WO2014161570A1 - Antibodies against human il17 and uses thereof - Google Patents

Antibodies against human il17 and uses thereof Download PDF

Info

Publication number
WO2014161570A1
WO2014161570A1 PCT/EP2013/056971 EP2013056971W WO2014161570A1 WO 2014161570 A1 WO2014161570 A1 WO 2014161570A1 EP 2013056971 W EP2013056971 W EP 2013056971W WO 2014161570 A1 WO2014161570 A1 WO 2014161570A1
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
seq
diseases
antibody according
antibodies
Prior art date
Application number
PCT/EP2013/056971
Other languages
French (fr)
Inventor
Johannes Auer
Nikolaos Dimoudis
Guy Georges
Hendrik Knoetgen
Ekkehard Moessner
Original Assignee
Roche Glycart Ag
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Roche Glycart Ag filed Critical Roche Glycart Ag
Priority to PCT/EP2013/056971 priority Critical patent/WO2014161570A1/en
Publication of WO2014161570A1 publication Critical patent/WO2014161570A1/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
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/33Crossreactivity, e.g. for species or epitope, or lack of said crossreactivity
    • 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
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/94Stability, e.g. half-life, pH, temperature or enzyme-resistance

Definitions

  • the present invention relates to antibodies against human IL17A (IL17 antibody), methods for their production, pharmaceutical compositions containing said antibodies, and uses thereof.
  • IL17 antibody human IL17A
  • SEQ ID NO: 22) is a pro-inflammatory cytokine produced by a subset of memory T cells (called Thl7) that has been implicated in the pathogenesis of MS.
  • IL-17A plays a role in the induction of other inflammatory cytokines, chemokines and adhesion molecules. Treatment of animals with IL-17A neutralizing antibodies decreases disease incidence and severity in autoimmune encephalomyelitis
  • IL-17A is over-expressed in the cerebrospinal fluid of MS patients (Hellings, P.W., et al, Am. J. Resp. Cell Mol. Biol. 28 (2003) 42-50; Matusevicius, D., et al, Multiple Sclerosis 5 (1999) 101-104; WO 2005/051422).
  • IL-17A neutralizing antibodies reduce severity and incidence of mouse RA model of collagen induced arthritis, and high levels of IL-17A can be detected in the synovial fluid of inflamed joints from RA patients (Ziolkowska, M., et al, J. Immunol.
  • WO 96/17939 US 5,716,623; WO 95/18826; WO 97/15320; WO 99/35276 and WO 00/69436 WO 95/18826 US 6,274,711, US 6,274,711, WO 97/15320, US 6,063,372, WO 2006/013107 and WO 2008/02115 relate to IL-17A and antibodies against IL-17A.
  • WO 2010/102251 relates IL17 binding proteins.
  • One aspect of the invention is an antibody that specifically binds to human IL17 characterized in comprising a CDR1H of SEQ ID NO: l, CDR2H of SEQ ID NO:2, CDR3H of SEQ ID NO:3, and CDRIL of SEQ ID NO:4, CDR2L of SEQ ID NO:5, CDR3L of SEQ ID NO:6.
  • such IL17 antibody is a chimeric or humanized variant.
  • the antibody is characterized in comprising a) a variable heavy chain domain (VH) of SEQ ID NO:9, or of SEQ ID NO: 10, and b) a variable light chain domain (VL) of SEQ ID NO: 11 , or of SEQ ID NO: 12.
  • the antibody is characterized in comprising a) a variable heavy chain domain (VH) of SEQ ID NO: 10, and b) a variable light chain domain (VL) of SEQ ID NO: 12.
  • the antibody is characterized in comprising a) a variable heavy chain domain (VH) of SEQ ID NO:9, and b) a variable light chain domain (VL) of SEQ ID NO: 11.
  • VH variable heavy chain domain
  • VL variable light chain domain
  • the antibody is characterized in being of IgGl or IgG4 subclass.
  • the antibody which binds to human IL17 and being characterized by the above mentioned amino acid sequences and amino acid sequence fragments is of human IgGl isotype, in one embodiment with mutations L234A and L235A, in one embodiment with mutations L234A, L235A and P329G (numbering according to the EU index of Kabat).
  • the antibody which binds to human IL17 and being characterized by the above mentioned amino acid sequences and amino acid sequence fragments is of human IgG4 isotype, in one embodiment with mutations S228P and L235E, in one embodiment with mutations S228P, L235E and P329G (numbering according to the EU index of Kabat).
  • a further embodiment of the invention is a pharmaceutical composition comprising an antibody according to the invention.
  • a further embodiment of the invention is the use of an antibody according to the invention for the manufacture of a pharmaceutical composition.
  • a further embodiment of the invention is a nucleic acid encoding an antibody according to the invention.
  • a further embodiment of the invention is a nucleic acid encoding a heavy chain variable domain and/or a light chain variable domain of an antibody according to the invention.
  • the invention further provides expression vectors containing nucleic acid according to the invention capable of expressing said nucleic acid in a prokaryotic or eukaryotic host cell, and host cells containing such vectors for the recombinant production of such an antibody.
  • the invention further comprises a prokaryotic or eukaryotic host cell comprising a vector according to the invention.
  • the invention further comprises a method for the production of a recombinant chimeric or humanized antibody according to the invention, characterized by expressing a nucleic acid according to the invention in a prokaryotic or eukaryotic host cell and recovering said antibody from said cell or the cell culture supernatant.
  • the invention further comprises the antibody obtainable by such a recombinant method.
  • Antibodies according to the invention show benefits for patients in need of an IL17 targeting therapy.
  • the antibodies according to the invention have valuable properties causing a benefit for a patient suffering from a cancer disease, especially suffering from colon, lung, or pancreatic cancer or from inflammatory diseases, especially from autoimmune diseases, rheumatoid arthritis, psoratic arthritis, muscle diseases, e.g. muscular dystrophy, multiple sclerosis, chronic kidney diseases, bone diseases, e.g. bone degeneration in multiple myeloma, systemic lupus erythematosus, lupus nephritis, and vascular injury.
  • a further embodiment of the invention is an antibody according to the invention for use in the treatment of cancer, inflammatory diseases, autoimmune diseases, rheumatoid arthritis, psoratic arthritis, muscle diseases, e.g. muscular dystrophy, multiple sclerosis, chronic kidney diseases, bone diseases, e.g. bone degeneration in multiple myeloma, systemic lupus erythematosus, lupus nephritis, and vascular injury, especially for the treatment of systemic lupus erythematosus, lupus nephritis.
  • a further embodiment of the invention is an antibody according to the invention for manufacture of a medicament for the treatment of cancer, inflammatory diseases, autoimmune diseases, rheumatoid arthritis, psoratic arthritis, muscle diseases, e.g. muscular dystrophy, multiple sclerosis, chronic kidney diseases, bone diseases, e.g. bone degeneration in multiple myeloma, systemic lupus erythematosus, lupus nephritis, and vascular injury, especially for the treatment of systemic lupus erythematosus, lupus nephritis.
  • the invention further provides a method for treating a patient suffering from cancer, especially from colon, lung, or pancreatic cancer or from inflammatory diseases, especially from autoimmune diseases, rheumatoid arthritis, psoratic arthritis, muscle diseases, e.g. muscular dystrophy, multiple sclerosis, chronic kidney diseases, bone diseases, e.g. bone degeneration in multiple myeloma, systemic lupus erythematosus, lupus nephritis, and vascular injury, comprising administering to a patient diagnosed as having such a disease (and therefore being in need of such a therapy) an effective amount of the antibody which binds to human IL17 according to the invention.
  • the antibody is administered preferably in a pharmaceutical composition.
  • the invention further comprises a pharmaceutical composition
  • a pharmaceutical composition comprising an antibody according to the invention, optionally together with a buffer and/or an adjuvant useful for the formulation of antibodies for pharmaceutical purposes.
  • the invention further provides a pharmaceutical composition comprising an antibody according to the invention in a pharmaceutically acceptable carrier.
  • the pharmaceutical composition may be included in an article of manufacture or kit.
  • the antibodies according to the invention show benefits for human patients in need of a human IL17 targeting therapy and have valuable properties.
  • the antibodies according to the invention are characterized by one or more of the following properties (as determined in Examples 4, 5 and 6): the antibody that specifically binds to human IL17 according to the invention a) shows no cross reactivity with IL17B, IL17C, IL17D, IL17F (which means that the binding to IL17B, IL17C, IL17D and IL17F is 0% compared to the binding to IL17A , which is set as 100%); b) inhibits IL17 induced IL8 cytokine stimulation of CCD-25SK cells with an IC50 value of 2.0 nM or lower( e.g.
  • c) inhibits IL17 induced IL6 cytokine stimulation of CCD-25SK cells with an IC50 value of 5.0 nM or lower ( e.g. with an IC50 value between 5.0 nM and 0.0 nM); preferably with an IC50 value of 2.0 nM or lower; d) inhibits IL17 induced IL6 cytokine stimulation of human fibroblast-like synoviocytes-rheumatoid arthritis (HFLS-RA) with an IC50 value of 2.0 nM or lower (e.g.
  • IC50 value between 2.0 nM and 0.0 nM
  • IC50 value 1.0 nM or lower
  • antibody encompasses the various forms of antibody structures including but not being limited to whole antibodies and antibody fragments.
  • the antibody according to the invention is preferably a humanized antibody, chimeric antibody, or further genetically engineered antibody as long as the characteristic properties according to the invention are retained.
  • Antibody fragments comprise a portion of a full length antibody, preferably the variable domain(s) thereof, or at least the antigen binding portion thereof. Examples of antibody fragments include diabodies, single-chain antibody molecules, and multispecific antibodies (e.g. bispecific antibodies) formed or derived from antibody fragments. scFv antibodies are, e.g., described in Huston, J.S., Methods in Enzymol. 203 (1991) 46-52.
  • antibody fragments comprise single chain polypeptides having the characteristics of a VH domain, namely being able to assemble together with a VL domain, or of a VL domain binding to IL-17, namely being able to assemble together with a VH domain to a functional antigen binding site and thereby providing the properties of an antibody according to the invention.
  • the terms “monoclonal antibody” or “monoclonal antibody composition” as used herein refer to a preparation of antibody molecules of a single amino acid composition.
  • the term “humanized antibody” refers to antibodies in which the framework and/or “complementary determining regions” (CDR) have been modified to comprise the CDR of an immunoglobulin of different species as compared to that of the parent immunoglobulin.
  • CDR complementary determining regions
  • a non-human (e.g. mouse, rabbit or hamster) CDR is grafted into the framework region of a human antibody to prepare the "humanized antibody”. See, e.g., Riechmann, L., et al, Nature 332 (1988) 323- 327; and Neuberger, M.S., et al., Nature 314 (1985) 268-270.
  • chimeric antibody refers to a monoclonal antibody comprising a variable region, i.e., binding region, from mouse and at least a portion of a constant region derived from a different source or species, usually prepared by recombinant DNA techniques. Chimeric antibodies comprising for example a mouse variable region and a human constant region. Such mouse/human chimeric antibodies are the product of expressed immunoglobulin genes comprising DNA segments encoding rat immunoglobulin variable regions and DNA segments encoding human immunoglobulin constant regions.
  • Other forms of "chimeric antibodies” encompassed by the present invention are those in which the class or subclass has been modified or changed from that of the original antibody.
  • Such “chimeric” antibodies are also referred to as "class-switched antibodies.”
  • Methods for producing chimeric antibodies involve conventional recombinant DNA and gene transfection techniques now well known in the art. See, e.g., Morrison, S.L., et al, Proc. Natl. Acad Sci. USA 81 (1984) 6851-6855; US Patent Nos. 5,202,238 and 5,204,244.
  • IL-17A Human IL-17A (CTLA-8, Swiss Prot Q16552, further named as IL-17. IL17; SEQ ID NO: 22)) is a pro-inflammatory cytokine produced by a subset of memory T cells (called Thl7) that has been implicated in the pathogenesis of MS. IL-17A plays a role in the induction of other inflammatory cytokines, chemokines and adhesion molecules. Treatment of animals with IL-17A neutralizing antibodies decreases disease incidence and severity in autoimmune encephalomyelitis ( Komiyama, Y., et al, J. Immunol. 177 (2006) 566-573). IL-17A is over-expressed in the cerebrospinal fluid of MS patients (Hellings, P.W., et al, Am. J. Resp. Cell
  • IL-17A neutralizing antibodies reduce severity and incidence of mouse RA model of collagen induced arthritis, and high levels of IL-17A can be detected in the synovial fluid of inflamed joints from RA patients (Ziolkowska, M., et al, J. Immunol. 164 (2000) 2832-2838; Kotake, S., et al, J. Clin. Invest. 103 (1999) 1345-1352; Hellings, P.W., et al, Am. J. Resp. Cell Mol. Biol. 28 (2003) 42-50).
  • the terms, “binds to”, “binding” or “specifically binding” refers to the binding of the antibody to an epitope of human IL17with sufficient affinity such that the antibody is useful as a therapeutic agent in targeting human IL17 according to the invention.
  • the binding of the antibody to an epitope of the antigen human IL17 can be measured in an in vitro assay, preferably in an plasmon resonance assay (e.g. BIAcore, GE-Healthcare Uppsala, Sweden) with purified wild-type human antigen (preferably with IL17A homodimer) ( see e.g. Example 9).
  • the affinity of the binding is defined by the terms ka (rate constant for the association of the antibody from the antibody/antigen complex), kd (dissociation constant), and KD (kd/ka).
  • An antibody comprising a that specifically binds to human IL17 refers to an antibody binds to human IL17 with a binding affinity (KD) of 1.0 x 10 "8 M or less, e.g. from 1.0 x 10 "8 M to 1.0 x 10 "13 M, in one embodiment from 1.0 x 10 "9 M to 1.0 x 10 13 M.
  • epitope denotes a protein determinant capable of specifically binding to an antibody.
  • Epitopes usually consist of chemically active surface groupings of molecules such as amino acids or sugar side chains and usually epitopes have specific three dimensional structural characteristics, as well as specific charge characteristics. Conformational and nonconformational epitopes are distinguished in that the binding to the former but not the latter is lost in the presence of denaturing solvents.
  • One embodiment of the invention is an antibody that specifically binds to human IL17, characterized in that the antibody binds to the same epitope on human IL17 as an antibody which comprises a CDR1H of SEQ ID NO: l, CDR2H of SEQ ID NO:
  • One embodiment of the invention is an antibody that specifically binds to human IL17, characterized in that the antibody binds to the same epitope on human IL17 as an antibody which comprises a variable heavy chain domain (VH) of SEQ ID NO: 1
  • One embodiment of the invention is an antibody that specifically binds to human IL17, characterized in that the antibody competes for binding to the same epitope on human IL17 as an antibody which comprises a CDR1H of SEQ ID NO: l, CDR2H of SEQ ID NO:2, CDR3H of SEQ ID NO:3, and CDRIL of SEQ ID NO:4, CDR2L of SEQ ID NO:5, CDR3L of SEQ ID NO:6.
  • One embodiment of the invention is an antibody that specifically binds to human IL17, characterized in that the antibody competes for binding to the same epitope on human IL17 as an antibody which comprises a variable heavy chain domain
  • VH variable light chain domain of SEQ ID NO:10
  • VH variable light chain domain of SEQ ID NO: 12.
  • variable domain (variable domain of a light chain (V L ), variable domain of a heavy chain (V R )) as used herein denotes each of the pair of light and heavy chain domains which are involved directly in binding the antibody to the antigen.
  • the variable light and heavy chain domains have the same general structure and each domain comprises four framework (FR) regions whose sequences are widely conserved, connected by three "hypervariable regions” (or complementary determining regions, CDRs).
  • the framework regions adopt a ⁇ -sheet conformation and the CDRs may form loops connecting the ⁇ -sheet structure.
  • the CDRs in each chain are held in their three-dimensional structure by the framework regions and form together with the CDRs from the other chain the antigen binding site.
  • the antibody's heavy and light chain CDR3 regions play a particularly important role in the binding specificity/affinity of the antibodies according to the invention and therefore provide a further object of the invention.
  • antigen-binding portion of an antibody when used herein refers to the amino acid residues of an antibody which are responsible for antigen-binding.
  • the antigen-binding portion of an antibody comprises amino acid residues from the
  • CDRs complementary determining regions
  • FR Framework regions
  • FR3 of the heavy chain is the region which contributes most to antigen binding and defines the antibody's properties.
  • CDR and FR regions are determined according to the standard definition of Kabat, E.A., et al, Sequences of Proteins of Immunological Interest, 5th ed., Public Health Service, National Institutes of Health, Bethesda, MD (1991) and/or those residues from a "hypervariable loop".
  • CDRlH denotes the CDR1 region of the heavy chain variable region calculated according to Kabat (Kabat, E.A., et al., Sequences of Proteins of Immunological Interest, 5th ed., Public Health Service, National Institutes of
  • CDR2L, CDR3H, etc. mean the respective regions from the heavy(H) or light(L) chain.
  • an antigen binding site characterized by comprising CDR1H of SEQ ID NO:3 means that the antigen binding site comprises this amino acid sequence as a heavy chain variable chain CDR1 region in its variable heavy chain.
  • an antigen binding site characterized by comprising CDR1H of SEQ ID NO: l, CDR2H of SEQ ID NO:2, CDR3H of SEQ ID NO:3 means that the antigen binding sites comprises in its heavy chain as sequence of CDR1 SEQ ID NO: l, as sequence of CDR2 SEQ ID NO:2, and as sequence of CDR3 SEQ ID NO:3.
  • the terms "nucleic acid” or “nucleic acid molecule” as used herein are intended to include DNA molecules and RNA molecules.
  • a nucleic acid molecule may be single-stranded or double-stranded, but preferably is double-stranded DNA.
  • amino acid denotes the group of naturally occurring carboxy a-amino acids comprising alanine (three letter code: ala, one letter code: A), arginine (arg, R), asparagine (asn, N), aspartic acid (asp, D), cysteine (cys, C), glutamine (gin, Q), glutamic acid (glu, E), glycine (gly, G), histidine (his, H), isoleucine (ile, I), leucine (leu, L), lysine (lys, K), methionine (met, M), phenylalanine (phe, F), proline (pro, P), serine (ser, S), threonine (thr, T), tryptophan (trp, W), tyrosine (tyr, Y), and valine (val, V).
  • a nucleic acid is "operably linked" when it is placed into a functional relationship with another nucleic acid.
  • DNA for a presequence or secretory leader is operably linked to DNA for a polypeptide if it is expressed as a preprotein that participates in the secretion of the polypeptide;
  • a promoter or enhancer is operably linked to a coding sequence if it affects the transcription of the sequence; or
  • a ribosome binding site is operably linked to a coding sequence if it is positioned so as to facilitate translation.
  • "operably linked” means that the DNA sequences being linked are colinear, and, in the case of a secretory leader, contiguous and in reading frame. However, enhancers do not have to be contiguous. Linking is accomplished by ligation at convenient restriction sites. If such sites do not exist, synthetic oligonucleotide adaptors or linkers are used in accordance with conventional practice.
  • the expressions "cell”, “cell line”, and “cell culture” are used interchangeably and all such designations include progeny.
  • the words “transformants” and “transformed cells” include the primary subject cell and cultures derived therefrom without regard for the number of transfers. It is also understood that all progeny may not be precisely identical in DNA content, due to deliberate or inadvertent mutations. Variant progeny that have the same function or biological activity as screened for in the originally transformed cell are included.
  • the "Fc part” of an antibody is not involved directly in binding of an antibody to an antigen, but exhibits various effector functions.
  • An “Fc part of an antibody” is a term well known to the skilled artisan and defined on the basis of papain cleavage of antibodies. Depending on the amino acid sequence of the constant region of their heavy chains, antibodies or immunoglobulins are divided into the classes: IgA, IgD, IgE, IgG and IgM, and several of these may be further divided into subclasses
  • immunoglobulins are called ⁇ , ⁇ , ⁇ , ⁇ , and ⁇ , respectively.
  • the Fc part 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.
  • ADCC antibody- dependent cell-mediated cytotoxicity
  • CDC complement-dependent cytotoxicity
  • Complement activation is initiated by binding of complement factor Clq to the Fc part of most IgG antibody subclasses.
  • binding sites in the Fc part are known in the state of the art and described, e.g., by Boackle, R.J., et al, Nature 282 (1979) 742-743; Lukas, T.J., et al, J. Immunol. 127 (1981) 2555-2560; Brunhouse, R., and Cebra, J.J., Mol. Immunol. 16 (1979) 907-917; Burton, D.R., et al, Nature 288 (1980) 338-344; T Subscribesen, J.E., et al, Mol. Immunol. 37 (2000) 995-1004; Idusogie, E.E., et al, J. Immunol. 164 (2000) 4178-4184; Hezareh, M., et al, J.
  • binding sites are, e.g., L234, L235, D270, N297, E318, K320, K322, P331, and P329 (numbering according to EU index of Kabat used for the numbering of the constant domains, Kabat, E.A., et al., Sequences of Proteins of Immunological Interest, 5th ed., Public Health Service, National Institutes of
  • Antibodies of subclass IgGl, IgG2 and IgG3 usually show complement activation and Clq and C3 binding, whereas IgG4 does not activate the complement system and does not bind Clq and C3.
  • the antibody according to the invention is characterized in that the constant chains are of human origin.
  • constant chains are well known in the state of the art and e.g. described by Kabat (see e.g. Johnson, G., and Wu, T.T., Nucleic Acids Res. 28 (2000) 214-218 and Kabat, E.A., et al, Sequences of Proteins of Immunological Interest, 5th ed., Public Health Service, National Institutes of Health, Bethesda, MD (1991)).
  • useful human heavy chain constant regions comprises an amino acid sequence SEQ ID NO: 15, SEQ ID NO:
  • a useful human light chain constant region comprises an amino acid sequence of a kappa-light chain constant region of SEQ ID NO: 13 or an amino acid sequence of a lambda-light chain constant region of SEQ ID NO: 14.
  • the antibody according to the invention comprises a Fc part derived from human origin and preferably all other parts of the human constant regions.
  • Fc part derived from human origin denotes a Fc part which is either a Fc part of a human antibody of the subclass IgGl, IgG2, IgG3 or IgG4, in one embodiment a Fc part from human IgGl subclass, a mutated Fc part from human IgGl subclass (preferably with a mutations L234A and L235A, or L234A, L235A and P329G ), a Fc part from human IgG4 subclass or a mutated Fc part from human IgG4 subclass (preferably with a mutations S228P and L235E, or
  • the antibody comprise the human heavy chain constant regions of SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17 or SEQ ID NO: 18 (human IgGl subclass allotypes (Caucasian and Afroamerican or mutants L234A/L235A, and L234A/L235A/P329G), SEQ ID NO: 19, SEQ ID NO: 20, or SEQ ID NO: 21 (human IgG4 subclass or mutants L234A/L235A, and L234A/L235A/P329G) (numbering according to the EU index of Kabat, E.A., et al, Sequences of Proteins of Immunological Interest, 5th ed., Public Health Service, National Institutes of Health, Bethesda, MD (1991).
  • human heavy chain constant regions can comprise additionally modifications and or mutations.
  • the antibody according to the invention is of human IgGl subclass or of human IgG4 subclass. In one embodiment the antibody according to the invention is of human IgGl subclass. In one embodiment the antibody according to the invention is of human IgG4 subclass.
  • a further embodiment of the invention is a method for the production of an antibody according to the invention, characterized in that the sequence of a nucleic acid encoding the heavy chain of an antibody according to the invention and the nucleic acid encoding the light chain of said antibody are inserted into one or two expression vector(s), said vector(s) is/are inserted in a eukaryotic host cell, the encoded antibody is expressed and recovered from the host cell or the supernatant.
  • the antibodies according to the invention are preferably produced by recombinant means. Such methods are widely known in the state of the art and comprise protein expression in prokaryotic and eukaryotic cells with subsequent isolation of the antibody polypeptide and usually purification to a pharmaceutically acceptable purity.
  • protein expression nucleic acids encoding light and heavy chains or fragments thereof are inserted into expression vectors by standard methods. Expression is performed in appropriate prokaryotic or eukaryotic host cells, such as
  • CHO cells NSO cells, SP2/0 cells, HEK293 cells, COS cells, yeast, or E. coli cells, and the antibody is recovered from the cells (from the supernatant or after cells lysis).
  • the antibodies may be present in whole cells, in a cell lysate, or in a partially purified, or substantially pure form. Purification is performed in order to eliminate other cellular components or other contaminants, e.g., other cellular nucleic acids or proteins, by standard techniques, including column chromatography and others well known in the art. See Ausubel, F., et al. (eds.), Current Protocols in Molecular Biology, Greene Publishing and Wiley Interscience, New York (1987). Expression in NSO cells is described by, e.g., Barnes, L.M., et al, Cytotechnology
  • a preferred transient expression system (HEK 293) is described by Schlaeger, E.-J., and Christensen, K., in Cytotechnology 30 (1999) 71-83, and by Schlaeger, E.-J., in J. Immunol. Methods 194 (1996) 191-199.
  • Monoclonal antibodies are suitably separated from the culture medium by conventional immunoglobulin purification procedures such as, for example, protein A-Sepharose, hydroxylapatite chromatography, dialysis, or affinity chromatography.
  • DNA and R A encoding the monoclonal antibodies is readily isolated and sequenced using conventional procedures.
  • the hybridoma cells can serve as a source of such DNA and RNA.
  • the DNA may be inserted into expression vectors, which are then transfected into host cells, such as HEK 293 cells, CHO cells, or myeloma cells that do not otherwise produce immunoglobulin protein, to obtain the synthesis of recombinant monoclonal antibodies in the host cells.
  • host cells such as HEK 293 cells, CHO cells, or myeloma cells that do not otherwise produce immunoglobulin protein.
  • Nucleic acid molecules encoding amino acid sequence variants of anti-IL17 antibodies of the invention are prepared by a variety of methods known in the art.
  • These methods include, but are not limited to, isolation from a natural source (in the case of naturally occurring amino acid sequence variants) or preparation by oligonucleotide-mediated (or site-directed) mutagenesis, PCR mutagenesis, and cassette mutagenesis of an earlier prepared variant or a non-variant version of anti-
  • the heavy and light chain variable domains according to the invention are combined with sequences of promoter, translation initiation, constant region, 3' untranslated region, polyadenylation, and transcription termination to form expression vector constructs.
  • the heavy and light chain expression constructs can be combined into a single vector, co-transfected, serially transfected, or separately transfected into host cells which are then fused to form a single host cell expressing both chains.
  • the anti-IL17 antibodies of the invention have valuable properties such as good developability and producibility in high yields (the antibodies contain e.g. no hotspots, for which the production conditions are often more restricted), good titers and yields and are producible in high amounts and with relatively low impurities (> 90% Monomer after Protein A (SE-HPLC) with an estimated purity after 2nd column (ESI-MS) > 95%) (see Example 2 and 3).
  • compositions of an IL17 antibody as described herein are prepared by mixing such antibody having the desired degree of purity with one or more optional pharmaceutically acceptable carriers (Remington's Pharmaceutical Sciences, 16th edition, Osol, A. (ed.) (1980)), in the form of lyophilized formulations or aqueous solutions.
  • Pharmaceutically acceptable carriers are generally nontoxic to recipients at the dosages and concentrations employed, and include, but are not limited to: buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyl dimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride; benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as poly(vinylpyrrolidone); amino acids such as glycine, glutamine, asparagine, histidine, arg
  • sHASEGP soluble neutral-active hyaluronidase glycoproteins
  • rhuPH20 HYLENEX®, Baxter International, Inc.
  • Certain exemplary sHASEGPs and methods of use, including rhuPH20, are described in US Patent Publication Nos. 2005/0260186 and 2006/0104968.
  • a sHASEGP is combined with one or more additional glycosaminoglycanases such as chondroitinases.
  • Exemplary lyophilized antibody formulations are described in US Patent No. 6,267,958.
  • Aqueous antibody formulations include those described in US Patent No. 6,171,586 and WO 2006/044908, the latter formulations including a histidine-acetate buffer.
  • the formulation herein may also contain more than one active ingredients as necessary for the particular indication being treated, preferably those with complementary activities that do not adversely affect each other.
  • the antibodies specifically binding to human IL17 according to the invention have especially valuable properties such as low viscosity and high stability (so that they can be produced without high aggregation and in good yields) (see Example 10). Such antibodies with their low viscosity and high stability are especially useful in highly concentrated formulations/compositions which can be used e.g. in a subcutaneous administration.
  • the antibodies according to the invention may be administered as the sole active ingredient or in conjunction with, e.g. as an adjuvant to or in combination to, other drugs e.g. immunosuppressive or immunomodulating agents or other anti- inflammatory agents, e.g. for the treatment or prevention of diseases mentioned above.
  • other drugs e.g. immunosuppressive or immunomodulating agents or other anti- inflammatory agents, e.g. for the treatment or prevention of diseases mentioned above.
  • the antibodies as described herein may be used in combination with DMARD, e.g.
  • Gold salts sulphasalazine, antimalarias, methotrexate, D-penicillamine, azathioprine, mycophenolic acid, cyclosporine A, tacrolimus, sirolimus, minocycline, lefiunomide, glococorticoids; a calcineurin inhibitor, e.g. cyclosporin A or FK 506; a modulator of lymphocyte recirculation, e.g. FTY720 and FTY720 analogs; a mTOR inhibitor, e.g.
  • rapamycin 40-O-(2-hydroxyethyl)- rapamycin, CCI779, ABT578, AP23573 or TAFA-93; an ascomycin having immunosuppressive properties, e.g. ABT-281, ASM981, etc.; corticosteroids; cyclo-phosphamide; azathioprene; methotrexate; lefiunomide; mizoribine; mycophenolic acid; myco-phenolate mofetil; 15-deoxyspergualine or an immunosuppressive homologue, analogue or derivative thereof; immunosuppressive monoclonal antibodies, e.g., monoclonal antibodies to leukocyte receptors, e.g., MHC, CD2, CD3, CD4, CD7, CD8, CD25, CD28, CD40.
  • immunosuppressive monoclonal antibodies e.g., monoclonal antibodies to leukocyte receptors, e.g., MHC, CD2, CD3, CD4, CD7
  • CD45, CD58, CD80, CD86 or their ligands other immunomodulatory compounds, e.g. a recombinant binding molecule having at least a portion of the extracellular domain of CTLA4 or a mutant thereof, e.g. an at least extracellular portion of CTLA4 or a mutant thereof joined to a non-CTLA4 protein sequence, e.g. CTLA4Ig (for ex. designated ATCC 68629) or a mutant thereof, e.g. LEA29Y; adhesion molecule inhibitors, e.g. LFA-1 antagonists, ICAM-1 or -3 antagonists, VCAM-4 antagonists or VLA-4 antagonists; or a chemotherapeutic agent, e.g.
  • other immunomodulatory compounds e.g. a recombinant binding molecule having at least a portion of the extracellular domain of CTLA4 or a mutant thereof, e.g. an at least extracellular portion of CTLA4 or a mutant thereof joined to a non-CTLA
  • anti TNF agents e.g. monoclonal antibodies to TNF, e.g. infliximab, adalimumab, CDP870, or receptor constructs to TNF-RI or TNF-RII, e.g. Etanercept, PEG-TNF-RI; blockers of proinflammatory cytokines, IL-1 blockers, e.g. Anakinra or IL-1 trap, AAL160, ACZ 885, IL-6 blockers; chemokines blockers, e.g. inhibitors or activators of proteases, e.g. metalloproteases, anti-IL-15 antibodies, anti-IL-6 antibodies, anti-IL-23 antibodies, anti-CD20 antibodies, NSAIDs, such as aspirin or an anti-infectious agent (the list not limited to the agent mentioned).
  • anti TNF agents e.g. monoclonal antibodies to TNF, e.g. infliximab, adalimumab, CDP8
  • An antibody according to the present invention may be provided in combination or addition with one or more of the following agents: - an antagonist of cytokine function, (e.g. an agent which act on cytokine signaling pathways such as a modulator of the SOCS system) , such as an alpha-, beta-, and/or gamma-interferon; modulators of insulin-like growth factor type I (IGF- I) , its receptors and associated binding proteins; interleukins (IL) e.g.
  • agents e.g. an agent which act on cytokine signaling pathways such as a modulator of the SOCS system
  • IGF- I insulin-like growth factor type I
  • IL interleukins
  • IL-1 to 33 one or more of IL-1 to 33, and/or an interleukin antagonist or inhibitor such as anakinra; inhibitors of receptors of interleukin family members or inhibitors of specific subunits of such receptors; a tumor necrosis factor alpha (TNF-. alpha.) inhibitor such as an anti-TNF monoclonal antibody (for example infliximab; adalimumab, and/or CDP-870) , and/or a TNF receptor antagonist e.g. an immunoglobulin molecule (such as etanercept) and/or a low-molecular-weight agent such as pentoxyfylline;
  • TNF-. alpha. tumor necrosis factor alpha
  • an anti-TNF monoclonal antibody for example infliximab; adalimumab, and/or CDP-870
  • TNF receptor antagonist e.g. an immunoglobulin molecule (such as etanercept)
  • a modulator of B cells e.g. a monoclonal antibody targeting B- lymphocytes (such as CD20 (rituximab) or MRA-aIL16R) or T-lymphocytes (e.g. CTLA4-Ig, HuMax 11-15 or Abatacept);
  • B- lymphocytes such as CD20 (rituximab) or MRA-aIL16R
  • T-lymphocytes e.g. CTLA4-Ig, HuMax 11-15 or Abatacept
  • a modulator that inhibits osteoclast activity for example an antibody to RANKL
  • a modulator of chemokine or chemokine receptor function such as an antagonist of CCR1, CCR2, CCR2A, CCR2B, CCR3, CCR4, CCR5, CCR6, CCR7, CCR8, CCR9, CCRIO and CCRI1 (for the C-C family); CXCR1, CXCR2, CXCR3, CXCR4 and CXCR5 and CXCR6 (for the C-X-C family) and CX3CR1 for the C-X3-C family
  • MMPs matrix metalloproteases
  • stromelysins i.e., one or more of the stromelysins, the collagenases, and the gelatinases, as well as aggrecanase; especially collagenase-1 (MMP1), collagenase-2 (MMP8), collagenase-3 (MMP13) , stromelysin-1 (MMP3)
  • an agent such as doxycycline; a leukotriene biosynthesis inhibitor, 5 -lipoxygenase (5-LO) inhibitor or 5- lipoxygenase activating protein (FLAP) antagonist such as; zileuton; ABT-761; fenleuton; tepoxalin; Abbott-79175; Abbott- 85761; N- (5 -substituted) - thiophene-2-alkylsulfonamides; 2, 6-di-tert- butylphenolhydrazones; methoxytetrahydropyrans such as Zeneca ZD- 2138; the compound SB-210661; a pyridinyl-substituted 2- cyanonaphthalene compound such as L-739,010; a 2- cyanoquinoline compound such as L-746,530; indole and/or a quinoline compound such as MK-591, MK-886, and/or BAY x 1005;
  • LT B4, LTC4, LTD4, and LTE4 a receptor antagonist for leukotrienes (LT) B4, LTC4, LTD4, and LTE4, selected from the group consisting of the phenothiazin-3-ls such as L-651,392; amidino compounds such as CGS-25019c; benzoxalamines such as ontazolast; benzenecarboximidamides such as BIIL 284/260; and compounds such as zafirlukast, ablukast, montelukast, pranlukast, verlukast (MK-679) , RG- 12525, Ro-245913, iralukast (CGP 45715A) , and BAY x 7195; a phosphodiesterase (PDE) inhibitor such as a methylxanthanine, e.g. theophylline and/or aminophylline; and/or a selective PDE isoenzyme inhibitor e
  • a histamine type 1 receptor antagonist such as cetirizine, loratadine, desloratadine, fexofenadine, acrivastine, terfenadine, astemizole, azelastine, levocabastine, chlorpheniramine, promethazine, cyclizine, and/or mizolastine (generally applied orally, topically or parenterally);
  • a proton pump inhibitor such as omeprazole
  • gastroprotective histamine type 2 receptor antagonist a proton pump inhibitor (such as omeprazole) or gastroprotective histamine type 2 receptor antagonist
  • an antagonist of the histamine type 4 receptor an alpha-l/alpha-2 adrenoceptor agonist vasoconstrictor sympathomimetic agent, such as propylhexedrine, phenylephrine, phenylpropanolamine, ephedrine, pseudoephedrine, naphazoline hydrochloride, oxymetazoline hydrochloride, tetrahydrozoline hydrochloride, xylometazoline hydrochloride, tramazoline hydrochloride, and ethylnorepinephrine hydrochloride; an anticholinergic agent, e.g.
  • a muscarinic receptor (Ml, M2, and M3) antagonist such as atropine, hyoscine, glycopyrrrolate, ipratropium bromide, tiotropium bromide, oxitropium bromide, pirenzepine, and telenzepine;
  • beta-adrenoceptor agonist such as isoprenaline, salbutamol, formoterol, salmeterol, terbutaline, orciprenaline, bitolterol mesylate, and/or pirbuterol, e.g. a chiral enantiomer thereof;
  • a chromone e.g. sodium cromoglycate and/or nedocromil sodium
  • a glucocorticoid such as flunisolide, triamcinolone acetonide, beclomethasone dipropionate, budesonide, fluticasone propionate, ciclesonide, and/or mometasone furoate
  • an agent that modulate nuclear hormone receptors such as a PPAR
  • anti-inflammatory agent e.g. thalidomide or a derivative thereof, a retinoid, dithranol, and/or calcipotriol;
  • aminosalicylates and sulfapyridine such as sulfasalazine, mesalazine, balsalazide, and olsalazine; and immunomodulatory agents such as the thiopurines, and corticosteroids such as budesonide;
  • an antibacterial agent e.g. a penicillin derivative, a tetracycline, a macrolide, a beta-lactam, a fluoroquinolone, metronidazole, and/or an inhaled aminoglycoside
  • an antiviral agent e.g. a penicillin derivative, a tetracycline, a macrolide, a beta-lactam, a fluoroquinolone, metronidazole, and/or an inhaled aminoglycoside
  • an antiviral agent e.g.
  • acyclovir famciclovir, valaciclovir, ganciclovir, cidofovir; amantadine, rimantadine; ribavirin; zanamavir and/or oseltamavir; a protease inhibitor such as indinavir, nelfmavir, ritonavir, and/or saquinavir; a nucleoside reverse transcriptase inhibitor such as didanosine, lamivudine, stavudine, zalcitabine, zidovudine; a non-nucleoside reverse transcriptase inhibitor such as nevirapine, efavirenz; a cardiovascular agent such as a calcium channel blocker, beta- adrenoceptor blocker, angiotensin-converting enzyme (ACE) inhibitor, angiotensin-2 receptor antagonist; lipid lowering agent such as a statin, and/or fibrate; a modulator of blood cell morphology
  • a CNS agent such as an antidepressant (such as sertraline) , anti-Parkinsonian drug (such as deprenyl, L-dopa, ropinirole, pramipexole, MAOB inhibitor such as selegine and rasagiline, comP inhibitor such as tasmar, A-2 inhibitor, dopamine reuptake inhibitor, NMDA antagonist, nicotine agonist, dopamine agonist and/or inhibitor of neuronal nitric oxide synthase), and an anti- Alzheimer's drug such as donepezil, rivastigmine, tacrine, COX-2 inhibitor, propentofylline or metrifonate; an agent for the treatment of acute and chronic pain, e.g.
  • an antidepressant such as sertraline
  • anti-Parkinsonian drug such as deprenyl, L-dopa, ropinirole, pramipexole, MAOB inhibitor such as selegine and rasagiline
  • analgesic such as an opioid analogue or derivative, carbamazepine, phenytoin, sodium valproate, amitryptiline or other antidepressant agent, paracetamol, or nonsteroidal anti-inflammatory agent; a parenterally or topically-applied (including inhaled) local anaesthetic agent such as lignocaine or an analogue thereof; an anti-osteoporosis agent e.g.
  • a hormonal agent such as raloxifene, or a biphosphonate such as alendronate
  • a tryptase inhibitor e.g., a platelet activating factor (PAF) antagonist
  • PAF platelet activating factor
  • ICE interleukin converting enzyme
  • IMPDH IMPDH
  • adhesion molecule inhibitors including VLA-4 antagonist
  • a cathepsin e.g.
  • an inhibitor of tyrosine kinases might include Gefitinib, Imatinib mesylate), a serine / threonine kinase (e.g. an inhibitor of MAP kinase such as p38, JNK, protein kinases A, B and C and IKK), or a kinase involved in cell cycle regulation (e.g. a cylin dependent kinase) ; (viii) a glucose-6 phosphate dehydrogenase inhibitor; (ix) a kinin-B. subl . - and/or B.sub2.
  • a serine / threonine kinase e.g. an inhibitor of MAP kinase such as p38, JNK, protein kinases A, B and C and IKK
  • a kinase involved in cell cycle regulation e.g. a cylin dependent kinase
  • -receptor antagonist e.g., an anti-gout agent, e.g., colchicine;
  • a xanthine oxidase inhibitor e.g., allopurinol;
  • a uricosuric agent e.g., probenecid, sulfinpyrazone, and/or benzbromarone;
  • a growth hormone secretagogue e.g., transforming growth factor (TGF.beta.) ;
  • PDGF platelet-derived growth factor
  • fibroblast growth factor e.g., basic fibroblast growth factor (bFGF)
  • GM-CSF granulocyte macrophage colony stimulating factor
  • capsaicin cream e.g., capsaicin cream
  • a tachykinin NK. subl. and/or NK.sub3. receptor antagonist such NKP-608C, SB-233412 (talnetant) , and/or D-4418;
  • an elastase inhibitor e.g. UT-77 and/or ZD-
  • TACE TNF-alpha converting enzyme inhibitor
  • iNOS induced nitric oxide synthase
  • a chemoattractant receptor- homologous molecule expressed on TH2 cells such as a CRTH2 antagonist
  • an inhibitor of a P38 a P38
  • an agent modulating the function of Toll-like receptors TLR
  • an agent modulating the activity of purinergic receptors such as P2X7
  • an inhibitor of transcription factor activation such as NFkB, API, and/or STATS.
  • An inhibitor may be specific or may be a mixed inhibitor, e.g. an inhibitor targeting more than one of the molecules (e.g. receptors) or molecular classes mentioned above.
  • the antibody could also be used in association with a chemotherapeutic agent or another tyrosine kinase inhibitor in coadministration or in the form of an immuno- conjugat. Fragments of said antibody could also be used in bispecific antibodies obtained by recombinant mechanisms or biochemical coupling, and then associating the specificity of the above described antibody with the specificity of other antibodies able to recognize other molecules involved in the activity for which IL-17 is associated.
  • an antibody of the invention may be combined with one or more agents such as:- Non-steroidal anti-inflammatory agents (hereinafter NSAIDs) including non-selective cyclo- oxygenase (COX)-I / COX-2 inhibitors whether applied topically or systemically (such as piroxicam, diclofenac, propionic acids such as naproxen, flurbiprofen, fenoprofen, ketoprofen and ibuprofen, fenamates such as mefenamic acid, indomethacin, sulindac, azapropazone, pyrazolones such as phenylbutazone, salicylates such as aspirin); selective COX-2 inhibitors (such as meloxicam, celecoxib, rofecoxib, valdecoxib, lumarocoxib, parecoxib and etoricoxib) ; cyclo- oxygenase inhibiting ni
  • COX-2 inhibitors
  • An antibody of the invention can also be used in combination with an existing therapeutic agent for the treatment of cancer.
  • Suitable agents to be used in combination include: (i) antiproliferative/antineoplastic drugs and combinations thereof, as used in medical oncology, such as alkylating agents (for example cis- platin, carboplatin, cyclophosphamide, nitrogen mustard, melphalan, chlorambucil, busulphan and nitrosoureas); antimetabolites (for example antifolates such as fluoropyrimidines like 5-fluorouracil and tegafur, raltitrexed, methotrexate, cytosine arabinoside, hydroxyurea, gemcitabine and paclitaxel; antitumour antibiotics (for example anthracyclines like adriamycin, bleomycin, doxorubicin, daunomycin, epirubicin, idarubicin, mitomycin-C, dactinomycin and
  • LHRH antagonists or LHRH agonists for example goserelin, leuprorelin and buserelin
  • progestogens for example megestrol acetate
  • aromatase inhibitors for example as anastrozole, letrozole, vorazole and exemestane
  • inhibitors of 5.alpha.- reductase such as finasteride
  • Agents which inhibit cancer cell invasion for example metalloproteinase inhibitors like marimastat and inhibitors of urokinase plasminogen activator receptor function);
  • inhibitors of growth factor function include growth factor antibodies, growth factor receptor antibodies (for example the anti-erbb2 antibody trastuzumab and the anti-erbbl antibody cetuximab [C225] ), farnesyl transferase inhibitors, tyrosine kinase inhibitors and serine/threonine kinase inhibitors, for example inhibitors of the epidermal growth factor family (for example EGFR family tyrosine kinase inhibitors such as N- (3- chloro-4-fluorophenyl) -7-methoxy-6- (3- morpholinopropoxy) quinazolin-4-amine (gefitinib, AZD1839), N- (3- ethynylphenyl) -6, 7
  • Such active ingredients are suitably present in combination in amounts that are effective for the purpose intended.
  • Active ingredients may be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsules and poly-(methyl methacrylate) microcapsules, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) or in macroemulsions.
  • colloidal drug delivery systems for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules
  • Sustained-release preparations may be prepared. Suitable examples of sustained- release preparations include semi-permeable matrices of solid hydrophobic polymers containing the antibody, which matrices are in the form of shaped articles, e.g. films, or microcapsules.
  • compositions to be used for in vivo administration are generally sterile. Sterility may be readily accomplished, e.g., by filtration through sterile filtration membranes.
  • the ⁇ I117> antibodies according to the invention have one or more of the following properties (as determined in Examples 4, 5 and 6): the antibody that specifically binds to human IL17 according to the invention a) shows no cross reactivity with IL17B, IL17C, IL17D, IL17F (which means that the binding to IL17B, IL17C, IL17D and IL17F is 0% compared to the binding to IL 17A , which is set as 100%); b) inhibits IL17 induced IL8 cytokine stimulation of CCD-25SK cells with an IC50 value of 2.0 nM or lower( e.g.
  • c) inhibits IL17 induced IL6 cytokine stimulation of CCD-25SK cells with an IC50 value of 5.0 nM or lower ( e.g. with an IC50 value between 5.0 nM and 0.0 nM); preferably with an IC50 value of 2.0 nM or lower; d) inhibits IL17 induced IL6 cytokine stimulation of human fibroblast- like synoviocytes-rheumatoid arthritis (HFLS-RA) with an IC50 value of 2.0 nM or lower (e.g.
  • IC50 value between 2.0 nM and 0.0 nM
  • IC50 value preferably with an IC50 value of 1.0 nM or lower
  • e) inhibits IL17 induced IL8 cytokine stimulation of human fibroblast-like synoviocytes-rheumatoid arthritis (HFLS-RA)with an IC50 value of 1.5 nM or lower ( e.g. with an IC50 value between 1.5 nM and 0.0 nM); preferably with an IC50 value of 1.0 nM or lower.
  • HFLS-RA human fibroblast-like synoviocytes-rheumatoid arthritis
  • the invention comprises a method for the treatment of a patient in need of therapy, characterized by administering to the patient a therapeutically effective amount of an antibody according to the invention.
  • the invention comprises the use of an antibody according to the invention for the preparation of a medicament for the treatment of cancer, especially colon, lung, or pancreatic cancer or for the treatment of autoimmune diseases, rheumatoid arthritis, psoratic arthritis, muscle diseases, e.g. muscular dystrophy, multiple sclerosis, chronic kidney diseases, bone diseases, e.g. bone degeneration in multiple myeloma, systemic lupus erythematosus, lupus nephritis, and vascular injury.
  • autoimmune diseases rheumatoid arthritis, psoratic arthritis
  • muscle diseases e.g. muscular dystrophy, multiple sclerosis, chronic kidney diseases
  • bone diseases e.g. bone degeneration in multiple myeloma, systemic lupus erythematosus, lupus nephritis, and vascular injury.
  • the invention comprises the use of an antibody according to the invention for the preparation of a medicament for the treatment of systemic lupus erythematosus, or lupus nephritis.
  • the invention comprises the use of an antibody according to the invention for the treatment of cancer or inflammatory diseases, preferably for the treatment of colon, lung, or pancreatic cancer or for the treatment of autoimmune diseases, rheumatoid arthritis, psoratic arthritis, muscle diseases, e.g. muscular dystrophy, multiple sclerosis, chronic kidney diseases, bone diseases, e.g. bone degeneration in multiple myeloma, systemic lupus erythematosus, lupus nephritis, and vascular injury.
  • autoimmune diseases e.g. muscular dystrophy, multiple sclerosis, chronic kidney diseases
  • bone diseases e.g. bone degeneration in multiple myeloma, systemic lupus erythematosus, lupus nephritis, and vascular injury.
  • the invention comprises the use of an antibody according to the invention for the treatment of cancer or inflammatory diseases, preferably for the treatment of systemic lupus erythematosus, or lupus nephritis.
  • the invention comprises the use of the antibodies specifically binding to human IL17 according to the invention for the treatment (or the antibodies for use in the treatment) of a patient suffering from cancer, especially from colon, lung, or pancreatic cancer or from autoimmune diseases, rheumatoid arthritis, psoratic arthritis, psoriasis, muscle diseases, e.g. muscular dystrophy, multiple sclerosis, chronic kidney diseases, bone diseases, e.g.
  • the invention comprises the use of the antibodies specifically binding to and human IL17 according to the invention for the treatment (or the antibodies for use in the treatment) of a patient suffering from systemic lupus erythematosus or lupus nephritis.
  • the invention comprises the use of the antibodies specifically binding to and human IL17 according to the invention for the treatment (or the antibodies for use in the treatment) of a variety of inflammatory, immune and proliferative disorders, including rheumatoid arthritis (RA), osteoarthritis, rheumatoid arthritis osteoporosis, inflammatory fibrosis (e.g. scleroderma, lung fibrosis, and cirrhosis), gingivitis, periodontitis or other inflammatory periodontal diseases, inflammatory bowel disorders (e.g.
  • Anti-Ill 7 antibodies of the invention are particularly useful for the treatment, prevention, or amelioration of autoimmune disease and of inflammatory conditions, in particular inflammatory conditions with an aetiology including an autoimmune component such as arthritis (for example rheumatoid arthritis, arthritis chronica progrediente and arthritis deformans) and rheumatic diseases, including inflammatory conditions and rheumatic diseases involving bone loss, inflammatory pain, spondyloarhropathies including ankolysing spondylitis, Reiter syndrome, reactive arthritis, psoriatic arthritis, and enterophathis arthritis, hypersensitivity (including both airways hypersensitivity and dermal hypersensitivity) and allergies.
  • arthritis for example rheumatoid arthritis, arthritis chronica progrediente and arthritis deformans
  • rheumatic diseases including inflammatory conditions and rheumatic diseases involving bone loss, inflammatory pain, spondyloarhropathies including ankolysing spondylitis, Reiter syndrome,
  • autoimmune haematological disorders including e.g. hemolytic anaemia, aplastic anaemia, pure red cell anaemia and idiopathic thrombocytopenia
  • systemic lupus erythematosus inflammatory muscle disorders, polychondritis, sclerodoma, Wegener granulomatosis, dermatomyositis, chronic active hepatitis, myasthenia gravis, psoriasis, Steven- Johnson syndrome, idiopathic sprue, autoimmune inflammatory bowel disease (including e.g.
  • ulcerative colitis Crohn's disease and Irritable Bowel Syndrome
  • endocrine ophthalmopathy Graves' disease, sarcoidosis, multiple sclerosis, primary biliary cirrhosis, juvenile diabetes (diabetes mellitus type I), uveitis (anterior and posterior), keratoconjunctivitis sicca and vernal keratoconjunctivitis, interstitial lung fibrosis, psoriatic arthritis and glomerulonephritis (with and without nephrotic syndrome, e.g.
  • the antibodies according to the invention are also useful for the treatment, prevention, or amelioration of asthma, bronchitis, pneumoconiosis, pulmonary emphysema, and other obstructive or inflammatory diseases of the airways.
  • the invention comprises also a method for the treatment of a patient suffering from such disease.
  • the invention further provides a method for the manufacture of a pharmaceutical composition comprising an effective amount of an antibody according to the invention together with a pharmaceutically acceptable carrier and the use of the antibody according to the invention for such a method.
  • the invention also provides the use of an antibody according to the invention in an effective amount for the manufacture of a pharmaceutical agent, preferably together with a pharmaceutically acceptable carrier, for the treatment of a patient suffering from cancer, especially from colon, lung, or pancreatic cancer or from autoimmune diseases, rheumatoid arthritis, psoratic arthritis, muscle diseases, e.g. muscular dystrophy, multiple sclerosis, chronic kidney diseases, bone diseases, e.g. bone degeneration in multiple myeloma, systemic lupus erythematosus, lupus nephritis, and vascular injury.
  • autoimmune diseases rheumatoid arthritis
  • psoratic arthritis muscle diseases, e.g. muscular dystrophy, multiple sclerosis, chronic kidney diseases
  • bone diseases e.g. bone degeneration in multiple myeloma, systemic lupus erythematosus, lupus nephritis, and vascular injury.
  • the invention also provides the use of an antibody according to the invention in an effective amount for the manufacture of a pharmaceutical agent, preferably together with a pharmaceutically acceptable carrier, for the treatment of a patient suffering from of autoimmune disease and of inflammatory conditions, in particular inflammatory conditions with an aetiology including an autoimmune component such as arthritis (for example rheumatoid arthritis, arthritis chronica progrediente and arthritis deformans) and rheumatic diseases, including inflammatory conditions and rheumatic diseases involving bone loss, inflammatory pain, spondyloarhropathies including ankolsing spondylitis, Reiter syndrome, reactive arthritis, psoriatic arthritis, and enterophathis arthritis, hypersensitivity (including both airways hypersensitivity and dermal hypersensitivity) and allergies.
  • arthritis for example rheumatoid arthritis, arthritis chronica progrediente and arthritis deformans
  • rheumatic diseases including inflammatory conditions and rheumatic diseases involving bone loss, inflammatory pain, spondy
  • autoimmune haemato logical disorders including e.g. hemolytic anaemia, aplastic anaemia, pure red cell anaemia and idiopathic thrombocytopenia
  • systemic lupus erythematosus inflammatory muscle disorders, polychondritis, sclerodoma, Wegener granulomatosis, dermatomyositis, chronic active hepatitis, myasthenia gravis, psoriasis, Steven- Johnson syndrome, idiopathic sprue, autoimmune inflammatory bowel disease (including e.g. ulcerative colitis,
  • the antibodies according to the invention are also useful for the treatment, prevention, or amelioration of asthma, bronchitis, pneumoconiosis, pulmonary emphysema, and other obstructive or inflammatory diseases of the airways.
  • the present invention provides a composition, e.g. a pharmaceutical composition, containing an IL17 antibody as described herein, formulated together with a pharmaceutically acceptable carrier, e.g., for use in any of the above therapeutic methods.
  • a pharmaceutically acceptable carrier e.g., for use in any of the above therapeutic methods.
  • pharmaceutically acceptable carrier includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption/resorption delaying agents, and the like that are physiologically compatible.
  • the carrier is suitable for injection or infusion.
  • a composition of the present invention can be administered by a variety of methods known in the art. As will be appreciated by the skilled artisan, the route and/or mode of administration will vary depending upon the desired results.
  • Pharmaceutically acceptable carriers include sterile aqueous solutions or dispersions and sterile powders for the preparation of sterile injectable solutions or dispersion.
  • the use of such media and agents for pharmaceutically active substances is known in the art.
  • the carrier can be, for example, an isotonic buffered saline solution.
  • a pharmaceutical formulation comprises any of the IL17 antibodies provided herein and at least one additional therapeutic agent, e.g., as described below.
  • Antibodies of the invention can be used either alone or in combination with other agents in a therapy.
  • an antibody of the invention may be coadministered with at least one additional therapeutic agent.
  • an additional therapeutic agent is a. immunosuppressive or immunomodulating agents or other anti-inflammatory agents.
  • the antibodies as described herein may be used in combination with DMARD, e.g. Gold salts, sulphasalazine, antimalarias, methotrexate, D-penicillamine, azathioprine, mycophenolic acid, cyclosporine A, tacrolimus, sirolimus, minocycline, leflunomide, glococorticoids; a calcineurin inhibitor, e.g.
  • cyclosporin A or FK 506 a modulator of lymphocyte recirculation, e.g. FTY720 and FTY720 analogs; a mTOR inhibitor, e.g. rapamycin, 40-O-(2-hydroxyethyl)-rapamycin, CCI779, ABT578, AP23573 or TAFA-93; an ascomycin having immuno-suppressive properties, e.g.
  • ABT-281, ASM981, etc. corticosteroids; cyclophosphamide; azathioprene; methotrexate; leflunomide; mizoribine; mycophenolic acid; myc-phenol-late mofetil; 15-deoxyspergualine or an immunosuppressive homologue, analogue or derivative thereof; immunosuppressive monoclonal antibodies, e.g., monoclonal antibodies to leukocyte receptors, e.g., MHC, CD2, CD3, CD4, CD7, CD8, CD25, CD28, CD40. CD45, CD58, CD80, CD86 or their ligands; other immunomodulatory compounds, e.g.
  • a recombinant binding molecule having at least a portion of the extracellular domain of CTLA4 or a mutant thereof, e.g. an at least extracellular portion of CTLA4 or a mutant thereof joined to a non-CTLA4 protein sequence, e.g. CTLA4Ig (for ex. designated ATCC 68629) or a mutant thereof, e.g. LEA29Y; adhesion molecule inhibitors, e.g. LFA-1 antagonists, ICAM-1 or -3 antagonists, VCAM-4 antagonists or VLA-4 antagonists; or a chemotherapeutic agent, e.g.
  • a recombinant binding molecule having at least a portion of the extracellular domain of CTLA4 or a mutant thereof, e.g. an at least extracellular portion of CTLA4 or a mutant thereof joined to a non-CTLA4 protein sequence, e.g. CTLA4Ig (for ex. designated ATCC 68629) or a mutant thereof, e.g. LEA
  • anti TNF agents e.g. monoclonal antibodies to TNF, e.g. infliximab, adalimumab, CDP870, or receptor constructs to TNF-RI or TNF-RII, e.g. Etanercept, PEG-TNF-RI
  • chemokines blockers e.g. inhibitors or activators of proteases, e.g. metalloproteases, anti-IL-15 antibodies, anti-IL-6 antibodies, anti-IL-23 antibodies, anti-CD20 antibodies, NSAIDs, such as aspirin or an anti-infectious agent (the list not limited to the agent mentioned).
  • Such combination therapies noted above encompass combined administration (where two or more therapeutic agents are included in the same or separate formulations), and separate administration, in which case, administration of the antibody of the invention can occur prior to, simultaneously, and/or following, administration of the additional therapeutic agent and/or adjuvant.
  • An antibody of the invention can be administered by any suitable means, including parenteral, intrapulmonary, and intranasal, and, if desired for local treatment, intralesional administration.
  • Parenteral infusions include intramuscular, intravenous, intraarterial, intraperitoneal, or subcutaneous administration. Dosing can be by any suitable route, e.g. by injections, such as intravenous or subcutaneous injections, depending in part on whether the administration is brief or chronic.
  • Various dosing schedules including but not limited to single or multiple administrations over various time- points, bolus administration, and pulse infusion are contemplated herein.
  • Antibodies of the invention would be formulated, dosed, and administered in a fashion consistent with good medical practice. Factors for consideration in this context include the particular disorder being treated, the particular mammal being treated, the clinical condition of the individual patient, the cause of the disorder, the site of delivery of the agent, the method of administration, the scheduling of administration, and other factors known to medical practitioners.
  • the antibody need not be, but is optionally formulated with one or more agents currently used to prevent or treat the disorder in question. The effective amount of such other agents depends on the amount of antibody present in the formulation, the type of disorder or treatment, and other factors discussed above.
  • an antibody of the invention when used alone or in combination with one or more other additional therapeutic agents, will depend on the type of disease to be treated, the type of antibody, the severity and course of the disease, whether the antibody is administered for preventive or therapeutic purposes, previous therapy, the patient's clinical history and response to the antibody, and the discretion of the attending physician.
  • the antibody is suitably administered to the patient at one time or over a series of treatments.
  • about 1 ⁇ g/kg to 15 mg/kg (e.g. 0.5mg/kg - 10 mg/kg) of antibody can be an initial candidate dosage for administration to the patient, whether, for example, by one or more separate administrations, or by continuous infusion.
  • One typical daily dosage might range from about 1 ⁇ g/kg to 100 mg/kg or more, depending on the factors mentioned above.
  • the treatment would generally be sustained until a desired suppression of disease symptoms occurs.
  • One exemplary dosage of the antibody would be in the range from about 0.05 mg/kg to about 10 mg/kg.
  • one or more doses of about 0.5 mg/kg, 2.0 mg/kg, 4.0 mg/kg or 10 mg/kg (or any combination thereof) may be administered to the patient.
  • Such doses may be administered intermittently, e.g. every week or every three weeks (e.g. such that the patient receives from about two to about twenty, or e.g. about six doses of the antibody).
  • An initial higher loading dose, followed by one or more lower doses may be administered.
  • An exemplary dosing regimen comprises administering. However, other dosage regimens may be useful. The progress of this therapy is easily monitored by conventional techniques and assays.
  • the compounds of the present invention which may be used in a suitable hydrated form, and/or the pharmaceutical compositions of the present invention, are formulated into pharmaceutically acceptable dosage forms by conventional methods known to those of skill in the art.
  • Actual dosage levels of the active ingredients in the pharmaceutical compositions of the present invention may be varied so as to obtain an amount of the active ingredient which is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient (effective amount).
  • the selected dosage level will depend upon a variety of pharmacokinetic factors including the activity of the particular compositions of the present invention employed, or the ester, salt or amide thereof, the route of administration, the time of administration, the rate of excretion of the particular compound being employed, other drugs, compounds and/or materials used in combination with the particular compositions employed, the age, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors well known in the medical arts.
  • an article of manufacture containing materials useful for the treatment, prevention and/or diagnosis of the disorders described above comprises a container and a label or package insert on or associated with the container.
  • Suitable containers include, for example, bottles, vials, syringes, IV solution bags, etc.
  • the containers may be formed from a variety of materials such as glass or plastic.
  • the container holds a composition which is by itself or combined with another composition effective for treating, preventing and/or diagnosing the condition and may have a sterile access port (for example the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle).
  • At least one active agent in the composition is an antibody of the invention.
  • the label or package insert indicates that the composition is used for treating the condition of choice.
  • the article of manufacture may comprise (a) a first container with a composition contained therein, wherein the composition comprises an antibody of the invention; and (b) a second container with a composition contained therein, wherein the composition comprises a further cytotoxic or otherwise therapeutic agent.
  • the article of manufacture in this embodiment of the invention may further comprise a package insert indicating that the compositions can be used to treat a particular condition.
  • the article of manufacture may further comprise a second (or third) container comprising a pharmaceutically- acceptable buffer, such as bacteriostatic water for injection (BWFI), phosphate- buffered saline, Ringer's solution and dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, and syringes. Description of the Sequences
  • An antibody that specifically binds to human IL17 characterized in comprising a CDRIH of SEQ ID NO: l, CDR2H of SEQ ID NO:2, CDR3H of SEQ ID NO:3, and CDRIL of SEQ ID NO:4, CDR2L of SEQ ID NO:5, CDR3L of SEQ ID NO:6.
  • the antibody according to any of embodiments 1 to 2 characterized in comprising a) a variable heavy chain domain (VH) of SEQ ID NO:9, or of SEQ ID NO: 10, and b) a variable light chain domain (VL) of SEQ ID NO: 11, or of SEQ ID NO: 12. 4.
  • the antibody according to any of embodiments 1 to 2 characterized in comprising a) a variable heavy chain domain (VH) of SEQ ID NO: 10, and b) a variable light chain domain (VL) of SEQ ID NO: 12.
  • the antibody according to any of embodiments 1 to 2 characterized in comprising a) a variable heavy chain domain (VH) of SEQ ID NO:9, and b) a variable light chain domain (VL) of SEQ ID NO: 11.
  • the antibody according to any of the preceding embodiments characterized in being of IgGl or IgG4 subclass.
  • the antibody according to any of the preceding embodiments characterized in being of IgG4 subclass with the mutations S228P and L235E (numbering according to the EU index of Kabat). 10. The antibody according to any of the preceding embodiments, characterized in being of IgG4 subclass with the mutations S228P, L235E and P329G (numbering according to the EU index of Kabat).
  • composition characterized by comprising an antibody according to embodiments 1 to 10.
  • an antibody according to embodiments 1 to 10 for manufacture of a medicament for the treatment of cancer, or inflammatory diseases, autoimmune diseases, rheumatoid arthritis, psoratic arthritis, muscle diseases, e.g. muscular dystrophy, multiple sclerosis, chronic kidney diseases, bone diseases, e.g. bone degeneration in multiple myeloma, systemic lupus erythematosus, lupus nephritis, and vascular injury.
  • Expression vectors characterized by comprising a nucleic acid according to embodiment 15 for the expression of the antibody according to embodiments 1 to 10 in a prokaryotic or eukaryotic host cell.
  • Prokaryotic or eukaryotic host cell comprising a vector according to embodiment 16.
  • Desired gene segments can be prepared from oligonucleotides made by chemical synthesis.
  • the gene segments which are flanked by singular restriction endonuclease cleavage sites, were assembled by annealing and ligation of oligonucleotides including PCR amplification and subsequently cloned via the indicated restriction sites e.g. Kpnl/ Sacl or Ascl/Pacl into a pPCRScript (Stratagene) based pGA4 cloning vector.
  • the DNA sequences of the subcloned gene fragments were confirmed by DNA sequencing.
  • Gene synthesis fragments were ordered according to given specifications at Geneart (Regensburg, Germany). All gene segments encoding light and heavy chains of IL-17 antibodies were synthesized with a 5 '-end DNA sequence coding for a leader peptide which targets proteins for secretion in eukaryotic cells, and unique restriction sites at the 5' and 3' ends of the synthesized gene.
  • DNA sequences were determined by double strand sequencing performed at
  • variants of expression plasmids for transient expression e.g. in HEK293 EBNA or HEK293-F cells
  • stable expression e.g. in CHO cells
  • a cDNA organization with a CMV-Intron A promoter or on a genomic organization with a CMV promoter were applied.
  • IgG4_SPLE In case of IgG4_SPLE the intron between the CHI domain and the hinge domain was removed, keeping the remainder of the antibody gene in a genomic organization. The intron-deleted version of IgG4_SPLE no longer shows hingeless antibodies as a result of a splice artefact commonly seen in IgG4_SPLE encoded in total genomic organization.
  • the vectors contained: an origin of replication which allows replication of this plasmid in E. coli, and
  • the transcription unit of the antibody gene is composed of the following elements:
  • human antibody chain (heavy chain, modified heavy chain or light chain) either as cDNA or as genomic organization with an the
  • Recombinant immunoglobulin variants were expressed by transient transfection of human embryonic kidney 293 cells using the FreeStyleTM 293 Expression System according to the manufacturer's instruction (Invitrogen, USA). Briefly, suspension HEK293 cells were cultivated in FreeStyleTM 293 Expression medium at 37°C/8 % C0 2 and the cells were seeded in fresh medium at a density of l-2xl0 6 viable cells/ml on the day of transfection.
  • DNA-293fectinTM complexes were prepared in Opti-MEM ® I medium (Invitrogen, USA) using 325 ⁇ of HEK293 and 250 ⁇ g of heavy and light chain plasmid DNA in a 1 : 1 molar ratio for a 250 ml final transfection volume for monospecific antibodies. "For expression yield and product quality optimization the ratio can be varied.
  • DNA-293fectin complexes were prepared in Opti-MEM ® I medium (Invitrogen, USA) using 325 ⁇ of 293fectinTM (Invitrogen, Germany) and 250 ⁇ g of "Knobs-into-hole" heavy chain 1 and 2 and light chain 1 and 2 plasmid DNA in a 1 : 1 : 1 : 1 molar ratio for a 250 ml final transfection volume.
  • Antibody containing cell culture supematants were harvested 7 days after transfection by centrifugation at 14000 g for 30 minutes and filtered through a sterile filter (0.22 ⁇ ). Supematants were stored at -20° C until purification.
  • the protein concentration of purified antibodies and derivatives was determined by determining the optical density (OD) at 280 nm, using the molar extinction coefficient calculated on the basis of the amino acid sequence according to Pace, C.N., et. al, Protein Science 4 (1995) 2411-1423.
  • the concentration of antibodies and derivatives in cell culture supematants was estimated by immunoprecipitation with Protein A Agarose-beads (Roche). 60 ⁇ ⁇
  • Protein A Agarose beads are washed three times in TBS-NP40 (50 mM Tris, pH 7.5, 150 mM NaCl, 1% Nonidet-P40). Subsequently, 1 -15 mL cell culture supernatant are applied to the Protein A Agarose beads pre-equilibrated in TBS-NP40. After incubation for at 1 h at room temperature the beads are washed on an Ultrafree-MC-filter column (Amicon] once with 0.5 mL TBS-NP40, twice with 0.5 mL 2x phosphate buffered saline (2xPBS, Roche) and briefly four times with 0.5 mL 100 mM Na-citrate pH 5,0.
  • TBS-NP40 50 mM Tris, pH 7.5, 150 mM NaCl, 1% Nonidet-P40.
  • 1 -15 mL cell culture supernatant are applied to the Protein A Agarose beads pre-equilibrated in TBS-NP40.
  • Bound antibody is eluted by addition of 35 ⁇ NuPAGE® LDS Sample Buffer (Invitrogen). Half of the sample is combined with NuPAGE® Sample Reducing Agent or left unreduced, respectively, and heated for 10 min at 70°C. Consequently, 20 ⁇ are applied to an 4-12%
  • NuPAGE® Bis-Tris SDS-PAGE (Invitrogen) (with MOPS buffer for non-reduced SDS-PAGE and MES buffer with NuPAGE® Antioxidant running buffer additive (Invitrogen) for reduced SDS-PAGE) and stained with Coomassie Blue.
  • the concentration of antibodies and derivatives in cell culture supematants was measured by Protein A-HPLC chromatography. Briefly, cell culture supematants containing antibodies and derivatives that bind to Protein A were applied to a HiTrap Protein A column (GE Healthcare) in 50 mM K2HP04, 300 mM NaCl, pH 7.3 and eluted from the matrix with 550 mM acetic acid, pH 2.5 on a Dionex HPLC-System. The eluted protein was quantified by UV absorbance and integration of peak areas. A purified standard IgGl antibody served as a standard.
  • the concentration of antibodies and derivatives in cell culture supematants was measured by Sandwich-IgG-ELISA. Briefly, StreptaWell High Bind Strepatavidin A-96 well microtiter plates (Roche) were coated with 100 ⁇ biotinylated anti-human IgG capture molecule F(ab')2 ⁇ h-Fcgamma> BI (Dianova) at 0.1 ⁇ g/mL for 1 h at room temperature or alternatively over night at
  • Antibodies were purified from cell culture supematants by affinity chromatography using Protein A-SepharoseTM (GE Healthcare, Sweden) and Superdex200 size exclusion chromatography. Briefly, sterile filtered cell culture supematants were ap lied on a HiTrap ProteinA HP (5 ml) column equilibrated with PBS buffer (10 mM Na 2 HP0 4 , 1 mM KH 2 P0 4 , 137 mM NaCl and 2.7 mM KC1, pH 7.4). Unbound proteins were washed out with equilibration buffer.
  • PBS buffer 10 mM Na 2 HP0 4 , 1 mM KH 2 P0 4 , 137 mM NaCl and 2.7 mM KC1, pH 7.4
  • Antibody and antibody variants were eluted with 0.1 M citrate buffer, pH 2.8, and the protein containing fractions were neutralized with 0.1 ml 1 M Tris, pH 8.5. Then, the eluted protein fractions were pooled, concentrated with an Amicon Ultra centrifugal filter device (MWCO: 30 K, Millipore) to a volume of 3 ml and loaded on a Superdex200 HiLoad 120 ml 16/60 gel filtration column (GE Healthcare, Sweden) equilibrated with 20mM Histidin, 140 mM NaCl, pH 6.0. Fractions containing purified antibodies with less than 5 % high molecular weight aggregates were pooled and stored as 1.0 mg/ml aliquots at -80°C.
  • MWCO Amicon Ultra centrifugal filter device
  • the NuPAGE® Pre-Cast gel system (Invitrogen) was used according to the manufacturer's instruction. In particular, 4-20 % NuPAGE® No vex® TRIS-Glycine Pre-Cast gels and a Novex® TRIS-Glycine SDS running buffer were used. Reducing of samples was achieved by adding NuPAGE® sample reducing agent prior to running the gel.
  • Protein A purified antibodies were applied to a Tosoh TSKgel G3000SW column in 300 mM NaCl, 50 mM KH2P04/K2HP04, pH 7.5 on an Agilent HPLC 1100 system or to a Superdex 200 column (GE Healthcare) in 2 x PBS on a Dionex HPLC-System.
  • the eluted protein was quantified by UV absorbance and integration of peak areas.
  • BioRad Gel Filtration Standard 151-1901 served as a standard.
  • the total deglycosylated mass of the antibodies was determined and confirmed via electrospray ionization mass spectrometry (ESI-MS). Moreover potential sideproducts mispairing were detected and relatively quantified. Briefly, 100 ⁇ g purified antibodies at a protein concentration of up to 3 mg/ml were deglycosylated with 14 or 28 U N-Glycosidase F (Roche) in 100 mM NaH2P04/Na2HP04, pH 7 at 37 or 45°C for 16 or 2 h and subsequently desalted via HPLC on a Sephadex G25 column (GE Healthcare). The mass of the respective heavy and light chains was determined by ESI-MS after deglycosylation and reduction.
  • ESI-MS electrospray ionization mass spectrometry
  • Immunization was performed within 20 weeks using 5 female Balb/c mice using 250 (lx) and 100 ⁇ g (3x) recombinant human IL17 from Peprotech (http://www.peprotech.com; Cat.No.: 200-17 in 1% PBS with 1% Albumin) per mouse. Hybridoma generation.
  • the mouse lymphocytes were isolated and fused with a mouse myeloma cell line using PEG based standard protocols to generate hybridomas.
  • the resulting hybridomas were then screened for the production of antigen- specific antibodies. From resulting hybridomas mouse clone ⁇ IL17> 9C6- 2B6 was selected using the binding to IL-17 subtypes measured by ELISA (see
  • Example 4 Humanization of mouse clone ⁇ IL17> 9C6-2B6 resulted in the humanized variants ⁇ IL17> 9C6-2B6-134 (with the humanized variant of VH, ⁇ IL17> 9C6-2B6-HC134 and humanized variant of VL, ⁇ IL17> 9C6-2B6-LC134 of SEQ ID No.
  • ⁇ IL-17> antibodies Light and heavy chains of the antibodies were constructed in genomic, partly genomic or cDNA-derived expression vectors as described. The plasmids were amplified in E. coli, purified, and subsequently HEK293 cells were transfected for transient expression of recombinant proteins. After 7 days of cultivation, the supematants of HEK293 cells were harvested, filtered and the antibodies were purified.
  • Antibodies were purified from cell culture supematants by affinity chromatography using MabSelect SuReTM (GE Healthcare, Sweden). The subsequent chromatographic steps (size exclusion chromatography (Superdex200 HiLoad 120 ml 16/60 gel filtration column, GE Healthcare, Sweden) or ion exchange chromatography (MacroPrep CHT Typell 10 ml, Bio-Rad plus size exclusion chromatography) was chosen in respect to the individual product related sideproducts of the antibodies after MabSelect SuReTM chromatography.
  • the antibody pools were prepared for hydrophobic interaction chromatography by rebuffering in 10 mM NaH 2 P0 4 , 20 mM MES, 50 mM NaCl, 0.1 mM CaCl 2 , pH 7.5.
  • equilibration buffer 10 mM NaH 2 P0 4 , 20 mM MES, 50 mM NaCl, 0.1 mM CaCl 2 , pH 7.5
  • the antibodies were applied to the CHT column, washed with equilibration buffer and eluted in an linear gradient to 10 mM NaH 2 P0 4 , 20 mM MES, 500 mM NaCl, 0.1 mM CaCl 2 , pH 7.5.
  • the antibody containing fractions (from ion exchange chromatography or MabSelect SuRe affinity chromatography) were pooled and further purified by size exclusion chromatography using a Superdex 200 26/60 GL (GE Healthcare, Sweden) column equilibrated with 20 mM histidine, 140 mM NaCl, pH 6.0.
  • the antibody containing fractions were pooled, concentrated to the required concentration using Vivaspin ultrafiltration devices (Sartorius Stedim Biotech S.A., France) and stored at -80°C.
  • Size exclusion chromatography for the determination of the aggregation and oligomeric state of antibodies was performed by HPLC chromatography. Briefly, purified antibodies at various levels of the purification process were applied to a Tosoh TSKgel G3000SW column in 300 mM NaCl, 50 mM KH2P04/K2HP04, H 7.5 on an Agilent HPLC 1100 system or to a Superdex 200 column (GE Healthcare) in 2 x PBS on a Dionex HPLC-System. The eluted protein was quantified by UV absorbance and integration of peak areas. BioRad Gel Filtration Standard 151-1901 served as a standard. Table 1: Yield, purity profile and aggregation tendencies (aggregate tendencies are reflected by % monomers after Protein A)
  • Binding to IL-17 and Crossreactivity with IL17 subtypes measured by ELISA NUNC® Maxisorp plates (96-well) are coated with recombinant human IL-17 (Peprotech # 200-17, www.peprotech.com) at a concentration of 0 ⁇ g/ml in PBS (lOOml/well). Plates are incubated at 37°C on an orbital shaker with agitation for 2 hours. Thereafter coating solution is removed and ⁇ /well PBSTC (phosphate buffered saline, 0,05%> Tween®20, 2%> chicken serum) is added. Plates are incubated at room temperature for 1 hour.
  • PBSTC phosphate buffered saline, 0,05%> Tween®20, 2%> chicken serum
  • Blocking solution is removed and samples (blank: PBSTC, samples (10 ⁇ g/ml in PBS): anti-human IL-17 antibodies ⁇ IL17> 9C6-2B6, ⁇ IL17> 9C6-2B6-134, ⁇ IL17> 9C6-2B6-136, Mab 16-7178-85 of eBioscience (www.ebioscience.com); MAB 317 of R&D Systems (www.rndsystems.com), NVP-AIN-497 (WO 2006/013107); are added to the plate ( ⁇ /well). Plates are incubated at room temperature with agitation.
  • Second antibody is removed, plates are washed three times with 200 ⁇ 1 ⁇ 11 PBST (phosphate buffered saline, 0,05% Tween®20) and ⁇ /well ABTS® (Roche Diagnostics GmbH) is added. Optical density is measured at 405/492nm in relation to IL-17A binding (set as 100%). Binding to other human IL-17 subtypes (IL-17B, IL-17C, IL-17D, IL-17E and IL-17F) were performed with the same assay format. Results are shown in table 4. The results show that antibody with the most similar binding behavior against the different IL17 subtypes is Mab MAB 317 of R&D Systems (www.rndsystems.com).
  • the assay is performed as detection of hIL-8 production of CCD-25SK cells (skin fibroblasts, ATCC No: CRL-1474) after stimulation with IL-17A and TNF-alpha with preincubation of anti-IL-17 antibodies.
  • CCD-25SK cells have the IL-17 receptor.
  • Soluble IL-17A binds to the these IL-17 receptor.
  • Antibodies against IL-17A bind to IL-17A.
  • the mechanism is only working in the presence of TNFalpha.
  • the cells produce hIL-6 and hIL-8 which can be detected by ELISA as a read out.
  • the measured hIL-6 and hIL-8 give the information in which concentrations anti-IL-17 antibodies inhibit the stimulation of CCD-25SK cells by IL-17.
  • CCD-25SK cells were seeded with a cell density of 2,5x10 4 cells/well in a 48-well plate (volume 0,45ml/well) and incubated for 24h at 37°C and 5%C0 2 . After overnight incubation the cells were treated with anti-IL-17 antibodies for 30 minutes with end concentrations of 9000; 3000;1000; 333,3; 111,1; 37,03; 12,34; and 4,11 ng/ml. Each antibody dilution series was made with medium, 50 ⁇ 1 ⁇ 11 (lOx concentrated). After 30 min the cells were stimulated with a mixture of lOng/ml IL-17A and 50pg/ml TNF-alpha.
  • hIL-6 and hIL-8 ELISA was performed as follows. ⁇ diluted capture antibody was added to each well and incubated overnight at 4°C. Dilutions were made with coating buffer. Plates were aspirated, washed with 200 ⁇ 1 ⁇ 11 for 3 times, blocked with 200 ⁇ 1 ⁇ 11 assay diluent, and incubated for lh at RT. The plates were aspirated and washed with 200 ⁇ 1 ⁇ 11 for 3 times.
  • Standard dilution series 400pg/ml; 200pg/ml; lOOpg/ml; 50pg/ml; 25pg/ml; 12,5pg/ml; 6,3pg/ml and assay diluent as negative control.
  • Sample dilution was 1 :200. Plates were aspirated and washed with 250 ⁇ 1 ⁇ 11 for 4 times. ⁇ conjugate was added to each well. The conjugate was prepared with detection antibody and enzyme reagent 1 :250 diluted in assay diluent. Plates were aspirated and washed with 250 ⁇ 1 ⁇ 11 for 6 times.
  • the ⁇ IL-17> antibodies were tested for inhibition of an IL-17-induced production of pro-inflammatory cytokines (e.g. IL-6, -8) by human adult fibroblast-like synoviocytes obtained from RA patients (human fibroblast-like synoviocytes- rheumatoid arthritis (HFLS-RA)). After establishment of a dose-response response of different RA-FLS donors, the potency of the antibodies was assessed.
  • pro-inflammatory cytokines e.g. IL-6, -8
  • human adult fibroblast-like synoviocytes obtained from RA patients (human fibroblast-like synoviocytes- rheumatoid arthritis (HFLS-RA)
  • HFLS-RA human fibroblast-like synoviocytes- rheumatoid arthritis
  • HFLS-RA (Cat. #408RA-05a) were purchased from Cell Applications Inc. (San Diego, CA, USA; German distributor: tebu-bio, Offenbach, Germany). They are cryopreserved at second passage and can be cultured and propagated at least 5 population doublings. HFLS are long known for their role in joint destruction by producing cytokines and metalloproteinases that contribute to cartilage degradation (Firestein, G.S., et al, J. Immunol.
  • Proinflammatory cytokines induce the proliferation, collagenase and aggrecanase production and GM-CSF secretion on HFLS (Alvaro, J.M., et al, J. Clin. Immunol. 13(3) (1993) 212; Yamanishi, Y., et al, J. Immunol. 168(3) (2002) 1405).
  • Cells were thawed, expanded in Synoviocyte Growth Medium (Cell Applications, Inc.; Cat. #415-500), detached with Accutase (PAA Laboratories GmbH, Pasching, Austria; Cat. #L11-007) before approx. 2xl0 4
  • HFLS-RA cells/well were seeded in 200 ⁇ 1 ⁇ 11 medium in 96wF cell culture plates (Costar/Corning Life Sciences, Amsterdam, The Netherlands; Cat. #3596).
  • cytokines and optionally antibodies
  • medium was removed and 150 ⁇ 1 ⁇ of the corresponding cytokine (optional: antibody) dilution was added: 0-10 ⁇ g/ml rec. human IL-17A (PeproTech, Hamburg, Germany; Cat. #200-17); 0-25 ⁇ g/ml rec. human TWEAK (R&D Systems, Wiesbaden, Germany; Cat. #1090-TW/CF), or 0- ⁇ g/ml rec. human TNFa (R&D Systems; Cat.
  • Cynomolgous monkey (Maccaca Fasicularis) cytokine release assay inhibition of cynomolgous IL-17A induced IL-6 and IL-8 production
  • Cynomolgous dermal fibroblasts (CDF) cells produce cynomolgous IL-6 and IL-8 in response to human or cynomolgous IL-17A stimulation.
  • the assay is performed to measure the inhibition of this cynomolgous IL-17A stimulated IL-6 and IL-8 production by CDF cells following preincubation of the cells with anti-IL-17 antibodies raised against human IL-17 prior to stimulation.
  • CDF cells are seeded with a cell density of 2xl0 5 cells/ml in a volume of 0.5ml in a
  • the cells are stimulated with lOOng/ml IL-17A (50 ⁇ 1 of lOOOng/ml lOx concentration) and incubated overnight (18h) at 37°C and 5%C0 2 . After the incubation period, supematants are transferred into fresh tubes and either analyzed immediately or stored at -80°C until analysis by ELISA.
  • hIL-6 and hIL-8 ELISA were shown to be cross-reactive with their respective cynomolgous cytokines and are used to quantitate cytokine levels.
  • ⁇ diluted capture antibody is added to each well and incubated overnight at 4°C. Dilutions are made with coating buffer.
  • Plates are aspirated, washed with 200 ⁇ 1 ⁇ 11 for 3 times, blocked with 200 ⁇ 1 ⁇ 11 assay diluent, and incubated for lh at RT. The plates are aspirated and washed with 200 ⁇ 1 ⁇ 11 for 3 times. ⁇ standard and samples are added and incubated for 2h at RT according to the manufacturer's instructions. Plates are aspirated and washed with 250 ⁇ 1 ⁇ 11 for at least 3 times. ⁇ conjugate is added to each well. The conjugate is prepared with detection antibody and enzyme reagent 1 :250 diluted in assay diluent. Plates are aspirated and washed with 250 ⁇ 1 ⁇ 11 for at least 3 times. ⁇ substrate was added to each well and incubated until sufficient color had developed for reading. After incubation the reaction is stopped with 50 ⁇ 1 ⁇ 11 1M H 2 S0 4 and read on the plate reader at a wavelength of 450nm within 30 min.
  • the ⁇ IL17> binding affinity of antibodies was measured by Surface Plasmon Resonance (SPR) using a BIAcore® T100 instrument (GE Healthcare) at 25°C.
  • SPR-technology is based on the measurement of the refractive index close to the surface of a gold coated biosensor chip. Changes in the refractive index indicate mass changes on the surface caused by the interaction of immobilized ligand with analyte injected in solution. The mass increases if molecules bind immobilized ligands on the surface, and vice versa, the mass decreases in case of dissociation of the analyte from the immobilized ligand (reflecting complex dissociation).
  • SPR allows a continuous real-time monitoring of ligand/analyte binding and thus the determination of the association rate constant (ka), the dissociation rate constant (kd), and of the equilibrium constant (KD).
  • the dissociation phase was monitored for up to 5 min and triggered by switching from the sample solution to running buffer.
  • the surface was regenerated by two times 60 sec. washing with a glycin pH 2.1 solution at a flow rate of 30 ⁇ /min.
  • the Langmuir 1 :1 model was used. Results are shown in Table 17 below.
  • Viscosity measurement is essentially performed as described in He, F., et al, Analytical Biochemistry 399(1) (2009) 141-3. Briefly, samples are concentrated to various protein concentrations in 200 mM arginine succinate, pH 5.5, before polystyrene latex beads (300 nm diameter) and Polysorbate 20 (0.02 % v/v) are added. Samples are transferred into an optical 384-well plate by centrifugation through a 0.4 ⁇ filter plate and covered with paraffine oil. The apparent diameter of the latex beads is determined by dynamic light scattering at 25 °C.
  • HMW high molecular weight
  • Table 8 Stability of bispecific antibodies was assessed by DLS and SE.-HPLC.

Abstract

The present invention relates to antibodies against human IL17A (IL17 antibody), methods for their production, pharmaceutical compositions containing said antibodies, and uses thereof.

Description

Antibodies against human IL17 and uses thereof
The present invention relates to antibodies against human IL17A (IL17 antibody), methods for their production, pharmaceutical compositions containing said antibodies, and uses thereof.
Background of the Invention Human IL-17A (CTLA-8, Swiss Prot Q 16552, further named as IL-17 or IL17;
SEQ ID NO: 22)) is a pro-inflammatory cytokine produced by a subset of memory T cells (called Thl7) that has been implicated in the pathogenesis of MS. IL-17A plays a role in the induction of other inflammatory cytokines, chemokines and adhesion molecules. Treatment of animals with IL-17A neutralizing antibodies decreases disease incidence and severity in autoimmune encephalomyelitis
(Komiyama, Y., et al, J. Immunol. 177 (2006) 566-573). IL-17A is over-expressed in the cerebrospinal fluid of MS patients (Hellings, P.W., et al, Am. J. Resp. Cell Mol. Biol. 28 (2003) 42-50; Matusevicius, D., et al, Multiple Sclerosis 5 (1999) 101-104; WO 2005/051422). In addition, IL-17A neutralizing antibodies reduce severity and incidence of mouse RA model of collagen induced arthritis, and high levels of IL-17A can be detected in the synovial fluid of inflamed joints from RA patients (Ziolkowska, M., et al, J. Immunol. 164 (2000) 2832-2838; Kotake, S., et al, J. Clin. Invest. 103 (1999) 1345-1352; Hellings, P.W., et al, Am. J. Resp. Cell Mol. Biol. 28 (2003) 42-50). WO 96/17939, US 5,716,623; WO 95/18826; WO 97/15320; WO 99/35276 and WO 00/69436 WO 95/18826 US 6,274,711, US 6,274,711, WO 97/15320, US 6,063,372, WO 2006/013107 and WO 2008/02115 relate to IL-17A and antibodies against IL-17A. WO 2010/102251 relates IL17 binding proteins.
Summary of the Invention One aspect of the invention is an antibody that specifically binds to human IL17 characterized in comprising a CDR1H of SEQ ID NO: l, CDR2H of SEQ ID NO:2, CDR3H of SEQ ID NO:3, and CDRIL of SEQ ID NO:4, CDR2L of SEQ ID NO:5, CDR3L of SEQ ID NO:6.
In one embodiment such IL17 antibody is a chimeric or humanized variant. In one embodiment the antibody is characterized in comprising a) a variable heavy chain domain (VH) of SEQ ID NO:9, or of SEQ ID NO: 10, and b) a variable light chain domain (VL) of SEQ ID NO: 11 , or of SEQ ID NO: 12. In one embodiment the antibody is characterized in comprising a) a variable heavy chain domain (VH) of SEQ ID NO: 10, and b) a variable light chain domain (VL) of SEQ ID NO: 12. In one embodiment the antibody is characterized in comprising a) a variable heavy chain domain (VH) of SEQ ID NO:9, and b) a variable light chain domain (VL) of SEQ ID NO: 11. In one embodiment the antibody is characterized in being of IgGl or IgG4 subclass.
In one embodiment the antibody which binds to human IL17 and being characterized by the above mentioned amino acid sequences and amino acid sequence fragments is of human IgGl isotype, in one embodiment with mutations L234A and L235A, in one embodiment with mutations L234A, L235A and P329G (numbering according to the EU index of Kabat).
In one embodiment the antibody which binds to human IL17 and being characterized by the above mentioned amino acid sequences and amino acid sequence fragments is of human IgG4 isotype, in one embodiment with mutations S228P and L235E, in one embodiment with mutations S228P, L235E and P329G (numbering according to the EU index of Kabat).
A further embodiment of the invention is a pharmaceutical composition comprising an antibody according to the invention.
A further embodiment of the invention is the use of an antibody according to the invention for the manufacture of a pharmaceutical composition.
A further embodiment of the invention is a nucleic acid encoding an antibody according to the invention. A further embodiment of the invention is a nucleic acid encoding a heavy chain variable domain and/or a light chain variable domain of an antibody according to the invention.
The invention further provides expression vectors containing nucleic acid according to the invention capable of expressing said nucleic acid in a prokaryotic or eukaryotic host cell, and host cells containing such vectors for the recombinant production of such an antibody.
The invention further comprises a prokaryotic or eukaryotic host cell comprising a vector according to the invention. The invention further comprises a method for the production of a recombinant chimeric or humanized antibody according to the invention, characterized by expressing a nucleic acid according to the invention in a prokaryotic or eukaryotic host cell and recovering said antibody from said cell or the cell culture supernatant. The invention further comprises the antibody obtainable by such a recombinant method.
Antibodies according to the invention show benefits for patients in need of an IL17 targeting therapy. The antibodies according to the invention have valuable properties causing a benefit for a patient suffering from a cancer disease, especially suffering from colon, lung, or pancreatic cancer or from inflammatory diseases, especially from autoimmune diseases, rheumatoid arthritis, psoratic arthritis, muscle diseases, e.g. muscular dystrophy, multiple sclerosis, chronic kidney diseases, bone diseases, e.g. bone degeneration in multiple myeloma, systemic lupus erythematosus, lupus nephritis, and vascular injury.
A further embodiment of the invention is an antibody according to the invention for use in the treatment of cancer, inflammatory diseases, autoimmune diseases, rheumatoid arthritis, psoratic arthritis, muscle diseases, e.g. muscular dystrophy, multiple sclerosis, chronic kidney diseases, bone diseases, e.g. bone degeneration in multiple myeloma, systemic lupus erythematosus, lupus nephritis, and vascular injury, especially for the treatment of systemic lupus erythematosus, lupus nephritis. A further embodiment of the invention is an antibody according to the invention for manufacture of a medicament for the treatment of cancer, inflammatory diseases, autoimmune diseases, rheumatoid arthritis, psoratic arthritis, muscle diseases, e.g. muscular dystrophy, multiple sclerosis, chronic kidney diseases, bone diseases, e.g. bone degeneration in multiple myeloma, systemic lupus erythematosus, lupus nephritis, and vascular injury, especially for the treatment of systemic lupus erythematosus, lupus nephritis.
The invention further provides a method for treating a patient suffering from cancer, especially from colon, lung, or pancreatic cancer or from inflammatory diseases, especially from autoimmune diseases, rheumatoid arthritis, psoratic arthritis, muscle diseases, e.g. muscular dystrophy, multiple sclerosis, chronic kidney diseases, bone diseases, e.g. bone degeneration in multiple myeloma, systemic lupus erythematosus, lupus nephritis, and vascular injury, comprising administering to a patient diagnosed as having such a disease (and therefore being in need of such a therapy) an effective amount of the antibody which binds to human IL17 according to the invention. The antibody is administered preferably in a pharmaceutical composition.
The invention further comprises a pharmaceutical composition comprising an antibody according to the invention, optionally together with a buffer and/or an adjuvant useful for the formulation of antibodies for pharmaceutical purposes.
The invention further provides a pharmaceutical composition comprising an antibody according to the invention in a pharmaceutically acceptable carrier. In one embodiment, the pharmaceutical composition may be included in an article of manufacture or kit.
The antibodies according to the invention show benefits for human patients in need of a human IL17 targeting therapy and have valuable properties. In one embodiment the antibodies according to the invention are characterized by one or more of the following properties (as determined in Examples 4, 5 and 6): the antibody that specifically binds to human IL17 according to the invention a) shows no cross reactivity with IL17B, IL17C, IL17D, IL17F ( which means that the binding to IL17B, IL17C, IL17D and IL17F is 0% compared to the binding to IL17A , which is set as 100%); b) inhibits IL17 induced IL8 cytokine stimulation of CCD-25SK cells with an IC50 value of 2.0 nM or lower( e.g. with an IC50 value between 2.0 nM and 0.0 nM); c) inhibits IL17 induced IL6 cytokine stimulation of CCD-25SK cells with an IC50 value of 5.0 nM or lower ( e.g. with an IC50 value between 5.0 nM and 0.0 nM); preferably with an IC50 value of 2.0 nM or lower; d) inhibits IL17 induced IL6 cytokine stimulation of human fibroblast-like synoviocytes-rheumatoid arthritis (HFLS-RA) with an IC50 value of 2.0 nM or lower ( e.g. with an IC50 value between 2.0 nM and 0.0 nM); preferably with an IC50 value of 1.0 nM or lower; e) inhibits IL17 induced IL8 cytokine stimulation of human fibroblast-like synoviocytes-rheumatoid arthritis (HFLS-RA)with an IC50 value of 1.5 nM or lower ( e.g. with an IC50 value between 1.5 nM and 0.0 nM); preferably with an
IC50 value of 1.0 nM or lower and/or; f) binds to IL-17 with a KD value of binding affinity of 0.3 nM or lower. Detailed Description of the Invention
The term "antibody" encompasses the various forms of antibody structures including but not being limited to whole antibodies and antibody fragments. The antibody according to the invention is preferably a humanized antibody, chimeric antibody, or further genetically engineered antibody as long as the characteristic properties according to the invention are retained. "Antibody fragments" comprise a portion of a full length antibody, preferably the variable domain(s) thereof, or at least the antigen binding portion thereof. Examples of antibody fragments include diabodies, single-chain antibody molecules, and multispecific antibodies (e.g. bispecific antibodies) formed or derived from antibody fragments. scFv antibodies are, e.g., described in Huston, J.S., Methods in Enzymol. 203 (1991) 46-52. In addition, antibody fragments comprise single chain polypeptides having the characteristics of a VH domain, namely being able to assemble together with a VL domain, or of a VL domain binding to IL-17, namely being able to assemble together with a VH domain to a functional antigen binding site and thereby providing the properties of an antibody according to the invention.
The terms "monoclonal antibody" or "monoclonal antibody composition" as used herein refer to a preparation of antibody molecules of a single amino acid composition. The term "humanized antibody" refers to antibodies in which the framework and/or "complementary determining regions" (CDR) have been modified to comprise the CDR of an immunoglobulin of different species as compared to that of the parent immunoglobulin. In a preferred embodiment, a non-human ( e.g. mouse, rabbit or hamster) CDR is grafted into the framework region of a human antibody to prepare the "humanized antibody". See, e.g., Riechmann, L., et al, Nature 332 (1988) 323- 327; and Neuberger, M.S., et al., Nature 314 (1985) 268-270.
The term "chimeric antibody" refers to a monoclonal antibody comprising a variable region, i.e., binding region, from mouse and at least a portion of a constant region derived from a different source or species, usually prepared by recombinant DNA techniques. Chimeric antibodies comprising for example a mouse variable region and a human constant region. Such mouse/human chimeric antibodies are the product of expressed immunoglobulin genes comprising DNA segments encoding rat immunoglobulin variable regions and DNA segments encoding human immunoglobulin constant regions. Other forms of "chimeric antibodies" encompassed by the present invention are those in which the class or subclass has been modified or changed from that of the original antibody. Such "chimeric" antibodies are also referred to as "class-switched antibodies." Methods for producing chimeric antibodies involve conventional recombinant DNA and gene transfection techniques now well known in the art. See, e.g., Morrison, S.L., et al, Proc. Natl. Acad Sci. USA 81 (1984) 6851-6855; US Patent Nos. 5,202,238 and 5,204,244.
Human IL-17A (CTLA-8, Swiss Prot Q16552, further named as IL-17. IL17; SEQ ID NO: 22)) is a pro-inflammatory cytokine produced by a subset of memory T cells (called Thl7) that has been implicated in the pathogenesis of MS. IL-17A plays a role in the induction of other inflammatory cytokines, chemokines and adhesion molecules. Treatment of animals with IL-17A neutralizing antibodies decreases disease incidence and severity in autoimmune encephalomyelitis (Komiyama, Y., et al, J. Immunol. 177 (2006) 566-573). IL-17A is over-expressed in the cerebrospinal fluid of MS patients (Hellings, P.W., et al, Am. J. Resp. Cell
Mol. Biol. 28 (2003) 42-50; Matusevicius, D., et al, Multiple Sclerosis 5 (1999) 101-104; WO 2005/051422). In addition, IL-17A neutralizing antibodies reduce severity and incidence of mouse RA model of collagen induced arthritis, and high levels of IL-17A can be detected in the synovial fluid of inflamed joints from RA patients (Ziolkowska, M., et al, J. Immunol. 164 (2000) 2832-2838; Kotake, S., et al, J. Clin. Invest. 103 (1999) 1345-1352; Hellings, P.W., et al, Am. J. Resp. Cell Mol. Biol. 28 (2003) 42-50).
As used herein, the terms, "binds to", "binding" or "specifically binding" refers to the binding of the antibody to an epitope of human IL17with sufficient affinity such that the antibody is useful as a therapeutic agent in targeting human IL17 according to the invention. The binding of the antibody to an epitope of the antigen human IL17 can be measured in an in vitro assay, preferably in an plasmon resonance assay (e.g. BIAcore, GE-Healthcare Uppsala, Sweden) with purified wild-type human antigen (preferably with IL17A homodimer) ( see e.g. Example 9). The affinity of the binding is defined by the terms ka (rate constant for the association of the antibody from the antibody/antigen complex), kd (dissociation constant), and KD (kd/ka). An antibody comprising a that specifically binds to human IL17 refers to an antibody binds to human IL17 with a binding affinity (KD) of 1.0 x 10"8 M or less, e.g. from 1.0 x 10"8 M to 1.0 x 10"13 M, in one embodiment from 1.0 x 10"9 M to 1.0 x 1013 M.
The term "epitope" denotes a protein determinant capable of specifically binding to an antibody. Epitopes usually consist of chemically active surface groupings of molecules such as amino acids or sugar side chains and usually epitopes have specific three dimensional structural characteristics, as well as specific charge characteristics. Conformational and nonconformational epitopes are distinguished in that the binding to the former but not the latter is lost in the presence of denaturing solvents.
One embodiment of the invention is an antibody that specifically binds to human IL17, characterized in that the antibody binds to the same epitope on human IL17 as an antibody which comprises a CDR1H of SEQ ID NO: l, CDR2H of SEQ ID
NO:2, CDR3H of SEQ ID NO:3, and CDRIL of SEQ ID NO:4, CDR2L of SEQ ID NO:5, CDR3L of SEQ ID NO:6.
One embodiment of the invention is an antibody that specifically binds to human IL17, characterized in that the antibody binds to the same epitope on human IL17 as an antibody which comprises a variable heavy chain domain (VH) of SEQ ID
NO: 10, and variable light chain domain of SEQ ID NO: 12.
One embodiment of the invention is an antibody that specifically binds to human IL17, characterized in that the antibody competes for binding to the same epitope on human IL17 as an antibody which comprises a CDR1H of SEQ ID NO: l, CDR2H of SEQ ID NO:2, CDR3H of SEQ ID NO:3, and CDRIL of SEQ ID NO:4, CDR2L of SEQ ID NO:5, CDR3L of SEQ ID NO:6.
One embodiment of the invention is an antibody that specifically binds to human IL17, characterized in that the antibody competes for binding to the same epitope on human IL17 as an antibody which comprises a variable heavy chain domain
(VH) of SEQ ID NO:10, and variable light chain domain of SEQ ID NO: 12.
Antibodies which compete for binding to the same epitope (and thus are likely to bind to the same epitope) cane identified by Surface Plasmon Resonance competition assay (e.g. via BIACORE epitope binning mode). The "variable domain" (variable domain of a light chain (VL), variable domain of a heavy chain (VR)) as used herein denotes each of the pair of light and heavy chain domains which are involved directly in binding the antibody to the antigen. The variable light and heavy chain domains have the same general structure and each domain comprises four framework (FR) regions whose sequences are widely conserved, connected by three "hypervariable regions" (or complementary determining regions, CDRs). The framework regions adopt a β-sheet conformation and the CDRs may form loops connecting the β-sheet structure. The CDRs in each chain are held in their three-dimensional structure by the framework regions and form together with the CDRs from the other chain the antigen binding site. The antibody's heavy and light chain CDR3 regions play a particularly important role in the binding specificity/affinity of the antibodies according to the invention and therefore provide a further object of the invention.
The term "antigen-binding portion of an antibody" when used herein refers to the amino acid residues of an antibody which are responsible for antigen-binding. The antigen-binding portion of an antibody comprises amino acid residues from the
"complementary determining regions" or "CDRs". "Framework" or "FR" regions are those variable domain regions other than the hypervariable region residues as herein defined. Therefore, the light and heavy chain variable domains of an antibody comprise from N- to C-terminus the domains FR1, CDR1, FR2, CDR2, FR3, CDR3, and FR4. Especially, CDR3 of the heavy chain is the region which contributes most to antigen binding and defines the antibody's properties. CDR and FR regions are determined according to the standard definition of Kabat, E.A., et al, Sequences of Proteins of Immunological Interest, 5th ed., Public Health Service, National Institutes of Health, Bethesda, MD (1991) and/or those residues from a "hypervariable loop".
The term"CDRlH" denotes the CDR1 region of the heavy chain variable region calculated according to Kabat (Kabat, E.A., et al., Sequences of Proteins of Immunological Interest, 5th ed., Public Health Service, National Institutes of
Health, Bethesda, MD (1991)). CDR2L, CDR3H, etc. mean the respective regions from the heavy(H) or light(L) chain. For example, an antigen binding site characterized by comprising CDR1H of SEQ ID NO:3 means that the antigen binding site comprises this amino acid sequence as a heavy chain variable chain CDR1 region in its variable heavy chain. For example, an antigen binding site characterized by comprising CDR1H of SEQ ID NO: l, CDR2H of SEQ ID NO:2, CDR3H of SEQ ID NO:3 means that the antigen binding sites comprises in its heavy chain as sequence of CDR1 SEQ ID NO: l, as sequence of CDR2 SEQ ID NO:2, and as sequence of CDR3 SEQ ID NO:3. The terms "nucleic acid" or "nucleic acid molecule" as used herein are intended to include DNA molecules and RNA molecules. A nucleic acid molecule may be single-stranded or double-stranded, but preferably is double-stranded DNA.
The term "amino acid" as used within this application denotes the group of naturally occurring carboxy a-amino acids comprising alanine (three letter code: ala, one letter code: A), arginine (arg, R), asparagine (asn, N), aspartic acid (asp, D), cysteine (cys, C), glutamine (gin, Q), glutamic acid (glu, E), glycine (gly, G), histidine (his, H), isoleucine (ile, I), leucine (leu, L), lysine (lys, K), methionine (met, M), phenylalanine (phe, F), proline (pro, P), serine (ser, S), threonine (thr, T), tryptophan (trp, W), tyrosine (tyr, Y), and valine (val, V). A nucleic acid is "operably linked" when it is placed into a functional relationship with another nucleic acid. For example, DNA for a presequence or secretory leader is operably linked to DNA for a polypeptide if it is expressed as a preprotein that participates in the secretion of the polypeptide; a promoter or enhancer is operably linked to a coding sequence if it affects the transcription of the sequence; or a ribosome binding site is operably linked to a coding sequence if it is positioned so as to facilitate translation. Generally, "operably linked" means that the DNA sequences being linked are colinear, and, in the case of a secretory leader, contiguous and in reading frame. However, enhancers do not have to be contiguous. Linking is accomplished by ligation at convenient restriction sites. If such sites do not exist, synthetic oligonucleotide adaptors or linkers are used in accordance with conventional practice.
As used herein, the expressions "cell", "cell line", and "cell culture" are used interchangeably and all such designations include progeny. Thus, the words "transformants" and "transformed cells" include the primary subject cell and cultures derived therefrom without regard for the number of transfers. It is also understood that all progeny may not be precisely identical in DNA content, due to deliberate or inadvertent mutations. Variant progeny that have the same function or biological activity as screened for in the originally transformed cell are included. The "Fc part" of an antibody is not involved directly in binding of an antibody to an antigen, but exhibits various effector functions. An "Fc part of an antibody" is a term well known to the skilled artisan and defined on the basis of papain cleavage of antibodies. Depending on the amino acid sequence of the constant region of their heavy chains, antibodies or immunoglobulins are divided into the classes: IgA, IgD, IgE, IgG and IgM, and several of these may be further divided into subclasses
(isotypes; the expressions "isotype" or "subclass" are used interchangeable herein), e.g. IgGl, IgG2, IgG3, and IgG4, IgAl, and IgA2. According to the heavy chain constant regions the different classes of immunoglobulins are called α, δ, ε, γ, and μ, respectively. The Fc part 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. Complement activation (CDC) is initiated by binding of complement factor Clq to the Fc part of most IgG antibody subclasses. While the influence of an antibody on the complement system is dependent on certain conditions, binding to Clq is caused by defined binding sites in the Fc part. Such binding sites are known in the state of the art and described, e.g., by Boackle, R.J., et al, Nature 282 (1979) 742-743; Lukas, T.J., et al, J. Immunol. 127 (1981) 2555-2560; Brunhouse, R., and Cebra, J.J., Mol. Immunol. 16 (1979) 907-917; Burton, D.R., et al, Nature 288 (1980) 338-344; Thommesen, J.E., et al, Mol. Immunol. 37 (2000) 995-1004; Idusogie, E.E., et al, J. Immunol. 164 (2000) 4178-4184; Hezareh, M., et al, J.
Virology 75 (2001) 12161-12168; Morgan, A., et al, Immunology 86 (1995) 319- 324; EP 0 307 434. Such binding sites are, e.g., L234, L235, D270, N297, E318, K320, K322, P331, and P329 (numbering according to EU index of Kabat used for the numbering of the constant domains, Kabat, E.A., et al., Sequences of Proteins of Immunological Interest, 5th ed., Public Health Service, National Institutes of
Health, Bethesda, MD (1991)). Antibodies of subclass IgGl, IgG2 and IgG3 usually show complement activation and Clq and C3 binding, whereas IgG4 does not activate the complement system and does not bind Clq and C3.
In one embodiment the antibody according to the invention is characterized in that the constant chains are of human origin. Such constant chains are well known in the state of the art and e.g. described by Kabat (see e.g. Johnson, G., and Wu, T.T., Nucleic Acids Res. 28 (2000) 214-218 and Kabat, E.A., et al, Sequences of Proteins of Immunological Interest, 5th ed., Public Health Service, National Institutes of Health, Bethesda, MD (1991)). For example, useful human heavy chain constant regions comprises an amino acid sequence SEQ ID NO: 15, SEQ ID
NO: 16, SEQ ID NO: 17 or SEQ ID NO: 18 (human IgGl subclass allotypes (Caucasian and Afroamerican or mutants L234A/L235A, and L234A/L235A/P329G), SEQ ID NO: 19, SEQ ID NO: 20, or SEQ ID NO: 21 (human IgG4 subclass or mutants L234A/L235A, and L234A/L235A/P329G). For example, a useful human light chain constant region comprises an amino acid sequence of a kappa-light chain constant region of SEQ ID NO: 13 or an amino acid sequence of a lambda-light chain constant region of SEQ ID NO: 14. In one embodiment the antibody according to the invention comprises a Fc part derived from human origin and preferably all other parts of the human constant regions. As used herein the term "Fc part derived from human origin" denotes a Fc part which is either a Fc part of a human antibody of the subclass IgGl, IgG2, IgG3 or IgG4, in one embodiment a Fc part from human IgGl subclass, a mutated Fc part from human IgGl subclass (preferably with a mutations L234A and L235A, or L234A, L235A and P329G ), a Fc part from human IgG4 subclass or a mutated Fc part from human IgG4 subclass (preferably with a mutations S228P and L235E, or
S228P, L235E and P329G). In one embodiment the antibody comprise the human heavy chain constant regions of SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17 or SEQ ID NO: 18 (human IgGl subclass allotypes (Caucasian and Afroamerican or mutants L234A/L235A, and L234A/L235A/P329G), SEQ ID NO: 19, SEQ ID NO: 20, or SEQ ID NO: 21 (human IgG4 subclass or mutants L234A/L235A, and L234A/L235A/P329G) (numbering according to the EU index of Kabat, E.A., et al, Sequences of Proteins of Immunological Interest, 5th ed., Public Health Service, National Institutes of Health, Bethesda, MD (1991). These human heavy chain constant regions can comprise additionally modifications and or mutations. In one embodiment the antibody according to the invention is of human IgGl subclass or of human IgG4 subclass. In one embodiment the antibody according to the invention is of human IgGl subclass. In one embodiment the antibody according to the invention is of human IgG4 subclass.
A further embodiment of the invention is a method for the production of an antibody according to the invention, characterized in that the sequence of a nucleic acid encoding the heavy chain of an antibody according to the invention and the nucleic acid encoding the light chain of said antibody are inserted into one or two expression vector(s), said vector(s) is/are inserted in a eukaryotic host cell, the encoded antibody is expressed and recovered from the host cell or the supernatant.
The antibodies according to the invention are preferably produced by recombinant means. Such methods are widely known in the state of the art and comprise protein expression in prokaryotic and eukaryotic cells with subsequent isolation of the antibody polypeptide and usually purification to a pharmaceutically acceptable purity. For the protein expression nucleic acids encoding light and heavy chains or fragments thereof are inserted into expression vectors by standard methods. Expression is performed in appropriate prokaryotic or eukaryotic host cells, such as
CHO cells, NSO cells, SP2/0 cells, HEK293 cells, COS cells, yeast, or E. coli cells, and the antibody is recovered from the cells (from the supernatant or after cells lysis).
Recombinant production of antibodies is well-known in the state of the art and described, for example, in the review articles of Makrides, S.C., Protein Expr. Purif.
17 (1999) 183-202; Geisse, S., et al, Protein Expr. Purif. 8 (1996) 271-282; Kaufman, R.J., Mol. Biotechnol. 16 (2000) 151-160; Werner, R.G., Arzneimittelforschung (Drug Res.) 48 (1998) 870-880.
The antibodies may be present in whole cells, in a cell lysate, or in a partially purified, or substantially pure form. Purification is performed in order to eliminate other cellular components or other contaminants, e.g., other cellular nucleic acids or proteins, by standard techniques, including column chromatography and others well known in the art. See Ausubel, F., et al. (eds.), Current Protocols in Molecular Biology, Greene Publishing and Wiley Interscience, New York (1987). Expression in NSO cells is described by, e.g., Barnes, L.M., et al, Cytotechnology
32 (2000) 109-123; Barnes, L.M., et al, Biotech. Bioeng. 73 (2001) 261-270. Transient expression is described by, e.g., Durocher, Y., et al, Nucl. Acids. Res. 30 (2002) E9. Cloning of variable domains is described by Orlandi, R., et al, Proc. Natl. Acad. Sci. USA 86 (1989) 3833-3837; Carter, P., et al, Proc. Natl. Acad. Sci. USA 89 (1992) 4285-4289; Norderhaug, L., et al, J. Immunol. Methods 204 (1997) 77-87. A preferred transient expression system (HEK 293) is described by Schlaeger, E.-J., and Christensen, K., in Cytotechnology 30 (1999) 71-83, and by Schlaeger, E.-J., in J. Immunol. Methods 194 (1996) 191-199. Monoclonal antibodies are suitably separated from the culture medium by conventional immunoglobulin purification procedures such as, for example, protein A-Sepharose, hydroxylapatite chromatography, dialysis, or affinity chromatography. DNA and R A encoding the monoclonal antibodies is readily isolated and sequenced using conventional procedures. The hybridoma cells can serve as a source of such DNA and RNA. Once isolated, the DNA may be inserted into expression vectors, which are then transfected into host cells, such as HEK 293 cells, CHO cells, or myeloma cells that do not otherwise produce immunoglobulin protein, to obtain the synthesis of recombinant monoclonal antibodies in the host cells. Nucleic acid molecules encoding amino acid sequence variants of anti-IL17 antibodies of the invention are prepared by a variety of methods known in the art. These methods include, but are not limited to, isolation from a natural source (in the case of naturally occurring amino acid sequence variants) or preparation by oligonucleotide-mediated (or site-directed) mutagenesis, PCR mutagenesis, and cassette mutagenesis of an earlier prepared variant or a non-variant version of anti-
IL17 antibody.
The heavy and light chain variable domains according to the invention are combined with sequences of promoter, translation initiation, constant region, 3' untranslated region, polyadenylation, and transcription termination to form expression vector constructs. The heavy and light chain expression constructs can be combined into a single vector, co-transfected, serially transfected, or separately transfected into host cells which are then fused to form a single host cell expressing both chains.
The anti-IL17 antibodies of the invention have valuable properties such as good developability and producibility in high yields (the antibodies contain e.g. no hotspots, for which the production conditions are often more restricted), good titers and yields and are producible in high amounts and with relatively low impurities (> 90% Monomer after Protein A (SE-HPLC) with an estimated purity after 2nd column (ESI-MS) > 95%) (see Example 2 and 3). Pharmaceutical Compositions
Pharmaceutical compositions of an IL17 antibody as described herein are prepared by mixing such antibody having the desired degree of purity with one or more optional pharmaceutically acceptable carriers (Remington's Pharmaceutical Sciences, 16th edition, Osol, A. (ed.) (1980)), in the form of lyophilized formulations or aqueous solutions. Pharmaceutically acceptable carriers are generally nontoxic to recipients at the dosages and concentrations employed, and include, but are not limited to: buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyl dimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride; benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as poly(vinylpyrrolidone); amino acids such as glycine, glutamine, asparagine, histidine, arginine, or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugars such as sucrose, mannitol, trehalose or sorbitol; salt-forming counter-ions such as sodium; metal complexes (e.g. Zn-protein complexes); and/or non-ionic surfactants such as polyethylene glycol (PEG). Exemplary pharmaceutically acceptable carriers herein further include interstitial drug dispersion agents such as soluble neutral-active hyaluronidase glycoproteins (sHASEGP), for example, human soluble PH-20 hyaluronidase glycoproteins, such as rhuPH20 (HYLENEX®, Baxter International, Inc.). Certain exemplary sHASEGPs and methods of use, including rhuPH20, are described in US Patent Publication Nos. 2005/0260186 and 2006/0104968. In one aspect, a sHASEGP is combined with one or more additional glycosaminoglycanases such as chondroitinases.
Exemplary lyophilized antibody formulations are described in US Patent No. 6,267,958. Aqueous antibody formulations include those described in US Patent No. 6,171,586 and WO 2006/044908, the latter formulations including a histidine-acetate buffer.
The formulation herein may also contain more than one active ingredients as necessary for the particular indication being treated, preferably those with complementary activities that do not adversely affect each other. The antibodies specifically binding to human IL17 according to the invention have especially valuable properties such as low viscosity and high stability (so that they can be produced without high aggregation and in good yields) (see Example 10). Such antibodies with their low viscosity and high stability are especially useful in highly concentrated formulations/compositions which can be used e.g. in a subcutaneous administration.
The antibodies according to the invention may be administered as the sole active ingredient or in conjunction with, e.g. as an adjuvant to or in combination to, other drugs e.g. immunosuppressive or immunomodulating agents or other anti- inflammatory agents, e.g. for the treatment or prevention of diseases mentioned above. For example, the antibodies as described herein may be used in combination with DMARD, e.g. Gold salts, sulphasalazine, antimalarias, methotrexate, D-penicillamine, azathioprine, mycophenolic acid, cyclosporine A, tacrolimus, sirolimus, minocycline, lefiunomide, glococorticoids; a calcineurin inhibitor, e.g. cyclosporin A or FK 506; a modulator of lymphocyte recirculation, e.g. FTY720 and FTY720 analogs; a mTOR inhibitor, e.g. rapamycin, 40-O-(2-hydroxyethyl)- rapamycin, CCI779, ABT578, AP23573 or TAFA-93; an ascomycin having immunosuppressive properties, e.g. ABT-281, ASM981, etc.; corticosteroids; cyclo-phosphamide; azathioprene; methotrexate; lefiunomide; mizoribine; mycophenolic acid; myco-phenolate mofetil; 15-deoxyspergualine or an immunosuppressive homologue, analogue or derivative thereof; immunosuppressive monoclonal antibodies, e.g., monoclonal antibodies to leukocyte receptors, e.g., MHC, CD2, CD3, CD4, CD7, CD8, CD25, CD28, CD40. CD45, CD58, CD80, CD86 or their ligands; other immunomodulatory compounds, e.g. a recombinant binding molecule having at least a portion of the extracellular domain of CTLA4 or a mutant thereof, e.g. an at least extracellular portion of CTLA4 or a mutant thereof joined to a non-CTLA4 protein sequence, e.g. CTLA4Ig (for ex. designated ATCC 68629) or a mutant thereof, e.g. LEA29Y; adhesion molecule inhibitors, e.g. LFA-1 antagonists, ICAM-1 or -3 antagonists, VCAM-4 antagonists or VLA-4 antagonists; or a chemotherapeutic agent, e.g. paclitaxel, gemcitabine, cisplatinum, doxorubicin or 5-fiuorouracil; anti TNF agents, e.g. monoclonal antibodies to TNF, e.g. infliximab, adalimumab, CDP870, or receptor constructs to TNF-RI or TNF-RII, e.g. Etanercept, PEG-TNF-RI; blockers of proinflammatory cytokines, IL-1 blockers, e.g. Anakinra or IL-1 trap, AAL160, ACZ 885, IL-6 blockers; chemokines blockers, e.g. inhibitors or activators of proteases, e.g. metalloproteases, anti-IL-15 antibodies, anti-IL-6 antibodies, anti-IL-23 antibodies, anti-CD20 antibodies, NSAIDs, such as aspirin or an anti-infectious agent (the list not limited to the agent mentioned).
An antibody according to the present invention may be provided in combination or addition with one or more of the following agents: - an antagonist of cytokine function, (e.g. an agent which act on cytokine signaling pathways such as a modulator of the SOCS system) , such as an alpha-, beta-, and/or gamma-interferon; modulators of insulin-like growth factor type I (IGF- I) , its receptors and associated binding proteins; interleukins (IL) e.g. one or more of IL-1 to 33, and/or an interleukin antagonist or inhibitor such as anakinra; inhibitors of receptors of interleukin family members or inhibitors of specific subunits of such receptors; a tumor necrosis factor alpha (TNF-. alpha.) inhibitor such as an anti-TNF monoclonal antibody (for example infliximab; adalimumab, and/or CDP-870) , and/or a TNF receptor antagonist e.g. an immunoglobulin molecule (such as etanercept) and/or a low-molecular-weight agent such as pentoxyfylline;
- a modulator of B cells, e.g. a monoclonal antibody targeting B- lymphocytes (such as CD20 (rituximab) or MRA-aIL16R) or T-lymphocytes (e.g. CTLA4-Ig, HuMax 11-15 or Abatacept);
- a modulator that inhibits osteoclast activity, for example an antibody to RANKL; - a modulator of chemokine or chemokine receptor function such as an antagonist of CCR1, CCR2, CCR2A, CCR2B, CCR3, CCR4, CCR5, CCR6, CCR7, CCR8, CCR9, CCRIO and CCRI1 (for the C-C family); CXCR1, CXCR2, CXCR3, CXCR4 and CXCR5 and CXCR6 (for the C-X-C family) and CX3CR1 for the C-X3-C family; - an inhibitor of matrix metalloproteases (MMPs), i.e., one or more of the stromelysins, the collagenases, and the gelatinases, as well as aggrecanase; especially collagenase-1 (MMP1), collagenase-2 (MMP8), collagenase-3 (MMP13) , stromelysin-1 (MMP3) , stromelysin-2 (MMP10), and/or stromelysin- 3 (MMP11) and/or MMP9 and/or MMP12, e.g. an agent such as doxycycline; a leukotriene biosynthesis inhibitor, 5 -lipoxygenase (5-LO) inhibitor or 5- lipoxygenase activating protein (FLAP) antagonist such as; zileuton; ABT-761; fenleuton; tepoxalin; Abbott-79175; Abbott- 85761; N- (5 -substituted) - thiophene-2-alkylsulfonamides; 2, 6-di-tert- butylphenolhydrazones; methoxytetrahydropyrans such as Zeneca ZD- 2138; the compound SB-210661; a pyridinyl-substituted 2- cyanonaphthalene compound such as L-739,010; a 2- cyanoquinoline compound such as L-746,530; indole and/or a quinoline compound such as MK-591, MK-886, and/or BAY x 1005;
- a receptor antagonist for leukotrienes (LT) B4, LTC4, LTD4, and LTE4, selected from the group consisting of the phenothiazin-3-ls such as L-651,392; amidino compounds such as CGS-25019c; benzoxalamines such as ontazolast; benzenecarboximidamides such as BIIL 284/260; and compounds such as zafirlukast, ablukast, montelukast, pranlukast, verlukast (MK-679) , RG- 12525, Ro-245913, iralukast (CGP 45715A) , and BAY x 7195; a phosphodiesterase (PDE) inhibitor such as a methylxanthanine, e.g. theophylline and/or aminophylline; and/or a selective PDE isoenzyme inhibitor e.g. a PDE4 inhibitor and/or inhibitor of the isoform PDE4D, and/or an inhibitor of PDE5;
- a histamine type 1 receptor antagonist such as cetirizine, loratadine, desloratadine, fexofenadine, acrivastine, terfenadine, astemizole, azelastine, levocabastine, chlorpheniramine, promethazine, cyclizine, and/or mizolastine (generally applied orally, topically or parenterally);
- a proton pump inhibitor (such as omeprazole) or gastroprotective histamine type 2 receptor antagonist;
- an antagonist of the histamine type 4 receptor; an alpha-l/alpha-2 adrenoceptor agonist vasoconstrictor sympathomimetic agent, such as propylhexedrine, phenylephrine, phenylpropanolamine, ephedrine, pseudoephedrine, naphazoline hydrochloride, oxymetazoline hydrochloride, tetrahydrozoline hydrochloride, xylometazoline hydrochloride, tramazoline hydrochloride, and ethylnorepinephrine hydrochloride; an anticholinergic agent, e.g. a muscarinic receptor (Ml, M2, and M3) antagonist such as atropine, hyoscine, glycopyrrrolate, ipratropium bromide, tiotropium bromide, oxitropium bromide, pirenzepine, and telenzepine;
- a beta-adrenoceptor agonist (including beta receptor subtypes 1- 4) such as isoprenaline, salbutamol, formoterol, salmeterol, terbutaline, orciprenaline, bitolterol mesylate, and/or pirbuterol, e.g. a chiral enantiomer thereof;
- a chromone, e.g. sodium cromoglycate and/or nedocromil sodium; - a glucocorticoid, such as flunisolide, triamcinolone acetonide, beclomethasone dipropionate, budesonide, fluticasone propionate, ciclesonide, and/or mometasone furoate; an agent that modulate nuclear hormone receptors such as a PPAR;
- an immunoglobulin (Ig) or Ig preparation or an antagonist or antibody modulating Ig function such as anti-IgE (e.g. omalizumab) ;
- other systemic or topically-applied anti-inflammatory agent, e.g. thalidomide or a derivative thereof, a retinoid, dithranol, and/or calcipotriol;
- combinations of aminosalicylates and sulfapyridine such as sulfasalazine, mesalazine, balsalazide, and olsalazine; and immunomodulatory agents such as the thiopurines, and corticosteroids such as budesonide;
- an antibacterial agent e.g. a penicillin derivative, a tetracycline, a macrolide, a beta-lactam, a fluoroquinolone, metronidazole, and/or an inhaled aminoglycoside; and/or an antiviral agent e.g. acyclovir, famciclovir, valaciclovir, ganciclovir, cidofovir; amantadine, rimantadine; ribavirin; zanamavir and/or oseltamavir; a protease inhibitor such as indinavir, nelfmavir, ritonavir, and/or saquinavir; a nucleoside reverse transcriptase inhibitor such as didanosine, lamivudine, stavudine, zalcitabine, zidovudine; a non-nucleoside reverse transcriptase inhibitor such as nevirapine, efavirenz; a cardiovascular agent such as a calcium channel blocker, beta- adrenoceptor blocker, angiotensin-converting enzyme (ACE) inhibitor, angiotensin-2 receptor antagonist; lipid lowering agent such as a statin, and/or fibrate; a modulator of blood cell morphology such as pentoxyfylline; a thrombolytic, and/or an anticoagulant e.g. a platelet aggregation inhibitor;
- a CNS agent such as an antidepressant (such as sertraline) , anti-Parkinsonian drug (such as deprenyl, L-dopa, ropinirole, pramipexole, MAOB inhibitor such as selegine and rasagiline, comP inhibitor such as tasmar, A-2 inhibitor, dopamine reuptake inhibitor, NMDA antagonist, nicotine agonist, dopamine agonist and/or inhibitor of neuronal nitric oxide synthase), and an anti- Alzheimer's drug such as donepezil, rivastigmine, tacrine, COX-2 inhibitor, propentofylline or metrifonate; an agent for the treatment of acute and chronic pain, e.g. a centrally or peripherally-acting analgesic such as an opioid analogue or derivative, carbamazepine, phenytoin, sodium valproate, amitryptiline or other antidepressant agent, paracetamol, or nonsteroidal anti-inflammatory agent; a parenterally or topically-applied (including inhaled) local anaesthetic agent such as lignocaine or an analogue thereof; an anti-osteoporosis agent e.g. a hormonal agent such as raloxifene, or a biphosphonate such as alendronate; (i) a tryptase inhibitor; (ii) a platelet activating factor (PAF) antagonist; (iii) an interleukin converting enzyme (ICE) inhibitor; (iv) an IMPDH inhibitor; (v) an adhesion molecule inhibitors including VLA-4 antagonist; (vi) a cathepsin; (vii) a kinase inhibitor e.g. an inhibitor of tyrosine kinases (such as Btk, Itk, Jak3 MAP examples of inhibitors might include Gefitinib, Imatinib mesylate), a serine / threonine kinase (e.g. an inhibitor of MAP kinase such as p38, JNK, protein kinases A, B and C and IKK), or a kinase involved in cell cycle regulation (e.g. a cylin dependent kinase) ; (viii) a glucose-6 phosphate dehydrogenase inhibitor; (ix) a kinin-B. subl . - and/or B.sub2. -receptor antagonist; (x) an anti-gout agent, e.g., colchicine; (xi) a xanthine oxidase inhibitor, e.g., allopurinol; (xii) a uricosuric agent, e.g., probenecid, sulfinpyrazone, and/or benzbromarone; (xiii) a growth hormone secretagogue; (xiv) transforming growth factor (TGF.beta.) ;
(xv) platelet-derived growth factor (PDGF) ; (xvi) fibroblast growth factor, e.g., basic fibroblast growth factor (bFGF) ; (xvii) granulocyte macrophage colony stimulating factor (GM-CSF) ; (xviii) capsaicin cream; (xix) a tachykinin NK. subl. and/or NK.sub3. receptor antagonist such NKP-608C, SB-233412 (talnetant) , and/or D-4418; (xx) an elastase inhibitor e.g. UT-77 and/or ZD-
0892; (xxi) a TNF-alpha converting enzyme inhibitor (TACE) ; (xxii) induced nitric oxide synthase (iNOS) inhibitor or (xxiii) a chemoattractant receptor- homologous molecule expressed on TH2 cells, (such as a CRTH2 antagonist) (xxiv) an inhibitor of a P38 (xxv) agent modulating the function of Toll-like receptors (TLR) and (xxvi) an agent modulating the activity of purinergic receptors such as P2X7; (xxvii) an inhibitor of transcription factor activation such as NFkB, API, and/or STATS.
An inhibitor may be specific or may be a mixed inhibitor, e.g. an inhibitor targeting more than one of the molecules (e.g. receptors) or molecular classes mentioned above.
The antibody could also be used in association with a chemotherapeutic agent or another tyrosine kinase inhibitor in coadministration or in the form of an immuno- conjugat. Fragments of said antibody could also be used in bispecific antibodies obtained by recombinant mechanisms or biochemical coupling, and then associating the specificity of the above described antibody with the specificity of other antibodies able to recognize other molecules involved in the activity for which IL-17 is associated. For treatment of an inflammatory disease, an antibody of the invention may be combined with one or more agents such as:- Non-steroidal anti-inflammatory agents (hereinafter NSAIDs) including non-selective cyclo- oxygenase (COX)-I / COX-2 inhibitors whether applied topically or systemically (such as piroxicam, diclofenac, propionic acids such as naproxen, flurbiprofen, fenoprofen, ketoprofen and ibuprofen, fenamates such as mefenamic acid, indomethacin, sulindac, azapropazone, pyrazolones such as phenylbutazone, salicylates such as aspirin); selective COX-2 inhibitors (such as meloxicam, celecoxib, rofecoxib, valdecoxib, lumarocoxib, parecoxib and etoricoxib) ; cyclo- oxygenase inhibiting nitric oxide donors (CINODs) ; glucocorticosteroids (whether administered by topical, oral, intramuscular, intravenous, or intra- articular routes) ; methotrexate, leflunomide; hydroxychloroquine, d- penicillamine, auranofin or other parenteral or oral gold preparations ; analgesics; diacerein; intra-articular therapies such as hyaluronic acid derivatives; and nutritional supplements such as glucosamine.
An antibody of the invention can also be used in combination with an existing therapeutic agent for the treatment of cancer. Suitable agents to be used in combination include: (i) antiproliferative/antineoplastic drugs and combinations thereof, as used in medical oncology, such as alkylating agents (for example cis- platin, carboplatin, cyclophosphamide, nitrogen mustard, melphalan, chlorambucil, busulphan and nitrosoureas); antimetabolites (for example antifolates such as fluoropyrimidines like 5-fluorouracil and tegafur, raltitrexed, methotrexate, cytosine arabinoside, hydroxyurea, gemcitabine and paclitaxel; antitumour antibiotics (for example anthracyclines like adriamycin, bleomycin, doxorubicin, daunomycin, epirubicin, idarubicin, mitomycin-C, dactinomycin and mithramycin); antimitotic agents (for example vinca alkaloids like vincristine, vinblastine, vindesine and vinorelbine and taxoids like taxol and taxotere); and topoisomerase inhibitors (for example epipodophyllotoxins like etoposide and teniposide, amsacrine, topotecan and camptothecins); (ii) cytostatic agents such as antioestrogens (for example tamoxifen, toremifene, raloxifene, droloxifene and iodoxyfene), oestrogen receptor down regulators (for example fulvestrant), antiandrogens
(for example bicalutamide, flutamide, nilutamide and cyproterone acetate), LHRH antagonists or LHRH agonists (for example goserelin, leuprorelin and buserelin), progestogens (for example megestrol acetate), aromatase inhibitors (for example as anastrozole, letrozole, vorazole and exemestane) and inhibitors of 5.alpha.- reductase such as finasteride;
(iii) Agents which inhibit cancer cell invasion (for example metalloproteinase inhibitors like marimastat and inhibitors of urokinase plasminogen activator receptor function); (iv) inhibitors of growth factor function, for example such inhibitors include growth factor antibodies, growth factor receptor antibodies (for example the anti-erbb2 antibody trastuzumab and the anti-erbbl antibody cetuximab [C225] ), farnesyl transferase inhibitors, tyrosine kinase inhibitors and serine/threonine kinase inhibitors, for example inhibitors of the epidermal growth factor family (for example EGFR family tyrosine kinase inhibitors such as N- (3- chloro-4-fluorophenyl) -7-methoxy-6- (3- morpholinopropoxy) quinazolin-4-amine (gefitinib, AZD1839), N- (3- ethynylphenyl) -6, 7-bis (2-methoxyethoxy) quinazolin-4-amine (erlotinib, OSI-774) and 6-acrylamido-N- (3-chloro-4- fluorophenyl) -7- (3- morpholinopropoxy) quinazolin-4-amine (CI 1033)), for example inhibitors of the platelet-derived growth factor family and for example inhibitors of the hepatocyte growth factor family; (v) antiangiogenic agents such as those which inhibit the effects of vascular endothelial growth factor, (for example the anti-vascular endothelial cell growth factor antibody bevacizumab, compounds such as those disclosed in International Patent Applications WO 97/22596, WO 97/30035, WO 97/32856 and WO 98/13354, each of which is incorporated herein in its entirety) and compounds that work by other mechanisms (for example linomide, inhibitors of integrin .alpha.v.beta.3 function and angiostatin) ; (vi) vascular damaging agents such as combretastatin A4 and compounds disclosed in International Patent Applications WO 99/02166, WO 00/40529, WO 00/41669, WO 01/92224, WO 02/04434 and WO 02/08213, each of which is incorporated herein in its entirety; (vii) antisense therapies, for example those which are directed to the targets listed above, such as ISIS 2503, an anti-ras antisense; (viii) gene therapy approaches, including for example approaches to replace aberrant genes such as aberrant p53 or aberrant BRCA1 or BRCA2, GDEPT (gene directed enzyme pro-drug therapy) approaches such as those using cytosine deaminase, thymidine kinase or a bacterial nitroreductase enzyme and approaches to increase patient tolerance to chemotherapy or radiotherapy such as multi-drug resistance gene therapy; and (ix) immunotherapeutic approaches, including for example ex vivo and in vivo approaches to increase the immunogenicity of patient tumor cells, such as transfection with cytokines such as interleukin 2, interleukin 4 or granulocyte macrophage colony stimulating factor, approaches to decrease T cell anergy, approaches using transfected immune cells such as cytokine transfected dendritic cells, approaches using cytokine transfected tumor cell lines and approaches using anti -idiotypic antibodies.
Such active ingredients are suitably present in combination in amounts that are effective for the purpose intended.
Active ingredients may be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsules and poly-(methyl methacrylate) microcapsules, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) or in macroemulsions. Such techniques are disclosed in Remington's Pharmaceutical Sciences, 16th edition, Osol, A. (ed.) (1980).
Sustained-release preparations may be prepared. Suitable examples of sustained- release preparations include semi-permeable matrices of solid hydrophobic polymers containing the antibody, which matrices are in the form of shaped articles, e.g. films, or microcapsules.
The compositions to be used for in vivo administration are generally sterile. Sterility may be readily accomplished, e.g., by filtration through sterile filtration membranes. The <I117> antibodies according to the invention have one or more of the following properties (as determined in Examples 4, 5 and 6): the antibody that specifically binds to human IL17 according to the invention a) shows no cross reactivity with IL17B, IL17C, IL17D, IL17F ( which means that the binding to IL17B, IL17C, IL17D and IL17F is 0% compared to the binding to IL 17A , which is set as 100%); b) inhibits IL17 induced IL8 cytokine stimulation of CCD-25SK cells with an IC50 value of 2.0 nM or lower( e.g. with an IC50 value between 2.0 nM and 0.0 nM); c) inhibits IL17 induced IL6 cytokine stimulation of CCD-25SK cells with an IC50 value of 5.0 nM or lower ( e.g. with an IC50 value between 5.0 nM and 0.0 nM); preferably with an IC50 value of 2.0 nM or lower; d) inhibits IL17 induced IL6 cytokine stimulation of human fibroblast- like synoviocytes-rheumatoid arthritis (HFLS-RA) with an IC50 value of 2.0 nM or lower ( e.g. with an IC50 value between 2.0 nM and 0.0 nM); preferably with an IC50 value of 1.0 nM or lower; and/or e) inhibits IL17 induced IL8 cytokine stimulation of human fibroblast-like synoviocytes-rheumatoid arthritis (HFLS-RA)with an IC50 value of 1.5 nM or lower ( e.g. with an IC50 value between 1.5 nM and 0.0 nM); preferably with an IC50 value of 1.0 nM or lower.
The invention comprises a method for the treatment of a patient in need of therapy, characterized by administering to the patient a therapeutically effective amount of an antibody according to the invention.
The invention comprises the use of an antibody according to the invention for the preparation of a medicament for the treatment of cancer, especially colon, lung, or pancreatic cancer or for the treatment of autoimmune diseases, rheumatoid arthritis, psoratic arthritis, muscle diseases, e.g. muscular dystrophy, multiple sclerosis, chronic kidney diseases, bone diseases, e.g. bone degeneration in multiple myeloma, systemic lupus erythematosus, lupus nephritis, and vascular injury.
The invention comprises the use of an antibody according to the invention for the preparation of a medicament for the treatment of systemic lupus erythematosus, or lupus nephritis.
The invention comprises the use of an antibody according to the invention for the treatment of cancer or inflammatory diseases, preferably for the treatment of colon, lung, or pancreatic cancer or for the treatment of autoimmune diseases, rheumatoid arthritis, psoratic arthritis, muscle diseases, e.g. muscular dystrophy, multiple sclerosis, chronic kidney diseases, bone diseases, e.g. bone degeneration in multiple myeloma, systemic lupus erythematosus, lupus nephritis, and vascular injury.
The invention comprises the use of an antibody according to the invention for the treatment of cancer or inflammatory diseases, preferably for the treatment of systemic lupus erythematosus, or lupus nephritis. The invention comprises the use of the antibodies specifically binding to human IL17 according to the invention for the treatment (or the antibodies for use in the treatment) of a patient suffering from cancer, especially from colon, lung, or pancreatic cancer or from autoimmune diseases, rheumatoid arthritis, psoratic arthritis, psoriasis, muscle diseases, e.g. muscular dystrophy, multiple sclerosis, chronic kidney diseases, bone diseases, e.g. bone degeneration in multiple myeloma, systemic lupus erythematosus, lupus nephritis, and vascular injury. The invention comprises the use of the antibodies specifically binding to and human IL17 according to the invention for the treatment (or the antibodies for use in the treatment) of a patient suffering from systemic lupus erythematosus or lupus nephritis.
The invention comprises the use of the antibodies specifically binding to and human IL17 according to the invention for the treatment (or the antibodies for use in the treatment) of a variety of inflammatory, immune and proliferative disorders, including rheumatoid arthritis (RA), osteoarthritis, rheumatoid arthritis osteoporosis, inflammatory fibrosis (e.g. scleroderma, lung fibrosis, and cirrhosis), gingivitis, periodontitis or other inflammatory periodontal diseases, inflammatory bowel disorders (e.g. Crohn's disease, ulcerative colitis and inflammatory bowel disease), asthma (including allergic asthma), allergies, chronic obstructive pulmonary disease (COPD), multiple sclerosis, psoriasis and cancer, ankylosing spondylitis, systemic scleroris, psioriatic arthritis, inflammatory arthritis, osteoarthritis, inflammatory joint disease, autoimmune disease including autoimmune vasculitis, multiple sclerosis, lupus, diabetes (e.g., insulin diabetes), inflammatory bowel disease, transplant rejection, graft vs. host disease, and inflammatory conditions resulting from strain, sprain, cartilage damage, trauma, orthopedic surgery, infection or other disease processes. Other diseases influenced by the dysfunction of the immune system are encompassed within the scope of the invention, including but not limited to, allergies.
Anti-Ill 7 antibodies of the invention are particularly useful for the treatment, prevention, or amelioration of autoimmune disease and of inflammatory conditions, in particular inflammatory conditions with an aetiology including an autoimmune component such as arthritis (for example rheumatoid arthritis, arthritis chronica progrediente and arthritis deformans) and rheumatic diseases, including inflammatory conditions and rheumatic diseases involving bone loss, inflammatory pain, spondyloarhropathies including ankolysing spondylitis, Reiter syndrome, reactive arthritis, psoriatic arthritis, and enterophathis arthritis, hypersensitivity (including both airways hypersensitivity and dermal hypersensitivity) and allergies. Specific auto-immune diseases for which the antibodies as described herein may be employed include autoimmune haematological disorders (including e.g. hemolytic anaemia, aplastic anaemia, pure red cell anaemia and idiopathic thrombocytopenia), systemic lupus erythematosus, inflammatory muscle disorders, polychondritis, sclerodoma, Wegener granulomatosis, dermatomyositis, chronic active hepatitis, myasthenia gravis, psoriasis, Steven- Johnson syndrome, idiopathic sprue, autoimmune inflammatory bowel disease (including e.g. ulcerative colitis, Crohn's disease and Irritable Bowel Syndrome), endocrine ophthalmopathy, Graves' disease, sarcoidosis, multiple sclerosis, primary biliary cirrhosis, juvenile diabetes (diabetes mellitus type I), uveitis (anterior and posterior), keratoconjunctivitis sicca and vernal keratoconjunctivitis, interstitial lung fibrosis, psoriatic arthritis and glomerulonephritis (with and without nephrotic syndrome, e.g. including idiopathic nephrotic syndrome or minimal change nephropathy), tumors, multiple sclerosis, inflammatory disease of skin and cornea, myositis, loosening of bone implants, metabolic disorders, such as atherosclerosis, diabetes, and dislipidemia. The antibodies according to the invention are also useful for the treatment, prevention, or amelioration of asthma, bronchitis, pneumoconiosis, pulmonary emphysema, and other obstructive or inflammatory diseases of the airways.
The invention comprises also a method for the treatment of a patient suffering from such disease. The invention further provides a method for the manufacture of a pharmaceutical composition comprising an effective amount of an antibody according to the invention together with a pharmaceutically acceptable carrier and the use of the antibody according to the invention for such a method.
The invention also provides the use of an antibody according to the invention in an effective amount for the manufacture of a pharmaceutical agent, preferably together with a pharmaceutically acceptable carrier, for the treatment of a patient suffering from cancer, especially from colon, lung, or pancreatic cancer or from autoimmune diseases, rheumatoid arthritis, psoratic arthritis, muscle diseases, e.g. muscular dystrophy, multiple sclerosis, chronic kidney diseases, bone diseases, e.g. bone degeneration in multiple myeloma, systemic lupus erythematosus, lupus nephritis, and vascular injury.
The invention also provides the use of an antibody according to the invention in an effective amount for the manufacture of a pharmaceutical agent, preferably together with a pharmaceutically acceptable carrier, for the treatment of a patient suffering from of autoimmune disease and of inflammatory conditions, in particular inflammatory conditions with an aetiology including an autoimmune component such as arthritis (for example rheumatoid arthritis, arthritis chronica progrediente and arthritis deformans) and rheumatic diseases, including inflammatory conditions and rheumatic diseases involving bone loss, inflammatory pain, spondyloarhropathies including ankolsing spondylitis, Reiter syndrome, reactive arthritis, psoriatic arthritis, and enterophathis arthritis, hypersensitivity (including both airways hypersensitivity and dermal hypersensitivity) and allergies. Specific auto-immune diseases for which the antibodies as described herein may be employed include autoimmune haemato logical disorders (including e.g. hemolytic anaemia, aplastic anaemia, pure red cell anaemia and idiopathic thrombocytopenia), systemic lupus erythematosus, inflammatory muscle disorders, polychondritis, sclerodoma, Wegener granulomatosis, dermatomyositis, chronic active hepatitis, myasthenia gravis, psoriasis, Steven- Johnson syndrome, idiopathic sprue, autoimmune inflammatory bowel disease (including e.g. ulcerative colitis,
Crohn's disease and Irritable Bowel Syndrome), endocrine ophthalmopathy, Graves' disease, sarcoidosis, multiple sclerosis, primary biliary cirrhosis, juvenile diabetes (diabetes mellitus type I), uveitis (anterior and posterior), keratoconjunctivitis sicca and vernal keratoconjunctivitis, interstitial lung fibrosis, psoriatic arthritis and glomerulonephritis (with and without nephrotic syndrome, e.g. including idiopathic nephrotic syndrome or minimal change nephropathy), tumors, multiple sclerosis, inflammatory disease of skin and cornea, myositis, loosening of bone implants, metabolic disorders, such as atherosclerosis, diabetes, and dislipidemia. The antibodies according to the invention are also useful for the treatment, prevention, or amelioration of asthma, bronchitis, pneumoconiosis, pulmonary emphysema, and other obstructive or inflammatory diseases of the airways.
In another aspect, the present invention provides a composition, e.g. a pharmaceutical composition, containing an IL17 antibody as described herein, formulated together with a pharmaceutically acceptable carrier, e.g., for use in any of the above therapeutic methods.
As used herein, "pharmaceutically acceptable carrier" includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption/resorption delaying agents, and the like that are physiologically compatible. Preferably, the carrier is suitable for injection or infusion. A composition of the present invention can be administered by a variety of methods known in the art. As will be appreciated by the skilled artisan, the route and/or mode of administration will vary depending upon the desired results.
Pharmaceutically acceptable carriers include sterile aqueous solutions or dispersions and sterile powders for the preparation of sterile injectable solutions or dispersion. The use of such media and agents for pharmaceutically active substances is known in the art. In addition to water, the carrier can be, for example, an isotonic buffered saline solution.
In another embodiment, a pharmaceutical formulation comprises any of the IL17 antibodies provided herein and at least one additional therapeutic agent, e.g., as described below.
Antibodies of the invention can be used either alone or in combination with other agents in a therapy. For instance, an antibody of the invention may be coadministered with at least one additional therapeutic agent. In certain embodiments, an additional therapeutic agent is a. immunosuppressive or immunomodulating agents or other anti-inflammatory agents. For example, the antibodies as described herein may be used in combination with DMARD, e.g. Gold salts, sulphasalazine, antimalarias, methotrexate, D-penicillamine, azathioprine, mycophenolic acid, cyclosporine A, tacrolimus, sirolimus, minocycline, leflunomide, glococorticoids; a calcineurin inhibitor, e.g. cyclosporin A or FK 506; a modulator of lymphocyte recirculation, e.g. FTY720 and FTY720 analogs; a mTOR inhibitor, e.g. rapamycin, 40-O-(2-hydroxyethyl)-rapamycin, CCI779, ABT578, AP23573 or TAFA-93; an ascomycin having immuno-suppressive properties, e.g. ABT-281, ASM981, etc.; corticosteroids; cyclophosphamide; azathioprene; methotrexate; leflunomide; mizoribine; mycophenolic acid; myc-phenol-late mofetil; 15-deoxyspergualine or an immunosuppressive homologue, analogue or derivative thereof; immunosuppressive monoclonal antibodies, e.g., monoclonal antibodies to leukocyte receptors, e.g., MHC, CD2, CD3, CD4, CD7, CD8, CD25, CD28, CD40. CD45, CD58, CD80, CD86 or their ligands; other immunomodulatory compounds, e.g. a recombinant binding molecule having at least a portion of the extracellular domain of CTLA4 or a mutant thereof, e.g. an at least extracellular portion of CTLA4 or a mutant thereof joined to a non-CTLA4 protein sequence, e.g. CTLA4Ig (for ex. designated ATCC 68629) or a mutant thereof, e.g. LEA29Y; adhesion molecule inhibitors, e.g. LFA-1 antagonists, ICAM-1 or -3 antagonists, VCAM-4 antagonists or VLA-4 antagonists; or a chemotherapeutic agent, e.g. paclitaxel, gemcitabine, cisplatinum, doxorubicin or 5-fluorouracil; anti TNF agents, e.g. monoclonal antibodies to TNF, e.g. infliximab, adalimumab, CDP870, or receptor constructs to TNF-RI or TNF-RII, e.g. Etanercept, PEG-TNF-RI; blockers of proinflammatory cytokines, IL-1 blockers, e.g. Anakinra or IL-1 trap, AAL160, ACZ 885, IL-6 blockers; chemokines blockers, e.g. inhibitors or activators of proteases, e.g. metalloproteases, anti-IL-15 antibodies, anti-IL-6 antibodies, anti-IL-23 antibodies, anti-CD20 antibodies, NSAIDs, such as aspirin or an anti-infectious agent (the list not limited to the agent mentioned).
Such combination therapies noted above encompass combined administration (where two or more therapeutic agents are included in the same or separate formulations), and separate administration, in which case, administration of the antibody of the invention can occur prior to, simultaneously, and/or following, administration of the additional therapeutic agent and/or adjuvant.
An antibody of the invention (and any additional therapeutic agent) can be administered by any suitable means, including parenteral, intrapulmonary, and intranasal, and, if desired for local treatment, intralesional administration. Parenteral infusions include intramuscular, intravenous, intraarterial, intraperitoneal, or subcutaneous administration. Dosing can be by any suitable route, e.g. by injections, such as intravenous or subcutaneous injections, depending in part on whether the administration is brief or chronic. Various dosing schedules including but not limited to single or multiple administrations over various time- points, bolus administration, and pulse infusion are contemplated herein.
Antibodies of the invention would be formulated, dosed, and administered in a fashion consistent with good medical practice. Factors for consideration in this context include the particular disorder being treated, the particular mammal being treated, the clinical condition of the individual patient, the cause of the disorder, the site of delivery of the agent, the method of administration, the scheduling of administration, and other factors known to medical practitioners. The antibody need not be, but is optionally formulated with one or more agents currently used to prevent or treat the disorder in question. The effective amount of such other agents depends on the amount of antibody present in the formulation, the type of disorder or treatment, and other factors discussed above. These are generally used in the same dosages and with administration routes as described herein, or about from 1 to 99% of the dosages described herein, or in any dosage and by any route that is empirically/clinically determined to be appropriate. For the prevention or treatment of disease, the appropriate dosage of an antibody of the invention (when used alone or in combination with one or more other additional therapeutic agents) will depend on the type of disease to be treated, the type of antibody, the severity and course of the disease, whether the antibody is administered for preventive or therapeutic purposes, previous therapy, the patient's clinical history and response to the antibody, and the discretion of the attending physician. The antibody is suitably administered to the patient at one time or over a series of treatments. Depending on the type and severity of the disease, about 1 μg/kg to 15 mg/kg (e.g. 0.5mg/kg - 10 mg/kg) of antibody can be an initial candidate dosage for administration to the patient, whether, for example, by one or more separate administrations, or by continuous infusion. One typical daily dosage might range from about 1 μg/kg to 100 mg/kg or more, depending on the factors mentioned above. For repeated administrations over several days or longer, depending on the condition, the treatment would generally be sustained until a desired suppression of disease symptoms occurs. One exemplary dosage of the antibody would be in the range from about 0.05 mg/kg to about 10 mg/kg. Thus, one or more doses of about 0.5 mg/kg, 2.0 mg/kg, 4.0 mg/kg or 10 mg/kg (or any combination thereof) may be administered to the patient. Such doses may be administered intermittently, e.g. every week or every three weeks (e.g. such that the patient receives from about two to about twenty, or e.g. about six doses of the antibody). An initial higher loading dose, followed by one or more lower doses may be administered. An exemplary dosing regimen comprises administering. However, other dosage regimens may be useful. The progress of this therapy is easily monitored by conventional techniques and assays. Regardless of the route of administration selected, the compounds of the present invention, which may be used in a suitable hydrated form, and/or the pharmaceutical compositions of the present invention, are formulated into pharmaceutically acceptable dosage forms by conventional methods known to those of skill in the art. Actual dosage levels of the active ingredients in the pharmaceutical compositions of the present invention may be varied so as to obtain an amount of the active ingredient which is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient (effective amount). The selected dosage level will depend upon a variety of pharmacokinetic factors including the activity of the particular compositions of the present invention employed, or the ester, salt or amide thereof, the route of administration, the time of administration, the rate of excretion of the particular compound being employed, other drugs, compounds and/or materials used in combination with the particular compositions employed, the age, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors well known in the medical arts.
Articles of Manufacture
In another aspect of the invention, an article of manufacture containing materials useful for the treatment, prevention and/or diagnosis of the disorders described above is provided. The article of manufacture comprises a container and a label or package insert on or associated with the container. Suitable containers include, for example, bottles, vials, syringes, IV solution bags, etc. The containers may be formed from a variety of materials such as glass or plastic. The container holds a composition which is by itself or combined with another composition effective for treating, preventing and/or diagnosing the condition and may have a sterile access port (for example the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle). At least one active agent in the composition is an antibody of the invention. The label or package insert indicates that the composition is used for treating the condition of choice. Moreover, the article of manufacture may comprise (a) a first container with a composition contained therein, wherein the composition comprises an antibody of the invention; and (b) a second container with a composition contained therein, wherein the composition comprises a further cytotoxic or otherwise therapeutic agent. The article of manufacture in this embodiment of the invention may further comprise a package insert indicating that the compositions can be used to treat a particular condition. Alternatively, or additionally, the article of manufacture may further comprise a second (or third) container comprising a pharmaceutically- acceptable buffer, such as bacteriostatic water for injection (BWFI), phosphate- buffered saline, Ringer's solution and dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, and syringes. Description of the Sequences
SEQ ID NO: 1 CDR1H <IL17> 9C6-2B6
SEQ ID NO: 2 CDR2H <IL17> 9C6-2B6
SEQ ID NO: 3 CDR3H <IL17> 9C6-2B6
SEQ ID NO: 4 CDR1L <IL17> 9C6-2B6
SEQ ID NO: 5 CDR2L <IL17> 9C6-2B6
SEQ ID NO: 6 CDR3L <IL17> 9C6-2B6
SEQ ID NO: 7 Mouse variable heavy chain domain (VH), <IL17> 9C6- 2B6
SEQ ID NO: 8 Mouse variable light chain domain (VL) <IL17> 9C6-2B6
SEQ ID NO: 9 Humanized variant of VH, <IL17> 9C6-2B6-HC134
SEQ ID NO: 10 Humanized variant of VH, <IL17> 9C6-2B6-HC136
SEQ ID NO: 11 Humanized variant of VL, <IL17> 9C6-2B6-LC134
SEQ ID NO: 12 Humanized variant of VL, <IL17> 9C6-2B6-LC136
SEQ ID NO: 13 Human kappa light chain constant region
SEQ ID NO: 14 Human lambda light chain constant region lambda
SEQ ID NO: 15 Human IgGl (Caucasian Allotype) constant region
SEQ ID NO: 16 Human IgGl (Afroamerican Allotype) constant region
SEQ ID NO: 17 Human IgGl L234A/L235A Mutant (Caucasian Allotype)
SEQ ID NO: 18 Human IgGl L234A/L235A/P329G Mutant (Caucasian
Allotype)
SEQ ID NO: 19 Human IgG4 constant region SEQ ID NO: 20 Human IgG4 S228P/L235E Mutant
SEQ ID NO: 21 Human IgG4 S228P/L235E/P329G Mutant
SEQ ID NO: 22 Human IL17 (IL17A)
SEQ ID NO: 23 Human IL17B
SEQ ID NO: 24 Human IL17C
SEQ ID NO: 25 Human IL17D
SEQ ID NO: 26 Human IL17E
SEQ ID NO: 27 Human IL17F
SEQ ID NO 28 Human IL6
SEQ ID NO 29 Human IL8
In the following embodiments of the invention are listed:
An antibody that specifically binds to human IL17, characterized in comprising a CDRIH of SEQ ID NO: l, CDR2H of SEQ ID NO:2, CDR3H of SEQ ID NO:3, and CDRIL of SEQ ID NO:4, CDR2L of SEQ ID NO:5, CDR3L of SEQ ID NO:6.
A chimeric or humanized variant of the antibody according to embodiment 1.
The antibody according to any of embodiments 1 to 2, characterized in comprising a) a variable heavy chain domain (VH) of SEQ ID NO:9, or of SEQ ID NO: 10, and b) a variable light chain domain (VL) of SEQ ID NO: 11, or of SEQ ID NO: 12. 4. The antibody according to any of embodiments 1 to 2, characterized in comprising a) a variable heavy chain domain (VH) of SEQ ID NO: 10, and b) a variable light chain domain (VL) of SEQ ID NO: 12. 5. The antibody according to any of embodiments 1 to 2, characterized in comprising a) a variable heavy chain domain (VH) of SEQ ID NO:9, and b) a variable light chain domain (VL) of SEQ ID NO: 11.
6. The antibody according to any of the preceding embodiments, characterized in being of IgGl or IgG4 subclass.
7. The antibody according to any of the preceding embodiments, characterized in being of IgGl subclass with the mutations L234A and L235A (numbering according to the EU index of Kabat).
8. The antibody according to any of the preceding embodiments, characterized in being of IgGl subclass with the mutations L234A, L235A and P329G
(numbering according to the EU index of Kabat).
9. The antibody according to any of the preceding embodiments, characterized in being of IgG4 subclass with the mutations S228P and L235E (numbering according to the EU index of Kabat). 10. The antibody according to any of the preceding embodiments, characterized in being of IgG4 subclass with the mutations S228P, L235E and P329G (numbering according to the EU index of Kabat).
11. Pharmaceutical composition characterized by comprising an antibody according to embodiments 1 to 10. 12. Use of an antibody according to embodiments 1 to 10 for the manufacture of a pharmaceutical composition.
13. An antibody according to embodiments 1 to 10 for use in the treatment of cancer, or inflammatory diseases, autoimmune diseases, rheumatoid arthritis, psoratic arthritis, muscle diseases, e.g. muscular dystrophy, multiple sclerosis, chronic kidney diseases, bone diseases, e.g. bone degeneration in multiple myeloma, systemic lupus erythematosus, lupus nephritis, and vascular injury.
14. Use of an antibody according to embodiments 1 to 10 for manufacture of a medicament for the treatment of cancer, or inflammatory diseases, autoimmune diseases, rheumatoid arthritis, psoratic arthritis, muscle diseases, e.g. muscular dystrophy, multiple sclerosis, chronic kidney diseases, bone diseases, e.g. bone degeneration in multiple myeloma, systemic lupus erythematosus, lupus nephritis, and vascular injury.
15. Nucleic acid encoding an antibody according to embodiments 1 to 10.
16. Expression vectors characterized by comprising a nucleic acid according to embodiment 15 for the expression of the antibody according to embodiments 1 to 10 in a prokaryotic or eukaryotic host cell.
17. Prokaryotic or eukaryotic host cell comprising a vector according to embodiment 16.
18. Method for the production of a recombinant antibody according to embodiments 1 to 10, characterized by expressing a nucleic acid according to embodiment 15 in a prokaryotic or eukaryotic host cell and recovering said antibody from said cell or the cell culture supernatant.
19. Method for the treatment of a patient suffering from cancer or from inflammatory diseases, autoimmune diseases, rheumatoid arthritis, psoratic arthritis, muscle diseases, e.g. muscular dystrophy, multiple sclerosis, chronic kidney diseases, bone diseases, e.g. bone degeneration in multiple myeloma, systemic lupus erythematosus, lupus nephritis, and vascular injury, characterized by administering to the patient an antibody according to embodiments 1 to 10.
The following examples and sequence listing are provided to aid the understanding of the present invention, the true scope of which is set forth in the appended claims. It is understood that modifications can be made in the procedures set forth without departing from the spirit of the invention. Examples
Materials & general methods
General information regarding the nucleotide sequences of human immunoglobulins light and heavy chains is given in: Kabat, E.A., et al, Sequences of Proteins of Immunological Interest, 5th ed., Public Health Service, National Institutes of Health, Bethesda, MD (1991). Amino acids of antibody constant chains are numbered and referred to according to EU index according to Kabat (Kabat, E.A., et al, Sequences of Proteins of Immunological Interest, 5th ed., Public Health Service, National Institutes of Health, Bethesda, MD, (1991)).
Recombinant DNA techniques
Standard methods were used to manipulate DNA as described in Sambrook, J., et al., Molecular Cloning: A Laboratory Manual; Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York (1989). The molecular biological reagents were used according to the manufacturer's instructions.
Gene synthesis
Desired gene segments can be prepared from oligonucleotides made by chemical synthesis. The gene segments, which are flanked by singular restriction endonuclease cleavage sites, were assembled by annealing and ligation of oligonucleotides including PCR amplification and subsequently cloned via the indicated restriction sites e.g. Kpnl/ Sacl or Ascl/Pacl into a pPCRScript (Stratagene) based pGA4 cloning vector. The DNA sequences of the subcloned gene fragments were confirmed by DNA sequencing.
Gene synthesis fragments were ordered according to given specifications at Geneart (Regensburg, Germany). All gene segments encoding light and heavy chains of IL-17 antibodies were synthesized with a 5 '-end DNA sequence coding for a leader peptide which targets proteins for secretion in eukaryotic cells, and unique restriction sites at the 5' and 3' ends of the synthesized gene.
DNA sequence determination
DNA sequences were determined by double strand sequencing performed at
MediGenomix GmbH (Martinsried, Germany) or Sequiserve GmbH (Vaterstetten, Germany). DNA and protein sequence analysis and sequence data management
The GCG's (Genetics Computer Group, Madison, Wisconsin) software package version 10.2 and Infomax's Vector NTl Advance suite version 11.5 was used for sequence creation, mapping, analysis, annotation and illustration. Expression vectors
For the expression of the described antibodies variants of expression plasmids for transient expression (e.g. in HEK293 EBNA or HEK293-F cells) or for stable expression (e.g. in CHO cells) based either on a cDNA organization with a CMV-Intron A promoter or on a genomic organization with a CMV promoter were applied.
In case of IgG4_SPLE the intron between the CHI domain and the hinge domain was removed, keeping the remainder of the antibody gene in a genomic organization. The intron-deleted version of IgG4_SPLE no longer shows hingeless antibodies as a result of a splice artefact commonly seen in IgG4_SPLE encoded in total genomic organization.
Beside the antibody expression cassette the vectors contained: an origin of replication which allows replication of this plasmid in E. coli, and
a β-lactamase gene which confers ampicillin resistance in E. coli.
The transcription unit of the antibody gene is composed of the following elements:
- unique restriction site(s) at the 5 ' end
the immediate early enhancer and promoter from the human
cytomegalovirus,
followed by the Intron A sequence in the case of the cDNA organization, - a 5 '-untranslated region of a human antibody gene,
an immunoglobulin heavy chain signal sequence,
the human antibody chain (heavy chain, modified heavy chain or light chain) either as cDNA or as genomic organization with an the
immunoglobulin exon-intron organization
- a 3 ' untranslated region with a polyadenylation signal sequence, and
unique restriction site(s) at the 3 ' end. For transient and stable transfections larger quantities of the plasmids were prepared by plasmid preparation from transformed E. coli cultures (Nucleobond AX, Macherey-Nagel). Cell culture techniques
Standard cell culture techniques were used as described in Current Protocols in Cell Biology (2000), Bonifacino, J.S., Dasso, M., Harford, J.B., Lippincott- Schwartz, J. and Yamada, K.M. (eds.), John Wiley & Sons, Inc.. Transient transfections in HEK293 system
Recombinant immunoglobulin variants were expressed by transient transfection of human embryonic kidney 293 cells using the FreeStyle™ 293 Expression System according to the manufacturer's instruction (Invitrogen, USA). Briefly, suspension HEK293 cells were cultivated in FreeStyle™ 293 Expression medium at 37°C/8 % C02 and the cells were seeded in fresh medium at a density of l-2xl06 viable cells/ml on the day of transfection. DNA-293fectin™ complexes were prepared in Opti-MEM® I medium (Invitrogen, USA) using 325 μΐ of HEK293 and 250 μg of heavy and light chain plasmid DNA in a 1 : 1 molar ratio for a 250 ml final transfection volume for monospecific antibodies. "For expression yield and product quality optimization the ratio can be varied. DNA-293fectin complexes were prepared in Opti-MEM® I medium (Invitrogen, USA) using 325 μΐ of 293fectin™ (Invitrogen, Germany) and 250 μg of "Knobs-into-hole" heavy chain 1 and 2 and light chain 1 and 2 plasmid DNA in a 1 : 1 : 1 : 1 molar ratio for a 250 ml final transfection volume. Antibody containing cell culture supematants were harvested 7 days after transfection by centrifugation at 14000 g for 30 minutes and filtered through a sterile filter (0.22 μιη). Supematants were stored at -20° C until purification.
Protein determination
The protein concentration of purified antibodies and derivatives was determined by determining the optical density (OD) at 280 nm, using the molar extinction coefficient calculated on the basis of the amino acid sequence according to Pace, C.N., et. al, Protein Science 4 (1995) 2411-1423.
Antibody concentration determination in supematants
The concentration of antibodies and derivatives in cell culture supematants was estimated by immunoprecipitation with Protein A Agarose-beads (Roche). 60 μΐ^
Protein A Agarose beads are washed three times in TBS-NP40 (50 mM Tris, pH 7.5, 150 mM NaCl, 1% Nonidet-P40). Subsequently, 1 -15 mL cell culture supernatant are applied to the Protein A Agarose beads pre-equilibrated in TBS-NP40. After incubation for at 1 h at room temperature the beads are washed on an Ultrafree-MC-filter column (Amicon] once with 0.5 mL TBS-NP40, twice with 0.5 mL 2x phosphate buffered saline (2xPBS, Roche) and briefly four times with 0.5 mL 100 mM Na-citrate pH 5,0. Bound antibody is eluted by addition of 35 μΐ NuPAGE® LDS Sample Buffer (Invitrogen). Half of the sample is combined with NuPAGE® Sample Reducing Agent or left unreduced, respectively, and heated for 10 min at 70°C. Consequently, 20 μΐ are applied to an 4-12%
NuPAGE® Bis-Tris SDS-PAGE (Invitrogen) (with MOPS buffer for non-reduced SDS-PAGE and MES buffer with NuPAGE® Antioxidant running buffer additive (Invitrogen) for reduced SDS-PAGE) and stained with Coomassie Blue.
The concentration of antibodies and derivatives in cell culture supematants was measured by Protein A-HPLC chromatography. Briefly, cell culture supematants containing antibodies and derivatives that bind to Protein A were applied to a HiTrap Protein A column (GE Healthcare) in 50 mM K2HP04, 300 mM NaCl, pH 7.3 and eluted from the matrix with 550 mM acetic acid, pH 2.5 on a Dionex HPLC-System. The eluted protein was quantified by UV absorbance and integration of peak areas. A purified standard IgGl antibody served as a standard.
Alternatively, the concentration of antibodies and derivatives in cell culture supematants was measured by Sandwich-IgG-ELISA. Briefly, StreptaWell High Bind Strepatavidin A-96 well microtiter plates (Roche) were coated with 100 μΕΛνεΙΙ biotinylated anti-human IgG capture molecule F(ab')2<h-Fcgamma> BI (Dianova) at 0.1 μg/mL for 1 h at room temperature or alternatively over night at
4°C and subsequently washed three times with 200 μΕΛνεΙΙ PBS, 0.05% Tween (PBST, Sigma). 100 μΕΛνεΙΙ of a dilution series in PBS (Sigma) of the respective antibody containing cell culture supematants was added to the wells and incubated for 1-2 h on a microtiterplate shaker at room temperature. The wells were washed three times with 200 μΕΛνεΙΙ PBST and bound antibody was detected with 100 μΐ
F(ab')2<hFcgamma>POD (Dianova) at 0.1 μg/mL as detection antibody for 1-2 h on a microtiterplate shaker at room temperature. Unbound detection antibody was washed away three times with 200 μΕΛνεΙΙ PBST and the bound detection antibody was detected by addition of 100 μΐ^ ABTS/well. Determination of absorbance was performed on a Tecan Fluor Spectrometer at a measurement wavelength of 405 nm (reference wavelength 492 nm).
Purification of antibodies
Antibodies were purified from cell culture supematants by affinity chromatography using Protein A-Sepharose™ (GE Healthcare, Sweden) and Superdex200 size exclusion chromatography. Briefly, sterile filtered cell culture supematants were ap lied on a HiTrap ProteinA HP (5 ml) column equilibrated with PBS buffer (10 mM Na2HP04, 1 mM KH2P04, 137 mM NaCl and 2.7 mM KC1, pH 7.4). Unbound proteins were washed out with equilibration buffer. Antibody and antibody variants were eluted with 0.1 M citrate buffer, pH 2.8, and the protein containing fractions were neutralized with 0.1 ml 1 M Tris, pH 8.5. Then, the eluted protein fractions were pooled, concentrated with an Amicon Ultra centrifugal filter device (MWCO: 30 K, Millipore) to a volume of 3 ml and loaded on a Superdex200 HiLoad 120 ml 16/60 gel filtration column (GE Healthcare, Sweden) equilibrated with 20mM Histidin, 140 mM NaCl, pH 6.0. Fractions containing purified antibodies with less than 5 % high molecular weight aggregates were pooled and stored as 1.0 mg/ml aliquots at -80°C.
SDS-PAGE
The NuPAGE® Pre-Cast gel system (Invitrogen) was used according to the manufacturer's instruction. In particular, 4-20 % NuPAGE® No vex® TRIS-Glycine Pre-Cast gels and a Novex® TRIS-Glycine SDS running buffer were used. Reducing of samples was achieved by adding NuPAGE® sample reducing agent prior to running the gel.
Analytical size exclusion chromatography
Size exclusion chromatography for the determination of the aggregation and oligomeric state of antibodies was performed by HPLC chromatography. Briefly,
Protein A purified antibodies were applied to a Tosoh TSKgel G3000SW column in 300 mM NaCl, 50 mM KH2P04/K2HP04, pH 7.5 on an Agilent HPLC 1100 system or to a Superdex 200 column (GE Healthcare) in 2 x PBS on a Dionex HPLC-System. The eluted protein was quantified by UV absorbance and integration of peak areas. BioRad Gel Filtration Standard 151-1901 served as a standard.
Mass spectrometry
The total deglycosylated mass of the antibodies was determined and confirmed via electrospray ionization mass spectrometry (ESI-MS). Moreover potential sideproducts mispairing were detected and relatively quantified. Briefly, 100 μg purified antibodies at a protein concentration of up to 3 mg/ml were deglycosylated with 14 or 28 U N-Glycosidase F (Roche) in 100 mM NaH2P04/Na2HP04, pH 7 at 37 or 45°C for 16 or 2 h and subsequently desalted via HPLC on a Sephadex G25 column (GE Healthcare). The mass of the respective heavy and light chains was determined by ESI-MS after deglycosylation and reduction. In brief, 50 μg antibody in 115 μΐ were incubated at 37°C for 30 min with 60 μΐ 0.5 M TCEP in 4 M Guanidine-hydrochloride and 50 μΐ 8 M Guanidine -hydrochloride and subsequently desalted. The total mass and the mass of the reduced heavy and light chains were determined via ESI-MS on a maXis UHR-TOF (Bruker) MS system equipped with a TriVersa NanoMate (Advion) source.
Example 1
Generation of IL17 antibodies
Immunization was performed within 20 weeks using 5 female Balb/c mice using 250 (lx) and 100 μg (3x) recombinant human IL17 from Peprotech (http://www.peprotech.com; Cat.No.: 200-17 in 1% PBS with 1% Albumin) per mouse. Hybridoma generation. The mouse lymphocytes were isolated and fused with a mouse myeloma cell line using PEG based standard protocols to generate hybridomas. The resulting hybridomas were then screened for the production of antigen- specific antibodies. From resulting hybridomas mouse clone <IL17> 9C6- 2B6 was selected using the binding to IL-17 subtypes measured by ELISA (see
Example 4) and a cytokine release assay (via the inhibition of IL-17A induced hIL-6 and hIL-8 release (see Example 5). Humanization of mouse clone <IL17> 9C6-2B6 resulted in the humanized variants <IL17> 9C6-2B6-134 (with the humanized variant of VH, <IL17> 9C6-2B6-HC134 and humanized variant of VL, <IL17> 9C6-2B6-LC134 of SEQ ID No. 9 and 11) and <IL17> 9C6-2B6 (with the humanized variant of VH, <IL17> 9C6-2B6-HC136 and humanized variant of VL, <IL17> 9C6-2B6-LC136 of SEQ ID No. 10 and 12).
Example 2
Expression and purification of < IL-17> antibodies Light and heavy chains of the antibodies were constructed in genomic, partly genomic or cDNA-derived expression vectors as described. The plasmids were amplified in E. coli, purified, and subsequently HEK293 cells were transfected for transient expression of recombinant proteins. After 7 days of cultivation, the supematants of HEK293 cells were harvested, filtered and the antibodies were purified.
Antibodies were purified from cell culture supematants by affinity chromatography using MabSelect SuRe™ (GE Healthcare, Sweden). The subsequent chromatographic steps (size exclusion chromatography (Superdex200 HiLoad 120 ml 16/60 gel filtration column, GE Healthcare, Sweden) or ion exchange chromatography (MacroPrep CHT Typell 10 ml, Bio-Rad plus size exclusion chromatography) was chosen in respect to the individual product related sideproducts of the antibodies after MabSelect SuRe™ chromatography.
Briefly, sterile filtered cell culture supematants were captured on a MabSelect SuRe resin equilibrated with PBS buffer (10 mM Na2HP04, 1 mM KH2P04,
137 mM NaCl and 2.7 mM KC1, pH 7.4), washed with equilibration buffer and eluted with 25 mM sodium citrate at pH 3.0. The eluted protein fractions were pooled and neutralized with 2M Tris, pH 9.0. The antibody pools were prepared for hydrophobic interaction chromatography by rebuffering in 10 mM NaH2P04, 20 mM MES, 50 mM NaCl, 0.1 mM CaCl2, pH 7.5. After equilibration of the CHT column with equilibration buffer (10 mM NaH2P04, 20 mM MES, 50 mM NaCl, 0.1 mM CaCl2, pH 7.5), the antibodies were applied to the CHT column, washed with equilibration buffer and eluted in an linear gradient to 10 mM NaH2P04, 20 mM MES, 500 mM NaCl, 0.1 mM CaCl2, pH 7.5. The antibody containing fractions (from ion exchange chromatography or MabSelect SuRe affinity chromatography) were pooled and further purified by size exclusion chromatography using a Superdex 200 26/60 GL (GE Healthcare, Sweden) column equilibrated with 20 mM histidine, 140 mM NaCl, pH 6.0. The antibody containing fractions were pooled, concentrated to the required concentration using Vivaspin ultrafiltration devices (Sartorius Stedim Biotech S.A., France) and stored at -80°C.
Example 3
SDS-CE and analytical SEC of antibodies SDS-CE
Purity, antibody integrity and molecular weight of antibodies were analyzed by CE-SDS using microfiuidic Labchip technology (Caliper Life Science, USA). 5 μΐ of protein solution was prepared for CE-SDS analysis using the HT Protein Express Reagent Kit according manufacturer's instructions and analyzed on LabChip GXII system using a HT Protein Express Chip. Data were analyzed using LabChip GX Software. Analytical size exclusion chromatography
Size exclusion chromatography for the determination of the aggregation and oligomeric state of antibodies was performed by HPLC chromatography. Briefly, purified antibodies at various levels of the purification process were applied to a Tosoh TSKgel G3000SW column in 300 mM NaCl, 50 mM KH2P04/K2HP04, H 7.5 on an Agilent HPLC 1100 system or to a Superdex 200 column (GE Healthcare) in 2 x PBS on a Dionex HPLC-System. The eluted protein was quantified by UV absorbance and integration of peak areas. BioRad Gel Filtration Standard 151-1901 served as a standard. Table 1: Yield, purity profile and aggregation tendencies (aggregate tendencies are reflected by % monomers after Protein A)
Figure imgf000043_0001
Example 4
Binding to IL-17 and Crossreactivity with IL17 subtypes measured by ELISA NUNC® Maxisorp plates (96-well) are coated with recombinant human IL-17 (Peprotech # 200-17, www.peprotech.com) at a concentration of 0^g/ml in PBS (lOOml/well). Plates are incubated at 37°C on an orbital shaker with agitation for 2 hours. Thereafter coating solution is removed and ΙΟΟμΙ/well PBSTC (phosphate buffered saline, 0,05%> Tween®20, 2%> chicken serum) is added. Plates are incubated at room temperature for 1 hour. Blocking solution is removed and samples (blank: PBSTC, samples (10μg/ml in PBS): anti-human IL-17 antibodies <IL17> 9C6-2B6, <IL17> 9C6-2B6-134, <IL17> 9C6-2B6-136, Mab 16-7178-85 of eBioscience (www.ebioscience.com); MAB 317 of R&D Systems (www.rndsystems.com), NVP-AIN-497 (WO 2006/013107); are added to the plate (ΙΟΟμΙ/well). Plates are incubated at room temperature with agitation. Samples are removed, plates are washed three times with 200μ1Λνε11 PBST (phosphate buffered saline, 0,05%> Tween® 20) and second antibody (Goat anti-mouse IgG, Fc gamma, HRP conjugate; Chemicon AP127P, www.millipore.com) for the detection of mouse antibodies or Goat Anti-human IgG, Fc gamma, HRP conjugate (Chemicon AP113P) for the detection of humanized antibodies is added. The second antibody is diluted 1 : 10000 in PBSTC and plates are incubated for 1 hour at room temperature with agitation. Second antibody is removed, plates are washed three times with 200μ1Λνε11 PBST (phosphate buffered saline, 0,05% Tween®20) and ΙΟΟμΙ/well ABTS® (Roche Diagnostics GmbH) is added. Optical density is measured at 405/492nm in relation to IL-17A binding (set as 100%). Binding to other human IL-17 subtypes (IL-17B, IL-17C, IL-17D, IL-17E and IL-17F) were performed with the same assay format. Results are shown in table 4. The results show that antibody with the most similar binding behavior against the different IL17 subtypes is Mab MAB 317 of R&D Systems (www.rndsystems.com).
Table 2:
Figure imgf000044_0001
Example 5
Cytokine release assay, inhibition of IL-17A induced hIL-6 and hIL-8 release in CCD-25SK cells
The assay is performed as detection of hIL-8 production of CCD-25SK cells (skin fibroblasts, ATCC No: CRL-1474) after stimulation with IL-17A and TNF-alpha with preincubation of anti-IL-17 antibodies. CCD-25SK cells have the IL-17 receptor. Soluble IL-17A binds to the these IL-17 receptor. Antibodies against IL-17A bind to IL-17A. The mechanism is only working in the presence of TNFalpha. Through the binding of IL-17A to the IL-17 receptor, the cells produce hIL-6 and hIL-8 which can be detected by ELISA as a read out. The measured hIL-6 and hIL-8 give the information in which concentrations anti-IL-17 antibodies inhibit the stimulation of CCD-25SK cells by IL-17.
CCD-25SK cells were seeded with a cell density of 2,5x104 cells/well in a 48-well plate (volume 0,45ml/well) and incubated for 24h at 37°C and 5%C02. After overnight incubation the cells were treated with anti-IL-17 antibodies for 30 minutes with end concentrations of 9000; 3000;1000; 333,3; 111,1; 37,03; 12,34; and 4,11 ng/ml. Each antibody dilution series was made with medium, 50μ1Λνε11 (lOx concentrated). After 30 min the cells were stimulated with a mixture of lOng/ml IL-17A and 50pg/ml TNF-alpha. 50μ1Λνε11 (lOx concentrated) and incubated for 24h at 37°C and 5%C02. After overnight incubation the supernatants were transferred to 96-well plates and frozen at -20°C as intermediates for hIL-8 ELISA. hIL-6 and hIL-8 ELISA was performed as follows. ΙΟΟμΙ diluted capture antibody was added to each well and incubated overnight at 4°C. Dilutions were made with coating buffer. Plates were aspirated, washed with 200μ1Λνε11 for 3 times, blocked with 200μ1Λνε11 assay diluent, and incubated for lh at RT. The plates were aspirated and washed with 200μ1Λνε11 for 3 times. ΙΟΟμΙ standard and samples were added and incubated for 2h at RT. Standard dilution series: 400pg/ml; 200pg/ml; lOOpg/ml; 50pg/ml; 25pg/ml; 12,5pg/ml; 6,3pg/ml and assay diluent as negative control. Sample dilution was 1 :200. Plates were aspirated and washed with 250μ1Λνε11 for 4 times. ΙΟΟμΙ conjugate was added to each well. The conjugate was prepared with detection antibody and enzyme reagent 1 :250 diluted in assay diluent. Plates were aspirated and washed with 250μ1Λνε11 for 6 times. ΙΟΟμΙ substrate was added to each well and incubated for 12 minutes. After incubation the reaction was stopped with 50μ1Λνε11 1M H2S04. Read out was performed at 450nm within 30 min with λ correction at 570nm. Results are shown in table 5 (IC50 values measured in relation to a maximal inhibition of 80%).
Table 3:
Figure imgf000045_0001
Example 6
Synoviocyte cytokine release assay, inhibition of IL-17A induced hIL-6 and hIL-8
The < IL-17> antibodies were tested for inhibition of an IL-17-induced production of pro-inflammatory cytokines (e.g. IL-6, -8) by human adult fibroblast-like synoviocytes obtained from RA patients (human fibroblast-like synoviocytes- rheumatoid arthritis (HFLS-RA)). After establishment of a dose-response response of different RA-FLS donors, the potency of the antibodies was assessed.
HFLS-RA (Cat. #408RA-05a) were purchased from Cell Applications Inc. (San Diego, CA, USA; German distributor: tebu-bio, Offenbach, Germany). They are cryopreserved at second passage and can be cultured and propagated at least 5 population doublings. HFLS are long known for their role in joint destruction by producing cytokines and metalloproteinases that contribute to cartilage degradation (Firestein, G.S., et al, J. Immunol. 149 (1992) 1054, Firestein, G.S., et al, Arthritis and Rheumatism 37(5) (1994) 644) Proinflammatory cytokines induce the proliferation, collagenase and aggrecanase production and GM-CSF secretion on HFLS (Alvaro, J.M., et al, J. Clin. Immunol. 13(3) (1993) 212; Yamanishi, Y., et al, J. Immunol. 168(3) (2002) 1405). Cells were thawed, expanded in Synoviocyte Growth Medium (Cell Applications, Inc.; Cat. #415-500), detached with Accutase (PAA Laboratories GmbH, Pasching, Austria; Cat. #L11-007) before approx. 2xl04
HFLS-RA cells/well were seeded in 200μ1Λνε11 medium in 96wF cell culture plates (Costar/Corning Life Sciences, Amsterdam, The Netherlands; Cat. #3596).
Cells were pre-cultured for two days at 37°C, 5 %C02 before cytokines (and optionally antibodies) were added. Prior to the cytokine addition, medium was removed and 150μ1Λν of the corresponding cytokine (optional: antibody) dilution was added: 0-10μg/ml rec. human IL-17A (PeproTech, Hamburg, Germany; Cat. #200-17); 0-25 μg/ml rec. human TWEAK (R&D Systems, Wiesbaden, Germany; Cat. #1090-TW/CF), or 0-^g/ml rec. human TNFa (R&D Systems; Cat. #210- TA/CF) was titrated in ten 1 : 10 dilution steps to obtain an ED50 value for the indicated cytokines. TWEAK was used as negative and TNF was used as positive control. The cells were incubated for 6h, 24h and 72h at 37°C, 5 % C02, whereas 72 hrs was used for the following experiments as this incubation time gave the most robust cytokine response (on a protein level). Cells were pre-incubated with several antibodies at different concentrations
Figure imgf000046_0001
for 30 min at 37°C, 5 % C02 before stimulation with 100ng/ml-^g/ml TWEAK for additional
72 hrs was applied. Table 4: Effective dose (ED50) values in ng/ml of cytokine induction by IL-17, Tweak and TNFalpha of synovial fibroblasts.
Figure imgf000047_0001
Table 5: Inhibitory concentration (IC50) in nM of <IL-17> antibodies after cytokine induction by IL-17 of synovial fibroblasts
Figure imgf000047_0002
Example 7
Crossreactivity with Cynomolgous IL-17A (Binding Assay)
Relative binding to human and cynomolgous IL17A was determined. The binding assay was performed according to example 2. The results for two separates experiments (one in which mouse IL17 antibodies were compared and one in which human and humanized IL17 antibodies were compared) are shown in tables 6a and 6b.
Table 6a:
IL 17 Antibody Relative binding to Relative binding to (mouse) human IL-17A in % cynomolgous IL-17A in %
Mab 317 (R&D) 100 100 eBio64CAP17 122 124
9C6/2B6 127 134 Table 6b:
Figure imgf000048_0001
Example 8
Cynomolgous monkey (Maccaca Fasicularis) cytokine release assay, inhibition of cynomolgous IL-17A induced IL-6 and IL-8 production Cynomolgous dermal fibroblasts (CDF) cells produce cynomolgous IL-6 and IL-8 in response to human or cynomolgous IL-17A stimulation. The assay is performed to measure the inhibition of this cynomolgous IL-17A stimulated IL-6 and IL-8 production by CDF cells following preincubation of the cells with anti-IL-17 antibodies raised against human IL-17 prior to stimulation. CDF cells are seeded with a cell density of 2xl05 cells/ml in a volume of 0.5ml in a
48-well plate, and incubated overnight at 37°C and 5%C02 to adhere. After overnight incubation, the media is replaced with 400μ1 fresh media and the cells are treated with anti-IL-17 antibodies for 30 minutes across a range of antibody concentrations (10000, 3000, 1000, 300, 100, 30, 10, 3, 0 ng/ml). Each antibody dilution series is made with medium using 50μ1Λνε11 (ΙΟχ concentrated). After
30 min the cells are stimulated with lOOng/ml IL-17A (50μ1 of lOOOng/ml lOx concentration) and incubated overnight (18h) at 37°C and 5%C02. After the incubation period, supematants are transferred into fresh tubes and either analyzed immediately or stored at -80°C until analysis by ELISA. hIL-6 and hIL-8 ELISA were shown to be cross-reactive with their respective cynomolgous cytokines and are used to quantitate cytokine levels. For the ELISA's ΙΟΟμΙ diluted capture antibody is added to each well and incubated overnight at 4°C. Dilutions are made with coating buffer. Plates are aspirated, washed with 200μ1Λνε11 for 3 times, blocked with 200μ1Λνε11 assay diluent, and incubated for lh at RT. The plates are aspirated and washed with 200μ1Λνε11 for 3 times. ΙΟΟμΙ standard and samples are added and incubated for 2h at RT according to the manufacturer's instructions. Plates are aspirated and washed with 250μ1Λνε11 for at least 3 times. ΙΟΟμΙ conjugate is added to each well. The conjugate is prepared with detection antibody and enzyme reagent 1 :250 diluted in assay diluent. Plates are aspirated and washed with 250μ1Λνε11 for at least 3 times. ΙΟΟμΙ substrate was added to each well and incubated until sufficient color had developed for reading. After incubation the reaction is stopped with 50μ1Λνε11 1M H2S04 and read on the plate reader at a wavelength of 450nm within 30 min.
Example 9
Binding affinity of the < IL17> antibodies
The <IL17> binding affinity of antibodies was measured by Surface Plasmon Resonance (SPR) using a BIAcore® T100 instrument (GE Healthcare) at 25°C. The BIAcore® system is well established for the study of molecule interactions. SPR-technology is based on the measurement of the refractive index close to the surface of a gold coated biosensor chip. Changes in the refractive index indicate mass changes on the surface caused by the interaction of immobilized ligand with analyte injected in solution. The mass increases if molecules bind immobilized ligands on the surface, and vice versa, the mass decreases in case of dissociation of the analyte from the immobilized ligand (reflecting complex dissociation). SPR allows a continuous real-time monitoring of ligand/analyte binding and thus the determination of the association rate constant (ka), the dissociation rate constant (kd), and of the equilibrium constant (KD).
IL17 binding affinity
Around 12000 resonance units (RU) of the capturing system (10 μg/ml goat anti human F(ab')2; Order Code: 28958325; GE Healthcare Bio-Sciences AB, Schweden) were coupled on a CM5 chip at pH 5.0 by using an amine coupling kit supplied by the GE Healthcare. The sample and system buffer was PBS-T (10 mM phosphate buffered saline including 0.05% Tween20 ) pH 7.4. The antibody was captured by injecting a 50nM solution for 1 min at a flow of 10 μΐ/min. Association was measured by injection human IL17 in various concentrations in solution for 3 min at a flow of 30 μΐ/min starting with 50nM in 1 : 1 dilutions. The dissociation phase was monitored for up to 5 min and triggered by switching from the sample solution to running buffer. The surface was regenerated by two times 60 sec. washing with a glycin pH 2.1 solution at a flow rate of 30 μΐ/min. Bulk refractive index differences were corrected by subtracting the response obtained from a goat anti human F(ab')2 surface. Blank injections are also subtracted (= double referencing). For calculation of apparent KD and other kinetic parameters the Langmuir 1 :1 model was used. Results are shown in Table 17 below.
Table 7: Binding affinity to human IL17
Figure imgf000050_0001
Example 10
Small-scale Dynamic Light Scattering (DLS) -based stability and viscosity measurement of <IL-17> antibodies
Viscosity measurement is essentially performed as described in He, F., et al, Analytical Biochemistry 399(1) (2009) 141-3. Briefly, samples are concentrated to various protein concentrations in 200 mM arginine succinate, pH 5.5, before polystyrene latex beads (300 nm diameter) and Polysorbate 20 (0.02 % v/v) are added. Samples are transferred into an optical 384-well plate by centrifugation through a 0.4 μιη filter plate and covered with paraffine oil. The apparent diameter of the latex beads is determined by dynamic light scattering at 25 °C. The viscosity of the solution can be calculated as η = /7o(n h,o) (η-' viscosity; η0: viscosity of water; rh: apparent hydrodynamic radius of the latex beads; rbfi: hydrodynamic radius of the latex beads in water.
To allow comparison of various samples at the same concentration, viscosity- concentration data were fitted with the Mooney equation (Equation 1) [(Mooney,
Colloid Sci (1951); Monkos, Biochem. Biophys. Acta (1997)] and data interpolated accordingly. f ΞΦ
η = η0 exp
(S: hydrodynamic interaction parameter of the protein; K: self-crowding factor; : volume fraction of the dissolved protein) Stability of bispecific antibodies
Samples are concentrated to a final concentration of 150 mg/mL in 200 mM arginine succinate, pH 5.5, sterile filtered and quiescently stored at 40°C for 4 days. Before and after storage, the content of high molecular weight (HMW) species is determined by size-exclusion chromatography. The difference in HMW content between the stored sample and a sample measured immediately after preparation is reported as "HMW increase".
Table 8: Stability of bispecific antibodies was assessed by DLS and SE.-HPLC.
<IL17> <IL17>
9C6-2B6- 9C6-2B6- 134 136
DLS aggregation onset ~66°C ~58°C
temperature (= aggregation
temperature)
4 d storage at 40°C at 150 n.d. n.d.
mg/ml (HMW increase)

Claims

Patent Claims
An antibody that specifically binds to human IL17, characterized in comprising a CDR1H of SEQ ID NO: l, CDR2H of SEQ ID NO:2, CDR3H of SEQ ID NO:3, and CDR1L of SEQ ID NO:4, CDR2L of SEQ ID NO:5, CDR3L of SEQ ID NO:6.
A chimeric or humanized variant of the antibody according to claim 1. The antibody according to any of claims 1 to 2, characterized in comprising a) a variable heavy chain domain (VH) of SEQ ID NO:9, or of SEQ ID NO: 10, and b) a variable light chain domain (VL) of SEQ ID NO: 11, or of SEQ ID NO: 12.
The antibody according to any of claims 1 to 2, characterized in comprising a) a variable heavy chain domain (VH) of SEQ ID NO: 10, and b) a variable light chain domain (VL) of SEQ ID NO: 12.
The antibody according to any of claims 1 to 2, characterized in comprising a) a variable heavy chain domain (VH) of SEQ ID NO:9, and b) a variable light chain domain (VL) of SEQ ID NO: 11.
The antibody according to any of the preceding claims, characterized in being of IgGl or IgG4 subclass.
The antibody according to any of the preceding claims, characterized in being of IgGl subclass with the mutations L234A and L235A (numbering according to the EU index of Kabat).
8. The antibody according to any of the preceding claims, characterized in being of IgGl subclass with the mutations L234A, L235A and P329G (numbering according to the EU index of Kabat).
9. The antibody according to any of the preceding claims, characterized in being of IgG4 subclass with the mutations S228P and L235E (numbering according to the EU index of Kabat).
10. The antibody according to any of the preceding claims, characterized in being of IgG4 subclass with the mutations S228P, L235E and P329G (numbering according to the EU index of Kabat).
11. Pharmaceutical composition characterized by comprising an antibody according to claims 1 to 10.
12. Use of an antibody according to claims 1 to 10 for the manufacture of a pharmaceutical composition.
13. An antibody according to claims 1 to 10 for use in the treatment of cancer, or inflammatory diseases, autoimmune diseases, rheumatoid arthritis, psoratic arthritis, muscle diseases, e.g. muscular dystrophy, multiple sclerosis, chronic kidney diseases, bone diseases, e.g. bone degeneration in multiple myeloma, systemic lupus erythematosus, lupus nephritis, and vascular injury.
14. Use of an antibody according to claims 1 to 10 for manufacture of a medicament for the treatment of cancer, or inflammatory diseases, autoimmune diseases, rheumatoid arthritis, psoratic arthritis, muscle diseases, e.g. muscular dystrophy, multiple sclerosis, chronic kidney diseases, bone diseases, e.g. bone degeneration in multiple myeloma, systemic lupus erythematosus, lupus nephritis, and vascular injury.
15. Nucleic acid encoding an antibody according to claims 1 to 10.
16. Expression vectors characterized by comprising a nucleic acid according to claim 15 for the expression of the antibody according to claims 1 to 10 in a prokaryotic or eukaryotic host cell.
17. Prokaryotic or eukaryotic host cell comprising a vector according to claim 16.
18. Method for the production of a recombinant antibody according to claims 1 to 10, characterized by expressing a nucleic acid according to claim 15 in a prokaryotic or eukaryotic host cell and recovering said antibody from said cell or the cell culture supernatant. Method for the treatment of a patient suffering from cancer or from inflammatory diseases, autoimmune diseases, rheumatoid arthritis, psoratic arthritis, muscle diseases, e.g. muscular dystrophy, multiple sclerosis, chronic kidney diseases, bone diseases, e.g. bone degeneration in multiple myeloma, systemic lupus erythematosus, lupus nephritis, and vascular injury, characterized by administering to the patient an antibody according to claims 1 to 10.
PCT/EP2013/056971 2013-04-03 2013-04-03 Antibodies against human il17 and uses thereof WO2014161570A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
PCT/EP2013/056971 WO2014161570A1 (en) 2013-04-03 2013-04-03 Antibodies against human il17 and uses thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/EP2013/056971 WO2014161570A1 (en) 2013-04-03 2013-04-03 Antibodies against human il17 and uses thereof

Publications (1)

Publication Number Publication Date
WO2014161570A1 true WO2014161570A1 (en) 2014-10-09

Family

ID=48045533

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2013/056971 WO2014161570A1 (en) 2013-04-03 2013-04-03 Antibodies against human il17 and uses thereof

Country Status (1)

Country Link
WO (1) WO2014161570A1 (en)

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017215524A1 (en) * 2016-06-12 2017-12-21 三生国健药业(上海)股份有限公司 Anti-human interleukin-17a monoclonal antibody, and preparation method and use thereof
WO2018002339A1 (en) * 2016-07-01 2018-01-04 Alligator Bioscience Ab Bispecific antibodies directed against ox40 and a tumor-associated antigen
EP3215527A4 (en) * 2014-11-05 2018-04-18 Annexon, Inc. Humanized anti-complement factor c1q antibodies and uses thereof
US10227398B2 (en) 2013-07-09 2019-03-12 Annexon, Inc. Anti-complement factor C1q antibodies and uses thereof
JP2020511159A (en) * 2017-03-10 2020-04-16 スージョウ カノヴァ バイオファーマシューティカル カンパニーリミテッドSuzhou Kanova Biopharmaceutical Co., Ltd. Monoclonal antibodies reactive with IL-17A and IL-17F, and uses thereof
US10723788B2 (en) 2015-11-24 2020-07-28 Annexon, Inc. Anti-complement factor C1q Fab fragments and uses thereof
EP3689907A1 (en) 2019-01-31 2020-08-05 Numab Therapeutics AG Antibodies targeting il-17a and methods of use thereof
WO2020157305A1 (en) 2019-01-31 2020-08-06 Numab Therapeutics AG Multispecific antibodies having specificity for tnfa and il-17a, antibodies targeting il-17a, and methods of use thereof
WO2021018035A1 (en) * 2019-07-26 2021-02-04 神州细胞工程有限公司 Humanized anti-il17a antibody and use thereof
CN116063493A (en) * 2022-07-22 2023-05-05 北京绎源生物科技有限公司 Pharmaceutical composition comprising antibody and mesenchymal stem cell exosomes and method for preparing the same
RU2804963C2 (en) * 2019-07-26 2023-10-09 Синоселлтех Лтд. Humanized anti-il17a antibody and its application

Citations (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0307434A1 (en) 1987-03-18 1989-03-22 Medical Res Council Altered antibodies.
US5202238A (en) 1987-10-27 1993-04-13 Oncogen Production of chimeric antibodies by homologous recombination
US5204244A (en) 1987-10-27 1993-04-20 Oncogen Production of chimeric antibodies by homologous recombination
WO1995018826A2 (en) 1994-01-05 1995-07-13 Schering Corporation Purified primate ctla-8 antigens and related reagents
WO1996017939A1 (en) 1994-12-07 1996-06-13 Immunex Corporation Isolated herpesvirus saimiri proteins that bind mhc class ii molecules
WO1997015320A1 (en) 1995-10-27 1997-05-01 Schering Corporation Novel uses of mammalian ctla-8 and related reagents
WO1997022596A1 (en) 1995-12-18 1997-06-26 Zeneca Limited Quinazoline derivatives
WO1997030035A1 (en) 1996-02-13 1997-08-21 Zeneca Limited Quinazoline derivatives as vegf inhibitors
WO1997032856A1 (en) 1996-03-05 1997-09-12 Zeneca Limited 4-anilinoquinazoline derivatives
WO1998013354A1 (en) 1996-09-25 1998-04-02 Zeneca Limited Quinazoline derivatives and pharmaceutical compositions containing them
WO1999002166A1 (en) 1997-07-08 1999-01-21 Angiogene Pharmaceuticals Ltd. Use of colchinol derivatives as vascular damaging agents
WO1999035276A1 (en) 1998-01-09 1999-07-15 Schering Corporation Mammalian matrix-type metalloprotease
US6063372A (en) 1995-10-27 2000-05-16 Inserm, Institut National De La Sante Et De La Recherche Medicale Uses of mammalian CTLA-8 and related reagents
WO2000040529A1 (en) 1999-01-07 2000-07-13 Angiogene Pharmaceuticals Ltd. Colchinol derivatives as vascular damaging agents
WO2000041669A2 (en) 1999-01-15 2000-07-20 Angiogene Pharmaceuticals Ltd. Benzimidazole vascular damaging agents
WO2000069436A1 (en) 1999-05-14 2000-11-23 Sepracor Inc. Methods of using and compositions comprising n-desmethylzolpidem
US6171586B1 (en) 1997-06-13 2001-01-09 Genentech, Inc. Antibody formulation
US6267958B1 (en) 1995-07-27 2001-07-31 Genentech, Inc. Protein formulation
US6274711B1 (en) 1993-06-14 2001-08-14 Inserm, Institut National De La Sante Et De La Recherche Medicale Purified mammalian CTLA-8 antigens and related reagents
WO2001092224A1 (en) 2000-05-31 2001-12-06 Astrazeneca Ab Indole derivatives with vascular damaging activity
WO2002004434A1 (en) 2000-07-07 2002-01-17 Angiogene Pharmaceuticals Limited Colchinol derivatives as vascular damaging agents
WO2002008213A1 (en) 2000-07-07 2002-01-31 Angiogene Pharmaceuticals Limited Colchinol derivatives as angiogenesis inhibitors
WO2005051422A1 (en) 2003-11-21 2005-06-09 Celltech R & D Limited Method for the treatment of multiple sclerosis by inhibiting il-17 activity
US20050260186A1 (en) 2003-03-05 2005-11-24 Halozyme, Inc. Soluble glycosaminoglycanases and methods of preparing and using soluble glycosaminoglycanases
WO2006013107A1 (en) 2004-08-05 2006-02-09 Novartis Ag Il-17 antagonistic antibodies
WO2006044908A2 (en) 2004-10-20 2006-04-27 Genentech, Inc. Antibody formulation in histidine-acetate buffer
US20060104968A1 (en) 2003-03-05 2006-05-18 Halozyme, Inc. Soluble glycosaminoglycanases and methods of preparing and using soluble glycosaminogly ycanases
WO2008002115A1 (en) 2006-06-27 2008-01-03 Vladimir Guzenko Diaphragm hydrounit for converting gravitational force into a torque for a fuel-less engine and said engine
WO2010102251A2 (en) 2009-03-05 2010-09-10 Abbott Laboratories Il-17 binding proteins

Patent Citations (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0307434A1 (en) 1987-03-18 1989-03-22 Medical Res Council Altered antibodies.
US5202238A (en) 1987-10-27 1993-04-13 Oncogen Production of chimeric antibodies by homologous recombination
US5204244A (en) 1987-10-27 1993-04-20 Oncogen Production of chimeric antibodies by homologous recombination
US6274711B1 (en) 1993-06-14 2001-08-14 Inserm, Institut National De La Sante Et De La Recherche Medicale Purified mammalian CTLA-8 antigens and related reagents
WO1995018826A2 (en) 1994-01-05 1995-07-13 Schering Corporation Purified primate ctla-8 antigens and related reagents
WO1996017939A1 (en) 1994-12-07 1996-06-13 Immunex Corporation Isolated herpesvirus saimiri proteins that bind mhc class ii molecules
US5716623A (en) 1994-12-07 1998-02-10 Immunex Corporation Isolated Herpesvirus saimiri proteins that bind MHC Class II molecules
US6267958B1 (en) 1995-07-27 2001-07-31 Genentech, Inc. Protein formulation
US6063372A (en) 1995-10-27 2000-05-16 Inserm, Institut National De La Sante Et De La Recherche Medicale Uses of mammalian CTLA-8 and related reagents
WO1997015320A1 (en) 1995-10-27 1997-05-01 Schering Corporation Novel uses of mammalian ctla-8 and related reagents
WO1997022596A1 (en) 1995-12-18 1997-06-26 Zeneca Limited Quinazoline derivatives
WO1997030035A1 (en) 1996-02-13 1997-08-21 Zeneca Limited Quinazoline derivatives as vegf inhibitors
WO1997032856A1 (en) 1996-03-05 1997-09-12 Zeneca Limited 4-anilinoquinazoline derivatives
WO1998013354A1 (en) 1996-09-25 1998-04-02 Zeneca Limited Quinazoline derivatives and pharmaceutical compositions containing them
US6171586B1 (en) 1997-06-13 2001-01-09 Genentech, Inc. Antibody formulation
WO1999002166A1 (en) 1997-07-08 1999-01-21 Angiogene Pharmaceuticals Ltd. Use of colchinol derivatives as vascular damaging agents
WO1999035276A1 (en) 1998-01-09 1999-07-15 Schering Corporation Mammalian matrix-type metalloprotease
WO2000040529A1 (en) 1999-01-07 2000-07-13 Angiogene Pharmaceuticals Ltd. Colchinol derivatives as vascular damaging agents
WO2000041669A2 (en) 1999-01-15 2000-07-20 Angiogene Pharmaceuticals Ltd. Benzimidazole vascular damaging agents
WO2000069436A1 (en) 1999-05-14 2000-11-23 Sepracor Inc. Methods of using and compositions comprising n-desmethylzolpidem
WO2001092224A1 (en) 2000-05-31 2001-12-06 Astrazeneca Ab Indole derivatives with vascular damaging activity
WO2002004434A1 (en) 2000-07-07 2002-01-17 Angiogene Pharmaceuticals Limited Colchinol derivatives as vascular damaging agents
WO2002008213A1 (en) 2000-07-07 2002-01-31 Angiogene Pharmaceuticals Limited Colchinol derivatives as angiogenesis inhibitors
US20050260186A1 (en) 2003-03-05 2005-11-24 Halozyme, Inc. Soluble glycosaminoglycanases and methods of preparing and using soluble glycosaminoglycanases
US20060104968A1 (en) 2003-03-05 2006-05-18 Halozyme, Inc. Soluble glycosaminoglycanases and methods of preparing and using soluble glycosaminogly ycanases
WO2005051422A1 (en) 2003-11-21 2005-06-09 Celltech R & D Limited Method for the treatment of multiple sclerosis by inhibiting il-17 activity
WO2006013107A1 (en) 2004-08-05 2006-02-09 Novartis Ag Il-17 antagonistic antibodies
WO2006044908A2 (en) 2004-10-20 2006-04-27 Genentech, Inc. Antibody formulation in histidine-acetate buffer
WO2008002115A1 (en) 2006-06-27 2008-01-03 Vladimir Guzenko Diaphragm hydrounit for converting gravitational force into a torque for a fuel-less engine and said engine
WO2010102251A2 (en) 2009-03-05 2010-09-10 Abbott Laboratories Il-17 binding proteins

Non-Patent Citations (48)

* Cited by examiner, † Cited by third party
Title
"Current Protocols in Cell Biology", 2000, JOHN WILEY & SONS
"Current Protocols in Molecular Biology", 1987, GREENE PUBLISHING AND WILEY INTERSCIENCE
"Remington's Pharmaceutical Sciences", 1980
ALVARO, J.M. ET AL., J. CLIN. IMMUNOL., vol. 13, no. 3, 1993, pages 212
BARNES, L.M. ET AL., BIOTECH. BIOENG., vol. 73, 2001, pages 261 - 270
BARNES, L.M. ET AL., CYTOTECHNOLOGY, vol. 32, 2000, pages 109 - 123
BOACKLE, R.J. ET AL., NATURE, vol. 282, 1979, pages 742 - 743
BOSMANN MARKUS ET AL: "Therapeutic potential of targeting IL-17 and IL-23 in sepsis.", CLINICAL AND TRANSLATIONAL MEDICINE 2012, vol. 1, no. 1, 4, 2012, pages 1 - 5, XP002717294, ISSN: 2001-1326 *
BRUNHOUSE, R.; CEBRA, J.J., MOL. IMMUNOL., vol. 16, 1979, pages 907 - 917
BURTON, D.R. ET AL., NATURE, vol. 288, 1980, pages 338 - 344
CARTER, P. ET AL., PROC. NATL. ACAD. SCI. USA, vol. 89, 1992, pages 4285 - 4289
DUROCHER, Y. ET AL., NUCL. ACIDS. RES., vol. 30, 2002, pages E9
FIRESTEIN, G.S. ET AL., ARTHRITIS AND RHEUMATISM, vol. 37, no. 5, 1994, pages 644
FIRESTEIN, G.S. ET AL., J. IMMUNOL., vol. 149, 1992, pages 1054
GEISSE, S. ET AL., PROTEIN EXPR. PURIF., vol. 8, 1996, pages 271 - 282
HE, F. ET AL., ANALYTICAL BIOCHEMISTRY, vol. 399, no. 1, 2009, pages 141 - 3
HELLINGS, P.W. ET AL., AM. J. RESP. CELL MOL. BIOL., vol. 28, 2003, pages 42 - 50
HEZAREH, M. ET AL., J. VIROLOGY, vol. 75, 2001, pages 12161 - 12168
HUSTON, J.S., METHODS IN ENZYMOL., vol. 203, 1991, pages 46 - 52
IDUSOGIE, E.E. ET AL., J. IMMUNOL., vol. 164, 2000, pages 4178 - 4184
JOHANSEN C ET AL: "Characterization of the interleukin-17 isoforms and receptors in lesional psoriatic skin.", THE BRITISH JOURNAL OF DERMATOLOGY FEB 2009, vol. 160, no. 2, February 2009 (2009-02-01), pages 319 - 324, XP002717293, ISSN: 1365-2133 *
JOHNSON, G.; WU, T.T., NUCLEIC ACIDS RES., vol. 28, 2000, pages 214 - 218
KABAT, E.A. ET AL.: "Sequences of Proteins of Immunological Interest", 1991, NATIONAL INSTITUTES OF HEALTH
KABAT, E.A. ET AL.: "Sequences of Proteins of Immunological Interest", 1991, PUBLIC HEALTH SERVICE, NATIONAL INSTITUTES OF HEALTH
KAUFMAN, R.J., MOL. BIOTECHNOL., vol. 16, 2000, pages 151 - 160
KOMIYAMA, Y. ET AL., J. IMMUNOL., vol. 177, 2006, pages 566 - 573
KOTAKE, S. ET AL., J. CLIN. INVEST., vol. 103, 1999, pages 1345 - 1352
LUKAS, T.J. ET AL., J. IMMUNOL., vol. 127, 1981, pages 2555 - 2560
MAKRIDES, S.C., PROTEIN EXPR. PURIF., vol. 17, 1999, pages 183 - 202
MATUSEVICIUS, D. ET AL., MULTIPLE SCLEROSIS, vol. 5, 1999, pages 101 - 104
MONKOS, BIOCHEM. BIOPHYS. ACTA, 1997
MOONEY, COLLOID SCI, 1951
MORGAN, A. ET AL., IMMUNOLOGY, vol. 86, 1995, pages 319 - 324
MORRISON, S.L. ET AL., PROC. NATL. ACAD SCI. USA, vol. 81, 1984, pages 6851 - 6855
NEUBERGER, M.S. ET AL., NATURE, vol. 314, 1985, pages 268 - 270
NORDERHAUG, L. ET AL., J. IMMUNOL. METHODS, vol. 204, 1997, pages 77 - 87
ORLANDI, R. ET AL., PROC. NATL. ACAD. SCI. USA, vol. 86, 1989, pages 3833 - 3837
PACE, C.N., PROTEIN SCIENCE, vol. 4, 1995, pages 2411 - 1423
R&D SYSTEMS: "Monoclonal Anti-human IL-17 Antibody", ANNOUNCEMENT R&D SYSTEMS, XX, XX, 11 January 2004 (2004-01-11), pages 1 - 2, XP002351567 *
RIECHMANN, L. ET AL., NATURE, vol. 332, 1988, pages 323 - 327
SAMBROOK, J. ET AL.: "Molecular Cloning: A Laboratory Manual", 1989, COLD SPRING HARBOR LABORATORY PRESS
SCHLAEGER, E.-J., J. IMMUNOL. METHODS, vol. 194, 1996, pages 191 - 199
SCHLAEGER, E.-J.; CHRISTENSEN, K., CYTOTECHNOLOGY, vol. 30, 1999, pages 71 - 83
THOMMESEN, J.E. ET AL., MOL. IMMUNOL., vol. 37, 2000, pages 995 - 1004
WERNER, R.G., ARZNEIMITTELFORSCHUNG (DRUG RES., vol. 48, 1998, pages 870 - 880
WITOWSKI J ET AL: "Interleukin-17: a mediator of inflammatory responses", CMLS CELLULAR AND MOLECULAR LIFE SCIENCES, BIRKHAUSER VERLAG, HEIDELBERG, DE, vol. 61, no. 5, 1 March 2004 (2004-03-01), pages 567 - 579, XP002318174, ISSN: 1420-682X, DOI: 10.1007/S00018-003-3228-Z *
YAMANISHI, Y. ET AL., J. IMMUNOL., vol. 168, no. 3, 2002, pages 1405
ZIOLKOWSKA, M. ET AL., J. IMMUNOL., vol. 164, 2000, pages 2832 - 2838

Cited By (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10927167B2 (en) 2013-07-09 2021-02-23 Annexon, Inc. Anti-complement factor C1Q antibodies and uses thereof
US10227398B2 (en) 2013-07-09 2019-03-12 Annexon, Inc. Anti-complement factor C1q antibodies and uses thereof
US11649279B2 (en) 2013-07-09 2023-05-16 Annexon, Inc. Anti-complement factor C1Q antibodies and uses thereof
US10590190B2 (en) 2013-07-09 2020-03-17 Annexon, Inc. Anti-complement factor C1q antibodies and uses thereof
EP4295911A3 (en) * 2014-11-05 2024-03-27 Annexon, Inc. Humanized anti-complement factor c1q antibodies and uses thereof
EP3215527A4 (en) * 2014-11-05 2018-04-18 Annexon, Inc. Humanized anti-complement factor c1q antibodies and uses thereof
US10316081B2 (en) 2014-11-05 2019-06-11 Annexon, Inc. Humanized anti-complement factor C1Q antibodies
US10723788B2 (en) 2015-11-24 2020-07-28 Annexon, Inc. Anti-complement factor C1q Fab fragments and uses thereof
WO2017215524A1 (en) * 2016-06-12 2017-12-21 三生国健药业(上海)股份有限公司 Anti-human interleukin-17a monoclonal antibody, and preparation method and use thereof
JP2019525730A (en) * 2016-06-12 2019-09-12 三生国健薬業(上海)股▲ふん▼有限公司Sunshine Guojian Pharmaceut Ical(Shanghai)Co.,Ltd Anti-human interleukin-17A monoclonal antibody, production method and use thereof
WO2018002339A1 (en) * 2016-07-01 2018-01-04 Alligator Bioscience Ab Bispecific antibodies directed against ox40 and a tumor-associated antigen
US10858427B2 (en) 2017-03-10 2020-12-08 Suzhou Kanova Biopharmaceutical Co., Ltd. Monoclonal antibody against both IL-17A and IL-17F and use of the same
JP2020511159A (en) * 2017-03-10 2020-04-16 スージョウ カノヴァ バイオファーマシューティカル カンパニーリミテッドSuzhou Kanova Biopharmaceutical Co., Ltd. Monoclonal antibodies reactive with IL-17A and IL-17F, and uses thereof
EP3689907A1 (en) 2019-01-31 2020-08-05 Numab Therapeutics AG Antibodies targeting il-17a and methods of use thereof
WO2020157305A1 (en) 2019-01-31 2020-08-06 Numab Therapeutics AG Multispecific antibodies having specificity for tnfa and il-17a, antibodies targeting il-17a, and methods of use thereof
WO2021018035A1 (en) * 2019-07-26 2021-02-04 神州细胞工程有限公司 Humanized anti-il17a antibody and use thereof
RU2804963C2 (en) * 2019-07-26 2023-10-09 Синоселлтех Лтд. Humanized anti-il17a antibody and its application
CN116063493A (en) * 2022-07-22 2023-05-05 北京绎源生物科技有限公司 Pharmaceutical composition comprising antibody and mesenchymal stem cell exosomes and method for preparing the same
CN116063493B (en) * 2022-07-22 2023-09-01 贵州省赛尔生物科技有限公司 Pharmaceutical composition comprising antibody and mesenchymal stem cell exosomes and method for preparing the same

Similar Documents

Publication Publication Date Title
US9714292B2 (en) Bispecific antibodies against human TWEAK and human IL17 and uses thereof
WO2014161570A1 (en) Antibodies against human il17 and uses thereof
US9416182B2 (en) Anti-TNF-anti-IL-17 bispecific antibodies
AU2011311673B2 (en) Antibodies against human TWEAK and uses thereof
TW201023882A (en) Antibodies against human iL17 and uses thereof
US8883976B2 (en) Antibodies against human tweak and uses thereof
WO2012007495A1 (en) Antibodies specifically binding to human tslpr and methods of use
US20190040156A1 (en) ANTI-TNF- / ANTI-IL-23 IgG BISPECIFIC ANTIBODIES
JP7415939B2 (en) Anti-human Fn14 antibody
KR20230054451A (en) Methods of Treating Autoimmune Diseases Using Antagonistic CD40 Monoclonal Antibodies

Legal Events

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

Ref document number: 13713886

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 13713886

Country of ref document: EP

Kind code of ref document: A1