WO2013109829A1 - Anticorps anti-adamts4 et méthodes de traitement - Google Patents

Anticorps anti-adamts4 et méthodes de traitement Download PDF

Info

Publication number
WO2013109829A1
WO2013109829A1 PCT/US2013/022062 US2013022062W WO2013109829A1 WO 2013109829 A1 WO2013109829 A1 WO 2013109829A1 US 2013022062 W US2013022062 W US 2013022062W WO 2013109829 A1 WO2013109829 A1 WO 2013109829A1
Authority
WO
WIPO (PCT)
Prior art keywords
seq
antigen binding
antibody
binding protein
amino acid
Prior art date
Application number
PCT/US2013/022062
Other languages
English (en)
Inventor
Jonathan David Larkin
Original Assignee
Glaxosmithkline Intellectual Property Development Ltd
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 Glaxosmithkline Intellectual Property Development Ltd filed Critical Glaxosmithkline Intellectual Property Development Ltd
Publication of WO2013109829A1 publication Critical patent/WO2013109829A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/40Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against enzymes
    • 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
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value

Definitions

  • This invention relates to methods for inhibiting aggrecanase, in particular
  • ADAMTS4 thereby reducing the break down of extracellular matrix aggrecan in cartilage and other tissues.
  • Cartilage is an avascular tissue populated by specialized cells termed chondrocytes, which respond to diverse mechanical and biochemical stimuli. Cartilage is present in the linings of joints, interstitial connective tissues, and basement membranes, and is composed of an extracellular matrix comprised of several matrix components including type II collagen, proteoglycans, fibronectin and laminin.
  • the ensuing response may be either anabolic (leading to matrix production and/or repair) or catabolic (leading to matrix degradation, cellular apoptosis, loss of function, and pain).
  • chondrocytes decrease matrix production and increase production of multiple matrix degrading enzymes.
  • matrix degrading enzymes include aggrecanases (ADAMTSs) and matrix metalloproteases (MMPs). The activities of these enzymes result in the degradation of the cartilage matrix.
  • ADAMTSs aggrecanases
  • MMPs matrix metalloproteases
  • ADAMTSs Aggrecanases
  • MMPs matrix metalloproteases
  • proteoglycan staining is observed in the superficial zone in early OA and adjacent to areas of cartilage erosion in moderate to severe OA.
  • Inflammatory arthritic conditions such as rheumatoid arthritis, psoriatic arthritis and lyme arthritis, undergo similar cartilage degradation outcomes, but the etiologic mechanisms of each disease may differ.
  • ADAMTS4 shown in Figure 5 as SEQ ID NO:29
  • ADAMTS5 shown in Figure 4 as SEQ ID NO: 30
  • SEQ ID NO: 30 Human ADAMTS4 and ADAMTS5 have been shown to cleave aggrecan between amino acids E373 and A374 producing the neoepitope ARGSVIL (SEQ ID NO: l).
  • Excessive degradation of extracellular matrix is implicated in the pathogenesis of many diseases and conditions, including pain, chronic pain, neuropathic pain, postoperative pain, rheumatoid arthritis, osteoarthritis, sports injuries, erosive arthritis, ankylosing spondylosis, neuralgia, neuropathies, algesia, nerve injury, ischaemia, neurodegeneration, cartilage degeneration, stroke, incontinence, inflammatory disorders, irritable bowel syndrome, periodontal disease, chronic kidney disease, aberrant angiogenesis, tumor invasion and metastasis, corneal ulceration, and in complications of diabetes.
  • diseases and conditions including pain, chronic pain, neuropathic pain, postoperative pain, rheumatoid arthritis, osteoarthritis, sports injuries, erosive arthritis, ankylosing spondylosis, neuralgia, neuropathies, algesia, nerve injury, ischaemia, neurodegeneration, cartilage degeneration, stroke, incontinence, inflammatory disorders,
  • the present invention provides isolated polypeptides comprising at least one variable domain capable of binding and/or neutralizing human ADAMTS4.
  • compositions comprising at least one polypeptide of the present invention.
  • Figure 1 In vitro Inhibition of ARGSVIL (SEQ ID NO: 1) Neoepitope Generation by ADAMTS4 mAbs.
  • Figure 2 In vitro Concentration Dependent Inhibition of ARGSVIL (SEQ ID NO: l) Neoepitope Generation by anti-ADAMTS4 murine mAbs.
  • Figure 3 ADAMTS4 mAb CDR amino acid alignments.
  • Figure 4 Amino Acid sequence of human ADAMTS5 (SEQ ID NO:29).
  • Figure 5 Amino Acid sequence of Human ADAMTS4 (SEQ ID NO:30).
  • Figure 6 Structure modeling predicts Ag/Ab interaction sites.
  • Polynucleotide generally refers to any polyribonucleotide or
  • polydeoxribonucleotide which may be unmodified RNA or DNA or modified RNA or DNA.
  • Polynucleotides include, without limitation single- and double-stranded DNA, DNA that is a mixture of single- and double-stranded regions, single- and double-stranded RNA, and RNA that is mixture of single- and double-stranded regions, hybrid molecules comprising DNA and RNA that may be single-stranded or, more typically, double- stranded or a mixture of single- and double-stranded regions.
  • polynucleotide refers to triple-stranded regions comprising RNA or DNA or both RNA and DNA.
  • the term polynucleotide also includes DNAs or RNAs containing one or more modified bases and DNAs or RNAs with backbones modified for stability or for other reasons.
  • Modified bases include, for example, tritylated bases and unusual bases such as inosine. A variety of modifications has been made to DNA and RNA; thus,
  • polynucleotide embraces chemically, enzymatically or metabolically modified forms of polynucleotides as typically found in nature, as well as the chemical forms of DNA and RNA characteristic of viruses and cells. "Polynucleotide” also embraces relatively short polynucleotides, often referred to as oligonucleotides.
  • Polypeptide refers to any peptide or protein comprising two or more amino acids joined to each other by peptide bonds or modified peptide bonds, i.e., peptide isosteres.
  • Polypeptide refers to both short chains, commonly referred to as peptides, oligopeptides or oligomers, and to longer chains, generally referred to as proteins. Polypeptides may contain amino acids other than the 20 gene-encoded amino acids.
  • Polypeptides include amino acid sequences modified either by natural processes, such as posttranslational processing, or by chemical modification techniques that are well known in the art. Such modifications are well described in basic texts and in more detailed monographs, as well as in a voluminous research literature. Modifications can occur anywhere in a
  • polypeptide including the peptide backbone, the amino acid side-chains and the amino or carboxyl termini. It will be appreciated that the same type of modification may be present in the same or varying degrees at several sites in a given polypeptide. Also, a given polypeptide may contain many types of modifications. Polypeptides may be branched as a result of ubiquitination, and they may be cyclic, with or without branching. Cyclic, branched and branched cyclic polypeptides may result from posttranslation natural processes or may be made by synthetic methods.
  • Modifications include acetylation, acylation, ADP-ribosylation, amidation, covalent attachment of flavin, covalent attachment of a heme moiety, covalent attachment of a nucleotide or nucleotide derivative, covalent attachment of a lipid or lipid derivative, covalent attachment of
  • phosphotidylinositol cross-linking, cyclization, disulfide bond formation, demethylation, formation of covalent cross-links, formation of cysteine, formation of pyroglutamate, formylation, gamma-carboxylation, glycosylation, GPI anchor formation, hydroxylation, iodination, methylation, myristoylation, oxidation, proteolytic processing,
  • Variant is a polynucleotide or polypeptide that differs from a reference polynucleotide or polypeptide respectively, but retains essential properties.
  • a typical variant of a polynucleotide differs in nucleotide sequence from another, reference polynucleotide. Changes in the nucleotide sequence of the variant may or may not alter the amino acid sequence of a polypeptide encoded by the reference polynucleotide. Nucleotide changes may result in amino acid substitutions, additions, deletions, fusions and truncations in the polypeptide encoded by the reference sequence, as discussed below.
  • a typical variant of a polypeptide differs in amino acid sequence from another, reference polypeptide. Generally, differences are limited so that the sequences of the reference polypeptide and the variant are closely similar overall and, in many regions, identical.
  • a variant and reference polypeptide may differ in amino acid sequence by one or more substitutions, additions, deletions in any combination.
  • a substituted or inserted amino acid residue may or may not be one encoded by the genetic code.
  • a variant of a polynucleotide or polypeptide may be a naturally occurring such as an allelic variant, or it may be a variant that is not known to occur naturally. Non-naturally occurring variants of polynucleotides and polypeptides may be made by mutagenesis techniques or by direct synthesis.
  • Isolated means altered “by the hand of man” from its natural state, i.e., if it occurs in nature, it has been changed or removed from its original environment, or both.
  • a polynucleotide or a polypeptide naturally present in a living organism is not “isolated,” but the same polynucleotide or polypeptide separated from the coexisting materials of its natural state is “isolated,” including, but not limited to, when such polynucleotide or polypeptide is introduced back into a cell.
  • nucleic acid or polynucleotide e.g., an R A, DNA or a mixed polymer
  • an isolated or substantially pure nucleic acid or polynucleotide is one which is substantially separated from other cellular components that naturally accompany the native polynucleotide in its natural host cell, e.g., ribosomes, polymerases and genomic sequences with which it is naturally associated.
  • the term embraces a nucleic acid or polynucleotide that (1) has been removed from its naturally occurring environment, (2) is not associated with all or a portion of a
  • isolated polynucleotide in which the "isolated polynucleotide” is found in nature (3) is operatively linked to a polynucleotide which it is not linked to in nature, or (4) does not occur in nature.
  • isolated or substantially pure also can be used in reference to recombinant or cloned DNA isolates, chemically synthesized polynucleotide analogs, or polynucleotide analogs that are biologically synthesized by heterologous systems.
  • isolated does not necessarily require that the nucleic acid or polynucleotide so described has itself been physically removed from its native
  • an endogenous nucleic acid sequence in the genome of an organism is deemed “isolated” herein if a heterologous sequence is placed adjacent to the endogenous nucleic acid sequence, such that the expression of this endogenous nucleic acid sequence is altered, for example, increased, decreased or eliminated.
  • a heterologous sequence is a sequence that is not naturally adjacent to the endogenous nucleic acid sequence, whether or not the heterologous sequence is itself endogenous (originating from the same host cell or progeny thereof) or exogenous (originating from a different host cell or progeny thereof).
  • a promoter sequence can be substituted (e.g., by homologous recombination) for the native promoter of a gene in the genome of a host cell, such that this gene has an altered expression pattern.
  • This gene would now become “isolated” because it is separated from at least some of the sequences that naturally flank it.
  • a nucleic acid is also considered “isolated” if it contains any modifications that do not naturally occur to the corresponding nucleic acid in a genome.
  • an endogenous coding sequence is considered “isolated” if it contains an insertion, deletion or a point mutation introduced artificially, e.g., by human intervention.
  • An "isolated nucleic acid” also includes a nucleic acid integrated into a host cell chromosome at a heterologous site and a nucleic acid construct present as an episome.
  • an "isolated nucleic acid” can be substantially free of other cellular material, or substantially free of culture medium when produced by recombinant techniques, or substantially free of chemical precursors or other chemicals when chemically synthesized.
  • inflammation mediators include any compound capable of triggering an inflammatory process.
  • inflammation generally refers to the process of reaction of vascularized living tissue to injury. This process includes but is not limited to increased blood flow, increased vascular permeability, and leukocytic exudation.
  • inflammatory mediators include, but are not limited to prostaglandins (e.g. PGE2), leukotrienes (e.g. LTB4), inflammatory cytokines, such as tumour necrosis factor alpha (TNFa), interleukin 1 (IL-1), and interleukin 6 (IL-6); nitric oxide (NO), metalloproteinases, and heat shock proteins.
  • PGE2 prostaglandins
  • LTB4 leukotrienes
  • inflammatory cytokines such as tumour necrosis factor alpha (TNFa), interleukin 1 (IL-1), and interleukin 6 (IL-6); nitric oxide (NO), metalloproteinases, and heat shock proteins.
  • PGE2 prostaglandins
  • LTB4 leukotrienes
  • inflammatory cytokines such as tumour necrosis factor alpha (TNFa), interleukin 1 (IL-1), and interleukin 6 (IL-6); nitric oxide (NO), metall
  • matrix protein includes proteins released from cells to form the extracellular matrix of cartilage and other tissues.
  • the extracellular matrix of cartilage consists of proteoglycans, belonging to several distinct proteoglycan families. These include, but are not limited to, perlecan and the hyalectans, exemplified by aggrecan and versican, and the small leucine-rich family of proteoglycans, including decorin, biglycan and fibromodulin.
  • the extracellular matrix also consists of hybrid collagen fibers comprised of three collagen isotypes, namely type II, type IX, and type XI collagens, along with accessory proteins such as cartilage oligeromeric matrix protein (COMP), link protein, and fibronectin.
  • COMP cartilage oligeromeric matrix protein
  • Cartilage also contains hyaluronin which forms a noncovalent association with the hyalectins.
  • a specialized pericellular matrix surrounds the chondrocyte which consists of proteoglycans, type VI collagen and collagen receptor proteins, such as anchorin.
  • matrix degrading enzymes refers to enzymes able to cleave extracellular matrix proteins. Cartilage extracellular matrix turnover is regulated by matrix metalloproteases (MMPs) which are synthesized as latent proenzymes that require activation in order to degrade cartilage extracellular matrix proteins.
  • MMPs matrix metalloproteases
  • Three classes of enzymes are believed to regulate the turnover of extracellular matrix proteins, namely collagenases (including, but not limited to, MMP-13), responsible for the degradation of native collagen fibers, stromelysins (including, but not limited to, MMP-3) which degrade proteoglycan and type IX collagen, and gelatinases (including, but not limited to, MMP-2 and MMP-9) which degrade denatured collagen.
  • MMP-13 matrix metalloproteases
  • ADAMTS metalloproteinases
  • ADAMTS4 aggrecanase-1
  • ADAMTS-5 aggrecanase-2
  • ADAMTS-5 aggrecanase-2
  • reducing aggrecanase activity refers to a decrease in any and/or all of the activities associated with at least one naturally occurring aggrecanase, including but not limited to ADAMTS4 and ADAMTS5.
  • reducing at least one ADAMTS4 activity refers to a decrease in any and/or all of the activities associated with naturally occurring ADAMTS4.
  • reducing ADAMTS4 in a mammal activity can be measured after administration of at least one polypeptide capable of binding to ADAMTS4 to a subject and compared with ADAMTS4 activity in the same subject prior to the administration of the polypeptide capable of binding to ADAMTS4 or in comparison to a second subject who is administered placebo.
  • reducing at least one ADAMTS4 includes the reduction of at least one or more enzyme activity.
  • a reduction in at least one ADAMTS4 activity includes a complete abrogation of at least one ADAMTS4.
  • Also included within this definition is a reduced amount of at least one enzyme activity. That is, ADAMTS4 may have more than one activity which is maintained the while a second activity of the same enzyme is reduced.
  • diseases associated with cartilage and/or extracellular matrix degradation include, but are not limited to cancer, pain, chronic pain, neuropathic pain, postoperative pain, osteoarthritis, sports injuries, erosive arthritis, rheumatoid arthritis, psoriatic arthritis, Lyme arthritis, juvenile arthritis, ankylosing spondylosis, neuralgia, neuropathies, algesia, nerve injury, ischaemia, neurodegeneration, inflammatory diseases, cartilage degeneration, chronic kidney disease, diseases affecting the larynx, trachea, auditory canal, intervertebral discs, ligaments, tendons, joint capsules or bone
  • co-administration or “co-administering” as used herein refers to administration of two or more compounds to the same patient. Co-administration of such compounds may be simultaneous or at about the same time (e.g., within the same hour) or it may be within several hours or days of one another. For example, a first compound may be administered once weekly while a second compound is co-administered daily.
  • Attenuate refers to a normalization (i.e., either an increase or decrease) of the amount of matrix degrading enzyme, inflammatory mediator, or matrix protein produced and/or released by a cell, following exposure to a catabolic stimulus.
  • matrix degrading enzymes e.g. MMP-13, ADAMTS4
  • reactive oxygen species e.g. NO
  • a “domain antibody” or “dAb” may be considered the same as a “single variable domain” which is capable of binding to an antigen.
  • a single variable domain may be a human antibody variable domain, but also includes single antibody variable domains from other species such as rodent (for example, as disclosed in WO 00/29004), nurse shark and Camelid V HH dAbs.
  • Camelid V HH are immunoglobulin single variable domain
  • V HH domains may be humanized according to standard techniques available in the art, and such domains are considered to be "domain antibodies.”
  • V H includes camelid V HH domains.
  • single variable domain refers to an antigen binding protein variable domain (for example, V H , V HH , V l ) that specifically binds an antigen or epitope independently of a different variable region or domain.
  • antigen binding protein refers to antibodies, antibody fragments and other protein constructs, such as domains, but not limited to, variable domains and domain antibodies, which are capable of binding to an antigen.
  • the antigen binding domain of an antibody comprises two separate regions: a heavy chain variable domain (v H ) and a light chain variable domain (v L : which can be either V K or V ).
  • the antigen binding site itself is formed by six polypeptide loops: three from v H domain (HI, H2 and H3) and three from y L domain (LI, L2 and L3).
  • Bispecific antibodies comprising complementary pairs of v H an d V L regions are known in the art. These bispecific antibodies must comprise two pairs of v H an d V L S > each V I V L P a i r binding to a single antigen or epitope. Methods described involve hybrid hybridomas (Milstein & Cuello AC, Nature 305 :537-40), minibodies (Hu et al, (1996) Cancer Res 56:3055-3061;), diabodies (HoUiger et al, (1993) Proc. Natl. Acad. Sci. USA 90, 6444-6448; WO 94/13804), chelating recombinant antibodies (CRAbs; Neri et al, (1995) J. Mol. Biol.
  • each antibody species comprises two antigen-binding sites, each fashioned by a complementary pair of V H an d V L domains. Each antibody is thereby able to bind to two different antigens or epitopes at the same time, with the binding to EACH antigen or epitope mediated by a V H an d its complementary V L domain.
  • inactive V I V L P arr s can greatly reduce the fraction of bispecific IgG.
  • WO 02/02773 (Abbott Laboratories) describes antibody molecules with "dual specificity.”
  • the antibody molecules referred to are antibodies raised or selected against multiple antigens, such that their specificity spans more than a single antigen.
  • Each complementary V J V L P arr m me antibodies of WO 02/02773 specifies a single binding specificity for two or more structurally related antigens; the V H and v L domains in such complementary pairs do not each possess a separate specificity.
  • the antibodies thus have a broad single specificity which encompasses two antigens, which are structurally related.
  • natural autoantibodies have been described that are polyreactive (Casali & Notkins, Ann. Rev. Immunol.
  • Protein engineering methods have been suggested that may have a bearing on this.
  • a catalytic antibody could be created with a binding activity to a metal ion through one variable domain, and to a hapten (substrate) through contacts with the metal ion and a complementary variable domain (Barbas et al., 1993 Proc. Natl. Acad. Sci USA 90, 6385-6389).
  • the binding and catalysis of the substrate is proposed to require the binding of the metal ion (second antigen).
  • the binding to the V I V L pairing relates to a single but multi- component antigen.
  • Single heavy chain variable domains have also been described, derived from natural antibodies which are normally associated with light chains (from monoclonal antibodies or from repertoires of domains; see EP-A-0368684). These heavy chain variable domains have been shown to interact specifically with one or more related antigens but have not been combined with other heavy or light chain variable domains to create a ligand with a specificity for two or more different antigens. Furthermore, these single domains have been shown to have a very short in vivo half-life. Therefore such domains are of limited therapeutic value.
  • the term "antibody” includes immunoglobulin molecules comprising four polypeptide chains, two heavy (H) chains and two light (L) chains inter- connected by disulfide bonds.
  • Each heavy chain is comprised of a heavy chain variable region (abbreviated herein as V H ) and a heavy chain constant region.
  • the heavy chain constant region is comprised of three domains, CHI, CH2 and CH3.
  • Each light chain is comprised of a light chain variable region (abbreviated herein as V L ) and a light chain constant region.
  • the light chains of antibodies (immunoglobulins) from any vertebrate species can be assigned to one of two clearly distinct types, called kappa ( ⁇ ) and lambda ( ⁇ ), based on the amino acid sequences of their constant domains.
  • the variable regions of kappa light chains are referred to herein as VK.
  • the expression V L is intended to include both the variable regions from kappa-type light chains (VK) and from lambda-type light chains.
  • the light chain constant region is comprised of one domain, CL.
  • the V H and V L regions include regions of hypervariability, termed complementarity determining regions (CDRs), interspersed with regions that are more conserved, termed framework regions (FR).
  • CDRs complementarity determining regions
  • Each V H and V L is composed of three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1 - CDR1 - FR2 - CDR2 - FR3 - CDR3 - FR4.
  • antibodies can be assigned to different classes. There are five major classes of intact antibodies: IgA, IgD, IgE, IgG, and IgM, and several of these may be further divided into subclasses (isotypes), e.g., IgGl, IgG2, IgG3, IgG4, IgA, and IgA2.
  • the heavy-chain constant domains that correspond to the different classes of antibodies are called ⁇ , ⁇ , ⁇ , ⁇ , and ⁇ , respectively.
  • the subunit structures and three-dimensional configurations of different classes of immunoglobulins are well known.
  • the present invention includes antibodies of any of the aforementioned classes or subclasses
  • antibody as used herein is also intended to encompass antibodies, digestion fragments, specified portions and variants thereof, including antibody mimetics or comprising portions of antibodies that mimic the structure and/or function of an antibody or specified fragment or portion thereof, including single chain antibodies and fragments thereof; each containing at least one CDR.
  • Functional fragments include antigen binding fragments that bind to an ADAMTS4 antigen.
  • antibody fragments capable of binding to ADAMTS4 a portion thereof including, but not limited to Fab ⁇ e.g., by papain digestion), facb ⁇ e.g., by plasmin digestion), pFc' ⁇ e.g., by pepsin or plasmin digestion), Fd ⁇ e.g., by pepsin digestion, partial reduction and reaggregation), Fv or scFv ⁇ e.g., by molecular biology techniques) fragments, are encompassed by the present invention.
  • Antibody fragments are also intended to include, e.g., domain deleted antibodies, diabodies, linear antibodies, single-chain antibody molecules, and
  • multispecific antibodies formed from antibody fragments.
  • the term "monoclonal antibody,” as used herein, refers to an antibody obtained from a population of substantially homogeneous antibodies, e.g., the individual antibodies comprising the population are substantially identical except for possible naturally occurring mutations or minor post-translational variations that may be present.
  • Monoclonal antibodies are highly specific, being directed against a single antigenic site. Furthermore, in contrast to conventional (polyclonal) antibody preparations which typically include different antibodies directed against different determinants (epitopes), each monoclonal antibody is directed against a single determinant on the antigen.
  • the modifier "monoclonal” indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method.
  • the monoclonal antibodies of the present invention are preferably made by recombinant DNA methods or are obtained by screening methods as described elsewhere herein.
  • immunoglobulins in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is identical with or homologous to corresponding sequences in antibodies derived from another species (e.g., mouse or rat) or belonging to another antibody class or subclass, as well as fragments of such antibodies, so long as they exhibit the desired biological activity (Morrison et al, Proc. Natl. Acad. Sci. USA 57:6851-6855 (1984)).
  • Chimeric antibodies of interest herein include "primatized" antibodies comprising variable domain antigen- binding sequences derived from a non-human primate ⁇ e.g., Old World Monkey, such as baboon, rhesus or cynomolgus monkey) and human constant region sequences (U.S. Pat. No. 5,693,780).
  • a non-human primate e.g., Old World Monkey, such as baboon, rhesus or cynomolgus monkey
  • human constant region sequences U.S. Pat. No. 5,693,780
  • the present invention includes, for example, chimeric monoclonal antibodies comprising a chimeric heavy chain and/or a chimeric light chain.
  • the chimeric heavy chain may comprise any of the heavy chain variable (V H ) regions described herein or mutants or variants thereof fused to a heavy chain constant region of a non-human or a human antibody.
  • the chimeric light chain may comprise any of the light chain variable (V L ) regions described herein or mutants or variants thereof fused to a light chain constant region of a non-human or a human antibody.
  • human antibody includes antibodies having variable and constant regions corresponding to human germline immunoglobulin sequences as described by Kabat et al. (See Kabat, et al. (1991) Sequences of Proteins of
  • the human antibodies of the invention may include amino acid residues not encoded by human germline immunoglobulin sequences ⁇ e.g. , mutations introduced by random or site-specific mutagenesis in vitro or by somatic mutation in vivo), for example in the CDRs and in particular CDR3.
  • the human antibody can have at least one position replaced with an amino acid residue, e.g., an activity enhancing amino acid residue which is not encoded by the human germline immunoglobulin sequence.
  • the human antibody can have up to twenty positions replaced with amino acid residues which are not part of the human germline
  • immunoglobulin sequence In other embodiments, up to ten, up to five, up to three or up to two positions are replaced.
  • human antibody as used herein, is not intended to include antibodies in which CDR sequences derived from the germline of another mammalian species, such as a mouse, have been grafted onto human framework sequences.
  • recombinant human antibody includes human antibodies that are prepared, expressed, created or isolated by recombinant means, such as antibodies expressed using a recombinant expression vector transfected into a host cell, antibodies isolated from a recombinant, combinatorial human antibody library, antibodies isolated from an animal that is transgenic for human immunoglobulin genes, or antibodies prepared, expressed, created or isolated by any other means that involves splicing of human immunoglobulin gene sequences to other DNA sequences.
  • recombinant human antibodies have variable and constant regions derived from human germline immunoglobulin sequences ⁇ See Kabat, E. A., et al. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S.
  • recombinant human antibodies include human germline immunoglobulin sequence that have been subjected to in vitro mutagenesis (or, when an animal transgenic for human Ig sequences is used, in vivo somatic mutagenesis) and thus the amino acid sequences of the V H and V L regions of the recombinant antibodies are sequences that, while derived from and related to human germline V H and V L sequences, may not naturally exist within the human antibody germline repertoire in vivo. In certain embodiments, however, such recombinant antibodies are the result of selective mutagenesis approach or backmutation or both.
  • the antibodies of the present invention may be isolated antibodies.
  • An "isolated antibody,” as used herein, includes an antibody that is substantially free of other antibodies having different antigenic specificities. Moreover, an isolated antibody may be substantially free of other cellular material and/or chemicals.
  • Intact antibodies include heteromultimeric glycoproteins comprising at least two heavy and two light chains. Aside from IgM, intact antibodies are usually
  • heterotetrameric glycoproteins of approximately 150Kda, composed of two identical light
  • each light chain is linked to a heavy chain by one covalent disulfide bond while the number of disulfide linkages between the heavy chains of different immunoglobulin isotypes varies.
  • Each heavy and light chain also has intrachain disulfide bridges.
  • Each heavy chain has at one end a variable domain (V H ) followed by a number of constant regions.
  • Each light chain has a variable domain (V L ) and a constant region at its other end; the constant region of the light chain is aligned with the first constant region of the heavy chain and the light chain variable domain is aligned with the variable domain of the heavy chain.
  • the light chains of antibodies from most vertebrate species can be assigned to one of two types called Kappa and Lambda based on the amino acid sequence of the constant region.
  • human antibodies can be assigned to five different classes, IgA, IgD, IgE, IgG and IgM.
  • IgG and IgA can be further subdivided into subclasses, IgGl, IgG2, IgG3 and IgG4; and IgAl and IgA2.
  • Species variants exist with mouse and rat having at least IgG2a, IgG2b.
  • variable domain of the antibody confers binding specificity upon the antibody with certain regions displaying particular variability called complementarity determining regions (CDRs).
  • CDRs complementarity determining regions
  • FR Framework regions
  • the variable domains of intact heavy and light chains each comprise four FR connected by three CDRs.
  • the CDRs in each chain are held together in close proximity by the FR regions and with the CDRs from the other chain contribute to the formation of the antigen binding site of antibodies.
  • the constant regions are not directly involved in the binding of the antibody to the antigen but exhibit various effector functions such as participation in antibody dependent cell-mediated cytotoxicity (ADCC), phagocytosis via binding to Fey receptor, half-life/clearance rate via neonatal Fc receptor (FcRn) and complement dependent cytotoxicity via the Clq component of the complement cascade.
  • ADCC antibody dependent cell-mediated cytotoxicity
  • FcRn neonatal Fc receptor
  • complement dependent cytotoxicity via the Clq component of the complement cascade.
  • Human antibodies may be produced by a number of methods known to those of skill in the art. Human antibodies can be made by the hybridoma method using human myeloma or mouse-human heteromyeloma cells lines see Kozbor J.Immunol 133, 3001, (1984) and Brodeu ⁇ Monoclonal Antibody Production Techniques and Applications, pp51-63 (Marcel Dekker Inc, 1987). Alternative methods include the use of phage libraries or transgenic mice both of which utilize human V region repertories (see Winter G, (1994), Annu.Rev.Immunol 12,433-455, Green LL (1999), J.Immunol.methods 231, 11-23).
  • mice Several strains of transgenic mice are now available wherein their mouse immunoglobulin loci has been replaced with human immunoglobulin gene segments (see Tomizuka K, (2000) PNAS 97,722-727; Fishwild D.M (1996) Nature Biotechnol. 14,845- 851, Mendez MJ, 1997, Nature Genetics, 15,146-156). Upon antigen challenge such mice are capable of producing a repertoire of human antibodies from which antibodies of interest can be selected.
  • TrimeraTM system (see Eren R et al, (1998) Immunology 93: 154-161) where human lymphocytes are transplanted into irradiated mice
  • SAM Selected Lymphocyte Antibody System
  • SLAM Selected Lymphocyte Antibody System
  • human (or other species) lymphocytes are effectively put through a massive pooled in vitro antibody generation procedure followed by deconvulated, limiting dilution and selection procedure and the Xenomouse IITM (Abgenix Inc).
  • An alternative approach is available from Morphotek Inc using the MorphodomaTM technology.
  • Phage display technology can be used to produce human antibodies (and fragments thereof), see McCafferty; Nature, 348, 552-553 (1990) and Griffiths AD et al (1994)
  • antibody V domain genes are cloned in frame into either a major or minor coat of protein gene of a filamentous bacteriophage such as M13 or fd and displayed (usually with the aid of a helper phage) as functional antibody fragments on the surface of the phage particle. Selections based on the functional properties of the antibody result in selection of the gene encoding the antibody exhibiting those properties.
  • the phage display technique can be used to select antigen specific antibodies from libraries made from human B cells taken from individuals afflicted with a disease or disorder described above or alternatively from unimmunized human donors (see Marks; J. Mol. Bio. 222,581-597, 1991). Where an intact human antibody is desired comprising a Fc domain it is necessary to redone the phage displayed derived fragment into a mammalian expression vectors comprising the desired constant regions and establishing stable expressing cell lines.
  • affinity maturation may be used to improve binding affinity wherein the affinity of the primary human antibody is improved by sequentially replacing the H and L chain V regions with naturally occurring variants and selecting on the basis of improved binding affinities.
  • Variants of this technique such as "epitope imprinting" are now also available see WO 93/06213. See also Waterhouse; Nucl.Acids Res 21, 2265-2266 (1993).
  • the antigen binding proteins of the present invention have an affinity of at least about 5xl0 4 liter/mole, lxl 0 5 liter/mole, 5xl0 5 liter/mole, or lxl 0 6 liter/mole as measured by an association constant (Ka).
  • the antigen binding proteins of the present invention binds to a neutralizing epitope of human ADAMTS4 with a dissociation constant (Kd) of less than about 5xl0 ⁇ 4 liter/second, lxlO "5 liter/second, 5xl0 ⁇ 5 liter/second, or lxlO "6 liter/second.
  • immunogenicity which is the immune system of the patient may recognize the non-human intact antibody as non-self and mount a neutralizing response. This reaction is particularly evident upon multiple administration of the non-human antibody to a human patient.
  • Various techniques have been developed over the years to overcome these problems and generally involve reducing the composition of non-human amino acid sequences in the intact antibody whilst retaining the relative ease in obtaining non-human antibodies from an immunized animal e.g. mouse, rat or rabbit. Broadly two approaches have been used to achieve this. The first are chimeric antibodies, which generally comprise a non-human (e.g. rodent such as mouse) variable domain fused to a human constant region.
  • Chimeric antibodies are typically produced using recombinant DNA methods.
  • DNA encoding the antibodies e.g. cDNA
  • cDNA DNA encoding the antibodies
  • conventional procedures e.g. by using oligonucleotide probes that are capable of binding specifically to genes encoding the H and L chains of the antibody of the invention.
  • Hybridoma cells serve as a typical source of such DNA. Once isolated, the DNA is placed into expression vectors which are then trans fected into host cells such as E. coli, COS cells, CHO cells or myeloma cells that do not otherwise produce immunoglobulin protein to obtain synthesis of the antibody.
  • the DNA may be modified by substituting the coding sequence for human L and H chains for the corresponding non-human (e.g. murine) H and L constant regions see e.g. Morrison; PNAS 81, 6851 (1984).
  • the second approach involves the generation of humanized antibodies wherein the non-human content of the antibody is reduced by humanizing the variable regions.
  • Humanized antibodies can be made by CDR grafting.
  • CDRs build loops close to the antibody's N-terminus where they form a surface mounted in a scaffold provided by the framework regions.
  • Antigen-binding specificity of the antibody is mainly defined by the topography and by the chemical characteristics of its CDR surface. These features are in turn determined by the conformation of the individual CDRs, by the relative disposition of the CDRs, and by the nature and disposition of the side chains of the residues comprising the CDRs.
  • a large decrease in immunogenicity can be achieved by grafting only the CDRs of a non-human (e.g.
  • human V regions showing the greatest sequence homology to the non-human donor antibody are chosen from a database in order to provide the human framework (FR).
  • the selection of human FRs can be made either from human consensus or individual human antibodies. Where necessary key residues from the donor antibody are substituted into the human acceptor framework to preserve CDR conformations. Computer modeling of the antibody maybe used to help identify such structurally important residues, see W099/48523.
  • humanization may be achieved by a process of "veneering."
  • Veneering A statistical analysis of unique human and murine immunoglobulin heavy and light chain variable regions revealed that the precise patterns of exposed residues are different in human and murine antibodies, and most individual surface positions have a strong preference for a small number of different residues (see Padlan E.A. et al; (1991)
  • the present invention provides isolated antigen binding proteins, comprising at least one first immunoglobulin variable domain capable of binding to an aggrecanase.
  • the aggrecanase is human ADAMTS4.
  • the antigen binding protein is an antibody or fragment thereof.
  • the antibody specifically binds to ADAMTS4.
  • the antibody may be a monoclonal antibody or fragment thereof.
  • the monoclonal antibodies or fragment thereof of the present invention are mouse, chimeric, humanized, or fully human.
  • the antigen binding protein comprises at least one complementarity determining region.
  • the antigen binding protein of the present invention is a monoclonal antibody comprising a heavy chain comprising CDRH1, CDRH2 and CDRH3, wherein the complementarity determining regions (CDRs) of the heavy chain are selected from the group consisting of:
  • CDRH1 having at least about 70, 75, 80, 85, 90, 95, or 98% sequence identity to amino acid sequence GDTFTIYDIN (SEQ ID NO:2);
  • CDRH2 having at least about 70, 75, 80, 85, 90, 95, or 98% sequence identity to amino acid sequence WIYPRDGSTKYNEKFKGKA (SEQ ID NO:3); and CDRH3 having at least about 80%> sequence identity to amino acid sequence GWFVY (SEQ ID NO:4);
  • CDRH1 having at least about 70, 75, 80, 85, 90, 95, or 98% sequence identity to amino acid sequence GFIFTIYDIN (SEQ ID NO: 8);
  • CDRH2 having at least about 70, 75, 80, 85, 90, 95, or 98% sequence identity to amino acid sequence WIYPRDGSTKYNEKFKGKA (SEQ ID NO:3);
  • CDRH3 having at least about 80%> sequence identity to amino acid sequence GWFAY (SEQ ID NO:9); and CDRH1 having at least about 70, 75, 80, 85, 90, 95, or 98% sequence identity to amino acid sequence GYTFTDYEIH (SEQ ID NO: 10);
  • CDRH2 having at least about 70, 75, 80, 85, 90, 95, or 98% sequence identity to amino acid sequence PIDPETGNTAYNQKFKG (SEQ ID NO: 11); and CDRH3 having at least about 70, 75, 80, 85, 90, 95, or 98% sequence identity to amino acid sequence EGLRGHWYFDV (SEQ ID NO: 12); and
  • CDRH1 having at least about 70, 75, 80, 85, 90, 95, or 98% sequence identity to amino acid sequence GFSLTTHGVN (SEQ ID NO: 16);
  • CDRH2 having at least about 70, 75, 80, 85, 90, 95, or 98% sequence identity to amino acid sequence VIWDDGNINYHSGL (SEQ ID NO: 17); and CDRH3 having at least about 70, 75, 80, 85, 90, 95, or 98% sequence identity to amino acid sequence PGSYPYYFDY (SEQ ID NO: 18).
  • the antigen binding protein of the present invention is a monoclonal antibody comprising a light chain comprising CDRLl, CDRL2 and CDRL3, wherein the complementarity determining regions (CDRs) of the light chain are selected from the group consisting of: CDRLl having at least about 70, 75, 80, 85, 90, 95, or 98% sequence identity to amino acid sequence SASSSVSDMH (SEQ ID NO:5);
  • CDRL2 having at least about 70, 75, 80, 85, 90, 95, or 98% sequence identity to amino acid sequence DTSKLAS (SEQ ID NO:6);
  • CDRL3 having at least about 70, 75, 80, 85, 90, 95, or 98% sequence identity to amino acid sequence QQWSSYPFT (SEQ ID NO:7);
  • CDRLl having at least about 70, 75, 80, 85, 90, 95, or 98% sequence identity to amino acid sequence SASLSVTYMY (SEQ ID NO: 13);
  • CDRL2 having at least about 70, 75, 80, 85, 90, 95, or 98% sequence identity to amino acid sequence DTSNLAS (SEQ ID NO: 14);
  • CDRL3 having at least about 70, 75, 80, 85, 90, 95, or 98% sequence identity to amino acid sequence QQWSYYPVT (SEQ ID NO: 15); and CDRLl having at least about 70, 75, 80, 85, 90, 95, or 98% sequence identity to amino acid sequence KASQNVGTAVA (SEQ ID NO: 19);
  • CDRL2 having at least about 70, 75, 80, 85, 90, 95, or 98% sequence identity to amino acid sequence SASNRFT (SEQ ID NO:20);
  • CDRL3 having at least about 70, 75, 80, 85, 90, 95, or 98% sequence identity to amino acid sequence QQYSTYPLT (SEQ ID NO:21).
  • the antigen binding protein comprises at least one complementarity determining region.
  • the antigen binding protein of the present invention is a monoclonal antibody comprising a heavy chain comprising CDRH1, CDRH2 and CDRH3 and a light chain comprising CDRLl, CDRL2 and CDRL3, wherein the complementarity determining regions (CDRs) of the heavy chain are selected from the group consisting of:
  • CDRH1 having at least about 70, 75, 80, 85, 90, 95, or 98% sequence identity to amino acid sequence GDTFTIYDIN (SEQ ID NO:2);
  • CDRH2 having at least about 70, 75, 80, 85, 90, 95, or 98% sequence identity to amino acid sequence WIYPRDGSTKYNEKFKGKA (SEQ ID NO:3); and CDRH3 having at least about 80%> sequence identity to amino acid sequence GWFVY (SEQ ID NO:4); and
  • the complementarity determining regions of the light chain are selected from the group consisting of:
  • CDRLl having at least about 70, 75, 80, 85, 90, 95, or 98% sequence identity to amino acid sequence SASSSVSDMH (SEQ ID NO:5);
  • CDRL2 having at least about 70, 75, 80, 85, 90, 95, or 98% sequence identity to amino acid sequence DTSKLAS (SEQ ID NO:6);
  • CDRL3 having at least about 70, 75, 80, 85, 90, 95, or 98% sequence identity to amino acid sequence QQWSSYPFT (SEQ ID NO:7).
  • CDRs complementarity determining regions
  • CDRH1 having at least about 70, 75, 80, 85, 90, 95, or 98% sequence identity to amino acid sequence GFIFTIYDIN (SEQ ID NO: 8); CDRH2 having at least about 70, 75, 80, 85, 90, 95, or 98% sequence identity to amino acid sequence WIYPRDGSTKYNEKFKGKA (SEQ ID NO:3); and CDRH3 having at least about 80% sequence identity to amino acid sequence GWFAY (SEQ ID NO:9); and
  • the complementarity determining regions of the light chain are selected from the group consisting of:
  • CDRL1 having at least about 70, 75, 80, 85, 90, 95, or 98% sequence identity to amino acid sequence SASSSVSDMH (SEQ ID NO:5);
  • CDRL2 having at least about 70, 75, 80, 85, 90, 95, or 98% sequence identity to amino acid sequence DTSKLAS (SEQ ID NO:6);
  • CDRL3 having at least about 70, 75, 80, 85, 90, 95, or 98% sequence identity to amino acid sequence QQWSSYPFT (SEQ ID NO:7).
  • CDRs complementarity determining regions
  • CDRH1 having at least about 70, 75, 80, 85, 90, 95, or 98% sequence identity to amino acid sequence GYTFTDYEIH (SEQ ID NO: 10);
  • CDRH2 having at least about 70, 75, 80, 85, 90, 95, or 98% sequence identity to amino acid sequence PIDPETGNTAYNQKFKG (SEQ ID NO: 11); and CDRH3 having at least about 70, 75, 80, 85, 90, 95, or 98% sequence identity to amino acid sequence EGLRGHWYFDV (SEQ ID NO: 12); and the complementarity determining regions of the light chain are selected from the group consisting of:
  • CDRL1 having at least about 70, 75, 80, 85, 90, 95, or 98% sequence identity to amino acid sequence SASLSVTYMY (SEQ ID NO: 13);
  • CDRL2 having at least about 70, 75, 80, 85, 90, 95, or 98% sequence identity to amino acid sequence DTSNLAS (SEQ ID NO: 14);
  • CDRL3 having at least about 70, 75, 80, 85, 90, 95, or 98% sequence identity to amino acid sequence QQWSYYPVT (SEQ ID NO: 15).
  • complementarity determining regions (CDRs) of the heavy chain are selected from the group consisting of: CDRH1 having at least about 70, 75, 80, 85, 90, 95, or 98% sequence identity to amino acid sequence GFSLTTHGVN (SEQ ID NO: 16);
  • CDRH2 having at least about 70, 75, 80, 85, 90, 95, or 98% sequence identity to amino acid sequence VIWDDGNINYHSGL (SEQ ID NO: 17); and CDRH3 having at least about 70, 75, 80, 85, 90, 95, or 98% sequence identity to amino acid sequence PGSYPYYFDY (SEQ ID NO: 18); and
  • the complementarity determining regions of the light chain are selected from the group consisting of:
  • CDRLl having at least about 70, 75, 80, 85, 90, 95, or 98% sequence identity to amino acid sequence KASQNVGTAVA (SEQ ID NO : 19);
  • CDRL2 having at least about 70, 75, 80, 85, 90, 95, or 98% sequence identity to amino acid sequence SASNRFT (SEQ ID NO:20);
  • CDRL3 having at least about 70, 75, 80, 85, 90, 95, or 98% sequence identity to amino acid sequence QQYSTYPLT (SEQ ID NO:21).
  • the antigen binding protein comprises at least one complementarity determining region.
  • the antigen binding protein of the present invention is a monoclonal antibody comprising a heavy chain comprising CDRHl , CDRH2 and CDRH3 and a light chain comprising CDRLl , CDRL2 and CDRL3, wherein the complementarity determining regions (CDRs) of the heavy chain are selected from the group consisting of:
  • the complementarity determining regions of the light chain are selected from the group consisting of:
  • CDRL2 having 100% sequence identity to amino acid sequence DTSKLAS (SEQ ID NO:6); and CDRL3 having 100% sequence identity to amino acid sequence QQWSSYPFT (SEQ ID NO:7).
  • CDRs complementarity determining regions
  • the complementarity determining regions of the light chain are selected from the group consisting of:
  • CDRL1 having 100% sequence identity to amino acid sequence
  • CDRL2 having 100% sequence identity to amino acid sequence DTSKLAS (SEQ ID NO:6);
  • CDRL3 having 100% sequence identity to amino acid sequence QQWSSYPFT (SEQ ID NO:7).
  • CDRs complementarity determining regions
  • the complementarity determining regions of the light chain are selected from the group consisting of:
  • CDRL1 having 100% sequence identity to amino acid sequence
  • SASLSVTYMY (SEQ ID NO: 13); CDRL2 having 100% sequence identity to amino acid sequence DTSNLAS (SEQ ID NO: 14); and
  • CDRL3 having 100% sequence identity to amino acid sequence
  • CDRs complementarity determining regions
  • GFSLTTHGVN (SEQ ID NO: 16);
  • the complementarity determining regions of the light chain are selected from the group consisting of:
  • CDRL1 having 100% sequence identity to amino acid sequence
  • CDRL2 having 100% sequence identity to amino acid sequence SASNRFT (SEQ ID NO:20).
  • CDRL3 having 100% sequence identity to amino acid sequence QQYSTYPLT (SEQ ID NO:21).
  • any amino acid of any of the aforementioned CDRs is substituted at one position by an amino acid selected from histidine, isoleucine, leucine, lysine, methionine, phenylalanine, threonine, tryptophan, valine, alanine, arginine, aspartic acid, cysteine, cystine, glutamic acid, glutamine, glycine, ornithine, proline, serine, taurine, and tyrosine.
  • an amino acid selected from histidine, isoleucine, leucine, lysine, methionine, phenylalanine, threonine, tryptophan, valine, alanine, arginine, aspartic acid, cysteine, cystine, glutamic acid, glutamine, glycine, ornithine, proline, serine, taurine, and tyrosine.
  • the antigen binding protein is a Fab or F(ab')2 fragment.
  • the first immunoglobulin variable domain is a single chain variable domain.
  • an antibody according to the invention described herein comprising a constant domain region such that the antibody has reduced ADCC and/or complement activation or effector functionality.
  • the constant domain may comprise a naturally disabled constant region of IgG2 or IgG4 isotype or a mutated IgGl constant domain. Examples of suitable modifications are described in EP0307434. One example comprises the substitutions of alanine residues at positions 235 and 237 (EU index numbering).
  • the antigen binding protein or a fragment thereof comprises an antibody V H domain comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 22, 24, 26, and 28.
  • the antigen binding protein or a fragment thereof comprises an antibody V L domain comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 23, 25, and 27.
  • the antigen binding protein or a fragment thereof comprises an antibody V H domain comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 22, 24, 26, and 28 and a V L domain comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 23, 25, and 27.
  • the antigen binding protein or a fragment thereof comprises an antibody V H domain comprising SEQ ID NO: 22 and a V L domain comprising SEQ ID NO: 23.
  • the antigen binding protein or a fragment thereof comprises an antibody V H domain comprising SEQ ID NO: 24 and a V L domain comprising SEQ ID NO: 23.
  • the antigen binding protein or a fragment thereof comprises an antibody V H domain comprising SEQ ID NO: 26 and a V L domain comprising SEQ ID NO: 25.
  • the antigen binding protein or a fragment thereof comprises an antibody V H domain comprising SEQ ID NO: 28 and a V L domain comprising SEQ ID NO: 27.
  • the antigen binding protein or a fragment thereof comprises an antibody heavy chain comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 22, 24, 26, and 28.
  • the antigen binding protein or a fragment thereof comprises an antibody light chain comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 23, 25, and 27. In one embodiment the antigen binding protein or a fragment thereof comprises an antibody heavy chain comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 22, 24, 26, and 28 and a light chain comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 23, 25, and 27.
  • the antigen binding protein or a fragment thereof comprises an antibody heavy chain comprising SEQ ID NO: 22 and a light chain comprising SEQ ID NO: 23.
  • the antigen binding protein or a fragment thereof comprises an antibody heavy chain comprising SEQ ID NO: 24 and a light chain comprising SEQ ID NO: 23.
  • the antigen binding protein or a fragment thereof comprises an antibody heavy chain comprising SEQ ID NO: 26 and a light chain comprising SEQ ID NO: 25.
  • the antigen binding protein or a fragment thereof comprises an antibody heavy chain comprising SEQ ID NO: 28 and a light chain comprising SEQ ID NO: 27.
  • Another aspect of the invention includes an antibody that competes for binding to ADAMTS4 with any one of the antibodies listed in Figure 3. These include the antibodies 4D8.1E1, 7C7.1H1, 7E8.1E3, and 7G4.1D6.
  • the antigen binding protein of the present invention can be characterized by a dissociation constant equal or less than about 9.0 x 10 " 9 M for human ADAMTS4, in some instances it is less than or equal to about 2.5 x 10 " 10 M.
  • Antigen binding protein affinity for a target such as human ADAMTS4 can be measured by surface plasmon resonance such as but not limited to BIACORE® or Octet®.
  • BIAcore kinetic analysis can be used to determine the binding on and off rates of antibodies or fragments thereof to an ADAMTS4 antigen.
  • BIAcore kinetic analysis comprises analyzing the binding and dissociation of an ADAMTS4 antigen from chips with immobilized antibodies or fragments thereof on their surface (see the Example section infra).
  • the present invention also provides antigen binding proteins that block and/or reduce at least one activity ADAMTS4.
  • the antigen binding proteins of the present invention blocks and/or reduces the cleavage of aggrecan by ADAMTS4 at the Glu 373 -Ala 374 cleavage site.
  • the antigen binding proteins of the present invention are capable of penetrating cartilage, even when administered by a non-articular route of administration.
  • the antigen binding proteins of the present invention may be administered intravenously, intramuscularly, intra-articularly, subcutaneously, orally, intranasally, and/or by peritoneal administration.
  • isolated polynucleotides encoding an antigen binding protein of this invention are also provided. Also provided are host cells comprising the polynucleotides encoding the antigen binding proteins of the present invention and methods of expressing the antigen binding proteins form said host cells. In addition, methods are provided for making the antigen binging proteins of the present invention.
  • Methods of making vectors, host cells and antibodies of the present invention include using conventional expression vectors or recombinant plasmids produced by placing coding sequences for the antibody in operative association with conventional regulatory control sequences capable of controlling the replication and expression in, and/or secretion from, a host cell.
  • Regulatory sequences include promoter sequences, e.g., CMV promoter, and signal sequences, which can be derived from other known antibodies.
  • a second expression vector can be produced having a DNA sequence which encodes a complementary antibody light or heavy chain.
  • this second expression vector is identical to the first except insofar as the coding sequences and selectable markers are concerned, so to ensure as far as possible that each polypeptide chain is functionally expressed.
  • the heavy and light chain coding sequences for the altered antibody may reside on a single vector.
  • a selected host cell is co-transfected by conventional techniques with both the first and second vectors (or simply transfected by a single vector) to create the transfected host cell of the invention comprising both the recombinant or synthetic light and heavy chains.
  • the transfected cell is then cultured by conventional techniques to produce the engineered antibody of the invention.
  • the antibody which includes the association of both the recombinant heavy chain and/or light chain is screened from culture by appropriate assay, such as ELISA or RIA. Similar conventional techniques may be employed to construct other altered antibodies and molecules.
  • Suitable vectors for the cloning and subcloning steps employed in the methods and construction of the compositions of this invention may be selected by one of skill in the art.
  • the conventional pUC series of cloning vectors may be used.
  • One vector, pUC19 is commercially available from supply houses, such as Amersham (Buckinghamshire, United Kingdom) or Pharmacia (Uppsala, Sweden).
  • any vector which is capable of replicating readily has an abundance of cloning sites and selectable genes (e.g., antibiotic resistance), and is easily manipulated may be used for cloning.
  • the selection of the cloning vector is not a limiting factor in this invention.
  • the vectors employed for expression of the antibodies may be selected by one of skill in the art from any conventional vector.
  • the vectors also contain selected regulatory sequences (such as CMV or RSV promoters) which direct the replication and expression of heterologous DNA sequences in selected host cells. These vectors contain the above described DNA sequences which code for the antibody or altered
  • the vectors may incorporate the selected immunoglobulin sequences modified by the insertion of desirable restriction sites for ready manipulation.
  • the expression vectors may also be characterized by genes suitable for amplifying expression of the heterologous DNA sequences, e.g., the mammalian dihydrofolate reductase gene (DHFR).
  • DHFR mammalian dihydrofolate reductase gene
  • Other preferable vector sequences include a poly A signal sequence, such as from bovine growth hormone (BGH) and the betaglobin promoter sequence (betaglopro).
  • BGH bovine growth hormone
  • betaglopro betaglobin promoter sequence
  • replicons e.g. replicons, selection genes, enhancers, promoters, signal sequences and the like
  • selection genes e.g. replicons, selection genes, enhancers, promoters, signal sequences and the like
  • Other appropriate expression vectors of which numerous types are known in the art for mammalian, bacterial, insect, yeast, and fungal expression may also be selected for this purpose.
  • the present invention also encompasses a cell line transfected with a recombinant plasmid containing the coding sequences of the antibodies or altered immunoglobulin molecules thereof.
  • Host cells useful for the cloning and other manipulations of these cloning vectors are also conventional. However, most desirably, cells from various strains of E. coli are used for replication of the cloning vectors and other steps in the construction of altered antibodies of this invention.
  • Suitable host cells or cell lines for the expression of the antibody of the invention are preferably mammalian cells such as NS0, Sp2/0, CHO (e.g. DG44), COS, a fibroblast cell (e.g., 3T3), and myeloma cells, and more preferably a CHO or a myeloma cell.
  • Human cells may be used, thus enabling the molecule to be modified with human glycosylation patterns.
  • other eukaryotic cell lines may be employed.
  • the selection of suitable mammalian host cells and methods for transformation, culture, amplification, screening and product production and purification are known in the art. See, e.g., Sambrook et al, cited above.
  • Bacterial cells may prove useful as host cells suitable for the expression of the recombinant Fabs of the present invention (see, e.g., Pluckthun, A., Immunol. Rev., 130: 151-188 (1992)).
  • any recombinant Fab produced in a bacterial cell would have to be screened for retention of antigen binding ability. If the molecule expressed by the bacterial cell was produced in a properly folded form, that bacterial cell would be a desirable host.
  • various strains of E. coli used for expression are well-known as host cells in the field of biotechnology.
  • Various strains of B. subtilis, Streptomyces, other bacilli and the like may also be employed in this method.
  • strains of yeast cells known to those skilled in the art are also available as host cells, as well as insect cells, e.g. Drosophila and Lepidoptera and viral expression systems. See, e.g. Miller et al., Genetic Engineering, 8:277-298, Plenum Press (1986) and references cited therein.
  • the transfection methods required to produce the host cells of the invention, and culture methods necessary to produce the antibody of the invention from such host cell are all conventional techniques.
  • the culture method of the present invention is a serum-free culture method, usually by culturing cells serum-free in suspension.
  • the antibodies of the invention may be purified from the cell culture contents according to standard procedures of the art, including ammonium sulfate precipitation, affinity columns, column chromatography, gel electrophoresis and the like. Such techniques are within the skill of the art and do not limit this invention. For example, preparation of altered antibodies are described in WO 99/58679 and WO 96/16990.
  • Yet another method of expression of the antibodies may utilize expression in a transgenic animal, such as described in U. S. Patent No. 4,873,316.
  • This relates to an expression system using the animal's casein promoter which when transgenically incorporated into a mammal permits the female to produce the desired recombinant protein in its milk.
  • a method of producing an antibody of the invention comprises the step of culturing a host cell transformed or transfected with a vector encoding the light and/or heavy chain of the antibody of the invention and recovering the antibody thereby produced.
  • the present invention provides a method of inhibiting ADAM and ADAMTS activity by providing a molecule that simultaneously binds both catalytic and disintegrin domains.
  • the ADAMTS is ADAMTS 4.
  • ADAMTS4 and the ADAMTS4 mAb 7E8.1E3 suggest simultaneous antibody/antigen interactions between both the catalytic and disintegrin domains of ADAMTS4.
  • the catalytic and disintegrin domains of ADAM and ADAMTS proteases are separated by a hinge region that imparts flexibility between the domains which may act to regulate function or allow for substrate localization to the catalytic site.
  • the high affinity mAb binding observed at this domain spanning epitope likely 'locks' the catalytic and disintegrin domains of ADAMTS4 together thereby neutralizing enzymatic activity.
  • the molecule is an antibody that binds to both the disintegrin and catalytic domains simultaneously.
  • the molecule is an antibody or antibody fragment of the present invention.
  • the present invention concerns an antibody which neutralizes the enzymatic activity of AMAMTS4, and in which the antibody simultaneously binds to catalytic and disintegrin domains with a KD of less than about lxl 0 "9 or 2x10 ⁇ 10 as measured by BiaCore or Octet QK.
  • compositions comprising at least one of the antigen binding proteins described herein.
  • the current invention also provides use of at least one antigen binding protein to ADAMTS4 in the manufacture of a medicament for reducing at least one ADAMTS4 activity in a human.
  • the present invention provides use of at least one antigen binding protein to ADAMTS4 for reducing at least one activity of ADAMTS4in a human comprising administering to a patient in need thereof a composition comprising at least one antigen binding protein to ADAMTS4.
  • compositions of the present invention may further comprise a second antigen binding protein.
  • the second antigen binding protein may be a monoclonal antibody.
  • the second monoclonal antibody binds at least one antigen selected from the group consisting of ADAMTS4, ADAMTS5, NGF, OSM, TNF-a, IL-6, VIP, TRPV1, TRPV4, ADAMTS1, Aggrecan, Collagen II, RANKL, and/or IL-1.
  • the pharmaceutical compositions of the present invention may comprise a first antigen binding protein, which may be a monoclonal antibody to ADAMTS4 and a second monoclonal antibody, which may also bind
  • a pharmaceutical composition of the present invention may comprise a first antigen binding protein, which is a monoclonal antibody that binds to ADAMTS4 and a second antigen binding protein, which is a monoclonal antibody that binds one of the following: ADAMTS5, NGF, OSM, TNF-a, IL-6, VIP, TRPV1, TRPV4, ADAMTS1, Aggrecan, Collagen II, RANKL, and/or IL-1.
  • a first antigen binding protein which is a monoclonal antibody that binds to ADAMTS4
  • a second antigen binding protein which is a monoclonal antibody that binds one of the following: ADAMTS5, NGF, OSM, TNF-a, IL-6, VIP, TRPV1, TRPV4, ADAMTS1, Aggrecan, Collagen II, RANKL, and/or IL-1.
  • the patient is suffering from a disease of the cartilage.
  • a patient may be suffering from one or more diseases chosen from the group consisting of: cancer, pain, chronic pain, neuropathic pain, postoperative pain, osteoarthritis, sports injuries, erosive arthritis, rheumatoid arthritis, psoriatic arthritis, Lyme arthritis, juvenile arthritis, ankylosing spondylosis, neuralgia, neuropathies, algesia, nerve injury, ischaemia, neurodegeneration, inflammatory diseases, cartilage degeneration, chronic kidney disease, diseases affecting the larynx, trachea, auditory canal, intervertebral discs, ligaments, tendons, joint capsules or bone development, invertebral disc degeneration, osteopenia, or periodontal diseases, acute joint injury, and/or a disease related to joint destruction.
  • the patient is suffering from osteoarthritis.
  • administering at least one dose of said pharmaceutical composition reduces cartilage degradation in said patient. In another embodiment, administering at least one dose of said pharmaceutical composition inhibits and/or reduces aggrecan cleavage in said patient.
  • compositions capable of treating disease associated with cartilage degradation or alleviating the symptoms produced thereby and formulated for the methods and uses described herein.
  • the present invention provides an
  • ADAMTS4 antibody for use in the treatment of diseases of the cartilage, for
  • Antigen-binding proteins of the present invention can be co-administered with other therapeutics in the same dose or separately.
  • the present invention also provides ADAMTS4 antibodies or fragments thereof for all of the methods and uses described herein.
  • patient refers to a human or other animal.
  • treatment means: (1) the amelioration of the condition being treated or one or more of the biological manifestations of the condition being treated, (2) the interference with (a) one or more points in the biological cascade that leads to or is responsible for the condition being treated or (b) one or more of the biological
  • prevention is not an absolute term. In medicine, “prevention” is understood to refer to the prophylactic administration of a drug to substantially diminish the likelihood or severity of a condition or biological manifestation thereof, or to delay the onset of such condition or biological manifestation thereof.
  • effective amount means an amount of at least one antigen binding protein sufficient to significantly induce a positive modification in the condition to be treated but low enough to avoid serious side effects (at a reasonable benefit/risk ratio) within the scope of sound medical judgment.
  • a safe and effective amount of at least one antigen binding protein of the invention will vary with the particular compound chosen (e.g.
  • the antigen binding proteins of the invention may be administered by any suitable route of administration, including both systemic administration and topical administration.
  • Systemic administration includes oral administration, parenteral administration, transdermal administration, rectal administration, and administration by inhalation.
  • Parenteral administration refers to routes of administration other than enteral, transdermal, or by inhalation, and is typically by injection or infusion.
  • Parenteral administration includes intravenous, intramuscular, and subcutaneous injection or infusion, including intraarticular administration.
  • Inhalation refers to administration into the patient's lungs whether inhaled through the mouth or through the nasal passages.
  • Topical administration includes application to the skin as well as intraocular, otic, intravaginal, and intranasal administration.
  • the antigen binding proteins of the invention may be administered once or according to a dosing regimen wherein a number of doses are administered at varying intervals of time for a given period of time. For example, doses may be administered one, two, three, or four times per day. Doses may be administered until the desired therapeutic effect is achieved or indefinitely to maintain the desired therapeutic effect. Suitable dosing regimens for a antigen binding protein of the invention depend on the
  • suitable dosing regimens including the duration such regimens are administered, for a compound of the invention depend on the condition being treated, the severity of the condition being treated, the age and physical condition of the patient being treated, the medical history of the patient to be treated, the nature of concurrent therapy, the desired therapeutic effect, and like factors within the knowledge and expertise of the skilled artisan. It will be further understood by such skilled artisans that suitable dosing regimens may require adjustment given an individual patient's response to the dosing regimen or over time as individual patient needs change.
  • the antibody is used to deliver a drug to the cartilage.
  • the invention is a method of delivering a drug to cartilage comprising linking the drug to an antibody of the present invention.
  • Such delivery can be conducted in vitro, ex vivo, or in vivo.
  • the antibody is used to deliver a growth factor to the cartilage which would promote the growth of new cartilage.
  • growth factors include Bone Morphogenic proteins, particularly BMP-7.
  • BMP-7 Bone Morphogenic proteins
  • Human ADAMTS5 and ADAMTS4 proteins were produced in transfected CHO cells and/or BacMam® transduced HEK293 cells and isolated by conventional chromatography methods.
  • ADAMTS5 (truncated, full length, Cat, Cat/dis domains). Immunogenicity was tested on sera from serial bleeds.
  • Splenocytes and lymph nodes were isolated and fused to mouse myeloma cells using a P3X63/Ag8.653-derived fusion partner. Immortalized antibody producing cells were generated. HAT selection was used to deselect unfused myeloma cells.
  • Resulting hybridoma supernatants from active cultures were screened for specific binding and neutralization of recombinant human ADAMTS5 and ADAMTS4. Hits were identified, confirmed and cloned to monoclonality either by limiting dilution or growth in semi-solid media.
  • Monoclonal antibodies with desired characteristics were scaled up in liquid culture and the antibody was purified by standard chromatography methods. Resulting purified antibody clones were then further characterized for binding affinity and functional potency.
  • ADAMTS4 mAbs were characterized for neutralization potency using in vitro aggrecan substrate cleavage assays (Table 1). ADAMTS4 mAbs were characterized for affinity using both Octet QK (Table 1) and BiaCore (comparable, but not shown) technologies. Antibodies were also tested for cross-reactivity to Human ADAMTS1, ADAMTS5, ADAMTS13, by celTRF and Octet QK, all of which were negative (not shown). All mAbs were also assessed for orthologue cross-reactivity by binding and neutralization against mouse, canine, and cynomolgus monkey ADAMTS4 (not shown). Binding was also detected against rat ADAMTS4 (not shown). Table 1: ADAMTS4/ADAMTS5 mAb Comparison
  • Example 2 Based on the characteristics identified in Example 2, four monoclonal antibodies were identified. The variable regions of these antibodies were sequenced and are shown below ( Figure 3). Heavy chain variable regions for mAb designates 4D8.1E1, 7C7.1H1, 7E8.1E3, and 7G4.1D6 are represented by SEQ ID NOs: 22, 24, 26, and 28, respectively. Light chain variable regions for mAb designates 7C7.1H1, 7E8.1E3, and 7G4.1D6, and are represented by SEQ ID NOs: 23, 25, and 27, respectively.
  • Donor human OA cartilage was obtained from knee replacement surgeries.
  • Cartilage was processed from the bone and cut into 3mm diameter discs. Discs were randomized and cultured in 96-well plates. Endogenous disease factors in the tissues were allowed to progress for cartilage degradation ex vivo. Samples were treated with the following: matched control IgG isotype, select anti-ADAMTS 5 antibodies (designated as 7B4.1E11 and 12F4.1H7), select anti-ADAMTS 4 antibodies (designated as 7E8.1E3 and 7C7.1H1), or a known aggrecanase/MMP inhibitor, shown as GSK571949 (CAS number 329040-94-0) below. Each treatment condition was tested in multiples of 8 on each donor plate. Inhibition of ARGSVIL (SEQ ID NO: 1) neoepitope release was measured for each sample ( Figure 1).
  • ARGSVIL SEQ ID NO: 1
  • cleavage of aggrecan by aggrecanase typically occurs at a conserved region within the interglobular domain of aggrecan. Enzyme cleavage will produce a released fragment containing a neoepitope with an N-terminal amino acid sequence (ARGSVIL) from aggrecan. This cleavage neoepitope can be detected and quantified using a monoclonal antibody which specifically binds to the cleaved forms, but not intact aggrecan.
  • a summary of percent inhibition of ARGSVIL release is shown in Figure 1.
  • Example 5 Monoclonal antibodies penetrate cartilage in vitro and in vivo
  • assessing the ability of a mAb to penetrate human cartilage was performed on ex-vivo tissue using mAbs with multiple specificities, including selected anti-ADAMTS4 mAbs.
  • Full thickness cartilage plugs, spanning synovial surface through sub-chondral bone, from knee replacement surgical specimen were placed in tissue culture for defined durations in the presence of mouse monoclonal antibodies with specificities for human proteins located on the surface of chondrocytes or non-specific isotype controls.
  • tissue culture for defined durations in the presence of mouse monoclonal antibodies with specificities for human proteins located on the surface of chondrocytes or non-specific isotype controls.
  • At the end of each timepoint tissues were processed for full thickness assessment, sectioned and stained using a FITC-labeled anti-mouse detection antibody. Penetration is defined by the depth and intensity of chondrocyte staining within the cartilage tissue.
  • mAb penetration was observed to be a concentration and time-dependent process primarily originating from the synovial surface of the cartilage and proceeding to full thickness penetration within 3-4 days dependent on concentration (not shown). No staining was observed for cartilage plugs treated with isotype control mAbs (not shown).
  • NIR labeled monoclonal antibodies were systemically
  • Example 6 Crystallography structure modeling of antigen-antibody interaction - Implications for epitope and MOA

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Immunology (AREA)
  • Peptides Or Proteins (AREA)

Abstract

La présente invention concerne une protéine de liaison à un antigène isolée qui comporte au moins un premier domaine variable d'immunoglobuline apte à se lier à ADAMTS4 humain. L'invention concerne également des protéines de liaison à un antigène de la présente invention qui sont des anticorps monoclonaux, des compositions pharmaceutiques comportant lesdites protéines de liaison à un antigène, et des méthodes de traitement.
PCT/US2013/022062 2012-01-20 2013-01-18 Anticorps anti-adamts4 et méthodes de traitement WO2013109829A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201261588847P 2012-01-20 2012-01-20
US61/588,847 2012-01-20

Publications (1)

Publication Number Publication Date
WO2013109829A1 true WO2013109829A1 (fr) 2013-07-25

Family

ID=48799668

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2013/022062 WO2013109829A1 (fr) 2012-01-20 2013-01-18 Anticorps anti-adamts4 et méthodes de traitement

Country Status (1)

Country Link
WO (1) WO2013109829A1 (fr)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015056808A1 (fr) 2013-10-15 2015-04-23 Genefrontier Corporation Anticorps humain contre les espèces adamts de type agrécanase pour des agents thérapeutiques contre des maladies associées à l'agrécanase
EP2678027A4 (fr) * 2011-02-24 2015-09-02 Glaxo Group Ltd Procédés d'identification d'une population de patients
WO2018220236A1 (fr) * 2017-06-02 2018-12-06 Merck Patent Gmbh Polypeptides se liant à adamts5, mmp13 et à l'aggrécane
RU2786659C2 (ru) * 2017-06-02 2022-12-23 Мерк Патент Гмбх Полипептиды, связывающиеся с adamts5, mmp13 и аггреканом

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000005391A1 (fr) * 1998-07-21 2000-02-03 Dow Agrosciences Llc Regulation negative de proteines vegetales a mediation par anticorps
US20050281828A1 (en) * 2003-03-04 2005-12-22 Bowdish Katherine S Method of treating autoimmune disease by inducing antigen presentation by tolerance inducing antigen presenting cells
WO2007147213A1 (fr) * 2006-06-22 2007-12-27 Walter And Eliza Hall Institute Of Medical Research Structure de l'ectodomaine du récepteur de l'insuline
US20080008719A1 (en) * 2004-07-10 2008-01-10 Bowdish Katherine S Methods and compositions for the treatment of prostate cancer
US20090017039A1 (en) * 2005-07-08 2009-01-15 Biogen Ides Ma Inc SP35 Antibodies and uses thereof
US20100254997A1 (en) * 2007-06-22 2010-10-07 Genera Istrazivanja D.O.O. ADAMTS4 as a blood biomarker and therapeutic target for chronic renal failure
WO2011002968A2 (fr) * 2009-07-02 2011-01-06 Glaxo Group Limited Polypeptides et procédé de traitement
WO2011139985A1 (fr) * 2010-05-03 2011-11-10 Genentech, Inc. Compositions et méthodes de diagnostic et de traitement d'une tumeur

Patent Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000005391A1 (fr) * 1998-07-21 2000-02-03 Dow Agrosciences Llc Regulation negative de proteines vegetales a mediation par anticorps
US20050281828A1 (en) * 2003-03-04 2005-12-22 Bowdish Katherine S Method of treating autoimmune disease by inducing antigen presentation by tolerance inducing antigen presenting cells
US20080008719A1 (en) * 2004-07-10 2008-01-10 Bowdish Katherine S Methods and compositions for the treatment of prostate cancer
US20090017039A1 (en) * 2005-07-08 2009-01-15 Biogen Ides Ma Inc SP35 Antibodies and uses thereof
WO2007147213A1 (fr) * 2006-06-22 2007-12-27 Walter And Eliza Hall Institute Of Medical Research Structure de l'ectodomaine du récepteur de l'insuline
US20100267927A1 (en) * 2006-06-22 2010-10-21 Garrett Thomas P J Structure of the Insulin Receptor Ectodomain
US20100254997A1 (en) * 2007-06-22 2010-10-07 Genera Istrazivanja D.O.O. ADAMTS4 as a blood biomarker and therapeutic target for chronic renal failure
WO2011002968A2 (fr) * 2009-07-02 2011-01-06 Glaxo Group Limited Polypeptides et procédé de traitement
WO2011139985A1 (fr) * 2010-05-03 2011-11-10 Genentech, Inc. Compositions et méthodes de diagnostic et de traitement d'une tumeur

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2678027A4 (fr) * 2011-02-24 2015-09-02 Glaxo Group Ltd Procédés d'identification d'une population de patients
WO2015056808A1 (fr) 2013-10-15 2015-04-23 Genefrontier Corporation Anticorps humain contre les espèces adamts de type agrécanase pour des agents thérapeutiques contre des maladies associées à l'agrécanase
CN105849128A (zh) * 2013-10-15 2016-08-10 基因先端领域株式会社 用于聚集蛋白聚糖酶相关疾病治疗的针对聚集蛋白聚糖酶型adamts种类的人抗体
JP2016538839A (ja) * 2013-10-15 2016-12-15 ジーンフロンティア株式会社 アグリカナーゼ関連疾患の治療のためのアグリカナーゼ型adamts種に対するヒト抗体
EP3057992A4 (fr) * 2013-10-15 2017-05-31 GeneFrontier Corporation Anticorps humain contre les espèces adamts de type agrécanase pour des agents thérapeutiques contre des maladies associées à l'agrécanase
CN105849128B (zh) * 2013-10-15 2019-10-11 基因先端领域株式会社 用于聚集蛋白聚糖酶相关疾病治疗的针对聚集蛋白聚糖酶型adamts种类的人抗体
AU2014335251B2 (en) * 2013-10-15 2020-04-30 Genefrontier Corporation Human antibody against aggrecanase-type ADAMTS species for therapeutics of aggrecanase-related diseases
US10640573B2 (en) 2013-10-15 2020-05-05 Genefrontier Corporation Human antibody against aggrecanase-type ADAMTS species for therapeutics of aggrecanase-related diseases
WO2018220236A1 (fr) * 2017-06-02 2018-12-06 Merck Patent Gmbh Polypeptides se liant à adamts5, mmp13 et à l'aggrécane
RU2786659C2 (ru) * 2017-06-02 2022-12-23 Мерк Патент Гмбх Полипептиды, связывающиеся с adamts5, mmp13 и аггреканом
US11813307B2 (en) 2017-06-02 2023-11-14 Merck Patent Gmbh Polypeptides binding ADAMTS5, MMP13 and aggrecan

Similar Documents

Publication Publication Date Title
US20120095193A1 (en) Polypeptides and method of treatment
US11820815B2 (en) Human antibodies to human delta like ligand 4
JP4851944B2 (ja) 新規抗il13抗体およびその使用
KR102693806B1 (ko) 항-c10orf54 항체들 및 그들의 용도들
TWI516501B (zh) Pcsk9拮抗劑類
CN111909273B (zh) 人源化的抗-cxcr5抗体、其衍生物及它们的应用
RU2493167C2 (ru) АНТИТЕЛА-АНТАГОНИСТЫ ПРОТИВ Notch3 И ИХ ПРИМЕНЕНИЕ ДЛЯ ПРОФИЛАКТИКИ И ЛЕЧЕНИЯ СВЯЗАННЫХ С Notch3 ЗАБОЛЕВАНИЙ
KR20100118606A (ko) Rgm a 단백질에 대한 모노클로날 항체 및 이의 용도
JP2021510064A (ja) 細胞傷害誘導治療剤
AU2018361957B2 (en) Anti-ApoC3 antibodies and methods of use thereof
WO2013109829A1 (fr) Anticorps anti-adamts4 et méthodes de traitement
RU2781074C2 (ru) АНТИ-ApoC3 АНТИТЕЛА И СПОСОБЫ ИХ ПРИМЕНЕНИЯ

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: 13738508

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: 13738508

Country of ref document: EP

Kind code of ref document: A1