WO2011053763A2 - Il-17a antagonists - Google Patents

Il-17a antagonists Download PDF

Info

Publication number
WO2011053763A2
WO2011053763A2 PCT/US2010/054662 US2010054662W WO2011053763A2 WO 2011053763 A2 WO2011053763 A2 WO 2011053763A2 US 2010054662 W US2010054662 W US 2010054662W WO 2011053763 A2 WO2011053763 A2 WO 2011053763A2
Authority
WO
WIPO (PCT)
Prior art keywords
seq
nos
amino acid
human
xaa
Prior art date
Application number
PCT/US2010/054662
Other languages
French (fr)
Other versions
WO2011053763A3 (en
Inventor
Michael Naso
Raymond Sweet
Jinquan Luo
Sheng-Jiun Wu
Merle Elloso
Daniela Della Ducata
Robert Rauchenberger
Mark Rutz
Juan Carlos Almagro
Susann Taudte
Bingyuan Wu
Galina Obmolova
Thomas Malia
Original Assignee
Centocor Ortho Biotech Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=43922991&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2011053763(A2) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority to UAA201206558A priority Critical patent/UA114070C2/en
Priority to MX2012005086A priority patent/MX2012005086A/en
Priority to CN201080059869.3A priority patent/CN102905727B/en
Priority to EA201290254A priority patent/EA029283B1/en
Priority to KR1020127013837A priority patent/KR101836217B1/en
Priority to CA2779257A priority patent/CA2779257C/en
Priority to ES10827516T priority patent/ES2728115T3/en
Application filed by Centocor Ortho Biotech Inc. filed Critical Centocor Ortho Biotech Inc.
Priority to JP2012537102A priority patent/JP5922025B2/en
Priority to EP10827516.5A priority patent/EP2493506B1/en
Priority to AU2010313304A priority patent/AU2010313304B2/en
Priority to NZ599737A priority patent/NZ599737A/en
Priority to BR112012010280-0A priority patent/BR112012010280B1/en
Publication of WO2011053763A2 publication Critical patent/WO2011053763A2/en
Publication of WO2011053763A3 publication Critical patent/WO2011053763A3/en
Priority to IL219390A priority patent/IL219390B/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
    • C07K16/244Interleukins [IL]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/713Double-stranded nucleic acids or oligonucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/56Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/59Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/04Drugs for disorders of the respiratory system for throat disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/04Drugs for skeletal disorders for non-specific disorders of the connective tissue
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/33Crossreactivity, e.g. for species or epitope, or lack of said crossreactivity
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/34Identification of a linear epitope shorter than 20 amino acid residues or of a conformational epitope defined by amino acid residues
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • 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

  • the present invention relates to interleukin-17A (IL-17A) antibody antagonists, polynucleotides encoding IL-17A antibody antagonists or fragments thereof, and methods of making and using the foregoing.
  • IL-17A interleukin-17A
  • Interleukin-17A is a cytokine secreted by activated Thl7 cells, CD8 + T cells, ⁇ T cells and NK cells in response to cytokines such as IL-23 and TGF- ⁇ , and regulates production of mediators such as antimicrobial peptides (defensins), proinflamatory cytokines and chemokines from multiple cell types such as fibroblasts and synoviocytes that are involved in neutrophil biology, inflammation, organ destruction and host defense (reviewed in Weaver et al . , Annu. Rev. Immunol. 25:821-52, 2007; Aggarwal et al . , J. Biol. Chem.
  • IL-17A synergizes with other cytokines, such as TNF-a and IL- ⁇ to potentiate the pro-inflammatory environment.
  • the IL-17A cytokine family consists of six homologs designated IL-17A, B, C, D, E and F, each with divergent and distinct biological roles (Kawaguchi et al . , J. Allergy Clin. Immunol. 114:1265-73, 2004; Rolls and Linden, Immunity 21:467-76, 2004; Moseley et al . , Cytokine Growth Factor Rev. 14:155-74, 2003) .
  • IL-17F is most homologous to IL- 17A and shares many similar functional properties such as induction of neutrophilia in the lung and induction of proinflammatory cytokines; however, in man, IL-17F is about 10-fold less potent than IL-17A (Moseley et al . , Cytokine Growth Factor Rev. 14:155-74, 2003; Rolls et al . , Immunity, 21: 467-76, 2004; McAllister et al . , J. Immunol. 175:404-12, 2005) .
  • IL-17A and IL- 17F can also form heterodimers , which have intermediate bioactivity in vitro (Wright et al . , J. Biol. Chem. 282:13447-55, 2007) .
  • IL-17A mediates it effects by interacting with the
  • IL-17F Interleukin-17 receptor A (IL-17RA) and receptor C (IL-17RC) (Moseley et al . , Cytokine Growth Factor Rev. 14:155-74, 2003; Toy et al . , J. Immunol. 177:36-9, 2006) .
  • IL-17F signals through the same receptors, although IL-17F affinity to the receptors is significantly lower (Kuestner et al . , J. Immunol. 179:5462-73,
  • Crystal structures of human IL-17F and human IL-17F/IL- 17RA complex identified a putative receptor-binding cavity in the IL-17F homodimer (Hymowitz et al . , EMBO J. 20:5332-41, 2001; Ely et al . , Nat. Immunology 10:1245-51, 2009) .
  • a similar cavity was identified in the crystal structure of human IL-17A in complex with a neutralizing Fab, although the cavity was partially occupied (Gerhardt et al . , J. Mol. Biol. 394:905-21, 2009) .
  • asthma asthma
  • psoriasis psoriasis
  • dermal hypersensitivity including atopic dermatitis (Toda et al . , J. Allergy Clin. Immunol. 111:875-81, 2003)
  • systemic sclerosis Fejimoto et al . , J. Dermatolog. Sci. 50:240-42, 2008
  • inflammatory bowel diseases including ulcerative colitis and Crohn's disease (Holtta et al., Inflamm. Bowel Dis. 14:1175-84, 2008; Zhang et al . , Inflamm. Bowel Dis. 12:382-88, 2006), and pulmonary diseases including chronic obstructive pulmonary disease (Curtis et al., Proc. Am. Thorac . Soc. 4:512-21, 2007) .
  • Antibodies to IL-17A have been proposed for use in the treatment of IL-17A mediated diseases and disorders (PCT Publ . Nos: WO08/021156, WO07/070750, WO07/149032, WO06/054059,
  • Fig. 1 A-H. CDR sequences of Family 2, 6a, 6b, 19a, and 19b IL-17A antibody antagonists.
  • Fig. 2 Exemplary A) IGLV3 and IGLJ; and B) IGHV3 and IGHJ germline genes as scaffolds for grafting paratope residues.
  • mAb6785 sequence is shown above. CDR regions are underlined and core contact sites in mAb6785 light chain (Y31, D49, Y90, F92, F93) and heavy chain (S52, T54, F57, Y59, Q99, L100 and T101) are denoted by an asterix "*".
  • Framework 4 regions in B) are double underlined. Sequence shown are *01 alleles unless specifically
  • Fig. 3 Rabat and Chotia numbering for select antibody A) light and B) heavy chains. Locations of Rabat CDRs and Chothia HVs are highlighted in gray.
  • Fig. 4 Competitive binding assays of labeled A) and B) mAbl926; C) mAb317; D) mAb3171; E) and F) mAb7357 with IL-17A in an ELI SA format.
  • FIG. 5 A) H/D exchange maps of the IL-17A complexed with different anti-IL-17A mAbs . Numbering above the protective blocks corresponds to mature IL-17A (SEQ ID NO: 105) sequence numbering .
  • Fig. 6. A) The overall molecular structure of IL-17A/ Fab6468 complex. The dimer of IL-17A is shown dark gray and light gray. The two Fab molecules are shown in dark gray and light gray, repectively; B) Comparison of monomer of IL-17A (light gray) and IL-17F (dark gray) ; C) Dimer of IL-17A (light and dark gray) ; D) Dimer of IL-17F (light and dark gray) .
  • Fig. 7. The two binding sites and the core epitope on IL- 17A for Fab6468. Protomers 1 and 2 are dark and light gray, respectively. The core epitope is indicated by the black oval. The broken line represents disordered residues.
  • FIG. 8 Comparison of IL-17A and IL-17F putative receptor binding pockets.
  • Fig. 9 Binding specificity of mAbl926 to different species of IL-17A proteins in an ELISA format.
  • One aspect of the invention is an isolated antibody or fragment thereof, wherein the antibody binds specifically to human IL-17A having the sequence shown in SEQ ID NO: 105 at amino acid residues 56-68 (SEQ ID NO: 157) and 100-116 (SEQ ID NO:
  • Another aspect of the invention is an isolated antibody or fragment thereof, wherein the antibody binds specifically to a P2 pocket cavity on human IL-17A, the P2 pocket cavity comprising amino acid residues V22, V24, L26, 128, Y62, L99, R101, F110, and LI 12 of SEQ ID NO: 105.
  • Another aspect of the invention is an isolated antibody or fragment that binds specifically to human IL-17A that competes for human IL-17A binding with a monoclonal antibody comprising the amino acid sequences of certain heavy chain complementarity determining regions (CDR) 1, 2 and 3 (HCDR1, HCDR2, HCDR3) , the amino acid sequences of certain light chain complementarity determining reigons (CDR) 1, 2 and 3 (LCDR1, LCDR2, LCDR3) , the amino acid sequences of certain heavy chain variable regions (VH) or the amino acid sequences of certain light chain variable regions (VL) .
  • CDR heavy chain complementarity determining regions
  • LCDR1, LCDR2, LCDR3 amino acid sequences of certain light chain complementarity determining reigons
  • VH amino acid sequences of certain heavy chain variable regions
  • VL amino acid sequences of certain light chain variable regions
  • Another aspect of the invention is an isolated antibody or fragment that binds specifically to human IL-17A, comprising certain heavy chain variable region paratope amino acid residues and certain light chain variable region paratope amino acid residues that interact with certain residues of human IL-17A having the amino acid sequence shown in SEQ ID NO: 105.
  • Another aspect of the invention is an isolated antibody or fragment that binds specifically to human IL-17A, comprising a heavy chain variable region and a light chain variable region, wherein the antibody comprises a heavy chain variable region paratope selected from Chothia residues F56 and Y58; and a light chain variable region paratope selected from Chothia residues Y91, F93 and F94.
  • Another aspect of the invention is an isolated antibody or fragment that binds specifically human IL-17A, comprising a heavy chain variable region (VH) and a light chain variable region (VL) , wherein the antibody comprises the amino acid sequences of certain heavy chain complementarity determining regions (CDR) 1,
  • HCDR1 the amino acid sequences of certain light chain complementarity determining reigons (CDR) 1,
  • LCDR1 LCDR2, LCDR3
  • VH amino acid sequences of certain heavy chain variable regions
  • VL amino acid sequences of certain light chain variable regions
  • Another aspect of the invention is an isolated antibody or fragment that specifically binds human IL-17A, wherein the antibody comprises the amino acid sequences of certain heavy chains and the amino acid sequences of certain light chains.
  • Another aspect of the invention is a pharmaceutical composition
  • a pharmaceutical composition comprising the isolated antibody or fragment of the invention and a pharmaceutically acceptable carrier.
  • Another aspect of the invention is an isolated antibody heavy chain comprising the amino acid sequence shown in SEQ ID NOs : 67, 68, 69, 81, 82, 83, 84, 85, 86, 92, 93, 94, 95, 96, 97, 98, 99, or 100.
  • Another aspect of the invention is an isolated antibody light chain comprising the amino acid sequence shown in SEQ ID NOs: 76, 77, 78, 79, 80, 87, 88, 89, 90, or 91.
  • Another aspect of the invention is an isolated
  • polynucleotide encoding an antibody heavy chain comprising the amino acid sequence shown in SEQ ID NO: 67, 68, 69, 81, 82, 83, 84, 85, 86, 92, 93, 94, 95, 96, 97, 98, 99, or 100.
  • Another aspect of the invention is an isolated
  • polynucleotide encoding an antibody light chain comprising the amino acid sequence shown in SEQ ID NO: 76, 77, 78, 79, 80, 87, 88, 89, 90, or 91.
  • Another aspect of the invention is a vector comprising at least one polynucleotide of the invention.
  • Another aspect of the invention is a host cell comprising the vector of the invention.
  • Another aspect of the invention os a method of inhibiting interaction of human IL-17A with IL-17RA comprising: providing a human IL-17A and IL-17RA; and
  • Another aspecf of the invention si a method of inhibiting IL-17A biological activity, comprising: providing a human IL17-A and IL-17RA; and contacting the human IL-17A with an antagonist that binds the human IL-17A at at least one amino acid residue selected from the group consisting of V22, V24, L26, 128, Y62, L99, R101, F110, and L112.
  • Another aspect of the invention is a method of treating an inflammatory condition comprising administering a therapeutically effective amount of the isolated antibody of claim 3 or 7 to a patient in need thereof for a time sufficient to treat the inflammatory condition.
  • antagonist means a molecule that partially or completely inhibits, by any mechanism, IL-17A activity.
  • exemplary antagonists are antibodies, fusion proteins, peptides, peptidomimetics , nucleic acids, oligonucleotides and small molecules.
  • the agent can be identified using well known assays for IL-17A activity described below.
  • IL-17A antibody antagonist or an "antibody reactive with IL-17A” as used herein refers to an antibody that is capable of, directly or indirectly, reducing or inhibiting IL- 17A biological activity, blocking binding of IL-17A to its receptor, or inhibiting IL-17A receptor activation.
  • an antibody reactive with IL-17A can bind directly to IL-17A and neutralize IL-17A activity, i.e, block IL-17A signaling to reduce cytokine and chemokine release.
  • IL-17A refers to a human IL-17A polypeptide having an amino acid sequence shown in GenBank Acc . No. NP_002181.
  • SEQ ID NO: 105 shows the amino acid sequence of the mature human IL-17A.
  • IL-17A in vivo forms homodimers of two monomers, which are designated monomer A and monomer B, or protomer A and protomer B, or protomer 1 and protomer 2, or chain A and chain B.
  • IL-17A can also form a heterodimer with IL-17F.
  • the term "IL-17A" comprises the monomer, the homodimer, and the heterodimer forms.
  • IL 17Amut6 refers to a variant of IL-17A having A70Q and A132Q substitutions.
  • the amino acid sequence of the mature IL-17Amut6 is shown in SEQ ID NO: 106, and the cDNA sequence in SEQ ID NO: 112.
  • IL-17A and IL-17Amut6 have comparable activities (PCT. Pat Appl. No. WO09/003096) .
  • IL-17A receptor comprises both receptor polypeptides, IL-17RA (GenBank Acc no: NP_055154, SEQ I NO: 107) and IL-17RC (GenBank Acc No NP_703191, SEQ ID NO: 113), and homodimers or heterodimers of the two polypeptides.
  • antibodies as used herein is meant in a broad sense and includes immunoglobulin molecules including polyclonal antibodies, monoclonal antibodies including murine, human, human adapted, humanized and chimeric monoclonal antibodies, antibody fragments, multispecific antibodies formed from at least two intact antibodies, dimeric, tetrameric or multimeric antibodies.
  • mAb monoclonal antibody
  • Monoclonal antibodies are highly specific, typically being directed against a single epitope.
  • the modifier “monoclonal” indicates the substantially homogeneous character of the antibody and does not require production of the antibody by any particular method.
  • Immunoglobulins can be assigned to five major classes, namely IgA, IgD, IgE, IgG and IgM, depending on the heavy chain constant domain amino acid sequence.
  • IgA and IgG are further sub-classified as the isotypes IgA x , IgA 2 , IgGi, IgG 2 , IgG 3 and IgG 4 .
  • Antibody light chains of any vertebrate species can be assigned to one of two clearly distinct types, namely kappa ( ⁇ ) and lambda ( ⁇ ) , based on the amino acid sequences of their constant domains .
  • the term "antibody fragments" comprise at least a portion of an immunoglobulin molecule, such as a heavy chain
  • HCDR complementarity determining region
  • LCDR complementarity determining region
  • VH heavy chain variable region
  • VL light chain variable region
  • CH heavy chain constant region
  • CL light chain constant region
  • FR framework region
  • An antibody may be a Fab, F(ab'), F(ah') 2 , scFv, dsFv, or diabody. Structures of the above mentioned antibody fragments, and techniques for the preparation and use of the antibodies and fragments thereof are well known in the art.
  • An antibody variable region consists of a "framework" region interrupted by three "antigen-binding sites".
  • the antigen-binding sites are defined using various terms: (i)
  • Complementarity Determining Regions are based on sequence variability (Wu and Rabat, J. Exp. Med. 132:211-250, 1970; Rabat et al . , Sequences of Proteins of
  • Chothia residues as used herein are the antibody VL and VH residues numbered according to Al-Lazikani (Al-Lazikani et al . , J. Mol. Biol. 273:927-48, 1997) . Correspondence between the two most used numbering systems, Rabat (Rabat et al.,
  • polypeptide numbering is shown in Figure 3 for exemplary
  • Framework or “framework sequences” are the remaining sequences of a variable region other than those defined to be antigen-binding site. Because the antigen-binding site can be defined by various terms as described above, the exact amino acid sequence of a framework depends on how the antigen-binding site was defined.
  • substantially identical means that the two antibody or antibody fragment amino acid sequences being compared are identical or have "insubstantial differences.” Insubstantial differences are substitutions of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 amino acids in an antibody or antibody fragment amino acid sequence that do not adversely affect antibody properties. Amino acid sequences substantially identical to the sequences disclosed herein are also part of this application. In some embodiments, the sequence identity can be about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or higher.
  • Percent identity can be determined for example by pairwise alignment using the default settings of the AlignX module of Vector NTI v.9.0.0 (Invitrogen, Carslbad, CA) .
  • the protein sequences of the present invention can be used as a query sequence to perform a search against public or patent databases to, for example, identify related sequences.
  • Exemplary programs used to perform such searches are the XBLAST or BLASTP programs (http //www ncbi nlm/nih gov), or the GenomeQuestTM (GenomeQuest , Westborough, MA) suite using the default settings.
  • inflammatory condition refers to acute or chronic localized or systemic responses to harmful stimuli, such as pathogens, damaged cells, physical injury or irritants, that are mediated in part by the activity of
  • cytokines e.g., neutrophils, monocytes, lymphocytes, macrophages
  • inflammatory cells e.g., neutrophils, monocytes, lymphocytes, macrophages
  • IL-17A-mediated inflammatory condition refers to an inflammatory condition resulting at least partially from IL-17A biological activity, or caused by IL-17A activity.
  • exemplary IL-17A-mediated inflammatory conditions are psoriasis and rheumatoid arthritis.
  • IL-17A-mediated condition encompasses all diseases and medical conditions in which IL-17A plays a role, whether directly or indirectly, in the disease or medical condition, including the causation, development,
  • epitope as used herein means a portion of an antigen to which an antibody specifically binds.
  • Epitopes usually consist of chemically active (such as polar, non-polar or hydrophobic) surface groupings of moieties such as amino acids or polysaccharide side chains and can have specific three- dimensional structural characteristics, as well as specific charge characteristics.
  • An epitope can be composed of either or both contiguous or discontiguous amino acids that form a
  • paratope as used herein means a portion of an antibody to which an antigen specifically binds.
  • a paratope can be linear in nature or can be discontinuous, formed by a spatial relationship between non-contiguous amino acids of an antibody rather than a linear series of amino acids.
  • a "light chain paratope” and a “heavy chain paratope” or “light chain paratope amino acid residues” and “heavy chain paratope amino acid residues” refer to antibody light chain and heavy chain residues in contact with an antigen, respectively.
  • the term "specific binding” as used herein refers to antibody binding to a predetermined antigen with greater affinity than for other antigens or proteins.
  • the antibody binds with a dissociation constant (K D ) of 10 ⁇ 7 M or less, and binds to the predetermined antigen with a K D that is at least ten fold less than its K D for binding to a non-specific antigen (e.g., BSA, casein, or any other specified polypeptide) other than the predetermined antigen.
  • K D dissociation constant
  • the phrases “an antibody recognizing an antigen” and “an antibody specific for an antigen” are used interchangeably herein with the term “an antibody specifically binding to an antigen” or "an antigen specific antibody” e.g. an IL-17A specific antibody.
  • the dissociation constant can be measured using standard procedures .
  • IL-17A biological activity or "IL-17A
  • IL-17A activation refers to any activity occurring as a result of IL-17A binding to the IL-17A receptor.
  • Exemplary IL- 17A biological activities result in increased secretion of IL-6 or IL-8, NF- ⁇ activation, or regulation of downstream kinases sucn as ERK1, ERK2 and p38 upon binding to the IL-17A receptor.
  • the release of cytokines and chemokines from cells, tissues or in circulation, NF- ⁇ activation, or kinase phosporylation events can be measured using well known methods, for example
  • vector means a polynucleotide capable of being duplicated within a biological system or that can be moved between such systems.
  • Vector polynucleotides typically contain elements, such as origins of replication, polyadenylation signal or selection markers, that function to facilitate the duplication or maintenance of these polynucleotides in a biological system.
  • examples of such biological systems may include a cell, virus, animal, plant, and reconstituted biological systems utilizing biological components capable of duplicating a vector.
  • the polynucleotide comprising a vector may be DNA or RNA molecules or a hybrid of these.
  • expression vector means a vector that can be utilized in a biological system or in a reconstituted biological system to direct the translation of a polypeptide encoded by a polynucleotide sequence present in the expression vector.
  • polynucleotide means a molecule comprising a chain of nucleotides covalently linked by a sugar-phosphate backbone or other equivalent covalent chemistry. Double and single-stranded DNAs and RNAs are typical examples of
  • polypeptide or "protein” means a molecule that comprises at least two amino acid residues linked by a peptide bond to form a polypeptide. Small polypeptides of less than 50 amino acids may be referred to as "peptides”.
  • the present invention provides IL-17A antibody antagonists capable of inhibiting IL-17A biological activity and uses of such antibodies.
  • exemplary mechanisms by which IL-17A activation may be inhibited by such antibodies include in vitro, in vivo or in situ inhibition of IL-17A homo-or heterodimerization, and blocking binding of IL-17A to the IL-17A receptor, inhibition of receptor dimerization, inhibition of kinase activity of
  • antibody antagonists capable of inhibiting IL-17A activation by other mechanisms are also within the scope of the various aspects and embodiments of the invention. These antagonists are useful as research reagents, diagnostic reagents and therapeutic agents.
  • the invention provides novel antigen-binding sites derived from human immunoglobulin gene libraries.
  • the structure for carrying an antigen-binding site is generally an antibody heavy or light chain or portion thereof.
  • the invention provides an isolated antibody or fragment thereof that binds specifically to human IL-17A, comprising a heavy chain variable region (VH) and a light chain variable region (VL) , wherein the antibody comprises the heavy chain complementarity determining region (CDR) 1, 2 and 3 (HCDR1, HCDR2 and HCDR3) amino acid sequences and the light chain complementarity determining region (CDR) 1, 2 and 3 (LCDR1, LCDR2 and LCDR3) amino acid sequences as shown in Table la.
  • the invention provides an isolated antibody or fragment that binds specifically to human IL-17A, comprising a VH and a VL, wherein the antibody comprises the HCDR1, HCDR2 and HCDR3 amino acid sequences as shown in SEQ ID NOs: 23, 35 and 52, wherein the HCDR2 of SEQ ID NO: 35 is further defined as shown in Formula (I) :
  • Xaai may be His, Met, Arg, Ser or Tyr;
  • Xaa 2 may be Trp, Thr or Tyr
  • Xaa 3 may be Tyr, Phe, Ser or Asp.
  • the invention provides an isolated antibody or fragment that binds specifically to human IL-17A, comprising a VH and a VL, wherein the antibody comprises the LCDR1, LCDR2 and LCDR3 amino acid sequences as shown in SEQ ID NOs: 2, 5 and 11, wherein the LCDR3 of SEQ ID NO: 11 is further defined as shown in Formula (II) :
  • Xaa 4 may be His or Gin
  • Xaa 5 may be Phe or Gly
  • Xaa 6 may be Thr, Val or Asn
  • Xaa 7 may be lie, Thr or Tyr
  • Xaa 8 may be Pro or Arg
  • Xaa 9 may be Ser or Pro
  • Xaaio may be His, Phe or Leu.
  • the invention provides an isolated antibody or fragment that binds specifically to binding human IL- 17A, comprising a VH and a VL, wherein the antibody comprises the LCDR1, LCDR2, and LCDR3 amino acid sequences as shown in SEQ ID NOs: 2, 5 and 17, wherein the LCDR3 of SEQ ID NO: 17 is further shown in Formula (HI) :
  • Xaa may be Gin or Thr
  • Xaa ⁇ 2 may be Ser or Tyr
  • Xaa i3 may be Asn, Arg, Val or Tyr;
  • Xaai 4 may be His or Ser
  • Xaais may be He, Thr, Leu, Ala or Ser;
  • Xaais may be Pro , Leu or Ser
  • Xaa i7 may be Pro , Ser, Phe or Leu;
  • Xaaie may be Ala, Leu or Asp.
  • the invention provides an isolatec body or fragment that binds specifically to human IL-17A, comprising a VH and a VL, wherein the antibody comprises the HCDR1, HCDR2 and HCDR3 amino acid sequences as shown in SEQ ID NOs: 24, 36 and 57, wherein the HCDR3 of SEQ ID NO: 57 is further defined as shown in Formula (IV) :
  • Xaaig is Met, lie, Leu or Thr.
  • the invention provides an isolated antibody or fragment that binds specifically to human IL-17A, comprising a VH and a VL, wherein the antibody comprises the LCDR1, LCDR2 and LCDR3 amino acid sequences as shown in SEQ ID NOs: 3, 6 and 22, wherein the LCDR3 of SEQ ID NO: 22 is further defined as shown in Formula (V) :
  • Xaa 20 is Met, Leu, Thr or Tyr.
  • the invention provides an isolated antibody or fragment that binds specifically to human IL-17A, comprising a VH and a VL, wherein the antibody comprises the HCDR1, HCDR2 and HCDR3 amino acid sequences as shown in SEQ ID NOs: 25, 46 and 61, wherein the HCDR2 of SEQ ID NO: 46 is further defined as shown in Formula (VI) :
  • Xaa 2 i may be Ala, Gly, Thr or Val;
  • Xaa 22 may be Asn or Ser
  • Xaa 23 may be Gly, Met, Lys, lie, Leu or His;
  • Xaa 24 may be Leu, Asp, Ala, His, Thr, Gly or Ser;
  • Xaa 25 may be Gly or Ser
  • Xaa 26 may be Thr, Gly, Tyr or Asp;
  • Xaa 2 7 may be His, Trp, Tyr or Phe;
  • Xaa 28 may be Lys, Thr or lie;
  • Xaa 2 g may be Tyr, Phe or Asn, and
  • HCDR3 of SEQ ID NO: 61 is defined as shown in Formula (VII) :
  • Xaa 30 may be Met, Leu or Thr.
  • the invention provides an isolated antibody or fragment that binds specifically to human IL-17A, comprising a VH and a VL, wherein the antibody comprises the HCDR1, HCDR2 and HCDR3 amino acid sequences as shown in SEQ ID NOs: 25, 51 and 58, wherein the HCDR2 of SEQ ID NO: 51 is further defined as shown in Formula (VIII) :
  • Xaa 3 i may be Ala, Lys, Met or His;
  • Xaa 32 may be Asn, Met, Thr or Arg;
  • Xaa 33 may be Gly or Asp
  • Xaa 34 may be Arg, His or Asn
  • Xaa 35 may be Asp or Gly.
  • Antibodies whose antigen-binding site amino acid sequences are substantially identical to those shown in Table la are encompassed within the scope of the invention. Typically, this involves one or more amino acid substitutions with an amino acid having similar charge or hydrophobic or stereochemical characteristics, and are made to improve antibody properties, for example stability or affinity.
  • a conservative substitution may involve a substitution of a native amino acid residue with a nonnative residue such that there is little or no effect on the polarity or charge of the amino acid residue at that position.
  • any native residue in the polypeptide may also be substituted with alanine, as has been previously described for alanine scanning mutagenesis (MacLennan et al., Acta Physiol. Scand. Suppl . 643:55-67, 1998; Sasaki et al . , Adv. Biophys. 35:1-24, 1998) .
  • Conservative substitutions will produce molecules having functional and chemical
  • Non-conservative substitutions in the functional and/or chemical characteristics of the molecules may be accomplished by selecting substitutions in the amino acid sequence that differ significantly in their effect on maintaining
  • amino acid substitutions can be used to identify residues important for the function of the antibodies, such as residues affecting affinity, or residues that impart undesireable properties such as aggregation.
  • Substitutions in the framework regions, in contrast to antigen- binding sites may also be made as long as they do not adversely affect the properties of the antibody.
  • Framework substitutions can be made for example at the Vernier Zone residues (US. Pat. No. 6,649,055) to improve antibody affinity or stability.
  • Substitutions can also be made at those framework positions in the antibody that differ in sequence when compared to the homologous human germline gene sequences to reduce possible immunogeneicity . These modifications can be done for example to antibodies derived from de novo antibody libraries, such as pIX libraries.
  • amino acid substitutions also encompass non-natural amino acid residues which are typically incorporated by chemical peptide synthesis rather than by synthesis in biological systems. Amino acid substitutions can be done for example by PCR mutagenesis (US Pat. No. 4,683,195) .
  • variants can be generated using well known methods, for example using random (NNK) or non-random codons, for example DVK codons, which encode 11 amino acids (ACDEGKNRSYW) , and screening the libraries or variants with desired properties, as shown in Example 1.
  • Figure 1 shows substitutions made to five parent IL-17A antibody antagonists within the LCDR3, HCDR2 and HCDR3 regions to improve antibody properties .
  • the invention provides an isolated antibody or fragment that binds specifically to human IL-17A, comprising a VH and a VL, wherein the antibody comprises certain VH and VL sequences, and also provides for each isolated VH and VL as shown in Table 2.
  • alternative embodiments may comprise single heavy chain variable regions or single light chain variable regions, single full length antibody chains, or CDR1, CDR2 and CDR3 regions from one antibody chain, either heavy or light.
  • the single variable region, full length antibody chain or CDR1, CDR2 and CDR3 region of one chain can be used to screen for
  • the screening may be accomplished by phage display screening methods using, e.g., a hierarchical dual combinatorial approach disclosed in PCT Publ. No. WO92/01047.
  • phage display screening methods using, e.g., a hierarchical dual combinatorial approach disclosed in PCT Publ. No. WO92/01047.
  • an individual colony containing either a H or L chain clone is used to infect a complete library of clones encoding the other chain (L or H) , and the resulting two-chain specific antigen-binding domain is selected in accordance with phage display techniques as
  • the invention provides an isolated antibody or fragment that binds specifically to human IL-17A, comprising a VH and a VL having amino acid sequences at least 90% identical to the VH and VL amino acid sequences shown in Table 2.
  • the invention provides an isolated antibody or fragment that binds specifically to human IL-17A, comprising a VH and a VL having amino acid sequences at least 95% identical to the VH and VL amino acid sequences shown in Table 2.
  • the invention provides an isolated antibody or fragment having certain heavy chain and light chain amino acid sequences as shown in Table 2.
  • polypeptides encoding antibody chains can be numbered based on Rabat's or Chothia' s numbering (Rabat et al . , sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md., 1991; Chothia and Lesk, Mol . Biol.
  • the invention provides an isolated antibody or fragment that binds specifically to human IL-17A, comprising a VH and a VL, wherein the antibody comprises a heavy chain variable region paratope selected from Chothia residues S51, T53, F56, Y58, Q95, L96 and T97 and a light chain variable region paratope selected from Chothia residues Y32, D50, Y91, F93 and F94.
  • the heavy chain paratope and the light chain paratope Chothia residues correspond to heavy chain residues S52, T54, F57, Y59, Q99, L100 and T101 of SEQ ID NO: 86 and light chain residues Y31, D49, Y90, F92 and F93 of SEQ ID NO: 79.
  • the invention provides an isolated antibody or fragment that binds specifically to human IL-17A, comprising heavy chain variable region paratope amino acid residues that interact with residues of human IL-17A having the amino acid sequence shown in SEQ ID NO: 105, comprising:
  • a lysine residue that interacts with E57 of human IL-17A a tyrosine residue that interacts with P59, E60 or R101 of human IL-17A;
  • the invention provides an isolated antibody or fragment that binds specifically to human IL-17A, comprising light chain variable region paratope amino acid residues that interact with residues of human IL-17A having the amino acid sequence shown in SEQ ID NO: 105, comprising:
  • a first tyrosine residue that interacts with P59, S64 or R101 of human IL-17A a second phenylalanine residue that interacts with P59, E60, R61, Y62, R101 or F110 of human IL-17A; and
  • the invention provides an isolated antibody or fragment that binds specifically to human IL-17A, comprising heavy chain variable region paratope amino acid residues and light chain variable region paratope amino acid residues that interact with residues of human IL-17A having the amino acid sequence shown in SEQ ID NO: 105, comprising:
  • the invention provides an isolated antibody or fragment that binds specifically to human IL-17A, comprising a heavy chain variable region and a light chain variable region, wherein the antibody comprises:
  • a heavy chain variable region paratope selected from
  • a light chain variable region paratope selected from
  • the heavy chain paratope Chothia residues F56 and Y58 and the light chain paratope Chothia residues Y91, F92 and F94 are residues in direct contact with IL-17A residues L26, Y62, R101 and F110. These IL-17A residues are part of both the Fab6468 epitope and the P2 pocket cavity (see below) . While not wishing to be bound to any particular theory, it is believed that interaction between the Fab6468 and IL-17A at these select residues may be sufficient for the antibody to block IL-17A activity .
  • Fully human mAbs lacking any non-human sequences can be prepared and optimized from phage display libraries by techniques referenced in, e.g., Knappik et al . , J. Mol. Biol. 296:57-86, 2000; and Krebs et al . , J. Immunol. Meth. 254:67-84 2001.
  • the antibodies of the invention are isolated from libraries expressing antibody heavy and light chain variable regions as fusion proteins with bacteriophage pIX coat protein.
  • the antibody libraries are screened for bining to human IL-17mut6 (SEQ ID NO: 105), and the obtained positive clones are further characterized, the Fabs isolated from the clone lysates, and expressed as full length IgGs. Exemplary antibody libraries and screening methods are described in Shi et al . , J. Mol. Biol.
  • the resulting mAbs can further be modified in their framework regions to change certain framework residues to those present in a matching human germline, as exemplified within.
  • Anibodies of the invention binding specific IL-17A epitopes can be made by immunizing humanized mice expressing human immunoglobulin loci (Lonberg et al . , Nature 368:856-9, 1994;
  • mice with the peptides encoding the epitopes, for example peptide 56 NEDPERYPSVIWE 68 (SEQ ID NO: 157) or 100 RREPPHCPNSFRLEKILn 6 (SEQ ID NO: 158) and using the hybrodima method of Kohler et al . , Nature 256:495-97.
  • the resulting antibodies are tested for their binding to the epitope using standard methods.
  • the identified mAbs can further be modified by incorporating altered framework support residues to preserve binding affinity by techniques such as those disclosed in Queen et al . , Proc. Natl. Acad. Sci. (USA), 86:10029-32, 1989 and Hodgson et al., Bio/Technology, 9:421, 1991.
  • Isolated antibodies having certain paratope residues that bind specifically to human IL-17A can be made, for example, by grafting the paratope residues into a suitable scaffold,
  • Exemplary scaffolds are amino acid sequences of human antibody variable regions encoded by human germline genes.
  • the scaffolds can be selected based on for example overall sequence homology, % identity between the paratope residues, or canonical structure class identity between the scaffold and an exemplary antibody, such as mAb6785.
  • Human antibody germline genes are disclosed in, for example, Tomlinson et al . , J. Mol. Biol
  • Consensus human framework regions can also be used, e.g., as described in U.S. Pat. No. 6,054,297. Selection of suitable scaffold can be done for example according to methods described in PCT Publ . No.
  • scaffolds onto which the paratope residues are grafted are the genes encoded by the ⁇ 3, Vh3, J , and the Jh frameworks.
  • VK3 genes are IGLV3-1, IGLV3-9, IGLV3-10, IGLV3-12, IGLV3-16, IGLV3-19, IGLV3-21, IGLV3-22, IGLV3-25, IGLV3-27, and IGLV3-32 (IMGT nomenclature, *01 alleles), (SEQ ID NOs : 117-127, respectively) .
  • Exemplary J genes are IGLJ1, IGLJ2, IGLJ3, IGLJ4, IGLJ5, IGLJ6, and IGLJ7 (SEQ ID NOs: 128-134,
  • Vh3 genes are IGHV3-7, IGHV3-9, IGHV3- 11, IGHV3-16, IGHV3-19, IGHV3-20, IGHV3-21, IGHV3-23, IGHV3-30, IGHV3-30*03, IGHV3-33, IGHV3-45, IGHV3-48, IGHV3-64, and IGHV3-74 (IMGT nomenclature, *01 alleles except when different allele is specificed) (SEQ ID NO : s 135-150, respectively) .
  • Exemplary Jh genes are IGHJ1, IGHJ2, IGHJ3, IGHJ4, IGHJ5, and IGHJ6 (SEQ ID NO : s 151-156, respectively) .
  • the germline J-regions are used in their entirety or in part to select FR4 sequences.
  • the mAb6785 light chain paratope residues can be grafted into a ⁇ 3 protein framework encoded by IGLV3-1 (SEQ ID NO: 117) that is joined to the J region sequence encoded by IGLJ2 (SEQ ID NO: 129) with insertion of a single amino acid residue between the IGLV3-1 and IGLJ2 sequences, for example methionine.
  • the ⁇ 3 protein framework encoded by IGLV3-1 may contain additional
  • substitutions for example a substitution of cysteine residue at position 33 of SEQ ID NO: 117 (“ACW”) with for example
  • sequences from other exemplary functional ⁇ 3 and genes can be used for grafting mAb6785 light chain paratope residues with the insertion of zero, one, or two amino acid residues between the carboxy- terminus encoded by the ⁇ 3 genes and the amino terminus encoded by the genes, such that the length of the CDR3 region is 11 amino acids.
  • methionine and isoleucine can be inserted between IGLV3-22 (SEQ ID NO: 124) and IGLJ2 (SEq ID NO: 129) .
  • Figure 2A shows alignment of exemplary light chain scaffolds that can be used for grafting.
  • the mAb6785 heavy chain paratope residues can be grafted onto for example a Vh3 framework encoded by IGHV3-23 (SEQ ID NO: 142), that is joined to the J region FR4 sequence (11 C-terminal amino acids e.g.
  • WGQGTLVTVSS of IGHJ1 (SEQ ID NO: 151), with the insertion of about about 5-7 residues, for example 6 residues, constituting HCDR3, between the V and the J regions.
  • the inserted HCDR3 about 5-7 residues include insertion of glutamine, leucine and
  • threonine e.g. 3 of the paratope residues from mAb6785 Vh (Table 10) .
  • Sequences from other exemplary functional Vh3 and Jh genes can be used for grafting mAb6785 heavy chain paratope residues.
  • one C-terminal amino acid from the Vh3 gene may be deleted before insertion of the about 5-7 residues constituting the HCDR3 so that only FR3 sequences are included in the
  • the extended paratope residues of mAb6785 can be used in place of the core paratope residues.
  • the paratope-grafted engineered antibodies can further be modified by substitutions of the Vernier Zone residues (U.S. Pat. No. 6,639,055) or the Affinity Determining Residues (U.S. Pat. Appl. No. 2010/0261620; Cobaugh et al . , J Mol Biol. 378:622- 33, 2008) to improve antibody properties for example affinity.
  • the framework amino acid sequence in the paratope-grafted antibody may be 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% identical to the mAb6785 framework sequences.
  • Allelic variants of the exemplary germline gene frameworks can be used in place of the V and J region protein sequences. Th sequences of the allelic variants are well known and can be obtained at the International ImMunoGeneTics (IMGT) database
  • Sequences from the antigen-binding sites can be grafted in addition to the paratope residues using standard methods. For example, a complete HCDR3 or LCDR3 may be grafted.
  • Another embodiment of the invention is an isolated antibody or fragment that binds specifically to human IL-17A that competes for human IL-17A binding with a monoclonal antibody comprising certain HCDR1, HCDR2 and HCDR3 , and LCDR1, LCDR2 and LCDR3 amino acid sequences.
  • Examplary monoclonal antibodies of the invention are an isolated antibody comprising HCDR1, HCDR2 and HCDR3 amino acid sequences as shown in SEQ ID NOs: 25, 43 and 60 and the LCDR1, LCDR2 and LCDR3 amino acid sequences as shown in SEQ ID NOs: 3, 6 and 18.
  • IL-17A Competition between specific binding to IL-17A can be assayed in vitro using well known methods. For example, binding of MSD Sulfo-TagTM NHS-ester -labeled antibody to IL-17A in the presence of an unlableled antibody can be assessed by ELISA.
  • Another embodiment of the invention is an isolated antibody or antibody or fragment thereof, wherein the antibody binds specifically to human IL-17A having the sequence shown in SEQ ID NO: 105 at amino acid residues 56-68 (SEQ ID NO: 157) and 100-116 (SEQ ID NO: 158); or at residues L26, R55, E57, P59, E60, R61, Y62, S64, V65, W67, R101, E102, P103 and F110.
  • Hydrogen-deuterium (H/D) exchange can be carried out with the antigen and antibody complex to map regions on the antigen that may be bound by the antibody. Segment and point mutagenesis of the antigen can be used to locate amino acids important for antibody binding. Co-crystal structure of antibody-antigen complex is used to identify residues contributing to the epitope and paratope.
  • Another embodiment of the invention is an isolated antibody or fragment thereof, wherein the antibody binds specifically to a P2 pocket cavity on IL-17A, the P2 pocket cavity comprising of amino acid residues V22, V24, L26, 128, Y62, L99, R101, F110, and LI 12 of SEQ ID NO: 105.
  • Co-crystal structure of IL-17A homodimer with the anti-IL- 17A Fab6468 identified a hydrophobic P2 pocket cavity on the surface of the IL-17A homodimer, which is likely to be involved in IL-17RA binding (see Examples) .
  • the "P2 pocket cavity” as used herein refers to a tertiary hydrophobic structural cavity on IL-17A homodimer, where the surface exposed residues in the P2 pocket are V24, L26, 128, Y62, L99, R101, F110 and L112 on monomer A and V22, V24 and L112 on monomer B, and vice versa.
  • Select antibodies of the invention reactive with IL-17A for example Fab6468, have direct contacts with the P2 pocket cavity residues L26, Y62, R101 and F110, which residues are also part of the Fab6468 epitope. While not wishing to be bound by any particular theory, it is assumed that the antibodies of the invention binding the select IL-17A P2 pocket cavity residues block interaction between IL-17A and IL-17RA. Based on co- crystal structure, the phenlylalanine motif (FF) at residues 93 and 94 in a light chain (SEQ ID NO: 79) of Fab6468 blocks the IL- 17A/ IL-17RA interaction, and thus is a P2 pocket cavity blocker. Other P2 pocket cavity blocker antagonists are also within the scope of this invention, such as novel peptides or small
  • inhibiting interaction of human IL-17A with IL-17RA comprising: providing human IL-17A and IL-17RA;
  • Human IL-17A and IL-17RA can be provided as isolated proteins or fusion proteins.
  • Human IL-17A homodimer can be purified from media of activated Thl7 cells prepared by in vitro stimulation of naive CD4 T cells by two anti-CD3/anti-CD28 stimulation in the presence of IL-2, IL-23 and IL- ⁇ .
  • the IL-17RA can be associated with cells or cell membranes, can be native or overexpressed, or can be a fragment of IL-17RA, for example the extracellular domain of the receptor.
  • the IL-17RA can be a human IL-17RA, or IL-17RA from other species such as from mouse, rat or monkey.
  • Antagonists binding to human IL-17A residues V22, V24, L26, 128, Y62, L99, R101, F110, and LI 12 can be identified by the ability of the antagonist to replace Fab6468 binding to IL-17A, by mutagenesis studies or by co-crystal structures.
  • Fusion proteins of human IL-17A and IL-17RA can be made by well known methods.
  • Exemplary fusion protein is a soluble IL-17RA fused to an immunoglobulin Fc domain.
  • Another aspect of the invention is an isolated
  • polynucleotide encoding any of the antibody heavy chains or the antibody light chains or fragments thereof of the invention or their complement. Certain exemplary polynucleotides are
  • polynucleotides which, given the degeneracy of the genetic code or codon preferences in a given expression system, encode the antibody antagonists of the invention are also within the scope of the invention.
  • Exemplary polynuceotides are shown in SEQ ID NOs : 101, 102, 103 and 104.
  • Exemplary antibody antagonists may be antibodies of the IgG, IgD, IgE, IgA or IgM isotypes. Additionally, such antibody antagonists can be post-translationally modified by processes such as glycosylation, isomerization, deglycosylation or non- naturally occurring covalent modification such as the addition of polyethylene glycol (PEG) moieties (pegylation) and lipidation. Such modifications may occur in vivo or in vitro.
  • the antibodies of the invention can be conjugated to polyethylene glycol (PEGylated) to improve their pharmacokinetic profiles. Conjugation can be carried out by techniques known to those skilled in the art. Conjugation of therapeutic antibodies with PEG has been shown to enhance pharmacodynamics while not
  • the "Fc” of an antibody is not involved directly in binding of an antibody to an antigen, but exhibits various effector functions.
  • An antibody “Fc” is a term well known and is defined on the basis of papain cleavage of antibodies.
  • the Fc of an antibody is directly involved in ADCC (antibody-dependent cell-mediated cytotoxicity) and CDC
  • complement and Fc receptor binding sites are well known and include for example L234, L235, D270, N297, E318, K320, K322, P331, and P329 (numbering according to EU index of Rabat) (Brekke et al . , Eur. J. Immunol. 24:2542-7, 1995; US. Pat. Nos. 5,624,821, 7,597,889, Canfield and Morrison, J. Exp. Med. 173:1483-91, 1991) .
  • mutation of Leu234/Leu235 in the hinge region of IgGl to L234A/L235A or Phe235/Leu236 in the hinge region of IgG4 to P235A/L236A minimizes FcR binding and reduces the ability of the immunoglobulin to mediate complement dependent cytotoxicity and ADCC.
  • a Ser to Pro substitution in the Cys-Pro-Ser-Cys (CPSC) motif in the hinge region of IgG4 heavy chains capable of forming either inter- or intra-heavy chain disulfide bonds in vivo via action of isomerases results in "IgGl-like behavior", i.e., the Pro- substituted molecules are unable to form intra-heavy chain disulfide bonds.
  • the location of the CPSC motif is typically found at residue 228 of a mature heavy chain but can change depending on CDR lengths .
  • An exemplary IgGl Fc region having the Leu234/Leu235 residues has an amino acid sequence shown in SEQ ID NO: 114, wherein the residues L117 and L118 correspond to the Leu234/Leu235 residues in the mature heavy chain.
  • An exemplary IgG4 Fc region having the Cys-Pro-Ser-Cys (CPSC) motif and the Leu234/Leu235 residues has an amino acid sequence shown in SEQ ID NO: 115, where the CPSC motif is located at residues 106-109 and the Leu234/Leu235 residues at positions 122 and 123.
  • CPSC Cys-Pro-Ser-Cys
  • Antibodies or fragments thereof of the invention modified to improve stability, selectivity, cross-reactivity, affinity, immunogenicity or other desirable biological or biophysical property are within the scope of the invention.
  • Stability of an antibody is influenced by a number of factors, including (1) core packing of individual domains that affects their intrinsic stability, (2) protein/protein interface interactions that have impact upon the HC and LC pairing, (3) burial of polar and charged residues, (4) H-bonding network for polar and charged residues; and (5) surface charge and polar residue distribution among other intra- and inter-molecular forces (Worn et al . , J. Mol. Biol. 305:989-1010, 2001) .
  • Potential structure including (1) core packing of individual domains that affects their intrinsic stability, (2) protein/protein interface interactions that have impact upon the HC and LC pairing, (3) burial of polar and charged residues, (4) H-bonding network for polar and charged residues; and (5) surface charge and polar residue distribution among other intra- and inter-molecular forces
  • destabilizing residues may be identified based upon the crystal structure of the antibody or by molecular modeling in certain caases, and the effect of the residues on antibody stability can be tested by generating and evaluating variants harboring mutations in the identified residues.
  • One of the ways to increase antibody stability is to raise the thermal transition midpoint (Tm) as measured by differential scanning calorimetry
  • the protein Tm is correlated with its stability and inversely correlated with its susceptibility to unfolding and denaturation in solution and the degradation processes that depend on the tendency of the protein to unfold
  • the antibody antagonists of the invention may bind IL-17A with a K d less than or equal to about 10 "7 , 10 "8 , 10 “9 , 10 "10 , 10 “11 or 10 " M.
  • the affinity of a given molecule for IL-17A, such as an antibody can be determined experimentally using any suitable method. Such methods may utilize Biacore or KinExA
  • Antibody antagonists binding human IL-17A with a desired affinity can be selected from libraries of variants or fragments by techniques including antibody affinity maturation. Antibody antagonists can be identified based on their inhibition of IL-17A biological activity using any suitable method. Such methods may utilize reporter-gene assays or assays measuring cytokine production using well known methods and as described in the application .
  • Another embodiment of the invention is a vector comprising at least one polynucleotide of the invention.
  • Such vectors may be plasmid vectors, viral vectors, vectors for baculovirus expression, transposon based vectors or any other vector suitable for introduction of the polynucleotides of the invention into a given organism or genetic background by any means.
  • polynucleotides of the invention comprising any of the polynucleotides of the invention such as a polynucleotide encoding a polypeptide comprising an
  • immunoglobulin heavy chain variable region having the amino acid sequence shown in SEQ ID NOs : 67-75 and 81-86 or an
  • immunoglobulin light chain variable region having the amino acid sequence shown in SEQ ID NOs: 62-66 and 76-80 or an an
  • Such host cells may be eukaryotic cells, bacterial cells, plant cells or archeal cells.
  • Exemplary eukaryotic cells may be of mammalian, insect, avian or other animal origins.
  • Mammalian eukaryotic cells include immortalized cell lines such as hybridomas or myeloma cell lines such as SP2/0 (American Type Culture Collection
  • Hamster Ovary (CHO) cells such as CHO-K1SV (Lonza Biologies, Walkersville, MD) , CHO-K1 (ATCC CRL-61) or DG44.
  • Another embodiment of the invention is a method of making an antibody reactive with IL-17A comprising culturing a host cell of the invention and recovering the antibody produced by the host cell.
  • Methods of making antibodies and purifying them are well known in the art.
  • the engineered family 2, 6a, 6b, 19a and 19b heavy chain sequences can include an N-terminal leader sequence such as MAWVWTLLFLMAAAQSIQA (SEQ ID NO:109) .
  • nucleotide sequences encoding the heavy chain of candidate mAb6785 (family 19) with a leader sequence and the mature form (without a leader sequence) are shown in SEQ ID NOs: 101 and 102, respectively.
  • the light chain sequences of the family 2, 6a, 6b antibodies of the invention can include an N-terminal leader sequence such as MGVPTQVLGLLLLWLTDARC (SEQ ID NO: 110) and the light chain
  • sequences of the family 19a and 19b antibodies of the invention can include an N-terminal leader sequence such as
  • MAWSPLLLTLLAHCTGSWA (SEQ ID NO: 116) .
  • Exemplary nucleotide sequences encoding the light chain of codon optimized mAb6785 with a leader sequence and the mature form (without a leader sequence) are shown in SEQ ID NOs: 103 and 104, respectively.
  • Another embodiment of the invention is a hybridoma cell line that produces an antibody of the invention.
  • IL-17A antagonists of the invention may be utilized in any therapy where it is desired to reduce the effects of IL-17A in the animal patient.
  • IL- 17A may be circulating in the body or may be present in an undesirably high level localised at a particular site in the body, for example a site of inflammation.
  • the antagonists of the invention provide beneficial therapy by preventing or reducing IL-17A binding to its receptor, or homo- or heterodimerization of IL-17A.
  • the methods of the invention may be used to treat an animal patient belonging to any classification. Examples of such animals include mammals such as humans, rodents, dogs, cats and farm animals.
  • Antibodies of the invention may be useful for the
  • IL-17A mediated conditions such as inflammatory conditions, allergies and allergic conditions, hypersensitivity reactions, autoimmune diseases, severe
  • IL-17A mediated conditions are inflammatory conditions, immune and proliferative disorders, including rheumatoid arthritis (RA) , ankylosing spondylitis, psoriatic arthritis, osteoarthritis, osteoporosis, uveitis, inflammatory fibrosis (e.g., scleroderma, lung fibrosis, and cirrhosis), inflammatory bowel disorders (e.g., Crohn's disease, ulcerative colitis and inflammatory bowel disease) , asthma (including allergic asthma) , allergies, COPD, multiple sclerosis, psoriasis, systemic lupus erythematosus, diabetes and cancer.
  • RA rheumatoid arthritis
  • ankylosing spondylitis psoriatic arthritis
  • osteoarthritis osteoarthritis
  • osteoporosis uveitis
  • inflammatory fibrosis e.g., scleroderma, lung fibrosis,
  • Inflammatory pulmonary condition is an example of an inflammatory condition.
  • Exemplary inflammatory pulmonary conditions include infection-induced pulmonary conditions including those associated with viral, bacterial, fungal, parasite or prion infections; allergen-induced pulmonary
  • pollutant-induced pulmonary conditions such as asbestosis, silicosis, or berylliosis
  • gastric aspiration-induced pulmonary conditions immune dysregulation, inflammatory
  • fibrosis and physical trauma-induced pulmonary conditions, such as ventilator injury.
  • These inflammatory conditions also include asthma, emphysema, bronchitis, chronic obstructive pulmonary disease (COPD) , sarcoidosis, histiocytosis, lymphangiomyomatosis , acute lung injury, acute respiratory distress syndrome, chronic lung disease, bronchopulmonary dysplasia, community-acquired pneumonia, nosocomial pneumonia, ventilator -associated
  • infection- associated inflammatory diseases may include viral or bacterial pneumonia, including severe pneumonia, cystic fibrosis,
  • IL-17A has been shown to regulate neutrophilic inflammation in the lungs - a hallmark of severe asthma as well as COPD - owing to the capacity of IL-17A to induce factors important in neutrophil recruitment, survival and activation from lung resident
  • epithelial cells e.g, IL-6, IL-8, GM-CSF, G-CSF.
  • antibodies aof the present invention suppress IL-6, IL-8, and GM- CSF secretion from lung epithelial cells, and thus may be beneficial in the therapeutic or prophylactic treatment of subjects with pulmonary inflammatory conditions, such as asthma and COPD.
  • pulmonary inflammatory conditions such as asthma and COPD.
  • Commonly used animal models for asthma and airway inflammation include the ovalbumin challenge model and
  • antagonists of the present invention to any of these models can be used to evaluate the use of those antagonists to ameliorate symptoms and alter the course of asthma, airway inflammation, COPD and the like.
  • Psoriasis is another example of an inflammatory condition.
  • Psoriasis is characterized by T cell mediated hyperproliferation of keratinocytes coupled with an inflammatory infiltrate.
  • the inflammation and hyperproliferation of psoriatic tissue is associated with a different histological, antigenic, and cytokine profile than normal skin.
  • cytokines associated with psoriasis are: TNF , IL-19, IL-18, IL-15, IL-12, IL-7, IFNy, IL- 17A and IL-23 (Gudjonsson et al . , Clin. Exp. Immunol. 135:1-8, 2004) .
  • IL-17A has been found overexpressed in psoriatic lesions (US Pat. No. 7,776,540) and positive outcomes in patients treated with anti-human IL-17A therapies have been
  • IL-17A IL-17A signaling may perpetuate inflammation and further tissue damage in the inflamed joint.
  • CIA collagen-induced arthritis
  • mice develop chronic inflammatory arthritis that closely resembles human rheumatoid arthritis.
  • Administration of the IL-17A antibodies of the present invention to the CIA model mice can be used to evaluate the use of these antagonists to ameliorate symptoms and alter the course of diseases.
  • Exemplary gastrointestinal inflammatory conditions are inflammatory bowel disease (IBD), ulcerative colitis (UC) and Crohn's disease (CD), colitis induced by environmental insults
  • gastrointestinal inflammation e.g., colitis
  • a therapeutic regimen such as administration of chemotherapy, radiation therapy, and the like
  • infections colitis ischemic colitis, collagenous or lymphocytic colitis, necrotizing enterocolitis, colitis in conditions such as chronic granulomatous disease or celiac disease, food allergies, gastritis, infectious gastritis or enterocolitis (e.g., Helicobacter pylori-infected chronic active gastritis) and other forms of gastrointestinal inflammation caused by an infectious agent.
  • TNBS TNBS acid/ethanol
  • DSS dextran sulfate sodium
  • Another model involves the adoptive transfer of naive CD45RB hl9h CD4 T cells to RAG or SCID mice.
  • donor naive T cells attack the recipient gut causing chronic bowel inflammation and symptoms similar to human inflammatory bowel diseases (Read and Powrie, Curr. Protoc . Immunol. Chapter 15 unit 15.13, 2001) .
  • the administration of antagonists of the present invention in any of these models can be used to evaluate the potential efficacy of those antagonists to ameliorate symptoms and alter the course of diseases associated with inflammation in the gut, such as inflammatory bowel disease.
  • Renal fibrosis can develop from either an acute insult (ex. graft ischemia/reperfusion) (Freese et al . , Nephrol. Dial.
  • the pathogenesis is typically characterized by an initial inflammatory response followed by sustained fibrogenesis of the glomerular filtration apparatus and tubular interstitium (Liu, Kidney Int. 69:213-7, 2006) .
  • Tubulointerstitial fibrosis has been shown to play a critical role in the pathogenesis of renal injury to end-stage renal failure and the proximal tubule cell has been revealed as a central mediator (Phillips and
  • tubulointerstitial compartment is mediated in part by activation of resident fibroblasts, which secrete pro-inflammatory cytokines that stimulate the proximal tubule epithelium to secrete local inflammatory and fibrogenic mediators. Additionally, chemotactic cytokines are secreted by fibroblasts and epithelial cells and provide a directional gradient guiding the infiltration of monocytes/macrophages and T-cells into the tubulointerstitium.
  • the inflammatory infiltrate produces additional fibrogenic and inflammatory cytokines that further activate fibroblast and epithelial cytokine release while also stimulating the epithelium to undergo a phenotypic transition in which the cells deposit excess extracellular matrix components (Simonson, Kidney Int.
  • IL-17A has been shown to be upregulated during human renal allograft rejection (Van Kooten et al . , J. Am. Soc. Nephrol. 9:1526-34, 1998; Loong et al . , J. Path. 197:322-32, 2002) .
  • IL-17A stimulates the production of the pro-inflammatory mediators IL-6, IL-8, complement component C3, and RANTES by proximal tubular epithelium (Van Kooten et al . , J. Am. Soc.
  • liver fibrosis including but not limited to alcohol-induced cirrhosis, viral-induced cirrhosis, autoimmune-induced hepatitis
  • lung fibrosis including but not limited to scleroderma, idiopathic pulmonary fibrosis
  • kidney fibrosis including but not limited to scleroderma, diabetic nephritis, glomerular nehpritis, lupus nephritis
  • dermal fibrosis including but not limited to scleroderma, hypertrophic and keloid scarring, burns
  • the fibrosis can be organ specific fibrosis or systemic fibrosis.
  • the organ specific fibrosis can be associated with lung fibrosis, liver fibrosis, kidney fibrosis, heart fibrosis, vascular fibrosis, skin fibrosis, eye fibrosis or bone marrow fibrosis.
  • the lung fibrosis can be associated with idiopathic pulmonary fibrosis, drug induced pulmonary fibrosis, asthma, sarcoidosis or chronic obstructive pulmonary disease.
  • the liver fibrosis can be associated with cirrhosis, schistomasomiasis or cholangitis.
  • the cirrhosis can be selected from alcoholic cirrhosis, posthepatitis C cirrhosis, primary biliary cirrhosis.
  • cholangitis can be sclerosing cholangitis.
  • the kidney fibrosis can be associated with diabetic nephropathy or lupus
  • the heart fibrosis can be associated with myocardial infarction.
  • the vascular fibrosis can be associated with postangioplasty arterial restenosis or atherosclerosis.
  • the skin fibrosis can be associated with burn scarring, hypertrophic scarring, keloid, or nephrogenic fibrosing dermatopathy .
  • the eye fibrosis can be associated with retro-orbital fibrosis,
  • the bone marrow fibrosis can be associated with idiopathic
  • the systemic fibrosis can be systemic sclerosis or graft versus host disease.
  • inflammatory conditions and neuropathies which may be prevented or treated by the methods of the invention are those caused by autoimmune diseases.
  • These conditions and neuropathies include multiple sclerosis, systemic lupus erythematous, and neurodegenerative and central nervous system (CNS) disorders including Alzheimer's disease, Parkinson's disease, Huntington's disease, bipolar disorder and Amyotrophic Lateral Sclerosis (ALS), liver diseases including primary biliary cirrhosis, primary sclerosing cholangitis, non-alcoholic fatty liver disease/steatohepatitis, fibrosis, hepatitis C virus (HCV) and hepatitis B virus (HBV) , diabetes and insulin resistance, cardiovascular disorders including atherosclerosis, cerebral hemorrhage, stroke and myocardial infarction, arthritis,
  • CNS central nervous system
  • ALS neurodegenerative and central nervous system
  • liver diseases including primary biliary cirrhosis, primary sclerosing cholangitis, non-alcoholic fatty liver
  • rheumatoid arthritis psoriatic arthritis and juvenile rheumatoid arthritis (JRA) , osteoporosis, osteoarthritis, pancreatitis, fibrosis, encephalitis, psoriasis, Giant cell arteritis,
  • HIV immunodeficiency virus
  • transplant cancer
  • allergies endocrine diseases
  • wound repair other autoimmune disorders
  • airway hyperresponsiveness and cell virus, or prion-mediated infections or disorders.
  • the "therapeutically effective amount" of the agent effective in the treatment of conditions where suppression of IL- 17A activity is desirable can be determined by standard research techniques.
  • the dosage of the agent that will be effective in the treatment of an inflammatory condition such as asthma, Crohn' s Disease, ulcerative colitis or rheumatoid arthritis can be determined by administering the agent to relevant animal models, such as the models described herein.
  • in vitro assays can optionally be employed to help identify optimal dosage ranges. Selection of a particular effective dose can be determined (e.g., via clinical trials) by those skilled in the art based upon the consideration of several factors. Such factors include the disease to be treated or prevented, the symptoms involved, the patient's body mass, the patient's immune status and other factors known by the skilled artisan. The precise dose to be employed in the formulation will also depend on the route of administration, and the severity of disease, and should be decided according to the judgment of the practitioner and each patient's circumstances. Effective doses can be extrapolated from dose-response curves derived from in vitro or animal model test systems.
  • the mode of administration for therapeutic use of the agent of the invention may be any suitable route that delivers the agent to the host.
  • Pharmaceutical compositions of these agents are particularly useful for parenteral administration, e.g., intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous or intranasal .
  • the agent of the invention may be prepared as
  • compositions containing an effective amount of the agent as an active ingredient in a pharmaceutically acceptable carrier.
  • carrier refers to a diluent, adjuvant, excipient, or vehicle with which the active compound is
  • Such pharmaceutical vehicles can be liquids, such as water and oils, including those of petroleum, animal,
  • compositions may contain pharmaceutically acceptable auxiliary substances as required to approximate physiological conditions such as pH adjusting and buffering agents, stabilizing, thickening, lubricating and coloring agents, etc.
  • concentration of the agent of the invention in such pharmaceutical formulation can vary widely, i.e., from less than about 0.5%, usually at or at least about 1% to as much as 15 or 20% by weight and will be selected primarily based on required dose, fluid volumes, viscosities, etc., according to the
  • a pharmaceutical composition of the invention for intramuscular injection could be prepared to contain 1 ml sterile buffered water, and between about 1 ng to about 100 mg, e.g.
  • a pharmaceutical composition of the invention for intravenous infusion could be made up to contain about 250 ml of sterile Ringer's solution, and about 1 mg to about 30 mg and preferably 5 mg to about 25 mg of an antagonist of the invention.
  • compositions are well known and are described in more detail in, for example, "Remington's Pharmaceutical Science", 15th ed., Mack Publishing Company, Easton, PA.
  • the antibody antagonists of the invention can be any antibody antagonists of the invention.
  • the MorphoSys Human Combinatorial Antibody Library (HuCAL®) Gold phage display library (Morphosys AG, Martinsried, Germany) was used as a source of human antibody fragments and was panned in subpools in solution.
  • the sublibraries were selected against biotinylated mature His6- tagged IL-17A A132Q and A70Q variant (IL-17Amut6) (SEQ ID NO: 106) .
  • the amplified output of round 1 was selected against biotinylated His6-tagged IL-17Amut6 in the presence or absence of other IL-17A family members as a
  • the amplified output of round 2 was divided in two pools. The first pool was panned as in round 1. The clones in the second pool were further diversified in either HCDR2 or LCDR3, depending on the sublibrary used in the initial selections, and then carried through 2 additional rounds of panning against IL- 17Amut6 to give a second source of clones for screening.
  • Fabs from clone lysates were captured in ELISA plate wells coated with sheep anti-human Fd antibody and screened for binding to
  • biotinylated IL-17Amut6 Crude lysates of positive clones were screened for inhibition of IL-17Amut6 binding to recombinant human IL-17RA receptor (SEQ ID NO: 107) .
  • the Fabs were tested for their inhibition of IL-17Amut6 and cynoIL-17A binding to recombinant human IL-17RA receptor, and their binding to IL-17Amut6. All tested Fabs inhibited both IL- 17Amut6 and cynoIL-17A binding to the IL-17RA. Affninty of the Fabs to IL-17Amut6 was measured using the SET assay (Table 4) . From the identified Fabs, candidates from families 2, 6a, 6b, 19a and 19b were seleced for further characterization.
  • the selected MOR# Fabs were converted and expressed as mAbs in a human IgGl format, and given corresponding MORmAb
  • the generated MORmAbs were tested for expression and aggregation, their ability to inhibit human and cyno IL-17A binding to human IL-17RA, and IL-8 secretion from NHDF cells.
  • Table 5 shows IC50 values for select assays for the MORmAbs. None of the tested MORmAbs (MORmAb#s 7702, 7708, 7785, 7786, 7706, 7775, 7700, 8095, 8096, 8097, 8098, 7768) cross-reacted with other IL-17 family members.
  • variable regions were further engineered in their variable regions to change certain framework residues to those present in a matching human germline and to change codons to those most frequently occurring in highly expressed mammalian proteins.
  • family 2 VL, L11V and V85T (linear sequence) substitutions were made, converting the framework to an exact match with the VK-1 germline Vb-L5 (IGKV1-12*01) .
  • An exemplary variable region with the V11V and V85T substitutions is the variable region having the amino acid sequence shown in SEQ ID NO: 76.
  • DIE, V59I and T86V substitutions were made (linear sequence) , converting the framework to exact match with the Vk-3 germline Vb-L6 (IGKV3-11*01) .
  • An exemplary variable region with the DIE, V59I and T86V substitutions is the variable region having the amino acid sequence shown in SEQ ID NO: 77.
  • family 6a and 6b VH a G44S substitution was made
  • variable region with the G44S substitution is the variable region having the amino acid sequence shown in SEQ ID NO: 81.
  • amino acids 1-3 DIE
  • Exemplary variable region with the QSV substitution is a variable region having the amino acid sequence shown in SEQ ID NO: 79.
  • family 19a and 19b VH a V5L substitution was made to give a close match the Vh-3 germline Vb 3-23 (IGHV3-23*01. Also, in this process the heavy chain constant region amino acid sequence residues 353-357
  • variable region with the V5L substitution is a variable region having the amino acid sequence shown in SEQ ID NO: 86.
  • substitutions is a heavy chain having the amino acid sequence shown in SEQ ID NO: 100.
  • the engineered antibodies were given mAb numbers.
  • the engineered mAbs were characterized as described above for the MORmAbs.
  • the IC50 values (pM) measured using indicated assays are shown in Table 6. Table 6.
  • Anti-IL-17 antibody inhibits cytokine secretion in NHBE cells
  • IL-17A has been shown to regulate neutrophilic inflammation in the lungs, a hallmark of severe asthma as well as COPD, owing to the capacity of IL-17A to induce factors important in
  • neutrophil recruitment, survival and activation e.g, IL-6, IL-8, GM-CSF
  • Anti-IL-17 antibody inhibits the biological activity of IL-17A/F heterodimer
  • NHDF Normal Human Dermal Fibroblasts
  • rhIL-17A/F heterodimer concentration (1.47 nM) of rhIL-17A/F heterodimer (R&D Systems) was pre-incubated with a dilution series (30 ⁇ g/mL - 0.5 ng/mL) of mAb6785 or control antibodies for 10 minutes at room
  • mAb6785 inhibited IL-17A/F heterodimer -induced IL-6 production by NHDF with EC50 2 ⁇ 2.5 nM.
  • Electrochemiluminescence (ECL) based affinity determination in solution and data evaluation were basically performed as described previously (Haenel et al . , Anal Biochem 339:182-4, 2005) .
  • a defined fixed concentration of purified Fab (10-100 pM) was incubated with increasing concentrations of IL-17Amut6 (highest concentration of 5 nM) in solution until chemical equilibrium was achieved.
  • the signal was detected with 100 ⁇ of 1:10,000 dilution of 1 mg/ml SA-HRP (Jackson Immunoresearch, West Grove, PA) for 20 minutes at room temperature (RT) followed by 100 ⁇ /well of OPD substrate (Sigma-Aldrich Corp., St. Louis, MO) .
  • the plates were read at 492 nm (Envision, PerkinElmer, Waltham, MA) .
  • Fab binding to IL- 17RA was tested as described for mAbs.
  • NHDF NHDF
  • IL-17mut6 samples with no added antibody were included as control samples, while samples consisting of TNF-a or culture medium only were included as negative controls. Cells were incubated for 24 hours
  • biotinylated cytokine was prepared at 2x final concentration. 100 ⁇ of cytokines at 2x final concentration were mixed (30-0 ⁇ g/ml final concentration) with 100 ⁇ of biotinylated IL-17mut6 at 2x final concentration (25 ng/ml final concentration) in assay buffer. Recombinant human IL-23 (R&D Systems, Minneapolis, MN) was used as a negative control, buffer only sample as background control, and IL-17mut6 as positive control. 100 ⁇ per well in duplicate of cytokine/biotinylated IL-17mut6 mixture was added to the plate and incubated for 1-2 hours.
  • Affinity measurements using Surface Plasmon Resonance (SPR) were performed using a Biacore 3000 optical biosensor (Biacore) .
  • Selected Fabs (-30 RU) or mAbs (-50 RU) were captured onto the sensor chip surface using a sheep anti-Fd antibody or an anti- human Fc antibody for Fab or mAb capture, respectively.
  • Capture of Fab or mAb was followed by injection of huIL-17mut6 or cyno IL-17A in solution (0.2 to 49 nM) .
  • Antibody epitopes were deduced by a combination of
  • mAbl926, MORmAb7700, MORmAb7706, MORmAb7708, mAb7357 (a mouse anti-human IL-17A neutralizing antibody derived from hybridoma C1863)
  • mAb2832 a mouse/human chimeric anti-human IL- 17A neutralizing antibody derived from hybridoma C1861
  • mAb317 mouse anti- human IL-17A antibody, R&D Systems, Minneapolis, MN
  • mAb3171 mouse anti-human IL-17A antibody, R&D Systems, Minneapolis, MN
  • mAbeBI016-7178 (a mouse anti-human IL-17A antibody, e-Bioscience , San Diego, CA) .
  • the three commercial antibodies showed varying degrees of neutralizing activity.
  • IL-17Amut6 protein For competitive ELISA, 5 ⁇ (20 g/ml) of IL-17Amut6 protein was coated on MSD HighBind plate (Meso Scale Discovery, Gaithersburg, MD) per well for 2 hr at room temperature. 150 ⁇ of 5% MSD Blocker A buffer (Meso Scale Discovery, Gaithersburg, MD) was added to each well and incubated for 2 hr at room temperature. Plates were washed with 0.1 M HEPES buffer (pH 7.4) . Labeled antibody (MDS fluorescence dye), 10 nM, was incubated with increasing concentrations of competitor antibodies (1 nM - 2 ⁇ ) , and 25 ⁇ of the mixture was added to the
  • Bin A mAbl926, MORmAb7706, and MORmAb7708
  • Bin B eBiol6-7178 and mAb7357
  • Bin C mAb317
  • Bin D mAb3171.
  • the back-exchanged IL-17Amut6 protein was eluted from the column and localization of deuterium containing fragments was determined by protease digestion and mass spec analysis. Regions bound to the antibody were inferred to be those sites relatively protected from exchange and thus containing a higher fraction of deuterium, compared to IL-17Amut6 not complexed with antibody.
  • H/D exchange perturbation maps of IL-17Amut6 are shown in Figure 5. The numbers on top of the bars refer to IL-17Amut6 resiudes.
  • MORmAb7700, MORmAb7706 and MORmAb7708 showed varying degrees of differential exchange for three segments of IL-17A (SEQ ID NO: 105) 45 NRS TS PWNLH 54 (SEQ ID NO: 159), 56 NEDPERYPSVIWE 68 (SEQ ID NO: 157) and i 00 RREPPHCPNSFRLEKILii 6 (SEQ ID NO: 158), indicating protection by the antibodies.
  • the 56 NEDPERYPSVIWE 68 (SEQ ID NO: 157) fragment was strongly protected by MORmAb7708, weakly protected by MORmAb7700, and not protected by MORmAb7706. The overlap in the fragment protection patterns of these
  • Bin A antibodies (MORmAb7700, MORmAb7706 and MORmAb7708) bound in the region of peptide segments 45 NRS TS PWNLH 54 , (SEQ ID NO: 159), 56 NEDPERYPSVIWE 68 (SEQ ID NO: 157) and i 00 RREPPHCPNSFRLEKILii 6 (SEQ ID NO: 158), of SEQ ID NO: 105, and Bin B antibodies (mAb7357 and mAbeBiol 6-7178 ) bound in region of peptide segment
  • the IL-17A/Fab6468 complex was prepared by mixing IL- 17Amut6 and Fab6468 in 1:1.1 molar ratio in 20 mM MES pH 6.5, 0.2 M NaCl, and 10% glycerol and incubated over night at 4°C.
  • the complex was purified from excess un-complexed Fab using size exclusion chromatography (SEC) on a Superdex 200 10/300 GL column (GE Healthcare, Piscataway, NJ) in 20 mM MES pH 6.5, 0.2 M NaCl, and 10% glycerol. Fractions corresponding to the complex were pooled and concentrated with an Amicon Ultra 10000 MWCO device to 4.6 mg/ml .
  • MMS microseed-matrix screening
  • Crystallographica Section D 63:550-4, 2007
  • Diffraction quality crystals were obtained from the MMS screen in 0.1 M Sodium Acetate pH 5.5, 12% PEG MME 5000 and 0.2 M Lithium
  • the crystal was soaked for a few seconds in the mother liquor supplemented with 24% glycerol, and flash frozen in the stream of nitrogen at 95°K.
  • diffraction data were collected and processed using a Rigaku MicroMaxTM-007HF microfocus X-ray generator equipped with an OsmicTM VariMaxTM confocal optics, Saturn 944 CCD detector, and an X-streamTM 2000 cryocooling system (Rigaku, Woodlands, TX) .
  • the crystal structure of IL-17A/Fab6468 was determined by molecular replacement using Phaser (Read, Acta Crystallogr D Biol Crystallogr, 57:1373-82, 2001) .
  • the search models were IL-17F (PDB ID 1JPY) (Hymowitz et al . , EMBO J., 20:5332-41, 2001) and a homology model for the Fv (VH/VL) , which was constructed based upon the anti-IL-13 antibody CNTO607 (PDB ID 3G6A) (Teplyakov et al., J. Mol. Biol. 389:115-23, 2009) for both the VH and VL, using Modeller (Accelrys, CA) .
  • the two constant domains CL/CH1 were taken from PDB ID 8FAB (Strong et al . , Biochemistry,
  • the structure of the complex was determined to high
  • IL-17A was a nearly symmetrical homo-dimer in the crystal and bound two Fab molecules .
  • the antibody-antigen interactions were largely hydrophobic and in contrast to most antibodies, the light chain CDRs made a number of the important contacts.
  • the overall molecular structure of IL-17A/Fab6468 complex is shown in Figure 6A.
  • the monomer of the IL-17A dimer adopted the overall topology of a cystine knot ( Figure 6B) .
  • the two monomers were very similar with a C RMSD of 0.54 A for 77 backbone Ca atoms.
  • IL-17A monomer cystine knot was very similar to that of IL-17F with an rmsd of 0.71 A for 76 Ca atoms (Figure 6B) .
  • Each IL-17A monomer was stabilized by three disulfide bonds.
  • chain B three intra- chain disulfide bonds were observed (C 10 -C 106 , C 71 -C 121 , C 76 -C 123 ) , whereas for chain A the C 10 -C 106 disulfide bond was not observed due to disorder in these segments of the monomer.
  • the latter two disulfide bonds stabilized the cystine knot architecture, analogous to IL-17F and NGF.
  • the structural model for chain B of IL-17A included all residues 10-128 (residues 1-9 were disordered) , whereas for chain A residues only residues 21- 29, 41-104 and 109-127 were observed and the other residues 1-20, 30-40, 105-108 and 128 were missing due to disorder in the structure.
  • residues 1-2 of both the light chains were disordered or had poor electron density.
  • the C- terminal 3 residues of both the heavy and light chains, including the inter-chain disulfide bonds as well as the His tag on the heavy chain were disordered.
  • the two N-terminal segments of IL-17A formed two interlocked monomers, which also gave rise to an apparent dimer of 26 kD on non-reducing SDS-PAGE.
  • the dimer of the IL-17A was nearly symmetrical for the four main ⁇ -strands (strands 1-4) ( Figure 6C) .
  • the C rmsd for 76 residues is 0.71 A.
  • the slight asymmetry came from two sources. First, the chain A contained a number of disordered segments, mainly in N-terminus . Only a short ⁇ -strand (strand 0, residues 22-26) was apparently ordered, whereas residues 10-40 of chain B were ordered with a helical segment (residues 12-16) and a ⁇ - strand (strand 0, residues 21-25) .
  • the contact residues from the antibodies in the two sites were not all identical.
  • the residues involved in identical contacts to the core epitope residues are referred to as the "core paratope", which was composed of the following residues: Light Chain (LC) : Y31, D49, Y90, F92, F93 (SEQ ID NO: 79); and heavy chain (HC) : S52, T54, F57, Y59, Q99, L100 and T101 (SEQ ID NO: 86) (Table 10) .
  • the core paratope residues are shown in bold in Table 10.
  • the additional "extended paratope" residues identified in one monomer binding a specific IL-17A residue are shown in parenthesis.
  • H/D protection data for the MORmAb7700 was in agreement with the co-crystal studies, as all core epitope residues identified in the co-crystal structure except L26 were within or at the borders of two of the protected segments identified by H/D exchange, 56 NEDPERYPSVIWE 68 (SEQ ID NO: 157) and
  • RREPPHCPNSFRLEKIL 116 (SEQ ID NO: 158) for the MORmAb7700. All of the MORmAb7700 antibody derivatives, including MORmAb8302 and mAbl926, are assumed to have the same binding specificity as Fab6468 since they differ at most by one residue in the N- terminal region of VH (see Example 1), 3 residues at the N- terminus of VL (see Example 1) , and 3 CDR residues (one each in H2 , H3 and L3, Table la) , none of which are part of the antibody paratope .
  • the IL-17A structure characterized in this invention is very similar to the previously published structure, except that due to missing segments, the P2 pocket cavity (see below) was not identified in the previous work (structure 2VXS, available at the Protein DataBank http //www rcsb org/pdb/home/home do; Gerhardt et al., J. Mol. Biol. 394:905-21, 2009) .
  • Fab6468 would inhibit IL-17A function by blocking its interactions with IL-17RA and by analogy, IL-17RC, though the mode of interaction between IL-17RC and IL-17A is not known at the molecular level.
  • the PI pocket which is analogous to a pocket first discovered in IL-17F (Hymowitz et al . , EMBO J, 20: 5332-41, 2001), is composed of residues Q94, E95, L97 and K114 of monomer A and L53, Y62, P63, V65, 166, W67, 196, V117 and V119 of monomer B, and vice versa.
  • the PI pocket On one side of the dimer, the PI pocket is partially covered by the segment 30-40, whereas on the other side it was completely open due to the segment being disordered.
  • the P2 pocket is also composed of residues from both chains: V24, L26, 128, Y62, L99, R101, F110 and LI 12 of monomer A and V22, V24 and L112 of monomer B, and vice versa.
  • the FF motif of IL-17F is likely a structural discriminant for human IL-17A and IL-17F interactions with receptors IL-17RA and IL-17RC. It is likely that both of these largely hydrophobic pockets (PI and P2 ) are required for IL-17A binding to IL-17RA.
  • the recent crystal structure of IL-17F/IL- 17RA complex shows that the FF motif is displaced by IL-17RA (Ely et al . , Nat. Immunology 10:1245-51, 2009) .
  • IL-17A and IL-17F provide a basis for dissecting their interactions with respective receptors. Furthermore, it is conceivable that peptides, peptidomimetics and small molecules can be designed to bind in either or both pockets to block IL-17A and/or IL-17F from interacting with their receptors. Since the FF motif present in Fab6468 (residues F92 and F93 in SEQ ID NO: 79) binds P2 pocket residues L26, R61, L99, R101 and R102, the Fab 6468 structure could be used to select and optimize additional IL-17A
  • antagonists such as peptides from randomized or designed peptide libraries using phage display.
  • residues lining the PI and P2 pockets are well
  • IL-17A/F heterodimer would adopt a nearly identical overall structure when compared to the IL-17A homodimer alone. Therfore, it is likely that the PI and P2 pockets are present in the IL-17A/F heterodimer with similar overall topology and constitute its receptor binding sites.
  • IL-17A is likely that the PI and P2 pockets are present in the IL-17A/F heterodimer with similar overall topology and constitute its receptor binding sites.
  • mAbtrl926 To evaluate cross-species binding specificity of mAbtrl926, a binding ELISA was performed with different IL-17A proteins coated on micro-titer plates. Human, mouse and rat IL-17A proteins were coated on the micro-titer plates. Serial dilutions of labeled mAbl926 were incubated at 37°C for 2 hours. Following incubation, micro-titer plates were washed thoroughly, and bound labeled mAbl926 was detected. mAb 1926 bound to human IL-17A much stronger than to rat or mouse IL-17A proteins ( Figure 9) .

Abstract

Interleukin-17A (IL-17A) antibody antagonists, polynucleotides encoding IL-17A antibody antagonists or fragments thereof, and methods of making and using the foregoing are disclosed.

Description

IL-17A Antagonists
Field of the Invention
The present invention relates to interleukin-17A (IL-17A) antibody antagonists, polynucleotides encoding IL-17A antibody antagonists or fragments thereof, and methods of making and using the foregoing.
Background of the Invention
Interleukin-17A (IL-17A, CTLA-8, IL-17) is a cytokine secreted by activated Thl7 cells, CD8+ T cells, γδ T cells and NK cells in response to cytokines such as IL-23 and TGF-β, and regulates production of mediators such as antimicrobial peptides (defensins), proinflamatory cytokines and chemokines from multiple cell types such as fibroblasts and synoviocytes that are involved in neutrophil biology, inflammation, organ destruction and host defense (reviewed in Weaver et al . , Annu. Rev. Immunol. 25:821-52, 2007; Aggarwal et al . , J. Biol. Chem. 278:1910-4, 2003; Mangan et al . , Nature 441:231-4, 2006) . IL-17A synergizes with other cytokines, such as TNF-a and IL-Ιβ to potentiate the pro-inflammatory environment.
The IL-17A cytokine family consists of six homologs designated IL-17A, B, C, D, E and F, each with divergent and distinct biological roles (Kawaguchi et al . , J. Allergy Clin. Immunol. 114:1265-73, 2004; Rolls and Linden, Immunity 21:467-76, 2004; Moseley et al . , Cytokine Growth Factor Rev. 14:155-74, 2003) . Of the family members, IL-17F is most homologous to IL- 17A and shares many similar functional properties such as induction of neutrophilia in the lung and induction of proinflammatory cytokines; however, in man, IL-17F is about 10-fold less potent than IL-17A (Moseley et al . , Cytokine Growth Factor Rev. 14:155-74, 2003; Rolls et al . , Immunity, 21: 467-76, 2004; McAllister et al . , J. Immunol. 175:404-12, 2005) . IL-17A and IL- 17F can also form heterodimers , which have intermediate bioactivity in vitro (Wright et al . , J. Biol. Chem. 282:13447-55, 2007) .
IL-17A mediates it effects by interacting with the
Interleukin-17 receptor A (IL-17RA) and receptor C (IL-17RC) (Moseley et al . , Cytokine Growth Factor Rev. 14:155-74, 2003; Toy et al . , J. Immunol. 177:36-9, 2006) . IL-17F signals through the same receptors, although IL-17F affinity to the receptors is significantly lower (Kuestner et al . , J. Immunol. 179:5462-73,
2007) . Crystal structures of human IL-17F and human IL-17F/IL- 17RA complex identified a putative receptor-binding cavity in the IL-17F homodimer (Hymowitz et al . , EMBO J. 20:5332-41, 2001; Ely et al . , Nat. Immunology 10:1245-51, 2009) . A similar cavity was identified in the crystal structure of human IL-17A in complex with a neutralizing Fab, although the cavity was partially occupied (Gerhardt et al . , J. Mol. Biol. 394:905-21, 2009) .
Inappropriate or excessive production of IL-17A is
associated with the pathology of various diseases and disorders, including rheumatoid arthritis (Lubberts, Cytokine 41:84-91,
2008) , airway hypersensitivity including allergic airway disease such as asthma (reviewed in Linden, Curr. Opin. Investig. Drugs. 4:1304-12, 2003; Ivanov, Trends Pharmacol. Sci. 30:95-103, 2009), psoriasis (Johansen et al., Br. J. Dermatol. 160:319-24, 2009), dermal hypersensitivity including atopic dermatitis (Toda et al . , J. Allergy Clin. Immunol. 111:875-81, 2003), systemic sclerosis (Fujimoto et al . , J. Dermatolog. Sci. 50:240-42, 2008),
inflammatory bowel diseases including ulcerative colitis and Crohn's disease (Holtta et al., Inflamm. Bowel Dis. 14:1175-84, 2008; Zhang et al . , Inflamm. Bowel Dis. 12:382-88, 2006), and pulmonary diseases including chronic obstructive pulmonary disease (Curtis et al., Proc. Am. Thorac . Soc. 4:512-21, 2007) .
Antibodies to IL-17A have been proposed for use in the treatment of IL-17A mediated diseases and disorders (PCT Publ . Nos: WO08/021156, WO07/070750, WO07/149032, WO06/054059,
WO06/013107, WO08/001063, WO10/034443; US Pat. Appl . Nos.
US2008/095775, US2009/0175881; ) . As the pharmacokinetic, efficacy and safety profiles of antibody therapeutics will be dependent on specific compositions, there is a need for improved antibodies to human IL-17A that are suitable for use in the treatment of IL-17A mediated diseases and disorders.
Brief Description of the Drawings
Fig. 1. A-H. CDR sequences of Family 2, 6a, 6b, 19a, and 19b IL-17A antibody antagonists.
Fig. 2. Exemplary A) IGLV3 and IGLJ; and B) IGHV3 and IGHJ germline genes as scaffolds for grafting paratope residues. mAb6785 sequence is shown above. CDR regions are underlined and core contact sites in mAb6785 light chain (Y31, D49, Y90, F92, F93) and heavy chain (S52, T54, F57, Y59, Q99, L100 and T101) are denoted by an asterix "*". Framework 4 regions in B) are double underlined. Sequence shown are *01 alleles unless specifically
indicated otherwise.
Fig. 3. Rabat and Chotia numbering for select antibody A) light and B) heavy chains. Locations of Rabat CDRs and Chothia HVs are highlighted in gray.
Fig. 4. Competitive binding assays of labeled A) and B) mAbl926; C) mAb317; D) mAb3171; E) and F) mAb7357 with IL-17A in an ELI SA format.
Fig. 5. A) H/D exchange maps of the IL-17A complexed with different anti-IL-17A mAbs . Numbering above the protective blocks corresponds to mature IL-17A (SEQ ID NO: 105) sequence numbering .
Fig. 6. A) The overall molecular structure of IL-17A/ Fab6468 complex. The dimer of IL-17A is shown dark gray and light gray. The two Fab molecules are shown in dark gray and light gray, repectively; B) Comparison of monomer of IL-17A (light gray) and IL-17F (dark gray) ; C) Dimer of IL-17A (light and dark gray) ; D) Dimer of IL-17F (light and dark gray) . Fig. 7. The two binding sites and the core epitope on IL- 17A for Fab6468. Protomers 1 and 2 are dark and light gray, respectively. The core epitope is indicated by the black oval. The broken line represents disordered residues.
Fig. 8. Comparison of IL-17A and IL-17F putative receptor binding pockets. A) Front and Back views of the the PI and P2 pockets of IL-17A. The FF motif of mAb6468 light chain CDR3 is shown in the P2 pocket. B) IL-17F with N-terminal FF motif in the P2 pocket. C) Sequence alignment of IL-17A and IL-17F and the conservation of PI and P2 pockets.
Fig. 9. Binding specificity of mAbl926 to different species of IL-17A proteins in an ELISA format.
Summary of the invention
One aspect of the invention is an isolated antibody or fragment thereof, wherein the antibody binds specifically to human IL-17A having the sequence shown in SEQ ID NO: 105 at amino acid residues 56-68 (SEQ ID NO: 157) and 100-116 (SEQ ID NO:
158); or at residues L26, R55, E57, P59, E60, R61, Y62, S64, V65, W67, R101, E102, P103 and F110.
Another aspect of the invention is an isolated antibody or fragment thereof, wherein the antibody binds specifically to a P2 pocket cavity on human IL-17A, the P2 pocket cavity comprising amino acid residues V22, V24, L26, 128, Y62, L99, R101, F110, and LI 12 of SEQ ID NO: 105.
Another aspect of the invention is an isolated antibody or fragment that binds specifically to human IL-17A that competes for human IL-17A binding with a monoclonal antibody comprising the amino acid sequences of certain heavy chain complementarity determining regions (CDR) 1, 2 and 3 (HCDR1, HCDR2, HCDR3) , the amino acid sequences of certain light chain complementarity determining reigons (CDR) 1, 2 and 3 (LCDR1, LCDR2, LCDR3) , the amino acid sequences of certain heavy chain variable regions (VH) or the amino acid sequences of certain light chain variable regions (VL) .
Another aspect of the invention is an isolated antibody or fragment that binds specifically to human IL-17A, comprising certain heavy chain variable region paratope amino acid residues and certain light chain variable region paratope amino acid residues that interact with certain residues of human IL-17A having the amino acid sequence shown in SEQ ID NO: 105.
Another aspect of the invention is an isolated antibody or fragment that binds specifically to human IL-17A, comprising a heavy chain variable region and a light chain variable region, wherein the antibody comprises a heavy chain variable region paratope selected from Chothia residues F56 and Y58; and a light chain variable region paratope selected from Chothia residues Y91, F93 and F94.
Another aspect of the invention is an isolated antibody or fragment that binds specifically human IL-17A, comprising a heavy chain variable region (VH) and a light chain variable region (VL) , wherein the antibody comprises the amino acid sequences of certain heavy chain complementarity determining regions (CDR) 1,
2 and 3 (HCDR1 , HCDR2, HCDR3), the amino acid sequences of certain light chain complementarity determining reigons (CDR) 1,
2 and 3 (LCDR1 , LCDR2, LCDR3), the amino acid sequences of certain heavy chain variable regions (VH) or the amino acid sequences of certain light chain variable regions (VL) .
Another aspect of the invention is an isolated antibody or fragment that specifically binds human IL-17A, wherein the antibody comprises the amino acid sequences of certain heavy chains and the amino acid sequences of certain light chains.
Another aspect of the invention is a pharmaceutical composition comprising the isolated antibody or fragment of the invention and a pharmaceutically acceptable carrier.
Another aspect of the invention is an isolated antibody heavy chain comprising the amino acid sequence shown in SEQ ID NOs : 67, 68, 69, 81, 82, 83, 84, 85, 86, 92, 93, 94, 95, 96, 97, 98, 99, or 100.
Another aspect of the invention is an isolated antibody light chain comprising the amino acid sequence shown in SEQ ID NOs: 76, 77, 78, 79, 80, 87, 88, 89, 90, or 91.
Another aspect of the invention is an isolated
polynucleotide encoding an antibody heavy chain comprising the amino acid sequence shown in SEQ ID NO: 67, 68, 69, 81, 82, 83, 84, 85, 86, 92, 93, 94, 95, 96, 97, 98, 99, or 100.
Another aspect of the invention is an isolated
polynucleotide encoding an antibody light chain comprising the amino acid sequence shown in SEQ ID NO: 76, 77, 78, 79, 80, 87, 88, 89, 90, or 91.
Another aspect of the invention is a vector comprising at least one polynucleotide of the invention.
Another aspect of the invention is a host cell comprising the vector of the invention.
Another aspect of the invention os a method of inhibiting interaction of human IL-17A with IL-17RA comprising: providing a human IL-17A and IL-17RA; and
contacting the human IL-17A with an antagonist that binds the human IL-17A at at least one amino acid residue selected from the group consisting of V22, V24, L26, 128, Y62, L99, R101, F110, and L112.
Another aspecf of the invention si a method of inhibiting IL-17A biological activity, comprising: providing a human IL17-A and IL-17RA; and contacting the human IL-17A with an antagonist that binds the human IL-17A at at least one amino acid residue selected from the group consisting of V22, V24, L26, 128, Y62, L99, R101, F110, and L112.
Another aspect of the invention is a method of treating an inflammatory condition comprising administering a therapeutically effective amount of the isolated antibody of claim 3 or 7 to a patient in need thereof for a time sufficient to treat the inflammatory condition. Detailed Description of the Invention
All publications, including but not limited to patents and patent applications, cited in this specification are herein incorporated by reference as though fully set forth.
It is to be understood that the terminology used herein is for the purpose of describing particular embodiments only and is not intended to be limiting. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which the invention pertains .
Although any methods and materials similar or equivalent to those described herein can be used in the practice for testing of the present invention, exemplary materials and methods are described herein. In describing and claming the present
invention, the following terminology will be used.
The term "antagonist" as used herein means a molecule that partially or completely inhibits, by any mechanism, IL-17A activity. Exemplary antagonists are antibodies, fusion proteins, peptides, peptidomimetics , nucleic acids, oligonucleotides and small molecules. The agent can be identified using well known assays for IL-17A activity described below.
The term "IL-17A antibody antagonist" or an "antibody reactive with IL-17A" as used herein refers to an antibody that is capable of, directly or indirectly, reducing or inhibiting IL- 17A biological activity, blocking binding of IL-17A to its receptor, or inhibiting IL-17A receptor activation. For example, an antibody reactive with IL-17A can bind directly to IL-17A and neutralize IL-17A activity, i.e, block IL-17A signaling to reduce cytokine and chemokine release.
The term "IL-17A" (CTLA-8, IL-17, interleukin- 17A) refers to a human IL-17A polypeptide having an amino acid sequence shown in GenBank Acc . No. NP_002181. SEQ ID NO: 105 shows the amino acid sequence of the mature human IL-17A. IL-17A in vivo forms homodimers of two monomers, which are designated monomer A and monomer B, or protomer A and protomer B, or protomer 1 and protomer 2, or chain A and chain B. IL-17A can also form a heterodimer with IL-17F. The term "IL-17A" comprises the monomer, the homodimer, and the heterodimer forms. The term "IL 17Amut6" refers to a variant of IL-17A having A70Q and A132Q substitutions. The amino acid sequence of the mature IL-17Amut6 is shown in SEQ ID NO: 106, and the cDNA sequence in SEQ ID NO: 112. IL-17A and IL-17Amut6 have comparable activities (PCT. Pat Appl. No. WO09/003096) .
The term "IL-17A receptor" as used herein comprises both receptor polypeptides, IL-17RA (GenBank Acc no: NP_055154, SEQ I NO: 107) and IL-17RC (GenBank Acc No NP_703191, SEQ ID NO: 113), and homodimers or heterodimers of the two polypeptides.
The term "antibodies" as used herein is meant in a broad sense and includes immunoglobulin molecules including polyclonal antibodies, monoclonal antibodies including murine, human, human adapted, humanized and chimeric monoclonal antibodies, antibody fragments, multispecific antibodies formed from at least two intact antibodies, dimeric, tetrameric or multimeric antibodies.
The term "monoclonal antibody" (mAb) as used herein means an antibody (or antibody fragment) obtained from a population of substantially homogeneous antibodies . Monoclonal antibodies are highly specific, typically being directed against a single epitope. The modifier "monoclonal" indicates the substantially homogeneous character of the antibody and does not require production of the antibody by any particular method.
Immunoglobulins can be assigned to five major classes, namely IgA, IgD, IgE, IgG and IgM, depending on the heavy chain constant domain amino acid sequence. IgA and IgG are further sub-classified as the isotypes IgAx, IgA2, IgGi, IgG2, IgG3 and IgG4. Antibody light chains of any vertebrate species can be assigned to one of two clearly distinct types, namely kappa (κ) and lambda (λ) , based on the amino acid sequences of their constant domains . The term "antibody fragments" comprise at least a portion of an immunoglobulin molecule, such as a heavy chain
complementarity determining region (HCDR) , a light chain
complementarity determining region (LCDR) , a heavy chain variable region (VH) , a light chain variable region (VL) , a heavy chain constant region (CH) , a light chain constant region (CL) , or a framework region (FR) from either antibody heavy or light chain. An antibody may be a Fab, F(ab'), F(ah')2, scFv, dsFv, or diabody. Structures of the above mentioned antibody fragments, and techniques for the preparation and use of the antibodies and fragments thereof are well known in the art.
An antibody variable region consists of a "framework" region interrupted by three "antigen-binding sites". The antigen-binding sites are defined using various terms: (i)
Complementarity Determining Regions (CDRs) , three in the VH (HCDR1, HCDR2, HCDR3), and three in the VL (LCDR1, LCDR2, LCDR3), are based on sequence variability (Wu and Rabat, J. Exp. Med. 132:211-250, 1970; Rabat et al . , Sequences of Proteins of
Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md., 1991) . (ii) "Hypervariable regions", "HVR" , or "HV", three in the VH (HI, H2, H3) and three in the VL (LI, L2, L3) , refer to the regions of an antibody variable domains which are hypervariable in structure as defined by Chothia and Lesk (Chothia and Lesk, Mol . Biol. 196:901-917, 1987) . Other terms include "IMGT-CDRs" (Lefranc et al . , Dev. Comparat . Immunol. 27:55-77, 2003) and "Specificity Determining Residue Usage" (SDRU) (Almagro, Mol. Recognit. 17:132-143, 2004) . The International ImMunoGeneTics (IMGT) database
(http://www imgt org) provides a standardized numbering and definition of antigen-binding sites. The correspondence between CDRs, HVs and IMGT delineations is described in Lefranc et al . , Dev. Comparat. Immunol. 27:55-77, 2003.
"Chothia residues" as used herein are the antibody VL and VH residues numbered according to Al-Lazikani (Al-Lazikani et al . , J. Mol. Biol. 273:927-48, 1997) . Correspondence between the two most used numbering systems, Rabat (Rabat et al.,
Sequences of Immunological Interest, 5th Ed. Public Health
Service, NIH, Bethesda, MD, 1991) and Chothia (Chothia and Lesk, Mol. Biol. 196:901-17, 1987) in relation to sequential
polypeptide numbering is shown in Figure 3 for exemplary
antibodies of the invention.
"Framework" or "framework sequences" are the remaining sequences of a variable region other than those defined to be antigen-binding site. Because the antigen-binding site can be defined by various terms as described above, the exact amino acid sequence of a framework depends on how the antigen-binding site was defined.
The term "substantially identical" as used herein means that the two antibody or antibody fragment amino acid sequences being compared are identical or have "insubstantial differences." Insubstantial differences are substitutions of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 amino acids in an antibody or antibody fragment amino acid sequence that do not adversely affect antibody properties. Amino acid sequences substantially identical to the sequences disclosed herein are also part of this application. In some embodiments, the sequence identity can be about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or higher. Percent identity can be determined for example by pairwise alignment using the default settings of the AlignX module of Vector NTI v.9.0.0 (Invitrogen, Carslbad, CA) . The protein sequences of the present invention can be used as a query sequence to perform a search against public or patent databases to, for example, identify related sequences. Exemplary programs used to perform such searches are the XBLAST or BLASTP programs (http //www ncbi nlm/nih gov), or the GenomeQuest™ (GenomeQuest , Westborough, MA) suite using the default settings.
The term "in combination with" as used herein means that the described agents can be administered to an animal together in a mixture, concurrently as single agents or sequentially as single agents in any order.
The term "inflammatory condition" as used herein refers to acute or chronic localized or systemic responses to harmful stimuli, such as pathogens, damaged cells, physical injury or irritants, that are mediated in part by the activity of
cytokines, chemokines, or inflammatory cells (e.g., neutrophils, monocytes, lymphocytes, macrophages) and is characterized in most instances by pain, redness, swelling, and impairment of tissue function .
The term "IL-17A-mediated inflammatory condition" as used herein refers to an inflammatory condition resulting at least partially from IL-17A biological activity, or caused by IL-17A activity. Exemplary IL-17A-mediated inflammatory conditions are psoriasis and rheumatoid arthritis.
The term "IL-17A-mediated condition" as used herein encompasses all diseases and medical conditions in which IL-17A 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.
The term "epitope" as used herein means a portion of an antigen to which an antibody specifically binds. Epitopes usually consist of chemically active (such as polar, non-polar or hydrophobic) surface groupings of moieties such as amino acids or polysaccharide side chains and can have specific three- dimensional structural characteristics, as well as specific charge characteristics. An epitope can be composed of either or both contiguous or discontiguous amino acids that form a
conformational spatial unit. For a discontiguous epitope, amino acids from differing portions of the linear sequence of the antigen come in close proximity in 3-dimensional space through the folding of the protein molecule.
The term "paratope" as used herein means a portion of an antibody to which an antigen specifically binds. A paratope can be linear in nature or can be discontinuous, formed by a spatial relationship between non-contiguous amino acids of an antibody rather than a linear series of amino acids. A "light chain paratope" and a "heavy chain paratope" or "light chain paratope amino acid residues" and "heavy chain paratope amino acid residues" refer to antibody light chain and heavy chain residues in contact with an antigen, respectively.
The term "specific binding" as used herein refers to antibody binding to a predetermined antigen with greater affinity than for other antigens or proteins. Typically, the antibody binds with a dissociation constant (KD) of 10~7 M or less, and binds to the predetermined antigen with a KD that is at least ten fold less than its KD for binding to a non-specific antigen (e.g., BSA, casein, or any other specified polypeptide) other than the predetermined antigen. The phrases "an antibody recognizing an antigen" and "an antibody specific for an antigen" are used interchangeably herein with the term "an antibody specifically binding to an antigen" or "an antigen specific antibody" e.g. an IL-17A specific antibody. The dissociation constant can be measured using standard procedures .
The term "IL-17A biological activity" or "IL-17A
activation" as used herein refers to any activity occurring as a result of IL-17A binding to the IL-17A receptor. Exemplary IL- 17A biological activities result in increased secretion of IL-6 or IL-8, NF-κΒ activation, or regulation of downstream kinases sucn as ERK1, ERK2 and p38 upon binding to the IL-17A receptor. The release of cytokines and chemokines from cells, tissues or in circulation, NF-κΒ activation, or kinase phosporylation events can be measured using well known methods, for example
immunoassays, immunoblotting, or reporter gene systems (Yao et al . , Immunity 3:811-21, 1995; Awane et al . , J. Immunol. 162:5337- 44, 1999) .
The term "vector" means a polynucleotide capable of being duplicated within a biological system or that can be moved between such systems. Vector polynucleotides typically contain elements, such as origins of replication, polyadenylation signal or selection markers, that function to facilitate the duplication or maintenance of these polynucleotides in a biological system. Examples of such biological systems may include a cell, virus, animal, plant, and reconstituted biological systems utilizing biological components capable of duplicating a vector. The polynucleotide comprising a vector may be DNA or RNA molecules or a hybrid of these.
The term "expression vector" means a vector that can be utilized in a biological system or in a reconstituted biological system to direct the translation of a polypeptide encoded by a polynucleotide sequence present in the expression vector.
The term "polynucleotide" means a molecule comprising a chain of nucleotides covalently linked by a sugar-phosphate backbone or other equivalent covalent chemistry. Double and single-stranded DNAs and RNAs are typical examples of
polynucleotides .
The term "polypeptide" or "protein" means a molecule that comprises at least two amino acid residues linked by a peptide bond to form a polypeptide. Small polypeptides of less than 50 amino acids may be referred to as "peptides".
Conventional one and three-letter amino acid codes are used herein as follows:
Figure imgf000015_0001
Figure imgf000016_0001
Compositions of matter
The present invention provides IL-17A antibody antagonists capable of inhibiting IL-17A biological activity and uses of such antibodies. Exemplary mechanisms by which IL-17A activation may be inhibited by such antibodies include in vitro, in vivo or in situ inhibition of IL-17A homo-or heterodimerization, and blocking binding of IL-17A to the IL-17A receptor, inhibition of receptor dimerization, inhibition of kinase activity of
downstream signaling pathways, or inhibition of IL-17A mRNA transcription. Other antibody antagonists capable of inhibiting IL-17A activation by other mechanisms are also within the scope of the various aspects and embodiments of the invention. These antagonists are useful as research reagents, diagnostic reagents and therapeutic agents.
The invention provides novel antigen-binding sites derived from human immunoglobulin gene libraries. The structure for carrying an antigen-binding site is generally an antibody heavy or light chain or portion thereof.
The invention provides an isolated antibody or fragment thereof that binds specifically to human IL-17A, comprising a heavy chain variable region (VH) and a light chain variable region (VL) , wherein the antibody comprises the heavy chain complementarity determining region (CDR) 1, 2 and 3 (HCDR1, HCDR2 and HCDR3) amino acid sequences and the light chain complementarity determining region (CDR) 1, 2 and 3 (LCDR1, LCDR2 and LCDR3) amino acid sequences as shown in Table la.
Table la.
Figure imgf000017_0001
Figure imgf000018_0002
In certain embodiments, the invention provides an isolated antibody or fragment that binds specifically to human IL-17A, comprising a VH and a VL, wherein the antibody comprises the HCDR1, HCDR2 and HCDR3 amino acid sequences as shown in SEQ ID NOs: 23, 35 and 52, wherein the HCDR2 of SEQ ID NO: 35 is further defined as shown in Formula (I) :
Figure imgf000018_0001
wherein
Xaai may be His, Met, Arg, Ser or Tyr;
Xaa2 may be Trp, Thr or Tyr; and
Xaa3 may be Tyr, Phe, Ser or Asp. In other embodiments, the invention provides an isolated antibody or fragment that binds specifically to human IL-17A, comprising a VH and a VL, wherein the antibody comprises the LCDR1, LCDR2 and LCDR3 amino acid sequences as shown in SEQ ID NOs: 2, 5 and 11, wherein the LCDR3 of SEQ ID NO: 11 is further defined as shown in Formula (II) :
Figure imgf000019_0001
wherein
Xaa4 may be His or Gin;
Xaa5 may be Phe or Gly;
Xaa6 may be Thr, Val or Asn;
Xaa7 may be lie, Thr or Tyr;
Xaa8 may be Pro or Arg;
Xaa9 may be Ser or Pro; and
Xaaio may be His, Phe or Leu.
In other embodiments, the invention provides an isolated antibody or fragment that binds specifically to binding human IL- 17A, comprising a VH and a VL, wherein the antibody comprises the LCDR1, LCDR2, and LCDR3 amino acid sequences as shown in SEQ ID NOs: 2, 5 and 17, wherein the LCDR3 of SEQ ID NO: 17 is further shown in Formula (HI) :
Figure imgf000019_0002
wherein
Xaa may be Gin or Thr;
Xaa^2 may be Ser or Tyr;
Xaai3 may be Asn, Arg, Val or Tyr;
Xaai4 may be His or Ser;
Xaais may be He, Thr, Leu, Ala or Ser;
Xaais may be Pro , Leu or Ser;
Xaai7 may be Pro , Ser, Phe or Leu; and
Xaaie may be Ala, Leu or Asp.
In other embodiments, the invention provides an isolatec body or fragment that binds specifically to human IL-17A, comprising a VH and a VL, wherein the antibody comprises the HCDR1, HCDR2 and HCDR3 amino acid sequences as shown in SEQ ID NOs: 24, 36 and 57, wherein the HCDR3 of SEQ ID NO: 57 is further defined as shown in Formula (IV) :
Figure imgf000020_0002
wherein
Xaaig is Met, lie, Leu or Thr.
In other embodiments, the invention provides an isolated antibody or fragment that binds specifically to human IL-17A, comprising a VH and a VL, wherein the antibody comprises the LCDR1, LCDR2 and LCDR3 amino acid sequences as shown in SEQ ID NOs: 3, 6 and 22, wherein the LCDR3 of SEQ ID NO: 22 is further defined as shown in Formula (V) :
Figure imgf000020_0003
wherein
Xaa20 is Met, Leu, Thr or Tyr.
In other embodiments, the invention provides an isolated antibody or fragment that binds specifically to human IL-17A, comprising a VH and a VL, wherein the antibody comprises the HCDR1, HCDR2 and HCDR3 amino acid sequences as shown in SEQ ID NOs: 25, 46 and 61, wherein the HCDR2 of SEQ ID NO: 46 is further defined as shown in Formula (VI) :
Figure imgf000020_0001
wherein
Xaa2i may be Ala, Gly, Thr or Val;
Xaa22 may be Asn or Ser;
Xaa23 may be Gly, Met, Lys, lie, Leu or His; Xaa24 may be Leu, Asp, Ala, His, Thr, Gly or Ser;
Xaa25 may be Gly or Ser;
Xaa26 may be Thr, Gly, Tyr or Asp; Xaa27 may be His, Trp, Tyr or Phe;
Xaa28 may be Lys, Thr or lie; and
Xaa2g may be Tyr, Phe or Asn, and
the HCDR3 of SEQ ID NO: 61 is defined as shown in Formula (VII) :
Figure imgf000021_0001
wherein
Xaa30 may be Met, Leu or Thr.
In other embodiments, the invention provides an isolated antibody or fragment that binds specifically to human IL-17A, comprising a VH and a VL, wherein the antibody comprises the HCDR1, HCDR2 and HCDR3 amino acid sequences as shown in SEQ ID NOs: 25, 51 and 58, wherein the HCDR2 of SEQ ID NO: 51 is further defined as shown in Formula (VIII) :
Figure imgf000021_0002
wherein
Xaa3i may be Ala, Lys, Met or His;
Xaa32 may be Asn, Met, Thr or Arg;
Xaa33 may be Gly or Asp;
Xaa34 may be Arg, His or Asn; and
Xaa35 may be Asp or Gly.
Antibodies whose antigen-binding site amino acid sequences are substantially identical to those shown in Table la (SEQ ID NOs: 1-61) are encompassed within the scope of the invention. Typically, this involves one or more amino acid substitutions with an amino acid having similar charge or hydrophobic or stereochemical characteristics, and are made to improve antibody properties, for example stability or affinity. For example, a conservative substitution may involve a substitution of a native amino acid residue with a nonnative residue such that there is little or no effect on the polarity or charge of the amino acid residue at that position. Furthermore, any native residue in the polypeptide may also be substituted with alanine, as has been previously described for alanine scanning mutagenesis (MacLennan et al., Acta Physiol. Scand. Suppl . 643:55-67, 1998; Sasaki et al . , Adv. Biophys. 35:1-24, 1998) . Conservative substitutions will produce molecules having functional and chemical
characteristics similar to those of the molecule from which such modifications are made. Non-conservative substitutions in the functional and/or chemical characteristics of the molecules may be accomplished by selecting substitutions in the amino acid sequence that differ significantly in their effect on maintaining
(1) the structure of the molecular backbone in the area of the substitution, for example, as a sheet or helical conformation,
(2) the charge or hydrophobicity of the molecule at the target site, or (3) the size of the molecule. Desired amino acid substitutions (whether conservative or non-conservative) can be determined by those skilled in the art at the time such
substitutions are desired. For example, amino acid substitutions can be used to identify residues important for the function of the antibodies, such as residues affecting affinity, or residues that impart undesireable properties such as aggregation.
Exemplary amino acid substitutions are shown in Table lb, and Figure 1.
Substitutions in the framework regions, in contrast to antigen- binding sites may also be made as long as they do not adversely affect the properties of the antibody. Framework substitutions can be made for example at the Vernier Zone residues (US. Pat. No. 6,649,055) to improve antibody affinity or stability. Substitutions can also be made at those framework positions in the antibody that differ in sequence when compared to the homologous human germline gene sequences to reduce possible immunogeneicity . These modifications can be done for example to antibodies derived from de novo antibody libraries, such as pIX libraries.
Figure imgf000023_0001
Conservative amino acid substitutions also encompass non- naturally occurring amino acid residues which are typically incorporated by chemical peptide synthesis rather than by synthesis in biological systems. Amino acid substitutions can be done for example by PCR mutagenesis (US Pat. No. 4,683,195) .
Libraries of variants can be generated using well known methods, for example using random (NNK) or non-random codons, for example DVK codons, which encode 11 amino acids (ACDEGKNRSYW) , and screening the libraries or variants with desired properties, as shown in Example 1. Figure 1 shows substitutions made to five parent IL-17A antibody antagonists within the LCDR3, HCDR2 and HCDR3 regions to improve antibody properties . Improved
properties, such as affinity or stability can be measured by well known methods . In other embodiments, the invention provides an isolated antibody or fragment that binds specifically to human IL-17A, comprising a VH and a VL, wherein the antibody comprises certain VH and VL sequences, and also provides for each isolated VH and VL as shown in Table 2.
Table 2
Figure imgf000024_0001
Figure imgf000024_0002
Although the embodiments illustrated in the Examples comprise pairs of variable regions, pairs of full length antibody chains, or pairs of CDR1, CDR2 and CDR3 regions, one from a heavy chain and one from a light chain, a skilled artisan will
recognize that alternative embodiments may comprise single heavy chain variable regions or single light chain variable regions, single full length antibody chains, or CDR1, CDR2 and CDR3 regions from one antibody chain, either heavy or light. The single variable region, full length antibody chain or CDR1, CDR2 and CDR3 region of one chain can be used to screen for
corresponding domains in another chain, the two chains capable of forming an antibody that specifically binds IL-17A. The screening may be accomplished by phage display screening methods using, e.g., a hierarchical dual combinatorial approach disclosed in PCT Publ. No. WO92/01047. In this approach, an individual colony containing either a H or L chain clone is used to infect a complete library of clones encoding the other chain (L or H) , and the resulting two-chain specific antigen-binding domain is selected in accordance with phage display techniques as
described.
In other embodiments, the invention provides an isolated antibody or fragment that binds specifically to human IL-17A, comprising a VH and a VL having amino acid sequences at least 90% identical to the VH and VL amino acid sequences shown in Table 2.
In other embodiments, the invention provides an isolated antibody or fragment that binds specifically to human IL-17A, comprising a VH and a VL having amino acid sequences at least 95% identical to the VH and VL amino acid sequences shown in Table 2.
In another aspect, the invention provides an isolated antibody or fragment having certain heavy chain and light chain amino acid sequences as shown in Table 2. In addition to numbering antibody residues sequentially, polypeptides encoding antibody chains can be numbered based on Rabat's or Chothia' s numbering (Rabat et al . , sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md., 1991; Chothia and Lesk, Mol . Biol.
196:901-917, 1987) . Examples of correspondence between
sequential, Rabat and Chotia numbering for a select antibody chains are shown in Figure 3. The positions highlighted in gray indicate antibody CDR regions.
In other embodiments, the invention provides an isolated antibody or fragment that binds specifically to human IL-17A, comprising a VH and a VL, wherein the antibody comprises a heavy chain variable region paratope selected from Chothia residues S51, T53, F56, Y58, Q95, L96 and T97 and a light chain variable region paratope selected from Chothia residues Y32, D50, Y91, F93 and F94. The heavy chain paratope and the light chain paratope Chothia residues correspond to heavy chain residues S52, T54, F57, Y59, Q99, L100 and T101 of SEQ ID NO: 86 and light chain residues Y31, D49, Y90, F92 and F93 of SEQ ID NO: 79.
In other embodiments, the invention provides an isolated antibody or fragment that binds specifically to human IL-17A, comprising heavy chain variable region paratope amino acid residues that interact with residues of human IL-17A having the amino acid sequence shown in SEQ ID NO: 105, comprising:
a first threonine residue that interacts with R55 or E57 of human IL-17A;
a glutamine residue that interacts with R55 or E57 of human IL-17A;
a lysine residue that interacts with E57 of human IL-17A; a tyrosine residue that interacts with P59, E60 or R101 of human IL-17A;
a phenylalanene residue that interacts with E60, R101, E102 or P103 of human IL-17A;
a serine residue that interacts with E60 of human IL-17A; and
a second threonine residue that interacts with E60 of human IL-17A.
In other embodiments, the invention provides an isolated antibody or fragment that binds specifically to human IL-17A, comprising light chain variable region paratope amino acid residues that interact with residues of human IL-17A having the amino acid sequence shown in SEQ ID NO: 105, comprising:
a first phenylalanine residue that interacts with L26 of human IL-17A;
an aspartic acid residue that interacts with R55 or W67 of human IL-17A;
a first tyrosine residue that interacts with P59, S64 or R101 of human IL-17A; a second phenylalanine residue that interacts with P59, E60, R61, Y62, R101 or F110 of human IL-17A; and
a second tyrosine residue that interacts with V65 of human IL-17A.
In another embodiment, the invention provides an isolated antibody or fragment that binds specifically to human IL-17A, comprising heavy chain variable region paratope amino acid residues and light chain variable region paratope amino acid residues that interact with residues of human IL-17A having the amino acid sequence shown in SEQ ID NO: 105, comprising:
a tyrosine residue in the heavy chain variable region that interacts with R101 of human IL-17A;
a phenylalanine residue in the heavy chain variable region that interacts with R101, of human IL-17A;
a first phenylalanine residue in the light chain variable region that interacts with Y62 and R101 of human IL-17A;
a second phenylalanine residue in the light chain variable region that interacts with L26 and F110 of human IL-17A; and a tyrosine residue in the light chain variable region that interacts with R101 of human IL-17A.
In another embodiment, the invention provides an isolated antibody or fragment that binds specifically to human IL-17A, comprising a heavy chain variable region and a light chain variable region, wherein the antibody comprises:
a heavy chain variable region paratope selected from
Chothia residues F56 and Y58; and
a light chain variable region paratope selected from
Chothia residues Y91, F93 and F94.
The heavy chain paratope Chothia residues F56 and Y58 and the light chain paratope Chothia residues Y91, F92 and F94 are residues in direct contact with IL-17A residues L26, Y62, R101 and F110. These IL-17A residues are part of both the Fab6468 epitope and the P2 pocket cavity (see below) . While not wishing to be bound to any particular theory, it is believed that interaction between the Fab6468 and IL-17A at these select residues may be sufficient for the antibody to block IL-17A activity .
Fully human mAbs lacking any non-human sequences can be prepared and optimized from phage display libraries by techniques referenced in, e.g., Knappik et al . , J. Mol. Biol. 296:57-86, 2000; and Krebs et al . , J. Immunol. Meth. 254:67-84 2001. In an exemplary method, the antibodies of the invention are isolated from libraries expressing antibody heavy and light chain variable regions as fusion proteins with bacteriophage pIX coat protein. The antibody libraries are screened for bining to human IL-17mut6 (SEQ ID NO: 105), and the obtained positive clones are further characterized, the Fabs isolated from the clone lysates, and expressed as full length IgGs. Exemplary antibody libraries and screening methods are described in Shi et al . , J. Mol. Biol.
397:385-96, 2010; PCT Pat. Appl . No. WO09/085462, and U.S. Ser. No. 12/546850; U.S. Pat. Nos . 5,223,409, 5,969,108, and
5, 885, 793) .
The resulting mAbs can further be modified in their framework regions to change certain framework residues to those present in a matching human germline, as exemplified within.
Anibodies of the invention binding specific IL-17A epitopes can be made by immunizing humanized mice expressing human immunoglobulin loci (Lonberg et al . , Nature 368:856-9, 1994;
Fishwild et al . , Nature Biotechnology 14:845-51, 1996; Mendez et al., Nature Genetics 15:146-56, 1997, US. Pat. Nos. 5,770,429, 7,041,870, and 5,939,598) or Balb/c mice with the peptides encoding the epitopes, for example peptide 56NEDPERYPSVIWE68 (SEQ ID NO: 157) or 100RREPPHCPNSFRLEKILn6 (SEQ ID NO: 158) and using the hybrodima method of Kohler et al . , Nature 256:495-97. The resulting antibodies are tested for their binding to the epitope using standard methods. The identified mAbs can further be modified by incorporating altered framework support residues to preserve binding affinity by techniques such as those disclosed in Queen et al . , Proc. Natl. Acad. Sci. (USA), 86:10029-32, 1989 and Hodgson et al., Bio/Technology, 9:421, 1991.
Isolated antibodies having certain paratope residues (eg., the core paratope residues defined in Table 10) that bind specifically to human IL-17A can be made, for example, by grafting the paratope residues into a suitable scaffold,
assembling the engineered scaffolds into full antibodies, expressing the resulting antibodies, and testing the antibodies for binding to IL-17A or for an effect on IL-17A biological activity. Exemplary scaffolds are amino acid sequences of human antibody variable regions encoded by human germline genes. The scaffolds can be selected based on for example overall sequence homology, % identity between the paratope residues, or canonical structure class identity between the scaffold and an exemplary antibody, such as mAb6785. Human antibody germline genes are disclosed in, for example, Tomlinson et al . , J. Mol. Biol
227:776-798, and at the International ImMunoGeneTics (IMGT) database (http :// www imgt org) . Consensus human framework regions can also be used, e.g., as described in U.S. Pat. No. 6,054,297. Selection of suitable scaffold can be done for example according to methods described in PCT Publ . No.
WO10/045340.
Exemplary human germline genes that can be used as
scaffolds onto which the paratope residues are grafted are the genes encoded by the νλ3, Vh3, J , and the Jh frameworks.
Exemplary VK3 genes are IGLV3-1, IGLV3-9, IGLV3-10, IGLV3-12, IGLV3-16, IGLV3-19, IGLV3-21, IGLV3-22, IGLV3-25, IGLV3-27, and IGLV3-32 (IMGT nomenclature, *01 alleles), (SEQ ID NOs : 117-127, respectively) . Exemplary J genes are IGLJ1, IGLJ2, IGLJ3, IGLJ4, IGLJ5, IGLJ6, and IGLJ7 (SEQ ID NOs: 128-134,
respectively) . Exemplary Vh3 genes are IGHV3-7, IGHV3-9, IGHV3- 11, IGHV3-16, IGHV3-19, IGHV3-20, IGHV3-21, IGHV3-23, IGHV3-30, IGHV3-30*03, IGHV3-33, IGHV3-45, IGHV3-48, IGHV3-64, and IGHV3-74 (IMGT nomenclature, *01 alleles except when different allele is specificed) (SEQ ID NO : s 135-150, respectively) . Exemplary Jh genes are IGHJ1, IGHJ2, IGHJ3, IGHJ4, IGHJ5, and IGHJ6 (SEQ ID NO : s 151-156, respectively) . The germline J-regions are used in their entirety or in part to select FR4 sequences. For example, the mAb6785 light chain paratope residues can be grafted into a νλ3 protein framework encoded by IGLV3-1 (SEQ ID NO: 117) that is joined to the J region sequence encoded by IGLJ2 (SEQ ID NO: 129) with insertion of a single amino acid residue between the IGLV3-1 and IGLJ2 sequences, for example methionine. The νλ3 protein framework encoded by IGLV3-1 may contain additional
substitutions, for example a substitution of cysteine residue at position 33 of SEQ ID NO: 117 ("ACW") with for example
asparagine; and substitution of residues 1-3 of SEQ ID NO: 117 ("SYE") with an amino-terminal sequence common to other lambda chain families, such as "QSV" of IGLV1 family. Sequences from other exemplary functional νλ3 and genes can be used for grafting mAb6785 light chain paratope residues with the insertion of zero, one, or two amino acid residues between the carboxy- terminus encoded by the νλ3 genes and the amino terminus encoded by the genes, such that the length of the CDR3 region is 11 amino acids. For example, methionine and isoleucine can be inserted between IGLV3-22 (SEQ ID NO: 124) and IGLJ2 (SEq ID NO: 129) . Figure 2A shows alignment of exemplary light chain scaffolds that can be used for grafting. The mAb6785 heavy chain paratope residues can be grafted onto for example a Vh3 framework encoded by IGHV3-23 (SEQ ID NO: 142), that is joined to the J region FR4 sequence (11 C-terminal amino acids e.g.
"WGQGTLVTVSS") of IGHJ1 (SEQ ID NO: 151), with the insertion of about about 5-7 residues, for example 6 residues, constituting HCDR3, between the V and the J regions. The inserted HCDR3 about 5-7 residues include insertion of glutamine, leucine and
threonine, e.g. 3 of the paratope residues from mAb6785 Vh (Table 10) . Sequences from other exemplary functional Vh3 and Jh genes can be used for grafting mAb6785 heavy chain paratope residues. In some cases, one C-terminal amino acid from the Vh3 gene may be deleted before insertion of the about 5-7 residues constituting the HCDR3 so that only FR3 sequences are included in the
scaffold. Sequences from other Vh3 genes that encode a CDR2 of 17 residues (residues 50-66 of IGHV3-23 (SEQ Id NO: 142) can also be used, and the FR4 sequences of other Jh genes can be
substituted in place of IGJH1.
The specific binding to human IL-17A and biological activity of the resulting antibody can be evaluated using standard methods. Alignments of the mAb6785 light chain variable regions and heavy chain variable regions with the exemplary Vh3, νλ3, or Jh genes are shown in Figures 2A and 2B.
Alternatively, the extended paratope residues of mAb6785, as defined in Table 10, can be used in place of the core paratope residues. The paratope-grafted engineered antibodies can further be modified by substitutions of the Vernier Zone residues (U.S. Pat. No. 6,639,055) or the Affinity Determining Residues (U.S. Pat. Appl. No. 2010/0261620; Cobaugh et al . , J Mol Biol. 378:622- 33, 2008) to improve antibody properties for example affinity. As long as the paratope-grafted antibody retains binding to IL- 17A, the framework amino acid sequence in the paratope-grafted antibody may be 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% identical to the mAb6785 framework sequences. Allelic variants of the exemplary germline gene frameworks can be used in place of the V and J region protein sequences. Th sequences of the allelic variants are well known and can be obtained at the International ImMunoGeneTics (IMGT) database
(http :// www imgt org) .
Sequences from the antigen-binding sites can be grafted in addition to the paratope residues using standard methods. For example, a complete HCDR3 or LCDR3 may be grafted.
Another embodiment of the invention is an isolated antibody or fragment that binds specifically to human IL-17A that competes for human IL-17A binding with a monoclonal antibody comprising certain HCDR1, HCDR2 and HCDR3 , and LCDR1, LCDR2 and LCDR3 amino acid sequences. Examplary monoclonal antibodies of the invention are an isolated antibody comprising HCDR1, HCDR2 and HCDR3 amino acid sequences as shown in SEQ ID NOs: 25, 43 and 60 and the LCDR1, LCDR2 and LCDR3 amino acid sequences as shown in SEQ ID NOs: 3, 6 and 18.
Competition between specific binding to IL-17A can be assayed in vitro using well known methods. For example, binding of MSD Sulfo-Tag™ NHS-ester -labeled antibody to IL-17A in the presence of an unlableled antibody can be assessed by ELISA.
Another embodiment of the invention is an isolated antibody or antibody or fragment thereof, wherein the antibody binds specifically to human IL-17A having the sequence shown in SEQ ID NO: 105 at amino acid residues 56-68 (SEQ ID NO: 157) and 100-116 (SEQ ID NO: 158); or at residues L26, R55, E57, P59, E60, R61, Y62, S64, V65, W67, R101, E102, P103 and F110.
Several well known methodologies can be employed to determine the binding epitope of the antibodies of the
invention. For example, when the structures of both individual components are known, in silico protein-protein docking can be carried out to identify compatible sites of interaction.
Hydrogen-deuterium (H/D) exchange can be carried out with the antigen and antibody complex to map regions on the antigen that may be bound by the antibody. Segment and point mutagenesis of the antigen can be used to locate amino acids important for antibody binding. Co-crystal structure of antibody-antigen complex is used to identify residues contributing to the epitope and paratope.
Previously described anti-IL-17A antibodies bind to epitopes on IL-17A distinct from the epitope for Fab6468
described in the present invention. Antibodies binding human IL- 17A (SEQ ID NO: 105) residues 74-85, 46-53, 71-87, 80-86, 11-18, 29-41 or 54-62 have been described (PCT Publ . Nos. WO08/021156, WO07/106769, WO07/149032, WO07/070750; US Appl . No.
US2008 /095775 , respectively) . Conformational epitopes have been described in PCT Publ . No. WO09/130459 and Gerhardt et al . , J. Mol. Biol: 394:901-21, 2009.
Another embodiment of the invention is an isolated antibody or fragment thereof, wherein the antibody binds specifically to a P2 pocket cavity on IL-17A, the P2 pocket cavity comprising of amino acid residues V22, V24, L26, 128, Y62, L99, R101, F110, and LI 12 of SEQ ID NO: 105.
Co-crystal structure of IL-17A homodimer with the anti-IL- 17A Fab6468 identified a hydrophobic P2 pocket cavity on the surface of the IL-17A homodimer, which is likely to be involved in IL-17RA binding (see Examples) . The "P2 pocket cavity" as used herein refers to a tertiary hydrophobic structural cavity on IL-17A homodimer, where the surface exposed residues in the P2 pocket are V24, L26, 128, Y62, L99, R101, F110 and L112 on monomer A and V22, V24 and L112 on monomer B, and vice versa. Select antibodies of the invention reactive with IL-17A, for example Fab6468, have direct contacts with the P2 pocket cavity residues L26, Y62, R101 and F110, which residues are also part of the Fab6468 epitope. While not wishing to be bound by any particular theory, it is assumed that the antibodies of the invention binding the select IL-17A P2 pocket cavity residues block interaction between IL-17A and IL-17RA. Based on co- crystal structure, the phenlylalanine motif (FF) at residues 93 and 94 in a light chain (SEQ ID NO: 79) of Fab6468 blocks the IL- 17A/ IL-17RA interaction, and thus is a P2 pocket cavity blocker. Other P2 pocket cavity blocker antagonists are also within the scope of this invention, such as novel peptides or small
molecules. These can be modeled based on the IL-17A/Fab6468 co- structure, and screened for their ability to replace Fab6468 binding to IL-17A. For example, peptide inhibitors can be screened from random peptide libraries that have incorporated the FF motif (for example libraries of XXXXFFXX; X indicated any amino acid; F = phenylalanine) and displayed on bacteriophage as a fusion with for example pill, pVII or pIX coat protein
(US5, 223, 409; Gao et al . , Proc . Natl. Acad. Sci. USA, 96:6025-30, 1999, Tornetta et al . , J. Immunol. Methods. 360:39-46, 2010; Shi et al., J. Mol. Biol. 397:385-96, 2010) and subsequently tested for their inhibition of Fab6468 binding to IL-17A, and inhibition of IL-17A activity.
Small molecules may be screened using libraries of
synthetic or natural compounds, or any combination thereof, and the resulting primary positive hits can be readily modified to produce structural analogs of the agents. Methods of making peptide libraries and pIX fusions, and screening the resulting libraries are well known.
Another embodiment of the invention of a method of
inhibiting interaction of human IL-17A with IL-17RA comprising: providing human IL-17A and IL-17RA; and
contacting human IL-17A with an antagonist that
specifically binds human IL-17A at at least one amino acid residue selected from the group consisting of: V22, V24, L26, 128, Y62, L99, R101, F110, and L112.
Another embodiment of the invention is a method of
inhibiting human IL-17A biological activity, comprising:
providing human IL17-A and IL-17RA; and
contacting human IL-17A with an antagonist that
specifically binds human IL-17A at at least one amino acid residue selected from the group consisting of: V22, V24, L26, 128, Y62, L99, R101, F110, and L112.
Human IL-17A and IL-17RA can be provided as isolated proteins or fusion proteins. Human IL-17A homodimer can be purified from media of activated Thl7 cells prepared by in vitro stimulation of naive CD4 T cells by two anti-CD3/anti-CD28 stimulation in the presence of IL-2, IL-23 and IL-Ιβ. The IL-17RA can be associated with cells or cell membranes, can be native or overexpressed, or can be a fragment of IL-17RA, for example the extracellular domain of the receptor. The IL-17RA can be a human IL-17RA, or IL-17RA from other species such as from mouse, rat or monkey. Antagonists binding to human IL-17A residues V22, V24, L26, 128, Y62, L99, R101, F110, and LI 12 can be identified by the ability of the antagonist to replace Fab6468 binding to IL-17A, by mutagenesis studies or by co-crystal structures. Fusion proteins of human IL-17A and IL-17RA can be made by well known methods. Exemplary fusion protein is a soluble IL-17RA fused to an immunoglobulin Fc domain.
Another aspect of the invention is an isolated
polynucleotide encoding any of the antibody heavy chains or the antibody light chains or fragments thereof of the invention or their complement. Certain exemplary polynucleotides are
disclosed herein, however, other polynucleotides which, given the degeneracy of the genetic code or codon preferences in a given expression system, encode the antibody antagonists of the invention are also within the scope of the invention. Exemplary polynuceotides are shown in SEQ ID NOs : 101, 102, 103 and 104.
Exemplary antibody antagonists may be antibodies of the IgG, IgD, IgE, IgA or IgM isotypes. Additionally, such antibody antagonists can be post-translationally modified by processes such as glycosylation, isomerization, deglycosylation or non- naturally occurring covalent modification such as the addition of polyethylene glycol (PEG) moieties (pegylation) and lipidation. Such modifications may occur in vivo or in vitro. For example, the antibodies of the invention can be conjugated to polyethylene glycol (PEGylated) to improve their pharmacokinetic profiles. Conjugation can be carried out by techniques known to those skilled in the art. Conjugation of therapeutic antibodies with PEG has been shown to enhance pharmacodynamics while not
interfering with function. See Deckert et al . , nt . J. Cancer 87:382-90, 2000; Knight et al . , Platelets 15:409-18, 2004; Leong et al . , Cytokine 16:106-19, 2001; and Yang et al . , Protein Eng. 16:761-70, 2003.
Pharmacokinetic properties of the antibodies of the invention can be enhanced through Fc modifications by techniques known to those skilled in the art. The "Fc" of an antibody is not involved directly in binding of an antibody to an antigen, but exhibits various effector functions. An antibody "Fc" is a term well known and is defined on the basis of papain cleavage of antibodies. The Fc of an antibody is directly involved in ADCC (antibody-dependent cell-mediated cytotoxicity) and CDC
(complement-dependent cytotoxicity) based on complement
activation, Clq binding and Fc receptor binding. Such complement and Fc receptor binding sites are well known and include for example L234, L235, D270, N297, E318, K320, K322, P331, and P329 (numbering according to EU index of Rabat) (Brekke et al . , Eur. J. Immunol. 24:2542-7, 1995; US. Pat. Nos. 5,624,821, 7,597,889, Canfield and Morrison, J. Exp. Med. 173:1483-91, 1991) . For example, mutation of Leu234/Leu235 in the hinge region of IgGl to L234A/L235A or Phe235/Leu236 in the hinge region of IgG4 to P235A/L236A minimizes FcR binding and reduces the ability of the immunoglobulin to mediate complement dependent cytotoxicity and ADCC. A Ser to Pro substitution in the Cys-Pro-Ser-Cys (CPSC) motif in the hinge region of IgG4 heavy chains capable of forming either inter- or intra-heavy chain disulfide bonds in vivo via action of isomerases (Aalberse and Schuurman, Immunology 105:9- 19, 2002), results in "IgGl-like behavior", i.e., the Pro- substituted molecules are unable to form intra-heavy chain disulfide bonds. The location of the CPSC motif is typically found at residue 228 of a mature heavy chain but can change depending on CDR lengths . An exemplary IgGl Fc region having the Leu234/Leu235 residues has an amino acid sequence shown in SEQ ID NO: 114, wherein the residues L117 and L118 correspond to the Leu234/Leu235 residues in the mature heavy chain. An exemplary IgG4 Fc region having the Cys-Pro-Ser-Cys (CPSC) motif and the Leu234/Leu235 residues has an amino acid sequence shown in SEQ ID NO: 115, where the CPSC motif is located at residues 106-109 and the Leu234/Leu235 residues at positions 122 and 123.
Antibodies or fragments thereof of the invention modified to improve stability, selectivity, cross-reactivity, affinity, immunogenicity or other desirable biological or biophysical property are within the scope of the invention. Stability of an antibody is influenced by a number of factors, including (1) core packing of individual domains that affects their intrinsic stability, (2) protein/protein interface interactions that have impact upon the HC and LC pairing, (3) burial of polar and charged residues, (4) H-bonding network for polar and charged residues; and (5) surface charge and polar residue distribution among other intra- and inter-molecular forces (Worn et al . , J. Mol. Biol. 305:989-1010, 2001) . Potential structure
destabilizing residues may be identified based upon the crystal structure of the antibody or by molecular modeling in certain caases, and the effect of the residues on antibody stability can be tested by generating and evaluating variants harboring mutations in the identified residues. One of the ways to increase antibody stability is to raise the thermal transition midpoint (Tm) as measured by differential scanning calorimetry
(DSC) . In general, the protein Tm is correlated with its stability and inversely correlated with its susceptibility to unfolding and denaturation in solution and the degradation processes that depend on the tendency of the protein to unfold
(Remmele et al . , Biopharm. 13:36-46, 2000) . A number of studies have found correlation between the ranking of the physical stability of formulations measured as thermal stability by DSC and physical stability measured by other methods (Gupta et al . , AAPS PharmSci. 5E8, 2003; Zhang et al . , J. Pharm. Sci. 93:3076- 89, 2004; Maa et al . , Int. J. Pharm., 140:155-68, 1996; Bedu-Addo et al . , Pharm. Res., 21:1353-61, 2004; Remmele et al . , Pharm. Res., 15:200-8, 1997) . Formulation studies suggest that a Fab Tm has implication for long-term physical stability of a
corresponding mAb. Differences in amino acids in either
framework or within the CDRs could have significant effects on the thermal stability of the Fab domain (Yasui, et al . , FEBS Lett. 353: 143-6, 1994) .
The antibody antagonists of the invention may bind IL-17A with a Kd less than or equal to about 10"7, 10"8, 10"9, 10"10, 10"11 or 10" M. The affinity of a given molecule for IL-17A, such as an antibody can be determined experimentally using any suitable method. Such methods may utilize Biacore or KinExA
instrumentation, ELISA or competitive binding assays known to those skilled in the art.
Antibody antagonists binding human IL-17A with a desired affinity can be selected from libraries of variants or fragments by techniques including antibody affinity maturation. Antibody antagonists can be identified based on their inhibition of IL-17A biological activity using any suitable method. Such methods may utilize reporter-gene assays or assays measuring cytokine production using well known methods and as described in the application .
Another embodiment of the invention is a vector comprising at least one polynucleotide of the invention. Such vectors may be plasmid vectors, viral vectors, vectors for baculovirus expression, transposon based vectors or any other vector suitable for introduction of the polynucleotides of the invention into a given organism or genetic background by any means.
Another embodiment of the invention is a host cell
comprising any of the polynucleotides of the invention such as a polynucleotide encoding a polypeptide comprising an
immunoglobulin heavy chain variable region having the amino acid sequence shown in SEQ ID NOs : 67-75 and 81-86 or an
immunoglobulin light chain variable region having the amino acid sequence shown in SEQ ID NOs: 62-66 and 76-80 or an an
immunoglobulin heavy chain having the amino acid sequence shown in SEQ ID NOs: 92-100 or an immunoglobulin light chain having the amino acid sequence shown in SEQ ID NOs: 87-91. Such host cells may be eukaryotic cells, bacterial cells, plant cells or archeal cells. Exemplary eukaryotic cells may be of mammalian, insect, avian or other animal origins. Mammalian eukaryotic cells include immortalized cell lines such as hybridomas or myeloma cell lines such as SP2/0 (American Type Culture Collection
(ATCC) , Manassas, VA, CRL-1581), NS0 (European Collection of Cell Cultures (ECACC) , Salisbury, Wiltshire, UK, ECACC No. 85110503), FO (ATCC CRL-1646) and Ag653 (ATCC CRL-1580) murine cell lines. An exemplary human myeloma cell line is U266 (ATTC CRL-TIB-196) . Other useful cell lines include those derived from Chinese
Hamster Ovary (CHO) cells such as CHO-K1SV (Lonza Biologies, Walkersville, MD) , CHO-K1 (ATCC CRL-61) or DG44.
Another embodiment of the invention is a method of making an antibody reactive with IL-17A comprising culturing a host cell of the invention and recovering the antibody produced by the host cell. Methods of making antibodies and purifying them are well known in the art. For expression, the engineered family 2, 6a, 6b, 19a and 19b heavy chain sequences can include an N-terminal leader sequence such as MAWVWTLLFLMAAAQSIQA (SEQ ID NO:109) .
Exemplary nucleotide sequences encoding the heavy chain of candidate mAb6785 (family 19) with a leader sequence and the mature form (without a leader sequence) are shown in SEQ ID NOs: 101 and 102, respectively. Likewise, for expression, the light chain sequences of the family 2, 6a, 6b antibodies of the invention can include an N-terminal leader sequence such as MGVPTQVLGLLLLWLTDARC (SEQ ID NO: 110) and the light chain
sequences of the family 19a and 19b antibodies of the invention can include an N-terminal leader sequence such as
MAWSPLLLTLLAHCTGSWA (SEQ ID NO: 116) . Exemplary nucleotide sequences encoding the light chain of codon optimized mAb6785 with a leader sequence and the mature form (without a leader sequence) are shown in SEQ ID NOs: 103 and 104, respectively.
Another embodiment of the invention is a hybridoma cell line that produces an antibody of the invention.
Methods of Treatment
IL-17A antagonists of the invention, for example IL-17A antibody antagonists, may be utilized in any therapy where it is desired to reduce the effects of IL-17A in the animal patient. IL- 17A may be circulating in the body or may be present in an undesirably high level localised at a particular site in the body, for example a site of inflammation. While not wishing to be bound by any particular theory, the antagonists of the invention provide beneficial therapy by preventing or reducing IL-17A binding to its receptor, or homo- or heterodimerization of IL-17A. The methods of the invention may be used to treat an animal patient belonging to any classification. Examples of such animals include mammals such as humans, rodents, dogs, cats and farm animals.
Antibodies of the invention may be useful for the
prophylaxis and treatment of IL-17A mediated conditions, such as inflammatory conditions, allergies and allergic conditions, hypersensitivity reactions, autoimmune diseases, severe
infections, and organ or tissue transplant rejection. The antibodies of the invention are also useful in the preparation of a medicament for such treatment, wherein the medicament is prepared for administration in dosages defined herein. Exemplary IL-17A mediated conditions are inflammatory conditions, immune and proliferative disorders, including rheumatoid arthritis (RA) , ankylosing spondylitis, psoriatic arthritis, osteoarthritis, osteoporosis, uveitis, inflammatory fibrosis (e.g., scleroderma, lung fibrosis, and cirrhosis), inflammatory bowel disorders (e.g., Crohn's disease, ulcerative colitis and inflammatory bowel disease) , asthma (including allergic asthma) , allergies, COPD, multiple sclerosis, psoriasis, systemic lupus erythematosus, diabetes and cancer. Positive outcomes in patients treated with anti-human IL-17A therapies have been described in rheumatoid arthritis, psoriasis and non-infectious uveitis (Genovese et al . , Arthritis Rheum. 62:929-39, 2010; Hueber et al . , Sci. Transl . Med. 2 : 52ra72. , 2010) .
Inflammatory pulmonary condition is an example of an inflammatory condition. Exemplary inflammatory pulmonary conditions include infection-induced pulmonary conditions including those associated with viral, bacterial, fungal, parasite or prion infections; allergen-induced pulmonary
conditions; pollutant-induced pulmonary conditions such as asbestosis, silicosis, or berylliosis; gastric aspiration-induced pulmonary conditions, immune dysregulation, inflammatory
conditions with genetic predisposition such as as cystic
fibrosis, and physical trauma-induced pulmonary conditions, such as ventilator injury. These inflammatory conditions also include asthma, emphysema, bronchitis, chronic obstructive pulmonary disease (COPD) , sarcoidosis, histiocytosis, lymphangiomyomatosis , acute lung injury, acute respiratory distress syndrome, chronic lung disease, bronchopulmonary dysplasia, community-acquired pneumonia, nosocomial pneumonia, ventilator -associated
pneumonia, sepsis, viral pneumonia, influenza infection,
parainfluenza infection, rotavirus infection, human
metapneumovirus infection, respiratory syncitial virus infection and aspergillus or other fungal infections. Exemplary infection- associated inflammatory diseases may include viral or bacterial pneumonia, including severe pneumonia, cystic fibrosis,
bronchitis, airway exacerbations and acute respiratory distress syndrome (ARDS) . Such infection-associated conditions may involve multiple infections such as a primary viral infection and a secondary bacterial infection. Dysregulated IL-17A production may play a role in the pathology of pulmonary diseases such as asthma and Chronic Obstructive Pulmonary Disease (COPD) (reviewed in Alcorn et al . , Annu . Rev. Physiol. 72:495-516, 2010) . IL-17A has been shown to regulate neutrophilic inflammation in the lungs - a hallmark of severe asthma as well as COPD - owing to the capacity of IL-17A to induce factors important in neutrophil recruitment, survival and activation from lung resident
epithelial cells (e.g, IL-6, IL-8, GM-CSF, G-CSF) . The
antibodies aof the present invention suppress IL-6, IL-8, and GM- CSF secretion from lung epithelial cells, and thus may be beneficial in the therapeutic or prophylactic treatment of subjects with pulmonary inflammatory conditions, such as asthma and COPD. Commonly used animal models for asthma and airway inflammation include the ovalbumin challenge model and
methacholine sensitization models (Hessel et al . , Eur. J. Pharmacol. 293:401-12, 1995) . Inhibition of cytokine and chemokine production from cultured human bronchial epithelial cells, bronchial fibroblasts or airway smooth muscle cells can also be used as in vitro models. The administration of
antagonists of the present invention to any of these models can be used to evaluate the use of those antagonists to ameliorate symptoms and alter the course of asthma, airway inflammation, COPD and the like.
Psoriasis is another example of an inflammatory condition. Psoriasis is characterized by T cell mediated hyperproliferation of keratinocytes coupled with an inflammatory infiltrate. The inflammation and hyperproliferation of psoriatic tissue is associated with a different histological, antigenic, and cytokine profile than normal skin. Among the cytokines associated with psoriasis are: TNF , IL-19, IL-18, IL-15, IL-12, IL-7, IFNy, IL- 17A and IL-23 (Gudjonsson et al . , Clin. Exp. Immunol. 135:1-8, 2004) . IL-17A has been found overexpressed in psoriatic lesions (US Pat. No. 7,776,540) and positive outcomes in patients treated with anti-human IL-17A therapies have been
described (Hueber et al., Sci. Transl. Med. 2: 52ra72., 2010) .
Arthritis, including osteoarthritis, rheumatoid arthritis, arthritic joints as a result of injury, and the like, are common inflammatory conditions, which would benefit from the therapeutic use of anti-inflammatory proteins, such as the antagonists of the present invention. Activation of IL-17A signaling may perpetuate inflammation and further tissue damage in the inflamed joint. Several animal models for rheumatoid arthritis are known. For example, in the collagen-induced arthritis (CIA) model, mice develop chronic inflammatory arthritis that closely resembles human rheumatoid arthritis. Administration of the IL-17A antibodies of the present invention to the CIA model mice can be used to evaluate the use of these antagonists to ameliorate symptoms and alter the course of diseases. Exemplary gastrointestinal inflammatory conditions are inflammatory bowel disease (IBD), ulcerative colitis (UC) and Crohn's disease (CD), colitis induced by environmental insults
(e.g., gastrointestinal inflammation (e.g., colitis) caused by or associated with (e.g., as a side effect) a therapeutic regimen, such as administration of chemotherapy, radiation therapy, and the like) , infections colitis, ischemic colitis, collagenous or lymphocytic colitis, necrotizing enterocolitis, colitis in conditions such as chronic granulomatous disease or celiac disease, food allergies, gastritis, infectious gastritis or enterocolitis (e.g., Helicobacter pylori-infected chronic active gastritis) and other forms of gastrointestinal inflammation caused by an infectious agent. Several animal models for gastrointestinal inflammatory conditions exist. Some of the most widely used models are the 2 , 4 , 6-trinitrobenesulfonic
acid/ethanol (TNBS ) -induced colitis model or the oxazalone model, which induce chronic inflammation and ulceration in the colon
(Neurath et al . , Intern. Rev. Immunol 19:51-62, 2000) . Another model uses dextran sulfate sodium (DSS), which induces an acute colitis manifested by bloody diarrhea, weight loss, shortening of the colon and mucosal ulceration with neutrophil infiltration. Another model involves the adoptive transfer of naive CD45RBhl9h CD4 T cells to RAG or SCID mice. In this model, donor naive T cells attack the recipient gut causing chronic bowel inflammation and symptoms similar to human inflammatory bowel diseases (Read and Powrie, Curr. Protoc . Immunol. Chapter 15 unit 15.13, 2001) . The administration of antagonists of the present invention in any of these models can be used to evaluate the potential efficacy of those antagonists to ameliorate symptoms and alter the course of diseases associated with inflammation in the gut, such as inflammatory bowel disease.
Renal fibrosis can develop from either an acute insult (ex. graft ischemia/reperfusion) (Freese et al . , Nephrol. Dial.
Transplant. 16:2401-6, 2001) or chronic condition (for example diabetes) (Ritz et al., Nephrol. Dial. Transplant. 11 Suppl 9:38- 44, 1996) . The pathogenesis is typically characterized by an initial inflammatory response followed by sustained fibrogenesis of the glomerular filtration apparatus and tubular interstitium (Liu, Kidney Int. 69:213-7, 2006) . Tubulointerstitial fibrosis has been shown to play a critical role in the pathogenesis of renal injury to end-stage renal failure and the proximal tubule cell has been revealed as a central mediator (Phillips and
Steadman, Histol. Histopathol. 17:247-52, 2002; Phillips, Chang Gung Med. J. 30:2-6, 2007) . Fibrogenesis in the
tubulointerstitial compartment is mediated in part by activation of resident fibroblasts, which secrete pro-inflammatory cytokines that stimulate the proximal tubule epithelium to secrete local inflammatory and fibrogenic mediators. Additionally, chemotactic cytokines are secreted by fibroblasts and epithelial cells and provide a directional gradient guiding the infiltration of monocytes/macrophages and T-cells into the tubulointerstitium. The inflammatory infiltrate produces additional fibrogenic and inflammatory cytokines that further activate fibroblast and epithelial cytokine release while also stimulating the epithelium to undergo a phenotypic transition in which the cells deposit excess extracellular matrix components (Simonson, Kidney Int.
71:846-54, 2007) . IL-17A has been shown to be upregulated during human renal allograft rejection (Van Kooten et al . , J. Am. Soc. Nephrol. 9:1526-34, 1998; Loong et al . , J. Path. 197:322-32, 2002) . IL-17A stimulates the production of the pro-inflammatory mediators IL-6, IL-8, complement component C3, and RANTES by proximal tubular epithelium (Van Kooten et al . , J. Am. Soc.
Nephrol. 9:1526-34, 1998; Woltman et al . , J. Am. Nephrol. 11:2044- 55, 2000) . These factors, in turn, mediate the recruitment of other inflammatory cell-types into the interstitium that
contribute to the maintenance of the inflammatory/immune response and, if not suppressed, the onset of fibrosis and chronic
allograft nephropathy (Racusen et al . , Kidney Int. 55:713-23, 1999; Mannon, Am. J. Transpl . 6:867-75, 2006) . Other exmplary fibrotic conditions may include liver fibrosis (including but not limited to alcohol-induced cirrhosis, viral-induced cirrhosis, autoimmune-induced hepatitis) ; lung fibrosis (including but not limited to scleroderma, idiopathic pulmonary fibrosis); kidney fibrosis (including but not limited to scleroderma, diabetic nephritis, glomerular nehpritis, lupus nephritis) ; dermal fibrosis (including but not limited to scleroderma, hypertrophic and keloid scarring, burns);
myelofibrosis; neurofibromatosis; fibroma; intestinal fibrosis; and fibrotic adhesions resulting from surgical procedures. The fibrosis can be organ specific fibrosis or systemic fibrosis. The organ specific fibrosis can be associated with lung fibrosis, liver fibrosis, kidney fibrosis, heart fibrosis, vascular fibrosis, skin fibrosis, eye fibrosis or bone marrow fibrosis. The lung fibrosis can be associated with idiopathic pulmonary fibrosis, drug induced pulmonary fibrosis, asthma, sarcoidosis or chronic obstructive pulmonary disease. The liver fibrosis can be associated with cirrhosis, schistomasomiasis or cholangitis. The cirrhosis can be selected from alcoholic cirrhosis, posthepatitis C cirrhosis, primary biliary cirrhosis. The
cholangitis can be sclerosing cholangitis. The kidney fibrosis can be associated with diabetic nephropathy or lupus
glomeruloschelerosis . The heart fibrosis can be associated with myocardial infarction. The vascular fibrosis can be associated with postangioplasty arterial restenosis or atherosclerosis. The skin fibrosis can be associated with burn scarring, hypertrophic scarring, keloid, or nephrogenic fibrosing dermatopathy . The eye fibrosis can be associated with retro-orbital fibrosis,
postcataract surgery or proliferative vitreoretinopathy . The bone marrow fibrosis can be associated with idiopathic
myelofibrosis or drug induced myelofibrosis. The systemic fibrosis can be systemic sclerosis or graft versus host disease.
Other inflammatory conditions and neuropathies, which may be prevented or treated by the methods of the invention are those caused by autoimmune diseases. These conditions and neuropathies include multiple sclerosis, systemic lupus erythematous, and neurodegenerative and central nervous system (CNS) disorders including Alzheimer's disease, Parkinson's disease, Huntington's disease, bipolar disorder and Amyotrophic Lateral Sclerosis (ALS), liver diseases including primary biliary cirrhosis, primary sclerosing cholangitis, non-alcoholic fatty liver disease/steatohepatitis, fibrosis, hepatitis C virus (HCV) and hepatitis B virus (HBV) , diabetes and insulin resistance, cardiovascular disorders including atherosclerosis, cerebral hemorrhage, stroke and myocardial infarction, arthritis,
rheumatoid arthritis, psoriatic arthritis and juvenile rheumatoid arthritis (JRA) , osteoporosis, osteoarthritis, pancreatitis, fibrosis, encephalitis, psoriasis, Giant cell arteritis,
ankylosing spondolytis, autoimmune hepatitis, human
immunodeficiency virus (HIV), inflammatory skin conditions, transplant, cancer, allergies, endocrine diseases, wound repair, other autoimmune disorders, airway hyperresponsiveness and cell, virus, or prion-mediated infections or disorders.
Administration/Pharmaceutical Compositions
The "therapeutically effective amount" of the agent effective in the treatment of conditions where suppression of IL- 17A activity is desirable can be determined by standard research techniques. For example, the dosage of the agent that will be effective in the treatment of an inflammatory condition such as asthma, Crohn' s Disease, ulcerative colitis or rheumatoid arthritis can be determined by administering the agent to relevant animal models, such as the models described herein.
In addition, in vitro assays can optionally be employed to help identify optimal dosage ranges. Selection of a particular effective dose can be determined (e.g., via clinical trials) by those skilled in the art based upon the consideration of several factors. Such factors include the disease to be treated or prevented, the symptoms involved, the patient's body mass, the patient's immune status and other factors known by the skilled artisan. The precise dose to be employed in the formulation will also depend on the route of administration, and the severity of disease, and should be decided according to the judgment of the practitioner and each patient's circumstances. Effective doses can be extrapolated from dose-response curves derived from in vitro or animal model test systems.
The mode of administration for therapeutic use of the agent of the invention may be any suitable route that delivers the agent to the host. Pharmaceutical compositions of these agents are particularly useful for parenteral administration, e.g., intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous or intranasal .
The agent of the invention may be prepared as
pharmaceutical compositions containing an effective amount of the agent as an active ingredient in a pharmaceutically acceptable carrier. The term "carrier" refers to a diluent, adjuvant, excipient, or vehicle with which the active compound is
administered. Such pharmaceutical vehicles can be liquids, such as water and oils, including those of petroleum, animal,
vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. For example, 0.4% saline and 0.3% glycine can be used. These solutions are sterile and generally free of particulate matter. They may be sterilized by conventional, well-known sterilization techniques (e.g.,
filtration) . The compositions may contain pharmaceutically acceptable auxiliary substances as required to approximate physiological conditions such as pH adjusting and buffering agents, stabilizing, thickening, lubricating and coloring agents, etc. The concentration of the agent of the invention in such pharmaceutical formulation can vary widely, i.e., from less than about 0.5%, usually at or at least about 1% to as much as 15 or 20% by weight and will be selected primarily based on required dose, fluid volumes, viscosities, etc., according to the
particular mode of administration selected. Thus, a pharmaceutical composition of the invention for intramuscular injection could be prepared to contain 1 ml sterile buffered water, and between about 1 ng to about 100 mg, e.g.
about 50 ng to about 30 mg or more preferably, about 5 mg to about 25 mg, of a IL-17A antibody antagonist of the invention. Similarly, a pharmaceutical composition of the invention for intravenous infusion could be made up to contain about 250 ml of sterile Ringer's solution, and about 1 mg to about 30 mg and preferably 5 mg to about 25 mg of an antagonist of the invention. Actual methods for preparing parenterally administrable
compositions are well known and are described in more detail in, for example, "Remington's Pharmaceutical Science", 15th ed., Mack Publishing Company, Easton, PA.
The antibody antagonists of the invention can be
lyophilized for storage and reconstituted in a suitable carrier prior to use. This technique has been shown to be effective with conventional immunoglobulins and protein preparations and art- known lyophilization and reconstitution techniques can be employed .
The present invention will now be described with reference to the following specific, non-limiting examples.
Example 1
Identification of anti-human IL-17A antagonistic mAbs
The MorphoSys Human Combinatorial Antibody Library (HuCAL®) Gold phage display library (Morphosys AG, Martinsried, Germany) was used as a source of human antibody fragments and was panned in subpools in solution. In the first round of panning, the sublibraries were selected against biotinylated mature His6- tagged IL-17A A132Q and A70Q variant (IL-17Amut6) (SEQ ID NO: 106) . In the second round, the amplified output of round 1 was selected against biotinylated His6-tagged IL-17Amut6 in the presence or absence of other IL-17A family members as a
competitor to bias against antibodies that were specific to IL- 17A. The amplified output of round 2 was divided in two pools. The first pool was panned as in round 1. The clones in the second pool were further diversified in either HCDR2 or LCDR3, depending on the sublibrary used in the initial selections, and then carried through 2 additional rounds of panning against IL- 17Amut6 to give a second source of clones for screening. Fabs from clone lysates were captured in ELISA plate wells coated with sheep anti-human Fd antibody and screened for binding to
biotinylated IL-17Amut6. Crude lysates of positive clones were screened for inhibition of IL-17Amut6 binding to recombinant human IL-17RA receptor (SEQ ID NO: 107) .
Select clones were chosen for further characterization as purified Fabs based on sequence scoring, affinity, and
representation of all sequence families, and were designated with MOR numbers. Additional variants for MOR7708, MOR7785, MOR7706, MOR7775 and MOR7700 were generated to replace Trp or Met residing in HCDR2, HCDR3, or LCDR3. Table 3 shows the generated variants.
Table 3.
Figure imgf000049_0001
The Fabs were tested for their inhibition of IL-17Amut6 and cynoIL-17A binding to recombinant human IL-17RA receptor, and their binding to IL-17Amut6. All tested Fabs inhibited both IL- 17Amut6 and cynoIL-17A binding to the IL-17RA. Affninty of the Fabs to IL-17Amut6 was measured using the SET assay (Table 4) . From the identified Fabs, candidates from families 2, 6a, 6b, 19a and 19b were seleced for further characterization.
Table 4
Figure imgf000050_0002
Figure imgf000050_0001
Example 2
Derivation, engineering and characterization of anti-IL-17A antagonistic mAbs
The selected MOR# Fabs were converted and expressed as mAbs in a human IgGl format, and given corresponding MORmAb
designation. The generated MORmAbs were tested for expression and aggregation, their ability to inhibit human and cyno IL-17A binding to human IL-17RA, and IL-8 secretion from NHDF cells. Table 5 shows IC50 values for select assays for the MORmAbs. None of the tested MORmAbs (MORmAb#s 7702, 7708, 7785, 7786, 7706, 7775, 7700, 8095, 8096, 8097, 8098, 7768) cross-reacted with other IL-17 family members.
Table 5.
Figure imgf000051_0001
Framework engineering of anti-IL-17A antagonistic mAbs
Based on activity and biophysical and biochemical properties, select MORmAbs were further engineered in their variable regions to change certain framework residues to those present in a matching human germline and to change codons to those most frequently occurring in highly expressed mammalian proteins. In family 2 VL, L11V and V85T (linear sequence) substitutions were made, converting the framework to an exact match with the VK-1 germline Vb-L5 (IGKV1-12*01) . An exemplary variable region with the V11V and V85T substitutions is the variable region having the amino acid sequence shown in SEQ ID NO: 76. In family 6a and 6b VL, DIE, V59I and T86V substitutions were made (linear sequence) , converting the framework to exact match with the Vk-3 germline Vb-L6 (IGKV3-11*01) . An exemplary variable region with the DIE, V59I and T86V substitutions is the variable region having the amino acid sequence shown in SEQ ID NO: 77. In family 6a and 6b VH, a G44S substitution was made
(linear sequence) to match the Vh-6 germline Vb 6-01 (IGHV6- 1*01) . An exemplary variable region with the G44S substitution is the variable region having the amino acid sequence shown in SEQ ID NO: 81. In family 19a and 19b VL, amino acids 1-3 (DIE) were substituted with QSV to replace the artificial kappa N- terminus with that of a lambda chain. Exemplary variable region with the QSV substitution is a variable region having the amino acid sequence shown in SEQ ID NO: 79. In family 19a and 19b VH, a V5L substitution was made to give a close match the Vh-3 germline Vb 3-23 (IGHV3-23*01. Also, in this process the heavy chain constant region amino acid sequence residues 353-357
(REEMT) were substituted with RDELT . An exemplary variable region with the V5L substitution is a variable region having the amino acid sequence shown in SEQ ID NO: 86. An exemplary heavy chain with the constant region 353-357 REEMT -> RDELT
substitutions is a heavy chain having the amino acid sequence shown in SEQ ID NO: 100. The engineered antibodies were given mAb numbers.
The corresponding designations and the sequence listings of the engineered and original variable regions and full length antibodies are listed in Table 2. The sequences of the CDRs within each family are shown in Figure 1.
The engineered mAbs were characterized as described above for the MORmAbs. The IC50 values (pM) measured using indicated assays are shown in Table 6. Table 6.
Figure imgf000053_0001
Affinity of select mAbs was assessed using Biacore. The results of the measurements are shown in Table 7.
Table 7.
Binding to human I L-17Amut6
Figure imgf000053_0002
Binding to cyno I L-17A
Figure imgf000053_0003
*dimers per anti-I L-1 7 Anti-IL-17 antibody inhibits cytokine secretion in NHBE cells
IL-17A has been shown to regulate neutrophilic inflammation in the lungs, a hallmark of severe asthma as well as COPD, owing to the capacity of IL-17A to induce factors important in
neutrophil recruitment, survival and activation (e.g, IL-6, IL-8, GM-CSF) . To determine whether anti-IL-17A antibodies of the invention can inhibit IL-17A-induced changes in lung resident cells, normal human bronchial epithelial (NHBE) cells were stimulated with human IL-17A for 48 hours in the presence of mAb6785. mAb6785 inhibited IL-17A-induced IL-6 and GM-CSF production by NHBE cells with IC50=619.0 ± 64.0 pM and 564 ± 86 pM, respectively.
Anti-IL-17 antibody inhibits the biological activity of IL-17A/F heterodimer
Normal Human Dermal Fibroblasts (NHDF; Lonza) cells were seeded into a 48-well flat bottom tissue culture plate at 10,000 cells per well in FGM-2 medium (Lonza) and incubated overnight
(31° , 5% C02) . Following incubation, 50 ng/mL final
concentration (1.47 nM) of rhIL-17A/F heterodimer (R&D Systems) was pre-incubated with a dilution series (30 μg/mL - 0.5 ng/mL) of mAb6785 or control antibodies for 10 minutes at room
temperature, and added to cells. Cells were incubated for 48h
(37°, 5% C02) and culture supernatants were collected and assayed by ELISA for IL-6 content using Human IL-6 Duo Sets (R&D Systems, Inc.) according to manufacturer's instructions. IC50 values were determined by non-linear regression using GraphPad Prism software
(GraphPad Software, Inc) . mAb6785 inhibited IL-17A/F heterodimer -induced IL-6 production by NHDF with EC50 2 ± 2.5 nM.
Methods
Determination of Picomolar Affinities Using Solution Equilibrium Titration (SET)
For KD determination by solution equilibrium titration
(SET) , monomer fractions (at least 90% monomer content, analyzed by analytical SEC; Superdex75 column, GE) of Fab protein were used .
Electrochemiluminescence (ECL) based affinity determination in solution and data evaluation were basically performed as described previously (Haenel et al . , Anal Biochem 339:182-4, 2005) . A defined fixed concentration of purified Fab (-10-100 pM) was incubated with increasing concentrations of IL-17Amut6 (highest concentration of 5 nM) in solution until chemical equilibrium was achieved. To quantify the unbound Fab in solution the samples were transferred to a Streptavidin MSD 384- well microtiter plate (Meso Scale Discovery, Gaithersburg, MD) with coated biotinylated IL-17Amut6. For detection, a ruthenium complex-labeled anti-human Fab/IgG antibody was applied and the plates were read with the Sector™ Imager 6000 (MSD) . Titration curves (concentration of free Fab as a function of antigen concentration) were plotted and fitted with Excel / XLfit software using the model described below.
For data evaluation for KD determination of Fab molecules, the following fit model was used (modified according to Abraham et al. J Mol Recognit. 9:456-461, 1996) :
y=Bmax- (Bmax/ (2*cFab) * (x +cFab+KD-sqrt ( (x+cFab+ KD) * (x+cFab+ KD) - 4*x*cFab) ) )
Whereas :
Bmax: maximum binding signal (at antigen concentration^ ) cFab: applied Fab concentration
x: applied total soluble antigen concentration (binding sites) sqrt: square root
KD: Equilibrium dissociation constant
Inhibition of IL-17A binding to IL-17RA (e.g. "IL-17RA
inhibition" assay
Clear maxisorp plates were coated with 100 μΐ/well of 2.5 μg/ml human IL-17RA-Fc (R&D Systems, Minneapolis, MN) in 0.1 M sodium carbonate-bicarbonate buffer, pH 9.4 and incubated overnight at 4°C. After blocking and washes, 25 ng/ml of biotinylated human IL-17mut6 (SEQ ID NO: 106) or cynomolgus IL- 17A (SEQ ID NO: 108) was pre-incubated with tested mAbs or control mAbs (30 to 0 μg/ml final concentration) in a combined volume of 100 μΐ for 5-10 minutes, and then added to plates. The signal was detected with 100 μΐ of 1:10,000 dilution of 1 mg/ml SA-HRP (Jackson Immunoresearch, West Grove, PA) for 20 minutes at room temperature (RT) followed by 100 μΐ/well of OPD substrate (Sigma-Aldrich Corp., St. Louis, MO) . The plates were read at 492 nm (Envision, PerkinElmer, Waltham, MA) . Fab binding to IL- 17RA was tested as described for mAbs.
Inhibition of IL-8 and IL-6 production from NHDF cells (e.g. "IL- 8 production" and "IL-6 production" assays)
Effect of inhibition of anti-IL-17A mAbs on IL-8 and IL-6 production was assessed in normal human dermal fibroblasts
(NHDF) . Cells were plated in a 48-well flat bottom tissue culture plate at 0.1x10s cells per well, 250 μΐ per well in FGM-2 medium and incubated overnight (37°, 5% C02) . Following
incubation, 0.1 ng/ml human TNF-a was added to all wells. 10 ng/ml IL-17mut6 or 25 ng/ml cynomolgus IL-17A was pre-incubated with tested mAbs or control mAbs (30-0 μg/ml final
concentration) in a combined volume of 250 μΐ for 10 minutes at RT, and then added to 250 μΐ of cells. In the assays, IL-17mut6 samples with no added antibody were included as control samples, while samples consisting of TNF-a or culture medium only were included as negative controls. Cells were incubated for 24 hours
(37°, 5% C02) and conditioned media were collected and assayed by ELISA for IL-6 and IL-8 using human IL-6 & IL-8 ELISA Duo Sets according to the manufacturer's instructions (R&D Systems,
Minneapolis, MN) . The Fabs were assayed as described fro the mAbs . Inhibition of IL-6 and G-CSF production from NHBE cells
Normal human bronchial epithelial (NHBE; Lonza) cells were seeded at 20,000 cells per well in BEGM media (Lonza) and incubated overnight (31° , 5% C02) . Following incubation, cells were stimulated with IL-17Amut6 for 48 hours in the presence of the tested antibodies at a range of concentrations (30 μg/mL - 0.5 ng.mL) . Supernatants were collected after the incubation and assayed for IL-6 or G-CSF content using a human IL-6- or G-CSF- specific ELISA (R&D Systems, Inc.) . IC50 values were determined by non-linear regression using GraphPad Prism software (GraphPad Software, Inc) .
Cross-reactivity with IL-17A family members
Clear maxisorp plates were coated with 100 μΐ/well of 5 μg/ml mAbs or isotype control mAbs in PBS, and incubated
overnight at 4°C. The plates were blocked with 200 μΐ/well for 1 hour with ELISA block buffer (1% BSA, 5% Sucrose in PBS with 0.05% NaN3) and washed three times with wash buffer (PBS, 0.01% Tween-20) . Competing cytokines were titrated in Assay Diluent Buffer (1% BSA in PBS) at 2x final concentration, and
biotinylated cytokine was prepared at 2x final concentration. 100 μΐ of cytokines at 2x final concentration were mixed (30-0 μg/ml final concentration) with 100 μΐ of biotinylated IL-17mut6 at 2x final concentration (25 ng/ml final concentration) in assay buffer. Recombinant human IL-23 (R&D Systems, Minneapolis, MN) was used as a negative control, buffer only sample as background control, and IL-17mut6 as positive control. 100 μΐ per well in duplicate of cytokine/biotinylated IL-17mut6 mixture was added to the plate and incubated for 1-2 hours. Plates were washed three times with wash buffer, and incubated with 100 μΐ of 1:10,000 dilution of 1 mg/ml SA-HRP (Jackson Immunoresearch, West Grove, PA) for 20 minutes at RT . Plates were washed three times with ELISA wash buffer. Following wash, 100 μΐ/well of OPD substrate (Sigma-Aldrich Corp., St. Louis, MO) was added to each well and incubated until the appropriate color change was detected. The reaction was stopped with the addition of 50 μΐ of 2N sulfuric acid, and the plates were read 492 nm using the Envision
instrument .
Affinity measurements - Biacore assay
Affinity measurements using Surface Plasmon Resonance (SPR) were performed using a Biacore 3000 optical biosensor (Biacore) . Selected Fabs (-30 RU) or mAbs (-50 RU) were captured onto the sensor chip surface using a sheep anti-Fd antibody or an anti- human Fc antibody for Fab or mAb capture, respectively. Capture of Fab or mAb was followed by injection of huIL-17mut6 or cyno IL-17A in solution (0.2 to 49 nM) .
Example 4
Epitope mapping
Antibody epitopes were deduced by a combination of
competition binding, H/D exchange analysis, and antibody-IL-17A co-structure (see Example 5) . The following antibodies were used: mAbl926, MORmAb7700, MORmAb7706, MORmAb7708, mAb7357 (a mouse anti-human IL-17A neutralizing antibody derived from hybridoma C1863) , mAb2832 (a mouse/human chimeric anti-human IL- 17A neutralizing antibody derived from hybridoma C1861), mAb317 (mouse anti- human IL-17A antibody, R&D Systems, Minneapolis, MN) and mAb3171 (mouse anti-human IL-17A antibody, R&D Systems, Minneapolis, MN) , and mAbeBI016-7178 (a mouse anti-human IL-17A antibody, e-Bioscience , San Diego, CA) . The three commercial antibodies showed varying degrees of neutralizing activity.
Competitive Epitope Binding
For competitive ELISA, 5 μΐ (20 g/ml) of IL-17Amut6 protein was coated on MSD HighBind plate (Meso Scale Discovery, Gaithersburg, MD) per well for 2 hr at room temperature. 150 μΐ of 5% MSD Blocker A buffer (Meso Scale Discovery, Gaithersburg, MD) was added to each well and incubated for 2 hr at room temperature. Plates were washed with 0.1 M HEPES buffer (pH 7.4) . Labeled antibody (MDS fluorescence dye), 10 nM, was incubated with increasing concentrations of competitor antibodies (1 nM - 2 μΜ) , and 25 μΐ of the mixture was added to the
designated wells. After 2-hour incubation with gentle shaking at RT, plates were washed as above, 150 μΐ diluted MSD Read Buffer T was added, and the plates were read with a MDS Sector Imager 6000.
Assays were carried out with labeled mAbl926, mAb317, mAb3171, or mAb7357 (Figure 4) . Based on competition assays, anti-IL-17A antibodies were assigned to four different bins. Bin A: mAbl926, MORmAb7706, and MORmAb7708; Bin B: eBiol6-7178 and mAb7357; Bin C: mAb317; Bin D: mAb3171.
H/D exchange analysis :
For H/D exchange, the procedure used to analyze the antibody perturbation was similar to that described previously (Hamuro et al . , J. Biomol. Techniques, 14:171-82, 2003; Horn et al . , Biochemistry, 45: 8488-98, 2006) with some modification. Recombinant IL-17Amut6 (expressed in HEK293E cells with C- terminal His-tag) was incubated in a deuterated water solution for pre-determined times resulting in deuterium incorporation at exchangeable hydrogen atoms. The deuterated IL-17Amut6 was captured on a column containing immobilized individual anti-IL- 17A mAbs and then washed with aqueous buffer. The back-exchanged IL-17Amut6 protein was eluted from the column and localization of deuterium containing fragments was determined by protease digestion and mass spec analysis. Regions bound to the antibody were inferred to be those sites relatively protected from exchange and thus containing a higher fraction of deuterium, compared to IL-17Amut6 not complexed with antibody. H/D exchange perturbation maps of IL-17Amut6 are shown in Figure 5. The numbers on top of the bars refer to IL-17Amut6 resiudes.
MORmAb7700, MORmAb7706 and MORmAb7708 showed varying degrees of differential exchange for three segments of IL-17A (SEQ ID NO: 105) 45NRS TS PWNLH54 (SEQ ID NO: 159), 56NEDPERYPSVIWE68 (SEQ ID NO: 157) and i00RREPPHCPNSFRLEKILii6 (SEQ ID NO: 158), indicating protection by the antibodies. The 56NEDPERYPSVIWE68 (SEQ ID NO: 157) fragment was strongly protected by MORmAb7708, weakly protected by MORmAb7700, and not protected by MORmAb7706. The overlap in the fragment protection patterns of these
antibodies is consistent with their cross-inhibition in the competition assays described above.
For both mAb7357 and mAbeBiol6-7178, strong protection was observed for 7iCRHLGCINADGNVDYHM87 (SEQ ID NO: 160) consistent with their cross-inhibition in the competition assays described above. Weak, and therefore inconclusive, differential exchange was observed for other fragments with mAb7357, mAb2832, mAb317 and mAb3171.
The H/D exchange studies localized the binding sites for two of the four competition groups defined above. Bin A antibodies (MORmAb7700, MORmAb7706 and MORmAb7708) bound in the region of peptide segments 45NRS TS PWNLH54 , (SEQ ID NO: 159), 56NEDPERYPSVIWE68 (SEQ ID NO: 157) and i00RREPPHCPNSFRLEKILii6 (SEQ ID NO: 158), of SEQ ID NO: 105, and Bin B antibodies (mAb7357 and mAbeBiol 6-7178 ) bound in region of peptide segment
71CRHLGCINADGNVDYHM87 (SEQ ID NO: 160) . mAb317 and mAb3171 bound to sites distinct from each other and from the bin A and bin B antibodies. However, the weak signals in the H/D exchange studies with both antibodies did not provide sufficient evidence to localize their epitopes on IL-17A.
Example 5
Co-crystal structure of IL-17A and anti-IL-17A antibody
The co-structure of IL-17Amut6 with the Fab6468, a recombinant His6-tagged Fab of mAb6785, was determined by X-ray crystallography. The amino acid sequence of the light chain of Fab6468 is shown in SEQ ID NO: 90, and the heavy chain amino acid sequence is shown in SEQ ID NO: 111. In the Example 5, the IL- 17A amino acid residues referred to indicate residues according to SEQ ID NO: 105, and the Fab6468 residues referred to indicate light chain variable region residues according to SEQ ID NO: 79 and heavy chain variable region residues according to SEQ ID NO: 86. The expression, refolding and purification of recombinant human IL-17Amut6 has been described (Wu et al . , Cytokine, ePub ahead of print Jul29) . Fab6468 was expressed in HEK-293F cells and purified using a similar method as described (Zhao et al . , Protein Expr Purif, 67:182-9, 2009) .
Crystallization of IL-17A/Fab6468 complex
The IL-17A/Fab6468 complex was prepared by mixing IL- 17Amut6 and Fab6468 in 1:1.1 molar ratio in 20 mM MES pH 6.5, 0.2 M NaCl, and 10% glycerol and incubated over night at 4°C. The complex was purified from excess un-complexed Fab using size exclusion chromatography (SEC) on a Superdex 200 10/300 GL column (GE Healthcare, Piscataway, NJ) in 20 mM MES pH 6.5, 0.2 M NaCl, and 10% glycerol. Fractions corresponding to the complex were pooled and concentrated with an Amicon Ultra 10000 MWCO device to 4.6 mg/ml .
Automated crystallization screening was performed using the Oryx4 automatic protein crystallization robot (Douglas
Instruments, East Garston, UK) dispensing equal volumes of protein and reservoir solution in a sitting drop format using Corning plate 3550 (Corning Inc., Corning, NY) . Initial
screening was performed with Hampton Crystal Screen HT (HR2-130, Hampton Research) and produced needle-like crystals from several conditions containing ammonium sulfate, PEGs at pH 4.5-4.6.
These small crystals were used to produce a seed stock for microseed-matrix screening (MMS) (D'Arcy et al . , Acta
Crystallographica Section D, 63:550-4, 2007) . Diffraction quality crystals were obtained from the MMS screen in 0.1 M Sodium Acetate pH 5.5, 12% PEG MME 5000 and 0.2 M Lithium
Sulfate.
X-ray data collection of IL-17A/Fab6468 complex
For X-ray data collection, the crystal was soaked for a few seconds in the mother liquor supplemented with 24% glycerol, and flash frozen in the stream of nitrogen at 95°K. X-ray
diffraction data were collected and processed using a Rigaku MicroMax™-007HF microfocus X-ray generator equipped with an Osmic™ VariMax™ confocal optics, Saturn 944 CCD detector, and an X-stream™ 2000 cryocooling system (Rigaku, Woodlands, TX) .
Diffraction intensities were detected over a 254° crystal rotation with the exposure time of 3 min per half-degree image to the maximum resolution of 2.2 A. The X-ray data were processed with the program D*TREK (Pflugrath, J. , Acta Crystallographica Section D, 55:1718-25, 1999) . The crystal belonged to the monoclinic space group P2X with a=73.40 A, b=64.04 A, c=145.61 A and β=95.39°. The X-ray data statistics are given in Table 8.
Figure imgf000062_0001
*Values for highest resolution shell are in ()'s.
Structure Determination
The crystal structure of IL-17A/Fab6468 was determined by molecular replacement using Phaser (Read, Acta Crystallogr D Biol Crystallogr, 57:1373-82, 2001) . The search models were IL-17F (PDB ID 1JPY) (Hymowitz et al . , EMBO J., 20:5332-41, 2001) and a homology model for the Fv (VH/VL) , which was constructed based upon the anti-IL-13 antibody CNTO607 (PDB ID 3G6A) (Teplyakov et al., J. Mol. Biol. 389:115-23, 2009) for both the VH and VL, using Modeller (Accelrys, CA) . The two constant domains CL/CH1 were taken from PDB ID 8FAB (Strong et al . , Biochemistry,
30:3739-48, 1991) . The structure refinement was carried with PHENIX (Adams et al . , J. Synchrotron. Radiat. 11:53-5, 2004) . The two-fold non-crystallographic symmetry was initially imposed in early stages of refinement but was relaxed in the final stages based upon Rfree · Model adjustment and manual rebuilding were done using COOT (Emsley et al . , Acta Crystallogr. D. Biol.
Crystallogr. 60:2126-32, 2004) . The final Rcryst and Rfree were 23.4% and 29.7%, respectively, for all 61,706 independent reflections to 2.2A. The refinement statistics are given in Table 9.
Figure imgf000063_0001
The IL-17A/Fab6468 complex structure
The structure of the complex was determined to high
resolution (~ 2.2 A) . IL-17A was a nearly symmetrical homo-dimer in the crystal and bound two Fab molecules . The antibody-antigen interactions were largely hydrophobic and in contrast to most antibodies, the light chain CDRs made a number of the important contacts. The overall molecular structure of IL-17A/Fab6468 complex is shown in Figure 6A. The monomer of the IL-17A dimer adopted the overall topology of a cystine knot (Figure 6B) . The two monomers were very similar with a C RMSD of 0.54 A for 77 backbone Ca atoms. The overall architecture of IL-17A monomer cystine knot was very similar to that of IL-17F with an rmsd of 0.71 A for 76 Ca atoms (Figure 6B) . Each IL-17A monomer was stabilized by three disulfide bonds. For chain B, three intra- chain disulfide bonds were observed (C10-C106, C71-C121, C76-C123) , whereas for chain A the C10-C106 disulfide bond was not observed due to disorder in these segments of the monomer. The latter two disulfide bonds (C71-C121, C76-C123) stabilized the cystine knot architecture, analogous to IL-17F and NGF. The structural model for chain B of IL-17A included all residues 10-128 (residues 1-9 were disordered) , whereas for chain A residues only residues 21- 29, 41-104 and 109-127 were observed and the other residues 1-20, 30-40, 105-108 and 128 were missing due to disorder in the structure. For the two Fabs, residues 1-2 of both the light chains were disordered or had poor electron density. The C- terminal 3 residues of both the heavy and light chains, including the inter-chain disulfide bonds as well as the His tag on the heavy chain were disordered.
The ordered N-terminal segment of IL-17A (chain B)
contained a short helical element (residues 8-12) . It folded back towards the loop 3-4 of the same monomer and formed an intra-chain disulfide bond (C10-C106) . In contrast, the equivalent segment of IL-17F reached over to the other monomer of the dimer and formed an inter-chain disulfide bond and linked the two monomers covalently. The ordered parts of the segments 17-39 of the two IL-17A monomers were swapped, as in IL-17F. This swapping resulted in a cross-over for these parts of the IL-17A dimer. Combined with the intra-molecular disulfide bond (C10- C106) , the two N-terminal segments of IL-17A formed two interlocked monomers, which also gave rise to an apparent dimer of 26 kD on non-reducing SDS-PAGE.
The dimer of the IL-17A was nearly symmetrical for the four main β-strands (strands 1-4) (Figure 6C) . The C rmsd for 76 residues is 0.71 A. The slight asymmetry came from two sources. First, the chain A contained a number of disordered segments, mainly in N-terminus . Only a short β-strand (strand 0, residues 22-26) was apparently ordered, whereas residues 10-40 of chain B were ordered with a helical segment (residues 12-16) and a β- strand (strand 0, residues 21-25) . Second, while the cystine knotted four main β-strands of the two monomers (40-128) of the IL-17A were related by a two-fold symmetry of rotation, the ordered parts of strand 0 did not superimpose well when the main body is overlaid (not shown) . Whether this was an artifact of protein refolding or such an arrangement exists in nature is not clear without further investigation. The bioactivity of this species was similar to that of a reference IL-17A from a
commercial source (also produced in E. coli) (R&D Systems,
Minneapolis, MN) suggesting that the inter- or intra- chain disulfide linkage for the C10-C106 is not important for its receptor binding. Nevertheless, the current structure suggests that the N-terminal segments (1-20) and (30-39) are very flexible and their structures do not impact the activity of the folded IL- 17A dimer.
The epitope and the paratope
The residues involved in binding between IL-17A and Fab6468 are listed in Table 10. Due to the missing residues in protomer A in the IL-17A and the slight asymmetrical nature of the IL-17A dimer, all epitope residues from the two contact sites were not identical (Table 10 and Figure 7) . However, there was a core set of residues as well as their interactions that were identical. These residues were L26, R55, E57, P59, E60, R61, Y62, S64, V65, W67, R101, E102, P103 and F110 of IL-17A SEQ ID NO: 105
(highlighted black in Table 10), and they constitute the core epitope for Fab6468.
Table 10. Core epitope residues are highlighted in black. Core paratope residues are bolded. Extended paratope residues are shown in parenthesis.
Figure imgf000066_0001
Similarly, the contact residues from the antibodies in the two sites were not all identical. The residues involved in identical contacts to the core epitope residues are referred to as the "core paratope", which was composed of the following residues: Light Chain (LC) : Y31, D49, Y90, F92, F93 (SEQ ID NO: 79); and heavy chain (HC) : S52, T54, F57, Y59, Q99, L100 and T101 (SEQ ID NO: 86) (Table 10) . The core paratope residues are shown in bold in Table 10. The additional "extended paratope" residues identified in one monomer binding a specific IL-17A residue are shown in parenthesis.
The H/D protection data for the MORmAb7700 was in agreement with the co-crystal studies, as all core epitope residues identified in the co-crystal structure except L26 were within or at the borders of two of the protected segments identified by H/D exchange, 56NEDPERYPSVIWE68 (SEQ ID NO: 157) and
100RREPPHCPNSFRLEKIL116 (SEQ ID NO: 158) for the MORmAb7700. All of the MORmAb7700 antibody derivatives, including MORmAb8302 and mAbl926, are assumed to have the same binding specificity as Fab6468 since they differ at most by one residue in the N- terminal region of VH (see Example 1), 3 residues at the N- terminus of VL (see Example 1) , and 3 CDR residues (one each in H2 , H3 and L3, Table la) , none of which are part of the antibody paratope .
The IL-17A structure characterized in this invention is very similar to the previously published structure, except that due to missing segments, the P2 pocket cavity (see below) was not identified in the previous work (structure 2VXS, available at the Protein DataBank http //www rcsb org/pdb/home/home do; Gerhardt et al., J. Mol. Biol. 394:905-21, 2009) .
The crystal structure of the human IL-17F in complex with IL-17RA has been reported (Ely et al . , Nat. Immunology, 10:1245- 51, 2009) . Because of the sequence and structural similarities between IL-17A and IL-17F, it is likely that IL-17A will interact with the IL-17RA in a similar manner to IL-17F. Molecular modeling by overlaying the IL-17A structure in complex with Fab6468 obtained in this study onto the IL-17F in the reported IL-17F/IL-17RA complex showed that segments of Fab6468 would have steric clashes with IL-17RA. One of these segments localize around the FF motif (residues 92 and 93 of SEQ ID NO: 79) in the light chain CDR3 of Fab6468. Thus, not wishing to be bound by any particular theory, it is suggested that Fab6468 would inhibit IL-17A function by blocking its interactions with IL-17RA and by analogy, IL-17RC, though the mode of interaction between IL-17RC and IL-17A is not known at the molecular level.
The significant differences in the affinities of IL-17A and IL-17F for IL-17RA suggest there may be significant differences in the details of IL-17A and IL-17RA interactions, the extent of which will only be available when the co-crystal structure of IL- 17A/IL-17RA is determined. This is implicated by the
identification of the P2 pocket cavity in this study, which is only partially identified in IL-17F analogous region in the reported IL-17F/IL-17RA crystal structure (Ely et al . , Nat.
Immunology, 10:1245-51, 2009) .
Two deep, largely hydrophobic pockets were identified on the surface of IL-17A along the dimer interface (Figure 8A, 5B) . The PI pocket, which is analogous to a pocket first discovered in IL-17F (Hymowitz et al . , EMBO J, 20: 5332-41, 2001), is composed of residues Q94, E95, L97 and K114 of monomer A and L53, Y62, P63, V65, 166, W67, 196, V117 and V119 of monomer B, and vice versa. On one side of the dimer, the PI pocket is partially covered by the segment 30-40, whereas on the other side it was completely open due to the segment being disordered. Since this segment appears to be flexible, the PI pocket would be accessible by other molecules. The P2 pocket is also composed of residues from both chains: V24, L26, 128, Y62, L99, R101, F110 and LI 12 of monomer A and V22, V24 and L112 of monomer B, and vice versa.
Though the details of the P2 pockets are slightly
different due to the asymmetry of the IL-17A dimer as described above, the overall geometry of the two P2 pockets is very similar. The two sets of residues lining the PI and P2 pockets are very well conserved between IL-17A and IL-17F (Figure 8C) . However, in the IL-17F structure, the P2 pocket is occupied by residues F10 and Fll (Fi0Fn motif) (Figure 8B) (Hymowitz et al . , EMBO J, 20: 5332-41, 2001) . The FF motif is absent in human IL- 17A; instead, the corresponding amino acid residues are 14 and P5 (residues 4 and 5 in SEQ ID NO: 105) (Figure 8C) . These residues are not likely to bind in the P2 pocket as well as the FF motif because they are much smaller than the phenylalanine residues, and most likely will not have sufficient affinity for the P2 pocket. Thus, the FF motif of IL-17F is likely a structural discriminant for human IL-17A and IL-17F interactions with receptors IL-17RA and IL-17RC. It is likely that both of these largely hydrophobic pockets (PI and P2 ) are required for IL-17A binding to IL-17RA. The recent crystal structure of IL-17F/IL- 17RA complex shows that the FF motif is displaced by IL-17RA (Ely et al . , Nat. Immunology 10:1245-51, 2009) . The energetic penalty of FF motif eviction from P2 likely results in lower binding affinity. This is consistent with the observations that IL-17RA binds IL-17A with high affinity but IL-17F with low affinity in humans (Kuestner et al . , J Immunol, 179:5462-73, 2007), and potentially could explain the differences in IL-17A and IL-17F potencies. In mice, the FF motif is absent in both IL-17A and IL-17F, and is replaced by residues IP and AL, respectively. The AL and IP residue pairs are small and likely to have low affinity for the P2 pocket. Thus, P2 would be available in mouse IL-17A and IL-17F for IL-17RA binding. Both mouse IL-17A and IL-17F bind mouse IL-17RA with similar affinity (Kuestner et al . , J Immunol, 179:5462-73, 2007), consistent with the present
suggestion that availability of P2 pocket for binding increases affinity of the ligands.
Overall, the structural differences observed between IL-17A and IL-17F provides a basis for dissecting their interactions with respective receptors. Furthermore, it is conceivable that peptides, peptidomimetics and small molecules can be designed to bind in either or both pockets to block IL-17A and/or IL-17F from interacting with their receptors. Since the FF motif present in Fab6468 (residues F92 and F93 in SEQ ID NO: 79) binds P2 pocket residues L26, R61, L99, R101 and R102, the Fab 6468 structure could be used to select and optimize additional IL-17A
antagonists, such as peptides from randomized or designed peptide libraries using phage display. The residues lining the PI and P2 pockets are well
conserved between IL-17A and IL-17F and molecular modeling suggests that an IL-17A/F heterodimer would adopt a nearly identical overall structure when compared to the IL-17A homodimer alone. Therfore, it is likely that the PI and P2 pockets are present in the IL-17A/F heterodimer with similar overall topology and constitute its receptor binding sites. Thus IL-17A
antagonists binding to the P2 pocket residues could bind and antagonize the IL-17A/F heterodimer.
Example 6
Cross-species binding specificity
To evaluate cross-species binding specificity of mAbtrl926, a binding ELISA was performed with different IL-17A proteins coated on micro-titer plates. Human, mouse and rat IL-17A proteins were coated on the micro-titer plates. Serial dilutions of labeled mAbl926 were incubated at 37°C for 2 hours. Following incubation, micro-titer plates were washed thoroughly, and bound labeled mAbl926 was detected. mAb 1926 bound to human IL-17A much stronger than to rat or mouse IL-17A proteins (Figure 9) . This reduced binding to rat and mouse IL-17A is consistent with these proteins both differing from human IL-17A at 7 positions of the Fab6468 extended epitope (Table 10) . In addition, there is a one amino acid insertion in rat and mouse IL-17A between residues 40 and 41 of human IL-17A, a position close to part of the Fab 6468 epitope.

Claims

1. An isolated antibody or fragment thereof, wherein the antibody binds specifically to human IL-17A having the sequence shown in SEQ ID NO: 105 at amino acid residues 56-68 (SEQ ID NO: 157) and 100-116 (SEQ ID NO: 158); or at residues L26, R55, E57, P59, E60, R61, Y62, S64, V65, W67, R101, E102, P103 and F110.
2. An isolated antibody or fragment thereof, wherein the antibody binds specifically to a P2 pocket cavity on human IL-17A, the P2 pocket cavity comprising amino acid residues V22, V24, L26, 128, Y62, L99, R101, F110, and L112 of SEQ ID NO: 105.
3. An isolated antibody or fragment that binds specifically to human IL-17A that competes for human IL-17A binding with a monoclonal antibody comprising the heavy chain CDR 1, 2 and 3 (HCDR1, HCDR2 and HCDR3) amino acid sequences as shown in SEQ ID NOs : 25, 43 and 60, respectively, and the light chain CDR 1, 2 and 3 (LCDR1, LCDR2 and LCDR3) amino acid sequences as shown in SEQ ID NOs: 3, 6 and 18, respectively.
4. An isolated antibody or fragment that binds specifically to human IL-17A, comprising heavy chain variable region paratope amino acid residues that interact with residues of human IL- 17A having the amino acid sequence shown in SEQ ID NO: 105, comprising :
a. a first threonine residue that interacts with R55 or E57 of human IL-17A;
b. a glutamine residue that interacts with R55 or E57 of human IL-17A;
c. a lysine residue that interacts with E57 of human IL-17A; d. a tyrosine residue that interacts with P59, E60 or R101 of human IL-17A;
e. a phenylalanine residue that interacts with E60, R101, E102 or P103 of human IL-17A;
f. a serine residue that interacts with E60 of human IL-17A; and
g. a second threonine residue that interacts with E60 of human IL-17A.
5. An isolated antibody or fragment that binds speficially to
human IL-17A, comprising light chain variable region paratope amino acid residues that interact with residues of human IL- 17A having the amino acid sequence shown in SEQ ID NO: 105, comprising :
a. a first phenylalanine residue that interacts with L26 of human IL-17A;
b. an aspartic acid residue that interacts with R55 or W67 of human IL-17A;
c. a first tyrosine residue that interacts with P59, S64 or R101 of human IL-17A;
d. a second phenylalanine residue that interacts with P59, E60, R61, Y62, R101 or F110 of human IL-17A; and e. a second tyrosine residue that interacts with V65 of
human IL-17A.
6. An isolated antibody or fragment that binds specifically to human IL-17A, comprising the heavy chain variable region paratope of claim 4 and the light chain variable region paratope of claim 5.
7. An isolated antibody or fragment that binds specifically to human IL-17A, comprising a heavy chain variable region and a light chain variable region, wherein the antibody comprises: a. a heavy chain variable region paratope selected from
Chothia residues S51, T53, F56, Y58, Q95, L96 and T97; or b. a light chain variable region paratope selected from
Chothia residues Y32, D50, Y91, F93 and F94; or
c. a heavy chain variable region paratope selected from Chothia residues S51, T53, F56, Y58, Q95, L96 and T97 and d. a light chain variable region paratope selected from
Chothia residues Y32, D50, Y91, F93 and F94.
8. An isolated antibody or fragment that binds specifically to human IL-17A, comprising heavy chain variable region paratope amino acid residues and light chain variable region paratope amino acid residues that interact with residues of human IL- 17A having the amino acid sequence shown in SEQ ID NO: 105, comprising :
a. a tyrosine residue in the heavy chain variable region that interacts with R101 of human IL-17A;
b. a phenylalanine residue in the heavy chain variable
region that interacts with R101 of human IL-17A; c. a first phenylalanine residue in the light chain variable region that interacts with Y62 and R101 of human IL-17A; d. a second phenylalanine residue in the light chain
variable region that interacts with L26 and F110 of human IL-17A; and
e. a tyrosine residue in the light chain variable region that interacts with R101 of human IL-17A.
9. An isolated antibody or fragment that binds specifically to human IL-17A, comprising a heavy chain variable region and a light chain variable region, wherein the antibody comprises: a. a heavy chain variable region paratope selected from
Chothia residues F56 and Y58; and
b. a light chain variable region paratope selected from
Chothia residues Y91, F93 and F94.
10. An isolated antibody or fragment thereof that binds
specifically to human IL-17A, comprising a heavy chain variable region (VH) and a light chain variable region (VL) , wherein the antibody comprises:
a. the heavy chain complementarity determining region (CDR) 1, 2 and 3 (HCDR1, HCDR2, HCDR3) amino acid sequences as shown in SEQ ID NOs: 23, 35 and 52, respectively, wherein the HCDR2 of SEQ ID NO: 35 is further defined as shown in Formula ( I ) :
Figure imgf000074_0002
wherein
Xaai may be His, Met, Arg, Ser or Tyr;
Xaa2 may be Trp, Thr or Tyr; and
Xaa3 may be Tyr, Phe, Ser or Asp; or
b. the HCDR1, HCDR2, and HCDR3 amino acid sequences as shown in SEQ ID NOs: 23, 35 and 52, respectively, wherein the HCDR2 of SEQ ID NO: 35 is further defined as shown in Formula ( I ) :
Figure imgf000074_0003
wherein
Xaai may be His, Met, Arg, Ser or Tyr;
Xaa2 may be Trp, Thr or Tyr; and
Xaa3 may be Tyr, Phe, Ser or Asp; and the light chain complementarity determining reigon (CDR) 1, 2 and 3 (LCDR1, LCDR2, LCDR3) amino acid sequences as shown in SEQ ID NOs: 1, 4 and 7,
respectively; or c. the LCDR1, LCDR2, and LCDR3 amino acid sequences as shown in SEQ ID NOs: 2, 5 and 11, respectively, wherein the LCDR3 of SEQ ID NO: 11 is further defined as shown in Formula (II) :
Figure imgf000074_0001
wherein
Xaa4 may be His or Gin;
Xaa5 may be Phe or Gly;
Xaa6 may be Thr, Val or Asn;
Xaa7 may be lie, Thr or Tyr; Xaa8 may be Pro or Arg;
Xaa9 may be Ser or Pro; and
Xaaio may be His, Phe or Leu; or
d. the LCDR1, LCDR2, and LCDR3 amino acid sequences as shown in SEQ ID NOs: 2, 5 and 11, respectively, wherein the LCDR3 of SEQ ID NO: 11 is further defined as shown in Formula (II) :
Figure imgf000075_0001
wherein
Xaa4 may be His or Gin;
Xaa5 may be Phe or Gly;
Xaa6 may be Thr, Val or Asn;
Xaa7 may be lie, Thr or Tyr;
Xaa8 may be Pro or Arg;
Xaa9 may be Ser or Pro; and
Xaaio may be His, Phe or Leu; and
the HCDR1, HCDR2, and HCDR3 amino acid sequences as shown in SEQ ID NOs: 24, 36 and 53, respectively; or e. the HCDR1, HCDR2, and HCDR3 amino acid sequences as shown in SEQ ID NOs: 24, 36 and 57, respectively, wherein the HCDR3 of SEQ ID NO: 57 is further defined as shown in Formula (IV) :
Figure imgf000075_0002
wherein
Xaaig is Met, lie, Leu or Thr; or
f. the LCDR1, LCDR2, and LCDR3 amino acid sequences as shown in SEQ ID NOs: 2, 5 and 17, respectively, wherein the LCDR3 of SEQ ID NO: 17 is further defined as shown in Formula (III) :
Figure imgf000075_0003
wherein Xaa may be Gin or Thr;
Xaai2 may be Ser or Tyr;
Xaai3 may be Asn, Arg, Val or Tyr;
Xaai4 may be His or Ser;
Xaai5 may be lie, Thr, Leu, Ala or Ser;
Xaai6 may be Pro, Leu or Ser;
Xaai7 may be Pro, Ser, Phe or Leu; and
Xaaie may be Ala, Leu or Asp; or
g. the HCDR1, HCDR2 and HCDR3 amino acid sequences as shown in SEQ ID NOs: 24, 36 and 57, respectively, wherein the HCDR3 of SEQ ID NO: 57 is further defined as shown in Formula (IV) :
Figure imgf000076_0001
wherein
Xaaig is Met, lie, Leu or Thr; and
the LCDR1, LCDR2, and LCDR3 amino acid sequences as shown in SEQ ID NOs: 2, 5 and 17, respectively, wherein the LCDR3 of SEQ ID NO: 17 is further defined as shown in Formula (III) :
Figure imgf000076_0002
wherein
Xaan may be Gin or Thr;
Xaai2 may be Ser or Tyr;
Xaai3 may be Asn, Arg, Val or Tyr;
Xaai4 may be His or Ser;
Xaai5 may be lie, Thr, Leu, Ala or Ser; Xaai 6 may be Pro, Leu or Ser;
Xaai7 may be Pro, Ser, Phe or Leu; and Xaaie may be Ala, Leu or Asp; or
h. the HCDR1, HCDR2 and HCDR3 amino acid sequences as shown in SEQ ID NOs: 25, 46 and 61, respectively, wherein the HCDR2 of SEQ ID NO: 46 is further defined as shown in Formula (VI) :
Figure imgf000077_0001
wherein
Xaa2i may be Ala, Gly, Thr or Val;
Xaa22 may be Asn or Ser;
Xaa23 may be Gly, Met, Lys, lie, Leu or His;
Xaa24 may be Leu, Asp, Ala, His, Thr, Gly or Ser;
Xaa25 may be Gly or Ser;
Xaa26 may be Thr, Gly, Tyr or Asp;
Xaa27 may be His, Trp, Tyr or Phe;
Xaa28 may be Lys, Thr or lie; and
Xaa29 may be Tyr, Phe or Asn; and
the HCDR3 of SEQ ID NO: 61 is further defined as shown in Formula (VII) :
Figure imgf000077_0003
wherein
Xaa30 may be Met, Leu or Thr; or
the LCDR1, LCDR2, and LCDR3 amino acid sequences as shown in SEQ ID NOs: 3, 6 and 22, respectively, wherein the LCDR3 of SEQ ID NO: 22 is further defined as shown in Formula (V) :
Figure imgf000077_0002
wherein
Xaa20 is Met, Leu, Thr or Tyr; or
the HCDR1, HCDR2 and HCDR3 amino acid sequences as shown in SEQ ID NOs: 25, 46 and 61, respectively, wherein the HCDR2 of SEQ ID NO: 46 is further defined as shown in Formula (VI) :
Figure imgf000077_0004
Figure imgf000078_0001
wherein
Xaa2i may be Ala, Gly, Thr or Val;
Xaa22 may be Asn or Ser;
Xaa23 may be Gly, Met, Lys, lie, Leu or His;
Xaa24 may be Leu, Asp, Ala, His, Thr, Gly or Ser;
Xaa25 may be Gly or Ser;
Xaa26 may be Thr, Gly, Tyr or Asp;
Xaa27 may be His, Trp, Tyr or Phe;
Xaa28 may be Lys, Thr or lie; and
Xaa29 may be Tyr, Phe or Asn; and
the HCDR3 of SEQ ID NO: 61 is further defined as shown in Formula (VII) :
Figure imgf000078_0002
wherein
Xaa30 may be Met, Leu or Thr; and
the LCDR1, LCDR2, and LCDR3 amino acid sequences as shown in SEQ ID NOs: 3, 6 and 22, respectively, wherein the LCDR3 of SEQ ID NO: 22 is further defined as shown in Formula (V) :
Figure imgf000078_0003
wherein
Xaa20 is Met, Leu, Thr or Tyr; or the HCDR1, HCDR2 and HCDR3 amino acid sequences as shown in SEQ ID NOs: 25, 51 and 58, respectively, wherein the HCDR2 of SEQ ID NO: 51 is further defined as shown in Formula (VIII) :
Figure imgf000078_0004
wherein
Xaa3i may be Ala, Lys, Met or His;
Xaa32 may be Asn, Met, Thr or Arg; Xaa33 may be Gly or Asp;
Xaa34 may be Arg, His or Asn; and
Xaa35 may be Asp or Gly; or
1. the HCDRl, HCDR2 and HCDR3 amino acid sequences as shown in SEQ ID NOs: 25, 51 and 58, respectively, wherein the HCDR2 of SEQ ID NO: 51 is further defined as shown in Formula (VIII) :
Figure imgf000079_0001
wherein
Xaa3i may be Ala, Lys, Met or His;
Xaa32 may be Asn, Met, Thr or Arg;
Xaa33 may be Gly or Asp;
Xaa34 may be Arg, His or Asn; and
Xaa35 may be Asp or Gly; and the LCDRl, LCDR2, and LCDR3 amino acid sequences as shown in SEQ ID NOs: 3, 6 and 18, respectively.
An isolated antibody or fragment thereof that binds specifically to human IL-17A, comprising a VH and a VL region, wherein the antibody comprises the HCDRl, HCDR2 and HCDR3 amino acid sequences as shown in
a . SEQ ID NOs : 23, 26 and 52, respectively;
b. SEQ ID NOs : 23, 27 and 52, respectively;
c . SEQ ID NOs : 23, 28 and 52, respectively;
d. SEQ ID NOs : 23, 29 and 52, respectively;
e . SEQ ID NOs : 23, 30 and 52, respectively;
f . SEQ ID NOs : 23, 31 and 52, respectively;
g. SEQ ID NOs : 23, 32 and 52, respectively;
h. SEQ ID NOs : 23, 33 and 52, respectively; or
i . SEQ ID NOs : 23, 34 and 52, respectively .
12. The isolated antibody or fragment of claim 11, wherein the antibody further comprises the LCDRl, LCDR2, and LCDR3 amino acid sequences as shown in SEQ ID NOs: 1, 4 and 7, respectively .
13. An isolated antibody or fragment that binds specifically to human IL-17A, comprising a VH and a VL, wherein in the antibody comprises the LCDRl, LCDR2, and LCDR3 amino acid sequences as shown in SEQ ID NOs: 1, 4 and 7, respectively.
14. An isolated antibody or fragment binds specifically to
human IL-17A, comprising a Vh and a VL, wherein the antibody comprises the LCDRl, LCDR2, and LCDR3 amino acid sequences as shown in:
a. SEQ ID NOs: 2, 5 and 8, respectively;
b. SEQ ID NOs: 2, 5 and 9, respectively; or
c. SEQ ID NOs: 2, 5 and 10, respectively.
15. The isolated antibody or fragment of claim 14, wherein the antibody further comprises the HCDRl, HCDR2 and HCDR3 amino acid sequences as shown in SEQ ID NOs: 24, 36 and 53, respectively .
16. An isolated antibody or fragment that binds specifically to human IL-17A, comprising a VH and a VL, wherein the antibody comprises the HCDRl, HCDR2 and HCDR3 amino acid sequences as shown in:
a. SEQ ID NOs: 24, 36 and 53, respectively;
b. SEQ ID NOs: 24, 36 and 54, respectively;
c. SEQ ID NOs: 24, 36 and 55, respectively; or
d. SEQ ID NOs: 24, 36 and 56, respectively.
17. The isolated antibody or fragment of claim 16, wherein the antibody further comprises the LCDRl, LCDR2, and LCDR3 amino acid sequences as shown in:
a. SEQ ID NOs: 2, 5 and 12, respectively;
b. SEQ ID NOs: 2, 5 and 13, respectively;
c. SEQ ID NOs: 2, 5 and 14, respectively;
d. SEQ ID NOs: 2, 5 and 15, respectively; or
e. SEQ ID NOs: 2, 5 and 16, respectively.
18. An isolated antibody or fragment that binds specifically to human IL-17A, comprising a VH and a VL, wherein the antibody comprises the LCDRl, LCDR2, and LCDR3 amino acid sequences as shown in:
a. SEQ ID NOs : 2, 5 and 12, respectively;
b. SEQ ID NOs: 2, 5 and 13, respectively;
c. SEQ ID NOs: 2, 5 and 14, respectively;
d. SEQ ID NOs: 2, 5 and 15, respectively;
e. SEQ ID NOs: 2, 5 and 16, respectively.
An isolated antibody or fragment binds specifically to human IL-17A, comprising a VH and a VL, wherein the antibody comprises the HCDR1, HCDR2 and HCDR3 amino acid sequences as shown in:
a . SEQ ID NOs : 25, 37 and 58, respectively;
b. SEQ ID NOs : 25, 38 and 58, respectively;
c . SEQ ID NOs : 25, 39 and 58, respectively;
d. SEQ ID NOs : 25, 40 and 58, respectively;
e . SEQ ID NOs : 25, 41 and 58, respectively;
f . SEQ ID NOs : 25, 42 and 58, respectively;
g. SEQ ID NOs : 25, 43 and 58, respectively;
h. SEQ ID NOs : 25, 41 and 59, respectively;
i . SEQ ID NOs : 25, 41 and 60, respectively;
j · SEQ ID NOs : 25, 43 and 60, respectively;
k. SEQ ID NOs : 25, 44 and 58, respectively; or
1. SEQ ID NOs : 25, 45 and 58, respectively .
20. The isolated antibody or fragment of claim 19, wherein the antibody further comprises the LCDRl, LCDR2, and LCDR3 amino acid sequences as shown in:
a. SEQ ID NOs: 3, 6 and 18, respectively;
b. SEQ ID NOs: 3, 6 and 19, respectively;
c. SEQ ID NOs: 3, 6 and 20, respectively; and
d. SEQ ID NOs: 3, 6 and 21, respectively.
21. An isolated antibody or fragment binds specifically to
human IL-17A, comprising a VH and a VL, wherein the antibody comprises the LCDRl, LCDR2, and LCDR3 amino acid sequences as shown in:
a. SEQ ID NOs: 3, 6 and 18, respectively;
b. SEQ ID NOs: 3, 6 and 19, respectively;
c. SEQ ID NOs: 3, 6 and 20, respectively; or
d. SEQ ID NOs: 3, 6 and 21, respectively.
22. An isolated antibody or fragment that binds specifically to human IL-17A, comprising a VH and a VL, wherein the antibody comprises the HCDR1, HCDR2 and HCDR3 amino acid sequences as shown in : :
a. SEQ ID NOs: 25, 47 and 58, respectively;
b. SEQ ID NOs: 25, 48 and 58, respectively;
c. SEQ ID NOs: 25, 49 and 58, respectively; or
d. SEQ ID NOs: 25, 50 and 58, respectively.
23. The isolated antibody or fragment of claim 22, wherein the antibody further comprises the LCDRl, LCDR2, and LCDR3 amino acid sequences as shown in SEQ ID NOs: 3, 6 and 18,
respectively .
24. An isolated antibody or fragment that binds specifically to human IL-17A comprising a VH and a VL, wherein the antibody comprises :
a. the HCDR1, HCDR2 and HCDR3 amino acid sequences as shown in SEQ ID NOs: 25, 43 and 60, respectively; or
b. the LCDRl, LCDR2, and LCDR3 amino acid sequences as shown in SEQ ID NOs: 3, 6 and 18, respectively; or
c. the HCDR1, HCDR2 and HCDR3 amino acid sequences as shown in SEQ ID NOs: 25, 43 and 60, respectively and
the LCDRl, LCDR2, and LCDR3 amino acid sequences as shown in SEQ ID NOs: 3, 6 and 18, respectively; or
d. the VH of SEQ ID NO: 86; or
e. the VL of SEQ ID NO: 79; or
f. the VH of SEQ ID NO: 86 and
the VL of SEQ ID NO: 79.
25. An isolated antibody or fragment that binds specifically to human IL-17A, comprising a VH and a VL, wherein the antibody comprises the VH having an amino acid sequence shown in SEQ ID NOs: 67, 68, 69, 81, 82, 83, 84, 85, or 86.
26. The isolated antibody or fragment of claim 25, wherein the antibody further comprises the VL having an amino acid sequence shown in SEQ ID NOs: 76, 77, 78, 79, or 80.
27. An isolated antibody or fragment that binds specifically to human IL-17A, comprising a VH and a VL, wherein the antibody comprises the VL having an amino acid sequence shown in SEQ ID NOs: 76, 77, 78, 79, or 80.
28. An isolated antibody or fragment that binds specifically to human IL-17A, comprising a VH and a VL, wherein the antibody comprises the VH which is at least 90% identical to the VH having an amino acid sequence shown in SEQ ID NOs: 67, 68, 69, 81, 82, 83, 84, 85, or 86.
29. An isolated antibody or fragment that binds specifically to human IL-17A, comprising a VH and a VL, wherein the antibody comprises the VL which is at least 90% identical to the variable region having an amino acid sequence shown in SEQ ID NOs: 76, 77, 78, 79, or 80.
30. An isolated antibody or fragment that binds specifically to human IL-17A, comprising an antibody heavy chain having an amino acid sequence shown in SEQ ID NOs: 92, 93, 94, 95, 96, 97, 98, 99, or 100.
31. An isolated antibody or fragment that binds specifically to IL-17A, comprising an antibody light chain having an amino acid sequence shown in SEQ ID NOs: 87, 88, 89, 90, or 91.
32. The isolated antibody or fragment of claim 3 or 7, wherein the antibody is fully human.
33. The isolated antibody or fragment of claim 3 or 7, wherein the antibody is conjugated to polyethylene glycol.
34. The isolated antibody or fragment of claim 3 or 7, having an IgGl or IgG4 isotype.
35. The isolated antibody or fragment of claim 3 or 7, wherein the Fc domain comprises S229P, P235A or L236A mutations in the Fc domain.
36. A pharmaceutical composition comprising the isolated
antibody or fragment of claim 3 or 7 and a pharmaceutically acceptable carrier.
37. An isolated antibody heavy chain comprising the amino acid sequence shown in SEQ ID NOs : 67, 68, 69, 81, 82, 83, 84, 85, 86, 92, 93, 94, 95, 96, 97, 98, 99, or 100.
38. An isolated antibody light chain comprising the amino acid sequence shown in SEQ ID NOs: 76, 77, 78, 79, 80, 87, 88, 89, 90, or 91.
39. An isolated polynucleotide encoding an antibody heavy chain comprising the amino acid sequence shown in SEQ ID NO: 67, 68, 69, 81, 82, 83, 84, 85, 86, 92, 93, 94, 95, 96, 97, 98, 99, or 100.
40. An isolated polynucleotide encoding an antibody light chain comprising the amino acid sequence shown in SEQ ID NO: 76, 77, 78, 79, 80, 87, 88, 89, 90, or 91.
41. A vector comprising at least one polynucleotide of claim 39 or 40.
42. A host cell comprising the vector of claim 40.
43. A method of inhibiting interaction of human IL-17A with IL- 17RA comprising:
a. providing human IL-17A and IL-17RA; and
b. contacting human IL-17A with an antagonist that binds human IL-17A at at least one amino acid residue selected from the group consisting of V22, V24, L26, 128, Y62, L99, R101, F110, and L112.
44. A method of inhibiting IL-17A biological activity,
comprising :
a. providing human IL17-A and IL-17RA; and
b. contacting human IL-17A with an antagonist that binds human IL-17A at at least one amino acid residue selected from the group consisting of V22, V24, L26, 128, Y62, L99, R101, F110, and L112.
45. A method of claim 43 or 44, wherein the antagonist is an antibody, a peptide, or a small molecule.
46. A method of treating an inflammatory condition, comprising administering a therapeutically effective amount of the isolated antibody of claim 3 or 7 to a patient in need thereof for a time sufficient to treat the inflammatory condition.
47. The method of claim 46, wherein the inflammatory condition affects the respiratory tract, lung, the gastrointestinal tract, small intestine, large intestine, colon, rectum, joint, bone and synovial tissue, cartilage, epithelium, endothelium, hepatic tissue, or skin.
48. The method of claim 46, wherein the inflammatory condition is an inflammatory pulmonary condition, asthma, chronic obstructive pulmonary disease (COPD) , an inflammatory bowel disease, an autoimmune disease, rheumatoid arthritis,
psoriasis or systemic sclerosis.
PCT/US2010/054662 2009-10-30 2010-10-29 Il-17a antagonists WO2011053763A2 (en)

Priority Applications (13)

Application Number Priority Date Filing Date Title
BR112012010280-0A BR112012010280B1 (en) 2009-10-30 2010-10-29 ISOLATED ANTIBODY OR FRAGMENT OF THE SAME THAT SPECIFICALLY LINKS TO HUMAN IL-17A, ITS USE, AND PHARMACEUTICAL COMPOSITION
ES10827516T ES2728115T3 (en) 2009-10-30 2010-10-29 IL-17A antagonists
CN201080059869.3A CN102905727B (en) 2009-10-30 2010-10-29 IL-17A Antagonist
EA201290254A EA029283B1 (en) 2009-10-30 2010-10-29 Il-17a antagonists
JP2012537102A JP5922025B2 (en) 2009-10-30 2010-10-29 IL-17A antagonist
CA2779257A CA2779257C (en) 2009-10-30 2010-10-29 Il-17a antagonists
MX2012005086A MX2012005086A (en) 2009-10-30 2010-10-29 Il-17a antagonists.
UAA201206558A UA114070C2 (en) 2009-10-30 2010-10-29 ANTIBODY OR FRAGMENT THAT SPECIFICALLY BECOMES TO HUMAN IL-17A
KR1020127013837A KR101836217B1 (en) 2009-10-30 2010-10-29 Il-17a antagonists
EP10827516.5A EP2493506B1 (en) 2009-10-30 2010-10-29 Il-17a antagonists
AU2010313304A AU2010313304B2 (en) 2009-10-30 2010-10-29 IL-17A antagonists
NZ599737A NZ599737A (en) 2009-10-30 2010-10-29 Il-17a antagonists
IL219390A IL219390B (en) 2009-10-30 2012-04-24 Il-17a antagonists

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US25686209P 2009-10-30 2009-10-30
US61/256,862 2009-10-30
US31091910P 2010-03-05 2010-03-05
US61/310,919 2010-03-05

Publications (2)

Publication Number Publication Date
WO2011053763A2 true WO2011053763A2 (en) 2011-05-05
WO2011053763A3 WO2011053763A3 (en) 2011-10-06

Family

ID=43922991

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2010/054662 WO2011053763A2 (en) 2009-10-30 2010-10-29 Il-17a antagonists

Country Status (21)

Country Link
US (1) US8519107B2 (en)
EP (1) EP2493506B1 (en)
JP (1) JP5922025B2 (en)
KR (1) KR101836217B1 (en)
CN (1) CN102905727B (en)
AU (1) AU2010313304B2 (en)
BR (1) BR112012010280B1 (en)
CA (1) CA2779257C (en)
CL (1) CL2012001141A1 (en)
CO (1) CO6541541A2 (en)
CR (1) CR20120298A (en)
EA (1) EA029283B1 (en)
EC (1) ECSP12011871A (en)
ES (1) ES2728115T3 (en)
GT (1) GT201200132A (en)
IL (1) IL219390B (en)
MX (1) MX2012005086A (en)
NI (1) NI201200080A (en)
NZ (1) NZ599737A (en)
PE (1) PE20121363A1 (en)
WO (1) WO2011053763A2 (en)

Cited By (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014001368A1 (en) * 2012-06-25 2014-01-03 Orega Biotech Il-17 antagonist antibodies
CN103509118A (en) * 2012-06-15 2014-01-15 郭怀祖 Insulin-Fc fusion protein
WO2014122613A1 (en) * 2013-02-08 2014-08-14 Novartis Ag Anti-il-17a antibodies and their use in treating autoimmune and inflammatory disorders
WO2015191610A3 (en) * 2014-06-09 2016-02-04 Biomed Valley Discoveries, Inc. Combination therapies using agents that target tumor-associated stroma or tumor cells and other pathways
WO2015191583A3 (en) * 2014-06-09 2016-02-18 Biomed Valley Discoveries, Inc. Combination therapies targeting tumor-associated stroma or tumor cells and topoisomerase
WO2015191615A3 (en) * 2014-06-09 2016-02-18 Biomed Valley Discoveries, Inc. Combination therapies using agents that target tumor-associated stroma or tumor cells and alkylating agents
WO2015191617A3 (en) * 2014-06-09 2016-02-25 Biomed Valley Discoveries, Inc. Combination therapies using anti-metabolites and agents that target tumor-associated stroma or tumor cells
WO2015191568A3 (en) * 2014-06-09 2016-02-25 Biomed Valley Discoveries, Inc. Combination therapies using agents that target tumor-associated stroma or tumor cells and tumor vasculature
US9284283B2 (en) 2012-02-02 2016-03-15 Ensemble Therapeutics Corporation Macrocyclic compounds for modulating IL-17
WO2015191590A3 (en) * 2014-06-09 2016-03-24 Biomed Valley Discoveries, Inc. Combination therapies targeting tumor-associated stroma or tumor cells and microtubules
WO2015191602A3 (en) * 2014-06-09 2016-03-24 Biomed Valley Discoveries, Inc. Combination therapies targeting tumor-associated stroma or tumor cells
US9475873B2 (en) 2009-05-05 2016-10-25 Novimmune Sa Nucleic acids encoding anti-IL-17F antibodies and methods of use thereof
WO2016123329A3 (en) * 2015-01-28 2016-11-03 Genentech, Inc. Gene expression markers and treatment of multiple sclerosis
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
US9932403B2 (en) 2010-05-20 2018-04-03 Ablynx Nv Biological materials related to HER3
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
RU2663721C1 (en) * 2015-03-05 2018-08-08 Бейджинг Беттермаб Биотекнолоджи Ко., Лтд Monoclonal antibodies to human il-17 and their application
US10208113B2 (en) 2014-06-23 2019-02-19 Janssen Biotech, Inc. Interferon α and ω antibody antagonists
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
EP3689907A1 (en) * 2019-01-31 2020-08-05 Numab Therapeutics AG Antibodies targeting il-17a and methods of use thereof
EP3816184A4 (en) * 2018-05-17 2021-09-01 Jiangsu Qyuns Therapeutics Co., Ltd. Anti-human interleukin 17a monoclonal antibody and application thereof
WO2024028436A1 (en) * 2022-08-04 2024-02-08 The University Of Birmingham Novel interleukin-17a (il-17a)-derived peptide and neutralizing antibody (ab17-ipl-1)

Families Citing this family (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20160124165A (en) 2014-02-21 2016-10-26 제넨테크, 인크. Anti-il-13/il-17 bispecific antibodies and uses thereof
CN103936854B (en) * 2014-04-30 2016-08-17 北京精益泰翔技术发展有限公司 Anti-IL-17A monoclonal antibody and preparation and application thereof
WO2015191596A1 (en) * 2014-06-09 2015-12-17 Biomed Valley Discoveries, Inc. Combination therapies using platinum agents and agents that target tumor-associated stroma or tumor cells
EP3233915A1 (en) * 2014-12-15 2017-10-25 MorphoSys AG Antibodies for il-17c
CN106336459B (en) * 2015-07-13 2020-12-08 三生国健药业(上海)股份有限公司 Human interleukin-17A resisting monoclonal antibody, preparation method and application thereof
MA46681A (en) * 2016-01-28 2019-09-11 Janssen Biotech Inc ANTI-TNF-ALPHA / IL-17A BISPECIFIC ANTIBODIES ANTIBODIES AND ANTI-TNF-ALPHA ANTIBODIES AND METHODS FOR USING THEM
KR102466763B1 (en) * 2016-04-13 2022-11-11 오리맵스 리미티드 Anti-PSMA Antibodies and Uses Thereof
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
CN112203685A (en) * 2018-03-29 2021-01-08 瑞美德生物医药科技有限公司 Treatment of autoimmune and inflammatory disorders using antibodies that bind interleukin-17A (IL-17A)
CN114127108A (en) * 2019-07-30 2022-03-01 江苏恒瑞医药股份有限公司 Methods of treating autoimmune diseases with IL-17 antagonists
CN114729943A (en) 2019-09-30 2022-07-08 詹森药业有限公司 Compositions and methods for IL-17 target conjugation assays with macromolecular modulators
KR20220070514A (en) 2019-09-30 2022-05-31 얀센 파마슈티카 엔.브이. Compositions and Methods for IL-17 Target Binding Assays Using Small Molecule Modulators

Citations (31)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4683195A (en) 1986-01-30 1987-07-28 Cetus Corporation Process for amplifying, detecting, and/or-cloning nucleic acid sequences
WO1992001047A1 (en) 1990-07-10 1992-01-23 Cambridge Antibody Technology Limited Methods for producing members of specific binding pairs
US5223409A (en) 1988-09-02 1993-06-29 Protein Engineering Corp. Directed evolution of novel binding proteins
US5624821A (en) 1987-03-18 1997-04-29 Scotgen Biopharmaceuticals Incorporated Antibodies with altered effector functions
US5770429A (en) 1990-08-29 1998-06-23 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5885793A (en) 1991-12-02 1999-03-23 Medical Research Council Production of anti-self antibodies from antibody segment repertoires and displayed on phage
US5939598A (en) 1990-01-12 1999-08-17 Abgenix, Inc. Method of making transgenic mice lacking endogenous heavy chains
US6054297A (en) 1991-06-14 2000-04-25 Genentech, Inc. Humanized antibodies and methods for making them
US20020086014A1 (en) 1999-08-24 2002-07-04 Korman Alan J. Human CTLA-4 antibodies and their uses
US20030099647A1 (en) 2001-10-05 2003-05-29 Deshpande Rajendra V. Fully human antibody Fab fragments with human interferon-gamma neutralizing activity
US6639055B1 (en) 1991-06-14 2003-10-28 Genentech, Inc. Method for making humanized antibodies
US6649055B1 (en) 2002-05-07 2003-11-18 The United States Of America As Represented By The United States Department Of Energy Pump station for radioactive waste water
WO2006013107A1 (en) 2004-08-05 2006-02-09 Novartis Ag Il-17 antagonistic antibodies
US7041870B2 (en) 2000-11-30 2006-05-09 Medarex, Inc. Transgenic transchromosomal rodents for making human antibodies
WO2006054059A1 (en) 2004-11-19 2006-05-26 Ucb Pharma S.A. Neutralising antibody molecules having specificity for human il-17
WO2007070750A1 (en) 2005-12-13 2007-06-21 Eli Lilly And Company Anti-il-17 antibodies
WO2007076524A2 (en) 2005-12-29 2007-07-05 Centocor, Inc. Human anti-il-23 antibodies, compositions, methods and uses
WO2007106769A2 (en) 2006-03-10 2007-09-20 Zymogenetics, Inc. Antibodies that bind both il-17a and il-17f 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
WO2008021156A2 (en) 2006-08-11 2008-02-21 Schering Corporation Antibodies to il-17a
US20080095775A1 (en) 2006-06-13 2008-04-24 Lewis Katherine E Il-17 and il-23 antagonists and methods of using the same
US20080095770A1 (en) 2006-08-09 2008-04-24 Glycart Biotechnology Ag Antigen binding molecules that bind EGFR, vectors encoding same, and uses thereof
WO2009003096A2 (en) 2007-06-26 2008-12-31 Centocor, Inc. Il-17 mutein proteins, antibodies, compositions, methods and uses
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
WO2009085462A1 (en) 2007-12-19 2009-07-09 Centocor, Inc. Design and generation of human de novo pix phage display libraries via fusion to pix or pvii, vectors, antibodies and methods
US7597889B1 (en) 1998-05-08 2009-10-06 Cambridge Enterprise Limited Binding molecules derived from immunoglobulins which do not trigger complement mediated lysis
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
WO2010045340A1 (en) 2008-10-14 2010-04-22 Centocor Ortho Biotech Inc. Methods of humanizing and affinity-maturing antibodies
US7776540B2 (en) 2004-05-03 2010-08-17 Schering Corporation Method of treating skin inflammation

Family Cites Families (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1051620B1 (en) * 1998-10-30 2006-06-07 Jonathan L. Miller Variable heavy chain and variable light chain regions of antibodies to human platelet glycoprotein ib alpha
US20050208558A1 (en) * 1999-10-19 2005-09-22 Applera Corporation Detection kits, such as nucleic acid arrays, for detecting the expression or 10,000 or more Drosophila genes and uses thereof
DE60237969D1 (en) * 2001-04-24 2010-11-25 Bayer Corp HUMAN ANTIBODIES AGAINST TIMP-1
WO2002102854A2 (en) * 2001-06-20 2002-12-27 Morphosys Ag Antibodies that block receptor protein tyrosine kinase activation, methods of screening for and uses thereof
US20030226155A1 (en) * 2001-08-30 2003-12-04 Biorexis Pharmaceutical Corporation Modified transferrin-antibody fusion proteins
US20050150002A1 (en) * 2004-01-02 2005-07-07 Dellapenna Dean Novel carotenoid hydroxylases for use in engineering carotenoid metabolism in plants
AR048098A1 (en) * 2004-03-15 2006-03-29 Wyeth Corp CALIQUEAMYCIN CONJUGATES
US20080014172A1 (en) 2004-05-28 2008-01-17 Applied Research Systems Ars Holding N.V. Use of Il-17 in the Treatment of Fertility-Related Disorders
US8088976B2 (en) * 2005-02-24 2012-01-03 Monsanto Technology Llc Methods for genetic control of plant pest infestation and compositions thereof
GB0525214D0 (en) * 2005-12-12 2006-01-18 Bioinvent Int Ab Biological materials and uses thereof
US20110008345A1 (en) 2007-11-30 2011-01-13 Claire Ashman Antigen-binding constructs

Patent Citations (35)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4683195B1 (en) 1986-01-30 1990-11-27 Cetus Corp
US4683195A (en) 1986-01-30 1987-07-28 Cetus Corporation Process for amplifying, detecting, and/or-cloning nucleic acid sequences
US5624821A (en) 1987-03-18 1997-04-29 Scotgen Biopharmaceuticals Incorporated Antibodies with altered effector functions
US5223409A (en) 1988-09-02 1993-06-29 Protein Engineering Corp. Directed evolution of novel binding proteins
US5939598A (en) 1990-01-12 1999-08-17 Abgenix, Inc. Method of making transgenic mice lacking endogenous heavy chains
US5969108A (en) 1990-07-10 1999-10-19 Medical Research Council Methods for producing members of specific binding pairs
WO1992001047A1 (en) 1990-07-10 1992-01-23 Cambridge Antibody Technology Limited Methods for producing members of specific binding pairs
US5770429A (en) 1990-08-29 1998-06-23 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US6054297A (en) 1991-06-14 2000-04-25 Genentech, Inc. Humanized antibodies and methods for making them
US6639055B1 (en) 1991-06-14 2003-10-28 Genentech, Inc. Method for making humanized antibodies
US5885793A (en) 1991-12-02 1999-03-23 Medical Research Council Production of anti-self antibodies from antibody segment repertoires and displayed on phage
US7597889B1 (en) 1998-05-08 2009-10-06 Cambridge Enterprise Limited Binding molecules derived from immunoglobulins which do not trigger complement mediated lysis
US20020086014A1 (en) 1999-08-24 2002-07-04 Korman Alan J. Human CTLA-4 antibodies and their uses
US7041870B2 (en) 2000-11-30 2006-05-09 Medarex, Inc. Transgenic transchromosomal rodents for making human antibodies
US20030099647A1 (en) 2001-10-05 2003-05-29 Deshpande Rajendra V. Fully human antibody Fab fragments with human interferon-gamma neutralizing activity
US6649055B1 (en) 2002-05-07 2003-11-18 The United States Of America As Represented By The United States Department Of Energy Pump station for radioactive waste water
US7776540B2 (en) 2004-05-03 2010-08-17 Schering Corporation Method of treating skin inflammation
WO2006013107A1 (en) 2004-08-05 2006-02-09 Novartis Ag Il-17 antagonistic antibodies
WO2006054059A1 (en) 2004-11-19 2006-05-26 Ucb Pharma S.A. Neutralising antibody molecules having specificity for human il-17
WO2007070750A1 (en) 2005-12-13 2007-06-21 Eli Lilly And Company Anti-il-17 antibodies
WO2007076524A2 (en) 2005-12-29 2007-07-05 Centocor, Inc. Human anti-il-23 antibodies, compositions, methods and uses
WO2007106769A2 (en) 2006-03-10 2007-09-20 Zymogenetics, Inc. Antibodies that bind both il-17a and il-17f and methods of using the same
US20080095775A1 (en) 2006-06-13 2008-04-24 Lewis Katherine E 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
US20080095770A1 (en) 2006-08-09 2008-04-24 Glycart Biotechnology Ag Antigen binding molecules that bind EGFR, vectors encoding same, and uses thereof
US20090175881A1 (en) 2006-08-11 2009-07-09 Presta Leonard G Antibodies to il-17a
WO2008021156A2 (en) 2006-08-11 2008-02-21 Schering Corporation Antibodies to il-17a
WO2009003096A2 (en) 2007-06-26 2008-12-31 Centocor, Inc. Il-17 mutein proteins, antibodies, compositions, methods and uses
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
WO2009085462A1 (en) 2007-12-19 2009-07-09 Centocor, Inc. Design and generation of human de novo pix phage display libraries via fusion to pix or pvii, vectors, antibodies and methods
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
WO2010045340A1 (en) 2008-10-14 2010-04-22 Centocor Ortho Biotech Inc. Methods of humanizing and affinity-maturing antibodies
US20100261620A1 (en) 2008-10-14 2010-10-14 Juan Carlos Almagro Methods of Humanizing and Affinity-Maturing Antibodies

Non-Patent Citations (103)

* Cited by examiner, † Cited by third party
Title
.: "Remington's Pharmaceutical Science, 15th ed.", MACK PUBLISHING COMPANY
AALBERSE; SCHUURMAN, IMMUNOLOGY, vol. 105, 2002, pages 9 - 19
ABRAHAM ET AL., J MOL RECOGNIT., vol. 9, 1996, pages 456 - 461
ADAMS ET AL., J. SYNCHROTRON. RADIAT., vol. 11, 2004, pages 53 - 5
AGGARWAL ET AL., J. BIOL. CHEM., vol. 278, 2003, pages 1910 - 4
ALCORN ET AL., ANNU. REV. PHYSIOL., vol. 72, 2010, pages 495 - 516
AL-LAZIKANI ET AL., J. MOL. BIOL., vol. 273, 1997, pages 927 - 48
ALMAGRO, MOL. RECOGNIT, vol. 17, 2004, pages 132 - 143
AWANE ET AL., J. IMMUNOL., vol. 162, 1999, pages 5337 - 44
BEDU-ADDO ET AL., PHARM. RES., vol. 21, 2004, pages 1353 - 61
BREKKE ET AL., EUR. J. IMMUNOL., vol. 24, 1995, pages 2542 - 7
CANFIELD; MORRISON, J. EXP. MED., vol. 173, 1991, pages 1483 - 91
CHOTHIA; LESK, MOL. BIOL, vol. 196, 1987, pages 901 - 917
CHOTHIA; LESK, MOL. BIOL., vol. 196, 1987, pages 901 - 17
CHOTHIA; LESK, MOL. BIOL., vol. 196, 1987, pages 901 - 917
COBAUGH ET AL., J MOL BIOL., vol. 378, 2008, pages 622 - 33
CURTIS ET AL., PROC. AM. THORAC. SOC., vol. 4, 2007, pages 512 - 21
D'ARCY ET AL., ACTA CRYSTALLOGRAPHICA SECTION D, vol. 63, 2007, pages 550 - 4
DECKERT ET AL., NT. J. CANCER, vol. 87, 2000, pages 382 - 90
ELY ET AL., NAT. IMMUNOLOGY, vol. 10, 2009, pages 1245 - 51
EMSLEY ET AL., ACTA CRYSTALLOGR. D. BIOL. CRYSTALLOGR., vol. 60, 2004, pages 2126 - 32
FISHWILD ET AL., NATURE BIOTECHNOLOGY, vol. 14, 1996, pages 845 - 51
FREESE ET AL., NEPHROL. DIAL. TRANSPLANT., vol. 16, 2001, pages 2401 - 6
FUJIMOTO ET AL., J. DERMATOLOG. SCI., vol. 50, 2008, pages 240 - 42
GAO ET AL., PROC. NATL. ACAD. SCI. USA, vol. 96, 1999, pages 6025 - 30
GENOVESE ET AL., ARTHRITIS RHEUM., vol. 62, 2010, pages 929 - 39
GERHARDT ET AL., J. MOL. BIOL, vol. 394, 2009, pages 901 - 21
GERHARDT ET AL., J. MOL. BIOL., vol. 394, 2009, pages 905 - 21
GUDJONSSON ET AL., CLIN. EXP. IMMUNOL., vol. 135, 2004, pages 1 - 8
GUPTA ET AL., AAPS PHARMSCI., vol. 5, 2003, pages E8
HAENEL ET AL., ANAL BIOCHEM, vol. 339, 2005, pages 182 - 4
HAMURO ET AL., J. BIOMOL. TECHNIQUES, vol. 14, 2003, pages 171 - 82
HESSEL ET AL., EUR. J. PHARMACOL., vol. 293, 1995, pages 401 - 12
HODGSON ET AL., BIO/TECHNOLOGY, vol. 9, 1991, pages 421
HOLTTA ET AL., INFLAMM. BOWEL DIS., vol. 14, 2008, pages 1175 - 84
HORN ET AL., BIOCHEMISTRY, vol. 45, 2006, pages 8488 - 98
HUEBER ET AL., SCI. TRANSL. MED., vol. 2, 2010, pages 52 - 72
HYMOWITZ ET AL., EMBO J, vol. 20, 2001, pages 5332 - 41
HYMOWITZ ET AL., EMBO J., vol. 20, 2001, pages 5332 - 41
IVANOV, TRENDS PHARMACOL. SCI., vol. 30, 2009, pages 95 - 103
JOHANSEN ET AL., BR. J. DERMATOL., vol. 160, 2009, pages 319 - 24
KABAT ET AL.: "Sequences of Immunological Interest, 5th ed.", 1991, PUBLIC HEALTH SERVICE, NIH
KABAT ET AL.: "Sequences of Proteins of Immunological Interest, 5th ed.", 1991, PUBLIC HEALTH SERVICE, NATIONAL INSTITUTES OF HEALTH
KAWAGUCHI ET AL., J. ALLERGY CLIN. IMMUNOL., vol. 114, 2004, pages 1265 - 73
KNAPPIK ET AL., J. MOL. BIOL., vol. 296, 2000, pages 57 - 86
KNIGHT ET AL., PLATELETS, vol. 15, 2004, pages 409 - 18
KOHLER ET AL., NATURE, vol. 256, pages 495 - 97
KOLLS ET AL., IMMUNITY, vol. 21, 2004, pages 467 - 76
KOLLS; LINDEN, IMMUNITY, vol. 21, 2004, pages 467 - 76
KREBS ET AL., J. IMMUNOL. METH, vol. 254, 2001, pages 67 - 84
KUESTNER ET AL., J IMMUNOL, vol. 179, 2007, pages 5462 - 73
KUESTNER ET AL., J. IMMUNOL., vol. 179, 2007, pages 5462 - 73
LEFRANC ET AL., DEV. COMPARAT. IMMUNOL., vol. 27, 2003, pages 55 - 77
LEONG ET AL., CYTOKINE, vol. 16, 2001, pages 106 - 19
LINDEN, CURR. OPIN. INVESTIG. DRUGS, vol. 4, 2003, pages 1304 - 12
LIU, KIDNEY INT., vol. 69, 2006, pages 213 - 7
LONBERG ET AL., NATURE, vol. 368, 1994, pages 856 - 9
LOONG ET AL., J. PATH., vol. 197, 2002, pages 322 - 32
LUBBERTS, CYTOKINE, vol. 41, 2008, pages 84 - 91
MAA ET AL., INT. J. PHARM., vol. 140, 1996, pages 155 - 68
MACLENNAN ET AL., ACTA PHYSIOL. SCAND. SUPPL., vol. 643, 1998, pages 55 - 67
MANGAN ET AL., NATURE, vol. 441, 2006, pages 231 - 4
MANNON, AM. J. TRANSPL., vol. 6, 2006, pages 867 - 75
MCALLISTER ET AL., J. IMMUNOL., vol. 175, 2005, pages 404 - 12
MENDEZ ET AL., NATURE GENETICS, vol. 15, 1997, pages 146 - 56
MOSELEY ET AL., CYTOKINE GROWTH FACTOR REV, vol. 14, 2003, pages 155 - 74
MOSELEY ET AL., CYTOKINE GROWTH FACTOR REV., vol. 14, 2003, pages 155 - 74
NEURATH ET AL., INTERN. REV. IMMUNOL, vol. 19, 2000, pages 51 - 62
PANKA ET AL., PROC. NATL. ACAD. SCI., vol. 85, 1988, pages 3080 - 84
PFLUGRATH, J., ACTA CRYSTALLOGRAPHICA SECTION D, vol. 55, 1999, pages 1718 - 25
PHILLIPS; CHANG; GUNG, MED. J., vol. 30, 2007, pages 2 - 6
PHILLIPS; STEADMAN, HISTOL. HISTOPATHOL., vol. 17, 2002, pages 247 - 52
QUEEN ET AL., PROC. NATL. ACAD. SCI. (USA, vol. 86, 1989, pages 10029 - 32
RACUSEN ET AL., KIDNEY INT., vol. 55, 1999, pages 713 - 23
READ, ACTA CRYSTALLOGR D BIOL CRYSTALLOGR, vol. 57, 2001, pages 1373 - 82
READ; POWRIE: "Curr. Protoc. Immunol.", 2001, article "Chapter 15 unit 15.13"
REMMELE ET AL., BIOPHARM., vol. 13, 2000, pages 36 - 46
REMMELE ET AL., PHARM. RES., vol. 15, 1997, pages 200 - 8
RITZ ET AL., NEPHROL. DIAL. TRANSPLANT., vol. 11, no. 9, 1996, pages 38 - 44
RUDIKOFF ET AL., PROC. NATL. ACAD. SCI., vol. 79, 1982, pages 1979 - 83
SASAKI ET AL., ADV. BIOPHYS., vol. 35, 1998, pages 1 - 24
See also references of EP2493506A4
SHI ET AL., J. MOL. BIOL., vol. 397, 2010, pages 385 - 96
SIMONSON, KIDNEY INT., vol. 71, 2007, pages 846 - 54
STRONG ET AL., BIOCHEMISTRY, vol. 30, 1991, pages 3739 - 48
TEPLYAKOV ET AL., J. MOL. BIOL., vol. 389, 2009, pages 115 - 23
TODA ET AL., J. ALLERGY CLIN. IMMUNOL., vol. 111, 2003, pages 875 - 81
TOMLINSON ET AL., J. MOL. BIOL, vol. 227, pages 776 - 798
TORNETTA ET AL., J. IMMUNOL. METHODS, vol. 360, 2010, pages 39 - 46
TOY ET AL., J. IMMUNOL., vol. 177, 2006, pages 36 - 9
VAN KOOTEN ET AL., J. AM. SOC. NEPHROL., vol. 9, 1998, pages 1526 - 34
WEAVER ET AL., ANNU. REV. IMMUNOL., vol. 25, 2007, pages 821 - 52
WOLTMAN ET AL., J. AM. NEPHROL., vol. 11, 2000, pages 2044 - 55
WORN ET AL., J. MOL. BIOL., vol. 305, 2001, pages 989 - 1010
WRIGHT ET AL., J. BIOL. CHEM., vol. 282, 2007, pages 13447 - 55
WU ET AL., CYTOKINE
WU; KABAT, J. EXP. MED., vol. 132, 1970, pages 211 - 250
YANG ET AL., PROTEIN ENG., vol. 16, 2003, pages 761 - 70
YAO ET AL., IMMUNITY, vol. 3, 1995, pages 811 - 21
YASUI ET AL., FEBS LETT., vol. 353, 1994, pages 143 - 6
ZHANG ET AL., INFLAMM. BOWEL DIS., vol. 12, 2006, pages 382 - 88
ZHANG ET AL., J. PHARM. SCI., vol. 93, 2004, pages 3076 - 89
ZHAO ET AL., PROTEIN EXPR PURIF, vol. 67, 2009, pages 182 - 9

Cited By (55)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9475873B2 (en) 2009-05-05 2016-10-25 Novimmune Sa Nucleic acids encoding anti-IL-17F antibodies and methods of use thereof
US9932403B2 (en) 2010-05-20 2018-04-03 Ablynx Nv Biological materials related to HER3
US10808032B2 (en) 2010-05-20 2020-10-20 Ablynx Nv Biological materials related to HER3
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
US9284283B2 (en) 2012-02-02 2016-03-15 Ensemble Therapeutics Corporation Macrocyclic compounds for modulating IL-17
CN103509118A (en) * 2012-06-15 2014-01-15 郭怀祖 Insulin-Fc fusion protein
CN103509118B (en) * 2012-06-15 2016-03-23 郭怀祖 insulin-Fc fusion protein
US9676847B2 (en) 2012-06-25 2017-06-13 Orega Biotech IL-17 antagonist antibodies
WO2014001368A1 (en) * 2012-06-25 2014-01-03 Orega Biotech Il-17 antagonist antibodies
JP2017018106A (en) * 2013-02-08 2017-01-26 ノバルティス アーゲー 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
AU2014213599B2 (en) * 2013-02-08 2017-02-09 Novartis Ag Anti-IL-17A antibodies and their use in treating autoimmune and inflammatory disorders
EP3656786A1 (en) * 2013-02-08 2020-05-27 Novartis AG Anti-il-17a antibodies and their use in treating autoimmune and inflammatory disorders
US9650439B2 (en) 2013-02-08 2017-05-16 Novartis Ag Anti-IL-17A antibodies and their use in treating autoimmune and inflammatory
EA031537B1 (en) * 2013-02-08 2019-01-31 Новартис Аг Anti-il-17a antibodies and their use in treating autoimmune and inflammatory disorders
KR101877109B1 (en) * 2013-02-08 2018-07-10 노파르티스 아게 Anti-il-17a antibodies and their use in treating autoimmune and inflammatory disorders
TWI632159B (en) * 2013-02-08 2018-08-11 諾華公司 Anti-il-17a antibodies and their use in treating autoimmune and inflammatory disorders
JP2016508508A (en) * 2013-02-08 2016-03-22 ノバルティス アーゲー Anti-IL-17A antibodies and their use in the treatment of autoimmune and inflammatory disorders
WO2014122613A1 (en) * 2013-02-08 2014-08-14 Novartis Ag Anti-il-17a antibodies and their use in treating autoimmune and inflammatory disorders
US10683349B2 (en) 2013-02-08 2020-06-16 Novartis Ag Anti-IL-17A antibodies and their use in treating autoimmune and inflammatory disorders
US9938342B2 (en) 2013-02-08 2018-04-10 Novartis Ag Nucleic acid encoding anti-IL-17A antibodies
AU2014213599C1 (en) * 2013-02-08 2017-09-07 Novartis Ag Anti-IL-17A antibodies and their use in treating autoimmune and inflammatory disorders
AU2014213599A1 (en) * 2013-02-08 2015-07-23 Novartis Ag Anti-IL-17A antibodies and their use in treating autoimmune and inflammatory disorders
EP3072905A4 (en) * 2013-11-18 2017-06-28 Shanghai Hengrui Pharmaceutical Co. Ltd. Il-17a conjugate and uses thereof
EP3670533A1 (en) * 2013-11-18 2020-06-24 Shanghai Hengrui Pharmaceutical Co. Ltd. Il-17a binding agent 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
WO2015191590A3 (en) * 2014-06-09 2016-03-24 Biomed Valley Discoveries, Inc. Combination therapies targeting tumor-associated stroma or tumor cells and microtubules
WO2015191615A3 (en) * 2014-06-09 2016-02-18 Biomed Valley Discoveries, Inc. Combination therapies using agents that target tumor-associated stroma or tumor cells and alkylating agents
WO2015191610A3 (en) * 2014-06-09 2016-02-04 Biomed Valley Discoveries, Inc. Combination therapies using agents that target tumor-associated stroma or tumor cells and other pathways
US11033620B2 (en) 2014-06-09 2021-06-15 Biomed Valley Discoveries, Inc. Combination therapies targeting tumor-associated stroma or tumor cells and microtubules
US11034757B2 (en) 2014-06-09 2021-06-15 Biomed Valley Discoveries, Inc. Combination therapies using agents that target tumor-associated stroma or tumor cells and tumor vasculature
US11033621B2 (en) 2014-06-09 2021-06-15 Biomed Valley Discoveries, Inc. Combination therapies targeting tumor-associated stroma or tumor cells
WO2015191583A3 (en) * 2014-06-09 2016-02-18 Biomed Valley Discoveries, Inc. Combination therapies targeting tumor-associated stroma or tumor cells and topoisomerase
US10758613B2 (en) 2014-06-09 2020-09-01 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services National Intstitutes Of Health Combination therapies using anti-metabolites and agents that target tumor-associated stroma or tumor cells
WO2015191602A3 (en) * 2014-06-09 2016-03-24 Biomed Valley Discoveries, Inc. Combination therapies targeting tumor-associated stroma or tumor cells
WO2015191568A3 (en) * 2014-06-09 2016-02-25 Biomed Valley Discoveries, Inc. Combination therapies using agents that target tumor-associated stroma or tumor cells and tumor vasculature
WO2015191617A3 (en) * 2014-06-09 2016-02-25 Biomed Valley Discoveries, Inc. Combination therapies using anti-metabolites and agents that target tumor-associated stroma or tumor cells
US10799584B2 (en) 2014-06-09 2020-10-13 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services National Institutes Of Health Combination therapies using agents that target tumor-associated stroma or tumor cells and alkylating agents
US10758526B2 (en) 2014-06-09 2020-09-01 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services National Institutes Of Health Combination therapies using agents that target tumor-associated stroma or tumor cells and other pathways
US10758614B2 (en) 2014-06-09 2020-09-01 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services National Institutes Of Health Combination therapies targeting tumor-associated stroma or tumor cells and topoisomerase
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
US11236391B2 (en) 2015-01-28 2022-02-01 Genentech, Inc. Gene expression markers and treatment of multiple sclerosis
WO2016123329A3 (en) * 2015-01-28 2016-11-03 Genentech, Inc. Gene expression markers and treatment of multiple sclerosis
RU2663721C1 (en) * 2015-03-05 2018-08-08 Бейджинг Беттермаб Биотекнолоджи Ко., Лтд Monoclonal antibodies to human il-17 and their application
US10392436B2 (en) 2015-03-05 2019-08-27 Beijing Bettermab Biotechnology Co., Ltd Anti-human IL-17 monoclonal antibodies and use thereof
EP3514176A4 (en) * 2016-09-14 2020-04-29 Beijing Hanmi Pharmaceutical Co., Ltd. Antibody specifically binding to il-17a and functional fragment thereof
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
US11072651B2 (en) 2016-09-14 2021-07-27 Beijing Hanmi Pharm. Co., Ltd. Antibody specifically binding to IL-17A, encoding nucleic acid, and method of using the antibody
EP3816184A4 (en) * 2018-05-17 2021-09-01 Jiangsu Qyuns Therapeutics Co., Ltd. Anti-human interleukin 17a monoclonal antibody and application thereof
EP3689907A1 (en) * 2019-01-31 2020-08-05 Numab Therapeutics AG Antibodies targeting il-17a and methods of use thereof
WO2024028436A1 (en) * 2022-08-04 2024-02-08 The University Of Birmingham Novel interleukin-17a (il-17a)-derived peptide and neutralizing antibody (ab17-ipl-1)

Also Published As

Publication number Publication date
ES2728115T3 (en) 2019-10-22
IL219390B (en) 2019-01-31
KR20120102662A (en) 2012-09-18
JP5922025B2 (en) 2016-05-25
EA029283B1 (en) 2018-03-30
IL219390A0 (en) 2012-06-28
GT201200132A (en) 2014-01-17
NZ599737A (en) 2015-02-27
PE20121363A1 (en) 2012-10-15
CN102905727A (en) 2013-01-30
EP2493506B1 (en) 2019-04-10
CO6541541A2 (en) 2012-10-16
NI201200080A (en) 2013-04-22
US8519107B2 (en) 2013-08-27
WO2011053763A3 (en) 2011-10-06
BR112012010280A2 (en) 2017-06-20
EP2493506A2 (en) 2012-09-05
BR112012010280B1 (en) 2020-09-24
CR20120298A (en) 2014-01-09
KR101836217B1 (en) 2018-03-08
CL2012001141A1 (en) 2012-11-30
CA2779257C (en) 2019-03-12
AU2010313304B2 (en) 2015-08-20
JP2013509193A (en) 2013-03-14
CN102905727B (en) 2016-12-07
EA201290254A1 (en) 2012-12-28
CA2779257A1 (en) 2011-05-05
AU2010313304A1 (en) 2012-05-24
EP2493506A4 (en) 2013-11-20
ECSP12011871A (en) 2012-08-31
US20110236390A1 (en) 2011-09-29
MX2012005086A (en) 2012-09-28

Similar Documents

Publication Publication Date Title
AU2010313304B2 (en) IL-17A antagonists
AU2017202610B2 (en) ST2L antagonists and methods of use
US10093725B2 (en) Anti-NGF antibodies and their use
AU2010286516B2 (en) Antikine antibodies that bind to multiple CC chemokines
MX2011001909A (en) Engineered anti-il-13 antibodies, compositions, methods and uses.
CA2871148C (en) Il-6 binding molecules
JP2016500656A (en) TOLL-like receptor 3 antagonists for treating metabolic and cardiovascular diseases
WO2015057906A1 (en) Cd200 receptor 1 agonists
AU2014346770B2 (en) Anti-CCL17 antibodies
US20190135912A1 (en) Interferon Alpha and Omega Antibody Antagonists
AU2017206283A1 (en) Anti-NGF antibodies and their use
AU2012323999A1 (en) IL-6 binding molecules

Legal Events

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

Ref document number: 201080059869.3

Country of ref document: CN

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 10827516

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2010313304

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 000591-2012

Country of ref document: PE

Ref document number: 2779257

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 1201002032

Country of ref document: TH

Ref document number: MX/A/2012/005086

Country of ref document: MX

Ref document number: 12012500862

Country of ref document: PH

WWE Wipo information: entry into national phase

Ref document number: 2012537102

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 12072188

Country of ref document: CO

WWE Wipo information: entry into national phase

Ref document number: 4244/DELNP/2012

Country of ref document: IN

ENP Entry into the national phase

Ref document number: 2010313304

Country of ref document: AU

Date of ref document: 20101029

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 20127013837

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: A201206558

Country of ref document: UA

Ref document number: 201290254

Country of ref document: EA

WWE Wipo information: entry into national phase

Ref document number: 2010827516

Country of ref document: EP

Ref document number: CR2012-000298

Country of ref document: CR

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112012010280

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 112012010280

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20120430