WO2016030488A1 - Treatment of celiac disease - Google Patents

Treatment of celiac disease Download PDF

Info

Publication number
WO2016030488A1
WO2016030488A1 PCT/EP2015/069704 EP2015069704W WO2016030488A1 WO 2016030488 A1 WO2016030488 A1 WO 2016030488A1 EP 2015069704 W EP2015069704 W EP 2015069704W WO 2016030488 A1 WO2016030488 A1 WO 2016030488A1
Authority
WO
WIPO (PCT)
Prior art keywords
kir3dl2
antibody
cells
agent
polypeptide
Prior art date
Application number
PCT/EP2015/069704
Other languages
French (fr)
Inventor
Cécile BONNAFOUS
Hélène Sicard
Renaud Buffet
Olivier Hermine
Original Assignee
Innate Pharma
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 Innate Pharma filed Critical Innate Pharma
Publication of WO2016030488A1 publication Critical patent/WO2016030488A1/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
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/34Identification of a linear epitope shorter than 20 amino acid residues or of a conformational epitope defined by amino acid residues
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • C07K2317/732Antibody-dependent cellular cytotoxicity [ADCC]

Definitions

  • This invention relates to the use of KIR3DL2-targeting agents for the diagnosis and treatment of celiac disease.
  • Celiac disease is an immune-based reaction to dietary gluten (storage protein for wheat, barley, and rye) that primarily affects the small intestine in those with a genetic predisposition and resolves with exclusion of gluten from the diet.
  • the presence of repeated motifs rich in glutamine and proline make gluten proteins a good substrate for tissue transglutaminases (TTG). Due to its deamidation properties, TTG introduces negative charges into the gluten peptides, thus increasing their affinity for the peptide pocket of HLA-DQ2/DQ8 molecules and promoting the formation of complexes that are recognized by CD4+ T lymphocytes.
  • the peptides in genetically predisposed individuals expressing HLA-DQ2/DQ8, thus activate CD4+ T cells which leads to an inflammatory process involving intraepithelial lymphocytes in the intestinal epithelium.
  • Celiac disease is usually detected by serologic testing of celiac-specific antibodies, notably to transglutaminase or gliadin. The diagnosis is confirmed by duodenal mucosal biopsies.
  • the treatment for celiac disease is primarily a gluten-free diet (GFD), which can be difficult to maintain on a lifelong basis, and involves significant monitoring and follow-up. Also, due to the nature and variety of the symptoms, many patients remain undiagnosed. Fur- thermore, even when patients are able to maintain a GFD over the long term, many patients have non-responsive disease, particularly in those diagnosed in adulthood.
  • GFD gluten-free diet
  • Type I is characterized by persisting villous atrophy with an increased number of intraepithelial lymphocytes (lELs) bearing a normal phenotype and polyclonal repertoire.
  • Type II is characterized by a clonal expansion of lELs lacking surface CD3 and generally CD8, and having intracellular CD3e.
  • Non-controlled CD such as RCD leads to complications including bone and autoimmune disease, and to lymphomas, notably enteropathy-associated T lymphoma (EATL).
  • RCD is often considered to be a low-grade intraepithelial T cell lymphoma, an intermediate between celiac disease and high grade invasive T cell lymphoma.
  • RCDII is generally considered a poor prognosis because it is frequently associated with over lymphoma such as EATL for which median survival is only 7 months.
  • KIR3DL2 is expressed on the surface of celiac disease (coeliac disease; CD), including celiac disease and advanced or refractory celiac disease stages. KIR3DL2 is also expressed on enteropathy-associated T-cell lym- phoma (EATL), a complication of CD. In KIR3DL2-positive celiac disease, membranar KIR3DL2 expression permits targeting with KIR3DL2-binding antibodies (e.g. as assessed by immunohistochemistry).
  • CD celiac disease
  • EATL enteropathy-associated T-cell lym- phoma
  • KIR3DL2 is expressed on few other tissues (only on a small fraction of healthy NK and T cells), permitting KIR3DL2 to serve as a marker and target for the detection and treatment of celiac disease, particularly refractory celiac disease. Accordingly, in one embodiment, provided is a method for treating or preventing a celiac disease or a complication thereof (e.g. a lymphoma, an EATL) in an individual, the method comprising administering to an individual a therapeutically active amount of a compound that binds a KIR3DL2 polypeptide.
  • the individual can be an individual having or suspected of having CD or who is susceptible to having CD.
  • the individual can be an individual having or suspected of having CD and who has a poor clinical prognosis, e.g. who has a high likelihood to progress to RCD, RCDI, or RCDII, or to have a complication such as EATL, or who has progressed to RCD, RCDI or RCDII .
  • the individual can be an individual having or suspected of having CD or who is susceptible to having CD.
  • a compound that binds a KIR3DL2 polypeptide for use in the treatment or prevention of celiac disease.
  • a method for treating an individual having a refractory, progressing or advanced celiac disease e.g.
  • the method comprising administering to an individual a therapeutically active amount of a compound that binds a KIR3DL2 polypeptide.
  • the compound that binds a KIR3DL2 polypeptide is capable of depleting a cell that expresses KIR3DL2 at its surface, e.g. a celiac disease cell that expresses KIR3DL2 on its surface (e.g., an intraepithelial lymphocyte).
  • the compound is a depleting anti-KIR3DL2 antibody.
  • a compound that binds a KIR3DL2 polypeptide and depletes KIR3DL2-expressing cells for use in the treatment or prevention of a celiac disease in an individual.
  • the treatment or prevention comprises admin- istration of the compound that binds a KIR3DL2 polypeptide to an individual having a celiac disease.
  • the individual has a refractory, progressing or advanced celiac disease, optionally wherein the celiac disease is an RCD, an RCDI, and RCDII, or a complication thereof, e.g. an enteropathy associated T cell lymphoma (EATL).
  • EATL enteropathy associated T cell lymphoma
  • the treatment or prevention of a celiac disease in an individual comprises:
  • celiac disease e.g. CD, RCD, RCDI, or RCDII
  • a complication thereof e.g. a lymphoma, an EATL
  • the celiac disease is RCDI. In one embodiment, the celiac disease is RCDII. In one embodiment, the celiac disease is RCDII, and the step of determining whether the individual has celiac disease comprises detecting the presence of abnormal lELs, optionally further wherein the abnormal lELs are abnormal lELs expressing KIR3DL2 at their surface.
  • the treatment or prevention of a celiac disease in an individual comprises:
  • a) determining the KIR3DL2 polypeptide status of cells e.g., intraepithelial lymphocytes, abnormal intraepithelial lymphocytes
  • cells e.g., intraepithelial lymphocytes, abnormal intraepithelial lymphocytes
  • a celiac disease e.g. CD, RCD, RCDI, or RCDII
  • a complication thereof e.g. a lymphoma, an EATL
  • b) upon a determination that the individual has KIR3DL2 polypeptides expressed on the surface of cells e.g., intraepithelial lymphocytes, abnormal intraepithelial lymphocytes
  • Step (b) can optionally comprise administering to the individual said compound that binds a KIR3DL2 polypeptide upon a determination that the individual has KIR3DL2 polypeptides that are prominently expressed on the surface of cells. Expression across a high percentage of ab- normal intraepithelial lymphocytes may occur with clonal expansion, e.g. in RCDII, permitting the disease to be particularly suitable to treatment with an anti-KIR3DL2 agent.
  • antibodies are provided that are particularly effective in diagnostic or prognostic assays to detect KIR3DL2 expression on abnormal lymphocytes, notably lELs, and notably in immunohistochemistry assays.
  • the antibodies are capable of detecting mem- branar KIR3DL2 in cases where prior antibodies were not able to detect such KIR3DL2 expression, including with specificity over KIR3DL1 .
  • a method comprising a KIR3DL2 detection step to identify patients having KIR3DL2+ celiac disease; these patients can thereafter be treated with a KIR3DL2-binding agent.
  • KIR3DL2 detection step permits KIR3DL2 therapy to be more precisely directed to patients without reliance on disease staging.
  • Such method also helps permit the prevention of advanced celiac disease (e.g. prevention of progression of celiac disease to an advanced stage, e.g. RCD or EATL) because patients can be treated as KIR3DL2 appears.
  • patients with celiac disease can be KIR3DL2-positive despite having a disease that has been shown to be - CD30-negative (the abnormal lELs do not express CD30 on their surface).
  • a CD30-negative celiac disease e.g. a RCD
  • methods of treating a CD30-negative celiac disease comprising administering a compound that binds a KIR3DL2 polypeptide to a patient having CD30-negative celiac disease.
  • the meth- ods or uses comprise administering a compound that binds a KIR3DL2 polypeptide to an individual having a celiac disease who is refractive to treatment, e.g. with a gluten free diet (GFT), with an immunomodulatory or immunosuppressive agent (e.g. an agent that inhibits the IL-15 signalling pathway, an anti-IL-15 antibody, a protein kinase inhibitor, an agent that inhibits JAK3 signalling), with a chemotherapeutic agent, and/or with a hematopoetic stem cell transplantation.
  • GFT gluten free diet
  • an immunomodulatory or immunosuppressive agent e.g. an agent that inhibits the IL-15 signalling pathway, an anti-IL-15 antibody, a protein kinase inhibitor, an agent that inhibits JAK3 signalling
  • chemotherapeutic agent e.g. an agent that inhibits the IL-15 signalling pathway, an anti-IL-15 antibody, a protein kinase inhibitor
  • a celiac disease a compound that binds a KIR3DL2 polypeptide can be administered in combination with a second treatment for CD (e.g., a gluten free diet (GFT), an immunomodulatory or immunosuppressive agent (e.g. an agent that inhibits the IL-15 signalling pathway, an anti-IL-15 antibody, a protein kinase inhibitor, an agent that inhibits JAK3 signalling), a chemotherapeutic agent, and/or a hematopoet- ic stem cell transplantation).
  • a second treatment for CD e.g., a gluten free diet (GFT), an immunomodulatory or immunosuppressive agent (e.g. an agent that inhibits the IL-15 signalling pathway, an anti-IL-15 antibody, a protein kinase inhibitor, an agent that inhibits JAK3 signalling), a chemotherapeutic agent, and/or a hematopoet- ic stem cell transplantation).
  • GFT gluten free diet
  • a method for detecting a celiac disease in an individual comprising detecting a KIR3DL2 nucleic acid or polypeptide in a biological sample (e.g. on a cell) from the individual (e.g. an individual having CD, suspected of having CD, having a symptom associated with CD, etc.).
  • a method for detecting a progressing, advanced, and/or refractory celiac disease in an individual comprising detecting a KIR3DL2 nucleic acid or polypeptide in a biological sample (e.g. on a cell) from the individual.
  • a determination that a biological sample expresses KIR3DL2 indicates that the patient has a celiac disease (or progressing, advanced, and/or refractory celiac disease).
  • a CD is an RCD, optionally an RCDI, optional- ly and RCDII.
  • a determination that a biological sample expresses KIR3DL2 can indicate that the patient is at risk of developing or has developed EATL, e.g. the patient has a celiac disease that is likely to give rise to/progress to EATL.
  • the method comprises determining the level of expression of a KIR3DL2 nucleic acid or polypeptide in a biological sample and comparing the level to a reference level (e.g.
  • detecting a KIR3DL2 polypeptide in a biological sample comprises detecting KIR3DL2 polypeptide expressed on the surface of an infiltrating lymphocyte (e.g., an I EL).
  • an infiltrating lymphocyte e.g., an I EL
  • a method comprising: (a) determining whether an individual has a celiac disease (e.g. CD, RCD, RCDI, RCDII); and (b) if the individual has a celiac disease, determining whether an individual has celiac disease cells (e.g., intraepithelial lymphocytes) that express a KIR3DL2 polypeptide.
  • the method may optionally further com- prise treating the individual with a therapeutically active amount of a compound that binds a KIR3DL2 polypeptide if the individual has celiac disease cells that express KIR3DL2 on their surface.
  • the celiac disease is RCD or RCDI.
  • the celiac disease is RCDII.
  • determining whether an individual has celiac disease cells that express a KIR3DL2 polypeptide comprises obtaining a biological sample from the individual that comprises celiac disease cells, bringing said cells into contact with an antibody that binds a KIR3DL2 polypeptide, and detecting whether cells express KIR3DL2 on their surface.
  • determining whether an individual has celiac disease cells that express KIR3DL2 comprises conducting an immunohistochemistry assay, e.g. an immunohistochemistry assay comprising obtaining from an individual a biological sample that comprises disease cells (e.g. abnormal lELs), fixing and sectioning said sample to obtain a tissue section, bringing said tissue section into contact with an antibody (e.g. an antibody that competes with antibody 12B1 1 for binding to a human KIR3DL2 polypeptide) and detecting expression of KIR3DL2 (e.g. detecting cells that express KIR3DL2).
  • the tissue section is a frozen tissue section.
  • determining whether an individual has celiac disease cells that express KIR3DL2 comprises conducting a flow cytometry assay. Both IHC and flow cytometry can detect surface expression of KIR3DL2.
  • a method of treating a patient with a celiac disease comprising a) determining the KIR3DL2 polypeptide status of disease cells (e.g. celiac disease cells; abnormal lymphocytes) within the patient, e.g. determining whether a KIR3DL2 polypeptide is prominently expressed on the surface of said disease cells, and b) administering a compound to the patient that specifically binds to a KIR3DL2 polypeptide that is prominently expressed in said disease cells (e.g. prominently expressed on the surface of disease cells).
  • determining the KIR3DL2 polypeptide status comprises determining whether a KIR3DL2 polypeptide that is prominently expressed on the surface of said malignant cells.
  • determining whether a KIR3DL2 polypeptide that is prominently expressed on the surface of said disease cells comprises obtaining from the individual a biological sample that comprises celiac disease cells (e.g., abnormal lymphocytes), bringing said cells into contact with an antibody that binds a KIR3DL2 polypeptide, and detecting cells that express KIR3DL2 (e.g. determining the number or portion of cells that express KIR3DL2).
  • celiac disease cells e.g., abnormal lymphocytes
  • the compound that binds a KIR3DL2 polypeptide is a compound that causes the death of a KIR3DL2-expressing cell.
  • the compound that binds a KIR3DL2 polypeptide is an antigen binding polypeptide, optionally an antibody (e.g. mono- clonal antibody), that binds a KIR3DL2 polypeptide, optionally a polypeptide or other compound that is a natural ligand of KIR3DL2 (e.g. an HLA polypeptide, or fragment or derivative thereof).
  • the antibody is a depleting polypeptide (antibody).
  • the antibody is an antibody that directs ADCC and/or CDC toward a KIR3DL2-expressing cell.
  • the antibody is an antibody that delivers a cytotoxic agent (e.g. small molecule) to a KIR3DL2-expressing cell.
  • the antibody used in any embodiment herein binds a KIR3DL2 polypeptide, optionally, wherein the antibody does not substantially bind to a KIR3DL1 polypeptide and has bivalent binding affinity (K D ) for a human KIR3DL2 polypeptide at of less than 10 "8 M.
  • the antibody binds a KIR3DL2 polypeptide in its D1 domain.
  • the antibody binds a KIR3DL2 polypeptide, wherein said antibody does not substantially bind to a KIR3DL1 polypeptide, and wherein said antibody binds to at least one residue in the segment corresponding to residues 99-192 of the mature KIR3DL2 polypeptide of SEQ ID NO: 1.
  • the antibody used herein competes for binding to a KIR3DL2 polypeptide with an antibody selected from the group consisting of: (a) an antibody having respectively a VH and VL region of SEQ ID NOS: 5 and 6
  • the antibody binds an epitope comprising residues P179 and/or residue S181 of the KIR3DL2 polypeptide of SEQ ID NO: 1 , and/or has reduced binding to a KIR3DL2 polypeptide having a mutation at residues P179 and/or residue S181 of SEQ ID NO: 1 , compared to a wild-type KIR3DL2 polypeptide of SEQ ID NO: 1 .
  • the antibody used herein binds an epitope comprising residues N99, H100, E130, H131 , F132, V178, H180, P182, Y183 and/or Q184 of SEQ ID NO: 1 , and/or has reduced binding to a KIR3DL2 polypeptide having a mutation at residues N99, H100, E130, H131 , F132, V178, H180, P182, Y183 and/or Q184 of SEQ ID NO: 1 , compared to a wild-type Kl R3DL2 polypeptide of SEQ I D NO: 1 .
  • the antibody binds an epitope comprising residues I60 and/or residue G62 of the KIR3DL2 polypeptide of SEQ ID NO: 1 , and/or has reduced binding to a KIR3DL2 polypeptide having a mutation at residues I60 and/or residue G62 of SEQ ID NO: 1 , compared to a wild-type KIR3DL2 polypeptide of SEQ ID NO: 1.
  • the antibody binds an epitope comprising residues P14, S15 and/or residue H23 of the KIR3DL2 polypeptide of SEQ ID NO: 1 , and/or has reduced binding to a KIR3DL2 polypeptide having a mutation at residues P14, S15 and/or residue H23 of SEQ ID NO: 1 , compared to a wild-type KIR3DL2 polypeptide of SEQ ID NO: 1.
  • the antibody used herein binds an epitope comprising resi- dues l60, G62, P14, S15 and/or residue H23 of SEQ ID NO: 1 .
  • the compound that binds a KIR3DL2 polypeptide is administered between once daily and once per month.
  • the composition is administered as monotherapy.
  • the composition is administered in combination with a second therapeutic agent.
  • the composition is administered in combination with an anti-celiac dis- ease treatment or agent.
  • a method of producing a composition for the treatment of celiac disease or for use in the prevention of celiac disease in a mammalian subject comprising the steps of: a) providing a plurality of test compositions; b) testing each compound for the ability to bind KIR3DL2 and/or cause the depletion of KIR3DL2-expressing cells; and c) selecting a com- pound which binds a KIR3DL2 polypeptide and/or causes the depletion of KIR3DL2- expressing cells as suitable for the treatment of celiac disease or for use in the prevention of celiac disease.
  • the method further comprises producing a quantity of the compound se- lected in step c), and/or formulation a quantity of the compound selected in step c) with a pharmaceutically acceptable excipient.
  • step b) further comprises testing said test composition for the ability to direct ADCC and/or CDC toward a KIR3DL2-expressing cell.
  • a celiac disease is a refractory ce- liac disease (RCD), optionally RCDI, optionally RCDII.
  • a method comprising: (a) determining whether an individual has celiac disease (e.g., CD, RCD, RCDI, or RCDII) or a complication thereof (e.g. a lymphoma, an EATL); and (b) if the individual has celiac disease or a complication thereof, treating the individual with a therapeutically active amount of a compound that binds a KIR3DL2 polypeptide.
  • celiac disease e.g., CD, RCD, RCDI, or RCDII
  • a complication thereof e.g. a lymphoma, an EATL
  • determining whether an individual has a celiac disease is made according to standard medical guidelines. In one embodiment of any of the methods herein, determining whether an individual has a celiac disease (e.g., CD, RCD, RCDI, or RCDII) comprises conducting a small bowel investigation, enteroscopy, small bowel imaging, a laboratory test, immunohistochemistry, molecular analysis, and/or flow cytometry.
  • determining whether an individual has a celiac disease comprises conducting a small bowel investigation, enteroscopy, small bowel imaging, a laboratory test, immunohistochemistry, molecular analysis, and/or flow cytometry.
  • determining whether an individual has a celiac disease comprises identifying a population of abnormal cells or abnormal numbers of cells (e.g., abnormal lELs).
  • said identification is by flow cytometry or immunohistochemistry.
  • the method further comprises sorting or isolating the population of abnormal cells.
  • determining whether an individual has or is susceptible of having a celiac disease comprises detecting a genetic predisposition.
  • a celiac disease e.g., CD, RCD, RCDI, or RCDII
  • a complication thereof e.g. a lymphoma, an EATL
  • determining whether an individual has or is susceptible of having a celiac disease comprises detecting a genetic predisposition.
  • the presence of HLA DQ heterodimers DQ2 and/or DQ8 is assessed, e.g., the HLA DQ2/DQ8 genotype is assessed, wherein presence of HLA DQ heterodimers DQ2 and/or DQ8 (e.g. HLA DQ2/DQ8 genotype) indicates a predisposition to CD.
  • detecting a genetic predisposition comprises contacting nucleic acid isolated from a subject with one or more oligonucleotides, wherein the contacting determines the presence of a genetic marker of predisposition to CD (e.g. CD, RCD, poor prognosis, disease pro- sion, etc.); thereby detecting whether an individual has or is susceptible of having a celiac disease.
  • a genetic marker of predisposition e.g. CD, RCD, poor prognosis, disease pro- sion, etc.
  • the method further comprises a step of assessing, following treatment with a compound that binds a KIR3DL2 polypeptide, whether the individual has an ameliora- tion in celiac disease, e.g., whether the individual has decreased numbers of celiac disease cells (e.g., abnormal lELs).
  • the celiac disease is a refractory celiac disease.
  • the celiac disease is an RCDI.
  • the celiac disease is an RCDII.
  • the individual having disease has a poor prognosis, e.g. for resistance to therapy, for occurrence of a complication such as a lymphoma or EATL, for survival.
  • the celiac disease is progressing disease. .
  • the celiac disease is susceptible of giving rise to a lymphoma, e.g. an EATL.
  • a method for diagnosing or monitoring a celiac disease in an individual comprising obtaining from an individual having (or suspect- ed of having, or susceptible to having) CD a biological sample that comprises cells (e.g., intraepithelial lymphocytes), bringing said cells into contact with an antibody that binds a human KIR3DL2 polypeptide, and detecting cells that express KIR3DL2.
  • the antibody that binds a KIR3DL2 polypeptide is an antibody that binds a human KIR3DL2 polypeptide but does not bind to a human KIR3DL1 polypeptide.
  • the antibody that binds a KIR3DL2 polypeptide competes with antibody 12B1 1 for binding to a human KIR3DL2 polypeptide (e.g. antibody 19H12).
  • a method for determining whether a KIR3DL2 polypeptide is expressed on the surface of a I EL comprising obtaining from an individual a bio- logical sample that comprises lELs, bringing said lELs into contact with an antibody that competes with antibody 12B1 1 for binding to a human KIR3DL2 polypeptide (e.g. antibody 19H12), and detecting lELs that express KIR3DL2.
  • the method further comprises determining whether the lELs express any one or more of: T cell receptor (TCR), CD3e, CD8, CD103 and CD45
  • a method for determining whether a KIR3DL2 polypeptide is expressed on the surface of a cell comprising obtaining from an individual (e.g. having, susceptible to or suspected of having celiac disease) a biological sample (e.g., tissue sample) that comprises cells, fixing and sectioning said sample to obtain a tissue section, bringing said tissue section into contact with an antibody that com- petes with antibody 12B1 1 for binding to a human KIR3DL2 polypeptide (e.g. antibody 19H12), and detecting expression of KIR3DL2 (e.g. detecting cells that express KIR3DL2).
  • the tissue section is a frozen tissue section.
  • antibodies having advantageous uses in diagnostic and prognostic methods for celiac disease and other diseases.
  • Provided is a method com- prising:
  • a method comprising:
  • the present disclosure further concerns a method for diagnosing a disease state mediated by pathogenic KIR3DL2-expressing cells, said method comprising the steps of combining with an ex vivo patient sample (from an individual having, susceptible to or suspected of having celiac disease) a composition comprising a conjugate or complex comprises an antibody that binds specifically to KIR3DL2 expressed on the surface of the pathogenic cells and an imaging agent, and detecting the pathogenic cells that express a receptor for the lig- and using flow cytometry.
  • the present disclosure further concerns a method of determining a prognosis of a celiac disease by detecting cells in an ex vivo patient sample, said method comprising the steps of: (a) combining with an ex vivo patient sample a composition comprising a conjugate or complex comprises an antibody (e.g. antibody 19H12) that binds specifically to KIR3DL2 expressed on the surface of the pathogenic cells and an imaging agent, (b) detecting the pathogenic cells that express a receptor for the ligand using flow cytometry, and (c) determining a prognosis for the celiac disease.
  • a composition comprising a conjugate or complex comprises an antibody (e.g. antibody 19H12) that binds specifically to KIR3DL2 expressed on the surface of the pathogenic cells and an imaging agent, (b) detecting the pathogenic cells that express a receptor for the ligand using flow cytometry, and (c) determining a prognosis for the celiac disease.
  • an antibody
  • the present disclosure further concerns a method for quantitating pathogenic cells from an individual having, susceptible to or suspected of having celiac disease, said method comprising the steps of: (a) combining, with an ex vivo patient sample, a conjugate or complex which comprises (i) an antibody that binds specifically to KIR3DL2 expressed on the surface of the pathogenic cells (e.g. antibody 19H12) and (ii) an imaging agent, and (b) quantitating said pathogenic cells in the ex vivo patient sample using flow cytometry.
  • a conjugate or complex which comprises (i) an antibody that binds specifically to KIR3DL2 expressed on the surface of the pathogenic cells (e.g. antibody 19H12) and (ii) an imaging agent, and (b) quantitating said pathogenic cells in the ex vivo patient sample using flow cytometry.
  • the antibody binds to a KIR3DL2 polypeptide on the surface of cells but not to a KIR3DL1 polypeptide.
  • said pathogenic cells are detected by single photon flow cytometry.
  • said pathogenic cells are detected by multiphoton flow cytometry.
  • the ex vivo patient sample is a patient body fluid.
  • the body fluid is selected from the group consisting of spinal fluid, lymph fluid, urine, mucus, and blood.
  • the pathogenic cells are lELs.
  • the antibody conjugated to an imaging agent is selected from the group consisting of anti-KIR3DL2-fluorescein, anti-KIR3DL2-Oregon Green, anti-KIR3DL2-rhodamine, anti- KIR3DL2- phycoerythrin, anti-KIR3DL2-cys-Texas Red, anti-KIR3DL2-AlexaFluor, and anti- KIR3DL2-Dyl_ight.
  • the imaging agent comprises a chromophore.
  • the chromophore is a fluorescent chromophore.
  • the chromophore comprises a compound selected from the group consisting of fluorescein, Oregon Green, rhodamine, phycoerythrin, Texas Red, DyLight 680, and AlexaFluor 488.
  • the methods further comprise the step of quantitating the pathogenic cells in the ex vivo patient sample.
  • the antibodies optionally bind to each of the KIR3DL2 polypeptides having the amino acid sequence shown in SEQ ID NOS: 1 , 27 and 29 (alleles_ * 002, * 001 and * 007, respectively). In one embodiment, the antibodies bind to each of the KIR3DL2 polypeptides having the amino acid sequence shown in SEQ ID NOS: 27 and 31 (alleles_ * 001 and * 009, respectively). In one embodiment, the antibodies bind to each of the KIR3DL2 polypeptides having the amino acid sequence shown in SEQ ID NOS: 27, 1 , 29 and 31 (alleles_ * 001 , * 002, * 007 and * 009, respectively).
  • the antibodies bind to each of the KIR3DL2 polypeptides having the amino acid sequence shown in SEQ ID NOS: 27, 1 , 2, 28 and 29 (alleles_ * 001 , * 002, * 003, * 005 and * 007, respectively). In one embodiment, the antibodies bind to each of the KIR3DL2 polypeptides having the amino acid sequence shown in SEQ ID NOS: 27, 1 , 29 and 30 (alleles_ * 001 , * 002, * 007 and * 008, respectively).
  • the antibodies bind to each of the KIR3DL2 polypeptides having the amino acid sequence shown in SEQ ID NOS: 27, 1 , 2, 28, 29 and 30 (alleles_ * 001 , * 002, * 003, * 005, * 007 and * 008, respectively).
  • the antibody binds an epitope comprising one, two, three, four, five or more of residues selected from the group consisting of: M128, E130, H131 , R145, V147, Q149, 1150, V178, P179, H180 and S181 (with reference to SEQ ID NO: 1 ), and/or the antibody may or may not have reduced binding to a KIR3DL2 polypeptide having a mutation at a residue selected from the group consisting of: M128, E130, H131 , R145, V147, Q149, 1150, V178, P179, H180 and/ S181 (with reference to SEQ ID NO: 1
  • the antibody binds an epitope comprising residues P179 and/or S181 of the KIR3DL2 polypeptide, and/or has reduced binding to a KIR3DL2 polypeptide having a mutation at residues P179 and/or S181 (with
  • the antibody binds an epitope comprising residues V178 and/or H180 of the KIR3DL2 polypeptide, and/or has reduced binding to a KIR3DL2 polypeptide having a mutation at residues V178 and/or H180 (with reference to SEQ ID NO: 1 , e.g. a V178A, H180S mutant).
  • the antibody binds an epitope comprising residues E130, H131 and/or R145 of the KIR3DL2 polypeptide, and/or have reduced binding to a KIR3DL2 polypeptide having a mutation at residues E130, H131 and/or R145 (with reference to SEQ ID NO: 1 , e.g. a E130S, H131 S, R145S mutant).
  • the antibody is an antibody that competes with, and/or that comprises the heavy and/or light chain CDRs 1 , 2 and/or 3 of, antibody 19H12 or 12B1 1.
  • Figure 1 shows staining of frozen tissue sections from RAJI-KIR3DL2 mouse tumor models and RAJI-KIR3DL2 cell lines, using AZ158 antibody (see WO2010/081890) or antibodies 12B1 1. While AZ158 was negative, tumors were positive when using 12B1 1 antibody at the same concentration (5 g/ml) of antibody.
  • Figure 2 shows staining of frozen tissue sections from cancer patients previously stained with AZ158, re-examined using antibody 12B1 1. Biopsies that had been KIR3DL2- negative with AZ158 were stained with 12B1 1 (i.e. becoming KIR3DL2-positive).
  • Figure 3 shows staining by anti-KIR3DL2 antibody on NK-/T- lymphoma cells, nasal type. The figure additionally shows that the KIR3DL2-positive cells express CD183 (CXCR3), CD56 and CD54 (ICAM).
  • KIR3DL2 polypeptides at the surface of celiac disease cells permits the development of therapeutic agents that are able to directly and specif- ically target pathogenic cells, as well as diagnostic agents that can be used to diagnose and monitor celiac disease.
  • a method for inhibiting celiac disease for delivering a molecule to a celiac disease cell (e.g. a toxic molecule, a detectable marker, etc.), for targeting, identifying or purifying a cell, for depleting, killing or eliminating a cell, for reducing cell proliferation, the method comprising exposing a cell, such as a celiac disease cell (e.g., an IEL, an abnormal IEL) which expresses a KIR3DL2 polypeptide, to a compound that binds a KIR3DL2 polypeptide.
  • a celiac disease cell e.g., an IEL, an abnormal IEL
  • cell proliferation can refer to any aspect of the growth or proliferation of cells, e.g., cell growth, cell division, or any aspect of the cell cycle.
  • the cell may be in cell culture (in vitro) or in a mammal (in vivo), e.g. a mammal suffering from celiac disease.
  • Also provided is a method for inducing the death of a cell or inhibiting the proliferation or activity of a celiac disease cell e.g., an IEL, an abnormal IEL
  • a celiac disease cell e.g., an IEL, an abnormal IEL
  • exposing the cell to an antigen-binding com- pound that binds a KIR3DL2 polypeptide in an amount effective to induce death and/or inhibit the proliferation of the cell.
  • Antibodies specific for KIR3DL2 can be used for a range of purposes for the diagnosis or treatment of celiac disease, including purifying KIR3DL2 or KIR3DL2-expressing cells in patients having celiac disease, suspected of having celiac disease or susceptible to celiac disease, targeting KIR3DL2-expressing cells for destruction in vivo, or specifically labeling/binding KIR3DL2 in vivo, ex vivo, or in vitro, cells in patients having celiac disease, suspected of having celiac disease or susceptible to celiac disease, including in methods such as immunoblotting, IHC analysis, i.e. on frozen biopsies, FACS analysis, and immunoprecipi- tation.
  • a or “an” may mean one or more.
  • the words “a” or “an” when used in conjunction with the word “comprising”, the words “a” or “an” may mean one or more than one.
  • another may mean at least a second or more.
  • treatment of celiac disease or the like is mentioned with reference to anti-KIR3DL2 binding agent (e.g. antibody), there is meant: (a) method of treatment of celiac disease, said method comprising the step of administering (for at least one treatment) an anti-KIR3DL2 binding agent, (e.g., in a pharmaceutically acceptable carrier material) to a warm-blooded animal, especially a human, in need of such treat- ment, in a dose that allows for the treatment of celiac disease, (a therapeutically effective amount), e.g, in a dose (amount) as specified hereinabove and herein below; (b) the use of an anti-KIR3DL2 binding agent for the treatment of celiac disease, or an anti-KIR3DL2 binding agent, for use in said treatment (especially in a human); (c) the use of an anti-KIR3DL2 binding agent for the manufacture of a pharmaceutical preparation for the treatment of celiac disease, a
  • biopsy as used herein is defined as removal of a tissue for the purpose of examination, such as to establish diagnosis.
  • types of biopsies include by application of suction, such as through a needle attached to a syringe; by instrumental removal of a fragment of tissue; by removal with appropriate instruments through an endoscope; by surgical excision, such as of the whole lesion; and the like.
  • antibody refers to polyclonal and monoclonal antibodies. Depending on the type of constant domain in the heavy chains, antibodies are assigned to one of five major classes: IgA, IgD, IgE, IgG, and IgM. Several of these are further divided into subclasses or isotypes, such as lgG1 , lgG2, lgG3, lgG4, and the like.
  • An exemplary im- munoglobulin (antibody) structural unit comprises a tetramer. Each tetramer is composed of two identical pairs of polypeptide chains, each pair having one "light" (about 25 kDa) and one "heavy" chain (about 50-70 kDa).
  • each chain defines a variable region of about 100 to 1 10 or more amino acids that is primarily responsible for antigen recognition.
  • the terms variable light chain (V L ) and variable heavy chain (V H ) refer to these light and heavy chains respectively.
  • the heavy-chain constant domains that correspond to the different classes of immunoglobulins are termed "alpha,” “delta,” “epsilon,” “gamma” and “mu,” respectively.
  • the subunit structures and three-dimensional configurations of different classes of immunoglobulins are well known.
  • IgG are the exemplary classes of antibodies employed herein because they are the most common antibodies in the physiological situation and be- cause they are most easily made in a laboratory setting.
  • an antibody is a monoclonal antibody.
  • Antibodies also includes any fragment or derivative of any of the herein described antibodies.
  • the term “specifically binds to” means that an antibody can bind in a competitive binding assay to the binding partner, e.g. KIR3DL2, as assessed using either recombinant forms of the proteins, epitopes therein, or native proteins present on the surface of isolated target cells.
  • a competitive binding assay to the binding partner, e.g. KIR3DL2, as assessed using either recombinant forms of the proteins, epitopes therein, or native proteins present on the surface of isolated target cells.
  • an antibody When an antibody is said to "compete with” a particular monoclonal antibody, it means that the antibody competes with the monoclonal antibody in a binding assay using either recombinant KIR3DL2 molecules or surface expressed KIR3DL2 molecules. For example, if a test antibody reduces the binding of AZ158, 19H12, 2B12 or 12B1 1 to a KIR3DL2 polypeptide or KIR3DL2-expressing cell in a binding assay, the antibody is said to "compete” respectively with AZ158, 19H12, 2B12 or 12B1 1.
  • affinity means the strength of the binding of an antibody to an epitope.
  • the affinity of an antibody is given by the dissociation constant Kd, defined as [Ab] x [Ag] / [Ab-Ag], where [Ab-Ag] is the molar concentration of the antibody-antigen complex, [Ab] is the molar concentration of the unbound antibody and [Ag] is the molar concentration of the unbound antigen.
  • Kd dissociation constant
  • a “determinant” designates a site of interaction or binding on a polypeptide.
  • epitope refers to an antigenic determinant, and is the area or region on an antigen to which an antibody binds.
  • a protein epitope may comprise amino acid residues directly involved in the binding as well as amino acid residues which are effectively blocked by the specific antigen binding antibody or peptide, i.e., amino acid residues within the "footprint” of the antibody. It is the simplest form or smallest structural area on a complex antigen molecule that can combine with e.g., an antibody or a receptor.
  • Epitopes can be linear or conformational/structural.
  • linear epitope is defined as an epitope composed of amino acid residues that are contiguous on the linear sequence of amino acids (primary structure).
  • conformational or structural epitope is defined as an epitope composed of amino acid residues that are not all contiguous and thus represent separated parts of the linear sequence of amino acids that are brought into proximity to one another by folding of the molecule (secondary, tertiary and/or quaternary structures).
  • a conformational epitope is dependent on the 3-dimensional structure.
  • 'conformational' is therefore often used interchangeably with 'structural'.
  • KIR3DL2-expressing cells means a process, method, or compound that can kill, eliminate, lyse or induce such killing, elimination or lysis, so as to negatively affect the number of KIR3DL2-expressing cells present in a sample or in a subject.
  • immunoconjugate refers to an antibody that is conjugated to another moiety (e.g. any non-antibody moiety, a therapeutic agent or a label).
  • agent is used herein to denote a chemical compound, a mixture of chem- ical compounds, a biological macromolecule, or an extract made from biological materials.
  • therapeutic agent refers to an agent that has biological activity.
  • cytotoxic agent encompass any compound that can slow down, halt, or reverse the proliferation of cells, decrease their activity in any detectable way, or directly or indirectly kill them. Cytotoxic agents can cause cell death primarily by interfering directly with the cell's functioning, and include, but are not limited to, alkylating agents, tumor necrosis factor inhibitors, DNA intercalators, microtubule inhibitors, kinase inhibitors, proteasome inhibitors and topoisomerase inhibitors.
  • a "toxic payload” as used herein refers to a sufficient amount of cytotoxic agent which, when delivered to a cell results in cell death.
  • Delivery of a toxic payload may be accomplished by administration of a sufficient amount of immunoconjugate comprising an antibody or antigen binding fragment and a cytotoxic agent. Delivery of a toxic payload may also be accomplished by administration of a sufficient amount of an immunoconjugate comprising a cytotoxic agent, wherein the immunoconjugate comprises a secondary antibody or antigen binding fragment thereof which recognizes and binds an antibody or antigen binding fragment.
  • human-suitable refers to any antibody, deri- vatized antibody, or antibody fragment that can be safely used in humans for, e.g. the therapeutic methods described herein.
  • Human-suitable antibodies include all types of humanized, chimeric, or fully human antibodies, or any antibodies in which at least a portion of the antibodies is derived from humans or otherwise modified so as to avoid the immune response that is generally provoked when native non-human antibodies are used.
  • a “humanized” or “human” antibody refers to an antibody in which the constant and variable framework region of one or more human immunoglobulins is fused with the binding region, e.g. the CDR, of an animal immunoglobulin.
  • Such antibodies are designed to maintain the binding specificity of the non-human antibody from which the binding regions are de- rived, but to avoid an immune reaction against the non-human antibody.
  • Such antibodies can be obtained from transgenic mice or other animals that have been "engineered” to produce specific human antibodies in response to antigenic challenge (see, e.g., Green et al. (1994) Nature Genet 7:13; Lonberg et al. (1994) Nature 368:856; Taylor et al.
  • a fully human an- tibody also can be constructed by genetic or chromosomal transfection methods, as well as phage display technology, all of which are known in the art (see, e.g., McCafferty et al. (1990) Nature 348:552-553).
  • Human antibodies may also be generated by in vitro activated B cells (see, e.g., U.S. Pat. Nos. 5,567,610 and 5,229,275, which are incorporated in their entirety by reference).
  • a “chimeric antibody” is an antibody molecule in which (a) the constant region, or a portion thereof, is altered, replaced or exchanged so that the antigen binding site (variable region) is linked to a constant region of a different or altered class, effector function and/or species, or an entirely different molecule which confers new properties to the chimeric antibody, e.g., an enzyme, toxin, hormone, growth factor, drug, etc.; or (b) the variable region, or a portion thereof, is altered, replaced or exchanged with a variable region having a different or altered antigen specificity.
  • Fc domain refers to a C-terminal fragment of an antibody heavy chain, e.g., from about amino acid (aa) 230 to about aa 450 of human ⁇ (gamma) heavy chain or its counterpart sequence in other types of antibody heavy chains (e.g., ⁇ , ⁇ , ⁇ and ⁇ for human antibodies), or a naturally occurring allotype thereof.
  • aa amino acid
  • gamma human ⁇
  • ⁇ and ⁇ for human antibodies e.g., ⁇ , ⁇ and ⁇ for human antibodies
  • the commonly accepted Kabat amino acid numbering for immunoglobulins is used throughout this disclosure (see Kabat et al. (1991 ) Sequences of Protein of Immunological Interest, 5th ed., United States Public Health Service, National Institute of Health, Bethesda, MD).
  • ADCC antibody-dependent cell-mediated cytotoxicity
  • NK natural killer
  • isolated refers to material that is substantially or essentially free from components which normally accompany it as found in its native state. Purity and homogeneity are typically determined using analytical chemistry techniques such as polyacrylamide gel electrophoresis or high performance liquid chromatography. A protein that is the predominant species present in a preparation is substantially purified.
  • polypeptide peptide
  • protein protein
  • amino acid polymers in which one or more amino acid residue is an artificial chemical mimetic of a corresponding naturally occurring amino acid, as well as to naturally occurring amino acid polymers and non-naturally occurring amino acid polymer.
  • recombinant when used with reference, e.g., to a cell, or nucleic acid, protein, or vector, indicates that the cell, nucleic acid, protein or vector, has been modified by the introduction of a heterologous nucleic acid or protein or the alteration of a native nucleic acid or protein, or that the cell is derived from a cell so modified.
  • recombinant cells express genes that are not found within the native (nonrecombinant) form of the cell or express native genes that are otherwise abnormally expressed, under expressed or not expressed at all.
  • Prominently expressed when referring to a KIR3DL2 polypeptide, means that the KIR3DL2 polypeptide is expressed in a substantial number of target cells (e.g. celiac disease cells, abnormal lELs) taken from a given patient. While the definition of the term “prominently expressed” is not bound by a precise percentage value, in most cases a receptor said to be “prominently expressed” will be present on at least 20%, 30%, 40%, 50°%, 60%, 70%, 80%, or more of the target cells (e.g. celiac disease cells, abnormal lELs) taken from a patient.
  • target cells e.g. celiac disease cells, abnormal lELs
  • antibody that "binds" a polypeptide or epitope designates an antibody that binds said determinant with specificity and/or affinity.
  • identity refers to the degree of sequence relatedness between polypeptides, as determined by the number of matches between strings of two or more amino acid residues. "Identity” measures the percent of identical matches between the smaller of two or more sequences with gap alignments (if any) addressed by a particular mathematical model or computer program (i.e., "algorithms”). Identity of related polypeptides can be readily calculated by known methods. Such methods include, but are not limited to, those described in Computational Molecular Biology, Lesk, A. M., ed., Oxford University Press, New York, 1988; Biocomputing: Informatics and Genome Projects, Smith, D.
  • Methods for determining identity are designed to give the largest match between the sequences tested. Methods of determining identity are described in publicly available com- puter programs. Computer program methods for determining identity between two sequences include the GCG program package, including GAP (Devereux et al., Nucl. Acid. Res. 12, 387 (1984); Genetics Computer Group, University of Wisconsin, Madison, Wis.), BLASTP, BLASTN, and FASTA (Altschul et al., J. Mol. Biol. 215, 403-410 (1990)). The BLASTX pro- gram is publicly available from the National Center for Biotechnology Information (NCBI) and other sources (BLAST Manual, Altschul et al. NCB/NLM/NIH Bethesda, Md. 20894; Altschul et al., supra). The well-known Smith Waterman algorithm may also be used to determine identity. Production of antibodies
  • KIR3DL2 (CD158k) is a disulphide-linked homodimer of three-lg domain molecules of about 140 kD, described in Pende et al. (1996) J. Exp. Med. 184: 505-518, the disclosure of which is incorporated herein by reference.
  • KIR3DL2 polypeptides Several allelic variants have been reported for KIR3DL2 polypeptides, each of these are encompassed by the term KIR3DL2.
  • the amino acid sequence of the mature human KIR3DL2 (allele * 002) is shown in SEQ ID NO: 1 , below, corresponding to Genbank accession no. AAB52520 in which the 21 amino acid residue leader sequence has been omitted.
  • KIR3DL2 (allele * 002) is shown in Genbank accession no. U30272.
  • the amino acid sequence of a human KIR3DL2 allele * 003 is shown below, corresponding to Genbank accession no. AAB36593:
  • nucleic acid or protein sequences sharing one or more biological properties or functions with wild type, full length KIR3DL2 respectively, and sharing at least 70%, 80%, 90%, 95%, 96%, 97%, 98%, 99%, or higher nucleotide or amino acid identity.
  • KIR3DL1 is a monomeric molecule of about 70 kD, described in Colonna and Samaridis (1995) Science 268 (5209), 405-408.
  • the cDNA encoding a KIR3DL1 (CD158e2) polypeptide (allele * 00101 ) is shown in Genbank accession no. L41269; the encoded amino acid sequence is shown in Genbank accession no. AAA69870.
  • a KIR3DL1 polypeptide referred to herein is allele * 00101.
  • Examples of antibodies that bind human KIR3DL2 include antibody AZ158, antibody 19H12, antibody 2B12 and antibody 12B1 1 . Further antibodies are provided in PCT/EP2013/069302 and PCT/EP2013/069293, both filed 17 September 2013, the disclosures of which antibodies are incorporated herein by reference.
  • AZ158 binds human KIR3DL2 as well as human KIR3DL1 and KIR3DS1 polypeptides; 19H12, 2B12 and 12B1 1 bind selectively to KIR3DL2 and do not bind KIR3DL1 (or KIR3DS1 ).
  • antibody AZ158 can be used, for example, as therapeutic agent administered to an individual for the elimina- tion of a KIR3DL2 expressing target, e.g. by induction of ADCC and/or CDC
  • antibody 12B1 1 and 19H12 will be advantageous over AZ158 for use in detection (e.g. in vitro assays) of KIR3DL2 expression on the surface of cells because 12B1 1 and 19H12 are both able to detect KIR3DL2-positive cells in detection assays, 12B1 1 is advantageous for immunohisto- chemistry assays using frozen tissue sections, while 19H12 is advantageous for flow cytome- try detection.
  • Each of 2B12, 19H12 and 12B1 1 are also suitable for use as therapeutic agent administered to an individual for the elimination of a KIR3DL2-expressing target cells.
  • 19H12 and 12B1 1 as well as other antibodies 15C1 1 , 19H12, 22B2, 18B10 and 13H1 disclosed in PCT/EP2013/069293 are capable of being internalized into cells via KIR3DL2 and can be used advantageously as an antibody-drug conjugate.
  • 2B12 and other antibodies such as an- tibodies 10F6, 18C6, 9E10, 10G5, 13H1 , 5H1 , 1 E2, 1 C3 and 20E9 disclosed in PCT/EP2013/069302 do no induce any KIR3DL2 internalization, thereby providing advantageous use when effector cell mediated activity is sought, e.g. for depleting antibodies that induce ADCC.
  • an antibody that binds essentially the same epitope or determinant as any of monoclonal antibodies AZ158, 19B12, 12B1 1 or 2B12; op- tionally the antibody comprises an antigen binding region of antibody AZ158, 19B12, 12B1 1 or 2B12.
  • antibody AZ158, 19B12, 12B1 1 or 2B12 can be characterized by its amino acid sequence and/or nucleic acid sequence encoding it.
  • the monoclonal antibody comprises the Fab or F(ab') 2 portion of AZ158, 19B12, 12B1 1 or 2B12.
  • a monoclonal antibody that comprises the heavy chain variable region of AZ158, 19B12, 12B1 1 or 2B12.
  • the monoclonal antibody comprises the three CDRs of the heavy chain variable region of AZ158, 19B12, 12B1 1 or 2B12.
  • a monoclonal antibody that further comprises the variable light chain variable region of AZ158, 19B12, 12B1 1 or 2B12 or one, two or three of the CDRs of the light chain variable region of AZ158, 19B12, 12B1 1 or 2B12.
  • any one or more of said light or heavy chain CDRs may contain one, two, three, four or five or more amino acid modifications (e.g. substitutions, insertions or deletions).
  • an antibody where any of the light and/or heavy chain variable regions comprising part or all of an antigen binding region of antibody AZ158, 19B12, 12B1 1 or 2B12 are fused to an im- munoglobulin constant region of the human IgG type, optionally a human constant region, optionally a human lgG1 or lgG3 isotype.
  • AZ158 binds human KIR3DL2 as well as human KIR3DL1 polypeptides, can be char- acterized as having the heavy and light chain variable regions or heavy and light chain region CDRs of SEQ ID NOS: 8 and 10, respectively, of PCT patent publication no. WO2010/081890).
  • the VH of AZ158 is shown below, with CDRs 1 , 2 and 3 underlined, respectively:
  • VL of AZ158 is shown below with CDRs 1 , 2 and 3 underlined, respectively: DIQMTQSPSS LSASLGGKVT ITCKASQDIN KYIAWYQHKP GKGPRLLIHY TSTLQPGIPS RFSGSGSGRD YSFSISNLEP EDITTYYCLQ YDNLWTFGGG TKLEIK
  • the anti-KIR3DL2 antibodies may include antibodies having variable region or CDR sequences from such AZ158 antibodies (e.g. a heavy and/or light chain variable region fused to a human constant region; a heavy chain variable region fused to a human lgG1 heavy chain constant region); alternatively, the anti-KIR3DL2 antibodies may be an antibody other than the antibodies having variable region or CDR sequences from a AZ158 antibody.
  • AZ158 antibodies e.g. a heavy and/or light chain variable region fused to a human constant region; a heavy chain variable region fused to a human lgG1 heavy chain constant region
  • the anti-KIR3DL2 antibodies may be an antibody other than the antibodies having variable region or CDR sequences from a AZ158 antibody.
  • amino acid sequence of the heavy chain variable region of antibody 19H12 is listed below:
  • amino acid sequence of the light chain variable region of antibody 19H12 is listed below:
  • a purified polypeptide which encodes an antibody comprising: a HCDR1 region comprising an amino acid sequence GYTFT- NFGMN as set forth in SEQ ID NO:9, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof (e.g., NFGMN (SEQ ID NO: 7), GYTFTN (SEQ ID NO: 8)), wherein one or more of these amino acids may be substituted by a different amino acid; a HCDR2 region comprising an amino acid sequence WINTYTGEPTYADDF as set forth in SEQ ID NO: 10, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof (e.g.
  • WINTYTGE (SEQ ID NO: 1 1 )), wherein one or more of these amino acids may be substituted by a different amino acid; a HCDR3 region comprising an amino acid sequence NGNFGYYFDY as set forth in SEQ ID NO: 12, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof, wherein one or more of these amino acids may be substituted by a different amino acid; a LCDR1 region comprising an amino acid sequence RSSQNIVHSNGNTYLE as set forth in SEQ ID NO: 13, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof, wherein one or more of these amino acids may be substituted by a different amino acid; a LCDR2 region comprising an amino acid sequence KVSNRFS as set forth in SEQ ID NO: 14, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof, wherein one or more of these amino acids may be substituted by a different amino acid; and/or a
  • an antibody that binds human KIR3DL2 comprising: (a) the heavy chain variable region of SEQ ID NO:5, wherein one, two, three or more amino acid residues may be substituted by a different amino acid; and/or
  • the heavy chain variable region which is at least 60%, 70%, 80%, 85%, 90% or 95% identical to the variable region having an amino acid sequence of SEQ ID NO:5, wherein one, two, three or more amino acid residues may be substituted by a different amino acid; and/or
  • the light chain variable region which is at least 60%, 70%, 80%, 85%, 90% or 95% identical to the variable region having an amino acid sequence of SEQ ID NO:6, where- in one, two, three or more amino acid residues may be substituted by a different amino acid.
  • amino acid sequence of the heavy chain variable region of antibody 12B1 1 is listed below:
  • amino acid sequence of the light chain variable region of antibody 12B1 1 is listed below:
  • a purified polypeptide which encodes an antibody where- in the antibody comprises: a HCDR1 region comprising an amino acid sequence GYTFT- NYGMN as set forth in SEQ ID NO: 20, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof (e.g. NYGMN (SEQ ID NO: 18), GYTFTN (SEQ ID NO: 19)), wherein one or more of these amino acids may be substituted by a different amino acid; a HCDR2 region comprising an amino acid sequence WINTYTGEPTYADDFKG as set forth in SEQ ID NO: 21 , or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof (e.g.
  • WINTYTGEPT (SEQ ID NO: 22)), wherein one or more of these amino acids may be substituted by a different amino acid; a HCDR3 region comprising an amino acid sequence GPWLAY as set forth in SEQ ID NO: 23, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof, wherein one or more of these amino acids may be substituted by a different amino acid; a LCDR1 region comprising an amino acid sequence KASQDINVYLS as set forth in SEQ ID NO: 24, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof, wherein one or more of these amino acids may be substituted by a different amino acid; a LCDR2 region comprising an amino acid sequence RAIRLVD as set forth in SEQ ID NO: 25, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous ami- no acids thereof, wherein one or more of these amino acids may be substituted by a different amino acid; a LCDR3 region compris
  • an antibody that binds human KIR3DL2 comprising:
  • the heavy chain variable region which is at least 60%, 70%, 80%, 85%, 90% or 95% identical to the variable region having an amino acid sequence of SEQ ID NO: 16, wherein one, two, three or more amino acid residues may be substituted by a different amino acid; and/or
  • the light chain variable region which is at least 60%, 70%, 80%, 85%, 90% or 95% identical to the variable region having an amino acid sequence of SEQ ID NO: 17, wherein one, two, three or more amino acid residues may be substituted by a different amino acid.
  • amino acid sequence of the heavy chain variable region of antibody 2B12 is listed below (Kabat definition CDRs underlined):
  • the amino acid sequence of the light chain variable region of antibody 2B12 is listed below (CDRs underlined): DIVMTQSH KF MSTS LG DRVSFTC KASQ DVSTAVAW Y Q Q K P G Q S P KL L I YWTSTRHTGVP DRFTGSGSGTDYTLTISSVQAEDLALY YCQQHYSTPWTFGGGTKLEI K
  • a purified polypeptide which encodes an antibody comprising: a HCDR1 region comprising an amino acid sequence GYTFT- TAGMQ as set forth in SEQ ID NO: 36, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof (e.g. GYTFTT (SEQ ID NO: 34), or TAGMQ (SEQ ID NO: 35)), wherein one or more of these amino acids may be substituted by a different amino acid; a HCDR2 region comprising an amino acid sequence WINSHSGVPKYAEDFK as set forth in SEQ ID NO: 37, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof (e.g.
  • WINSHSGVP (SEQ ID NO: 38)), wherein one or more of these amino acids may be substituted by a different amino acid; a HCDR3 region comprising an amino acid sequence GGDEGVMDYW as set forth in SEQ ID NO: 39, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof, wherein one or more of these amino acids may be substituted by a different amino acid; a LCDR1 region comprising an amino acid sequence KASQ DVSTAVA as set forth in SEQ ID NO: 40, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof, wherein one or more of these amino acids may be substituted by a different amino acid; a LCDR2 region comprising an amino acid sequence WTSTRHT as set forth in SEQ ID NO: 41 , or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof, wherein one or more of these amino acids may be substituted by a different amino acid; a LCDR3 region
  • an antibody that binds human KIR3DL2 comprising:
  • the heavy chain variable region which is at least 60%, 70%, 80%, 85%, 90% or 95% identical to the variable region having an amino acid sequence of SEQ ID NO: 32, wherein one, two, three or more amino acid residues may be substituted by a different amino acid; and/or
  • the light chain variable region which is at least 60%, 70%, 80%, 85%, 90% or 95% identical to the variable region having an amino acid sequence of SEQ ID NO: 33, wherein one, two, three or more amino acid residues may be substituted by a different amino acid.
  • any of the CDRs 1 , 2 and 3 of the heavy and light chains may be characterized by a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof, and/or as having an amino acid sequence that shares at least 50%, 60%, 70%, 80%, 85%, 90% or 95% sequence identity with the particular CDR or set of CDRs listed in the corresponding SEQ ID NO.
  • an antibody that competes for KIR3DL2 binding with a monoclonal antibody of (a) to (h), for any of the above antibodies.
  • the antibodies that are used bind substantially the same epitope as antibody 19H12 or 12B1 1.
  • the antibodies at least partially overlaps, or includes at least one residue in the segment corresponding to residues 1-192, residues 1 -98, or residues 99-192 of the KIR3DL2 polypeptide of SEQ ID NO: 1 (or a subsequence thereof).
  • all key residues of the epitope is in a segment corresponding to residues 1 -192, residues 1- 98 or residues 99-192 of the KIR3DL2 polypeptide of SEQ ID NO: 1.
  • the antibodies bind an epitope comprising 1 , 2, 3, 4, 5, 6, 7 or more residues in the segment corresponding to residues 1-192, 1 -98 or 99-192 of the KIR3DL2 polypeptide of SEQ ID NO: 1.
  • residues bound by the antibody are present on the surface of the of the KIR3DL2 polypeptide.
  • the antibodies bind an epitope comprising residues P179 and/or residue S181 of SEQ ID NO: 1.
  • the antibodies bind to an epitope comprising 1 , 2, 3, 4, 5, 6 or 7 or more residues selected from the group consisting of: N99, H100, E130, H131 , F132, V178, P179, H180, S181 , P182, Y183 and/or residue Q184 of SEQ ID NO: 1.
  • the antibody binds an epitope comprising residues I60 and/or residue
  • G62 of the KIR3DL2 polypeptide of SEQ ID NO: 1 and/or has reduced binding to a KIR3DL2 polypeptide having a mutation at residues I60 and/or residue G62 of SEQ ID NO: 1 , compared to a wild-type KIR3DL2 polypeptide of SEQ ID NO: 1 .
  • the antibody binds an epitope comprising residues P14, S15 and/or residue H23 of the KIR3DL2 polypeptide of SEQ ID NO: 1 , and/or has reduced binding to a KIR3DL2 polypeptide having a mutation at residues P14, S15 and/or residue H23 of SEQ ID NO: 1 , compared to a wild-type KIR3DL2 polypeptide of SEQ ID NO: 1.
  • the antibody binds an epitope comprising residues I60, G62, P14, S15 and/or residue H23 of SEQ ID NO: 1 .
  • the Examples section herein describes the testing of a series of mutant human KIR3DL2 polypeptides. Binding of anti-KIR3DL2 antibody to cells transfected with the KIR3DL2 mutants was measured and compared to the ability of anti-KIR3DL2 antibody to bind wild-type KIR3DL2 polypeptide (SEQ ID NO:1 ).
  • a reduction in binding between an anti- KIR3DL2 antibody and a mutant KIR3DL2 polypeptide as used herein means that there is a reduction in binding affinity (e.g., as measured by known methods such FACS testing of cells expressing a particular mutant, or by Biacore testing of binding to mutant polypeptides) and/or a reduction in the total binding capacity of the anti-KIR3DL2 antibody (e.g., as evidenced by a decrease in Bmax in a plot of anti-KIR3DL2 antibody concentration versus polypeptide concentration).
  • a significant reduction in binding indicates that the mutated residue is directly involved in binding to the anti-KIR3DL2 antibody or is in close proximity to the bind- ing protein when the anti-KIR3DL2 antibody is bound to KIR3DL2.
  • An antibody epitope will may thus include such residue and may include additional residues spatially adjacent to such residue.
  • a significant reduction in binding means that the binding affinity and/or capacity between an anti-KIR3DL2 antibody and a mutant KIR3DL2 polypeptide is reduced by greater than 40 %, greater than 50 %, greater than 55 %, greater than 60 %, greater than 65 %, greater than 70 %, greater than 75 %, greater than 80 %, greater than 85 %, greater than 90% or greater than 95% relative to binding between the antibody and a wild type KIR3DL2 polypeptide (e.g., the polypeptide shown in SEQ ID NO:1 ). In certain embodiments, binding is reduced below detectable limits.
  • a significant re-duction in binding is evidenced when binding of an anti-KIR3DL2 antibody to a mutant KIR3DL2 polypeptide is less than 50% (e.g., less than 45%, 40%, 35%, 30%, 25%, 20%, 15% or 10%) of the binding observed between the anti-KIR3DL2 antibody and a wild-type KIR3DL2 polypeptide (e.g., the extracellular domain shown in SEQ ID NO:1 ).
  • binding measurements can be made using a variety of binding assays known in the art. A specific example of one such assay is described in the Example section.
  • anti-KIR3DL2 antibodies exhibit significantly lower binding for a mutant KIR3DL2 polypeptide in which a residue in a wild-type KIR3DL2 polypeptide (e.g., SEQ ID NO:1 ) is substituted, e.g. the mutants as described in Example 1 .
  • a mutant KIR3DL2 polypeptide in which a residue in a wild-type KIR3DL2 polypeptide (e.g., SEQ ID NO:1 ) is substituted, e.g. the mutants as described in Example 1 .
  • the format is: Wild type residue: Position in polypeptide: Mutant residue, with the numbering of the residues as indicated in SEQ ID NO: 1.
  • the antibodies have reduced binding to a KIR3DL2 polypeptide having a substitution at residues N99, H100, E130, H131 , F132, V178, P179, H180, S181 , P182, Y183 and/or residue Q184 of SEQ ID NO: 1.
  • an anti-KIR3DL2 antibody binds a wild-type KIR3DL2 poly- peptide having a sequence of SEQ ID NO: 1 but has decreased binding to a mutant KIR3DL2 polypeptide having any one or more (e.g., 1 , 2, 3 or 4) of the following mutations: P179T and/or S181T (with reference to SEQ ID NO:1 ). In one embodiment, binding to the mutant KIR3DL2 is significantly reduced compared to binding to the wild-type KIR3DL2.
  • anti-KIR3DL2 antibodies that exhibit significant- ly lower binding for a mutant KIR3DL2 polypeptide in which a residue in a segment corresponding to residues 1-98, residues 99-292, or residues 99-192 (or a subsequence thereof) in a wild-type KIR3DL2 polypeptide (e.g., SEQ ID NO:1 ) is substituted with a different amino acid.
  • a mutant KIR3DL2 polypeptide in which a residue in a segment corresponding to residues 1-98, residues 99-292, or residues 99-192 (or a subsequence thereof) in a wild-type KIR3DL2 polypeptide (e.g., SEQ ID NO:1 ) is substituted with a different amino acid.
  • an antibody can compete with monoclonal antibody AZ158, 19H12, 2B12 or 12B1 1 and recognizes bind to, or have immunospecificity for substantially or essentially the same, or the same, epitope or "epitopic site" on a KIR3DL2 molecule as monoclonal antibody AZ158, 19H12, 2B12 or 12B1 1 .
  • the monoclonal antibody consists of, or is a derivative or fragment of, antibody AZ158, 19H12, 2B12 or 12B1 1.
  • suitable antibodies can recognize and be raised against any part of the KIR3DL2 polypeptide so long as the antibody binds KIR3DL2 and has the desired functionality.
  • any fragment of KIR3DL2, e.g., human KIR3DL2, or any combination of KIR3DL2 fragments can be used as immunogens to raise antibodies, and the antibodies can recognize epitopes at any location within the KIR3DL2 polypeptide, so long as they can do so on KIR3DL2 expressing NK cells as described herein.
  • the recognized epitopes are present on the cell surface, i.e.
  • the epitope is the epitope specifically recognized by antibody AZ158, 19H12, 2B12 or 12B1 1.
  • antibodies recognizing distinct epitopes within KIR3DL2 can be used in combination, e.g. to bind to KIR3DL2 polypeptides with maximum efficacy and breadth among different individuals.
  • the antibodies may be produced by a variety of techniques known in the art. Typically, they are produced by immunization of a non-human animal, optionally a mouse, with an immunogen comprising a KIR3DL2 polypeptide, optionally a human KIR3DL2 polypeptide.
  • the KIR3DL2 polypeptide may comprise the full length sequence of a human KIR3DL2 polypeptide, or a fragment or derivative thereof, typically an immunogenic fragment, i.e., a portion of the polypeptide comprising an epitope exposed on the surface of cells expressing a KIR3DL2 polypeptide, optionally the epitope recognized by the AZ158, 19H12, 2B12 or 12B1 1 antibody.
  • Such fragments typically contain at least about 7 consecutive amino acids of the mature polypeptide sequence, or at least about 10 consecutive amino acids thereof. Fragments typically are essentially derived from the extra-cellular domain of the receptor.
  • the immunogen comprises a wild-type human KIR3DL2 polypeptide in a lipid membrane, typically at the surface of a cell.
  • the immunogen comprises intact cells, particularly intact human cells, optionally treated or lysed.
  • the polypeptide is a recombinant KIR3DL2 polypeptide.
  • the step of immunizing a non-human mammal with an antigen may be carried out in any manner well known in the art for stimulating the production of antibodies in a mouse (see, for example, E. Harlow and D. Lane, Antibodies: A Laboratory Manual., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY (1988), the entire disclosure of which is herein incorporated by reference).
  • the immunogen is suspended or dissolved in a buffer, optionally with an adjuvant, such as complete or incomplete Freund's adjuvant.
  • an adjuvant such as complete or incomplete Freund's adjuvant.
  • the location and frequency of immunization sufficient to stimulate the pro- duction of antibodies is also well known in the art.
  • the non- human animals are injected intraperitoneal ⁇ with antigen on day 1 and again about a week later.
  • recall injections of the antigen around day 20 optionally with an adjuvant such as incomplete Freund's adjuvant.
  • the recall injections are performed intravenously and may be repeated for several consecutive days.
  • a booster in- jection at day 40 either intravenously or intraperitoneally, typically without adjuvant.
  • This protocol results in the production of antigen-specific antibody-producing B cells after about 40 days.
  • Other protocols may also be used as long as they result in the production of B cells expressing an antibody directed to the antigen used in immunization.
  • serum is obtained from an immunized non- human animal and the antibodies present therein isolated by well-known techniques.
  • the serum may be affinity purified using any of the immunogens set forth above linked to a solid support so as to obtain antibodies that react with KIR3DL2 polypeptides.
  • lymphocytes from a non-immunized non-human mammal are isolated, grown in vitro, and then exposed to the immunogen in cell culture.
  • the lym- phocytes are then harvested and the fusion step described below is carried out.
  • the next step is the isolation of splenocytes from the immunized non-human mammal and the subsequent fusion of those splenocytes with an immortalized cell in order to form an antibody-producing hybridoma.
  • the isolation of splenocytes from a non-human mammal is well-known in the art and typically involves re- moving the spleen from an anesthetized non-human mammal, cutting it into small pieces and squeezing the splenocytes from the splenic capsule through a nylon mesh of a cell strainer into an appropriate buffer so as to produce a single cell suspension.
  • the cells are washed, centrifuged and resuspended in a buffer that lyses any red blood cells.
  • the solution is again centrifuged and remaining lymphocytes in the pellet are finally resuspended in fresh buffer.
  • the lymphocytes can be fused to an immortal cell line.
  • This is typically a mouse myeloma cell line, although many other immortal cell lines useful for creating hybridomas are known in the art.
  • Murine myeloma lines include, but are not limited to, those derived from MOPC-21 and MPC-1 1 mouse tumors available from the Salk Institute Cell Distribution Center, San Diego, U. S. A., X63 Ag8653 and SP-2 cells available from the American Type Culture Collection, Rockville, Maryland U. S. A.
  • the fusion is effected using polyethylene glycol or the like.
  • the resulting hybridomas are then grown in selective media that contains one or more substances that inhibit the growth or survival of the unfused, parental myeloma cells.
  • the culture medium for the hybridomas typically will include hypoxanthine, aminopter- in, and thymidine (HAT medium), which substances prevent the growth of HGPRT-deficient cells.
  • HGPRT hypoxanthine guanine phosphoribosyl transferase
  • Hybridomas are typically grown on a feeder layer of macrophages.
  • the macrophages can be from littermates of the non-human mammal used to isolate splenocytes and are typically primed with incomplete Freund's adjuvant or the like several days before plating the hybridomas. Fusion methods are described in Goding, "Monoclonal Antibodies: Principles and Practice,” pp. 59-103 (Academic Press, 1986), the disclosure of which is herein incorporated by reference.
  • the cells are allowed to grow in the selection media for sufficient time for colony formation and antibody production. This is usually between about 7 and about 14 days.
  • the hybridoma colonies are then assayed for the production of antibodies that specifically bind to KIR3DL2 polypeptide gene products, optionally the epitope specifically recognized by antibody AZ158, 19H12, 2B12 or 12B1 1 .
  • the assay is typically a colorimetric ELISA-type assay, although any assay may be employed that can be adapted to the wells that the hybridomas are grown in. Other assays include radioimmunoassays or fluorescence activated cell sorting. The wells positive for the desired antibody production are examined to determine if one or more distinct colonies are present.
  • the cells may be re-cloned and grown to ensure that only a single cell has given rise to the colony producing the desired antibody.
  • the antibodies will also be tested for the ability to bind to KIR3DL2 polypeptides, e.g., KIR3DL2-expressing cells.
  • Hybridomas that are confirmed to produce a monoclonal antibody can be grown up in larger amounts in an appropriate medium, such as DMEM or RPMI-1640.
  • an appropriate medium such as DMEM or RPMI-1640.
  • the hybridoma cells can be grown in vivo as ascites tumors in an animal.
  • the growth media con- taining monoclonal antibody (or the ascites fluid) is separated away from the cells and the monoclonal antibody present therein is purified. Purification is typically achieved by gel electrophoresis, dialysis, chromatography using protein A or protein G-Sepharose, or an anti- mouse Ig linked to a solid support such as agarose or Sepharose beads (all described, for example, in the Antibody Purification Handbook, Biosciences, publication No. 18-1037-46, Edition AC, the disclosure of which is hereby incorporated by reference).
  • the bound antibody is typically eluted from protein A protein G columns by using low pH buffers (glycine or acetate buffers of pH 3.0 or less) with immediate neutralization of antibody-containing fractions. These fractions are pooled, dialyzed, and concentrated as needed.
  • low pH buffers glycine or acetate buffers of pH 3.0 or less
  • Antibodies may also be produced by selection of combinatorial libraries of immunoglobulins, as disclosed for instance in (Ward et al. Nature, 341 (1989) p. 544, the entire disclosure of which is herein incorporated by reference).
  • the identification of one or more antibodies that bind(s) to KIR3DL2, particularly substantially or essentially the same epitope as monoclonal antibody AZ158, 19H12, 2B12 or 12B1 1 can be readily determined using any one of a variety of immunological screening assays in which antibody competition can be assessed. Many such assays are routinely practiced and are well known in the art (see, e. g., U. S. Pat. No. 5,660,827, issued Aug. 26, 1997, which is specifically incorporated herein by reference). It will be understood that actual- ly determining the epitope to which an antibody described herein binds is not in any way required to identify an antibody that binds to the same or substantially the same epitope as the monoclonal antibody described herein.
  • test antibodies to be examined are obtained from different source animals, or are even of a different Ig isotype
  • a simple competition assay may be em- ployed in which the control (AZ158, 19H12, 2B12 or 12B1 1 , for example) and test antibodies are admixed (or pre-adsorbed) and applied to a sample containing KIR3DL2 polypeptides. Protocols based upon western blotting and the use of BIACORE analysis are suitable for use in such competition studies.
  • the control and varying amounts of test antibodies can simply be admixed during exposure to the KIR3DL2 antigen sample. As long as one can distinguish bound from free antibodies (e. g., by using separation or washing techniques to eliminate unbound antibodies) and AZ158, 19H 12, 2B12 or 12B1 1 from the test antibodies (e.
  • test antibodies by using species-specific or isotype-specific secondary antibodies or by specifically labeling AZ158, 19H 12, 2B12 or 12B1 1 with a detectable label) one can determine if the test antibodies reduce the binding of AZ158, 19H12, 2B12 or 12B1 1 to the antigens, indicating that the test antibody recognizes substantially the same epitope as AZ158, 19H12, 2B12 or 12B1 1 .
  • the binding of the (la- beled) control antibodies in the absence of a completely irrelevant antibody can serve as the control high value.
  • the control low value can be obtained by incubating the labeled (AZ158, 19H12, 2B12 or 12B1 1 ) antibodies with unlabelled antibodies of exactly the same type (AZ158, 19H12, 2B12 or 12B1 1 ), where competition would occur and reduce binding of the labeled antibodies.
  • a significant reduction in labeled antibody reactivity in the presence of a test antibody is indicative of a test antibody that recognizes substantially the same epitope, i.e., one that "cross-reacts" or competes with the labeled (AZ158, 19H 12, 2B12 or 12B1 1 ) antibody.
  • test antibody that reduces the binding of AZ158, 19H12, 2B12 or 12B1 1 to KIR3DL2 antigens by at least about 50%, such as at least about 60%, or more preferably at least about 80% or 90% (e. g., about 65-100%), at any ratio of AZ158, 19H12, 2B12 or 12B1 1 : test antibody between about 1 :10 and about 1 :100 is considered to be an antibody that binds to substantially the same epitope or determinant as AZ158, 19H12, 2B12 or 12B1 1.
  • test antibody will reduce the binding of AZ158, 19H12, 2B12 or 12B1 1 to the KIR3DL2 antigen by at least about 90% (e.g., about 95%).
  • ⁇ Competition can also be assessed by, for example, a flow cytometry test.
  • cells bearing a given KIR3DL2 polypeptide can be incubated first with AZ158, 19H12, 2B12 or 12B1 1 , for example, and then with the test antibody labeled with a fluorochrome or biotin.
  • the antibody is said to compete with AZ158, 19H12, 2B12 or 12B1 1 if the binding obtained upon preincubation with a saturating amount of AZ158, 19H12, 2B12 or 12B1 1 is about 80%, about 50%, about 40% or less (e.g., about 30%, 20% or 10%) of the binding (as measured by mean of fluorescence) obtained by the antibody without pre-incubation with AZ158, 19H 12, 2B12 or 12B1 1.
  • an antibody is said to compete with AZ158, 19H12, 2B12 or 12B1 1 if the binding obtained with a labeled AZ158, 19H12, 2B12 or 12B1 1 antibody (by a fluorochrome or biotin) on cells preincubated with a saturating amount of test antibody is about 80%, about 50%, about 40%, or less (e. g., about 30%, 20% or 10%) of the binding obtained without preincubation with the test antibody.
  • a simple competition assay in which a test antibody is pre-adsorbed and applied at saturating concentration to a surface onto which a KIR3DL2 antigen is immobilized may also be employed.
  • the surface in the simple competition assay is for example a BIACORE chip (or other media suitable for surface plasmon resonance analysis).
  • the control antibody e.g., AZ158, 19H12, 2B12 or 12B1 1
  • the control antibody is then brought into contact with the surface at a KIR3DL2- saturating concentration and the KIR3DL2 and surface binding of the control antibody is measured. This binding of the control antibody is compared with the binding of the control antibody to the KIR3DL2-containing surface in the absence of test antibody.
  • a significant reduction in binding of the KIR3DL2-containing surface by the control antibody in the presence of a test antibody indicates that the test antibody recognizes substantially the same epitope as the control antibody such that the test antibody "cross-reacts" with the control antibody.
  • Any test antibody that reduces the binding of control (such as AZ158, 19H12, 2B12 or 12B1 1 ) antibody to a KIR3DL2 antigen by at least about 30% or more, or about 40%, can be considered to be an antibody that binds to substantially the same epitope or de- terminant as a control (e.g., AZ158, 19H12, 2B12 or 12B1 1 ).
  • such a test anti- body will reduce the binding of the control antibody (e.g., AZ158, 19H 12, 2B12 or 12B1 1 ) to the KIR3DL2 antigen by at least about 50% (e. g., at least about 60%, at least about 70%, or more).
  • the order of control and test antibodies can be reversed: that is, the control antibody can be first bound to the surface and the test antibody is brought into contact with the surface thereafter in a competition assay.
  • the antibody having higher affinity for the KIR3DL2 antigen is bound to the surface first, as it will be expected that the decrease in binding seen for the second antibody (assuming the antibodies are cross-reacting) will be of greater magnitude.
  • assays are provided in, e.g., Saunal (1995) J. Immunol. Methods 183: 33-41 , the disclosure of which is in- corporated herein by reference.
  • an epitope region for an anti-KIR3DL2 antibody may be determined by epitope "foot-printing" using chemical modification of the exposed amines/carboxyls in the KIR3DL2 protein.
  • a foot-printing technique is the use of HXMS (hydrogen-deuterium exchange detected by mass spectrometry) wherein a hydrogen/deuterium exchange of receptor and ligand protein amide protons, binding, and back exchange occurs, wherein the backbone amide groups participating in protein binding are protected from back exchange and therefore will remain deuterated.
  • NMR nuclear magnetic resonance epitope mapping
  • the antigen typically is selectively isotopically labeled with 15N so that only signals corresponding to the antigen and no signals from the antigen binding peptide are seen in the NMR- spectrum.
  • Antigen signals originating from amino acids involved in the interaction with the antigen binding peptide typically will shift position in the spectrum of the complex compared to the spectrum of the free antigen, and the amino acids involved in the binding can be identified that way. See, e. g., Ernst Schering Res Found Workshop. 2004; (44): 149-67; Huang et Journal of Molecular Biology, Vol. 281 (1 ) pp. 61 -67 (1998); and Saito and Patterson, Methods. 1996 Jun; 9 (3): 516-24.
  • Epitope mapping/characterization also can be performed using mass spectrometry methods. See, e.g., Downward, J Mass Spectrom. 2000 Apr; 35 (4): 493-503 and Kiselar and Downard, Anal Chem. 1999 May 1 ; 71 (9): 1792-801.
  • Protease digestion techniques also can be useful in the context of epitope mapping and identification.
  • Antigenic determinant-relevant regions/sequences can be determined by protease digestion, e.g. by using trypsin in a ratio of about 1 :50 to KIR3DL2 or o/n digestion at and pH 7-8, followed by mass spectrometry (MS) analysis for peptide identification.
  • MS mass spectrometry
  • the peptides protected from trypsin cleavage by the anti-KIR3DL2 binder can subsequently be identified by comparison of samples subjected to trypsin digestion and samples incubated with antibody and then subjected to digestion by e.g. trypsin (thereby revealing a footprint for the binder).
  • Other enzymes like chymotrypsin, pepsin, etc. also or alternatively can be used in similar epitope characterization methods.
  • enzymatic digestion can provide a quick method for analyzing whether a potential antigenic determinant sequence is within a region of the KIR3DL2 polypeptide that is not surface exposed and, accordingly, most likely not relevant in terms of immunogenici- ty/antigenicity. See, e. g., Manca, Ann 1st Super Sanita. 1991 ; 27: 15-9 for a discussion of similar techniques.
  • Site-directed mutagenesis is another technique useful for elucidation of a binding epitope. For example, in “alanine-scanning", each residue within a protein segment is replaced with an alanine residue, and the consequences for binding affinity measured. If the mutation leads to a significant reduction in binding affinity, it is most likely involved in binding. Monoclonal antibodies specific for structural epitopes (i.e., antibodies which do not bind the unfolded protein) can be used to verify that the alanine-replacement does not influence overall fold of the protein. See, e.g. , Clackson and Wells, Science 1995; 267:383-386; and Wells, Proc Natl Acad Sci USA 1996; 93:1-6.
  • Electron microscopy can also be used for epitope "foot-printing".
  • Wang et al., Nature 1992; 355:275-278 used coordinated application of cryoelectron microscopy, three-dimensional image reconstruction, and X-ray crystallography to determine the physical footprint of a Fab-fragment on the capsid surface of native cowpea mosaic virus.
  • label-free assay for epitope evaluation include surface plasmon resonance (SPR, BIACORE) and reflectometric interference spectroscopy (RifS).
  • SPR surface plasmon resonance
  • RifS reflectometric interference spectroscopy
  • an antibody binding the same or substantially the same epitope as an antibody can be identified in one or more of the exemplary competition assays described herein. Once antibodies are identified that are capable of binding KIR3DL2 and/or having other desired properties, they will also typically be assessed, using standard methods including those described herein, for their ability to bind to other polypeptides, including unrelated polypeptides. Ideally, the antibodies only bind with substantial affinity to KIR3DL2, e.g., hu- man KIR3DL2, and do not bind at a significant level to unrelated polypeptides.
  • the antibodies are suitable for use in the present methods.
  • the binding of the antibodies to KIR3DL2-expressing cells can also be assessed in non-human primates, e.g. cynomolgus monkeys, or other mammals such as mice.
  • non-human primates e.g. cynomolgus monkeys
  • the disclosure also relates to methods of producing such antibodies, comprising: (a) immunizing a non-human mammal with an immunogen comprising a KIR3DL2 polypeptide; and (b) preparing antibodies from said immunized animal; and (c) selecting antibodies from step (b) that are capable of binding KIR3DL2.
  • the antibodies prepared according to the present methods are monoclonal antibodies.
  • the non-human animal used iesto produce antibodies is a mammal, such as a rodent, bovine, porcine, fowl, horse, rabbit, goat, or sheep.
  • the DNA encoding an antibody that binds an epitope present on KIR3DL2 polypeptides is isolated from the hybridoma and placed in an appropriate expression vector for transfection into an appropriate host. The host is then used for the recombinant production of the antibody, or variants thereof, such as a humanized version of that monoclonal antibody, active fragments of the antibody, chimeric antibodies comprising the antigen recognition portion of the antibody, or versions comprising a detectable moiety.
  • DNA encoding a monoclonal antibody can be readily isolated and sequenced using conventional procedures (e. g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of murine antibodies).
  • the DNA can be placed into expression vectors, which are then transfected into host cells such as E. coli cells, simian COS cells, Chinese hamster ovary (CHO) cells, or myeloma cells that do not otherwise produce immunoglobulin protein, to obtain the synthesis of monoclonal antibodies in the recombinant host cells.
  • DNA sequences can be modified for any of a large number of purposes, e.g., for humanizing antibodies, producing fragments or derivatives, or for modifying the sequence of the antibody, e.g., in the antigen binding site in order to optimize the binding specificity of the antibody.
  • an antigen-binding compound may be assessed for its ability to induce ADCC or CDC towards, inhibit the activity and/or proliferation of and/or cause the elimination of KIR3DL2-expressing target cells. Assessing the antigen-binding compound's ability to induce ADCC, CDC (complement dependent cytotoxicity) or generally lead to the elimination or inhibition of activity of KIR3DL2-expressing target cells, can be carried out at any suitable stage of the method. This assessment can be useful at one or more of the various steps involved in the identification, production and/or development of an antibody (or other compound) destined for therapeutic use.
  • activity may be assessed in the context of a screening method to identify candidate antigen-binding compounds, or in methods where an antigen-binding compound is selected and made human suitable (e.g. made chimeric or humanized in the case of an antibody), where a cell expressing the antigen- binding compound (e.g. a host cell expressing a recombinant antigen-binding compound) has been obtained and is assessed for its ability to produce functional antibodies (or other compounds), and/or where a quantity of antigen-binding compound has been produced and is to be assessed for activity (e.g. to test batches or lots of product).
  • the antigen- binding compound will be known to specifically bind to a KIR3DL2 polypeptide.
  • the step may involve testing a plurality (e.g., a very large number using high throughput screening methods or a smaller number) of antigen-binding compounds.
  • Testing CDC and ADCC can be carried out can be determined by various assays including those known in the art and those described in the experimental examples herein.
  • Testing ADCC typically involves assessing cell-mediated cytotoxicity in which a KIR3DL2- expressing target cell (e.g. a celiac disease cell or other KIR3DL2-expressing cell) with bound anti-KIR3DL2 antibody is recognized by an effector cell bearing Fc receptors, without the involvement of complement.
  • a cell which does not express a KIR3DL2 antigen can optionally be used as a control.
  • Activation of NK cell cytotoxicity is assessed by measuring an increase in cytokine production (e.g. IFN- ⁇ production) or cytotoxicity markers (e.g. CD107 mobilization).
  • the antibody will induce an increase in cytokine production, expression of cytotoxicity markers, or target cell lysis of at least 20%, 50%, 80%, 100%, 200% or 500% in the presence of target cells, compared to a control antibody (e.g. an antibody not binding to KIR3DL2, a KIR3DL2 antibody having murine constant regions).
  • lysis of target cells is detected, e.g. in a chromium release assay, for example the antibody will induce lysis of at least 10%, 20%, 30%, 40% or 50% of target cells.
  • Fragments and derivatives of antibodies can be produced by techniques that are known in the art. "Fragments” comprise a portion of the intact antibody, generally the antigen binding site or variable region.
  • antibody fragments include Fab, Fab', Fab'-SH, F (ab') 2, and Fv fragments; di- abodies; any antibody fragment that is a polypeptide having a primary structure consisting of one uninterrupted sequence of contiguous amino acid residues (referred to herein as a "single-chain antibody fragment” or "single chain polypeptide"), including without limitation (1 ) single-chain Fv molecules (2) single chain polypeptides containing only one light chain variable domain, or a fragment thereof that contains the three CDRs of the light chain variable domain, without an associated heavy chain moiety and (3) single chain polypeptides containing only one heavy chain variable region, or a fragment thereof containing the three CDRs of the heavy chain variable region, without an associated light chain moiety; and multispecific antibodies formed from antibody fragments. Included, inter alia, are a nanobody, domain antibody, single domain antibody or a "dAb”.
  • the DNA of a hybridoma producing an antibody can be modified prior to insertion into an expression vector, for example, by substituting the coding sequence for human heavy- and light-chain constant domains in place of the homologous non-human sequences (e.g., Morrison et al., PNAS pp. 6851 (1984)), or by covalently joining to the immunoglobulin coding sequence all or part of the coding sequence for a non- immunoglobulin polypeptide. In that manner, "chimeric” or "hybrid” antibodies are prepared that have the binding specificity of the original antibody. Typically, such non-immunoglobulin polypeptides are substituted for the constant domains of an antibody.
  • the antibody is humanized.
  • “Humanized” forms of antibodies are specific chimeric immunoglobulins, immunoglobulin chains or fragments thereof (such as Fv, Fab, Fab', F (ab') 2, or other antigen-binding subsequences of antibodies) which contain minimal sequence derived from the murine immunoglobulin.
  • humanized antibodies are human immunoglobulins (recipient antibody) in which residues from a complementary-determining region (CDR) of the recipient are re- placed by residues from a CDR of the original antibody (donor antibody) while maintaining the desired specificity, affinity, and capacity of the original antibody.
  • CDR complementary-determining region
  • humanized antibodies can comprise residues that are not found in either the recipient antibody or in the imported CDR or framework sequences. These modifications are made to further refine and optimize antibody performance.
  • the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the CDR regions correspond to those of the original antibody and all or substantially all of the FR re- gions are those of a human immunoglobulin consensus sequence.
  • the humanized antibody optimally also will comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin.
  • variable domains both light and heavy
  • sequence of the variable domain of an antibody is screened against the entire library of known human variable-domain sequences.
  • the human sequence which is closest to that of the mouse is then accepted as the human framework (FR) for the humanized antibody (Sims et al., J. Immunol. 151 , pp. 2296 (1993); Chothia and Lesk, J. Mol. 196, 1987, pp. 901 ).
  • Another method uses a particular framework from the consensus sequence of all human antibodies of a particular subgroup of light or heavy chains.
  • humanized antibodies are prepared by a process of analysis of the parental sequences and various conceptual humanized products using three-dimensional models of the parental and humanized sequences.
  • Three-dimensional immunoglobulin models are commonly available and are familiar to those skilled in the art.
  • Computer programs are available which illustrate and display probable three-dimensional structures of selected candidate immunoglobulin sequences. Inspection of these displays permits analysis of the likely role of the residues in the functioning of the candidate immunoglobulin sequence, i.e., the analysis of residues that influence the ability of the candidate immunoglobulin to bind its antigen.
  • FR residues can be selected and combined from the consensus and import sequences so that the desired antibody characteristic, such as increased affinity for the target antigen (s), is achieved.
  • the CDR residues are directly and most substantially in- volved in influencing antigen binding.
  • Another method of making "humanized” monoclonal antibodies is to use a Xen- oMouse (Abgenix, Fremont, CA) as the mouse used for immunization.
  • a XenoMouse is a murine host that has had its immunoglobulin genes replaced by functional human immunoglobulin genes.
  • antibodies produced by this mouse or in hybridomas made from the B cells of this mouse are already humanized.
  • the XenoMouse is described in United States Patent No. 6,162,963, which is herein incorporated in its entirety by reference.
  • Human antibodies may also be produced according to various other techniques, such as by using, for immunization, other transgenic animals that have been engineered to express a human antibody repertoire (Jakobovitz et al., Nature 362 (1993) 255), or by selec- tion of antibody repertoires using phage display methods. Such techniques are known to the skilled person and can be implemented starting from monoclonal antibodies as disclosed in the present application.
  • a KIR3DL2 binding compound e.g., an anti-KIR3DL2 antibody
  • the second moiety is a therapeutic agent, a toxic agent, and/or a detectable agent.
  • antibodies in underivatized or unmodified form are expected to inhibit the proliferation of the overproliferating cells or be cytotoxic towards overproliferating cells such as in those from a celiac disease patient, e.g., by direct- ing ADCC and/or CDC toward KIR3DL2-expressing celiac disease cells, it is also possible to prepare derivatized antibody immunoconjugates that are cytotoxic.
  • the KIR3DL2 specific antibodies are isolated and optionally otherwise modified (e.g. humanized), they will be derivatized to make them toxic to cells. In this way, administration of the antibody to celiac disease patients will lead to the relatively specific binding of the anti- body to overproliferating cells, thereby directly killing or inhibiting the cells underlying the disorder.
  • the antibodies will be directly derivatized with radioisotopes or other toxic compounds.
  • toxic agents used in immunoconjugates in develop- ment include, in particular, for example taxanes, anthracyclines, camptothecins, epothilones, mytomycins, combretastatins, vinca alkaloids, nitrogen mustards, maytansinoids, cali- cheamycins, duocarmycins, tubulysins, dolastatins and auristatins, enediynes, pyrrolobenzo- diazepines, ethylenimines, radioisotopes, therapeutic proteins and peptides, and toxins or fragments thereof.
  • any type of moiety with a cytotoxic or cytoinhibitory effect can be used in conjunction with the present antibodies to inhibit or kill specific NK receptor expressing cells, including radioisotopes, toxic proteins, toxic small molecules, such as drugs, toxins, im- munomodulators, hormones, hormone antagonists, enzymes, oligonucleotides, enzyme inhibitors, therapeutic radionuclides, angiogenesis inhibitors, chemotherapeutic drugs, vinca alkaloids, epidophyllotoxins, antimetabolites, alkylating agents, antibiotics, antimitotics, anti- angiogenic and apoptotoic agents, particularly doxorubicin, methotrexate, camptothecans, nitrogen mustards, gemcitabine, alkyl sulfonates, nitrosoureas, triazenes, folic acid analogs, pyrimidine analogs, purine analogs, platinum coordination complexes, Pseudomonas exotoxin, ricin, 5-
  • the antibody will be derivatized with a radioactive isotope, such as 1-131.
  • a radioactive isotope such as 1-131.
  • Any of a number of suitable radioactive isotopes can be used, including, but not limited to, lndium-1 1 1 , Lutetium-171 , Bismuth-212, Bismuth-213, Astatine-21 1 , Copper-62, Copper-64, Copper-67, Yttrium-90, lodine-125, lodine-131 , Phosphorus-32, Phosphorus-33, Scandium-47, Silver-1 1 1 , Gallium-67, Praseodymium-142, Samarium-153, Terbium-161 , Dysprosium-166, Holmium-166, Rhenium-186, Rhenium-188, Rhenium-189, Lead-212, Ra- dium-223, Actinium-225, lron-59, Selenium-75, Arsenic-77, St
  • the radionuclide may have a decay energy in the range of 20 to 6,000 keV, optionally in the ranges 60 to 200 keV for an Auger emitter, 100- 2,500 keV for a beta emitter, and 4,000-6,000 keV for an alpha emitter. Also provided are radionuclides that substantially decay with generation of alpha-particles.
  • the antibodies can also be made with modifications that increase their ability to bind Fc receptors which can affect effector functions such as antibody-dependent cytotoxicity, mast cell degranulation, and phagocytosis, as well as immunomodulatory signals such as regulation of lymphocyte proliferation and antibody secretion.
  • Typical modifications include modified human lgG1 constant regions comprising at least one amino acid modification (e.g. substitution, deletions, insertions), and/or altered types of glycosylation, e.g., hypofucosylation.
  • FcyRI CD64
  • FcyRI I CD32
  • FcyRI II CD 16
  • FcyRI CD64
  • FcyRIIA CD32A
  • FcvRIII CD 16
  • a modification may, for example, increase binding of the Fc domain to FcyRllla on effector (e.g. NK) cells.
  • Anti-KIR3DL2 antibodies may comprise an Fc domain (or portion thereof) of human lgG1 or lgG3 isotype, optionally modified. Residues 230-341 (Kabat EU) are the Fc CH2 region. Residues 342-447 (Kabat EU) are the Fc CH3 region. Anti-KIR3DL2 antibodies may comprise a variant Fc region having one or more amino acid modifications (e.g., substitutions, deletions, insertions) in one or more portions, which modifications increase the affinity and avidity of the variant Fc region for an FcyR (including activating and inhibitory FcyRs).
  • amino acid modifications e.g., substitutions, deletions, insertions
  • said one or more amino acid modifications increase the affinity of the variant Fc region for FcyRIIIA and/or FcyRIIA.
  • the variant Fc region further specifically binds FcyRIIB with a lower affinity than does the Fc region of the comparable parent antibody (i.e., an antibody having the same amino acid sequence as the antibody except for the one or more amino acid modifications in the Fc region).
  • the one or both of the histidine residues at amino acid positions 310 and 435 may be substituted, for example by lysine, alanine, glycine, valine, leucine, isoleucine, proline, methionine, tryptophan, phenylalanine, serine or threonine (see, e.g. PCT publication no. WO 2007/080277); such substituted constant regions provide decreased binding to the inhibitory FcyRIIB without decreasing binding to the activatory FcyRIIIA.
  • such modifications in- crease the affinity of the variant Fc region for FcyRIIIA and/or FcyRIIA and also enhance the affinity of the variant Fc region for FcyyRIIB relative to the parent antibody.
  • said one or more amino acid modifications increase the affinity of the variant Fc region for FcyRIIIA and/or FcyRIIA but do not alter the affinity of the variant Fc regions for FcyRIIB relative to the Fc region of the parent antibody.
  • said one or more amino acid modifications enhance the affinity of the variant Fc region for FcyRIIIA and FcyRIIA but reduce the affinity for FcyRIIB relative to the parent antibody. Increased affinity and/or avidity results in detectable binding to the FcyR or FcyR- related activity in cells that express low levels of the FcyR when binding activity of the parent molecule (without the modified Fc region) cannot be detected in the cells.
  • the affinities and binding properties of the molecules for an FcyR can be determined using in vitro assays (biochemical or immunological based assays) known in the art for determining antibody-antigen or Fc-FcyR interactions, i.e., specific binding of an antigen to an antibody or specific binding of an Fc region to an FcyR, respectively, including but not limited to ELISA assay, surface plasmon resonance assay, immunoprecipitation assays.
  • the molecules comprising a variant Fc region comprise at least one amino acid modification (for example, possessing 1 , 2, 3, 4, 5, 6, 7, 8, 9, or more amino acid modifications) in the CH3 domain of the Fc region. In other embodiments, the molecules comprising a variant Fc region comprise at least one amino acid modification (for example, possessing 1 , 2, 3, 4, 5, 6, 7, 8, 9, or more amino acid modifications) in the CH2 domain of the Fc region, which is defined as extending from amino acids 231 -341 . In some embodiments, the molecules comprise at least two amino acid modifications (for example, possessing 2, 3, 4, 5, 6, 7, 8, 9, or more amino acid modifications), wherein at least one such modification is in the CH3 region and at least one such modification is in the CH2 region. Amino acid modifications may be made for example in the hinge region. In a particular em- bodiment, the invention encompasses amino acid modification in the CH 1 domain of the Fc region, which is defined as extending from amino acids 216-230.
  • Anti-KIR3DL2 antibodies may comprise a variant Fc region, wherein the variant Fc region comprises at least one amino acid modification (for example, possessing 1 , 2, 3, 4, 5, 6, 7, 8, 9, or more amino acid modifications) relative to a wild-type Fc region, such that the molecule has an enhanced effector function relative to a molecule comprising a wild-type Fc region, optionally wherein the variant Fc region comprises a substitution at any one or more of positions 221 , 239, 243, 247, 255, 256, 258, 267, 268, 269, 270, 272, 276, 278, 280, 283, 285, 286, 289, 290, 292, 293, 294, 295, 296, 298, 300, 301 , 303, 305, 307, 308, 309, 310, 31 1 , 312, 316, 320, 322, 326, 329, 330, 332, 331 , 332, 333, 334, 335, 337, 338, 339, 340,
  • Anti-KIR3DL2 antibodies may comprise a variant Fc region, wherein the variant Fc region comprises at least one amino acid modification (for example, possessing 1 , 2, 3, 4, 5, 6, 7, 8, 9, or more amino acid modifications) relative to a wild-type Fc region, such that the molecule has an enhanced effector function relative to a molecule comprising a wild-type Fc region, optionally wherein the variant Fc region comprises a substitution at any one or more of positions 329, 298, 330, 332, 333 and/or 334 (e.g. S239D, S298A, A330L, I332E, E333A and/or K334A substitutions).
  • the variant Fc region comprises at least one amino acid modification (for example, possessing 1 , 2, 3, 4, 5, 6, 7, 8, 9, or more amino acid modifications) relative to a wild-type Fc region, such that the molecule has an enhanced effector function relative to a molecule comprising a wild-type Fc region, optionally wherein the variant Fc region comprises
  • antibodies having variant or wild-type Fc regions may have altered glycosylation patterns that increase Fc receptor binding ability of antibodies.
  • car- bohydrate modifications can be accomplished by, for example, expressing the antibody in a host cell with altered glycosylation machinery.
  • Cells with altered glycosylation machinery have been described in the art and can be used as host cells in which to express recombinant antibodies to thereby produce an antibody with altered glycosylation. See, for example, Shields, R.L. et al. (2002) J. Biol. Chem. 277:26733-26740; Umana et al. (1999) Nat. Bio- tech. 17:176-1 , as well as, European Patent No: EP 1 ,176,195; PCT Publications WO 06/133148; WO 03/035835; WO 99/54342, each of which is incorporated herein by reference in its entirety.
  • such antibodies with altered glycosylation are "glyco-optimized” such that the antibody has a particular N-glycan structure that produces certain desirable properties, including but not limited to, enhanced ADCC and effector cell receptor binding activity when compared to non-modified antibodies or antibodies having a naturally occurring constant region and produced by murine myeloma NSO and Chinese Hamster Ovary (CHO) cells (Chu and Robinson, Current Opinion Biotechnol. 2001 , 12: 180-7), HEK293T-expressed antibodies as produced herein in the Examples section, or other mammalian host cell lines common- ly used to produce recombinant therapeutic antibodies.
  • CHO Chinese Hamster Ovary
  • Monoclonal antibodies produced in mammalian host cells contain an N- linked glycosylation site at Asn297 of each heavy chain.
  • Glycans on antibodies are typically complex bi- atennary structures with very low or no bisecting N-acetylglucosamine (bisecting GlcNAc) and high levels of core fucosylation.
  • Glycan temini contain very low or no terminal sialic acid and variable amounts of galactose.
  • the important carbohydrate structures contributing to antibody activity are believed to be the fucose residues attached via alpha-1 ,6 linkage to the innermost N- acetylglucosamine (GlacNAc) residues of the Fc region N-linked oligosaccharides (Shields et al., 2002).
  • FcyR binding requires the presence of oligosaccharides covalently attached at the conserved Asn297 in the Fc region of human IgGI, lgG2 or lgG3 type.
  • Non-fucosylated oligosaccharides structures have recently been associated with dramatically increased in vitro ADCC activity.
  • “Asn 297” means amino acid asparagine located at about position 297 in the Fc region; based on minor sequence variations of antibodies, Asn297 can also be located some amino acids (usually not more than +3 amino acids) upstream or downstream.
  • antibodies produced in CHO cells contain about 2 to 6% in the population that are nonfucosylated.
  • YB2/0 rat myeloma
  • Lecl3 cell line a lectin mutant of CHO line which has a deficient GDP- mannose 4,6-dehydratase leading to the deficiency of GDP- fucose or GDP sugar intermediates that are the substrate of alpha6-fucosyltransferase have been reported to produce antibodies with 78 to 98% non-fucosylated species.
  • RNA interference or knock-out techniques can be employed to engineer cells to either decrease the FUT8 mRNA transcript levels or knock out gene expression entirely, and such antibodies have been reported to contain up to 70% non-fucosylated glycan.
  • An antibody that binds to KIR3DL2 may be glycosylated with a sugar chain at Asn297.
  • an antibody will comprise a constant region comprising at least one amino acid alteration in the Fc region that improves antibody binding to FcyRllla and/or ADCC.
  • an antibody composition comprises a chimeric, human or humanized antibody described herein, wherein at least 20, 30, 40, 50, 60, 75, 85, 90, 95% or substantially all of the antibody species in the composition have a constant region comprising a core carbohydrate structure (e.g. complex, hybrid and high mannose structures) which lacks fucose.
  • a core carbohydrate structure e.g. complex, hybrid and high mannose structures
  • an antibody composition which is free of antibodies comprising a core carbohydrate structure having fucose.
  • the core carbohydrate will preferably be a sugar chain at Asn297.
  • an antibody composition e.g. a composition comprising antibodies which bind to KIR3DL2, are glycosylated with a sugar chain at Asn297, wherein the antibodies are partially fucosylated.
  • Partially fucosylated antibodies are characterized in that the proportion of anti-KIR3DL2 antibodies in the composition that lack fucose within the sugar chain at Asn297 is between 20% and 90%, between 20% and 80%, between 20% and 50%, 55%, 60%, 70% or 75%, between 35% and 50%, 55%, 60%, 70% or 75%, or between 45% and 50%, 55%, 60%, 70% or 75%.
  • the antibody is of human IgGI or lgG3 type.
  • the sugar chain show can further show any characteristics (e.g. presence and proportion of complex, hybrid and high mannose structures), including the characteristics of N- linked glycans attached to Asn297 of an antibody from a human cell, or of an antibody re- combinantly expressed in a rodent cell, murine cell (e.g. CHO cell) or in an avian cell.
  • characteristics e.g. presence and proportion of complex, hybrid and high mannose structures
  • the antibody is expressed in a cell that is lacking in a fucosyl- transferase enzyme such that the cell line produces proteins lacking fucose in their core carbohydrates.
  • the cell lines Ms704, Ms705, and Ms709 lack the fucosyltransfer- ase gene, FUT8 (alpha (1 ,6) fucosyltransferase), such that antibodies expressed in the Ms704, Ms705, and Ms709 cell lines lack fucose on their core carbohydrates.
  • FUT8 alpha (1 ,6) fucosyltransferase
  • the antibody is expressed in a cell line with a functionally disrupted FUT8 gene, which encodes a fucosyl transferase, such that antibodies expressed in such a cell line exhibit hypofucosylation by reducing or eliminating the alpha 1 ,6 bond-related enzyme.
  • the antibody is expressed in cell lines engineered to express gly- coprotem-modifying glycosyl transferases (e.g., beta(l,4)-N-acetylglucosaminyl-transferase III (GnTHI)) such that antibodies expressed in the engineered cell lines exhibit increased bisecting GlcNac structures which results in increased ADCC activity of the antibodies (PCT Publication WO 99/54342 by Umana et al.; and Umana et al. (1999) Nat. Biotech. 17:176-180, the disclosures of which are incorporated herein by reference).
  • gly- coprotem-modifying glycosyl transferases e.g., beta(l,4)-N-acetylglucosaminyl-transferase III (GnTHI)
  • the antibody is expressed and the fucosyl residue(s) is cleaved using a fucosidase enzyme.
  • a fucosidase enzyme removes fucosyl residues from antibodies (Tarentino, et al. (1975) Biochem. 14:5516-5523).
  • a cell line producing an antibody can be treated with a glycosylation inhibi- tor; Zhou et al. Biotech, and Bioengin. 99: 652-665 (2008) described treatment of CHO cells with the alpha-mannosidase I inhibitor, kifunensine, resulting in the production of antibodies with non-fucosylated oligomannose-type N-glucans.
  • the antibody is expressed in a cell line which naturally has a low enzyme activity for adding fucosyl to the N-acetylglucosamine that binds to the Fc region of the antibody or does not have the enzyme activity, for example the rat myeloma cell line YB2/0 (ATCC CRL 1662).
  • cell lines include a variant CHO cell line, Led 3 cells, with reduced ability to attach fucosyl to Asn(297)-linked carbohydrates, also resulting in hypofucosylation of antibodies expressed in that host cell (WO 03/035835 (Presta et al); and Shields, RX. et al. (2002) J. Biol. Chem.
  • the antibody is expressed in an avian cell, e.g., a EBx® cell (Vivalis, France) which naturally yields antibodies with low fu- cose content e.g WO2008/142124.
  • Hypofucosylated glycans can also be produced in cell lines of plant origin, e.g. WO 07/084926A2 (Biolex Inc.), WO 08/006554 (Greenovation Biotech GMBH), the disclosures of which are incorporated herein by reference.
  • compositions include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carbox- ymethylcellulose, polyacrylates, waxes, polyethylene-polyoxypropylene- block polymers, polyethylene glycol and wool fat.
  • ion exchangers alumina, aluminum stearate, lecithin
  • serum proteins such as human serum albumin
  • buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate,
  • the antibodies may be employed in a method of modulating, e.g inhibiting, the activity of KIR3DL2-expressing cells in a patient. This method comprises the step of contacting said composition with said patient. Such method will be useful for both prophylaxis and therapeutic purposes.
  • compositions for use in administration to a patient, will be formulated for administration to the patient.
  • the compositions may be administered orally, parenterally, by inhala- tion spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir.
  • the used herein includes subcutaneous, intravenous, intramuscular, intra-articular, intra-synovial, intrasternal, intrathecal, intrahepatic, intralesional and intracranial injection or infusion techniques.
  • Sterile injectable forms of the compositions may be aqueous or an oleaginous sus- pension. These suspensions may be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, for example as a solution in 1 ,3-butanediol.
  • the acceptable vehicles and solvents that may be employed are water, Ringer's solution and iso- tonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose, any bland fixed oil may be employed including synthetic mono-or diglycerides.
  • Fatty acids such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically- acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions.
  • These oil solutions or suspensions may also contain a long-chain alcohol diluent or disper- sant, such as carboxymethyl cellulose or similar dispersing agents that are commonly used in the formulation of pharmaceutically acceptable dosage forms including emulsions and suspensions.
  • Other commonly used surfactants such as Tweens, Spans and other emulsify- ing agents or bioavailability enhancers which are commonly used in the manufacture of pharmaceutically acceptable solid, liquid, or other dosage forms may also be used for the purposes of formulation.
  • an antibody present in a pharmaceutical composition can be supplied at a concentration of 10 mg/mL in either 100 mg (10 mL) or 500 mg (50 mL) single-use vials.
  • the product is formulated for IV administration in 9.0 mg/mL sodium chloride, 7.35 mg/mL sodium citrate dihydrate, 0.7 mg/mL polysorbate 80, and Sterile Water for Injection.
  • An exemplary suitable dosage range for an antibody in a pharmaceutical composition may between about 1 mg/m 2 and 500 mg/m 2 .
  • schedules are exemplary and that an optimal schedule and regimen can be adapted taking into account the affinity and tolerability of the particular antibody in the pharmaceutical composition that must be determined in clinical tri- als.
  • a pharmaceutical composition for injection e.g., intramuscular, i.v.
  • the antibody compositions may further comprise another therapeutic agent, including agents normally utilized for the particular therapeutic purpose for which the antibody is being administered, notably for the treatment of a celiac disease.
  • the additional therapeutic agent will normally be present in the composition in amounts typically used for that agent in a monotherapy for the particular disease or condition being treated.
  • therapeutic agents include, but are not limited to anti-inflammation agents, steroids, immune system suppressors, antibiotics, antivirals and other antibodies and fragments thereof.
  • Celiac disease is usually detected by serologic testing of celiac-specific antibodies, notably the performance of tissue transglutaminase (TTG) antibody testing. The diagnosis is confirmed by duodenal mucosal biopsies.
  • TTG tissue transglutaminase
  • RCD Diagnosis of RCD is a more recent classification and requires specialised small bowel investigations, notably enteroscopy and small bowel imaging, and laboratory tests, notably immunohistochemistry, molecular analysis, and flow cytometry of isolated lELs.
  • Two types of RCD, RCD type I and RCD type II are generally distinguished according to the normal (RCDI) or abnormal (RCDII) phenotype of intestinal lELs.
  • RCDI histological examination is similar to that found in active celiac disease with villous atrophy and increased normal IEL.
  • RCDII an abnormal population of lELs is identified: over 25% of the CD103+ of CD45+ lELs lacking surface CD3-TCR complexes on flow cytometry, or more than 50% lELs expressing intracellular CD3e but no CD8 in formol-fixed sections and the presence of a detectable clonal rearrangement of the gamma chain of the TCD in duodenal biopsies.
  • KIR3DL2 is expressed in celiac disease, including in RCDI and RCDII. It is also shown that KIR3DL2 is expressed on enteropathy-associated T- cell lymphoma (EATL), a complication of CD (for EATL see also PCT/EP2014/053340 filed 20 February 2014, the disclosure of which is incorporated by reference).
  • EATL enteropathy-associated T- cell lymphoma
  • the methods comprise determining the level of expres- sion of a KIR3DL2 nucleic acid or polypeptide in a biological sample from a patient, e.g. in lymphocytes (e.g. intraepithelial lymphocytes) found in a biological sample.
  • lymphocytes e.g. intraepithelial lymphocytes
  • Abnormal lELs in celiac patients can be found in a sample obtained as a biopsy.
  • abnormal lELs can generally be found in and outside the intestine as cells have tendency to disseminate in and outside the intestine.
  • Abnormal lELs may be found for example in mesen- teric lymph nodes, blood, and bone marrow, and in different epitheliums such as lung and skin.
  • a high percentage of abnormal cells have been found to be predictive of abnormal circulating cells in peripheral blood.
  • the methods comprise: (a) determining whether an individual has a celiac disease (or a complication thereof, e.g. a lymphoma, an EATL); and (b) if the individual has a celiac disease (or a complication thereof), determining whether an individual has celiac disease cells (e.g. a population of abnormal lELs) that express a KIR3DL2 polypeptide.
  • the method further comprises determining whether disease cells also express other markers of abnormal lELs at their surface, for example determining whether cells are CD103+, CD45+, CD3-TCR-negative cells or intracellular CD3e+, CD8- cells.
  • KIR3DL2 expression at the surface of lELs can be used for example as a tool for monitoring disease, assessing disease status, as a prognostic tool for evaluating response to treatment (with an anti-KIR3DL2 agent or a different therapeutic agent), or as a tool for evaluating prognosis without treatment, e.g. the occurrence of compli- cations such as EATL.
  • the methods comprise determining the level of expression of a KIR3DL2 nucleic acid or polypeptide in a biological sample and comparing the level to a reference level (e.g. a value, weak cell surface staining, etc.) corresponding to a healthy(s) individual.
  • a reference level e.g. a value, weak cell surface staining, etc.
  • detecting a KIR3DL2 polypeptide in a biological sample comprises detect- ing KIR3DL2 polypeptide expressed on the surface of a lymphocyte.
  • a biological sample collected from a body compartment e.g., mesenteric lymph nodes, blood, and bone marrow, and in different epitheliums such as lung and skin, are brought into contact with an anti- KIR3DL2 antibody and lELs (e.g. the proportion of cells) expressing a KIR3DL2 polypeptide and/or other markers of abnormal lELs at their surface is measured.
  • the cells may be, for example CD103+, CD45+, CD3-TCR-negative cells or intracellular CD3e+, CD8- cells.
  • a finding that lELs express, or predominantly express, KIR3DL2 indicates that the disease can be treated with an anti-KIR3DL2 agent.
  • CD e.g. CD, RCD, RCDI, RCDII
  • a method for diagnosis of CD comprising bringing cells from a biological sample from an individual into contact with an anti- KIR3DL2 antibody and the proportion (e.g. percentage) of T cells expressing a KIR3DL2 polypeptide at their surface is measured, and comparing such proportion to the average propor- tion (e.g. percentage) of T cells expressing a KIR3DL2 polypeptide at their surface observed in non-celiac disease humans (e.g., in healthy humans), wherein a celiac disease-positive diagnosis is made when said measured proportion is significantly higher than said average proportion.
  • CD e.g. CD, RCD, RCDI, RCDII
  • the treat-ment involves administering anti-KIR3DL2 antibodies, anti-KIR3DL2 antibody compositions, and/or related compositions, to an individual having or susceptible to celiac disease.
  • the celiac disease is an aggressive or advanced celiac disease (e.g. RCD, RCDI, RCDII, or otherwise with a poor prognosis of survival).
  • the celiac disease is a non-cutaneous celiac disease.
  • the celiac disease is an aggressive T cell lymphoma.
  • the patient has relapsing or refractory disease.
  • the patient has a poor prognosis for disease progression (e.g. poor prognosis for survival) or has a poor prognosis for response to a therapy, e.g. gluten- free diet, stem cell transplantation, antibody therapy, chemotherapy, etc.
  • a therapy e.g. gluten- free diet, stem cell transplantation, antibody therapy, chemotherapy, etc.
  • Celiac disease diagnosis criteria can be those of standard medical guidelines, for example, according to the World Gastroenterology Organization (WGO) (see, e.g., World Gastroenterology Organization Practice Guidelines, Celiac Disease: World Gastroenterology Organization, 2007). See also, e.g., Rubio-Tapia et al. (2013) Am. J. Gastroenterology 108(5):656-677, the disclosures of which are incorporated herein by reference.
  • WGO World Gastroenterology Organization
  • a method of reducing progression of celiac disease in a mammalian host comprising administering an anti-KIR3DL2 antibody, an anti-KIR3DL2 antibody composition, or a related composition (e.g., a nucleic acid encoding an anti-KIR3DL2 antibody), in an amount sufficient to detectably reduce the progression of the celiac disease in the host.
  • the anti-KIR3DL2 antibody reduces the progression to or of lymphoma (e.g. EATL) in the host.
  • a method of treating celiac disease in an individual having a poor disease prognosis and/or who has relapsed, is resistant or is not responsive to therapy with a first therapeutic agent.
  • Disease diagnosis and progression can be defined by standard criteria, including for each subtype of CD (e.g. RCDI, RCDII) is typically based on examination of enteroscopy, small bowel imaging, peripheral blood or tissue biopsy for histological features supplemented by detailed immunohistochemistry, flow cytometry and molecular genetics. Progression is optionally determined by assessing the presence and/or numbers of abnormal lELs. Meth- ods for detecting disease progression can be achieved by any suitable technique, several examples of which are known in the art.
  • PCR and RT-PCR including for the diagnosis of complications such as lymphomas, detection of celiac disease associated genes or "markers"
  • biopsy including for the diagnosis of complications such as lymphomas, detection of celiac disease associated genes or "markers”
  • imaging techniques including for the biopsy of tissue and tissue samples
  • karyotyping and other chromosomal analysis including for the diagnosis of complications such as lymphomas, detection of celiac disease associated genes or "markers”
  • immunoassay/immunocytochemical detection techniques include histological and/or histopathology assays, cell kinetic studies and cell cycle analysis, flow cytometry, and physical examination techniques (e.g., for physical symptoms).
  • Delivering anti-KIR3DL2 antibodies to a subject can be used to reduce, treat, prevent, or otherwise ameliorate any suitable aspect of celiac disease.
  • the methods herein can be particularly useful in the reduction and/or amelioration of proliferation of abnormal lELs (e.g. percentage (abnormal lELs compared to healthy lymphocytes), number of abnormal lELs in circulation), and any parameter or symptom associated therewith (e.g. biomarkers).
  • a method of reducing the risk of disease progression, reducing the risk of lymphoma in a cell population that has undergone initiation (e.g. in a RCDI or RCDII patient), and/or providing a therapeutic regimen for reducing lymphoma pro- gression in a human patient having celiac disease which comprises administering to the patient one or more first treatments (e.g. induction therapy, such as a chemotherapeutic agent or an antibody) in an amount and regimen sufficient to achieve a response (partial or complete response), and then administering an amount of an Anti-KIR3DL2 antibody or related composition (or applying a combination administration method) to the patient.
  • first treatments e.g. induction therapy, such as a chemotherapeutic agent or an antibody
  • a method of increasing the likelihood of survival over a relevant period in a human patient diagnosed with celiac disease comprising administering to the patient a composition in an amount effective to improve the quality of life thereof.
  • methods described herein can be applied to significantly reduce the number of celiac disease cells in a vertebrate host, such that, for example, the total number of celiac disease cells (e.g., abnormal lELs) is reduced.
  • a method for killing e.g. either directly or indirectly causing death of) celiac disease cells in a vertebrate, such as a human patient.
  • the antibody compositions may be used in com- bined treatments with one or more other treatments or therapeutic agents, including treatments and agents normally utilized for the particular therapeutic purpose for which the antibody is being administered, notably for the treatment of a celiac disease.
  • the additional therapeutic agent will normally be administered in amounts and treatment regimens typically used for that agent in a monotherapy for the particular disease or condition being treated.
  • a second treatment may include a gluten free diet (GFT) or a hemato- poetic stem cell transplantation.
  • GFT gluten free diet
  • hemato- poetic stem cell transplantation a second treatment may include a gluten free diet (GFT) or a hemato- poetic stem cell transplantation.
  • a second therapeutic agent may for example include one or more chemotherapeutic drugs, antibodies that bind to a polypeptide selectively expressed on an abnormal IEL in a celiac disease patient, an immunomodulatory or immunosuppressive agent (e.g. an antibody that modulates the immune system, an agent that inhibits the IL-15 signalling pathway, an anti-IL-15 antibody, an agent that inhibits signalling by a protein tyrosine kinase, an agent that inhibits JAK3 signalling, STAT5 signalling and/or syk kinase), and/or a chemotherapeutic agent
  • an immunomodulatory or immunosuppressive agent e.g. an antibody that modulates the immune system, an agent that inhibits the IL-15 signalling pathway, an anti-IL-15 antibody, an agent that inhibits signalling by a protein tyrosine kinase, an agent that inhibits JAK3 signalling, STAT5 signalling and/or syk kinase
  • the KIR3DL2-binding compound and the second thera-guideic agent or treatment can be administered separately, together or sequentially, or in a cocktail.
  • the KIR3DL2-binding compound is administered prior to the administration of the second therapeutic agent or treatment.
  • the KIR3DL2- binding compound can be administered approximately 0 to 30 days prior to the administration of the second therapeutic agent.
  • an KIR3DL2-binding compound is administered from about 30 minutes to about 2 weeks, from about 30 minutes to about 1 week, from about 1 hour to about 2 hours, from about 2 hours to about 4 hours, from about 4 hours to about 6 hours, from about 6 hours to about 8 hours, from about 8 hours to 1 day, or from about 1 to 5 days prior to the administration of the second therapeutic agent.
  • a KIR3DL2-binding compound is administered concurrently with the admin- istration of the therapeutic agents.
  • a KIR3DL2-binding compound is administered after the administration of the second therapeutic agent.
  • a KIR3DL2-binding compound can be administered approximately 0 to 30 days after the administration of the second therapeutic agent or treatment.
  • an KIR3DL2-binding compound is administered from about 30 minutes to about 2 weeks, from about 30 minutes to about 1 week, from about 1 hour to about 2 hours, from about 2 hours to about 4 hours, from about 4 hours to about 6 hours, from about 6 hours to about 8 hours, from about 8 hours to 1 day, or from about 1 to 5 days after the administration of the second therapeutic agent.
  • Antibodies which bind KIR3DL2 but not closely related KIR3DL1 were generated by immunizing mice with recombinant KIR3DL2-Fc fusion protein.
  • Supernatant (SN) of the growing hybridomas were tested by flow cytometry on Sezary Syndrome cell lines (HUT78, COU-L) and HEK-293T/KIR3DL2 Domain 0 - eGFP.
  • Potentially interesting hybridomas se- lected from the initial screening were cloned by limiting dilution techniques in 96-wells plates.
  • the secondary screen involved selection of hybridomas of interest by testing supernatants of the subclones by flow cytometry on HUT78 , COU-L, HEK-293T/KIR3DL1 Domain 0 - eGFP and HEK-293T/KIR3DL2 Domain 0 - eGFP. Positive subclones were injected into mice to produce ascitis and antibodies of interest were purified before being tested in a Biacore assay using rec KIR3DL2 chips, followed by various assays formats based on binding to human KIR3DL2-expressing cells.
  • VH and VL sequences were then sub-cloned into the Lonza expression vectors (Double-Gene Vectors) using the InFu- sion system (Clontech) according to the manufacturer's instructions. After sequencing, vectors containing the VH and VL sequences were prepared as Maxiprep using the Promega PureYieldTM Plasmid Maxiprep System. Vectors were then used for HEK-293T cell transfec- tion using Invitrogen's Lipofectamine 2000 according to the manufacturer instructions. Antibodies generated included, inter alia, 19H12 and 12B1 1 .
  • Hut-78 cells were harvested and stained in PBS 1 X / BSA 0,2% / EDTA 2 mM buffer during 1 H at 4°C using 5 ⁇ g ml of increasing concentrations of the antibodies, includ- ing 19H12, 12B1 1 and AZ158 (previously identified) (0.006-200 Mg/ml). After two washes, staining data were acquired on a BD FACS Canto II and analyzed using the FlowJo software.
  • Antibodies were further tested for binding to a series of KIR3DL2 mutants.
  • Antibodies 19H12 and 12B1 1 did not show any loss of binding to unmutated wild type KIR3DL2 (WTaKIR3DL2), but lost binding to mutant 1 1 having P179T and S181T substitutions as well as to mutant 1 1A1 having V178A and H180S substitutions.
  • the principal epitope of these antibodies 19H12, 18B10 and 12B1 1 therefore includes residues P179, S181 , V178 and/or H180. These residues at positions 179 and 181 in mutant 1 1 correspond to the residues present at in KIR3DL1 (KIR3DL1 has T179 and T181 ).
  • Residues P179 and S181 in particular are within the D1 domain of KIR3DL2 and on the opposite face on the KIR3DL2 protein of the HLA-binding regions (i.e. the HLA binding pocket).
  • Each of antibodies 15C1 1 , 19H12, 18B10 and 12B1 1 had reduced binding (full loss of binding for 15C1 1 and 19H12) to mutant M1 1A4 having substitutions E130S, H131 S and R145S..
  • These residues at positions 179 and 181 in mutant 1 1 correspond to the residues present at in KIR3DL1 (KIR3DL1 has T179 and T181 ).
  • Residues P179 and S181 in particular are within the D1 domain of KIR3DL2 and on the opposite face on the KIR3DL2 protein of the HLA-binding regions (i.e. the HLA bind- ing pocket).
  • Surface-exposed residues adjacent to these mutated residues can also contribute to the epitopes of the antibodies, including for example residues N99, H100, E130, H131 , F132, V178, H180, P182, Y183, and Q184 (reference to SEQ ID NO: 1 ) located at the surface of KIR3DL2 in the region of the P179/S181 epitope but outside of the region of the KIR3DL2 mutations which did not result in loss of binding of the antibodies (e.g., mutant 5 (residue P66) and mutant 8 (residues V127)).
  • Antibody 2B12 and other antibodies disclosed in PCT/EP2013/069302 filed 17 September 2013 had loss of binding to mutants having I60N and G62S substitutions and decrease in binding to mutants having P14S, S15A and H23S substitutions, but did not lose binding to any other mutants.
  • the principal epitope of these antibodies therefore includes residues I60 and/or G62 (and the epitope optionally further includes one or more of P14, S15, and H23).
  • Residues 60 and 62 are within the DO domain of KIR3DL2.
  • Residues 14, 15, 23, 60 and 61 are within the DO domain of KIR3DL2.
  • Example 2 Antibodies are able to kill KIR3DL2 expressing targets via antibody dependent cellular cytotoxicity (ADCC)
  • Anti-KIR3DL2 mAbs tested at the same final concentration (10 g/ml), to kill KIR3DL2-transfected B221 target cells.
  • mice were made to express human KIR3DL2.
  • Immune compromised mice used for B221 -KIR3DL2 and RAJI-KIR3DL2 models were NOD-SCID purchased from Charles River Laboratories.
  • 5 million human B221 - KIR3DL2 or RAJI-KIR3DL2 tumor cells (in 100 ⁇ PBS as vehicle) were engrafted IV on Day 0 (DO), i.e. 1 day before treatment initiation (D1 ). From D1 , mice were treated IV with differ- ent doses of anti-KIR3DL2 mAbs (doses were adapted to mouse body weight) diluted in PBS, 2 injections per week for the duration of the whole experiment.
  • mice injected with the same dose of isotype control-matched mAbs directed against an irrelevant antigen mice injected with the same dose of isotype control-matched mAbs directed against an irrelevant antigen.
  • mice were weighed and observed for clinical signs every 2 to 5 days depending on the model. Percent of body weight changes were calculated as compared to body weight at DO before tumor engraftment or to the highest body weight reached during the experiment. Mouse deaths or important weight losses were recorded and used to draw survival Kaplan- Meier curves and calculate improvement in survival as compared to control groups of mice.
  • Example 4 Improved detection methods reveal KIR3DL2 positive tumors
  • Tumor biopsies from RAJI-KI R3DL2 models and RAJI-KIR3DL2 cell lines were obtained and staining was performed on frozen sample using AZ158 antibody (see WO2010/081890) or antibodies 12B1 1 (see Example 1 ).
  • KIR3DL2 was stained with anti- KIR3DL2 antibody by DAB chromogenic detection according to standard protocols, adapted for immunostaining with BenchMark XT Ventana Roche. For all staining control isotype (mlgG1 ) and control DAB were performed.
  • AZ158 was negative, tumors were positive when using 12B1 1 antibody at the same concentration (5 ⁇ g ml) of antibody (see Figure 1 ). Raising concentrations of antibody AZ158 (to 50 ⁇ g ml) generated extensive background staining that did not allow tumor samples to be differentiated from healthy tissue.
  • Tumor biopsies from PTCL patients were obtained and staining was performed on frozen samples.
  • KIR3DL2 was stained with anti-KIR3DL2 antibody 12B1 1 (mlgG1 ) by DAB chromogenic detection according to standard protocols, adapted for immunostaining with BenchMark XT Ventana Roche.
  • control isotype mlgG1
  • control DAB were performed.
  • CD30 was additionally stained.
  • Tumors 3, 4 and 5 were from the same patient.
  • Tumors 1 -5 are from patients having PTCL not otherwise specified.
  • Tumors 6-8 are mycosis fungoides samples, a cutaneous T cell lymphoma (CTCL).
  • CTCL cutaneous T cell lymphoma
  • Tumor sample characteristics are shown in Table B.
  • PTCL from each of the samples from patient from which tumor samples 3, 4 and 5 were obtained had strong membranar staining, with a high percentage of cells being KIR3DL2 positive.
  • KIR3DL2 was stained with anti-KIR3DL2 antibody linked to phycoerythrin (PE), Additional markers evaluated were hCD56 PE, hCD183/CXCR3 PE, hCD3 PE, hCD4 PE, hCD8 PE and CD54 / ICAM PE. Cells were harvested and stained using PE-labeled antibodies. After two washes, stainings were acquired on a BD FACS Canto II and analyzed using the FlowJo software.
  • PE phycoerythrin
  • Results are shown in Figure 3.
  • Anti-KIR3DL2 antibody showed strong staining on the MEC04 cells.
  • MEC04 cells were additionally positive for staining with CD183 (CXCR3), CD56 and CD54 (ICAM), but not CD3, CD4 or CD8 (the most common phenotype of extranodal NK-/T lymphomas are surface CD3- and CD56+).
  • NK-/T-lymphoma cells and in particular extranodal NK-/T cell lymphoma, nasal type, can therefore express KIR3DL2 at significant levels, thereby providing the possibility to treat such disease with anti-KIR3DL2 antibodies.
  • KIR3DL2-positive NK-/T lym- phoma tumors were found to express CD183 (CXCR3), CD56 and CD54 (ICAM), which may permit administration of anti-KIR3DL2 in poor prognosis patients, for example those having CXCR3 expression typically associated with poor disease prognosis.
  • IHC immunohistochemistry
  • Example 6 - KIR3DL2 is expressed in coeliac disease, refractory type 1 Sprue and refractory type 2 Sprue
  • Biopsies from patients with coeliac disease, refractory type 1 Sprue (RCD1 ) and refractory type 2 Sprue (RCDII) were obtained and staining was performed on frozen samples.
  • KIR3DL2 was stained with anti-KIR3DL2 antibody 12B1 1 (mlgG1 ) by DAB chromo- genic detection according to standard protocols, adapted for immunostaining with Bench- Mark XT Ventana Roche. For all staining control isotype (mlgG1 ) and control DAB were per- formed. Results are shown in Table C below.
  • KIR3DL2 is found expressed at all stages.
  • KIR3DL2 may be a relevant target for antibody-based therapies at all stages, from gluten-free diet responding coeliac disease to aggressive, lymphoma-stages like refractory type 2 sprue and EATL.
  • Depletion of KIR3DL2-expressing pathologic cells infiltrating digestive epithelia may reduce local inflammation, may improve digestive function that is strongly impaired in patient, and may prevent evolution of the disease from earlier to later stages. In particular, it may prevent evolution to EATL.
  • KIR3DL2 expression may also be useful to monitor coeliac disease progression towards refractory and tumor stages with poorer prognosis and improve patient therapeutic management. Table A

Abstract

This invention relates to the use of KIR3DL2-targeting agents for the diagnosis and treatment of celiac disease, including refractory celiac disease.

Description

TREATMENT OF CELIAC DISEASE
CROSS-REFERENCE TO RELATED APPLICATIONS
This application claims the benefit of U.S. Provisional Application Nos. 62/042,486, filed 27 August, 2014; all of which are incorporated herein by reference in their entirety; in- eluding any drawings.
REFERENCE TO SEQUENCE LISTING
The present application is being filed along with a Sequence Listing in electronic format. The Sequence Listing is provided as a file entitled "KIR-6_ST25", created 21 August 2015, which is 39 KB in size. The information in the electronic format of the Sequence List- ing is incorporated herein by reference in its entirety.
FIELD OF THE INVENTION
This invention relates to the use of KIR3DL2-targeting agents for the diagnosis and treatment of celiac disease.
BACKGROUND OF THE INVENTION
Celiac disease is an immune-based reaction to dietary gluten (storage protein for wheat, barley, and rye) that primarily affects the small intestine in those with a genetic predisposition and resolves with exclusion of gluten from the diet. The presence of repeated motifs rich in glutamine and proline make gluten proteins a good substrate for tissue transglutaminases (TTG). Due to its deamidation properties, TTG introduces negative charges into the gluten peptides, thus increasing their affinity for the peptide pocket of HLA-DQ2/DQ8 molecules and promoting the formation of complexes that are recognized by CD4+ T lymphocytes. The peptides, in genetically predisposed individuals expressing HLA-DQ2/DQ8, thus activate CD4+ T cells which leads to an inflammatory process involving intraepithelial lymphocytes in the intestinal epithelium.
Celiac disease is usually detected by serologic testing of celiac-specific antibodies, notably to transglutaminase or gliadin. The diagnosis is confirmed by duodenal mucosal biopsies. The treatment for celiac disease is primarily a gluten-free diet (GFD), which can be difficult to maintain on a lifelong basis, and involves significant monitoring and follow-up. Also, due to the nature and variety of the symptoms, many patients remain undiagnosed. Fur- thermore, even when patients are able to maintain a GFD over the long term, many patients have non-responsive disease, particularly in those diagnosed in adulthood. Consequently, there are many patients that have refractory celiac disease (RCD), whether due to bad observance of GFD or authentic RCD with persistent malabsorption and intestinal villous atrophy. RCD had been divided into two subgroups, Type I (RCDI) and Type II (RCDII). Type I is characterized by persisting villous atrophy with an increased number of intraepithelial lymphocytes (lELs) bearing a normal phenotype and polyclonal repertoire. Type II is characterized by a clonal expansion of lELs lacking surface CD3 and generally CD8, and having intracellular CD3e. Non-controlled CD such as RCD leads to complications including bone and autoimmune disease, and to lymphomas, notably enteropathy-associated T lymphoma (EATL). RCD is often considered to be a low-grade intraepithelial T cell lymphoma, an intermediate between celiac disease and high grade invasive T cell lymphoma. RCDII is generally considered a poor prognosis because it is frequently associated with over lymphoma such as EATL for which median survival is only 7 months.
There is therefore a need in the art for improved benefit to patients having celiac disease.
SUMMARY OF THE INVENTION
The present inventors have discovered that KIR3DL2 is expressed on the surface of celiac disease (coeliac disease; CD), including celiac disease and advanced or refractory celiac disease stages. KIR3DL2 is also expressed on enteropathy-associated T-cell lym- phoma (EATL), a complication of CD. In KIR3DL2-positive celiac disease, membranar KIR3DL2 expression permits targeting with KIR3DL2-binding antibodies (e.g. as assessed by immunohistochemistry). KIR3DL2 is expressed on few other tissues (only on a small fraction of healthy NK and T cells), permitting KIR3DL2 to serve as a marker and target for the detection and treatment of celiac disease, particularly refractory celiac disease. Accordingly, in one embodiment, provided is a method for treating or preventing a celiac disease or a complication thereof (e.g. a lymphoma, an EATL) in an individual, the method comprising administering to an individual a therapeutically active amount of a compound that binds a KIR3DL2 polypeptide. The individual can be an individual having or suspected of having CD or who is susceptible to having CD. The individual can be an individual having or suspected of having CD and who has a poor clinical prognosis, e.g. who has a high likelihood to progress to RCD, RCDI, or RCDII, or to have a complication such as EATL, or who has progressed to RCD, RCDI or RCDII . The individual can be an individual having or suspected of having CD or who is susceptible to having CD. In one aspect provided is a compound that binds a KIR3DL2 polypeptide, for use in the treatment or prevention of celiac disease. In one embodiment, provided is a method for treating an individual having a refractory, progressing or advanced celiac disease (e.g. RCD, RCDI or RCDII), the method comprising administering to an individual a therapeutically active amount of a compound that binds a KIR3DL2 polypeptide. In one aspect the compound that binds a KIR3DL2 polypeptide is capable of depleting a cell that expresses KIR3DL2 at its surface, e.g. a celiac disease cell that expresses KIR3DL2 on its surface (e.g., an intraepithelial lymphocyte). In one aspect the compound is a depleting anti-KIR3DL2 antibody. In one embodiment provided is a compound that binds a KIR3DL2 polypeptide and depletes KIR3DL2-expressing cells, for use in the treatment or prevention of a celiac disease in an individual. Optionally the treatment or prevention comprises admin- istration of the compound that binds a KIR3DL2 polypeptide to an individual having a celiac disease. In one embodiment of any of the therapeutic uses or celiac disease treatment or prevention methods herein, the individual has a refractory, progressing or advanced celiac disease, optionally wherein the celiac disease is an RCD, an RCDI, and RCDII, or a complication thereof, e.g. an enteropathy associated T cell lymphoma (EATL).
In one embodiment, the treatment or prevention of a celiac disease in an individual comprises:
a) determining whether the individual has celiac disease (e.g. CD, RCD, RCDI, or RCDII) or a complication thereof (e.g. a lymphoma, an EATL), and
b) upon a determination that the individual has celiac disease or a complication thereof, administering to the individual a compound that binds a KIR3DL2 polypeptide. In one embodiment, the celiac disease is RCDI. In one embodiment, the celiac disease is RCDII. In one embodiment, the celiac disease is RCDII, and the step of determining whether the individual has celiac disease comprises detecting the presence of abnormal lELs, optionally further wherein the abnormal lELs are abnormal lELs expressing KIR3DL2 at their surface.
In one embodiment, the treatment or prevention of a celiac disease in an individual comprises:
a) determining the KIR3DL2 polypeptide status of cells (e.g., intraepithelial lymphocytes, abnormal intraepithelial lymphocytes) within the individual having a celiac disease (e.g. CD, RCD, RCDI, or RCDII) or a complication thereof (e.g. a lymphoma, an EATL), and b) upon a determination that the individual has KIR3DL2 polypeptides expressed on the surface of cells (e.g., intraepithelial lymphocytes, abnormal intraepithelial lymphocytes), administering to the individual said compound that binds a KIR3DL2 polypeptide. Step (b) can optionally comprise administering to the individual said compound that binds a KIR3DL2 polypeptide upon a determination that the individual has KIR3DL2 polypeptides that are prominently expressed on the surface of cells. Expression across a high percentage of ab- normal intraepithelial lymphocytes may occur with clonal expansion, e.g. in RCDII, permitting the disease to be particularly suitable to treatment with an anti-KIR3DL2 agent.
Additionally, antibodies are provided that are particularly effective in diagnostic or prognostic assays to detect KIR3DL2 expression on abnormal lymphocytes, notably lELs, and notably in immunohistochemistry assays. The antibodies are capable of detecting mem- branar KIR3DL2 in cases where prior antibodies were not able to detect such KIR3DL2 expression, including with specificity over KIR3DL1 .
In another embodiment provided is a method comprising a KIR3DL2 detection step to identify patients having KIR3DL2+ celiac disease; these patients can thereafter be treated with a KIR3DL2-binding agent. Such method permits KIR3DL2 therapy to be more precisely directed to patients without reliance on disease staging. Such method also helps permit the prevention of advanced celiac disease (e.g. prevention of progression of celiac disease to an advanced stage, e.g. RCD or EATL) because patients can be treated as KIR3DL2 appears.
In a further aspect, it has been found that patients with celiac disease can be KIR3DL2-positive despite having a disease that has been shown to be - CD30-negative (the abnormal lELs do not express CD30 on their surface). Thus, provided are methods of treating a CD30-negative celiac disease, e.g. a RCD, comprising administering a compound that binds a KIR3DL2 polypeptide to a patient having CD30-negative celiac disease.
In another embodiment of treating an individual having a celiac disease, the meth- ods or uses comprise administering a compound that binds a KIR3DL2 polypeptide to an individual having a celiac disease who is refractive to treatment, e.g. with a gluten free diet (GFT), with an immunomodulatory or immunosuppressive agent (e.g. an agent that inhibits the IL-15 signalling pathway, an anti-IL-15 antibody, a protein kinase inhibitor, an agent that inhibits JAK3 signalling), with a chemotherapeutic agent, and/or with a hematopoetic stem cell transplantation. In other embodiments, a celiac disease a compound that binds a KIR3DL2 polypeptide can be administered in combination with a second treatment for CD (e.g., a gluten free diet (GFT), an immunomodulatory or immunosuppressive agent (e.g. an agent that inhibits the IL-15 signalling pathway, an anti-IL-15 antibody, a protein kinase inhibitor, an agent that inhibits JAK3 signalling), a chemotherapeutic agent, and/or a hematopoet- ic stem cell transplantation).
In one embodiment, provided is a method for detecting a celiac disease in an individual, the method comprising detecting a KIR3DL2 nucleic acid or polypeptide in a biological sample (e.g. on a cell) from the individual (e.g. an individual having CD, suspected of having CD, having a symptom associated with CD, etc.). In one embodiment, provided is a method for detecting a progressing, advanced, and/or refractory celiac disease in an individual, the method comprising detecting a KIR3DL2 nucleic acid or polypeptide in a biological sample (e.g. on a cell) from the individual. A determination that a biological sample expresses KIR3DL2 indicates that the patient has a celiac disease (or progressing, advanced, and/or refractory celiac disease). In one embodiment, a CD is an RCD, optionally an RCDI, optional- ly and RCDII. A determination that a biological sample expresses KIR3DL2 can indicate that the patient is at risk of developing or has developed EATL, e.g. the patient has a celiac disease that is likely to give rise to/progress to EATL. In one embodiment, the method comprises determining the level of expression of a KIR3DL2 nucleic acid or polypeptide in a biological sample and comparing the level to a reference level (e.g. a value, weak cell surface stain- ing, etc.) corresponding to a healthy individual. A determination that a biological sample expresses KIR3DL2 nucleic acid or polypeptide at a level that is increased compared to the reference level indicates that the patient has a celiac disease (or progressing, advanced, and/or refractory celiac disease, or is at risk of developing or has developed EATL). Optionally, detecting a KIR3DL2 polypeptide in a biological sample comprises detecting KIR3DL2 polypeptide expressed on the surface of an infiltrating lymphocyte (e.g., an I EL).
In one embodiment, provided is a method comprising: (a) determining whether an individual has a celiac disease (e.g. CD, RCD, RCDI, RCDII); and (b) if the individual has a celiac disease, determining whether an individual has celiac disease cells (e.g., intraepithelial lymphocytes) that express a KIR3DL2 polypeptide. The method may optionally further com- prise treating the individual with a therapeutically active amount of a compound that binds a KIR3DL2 polypeptide if the individual has celiac disease cells that express KIR3DL2 on their surface. In one embodiment, the celiac disease is RCD or RCDI. In one embodiment, the celiac disease is RCDII.
In one embodiment of any of the methods, determining whether an individual has celiac disease cells that express a KIR3DL2 polypeptide comprises obtaining a biological sample from the individual that comprises celiac disease cells, bringing said cells into contact with an antibody that binds a KIR3DL2 polypeptide, and detecting whether cells express KIR3DL2 on their surface.
Optionally, in any embodiment, determining whether an individual has celiac disease cells that express KIR3DL2 comprises conducting an immunohistochemistry assay, e.g. an immunohistochemistry assay comprising obtaining from an individual a biological sample that comprises disease cells (e.g. abnormal lELs), fixing and sectioning said sample to obtain a tissue section, bringing said tissue section into contact with an antibody (e.g. an antibody that competes with antibody 12B1 1 for binding to a human KIR3DL2 polypeptide) and detecting expression of KIR3DL2 (e.g. detecting cells that express KIR3DL2). In one embodiment, the tissue section is a frozen tissue section. In another embodiment, determining whether an individual has celiac disease cells that express KIR3DL2 comprises conducting a flow cytometry assay. Both IHC and flow cytometry can detect surface expression of KIR3DL2.
Also provided is a method of treating a patient with a celiac disease, the method comprising a) determining the KIR3DL2 polypeptide status of disease cells (e.g. celiac disease cells; abnormal lymphocytes) within the patient, e.g. determining whether a KIR3DL2 polypeptide is prominently expressed on the surface of said disease cells, and b) administering a compound to the patient that specifically binds to a KIR3DL2 polypeptide that is prominently expressed in said disease cells (e.g. prominently expressed on the surface of disease cells). Optionally, determining the KIR3DL2 polypeptide status comprises determining whether a KIR3DL2 polypeptide that is prominently expressed on the surface of said malignant cells. Optionally, determining whether a KIR3DL2 polypeptide that is prominently expressed on the surface of said disease cells comprises obtaining from the individual a biological sample that comprises celiac disease cells (e.g., abnormal lymphocytes), bringing said cells into contact with an antibody that binds a KIR3DL2 polypeptide, and detecting cells that express KIR3DL2 (e.g. determining the number or portion of cells that express KIR3DL2).
Preferably the compound that binds a KIR3DL2 polypeptide is a compound that causes the death of a KIR3DL2-expressing cell. Optionally, the compound that binds a KIR3DL2 polypeptide is an antigen binding polypeptide, optionally an antibody (e.g. mono- clonal antibody), that binds a KIR3DL2 polypeptide, optionally a polypeptide or other compound that is a natural ligand of KIR3DL2 (e.g. an HLA polypeptide, or fragment or derivative thereof). Optionally, the antibody is a depleting polypeptide (antibody). Optionally, the antibody is an antibody that directs ADCC and/or CDC toward a KIR3DL2-expressing cell. Optionally, the antibody is an antibody that delivers a cytotoxic agent (e.g. small molecule) to a KIR3DL2-expressing cell.
In one embodiment, the antibody used in any embodiment herein binds a KIR3DL2 polypeptide, optionally, wherein the antibody does not substantially bind to a KIR3DL1 polypeptide and has bivalent binding affinity (KD) for a human KIR3DL2 polypeptide at of less than 10"8 M. In one embodiment, the antibody binds a KIR3DL2 polypeptide in its D1 domain. In one embodiment, the antibody binds a KIR3DL2 polypeptide, wherein said antibody does not substantially bind to a KIR3DL1 polypeptide, and wherein said antibody binds to at least one residue in the segment corresponding to residues 99-192 of the mature KIR3DL2 polypeptide of SEQ ID NO: 1.
In one embodiment, the antibody used herein competes for binding to a KIR3DL2 polypeptide with an antibody selected from the group consisting of: (a) an antibody having respectively a VH and VL region of SEQ ID NOS: 5 and 6
(19H12),
(b) an antibody having respectively a VH and VL region of SEQ ID NOS: 16 and 17 (12B1 1 ); or
(b) an antibody having respectively a VH and VL region of SEQ ID NOS: 33 and 34
(2B12).
Optionally, the antibody binds an epitope comprising residues P179 and/or residue S181 of the KIR3DL2 polypeptide of SEQ ID NO: 1 , and/or has reduced binding to a KIR3DL2 polypeptide having a mutation at residues P179 and/or residue S181 of SEQ ID NO: 1 , compared to a wild-type KIR3DL2 polypeptide of SEQ ID NO: 1 .
Optionally, the antibody used herein binds an epitope comprising residues N99, H100, E130, H131 , F132, V178, H180, P182, Y183 and/or Q184 of SEQ ID NO: 1 , and/or has reduced binding to a KIR3DL2 polypeptide having a mutation at residues N99, H100, E130, H131 , F132, V178, H180, P182, Y183 and/or Q184 of SEQ ID NO: 1 , compared to a wild-type Kl R3DL2 polypeptide of SEQ I D NO: 1 .
Optionally, the antibody (e.g. antibody 2B12 or an antibody that competes therewith for binding to KIR3DL2) binds an epitope comprising residues I60 and/or residue G62 of the KIR3DL2 polypeptide of SEQ ID NO: 1 , and/or has reduced binding to a KIR3DL2 polypeptide having a mutation at residues I60 and/or residue G62 of SEQ ID NO: 1 , compared to a wild-type KIR3DL2 polypeptide of SEQ ID NO: 1. Optionally, the antibody binds an epitope comprising residues P14, S15 and/or residue H23 of the KIR3DL2 polypeptide of SEQ ID NO: 1 , and/or has reduced binding to a KIR3DL2 polypeptide having a mutation at residues P14, S15 and/or residue H23 of SEQ ID NO: 1 , compared to a wild-type KIR3DL2 polypeptide of SEQ ID NO: 1. Optionally, the antibody used herein binds an epitope comprising resi- dues l60, G62, P14, S15 and/or residue H23 of SEQ ID NO: 1 .
Optionally, the compound that binds a KIR3DL2 polypeptide is administered between once daily and once per month. Optionally, the composition is administered as monotherapy. Optionally, the composition is administered in combination with a second therapeutic agent. Optionally, the composition is administered in combination with an anti-celiac dis- ease treatment or agent.
In one embodiment, provided is a method of producing a composition for the treatment of celiac disease or for use in the prevention of celiac disease in a mammalian subject (e.g. in an individual who is susceptible to having CD), said method comprising the steps of: a) providing a plurality of test compositions; b) testing each compound for the ability to bind KIR3DL2 and/or cause the depletion of KIR3DL2-expressing cells; and c) selecting a com- pound which binds a KIR3DL2 polypeptide and/or causes the depletion of KIR3DL2- expressing cells as suitable for the treatment of celiac disease or for use in the prevention of celiac disease.
Optionally, the method further comprises producing a quantity of the compound se- lected in step c), and/or formulation a quantity of the compound selected in step c) with a pharmaceutically acceptable excipient.
Optionally, step b) further comprises testing said test composition for the ability to direct ADCC and/or CDC toward a KIR3DL2-expressing cell.
In one embodiment of any of the methods herein, a celiac disease is a refractory ce- liac disease (RCD), optionally RCDI, optionally RCDII.
In one embodiment, provided is a method comprising: (a) determining whether an individual has celiac disease (e.g., CD, RCD, RCDI, or RCDII) or a complication thereof (e.g. a lymphoma, an EATL); and (b) if the individual has celiac disease or a complication thereof, treating the individual with a therapeutically active amount of a compound that binds a KIR3DL2 polypeptide.
In one embodiment of any of the methods herein, determining whether an individual has a celiac disease is made according to standard medical guidelines. In one embodiment of any of the methods herein, determining whether an individual has a celiac disease (e.g., CD, RCD, RCDI, or RCDII) comprises conducting a small bowel investigation, enteroscopy, small bowel imaging, a laboratory test, immunohistochemistry, molecular analysis, and/or flow cytometry.
In one embodiment of any of the methods herein, determining whether an individual has a celiac disease (e.g., CD, RCD, RCDI, or RCDII) or a complication thereof comprises identifying a population of abnormal cells or abnormal numbers of cells (e.g., abnormal lELs). Optionally, said identification is by flow cytometry or immunohistochemistry. Optionally, the method further comprises sorting or isolating the population of abnormal cells.
In one embodiment of any of the methods herein, determining whether an individual has or is susceptible of having a celiac disease (e.g., CD, RCD, RCDI, or RCDII) or a complication thereof (e.g. a lymphoma, an EATL) comprises detecting a genetic predisposition. In one embodiment, the presence of HLA DQ heterodimers DQ2 and/or DQ8 is assessed, e.g., the HLA DQ2/DQ8 genotype is assessed, wherein presence of HLA DQ heterodimers DQ2 and/or DQ8 (e.g. HLA DQ2/DQ8 genotype) indicates a predisposition to CD. In one embodiment, detecting a genetic predisposition comprises contacting nucleic acid isolated from a subject with one or more oligonucleotides, wherein the contacting determines the presence of a genetic marker of predisposition to CD (e.g. CD, RCD, poor prognosis, disease progres- sion, etc.); thereby detecting whether an individual has or is susceptible of having a celiac disease.
Optionally, the method further comprises a step of assessing, following treatment with a compound that binds a KIR3DL2 polypeptide, whether the individual has an ameliora- tion in celiac disease, e.g., whether the individual has decreased numbers of celiac disease cells (e.g., abnormal lELs).
In one embodiment of any aspect herein, the celiac disease is a refractory celiac disease. In one embodiment, the celiac disease is an RCDI. In one embodiment, the celiac disease is an RCDII. In one embodiment, the individual having disease has a poor prognosis, e.g. for resistance to therapy, for occurrence of a complication such as a lymphoma or EATL, for survival. In one embodiment, the celiac disease is progressing disease. . In one embodiment, the celiac disease is susceptible of giving rise to a lymphoma, e.g. an EATL.
In one embodiment, provided is a method for diagnosing or monitoring a celiac disease in an individual, the method comprising obtaining from an individual having (or suspect- ed of having, or susceptible to having) CD a biological sample that comprises cells (e.g., intraepithelial lymphocytes), bringing said cells into contact with an antibody that binds a human KIR3DL2 polypeptide, and detecting cells that express KIR3DL2. Optionally, the antibody that binds a KIR3DL2 polypeptide is an antibody that binds a human KIR3DL2 polypeptide but does not bind to a human KIR3DL1 polypeptide. Optionally, the antibody that binds a KIR3DL2 polypeptide competes with antibody 12B1 1 for binding to a human KIR3DL2 polypeptide (e.g. antibody 19H12).
In one embodiment, provided is a method for determining whether a KIR3DL2 polypeptide is expressed on the surface of a I EL (e.g. an I EL from an individual having, susceptible to or suspected of having CD), the method comprising obtaining from an individual a bio- logical sample that comprises lELs, bringing said lELs into contact with an antibody that competes with antibody 12B1 1 for binding to a human KIR3DL2 polypeptide (e.g. antibody 19H12), and detecting lELs that express KIR3DL2. Optionally, the method further comprises determining whether the lELs express any one or more of: T cell receptor (TCR), CD3e, CD8, CD103 and CD45
In one embodiment, provided is a method for determining whether a KIR3DL2 polypeptide is expressed on the surface of a cell (e.g. an I EL), the method comprising obtaining from an individual (e.g. having, susceptible to or suspected of having celiac disease) a biological sample (e.g., tissue sample) that comprises cells, fixing and sectioning said sample to obtain a tissue section, bringing said tissue section into contact with an antibody that com- petes with antibody 12B1 1 for binding to a human KIR3DL2 polypeptide (e.g. antibody 19H12), and detecting expression of KIR3DL2 (e.g. detecting cells that express KIR3DL2). In one embodiment, the tissue section is a frozen tissue section.
In one embodiment, provided are antibodies having advantageous uses in diagnostic and prognostic methods for celiac disease and other diseases. Provided is a method com- prising:
(a) obtaining a biological sample of cells and bringing such cells into contact with antibody 19H12 or a derivative or fragment thereof, an antibody that competes therewith for binding to KIR3DL2, or an antibody that binds residues P179 and/or residue S181 on a KIR3DL2 polypeptide, optionally wherein said antibody is labelled with a detectable moiety;
(b) determining by flow cytometry whether said antibody binds to said cells, wherein binding indicates that the cells express KIR3DL2 on their surface.
In another embodiment, provided is a method comprising:
(a) obtaining a biological sample of cells, preparing frozen tissue section from such cells, and bringing such sections into contact with antibody 12B1 1 or a derivative or fragment thereof, an antibody that competes therewith for binding to KIR3DL2, or an antibody that binds residues P179 and/or residue S181 on a KIR3DL2 polypeptide, optionally wherein said antibody is labelled with a detectable moiety;
(b) determining whether said antibody binds to said cells, wherein binding indicates that the cells express KIR3DL2 on their surface.
The present disclosure further concerns a method for diagnosing a disease state mediated by pathogenic KIR3DL2-expressing cells, said method comprising the steps of combining with an ex vivo patient sample (from an individual having, susceptible to or suspected of having celiac disease) a composition comprising a conjugate or complex comprises an antibody that binds specifically to KIR3DL2 expressed on the surface of the pathogenic cells and an imaging agent, and detecting the pathogenic cells that express a receptor for the lig- and using flow cytometry.
The present disclosure further concerns a method of determining a prognosis of a celiac disease by detecting cells in an ex vivo patient sample, said method comprising the steps of: (a) combining with an ex vivo patient sample a composition comprising a conjugate or complex comprises an antibody (e.g. antibody 19H12) that binds specifically to KIR3DL2 expressed on the surface of the pathogenic cells and an imaging agent, (b) detecting the pathogenic cells that express a receptor for the ligand using flow cytometry, and (c) determining a prognosis for the celiac disease.
The present disclosure further concerns a method for quantitating pathogenic cells from an individual having, susceptible to or suspected of having celiac disease, said method comprising the steps of: (a) combining, with an ex vivo patient sample, a conjugate or complex which comprises (i) an antibody that binds specifically to KIR3DL2 expressed on the surface of the pathogenic cells (e.g. antibody 19H12) and (ii) an imaging agent, and (b) quantitating said pathogenic cells in the ex vivo patient sample using flow cytometry.
In any of the above flow-cytometry based methods, the antibody binds to a KIR3DL2 polypeptide on the surface of cells but not to a KIR3DL1 polypeptide. Optionally, said pathogenic cells are detected by single photon flow cytometry. Optionally, said pathogenic cells are detected by multiphoton flow cytometry. Optionally, wherein the ex vivo patient sample is a patient body fluid. Optionally, the body fluid is selected from the group consisting of spinal fluid, lymph fluid, urine, mucus, and blood. Optionally, the pathogenic cells are lELs. Optionally, the antibody conjugated to an imaging agent is selected from the group consisting of anti-KIR3DL2-fluorescein, anti-KIR3DL2-Oregon Green, anti-KIR3DL2-rhodamine, anti- KIR3DL2- phycoerythrin, anti-KIR3DL2-cys-Texas Red, anti-KIR3DL2-AlexaFluor, and anti- KIR3DL2-Dyl_ight. Optionally, the imaging agent comprises a chromophore. Optionally, the chromophore is a fluorescent chromophore. Optionally, the chromophore comprises a compound selected from the group consisting of fluorescein, Oregon Green, rhodamine, phycoerythrin, Texas Red, DyLight 680, and AlexaFluor 488. Optionally, the methods further comprise the step of quantitating the pathogenic cells in the ex vivo patient sample.
In any of the above flow-cytometry based methods, the antibodies optionally bind to each of the KIR3DL2 polypeptides having the amino acid sequence shown in SEQ ID NOS: 1 , 27 and 29 (alleles_*002, *001 and *007, respectively). In one embodiment, the antibodies bind to each of the KIR3DL2 polypeptides having the amino acid sequence shown in SEQ ID NOS: 27 and 31 (alleles_*001 and *009, respectively). In one embodiment, the antibodies bind to each of the KIR3DL2 polypeptides having the amino acid sequence shown in SEQ ID NOS: 27, 1 , 29 and 31 (alleles_*001 , *002, *007 and *009, respectively). In one embodiment, the antibodies bind to each of the KIR3DL2 polypeptides having the amino acid sequence shown in SEQ ID NOS: 27, 1 , 2, 28 and 29 (alleles_*001 , *002, *003, *005 and *007, respectively). In one embodiment, the antibodies bind to each of the KIR3DL2 polypeptides having the amino acid sequence shown in SEQ ID NOS: 27, 1 , 29 and 30 (alleles_*001 , *002, *007 and *008, respectively). In one embodiment, the antibodies bind to each of the KIR3DL2 polypeptides having the amino acid sequence shown in SEQ ID NOS: 27, 1 , 2, 28, 29 and 30 (alleles_*001 , *002, *003, *005, *007 and *008, respectively).
In any of the above flow-cytometry based methods, the antibody binds an epitope comprising one, two, three, four, five or more of residues selected from the group consisting of: M128, E130, H131 , R145, V147, Q149, 1150, V178, P179, H180 and S181 (with reference to SEQ ID NO: 1 ), and/or the antibody may or may not have reduced binding to a KIR3DL2 polypeptide having a mutation at a residue selected from the group consisting of: M128, E130, H131 , R145, V147, Q149, 1150, V178, P179, H180 and/ S181 (with reference to SEQ ID NO: 1 In any of the above flow-cytometry based methods, the antibody binds an epitope comprising residues P179 and/or S181 of the KIR3DL2 polypeptide, and/or has reduced binding to a KIR3DL2 polypeptide having a mutation at residues P179 and/or S181 (with reference to SEQ ID NO: 1 , e.g. a P179T, S181T mutant). In one aspect the antibody binds an epitope comprising residues V178 and/or H180 of the KIR3DL2 polypeptide, and/or has reduced binding to a KIR3DL2 polypeptide having a mutation at residues V178 and/or H180 (with reference to SEQ ID NO: 1 , e.g. a V178A, H180S mutant). In one aspect the antibody binds an epitope comprising residues E130, H131 and/or R145 of the KIR3DL2 polypeptide, and/or have reduced binding to a KIR3DL2 polypeptide having a mutation at residues E130, H131 and/or R145 (with reference to SEQ ID NO: 1 , e.g. a E130S, H131 S, R145S mutant). In any of the above flow-cytometry based methods, the antibody is an antibody that competes with, and/or that comprises the heavy and/or light chain CDRs 1 , 2 and/or 3 of, antibody 19H12 or 12B1 1.
These aspects are more fully described in, and additional aspects, features, and advantages will be apparent from, the description of the invention provided herein.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 shows staining of frozen tissue sections from RAJI-KIR3DL2 mouse tumor models and RAJI-KIR3DL2 cell lines, using AZ158 antibody (see WO2010/081890) or antibodies 12B1 1. While AZ158 was negative, tumors were positive when using 12B1 1 antibody at the same concentration (5 g/ml) of antibody.
Figure 2 shows staining of frozen tissue sections from cancer patients previously stained with AZ158, re-examined using antibody 12B1 1. Biopsies that had been KIR3DL2- negative with AZ158 were stained with 12B1 1 (i.e. becoming KIR3DL2-positive).
Figure 3 shows staining by anti-KIR3DL2 antibody on NK-/T- lymphoma cells, nasal type. The figure additionally shows that the KIR3DL2-positive cells express CD183 (CXCR3), CD56 and CD54 (ICAM).
DESCRIPTION OF THE INVENTION
The identification expression of KIR3DL2 polypeptides at the surface of celiac disease cells permits the development of therapeutic agents that are able to directly and specif- ically target pathogenic cells, as well as diagnostic agents that can be used to diagnose and monitor celiac disease.
Provided are methods of using the antigen-binding compounds; for example, provided is a method for inhibiting celiac disease, for delivering a molecule to a celiac disease cell (e.g. a toxic molecule, a detectable marker, etc.), for targeting, identifying or purifying a cell, for depleting, killing or eliminating a cell, for reducing cell proliferation, the method comprising exposing a cell, such as a celiac disease cell (e.g., an IEL, an abnormal IEL) which expresses a KIR3DL2 polypeptide, to a compound that binds a KIR3DL2 polypeptide. It will be appreciated that for the purposes herein, "cell proliferation" can refer to any aspect of the growth or proliferation of cells, e.g., cell growth, cell division, or any aspect of the cell cycle. The cell may be in cell culture (in vitro) or in a mammal (in vivo), e.g. a mammal suffering from celiac disease. Also provided is a method for inducing the death of a cell or inhibiting the proliferation or activity of a celiac disease cell (e.g., an IEL, an abnormal IEL) which expresses a KIR3DL2 polypeptide, comprising exposing the cell to an antigen-binding com- pound that binds a KIR3DL2 polypeptide in an amount effective to induce death and/or inhibit the proliferation of the cell.
Antibodies specific for KIR3DL2 can be used for a range of purposes for the diagnosis or treatment of celiac disease, including purifying KIR3DL2 or KIR3DL2-expressing cells in patients having celiac disease, suspected of having celiac disease or susceptible to celiac disease, targeting KIR3DL2-expressing cells for destruction in vivo, or specifically labeling/binding KIR3DL2 in vivo, ex vivo, or in vitro, cells in patients having celiac disease, suspected of having celiac disease or susceptible to celiac disease, including in methods such as immunoblotting, IHC analysis, i.e. on frozen biopsies, FACS analysis, and immunoprecipi- tation.
As used herein, "a" or "an" may mean one or more. As used in the claim(s), when used in conjunction with the word "comprising", the words "a" or "an" may mean one or more than one. As used herein "another" may mean at least a second or more.
Where "comprising" is used, this can optionally be replaced by "consisting essentially of" or by "consisting of".
Whenever within this whole specification "treatment of celiac disease" or the like is mentioned with reference to anti-KIR3DL2 binding agent (e.g. antibody), there is meant: (a) method of treatment of celiac disease, said method comprising the step of administering (for at least one treatment) an anti-KIR3DL2 binding agent, (e.g., in a pharmaceutically acceptable carrier material) to a warm-blooded animal, especially a human, in need of such treat- ment, in a dose that allows for the treatment of celiac disease, (a therapeutically effective amount), e.g, in a dose (amount) as specified hereinabove and herein below; (b) the use of an anti-KIR3DL2 binding agent for the treatment of celiac disease, or an anti-KIR3DL2 binding agent, for use in said treatment (especially in a human); (c) the use of an anti-KIR3DL2 binding agent for the manufacture of a pharmaceutical preparation for the treatment of celiac disease, a method of using an anti-KIR3DL2 binding agent for the manufacture of a pharmaceutical preparation for the treatment of celiac disease, comprising admixing an anti- KIR3DL2 binding agent with a pharmaceutically acceptable carrier, or a pharmaceutical preparation comprising an effective dose of an anti-KIR3DL2 binding agent that is appropriate for the treatment of celiac disease; or (d) any combination of a), b), and c), in accordance with the subject matter allowable for patenting in a country where this application is filed.
The term "biopsy" as used herein is defined as removal of a tissue for the purpose of examination, such as to establish diagnosis. Examples of types of biopsies include by application of suction, such as through a needle attached to a syringe; by instrumental removal of a fragment of tissue; by removal with appropriate instruments through an endoscope; by surgical excision, such as of the whole lesion; and the like.
The term "antibody," as used herein, refers to polyclonal and monoclonal antibodies. Depending on the type of constant domain in the heavy chains, antibodies are assigned to one of five major classes: IgA, IgD, IgE, IgG, and IgM. Several of these are further divided into subclasses or isotypes, such as lgG1 , lgG2, lgG3, lgG4, and the like. An exemplary im- munoglobulin (antibody) structural unit comprises a tetramer. Each tetramer is composed of two identical pairs of polypeptide chains, each pair having one "light" (about 25 kDa) and one "heavy" chain (about 50-70 kDa). The N-terminus of each chain defines a variable region of about 100 to 1 10 or more amino acids that is primarily responsible for antigen recognition. The terms variable light chain (VL) and variable heavy chain (VH) refer to these light and heavy chains respectively. The heavy-chain constant domains that correspond to the different classes of immunoglobulins are termed "alpha," "delta," "epsilon," "gamma" and "mu," respectively. The subunit structures and three-dimensional configurations of different classes of immunoglobulins are well known. IgG are the exemplary classes of antibodies employed herein because they are the most common antibodies in the physiological situation and be- cause they are most easily made in a laboratory setting. In one embodiment, an antibody is a monoclonal antibody. Provided are humanized, chimeric, human, or otherwise-human- suitable antibodies. "Antibodies" also includes any fragment or derivative of any of the herein described antibodies.
The term "specifically binds to" means that an antibody can bind in a competitive binding assay to the binding partner, e.g. KIR3DL2, as assessed using either recombinant forms of the proteins, epitopes therein, or native proteins present on the surface of isolated target cells. Competitive binding assays and other methods for determining specific binding are further described below and are well known in the art.
When an antibody is said to "compete with" a particular monoclonal antibody, it means that the antibody competes with the monoclonal antibody in a binding assay using either recombinant KIR3DL2 molecules or surface expressed KIR3DL2 molecules. For example, if a test antibody reduces the binding of AZ158, 19H12, 2B12 or 12B1 1 to a KIR3DL2 polypeptide or KIR3DL2-expressing cell in a binding assay, the antibody is said to "compete" respectively with AZ158, 19H12, 2B12 or 12B1 1.
The term "affinity", as used herein, means the strength of the binding of an antibody to an epitope. The affinity of an antibody is given by the dissociation constant Kd, defined as [Ab] x [Ag] / [Ab-Ag], where [Ab-Ag] is the molar concentration of the antibody-antigen complex, [Ab] is the molar concentration of the unbound antibody and [Ag] is the molar concentration of the unbound antigen. The affinity constant Ka is defined by 1/Kd. Methods for de- termining the affinity of mAbs can be found in Harlow, et al., Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1988), Coligan et al., eds., Current Protocols in Immunology, Greene Publishing Assoc. and Wiley Interscience, N.Y., (1992, 1993), and Muller, Meth. Enzymol. 92:589-601 (1983), which references are entirely incorporated herein by reference. One standard method well known in the art for de- termining the affinity of mAbs is the use of surface plasmon resonance (SPR) screening (such as by analysis with a BIAcore™ SPR analytical device).
A "determinant" designates a site of interaction or binding on a polypeptide.
The term "epitope" refers to an antigenic determinant, and is the area or region on an antigen to which an antibody binds. A protein epitope may comprise amino acid residues directly involved in the binding as well as amino acid residues which are effectively blocked by the specific antigen binding antibody or peptide, i.e., amino acid residues within the "footprint" of the antibody. It is the simplest form or smallest structural area on a complex antigen molecule that can combine with e.g., an antibody or a receptor. Epitopes can be linear or conformational/structural. The term "linear epitope" is defined as an epitope composed of amino acid residues that are contiguous on the linear sequence of amino acids (primary structure). The term "conformational or structural epitope" is defined as an epitope composed of amino acid residues that are not all contiguous and thus represent separated parts of the linear sequence of amino acids that are brought into proximity to one another by folding of the molecule (secondary, tertiary and/or quaternary structures). A conformational epitope is dependent on the 3-dimensional structure. The term 'conformational' is therefore often used interchangeably with 'structural'.
The term "depleting", "deplete" or "depletion", with respect to KIR3DL2-expressing cells means a process, method, or compound that can kill, eliminate, lyse or induce such killing, elimination or lysis, so as to negatively affect the number of KIR3DL2-expressing cells present in a sample or in a subject.
The terms "immunoconjugate", "antibody conjugate", "antibody drug conjugate", and "ADC" are used interchangeably and refer to an antibody that is conjugated to another moiety (e.g. any non-antibody moiety, a therapeutic agent or a label).
The term "agent" is used herein to denote a chemical compound, a mixture of chem- ical compounds, a biological macromolecule, or an extract made from biological materials. The term "therapeutic agent" refers to an agent that has biological activity.
The terms "toxic agent", "toxic moiety" and "cytotoxic agent" encompass any compound that can slow down, halt, or reverse the proliferation of cells, decrease their activity in any detectable way, or directly or indirectly kill them. Cytotoxic agents can cause cell death primarily by interfering directly with the cell's functioning, and include, but are not limited to, alkylating agents, tumor necrosis factor inhibitors, DNA intercalators, microtubule inhibitors, kinase inhibitors, proteasome inhibitors and topoisomerase inhibitors. A "toxic payload" as used herein refers to a sufficient amount of cytotoxic agent which, when delivered to a cell results in cell death. Delivery of a toxic payload may be accomplished by administration of a sufficient amount of immunoconjugate comprising an antibody or antigen binding fragment and a cytotoxic agent. Delivery of a toxic payload may also be accomplished by administration of a sufficient amount of an immunoconjugate comprising a cytotoxic agent, wherein the immunoconjugate comprises a secondary antibody or antigen binding fragment thereof which recognizes and binds an antibody or antigen binding fragment.
The term "human-suitable", with respect to an antibody, refers to any antibody, deri- vatized antibody, or antibody fragment that can be safely used in humans for, e.g. the therapeutic methods described herein. Human-suitable antibodies include all types of humanized, chimeric, or fully human antibodies, or any antibodies in which at least a portion of the antibodies is derived from humans or otherwise modified so as to avoid the immune response that is generally provoked when native non-human antibodies are used.
A "humanized" or "human" antibody refers to an antibody in which the constant and variable framework region of one or more human immunoglobulins is fused with the binding region, e.g. the CDR, of an animal immunoglobulin. Such antibodies are designed to maintain the binding specificity of the non-human antibody from which the binding regions are de- rived, but to avoid an immune reaction against the non-human antibody. Such antibodies can be obtained from transgenic mice or other animals that have been "engineered" to produce specific human antibodies in response to antigenic challenge (see, e.g., Green et al. (1994) Nature Genet 7:13; Lonberg et al. (1994) Nature 368:856; Taylor et al. (1994) Int Immun 6:579, the entire teachings of which are herein incorporated by reference). A fully human an- tibody also can be constructed by genetic or chromosomal transfection methods, as well as phage display technology, all of which are known in the art (see, e.g., McCafferty et al. (1990) Nature 348:552-553). Human antibodies may also be generated by in vitro activated B cells (see, e.g., U.S. Pat. Nos. 5,567,610 and 5,229,275, which are incorporated in their entirety by reference).
A "chimeric antibody" is an antibody molecule in which (a) the constant region, or a portion thereof, is altered, replaced or exchanged so that the antigen binding site (variable region) is linked to a constant region of a different or altered class, effector function and/or species, or an entirely different molecule which confers new properties to the chimeric antibody, e.g., an enzyme, toxin, hormone, growth factor, drug, etc.; or (b) the variable region, or a portion thereof, is altered, replaced or exchanged with a variable region having a different or altered antigen specificity.
The terms "Fc domain," "Fc portion," and "Fc region" refer to a C-terminal fragment of an antibody heavy chain, e.g., from about amino acid (aa) 230 to about aa 450 of human γ (gamma) heavy chain or its counterpart sequence in other types of antibody heavy chains (e.g., α, δ, ε and μ for human antibodies), or a naturally occurring allotype thereof. Unless otherwise specified, the commonly accepted Kabat amino acid numbering for immunoglobulins is used throughout this disclosure (see Kabat et al. (1991 ) Sequences of Protein of Immunological Interest, 5th ed., United States Public Health Service, National Institute of Health, Bethesda, MD).
The term "antibody-dependent cell-mediated cytotoxicity" or "ADCC" is a term well understood in the art, and refers to a cell-mediated reaction in which non- specific cytotoxic cells that express Fc receptors (FcRs) recognize bound antibody on a target cell and subsequently cause lysis of the target cell. Non-specific cytotoxic cells that mediate ADCC include natural killer (NK) cells, macrophages, monocytes, neutrophils, and eosinophils.
The terms "isolated", "purified" or "biologically pure" refer to material that is substantially or essentially free from components which normally accompany it as found in its native state. Purity and homogeneity are typically determined using analytical chemistry techniques such as polyacrylamide gel electrophoresis or high performance liquid chromatography. A protein that is the predominant species present in a preparation is substantially purified.
The terms "polypeptide," "peptide" and "protein" are used interchangeably herein to refer to a polymer of amino acid residues. The terms apply to amino acid polymers in which one or more amino acid residue is an artificial chemical mimetic of a corresponding naturally occurring amino acid, as well as to naturally occurring amino acid polymers and non-naturally occurring amino acid polymer.
The term "recombinant" when used with reference, e.g., to a cell, or nucleic acid, protein, or vector, indicates that the cell, nucleic acid, protein or vector, has been modified by the introduction of a heterologous nucleic acid or protein or the alteration of a native nucleic acid or protein, or that the cell is derived from a cell so modified. Thus, for example, recombinant cells express genes that are not found within the native (nonrecombinant) form of the cell or express native genes that are otherwise abnormally expressed, under expressed or not expressed at all.
"Prominently expressed", when referring to a KIR3DL2 polypeptide, means that the KIR3DL2 polypeptide is expressed in a substantial number of target cells (e.g. celiac disease cells, abnormal lELs) taken from a given patient. While the definition of the term "prominently expressed" is not bound by a precise percentage value, in most cases a receptor said to be "prominently expressed" will be present on at least 20%, 30%, 40%, 50°%, 60%, 70%, 80%, or more of the target cells (e.g. celiac disease cells, abnormal lELs) taken from a patient.
Within the context herein, the term antibody that "binds" a polypeptide or epitope designates an antibody that binds said determinant with specificity and/or affinity.
The term "identity" or "identical", when used in a relationship between the sequences of two or more polypeptides, refers to the degree of sequence relatedness between polypeptides, as determined by the number of matches between strings of two or more amino acid residues. "Identity" measures the percent of identical matches between the smaller of two or more sequences with gap alignments (if any) addressed by a particular mathematical model or computer program (i.e., "algorithms"). Identity of related polypeptides can be readily calculated by known methods. Such methods include, but are not limited to, those described in Computational Molecular Biology, Lesk, A. M., ed., Oxford University Press, New York, 1988; Biocomputing: Informatics and Genome Projects, Smith, D. W., ed., Academic Press, New York, 1993; Computer Analysis of Sequence Data, Part 1 , Griffin, A. M., and Griffin, H. G., eds., Humana Press, New Jersey, 1994; Sequence Analysis in Molecular Biology, von Heinje, G., Academic Press, 1987; Sequence Analysis Primer, Gribskov, M. and Devereux, J., eds., M. Stockton Press, New York, 1991 ; and Carillo et al., SIAM J. Applied Math. 48, 1073 (1988).
Methods for determining identity are designed to give the largest match between the sequences tested. Methods of determining identity are described in publicly available com- puter programs. Computer program methods for determining identity between two sequences include the GCG program package, including GAP (Devereux et al., Nucl. Acid. Res. 12, 387 (1984); Genetics Computer Group, University of Wisconsin, Madison, Wis.), BLASTP, BLASTN, and FASTA (Altschul et al., J. Mol. Biol. 215, 403-410 (1990)). The BLASTX pro- gram is publicly available from the National Center for Biotechnology Information (NCBI) and other sources (BLAST Manual, Altschul et al. NCB/NLM/NIH Bethesda, Md. 20894; Altschul et al., supra). The well-known Smith Waterman algorithm may also be used to determine identity. Production of antibodies
KIR3DL2 (CD158k) is a disulphide-linked homodimer of three-lg domain molecules of about 140 kD, described in Pende et al. (1996) J. Exp. Med. 184: 505-518, the disclosure of which is incorporated herein by reference. Several allelic variants have been reported for KIR3DL2 polypeptides, each of these are encompassed by the term KIR3DL2. The amino acid sequence of the mature human KIR3DL2 (allele *002) is shown in SEQ ID NO: 1 , below, corresponding to Genbank accession no. AAB52520 in which the 21 amino acid residue leader sequence has been omitted.
LMGGQDKPF LSARPSTWP RGGHVALQCH YRRGFNNFML YKEDRSHVPI FHGRIFQESF IMGPVTPAHA GTYRCRGSRP HSLTGWSAPS NPLVIMVTGN HRKPSLLAHP GPLLKSGETV ILQCWSDVMF EHFFLHRDGI SEDPSRLVGQ IHDGVSKANF SIGPLMPVLA GTYRCYGSVP HSPYQLSAPS DPLDIVITGL YEKPSLSAQP GPTVQAGENV TLSCSSWSSY DIYHLSREGE AHERRLRAVP KVNRTFQADF PLGPATHGGT YRCFGSFRAL PCVWSNSSDP LLVSVTGNPS SSWPSPTEPS SKSGICRHLH VLIGTSWIF LFILLLFFLL YRWCSNKKNA AVMDQEPAGD RTVNRQDSDE QDPQEVTYAQ LDHCVFIQRK ISRPSQRPKT PLTDTSVYTE LPNAEPRSKV VSCPRAPQSG LEGVF (SEQ ID NO: 1 ).
The cDNA of KIR3DL2 (allele *002) is shown in Genbank accession no. U30272. The amino acid sequence of a human KIR3DL2 allele *003 is shown below, corresponding to Genbank accession no. AAB36593:
MSLTVVSMAC VGFFLLQGAW PLMGGQDKPF LSARPSTWP RGGHVALQCH YRRGFNNFML YKEDRSHVPI FHGRIFQESF IMGPVTPAHA GTYRCRGSRP HSLTGWSAPS NPWIMVTGN HRKPSLLAHP GPLLKSGETV ILQCWSDVMF EHFFLHREGI SEDPSRLVGQ IHDGVSKANF SIGPLMPVLA GTYRCYGSVP HSPYQLSAPS DPLDIVITGL YEKPSLSAQP GPTVQAGENV TLSCSSWSSY DIYHLSREGE AHERRLRAVP KVNRTFQADF PLGPATHGGT YRCFGSFRAL PCVWSNSSDP LLVSVTGNPS SSWPSPTEPS SKSGICRHLH VLIGTSWIF LFILLLFFLL YRWCSNKKNA AVMDQEPAGD RTVNRQDSDE QDPQEVTYAQ LDHCVFIQRK ISRPSQRPKT PLTDTSVYTE LPNAEPRSKV VSCPRAPQSG LEGVF (SEQ ID NO: 2).
Also encompassed are any nucleic acid or protein sequences sharing one or more biological properties or functions with wild type, full length KIR3DL2 respectively, and sharing at least 70%, 80%, 90%, 95%, 96%, 97%, 98%, 99%, or higher nucleotide or amino acid identity.
Closely related KIR3DL1 (CD158e1 ) is a monomeric molecule of about 70 kD, described in Colonna and Samaridis (1995) Science 268 (5209), 405-408. The cDNA encoding a KIR3DL1 (CD158e2) polypeptide (allele *00101 ) is shown in Genbank accession no. L41269; the encoded amino acid sequence is shown in Genbank accession no. AAA69870. In one embodiment, a KIR3DL1 polypeptide referred to herein is allele *00101.
Examples of antibodies that bind human KIR3DL2 include antibody AZ158, antibody 19H12, antibody 2B12 and antibody 12B1 1 . Further antibodies are provided in PCT/EP2013/069302 and PCT/EP2013/069293, both filed 17 September 2013, the disclosures of which antibodies are incorporated herein by reference. AZ158 binds human KIR3DL2 as well as human KIR3DL1 and KIR3DS1 polypeptides; 19H12, 2B12 and 12B1 1 bind selectively to KIR3DL2 and do not bind KIR3DL1 (or KIR3DS1 ). While antibody AZ158 can be used, for example, as therapeutic agent administered to an individual for the elimina- tion of a KIR3DL2 expressing target, e.g. by induction of ADCC and/or CDC, antibody 12B1 1 and 19H12 will be advantageous over AZ158 for use in detection (e.g. in vitro assays) of KIR3DL2 expression on the surface of cells because 12B1 1 and 19H12 are both able to detect KIR3DL2-positive cells in detection assays, 12B1 1 is advantageous for immunohisto- chemistry assays using frozen tissue sections, while 19H12 is advantageous for flow cytome- try detection. Each of 2B12, 19H12 and 12B1 1 are also suitable for use as therapeutic agent administered to an individual for the elimination of a KIR3DL2-expressing target cells. 19H12 and 12B1 1 as well as other antibodies 15C1 1 , 19H12, 22B2, 18B10 and 13H1 disclosed in PCT/EP2013/069293 are capable of being internalized into cells via KIR3DL2 and can be used advantageously as an antibody-drug conjugate. 2B12 and other antibodies such as an- tibodies 10F6, 18C6, 9E10, 10G5, 13H1 , 5H1 , 1 E2, 1 C3 and 20E9 disclosed in PCT/EP2013/069302 do no induce any KIR3DL2 internalization, thereby providing advantageous use when effector cell mediated activity is sought, e.g. for depleting antibodies that induce ADCC.
In a specific embodiment, provided is an antibody that binds essentially the same epitope or determinant as any of monoclonal antibodies AZ158, 19B12, 12B1 1 or 2B12; op- tionally the antibody comprises an antigen binding region of antibody AZ158, 19B12, 12B1 1 or 2B12. In any of the embodiments herein, antibody AZ158, 19B12, 12B1 1 or 2B12 can be characterized by its amino acid sequence and/or nucleic acid sequence encoding it. In one embodiment, the monoclonal antibody comprises the Fab or F(ab')2 portion of AZ158, 19B12, 12B1 1 or 2B12. Also provided is a monoclonal antibody that comprises the heavy chain variable region of AZ158, 19B12, 12B1 1 or 2B12. According to one embodiment, the monoclonal antibody comprises the three CDRs of the heavy chain variable region of AZ158, 19B12, 12B1 1 or 2B12. Also provided is a monoclonal antibody that further comprises the variable light chain variable region of AZ158, 19B12, 12B1 1 or 2B12 or one, two or three of the CDRs of the light chain variable region of AZ158, 19B12, 12B1 1 or 2B12. Optionally any one or more of said light or heavy chain CDRs may contain one, two, three, four or five or more amino acid modifications (e.g. substitutions, insertions or deletions). Optionally, provided is an antibody where any of the light and/or heavy chain variable regions comprising part or all of an antigen binding region of antibody AZ158, 19B12, 12B1 1 or 2B12 are fused to an im- munoglobulin constant region of the human IgG type, optionally a human constant region, optionally a human lgG1 or lgG3 isotype.
Antibody AZ158
AZ158 binds human KIR3DL2 as well as human KIR3DL1 polypeptides, can be char- acterized as having the heavy and light chain variable regions or heavy and light chain region CDRs of SEQ ID NOS: 8 and 10, respectively, of PCT patent publication no. WO2010/081890). The VH of AZ158 is shown below, with CDRs 1 , 2 and 3 underlined, respectively:
QVQLKESGPG LVAPSQSLSI TCTVSGFSLT SFGVHWVRQP PGKGLEWLGV IWAGGSTNYN SALMSRLSIS KDNSKSQVFL KMNSLQNDDT AMYYCARGNS NHYVSS- FYYF DYWGQGTTLT VSS
(SEQ ID NO: 3).
The VL of AZ158 is shown below with CDRs 1 , 2 and 3 underlined, respectively: DIQMTQSPSS LSASLGGKVT ITCKASQDIN KYIAWYQHKP GKGPRLLIHY TSTLQPGIPS RFSGSGSGRD YSFSISNLEP EDITTYYCLQ YDNLWTFGGG TKLEIK
(SEQ ID NO: 4).
The anti-KIR3DL2 antibodies may include antibodies having variable region or CDR sequences from such AZ158 antibodies (e.g. a heavy and/or light chain variable region fused to a human constant region; a heavy chain variable region fused to a human lgG1 heavy chain constant region); alternatively, the anti-KIR3DL2 antibodies may be an antibody other than the antibodies having variable region or CDR sequences from a AZ158 antibody.
Antibody 19H12
The amino acid sequence of the heavy chain variable region of antibody 19H12 is listed below:
QIQLVQSGPELKKPGETVKISCKASGYTFTNFGMNWVKQAPGKGLK- WMGWINTYTGEPTYADDFKGRFAFSLETSASTAYLQINNLKNED- MATYFCARNGNFGYYFDYWGQGTTLTVSS
(SEQ ID NO: 5),
The amino acid sequence of the light chain variable region of antibody 19H12 is listed below:
DVLMTQTPLSLPVSLGDQASFSCRSSQNIVHSNGNTYLEWYLQKPGQSPSLLI- YKVSNRFSGVPDRFSGSGSGTDFTLKITRVEAEDLGVYYCFQGSHVPFTFGSGTKLEIK
(SEQ ID NO: 6)
In one aspect, provided is a purified polypeptide which encodes an antibody, wherein the antibody comprises: a HCDR1 region comprising an amino acid sequence GYTFT- NFGMN as set forth in SEQ ID NO:9, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof (e.g., NFGMN (SEQ ID NO: 7), GYTFTN (SEQ ID NO: 8)), wherein one or more of these amino acids may be substituted by a different amino acid; a HCDR2 region comprising an amino acid sequence WINTYTGEPTYADDF as set forth in SEQ ID NO: 10, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof (e.g. WINTYTGE (SEQ ID NO: 1 1 )), wherein one or more of these amino acids may be substituted by a different amino acid; a HCDR3 region comprising an amino acid sequence NGNFGYYFDY as set forth in SEQ ID NO: 12, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof, wherein one or more of these amino acids may be substituted by a different amino acid; a LCDR1 region comprising an amino acid sequence RSSQNIVHSNGNTYLE as set forth in SEQ ID NO: 13, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof, wherein one or more of these amino acids may be substituted by a different amino acid; a LCDR2 region comprising an amino acid sequence KVSNRFS as set forth in SEQ ID NO: 14, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof, wherein one or more of these amino acids may be substituted by a different amino acid; and/or a LCDR3 region comprising an amino acid sequence FQGSHVPFT as set forth in SEQ ID NO: 15, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof, wherein one or more of these amino acids may be deleted or substituted by a different amino acid, or where the sequence may comprise an insertion of one or more amino acids.
In another aspect, provided is an antibody that binds human KIR3DL2, comprising: (a) the heavy chain variable region of SEQ ID NO:5, wherein one, two, three or more amino acid residues may be substituted by a different amino acid; and/or
(b) the light chain variable region of SEQ ID NO:6, wherein one, two, three or more amino acid residues may be substituted by a different amino acid; and/or
(c) the heavy chain variable region of SEQ ID NO:5, wherein one, two, three or more amino acid residues may be substituted by a different amino acid; and the light chain variable region of SEQ ID NO:6, wherein one or more of these amino acids may be substituted by a different amino acid; and/or
(d) the heavy chain CDR 1 , 2 and 3 (HCDR1 , HCDR2, HCDR3) amino acid sequences as shown in SEQ ID NOS: 7-9, 10-1 1 and 12, respectively, wherein one, two, three or more amino acid residues of any CDR may be substituted by a different amino acid; and/or
(e) the light chain CDR 1 , 2 and 3 (LCDR1 , LCDR2, LCDR3) amino acid sequences as shown in SEQ ID NOS: 13, 14 or 15, respectively, wherein one, two, three or more amino acid residues of any CDR may be substituted by a different amino acid; and/or
(f) the heavy chain CDR 1 , 2 and 3 (HCDR1 , HCDR2, HCDR3) amino acid sequences as shown in SEQ ID NOS: 7, 8 or 9, 10 or 1 1 and 12, respectively, wherein one, two, three or more amino acid residues of any CDR may be substituted by a different amino acid; and the light chain CDR 1 , 2 and 3 (LCDR1 , LCDR2, LCDR3) amino acid sequences as shown in SEQ ID NOS: 13, 14 or 15, wherein one, two, three or more amino acid residues may be substituted by a different amino acid; and/or
(g) the heavy chain variable region which is at least 60%, 70%, 80%, 85%, 90% or 95% identical to the variable region having an amino acid sequence of SEQ ID NO:5, wherein one, two, three or more amino acid residues may be substituted by a different amino acid; and/or
(h) the light chain variable region which is at least 60%, 70%, 80%, 85%, 90% or 95% identical to the variable region having an amino acid sequence of SEQ ID NO:6, where- in one, two, three or more amino acid residues may be substituted by a different amino acid.
Antibody 12B11
The amino acid sequence of the heavy chain variable region of antibody 12B1 1 is listed below:
QLVQSGPELK PGETVKISCKASGYTFTNYGMNWVKQAPGKGLKWMGWINTY- TGEPTYADDFKGRFAFSLETSASTAYLQINNLKNEDTATYFCAHGPWLAY-
WGQGTLVTVS
(SEQ ID NO: 16).
The amino acid sequence of the light chain variable region of antibody 12B1 1 is listed below:
DIKMTQSPSSMYASLGERVTITCKASQDINVYLSWFQQKPGKSPKTLIYRAI R- LVDGVPSRFSGSGSGQDYSLTISSLDYEDMGIYYCLQYDELPYTFGGGTKLEIE
(SEQ ID NO: 17).
In one aspect, provided is a purified polypeptide which encodes an antibody, where- in the antibody comprises: a HCDR1 region comprising an amino acid sequence GYTFT- NYGMN as set forth in SEQ ID NO: 20, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof (e.g. NYGMN (SEQ ID NO: 18), GYTFTN (SEQ ID NO: 19)), wherein one or more of these amino acids may be substituted by a different amino acid; a HCDR2 region comprising an amino acid sequence WINTYTGEPTYADDFKG as set forth in SEQ ID NO: 21 , or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof (e.g. WINTYTGEPT (SEQ ID NO: 22)), wherein one or more of these amino acids may be substituted by a different amino acid; a HCDR3 region comprising an amino acid sequence GPWLAY as set forth in SEQ ID NO: 23, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof, wherein one or more of these amino acids may be substituted by a different amino acid; a LCDR1 region comprising an amino acid sequence KASQDINVYLS as set forth in SEQ ID NO: 24, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof, wherein one or more of these amino acids may be substituted by a different amino acid; a LCDR2 region comprising an amino acid sequence RAIRLVD as set forth in SEQ ID NO: 25, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous ami- no acids thereof, wherein one or more of these amino acids may be substituted by a different amino acid; a LCDR3 region comprising an amino acid sequence LQYDELPYT as set forth in SEQ ID NO: 26, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof, wherein one or more of these amino acids may be deleted or substituted by a different amino acid.
In another aspect, provided is an antibody that binds human KIR3DL2, comprising:
(a) the heavy chain variable region of SEQ ID NO: 16, wherein one, two, three or more amino acid residues may be substituted by a different amino acid; and/or
(b) the light chain variable region of SEQ ID NO: 17, wherein one, two, three or more amino acid residues may be substituted by a different amino acid; and/or
(c) the heavy chain variable region of SEQ ID NO: 16, wherein one or more amino acid residues may be substituted by a different amino acid; and the light chain variable region of SEQ ID NO: 17, wherein one, two, three or more of these amino acids may be substituted by a different amino acid; and/or
(d) the heavy chain CDR 1, 2 and 3 (HCDR1, HCDR2, HCDR3) amino acid se- quences as shown in SEQ ID NO: 18, 19 or 20, 21 or 22 and 23, respectively, wherein one, two, three or more o amino acid residues of any CDR may be substituted by a different amino acid; and/or
(e) the light chain CDR 1, 2 and 3 (LCDR1, LCDR2, LCDR3) amino acid sequences as shown in SEQ ID NO: 24, 25 and 26, wherein one, two, three or more amino acid resi- dues of any CDR may be substituted by a different amino acid; and/or
(f) the heavy chain CDR 1, 2 and 3 (HCDR1, HCDR2, HCDR3) amino acid sequences as shown in SEQ ID NO: 18, 19 or 20, 21 or 22 and 23, respectively, wherein one or more amino acid residues of any CDR may be substituted by a different amino acid; and the light chain CDRs 1, 2 and 3 (LCDR1, LCDR2, LCDR3) amino acid sequences as shown in SEQ ID NO: 24, 25 and 26, wherein one, two, three or more amino acid residues of any CDR may be substituted by a different amino acid; and/or
(g) the heavy chain variable region which is at least 60%, 70%, 80%, 85%, 90% or 95% identical to the variable region having an amino acid sequence of SEQ ID NO: 16, wherein one, two, three or more amino acid residues may be substituted by a different amino acid; and/or
(h) the light chain variable region which is at least 60%, 70%, 80%, 85%, 90% or 95% identical to the variable region having an amino acid sequence of SEQ ID NO: 17, wherein one, two, three or more amino acid residues may be substituted by a different amino acid.
Antibody 2B12
The amino acid sequence of the heavy chain variable region of antibody 2B12 is listed below (Kabat definition CDRs underlined):
QI QLVQSG PELKKPGETVRI SCKAS GYTFTTAG MQWVQKTP G K G LKWI GWI NS HSGVPKYAEDFKGRFAFSLETSASTAYLQI STLK N EDTATYFCARGG DEGVM DYWGQGTSVTVS
(SEQ ID NO: 32).
The amino acid sequence of the light chain variable region of antibody 2B12 is listed below (CDRs underlined): DIVMTQSH KF MSTS LG DRVSFTC KASQ DVSTAVAW Y Q Q K P G Q S P KL L I YWTSTRHTGVP DRFTGSGSGTDYTLTISSVQAEDLALY YCQQHYSTPWTFGGGTKLEI K
(SEQ ID NO: 33).
In one aspect, provided is a purified polypeptide which encodes an antibody, wherein the antibody comprises: a HCDR1 region comprising an amino acid sequence GYTFT- TAGMQ as set forth in SEQ ID NO: 36, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof (e.g. GYTFTT (SEQ ID NO: 34), or TAGMQ (SEQ ID NO: 35)), wherein one or more of these amino acids may be substituted by a different amino acid; a HCDR2 region comprising an amino acid sequence WINSHSGVPKYAEDFK as set forth in SEQ ID NO: 37, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof (e.g. WINSHSGVP (SEQ ID NO: 38)), wherein one or more of these amino acids may be substituted by a different amino acid; a HCDR3 region comprising an amino acid sequence GGDEGVMDYW as set forth in SEQ ID NO: 39, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof, wherein one or more of these amino acids may be substituted by a different amino acid; a LCDR1 region comprising an amino acid sequence KASQ DVSTAVA as set forth in SEQ ID NO: 40, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof, wherein one or more of these amino acids may be substituted by a different amino acid; a LCDR2 region comprising an amino acid sequence WTSTRHT as set forth in SEQ ID NO: 41 , or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof, wherein one or more of these amino acids may be substituted by a different amino acid; a LCDR3 region comprising an amino acid sequence QQHYSTPWT as set forth in SEQ ID NO: 42, or a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof, wherein one or more of these amino acids may be deleted or substituted by a different amino acid.
In another aspect, provided is an antibody that binds human KIR3DL2, comprising:
(a) the heavy chain variable region of SEQ ID NO: 32, wherein one, two, three or more amino acid residues may be substituted by a different amino acid; and/or
(b) the light chain variable region of SEQ ID NO: 33, wherein one, two, three or more amino acid residues may be substituted by a different amino acid; and/or
(c) the heavy chain variable region of SEQ ID NO: 32, wherein one or more amino acid residues may be substituted by a different amino acid; and the light chain variable region of SEQ ID NO: 33, wherein one, two, three or more of these amino acids may be substituted by a different amino acid; and/or
(d) the heavy chain CDR 1, 2 and 3 (HCDR1, HCDR2, HCDR3) amino acid se- quences as shown in SEQ ID NO: 34, 35 or 36, 37 or 38 and 39, respectively, wherein one, two, three or more o amino acid residues of any CDR may be substituted by a different amino acid; and/or
(e) the light chain CDR 1 , 2 and 3 (LCDR1 , LCDR2, LCDR3) amino acid sequences as shown in SEQ ID NO: 40, 41 and 42, wherein one, two, three or more amino acid residues of any CDR may be substituted by a different amino acid; and/or
(f) the heavy chain CDR 1 , 2 and 3 (HCDR1 , HCDR2, HCDR3) amino acid sequences as shown in SEQ ID NO: 34, 35 or 36, 37 or 38 and 39, respectively, wherein one or more amino acid residues of any CDR may be substituted by a different amino acid; and the light chain CDRs 1 , 2 and 3 (LCDR1 , LCDR2, LCDR3) amino acid sequences as shown in SEQ ID NO: 40, 41 and 42, wherein one, two, three or more amino acid residues of any CDR may be substituted by a different amino acid; and/or
(g) the heavy chain variable region which is at least 60%, 70%, 80%, 85%, 90% or 95% identical to the variable region having an amino acid sequence of SEQ ID NO: 32, wherein one, two, three or more amino acid residues may be substituted by a different amino acid; and/or
(h) the light chain variable region which is at least 60%, 70%, 80%, 85%, 90% or 95% identical to the variable region having an amino acid sequence of SEQ ID NO: 33, wherein one, two, three or more amino acid residues may be substituted by a different amino acid.
In another aspect of any of the embodiments herein, any of the CDRs 1 , 2 and 3 of the heavy and light chains may be characterized by a sequence of at least 4, 5, 6, 7, 8, 9 or 10 contiguous amino acids thereof, and/or as having an amino acid sequence that shares at least 50%, 60%, 70%, 80%, 85%, 90% or 95% sequence identity with the particular CDR or set of CDRs listed in the corresponding SEQ ID NO.
In another aspect, provided is an antibody that competes for KIR3DL2 binding with a monoclonal antibody of (a) to (h), for any of the above antibodies.
Antibody epitopes
While it will be appreciated that any suitable antibody can be used, in one aspect the antibodies that are used bind substantially the same epitope as antibody 19H12 or 12B1 1. In another embodiment, the antibodies at least partially overlaps, or includes at least one residue in the segment corresponding to residues 1-192, residues 1 -98, or residues 99-192 of the KIR3DL2 polypeptide of SEQ ID NO: 1 (or a subsequence thereof). In one embodiment, all key residues of the epitope is in a segment corresponding to residues 1 -192, residues 1- 98 or residues 99-192 of the KIR3DL2 polypeptide of SEQ ID NO: 1. In one embodiment, the antibodies bind an epitope comprising 1 , 2, 3, 4, 5, 6, 7 or more residues in the segment corresponding to residues 1-192, 1 -98 or 99-192 of the KIR3DL2 polypeptide of SEQ ID NO: 1. Preferably the residues bound by the antibody are present on the surface of the of the KIR3DL2 polypeptide.
Optionally, the antibodies bind an epitope comprising residues P179 and/or residue S181 of SEQ ID NO: 1. Optionally, the antibodies bind to an epitope comprising 1 , 2, 3, 4, 5, 6 or 7 or more residues selected from the group consisting of: N99, H100, E130, H131 , F132, V178, P179, H180, S181 , P182, Y183 and/or residue Q184 of SEQ ID NO: 1.
Optionally, the antibody binds an epitope comprising residues I60 and/or residue
G62 of the KIR3DL2 polypeptide of SEQ ID NO: 1 , and/or has reduced binding to a KIR3DL2 polypeptide having a mutation at residues I60 and/or residue G62 of SEQ ID NO: 1 , compared to a wild-type KIR3DL2 polypeptide of SEQ ID NO: 1 . Optionally, the antibody binds an epitope comprising residues P14, S15 and/or residue H23 of the KIR3DL2 polypeptide of SEQ ID NO: 1 , and/or has reduced binding to a KIR3DL2 polypeptide having a mutation at residues P14, S15 and/or residue H23 of SEQ ID NO: 1 , compared to a wild-type KIR3DL2 polypeptide of SEQ ID NO: 1. Optionally, the antibody binds an epitope comprising residues I60, G62, P14, S15 and/or residue H23 of SEQ ID NO: 1 .
The Examples section herein describes the testing of a series of mutant human KIR3DL2 polypeptides. Binding of anti-KIR3DL2 antibody to cells transfected with the KIR3DL2 mutants was measured and compared to the ability of anti-KIR3DL2 antibody to bind wild-type KIR3DL2 polypeptide (SEQ ID NO:1 ). A reduction in binding between an anti- KIR3DL2 antibody and a mutant KIR3DL2 polypeptide as used herein means that there is a reduction in binding affinity (e.g., as measured by known methods such FACS testing of cells expressing a particular mutant, or by Biacore testing of binding to mutant polypeptides) and/or a reduction in the total binding capacity of the anti-KIR3DL2 antibody (e.g., as evidenced by a decrease in Bmax in a plot of anti-KIR3DL2 antibody concentration versus polypeptide concentration). A significant reduction in binding indicates that the mutated residue is directly involved in binding to the anti-KIR3DL2 antibody or is in close proximity to the bind- ing protein when the anti-KIR3DL2 antibody is bound to KIR3DL2. An antibody epitope will may thus include such residue and may include additional residues spatially adjacent to such residue.
In some embodiments, a significant reduction in binding means that the binding affinity and/or capacity between an anti-KIR3DL2 antibody and a mutant KIR3DL2 polypeptide is reduced by greater than 40 %, greater than 50 %, greater than 55 %, greater than 60 %, greater than 65 %, greater than 70 %, greater than 75 %, greater than 80 %, greater than 85 %, greater than 90% or greater than 95% relative to binding between the antibody and a wild type KIR3DL2 polypeptide (e.g., the polypeptide shown in SEQ ID NO:1 ). In certain embodiments, binding is reduced below detectable limits. In some embodiments, a significant re- duction in binding is evidenced when binding of an anti-KIR3DL2 antibody to a mutant KIR3DL2 polypeptide is less than 50% (e.g., less than 45%, 40%, 35%, 30%, 25%, 20%, 15% or 10%) of the binding observed between the anti-KIR3DL2 antibody and a wild-type KIR3DL2 polypeptide (e.g., the extracellular domain shown in SEQ ID NO:1 ). Such binding measurements can be made using a variety of binding assays known in the art. A specific example of one such assay is described in the Example section.
In some embodiments, anti-KIR3DL2 antibodies are provided that exhibit significantly lower binding for a mutant KIR3DL2 polypeptide in which a residue in a wild-type KIR3DL2 polypeptide (e.g., SEQ ID NO:1 ) is substituted, e.g. the mutants as described in Example 1 . In the shorthand notation used here, the format is: Wild type residue: Position in polypeptide: Mutant residue, with the numbering of the residues as indicated in SEQ ID NO: 1.
Optionally, the antibodies have reduced binding to a KIR3DL2 polypeptide having a substitution at residues N99, H100, E130, H131 , F132, V178, P179, H180, S181 , P182, Y183 and/or residue Q184 of SEQ ID NO: 1.
In some embodiments, an anti-KIR3DL2 antibody binds a wild-type KIR3DL2 poly- peptide having a sequence of SEQ ID NO: 1 but has decreased binding to a mutant KIR3DL2 polypeptide having any one or more (e.g., 1 , 2, 3 or 4) of the following mutations: P179T and/or S181T (with reference to SEQ ID NO:1 ). In one embodiment, binding to the mutant KIR3DL2 is significantly reduced compared to binding to the wild-type KIR3DL2.
In some embodiments, anti-KIR3DL2 antibodies are provided that exhibit significant- ly lower binding for a mutant KIR3DL2 polypeptide in which a residue in a segment corresponding to residues 1-98, residues 99-292, or residues 99-192 (or a subsequence thereof) in a wild-type KIR3DL2 polypeptide (e.g., SEQ ID NO:1 ) is substituted with a different amino acid.
In one aspect, an antibody can compete with monoclonal antibody AZ158, 19H12, 2B12 or 12B1 1 and recognizes bind to, or have immunospecificity for substantially or essentially the same, or the same, epitope or "epitopic site" on a KIR3DL2 molecule as monoclonal antibody AZ158, 19H12, 2B12 or 12B1 1 . In other embodiments, the monoclonal antibody consists of, or is a derivative or fragment of, antibody AZ158, 19H12, 2B12 or 12B1 1.
It will be appreciated that, while antibodies may bind to the same epitope as anti- body AZ158, 19H12, 2B12 or 12B1 1 , suitable antibodies can recognize and be raised against any part of the KIR3DL2 polypeptide so long as the antibody binds KIR3DL2 and has the desired functionality. For example, any fragment of KIR3DL2, e.g., human KIR3DL2, or any combination of KIR3DL2 fragments, can be used as immunogens to raise antibodies, and the antibodies can recognize epitopes at any location within the KIR3DL2 polypeptide, so long as they can do so on KIR3DL2 expressing NK cells as described herein. In an embodiment, the recognized epitopes are present on the cell surface, i.e. they are accessible to antibodies present outside of the cell. Optionally, the epitope is the epitope specifically recognized by antibody AZ158, 19H12, 2B12 or 12B1 1. Further, antibodies recognizing distinct epitopes within KIR3DL2 can be used in combination, e.g. to bind to KIR3DL2 polypeptides with maximum efficacy and breadth among different individuals.
The antibodies may be produced by a variety of techniques known in the art. Typically, they are produced by immunization of a non-human animal, optionally a mouse, with an immunogen comprising a KIR3DL2 polypeptide, optionally a human KIR3DL2 polypeptide. The KIR3DL2 polypeptide may comprise the full length sequence of a human KIR3DL2 polypeptide, or a fragment or derivative thereof, typically an immunogenic fragment, i.e., a portion of the polypeptide comprising an epitope exposed on the surface of cells expressing a KIR3DL2 polypeptide, optionally the epitope recognized by the AZ158, 19H12, 2B12 or 12B1 1 antibody. Such fragments typically contain at least about 7 consecutive amino acids of the mature polypeptide sequence, or at least about 10 consecutive amino acids thereof. Fragments typically are essentially derived from the extra-cellular domain of the receptor. In one embodiment, the immunogen comprises a wild-type human KIR3DL2 polypeptide in a lipid membrane, typically at the surface of a cell. In one embodiment, the immunogen comprises intact cells, particularly intact human cells, optionally treated or lysed. In another embodiment, the polypeptide is a recombinant KIR3DL2 polypeptide.
The step of immunizing a non-human mammal with an antigen may be carried out in any manner well known in the art for stimulating the production of antibodies in a mouse (see, for example, E. Harlow and D. Lane, Antibodies: A Laboratory Manual., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY (1988), the entire disclosure of which is herein incorporated by reference). The immunogen is suspended or dissolved in a buffer, optionally with an adjuvant, such as complete or incomplete Freund's adjuvant. Methods for determining the amount of immunogen, types of buffers and amounts of adjuvant are well known to those of skill in the art and are not limiting in any way. These parameters may be different for different immunogens, but are easily elucidated.
Similarly, the location and frequency of immunization sufficient to stimulate the pro- duction of antibodies is also well known in the art. In a typical immunization protocol, the non- human animals are injected intraperitoneal^ with antigen on day 1 and again about a week later. This is followed by recall injections of the antigen around day 20, optionally with an adjuvant such as incomplete Freund's adjuvant. The recall injections are performed intravenously and may be repeated for several consecutive days. This is followed by a booster in- jection at day 40, either intravenously or intraperitoneally, typically without adjuvant. This protocol results in the production of antigen-specific antibody-producing B cells after about 40 days. Other protocols may also be used as long as they result in the production of B cells expressing an antibody directed to the antigen used in immunization.
For polyclonal antibody preparation, serum is obtained from an immunized non- human animal and the antibodies present therein isolated by well-known techniques. The serum may be affinity purified using any of the immunogens set forth above linked to a solid support so as to obtain antibodies that react with KIR3DL2 polypeptides.
In an alternate embodiment, lymphocytes from a non-immunized non-human mammal are isolated, grown in vitro, and then exposed to the immunogen in cell culture. The lym- phocytes are then harvested and the fusion step described below is carried out.
For exemplary monoclonal antibodies, the next step is the isolation of splenocytes from the immunized non-human mammal and the subsequent fusion of those splenocytes with an immortalized cell in order to form an antibody-producing hybridoma. The isolation of splenocytes from a non-human mammal is well-known in the art and typically involves re- moving the spleen from an anesthetized non-human mammal, cutting it into small pieces and squeezing the splenocytes from the splenic capsule through a nylon mesh of a cell strainer into an appropriate buffer so as to produce a single cell suspension. The cells are washed, centrifuged and resuspended in a buffer that lyses any red blood cells. The solution is again centrifuged and remaining lymphocytes in the pellet are finally resuspended in fresh buffer.
Once isolated and present in single cell suspension, the lymphocytes can be fused to an immortal cell line. This is typically a mouse myeloma cell line, although many other immortal cell lines useful for creating hybridomas are known in the art. Murine myeloma lines include, but are not limited to, those derived from MOPC-21 and MPC-1 1 mouse tumors available from the Salk Institute Cell Distribution Center, San Diego, U. S. A., X63 Ag8653 and SP-2 cells available from the American Type Culture Collection, Rockville, Maryland U. S. A. The fusion is effected using polyethylene glycol or the like. The resulting hybridomas are then grown in selective media that contains one or more substances that inhibit the growth or survival of the unfused, parental myeloma cells. For example, if the parental myeloma cells lack the enzyme hypoxanthine guanine phosphoribosyl transferase (HGPRT or HPRT), the culture medium for the hybridomas typically will include hypoxanthine, aminopter- in, and thymidine (HAT medium), which substances prevent the growth of HGPRT-deficient cells.
Hybridomas are typically grown on a feeder layer of macrophages. The macrophages can be from littermates of the non-human mammal used to isolate splenocytes and are typically primed with incomplete Freund's adjuvant or the like several days before plating the hybridomas. Fusion methods are described in Goding, "Monoclonal Antibodies: Principles and Practice," pp. 59-103 (Academic Press, 1986), the disclosure of which is herein incorporated by reference.
The cells are allowed to grow in the selection media for sufficient time for colony formation and antibody production. This is usually between about 7 and about 14 days.
The hybridoma colonies are then assayed for the production of antibodies that specifically bind to KIR3DL2 polypeptide gene products, optionally the epitope specifically recognized by antibody AZ158, 19H12, 2B12 or 12B1 1 . The assay is typically a colorimetric ELISA-type assay, although any assay may be employed that can be adapted to the wells that the hybridomas are grown in. Other assays include radioimmunoassays or fluorescence activated cell sorting. The wells positive for the desired antibody production are examined to determine if one or more distinct colonies are present. If more than one colony is present, the cells may be re-cloned and grown to ensure that only a single cell has given rise to the colony producing the desired antibody. Typically, the antibodies will also be tested for the ability to bind to KIR3DL2 polypeptides, e.g., KIR3DL2-expressing cells.
Hybridomas that are confirmed to produce a monoclonal antibody can be grown up in larger amounts in an appropriate medium, such as DMEM or RPMI-1640. Alternatively, the hybridoma cells can be grown in vivo as ascites tumors in an animal.
After sufficient growth to produce the desired monoclonal antibody, the growth media con- taining monoclonal antibody (or the ascites fluid) is separated away from the cells and the monoclonal antibody present therein is purified. Purification is typically achieved by gel electrophoresis, dialysis, chromatography using protein A or protein G-Sepharose, or an anti- mouse Ig linked to a solid support such as agarose or Sepharose beads (all described, for example, in the Antibody Purification Handbook, Biosciences, publication No. 18-1037-46, Edition AC, the disclosure of which is hereby incorporated by reference). The bound antibody is typically eluted from protein A protein G columns by using low pH buffers (glycine or acetate buffers of pH 3.0 or less) with immediate neutralization of antibody-containing fractions. These fractions are pooled, dialyzed, and concentrated as needed.
Positive wells with a single apparent colony are typically re-cloned and re-assayed to insure only one monoclonal antibody is being detected and produced. Antibodies may also be produced by selection of combinatorial libraries of immunoglobulins, as disclosed for instance in (Ward et al. Nature, 341 (1989) p. 544, the entire disclosure of which is herein incorporated by reference).
The identification of one or more antibodies that bind(s) to KIR3DL2, particularly substantially or essentially the same epitope as monoclonal antibody AZ158, 19H12, 2B12 or 12B1 1 , can be readily determined using any one of a variety of immunological screening assays in which antibody competition can be assessed. Many such assays are routinely practiced and are well known in the art (see, e. g., U. S. Pat. No. 5,660,827, issued Aug. 26, 1997, which is specifically incorporated herein by reference). It will be understood that actual- ly determining the epitope to which an antibody described herein binds is not in any way required to identify an antibody that binds to the same or substantially the same epitope as the monoclonal antibody described herein.
For example, where the test antibodies to be examined are obtained from different source animals, or are even of a different Ig isotype, a simple competition assay may be em- ployed in which the control (AZ158, 19H12, 2B12 or 12B1 1 , for example) and test antibodies are admixed (or pre-adsorbed) and applied to a sample containing KIR3DL2 polypeptides. Protocols based upon western blotting and the use of BIACORE analysis are suitable for use in such competition studies.
In certain embodiments, one pre-mixes the control antibodies (AZ158, 19H12, 2B12 or 12B1 1 , for example) with varying amounts of the test antibodies (e.g., about 1 :10 or about 1 :100) for a period of time prior to applying to the KIR3DL2 antigen sample. In other embodiments, the control and varying amounts of test antibodies can simply be admixed during exposure to the KIR3DL2 antigen sample. As long as one can distinguish bound from free antibodies (e. g., by using separation or washing techniques to eliminate unbound antibodies) and AZ158, 19H 12, 2B12 or 12B1 1 from the test antibodies (e. g., by using species-specific or isotype-specific secondary antibodies or by specifically labeling AZ158, 19H 12, 2B12 or 12B1 1 with a detectable label) one can determine if the test antibodies reduce the binding of AZ158, 19H12, 2B12 or 12B1 1 to the antigens, indicating that the test antibody recognizes substantially the same epitope as AZ158, 19H12, 2B12 or 12B1 1 . The binding of the (la- beled) control antibodies in the absence of a completely irrelevant antibody can serve as the control high value. The control low value can be obtained by incubating the labeled (AZ158, 19H12, 2B12 or 12B1 1 ) antibodies with unlabelled antibodies of exactly the same type (AZ158, 19H12, 2B12 or 12B1 1 ), where competition would occur and reduce binding of the labeled antibodies. In a test assay, a significant reduction in labeled antibody reactivity in the presence of a test antibody is indicative of a test antibody that recognizes substantially the same epitope, i.e., one that "cross-reacts" or competes with the labeled (AZ158, 19H 12, 2B12 or 12B1 1 ) antibody. Any test antibody that reduces the binding of AZ158, 19H12, 2B12 or 12B1 1 to KIR3DL2 antigens by at least about 50%, such as at least about 60%, or more preferably at least about 80% or 90% (e. g., about 65-100%), at any ratio of AZ158, 19H12, 2B12 or 12B1 1 : test antibody between about 1 :10 and about 1 :100 is considered to be an antibody that binds to substantially the same epitope or determinant as AZ158, 19H12, 2B12 or 12B1 1. For example such test antibody will reduce the binding of AZ158, 19H12, 2B12 or 12B1 1 to the KIR3DL2 antigen by at least about 90% (e.g., about 95%).
Competition can also be assessed by, for example, a flow cytometry test. In such a test, cells bearing a given KIR3DL2 polypeptide can be incubated first with AZ158, 19H12, 2B12 or 12B1 1 , for example, and then with the test antibody labeled with a fluorochrome or biotin. The antibody is said to compete with AZ158, 19H12, 2B12 or 12B1 1 if the binding obtained upon preincubation with a saturating amount of AZ158, 19H12, 2B12 or 12B1 1 is about 80%, about 50%, about 40% or less (e.g., about 30%, 20% or 10%) of the binding (as measured by mean of fluorescence) obtained by the antibody without pre-incubation with AZ158, 19H 12, 2B12 or 12B1 1. Alternatively, an antibody is said to compete with AZ158, 19H12, 2B12 or 12B1 1 if the binding obtained with a labeled AZ158, 19H12, 2B12 or 12B1 1 antibody (by a fluorochrome or biotin) on cells preincubated with a saturating amount of test antibody is about 80%, about 50%, about 40%, or less (e. g., about 30%, 20% or 10%) of the binding obtained without preincubation with the test antibody.
A simple competition assay in which a test antibody is pre-adsorbed and applied at saturating concentration to a surface onto which a KIR3DL2 antigen is immobilized may also be employed. The surface in the simple competition assay is for example a BIACORE chip (or other media suitable for surface plasmon resonance analysis). The control antibody (e.g., AZ158, 19H12, 2B12 or 12B1 1 ) is then brought into contact with the surface at a KIR3DL2- saturating concentration and the KIR3DL2 and surface binding of the control antibody is measured. This binding of the control antibody is compared with the binding of the control antibody to the KIR3DL2-containing surface in the absence of test antibody. In a test assay, a significant reduction in binding of the KIR3DL2-containing surface by the control antibody in the presence of a test antibody indicates that the test antibody recognizes substantially the same epitope as the control antibody such that the test antibody "cross-reacts" with the control antibody. Any test antibody that reduces the binding of control (such as AZ158, 19H12, 2B12 or 12B1 1 ) antibody to a KIR3DL2 antigen by at least about 30% or more, or about 40%, can be considered to be an antibody that binds to substantially the same epitope or de- terminant as a control (e.g., AZ158, 19H12, 2B12 or 12B1 1 ). For example, such a test anti- body will reduce the binding of the control antibody (e.g., AZ158, 19H 12, 2B12 or 12B1 1 ) to the KIR3DL2 antigen by at least about 50% (e. g., at least about 60%, at least about 70%, or more). It will be appreciated that the order of control and test antibodies can be reversed: that is, the control antibody can be first bound to the surface and the test antibody is brought into contact with the surface thereafter in a competition assay. For example, the antibody having higher affinity for the KIR3DL2 antigen is bound to the surface first, as it will be expected that the decrease in binding seen for the second antibody (assuming the antibodies are cross-reacting) will be of greater magnitude. Further examples of such assays are provided in, e.g., Saunal (1995) J. Immunol. Methods 183: 33-41 , the disclosure of which is in- corporated herein by reference.
Determination of whether an antibody binds within an epitope region can be carried out in ways known to the person skilled in the art. As one example of such mapping/characterization methods, an epitope region for an anti-KIR3DL2 antibody may be determined by epitope "foot-printing" using chemical modification of the exposed amines/carboxyls in the KIR3DL2 protein. One specific example of such a foot-printing technique is the use of HXMS (hydrogen-deuterium exchange detected by mass spectrometry) wherein a hydrogen/deuterium exchange of receptor and ligand protein amide protons, binding, and back exchange occurs, wherein the backbone amide groups participating in protein binding are protected from back exchange and therefore will remain deuterated. Relevant regions can be identified at this point by peptic proteolysis, fast microbore high-performance liquid chromatography separation, and/or electrospray ionization mass spectrometry. See, e. g., Ehring H, Analytical Biochemistry, Vol. 267 (2) pp. 252-259 (1999) Engen, J. R. and Smith, D. L. (2001 ) Anal. Chem. 73, 256A-265A. Another example of a suitable epitope identification technique is nuclear magnetic resonance epitope mapping (NMR), where typically the position of the signals in two-dimensional NMR spectra of the free antigen and the antigen complexed with the antigen binding peptide, such as an antibody, are compared. The antigen typically is selectively isotopically labeled with 15N so that only signals corresponding to the antigen and no signals from the antigen binding peptide are seen in the NMR- spectrum. Antigen signals originating from amino acids involved in the interaction with the antigen binding peptide typically will shift position in the spectrum of the complex compared to the spectrum of the free antigen, and the amino acids involved in the binding can be identified that way. See, e. g., Ernst Schering Res Found Workshop. 2004; (44): 149-67; Huang et Journal of Molecular Biology, Vol. 281 (1 ) pp. 61 -67 (1998); and Saito and Patterson, Methods. 1996 Jun; 9 (3): 516-24.
Epitope mapping/characterization also can be performed using mass spectrometry methods. See, e.g., Downward, J Mass Spectrom. 2000 Apr; 35 (4): 493-503 and Kiselar and Downard, Anal Chem. 1999 May 1 ; 71 (9): 1792-801. Protease digestion techniques also can be useful in the context of epitope mapping and identification. Antigenic determinant-relevant regions/sequences can be determined by protease digestion, e.g. by using trypsin in a ratio of about 1 :50 to KIR3DL2 or o/n digestion at and pH 7-8, followed by mass spectrometry (MS) analysis for peptide identification. The peptides protected from trypsin cleavage by the anti-KIR3DL2 binder can subsequently be identified by comparison of samples subjected to trypsin digestion and samples incubated with antibody and then subjected to digestion by e.g. trypsin (thereby revealing a footprint for the binder). Other enzymes like chymotrypsin, pepsin, etc., also or alternatively can be used in similar epitope characterization methods. Moreover, enzymatic digestion can provide a quick method for analyzing whether a potential antigenic determinant sequence is within a region of the KIR3DL2 polypeptide that is not surface exposed and, accordingly, most likely not relevant in terms of immunogenici- ty/antigenicity. See, e. g., Manca, Ann 1st Super Sanita. 1991 ; 27: 15-9 for a discussion of similar techniques.
Site-directed mutagenesis is another technique useful for elucidation of a binding epitope. For example, in "alanine-scanning", each residue within a protein segment is replaced with an alanine residue, and the consequences for binding affinity measured. If the mutation leads to a significant reduction in binding affinity, it is most likely involved in binding. Monoclonal antibodies specific for structural epitopes (i.e., antibodies which do not bind the unfolded protein) can be used to verify that the alanine-replacement does not influence overall fold of the protein. See, e.g. , Clackson and Wells, Science 1995; 267:383-386; and Wells, Proc Natl Acad Sci USA 1996; 93:1-6.
Electron microscopy can also be used for epitope "foot-printing". For example, Wang et al., Nature 1992; 355:275-278 used coordinated application of cryoelectron microscopy, three-dimensional image reconstruction, and X-ray crystallography to determine the physical footprint of a Fab-fragment on the capsid surface of native cowpea mosaic virus.
Other forms of "label-free" assay for epitope evaluation include surface plasmon resonance (SPR, BIACORE) and reflectometric interference spectroscopy (RifS). See, e.g., Fagerstam et al., Journal Of Molecular Recognition 1990;3:208-14; Nice et al., J. Chroma- togr. 1993; 646:159-168; Leipert et al., Angew. Chem. Int. Ed. 1998; 37:3308-331 1 ; Kroger et al., Biosensors and Bioelectronics 2002; 17:937-944.
It should also be noted that an antibody binding the same or substantially the same epitope as an antibody can be identified in one or more of the exemplary competition assays described herein. Once antibodies are identified that are capable of binding KIR3DL2 and/or having other desired properties, they will also typically be assessed, using standard methods including those described herein, for their ability to bind to other polypeptides, including unrelated polypeptides. Ideally, the antibodies only bind with substantial affinity to KIR3DL2, e.g., hu- man KIR3DL2, and do not bind at a significant level to unrelated polypeptides. However, it will be appreciated that, as long as the affinity for KIR3DL2 is substantially greater (e.g., 5x, 10x, 50x, 100x, 500x, 1000x, 10,000x, or more) than it is for other, unrelated polypeptides), then the antibodies are suitable for use in the present methods.
The binding of the antibodies to KIR3DL2-expressing cells can also be assessed in non-human primates, e.g. cynomolgus monkeys, or other mammals such as mice. Provided is an antibody, as well as fragments and derivatives thereof, wherein said antibody, fragment or derivative specifically bind KIR3DL2, and which furthermore bind KIR3DL2 from non- human primates, e.g., cynomolgus monkeys.
Upon immunization and production of antibodies in a vertebrate or cell, particular selection steps may be performed to isolate antibodies as claimed. In this regard, in a specific embodiment, the disclosure also relates to methods of producing such antibodies, comprising: (a) immunizing a non-human mammal with an immunogen comprising a KIR3DL2 polypeptide; and (b) preparing antibodies from said immunized animal; and (c) selecting antibodies from step (b) that are capable of binding KIR3DL2.
In one aspect of any of the embodiments, the antibodies prepared according to the present methods are monoclonal antibodies. In another aspect, the non-human animal used iesto produce antibodies is a mammal, such as a rodent, bovine, porcine, fowl, horse, rabbit, goat, or sheep.
According to an alternate embodiment, the DNA encoding an antibody that binds an epitope present on KIR3DL2 polypeptides is isolated from the hybridoma and placed in an appropriate expression vector for transfection into an appropriate host. The host is then used for the recombinant production of the antibody, or variants thereof, such as a humanized version of that monoclonal antibody, active fragments of the antibody, chimeric antibodies comprising the antigen recognition portion of the antibody, or versions comprising a detectable moiety.
DNA encoding a monoclonal antibody, e.g., antibody 19H12, 2B12 or 12B1 1 , can be readily isolated and sequenced using conventional procedures (e. g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of murine antibodies). Once isolated, the DNA can be placed into expression vectors, which are then transfected into host cells such as E. coli cells, simian COS cells, Chinese hamster ovary (CHO) cells, or myeloma cells that do not otherwise produce immunoglobulin protein, to obtain the synthesis of monoclonal antibodies in the recombinant host cells. As described elsewhere in the present specification, such DNA sequences can be modified for any of a large number of purposes, e.g., for humanizing antibodies, producing fragments or derivatives, or for modifying the sequence of the antibody, e.g., in the antigen binding site in order to optimize the binding specificity of the antibody.
Recombinant expression in bacteria of DNA encoding the antibody is well known in the art (see, for example, Skerra et al., Curr. Opinion in Immunol., 5, pp. 256 (1993); and Pluckthun, Immunol. 130, p. 151 (1992).
Once an antigen-binding compound is obtained it may be assessed for its ability to induce ADCC or CDC towards, inhibit the activity and/or proliferation of and/or cause the elimination of KIR3DL2-expressing target cells. Assessing the antigen-binding compound's ability to induce ADCC, CDC (complement dependent cytotoxicity) or generally lead to the elimination or inhibition of activity of KIR3DL2-expressing target cells, can be carried out at any suitable stage of the method. This assessment can be useful at one or more of the various steps involved in the identification, production and/or development of an antibody (or other compound) destined for therapeutic use. For example, activity may be assessed in the context of a screening method to identify candidate antigen-binding compounds, or in methods where an antigen-binding compound is selected and made human suitable (e.g. made chimeric or humanized in the case of an antibody), where a cell expressing the antigen- binding compound (e.g. a host cell expressing a recombinant antigen-binding compound) has been obtained and is assessed for its ability to produce functional antibodies (or other compounds), and/or where a quantity of antigen-binding compound has been produced and is to be assessed for activity (e.g. to test batches or lots of product). Generally the antigen- binding compound will be known to specifically bind to a KIR3DL2 polypeptide. The step may involve testing a plurality (e.g., a very large number using high throughput screening methods or a smaller number) of antigen-binding compounds.
Testing CDC and ADCC can be carried out can be determined by various assays including those known in the art and those described in the experimental examples herein. Testing ADCC typically involves assessing cell-mediated cytotoxicity in which a KIR3DL2- expressing target cell (e.g. a celiac disease cell or other KIR3DL2-expressing cell) with bound anti-KIR3DL2 antibody is recognized by an effector cell bearing Fc receptors, without the involvement of complement. A cell which does not express a KIR3DL2 antigen can optionally be used as a control. Activation of NK cell cytotoxicity is assessed by measuring an increase in cytokine production (e.g. IFN-γ production) or cytotoxicity markers (e.g. CD107 mobilization). In one embodiment, the antibody will induce an increase in cytokine production, expression of cytotoxicity markers, or target cell lysis of at least 20%, 50%, 80%, 100%, 200% or 500% in the presence of target cells, compared to a control antibody (e.g. an antibody not binding to KIR3DL2, a KIR3DL2 antibody having murine constant regions). In an- other example, lysis of target cells is detected, e.g. in a chromium release assay, for example the antibody will induce lysis of at least 10%, 20%, 30%, 40% or 50% of target cells.
Fragments and derivatives of antibodies (which are encompassed by the term "antibody" or "antibodies" as used in this application, unless otherwise stated or clearly contradicted by context) can be produced by techniques that are known in the art. "Fragments" comprise a portion of the intact antibody, generally the antigen binding site or variable region. Examples of antibody fragments include Fab, Fab', Fab'-SH, F (ab') 2, and Fv fragments; di- abodies; any antibody fragment that is a polypeptide having a primary structure consisting of one uninterrupted sequence of contiguous amino acid residues (referred to herein as a "single-chain antibody fragment" or "single chain polypeptide"), including without limitation (1 ) single-chain Fv molecules (2) single chain polypeptides containing only one light chain variable domain, or a fragment thereof that contains the three CDRs of the light chain variable domain, without an associated heavy chain moiety and (3) single chain polypeptides containing only one heavy chain variable region, or a fragment thereof containing the three CDRs of the heavy chain variable region, without an associated light chain moiety; and multispecific antibodies formed from antibody fragments. Included, inter alia, are a nanobody, domain antibody, single domain antibody or a "dAb".
In certain embodiments, the DNA of a hybridoma producing an antibody can be modified prior to insertion into an expression vector, for example, by substituting the coding sequence for human heavy- and light-chain constant domains in place of the homologous non-human sequences (e.g., Morrison et al., PNAS pp. 6851 (1984)), or by covalently joining to the immunoglobulin coding sequence all or part of the coding sequence for a non- immunoglobulin polypeptide. In that manner, "chimeric" or "hybrid" antibodies are prepared that have the binding specificity of the original antibody. Typically, such non-immunoglobulin polypeptides are substituted for the constant domains of an antibody.
Thus, according to another embodiment, the antibody is humanized. "Humanized" forms of antibodies according are specific chimeric immunoglobulins, immunoglobulin chains or fragments thereof (such as Fv, Fab, Fab', F (ab') 2, or other antigen-binding subsequences of antibodies) which contain minimal sequence derived from the murine immunoglobulin. For the most part, humanized antibodies are human immunoglobulins (recipient antibody) in which residues from a complementary-determining region (CDR) of the recipient are re- placed by residues from a CDR of the original antibody (donor antibody) while maintaining the desired specificity, affinity, and capacity of the original antibody.
In some instances, Fv framework residues of the human immunoglobulin may be replaced by corresponding non-human residues. Furthermore, humanized antibodies can comprise residues that are not found in either the recipient antibody or in the imported CDR or framework sequences. These modifications are made to further refine and optimize antibody performance. In general, the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the CDR regions correspond to those of the original antibody and all or substantially all of the FR re- gions are those of a human immunoglobulin consensus sequence. The humanized antibody optimally also will comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin. For further details see Jones et al., Nature, 321 , pp. 522 (1986); Reichmann et al, Nature, 332, pp. 323 (1988); Presta, Curr. Op. Struct. Biol., 2, pp. 593 (1992); Verhoeyen et Science, 239, pp. 1534; and U.S. Patent No. 4,816,567, the entire disclosures of which are herein incorporated by reference.) Methods for humanizing the antibodies are well known in the art.
The choice of human variable domains, both light and heavy, to be used in making the humanized antibodies is very important to reduce antigenicity. According to the so-called "best-fit" method, the sequence of the variable domain of an antibody is screened against the entire library of known human variable-domain sequences. The human sequence which is closest to that of the mouse is then accepted as the human framework (FR) for the humanized antibody (Sims et al., J. Immunol. 151 , pp. 2296 (1993); Chothia and Lesk, J. Mol. 196, 1987, pp. 901 ). Another method uses a particular framework from the consensus sequence of all human antibodies of a particular subgroup of light or heavy chains. The same frame- work can be used for several different humanized antibodies (Carter et al., PNAS 89, pp. 4285 (1992); Presta et al., J. Immunol., 151 , p. 2623 (1993)).
It is further important that antibodies be humanized with retention of high affinity for KIR3DL2 receptors and other favorable biological properties. To achieve this goal, according to one method, humanized antibodies are prepared by a process of analysis of the parental sequences and various conceptual humanized products using three-dimensional models of the parental and humanized sequences. Three-dimensional immunoglobulin models are commonly available and are familiar to those skilled in the art. Computer programs are available which illustrate and display probable three-dimensional structures of selected candidate immunoglobulin sequences. Inspection of these displays permits analysis of the likely role of the residues in the functioning of the candidate immunoglobulin sequence, i.e., the analysis of residues that influence the ability of the candidate immunoglobulin to bind its antigen. In this way, FR residues can be selected and combined from the consensus and import sequences so that the desired antibody characteristic, such as increased affinity for the target antigen (s), is achieved. In general, the CDR residues are directly and most substantially in- volved in influencing antigen binding.
Another method of making "humanized" monoclonal antibodies is to use a Xen- oMouse (Abgenix, Fremont, CA) as the mouse used for immunization. A XenoMouse is a murine host that has had its immunoglobulin genes replaced by functional human immunoglobulin genes. Thus, antibodies produced by this mouse or in hybridomas made from the B cells of this mouse, are already humanized. The XenoMouse is described in United States Patent No. 6,162,963, which is herein incorporated in its entirety by reference.
Human antibodies may also be produced according to various other techniques, such as by using, for immunization, other transgenic animals that have been engineered to express a human antibody repertoire (Jakobovitz et al., Nature 362 (1993) 255), or by selec- tion of antibody repertoires using phage display methods. Such techniques are known to the skilled person and can be implemented starting from monoclonal antibodies as disclosed in the present application.
A KIR3DL2 binding compound, e.g., an anti-KIR3DL2 antibody, may be further bound to a second moiety, wherein the antibody is capable of delivering the second moiety to a KIR3DL2-expressing cell. Optionally the second moiety is a therapeutic agent, a toxic agent, and/or a detectable agent.
While antibodies in underivatized or unmodified form, particularly of the lgG1 or lgG3 type are expected to inhibit the proliferation of the overproliferating cells or be cytotoxic towards overproliferating cells such as in those from a celiac disease patient, e.g., by direct- ing ADCC and/or CDC toward KIR3DL2-expressing celiac disease cells, it is also possible to prepare derivatized antibody immunoconjugates that are cytotoxic. In one embodiment, once the KIR3DL2 specific antibodies are isolated and optionally otherwise modified (e.g. humanized), they will be derivatized to make them toxic to cells. In this way, administration of the antibody to celiac disease patients will lead to the relatively specific binding of the anti- body to overproliferating cells, thereby directly killing or inhibiting the cells underlying the disorder.
Any of a large number of toxic moieties or strategies can be used to produce such antibodies. In certain embodiments, the antibodies will be directly derivatized with radioisotopes or other toxic compounds. Examples of toxic agents used in immunoconjugates in develop- ment include, in particular, for example taxanes, anthracyclines, camptothecins, epothilones, mytomycins, combretastatins, vinca alkaloids, nitrogen mustards, maytansinoids, cali- cheamycins, duocarmycins, tubulysins, dolastatins and auristatins, enediynes, pyrrolobenzo- diazepines, ethylenimines, radioisotopes, therapeutic proteins and peptides, and toxins or fragments thereof. Any type of moiety with a cytotoxic or cytoinhibitory effect can be used in conjunction with the present antibodies to inhibit or kill specific NK receptor expressing cells, including radioisotopes, toxic proteins, toxic small molecules, such as drugs, toxins, im- munomodulators, hormones, hormone antagonists, enzymes, oligonucleotides, enzyme inhibitors, therapeutic radionuclides, angiogenesis inhibitors, chemotherapeutic drugs, vinca alkaloids, epidophyllotoxins, antimetabolites, alkylating agents, antibiotics, antimitotics, anti- angiogenic and apoptotoic agents, particularly doxorubicin, methotrexate, camptothecans, nitrogen mustards, gemcitabine, alkyl sulfonates, nitrosoureas, triazenes, folic acid analogs, pyrimidine analogs, purine analogs, platinum coordination complexes, Pseudomonas exotoxin, ricin, 5-fluorouridine, ribonuclease (RNase), DNase I, Staphylococcal enterotoxin-A, pokeweed antiviral protein, gelonin, diphtherin toxin, Pseudomonas exotoxin, and Pseudo- monas endotoxin and others (see, e.g., Remington's Pharmaceutical Sciences, 19th Ed. (Mack Publishing Co. 1995); Goodman and Gilman's The Pharmacological Basis of Therapeutics (McGraw Hill, 2001 ); Pastan et al. (1986) Cell 47:641 ; Goldenberg (1994) Cancer Journal for Clinicians 44:43; U.S. Pat. No. 6,077,499; the entire disclosures of which are herein incorporated by reference).
In one embodiment, the antibody will be derivatized with a radioactive isotope, such as 1-131. Any of a number of suitable radioactive isotopes can be used, including, but not limited to, lndium-1 1 1 , Lutetium-171 , Bismuth-212, Bismuth-213, Astatine-21 1 , Copper-62, Copper-64, Copper-67, Yttrium-90, lodine-125, lodine-131 , Phosphorus-32, Phosphorus-33, Scandium-47, Silver-1 1 1 , Gallium-67, Praseodymium-142, Samarium-153, Terbium-161 , Dysprosium-166, Holmium-166, Rhenium-186, Rhenium-188, Rhenium-189, Lead-212, Ra- dium-223, Actinium-225, lron-59, Selenium-75, Arsenic-77, Strontium-89, Molybdenum-99, Rhodium-105, Palladium-109, Praseodymium-143, Promethium-149, Erbium-169, Iridium- 194, Gold-198, Gold-199, and Lead-21 1. The radionuclide may have a decay energy in the range of 20 to 6,000 keV, optionally in the ranges 60 to 200 keV for an Auger emitter, 100- 2,500 keV for a beta emitter, and 4,000-6,000 keV for an alpha emitter. Also provided are radionuclides that substantially decay with generation of alpha-particles.
In view of the ability of the anti-KIR3DL2 antibodies to induce ADCC and CDC, the antibodies can also be made with modifications that increase their ability to bind Fc receptors which can affect effector functions such as antibody-dependent cytotoxicity, mast cell degranulation, and phagocytosis, as well as immunomodulatory signals such as regulation of lymphocyte proliferation and antibody secretion. Typical modifications include modified human lgG1 constant regions comprising at least one amino acid modification (e.g. substitution, deletions, insertions), and/or altered types of glycosylation, e.g., hypofucosylation. Such modifications can affect interaction with Fc receptors: FcyRI (CD64), FcyRI I (CD32), and FcyRI II (CD 16). FcyRI (CD64), FcyRIIA (CD32A) and FcvRIII (CD 16) are activating (i.e. , immune system enhancing) receptors while FcyRIIB (CD32B) is an inhibiting (i.e., immune system dampening) receptor. A modification may, for example, increase binding of the Fc domain to FcyRllla on effector (e.g. NK) cells.
Anti-KIR3DL2 antibodies may comprise an Fc domain (or portion thereof) of human lgG1 or lgG3 isotype, optionally modified. Residues 230-341 (Kabat EU) are the Fc CH2 region. Residues 342-447 (Kabat EU) are the Fc CH3 region. Anti-KIR3DL2 antibodies may comprise a variant Fc region having one or more amino acid modifications (e.g., substitutions, deletions, insertions) in one or more portions, which modifications increase the affinity and avidity of the variant Fc region for an FcyR (including activating and inhibitory FcyRs). In some embodiments, said one or more amino acid modifications increase the affinity of the variant Fc region for FcyRIIIA and/or FcyRIIA. In another embodiment, the variant Fc region further specifically binds FcyRIIB with a lower affinity than does the Fc region of the comparable parent antibody (i.e., an antibody having the same amino acid sequence as the antibody except for the one or more amino acid modifications in the Fc region). For example, the one or both of the histidine residues at amino acid positions 310 and 435 may be substituted, for example by lysine, alanine, glycine, valine, leucine, isoleucine, proline, methionine, tryptophan, phenylalanine, serine or threonine (see, e.g. PCT publication no. WO 2007/080277); such substituted constant regions provide decreased binding to the inhibitory FcyRIIB without decreasing binding to the activatory FcyRIIIA. In some embodiments, such modifications in- crease the affinity of the variant Fc region for FcyRIIIA and/or FcyRIIA and also enhance the affinity of the variant Fc region for FcyyRIIB relative to the parent antibody. In other embodiments, said one or more amino acid modifications increase the affinity of the variant Fc region for FcyRIIIA and/or FcyRIIA but do not alter the affinity of the variant Fc regions for FcyRIIB relative to the Fc region of the parent antibody. In another embodiment, said one or more amino acid modifications enhance the affinity of the variant Fc region for FcyRIIIA and FcyRIIA but reduce the affinity for FcyRIIB relative to the parent antibody. Increased affinity and/or avidity results in detectable binding to the FcyR or FcyR- related activity in cells that express low levels of the FcyR when binding activity of the parent molecule (without the modified Fc region) cannot be detected in the cells.
The affinities and binding properties of the molecules for an FcyR can be determined using in vitro assays (biochemical or immunological based assays) known in the art for determining antibody-antigen or Fc-FcyR interactions, i.e., specific binding of an antigen to an antibody or specific binding of an Fc region to an FcyR, respectively, including but not limited to ELISA assay, surface plasmon resonance assay, immunoprecipitation assays.
In some embodiments, the molecules comprising a variant Fc region comprise at least one amino acid modification (for example, possessing 1 , 2, 3, 4, 5, 6, 7, 8, 9, or more amino acid modifications) in the CH3 domain of the Fc region. In other embodiments, the molecules comprising a variant Fc region comprise at least one amino acid modification (for example, possessing 1 , 2, 3, 4, 5, 6, 7, 8, 9, or more amino acid modifications) in the CH2 domain of the Fc region, which is defined as extending from amino acids 231 -341 . In some embodiments, the molecules comprise at least two amino acid modifications (for example, possessing 2, 3, 4, 5, 6, 7, 8, 9, or more amino acid modifications), wherein at least one such modification is in the CH3 region and at least one such modification is in the CH2 region. Amino acid modifications may be made for example in the hinge region. In a particular em- bodiment, the invention encompasses amino acid modification in the CH 1 domain of the Fc region, which is defined as extending from amino acids 216-230.
Any combination of Fc modifications can be made, for example any combination of different modifications disclosed in United States Patents Nos. US, 7,632,497; 7,521 ,542; 7,425,619; 7,416,727; 7,371 ,826; 7,355,008; 7,335,742; 7,332,581 ; 7, 183,387; 7, 122,637; 6,821 ,505and 6,737,056; in PCT Publications Nos. WO201 1/109400; WO 2008/105886; WO 2008/002933; WO 2007/021841 ; WO 2007/106707; WO 06/088494; WO 05/1 15452; WO 05/1 10474; WO 04/1032269; WO 00/42072; WO 06/088494; WO 07/024249; WO 05/047327; WO 04/099249 and WO 04/063351 ; and in Presta, L.G. et al. (2002) Biochem. Soc. Trans. 30(4):487-490; Shields, R.L. et al. (2002) J. Biol. Chem. 26; 277(30):26733- 26740 and Shields, R.L. et al. (2001 ) J. Biol. Chem. 276(9):6591 -6604).
Anti-KIR3DL2 antibodies may comprise a variant Fc region, wherein the variant Fc region comprises at least one amino acid modification (for example, possessing 1 , 2, 3, 4, 5, 6, 7, 8, 9, or more amino acid modifications) relative to a wild-type Fc region, such that the molecule has an enhanced effector function relative to a molecule comprising a wild-type Fc region, optionally wherein the variant Fc region comprises a substitution at any one or more of positions 221 , 239, 243, 247, 255, 256, 258, 267, 268, 269, 270, 272, 276, 278, 280, 283, 285, 286, 289, 290, 292, 293, 294, 295, 296, 298, 300, 301 , 303, 305, 307, 308, 309, 310, 31 1 , 312, 316, 320, 322, 326, 329, 330, 332, 331 , 332, 333, 334, 335, 337, 338, 339, 340, 359, 360, 370, 373, 376, 378, 392, 396, 399, 402, 404, 416, 419, 421 , 430, 434, 435, 437, 438 and/or 439. Anti-KIR3DL2 antibodies may comprise a variant Fc region, wherein the variant Fc region comprises at least one amino acid modification (for example, possessing 1 , 2, 3, 4, 5, 6, 7, 8, 9, or more amino acid modifications) relative to a wild-type Fc region, such that the molecule has an enhanced effector function relative to a molecule comprising a wild-type Fc region, optionally wherein the variant Fc region comprises a substitution at any one or more of positions 329, 298, 330, 332, 333 and/or 334 (e.g. S239D, S298A, A330L, I332E, E333A and/or K334A substitutions).
In one embodiment, antibodies having variant or wild-type Fc regions may have altered glycosylation patterns that increase Fc receptor binding ability of antibodies. Such car- bohydrate modifications can be accomplished by, for example, expressing the antibody in a host cell with altered glycosylation machinery. Cells with altered glycosylation machinery have been described in the art and can be used as host cells in which to express recombinant antibodies to thereby produce an antibody with altered glycosylation. See, for example, Shields, R.L. et al. (2002) J. Biol. Chem. 277:26733-26740; Umana et al. (1999) Nat. Bio- tech. 17:176-1 , as well as, European Patent No: EP 1 ,176,195; PCT Publications WO 06/133148; WO 03/035835; WO 99/54342, each of which is incorporated herein by reference in its entirety.
Generally, such antibodies with altered glycosylation are "glyco-optimized" such that the antibody has a particular N-glycan structure that produces certain desirable properties, including but not limited to, enhanced ADCC and effector cell receptor binding activity when compared to non-modified antibodies or antibodies having a naturally occurring constant region and produced by murine myeloma NSO and Chinese Hamster Ovary (CHO) cells (Chu and Robinson, Current Opinion Biotechnol. 2001 , 12: 180-7), HEK293T-expressed antibodies as produced herein in the Examples section, or other mammalian host cell lines common- ly used to produce recombinant therapeutic antibodies.
Monoclonal antibodies produced in mammalian host cells contain an N- linked glycosylation site at Asn297 of each heavy chain. Glycans on antibodies are typically complex bi- atennary structures with very low or no bisecting N-acetylglucosamine (bisecting GlcNAc) and high levels of core fucosylation. Glycan temini contain very low or no terminal sialic acid and variable amounts of galactose. For a review of effects of glycosylation on antibody function, see, e.g., Wright & Morrison, Trend Biotechnol.15:26- 31 (1997). Considerable work shows that changes to the sugar composition of the antibody glycan structure can alter Fc effector functions. The important carbohydrate structures contributing to antibody activity are believed to be the fucose residues attached via alpha-1 ,6 linkage to the innermost N- acetylglucosamine (GlacNAc) residues of the Fc region N-linked oligosaccharides (Shields et al., 2002).
FcyR binding requires the presence of oligosaccharides covalently attached at the conserved Asn297 in the Fc region of human IgGI, lgG2 or lgG3 type. Non-fucosylated oligosaccharides structures have recently been associated with dramatically increased in vitro ADCC activity. "Asn 297" means amino acid asparagine located at about position 297 in the Fc region; based on minor sequence variations of antibodies, Asn297 can also be located some amino acids (usually not more than +3 amino acids) upstream or downstream.
Historically, antibodies produced in CHO cells contain about 2 to 6% in the population that are nonfucosylated. YB2/0 (rat myeloma) and Lecl3 cell line (a lectin mutant of CHO line which has a deficient GDP- mannose 4,6-dehydratase leading to the deficiency of GDP- fucose or GDP sugar intermediates that are the substrate of alpha6-fucosyltransferase have been reported to produce antibodies with 78 to 98% non-fucosylated species. In other examples, RNA interference (RNAi) or knock-out techniques can be employed to engineer cells to either decrease the FUT8 mRNA transcript levels or knock out gene expression entirely, and such antibodies have been reported to contain up to 70% non-fucosylated glycan.
An antibody that binds to KIR3DL2 may be glycosylated with a sugar chain at Asn297. In one embodiment, an antibody will comprise a constant region comprising at least one amino acid alteration in the Fc region that improves antibody binding to FcyRllla and/or ADCC.
In one aspect, the antibodies are hypofucosylated in their constant region. Such antibodies may comprise an amino acid alteration or may not comprise an amino acid alteration but be produced or treated under conditions so as to yield such hypofucosylation. In one aspect, an antibody composition comprises a chimeric, human or humanized antibody described herein, wherein at least 20, 30, 40, 50, 60, 75, 85, 90, 95% or substantially all of the antibody species in the composition have a constant region comprising a core carbohydrate structure (e.g. complex, hybrid and high mannose structures) which lacks fucose. In one embodiment, provided is an antibody composition which is free of antibodies comprising a core carbohydrate structure having fucose. The core carbohydrate will preferably be a sugar chain at Asn297.
In one embodiment, an antibody composition, e.g. a composition comprising antibodies which bind to KIR3DL2, are glycosylated with a sugar chain at Asn297, wherein the antibodies are partially fucosylated. Partially fucosylated antibodies are characterized in that the proportion of anti-KIR3DL2 antibodies in the composition that lack fucose within the sugar chain at Asn297 is between 20% and 90%, between 20% and 80%, between 20% and 50%, 55%, 60%, 70% or 75%, between 35% and 50%, 55%, 60%, 70% or 75%, or between 45% and 50%, 55%, 60%, 70% or 75%. Optionally the antibody is of human IgGI or lgG3 type.
The sugar chain show can further show any characteristics (e.g. presence and proportion of complex, hybrid and high mannose structures), including the characteristics of N- linked glycans attached to Asn297 of an antibody from a human cell, or of an antibody re- combinantly expressed in a rodent cell, murine cell (e.g. CHO cell) or in an avian cell.
In one embodiment, the antibody is expressed in a cell that is lacking in a fucosyl- transferase enzyme such that the cell line produces proteins lacking fucose in their core carbohydrates. For example, the cell lines Ms704, Ms705, and Ms709 lack the fucosyltransfer- ase gene, FUT8 (alpha (1 ,6) fucosyltransferase), such that antibodies expressed in the Ms704, Ms705, and Ms709 cell lines lack fucose on their core carbohydrates. These cell lines were created by the targeted disruption of the FUT8 gene in CHO/DG44 cells using two replacement vectors (see U.S. Patent Publication No. 200401 10704 by Yamane et al.; and Yamane-Ohnuki et al. (2004) Biotechnol Bioeng 87:614-22, the disclosures of which are incorporated herein by reference). Other examples have included use of antisense suppres- sion, double-stranded RNA (dsRNA) interference, hairpin RNA (hpRNA) interference or in- tron-containing hairpin RNA (ihpRNA) interference to functionally disrupt the FUT8 gene. In one embodiment, the antibody is expressed in a cell line with a functionally disrupted FUT8 gene, which encodes a fucosyl transferase, such that antibodies expressed in such a cell line exhibit hypofucosylation by reducing or eliminating the alpha 1 ,6 bond-related enzyme.
In one embodiment, the antibody is expressed in cell lines engineered to express gly- coprotem-modifying glycosyl transferases (e.g., beta(l,4)-N-acetylglucosaminyl-transferase III (GnTHI)) such that antibodies expressed in the engineered cell lines exhibit increased bisecting GlcNac structures which results in increased ADCC activity of the antibodies (PCT Publication WO 99/54342 by Umana et al.; and Umana et al. (1999) Nat. Biotech. 17:176-180, the disclosures of which are incorporated herein by reference).
In another embodiment, the antibody is expressed and the fucosyl residue(s) is cleaved using a fucosidase enzyme. For example, the fucosidase alpha-L- fucosidase removes fucosyl residues from antibodies (Tarentino, et al. (1975) Biochem. 14:5516-5523). In other examples, a cell line producing an antibody can be treated with a glycosylation inhibi- tor; Zhou et al. Biotech, and Bioengin. 99: 652-665 (2008) described treatment of CHO cells with the alpha-mannosidase I inhibitor, kifunensine, resulting in the production of antibodies with non-fucosylated oligomannose-type N-glucans.
In one embodiment, the antibody is expressed in a cell line which naturally has a low enzyme activity for adding fucosyl to the N-acetylglucosamine that binds to the Fc region of the antibody or does not have the enzyme activity, for example the rat myeloma cell line YB2/0 (ATCC CRL 1662). Other example of cell lines include a variant CHO cell line, Led 3 cells, with reduced ability to attach fucosyl to Asn(297)-linked carbohydrates, also resulting in hypofucosylation of antibodies expressed in that host cell (WO 03/035835 (Presta et al); and Shields, RX. et al. (2002) J. Biol. Chem. 277:26733-26740, the disclosures of which are in- corporated herein by reference). In another embodiment, the antibody is expressed in an avian cell, e.g., a EBx® cell (Vivalis, France) which naturally yields antibodies with low fu- cose content e.g WO2008/142124. Hypofucosylated glycans can also be produced in cell lines of plant origin, e.g. WO 07/084926A2 (Biolex Inc.), WO 08/006554 (Greenovation Biotech GMBH), the disclosures of which are incorporated herein by reference. Antibody Formulations
Pharmaceutically acceptable carriers that may be used in these compositions include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carbox- ymethylcellulose, polyacrylates, waxes, polyethylene-polyoxypropylene- block polymers, polyethylene glycol and wool fat. The antibodies may be employed in a method of modulating, e.g inhibiting, the activity of KIR3DL2-expressing cells in a patient. This method comprises the step of contacting said composition with said patient. Such method will be useful for both prophylaxis and therapeutic purposes.
For use in administration to a patient, the composition will be formulated for administration to the patient. The compositions may be administered orally, parenterally, by inhala- tion spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir. The used herein includes subcutaneous, intravenous, intramuscular, intra-articular, intra-synovial, intrasternal, intrathecal, intrahepatic, intralesional and intracranial injection or infusion techniques.
Sterile injectable forms of the compositions may be aqueous or an oleaginous sus- pension. These suspensions may be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, for example as a solution in 1 ,3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution and iso- tonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose, any bland fixed oil may be employed including synthetic mono-or diglycerides. Fatty acids, such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically- acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions. These oil solutions or suspensions may also contain a long-chain alcohol diluent or disper- sant, such as carboxymethyl cellulose or similar dispersing agents that are commonly used in the formulation of pharmaceutically acceptable dosage forms including emulsions and suspensions. Other commonly used surfactants, such as Tweens, Spans and other emulsify- ing agents or bioavailability enhancers which are commonly used in the manufacture of pharmaceutically acceptable solid, liquid, or other dosage forms may also be used for the purposes of formulation.
Several monoclonal antibodies have been shown to be efficient in clinical situations, such as Rituxan™ (rituximab), Herceptin™ (Trastuzumab) or Xolair™ (Omalizumab), and similar administration regimens (i.e., formulations and/or doses and/or administration protocols) may be used with the antibodies. For example, an antibody present in a pharmaceutical composition can be supplied at a concentration of 10 mg/mL in either 100 mg (10 mL) or 500 mg (50 mL) single-use vials. The product is formulated for IV administration in 9.0 mg/mL sodium chloride, 7.35 mg/mL sodium citrate dihydrate, 0.7 mg/mL polysorbate 80, and Sterile Water for Injection. The pH is adjusted to 6.5. An exemplary suitable dosage range for an antibody in a pharmaceutical composition may between about 1 mg/m2 and 500 mg/m2. However, it will be appreciated that these schedules are exemplary and that an optimal schedule and regimen can be adapted taking into account the affinity and tolerability of the particular antibody in the pharmaceutical composition that must be determined in clinical tri- als. A pharmaceutical composition for injection (e.g., intramuscular, i.v.) could be prepared to contain sterile buffered water (e.g. 1 ml for intramuscular), and between about 1 ng to about 100 mg, e.g. about 50 ng to about 30 mg or more, e.g., about 5 mg to about 25 mg, of an an- ti-KIR3DL2 antibody.
According to another embodiment, the antibody compositions may further comprise another therapeutic agent, including agents normally utilized for the particular therapeutic purpose for which the antibody is being administered, notably for the treatment of a celiac disease. The additional therapeutic agent will normally be present in the composition in amounts typically used for that agent in a monotherapy for the particular disease or condition being treated. Such therapeutic agents include, but are not limited to anti-inflammation agents, steroids, immune system suppressors, antibiotics, antivirals and other antibodies and fragments thereof.
Diagnosis and treatment of disease
Celiac disease is usually detected by serologic testing of celiac-specific antibodies, notably the performance of tissue transglutaminase (TTG) antibody testing. The diagnosis is confirmed by duodenal mucosal biopsies.
Diagnosis of RCD is a more recent classification and requires specialised small bowel investigations, notably enteroscopy and small bowel imaging, and laboratory tests, notably immunohistochemistry, molecular analysis, and flow cytometry of isolated lELs. Two types of RCD, RCD type I and RCD type II are generally distinguished according to the normal (RCDI) or abnormal (RCDII) phenotype of intestinal lELs. In RCDI, histological examination is similar to that found in active celiac disease with villous atrophy and increased normal IEL. In RCDII, an abnormal population of lELs is identified: over 25% of the CD103+ of CD45+ lELs lacking surface CD3-TCR complexes on flow cytometry, or more than 50% lELs expressing intracellular CD3e but no CD8 in formol-fixed sections and the presence of a detectable clonal rearrangement of the gamma chain of the TCD in duodenal biopsies. In any of the methods of the invention,
The disclosure shows that KIR3DL2 is expressed in celiac disease, including in RCDI and RCDII. It is also shown that KIR3DL2 is expressed on enteropathy-associated T- cell lymphoma (EATL), a complication of CD (for EATL see also PCT/EP2014/053340 filed 20 February 2014, the disclosure of which is incorporated by reference).
Provided in one aspect are methods useful in the diagnosis, prognosis and monitoring of a celiac disease or a complication thereof (e.g. EATL) in an individual involving detection of KIR3DL2. In one embodiment, the methods comprise determining the level of expres- sion of a KIR3DL2 nucleic acid or polypeptide in a biological sample from a patient, e.g. in lymphocytes (e.g. intraepithelial lymphocytes) found in a biological sample. Abnormal lELs in celiac patients can be found in a sample obtained as a biopsy. Particularly in RCDII patients, abnormal lELs can generally be found in and outside the intestine as cells have tendency to disseminate in and outside the intestine. Abnormal lELs may be found for example in mesen- teric lymph nodes, blood, and bone marrow, and in different epitheliums such as lung and skin. A high percentage of abnormal cells have been found to be predictive of abnormal circulating cells in peripheral blood.
In one embodiment, the methods comprise: (a) determining whether an individual has a celiac disease (or a complication thereof, e.g. a lymphoma, an EATL); and (b) if the individual has a celiac disease (or a complication thereof), determining whether an individual has celiac disease cells (e.g. a population of abnormal lELs) that express a KIR3DL2 polypeptide. Optionally the method further comprises determining whether disease cells also express other markers of abnormal lELs at their surface, for example determining whether cells are CD103+, CD45+, CD3-TCR-negative cells or intracellular CD3e+, CD8- cells. A finding that lELs express, or predominantly express, KIR3DL2 indicates that the disease can be treated with an anti-KIR3DL2 agent. KIRDL2 expression at the surface of lELs can be used for example as a tool for monitoring disease, assessing disease status, as a prognostic tool for evaluating response to treatment (with an anti-KIR3DL2 agent or a different therapeutic agent), or as a tool for evaluating prognosis without treatment, e.g. the occurrence of compli- cations such as EATL.
In one embodiment, the methods comprise determining the level of expression of a KIR3DL2 nucleic acid or polypeptide in a biological sample and comparing the level to a reference level (e.g. a value, weak cell surface staining, etc.) corresponding to a healthy(s) individual. Optionally, detecting a KIR3DL2 polypeptide in a biological sample comprises detect- ing KIR3DL2 polypeptide expressed on the surface of a lymphocyte.
Also provided is a method for the assessment of the development level of a celiac disease (staging disease) permitting the evaluation of the proportion (e.g. percentage) of abnormal lELs present in a patient. According to this method, cells from a biological sample collected from a body compartment, e.g., mesenteric lymph nodes, blood, and bone marrow, and in different epitheliums such as lung and skin, are brought into contact with an anti- KIR3DL2 antibody and lELs (e.g. the proportion of cells) expressing a KIR3DL2 polypeptide and/or other markers of abnormal lELs at their surface is measured. The cells may be, for example CD103+, CD45+, CD3-TCR-negative cells or intracellular CD3e+, CD8- cells. A finding that lELs express, or predominantly express, KIR3DL2 indicates that the disease can be treated with an anti-KIR3DL2 agent.
Also provided is a method for diagnosis of CD (e.g. CD, RCD, RCDI, RCDII), comprising bringing cells from a biological sample from an individual into contact with an anti- KIR3DL2 antibody and the proportion (e.g. percentage) of T cells expressing a KIR3DL2 polypeptide at their surface is measured, and comparing such proportion to the average propor- tion (e.g. percentage) of T cells expressing a KIR3DL2 polypeptide at their surface observed in non-celiac disease humans (e.g., in healthy humans), wherein a celiac disease-positive diagnosis is made when said measured proportion is significantly higher than said average proportion.
Further provided are therapeutic methods for treating individuals having a celiac disease, susceptible to a celiac disease or having experienced a celiac disease, wherein the treat- ment involves administering anti-KIR3DL2 antibodies, anti-KIR3DL2 antibody compositions, and/or related compositions, to an individual having or susceptible to celiac disease. In one embodiment, the celiac disease is an aggressive or advanced celiac disease (e.g. RCD, RCDI, RCDII, or otherwise with a poor prognosis of survival). In one embodiment, the celiac disease is a non-cutaneous celiac disease. In one embodiment, the celiac disease is an aggressive T cell lymphoma. In one embodiment, the patient has relapsing or refractory disease. In one embodiment, the patient has a poor prognosis for disease progression (e.g. poor prognosis for survival) or has a poor prognosis for response to a therapy, e.g. gluten- free diet, stem cell transplantation, antibody therapy, chemotherapy, etc.
Celiac disease diagnosis criteria can be those of standard medical guidelines, for example, according to the World Gastroenterology Organization (WGO) (see, e.g., World Gastroenterology Organization Practice Guidelines, Celiac Disease: World Gastroenterology Organization, 2007). See also, e.g., Rubio-Tapia et al. (2013) Am. J. Gastroenterology 108(5):656-677, the disclosures of which are incorporated herein by reference.
In one exemplary aspect, provided is a method of reducing progression of celiac disease in a mammalian host, (e.g., a human patient) having a detectable level of abnormal lELs comprising administering an anti-KIR3DL2 antibody, an anti-KIR3DL2 antibody composition, or a related composition (e.g., a nucleic acid encoding an anti-KIR3DL2 antibody), in an amount sufficient to detectably reduce the progression of the celiac disease in the host. Preferably the anti-KIR3DL2 antibody reduces the progression to or of lymphoma (e.g. EATL) in the host.
In one exemplary aspect, provided is a method of treating celiac disease in an individual having a poor disease prognosis and/or who has relapsed, is resistant or is not responsive to therapy with a first therapeutic agent.
Disease diagnosis and progression can be defined by standard criteria, including for each subtype of CD (e.g. RCDI, RCDII) is typically based on examination of enteroscopy, small bowel imaging, peripheral blood or tissue biopsy for histological features supplemented by detailed immunohistochemistry, flow cytometry and molecular genetics. Progression is optionally determined by assessing the presence and/or numbers of abnormal lELs. Meth- ods for detecting disease progression can be achieved by any suitable technique, several examples of which are known in the art. Examples of suitable techniques include PCR and RT-PCR (including for the diagnosis of complications such as lymphomas, detection of celiac disease associated genes or "markers"), biopsy, imaging techniques, karyotyping and other chromosomal analysis, immunoassay/immunocytochemical detection techniques, histological and/or histopathology assays, cell kinetic studies and cell cycle analysis, flow cytometry, and physical examination techniques (e.g., for physical symptoms).
Delivering anti-KIR3DL2 antibodies to a subject (either by direct administration or expression from a nucleic acid therein, such as from a pox viral gene transfer vector comprising anti-KIR3DL2 antibody-encoding nucleic acid sequence(s)) and practicing the other methods herein can be used to reduce, treat, prevent, or otherwise ameliorate any suitable aspect of celiac disease. The methods herein can be particularly useful in the reduction and/or amelioration of proliferation of abnormal lELs (e.g. percentage (abnormal lELs compared to healthy lymphocytes), number of abnormal lELs in circulation), and any parameter or symptom associated therewith (e.g. biomarkers). Methods that reduce, prevent, or other- wise ameliorate such aspects of disease progression, independently and collectively, are advantageous features.
In another aspect, provided is a method of reducing the risk of disease progression, reducing the risk of lymphoma in a cell population that has undergone initiation (e.g. in a RCDI or RCDII patient), and/or providing a therapeutic regimen for reducing lymphoma pro- gression in a human patient having celiac disease, which comprises administering to the patient one or more first treatments (e.g. induction therapy, such as a chemotherapeutic agent or an antibody) in an amount and regimen sufficient to achieve a response (partial or complete response), and then administering an amount of an Anti-KIR3DL2 antibody or related composition (or applying a combination administration method) to the patient.
In a further aspect, provided is a method of increasing the likelihood of survival over a relevant period in a human patient diagnosed with celiac disease. In another aspect, provided is a method for improving the quality of life of a celiac disease patient comprising administering to the patient a composition in an amount effective to improve the quality of life thereof. In a further aspect, methods described herein can be applied to significantly reduce the number of celiac disease cells in a vertebrate host, such that, for example, the total number of celiac disease cells (e.g., abnormal lELs) is reduced. In a related sense, provided is a method for killing (e.g. either directly or indirectly causing death of) celiac disease cells in a vertebrate, such as a human patient.
According to another embodiment, the antibody compositions may be used in com- bined treatments with one or more other treatments or therapeutic agents, including treatments and agents normally utilized for the particular therapeutic purpose for which the antibody is being administered, notably for the treatment of a celiac disease. The additional therapeutic agent will normally be administered in amounts and treatment regimens typically used for that agent in a monotherapy for the particular disease or condition being treated.
For example, a second treatment may include a gluten free diet (GFT) or a hemato- poetic stem cell transplantation.
A second therapeutic agent may for example include one or more chemotherapeutic drugs, antibodies that bind to a polypeptide selectively expressed on an abnormal IEL in a celiac disease patient, an immunomodulatory or immunosuppressive agent (e.g. an antibody that modulates the immune system, an agent that inhibits the IL-15 signalling pathway, an anti-IL-15 antibody, an agent that inhibits signalling by a protein tyrosine kinase, an agent that inhibits JAK3 signalling, STAT5 signalling and/or syk kinase), and/or a chemotherapeutic agent
In the treatment methods, the KIR3DL2-binding compound and the second thera- peutic agent or treatment can be administered separately, together or sequentially, or in a cocktail. In some embodiments, the KIR3DL2-binding compound is administered prior to the administration of the second therapeutic agent or treatment. For example, the KIR3DL2- binding compound can be administered approximately 0 to 30 days prior to the administration of the second therapeutic agent. In some embodiments, an KIR3DL2-binding compound is administered from about 30 minutes to about 2 weeks, from about 30 minutes to about 1 week, from about 1 hour to about 2 hours, from about 2 hours to about 4 hours, from about 4 hours to about 6 hours, from about 6 hours to about 8 hours, from about 8 hours to 1 day, or from about 1 to 5 days prior to the administration of the second therapeutic agent. In some embodiments, a KIR3DL2-binding compound is administered concurrently with the admin- istration of the therapeutic agents. In some embodiments, a KIR3DL2-binding compound is administered after the administration of the second therapeutic agent. For example, a KIR3DL2-binding compound can be administered approximately 0 to 30 days after the administration of the second therapeutic agent or treatment. In some embodiments, an KIR3DL2-binding compound is administered from about 30 minutes to about 2 weeks, from about 30 minutes to about 1 week, from about 1 hour to about 2 hours, from about 2 hours to about 4 hours, from about 4 hours to about 6 hours, from about 6 hours to about 8 hours, from about 8 hours to 1 day, or from about 1 to 5 days after the administration of the second therapeutic agent. EXAMPLES
Example 1 - Generation of KIR3DL2 -selective antibodies
Antibodies which bind KIR3DL2 but not closely related KIR3DL1 were generated by immunizing mice with recombinant KIR3DL2-Fc fusion protein. Supernatant (SN) of the growing hybridomas were tested by flow cytometry on Sezary Syndrome cell lines (HUT78, COU-L) and HEK-293T/KIR3DL2 Domain 0 - eGFP. Potentially interesting hybridomas se- lected from the initial screening were cloned by limiting dilution techniques in 96-wells plates. The secondary screen involved selection of hybridomas of interest by testing supernatants of the subclones by flow cytometry on HUT78 , COU-L, HEK-293T/KIR3DL1 Domain 0 - eGFP and HEK-293T/KIR3DL2 Domain 0 - eGFP. Positive subclones were injected into mice to produce ascitis and antibodies of interest were purified before being tested in a Biacore assay using rec KIR3DL2 chips, followed by various assays formats based on binding to human KIR3DL2-expressing cells.
Sequences of the variable domains of heavy (VH) and light (VL) chain of antibodies were amplified by PCR from the cDNA of each antibody. Sequences amplified were run on agarose gel then purified using the Qiagen Gel Extraction kit. VH and VL sequences were then sub-cloned into the Lonza expression vectors (Double-Gene Vectors) using the InFu- sion system (Clontech) according to the manufacturer's instructions. After sequencing, vectors containing the VH and VL sequences were prepared as Maxiprep using the Promega PureYield™ Plasmid Maxiprep System. Vectors were then used for HEK-293T cell transfec- tion using Invitrogen's Lipofectamine 2000 according to the manufacturer instructions. Antibodies generated included, inter alia, 19H12 and 12B1 1 .
Competition assays were conducted by flow cytometry according to the methods described. Hut-78 cells were harvested and stained in PBS 1 X / BSA 0,2% / EDTA 2 mM buffer during 1 H at 4°C using 5 μg ml of increasing concentrations of the antibodies, includ- ing 19H12, 12B1 1 and AZ158 (previously identified) (0.006-200 Mg/ml). After two washes, staining data were acquired on a BD FACS Canto II and analyzed using the FlowJo software.
Increasing concentrations of (naked) 19H12, 12B1 1 , and AZ158 were used to shift labelled antibody bound to KIR3DL2 at the surface of HUT78 SS cell lines. Antibody AZ158 (anti-DO domain antibody) does not compete with the KIR3DL2 antibodies 19H12 or 12B1 1 for binding to KIR3DL2.
Antibodies were further tested for binding to a series of KIR3DL2 mutants. Antibodies 19H12 and 12B1 1 did not show any loss of binding to unmutated wild type KIR3DL2 (WTaKIR3DL2), but lost binding to mutant 1 1 having P179T and S181T substitutions as well as to mutant 1 1A1 having V178A and H180S substitutions. The principal epitope of these antibodies 19H12, 18B10 and 12B1 1 therefore includes residues P179, S181 , V178 and/or H180. These residues at positions 179 and 181 in mutant 1 1 correspond to the residues present at in KIR3DL1 (KIR3DL1 has T179 and T181 ). Residues P179 and S181 in particular are within the D1 domain of KIR3DL2 and on the opposite face on the KIR3DL2 protein of the HLA-binding regions (i.e. the HLA binding pocket). Each of antibodies 15C1 1 , 19H12, 18B10 and 12B1 1 had reduced binding (full loss of binding for 15C1 1 and 19H12) to mutant M1 1A4 having substitutions E130S, H131 S and R145S.. These residues at positions 179 and 181 in mutant 1 1 correspond to the residues present at in KIR3DL1 (KIR3DL1 has T179 and T181 ). Residues P179 and S181 in particular are within the D1 domain of KIR3DL2 and on the opposite face on the KIR3DL2 protein of the HLA-binding regions (i.e. the HLA bind- ing pocket). Surface-exposed residues adjacent to these mutated residues can also contribute to the epitopes of the antibodies, including for example residues N99, H100, E130, H131 , F132, V178, H180, P182, Y183, and Q184 (reference to SEQ ID NO: 1 ) located at the surface of KIR3DL2 in the region of the P179/S181 epitope but outside of the region of the KIR3DL2 mutations which did not result in loss of binding of the antibodies (e.g., mutant 5 (residue P66) and mutant 8 (residues V127)).
Antibody 2B12 and other antibodies disclosed in PCT/EP2013/069302 filed 17 September 2013 had loss of binding to mutants having I60N and G62S substitutions and decrease in binding to mutants having P14S, S15A and H23S substitutions, but did not lose binding to any other mutants. The principal epitope of these antibodies therefore includes residues I60 and/or G62 (and the epitope optionally further includes one or more of P14, S15, and H23). Residues 60 and 62 are within the DO domain of KIR3DL2. Residues 14, 15, 23, 60 and 61 are within the DO domain of KIR3DL2.
Example 2 - Antibodies are able to kill KIR3DL2 expressing targets via antibody dependent cellular cytotoxicity (ADCC)
Cell lysis through an ADCC mechanism was monitored in a radioactivity-based 51Cr release experiment (the level of radioactivity released from the preloaded target cells being proportional to their death). One million target cells were loaded with 51Cr for 1 hour at 37°C and washed 3 times. 3,000 cells were seeded per well (U-shaped bottom 96-well plates) and test mAbs are added at 10 or 20 μg/ml final concentration (or increasing concentrations if dose-response relationship is studied). Effector cells were added at a defined effectortarget ratio (in general 10:1 ) and the mixture was incubated at 37°C for 4 h. Supernatant is analyzed on a Lumaplate apparatus.
Anti-KIR3DL2 mAbs, tested at the same final concentration (10 g/ml), to kill KIR3DL2-transfected B221 target cells. The mAbs, including 19H12, were effective in medi- ating ADCC against KIR3DL2-expressing B221 targets.
Example 3 - Activity in mouse xenograft models of KIR3DL2 expressing human tumors
Tumor cells lines B221 and RAJI were made to express human KIR3DL2. Immune compromised mice used for B221 -KIR3DL2 and RAJI-KIR3DL2 models were NOD-SCID purchased from Charles River Laboratories. In the following models, 5 million human B221 - KIR3DL2 or RAJI-KIR3DL2 tumor cells (in 100 μΙ PBS as vehicle) were engrafted IV on Day 0 (DO), i.e. 1 day before treatment initiation (D1 ). From D1 , mice were treated IV with differ- ent doses of anti-KIR3DL2 mAbs (doses were adapted to mouse body weight) diluted in PBS, 2 injections per week for the duration of the whole experiment.
Control groups included, depending on the experiment:
PBS/placebo-treated mice as a control of normal/unaffected tumor growth;
mice injected with the same dose of isotype control-matched mAbs directed against an irrelevant antigen.
Mice were weighed and observed for clinical signs every 2 to 5 days depending on the model. Percent of body weight changes were calculated as compared to body weight at DO before tumor engraftment or to the highest body weight reached during the experiment. Mouse deaths or important weight losses were recorded and used to draw survival Kaplan- Meier curves and calculate improvement in survival as compared to control groups of mice.
The efficacy of lgG2b isotype murine anti-KIR3DL2 19H12 antibodies (given at 300 μg mouse, twice a week) was separately tested against SC B221 -KIR3DL2 xenografts or RAJI-KIR3DL2 xenografts (n = 6 NOD-SCID mice per group). Animals treated with anti- KIR3DL2 antibodies showed an increase in survival in comparison to mice treated with iso- type control-matched mAbs.
Example 4 - Improved detection methods reveal KIR3DL2 positive tumors
Tumor biopsies from RAJI-KI R3DL2 models and RAJI-KIR3DL2 cell lines were obtained and staining was performed on frozen sample using AZ158 antibody (see WO2010/081890) or antibodies 12B1 1 (see Example 1 ). KIR3DL2 was stained with anti- KIR3DL2 antibody by DAB chromogenic detection according to standard protocols, adapted for immunostaining with BenchMark XT Ventana Roche. For all staining control isotype (mlgG1 ) and control DAB were performed. Surprisingly while AZ158 was negative, tumors were positive when using 12B1 1 antibody at the same concentration (5 μg ml) of antibody (see Figure 1 ). Raising concentrations of antibody AZ158 (to 50 μg ml) generated extensive background staining that did not allow tumor samples to be differentiated from healthy tissue.
Next, tumor biopsies from cancer patients previously stained with AZ158 were reexamined using antibody 12B1 1 . Biopsies that had been KIR3DL2-negative with AZ158 were stained with 12B1 1 (i.e. becoming KIR3DL2-positive). Results are shown in Figure 2. Example 5 - KIR3DL2 is expressed in PTCL, including ortho visceral extranodal disease (NK/T lymphoma and EATL)
PTCL.
Tumor biopsies from PTCL patients were obtained and staining was performed on frozen samples. KIR3DL2 was stained with anti-KIR3DL2 antibody 12B1 1 (mlgG1 ) by DAB chromogenic detection according to standard protocols, adapted for immunostaining with BenchMark XT Ventana Roche. For all staining control isotype (mlgG1 ) and control DAB were performed. CD30 was additionally stained. Tumors 3, 4 and 5 were from the same patient. Tumors 1 -5 are from patients having PTCL not otherwise specified. Tumors 6-8 are mycosis fungoides samples, a cutaneous T cell lymphoma (CTCL).
Results are shown in Table A, below (LN = lymph node). Tumor sample characteristics are shown in Table B. PTCL from each of the samples from patient from which tumor samples 3, 4 and 5 were obtained had strong membranar staining, with a high percentage of cells being KIR3DL2 positive.
NK-/T-lymp oma and EA TL
MEC04 and SNK6 NK-/T-lymphoma cells were stained for KIR3DL2 expression using flow cytometry (FACS), together with characterization of various cell surface markers. KIR3DL2 was stained with anti-KIR3DL2 antibody linked to phycoerythrin (PE), Additional markers evaluated were hCD56 PE, hCD183/CXCR3 PE, hCD3 PE, hCD4 PE, hCD8 PE and CD54 / ICAM PE. Cells were harvested and stained using PE-labeled antibodies. After two washes, stainings were acquired on a BD FACS Canto II and analyzed using the FlowJo software.
Results are shown in Figure 3. Anti-KIR3DL2 antibody showed strong staining on the MEC04 cells. MEC04 cells were additionally positive for staining with CD183 (CXCR3), CD56 and CD54 (ICAM), but not CD3, CD4 or CD8 (the most common phenotype of extranodal NK-/T lymphomas are surface CD3- and CD56+).
NK-/T-lymphoma cells, and in particular extranodal NK-/T cell lymphoma, nasal type, can therefore express KIR3DL2 at significant levels, thereby providing the possibility to treat such disease with anti-KIR3DL2 antibodies. Additionally, KIR3DL2-positive NK-/T lym- phoma tumors were found to express CD183 (CXCR3), CD56 and CD54 (ICAM), which may permit administration of anti-KIR3DL2 in poor prognosis patients, for example those having CXCR3 expression typically associated with poor disease prognosis.
Studies were carried out by immunohistochemistry (IHC) to provide confirmation on patient samples and for different indications, by staining primary tumor cells from human pa- tients in frozen tissue sections with labelled anti-KIR3DL2 antibody. Briefly, cell lines known to be positive and negative for KIR3DL2 expression were used as positive and negative controls, respectively. In NK/T lymphomas, nasal type, 6 patient samples were tested, of which 5 samples were interpretable. 2 interpretable samples were positively stained which 3 were not, confirming that NK/T lymphomas express KIR3DL2. In samples from patients diagnosed with enteropathy-associated T cell lymphoma (EATL), of 5 interpretable sample, 2 were positively stained and 3 were negative for staining, confirming that EATL cells can express KIR3DL2.
Example 6 - KIR3DL2 is expressed in coeliac disease, refractory type 1 Sprue and refractory type 2 Sprue
Biopsies from patients with coeliac disease, refractory type 1 Sprue (RCD1 ) and refractory type 2 Sprue (RCDII) were obtained and staining was performed on frozen samples.
KIR3DL2 was stained with anti-KIR3DL2 antibody 12B1 1 (mlgG1 ) by DAB chromo- genic detection according to standard protocols, adapted for immunostaining with Bench- Mark XT Ventana Roche. For all staining control isotype (mlgG1 ) and control DAB were per- formed. Results are shown in Table C below.
Table C
Figure imgf000061_0001
In contrast to CD30, reported to be expressed only on EATL, but not on earlier stages of coeliac disease, including refractory ones such as type 1 and type 2 Sprue, KIR3DL2 is found expressed at all stages.
Thus, KIR3DL2 may be a relevant target for antibody-based therapies at all stages, from gluten-free diet responding coeliac disease to aggressive, lymphoma-stages like refractory type 2 sprue and EATL. Depletion of KIR3DL2-expressing pathologic cells infiltrating digestive epithelia may reduce local inflammation, may improve digestive function that is strongly impaired in patient, and may prevent evolution of the disease from earlier to later stages. In particular, it may prevent evolution to EATL. KIR3DL2 expression may also be useful to monitor coeliac disease progression towards refractory and tumor stages with poorer prognosis and improve patient therapeutic management. Table A
Tumor sample KIR3DL2 CD30 staining
Staining
Tumor 1 : Positive Negative
LN/Lymphoma peripheral T cells
Tumor 2: Negative Negative
Testis/Lymphoma peripheral T cells
Tumor 3: Negative Positive
Spleen/Lymphoma peripheral T cells
Tumor 4: Negative Positive
Spleen/Lymphoma peripheral T cells
Tumor 5: Negative Positive
Spleen/Lymphoma peripheral T cells
Tumor 6: Positive Positive
LN/Mycosis fungoides
Tumor 7: Positive Positive
LN/Mycosis fungoides
Tumor 8: Positive Positive
LN/Mycosis fungoides
Table B
Figure imgf000063_0001
All references, including publications, patent applications, and patents, cited herein are hereby incorporated by reference in their entirety and to the same extent as if each reference were individually and specifically indicated to be incorporated by reference and were set forth in its entirety herein (to the maximum extent permitted by law), regardless of any separately provided incorporation of particular documents made elsewhere herein.
The use of the terms "a" and "an" and "the" and similar referents in the context of describing the invention are to be construed to cover both the singular and the plural, unless otherwise indicated herein or clearly contradicted by context.
Unless otherwise stated, all exact values provided herein are representative of corresponding approximate values (e.g., all exact exemplary values provided with respect to a particular factor or measurement can be considered to also provide a corresponding approximate measurement, modified by "about," where appropriate).
The description herein of any aspect or embodiment of the invention using terms such as "comprising", "having," "including," or "containing" with reference to an element or elements is intended to provide support for a similar aspect or embodiment of the invention that "consists of", "consists essentially of", or "substantially comprises" that particular element or elements, unless otherwise stated or clearly contradicted by context (e.g., a composition described herein as comprising a particular element should be understood as also describing a composition consisting of that element, unless otherwise stated or clearly contradicted by context).
The use of any and all examples, or exemplary language (e.g., "such as") provided herein, is intended merely to better illuminate the invention and does not pose a limitation on the scope of the invention unless otherwise claimed. No language in the specification should be construed as indicating any non-claimed element as essential to the practice of the invention.

Claims

1. An antigen binding agent that binds a KIR3DL2 polypeptide, for use in the treatment or prevention of celiac disease in an individual.
2. The agent of claim 1 , wherein the individual has RCD type I .
3. The agent of claim 1 , wherein the individual has RCD type II.
4. The agent of claims 1 -3, wherein the treatment or prevention of celiac disease in an individual comprises:
a) determining the KIR3DL2 polypeptide status of abnormal lymphocytes within the individual having a celiac disease, and
b) upon a determination that the individual has KIR3DL2 polypeptide expressed on the surface of abnormal lymphocytes, administering to the individual said agent that binds a KIR3DL2 polypeptide.
5. The agent of claim 4, wherein determining the KIR3DL2 polypeptide status comprises determining whether a KIR3DL2 polypeptide is prominently expressed on the surface of said lymphocytes, and the agent that binds a KIR3DL2 polypeptide is administered if the individual is determined to have KIR3DL2 polypeptide prominently expressed on the surface of abnormal lymphocytes.
6. The agent of claims 4-5, wherein determining whether a KIR3DL2 polypeptide that is prominently expressed on the surface of said lymphocytes cells comprises obtaining from the individual a biological sample that comprises abnormal intraepithelial lymphocytes, bringing said cells into contact with an antibody that binds a KIR3DL2 polypeptide, and detecting cells that express KIR3DL2.
7. The agent of any one of the above claims, wherein the agent that binds a KIR3DL2 polypeptide is an antibody.
8. The agent of claims 6-7, wherein the antibody that binds a KIR3DL2 polypeptide competes with antibody 12B1 1 , 19H12 or 2B12 for binding to a human KIR3DL2 polypeptide.
9. The agent of any of the above claims, wherein treatment with the agent that binds a KIR3DL2 polypeptide is used in combination with treatment comprising administration of a second pharmaceutically active agent.
10. The agent of claims 7-9, wherein the antibody depletes KIR3DL2- expressing cells.
1 1 . The agent of claim 10, wherein the antibody comprises a human lgG1 or lgG3 constant region.
12. The agent of claims 10-1 1 , wherein the antibody comprises an Fc- engineered constant region.
13. The agent of claim 12, wherein the antibody comprises an amino acid modification that enhances binding to a human Fey receptor.
14. The agent of claims 7-13, wherein the antibody is linked to a toxic agent.
15. The agent of claims 7-14, wherein the anti-KIR3DL2 antibody binds human KIR3DL2 but does not bind to human KIR3DL1 .
16. The agent of claims 7-15, wherein the anti-KIR3DL2 antibody bind to the DO or D1 domain of human KIR3DL2.
17. The agent of claims 7-16, wherein the anti-KIR3DL2 antibody has reduced binding to a KIR3DL2 polypeptide having a mutation at residues P179 and/or residue S181 , compared to a wild-type KIR3DL2 polypeptide of SEQ ID NO: 1.
18. The agent of claims 7-16, wherein the anti-KIR3DL2 antibody has reduced binding to a KIR3DL2 polypeptide having a mutation at residues I60 and/or residue G62, compared to a wild-type KIR3DL2 polypeptide of SEQ ID NO: 1.
19. The agent of claims 7-18, wherein the anti-KIR3DL2 antibody competes with antibodies AZ158, 19H12, 2B12 or 12B1 1 for binding to human KIR3DL2.
20. The agent of claim 19, wherein the antibody comprises the VL and VH CDRs of 19H12, 2B12 or 12B1 1.
21. The agent of any of the above claims, wherein the antigen binding agent is administered as a pharmaceutically acceptable composition comprising a therapeutically effective amount of the antigen binding agent.
22. A method for diagnosing or monitoring a celiac disease in an individual, the method comprising obtaining from an individual a biological sample that comprises celiac disease cells, bringing said cells into contact with an antibody that binds a human KIR3DL2 polypeptide, and detecting cells that express KIR3DL2.
23. The method of claim 22, wherein the antibody that binds a KIR3DL2 polypeptide competes with antibody 12B1 1 or 19H12 for binding to a human KIR3DL2 polypeptide.
24. A method for determining whether a KIR3DL2 polypeptide is expressed on the surface of a cell from an individual having celiac disease (or for determining whether a patient having celiac disease has cells that express KIR3DL2), the method comprising obtaining from an individual having celiac disease a biological sample that comprises cells, bringing said cells into contact with an antibody that competes with antibody 19H12 for binding to a human KIR3DL2 polypeptide, and detecting cells that express KIR3DL2 using flow cytometry.
25. A method for determining whether a KIR3DL2 polypeptide is expressed on the surface of a cell from an individual having celiac disease (or for determining whether a patient having celiac disease has cells that express KIR3DL2), the method comprising obtaining from an individual having celiac disease a biological sample that comprises cells, preparing a tissue section from such cells, bringing said tissue section into contact with an antibody that competes with antibody 12B1 1 for binding to a human KIR3DL2 polypeptide, and detecting expression of KIR3DL2.
26. The method of claims 22-25, wherein the cells are abnormal lELs.
PCT/EP2015/069704 2014-08-27 2015-08-27 Treatment of celiac disease WO2016030488A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201462042486P 2014-08-27 2014-08-27
US62/042,486 2014-08-27

Publications (1)

Publication Number Publication Date
WO2016030488A1 true WO2016030488A1 (en) 2016-03-03

Family

ID=54014814

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2015/069704 WO2016030488A1 (en) 2014-08-27 2015-08-27 Treatment of celiac disease

Country Status (1)

Country Link
WO (1) WO2016030488A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018041989A1 (en) * 2016-09-02 2018-03-08 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for diagnosing and treating refractory celiac disease type 2
WO2018073363A1 (en) * 2016-10-21 2018-04-26 Innate Pharma Treatment with anti-kir3dl2 agents

Citations (40)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US5229275A (en) 1990-04-26 1993-07-20 Akzo N.V. In-vitro method for producing antigen-specific human monoclonal antibodies
US5567610A (en) 1986-09-04 1996-10-22 Bioinvent International Ab Method of producing human monoclonal antibodies and kit therefor
US5660827A (en) 1992-03-05 1997-08-26 Board Of Regents, The University Of Texas System Antibodies that bind to endoglin
WO1999054342A1 (en) 1998-04-20 1999-10-28 Pablo Umana Glycosylation engineering of antibodies for improving antibody-dependent cellular cytotoxicity
US6077499A (en) 1996-05-03 2000-06-20 Immunomedics, Inc. Targeted combination immunotherapy of cancer
WO2000042072A2 (en) 1999-01-15 2000-07-20 Genentech, Inc. Polypeptide variants with altered effector function
US6162963A (en) 1990-01-12 2000-12-19 Abgenix, Inc. Generation of Xenogenetic antibodies
EP1176195A1 (en) 1999-04-09 2002-01-30 Kyowa Hakko Kogyo Co., Ltd. Method for controlling the activity of immunologically functional molecule
WO2003035835A2 (en) 2001-10-25 2003-05-01 Genentech, Inc. Glycoprotein compositions
US6737056B1 (en) 1999-01-15 2004-05-18 Genentech, Inc. Polypeptide variants with altered effector function
US20040110704A1 (en) 2002-04-09 2004-06-10 Kyowa Hakko Kogyo Co., Ltd. Cells of which genome is modified
WO2004063351A2 (en) 2003-01-09 2004-07-29 Macrogenics, Inc. IDENTIFICATION AND ENGINEERING OF ANTIBODIES WITH VARIANT Fc REGIONS AND METHODS OF USING SAME
WO2004099249A2 (en) 2003-05-02 2004-11-18 Xencor, Inc. Optimized fc variants and methods for their generation
US6821505B2 (en) 1997-03-03 2004-11-23 Board Of Regents, The University Of Texas System Immunoglobin-like domains with increased half lives
WO2004103269A2 (en) 2002-10-18 2004-12-02 Macrogenics, Inc. Methods and compositions for vaccination comprising nucleic acid and/or polypeptide sequences of the genus borrelia
WO2005047327A2 (en) 2003-11-12 2005-05-26 Biogen Idec Ma Inc. NEONATAL Fc RECEPTOR (FcRn)-BINDING POLYPEPTIDE VARIANTS, DIMERIC Fc BINDING PROTEINS AND METHODS RELATED THERETO
WO2005110474A2 (en) 2004-05-10 2005-11-24 Macrogenics, Inc. HUMANIZED FcϜRIIB SPECIFIC ANTIBODIES AND METHODS OF USE THEREOF
WO2005115452A2 (en) 2004-04-16 2005-12-08 Macrogenics, Inc. Fcϝriib-specific antibodies and methods of use thereof
WO2006088494A2 (en) 2004-07-12 2006-08-24 Macrogenics, Inc. Identification and engineering of antibodies with variant fc regions and methods of using the same
WO2006133148A2 (en) 2005-06-03 2006-12-14 Genentech, Inc. Method of producing antibodies with modified fucosylation level
WO2007021841A2 (en) 2005-08-10 2007-02-22 Macrogenics, Inc. Identification and engineering of antibodies with variant fc regions and methods of using same
US7183387B1 (en) 1999-01-15 2007-02-27 Genentech, Inc. Polypeptide variants with altered effector function
WO2007024249A2 (en) 2004-11-10 2007-03-01 Macrogenics, Inc. Engineering fc antibody regions to confer effector function
WO2007080277A1 (en) 2005-12-16 2007-07-19 Lfb Biotechnologies Method for preparing antibodies selective for activating fc receptors
WO2007084926A2 (en) 2006-01-17 2007-07-26 Biolex Therapeutics, Inc. Compositions and methods for humanization and optimization of n-glycans in plants
WO2007106707A2 (en) 2006-03-10 2007-09-20 Macrogenics, Inc. Identification and engineering of antibodies with variant heavy chains and methods of using same
WO2008002933A2 (en) 2006-06-26 2008-01-03 Macrogenics, Inc. Combination of fcgammariib antibodies and cd20-specific antibodies and methods of use thereof
WO2008006554A2 (en) 2006-07-11 2008-01-17 Greenovation Biotech Gmbh Glyco-engineered antibodies
WO2008105886A2 (en) 2006-05-26 2008-09-04 Macrogenics, Inc. HUMANIZED FCγRIIB-SPECIFIC ANTIBODIES AND METHODS OF USE THEREOF
US7425619B2 (en) 2002-08-14 2008-09-16 Macrogenics, Inc. FcγRIIB specific antibodies and methods of use thereof
WO2008142124A1 (en) 2007-05-21 2008-11-27 Vivalis Recombinant protein production in avian ebx® cells
WO2010081890A1 (en) 2009-01-19 2010-07-22 Innate Pharma Anti-kir3d antibodies
WO2011109400A2 (en) 2010-03-04 2011-09-09 Macrogenics,Inc. Antibodies reactive with b7-h3, immunologically active fragments thereof and uses thereof
WO2012160448A2 (en) * 2011-05-25 2012-11-29 Innate Pharma, S.A. Anti-kir antibodies for the treatment of inflammatory disorders
WO2014044681A1 (en) * 2012-09-19 2014-03-27 Innate Pharma Kir3dl2 binding agents
WO2014044686A1 (en) * 2012-09-19 2014-03-27 Innate Pharma Kir3dl2 binding agents
WO2014125041A1 (en) * 2013-02-14 2014-08-21 Innate Pharma Treatment of peripheral t cell lymphoma
WO2014128221A1 (en) * 2013-02-20 2014-08-28 Innate Pharma A compound that specifically binds to kir3dl2 for use in the treatment of peripheral t cell lymphoma
WO2014191936A1 (en) * 2013-05-29 2014-12-04 Institut National De La Sante Et De La Recherche Medicale Kir3dl2 is a biomarker and a therapeutic target useful for respectively preventing and treating a subset of cutaneous and non-cutaneous peripheral t-cell lymphomas

Patent Citations (48)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US5567610A (en) 1986-09-04 1996-10-22 Bioinvent International Ab Method of producing human monoclonal antibodies and kit therefor
US6162963A (en) 1990-01-12 2000-12-19 Abgenix, Inc. Generation of Xenogenetic antibodies
US5229275A (en) 1990-04-26 1993-07-20 Akzo N.V. In-vitro method for producing antigen-specific human monoclonal antibodies
US5660827A (en) 1992-03-05 1997-08-26 Board Of Regents, The University Of Texas System Antibodies that bind to endoglin
US6077499A (en) 1996-05-03 2000-06-20 Immunomedics, Inc. Targeted combination immunotherapy of cancer
US6821505B2 (en) 1997-03-03 2004-11-23 Board Of Regents, The University Of Texas System Immunoglobin-like domains with increased half lives
WO1999054342A1 (en) 1998-04-20 1999-10-28 Pablo Umana Glycosylation engineering of antibodies for improving antibody-dependent cellular cytotoxicity
US6737056B1 (en) 1999-01-15 2004-05-18 Genentech, Inc. Polypeptide variants with altered effector function
US7183387B1 (en) 1999-01-15 2007-02-27 Genentech, Inc. Polypeptide variants with altered effector function
WO2000042072A2 (en) 1999-01-15 2000-07-20 Genentech, Inc. Polypeptide variants with altered effector function
US7416727B2 (en) 1999-01-15 2008-08-26 Genentech, Inc. Polypeptide variants with altered effector function
US7371826B2 (en) 1999-01-15 2008-05-13 Genentech, Inc. Polypeptide variants with altered effector function
US7335742B2 (en) 1999-01-15 2008-02-26 Genentech, Inc. Polypeptide variants with altered effector function
US7332581B2 (en) 1999-01-15 2008-02-19 Genentech, Inc. Polypeptide variants with altered effector function
US7122637B2 (en) 1999-01-15 2006-10-17 Genentech, Inc. Polypeptide variants with altered effector function
EP1176195A1 (en) 1999-04-09 2002-01-30 Kyowa Hakko Kogyo Co., Ltd. Method for controlling the activity of immunologically functional molecule
WO2003035835A2 (en) 2001-10-25 2003-05-01 Genentech, Inc. Glycoprotein compositions
US20040110704A1 (en) 2002-04-09 2004-06-10 Kyowa Hakko Kogyo Co., Ltd. Cells of which genome is modified
US7425619B2 (en) 2002-08-14 2008-09-16 Macrogenics, Inc. FcγRIIB specific antibodies and methods of use thereof
WO2004103269A2 (en) 2002-10-18 2004-12-02 Macrogenics, Inc. Methods and compositions for vaccination comprising nucleic acid and/or polypeptide sequences of the genus borrelia
WO2004063351A2 (en) 2003-01-09 2004-07-29 Macrogenics, Inc. IDENTIFICATION AND ENGINEERING OF ANTIBODIES WITH VARIANT Fc REGIONS AND METHODS OF USING SAME
US7355008B2 (en) 2003-01-09 2008-04-08 Macrogenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
WO2004099249A2 (en) 2003-05-02 2004-11-18 Xencor, Inc. Optimized fc variants and methods for their generation
WO2005047327A2 (en) 2003-11-12 2005-05-26 Biogen Idec Ma Inc. NEONATAL Fc RECEPTOR (FcRn)-BINDING POLYPEPTIDE VARIANTS, DIMERIC Fc BINDING PROTEINS AND METHODS RELATED THERETO
WO2005115452A2 (en) 2004-04-16 2005-12-08 Macrogenics, Inc. Fcϝriib-specific antibodies and methods of use thereof
WO2005110474A2 (en) 2004-05-10 2005-11-24 Macrogenics, Inc. HUMANIZED FcϜRIIB SPECIFIC ANTIBODIES AND METHODS OF USE THEREOF
US7521542B2 (en) 2004-05-10 2009-04-21 Macrogenics, Inc. Humanized FcγRIIB-specific antibodies and methods of use thereof
WO2006088494A2 (en) 2004-07-12 2006-08-24 Macrogenics, Inc. Identification and engineering of antibodies with variant fc regions and methods of using the same
WO2007024249A2 (en) 2004-11-10 2007-03-01 Macrogenics, Inc. Engineering fc antibody regions to confer effector function
US7632497B2 (en) 2004-11-10 2009-12-15 Macrogenics, Inc. Engineering Fc Antibody regions to confer effector function
WO2006133148A2 (en) 2005-06-03 2006-12-14 Genentech, Inc. Method of producing antibodies with modified fucosylation level
WO2007021841A2 (en) 2005-08-10 2007-02-22 Macrogenics, Inc. Identification and engineering of antibodies with variant fc regions and methods of using same
WO2007080277A1 (en) 2005-12-16 2007-07-19 Lfb Biotechnologies Method for preparing antibodies selective for activating fc receptors
WO2007084926A2 (en) 2006-01-17 2007-07-26 Biolex Therapeutics, Inc. Compositions and methods for humanization and optimization of n-glycans in plants
WO2007106707A2 (en) 2006-03-10 2007-09-20 Macrogenics, Inc. Identification and engineering of antibodies with variant heavy chains and methods of using same
WO2008105886A2 (en) 2006-05-26 2008-09-04 Macrogenics, Inc. HUMANIZED FCγRIIB-SPECIFIC ANTIBODIES AND METHODS OF USE THEREOF
WO2008002933A2 (en) 2006-06-26 2008-01-03 Macrogenics, Inc. Combination of fcgammariib antibodies and cd20-specific antibodies and methods of use thereof
WO2008006554A2 (en) 2006-07-11 2008-01-17 Greenovation Biotech Gmbh Glyco-engineered antibodies
WO2008142124A1 (en) 2007-05-21 2008-11-27 Vivalis Recombinant protein production in avian ebx® cells
WO2010081890A1 (en) 2009-01-19 2010-07-22 Innate Pharma Anti-kir3d antibodies
WO2011109400A2 (en) 2010-03-04 2011-09-09 Macrogenics,Inc. Antibodies reactive with b7-h3, immunologically active fragments thereof and uses thereof
WO2012160448A2 (en) * 2011-05-25 2012-11-29 Innate Pharma, S.A. Anti-kir antibodies for the treatment of inflammatory disorders
WO2014044681A1 (en) * 2012-09-19 2014-03-27 Innate Pharma Kir3dl2 binding agents
WO2014044686A1 (en) * 2012-09-19 2014-03-27 Innate Pharma Kir3dl2 binding agents
WO2014125041A1 (en) * 2013-02-14 2014-08-21 Innate Pharma Treatment of peripheral t cell lymphoma
WO2014128221A1 (en) * 2013-02-20 2014-08-28 Innate Pharma A compound that specifically binds to kir3dl2 for use in the treatment of peripheral t cell lymphoma
WO2014191936A1 (en) * 2013-05-29 2014-12-04 Institut National De La Sante Et De La Recherche Medicale Kir3dl2 is a biomarker and a therapeutic target useful for respectively preventing and treating a subset of cutaneous and non-cutaneous peripheral t-cell lymphomas

Non-Patent Citations (69)

* Cited by examiner, † Cited by third party
Title
"Antibody Purification Handbook", BIOSCIENCES
"Biocomputing: Informatics and Genome Projects", 1993, ACADEMIC PRESS
"Computational Molecular Biology", 1988, OXFORD UNIVERSITY PRESS
"Computer Analysis of Sequence Data", 1994, HUMANA PRESS
"Current Protocols in Immunology", 1992, GREENE PUBLISHING ASSOC. AND WILEY INTERSCIENCE
"Remington's Pharmaceutical Sciences", 1995, MACK PUBLISHING CO
"Sequence Analysis Primer", 1991, M. STOCKTON PRESS
"World Gastroenterology Organization Practice Guidelines", CELIAC DISEASE: WORLD GASTROENTEROLOGY ORGANIZATION, 2007
ALTSCHUL ET AL., J. MOL. BIOL., vol. 215, 1990, pages 403 - 410
CARILLO ET AL., SIAM J. APPLIED MATH., vol. 48, 1988, pages 1073
CARTER ET AL., PNAS, vol. 89, 1992, pages 4285
CHOTHIA; LESK, J. MOL., vol. 196, 1987, pages 901
CHU; ROBINSON, CURRENT OPINION BIOTECHNOL., vol. 12, 2001, pages 180 - 7
CLACKSON; WELLS, SCIENCE, vol. 267, 1995, pages 383 - 386
COLONNA; SAMARIDIS, SCIENCE, vol. 268, no. 5209, 1995, pages 405 - 408
DEVEREUX ET AL., NUCL. ACID. RES, vol. 12, 1984, pages 387
DI SABATINO ANTONIO ET AL: "How I treat enteropathy-associated T-cell lymphoma", BLOOD, vol. 119, no. 11, March 2012 (2012-03-01), pages 2458 - 2468, XP002746514, DOI: 10.1182/BLOOD-2011-10-385559 *
DOWNWARD, J MASS SPECTROM, vol. 35, no. 4, April 2000 (2000-04-01), pages 493 - 503
E. HARLOW; D. LANE: "Antibodies: A Laboratory Manual.", 1988, COLD SPRING HARBOR LABORATORY PRESS
EHRING H, ANALYTICAL BIOCHEMISTRY, vol. 267, no. 2, 1999, pages 252 - 259
ENGEN, J. R; SMITH, D. L, ANAL. CHEM., vol. 73, 2001, pages 256A - 265A
ERNST, SCHERING RES FOUND WORKSHOP, 2004, pages 149 - 67
FAGERSTAM ET AL., JOURNAL OF MOLECULAR RECOGNITION, vol. 3, 1990, pages 208 - 14
GODING: "Monoclonal Antibodies: Principles and Practice", 1986, ACADEMIC PRESS, pages: 59 - 103
GOLDENBERG, CANCER JOURNAL FOR CLINICIANS, vol. 44, 1994, pages 43
GOODMAN; GILMAN'S: "The Pharmacological Basis of Therapeutics", 2001, MCGRAW HILL
GREEN ET AL., NATURE GENET, vol. 7, 1994, pages 13
HARLOW ET AL.: "Antibodies: A Laboratory Manual", 1988, COLD SPRING HARBOR LABORATORY PRESS
HUANG, JOURNAL OF MOLECULAR BIOLOGY, vol. 281, no. 1, 1998, pages 61 - 67
JAKOBOVITZ ET AL., NATURE, vol. 362, 1993, pages 255
JONES ET AL., NATURE, vol. 321, 1986, pages 522
KISELAR; DOWNARD, ANAL CHEM., vol. 71, no. 9, 1 May 1999 (1999-05-01), pages 1792 - 801
KROGER ET AL., BIOSENSORS AND BIOELECTRONICS, vol. 17, 2002, pages 937 - 944
LEIPERT ET AL., ANGEW. CHEM. INT. ED, vol. 37, 1998, pages 3308 - 3311
LONBERG ET AL., NATURE, vol. 368, 1994, pages 856
MANCA, ANN 1ST SUPER SANITA., vol. 27, 1991, pages 15 - 9
MCCAFFERTY ET AL., NATURE, vol. 348, 1990, pages 552 - 553
MORRISON ET AL., PNAS, 1984, pages 6851
MULLER, METH. ENZYMOL., vol. 92, 1983, pages 589 - 601
NICE ET AL., J. CHROMA-TOGR., vol. 646, 1993, pages 159 - 168
PASTAN ET AL., CELL, vol. 47, 1986, pages 641
PENDE ET AL., J. EXP. MED., vol. 184, 1996, pages 505 - 518
PLUCKTHUN, IMMUNOL, vol. 130, 1992, pages 151
PLUGIS NICHOLAS M ET AL: "Therapeutic approaches for celiac disease.", BEST PRACTICE & RESEARCH. CLINICAL GASTROENTEROLOGY JUN 2015, vol. 29, no. 3, June 2015 (2015-06-01), pages 503 - 521, XP002746515, ISSN: 1532-1916 *
PRESTA ET AL., J. IMMUNOL., vol. 151, 1993, pages 2623
PRESTA, CURR. OP. STRUCT. BIOL., vol. 2, 1992, pages 593
PRESTA, L.G. ET AL., BIOCHEM. SOC. TRANS., vol. 30, no. 4, 2002, pages 487 - 490
REICHMANN ET AL., NATURE, vol. 332, 1988, pages 323
RUBIO-TAPIA ET AL., AM. J. GASTROENTEROLOGY, vol. 108, no. 5, 2013, pages 656 - 677
SAITO; PATTERSON, METHODS, vol. 9, no. 3, June 1996 (1996-06-01), pages 516 - 24
SAUNAL, J. IMMUNOL. METHODS, vol. 183, 1995, pages 33 - 41
SHIELDS, R.L ET AL., J. BIOL. CHEM., vol. 277, 2002, pages 26733 - 26740
SHIELDS, R.L. ET AL., J. BIOL. CHEM., vol. 276, no. 9, 2001, pages 6591 - 6604
SHIELDS, R.L. ET AL., J. BIOL. CHEM., vol. 277, no. 30, 26 December 2001 (2001-12-26), pages 26733 - 26740
SHIELDS, RX ET AL., J. BIOL. CHEM., vol. 277, 2002, pages 26733 - 26740
SIMS ET AL., J. IMMUNOL., vol. 151, 1993, pages 2296
SKERRA ET AL., CURR. OPINION IN IMMUNOL., vol. 5, 1993, pages 256
TARENTINO ET AL., BIOCHEM., vol. 14, 1975, pages 5516 - 5523
TAYLOR ET AL., INT IMMUN, vol. 6, 1994, pages 579
UMANA ET AL., NAT. BIOTECH., vol. 17, 1999, pages 176 - 1
UMANA ET AL., NAT. BIOTECH., vol. 17, 1999, pages 176 - 180
VERHOEYEN, SCIENCE, vol. 239, pages 1534
VON HEINJE, G.: "Sequence Analysis in Molecular Biology", 1987, ACADEMIC PRESS
WANG ET AL., NATURE, vol. 355, 1992, pages 275 - 278
WARD ET AL., NATURE, vol. 341, 1989, pages 544
WELLS, PROC NATL ACAD SCI USA, vol. 93, 1996, pages 1 - 6
WRIGHT; MORRISON, TREND BIOTECHNOL., vol. 15, 1997, pages 26 - 31
YAMANE-OHNUKI ET AL., BIOTECHNOL BIOENG, vol. 87, 2004, pages 614 - 22
ZHOU ET AL., BIOTECH. AND BIOENGIN, vol. 99, 2008, pages 652 - 665

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018041989A1 (en) * 2016-09-02 2018-03-08 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for diagnosing and treating refractory celiac disease type 2
WO2018073363A1 (en) * 2016-10-21 2018-04-26 Innate Pharma Treatment with anti-kir3dl2 agents
JP2020500161A (en) * 2016-10-21 2020-01-09 イナート・ファルマ・ソシエテ・アノニムInnate Pharma Pharma S.A. Treatment with anti-KIR3DL2 agent

Similar Documents

Publication Publication Date Title
US20210363248A1 (en) Compound that specifically binds to kir3dl2 for use in the treatment of peripheral t cell lymphoma
US11858990B2 (en) KIR3DL2 binding agents
US20190315857A1 (en) Treatment of peripheral t cell lymphoma
US20210087270A1 (en) Treatment with anti-kir3dl2 agents
WO2016030488A1 (en) Treatment of celiac disease
US20240124579A1 (en) Kir3dl2 binding agents

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: 15756899

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: 15756899

Country of ref document: EP

Kind code of ref document: A1