EP1096949A2 - Traitement de la maladie coeliaque a l'aide d'antagonistes de l'interleukine-15 - Google Patents

Traitement de la maladie coeliaque a l'aide d'antagonistes de l'interleukine-15

Info

Publication number
EP1096949A2
EP1096949A2 EP99933001A EP99933001A EP1096949A2 EP 1096949 A2 EP1096949 A2 EP 1096949A2 EP 99933001 A EP99933001 A EP 99933001A EP 99933001 A EP99933001 A EP 99933001A EP 1096949 A2 EP1096949 A2 EP 1096949A2
Authority
EP
European Patent Office
Prior art keywords
antagonist
cells
expression
fas
gliadin
Prior art date
Legal status (The legal status 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 status listed.)
Withdrawn
Application number
EP99933001A
Other languages
German (de)
English (en)
Inventor
Marco Londei
Sonia Quaratino
Luigi Maiuri
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
MATHILDA AND TERENCE KENNEDY INSTITUTE OF
Kennedy Trust for Rheumatology Research
Original Assignee
MATHILDA AND TERENCE KENNEDY INSTITUTE OF
Kennedy Institute of Rheumotology
Mathilda and Terence Kennedy Institute of Rheumatology
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 MATHILDA AND TERENCE KENNEDY INSTITUTE OF, Kennedy Institute of Rheumotology, Mathilda and Terence Kennedy Institute of Rheumatology filed Critical MATHILDA AND TERENCE KENNEDY INSTITUTE OF
Publication of EP1096949A2 publication Critical patent/EP1096949A2/fr
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
    • C07K16/244Interleukins [IL]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/20Interleukins [IL]
    • A61K38/2086IL-13 to IL-16
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • C07K14/5443IL-15
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0031Rectum, anus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding

Definitions

  • the invention relates to celiac disease, and in particular to the treatment of celiac disease.
  • CD Celiac disease
  • anti-endomysium antibodies 3 4.
  • the latter have been shown to be a specific marker for
  • lymphocytes (IEL) have been considered as the "key players" in this disease 1.
  • Interleukin 15 has recently gained a
  • IL-15 can induce mucosal T lymphocyte migration in celiac as well as
  • IL-15 has been reported to modulate the function of intestinal epithelial cells 12, the
  • a first aspect of the invention provides a method of treating an inflammatory
  • bowel disease such as celiac disease
  • a further aspect of the invention provides the use of an antagonist of IL-15 to treat an inflammatory bowel disease, such as celiac disease.
  • a still further aspect of the invention provides an antagonist of IL-15 for use in the
  • the antagonist of IL-15 activity interferes with the signal transduction of IL-15
  • IL-15 antagonists used in the invention are IL-15 antagonists used in the invention.
  • IL-15 preferably selected from the group consisting of (a) a mutein of mature, or native, IL-15
  • Antagonists for use in the invention also serve as antagonists for use in the invention.
  • IL-15 include monoclonal antibodies against IL-15.
  • the antagonist used is selected from mature, or native, simian IL-15 molecules
  • Such IL-15 muteins are capable of binding to the
  • IL-15R ⁇ subunit and are incapable of transducing a signal through the ⁇ or 7-subunits of
  • the antagonist is supplied in a pharmaceutically effective amount. That is, an
  • substitutions or deletions of amino acid residues Preferably, substitution and deletion
  • IL-15 binding of IL-15 to either or both of the ⁇ or 7-subunits of the IL-15 receptor complex.
  • Recombinant production of an IL-15 mutein first requires isolation of a DNA clone (i.e.,
  • cDNA that encodes an IL- 15 mutein.
  • cDNA clones are derived from primary cells or cell
  • cDNA library is made from the mRNA by reverse transcription.
  • a cDNA clone may be isolated and identified using the DNA sequence information provided herein to design a
  • SEQ ID NO:3 may have the sequence of nucleic acids 1-489 of SEQ ID NO:3 and SEQ ID NO:4.
  • the isolated cDNA is preferably in the form of an open reading frame uninterrupted by
  • Genomic DNA containing the relevant sequences containing the relevant sequences, or introns.
  • nucleotide sequences that encode mammalian IL-15 polypeptides can also be used as a
  • cDNA can be mutated utilising techniques known in the art to provide IL-15 antagonist
  • N-glycosylation sites in IL-15 are encompassed by the invention.
  • N-glycosylation sites in IL-15 are encompassed by the invention.
  • polypeptides are characterised by an amino acid triplet Asn-X-Y, wherein X is any amino
  • the simian IL-15 protein comprises two such triplets,
  • Recombinant expression vectors include synthetic or cDNA-derived DNA fragments
  • the DNA encoding an IL-15 mutein is operably linked to a
  • suitable transcriptional or translational regulatory or structural nucleotide sequence such as
  • regulatory elements as one derived from mammalian, microbial, viral or insect genes.
  • regulatory elements as one derived from mammalian, microbial, viral or insect genes.
  • sequences include, for example, a genetic sequence having a regulatory role in gene
  • expression e.g.. transcriptional promoters or enhancers
  • expression e.g. transcriptional promoters or enhancers
  • control transcription a sequence encoding suitable mRNA ribosomal binding sites
  • Nucleotide sequences are operably linked when the regulatory sequence functionally
  • a DNA sequence for a signal peptide secretory
  • leader may be operably linked to a structural gene DNA sequence for an IL- 15 mutein if
  • the signal peptide is expressed as part of a precursor amino acid sequence and participates
  • a promoter nucleotide sequence is operably
  • promoter nucleotide e.g., structural gene DNA
  • a ribosome binding site may be operably linked to a structural gene nucleotide
  • coding sequence e.g. IL-15 mutein
  • Suitable host cells for expression of an IL-15 mutein include prokaryotes, yeast or higher
  • Prokaryotes include gram negative or gram positive organisms, for example E. coli or bacilli. Suitable prokaryotic
  • hosts cells for transformation include, for example, E. coli. Bacillus subtilis. Salmonella
  • yeast such as S. cerevisiae
  • mammalian cell line such as Chinese Hamster
  • nucleotide sequence e.g., IL-15 mutein
  • the structural gene that encodes an IL- 15 mutein may include a leader sequence.
  • the leader may include a leader sequence.
  • sequence may enable improved extracellular secretion of translated polypeptide by a yeast
  • IL-15 muteins may be expressed in yeast host cells, preferably from the Saccharomyces
  • yeast e.g.. S. cerevisiae
  • Other genera of yeast such as Pichia or Kluyveromyces, may
  • yeast vectors will often contain an origin of replication sequence from
  • a 2m yeast plasmid a 2m yeast plasmid. an autonomously replicating sequence (ARS), a promoter region,
  • ARS autonomously replicating sequence
  • sequences for polyadenylation and sequences for transcription termination.
  • sequences for polyadenylation and sequences for transcription termination.
  • yeast vectors include an origin of replication sequence and selectable marker. Suitable
  • promoter sequences for yeast vectors include promoters for metallothionein,
  • hexokinase pyruvate decarboxylase, phosphofructokinase. glucose-6-phosphate
  • isomerase 3-phosphoglycerate mutase, pyruvate kinase, triosephosphate isomerase,
  • phosphoglucose isomerase phosphoglucose isomerase
  • glucokinase phosphoglucokinase
  • Yeast vectors can be assembled, for example, using DNA sequences from pBR322 for
  • DNA sequences that can be included in a yeast expression construct include a
  • the ADH2 promoter glucose-repressible ADH2 promoter and a-factor secretion leader.
  • yeast ⁇ -factor leader sequence directs secretion of
  • heterologous polypeptides The a-factor leader sequence is often inserted between the amino acids that are amino acids that are amino acids that are amino acids that are amino acids that are amino acids that are amino acids that are amino acids that are amino acids that are amino acids that are amino acids that are amino acids that are amino acids that are amino acids that are amino acids that are amino acids that are amino acids that are amino acids that are amino acids that are amino acids that are amino acids that are amino acids that are amino acids that are amino acids.
  • a leader sequence may
  • Yeast transformation protocols are known to those skilled in the art.
  • One such protocol is
  • yeast host cells transformed by vectors containing ADH2 promoter sequence may be
  • a rich medium is one
  • the IL- 15 mutein may include an
  • N-terminal methionine residue to facilitate expression of the recombinant polypeptide in a
  • the N-terminal Met may be cleaved from the expressed
  • nucleotide sequence are transfected or transformed into a suitable host microorganism or
  • Expression vectors transfected into prokaryotic host cells generally comprise one or more
  • a phenotypic selectable marker is, for example, a gene
  • prokaryotic host cells include a
  • selectable marker can comprise genetic elements of the cloning vector pBR322 (ATCC
  • pBR322 contains genes for ampicillin and tetracycline resistance and thus provides simple means for identifying transformed cells.
  • Promoter sequences are commonly used for recombinant prokaryotic host cell expression
  • Common promoter sequences include Mactamase (penicillinase), lactose
  • tryptophan (tip) promoter system (Goeddel et al., Nucl. Acids Res. 8:4057, 1980;
  • cell expression system employs a phage 1 PL promoter and a cI857ts thermolabile
  • Plasmid vectors available from the American Type Culture Collection are available from the American Type Culture Collection.
  • Mammalian or insect host cell culture systems also could be employed to express
  • Suitable mammalian host cell lines include the
  • COS-7 lines of monkey kidney cells (Gluzman et al, Cell 23:175, (1981); ATCC CRL
  • L cells C127 cells, 3T3 cells (ATCC CCL 163), CHO cells, HeLa cells (ATCC
  • nontranscribed elements such as an origin of replication, a promoter sequence, an
  • enhancer linked to the structural gene other 5' or 3' flanking nontranscribed sequences, such as ribosome binding sites, a polyadenylation site, splice donor and acceptor sites, and transcriptional termination sequences.
  • vectors may be provided by viral sources.
  • viral sources for example, commonly used mammalian cell
  • promoter sequences and enhancer sequences are derived from Polyoma.
  • Adenovirus 2 Adenovirus 3
  • Simian Virus 40 (SV40), and human cytomegalovirus. DNA sequences derived from the
  • SV40 viral genome for example. SV40 origin, early and late promoter, enhancer, splice,
  • polyadenylation sites may be used to provide the other genetic elements required for
  • promoters are particularly useful because both are easily obtained from a viral genome as a
  • Exemplar ⁇ ' mammalian expression vectors can be constructed as disclosed by Okayama
  • IL- 15 mutein polypeptides may be prepared by culturing transformed host cells
  • IL-15 mutein polypeptides under culture conditions necessary to express IL-15 mutein polypeptides.
  • the resulting expressed mutein may then be purified from culture media or cell extracts.
  • mutein may be concentrated using a commercially available protein concentration filter
  • the culture media can be applied to a purification matrix such as a
  • Phenyl Sepharose is the
  • an anion exchange resin can be employed, for example,
  • the matrices can be any suitable matrix or substrate having pendant diethylaminoethyl (DEAE) groups.
  • the matrices can be any suitable matrices.
  • gel filtration medium can be used.
  • hydrophobic RP-HPLC media e.g., silica gel having pendant butyl or
  • Recombinant protein produced in bacterial culture is usually isolated by initial disruption
  • Microbial cells can be disrupted by any convenient method, including freeze-thaw cycling, sonication. mechanical disruption, or
  • Transformed yeast host cells are preferably employed to express an IL-15 mutein as a
  • a mutein of IL-15 is used wherein at least one of the amino acid residues
  • residues 49-162 shown in SEQ ID NO:2) is deleted or substituted with a different
  • Asp56 can be deleted while Glnl56 is substituted with any
  • Asp56 can be substituted with any amino acid while
  • Gin 156 is deleted. Generally, substitution muteins are preferred, and more preferred are
  • muteins preferably include those wherein Asp56 is substituted by serine or cysteine; or
  • Gin 156 is substituted by serine or cysteine. or wherein both Asp56 and Gin 156
  • deletion muteins are each substituted with a serine or cysteine.
  • deletion muteins include those
  • the invention further encompasses muteins wherein amino
  • mature IL-15 polypeptides disclosed herein comprising the
  • muteins. may be modified by forming covalent or aggregative conjugates with other
  • Such moieties can include PEG, mPEG, dextran. PVP, PVA,
  • polyamino acids such as poly-L-lysine or polyhistidine, albumin and gelatin at specific
  • the IL-15 receptor complex while maintaining the high affinity of IL-15 for the IL-15R ⁇ .
  • IL-15 can be specifically glycosylated at sites that can interfere with binding
  • PEG PEG
  • dextran PEG
  • PVP Most preferred for use in the invention is PEG, wherein the molecular weight of the
  • PEG is preferably between about 1,000 to about 20,000. A molecular weight of about
  • PEG can be used in the form of succinimidyl succinate PEG (SS-PEG)
  • succinimidyl carbonate PEG which provides a urethane linkage and is stable
  • succinimidyl propionate PEG (SPA-PEG) provides an
  • SC-PEG and VS-PEG are preferred, and SC-PEG is most preferred due to its
  • the PEG moieties can be bonded to IL- 15 in strategic sites to take advantage of PEGOs
  • PEG moieties can be bonded to IL-15 by
  • PEGylation One method of site specific PEGylation is through methods of protein
  • the large molecular size of the PEG chain(s) conjugated to IL-15 is
  • PEGylation is carried out at either (1) about pH 9.0 and at molar ratios of SC-PEG to
  • lysine residue of approximately 1 :1 to 100:1, or greater; or (2) at about pH 7.0 and at
  • the extent of modification and heterogeneity of PEGylated IL- 15 can be determined using
  • MALDI matrix assisted laser desorption ionization mass spectrometry
  • human IL-15 has a molecular weight of about 13.000 and by using PEG having a
  • an antagonist according to the invention can take the form of a monoclonal
  • IL-15 7-subunits of the IL-15 receptor complex.
  • IL-15 7-subunits of the IL-15 receptor complex.
  • IL-15 peptides can be used to prepare antibodies that specifically bind to IL-15.
  • antibodies should be understood to include polyclonal antibodies, monoclonal antibodies, fragments thereof such as F(ab')2 and Fab
  • monoclonal antibody or binding partner may be readily determined by one of ordinary skill
  • monoclonal antibodies against IL-15 can be generated using the following
  • IL-15 can be used to generate monoclonal antibodies against IL-15 using techniques
  • mice are immunised with IL-15 as an immunogen emulsified in complete Freund's
  • RIBI adjuvant or RIBI adjuvant (RIBI Corp.. Hamilton. Montana), and injected in amounts
  • mice are periodically boosted thereafter on a weekly to bi-weekly immunisation
  • Serum samples are periodically taken by retro-orbital bleeding or tail-tip
  • spleen cells are sacrificed, spleen cells harvested, and spleen cells are fused to a murine myeloma cell
  • hybridoma cells which are plated in multiple microtiter plates in a HAT (hypoxanthine, aminopterin and thymidine) selective medium to inhibit proliferation of non-fused
  • HAT hypoxanthine, aminopterin and thymidine
  • myeloma cells and myeloma hybrids are myeloma cells and myeloma hybrids.
  • the hybridoma cells are screened by ELISA for reactivity against purified IL-15 by
  • a preferred screening technique is the antibody capture technique
  • hybridoma cells can be
  • antibody to protein A or protein G can also be used, as can affinity chromatography
  • microbodies can be found in WO 94/09817; and procedures to generate transgenic
  • a CTLL-2 proliferation assay is preferred for this purpose. See. Gillis and
  • the IL-15 antagonists are formulated according to known methods used to formulate the IL-15 antagonists.
  • IL-15 antagonists are formulated according to known methods used to formulate the IL-15 antagonists.
  • An IL-15 antagonist can be combined in
  • diluents e.g., Tris-HCl, acetate, phosphate
  • preservatives e.g., sodium bicarbonate
  • compositions can contain
  • polymeric compounds such as polyacetic acid, polyglycolic acid,
  • compositions will be described in detail below.
  • An IL-15 antagonist can also be conjugated to antibodies
  • tissue-specific receptors against tissue-specific receptors, ligands or antigens, or coupled to ligands of tissue-specific receptors.
  • the IL- 15 antagonist of the invention can be administered topically, orally, parenterally,
  • parenteral includes subcutaneous injections
  • compositions will typically contain an effective amount of an IL-15 antagonist, alone or in combination with an effective amount of any other active material. Such dosages and
  • compositions may vary depending upon many variables
  • Preliminary doses can be determined according to animal tests, and the
  • Treated CD intestine A: Treated CD intestine:
  • IE intraepithelial
  • intraepithelial lymphocytes are found within the surface epithelium after incubation with
  • FAS expression by enterocytes is detected after culture with IL-15.
  • the staining is mainly detected on the basolateral membranes and in the basal cytoplasm (B). ⁇ Original
  • enterocytes are TUNEL+ after challenge with gliadin (A) ; a dramatic decrease in
  • Ml 10 anti-IL-15 MoAb (B) (Original magnification, xl80)
  • Figure 7 Effect of IL-15 and IL-2 on the expression of immunological markers after 24 h
  • Figure 8 Effect of IL-15 on cell death in CACO-2 cell line simultaneously cultured with
  • IL-15 vs medium alone, or vs IL-7.
  • IL-2 gliadin.
  • IL-15+M3MoAb Trypan-Blue+ cells in
  • Figure 9 Effect of IL-15 on CACO-2 cell line: induction of apoptosis after 24 h of incubation with IL- 15. After 24 h of IL-15 treatment, incubation with FITC Annexin-V (green colour), and
  • propidium iodide (orange colour) leads some cells to show green colour and others to
  • non-CD controls (mean age 38.5, range 19-53) underwent duodenal endoscopy for
  • IL-15 and IL-7 were obtained from
  • IL-15 IL-7. IL-2 and IL-4 were added to the medium at the final concentration of 10
  • IL-15 10 mg/ml
  • ICAM-1 (Dako. Copenhagen. Denmark. 1 :400)
  • CD25 (Dako. 1:30), CD3 (Dako. 1 :200), CD8 (Dako, 1 :200), ⁇ l ⁇ (T Cell Diagnostics. Inc.
  • alkaline phosphatase/anti-alkaline phosphatase or perxoidase staining techniques
  • anti-CD3 or CD8 or yl ⁇ MoAbs were numbered per mm epithelium; the number of
  • EMA detection in culture supernatants EMA detection in culture supernatants.
  • Dulbecco ' s modified Eagle ' s medium containing 25 mmol/L glucose and
  • wortexing cells were loaded in a Neubaumer chamber (Carlo Erba. Milan, Italy) and counted.
  • TdT deoxynucleotidyl transferase
  • TUNEL method and visualised by peroxidase staining as previously described 34 .
  • PBS phosphate buffer
  • IL-15 induces migration and activation of T cells in treated celiac and control
  • biopsies of treated CD with IL-15 induced the migration of CD3+ cells both into the
  • SE subepithelial
  • IE intraepithelial
  • gliadin was also competent in inducing migration of ⁇ + cells to the IEC (Fig 1A). IL-7,
  • ICM-1 intercellular adhesion molecule- 1
  • IL-2 intercellular adhesion molecule- 1
  • IL-4 or IL-2 induced an intraepithelial increase in CD8+ cells in celiac and control intestine.
  • IL-15 also increases the number of intraepithelial CD94+ cells in celiacs only.
  • IL-7 was not found to increase CD94+ cells in celiac biopsies.
  • CD8+ (p ⁇ .001) and CD94+ (p ⁇ 0.05) cells in celiacs but not in controls.
  • IL-15 challenge induces epithelial changes in treated celiac but not in control
  • enterocytes were also detected in villus after IL-15 treatment. No Ki67 expression by
  • gliadin challenge was effective in enhancing epithelial expression of FAS in 8
  • enterocytes of non-CD controls low or undetectable FAS expression in all 8 tested cases
  • EMA antiendomysium antibodies
  • gliadin. did not induce EMA in any of the 5 tested non-CD control cases.
  • Anti-IL-15 neutralising antibodies control the epithelial changes and the production of EMA induced by IL-15 as well as by gliadin challenge in treated celiac intestine
  • MoAbS monoclonal antibodies
  • Ml 10 and Ml 11 Two MoAbs (Ml 10 and Ml 11)
  • MHO MoAb was effective in down regulating enterocytes expression (Fig 5 A-B). On the contrary, neutralising monoclonal antibodies were not effective in
  • Anti-human lactase MoAb mlacl used as isotype
  • the inventors have found that a 24 h of in vitro with gliadin challenge in untreated CD
  • intestine biopsies is effective in producing enterocytes's DNA fragmentation (Maiuri L. et).
  • LPMNC lamina limbal mononuclear cells
  • IL-15 the CACO-2 epithelial cell line
  • IL-15 and IL-2 but not IL-7 nor gliadin induces Ki67 antigen expression.
  • Ki67 expression was restricted to a lower number of cells (less than
  • IL-15 but not IL-2, IL-7 nor gliadin induces FAS and FAS-L expression
  • IL-15 but not IL-2, IL-7 nor gliadin induces cell death
  • CD has always been considered as the prototype of an immuno-mediated disease in which
  • gliadin a single antigen, gliadin, induces T cell activation leading to disease 1, 2 .
  • gliadin could be, by enlarge, controlled with neutralising anti-IL-15 monoclonal
  • gliadin has a central role in the pathogenesis of CD in 3 different ways. Firstly by
  • IL-15 has another unique characteristic, which further suggests a
  • CD potential role in CD: the ability to directly modulate small intestine epithelial cells 12 .
  • one treated CD biopsy was controlled by neutralising anti-IL-15 monoclonal antibodies.
  • IL-15 could control the induction of EMA by different, and not conflicting, ways for
  • tissue transglutaminase tissue transglutaminase
  • IL-15 fulfils the role of an agent unmasking a 'hidden' autoantigen (translutaminase). IL-15 may further influence the production of EMA acting
  • T and B cells 27,28, 29 as a locally available growth and differentiation factor for T and B cells 27,28, 29 .
  • Epithelial cells seem to be not involved, although small intestine epithelial cells
  • the first is the restricted ability of monocytic cells of celiacs to produce IL-15 after gliadin challenge.
  • the second being the specific effects of IL-15 on epithelial

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Engineering & Computer Science (AREA)
  • Biophysics (AREA)
  • Molecular Biology (AREA)
  • Genetics & Genomics (AREA)
  • Zoology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Immunology (AREA)
  • Biochemistry (AREA)
  • Epidemiology (AREA)
  • Oncology (AREA)
  • Communicable Diseases (AREA)
  • Toxicology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)
  • Medicinal Preparation (AREA)

Abstract

La présente invention concerne le traitement de maladies inflammatoires de l'intestin, telle que la maladie coeliaque, à l'aide d'antagonistes de l'interleukine-15 (IL-15). De préférence, les antagonistes sont des mutéines de l'IL-15, des anticorps contre l'IL-15 ou des molécules d'IL-15 liées à des groupes chimiques qui interfèrent avec l'aptitude de l'IL-15 à effectuer une transduction de signal via les sous-unités β ou η du complexe récepteur de l'IL-15, mais qui n'interfèrent pas avec la liaison de l'IL-15 avec l'IL-15Rα.
EP99933001A 1998-07-10 1999-07-09 Traitement de la maladie coeliaque a l'aide d'antagonistes de l'interleukine-15 Withdrawn EP1096949A2 (fr)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
GB9814892 1998-07-10
GBGB9814892.7A GB9814892D0 (en) 1998-07-10 1998-07-10 Treatment of celiac disease
PCT/GB1999/002201 WO2000002582A2 (fr) 1998-07-10 1999-07-09 Traitement de la maladie coeliaque a l'aide d'antagonistes de l'interleukine-15

Publications (1)

Publication Number Publication Date
EP1096949A2 true EP1096949A2 (fr) 2001-05-09

Family

ID=10835232

Family Applications (1)

Application Number Title Priority Date Filing Date
EP99933001A Withdrawn EP1096949A2 (fr) 1998-07-10 1999-07-09 Traitement de la maladie coeliaque a l'aide d'antagonistes de l'interleukine-15

Country Status (5)

Country Link
EP (1) EP1096949A2 (fr)
JP (1) JP2002520294A (fr)
CA (1) CA2333923A1 (fr)
GB (1) GB9814892D0 (fr)
WO (1) WO2000002582A2 (fr)

Families Citing this family (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7247304B2 (en) 2001-08-23 2007-07-24 Genmab A/S Methods of treating using anti-IL-15 antibodies
US7329405B2 (en) 2001-08-23 2008-02-12 Genmab A/S Human antibodies specific for interleukin 15 (IL-15)
RS51829B (sr) * 2001-08-23 2012-02-29 Genmab A/S. Ljudska antitela specifična za interleukin 15 (il-15)
CU23093A1 (es) 2002-10-09 2005-10-19 Ct Ingenieria Genetica Biotech Composición vacunal que comprende interleucina-15 (il-15)
EA015897B1 (ru) * 2003-02-26 2011-12-30 Генмаб А/С Применение человеческого моноклонального антитела к ил-15 в составе композиции и лекарственного препарата (варианты), композиция и лекарственный препарат, его включающие
US20050123542A1 (en) * 2003-11-06 2005-06-09 Genmab A/S Methods for treating disorders involving monocytes
WO2005099753A1 (fr) * 2004-04-16 2005-10-27 The Govenors Of The University Of Alberta Anticorps de jaune d'oeuf anti-gluten pour le traitement de la maladie caeliaque
CU23472A1 (es) 2004-09-17 2009-12-17 Ct Ingenieria Genetica Biotech Péptido antagonista de la interleucina-15
NZ569541A (en) 2006-01-13 2012-05-25 Us Gov Health & Human Serv Codon optimized IL-15 and IL-15R-alpha genes for expression in mammalian cells
US8282928B2 (en) 2007-05-30 2012-10-09 The Governors Of The University Of Alberta Anti-gluten egg yolk antibodies for the treatment of celiac disease
AU2010282280B2 (en) 2009-08-14 2016-06-09 The Government Of The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Use of IL-15 to increase thymic output and to treat lymphopenia
US20180002417A1 (en) * 2016-06-15 2018-01-04 Celimmune Llc Methods and Compositions for the Treatment of Celiac Disease, Non-Celiac Gluten Sensitivity, and Refractory Celiac Disease
WO2018041989A1 (fr) * 2016-09-02 2018-03-08 INSERM (Institut National de la Santé et de la Recherche Médicale) Procédés de diagnostic et de traitement de la maladie coeliaque réfractaire de type 2

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5574138A (en) * 1993-03-08 1996-11-12 Immunex Corporation Epithelium-derived T-cell factor
US5795966A (en) * 1995-02-22 1998-08-18 Immunex Corp Antagonists of interleukin-15
CA2252557A1 (fr) * 1996-04-26 1997-11-06 Beth Israel Deaconess Medical Center, Inc. Antagonistes de l'interleukine-15

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO0002582A2 *

Also Published As

Publication number Publication date
JP2002520294A (ja) 2002-07-09
WO2000002582A2 (fr) 2000-01-20
CA2333923A1 (fr) 2000-01-20
GB9814892D0 (en) 1998-09-09
WO2000002582A3 (fr) 2000-03-16

Similar Documents

Publication Publication Date Title
US7235240B2 (en) Antagonists of interleukin-15
CA2210491C (fr) Antagonistes de l'interleukine-15
US7803361B2 (en) Therapeutic use of interleukin-2 mutants
JP4787439B2 (ja) 免疫調節因子であるbaffレセプター(bcma)
US20100086517A1 (en) Enhancement of B cell proliferation by IL-15
CA2194299A1 (fr) Nouvelle cytokine designee lerk-5
WO2000002582A2 (fr) Traitement de la maladie coeliaque a l'aide d'antagonistes de l'interleukine-15
EP0724456A1 (fr) Anticorps diriges contre le cd40
JP4299887B2 (ja) 免疫学的疾患の治療のための治療剤としての、可溶性リンホトキシン―βレセプター、抗リンホトキシンレセプター抗体、および抗リンホトキシンリガンド抗体
US6884603B2 (en) Nucleic acids encoding IL13 mutants
WO2001025282A1 (fr) Mutants il13
MX2007004094A (en) Enhancement of b cell proliferation by il-15

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20010212

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE CH CY DE DK ES FI FR GB GR IE IT LI LU MC NL PT SE

GRAG Despatch of communication of intention to grant

Free format text: ORIGINAL CODE: EPIDOS AGRA

17Q First examination report despatched

Effective date: 20010605

GRAG Despatch of communication of intention to grant

Free format text: ORIGINAL CODE: EPIDOS AGRA

GRAH Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOS IGRA

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20020903