0417487D0 (en)
HK (3) HK1101277A1 (en)
HR (3) HRP20110758T4 (en)
HU (4) HUE038187T2 (en)
IL (1) IL180717A (en)
LT (3) LT2902039T (en)
LU (2) LU92768I2 (en)
MA (1) MA28982B1 (en)
MX (1) MX2007001338A (en)
MY (1) MY144925A (en)
NL (1) NL300749I2 (en)
NO (9) NO336279B1 (en)
NZ (1) NZ552658A (en)
PE (1) PE20060418A1 (en)
PL (4) PL2902039T3 (en)
PT (4) PT2364729E (en)
RU (2) RU2426741C3 (en)
SG (1) SG155186A1 (en)
SI (4) SI2364729T1 (en)
TN (1) TNSN07034A1 (en)
TR (1) TR201808057T4 (en)
TW (1) TWI359153B (en)
WO (1) WO2006013107A1 (en)
ZA (1) ZA200700242B (en)

Cited By (119)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007070750A1 (en) * 2005-12-13 2007-06-21 Eli Lilly And Company Anti-il-17 antibodies
EP1814915A1 (en) * 2004-11-19 2007-08-08 UCB Pharma, S.A. Neutralising antibody molecules having specificity for human il-17
WO2007117749A2 (en) * 2006-01-31 2007-10-18 Novartis Ag Il-17 antagonistic antibodies fpr treating cancer
WO2007147019A2 (en) 2006-06-13 2007-12-21 Zymogenetics, Inc. Il-17 and il-23 antagonists and methods of using the same
WO2007149032A1 (en) * 2006-06-23 2007-12-27 Astrazeneca Ab Antibody molecules for human il-17
WO2008001063A1 (en) * 2006-06-29 2008-01-03 Ucb Pharma S.A. Antibody molecules which bind human il-17
WO2008021156A3 (en) * 2006-08-11 2008-07-03 Schering Corp Antibodies to il-17a
WO2008106131A2 (en) 2007-02-28 2008-09-04 Schering Corporation Combination therapy for treatment of immune disorders
JP2009507023A (en) * 2005-09-01 2009-02-19 シェーリング コーポレイション Use of IL-23 and IL-17 antagonists to treat autoimmune ocular inflammatory diseases
WO2009082624A2 (en) * 2007-12-10 2009-07-02 Zymogenetics, Inc. Antagonists of il-17a, il-17f, and il-23 and methods of using the same
WO2009130459A2 (en) * 2008-04-23 2009-10-29 Ucb Pharma S.A. Epitopes of il-17a and il-17f and antibodies specific thereto
WO2010034443A1 (en) * 2008-09-29 2010-04-01 F. Hoffmann-La Roche Ag Antibodies against human il 17 and uses thereof
EP2182943A2 (en) * 2007-07-23 2010-05-12 Centocor Ortho Biotech Inc. Methods and compositions for treating fibrosis related disorders using il-17 antagonists
CN101932935A (en) * 2007-06-20 2010-12-29 先灵公司 Joint destruction biomarkers for anti-il-17a therapy of inflammatory joint disease
US7910703B2 (en) 2006-03-10 2011-03-22 Zymogenetics, Inc. Antagonists to IL-17A, IL-17F, and IL-23P19 and methods of use
US20110104236A1 (en) * 2008-01-09 2011-05-05 Reza Dana Therapeutic compositions for treatment of ocular inflammatory disorders
WO2011053763A2 (en) 2009-10-30 2011-05-05 Centocor Ortho Biotech Inc. Il-17a antagonists
US20110104172A1 (en) * 2008-04-29 2011-05-05 Micromet Ag Inhibitors of gm-csf and il-17 for therapy
WO2012045848A1 (en) 2010-10-08 2012-04-12 Novartis Ag Methods of treating psoriasis using il-17 antagonists
WO2012059598A2 (en) 2010-11-05 2012-05-10 Novartis Ag Methods of treating rheumatoid arthritis using il-17 antagonists
US8188249B2 (en) 2003-09-10 2012-05-29 Amgen Fremont Inc. Nucleic acid molecules encoding antibodies to M-CSF
WO2012082573A1 (en) 2010-12-13 2012-06-21 Novartis Ag Predictive methods and methods of treating arthritis using il-17 antagonists
EP2468302A1 (en) 2007-05-29 2012-06-27 Novartis AG New indications for anti-IL-1beta therapy
WO2012093254A1 (en) 2011-01-07 2012-07-12 Ucb Pharma S.A. Lipocalin 2 as a biomarker for il-17 inhibitor therapy efficacy
EP2485763A2 (en) * 2009-10-10 2012-08-15 The Board of Trustees of The Leland Stanford Junior University Il-17 family cytokine compositions and uses
WO2012125680A1 (en) 2011-03-16 2012-09-20 Novartis Ag Methods of treating vasculitis using an il-17 binding molecule
EP2514764A2 (en) 2006-10-18 2012-10-24 UCB Pharma, S.A. Antibody molecules which bind IL-17A and IL-17F
WO2013077907A1 (en) 2011-11-21 2013-05-30 Novartis Ag Methods of treating psoriatic arthritis (psa) using il-17 antagonists and psa response or non- response alleles
WO2013082282A1 (en) 2011-12-02 2013-06-06 lNOVARTIS AG Anti-il-1beta (interleukin-1beta) antibody-based prophylactic therapy to prevent complications leading to vaso-occlusion in sickle cell disease.
US8470992B2 (en) 2005-12-20 2013-06-25 Sbi Biotech Co., Ltd. Anti-ILT7 antibody
US20130202591A1 (en) * 2010-08-05 2013-08-08 Anaptysbio, Inc. Antibodies directed against il-17
WO2013150043A1 (en) 2012-04-05 2013-10-10 F. Hoffmann-La Roche Ag Bispecific antibodies against human tweak and human il17 and uses thereof
WO2013158821A2 (en) 2012-04-20 2013-10-24 Novartis Ag Methods of treating ankylosing spondylitis using il-17 antagonists
US8580265B2 (en) 2011-01-14 2013-11-12 Ucb Pharma S.A. Antibody molecules which bind IL-17A and IL-17F
US8586035B2 (en) 2007-02-12 2013-11-19 Merck Sharp & Dohme Corp. Use of IL-23 antagonists for treatment of infection
US8586714B2 (en) 2009-09-01 2013-11-19 Abbvie, Inc. Dual variable domain immunoglobulins and uses thereof
US8716450B2 (en) 2009-10-15 2014-05-06 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
US8722855B2 (en) 2009-10-28 2014-05-13 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
US8735546B2 (en) 2010-08-03 2014-05-27 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
US8822645B2 (en) 2008-07-08 2014-09-02 Abbvie Inc. Prostaglandin E2 dual variable domain immunoglobulins and uses thereof
WO2014155278A2 (en) 2013-03-26 2014-10-02 Novartis Ag Methods of treating autoimmune diseases using il-17 antagonists
WO2014161570A1 (en) 2013-04-03 2014-10-09 Roche Glycart Ag Antibodies against human il17 and uses thereof
WO2015022656A1 (en) 2013-08-15 2015-02-19 Novartis Ag Methods of treating generalized pustular psoriasis (gpp) using il-17 antagonists
US8980864B2 (en) 2013-03-15 2015-03-17 Moderna Therapeutics, Inc. Compositions and methods of altering cholesterol levels
US8987418B2 (en) 2013-03-15 2015-03-24 Abbvie Inc. Dual specific binding proteins directed against IL-1β and/or IL-17
US8999380B2 (en) 2012-04-02 2015-04-07 Moderna Therapeutics, Inc. Modified polynucleotides for the production of biologics and proteins associated with human disease
US9029508B2 (en) 2008-04-29 2015-05-12 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
US9035027B2 (en) 2008-06-03 2015-05-19 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
US9045551B2 (en) 2012-11-01 2015-06-02 Abbvie Inc. Anti-DLL4/VEGF dual variable domain immunoglobulin and uses thereof
US9046513B2 (en) 2010-08-26 2015-06-02 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
US9109026B2 (en) 2008-06-03 2015-08-18 Abbvie, Inc. Dual variable domain immunoglobulins and uses thereof
US9107886B2 (en) 2012-04-02 2015-08-18 Moderna Therapeutics, Inc. Modified polynucleotides encoding basic helix-loop-helix family member E41
US9120870B2 (en) 2011-12-30 2015-09-01 Abbvie Inc. Dual specific binding proteins directed against IL-13 and IL-17
US9181319B2 (en) 2010-08-06 2015-11-10 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US9186372B2 (en) 2011-12-16 2015-11-17 Moderna Therapeutics, Inc. Split dose administration
US9193788B2 (en) 2013-02-08 2015-11-24 Novartis Ag Anti-IL-17A antibodies and their uses
AU2014259523B2 (en) * 2010-11-05 2016-02-04 Novartis Ag Methods of treating psoriatic arthritis using IL-17 antagonists
US9283287B2 (en) 2012-04-02 2016-03-15 Moderna Therapeutics, Inc. Modified polynucleotides for the production of nuclear proteins
US9284283B2 (en) 2012-02-02 2016-03-15 Ensemble Therapeutics Corporation Macrocyclic compounds for modulating IL-17
WO2016038538A1 (en) 2014-09-10 2016-03-17 Novartis Ag Use of il-17 antagonists to inhibit the progression of structural damage in psoriatic arthritis patients
WO2016057841A1 (en) 2014-10-08 2016-04-14 Novartis Ag Compositions and methods of use for augmented immune response and cancer therapy
WO2016061142A1 (en) 2014-10-14 2016-04-21 Novartis Ag Antibody molecules to pd-l1 and uses thereof
US9334328B2 (en) 2010-10-01 2016-05-10 Moderna Therapeutics, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
WO2016103146A1 (en) 2014-12-22 2016-06-30 Novartis Ag Selective reduction of cysteine residues in il-17 antibodies
WO2016103153A1 (en) 2014-12-22 2016-06-30 Novartis Ag Pharmaceutical products and stable liquid compositions of il-17 antibodies
US9428535B2 (en) 2011-10-03 2016-08-30 Moderna Therapeutics, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US9464124B2 (en) 2011-09-12 2016-10-11 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US9475873B2 (en) 2009-05-05 2016-10-25 Novimmune Sa Nucleic acids encoding anti-IL-17F antibodies and methods of use thereof
US9481735B2 (en) 2009-03-05 2016-11-01 Abbvie Inc. IL-17 binding proteins
US9533047B2 (en) 2011-03-31 2017-01-03 Modernatx, Inc. Delivery and formulation of engineered nucleic acids
JP2017502924A (en) * 2013-11-18 2017-01-26 シャンハイ ヘンルイ ファーマスーティカル カンパニー リミテッドShanghai Hengrui Pharmaceutical Co., Ltd. IL-17A binding agent and use thereof
WO2017019894A1 (en) 2015-07-29 2017-02-02 Novartis Ag Combination therapies comprising antibody molecules to lag-3
WO2017019897A1 (en) 2015-07-29 2017-02-02 Novartis Ag Combination therapies comprising antibody molecules to tim-3
US9572897B2 (en) 2012-04-02 2017-02-21 Modernatx, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
US9597380B2 (en) 2012-11-26 2017-03-21 Modernatx, Inc. Terminally modified RNA
WO2017068472A1 (en) 2015-10-19 2017-04-27 Novartis Ag Methods of treating non-radiographic axial spondyloarthritis using interleukin-17 (il-17) antagonists
US9650437B2 (en) 2008-05-05 2017-05-16 Novimmune S.A. Nucleic acid encoding and method of producing anti-IL-17A/IL-17F cross-reactive antibodies
WO2017106656A1 (en) 2015-12-17 2017-06-22 Novartis Ag Antibody molecules to pd-1 and uses thereof
US9833410B2 (en) 2012-10-31 2017-12-05 Takeda Gmbh Lyophilized formulation comprising GM-CSF neutralizing compound
US9840554B2 (en) 2015-06-15 2017-12-12 Abbvie Inc. Antibodies against platelet-derived growth factor (PDGF)
WO2017221174A1 (en) 2016-06-22 2017-12-28 Novartis Ag Methods of treating vitiligo using interleukin-17 (il-17) antibodies
WO2018015880A1 (en) 2016-07-19 2018-01-25 Novartis Ag Methods of treating new-onset plaque type psoriasis using il-17 antagonists
US9919051B2 (en) 2012-10-31 2018-03-20 Amgen Research (Munich) Gmbh Liquid formulation comprising GM-CSF neutralizing compound
US9932403B2 (en) 2010-05-20 2018-04-03 Ablynx Nv Biological materials related to HER3
WO2018096467A1 (en) 2016-11-28 2018-05-31 Novartis Ag Methods of treating acne using interleukin-17 (il-17) antagonists
US10017568B2 (en) 2011-05-05 2018-07-10 Merck Patent Gmbh Polypeptides that bind to IL-17A, IL-17F and/or IL17-A/F and method of treatment using same
WO2018148585A1 (en) 2017-02-10 2018-08-16 Genentech, Inc. Anti-tryptase antibodies, compositions thereof, and uses thereof
WO2018158741A1 (en) 2017-03-03 2018-09-07 Novartis Ag Psoriasis disease modification following long-term treatment with an il-17 antagonist
US10093733B2 (en) 2014-12-11 2018-10-09 Abbvie Inc. LRP-8 binding dual variable domain immunoglobulin proteins
WO2018203289A1 (en) 2017-05-05 2018-11-08 Novartis Ag Methods of selectively treating asthma using il-17 antagonists
US10208113B2 (en) 2014-06-23 2019-02-19 Janssen Biotech, Inc. Interferon α and ω antibody antagonists
US10208349B2 (en) 2011-01-07 2019-02-19 Ucb Biopharma Sprl Lipocalin 2 as a biomarker for IL-17 inhibitor therapy efficacy
WO2019087133A1 (en) 2017-11-02 2019-05-09 Novartis Ag Method of treating tendinopathy using interleukin-17 (il-17)
WO2019097493A1 (en) 2017-11-20 2019-05-23 Novartis Ag Treating hidradenitis suppurativa with il-17 antagonists
WO2019108900A1 (en) 2017-11-30 2019-06-06 Novartis Ag Bcma-targeting chimeric antigen receptor, and uses thereof
US10323076B2 (en) 2013-10-03 2019-06-18 Modernatx, Inc. Polynucleotides encoding low density lipoprotein receptor
US10442869B2 (en) 2014-09-26 2019-10-15 Closed Joint Stock Company “Biocad” Anti-IL-17A Antibodies
WO2019229701A2 (en) 2018-06-01 2019-12-05 Novartis Ag Binding molecules against bcma and uses thereof
EP3689907A1 (en) 2019-01-31 2020-08-05 Numab Therapeutics AG Antibodies targeting il-17a and methods of use thereof
WO2020157305A1 (en) 2019-01-31 2020-08-06 Numab Therapeutics AG Multispecific antibodies having specificity for tnfa and il-17a, antibodies targeting il-17a, and methods of use thereof
US10745475B2 (en) 2013-08-30 2020-08-18 Takeda Gmbh Antibodies neutralizing GM-CSF for use in the treatment of rheumatoid arthritis or as analgesics
US10815291B2 (en) 2013-09-30 2020-10-27 Modernatx, Inc. Polynucleotides encoding immune modulating polypeptides
EP3738976A1 (en) 2015-01-12 2020-11-18 Affibody AB Il-17a-binding polypeptides
WO2021053591A1 (en) 2019-09-20 2021-03-25 Novartis Ag Methods of treating autoimmune diseases using interleukin-17 (il-17) antagonists
WO2021067195A1 (en) 2019-09-30 2021-04-08 Janssen Biotech, Inc. Compositions and methods for an il-17 target engagement assay with large molecule modulators
WO2021099924A1 (en) 2019-11-19 2021-05-27 Novartis Ag Methods of treating lupus nephritis using interleukin-17 (il-17) antagonists
WO2021111377A2 (en) 2019-12-06 2021-06-10 Novartis Ag Methods of treating lichen planus using interleukin-17 (il-17) antagonists
WO2021262876A1 (en) 2020-06-23 2021-12-30 Novartis Ag Methods of treating thyroid eye disease and graves' orbitopahy using interleukin-17 (il-17) antagonists
WO2022091056A1 (en) 2020-11-02 2022-05-05 Novartis Ag Interleukin-17 inhibitors
WO2022097060A1 (en) 2020-11-06 2022-05-12 Novartis Ag Cd19 binding molecules and uses thereof
WO2022117773A1 (en) 2020-12-02 2022-06-09 Fresenius Kabi Deutschland Gmbh Methods of selectively reducing antibodies
WO2022137132A1 (en) 2020-12-22 2022-06-30 Novartis Ag Methods for reducing the oxidation level of cysteine residues in a secreted recombinantly-expressed protein during cell culture
US11401326B2 (en) * 2016-05-24 2022-08-02 Assistance Publique—Hopitaus De Paris Method of treatment of human papillomaviruses related chronic infections
US11453717B2 (en) 2016-04-29 2022-09-27 Joint Stock Company “Biocad” Trispecific antibodies against IL-17A, IL-17F and other pro-inflammatory molecule
US11673950B2 (en) 2016-03-10 2023-06-13 Viela Bio, Inc. ILT7 binding molecules and methods of using the same
EP3976032A4 (en) * 2019-05-31 2023-06-14 Figene, LLC Concurrent activation of regenerative and tolerogenic processes by fibroblast-based compositions for the treatment of multiple sclerosis
WO2023209519A1 (en) 2022-04-25 2023-11-02 Novartis Ag Crystalline forms of an il-17 inhibitor
WO2023223263A1 (en) 2022-05-18 2023-11-23 Novartis Ag Methods of selectively treating tendinopathy using interleukin-17 (il-17) antagonists
WO2023223211A1 (en) 2022-05-16 2023-11-23 Novartis Ag Methods of treating giant cell arteritis using interleukin-17 (il-17) antagonists

Families Citing this family (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB0417487D0 (en) * 2004-08-05 2004-09-08 Novartis Ag Organic compound
AR060017A1 (en) 2006-01-13 2008-05-21 Novartis Ag COMPOSITIONS AND METHODS OF USE FOR DICKKOPF -1 ANTIBODIES
WO2009033091A1 (en) * 2007-09-06 2009-03-12 City Of Hope Treatment of th17-mediated autoimmune disease via inhibition of stat3
WO2009072802A2 (en) * 2007-12-03 2009-06-11 Amorepacific Corporation Composition for slimming
EA027071B1 (en) * 2009-04-27 2017-06-30 Новартис Аг ANTI-ActRIIB ANTIBODY AND COMPOSITION COMPRISING SAME
JP5947041B2 (en) * 2011-01-07 2016-07-06 第一三共ヘルスケア株式会社 Safe IL-17 production inhibitor
WO2012095507A1 (en) * 2011-01-13 2012-07-19 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and kits for predicting the risk of having a cardiovascular event in a subject
WO2012135415A1 (en) * 2011-03-29 2012-10-04 Glaxosmithkline Llc Buffer system for protein purification
US9580501B2 (en) 2011-12-16 2017-02-28 Synthon Biopharmaceuticals B.V. Anti-TNF alpha monoclonal secretory IgA antibodies and methods for treating inflammatory diseases
EP2852615B1 (en) 2012-05-22 2018-12-05 Bristol-Myers Squibb Company Il-17a/f il-23 bispecific antibodies and their uses
WO2014107737A2 (en) 2013-01-07 2014-07-10 Eleven Biotherapeutics, Inc. Local delivery of il-17 inhibitors for treating ocular disease
US9707154B2 (en) 2013-04-24 2017-07-18 Corning Incorporated Delamination resistant pharmaceutical glass containers containing active pharmaceutical ingredients
US9603882B2 (en) 2013-08-13 2017-03-28 Industrial Technology Research Institute Method for modulating Th17 cells and method for treating a disease related to modulation of Th17 cells
JP2017508446A (en) * 2014-01-28 2017-03-30 北京韓美薬品有限公司 Bifunctional fusion protein and production method and use thereof
WO2016123329A2 (en) 2015-01-28 2016-08-04 Genentech, Inc. Gene expression markers and treatment of multiple sclerosis
CN105315371B (en) 2015-03-05 2018-05-29 北京百特美博生物科技有限公司 Anti-human IL-17 monoclonal antibodies
MX2018000694A (en) 2015-07-16 2018-05-07 Lilly Co Eli Treatment of pruritus.
AU2017230091B2 (en) 2016-03-10 2022-04-07 Acceleron Pharma Inc. Activin type 2 receptor binding proteins and uses thereof
CN107488227A (en) * 2016-06-12 2017-12-19 三生国健药业(上海)股份有限公司 Anti-human IL-17 A monoclonal antibodies, its preparation method and application
CN109715660A (en) 2016-09-14 2019-05-03 北京韩美药品有限公司 A kind of antibody and its function fragment that can specifically combine IL-17A
CN108359011B (en) 2017-07-21 2019-06-25 华博生物医药技术(上海)有限公司 Target the antibody, preparation method and application of interleukin-17 A
JP2021519589A (en) * 2018-03-29 2021-08-12 アールイーエムディー バイオセラピューティクス,インコーポレイテッドREMD Biotherapeutics,Inc Treatment of autoimmune and inflammatory disorders with antibodies that bind to interleukin 17A (IL-17A)
WO2020024931A1 (en) * 2018-07-31 2020-02-06 Shen Weiqun Anti-il-17a antibodies and use thereof
WO2020064702A1 (en) 2018-09-25 2020-04-02 INSERM (Institut National de la Santé et de la Recherche Médicale) Use of antagonists of th17 cytokines for the treatment of bronchial remodeling in patients suffering from allergic asthma
PE20212185A1 (en) 2019-02-18 2021-11-11 Lilly Co Eli FORMULATION OF THERAPEUTIC ANTIBODIES
CN110179746A (en) 2019-05-17 2019-08-30 通化东宝生物科技有限公司 A kind of stable Su Jin monoclonal antibody injection and preparation method thereof
CN110343666B (en) * 2019-07-10 2023-05-30 通化东宝药业股份有限公司 Feed supplement culture medium for CHO cell culture and preparation method and application thereof
WO2021018035A1 (en) 2019-07-26 2021-02-04 神州细胞工程有限公司 Humanized anti-il17a antibody and use thereof
WO2021018191A1 (en) 2019-07-30 2021-02-04 江苏恒瑞医药股份有限公司 Method for treating autoimmune disease by il-17 antagonist
CN111040035B (en) * 2019-12-31 2022-05-24 南京融捷康生物科技有限公司 Antibody aiming at IL-17RA protein and preparation method and application thereof

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1988001649A1 (en) 1986-09-02 1988-03-10 Genex Corporation Single polypeptide chain binding molecules
WO1990007861A1 (en) 1988-12-28 1990-07-26 Protein Design Labs, Inc. CHIMERIC IMMUNOGLOBULINS SPECIFIC FOR p55 TAC PROTEIN OF THE IL-2 RECEPTOR
WO1995018826A2 (en) 1994-01-05 1995-07-13 Schering Corporation Purified primate ctla-8 antigens and related reagents
WO1997004097A2 (en) * 1995-07-19 1997-02-06 Genetics Institute, Inc. Human ctla-8 and uses of ctla-8-related proteins
JP2000186046A (en) * 1998-10-14 2000-07-04 Snow Brand Milk Prod Co Ltd Therapeutic agent for and diagnosis of chronic rheumatism and
WO2000069463A1 (en) * 1999-05-17 2000-11-23 Schering Corporation Compositions and methods for treating cell proliferation disorders
WO2002058717A2 (en) * 2000-10-18 2002-08-01 Immunex Corporation Methods for treating rheumatoid arthritis using il-17 antagonists

Family Cites Families (49)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB8601597D0 (en) 1986-01-23 1986-02-26 Wilson R H Nucleotide sequences
GB8717430D0 (en) 1987-07-23 1987-08-26 Celltech Ltd Recombinant dna product
GB8823869D0 (en) 1988-10-12 1988-11-16 Medical Res Council Production of antibodies
DK0710719T3 (en) 1990-01-12 2007-07-09 Amgen Fremont Inc Generation of xenogenic antibodies
US5625126A (en) 1990-08-29 1997-04-29 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
EP0814159B1 (en) 1990-08-29 2005-07-27 GenPharm International, Inc. Transgenic mice capable of producing heterologous antibodies
US5661016A (en) 1990-08-29 1997-08-26 Genpharm International Inc. Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US5633425A (en) 1990-08-29 1997-05-27 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5545806A (en) 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
US5770429A (en) 1990-08-29 1998-06-23 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US6274711B1 (en) * 1993-06-14 2001-08-14 Inserm, Institut National De La Sante Et De La Recherche Medicale Purified mammalian CTLA-8 antigens and related reagents
ATE279517T1 (en) 1995-03-23 2004-10-15 Immunex Corp IL-17 RECEPTOR
US6074849A (en) * 1995-07-19 2000-06-13 Genetics Institute, Inc. Polynucleotides encoding human CTLA-8 related proteins
ES2181915T3 (en) 1995-10-27 2003-03-01 Schering Corp CTLA-8 IN COMBINATION WITH G-CSF AND IL-6, AND USE OF CTLA-8 TO TREAT INFECTIONS.
ATE429241T1 (en) 1996-11-27 2009-05-15 Immunex Corp METHOD FOR REGULATING NITROGEN MONOXIDE GENERATION
US6849719B2 (en) 1997-09-17 2005-02-01 Human Genome Sciences, Inc. Antibody to an IL-17 receptor like protein
US6482923B1 (en) 1997-09-17 2002-11-19 Human Genome Sciences, Inc. Interleukin 17-like receptor protein
US6635443B1 (en) 1997-09-17 2003-10-21 Human Genome Sciences, Inc. Polynucleotides encoding a novel interleukin receptor termed interleukin-17 receptor-like protein
CA2309604C (en) 1997-11-10 2010-01-12 Cytimmune Sciences, Inc. Compositions and methods for targeted delivery of factors
US6562578B1 (en) * 1999-01-11 2003-05-13 Schering Corporation IL-17-like cytokine binding compounds and antibodies
AU2004799A (en) 1997-12-19 1999-07-12 Millennium Biotherapeutics, Inc. Novel embryo-derived interleukin related factor molecules and uses therefor
WO1999035263A2 (en) 1998-01-09 1999-07-15 Immunex Corporation Il-17rh dna and polypeptides
WO1999035267A1 (en) 1998-01-09 1999-07-15 Immunex Corporation Human and murine il-17d, cytokine related to interleukin-17: dna and polypeptides
AU2674199A (en) 1998-02-11 1999-08-30 Maxygen, Inc. Optimization of immunomodulatory properties of genetic vaccines
CA2328496C (en) 1998-05-15 2016-01-05 Genentech, Inc. Il-17 homologous polypeptides and therapeutic uses thereof
ES2300154T3 (en) 1998-09-17 2008-06-01 Zymogenetics, Inc. BETA-9 TRANSFORMATION GROWTH FACTOR IN MAMMALS (ZTGFSS9).
AU6277799A (en) 1998-10-02 2000-04-26 Eli Lilly And Company Il-17 homolog nucleic acids, polypeptides, vectors, host cells, methods and usesthereof
AU1071200A (en) 1998-10-19 2000-05-08 Biotech Australia Pty Limited Systems for oral delivery
DE29820466U1 (en) 1998-11-16 1999-04-08 Reutter Werner Prof Dr Med Recombinant glycoproteins and medicinal products containing them
US6734172B2 (en) 1998-11-18 2004-05-11 Pacific Northwest Research Institute Surface receptor antigen vaccines
ES2374429T3 (en) 1999-01-11 2012-02-16 Schering Corporation MAMMAL CYTOKINES RELATED TO INTERLEUQUINE 17. POLINUCLEOTIDES CODING THEM. APPLICATIONS.
WO2000042187A1 (en) 1999-01-11 2000-07-20 Schering Corporation Interleukin-17 related mammalian cytokine (il-171). polynucleotides encoding them. uses
EP1031346B1 (en) 1999-01-27 2002-05-02 Idea Ag Noninvasive vaccination through the skin
PL200919B1 (en) 1999-02-12 2009-02-27 Lexigen Pharm Corp Methods for treatment of tumors and metastases using a combination of anti-angiogenic and immuno therapies
US6036128A (en) 1999-05-17 2000-03-14 Common Ground Recycling, Inc. Tire shredding machine and method of using the same
WO2001015728A1 (en) 1999-08-27 2001-03-08 University Health Network Method for activating cytotoxic t-lymphocytes (ctls) in vivo : composition comprising antibody anti cd40 (or cd40l or cd40 binding protein) and an antigen
JP2001086046A (en) * 1999-09-09 2001-03-30 Nec Corp Portable telephone system with camera
US7220840B2 (en) * 2000-06-16 2007-05-22 Human Genome Sciences, Inc. Antibodies that immunospecifically bind to B lymphocyte stimulator protein
PT2281843T (en) 2000-06-16 2017-01-02 Human Genome Sciences Inc Antibodies that immunospecifically bind to blys
AU2001296229A1 (en) 2000-10-13 2002-04-29 Eli Lilly And Company Methods of using a human il-17-related polypeptide to treat disease
ATE501730T1 (en) 2001-01-25 2011-04-15 Zymogenetics Inc METHOD FOR TREATING PSORIASIS USING AN IL-17D ANTAGONIST
CN101508734A (en) * 2001-04-27 2009-08-19 协和发酵麒麟株式会社 Anti-CD40 monoclonal antibody
CA2467521A1 (en) 2001-11-16 2003-07-10 Human Genome Sciences, Inc. Antibodies that immunospecifically bind to blys
WO2004019866A2 (en) 2002-08-28 2004-03-11 Immunex Corporation Compositions and methods for treating cardiovascular disease
US7456264B2 (en) * 2003-05-21 2008-11-25 Medarex, Inc. Human monoclonal antibodies against Bacillus anthracis protective antigen
JP4833850B2 (en) 2003-11-21 2011-12-07 ユセベ ファルマ ソシエテ アノニム Method for treating multiple sclerosis by inhibiting IL-17 activity
US7501247B2 (en) 2004-05-03 2009-03-10 Schering Corporation Method of treating skin inflammation
GB0417487D0 (en) * 2004-08-05 2004-09-08 Novartis Ag Organic compound
WO2007027761A2 (en) 2005-09-01 2007-03-08 Schering Corporation Use of il-23 and il-17 antagonists to treat autoimmune ocular inflammatory disease

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1988001649A1 (en) 1986-09-02 1988-03-10 Genex Corporation Single polypeptide chain binding molecules
WO1990007861A1 (en) 1988-12-28 1990-07-26 Protein Design Labs, Inc. CHIMERIC IMMUNOGLOBULINS SPECIFIC FOR p55 TAC PROTEIN OF THE IL-2 RECEPTOR
WO1995018826A2 (en) 1994-01-05 1995-07-13 Schering Corporation Purified primate ctla-8 antigens and related reagents
WO1997004097A2 (en) * 1995-07-19 1997-02-06 Genetics Institute, Inc. Human ctla-8 and uses of ctla-8-related proteins
JP2000186046A (en) * 1998-10-14 2000-07-04 Snow Brand Milk Prod Co Ltd Therapeutic agent for and diagnosis of chronic rheumatism and
WO2000069463A1 (en) * 1999-05-17 2000-11-23 Schering Corporation Compositions and methods for treating cell proliferation disorders
WO2002058717A2 (en) * 2000-10-18 2002-08-01 Immunex Corporation Methods for treating rheumatoid arthritis using il-17 antagonists

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
CHABAUD M ET AL: "Contribution of Interleukin 17 to Synovium Matrix Destruction in Rheumatoid Arthritis", CYTOKINE, ACADEMIC PRESS LTD, PHILADELPHIA, PA, US, vol. 12, no. 7, July 2000 (2000-07-01), pages 1092 - 1099, XP002218621, ISSN: 1043-4666 *
DATABASE WPI Section Ch Week 200051, Derwent World Patents Index; Class B04, AN 2000-551579, XP002352043 *
DUMONT F J: "IL-17 cytokine/receptor families: Emerging targets for the modulation of inflammatory responses", EXPERT OPINION ON THERAPEUTIC PATENTS 01 MAR 2003 UNITED KINGDOM, vol. 13, no. 3, 1 March 2003 (2003-03-01), pages 287 - 303, XP002351568, ISSN: 1354-3776 *
LUBBERTS ERIK: "The role of IL-17 and family members in the pathogenesis of arthritis.", CURRENT OPINION IN INVESTIGATIONAL DRUGS (LONDON, ENGLAND : 2000) MAY 2003, vol. 4, no. 5, May 2003 (2003-05-01), pages 572 - 577, XP009056201, ISSN: 1472-4472 *
R&D SYSTEMS: "Monoclonal Anti-human IL-17 Antibody", R&D SYSTEMS CATALOGUE, 11 January 2004 (2004-01-11), XP002351567, Retrieved from the Internet <URL:http://cytokine.rndsystems.com/pdf/MAB317.pdf> [retrieved on 20051027] *

Cited By (293)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9718883B2 (en) 2003-09-10 2017-08-01 Amgen Fremont Inc. Antibodies to M-CSF
US8188249B2 (en) 2003-09-10 2012-05-29 Amgen Fremont Inc. Nucleic acid molecules encoding antibodies to M-CSF
US10280219B2 (en) 2003-09-10 2019-05-07 Amgen Fremont Inc. Antibodies to M-CSF
EP1814915A1 (en) * 2004-11-19 2007-08-08 UCB Pharma, S.A. Neutralising antibody molecules having specificity for human il-17
US8231875B2 (en) 2004-11-19 2012-07-31 Ucb Pharma S.A. Neutralising antibody molecules having specificity for human IL-17
US8329431B2 (en) 2004-11-19 2012-12-11 Ucb Pharma S.A. Nucleic acid molecules encoding neutralising antibodies having specificity for human IL-17
EP1814915B1 (en) * 2004-11-19 2014-01-08 UCB Pharma S.A. Neutralising antibodies having specificity for human il-17
US8865167B2 (en) 2004-11-19 2014-10-21 Ucb Pharma, S.A. Neutralising antibody molecules having specificity for human IL-17
JP2011184466A (en) * 2005-09-01 2011-09-22 Schering Corp Use of il-23 and il-17 antagonist to treat autoimmune ocular inflammatory disease
AU2006284841B2 (en) * 2005-09-01 2012-11-08 Merck Sharp & Dohme Corp. Use of IL-23 and IL-17 antagonists to treat autoimmune ocular inflammatory disease
JP2009507023A (en) * 2005-09-01 2009-02-19 シェーリング コーポレイション Use of IL-23 and IL-17 antagonists to treat autoimmune ocular inflammatory diseases
EP2322219A3 (en) * 2005-09-01 2013-06-05 Merck Sharp & Dohme Corp. Use of IL-23 and IL-17 antagonists to treat autoimmune ocular inflammatory disease
US20130323251A1 (en) * 2005-09-01 2013-12-05 The Government of the United States of America as represented by the Secretary of the Dep. of H.H.S. Use of il-23 and il-17 antagonists to treat autoimmune ocular inflammatory disease
US8524230B2 (en) 2005-09-01 2013-09-03 Merck Sharp & Dohme Corp. Use of IL-23 and IL-17 antagonists to treat autoimmune ocular inflammatory disease
EP2116258A1 (en) * 2005-09-01 2009-11-11 Schering Corporation Use of IL-23 and IL-17 antagonists to treat autoimmune ocular inflammatory disease
JP2014005285A (en) * 2005-09-01 2014-01-16 Merck Sharp & Dohme Corp Use of il-23 and il-17 antagonists to treat autoimmune ocular inflammatory disease
EP3366702A1 (en) * 2005-12-13 2018-08-29 Eli Lilly And Co. Anti-il-17 antibodies
WO2007070750A1 (en) * 2005-12-13 2007-06-21 Eli Lilly And Company Anti-il-17 antibodies
EP3808769A1 (en) * 2005-12-13 2021-04-21 Eli Lilly And Company Anti-il-17 antibodies
EP2481753A1 (en) * 2005-12-13 2012-08-01 Eli Lilly and Company Anti-IL-17 Antibodies
US7838638B2 (en) 2005-12-13 2010-11-23 Eli Lilly And Company Anti-IL-17 antibodies
EA014298B1 (en) * 2005-12-13 2010-10-29 Эли Лилли Энд Компани Anti-il-17 antibodies
US8110191B2 (en) 2005-12-13 2012-02-07 Eli Lilly And Company Anti-IL-17 antibodies
US8470992B2 (en) 2005-12-20 2013-06-25 Sbi Biotech Co., Ltd. Anti-ILT7 antibody
AU2007235199B2 (en) * 2006-01-31 2010-09-23 Novartis Ag IL-17 antagonistic antibodies for treating cancer
AU2007235199B8 (en) * 2006-01-31 2010-10-28 Novartis Ag IL-17 antagonistic antibodies for treating cancer
WO2007117749A3 (en) * 2006-01-31 2008-06-12 Novartis Ag Il-17 antagonistic antibodies fpr treating cancer
WO2007117749A2 (en) * 2006-01-31 2007-10-18 Novartis Ag Il-17 antagonistic antibodies fpr treating cancer
US8012477B2 (en) 2006-01-31 2011-09-06 Novartis Ag Methods of treating multiple myeloma using IL-17 binding molecules
US9464134B2 (en) 2006-03-10 2016-10-11 Zymogenetics, Inc. Polynucleotides encoding antagonists of IL-17A, IL-17F, and IL-23p19
US9994634B2 (en) 2006-03-10 2018-06-12 Zymogenetics, Inc. Use of an IL-17/IL-23 bispecific antibody for treating inflammation
US8333968B2 (en) 2006-03-10 2012-12-18 Zymogenetics, Inc. Methods of inhibiting inflammation with antagonists to IL-17A, IL-17F, and IL-23P19
US7910703B2 (en) 2006-03-10 2011-03-22 Zymogenetics, Inc. Antagonists to IL-17A, IL-17F, and IL-23P19 and methods of use
US8496936B2 (en) 2006-03-10 2013-07-30 Zymogenetics, Inc. Antagonists of IL-17A, IL-17F, and IL-23P19
US10562967B2 (en) 2006-03-10 2020-02-18 Zymogenetics, Inc. Treating inflammation with IL-17/IL-23 bispecific antibodies
US8992922B2 (en) 2006-03-10 2015-03-31 Zymogenetics, Inc. Antagonists of IL-17A, IL-17F, and IL-23P19
US8227579B2 (en) 2006-06-13 2012-07-24 Zymogenetics, Inc. IL-23 antagonists
US7790862B2 (en) 2006-06-13 2010-09-07 Zymogenetics, Inc. IL-17 and IL-23 antagonists and methods of using the same
WO2007147019A2 (en) 2006-06-13 2007-12-21 Zymogenetics, Inc. Il-17 and il-23 antagonists and methods of using the same
WO2007147019A3 (en) * 2006-06-13 2008-08-07 Zymogenetics Inc Il-17 and il-23 antagonists and methods of using the same
WO2007149032A1 (en) * 2006-06-23 2007-12-27 Astrazeneca Ab Antibody molecules for human il-17
JP2009540824A (en) * 2006-06-23 2009-11-26 アストラゼネカ アクチボラグ Antibody molecules against human IL-17
US8865166B2 (en) 2006-06-23 2014-10-21 Medimmune Limited Antibodies to IL-17A and uses thereof
US8057794B2 (en) 2006-06-29 2011-11-15 Ucb Pharma S.A. Antibody molecules which bind to human IL-17
WO2008001063A1 (en) * 2006-06-29 2008-01-03 Ucb Pharma S.A. Antibody molecules which bind human il-17
JP2013252147A (en) * 2006-06-29 2013-12-19 Ucb Pharma Sa Antibody molecule binding to human il-17
EA020113B1 (en) * 2006-06-29 2014-08-29 Юсб Фарма С.А. Neutralising antibodies which bind human il-17 and use thereof
JP2009540855A (en) * 2006-06-29 2009-11-26 ユセベ ファルマ ソシエテ アノニム Antibody molecules that bind to human IL-17
US8753843B2 (en) 2006-08-11 2014-06-17 Merck Sharp & Dohme Corp. Antibodies to IL-17A
AU2007284782B2 (en) * 2006-08-11 2012-11-15 Merck Sharp & Dohme Corp. Antibodies to IL-17A
CN101646690B (en) * 2006-08-11 2013-10-23 默沙东公司 Antibodies to il-17a
US7846443B2 (en) 2006-08-11 2010-12-07 Schering Corporation Antibodies to IL-17A
JP2013009680A (en) * 2006-08-11 2013-01-17 Merck Sharp & Dohme Corp Antibody to il-17a
KR101218484B1 (en) * 2006-08-11 2013-01-04 머크 샤프 앤드 돔 코포레이션 Antibodies to il-17a
WO2008021156A3 (en) * 2006-08-11 2008-07-03 Schering Corp Antibodies to il-17a
JP2010500028A (en) * 2006-08-11 2010-01-07 シェーリング コーポレイション Antibody to IL-17A
US8193319B2 (en) 2006-08-11 2012-06-05 Schering Corporation Antibodies to IL-17A
US8303953B2 (en) 2006-10-18 2012-11-06 Ucb Pharma S.A. Antibody molecules which bind IL-17A and IL-17F
US10308723B2 (en) 2006-10-18 2019-06-04 Ucb Biopharma Sprl Method of treating inflammation by administering antibody molecules which bind IL-17A and IL-17F
EP2514764A2 (en) 2006-10-18 2012-10-24 UCB Pharma, S.A. Antibody molecules which bind IL-17A and IL-17F
EP3524623A1 (en) 2006-10-18 2019-08-14 UCB Biopharma SPRL Antibody molecules which bind il-17a and il-17f
US9890219B2 (en) 2006-10-18 2018-02-13 Ucb Biopharma Sprl Methods of treating an IL-17A mediated pathological condition
US8586035B2 (en) 2007-02-12 2013-11-19 Merck Sharp & Dohme Corp. Use of IL-23 antagonists for treatment of infection
EP2417974A1 (en) 2007-02-28 2012-02-15 Schering Corporation Combination therapy for treatment of immune disorders
WO2008106131A2 (en) 2007-02-28 2008-09-04 Schering Corporation Combination therapy for treatment of immune disorders
EP4177268A2 (en) 2007-05-29 2023-05-10 Novartis AG New indications for anti-il-1beta therapy
EP2468302A1 (en) 2007-05-29 2012-06-27 Novartis AG New indications for anti-IL-1beta therapy
EP2468301A1 (en) 2007-05-29 2012-06-27 Novartis AG New indications for anti-IL-1-beta therapy
CN101932935A (en) * 2007-06-20 2010-12-29 先灵公司 Joint destruction biomarkers for anti-il-17a therapy of inflammatory joint disease
EP2182943A2 (en) * 2007-07-23 2010-05-12 Centocor Ortho Biotech Inc. Methods and compositions for treating fibrosis related disorders using il-17 antagonists
EP2182943A4 (en) * 2007-07-23 2010-10-06 Centocor Ortho Biotech Inc Methods and compositions for treating fibrosis related disorders using il-17 antagonists
JP2010534664A (en) * 2007-07-23 2010-11-11 セントコア・オーソ・バイオテツク・インコーポレーテツド Methods and compositions for treatment of fibrosis related diseases using IL-17 antagonists
WO2009082624A3 (en) * 2007-12-10 2009-10-29 Zymogenetics, Inc. Antagonists of il-17a, il-17f, and il-23 and methods of using the same
WO2009082624A2 (en) * 2007-12-10 2009-07-02 Zymogenetics, Inc. Antagonists of il-17a, il-17f, and il-23 and methods of using the same
US9309313B2 (en) * 2008-01-09 2016-04-12 The Schepens Eye Research Institute, Inc. Therapeutic compositions for treatment of ocular inflammatory disorders
US9872901B2 (en) 2008-01-09 2018-01-23 The Schepens Eye Research Institute, Inc. Therapeutic compositions for treatment of ocular inflammatory disorders
US11241497B2 (en) 2008-01-09 2022-02-08 The Schepens Eye Research Institute, Inc. Therapeutic compositions for treatment of ocular inflammatory disorders
AU2009204441B2 (en) * 2008-01-09 2015-03-05 The Schepens Eye Research Institute, Inc. Therapeutic compositions for treatment of ocular inflammatory disorders
US20110104236A1 (en) * 2008-01-09 2011-05-05 Reza Dana Therapeutic compositions for treatment of ocular inflammatory disorders
EP2975048A3 (en) * 2008-04-23 2016-05-11 UCB Biopharma SPRL Epitopes of il-17a and il-17f and antibodies specific thereto
WO2009130459A3 (en) * 2008-04-23 2010-11-18 Ucb Pharma S.A. Epitopes of il-17a and il-17f and antibodies specific thereto
US9045537B2 (en) 2008-04-23 2015-06-02 Ucb Pharma Sa Antibodies specific to IL-17A and IL-17F
US8679494B2 (en) 2008-04-23 2014-03-25 Ucb Pharma S.A. Antibodies specific to IL-17A and IL-17F
WO2009130459A2 (en) * 2008-04-23 2009-10-29 Ucb Pharma S.A. Epitopes of il-17a and il-17f and antibodies specific thereto
US9771420B2 (en) 2008-04-23 2017-09-26 Ucb Biopharma Sprl Methods of treating an IL-17A mediated pathological condition
EP2975048A2 (en) 2008-04-23 2016-01-20 UCB Biopharma SPRL Epitopes of il-17a and il-17f and antibodies specific thereto
US20110104172A1 (en) * 2008-04-29 2011-05-05 Micromet Ag Inhibitors of gm-csf and il-17 for therapy
AU2009242088B2 (en) * 2008-04-29 2014-08-21 Amgen Research (Munich) Gmbh Inhibitors of GM-CSF and IL-17 for therapy
US9029508B2 (en) 2008-04-29 2015-05-12 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
US9353180B2 (en) 2008-04-29 2016-05-31 Amgen Research (Munich) Gmbh Method of treatment by the administration of inhibitors of GM-CSF and IL-17
US9650437B2 (en) 2008-05-05 2017-05-16 Novimmune S.A. Nucleic acid encoding and method of producing anti-IL-17A/IL-17F cross-reactive antibodies
US9109026B2 (en) 2008-06-03 2015-08-18 Abbvie, Inc. Dual variable domain immunoglobulins and uses thereof
US9035027B2 (en) 2008-06-03 2015-05-19 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
US8822645B2 (en) 2008-07-08 2014-09-02 Abbvie Inc. Prostaglandin E2 dual variable domain immunoglobulins and uses thereof
TWI393570B (en) * 2008-09-29 2013-04-21 Roche Glycart Ag Antibodies against human il17 and uses thereof
US8003099B2 (en) 2008-09-29 2011-08-23 Roche Palo Alto Llc Antibodies against human IL17 and uses thereof
RU2539029C2 (en) * 2008-09-29 2015-01-10 Роше Гликарт Аг Human interleukin 17 (il-17) antibodies and using them
WO2010034443A1 (en) * 2008-09-29 2010-04-01 F. Hoffmann-La Roche Ag Antibodies against human il 17 and uses thereof
KR101318549B1 (en) 2008-09-29 2013-10-16 로슈 글리카트 아게 Antibodies against human il17 and uses thereof
US9481736B2 (en) 2009-03-05 2016-11-01 Abbvie, Inc. IL-17 binding proteins
US9663587B2 (en) 2009-03-05 2017-05-30 Abbvie Inc. IL-17 binding proteins
US9481735B2 (en) 2009-03-05 2016-11-01 Abbvie Inc. IL-17 binding proteins
US9475873B2 (en) 2009-05-05 2016-10-25 Novimmune Sa Nucleic acids encoding anti-IL-17F antibodies and methods of use thereof
US8586714B2 (en) 2009-09-01 2013-11-19 Abbvie, Inc. Dual variable domain immunoglobulins and uses thereof
EP2485763A4 (en) * 2009-10-10 2013-10-30 Univ Leland Stanford Junior Il-17 family cytokine compositions and uses
EP2485763A2 (en) * 2009-10-10 2012-08-15 The Board of Trustees of The Leland Stanford Junior University Il-17 family cytokine compositions and uses
US8716450B2 (en) 2009-10-15 2014-05-06 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
US8722855B2 (en) 2009-10-28 2014-05-13 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
US8519107B2 (en) 2009-10-30 2013-08-27 Janssen Biotech, Inc. IL-17A antibodies
WO2011053763A2 (en) 2009-10-30 2011-05-05 Centocor Ortho Biotech Inc. Il-17a antagonists
US10808032B2 (en) 2010-05-20 2020-10-20 Ablynx Nv Biological materials related to HER3
US9932403B2 (en) 2010-05-20 2018-04-03 Ablynx Nv Biological materials related to HER3
US8735546B2 (en) 2010-08-03 2014-05-27 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
US9493560B2 (en) 2010-08-03 2016-11-15 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
US9328166B2 (en) 2010-08-05 2016-05-03 Anaptysbio, Inc. Antibodies directed against IL-17
US20130202591A1 (en) * 2010-08-05 2013-08-08 Anaptysbio, Inc. Antibodies directed against il-17
US9447164B2 (en) 2010-08-06 2016-09-20 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US9937233B2 (en) 2010-08-06 2018-04-10 Modernatx, Inc. Engineered nucleic acids and methods of use thereof
US9181319B2 (en) 2010-08-06 2015-11-10 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US9046513B2 (en) 2010-08-26 2015-06-02 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
US9334328B2 (en) 2010-10-01 2016-05-10 Moderna Therapeutics, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US10064959B2 (en) 2010-10-01 2018-09-04 Modernatx, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US9701965B2 (en) 2010-10-01 2017-07-11 Modernatx, Inc. Engineered nucleic acids and methods of use thereof
US9657295B2 (en) 2010-10-01 2017-05-23 Modernatx, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US9717791B2 (en) 2010-10-08 2017-08-01 Novartis Ag Methods of treating psoriasis using IL-17 antibody
WO2012045848A1 (en) 2010-10-08 2012-04-12 Novartis Ag Methods of treating psoriasis using il-17 antagonists
CN107029234A (en) * 2010-10-08 2017-08-11 诺华有限公司 Utilize the psoriasic method of the antagonist for treating of IL 17
EP3299390A1 (en) 2010-10-08 2018-03-28 Novartis AG Methods of treating psoriasis using il-17 antagonists
US10583190B2 (en) 2010-10-08 2020-03-10 Novartis Ag Methods of treating psoriasis using IL-17 antibodies
KR101620771B1 (en) * 2010-10-08 2016-05-12 노파르티스 아게 Methods of treating psoriasis using il-17 antagonists
US11534490B2 (en) 2010-10-08 2022-12-27 Novartis Ag Methods of treating psoriasis using IL-17 antagonists
AU2011311482B2 (en) * 2010-10-08 2014-05-29 Novartis Ag Methods of treating psoriasis using IL-17 antagonists
EP3792281A1 (en) * 2010-10-08 2021-03-17 Novartis AG Methods of treating psoriasis using il-17 antagonists
CN103154031A (en) * 2010-10-08 2013-06-12 诺华有限公司 Methods of treating psoriasis using il-17 antagonists
RU2591083C2 (en) * 2010-10-08 2016-07-10 Новартис Аг Methods of treating psoriasis using il-17 antagonists
EP4137514A1 (en) 2010-10-08 2023-02-22 Novartis AG Methods of treating psoriasis using il-17 antagonists
US11534491B2 (en) 2010-11-05 2022-12-27 Novartis Ag Methods of treating ankylosing spondylitis using IL-17 antagonists
RU2582937C2 (en) * 2010-11-05 2016-04-27 Новартис Аг Methods of treating rheumatoid arthritis using il-17 antagonists
EP3912640A1 (en) 2010-11-05 2021-11-24 Novartis AG Pharmaceutical compositions comprising secukinumab
TWI618543B (en) * 2010-11-05 2018-03-21 諾華公司 Uses of il-17 antibodies in the manufacture of medicaments for treating ankylosing spondylitis
US9744234B2 (en) 2010-11-05 2017-08-29 Novartis Ag Methods of treating ankylosing spondylitis using IL-17 antagonists
TWI616204B (en) * 2010-11-05 2018-03-01 諾華公司 Uses of il-17 antibodies in the manufacture of medicaments for treating psoristic arthritis
TWI604851B (en) * 2010-11-05 2017-11-11 諾華公司 Use of il-17 antibodies in the manufacture of medicaments for treating ankylosing spondylitis
AU2014259526B2 (en) * 2010-11-05 2016-02-04 Novartis Ag Methods of treating ankylosing spondylitis using IL-17 antagonists
AU2014259523B2 (en) * 2010-11-05 2016-02-04 Novartis Ag Methods of treating psoriatic arthritis using IL-17 antagonists
US10363307B2 (en) 2010-11-05 2019-07-30 Novartis Ag Methods of treating psoriatic arthritis using IL-17 antagonists
EP3111954B1 (en) 2010-11-05 2019-04-03 Novartis Ag Methods of treating ankylosing spondylitis using anti-il-17 antibodies
EP3111954A1 (en) * 2010-11-05 2017-01-04 Novartis Ag Methods of treating ankylosing spondylitis or psoriatic arthritis using anti-il-17 antibodies
EP4116325A1 (en) 2010-11-05 2023-01-11 Novartis AG Methods of treating rheumatoid arthritis using il-17 antagonists
WO2012059598A2 (en) 2010-11-05 2012-05-10 Novartis Ag Methods of treating rheumatoid arthritis using il-17 antagonists
WO2012059598A3 (en) * 2010-11-05 2012-09-27 Novartis Ag Methods of treating rheumatoid arthritis using il-17 antagonists
EP3757126A1 (en) * 2010-11-05 2020-12-30 Novartis AG Methods of treating psoriatic arthritis using il-17 antagonists
EP3542820A1 (en) 2010-11-05 2019-09-25 Novartis AG Methods of treating psoriatic arthritis using il-17 antagonists
WO2012082573A1 (en) 2010-12-13 2012-06-21 Novartis Ag Predictive methods and methods of treating arthritis using il-17 antagonists
US11466324B2 (en) 2011-01-07 2022-10-11 UCB Biopharma SRL Lipocalin 2 as a biomarker for IL-17 inhibitor therapy efficacy
EP3534159A1 (en) 2011-01-07 2019-09-04 UCB Biopharma SPRL Lipocalin 2 as a biomarker for il-17 inhibitor therapy efficacy
US10208349B2 (en) 2011-01-07 2019-02-19 Ucb Biopharma Sprl Lipocalin 2 as a biomarker for IL-17 inhibitor therapy efficacy
WO2012093254A1 (en) 2011-01-07 2012-07-12 Ucb Pharma S.A. Lipocalin 2 as a biomarker for il-17 inhibitor therapy efficacy
US9988446B2 (en) 2011-01-14 2018-06-05 Ucb Biopharma Sprl Methods of treatment using antibodies which bind IL-17A and IL-17F
US11919950B2 (en) 2011-01-14 2024-03-05 UCB Biopharma SRL Expression vector encoding antibody molecule which binds IL-17A and IL-17F
US9034600B2 (en) 2011-01-14 2015-05-19 Ucb Biopharma Sprl DNA encoding antibody molecules which bind IL-17A and IL-17F
US8580265B2 (en) 2011-01-14 2013-11-12 Ucb Pharma S.A. Antibody molecules which bind IL-17A and IL-17F
WO2012125680A1 (en) 2011-03-16 2012-09-20 Novartis Ag Methods of treating vasculitis using an il-17 binding molecule
US9533047B2 (en) 2011-03-31 2017-01-03 Modernatx, Inc. Delivery and formulation of engineered nucleic acids
US9950068B2 (en) 2011-03-31 2018-04-24 Modernatx, Inc. Delivery and formulation of engineered nucleic acids
US10017568B2 (en) 2011-05-05 2018-07-10 Merck Patent Gmbh Polypeptides that bind to IL-17A, IL-17F and/or IL17-A/F and method of treatment using same
US11773159B2 (en) 2011-05-05 2023-10-03 Merck Patent Gmbh Polypeptides that bind to IL-17A, IL-17F and/or IL17-A/F and methods of treatment using same
US10829552B2 (en) 2011-05-05 2020-11-10 Merck Patent Gmbh Polypeptides that bind to IL-17A, IL-17F and/or IL17-A/F and methods of treatment using same
US9464124B2 (en) 2011-09-12 2016-10-11 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US10022425B2 (en) 2011-09-12 2018-07-17 Modernatx, Inc. Engineered nucleic acids and methods of use thereof
US10751386B2 (en) 2011-09-12 2020-08-25 Modernatx, Inc. Engineered nucleic acids and methods of use thereof
US9428535B2 (en) 2011-10-03 2016-08-30 Moderna Therapeutics, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
WO2013077907A1 (en) 2011-11-21 2013-05-30 Novartis Ag Methods of treating psoriatic arthritis (psa) using il-17 antagonists and psa response or non- response alleles
WO2013082282A1 (en) 2011-12-02 2013-06-06 lNOVARTIS AG Anti-il-1beta (interleukin-1beta) antibody-based prophylactic therapy to prevent complications leading to vaso-occlusion in sickle cell disease.
US9271996B2 (en) 2011-12-16 2016-03-01 Moderna Therapeutics, Inc. Formulation and delivery of PLGA microspheres
US9295689B2 (en) 2011-12-16 2016-03-29 Moderna Therapeutics, Inc. Formulation and delivery of PLGA microspheres
US9186372B2 (en) 2011-12-16 2015-11-17 Moderna Therapeutics, Inc. Split dose administration
US9120870B2 (en) 2011-12-30 2015-09-01 Abbvie Inc. Dual specific binding proteins directed against IL-13 and IL-17
US9284283B2 (en) 2012-02-02 2016-03-15 Ensemble Therapeutics Corporation Macrocyclic compounds for modulating IL-17
US9107886B2 (en) 2012-04-02 2015-08-18 Moderna Therapeutics, Inc. Modified polynucleotides encoding basic helix-loop-helix family member E41
US9878056B2 (en) 2012-04-02 2018-01-30 Modernatx, Inc. Modified polynucleotides for the production of cosmetic proteins and peptides
US9192651B2 (en) 2012-04-02 2015-11-24 Moderna Therapeutics, Inc. Modified polynucleotides for the production of secreted proteins
US9216205B2 (en) 2012-04-02 2015-12-22 Moderna Therapeutics, Inc. Modified polynucleotides encoding granulysin
US9675668B2 (en) 2012-04-02 2017-06-13 Moderna Therapeutics, Inc. Modified polynucleotides encoding hepatitis A virus cellular receptor 2
US10501512B2 (en) 2012-04-02 2019-12-10 Modernatx, Inc. Modified polynucleotides
US9220792B2 (en) 2012-04-02 2015-12-29 Moderna Therapeutics, Inc. Modified polynucleotides encoding aquaporin-5
US9782462B2 (en) 2012-04-02 2017-10-10 Modernatx, Inc. Modified polynucleotides for the production of proteins associated with human disease
US9221891B2 (en) 2012-04-02 2015-12-29 Moderna Therapeutics, Inc. In vivo production of proteins
US9814760B2 (en) 2012-04-02 2017-11-14 Modernatx, Inc. Modified polynucleotides for the production of biologics and proteins associated with human disease
US9827332B2 (en) 2012-04-02 2017-11-28 Modernatx, Inc. Modified polynucleotides for the production of proteins
US9828416B2 (en) 2012-04-02 2017-11-28 Modernatx, Inc. Modified polynucleotides for the production of secreted proteins
US9303079B2 (en) 2012-04-02 2016-04-05 Moderna Therapeutics, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
US9220755B2 (en) 2012-04-02 2015-12-29 Moderna Therapeutics, Inc. Modified polynucleotides for the production of proteins associated with blood and lymphatic disorders
US9233141B2 (en) 2012-04-02 2016-01-12 Moderna Therapeutics, Inc. Modified polynucleotides for the production of proteins associated with blood and lymphatic disorders
US9149506B2 (en) 2012-04-02 2015-10-06 Moderna Therapeutics, Inc. Modified polynucleotides encoding septin-4
US9587003B2 (en) 2012-04-02 2017-03-07 Modernatx, Inc. Modified polynucleotides for the production of oncology-related proteins and peptides
US9301993B2 (en) 2012-04-02 2016-04-05 Moderna Therapeutics, Inc. Modified polynucleotides encoding apoptosis inducing factor 1
US9572897B2 (en) 2012-04-02 2017-02-21 Modernatx, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
US8999380B2 (en) 2012-04-02 2015-04-07 Moderna Therapeutics, Inc. Modified polynucleotides for the production of biologics and proteins associated with human disease
US9114113B2 (en) 2012-04-02 2015-08-25 Moderna Therapeutics, Inc. Modified polynucleotides encoding citeD4
US9254311B2 (en) 2012-04-02 2016-02-09 Moderna Therapeutics, Inc. Modified polynucleotides for the production of proteins
US9050297B2 (en) 2012-04-02 2015-06-09 Moderna Therapeutics, Inc. Modified polynucleotides encoding aryl hydrocarbon receptor nuclear translocator
US9061059B2 (en) 2012-04-02 2015-06-23 Moderna Therapeutics, Inc. Modified polynucleotides for treating protein deficiency
US9255129B2 (en) 2012-04-02 2016-02-09 Moderna Therapeutics, Inc. Modified polynucleotides encoding SIAH E3 ubiquitin protein ligase 1
US9095552B2 (en) 2012-04-02 2015-08-04 Moderna Therapeutics, Inc. Modified polynucleotides encoding copper metabolism (MURR1) domain containing 1
US9089604B2 (en) 2012-04-02 2015-07-28 Moderna Therapeutics, Inc. Modified polynucleotides for treating galactosylceramidase protein deficiency
US9283287B2 (en) 2012-04-02 2016-03-15 Moderna Therapeutics, Inc. Modified polynucleotides for the production of nuclear proteins
US9714292B2 (en) 2012-04-05 2017-07-25 Hoffmann-La Roche Inc. Bispecific antibodies against human TWEAK and human IL17 and uses thereof
WO2013150043A1 (en) 2012-04-05 2013-10-10 F. Hoffmann-La Roche Ag Bispecific antibodies against human tweak and human il17 and uses thereof
WO2013158821A2 (en) 2012-04-20 2013-10-24 Novartis Ag Methods of treating ankylosing spondylitis using il-17 antagonists
US10758621B2 (en) 2012-10-31 2020-09-01 Amgen Research (Munich) Gmbh Liquid formulation comprising GM-CSF neutralizing compound
US9919051B2 (en) 2012-10-31 2018-03-20 Amgen Research (Munich) Gmbh Liquid formulation comprising GM-CSF neutralizing compound
US9833410B2 (en) 2012-10-31 2017-12-05 Takeda Gmbh Lyophilized formulation comprising GM-CSF neutralizing compound
US9944720B2 (en) 2012-11-01 2018-04-17 Abbvie Inc. Anti-DLL4/VEGF dual variable domain immunoglobulin and uses thereof
US9045551B2 (en) 2012-11-01 2015-06-02 Abbvie Inc. Anti-DLL4/VEGF dual variable domain immunoglobulin and uses thereof
US9163093B2 (en) 2012-11-01 2015-10-20 Abbvie Inc. Anti-DLL4/VEGF dual variable domain immunoglobulin and uses thereof
US9597380B2 (en) 2012-11-26 2017-03-21 Modernatx, Inc. Terminally modified RNA
US9938342B2 (en) 2013-02-08 2018-04-10 Novartis Ag Nucleic acid encoding anti-IL-17A antibodies
US10683349B2 (en) 2013-02-08 2020-06-16 Novartis Ag Anti-IL-17A antibodies and their use in treating autoimmune and inflammatory disorders
US9193788B2 (en) 2013-02-08 2015-11-24 Novartis Ag Anti-IL-17A antibodies and their uses
US9650439B2 (en) 2013-02-08 2017-05-16 Novartis Ag Anti-IL-17A antibodies and their use in treating autoimmune and inflammatory
US8980864B2 (en) 2013-03-15 2015-03-17 Moderna Therapeutics, Inc. Compositions and methods of altering cholesterol levels
US8987418B2 (en) 2013-03-15 2015-03-24 Abbvie Inc. Dual specific binding proteins directed against IL-1β and/or IL-17
US9062108B2 (en) 2013-03-15 2015-06-23 Abbvie Inc. Dual specific binding proteins directed against IL-1 and/or IL-17
WO2014155278A2 (en) 2013-03-26 2014-10-02 Novartis Ag Methods of treating autoimmune diseases using il-17 antagonists
WO2014161570A1 (en) 2013-04-03 2014-10-09 Roche Glycart Ag Antibodies against human il17 and uses thereof
EP4223312A1 (en) 2013-08-15 2023-08-09 Novartis AG Methods of treating plaque psoriasis using il-17 antagonists
US11351253B2 (en) * 2013-08-15 2022-06-07 Novartis Ag Methods of treating palmoplantar pustular psoriasis (PPP) using IL-17 antibody
WO2015022656A1 (en) 2013-08-15 2015-02-19 Novartis Ag Methods of treating generalized pustular psoriasis (gpp) using il-17 antagonists
US11795216B2 (en) 2013-08-30 2023-10-24 Takeda Pharmaceutical Company Limited Antibodies neutralizing GM-CSF for use in the treatment of rheumatoid arthritis or as analgesics
US10745475B2 (en) 2013-08-30 2020-08-18 Takeda Gmbh Antibodies neutralizing GM-CSF for use in the treatment of rheumatoid arthritis or as analgesics
US10815291B2 (en) 2013-09-30 2020-10-27 Modernatx, Inc. Polynucleotides encoding immune modulating polypeptides
US10323076B2 (en) 2013-10-03 2019-06-18 Modernatx, Inc. Polynucleotides encoding low density lipoprotein receptor
EP3072905A4 (en) * 2013-11-18 2017-06-28 Shanghai Hengrui Pharmaceutical Co. Ltd. Il-17a conjugate and uses thereof
JP2017502924A (en) * 2013-11-18 2017-01-26 シャンハイ ヘンルイ ファーマスーティカル カンパニー リミテッドShanghai Hengrui Pharmaceutical Co., Ltd. IL-17A binding agent and use thereof
EP3670533A1 (en) * 2013-11-18 2020-06-24 Shanghai Hengrui Pharmaceutical Co. Ltd. Il-17a binding agent and uses thereof
US10759854B2 (en) 2014-06-23 2020-09-01 Janssen Biotech, Inc. Interferon alpha and omega antibody antagonists
US10358491B2 (en) 2014-06-23 2019-07-23 Janssen Biotech, Inc. Interferon alpha and omega antibody antagonists
US10208113B2 (en) 2014-06-23 2019-02-19 Janssen Biotech, Inc. Interferon α and ω antibody antagonists
CN107073110A (en) * 2014-09-10 2017-08-18 诺华股份有限公司 The structural damage for suppressing psoriatic arthritis patient using the antagonists of IL 17 is in progress
US11278618B2 (en) 2014-09-10 2022-03-22 Novartis Ag Use of IL-17 antagonists to inhibit the progression of structural damage in psoriatic arthritis patients
WO2016038538A1 (en) 2014-09-10 2016-03-17 Novartis Ag Use of il-17 antagonists to inhibit the progression of structural damage in psoriatic arthritis patients
US10442869B2 (en) 2014-09-26 2019-10-15 Closed Joint Stock Company “Biocad” Anti-IL-17A Antibodies
WO2016057841A1 (en) 2014-10-08 2016-04-14 Novartis Ag Compositions and methods of use for augmented immune response and cancer therapy
EP4245376A2 (en) 2014-10-14 2023-09-20 Novartis AG Antibody molecules to pd-l1 and uses thereof
WO2016061142A1 (en) 2014-10-14 2016-04-21 Novartis Ag Antibody molecules to pd-l1 and uses thereof
US10093733B2 (en) 2014-12-11 2018-10-09 Abbvie Inc. LRP-8 binding dual variable domain immunoglobulin proteins
RU2765307C2 (en) * 2014-12-22 2022-01-28 Новартис Аг Selective reduction of cysteine residues in antibodies against il-17
US11292833B2 (en) 2014-12-22 2022-04-05 Novartis Ag Selective reduction of cysteine residues in IL-17 antibodies
EP4372005A2 (en) 2014-12-22 2024-05-22 Novartis AG Pharmaceutical products and stable liquid compositions of il-17 antibodies
RU2719563C2 (en) * 2014-12-22 2020-04-21 Новартис Аг Selective reduction of cysteine residues in antibodies against il-17
WO2016103146A1 (en) 2014-12-22 2016-06-30 Novartis Ag Selective reduction of cysteine residues in il-17 antibodies
WO2016103153A1 (en) 2014-12-22 2016-06-30 Novartis Ag Pharmaceutical products and stable liquid compositions of il-17 antibodies
CN114010780A (en) * 2014-12-22 2022-02-08 诺华股份有限公司 Selective reduction of cysteine residues in IL-17 antibodies
AU2015370515B2 (en) * 2014-12-22 2018-11-22 Novartis Ag Selective reduction of cysteine residues in IL-17 antibodies
EP3738976A1 (en) 2015-01-12 2020-11-18 Affibody AB Il-17a-binding polypeptides
US9840554B2 (en) 2015-06-15 2017-12-12 Abbvie Inc. Antibodies against platelet-derived growth factor (PDGF)
WO2017019894A1 (en) 2015-07-29 2017-02-02 Novartis Ag Combination therapies comprising antibody molecules to lag-3
EP3878465A1 (en) 2015-07-29 2021-09-15 Novartis AG Combination therapies comprising antibody molecules to tim-3
EP3964528A1 (en) 2015-07-29 2022-03-09 Novartis AG Combination therapies comprising antibody molecules to lag-3
WO2017019897A1 (en) 2015-07-29 2017-02-02 Novartis Ag Combination therapies comprising antibody molecules to tim-3
WO2017068472A1 (en) 2015-10-19 2017-04-27 Novartis Ag Methods of treating non-radiographic axial spondyloarthritis using interleukin-17 (il-17) antagonists
WO2017106656A1 (en) 2015-12-17 2017-06-22 Novartis Ag Antibody molecules to pd-1 and uses thereof
US11673950B2 (en) 2016-03-10 2023-06-13 Viela Bio, Inc. ILT7 binding molecules and methods of using the same
US11453717B2 (en) 2016-04-29 2022-09-27 Joint Stock Company “Biocad” Trispecific antibodies against IL-17A, IL-17F and other pro-inflammatory molecule
US11401326B2 (en) * 2016-05-24 2022-08-02 Assistance Publique—Hopitaus De Paris Method of treatment of human papillomaviruses related chronic infections
WO2017221174A1 (en) 2016-06-22 2017-12-28 Novartis Ag Methods of treating vitiligo using interleukin-17 (il-17) antibodies
WO2018015880A1 (en) 2016-07-19 2018-01-25 Novartis Ag Methods of treating new-onset plaque type psoriasis using il-17 antagonists
WO2018096467A1 (en) 2016-11-28 2018-05-31 Novartis Ag Methods of treating acne using interleukin-17 (il-17) antagonists
WO2018148585A1 (en) 2017-02-10 2018-08-16 Genentech, Inc. Anti-tryptase antibodies, compositions thereof, and uses thereof
WO2018158741A1 (en) 2017-03-03 2018-09-07 Novartis Ag Psoriasis disease modification following long-term treatment with an il-17 antagonist
WO2018203289A1 (en) 2017-05-05 2018-11-08 Novartis Ag Methods of selectively treating asthma using il-17 antagonists
WO2019087133A1 (en) 2017-11-02 2019-05-09 Novartis Ag Method of treating tendinopathy using interleukin-17 (il-17)
CN111372948A (en) * 2017-11-20 2020-07-03 诺华股份有限公司 Treatment of hidradenitis suppurativa with IL-17 antagonists
WO2019097493A1 (en) 2017-11-20 2019-05-23 Novartis Ag Treating hidradenitis suppurativa with il-17 antagonists
WO2019108900A1 (en) 2017-11-30 2019-06-06 Novartis Ag Bcma-targeting chimeric antigen receptor, and uses thereof
WO2019229701A2 (en) 2018-06-01 2019-12-05 Novartis Ag Binding molecules against bcma and uses thereof
EP3689907A1 (en) 2019-01-31 2020-08-05 Numab Therapeutics AG Antibodies targeting il-17a and methods of use thereof
WO2020157305A1 (en) 2019-01-31 2020-08-06 Numab Therapeutics AG Multispecific antibodies having specificity for tnfa and il-17a, antibodies targeting il-17a, and methods of use thereof
US11878037B2 (en) 2019-05-31 2024-01-23 Figene, Llc Concurrent activation of regenerative and tolerogenic processes by fibroblast-based compositions for the treatment of multiple sclerosis
EP3976032A4 (en) * 2019-05-31 2023-06-14 Figene, LLC Concurrent activation of regenerative and tolerogenic processes by fibroblast-based compositions for the treatment of multiple sclerosis
WO2021053591A1 (en) 2019-09-20 2021-03-25 Novartis Ag Methods of treating autoimmune diseases using interleukin-17 (il-17) antagonists
WO2021067195A1 (en) 2019-09-30 2021-04-08 Janssen Biotech, Inc. Compositions and methods for an il-17 target engagement assay with large molecule modulators
WO2021099924A1 (en) 2019-11-19 2021-05-27 Novartis Ag Methods of treating lupus nephritis using interleukin-17 (il-17) antagonists
WO2021111377A2 (en) 2019-12-06 2021-06-10 Novartis Ag Methods of treating lichen planus using interleukin-17 (il-17) antagonists
WO2021262876A1 (en) 2020-06-23 2021-12-30 Novartis Ag Methods of treating thyroid eye disease and graves' orbitopahy using interleukin-17 (il-17) antagonists
WO2022091056A1 (en) 2020-11-02 2022-05-05 Novartis Ag Interleukin-17 inhibitors
WO2022097060A1 (en) 2020-11-06 2022-05-12 Novartis Ag Cd19 binding molecules and uses thereof
WO2022117773A1 (en) 2020-12-02 2022-06-09 Fresenius Kabi Deutschland Gmbh Methods of selectively reducing antibodies
WO2022137132A1 (en) 2020-12-22 2022-06-30 Novartis Ag Methods for reducing the oxidation level of cysteine residues in a secreted recombinantly-expressed protein during cell culture
WO2023209519A1 (en) 2022-04-25 2023-11-02 Novartis Ag Crystalline forms of an il-17 inhibitor
WO2023223211A1 (en) 2022-05-16 2023-11-23 Novartis Ag Methods of treating giant cell arteritis using interleukin-17 (il-17) antagonists
WO2023223263A1 (en) 2022-05-18 2023-11-23 Novartis Ag Methods of selectively treating tendinopathy using interleukin-17 (il-17) antagonists

Also Published As

Publication number Publication date
PT1776142E (en) 2011-10-06
NO20150065L (en) 2007-03-30
ES2487533T3 (en) 2014-08-21
NZ552658A (en) 2009-11-27
PE20060418A1 (en) 2006-06-15
PL2902039T3 (en) 2018-09-28
NO341384B1 (en) 2017-10-30
EP1776142B1 (en) 2011-07-27
CA2573586A1 (en) 2006-02-09
DK2364729T6 (en) 2015-07-06
NO20171697A1 (en) 2007-03-30
AU2005268857B2 (en) 2010-01-28
TR201808057T4 (en) 2018-06-21
US7807155B2 (en) 2010-10-05
AU2005268857C1 (en) 2012-01-19
NO337286B1 (en) 2016-02-29
RU2011113153A (en) 2012-10-20
DK2902039T3 (en) 2018-07-16
US20190270804A1 (en) 2019-09-05
US8617552B2 (en) 2013-12-31
ES2367440T9 (en) 2021-04-28
MA28982B1 (en) 2007-11-01
ES2677245T3 (en) 2018-07-31
EP3409288A1 (en) 2018-12-05
CY1111963T1 (en) 2015-11-04
ES2487533T7 (en) 2015-07-14
MX2007001338A (en) 2008-03-11
ES2367440T7 (en) 2015-07-14
LTPA2015029I1 (en) 2022-07-11
KR20080029018A (en) 2008-04-02
BRPI0513078B1 (en) 2019-02-19
SI1776142T1 (en) 2011-11-30
HUS1500037I1 (en) 2017-10-30
AU2010201689A1 (en) 2010-05-20
ATE517924T1 (en) 2011-08-15
BR122017009404B1 (en) 2022-02-22
PT2902039T (en) 2018-07-17
NL300749I2 (en) 2017-11-02
JP2008507988A (en) 2008-03-21
US10344084B2 (en) 2019-07-09
EP2366405B1 (en) 2015-02-25
SG155186A1 (en) 2009-09-30
HUS1800040I1 (en) 2018-11-28
DK2366405T3 (en) 2015-05-11
AR050200A1 (en) 2006-10-04
ES2367440T3 (en) 2011-11-03
CY2018027I2 (en) 2020-05-29
NO20150064L (en) 2007-03-30
NO2018007I1 (en) 2018-02-14
CY1115444T1 (en) 2017-01-04
EP2366405A2 (en) 2011-09-21
BR122018075556B1 (en) 2022-10-18
AU2005268857A1 (en) 2006-02-09
US20140079719A1 (en) 2014-03-20
JP4682200B2 (en) 2011-05-11
PL1776142T3 (en) 2011-12-30
EP1776142A1 (en) 2007-04-25
EP2902039A1 (en) 2015-08-05
LTC1776142I2 (en) 2022-07-11
HK1207559A1 (en) 2016-02-05
US20120107325A1 (en) 2012-05-03
CY2018027I1 (en) 2020-05-29
DK1776142T3 (en) 2011-10-31
HRP20110758T4 (en) 2015-07-31
HUE038187T2 (en) 2018-10-29
RU2426741C2 (en) 2011-08-20
DK1776142T6 (en) 2015-07-06
IL180717A0 (en) 2007-06-03
ZA200700242B (en) 2008-08-27
US20230235038A1 (en) 2023-07-27
CY1120723T1 (en) 2019-12-11
EP2364729B3 (en) 2015-06-24
RU2426741C3 (en) 2017-11-15
AU2005268857B8 (en) 2010-05-27
TWI359153B (en) 2012-03-01
PL1776142T4 (en) 2018-04-30
BRPI0513078A (en) 2008-04-22
SI2902039T1 (en) 2018-07-31
BRPI0513078C1 (en) 2021-05-25
HK1256639A1 (en) 2019-09-27
NO2015023I2 (en) 2015-10-26
ECSP077198A (en) 2007-02-28
US20090280131A1 (en) 2009-11-12
HUE025815T2 (en) 2016-04-28
DK2364729T3 (en) 2014-07-21
NO20070985L (en) 2007-03-30
NO337129B1 (en) 2016-01-25
PL1776142T6 (en) 2016-06-30
PL2366405T3 (en) 2015-08-31
CY1116256T1 (en) 2017-02-08
LT2902039T (en) 2018-06-25
CA2573586C (en) 2013-06-25
EP2364729B1 (en) 2014-05-14
HRP20181069T1 (en) 2018-09-07
FR15C0048I1 (en) 2015-08-28
PL2364729T6 (en) 2016-06-30
IL180717A (en) 2012-07-31
TNSN07034A1 (en) 2008-06-02
TW200617025A (en) 2006-06-01
SI2366405T1 (en) 2015-07-31
NO336279B1 (en) 2015-07-06
HK1101277A1 (en) 2007-10-12
NO2022026I1 (en) 2022-06-24
RU2007108067A (en) 2008-09-10
LUC00088I2 (en) 2018-12-17
GB0417487D0 (en) 2004-09-08
ES2536228T3 (en) 2015-05-21
KR100852523B1 (en) 2008-08-14
PL2364729T3 (en) 2014-10-31
EP2364729A2 (en) 2011-09-14
NO20151787L (en) 2006-02-06
EP2902039B1 (en) 2018-04-11
CY2015030I2 (en) 2017-11-14
BRPI0513078B8 (en) 2019-08-27
EP2364729A3 (en) 2012-01-18
HRP20110758T1 (en) 2011-11-30
AU2010201689B2 (en) 2011-09-29
LTPA2018515I1 (en) 2018-10-25
EP1776142B3 (en) 2015-06-24
NO2016017I1 (en) 2016-08-23
KR20070036166A (en) 2007-04-02
CN101001645B (en) 2012-09-05
HRP20150480T1 (en) 2015-07-17
LU92768I2 (en) 2015-11-03
US20150152178A1 (en) 2015-06-04
US9765140B2 (en) 2017-09-19
US8119131B2 (en) 2012-02-21
CY2015030I1 (en) 2015-11-04
SI2364729T1 (en) 2014-08-29
FR15C0048I2 (en) 2015-11-20
PT2366405E (en) 2015-06-30
NO2015023I1 (en) 2015-11-02
CN101001645A (en) 2007-07-18
US20100215666A1 (en) 2010-08-26
US20170355762A1 (en) 2017-12-14
EP1776142B9 (en) 2016-09-07
PT2364729E (en) 2014-09-01
MY144925A (en) 2011-11-30
EP2366405A3 (en) 2012-01-18

Similar Documents

Publication Publication Date Title
US20230235038A1 (en) Interleukin-17 (il-17) antagonistic antibodies

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: DZP2005000160

Country of ref document: DZ

AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KM KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NG NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SM SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): BW GH GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LT LU LV MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

WWE Wipo information: entry into national phase

Ref document number: 200700242

Country of ref document: ZA

Ref document number: 250/DELNP/2007

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 12007500089

Country of ref document: PH

WWE Wipo information: entry into national phase

Ref document number: 2573586

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2005770286

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 180717

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 552658

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: MX/a/2007/001338

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 1020077002745

Country of ref document: KR

Ref document number: 07010436

Country of ref document: CO

WWE Wipo information: entry into national phase

Ref document number: 2005268857

Country of ref document: AU

Ref document number: 200580026569.4

Country of ref document: CN

Ref document number: 2007524286

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 1561/DELNP/2007

Country of ref document: IN

ENP Entry into the national phase

Ref document number: 2005268857

Country of ref document: AU

Date of ref document: 20050804

Kind code of ref document: A

WWP Wipo information: published in national office

Ref document number: 2005268857

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 11658344

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: DZP2007000160

Country of ref document: DZ

WWE Wipo information: entry into national phase

Ref document number: 2007108067

Country of ref document: RU

WWP Wipo information: published in national office

Ref document number: 1020077002745

Country of ref document: KR

WWP Wipo information: published in national office

Ref document number: 2005770286

Country of ref document: EP

ENP Entry into the national phase

Ref document number: PI0513078

Country of ref document: BR