US20140093509A1 - Screening Method for the Identification of Agents Capable of Activating CD4+CD25+ Regulatory T-Cells Through Interactions with the HIV-1 GP120 Binding Site on CD4 - Google Patents

Screening Method for the Identification of Agents Capable of Activating CD4+CD25+ Regulatory T-Cells Through Interactions with the HIV-1 GP120 Binding Site on CD4 Download PDF

Info

Publication number
US20140093509A1
US20140093509A1 US14/031,919 US201314031919A US2014093509A1 US 20140093509 A1 US20140093509 A1 US 20140093509A1 US 201314031919 A US201314031919 A US 201314031919A US 2014093509 A1 US2014093509 A1 US 2014093509A1
Authority
US
United States
Prior art keywords
seq
cells
treg cell
hiv
treg
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.)
Abandoned
Application number
US14/031,919
Inventor
Franz-Josef Schneider
Christian Becker
Tobias Bopp
Helmut Jonuleit
Edgar Schmitt
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.)
TCF GmbH
Original Assignee
TCF GmbH
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 TCF GmbH filed Critical TCF GmbH
Priority to US14/031,919 priority Critical patent/US20140093509A1/en
Assigned to BOEHRINGER INGELHEIM INTERNATIONAL GMBH reassignment BOEHRINGER INGELHEIM INTERNATIONAL GMBH ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BECKER, CHRISTIAN, BOPP, TOBIAS, JONULEIT, HELMUT, SCHMITT, EDGAR, SCHNEIDER, FRANZ-JOSEF
Assigned to JONULEIT, HELMUT reassignment JONULEIT, HELMUT ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BOEHRINGER INGELHEIM INTERNATIONAL GMBH
Assigned to TCF GMBH reassignment TCF GMBH ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: JONULEIT, HELMUT
Publication of US20140093509A1 publication Critical patent/US20140093509A1/en
Priority to US15/285,561 priority patent/US10729742B2/en
Priority to US16/912,199 priority patent/US20200316167A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • 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
    • A61K38/162Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from virus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • A61K39/21Retroviridae, e.g. equine infectious anemia virus
    • 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
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/14Prodigestives, e.g. acids, enzymes, appetite stimulants, antidyspeptics, tonics, antiflatulents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/18Drugs for disorders of the alimentary tract or the digestive system for pancreatic disorders, e.g. pancreatic enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/02Nasal agents, e.g. decongestants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/08Bronchodilators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/16Central respiratory analeptics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/04Antipruritics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/02Muscle relaxants, e.g. for tetanus or cramps
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/04Drugs for disorders of the muscular or neuromuscular system for myasthenia gravis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/14Decongestants or antiallergics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • 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
    • 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
    • A61P37/04Immunostimulants
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/14Drugs for disorders of the endocrine system of the thyroid hormones, e.g. T3, T4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5044Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics involving specific cell types
    • G01N33/5047Cells of the immune system
    • G01N33/505Cells of the immune system involving T-cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • 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
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/16011Human Immunodeficiency Virus, HIV
    • C12N2740/16033Use of viral protein as therapeutic agent other than vaccine, e.g. apoptosis inducing or anti-inflammatory
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/16011Human Immunodeficiency Virus, HIV
    • C12N2740/16111Human Immunodeficiency Virus, HIV concerning HIV env
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/16011Human Immunodeficiency Virus, HIV
    • C12N2740/16111Human Immunodeficiency Virus, HIV concerning HIV env
    • C12N2740/16122New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/005Assays involving biological materials from specific organisms or of a specific nature from viruses
    • G01N2333/08RNA viruses
    • G01N2333/15Retroviridae, e.g. bovine leukaemia virus, feline leukaemia virus, feline leukaemia virus, human T-cell leukaemia-lymphoma virus
    • G01N2333/155Lentiviridae, e.g. visna-maedi virus, equine infectious virus, FIV, SIV
    • G01N2333/16HIV-1, HIV-2
    • G01N2333/162HIV-1, HIV-2 env, e.g. gp160, gp110/120, gp41, V3, peptid T, DC4-Binding site
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants
    • G01N2333/70503Immunoglobulin superfamily, e.g. VCAMs, PECAM, LFA-3
    • G01N2333/7051T-cell receptor (TcR)-CD3 complex

Definitions

  • PCT/EP2008/051144 entitled “SPECIFIC ACTIVATION OF A REGULATORY T CELL AND ITS USE FOR TREATMENT OF ASTHMA, ALLERGIC DISEASE, AUTOIMMUNE DISEASE, GRAFT REJECTION AND FOR TOLERANCE INDUCTION”, filed Jan. 30, 2008, which claims priority to European Patent Application No. 07101604.2, entitled “SPECIFIC ACTIVATION OF A REGULATORY T CELL AND ITS USE FOR TREATMENT OF ASTHMA, ALLERGIC DISEASE, AUTOIMMUNE DISEASE, GRAFT REJECTION AND FOR TOLERANCE INDUCTION”, filed Feb.
  • an allergic disease, transplant rejection and an autoimmune disease have one fundamental principle in common, they all are triggered by an imbalanced immune system which reacts hyperactive against a specific exogenic and/or endogenic challenge and therewith contributes significantly to the disease status.
  • Effector T cell-directed immunomodulation therefore is the key to successful treatment of asthma, an autoimmune condition, prevention of graft vs. host disease (GVHD) and prevention of graft rejection.
  • GVHD graft vs. host disease
  • Treg cells regulatory T cells
  • CD4 + CD25 + Treg cells also designated as “naturally occurring Treg cells” (Sakaguchi 2005)
  • Tr1 Noncarolo et al. 2001
  • Th3 Th3
  • Tr1 and Th3 are induced in the periphery
  • CD4 + CD25 + Treg cells develop in the thymus and constitute 5-10% of peripheral CD4 + T cells in healthy man.
  • Tr1 and Th3 exert their suppressive activity by production of IL-10 and TGF-beta, respectively (Shevach 2002).
  • IPEX syndrome immunological hypertension, polyendo-crinopathy and enteropathy, X-linked
  • IPEX shows aggressive autoimmunity, severe eczema, elevated IgE levels, eosinophilia and food allergies and early death (Fontenot and Rudensky 2005).
  • Treg cells play an important role in asthma and autoimmune diseases and have a potential for treatment of GVHD and therewith transplantation tolerance (Robinson 2004, Sakaguchi 2005).
  • Treg cells are rare in peripheral blood clinical application of human Treg cells depends on highly expensive ex vivo expansion of Treg cells (Hoffmann et al. 2004, Horwitz et al. 2003, Tang et al. 2003, Zheng et al. 2004).
  • Bluestone and Tang went one step further: They are trying to solve the problem by not only increasing the amount of Treg cells for therapy but enhancing suppressive activity of Treg cells by activating Treg cells via triggering of the T cell receptor (TCR) by an anti-CD3 antibody (Bluestone and Tang 2004).
  • TCR T cell receptor
  • This approach resembles by far no Treg cell specific activation as anti-CD3 activates all T cell receptor-expressing cells which bears the obstacle that anti-CD3 treatment induces effector T cell function probably leading to uncontrolled proliferation and non-specific pro-inflammatory cytokine production and exaggeration of pathology.
  • Treg cells have to be highly purified and activated ex vivo with anti-CD3 which again is a highly expensive and laborious procedure.
  • the absence of a Treg cell specific marker makes it difficult to achieve high purity of Treg cells.
  • Treg cells possibly can be activated, e.g. nonspecifically via CD3 (Thornton and Shevach 1998), or via CD28/B7 pathway (Shevach 2002, Bluestone and Tang 2004, Hunig and Dennehy 2005), or via CD4 (WO04083247). So far all this solutions do not result in a specific Treg cell activation.
  • the present invention bases on the new finding that a specific epitope of CD4 triggers activation of Treg cells. Said epitope overlaps with the known HIV-1 gp120-binding site but surprisingly binding to this site causes Treg cell activation. This finding was totally unexpected for the following reasons: Until now Treg cell activation via CD4 was attributed to a different epitope to which the monoclonal antibody BF5 (WO04083247) binds.—Carriere et al., 1995 have investigated that the binding site of BF5 on CD4 is completely independent of the HIV-1 gp120-binding site.—Unexpectedly, despite reports in the literature on direct anergizing and blocking of CD4 on T cell function by HIV-1 gp120 (Diamond et al. 1988), we found an activating property of HIV-1 gp120 on CD4 of Treg cells.
  • the present invention discloses a physiologically active CD4 epitope on a Treg cell which triggers suppressive activity of a Treg cell.
  • the epitope of the present invention has been identified as a region on the human CD4 protein (SEQ ID NO.: 2) spanning amino acid position No. 54 to 84 of SEQ ID NO.:2. Those 31 amino acids are explicitly given in SEQ ID NO.:1.
  • a preferred peptide which is a CD4 fragment according to the present invention is selected from a group consisting of an isolated peptide spanning amino acid No. 1-31 of SEQ ID NO 1, No. 26 to 458 of SEQ ID NO.: 2, No. 26 to 419 of SEQ ID NO.:2, No. 26 to 207 of SEQ ID NO.: 2, No. 26 to 131 of SEQ ID NO.: 2 and No. 46 to 89 of SEQ ID NO.: 2. All said peptides are additionally characterized in that they all do harbor the critical amino acid Phenylalanine at residue 68 of SEQ ID NO.: 2.
  • Treg cell activator of the present invention which is useful for the treatment of a disease of the invention which is a disease in which increase of activated regulatory T cells (Treg cells) can improve clinical picture wherein such a disease is (i) a non-autoimmune inflammatory disease: asthma, allergic asthma, respiratory allergy, allergic rhinoconjunctivitis, allergic alveolitis, contact allergy, atopic dermatitis, neurodermatitis, food allergy, graft-versus-host disease, non-autoimmune inflammatory bowel disease, acute respiratory distress syndrome, acute inflammatory pancreatitis, burns, wound healing, skin scarring disorders, sarcoidosis, Behcet's disease or Sweet's syndrome; (ii) an autoimmune inflammatory disease: rheumatoid arthritis, rheumatic fever, systemic lupus erythematosus, ulcerative colitis, Crohn's disease, autoimmune
  • a “substance” of the present invention can be used in a method according to the present invention.
  • the meaning of the term substance according to the present invention includes but is not limited to a peptide, a scaffolded peptide, an antibody, a fragment of an antibody, a nucleic acid molecule, a ribozyme, an organic compound or an inorganic compound.
  • the new epitope serves as the basis for the synthesis of a new tool which can be used e.g. in a competition assay or screening assay according to the present invention for determining whether a substance can activate a regulatory T cell (Treg cell) via interaction with the epitope as set forth in SEQ ID NO.:1.
  • a tool is a peptide of the present invention which is an isolated peptide spanning amino acid No. 1-31 of SEQ ID NO 1, or No. 26 to 458 of SEQ ID NO.: 2, or No. 26 to 419 of SEQ ID NO.:2, or No. 26 to 207 of SEQ ID NO.: 2, or No. 26 to 131 of SEQ ID NO.: 2 or No.
  • SEQ ID NO.: 2 is an isolated peptide spanning amino acid No. 1 to 31 of SEQ ID NO.: 1 and having additional up-stream and/or downstream amino acids with the prerequisite that the additional amino acids do not hinder binding of a substance to the amino acid stretch as set forth in SEQ ID NO.: 1.
  • a preferred peptide according to the present invention consist of the peptide as set forth in SEQ ID NO.:1 and additionally consist of an additional up-stream amino acid or amino acid stretch which is selected from a group consisting of the amino acid or amino acid stretch as set forth in SEQ ID NO.: 2 at position 53, at position 52-53, at position 51-53, at position 50-53, at position 49-53, at position 48-53, at position 47-53, at position 46-53, at position 45-53, at position 44-53, at position 43-53, at position 42-53, at position 41-53, at position 40-53, at position 39-53, at position 38-53, at position 37-53, at position 36-53, at position 35-53, at position 34-53, at position 33-53, at position 32-53, at position 31-53, at position 30-53, at position 29-53, at position 28-53, at position 27-53, at position 26-53, at position 25-53, at position 24-53, at position 23-53, at position
  • a further preferred peptide according to the present invention consists of the peptide which is mentioned as “preferred peptide” in the paragraph above and additionally consists of at least one additional downstream amino acid as given in SEQ ID NO.: 2 at position 85, or additionally consists of amino acids as given in SEQ ID NO.:2 at position 85 to n, wherein n is an integer between 86-458, i.e.
  • the finding that the epitope given in SEQ ID NO.:1 is a key to activate a Treg cell is a link between two different up to now unrelated technical fields, namely that of (a) HIV-1 related diseases with (b) diseases according to the present invention e.g. autoimmune disease, allergy, asthma, graft rejection and a diseases due to lacking immunotolerance caused by organ transplantation or by therapeutical administration of a non-self or self biological entity to a human in need thereof and therewith allows a bundle of new uses as explained in the following:
  • the epitope given in SEQ ID NO.:1 is not only a further epitope which can be used to activate Treg cells. HIV-1 gp120 interacts with CD4 of T cells and therewith enables virus entry into a CD4 + cell (Klatzmann et al. 1984).
  • the finding that epitope given in SEQ ID NO.:1 harbors the high affinity binding site on CD4 to which human immune deficiency virus 1 (HIV-1) glycoprotein gp120 binds offers a further advantage. It provides the basis to bring together the findings of two different unrelated technical fields that of i.e. HIV-1 related diseases with diseases according to the present invention.
  • HIV-1 attachment or entry inhibitors are known in the art.
  • the keyhole which allows HIV-1 to enter the cell can be used as the keyhole to activate Treg cells. Therefore, substances known in the art to interfere with HIV-1 attachment and cell entry (Markovic and Clouse 2004, Castagna et al. 2005), like e.g. HIV-1 gp120 itself, derivatives thereof, peptidomimetics, antibodies, aptamers or any low molecular weight (LMW) compound directed against the binding site of HIV-1 gp120 on CD4 could possibly be useful to activate a Treg cell and therewith can be useful for the treatment of a disease according to the present invention (HIV-1 gp120 is well-known in the art and its amino acid sequence as well as the respective gene has been published since years (Muesing et al.
  • LMW low molecular weight
  • HIV-1 attachment inhibitor or entry inhibitor A substance which can interfere with HIV-1 attachment and/or cell entry is commonly named HIV-1 attachment inhibitor or entry inhibitor. Such a substance, do either bind to (i) HIV-1, or (ii) to CD4, or (iii) HIV-1 and CD4 or (iv) co-receptor e.g. CCR5 or CXCR4.
  • Such an inhibitor can be according to the present invention useful for the treatment of diseases according to the present invention like e.g. an autoimmune disease, an allergy, asthma or GVHD if it exerts property (ii) or (iii) and activates a Treg cell.
  • the present invention discloses several assays which allow to determine whether a substance identified in the technical field of HIV-1 research to interfere with HIV-1 attachment and/or cell entry can be useful in the other above-mentioned technical fields like that of autoimmune diseases or allergies or asthma or organ transplantation. Therefore, the present invention teaches a short cut for identifying a substance which can be useful for the treatment of a disease according to the present invention.
  • an appropriate pre-selected substance is (i) proven to interact at least with epitope (SEQ ID NO.:1), and/or is (ii) known from HIV-1 research as HIV-1 attachment inhibitor or entry inhibitor or synthetic mimetics of the CD4 binding site of HIV-1 gp120.
  • Such a type of assay for determining whether a substance can interact with a specific peptide, i.e. epitope is not restricted to in vitro assays since cellular assays for achieving such an information on a substance or other in vitro formats are well known in the art and broadly used.
  • the present invention concerns a method for determining whether a substance which can interfere with the interaction of HIV-1 gp120 with CD4 can be useful for positively influence a disease in which increase of activated regulatory T cells (Treg cells) can improve clinical picture comprising:
  • a disease according to the method above in which increase of activated regulatory T cells (Treg cells) can improve clinical picture is (i) a non-autoimmune inflammatory disease: asthma, allergic asthma, respiratory allergy, allergic rhinoconjunctivitis, allergic alveolitis, contact allergy, atopic dermatitis, neurodermatitis, food allergy, graft-versus-host disease, non-autoimmune inflammatory bowel disease, acute respiratory distress syndrome, acute inflammatory pancreatitis, burns, wound healing, skin scarring disorders, sarcoidosis, Behcet's disease or Sweet's syndrome; (ii) an autoimmune inflammatory disease: rheumatoid arthritis, rheumatic fever, systemic lupus erythematosus, ulcerative colitis, Crohn's disease, autoimmune inflammatory bowel disease, diabetes type I, gastritis, autoimmune atrophic gastritis, autoimmune hepatitis, Hashimoto's thyroiditis, thyreoiditis, multiple s
  • the solution of (a) does not only contain a Treg cell but also an inactivated syngenic CD3-depleted PBMC (peripheral blood mononuclear cell) or a dendritic cell (DC) and an allogeneic CD8 + T cell or an allogeneic CD4 + T cell.
  • PBMC peripheral blood mononuclear cell
  • DC dendritic cell
  • an allogeneic CD8 + T cell or an allogeneic CD4 + T cell do additionally increase intracellular cAMP amount and therewith lead to a more sensitive readout system.
  • the present invention concerns a method for determining whether a substance can activate a Treg cell via interaction with the HIV-1 gp120-binding site of CD4, comprising:
  • the present invention also provides for a method which allow to identify a substance which can interact with the HIV-1 gp120-binding site of CD4 and therewith can be used for pre-selecting a substance which can interact with the HIV-1 gp120-binding site of CD4.
  • the method comprises:
  • This pre-selecting method can additionally be performed according to the present invention if instead of CD4 a peptide is used which comprises the amino acid stretch as set forth in SEQ ID NO.1 or instead of CD4 a peptide is used as set forth in SEQ ID NO.:1.
  • a peptide is used selected from a group consisting of a peptide comprising the amino acid sequence as set forth in SEQ ID NO.: 3, SEQ ID NO.: 4, SEQ ID NO.: 5, SEQ ID NO.: 6, SEQ ID NO.: 7, SEQ ID NO.: 8, SEQ ID NO.: 9, and SEQ ID NO.: 10 or a peptide is used selected from a group consisting of a peptide consisting of the amino acid sequence as set forth in SEQ ID NO.: 3, SEQ ID NO.: 4, SEQ ID NO.: 5, SEQ ID NO.: 6, SEQ ID NO.: 7, SEQ ID NO.: 8, SEQ ID NO.: 9, and SEQ ID NO.: 10.
  • the present invention concerns a method for determining whether a substance can activate a regulatory T cell (Treg) via interaction with the epitope as set forth in SEQ ID NO: 1 comprising:
  • the peptide used is selected from a group consisting of an isolated peptide spanning amino acid No. 1-31 of SEQ ID NO 1, No. 26 to 458 of SEQ ID NO.: 2, No. 26 to 419 of SEQ ID NO.:2, No. 26 to 207 of SEQ ID NO.: 2, No. 26 to 131 of SEQ ID NO.: 2 and No. 46 to 89 of SEQ ID NO.: 2 or the peptide used is an isolated peptide spanning amino acid No. 1 to 31 of SEQ ID NO.: 1 and having additional up-stream and/or downstream amino acids with the prerequisite that the additional amino acids do not hinder binding of a substance to the amino acid stretch as set forth in SEQ ID NO.:1.
  • Said additional up-stream amino acid or amino acid stretch is preferably selected from a group consisting of the amino acid or amino acid stretch as set forth in SEQ ID NO.: 2 at position 53, at position 52-53, at position 51-53, at position 50-53, at position 49-53, at position 48-53, at position 47-53, at position 46-53, at position 45-53, at position 44-53, at position 43-53, at position 42-53, at position 41-53, at position 40-53, at position 39-53, at position 38-53, at position 37-53, at position 36-53, at position 35-53, at position 34-53, at position 33-53, at position 32-53, at position 31-53, at position 30-53, at position 29-53, at position 28-53, at position 27-53, at position 26-53, at position 25-53, at position 24-53, at position 23-53, at position 22-53, at position 21-53, at position 20-53, at position 19-53, at position 18-53, at
  • a more preferred peptide does additionally comprise downstream one or more amino acids as set forth in SEQ ID NO.: 2 at position 85, or at position 85 to n, wherein n is an integer between 86-458, i.e. position 85 to 86, 85 to 87, 85 to 88, 85 to 89, 85 to 90, 85 to 91, 85 to 92, 85 to 93, 85 to 94, 85 to 95, 85 to 96, 85 to 97, 85 to 98, 85 to 99, 85 to 100, 85 to 101, 85 to 102, 85 to 103, 85 to 104, 85 to 105, 85 to 106, 85 to 107, 85 to 108, 85 to 109, 85 to 110, 85 to 111, 85 to 112, 85 to 113, 85 to 114, 85 to 115, 85 to 116, 85 to 117, 85 to 118, 85 to 119, 85 to 120, 85 to 121, 85 to 122, 85 to 123, 85 to 124, 85 to 125, 85 to 126, 85
  • Treg cell activators which are structurally a peptide or a polypeptide, preferably an antibody of a binding fragment thereof or a scaffolded peptide.
  • the present invention further concerns a new antibody or a binding fragment thereof capable of binding to the peptide as set forth in SEQ ID NO.:1 with the proviso that the antibody or the antibody fragment is not OKT4A, OKT4D4, OKTcdr4a and not Leu3.
  • Said disclaimed antibodies all relate to a totally different technical filed, namely HIV-1 research which is not related to the activation of Treg cells and disease according to the present invention.
  • the present invention combines for the first time the technical field of (a) HIV-1 related diseases with (b) diseases according to the present invention e.g. autoimmune disease, allergy, asthma, graft rejection and a diseases due to lacking immunotolerance caused by organ transplantation or by therapeutical administration of a non-self or self biological entity to a human in need thereof.
  • a further preferred Treg cell activator peptide is a peptide selected from a group consisting of a peptide comprising the amino acid sequence as set forth in SEQ ID NO.: 3, SEQ ID NO.: 4, SEQ ID NO.: 5, SEQ ID NO.: 6, SEQ ID NO.: 7, SEQ ID NO.: 8, SEQ ID NO.: 9, and SEQ ID NO.: 10, or is a peptide selected from a group consisting of a peptide consisting of the amino acid sequence as set forth in SEQ ID NO.: 3, SEQ ID NO.: 4, SEQ ID NO.: 5, SEQ ID NO.: 6, SEQ ID NO.: 7, SEQ ID NO.: 8, SEQ ID NO.: 9, and SEQ ID NO.: 10.
  • a more preferred Treg cell activator polypeptide is selected from a group consisting of: HIV-1 gp120, NSC 13778 which chemical structure is disclosed in Yang et al. 2005 on page 6124 in FIG. 1 , peptide 2 which presents three HIV-1 gp120 fragments bound together through comformationally flexible scaffolds which chemical structure is disclosed in Franke et al. 2007 on page 4 at the bottom of the right column, monoclonal antibody OKT4A which binds to the HIV-1 gp120-binding region of CD4 as disclosed in Mizukami et al.
  • Treg cell activators can be used as a medicament and for the preparation of a medicament for the treatment of a disease selected from a group consisting of (i) a non-autoimmune inflammatory disease: asthma, allergic asthma, respiratory allergy, allergic rhinoconjunctivitis, allergic alveolitis, contact allergy, atopic dermatitis, neurodermatitis, food allergy, graft-versus-host disease, non-autoimmune inflammatory bowel disease, acute respiratory distress syndrome, acute inflammatory pancreatitis, burns, wound healing, skin scarring disorders, sarcoidosis, Behcet's disease, Sweet's syndrome; (ii) an autoimmune inflammatory disease: rheumatoid arthritis, rheumatic fever, systemic lupus erythematosus, ulcerative colitis, Crohn's disease, autoimmune inflammatory bowel disease, diabetes type I, gastritis, autoimmune atrophic gastritis, autoimmune hepatitis, Hashimoto's
  • a disease selected from a
  • Another embodiment of the present invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising at least one Treg cell activator according to the present invention—preferably HIV-1 gp120—as an active ingredient and can be formulated in conventional manner.
  • Methods for making such formulations can be found in manuals, e.g. “Remington Pharmaceutical Science”.
  • ingredients that are useful for formulating at least one substance according to the present invention are also found in WO99/18193, which is hereby incorporated by reference.
  • composition may be manufactured in a manner that is itself known, e.g. by mean of conventional mixing, dissolving, granulating, dragee-making, levitating, powdering, emulsifying, encapsulating, entrapping of lyophilizing processes.
  • the invention teaches a method for treating a disease which is characterized in that its clinical picture can be influenced positively by an increase of activated Treg cells which method comprises administering to a being preferably a human being in need of such a treatment a suitable amount of a pharmaceutical composition comprising at least one Treg cell activator according to the present invention, preferably HIV-1 gp120 or HIV-1 gp120 derived fragments and peptides thereof.
  • the present invention provides therefore for a method for treating (i) a non autoimmune inflammatory disease: asthma, allergic asthma, respiratory allergy, allergic rhinoconjunctivitis, allergic alveolitis, contact allergy, atopic dermatitis, neurodermatitis, food allergy, graft-versus-host disease, non-autoimmune inflammatory bowel disease, acute respiratory distress syndrome, acute inflammatory pancreatitis, burns, wound healing, skin scarring disorders, sarcoidosis, Behcet's disease or Sweet's syndrome; (ii) an autoimmune inflammatory disease: rheumatoid arthritis, rheumatic fever, systemic lupus erythematosus, ulcerative colitis, Crohn's disease, autoimmune inflammatory bowel disease, diabetes type I, gastritis, autoimmune atrophic gastritis, autoimmune hepatitis, Hashimoto's thyroiditis, thyreoiditis, multiple sclerosis, myasthenia gravis, autoimmune haemo
  • the present invention additionally, provides for a use of a Treg cell activator according to the present invention for reducing and/or preventing an unwanted immune reaction due to a exogenously administered self or exogenously administered non-autologous recombinant polypeptide and provides for a method for reducing or preventing an unwanted immune reaction comprising: (a) adding a sufficient amount of at least one Treg cell activator according to the present invention to a non-human animal, preferably a non-human primate.
  • a peptide of b) is selected from a group consisting of an isolated peptide spanning amino acid No. 26 to 458 of SEQ ID NO.: 2, amino acid No. 26 to 419 of SEQ ID NO.: 2, amino acid No. 26 to 207 of SEQ ID NO.: 2, amino acid No. 26 to 131 of SEQ ID NO.: 2, and amino acid No. 46 to 89 of SEQ ID NO.: 2. or the peptide used is an isolated peptide spanning amino acid No. 1 to 31 of SEQ ID NO.: 1 and having additional up-stream and/or downstream amino acids with the prerequisite that the additional amino acids do not hinder binding of a substance to the amino acid stretch as set forth in SEQ ID NO.: 1.
  • Said additional up-stream amino acid or amino acid stretch is preferably selected from a group consisting of the amino acid or amino acid stretch as set forth in SEQ ID NO.: 2 at position 53, at position 52-53, at position 51-53, at position 50-53, at position 49-53, at position 48-53, at position 47-53, at position 46-53, at position 45-53, at position 44-53, at position 43-53, at position 42-53, at position 41-53, at position 40-53, at position 39-53, at position 38-53, at position 37-53, at position 36-53, at position 35-53, at position 34-53, at position 33-53, at position 32-53, at position 31-53, at position 30-53, at position 29-53, at position 28-53, at position 27-53, at position 26-53, at position 25-53, at position 24-53, at position 23-53, at position 22-53, at position 21-53, at position 20-53, at position 19-53, at position 18-53, at
  • a more preferred peptide does additionally comprise downstream one or more amino acids as set forth in SEQ ID NO.: 2 at position 85, or at position 85 to n, wherein n is an integer between 86-458, i.e. position 85 to 86, 85 to 87, 85 to 88, 85 to 89, 85 to 90, 85 to 91, 85 to 92, 85 to 93, 85 to 94, 85 to 95, 85 to 96, 85 to 97, 85 to 98, 85 to 99, 85 to 100, 85 to 101, 85 to 102, 85 to 103, 85 to 104, 85 to 105, 85 to 106, 85 to 107, 85 to 108, 85 to 109, 85 to 110, 85 to 111, 85 to 112, 85 to 113, 85 to 114, 85 to 115, 85 to 116, 85 to 117, 85 to 118, 85 to 119, 85 to 120, 85 to 121, 85 to 122, 85 to 123, 85 to 124, 85 to 125, 85 to 126, 85
  • CD4/HIV-1 gp120 competition assay for determining whether a substance can bind at least to a peptide spanning epitope as set fourth in SEQ ID NO.: 1.
  • 96 well assay plates (Nunc, Germany) are coated overnight at 4° C. with CD4 (sCD4, Immunodiagnostics, USA) 100 ng per well in PBS, pH 7.4. Coated plates are saturated with PBS/3% BSA buffer and washed three times. To determine binding of a test substance sample is added for 1 hour in different concentrations. No test substance is added to control wells. After washing three times HIV-1 gp120-peroxidase conjugate (Immunodiagnostics, USA) is added to the plate for 1 h.
  • Unbound HIV-1 gp120-peroxidase conjugate is removed by washing three times. After washing, 3,3,5,5-tetramethylbenzidine chromogen substrate (Pierce, USA) for peroxidase is added and the optical density is read at 450 nm. A substance which interacts with the CD4 HIV-1 gp120-binding site will block the binding of labeled HIV-1 gp120 and is identifiable by a reduced signal compared to the controls.
  • Assay for determining whether e.g. HIV-1 gp120 or a substance which can interfere with HIV-1 gp120-binding to a CD4 + CD25 + Treg can activate a CD4 + CD25 + Treg and therewith can be useful in the treatment an autoimmune disease (e.g. Inflammatory Bowel Disease, Multiple Sclerosis, Rheumatoid Arthritis, Psoriasis, Diabetes Type I, Lupus Erythematosus, Phemphigus vulgaris, Thyreoiditis), other diseases with autoimmune aspects in their pathogenesis such as vitiligo, atopic dermatitis, an allergy (e.g. Allergic rhinitis,), asthma (e.g. allergic asthma), GVHD (graft-versus-host disease), graft rejection.
  • an autoimmune disease e.g. Inflammatory Bowel Disease, Multiple Sclerosis, Rheumatoid Arthritis, Psoriasis, Diabetes Type I, Lupus Ery
  • PBMC peripheral blood mononuclear cells
  • Blood from buffy coats is diluted 1:1 with PBS (phosphate buffered saline) containing 0.2% Liquemine (Sodium-Heparin) and 2 mM EDTA at room temperature.
  • PBS phosphate buffered saline
  • Liquemine Sodium-Heparin
  • 2 mM EDTA 2 mM EDTA
  • the diluted blood is thoroughly pipetted onto prepared Ficoll layers (30 ml diluted blood per 15 ml Ficoll layer per 50 ml tube) and centrifuged for 15 min. (minute) at 200 ⁇ g (with brake on) at room temperature. 8-10 ml of the upper fluid are carefully removed and the tubes centrifuged at 450 ⁇ g for 15 min. at room temperature (with brake off).
  • PBMC are collected from the interphase of each gradient, washed three times with 50 ml PBS/1 mM EDTA separately, than pooled and wash two more times. Finally, PBMC are re-diluted in X-VIVO-15 (Cambrex, Verviers, Belgium) in cell culture medium and counted.
  • antibody-coated magnetic beads are used (Miltenyi, Germany and or Dynal, Norway). Magnetic beads in this context are paramagnetic particles that are coupled to specific monoclonal antibodies. They are used to magnetically label the target cell population. The antibody-coated magnetic beads bind to the target cells. This labeled cell fraction is retained by magnetic force and can be recovered subsequently highly purified (positive selection). Before positive isolation of CD4 + CD25 ⁇ T helper cells PBMC are washed two times with 50 ml washing buffer according to the manufacturer's instructions (Miltenyi Germany).
  • CD4 microbeads (Miltenyi Germany, 2-4 ⁇ L microbeads/10 7 PBMC) are used according to the manufacturer's instructions.
  • the CD4 + fraction is isolated using a MACS separator (Miltenyi) according to the instructions of the manufacturer. MACS separator retains magnetic bead-labeled cells by magnetic force.
  • Contaminating CD4 + CD25 + Treg cells are depleted in a second step by using CD25 Dynabeads, (Dynal, Norway) according to the instructions of the manufacturer (details see 2.1.4) using (0.5 beads/cell). This depletion procedure results in highly purified CD4 + CD25 ⁇ T helper cells (negative selection).
  • CD4 + T helper cells are generated by using the negative isolation kit (Miltenyi Germany) according to the manufacturer's instructions. Before negative isolation of CD4 + T helper cells PBMC are washed two times with 50 ml washing buffer according to the manufacturer's instructions (Miltenyi Germany). For isolation of CD4 + T helper cells, PBMC are incubated with a cocktail of biotinylated CD45RO, CD8, CD14, CD16, CD19, CD56, CD36, CD123, anti-TCR ⁇ / ⁇ , and CD235a antibodies. These cells are subsequently magnetically labeled with Anti-Biotin Microbeads for depletion.
  • the CD4 + fraction is isolated using a MACS separator (Miltenyi) according to the instructions of the manufacturer. MACS separator retains magnetic bead-labeled cells by magnetic force. Contaminating CD4 + CD25 + Treg cells are depleted in a second step by using CD25 Dynabeads, (Dynal, Norway) according to the instructions of the manufacturer (details see 2.1.4) using 0.5 beads/cell.
  • MACS separator retains magnetic bead-labeled cells by magnetic force.
  • Contaminating CD4 + CD25 + Treg cells are depleted in a second step by using CD25 Dynabeads, (Dynal, Norway) according to the instructions of the manufacturer (details see 2.1.4) using 0.5 beads/cell.
  • CD8 + T effector cells Before positive isolation of CD8 + T effector cells PBMC are washed two times with 50 ml washing buffer according to the manufacturer's instructions (Miltenyi Germany). For isolation of CD8 + CD25 ⁇ T cells, CD8 microbeads (Miltenyi Germany, 2-4 ⁇ L microbeads/10 7 PBMC) are used according to the manufacturer's instructions. The CD8 + fraction is isolated using a MACS separator (Miltenyi) according to the instructions of the manufacturer. Contaminating CD8 + CD25 + T cells are depleted in a second step by using CD25 Dynabeads, (Dynal, Norway) according to the instructions of the manufacturer (details see 2.1.4) using (0.5 beads/cell). This depletion procedure results in highly purified CD8 + CD25 ⁇ T effector cells (negative selection).
  • CD25 + CD25 + Treg cells positive and negative selection is combined.
  • PBMC are washed with washing buffer according to the manufacturer's instructions (Miltenyi Germany) and subsequently incubated with CD25 microbeads (2 ⁇ L microbeads/10 7 PBMC) for 20 min. at 4° C. in isolation buffer (1 ⁇ 10 8 /ml) according to the manufacturer's instructions. Afterwards, the cells are washed two times in PBS. The CD25 + fraction is isolated using a MACS separator (Miltenyi) according to the instructions of the manufacturer.
  • the positively selected CD25 + fraction contains 65-80% CD4 + T cells and 20-35% contaminating CD19 + B cells, CD8 + T cells, and few CD14 + monocytes.
  • the contaminating cells are depleted with Dynabeads (Dynal, Norway).
  • the following amounts of beads are used: CD19 Dynabeads: 2 beads/cell, CD8 Dynabeads: 3 beads/cell, CD14 Dynabeads: 1 bead/cell.
  • Collected Dynabeads are washed two times in 15 ml tubes with depletion buffer using the magnetic particle concentrator (Dynal) according to the manufacturer's instructions.
  • the CD25 + PBMC fraction (5 ⁇ 10 7 /ml) is added in depletion buffer and incubated for 20 min.
  • Dendritic cells are generated from buffy coats of healthy volunteers.
  • PBMC (2.1.6) are plated in 6-well tissue culture plates at a density of 15 ⁇ 10 6 cells/well in 3 ml X-VIVO-15 (Cambrex, Verviers, Belgium) plus 1.5% heat-inactivated autologous plasma containing 800 U/ml GM-CSF (Leukomax; Novartis, Basel, Switzerland) and 1,000 U/ml IL-4 (Strathmann Biotec, Hamburg, Germany). Cultures are fed every other day (days 2, 4 and 6) by removing 1 ml of the medium and adding back 1 ml fresh medium with cytokines.
  • non-adherent cells were harvested and transferred to new 6 well plates and cultured further on in the presence of 10 ng/ml IL-1 ⁇ , 10 ng/ml TNF- ⁇ , 1,000 U/ml IL-6 (all from Strathmann, Biotech, Germany) and 1 ⁇ g/ml PGE 2 (Pharmacia-Upjohn, Uppsala, Sweden).
  • Mature CD83 + DC are harvested at day 9 of culture.
  • T cells are depleted from PBMC with CD3 Dynabeads (Dynal, Norway) by using 0.5 beads/cell. Collected Dynabeads are washed two times in 15 ml tubes with depletion buffer using the magnetic particle concentrator (Dynal) according to the manufacturer's instructions. PBMC (5 ⁇ 10 7 /ml) are added in depletion buffer and incubated for 20 min. at 4° C. on a shaker (sample mixer, Dynal). CD3 + cells are depleted according to the manufacturer's instructions by the use of the magnetic particle concentrator resulting in a purity of >98% CD3 ⁇ PBMC.
  • PBMC are isolated according to (2.1.1).
  • CD25-expressing regulatory T cells in the PBMC preparation are depleted with CD25 Dynabeads (Dynal, Norway) according to the instructions of the manufacturer (details see 2.1.4) by using 0.5 beads/cell.
  • Collected Dynabeads are washed two times in 15 ml tubes with depletion buffer using the magnetic particle concentrator (Dynal) according to the manufacturer's instructions.
  • PBMC (5 ⁇ 10 7 /ml) are added in depletion buffer and incubated for 20 min. at 4° C. on a shaker (sample mixer, Dynal).
  • CD25 + cells are depleted according to the manufacturer's instructions by the use of the magnetic particle concentrator resulting in a purity of >99% CD25-negative PBMC.
  • Coculture suppression assay A Mixed leukocyte reaction (MLR) Cocultures of CD4 + T helper cells or CD8 + T effector cells with CD4 + CD25 + Treg cells and allogeneic DC have to be performed to analyze the suppressive activity of CD4 + CD25 + Treg cells on CD4 + T helper cells or CD8 + T effector cells.
  • MLR Mixed leukocyte reaction
  • 1 ⁇ 10 5 /well CD4 + T helper cells (2.1.2) or CD8 + T effector cells (2.1.3) are cocultured with different numbers of CD4 + CD25 + Treg cells (2.1.4; ratio 1:1 to 1:4) and 1 ⁇ 10 4 /well DC in 96 well flat bottom culture plates in X-VIVO 15 (Cambrex, Verviers, Belgium) in the presence or absence of a CD4-binding compound e.g. HIV-1 gp120 (0.1-10 ⁇ g/ml).
  • a CD4-binding compound e.g. HIV-1 gp120 (0.1-10 ⁇ g/ml).
  • Mature dendritic cells (DC) generated as described (2.1.5) are from the same donor as CD4 + CD25 + Treg cells (syngenic) but allogeneic to the CD4 + T helper cells or CD8 + T effector cells are used for T cell stimulation.
  • CD4 + T helper cells or CD8 + T effector cells are activated by the allogeneic DC (MLR) resulting in a strong proliferation of the T cell subset.
  • MLR allogeneic DC
  • Non-activated CD4 + CD25 + Treg cells did not suppress this proliferation in absence of a Treg cell activating compound.
  • a functional activation of CD4 + CD25 + Treg cells by a CD4 ⁇ binding compound resulted in a reduced proliferation of CD4 + T helper cells or CD8 + T effector cells.
  • Proliferation is determined after 4 days of culture by adding 37 kBq 3H-Thymindine ( 3 H-Tdr) for additional 16 h.
  • Coculture Suppression Assay B Stimulation of CD8 + T effector cells with allogeneic PBMC and CD4 + CD25 + Treg cells from the same healthy volunteer.
  • HIV-1 gp120 on the function of CD4 + CD25 + Treg cells isolated CD4 + CD25 + Treg cells (2.1.4) are co-cultured with syngenic, T cell-depleted and irradiated (50 Gy) PBMC 2.1.6) and allogeneic CD8 + T effector cells (2.1.3) in presence of varying concentrations (0.1-10 ⁇ g/ml) of different HIV-1 gp 120 preparations. Briefly, 1 ⁇ 10 5 CD4 + CD25 + Treg cells are incubated with 3 ⁇ 10 5 syngenic T cell-depleted PBMC in the presence or absence of varying amounts of HIV-1 gp120.
  • CD4 is added in different concentrations (0.1-10 ⁇ g/ml) to isolated CD4 + CD25 ⁇ Treg cells (2.1.4) cocultured with syngenic T cell-depleted PBMC (2.1.6) and allogeneic CD8 + T effector cells (2.1.3) in the presence of varying amounts of the substance.
  • Enhanced proliferation of CD8 + effector cells resembles competitive binding to CD4 and blocked activation of a Treg cell via interaction with the epitope as set forth in SEQ ID NO:1. All cultures are performed in serum free X-VIVO-15 (Cambrex, Verviers, Belgium).
  • PBMC can be treated with Mitomycin C (Sigma, Germany). Briefly, 3 ⁇ 10 7 PBMC are incubated in 3 ml MEM/10% FCS/180 ⁇ g Mitomycin C for 30 min at 37° C. Afterwards cells are washed 5 ⁇ using MEM/10% FCS. Subsequently cells are subjected to the assay.
  • Mitomycin C Sigma, Germany
  • Coculture suppression assay C Stimulation of T cells in presence of pre-activated CD4 + CD25 + Treg cells and allogeneic PBMC
  • isolated CD4 + CD25 + Treg cells are pre-cultured in X-VIVO-15 for 16-48 h. alone, in presence of 0.5 ⁇ g/ml anti-CD3 monoclonal antibody (OKT-3) as positive control, or in presence of different concentrations of HIV-1 gp120. Afterwards, cells are washed intensively and added to cocultures of syngenic, irradiated (50 Gy) PBMC and allogeneic CD4 + T helper cells or CD8 + T effector cells. Proliferation is determined after additional 72 h.
  • a reduced incorporated radioactivity resembles suppressed proliferation and identifies CD8 + inhibition and therewith identifies a substance to activate a Treg cell via interaction with the epitope as set forth in SEQ ID NO.: 1.
  • T cell proliferation is measured.
  • cells are pulsed with 3 H-Tdr (37 kBq/well, MB Biomedicals), and incorporated radioactivity is measured by using a liquid scintillation counter (Betaplate, Wallac/PerkinElmer).
  • a Treg cell activating compound resting Treg cells cannot inhibit the proliferation of CD4 + T helper cells or CD8 + T effector cells (negative control).
  • CD4 + CD25 + Treg cells CD4 + T helper cells or CD8 + T effector cells
  • flow cytometry after labeling of these population with Vibrant CFDA SE Cell Tracer Kit (Invitrogen Life Technologies, San Diego, USA) according to the manufacturer's protocol.
  • CFDA labeling T cells (1 ⁇ 10 7 cells/ml) are incubated in PBS with 1 ⁇ M CFDA Vibrant CFDA SE Cell Tracer Kit (Invitrogen Life Technologies, San Diego, USA) at 37° C. for 30 min. Thereafter, cells are washed with X-VIVO-15 and incubated for 30 min. at 37° C. in the dark. After an additional wash the cells are counted and added to the cocultures. In addition, proliferation of cells is measured by flow cytometry after 4-6 days of culture.
  • CD8 + T effector cells are polyclonal restimulated with 2.4 ⁇ g/ml phytohemagglutinin (PHA, Sigma, Germany) and 1 ng/ml PMA for 5 h. in presence of monensin (BD GolgiStopTM, BD Biosciences Pharmingen 1.3 ⁇ M). Thereafter, cells are collected, washed with PBS, fixed and permeabilized according to the manufacturer's instruction (perm/fix solution, BD PharMingen, Germany) and stained with 0.5 ⁇ g/test of a cytokine-specific monoclonal antibody (anti-IFN- ⁇ , anti-IL-2, anti-TNF- ⁇ , all from BD PharMingen).
  • PHA phytohemagglutinin
  • PMA monensin
  • cytokine production of CD4 + T helper cells or CD8 + T effector cells is analyzed by flow cytometry.
  • Treg cells cannot inhibit the cytokine production of CD4 + T helper cells or CD8 + T effector cells (negative control).
  • an activating CD4-binding compound or anti-CD3 monoclonal antibodies positive control
  • the cytokine production of CD4 + T helper cells or CD8 + T effector cells is suppressed by functional activated Treg cells.
  • a reduced cytokine production identifies inhibition and therewith identifies that a CD4 + CD25 + Treg cell has been activated.
  • Treg cells inhibit the ability of CD8 + T effector cells and CD4 + T helper cells to express the ⁇ -chain of the IL-2 receptor, CD25. Therefore, the analysis of CD25 expression by flow cytometry is an additional method to evaluate the Treg cell activating potential of a reagent.
  • isolated CD4 + CD25 + Treg cells accordinging to 2.1.4
  • syngenic CD3-depleted PBMC accordinging to 2.1.6
  • allogeneic CD8 + T effector cells or allogeneic CD4 + T helper cells accordinging to 2.1.3, 2.1.2 and 2.2.1, 2.2.2 and 2.2.3).
  • alloreactive CD8 + T effector cells or allogeneic CD4 + T helper cells are stimulated with allogeneic PBMC from the same donor or DC as used in primary culture and expression of CD25 on alloreactive CD8 + T effector or allogeneic CD4 + T helper cells cells are analyzed 24 h. thereafter by flow cytometry.
  • Activation of Treg cells by the test compound results directly in an inhibited CD25 expression on the re-stimulated CD8 + T effector cells or CD4 + T helper cells.
  • Treg cells cannot inhibit the CD25 expression on re-stimulated CD8 + T effector cells or CD4 + T helper cells (negative control).
  • CD25 expression of CD8 + T effector cells or CD4+ T helper cells is suppressed by Treg cells.
  • a reduced CD25 expression identifies inhibition and therewith identifies that a CD4 + CD25 + Treg cell has been activated.
  • Treg cells results in strong increase of cytosolic (i.e. intracellular) cAMP. Therefore, the analysis of cAMP in Treg cells is an additional method to determine whether a substance which can interfere with HIV-1 gp120 binding to CD4 epitope of a CD4-positive cell can activate a Treg cell.
  • freshly isolated CD4 + CD25 + Treg cells (1 ⁇ 10 5 -1 ⁇ 10 6 /well) are incubated with anti-CD3 monoclonal antibody (OKT-3; 0.5 ⁇ g/ml) or HIV-1 gp120 (0.1-10 m/ml; Protein Science Corp., Meriden, Conn., USA) or left untreated for 16 hours.
  • a cAMP-specific ELISA (ParameterTM Cyclic AMP Assay, Cat. No. KGE002; R&D Systems, Wiesbaden, Germany) is applied. Treg cells are washed three times in ice-cold PBS and subsequently lysed (1 ⁇ 10 7 /ml) using lysis buffer supplied by the manufacturer and applied to the ELISA according to the manufacturer's recommendations.
  • NOD-Scid mice non-obese diabetic/severe combined immunodeficient mice (mutant allele: Prkdc Scid , strain: NOD.CB17-Prkdc Scid , (Shultz et al. 1995)) from the Central Laboratory Animal Facility of the University of Mainz (also purchasable from The Jackson Laboratory via Charles River Laboratories, Germany) are used as an animal model for transfer of peripheral human immune cells and thus for the in vivo analysis of immuno-regulatory interactions (Hesselton et al. 1995).
  • Human peripheral blood mononuclear cells (PBMC) transferred into NOD-Scid mice induce an accelerating age-dependent graft-versus-host disease (GVHD).
  • PBMC peripheral blood mononuclear cells
  • the human PBMC-induced disease is characterized by lack of weight increase and weight loss, decreased mobility, hunched posture, ruffled fur and organ inflammation in the treated animals (Kizilisik and Al-Sebayel 1997). Transfer of human PBMC results in growth arrest or weight loss within 30 to 40 days (depending on the number of cells transferred) after transfer compared to untreated mice. Lack of weight increase/weight loss is used as a parameter to score GVHD severity. Control mice did not receive any PBMC. Data see FIG. 3 .
  • HIV-1 gp120 or a substance which can interfere with HIV-1 gp120-binding is administered in addition to the injection of human PBMC.
  • Administration of HIV-1 gp120 or a substance which can interfere with HIV-1 gp120-binding induces an activation of human regulatory T cells in the PBMC fraction resulting in prevention of organ inflammation and prevention of growth arrest/weight loss similar to mice treated with human PBMC and additional human regulatory T cells or untreated mice. Lack of weight increase/weight loss is used as a parameter to score GVHD severity. Control mice did not receive any PBMC. Data see FIG. 4 .
  • NOD-Scid mice at the age of three to six days after birth are intraperitoneally injected with 1 ⁇ 10 7 human CD25-depleted PBMC (isolation of human CD25-depleted PBMC according to 2.1.7).
  • GVHD is induced by 1 ⁇ 10 7 human CD25-depleted PBMC at the same magnitude as by transfer of 1 ⁇ 10 7 human non CD25-depleted PBMC into NOD-Scid mice.
  • Mice receiving CD25-depleted PBMC and an additional administration of HIV gp120 are not protected from development of GVHD and weight loss.
  • Prevention of GVHD by HIV gp120 or a substance which can interfere with HIV-1 gp120-binding depends on regulatory T cells. Lack of weight increase/weight loss is used as a parameter to score GVHD severity. Data see FIG. 5 .
  • FIG. 1 HIV-1 gp120 treatment activates human CD4 + CD25 + Treg cells
  • Grey bars represent proliferation of CD8 + T cells and inactivated syngenic CD3-depleted PBMC, white bars represent the proliferation of CD4 + CD25 + Tregs and the black bars represent the proliferation of CD8 + T cells co-cultured with CD4 + CD25 + Tregs cells and inactivated syngenic CD3-depleted PBMC. Due to inactivation PBMC don't contribute to proliferation of the samples.
  • Tregs don't show a significant proliferation under all conditions (without additional stimulus, or anti-CD3, or HIV-1 gp120 MN, or HIV-1 gp120 LAV, or HIV-1 gp120 CM).
  • CD8 + T cells grey bars
  • CD8 + T cells co-cultured with Treg cells black bars
  • CD8 + T cells grey bars
  • CD8 + T cells co-cultured with Treg cells black bars
  • the effect is anti-CD3 dose-dependent.
  • HIV-1 gp120 MN Upon stimulation with HIV-1 gp120 MN, or HIV-1 gp120 LAV, or HIV-1 gp120 CM CD8 + T cells co-cultured with Treg cells (black bars) in contrast to CD8 + T cells without Tregs (grey bars) reduced proliferation indicating HIV-1 gp120 activated Tregs.
  • HIV-1 gp120 activates Tregs which subsequently exert suppressive activity on CD8 + T cells by reducing their proliferation.
  • the suppressive activity of Treg cells and therefore the reduced proliferation is HIV-1 gp120 dose-dependent.
  • FIG. 2 Cyclic AMP measurement in CD4+CD25+ Treg cells.
  • HIV-1 gp120 treatment augments cytosolic cAMP in human CD4 + CD25 + Treg cells.
  • CD4 + CD25 + regulatory T cells are left untreated ( ⁇ ) or stimulated with an anti-CD3 monoclonal Antibody (OKT-3; 1 ⁇ g/ml) or HIV-1 gp120 (14 ml).
  • OKT-3 anti-CD3 monoclonal Antibody
  • the CD4 + CD25 + regulatory T cells are lysed (1 ⁇ 10 7 /ml) and the cytosolic cAMP concentration of 1 ⁇ 10 6 T cells is assessed using a cAMP-specific ELISA.
  • CD4 + CD25 + regulatory T cells are left untreated ( ⁇ ) or stimulated with an anti-CD3 monoclonal Antibody (OKT-3; 1 ⁇ g/ml) or HIV-1 gp120 (1 ⁇ g/ml).
  • OKT-3 anti-CD3 monoclonal Antibody
  • HIV-1 gp120 1 ⁇ g/ml
  • the CD4 + CD25 + regulatory T cells are lysed (1 ⁇ 10 7 /ml) and the cytosolic cAMP concentration of 1 ⁇ 10 6 T cells is assessed using a cAMP-specific ELISA.
  • Bars represent the amount of cytosolic (intracellular) cyclic AMP (cAMP) in untreated Treg cells or Treg cells treated with different stimuli.
  • the upper bar represent Tregs left untreated ( ⁇ ) and shows the basal level of intracellular cAMP.
  • Tregs are activated and show an increase of intracellular cAMP compared to the untreated control (upper bar) as demonstrated with the second bar.
  • Treatment of Tregs with HIV-1 gp120 also induces an increase of intracellular cAMP as demonstrated with the lower bar. This demonstrated that activation of Treg cells with different stimuli induces an increase of cytosolic cAMP which can be used as readout for activation.
  • FIG. 3 Induction of GVHD by transfer of PBMC into NOD-Scid mice and prevention of GVHD by additional transfer of regulatory T cells (Tregs).
  • NOD-Scid mice Three to six days old NOD-Scid mice are intraperitoneally injected with 1 ⁇ 10 7 human PBMC without (circles) or together with 2.5 ⁇ 10 6 human regulatory T cells (triangles). Control mice (rhombi) did not receive any PBMC. Mice having received PBMC develop a fatal GVHD, do not grow and die. Animals having additionally received regulatory T cells are protected from development of GVHD and develop normally (3 mice per group). Lack of weight increase/weight loss is used as a parameter to score GVHD severity. The diagram shows the relative body weight at different time points after transfer.
  • FIG. 4 Induction of GVHD by transfer of PBMC into NOD-Scid mice and prevention of GVHD by additional injection of HIV gp120.
  • NOD-Scid mice Three to six days old NOD-Scid mice are intraperitoneally injected with 1 ⁇ 10 7 human PBMC without (circles) or together with 5 ⁇ g HIV gp120 (triangles). Control mice (rhombs) did not receive any PBMC. Mice having received PBMC develop a fatal GVHD, do not grow and die. Animals having additionally received HIV gp120 are protected from development of GVHD and develop normally (3 mice per group). Lack of weight increase/weight loss is used as a parameter to score GVHD severity. The diagram shows the relative body weight at different time points after transfer.
  • FIG. 5 Prevention of GVHD in NOD-Scid mice transferred with PBMC by additional injection of HIV gp120 depends on the presence of regulatory T cells.
  • NOD-Scid mice Three to six days old NOD-Scid mice are intraperitoneally injected with 1 ⁇ 10 7 human CD25-depleted PBMC without (circles) or together with 5 ⁇ g HIV gp120 (triangles). Control mice (rhombi) do not receive any PBMC (3 mice per group). Mice having received CD25-depleted PBMC develop a fatal GVHD, do not grow and die. Animals having received 1 ⁇ 10 7 human CD25-depleted PBMC and additionally HIV gp120 are not protected from development of GVHD, also do not grow and die. Lack of weight increase/weight loss is used as a parameter to score GVHD severity. The diagram shows the relative body weight at different time points after transfer.

Abstract

The present invention relates specific activation of a regulatory T cell via a specific CD4 epitope and uses thereof, e.g. for the treatment of an autoimmune disease or an allergy or asthma or graft rejection or tolerance induction.

Description

    PRIORITY CLAIM
  • In accordance with 37 C.F.R. 1.76, a claim of priority is included in an Application Data Sheet filed concurrently herewith. Accordingly, the present invention claims priority as a divisional of U.S. National Phase patent application Ser. No. 12/525,142; entitled “SCREENING METHOD FOR THE IDENTIFICATION OF AGENTS CAPABLE OF ACTIVATING CD4+CD25+REGULATORY T-CELLS THROUGH INTERACTIONS WITH THE HIV-1 GP120 BINDING SITE ON CD4”, filed Oct. 7, 2009; which claims priority to International PCT Patent Application No. PCT/EP2008/051144, entitled “SPECIFIC ACTIVATION OF A REGULATORY T CELL AND ITS USE FOR TREATMENT OF ASTHMA, ALLERGIC DISEASE, AUTOIMMUNE DISEASE, GRAFT REJECTION AND FOR TOLERANCE INDUCTION”, filed Jan. 30, 2008, which claims priority to European Patent Application No. 07101604.2, entitled “SPECIFIC ACTIVATION OF A REGULATORY T CELL AND ITS USE FOR TREATMENT OF ASTHMA, ALLERGIC DISEASE, AUTOIMMUNE DISEASE, GRAFT REJECTION AND FOR TOLERANCE INDUCTION”, filed Feb. 1, 2007 and 07122424.0, entitled “SPECIFIC ACTIVATION OF A REGULATORY T CELL AND ITS USE FOR TREATMENT OF ASTHMA, ALLERGIC DISEASE, AUTOIMMUNE DISEASE, GRAFT REJECTION AND FOR TOLERANCE INDUCTION”, filed Dec. 5, 2007. The contents of the above referenced applications are incorporated herein by reference.
  • INTRODUCTION
  • Asthma, an allergic disease, transplant rejection and an autoimmune disease have one fundamental principle in common, they all are triggered by an imbalanced immune system which reacts hyperactive against a specific exogenic and/or endogenic challenge and therewith contributes significantly to the disease status.
  • It is generally accepted that such aberrations of the immune system have a common pathophysiological mechanism triggered by hyper-responsive effector T cells playing a central role in immune reactivity. Effector T cell-directed immunomodulation therefore is the key to successful treatment of asthma, an autoimmune condition, prevention of graft vs. host disease (GVHD) and prevention of graft rejection.
  • T lymphocytes, designated as regulatory T cells (“Treg cells”) control immune responses by suppressing the effector function of CD4+ T cells and CD8+ T cells (Shevach 2002). Different subsets of Treg cells have been described. These include but are not limited to (i) CD4+CD25+Treg cells—also designated as “naturally occurring Treg cells” (Sakaguchi 2005), (ii) Tr1 (Roncarolo et al. 2001) and (iii) Th3 (Weiner 2001). Tr1 and Th3 are induced in the periphery, whereas CD4+CD25+ Treg cells develop in the thymus and constitute 5-10% of peripheral CD4+ T cells in healthy man. At least in vitro these cells are anergic, produce minimal amounts of cytokines and exert their suppressive effects only upon stimulation and in a strictly cell-contact dependent manner. Tr1 and Th3 exert their suppressive activity by production of IL-10 and TGF-beta, respectively (Shevach 2002).
  • Genetic defects that primarily affect Treg cell development or function should cause autoimmune and inflammatory aberrations. IPEX syndrome (immunodysregulation, polyendo-crinopathy and enteropathy, X-linked), a rare recessive disorder in humans is caused by a mutation in the gene of the transcription factor FOXP3 and subsequent absence of Treg cells. IPEX shows aggressive autoimmunity, severe eczema, elevated IgE levels, eosinophilia and food allergies and early death (Fontenot and Rudensky 2005).
  • Data from the literature show that Treg cells play an important role in asthma and autoimmune diseases and have a potential for treatment of GVHD and therewith transplantation tolerance (Robinson 2004, Sakaguchi 2005).
  • Therefore, attempts have been started to use Treg cells as a therapeutic agent for patients with established autoimmune disease (Horwitz et al. 2003). It is believed that said patients lack sufficient Treg cells or do have impaired Treg cell function resulting in misdirected and uncontrolled effector T cell activity. The previous thinking for solving this problem is administering Treg cells to a said patient. Since Treg cells are rare in peripheral blood clinical application of human Treg cells depends on highly expensive ex vivo expansion of Treg cells (Hoffmann et al. 2004, Horwitz et al. 2003, Tang et al. 2003, Zheng et al. 2004). Bluestone and Tang went one step further: They are trying to solve the problem by not only increasing the amount of Treg cells for therapy but enhancing suppressive activity of Treg cells by activating Treg cells via triggering of the T cell receptor (TCR) by an anti-CD3 antibody (Bluestone and Tang 2004). This approach resembles by far no Treg cell specific activation as anti-CD3 activates all T cell receptor-expressing cells which bears the obstacle that anti-CD3 treatment induces effector T cell function probably leading to uncontrolled proliferation and non-specific pro-inflammatory cytokine production and exaggeration of pathology. To circumvent this unwanted triggering of effector T cells Treg cells have to be highly purified and activated ex vivo with anti-CD3 which again is a highly expensive and laborious procedure. In addition, the absence of a Treg cell specific marker makes it difficult to achieve high purity of Treg cells.
  • Therefore, it is a primary goal to identify a substance which can activate Treg cells specifically without stimulating the immune system any further and therefore would provide a basis for a direct in vivo application without money consuming ex vivo treatments.
  • There is some information in the prior art how Treg cells possibly can be activated, e.g. nonspecifically via CD3 (Thornton and Shevach 1998), or via CD28/B7 pathway (Shevach 2002, Bluestone and Tang 2004, Hunig and Dennehy 2005), or via CD4 (WO04083247). So far all this solutions do not result in a specific Treg cell activation.
  • DESCRIPTION OF THE INVENTION
  • The present invention bases on the new finding that a specific epitope of CD4 triggers activation of Treg cells. Said epitope overlaps with the known HIV-1 gp120-binding site but surprisingly binding to this site causes Treg cell activation. This finding was totally unexpected for the following reasons: Until now Treg cell activation via CD4 was attributed to a different epitope to which the monoclonal antibody BF5 (WO04083247) binds.—Carriere et al., 1995 have investigated that the binding site of BF5 on CD4 is completely independent of the HIV-1 gp120-binding site.—Unexpectedly, despite reports in the literature on direct anergizing and blocking of CD4 on T cell function by HIV-1 gp120 (Diamond et al. 1988), we found an activating property of HIV-1 gp120 on CD4 of Treg cells.
  • The present invention discloses a physiologically active CD4 epitope on a Treg cell which triggers suppressive activity of a Treg cell. The epitope of the present invention has been identified as a region on the human CD4 protein (SEQ ID NO.: 2) spanning amino acid position No. 54 to 84 of SEQ ID NO.:2. Those 31 amino acids are explicitly given in SEQ ID NO.:1.
  • A preferred peptide which is a CD4 fragment according to the present invention is selected from a group consisting of an isolated peptide spanning amino acid No. 1-31 of SEQ ID NO 1, No. 26 to 458 of SEQ ID NO.: 2, No. 26 to 419 of SEQ ID NO.:2, No. 26 to 207 of SEQ ID NO.: 2, No. 26 to 131 of SEQ ID NO.: 2 and No. 46 to 89 of SEQ ID NO.: 2. All said peptides are additionally characterized in that they all do harbor the critical amino acid Phenylalanine at residue 68 of SEQ ID NO.: 2.
  • The finding that the epitope given in SEQ ID NO.:1 is a key to activate a Treg cell provides a basis for several uses, e.g.:
  • Methods for identification of a substance which can activate a regulatory T cell (Treg cell). Such a substance is designated “Treg cell activator” of the present invention which is useful for the treatment of a disease of the invention which is a disease in which increase of activated regulatory T cells (Treg cells) can improve clinical picture wherein such a disease is (i) a non-autoimmune inflammatory disease: asthma, allergic asthma, respiratory allergy, allergic rhinoconjunctivitis, allergic alveolitis, contact allergy, atopic dermatitis, neurodermatitis, food allergy, graft-versus-host disease, non-autoimmune inflammatory bowel disease, acute respiratory distress syndrome, acute inflammatory pancreatitis, burns, wound healing, skin scarring disorders, sarcoidosis, Behcet's disease or Sweet's syndrome; (ii) an autoimmune inflammatory disease: rheumatoid arthritis, rheumatic fever, systemic lupus erythematosus, ulcerative colitis, Crohn's disease, autoimmune inflammatory bowel disease, diabetes type I, gastritis, autoimmune atrophic gastritis, autoimmune hepatitis, Hashimoto's thyroiditis, thyreoiditis, multiple sclerosis, myasthenia gravis, autoimmune haemolytic anemia, Addison's disease, scleroderma, Goodpasture's syndrome, Guillain-Barre syndrome, Graves' disease, glomerulonephritis, psoriasis, pemphigus vulgaris, pemphigoid, vitiligo, idiopathic leukopenia, Sjogren's syndrome or Wegener's granulomatosis; (iii) an inflammatory disease due to organ transplantation; (iv) a bone marrow transplantation; or (v) a disease due to exogenously administered self or exogenously administered non-autologous recombinant polypeptide.
  • A “substance” of the present invention can be used in a method according to the present invention. The meaning of the term substance according to the present invention includes but is not limited to a peptide, a scaffolded peptide, an antibody, a fragment of an antibody, a nucleic acid molecule, a ribozyme, an organic compound or an inorganic compound.
  • In a second aspect the new epitope serves as the basis for the synthesis of a new tool which can be used e.g. in a competition assay or screening assay according to the present invention for determining whether a substance can activate a regulatory T cell (Treg cell) via interaction with the epitope as set forth in SEQ ID NO.:1. Such a tool is a peptide of the present invention which is an isolated peptide spanning amino acid No. 1-31 of SEQ ID NO 1, or No. 26 to 458 of SEQ ID NO.: 2, or No. 26 to 419 of SEQ ID NO.:2, or No. 26 to 207 of SEQ ID NO.: 2, or No. 26 to 131 of SEQ ID NO.: 2 or No. 46 to 89 of SEQ ID NO.: 2, or is an isolated peptide spanning amino acid No. 1 to 31 of SEQ ID NO.: 1 and having additional up-stream and/or downstream amino acids with the prerequisite that the additional amino acids do not hinder binding of a substance to the amino acid stretch as set forth in SEQ ID NO.: 1. A preferred peptide according to the present invention consist of the peptide as set forth in SEQ ID NO.:1 and additionally consist of an additional up-stream amino acid or amino acid stretch which is selected from a group consisting of the amino acid or amino acid stretch as set forth in SEQ ID NO.: 2 at position 53, at position 52-53, at position 51-53, at position 50-53, at position 49-53, at position 48-53, at position 47-53, at position 46-53, at position 45-53, at position 44-53, at position 43-53, at position 42-53, at position 41-53, at position 40-53, at position 39-53, at position 38-53, at position 37-53, at position 36-53, at position 35-53, at position 34-53, at position 33-53, at position 32-53, at position 31-53, at position 30-53, at position 29-53, at position 28-53, at position 27-53, at position 26-53, at position 25-53, at position 24-53, at position 23-53, at position 22-53, at position 21-53, at position 20-53, at position 19-53, at position 18-53, at position 17-53, at position 16-53, at position 15-53, at position 14-53, at position 13-53, at position 12-53, at position 11-53, at position 10-53, at position 9-53, at position 8-53, at position 7-53, at position 6-53, at position 5-53, at position 4-53, at position 3-53, at position 2-53, and at position 1-53.
  • A further preferred peptide according to the present invention consists of the peptide which is mentioned as “preferred peptide” in the paragraph above and additionally consists of at least one additional downstream amino acid as given in SEQ ID NO.: 2 at position 85, or additionally consists of amino acids as given in SEQ ID NO.:2 at position 85 to n, wherein n is an integer between 86-458, i.e. position 85 to 86, 85 to 87, 85 to 88, 85 to 89, 85 to 90, 85 to 91, 85 to 92, 85 to 93, 85 to 94, 85 to 95, 85 to 96, 85 to 97, 85 to 98, 85 to 99, 85 to 100, 85 to 101, 85 to 102, 85 to 103, 85 to 104, 85 to 105, 85 to 106, 85 to 107, 85 to 108, 85 to 109, 85 to 110, 85 to 111, 85 to 112, 85 to 113, 85 to 114, 85 to 115, 85 to 116, 85 to 117, 85 to 118, 85 to 119, 85 to 120, 85 to 121, 85 to 122, 85 to 123, 85 to 124, 85 to 125, 85 to 126, 85 to 127, 85 to 128, 85 to 129, 85 to 130, 85 to 131, 85 to 132, 85 to 133, 85 to 134, 85 to 135, 85 to 136, 85 to 137, 85 to 138, 85 to 139, 85 to 140, 85 to 141, 85 to 142, 85 to 143, 85 to 144, 85 to 145, 85 to 146, 85 to 147, 85 to 148, 85 to 149, 85 to 150, 85 to 151, 85 to 152, 85 to 153, 85 to 154, 85 to 155, 85 to 156, 85 to 157, 85 to 158, 85 to 159, 85 to 160, 85 to 161, 85 to 162, 85 to 163, 85 to 164, 85 to 165, 85 to 166, 85 to 167, 85 to 168, 85 to 169, 85 to 170, 85 to 171, 85 to 172, 85 to 173, 85 to 174, 85 to 175, 85 to 176, 85 to 177, 85 to 178, 85 to 179, 85 to 180, 85 to 181, 85 to 182, 85 to 183, 85 to 184, 85 to 185, 85 to 186, 85 to 187, 85 to 188, 85 to 189, 85 to 190, 85 to 191, 85 to 192, 85 to 193, 85 to 194, 85 to 195, 85 to 196, 85 to 197, 85 to 198, 85 to 199, 85 to 200, 85 to 201, 85 to 202, 85 to 203, 85 to 204, 85 to 205, 85 to 206, 85 to 207, 85 to 208, 85 to 209, 85 to 210, 85 to 211, 85 to 212, 85 to 213, 85 to 214, 85 to 215, 85 to 216, 85 to 217, 85 to 218, 85 to 219, 85 to 220, 85 to 221, 85 to 222, 85 to 223, 85 to 224, 85 to 225, 85 to 226, 85 to 227, 85 to 228, 85 to 229, 85 to 230, 85 to 231, 85 to 232, 85 to 233, 85 to 234, 85 to 235, 85 to 236, 85 to 237, 85 to 238, 85 to 239, 85 to 240, 85 to 241, 85 to 242, 85 to 243, 85 to 244, 85 to 245, 85 to 246, 85 to 247, 85 to 248, 85 to 249, 85 to 250, 85 to 251, 85 to 252, 85 to 253, 85 to 254, 85 to 255, 85 to 256, 85 to 257, 85 to 258, 85 to 259, 85 to 260, 85 to 261, 85 to 262, 85 to 263, 85 to 264, 85 to 265, 85 to 266, 85 to 267, 85 to 268, 85 to 269, 85 to 270, 85 to 271, 85 to 272, 85 to 273, 85 to 274, 85 to 275, 85 to 276, 85 to 277, 85 to 278, 85 to 279, 85 to 280, 85 to 281, 85 to 282, 85 to 283, 85 to 284, 85 to 285, 85 to 286, 85 to 287, 85 to 288, 85 to 289, 85 to 290, 85 to 291, 85 to 292, 85 to 293, 85 to 294, 85 to 295, 85 to 296, 85 to 297, 85 to 298, 85 to 299, 85 to 300, 85 to 301, 85 to 302, 85 to 303, 85 to 304, 85 to 305, 85 to 306, 85 to 307, 85 to 308, 85 to 309, 85 to 310, 85 to 311, 85 to 312, 85 to 313, 85 to 314, 85 to 315, 85 to 316, 85 to 317, 85 to 318, 85 to 319, 85 to 320, 85 to 321, 85 to 322, 85 to 323, 85 to 324, 85 to 325, 85 to 326, 85 to 327, 85 to 328, 85 to 329, 85 to 330, 85 to 331, 85 to 332, 85 to 333, 85 to 334, 85 to 335, 85 to 336, 85 to 337, 85 to 338, 85 to 339, 85 to 340, 85 to 341, 85 to 342, 85 to 343, 85 to 344, 85 to 345, 85 to 346, 85 to 347, 85 to 348, 85 to 349, 85 to 350, 85 to 351, 85 to 352, 85 to 353, 85 to 354, 85 to 355, 85 to 356, 85 to 357, 85 to 358, 85 to 359, 85 to 360, 85 to 361, 85 to 362, 85 to 363, 85 to 364, 85 to 365, 85 to 366, 85 to 367, 85 to 368, 85 to 369, 85 to 370, 85 to 371, 85 to 372, 85 to 373, 85 to 374, 85 to 375, 85 to 376, 85 to 377, 85 to 378, 85 to 379, 85 to 380, 85 to 381, 85 to 382, 85 to 383, 85 to 384, 85 to 385, 85 to 386, 85 to 387, 85 to 388, 85 to 389, 85 to 390, 85 to 391, 85 to 392, 85 to 393, 85 to 394, 85 to 395, 85 to 396, 85 to 397, 85 to 398, 85 to 399, 85 to 400, 85 to 401, 85 to 402, 85 to 403, 85 to 404, 85 to 405, 85 to 406, 85 to 407, 85 to 408, 85 to 409, 85 to 410, 85 to 411, 85 to 412, 85 to 413, 85 to 414, 85 to 415, 85 to 416, 85 to 417, 85 to 418, 85 to 419, 85 to 420, 85 to 421, 85 to 422, 85 to 423, 85 to 424, 85 to 425, 85 to 426, 85 to 427, 85 to 428, 85 to 429, 85 to 430, 85 to 431, 85 to 432, 85 to 433, 85 to 434, 85 to 435, 85 to 436, 85 to 437, 85 to 438, 85 to 439, 85 to 440, 85 to 441, 85 to 442, 85 to 443, 85 to 444, 85 to 445, 85 to 446, 85 to 447, 85 to 448, 85 to 449, 85 to 450, 85 to 451, 85 to 452, 85 to 453, 85 to 454, 85 to 455, 85 to 456, 85 to 457, or 85 to 458.
  • Additionally, the finding that the epitope given in SEQ ID NO.:1 is a key to activate a Treg cell is a link between two different up to now unrelated technical fields, namely that of (a) HIV-1 related diseases with (b) diseases according to the present invention e.g. autoimmune disease, allergy, asthma, graft rejection and a diseases due to lacking immunotolerance caused by organ transplantation or by therapeutical administration of a non-self or self biological entity to a human in need thereof and therewith allows a bundle of new uses as explained in the following:
  • The epitope given in SEQ ID NO.:1 is not only a further epitope which can be used to activate Treg cells. HIV-1 gp120 interacts with CD4 of T cells and therewith enables virus entry into a CD4+ cell (Klatzmann et al. 1984). The finding that epitope given in SEQ ID NO.:1 harbors the high affinity binding site on CD4 to which human immune deficiency virus 1 (HIV-1) glycoprotein gp120 binds (Jameson et al. 1988, Arthos et al. 1989) offers a further advantage. It provides the basis to bring together the findings of two different unrelated technical fields that of i.e. HIV-1 related diseases with diseases according to the present invention.
  • To alleviate the worldwide HIV-1 problem many efforts have been made to identify a substance which is able to inhibit HIV-1 entry into a CD4+ cell. As a result thereof, so called HIV-1 attachment or entry inhibitors are known in the art.
  • The keyhole which allows HIV-1 to enter the cell can be used as the keyhole to activate Treg cells. Therefore, substances known in the art to interfere with HIV-1 attachment and cell entry (Markovic and Clouse 2004, Castagna et al. 2005), like e.g. HIV-1 gp120 itself, derivatives thereof, peptidomimetics, antibodies, aptamers or any low molecular weight (LMW) compound directed against the binding site of HIV-1 gp120 on CD4 could possibly be useful to activate a Treg cell and therewith can be useful for the treatment of a disease according to the present invention (HIV-1 gp120 is well-known in the art and its amino acid sequence as well as the respective gene has been published since years (Muesing et al. 1985, Starcich et al. 1986, Jeffs et al. 1996). Additionally, methods for producing HIV-1 gp120 are known (Lasky et al. 1986, Leonard et al. 1990, Culp et al. 1991, Jeffs et al. 1996).
  • A substance which can interfere with HIV-1 attachment and/or cell entry is commonly named HIV-1 attachment inhibitor or entry inhibitor. Such a substance, do either bind to (i) HIV-1, or (ii) to CD4, or (iii) HIV-1 and CD4 or (iv) co-receptor e.g. CCR5 or CXCR4. Such an inhibitor can be according to the present invention useful for the treatment of diseases according to the present invention like e.g. an autoimmune disease, an allergy, asthma or GVHD if it exerts property (ii) or (iii) and activates a Treg cell. To determine whether such an inhibitor can be useful for a said disease the present invention discloses several assays which allow to determine whether a substance identified in the technical field of HIV-1 research to interfere with HIV-1 attachment and/or cell entry can be useful in the other above-mentioned technical fields like that of autoimmune diseases or allergies or asthma or organ transplantation. Therefore, the present invention teaches a short cut for identifying a substance which can be useful for the treatment of a disease according to the present invention.
  • Identification of a substance which can be used as a medicament in a specific disease usually depends on resource consuming high through put screenings (HTS). Determination whether a substance can activate a Treg cell currently depends on a cellular assay comprising a Treg cell. Since Treg cells can only be provided in small amounts large screening campaigns or even an HTS therewith are not feasible today. The teaching of the present invention allows to circumvent this obstacles since the present invention allows to pre-select substances which possible can activate a Treg cell. According to the present invention an appropriate pre-selected substance is (i) proven to interact at least with epitope (SEQ ID NO.:1), and/or is (ii) known from HIV-1 research as HIV-1 attachment inhibitor or entry inhibitor or synthetic mimetics of the CD4 binding site of HIV-1 gp120.
  • It is state of the art to determine whether a substance can interact with a given peptide and therewith with a given epitope even in an HTS format. Concerning the present invention this can be performed for example in an in vitro competition type assay comprising a peptide spanning at least amino acids as set forth in SEQ ID NO.: 1 mixed with an unlabeled substance to be tested and subsequently with a labeled substance which is known to bind the peptide (e.g. HIV-1 gp120) under conditions which allow binding of the peptide with the labeled substance. A substance which interact with the peptide will compete with the labeled substance and is identifiable by a rendered readout, which can be performed e.g. by measuring the bound or free labeled substance.
  • Such a type of assay for determining whether a substance can interact with a specific peptide, i.e. epitope is not restricted to in vitro assays since cellular assays for achieving such an information on a substance or other in vitro formats are well known in the art and broadly used.
  • In one embodiment the present invention concerns a method for determining whether a substance which can interfere with the interaction of HIV-1 gp120 with CD4 can be useful for positively influence a disease in which increase of activated regulatory T cells (Treg cells) can improve clinical picture comprising:
      • (a) providing a solution comprising a Treg cell, wherein a Treg cell is preferably a CD44CD25+ Treg cell or a Tr1 cell or a Th3 cell. Said solution does more preferably comprise additionally an inactivated syngenic CD3-depleted PBMC (peripheral blood mononuclear cell which has preferably been inactivated via irradiation or via mitomycin C) or a dendritic cell (DC) and an allogeneic CD8+ T cell or an allogeneic CD4+ T cell,
      • (b) adding a substance to be tested under conditions which allow interaction of the substance with a Treg cell,
      • (c) measuring whether a Treg cell has been activated, wherein an activated Treg cell identifies the substance as a Treg cell activator.
      • Said measuring can be performed using a suitable read-out system such as:
        • (i) measuring whether a CD8+ T cell has been suppressed—which preferably can be determined by measuring inhibited proliferation of the CD8+ T cell or by measuring reduced CD25 expression of the CD8+ T cell, or by measuring inhibited cytokine production of the CD8+ T cell wherein a suitable cytokine is IFNγ or IL2, or TNFα—wherein a suppressed CD8+ T cell identifies an activated Treg cell and therewith identifies the substance as a Treg cell activator, or by (ii) measuring whether a CD4+ T cell has been suppressed—which preferably can be determined by measuring inhibited proliferation of the CD4+ T cell or by measuring reduced CD25 expression of the CD4+ T cell, or by measuring inhibited cytokine production of the CD4+ T cell wherein a suitable cytokine is IFNγ or IL2, or TNFα—wherein a suppressed CD4+ T cell identifies an activated Treg cell and therewith identifies the substance as a Treg cell activator, or by (iii) measuring the amount of intracellular cAMP (i.e. cytosolic cAMP) and wherein an increased amount of intracellular cAMP is indicative for an activated Treg cell and therewith identifies the substance as a Treg cell activator.
  • A disease according to the method above in which increase of activated regulatory T cells (Treg cells) can improve clinical picture is (i) a non-autoimmune inflammatory disease: asthma, allergic asthma, respiratory allergy, allergic rhinoconjunctivitis, allergic alveolitis, contact allergy, atopic dermatitis, neurodermatitis, food allergy, graft-versus-host disease, non-autoimmune inflammatory bowel disease, acute respiratory distress syndrome, acute inflammatory pancreatitis, burns, wound healing, skin scarring disorders, sarcoidosis, Behcet's disease or Sweet's syndrome; (ii) an autoimmune inflammatory disease: rheumatoid arthritis, rheumatic fever, systemic lupus erythematosus, ulcerative colitis, Crohn's disease, autoimmune inflammatory bowel disease, diabetes type I, gastritis, autoimmune atrophic gastritis, autoimmune hepatitis, Hashimoto's thyroiditis, thyreoiditis, multiple sclerosis, myasthenia gravis, autoimmune haemolytic anemia, Addison's disease, scleroderma, Goodpasture's syndrome, Guillain-Barre syndrome, Graves' disease, glomerulonephritis, psoriasis, pemphigus vulgaris, pemphigoid, vitiligo, idiopathic leukopenia, Sjogren's syndrome or Wegener's granulomatosis; (iii) an inflammatory disease due to organ transplantation; (iv) a bone marrow transplantation; or (v) a disease due to exogenously administered self or exogenously administered non-autologous recombinant polypeptide.
  • As the readout systems as mentioned in (i) and (ii) above are methods in which several steps have to be performed a specific new test system has been invented to determine in only one step whether a Treg cell has been activated which is mentioned in (iii) above. Basing on the surprising finding that the activation status of a Treg cell strongly correlates with the amount of intracellular cAMP the present invention provides for a specific method for determining whether a Treg cell has been activated which comprises:
      • (a) providing a first solution comprising a Treg cell,
      • (b) providing a second solution comprising a Treg cell,
      • (c) manipulation the first solution by adding at least a test substance,
      • (d) determining the amount of intracellular cAMP of the first and the second solution
      • wherein an increased amount of intracellular cAMP of the first solution is indicative for an activated Treg cell.
  • In a more preferred method the solution of (a) does not only contain a Treg cell but also an inactivated syngenic CD3-depleted PBMC (peripheral blood mononuclear cell) or a dendritic cell (DC) and an allogeneic CD8+ T cell or an allogeneic CD4+ T cell. These cells when combined in one solution with a activated Treg cell do additionally increase intracellular cAMP amount and therewith lead to a more sensitive readout system.
  • In a further embodiment the present invention concerns a method for determining whether a substance can activate a Treg cell via interaction with the HIV-1 gp120-binding site of CD4, comprising:
      • (a) pre-selecting a substance which can interact with the HIV-1 gp120-binding site of CD4 (for a method for pre-selection please see below),
      • (b) providing a solution comprising a Treg cell wherein a Treg cell is preferably a CD4+CD25+ Treg cell or a Tr1 cell or a Th3 cell. Said solution does more preferably comprise additionally an inactivated syngenic CD3-depleted PBMC (peripheral blood mononuclear cell) which has preferably been inactivated via irradiation or via mitomycin C) or a dendritic cell (DC) and an allogeneic CD8+ T cell or an allogeneic CD4+ T cell,
      • (c) adding a pre-selected substance according to (a) under conditions which allow interaction of the substance with a Treg cell,
      • (d) measuring whether a Treg cell has been activated, wherein an activated Treg cell identifies the substance as a Treg cell activator. Said measuring can be performed using a suitable read-out system such as:
        • (i) measuring whether a CD8+ T cell has been suppressed—which preferably can be determined by measuring inhibited proliferation of the CD8+ T cell or by measuring reduced CD25 expression of the CD8+ T cell, or by measuring inhibited cytokine production of the CD8+ T cell wherein a suitable cytokine is IFNγ or IL2, or TNFα—wherein a suppressed CD8+ T cell identifies an activated Treg cell and therewith identifies the substance as a Treg cell activator, or by
        • (ii) measuring whether a CD4+ T cell has been suppressed—which preferably can be determined by measuring inhibited proliferation of the CD4+ T cell or by measuring reduced CD25 expression of the CD4+ T cell, or by measuring inhibited cytokine production of the CD4+ T cell wherein a suitable cytokine is IFNγ or IL2, or TNFα—wherein a suppressed CD4+ T cell identifies an activated Treg cell and therewith identifies the substance as a Treg cell activator, or by
      • (iii) measuring the amount of intracellular cAMP and wherein an increased amount of intracellular cAMP is indicative for an activated Treg cell and therewith identifies the substance as a Treg cell activator.
  • The present invention also provides for a method which allow to identify a substance which can interact with the HIV-1 gp120-binding site of CD4 and therewith can be used for pre-selecting a substance which can interact with the HIV-1 gp120-binding site of CD4.
  • The method comprises:
      • (a) providing a first solution comprising CD4,
      • (b) providing a second solution comprising CD4,
      • (c) adding to the first solution a substance to be tested and HIV-1 gp120 under conditions which allow binding of HIV-1 gp120 with CD4,
      • (d) adding to the second solution HIV-1 gp120 under conditions like (c) allowing binding of HIV-1 gp120 with CD4,
      • (e) measuring in the first and in the second solution whether the HIV-1 gp120 has bound to the CD4 wherein a reduced amount of bound HIV-1 gp120 in the first solution indicates that the substance can interact with HIV-1 gp120-binding site of CD4.
  • This pre-selecting method can additionally be performed according to the present invention if instead of CD4 a peptide is used which comprises the amino acid stretch as set forth in SEQ ID NO.1 or instead of CD4 a peptide is used as set forth in SEQ ID NO.:1. In a more preferred method according to the present invention instead of HIV-1 gp120 a peptide is used selected from a group consisting of a peptide comprising the amino acid sequence as set forth in SEQ ID NO.: 3, SEQ ID NO.: 4, SEQ ID NO.: 5, SEQ ID NO.: 6, SEQ ID NO.: 7, SEQ ID NO.: 8, SEQ ID NO.: 9, and SEQ ID NO.: 10 or a peptide is used selected from a group consisting of a peptide consisting of the amino acid sequence as set forth in SEQ ID NO.: 3, SEQ ID NO.: 4, SEQ ID NO.: 5, SEQ ID NO.: 6, SEQ ID NO.: 7, SEQ ID NO.: 8, SEQ ID NO.: 9, and SEQ ID NO.: 10.
  • In a further embodiment the present invention concerns a method for determining whether a substance can activate a regulatory T cell (Treg) via interaction with the epitope as set forth in SEQ ID NO: 1 comprising:
      • (a) providing a first solution comprising a Treg cell, wherein a Treg cell is preferably a CD4+CD25+ Treg cell or a Tr1 cell or a Th3 cell. Said solution does more preferably comprise additionally an inactivated syngenic CD3-depleted PBMC (peripheral blood mononuclear cell which has preferably been inactivated via irradiation or via mitomycin C) or a dendritic cell (DC) and an allogeneic CD8+ T cell or an allogeneic CD4+ T cell,
      • (b) adding a substance to be tested under conditions which allow interaction of the substance with a Treg cell,
      • (c) measuring whether a Treg cell of the first solution has been activated,
      • (d) providing a second solution comprising a Treg cell, wherein a Treg cell is preferably a CD4+CD25+Treg cell or a Tr1 cell or a Th3 cell. Said solution does more preferably comprise additionally an inactivated syngenic CD3-depleted PBMC (peripheral blood mononuclear cell which has preferably been inactivated via irradiation or via mitomycin C) or a dendritic cell (DC) and an allogeneic CD8+ T cell or an allogeneic CD4+ T cell, (e) adding the substance to be tested and a peptide comprising an amino acid sequence as set forth in SEQ ID NO: 1 under conditions like (b),
      • (f) measuring whether a Treg cell of the second solution has been activated,
      • Said measuring can be performed using a suitable read-out system such as:
        • (i) measuring whether a CD8+ T cell has been suppressed—which preferably can be determined by measuring inhibited proliferation of the CD8+ T cell or by measuring reduced CD25 expression of the CD8+ T cell, or by measuring inhibited cytokine production of the CD8+ T cell wherein a suitable cytokine is IFNγ or IL2, or TNFα wherein a suppressed CD8+ T cell identifies an activated Treg cell and therewith identifies the substance as a Treg cell activator, or by
        • (ii) measuring whether a CD4+ T cell has been suppressed—which preferably can be determined by measuring inhibited proliferation of the CD4+ T cell or by measuring reduced CD25 expression of the CD4+ T cell, or by measuring inhibited cytokine production of the CD4+ T cell wherein a suitable cytokine is IFN-γ or IL2, or TNFα wherein a suppressed CD4+ T cell identifies an activated Treg cell and therewith identifies the substance as a Treg cell activator, or by
        • (iii) measuring the amount of intracellular cAMP and wherein an increased amount of intracellular cAMP is indicative for an activated Treg cell and therewith identifies the substance as a Treg cell activator;
        • (g) comparing results obtained from (c) with those obtained from (0 wherein a reduced activation of (0 identifies the substance as a Treg cell activator which activates a Treg cell via interaction with the epitope as set forth in SEQ ID NO: 1.
  • In a more preferred method in step (e) of above mentioned method the peptide used is selected from a group consisting of an isolated peptide spanning amino acid No. 1-31 of SEQ ID NO 1, No. 26 to 458 of SEQ ID NO.: 2, No. 26 to 419 of SEQ ID NO.:2, No. 26 to 207 of SEQ ID NO.: 2, No. 26 to 131 of SEQ ID NO.: 2 and No. 46 to 89 of SEQ ID NO.: 2 or the peptide used is an isolated peptide spanning amino acid No. 1 to 31 of SEQ ID NO.: 1 and having additional up-stream and/or downstream amino acids with the prerequisite that the additional amino acids do not hinder binding of a substance to the amino acid stretch as set forth in SEQ ID NO.:1. Said additional up-stream amino acid or amino acid stretch is preferably selected from a group consisting of the amino acid or amino acid stretch as set forth in SEQ ID NO.: 2 at position 53, at position 52-53, at position 51-53, at position 50-53, at position 49-53, at position 48-53, at position 47-53, at position 46-53, at position 45-53, at position 44-53, at position 43-53, at position 42-53, at position 41-53, at position 40-53, at position 39-53, at position 38-53, at position 37-53, at position 36-53, at position 35-53, at position 34-53, at position 33-53, at position 32-53, at position 31-53, at position 30-53, at position 29-53, at position 28-53, at position 27-53, at position 26-53, at position 25-53, at position 24-53, at position 23-53, at position 22-53, at position 21-53, at position 20-53, at position 19-53, at position 18-53, at position 17-53, at position 16-53, at position 15-53, at position 14-53, at position 13-53, at position 12-53, at position 11-53, at position 10-53, at position 9-53, at position 8-53, at position 7-53, at position 6-53, at position 5-53, at position 4-53, at position 3-53, at position 2-53, and at position 1-53. A more preferred peptide does additionally comprise downstream one or more amino acids as set forth in SEQ ID NO.: 2 at position 85, or at position 85 to n, wherein n is an integer between 86-458, i.e. position 85 to 86, 85 to 87, 85 to 88, 85 to 89, 85 to 90, 85 to 91, 85 to 92, 85 to 93, 85 to 94, 85 to 95, 85 to 96, 85 to 97, 85 to 98, 85 to 99, 85 to 100, 85 to 101, 85 to 102, 85 to 103, 85 to 104, 85 to 105, 85 to 106, 85 to 107, 85 to 108, 85 to 109, 85 to 110, 85 to 111, 85 to 112, 85 to 113, 85 to 114, 85 to 115, 85 to 116, 85 to 117, 85 to 118, 85 to 119, 85 to 120, 85 to 121, 85 to 122, 85 to 123, 85 to 124, 85 to 125, 85 to 126, 85 to 127, 85 to 128, 85 to 129, 85 to 130, 85 to 131, 85 to 132, 85 to 133, 85 to 134, 85 to 135, 85 to 136, 85 to 137, 85 to 138, 85 to 139, 85 to 140, 85 to 141, 85 to 142, 85 to 143, 85 to 144, 85 to 145, 85 to 146, 85 to 147, 85 to 148, 85 to 149, 85 to 150, 85 to 151, 85 to 152, 85 to 153, 85 to 154, 85 to 155, 85 to 156, 85 to 157, 85 to 158, 85 to 159, 85 to 160, 85 to 161, 85 to 162, 85 to 163, 85 to 164, 85 to 165, 85 to 166, 85 to 167, 85 to 168, 85 to 169, 85 to 170, 85 to 171, 85 to 172, 85 to 173, 85 to 174, 85 to 175, 85 to 176, 85 to 177, 85 to 178, 85 to 179, 85 to 180, 85 to 181, 85 to 182, 85 to 183, 85 to 184, 85 to 185, 85 to 186, 85 to 187, 85 to 188, 85 to 189, 85 to 190, 85 to 191, 85 to 192, 85 to 193, 85 to 194, 85 to 195, 85 to 196, 85 to 197, 85 to 198, 85 to 199, 85 to 200, 85 to 201, 85 to 202, 85 to 203, 85 to 204, 85 to 205, 85 to 206, 85 to 207, 85 to 208, 85 to 209, 85 to 210, 85 to 211, 85 to 212, 85 to 213, 85 to 214, 85 to 215, 85 to 216, 85 to 217, 85 to 218, 85 to 219, 85 to 220, 85 to 221, 85 to 222, 85 to 223, 85 to 224, 85 to 225, 85 to 226, 85 to 227, 85 to 228, 85 to 229, 85 to 230, 85 to 231, 85 to 232, 85 to 233, 85 to 234, 85 to 235, 85 to 236, 85 to 237, 85 to 238, 85 to 239, 85 to 240, 85 to 241, 85 to 242, 85 to 243, 85 to 244, 85 to 245, 85 to 246, 85 to 247, 85 to 248, 85 to 249, 85 to 250, 85 to 251, 85 to 252, 85 to 253, 85 to 254, 85 to 255, 85 to 256, 85 to 257, 85 to 258, 85 to 259, 85 to 260, 85 to 261, 85 to 262, 85 to 263, 85 to 264, 85 to 265, 85 to 266, 85 to 267, 85 to 268, 85 to 269, 85 to 270, 85 to 271, 85 to 272, 85 to 273, 85 to 274, 85 to 275, 85 to 276, 85 to 277, 85 to 278, 85 to 279, 85 to 280, 85 to 281, 85 to 282, 85 to 283, 85 to 284, 85 to 285, 85 to 286, 85 to 287, 85 to 288, 85 to 289, 85 to 290, 85 to 291, 85 to 292, 85 to 293, 85 to 294, 85 to 295, 85 to 296, 85 to 297, 85 to 298, 85 to 299, 85 to 300, 85 to 301, 85 to 302, 85 to 303, 85 to 304, 85 to 305, 85 to 306, 85 to 307, 85 to 308, 85 to 309, 85 to 310, 85 to 311, 85 to 312, 85 to 313, 85 to 314, 85 to 315, 85 to 316, 85 to 317, 85 to 318, 85 to 319, 85 to 320, 85 to 321, 85 to 322, 85 to 323, 85 to 324, 85 to 325, 85 to 326, 85 to 327, 85 to 328, 85 to 329, 85 to 330, 85 to 331, 85 to 332, 85 to 333, 85 to 334, 85 to 335, 85 to 336, 85 to 337, 85 to 338, 85 to 339, 85 to 340, 85 to 341, 85 to 342, 85 to 343, 85 to 344, 85 to 345, 85 to 346, 85 to 347, 85 to 348, 85 to 349, 85 to 350, 85 to 351, 85 to 352, 85 to 353, 85 to 354, 85 to 355, 85 to 356, 85 to 357, 85 to 358, 85 to 359, 85 to 360, 85 to 361, 85 to 362, 85 to 363, 85 to 364, 85 to 365, 85 to 366, 85 to 367, 85 to 368, 85 to 369, 85 to 370, 85 to 371, 85 to 372, 85 to 373, 85 to 374, 85 to 375, 85 to 376, 85 to 377, 85 to 378, 85 to 379, 85 to 380, 85 to 381, 85 to 382, 85 to 383, 85 to 384, 85 to 385, 85 to 386, 85 to 387, 85 to 388, 85 to 389, 85 to 390, 85 to 391, 85 to 392, 85 to 393, 85 to 394, 85 to 395, 85 to 396, 85 to 397, 85 to 398, 85 to 399, 85 to 400, 85 to 401, 85 to 402, 85 to 403, 85 to 404, 85 to 405, 85 to 406, 85 to 407, 85 to 408, 85 to 409, 85 to 410, 85 to 411, 85 to 412, 85 to 413, 85 to 414, 85 to 415, 85 to 416, 85 to 417, 85 to 418, 85 to 419, 85 to 420, 85 to 421, 85 to 422, 85 to 423, 85 to 424, 85 to 425, 85 to 426, 85 to 427, 85 to 428, 85 to 429, 85 to 430, 85 to 431, 85 to 432, 85 to 433, 85 to 434, 85 to 435, 85 to 436, 85 to 437, 85 to 438, 85 to 439, 85 to 440, 85 to 441, 85 to 442, 85 to 443, 85 to 444, 85 to 445, 85 to 446, 85 to 447, 85 to 448, 85 to 449, 85 to 450, 85 to 451, 85 to 452, 85 to 453, 85 to 454, 85 to 455, 85 to 456, 85 to 457, or 85 to 458.
  • In the context of the present invention new and known substances have been examined in the assays according to the present invention. As a result thereof substances could be identified which can act as a Treg cell activator of the present invention, i.e. such a substance is able to activate a Treg cell via interaction with the Treg cell epitope as set forth in SEQ ID NO:1 which could be proven in vivo. The present invention therefore provides for Treg cell activators.
  • Disclosed are Treg cell activators according to the invention which are structurally a peptide or a polypeptide, preferably an antibody of a binding fragment thereof or a scaffolded peptide.
  • The present invention further concerns a new antibody or a binding fragment thereof capable of binding to the peptide as set forth in SEQ ID NO.:1 with the proviso that the antibody or the antibody fragment is not OKT4A, OKT4D4, OKTcdr4a and not Leu3. Said disclaimed antibodies all relate to a totally different technical filed, namely HIV-1 research which is not related to the activation of Treg cells and disease according to the present invention. Even the present invention combines for the first time the technical field of (a) HIV-1 related diseases with (b) diseases according to the present invention e.g. autoimmune disease, allergy, asthma, graft rejection and a diseases due to lacking immunotolerance caused by organ transplantation or by therapeutical administration of a non-self or self biological entity to a human in need thereof.
  • A further preferred Treg cell activator peptide is a peptide selected from a group consisting of a peptide comprising the amino acid sequence as set forth in SEQ ID NO.: 3, SEQ ID NO.: 4, SEQ ID NO.: 5, SEQ ID NO.: 6, SEQ ID NO.: 7, SEQ ID NO.: 8, SEQ ID NO.: 9, and SEQ ID NO.: 10, or is a peptide selected from a group consisting of a peptide consisting of the amino acid sequence as set forth in SEQ ID NO.: 3, SEQ ID NO.: 4, SEQ ID NO.: 5, SEQ ID NO.: 6, SEQ ID NO.: 7, SEQ ID NO.: 8, SEQ ID NO.: 9, and SEQ ID NO.: 10.
  • A more preferred Treg cell activator polypeptide is selected from a group consisting of: HIV-1 gp120, NSC 13778 which chemical structure is disclosed in Yang et al. 2005 on page 6124 in FIG. 1, peptide 2 which presents three HIV-1 gp120 fragments bound together through comformationally flexible scaffolds which chemical structure is disclosed in Franke et al. 2007 on page 4 at the bottom of the right column, monoclonal antibody OKT4A which binds to the HIV-1 gp120-binding region of CD4 as disclosed in Mizukami et al. 1988 on page 9273 right column line 19, monoclonal antibody OKT4D which binds to the HIV-1 gp120-binding region of CD4 as disclosed in Mizukami et al. 1988 on page 9273 right column line 19, monoclonal antibody OKTcdr4a which derives from the murine OKT4a as disclosed in Moreland et al. 1998 on page 222 right column line 1, monoclonal antibody Leu3 which binds to epitope overlapping the HIV-1 gp120-binding site of CD4 as disclosed in Lohmann et al. 1992 on page 3248 left column line 7, and monoclonal antibody MAX.12H5 which binds to the CDR2-like region of CD4 as disclosed in Repke et al. 1992 on page 1809 abstract line 11 and on page 1812 left column line 37.
  • Each of the above mentioned Treg cell activators according to the present invention can be used as a medicament and for the preparation of a medicament for the treatment of a disease selected from a group consisting of (i) a non-autoimmune inflammatory disease: asthma, allergic asthma, respiratory allergy, allergic rhinoconjunctivitis, allergic alveolitis, contact allergy, atopic dermatitis, neurodermatitis, food allergy, graft-versus-host disease, non-autoimmune inflammatory bowel disease, acute respiratory distress syndrome, acute inflammatory pancreatitis, burns, wound healing, skin scarring disorders, sarcoidosis, Behcet's disease, Sweet's syndrome; (ii) an autoimmune inflammatory disease: rheumatoid arthritis, rheumatic fever, systemic lupus erythematosus, ulcerative colitis, Crohn's disease, autoimmune inflammatory bowel disease, diabetes type I, gastritis, autoimmune atrophic gastritis, autoimmune hepatitis, Hashimoto's thyroiditis, thyreoiditis, multiple sclerosis, myasthenia gravis, autoimmune haemolytic anemia, Addison's disease, scleroderma, Goodpasture's syndrome, Guillain-Barre syndrome, Graves' disease, glomerulonephritis, psoriasis, pemphigus vulgaris, pemphigoid, vitiligo, idiopathic leukopenia, Sjogren's syndrome, Wegener's granulomatosis; (iii) an inflammatory disease due to organ transplantation; (iv) a bone marrow transplantation; and (v) a disease due to exogenously administered self or exogenously administered non-autologous recombinant polypeptide.
  • Another embodiment of the present invention relates to a pharmaceutical composition comprising at least one Treg cell activator according to the present invention—preferably HIV-1 gp120—as an active ingredient and can be formulated in conventional manner. Methods for making such formulations can be found in manuals, e.g. “Remington Pharmaceutical Science”. Examples for ingredients that are useful for formulating at least one substance according to the present invention are also found in WO99/18193, which is hereby incorporated by reference.
  • The composition may be manufactured in a manner that is itself known, e.g. by mean of conventional mixing, dissolving, granulating, dragee-making, levitating, powdering, emulsifying, encapsulating, entrapping of lyophilizing processes.
  • In a further aspect the invention teaches a method for treating a disease which is characterized in that its clinical picture can be influenced positively by an increase of activated Treg cells which method comprises administering to a being preferably a human being in need of such a treatment a suitable amount of a pharmaceutical composition comprising at least one Treg cell activator according to the present invention, preferably HIV-1 gp120 or HIV-1 gp120 derived fragments and peptides thereof. The present invention provides therefore for a method for treating (i) a non autoimmune inflammatory disease: asthma, allergic asthma, respiratory allergy, allergic rhinoconjunctivitis, allergic alveolitis, contact allergy, atopic dermatitis, neurodermatitis, food allergy, graft-versus-host disease, non-autoimmune inflammatory bowel disease, acute respiratory distress syndrome, acute inflammatory pancreatitis, burns, wound healing, skin scarring disorders, sarcoidosis, Behcet's disease or Sweet's syndrome; (ii) an autoimmune inflammatory disease: rheumatoid arthritis, rheumatic fever, systemic lupus erythematosus, ulcerative colitis, Crohn's disease, autoimmune inflammatory bowel disease, diabetes type I, gastritis, autoimmune atrophic gastritis, autoimmune hepatitis, Hashimoto's thyroiditis, thyreoiditis, multiple sclerosis, myasthenia gravis, autoimmune haemolytic anemia, Addison's disease, scleroderma, Goodpasture's syndrome, Guillain-Barre syndrome, Graves' disease, glomerulonephritis, psoriasis, pemphigus vulgaris, pemphigoid, vitiligo, idiopathic leukopenia, Sjogren's syndrome or Wegener's granulomatosis; (iii) an inflammatory disease due to organ transplantation; (iv) a bone marrow transplantation; or (v) a disease due to exogenously administered self or exogenously administered non-autologous recombinant polypeptide which method comprises administering to a being in need of such a treatment a suitable amount of a pharmaceutical composition comprising at least one Treg cell activator.
  • The present invention additionally, provides for a use of a Treg cell activator according to the present invention for reducing and/or preventing an unwanted immune reaction due to a exogenously administered self or exogenously administered non-autologous recombinant polypeptide and provides for a method for reducing or preventing an unwanted immune reaction comprising: (a) adding a sufficient amount of at least one Treg cell activator according to the present invention to a non-human animal, preferably a non-human primate.
  • In a further embodiment the present invention provides for a test system for determining whether a substance is a Treg cell activator according to the present invention comprising at least
  • a) a Treg cell and
  • b) a peptide spanning at least the epitope as set forth in SEQ ID NO.: 1.
  • In a preferred test system of the present invention a peptide of b) is selected from a group consisting of an isolated peptide spanning amino acid No. 26 to 458 of SEQ ID NO.: 2, amino acid No. 26 to 419 of SEQ ID NO.: 2, amino acid No. 26 to 207 of SEQ ID NO.: 2, amino acid No. 26 to 131 of SEQ ID NO.: 2, and amino acid No. 46 to 89 of SEQ ID NO.: 2. or the peptide used is an isolated peptide spanning amino acid No. 1 to 31 of SEQ ID NO.: 1 and having additional up-stream and/or downstream amino acids with the prerequisite that the additional amino acids do not hinder binding of a substance to the amino acid stretch as set forth in SEQ ID NO.: 1. Said additional up-stream amino acid or amino acid stretch is preferably selected from a group consisting of the amino acid or amino acid stretch as set forth in SEQ ID NO.: 2 at position 53, at position 52-53, at position 51-53, at position 50-53, at position 49-53, at position 48-53, at position 47-53, at position 46-53, at position 45-53, at position 44-53, at position 43-53, at position 42-53, at position 41-53, at position 40-53, at position 39-53, at position 38-53, at position 37-53, at position 36-53, at position 35-53, at position 34-53, at position 33-53, at position 32-53, at position 31-53, at position 30-53, at position 29-53, at position 28-53, at position 27-53, at position 26-53, at position 25-53, at position 24-53, at position 23-53, at position 22-53, at position 21-53, at position 20-53, at position 19-53, at position 18-53, at position 17-53, at position 16-53, at position 15-53, at position 14-53, at position 13-53, at position 12-53, at position 11-53, at position 10-53, at position 9-53, at position 8-53, at position 7-53, at position 6-53, at position 5-53, at position 4-53, at position 3-53, at position 2-53, and at position 1-53. A more preferred peptide does additionally comprise downstream one or more amino acids as set forth in SEQ ID NO.: 2 at position 85, or at position 85 to n, wherein n is an integer between 86-458, i.e. position 85 to 86, 85 to 87, 85 to 88, 85 to 89, 85 to 90, 85 to 91, 85 to 92, 85 to 93, 85 to 94, 85 to 95, 85 to 96, 85 to 97, 85 to 98, 85 to 99, 85 to 100, 85 to 101, 85 to 102, 85 to 103, 85 to 104, 85 to 105, 85 to 106, 85 to 107, 85 to 108, 85 to 109, 85 to 110, 85 to 111, 85 to 112, 85 to 113, 85 to 114, 85 to 115, 85 to 116, 85 to 117, 85 to 118, 85 to 119, 85 to 120, 85 to 121, 85 to 122, 85 to 123, 85 to 124, 85 to 125, 85 to 126, 85 to 127, 85 to 128, 85 to 129, 85 to 130, 85 to 131, 85 to 132, 85 to 133, 85 to 134, 85 to 135, 85 to 136, 85 to 137, 85 to 138, 85 to 139, 85 to 140, 85 to 141, 85 to 142, 85 to 143, 85 to 144, 85 to 145, 85 to 146, 85 to 147, 85 to 148, 85 to 149, 85 to 150, 85 to 151, 85 to 152, 85 to 153, 85 to 154, 85 to 155, 85 to 156, 85 to 157, 85 to 158, 85 to 159, 85 to 160, 85 to 161, 85 to 162, 85 to 163, 85 to 164, 85 to 165, 85 to 166, 85 to 167, 85 to 168, 85 to 169, 85 to 170, 85 to 171, 85 to 172, 85 to 173, 85 to 174, 85 to 175, 85 to 176, 85 to 177, 85 to 178, 85 to 179, 85 to 180, 85 to 181, 85 to 182, 85 to 183, 85 to 184, 85 to 185, 85 to 186, 85 to 187, 85 to 188, 85 to 189, 85 to 190, 85 to 191, 85 to 192, 85 to 193, 85 to 194, 85 to 195, 85 to 196, 85 to 197, 85 to 198, 85 to 199, 85 to 200, 85 to 201, 85 to 202, 85 to 203, 85 to 204, 85 to 205, 85 to 206, 85 to 207, 85 to 208, 85 to 209, 85 to 210, 85 to 211, 85 to 212, 85 to 213, 85 to 214, 85 to 215, 85 to 216, 85 to 217, 85 to 218, 85 to 219, 85 to 220, 85 to 221, 85 to 222, 85 to 223, 85 to 224, 85 to 225, 85 to 226, 85 to 227, 85 to 228, 85 to 229, 85 to 230, 85 to 231, 85 to 232, 85 to 233, 85 to 234, 85 to 235, 85 to 236, 85 to 237, 85 to 238, 85 to 239, 85 to 240, 85 to 241, 85 to 242, 85 to 243, 85 to 244, 85 to 245, 85 to 246, 85 to 247, 85 to 248, 85 to 249, 85 to 250, 85 to 251, 85 to 252, 85 to 253, 85 to 254, 85 to 255, 85 to 256, 85 to 257, 85 to 258, 85 to 259, 85 to 260, 85 to 261, 85 to 262, 85 to 263, 85 to 264, 85 to 265, 85 to 266, 85 to 267, 85 to 268, 85 to 269, 85 to 270, 85 to 271, 85 to 272, 85 to 273, 85 to 274, 85 to 275, 85 to 276, 85 to 277, 85 to 278, 85 to 279, 85 to 280, 85 to 281, 85 to 282, 85 to 283, 85 to 284, 85 to 285, 85 to 286, 85 to 287, 85 to 288, 85 to 289, 85 to 290, 85 to 291, 85 to 292, 85 to 293, 85 to 294, 85 to 295, 85 to 296, 85 to 297, 85 to 298, 85 to 299, 85 to 300, 85 to 301, 85 to 302, 85 to 303, 85 to 304, 85 to 305, 85 to 306, 85 to 307, 85 to 308, 85 to 309, 85 to 310, 85 to 311, 85 to 312, 85 to 313, 85 to 314, 85 to 315, 85 to 316, 85 to 317, 85 to 318, 85 to 319, 85 to 320, 85 to 321, 85 to 322, 85 to 323, 85 to 324, 85 to 325, 85 to 326, 85 to 327, 85 to 328, 85 to 329, 85 to 330, 85 to 331, 85 to 332, 85 to 333, 85 to 334, 85 to 335, 85 to 336, 85 to 337, 85 to 338, 85 to 339, 85 to 340, 85 to 341, 85 to 342, 85 to 343, 85 to 344, 85 to 345, 85 to 346, 85 to 347, 85 to 348, 85 to 349, 85 to 350, 85 to 351, 85 to 352, 85 to 353, 85 to 354, 85 to 355, 85 to 356, 85 to 357, 85 to 358, 85 to 359, 85 to 360, 85 to 361, 85 to 362, 85 to 363, 85 to 364, 85 to 365, 85 to 366, 85 to 367, 85 to 368, 85 to 369, 85 to 370, 85 to 371, 85 to 372, 85 to 373, 85 to 374, 85 to 375, 85 to 376, 85 to 377, 85 to 378, 85 to 379, 85 to 380, 85 to 381, 85 to 382, 85 to 383, 85 to 384, 85 to 385, 85 to 386, 85 to 387, 85 to 388, 85 to 389, 85 to 390, 85 to 391, 85 to 392, 85 to 393, 85 to 394, 85 to 395, 85 to 396, 85 to 397, 85 to 398, 85 to 399, 85 to 400, 85 to 401, 85 to 402, 85 to 403, 85 to 404, 85 to 405, 85 to 406, 85 to 407, 85 to 408, 85 to 409, 85 to 410, 85 to 411, 85 to 412, 85 to 413, 85 to 414, 85 to 415, 85 to 416, 85 to 417, 85 to 418, 85 to 419, 85 to 420, 85 to 421, 85 to 422, 85 to 423, 85 to 424, 85 to 425, 85 to 426, 85 to 427, 85 to 428, 85 to 429, 85 to 430, 85 to 431, 85 to 432, 85 to 433, 85 to 434, 85 to 435, 85 to 436, 85 to 437, 85 to 438, 85 to 439, 85 to 440, 85 to 441, 85 to 442, 85 to 443, 85 to 444, 85 to 445, 85 to 446, 85 to 447, 85 to 448, 85 to 449, 85 to 450, 85 to 451, 85 to 452, 85 to 453, 85 to 454, 85 to 455, 85 to 456, 85 to 457, or 85 to 458.
  • The following examples are meant to illustrate the present invention, however, shall not be construed as limitation. However, the Examples describe most preferred embodiments of the invention.
  • EXAMPLES
  • (1) CD4/HIV-1 gp120 competition assay for determining whether a substance can bind at least to a peptide spanning epitope as set fourth in SEQ ID NO.: 1. 96 well assay plates (Nunc, Germany) are coated overnight at 4° C. with CD4 (sCD4, Immunodiagnostics, USA) 100 ng per well in PBS, pH 7.4. Coated plates are saturated with PBS/3% BSA buffer and washed three times. To determine binding of a test substance sample is added for 1 hour in different concentrations. No test substance is added to control wells. After washing three times HIV-1 gp120-peroxidase conjugate (Immunodiagnostics, USA) is added to the plate for 1 h. Unbound HIV-1 gp120-peroxidase conjugate is removed by washing three times. After washing, 3,3,5,5-tetramethylbenzidine chromogen substrate (Pierce, USA) for peroxidase is added and the optical density is read at 450 nm. A substance which interacts with the CD4 HIV-1 gp120-binding site will block the binding of labeled HIV-1 gp120 and is identifiable by a reduced signal compared to the controls.
  • (2) Assay for determining whether e.g. HIV-1 gp120 or a substance which can interfere with HIV-1 gp120-binding to a CD4+CD25+ Treg can activate a CD4+CD25+ Treg and therewith can be useful in the treatment an autoimmune disease (e.g. Inflammatory Bowel Disease, Multiple Sclerosis, Rheumatoid Arthritis, Psoriasis, Diabetes Type I, Lupus Erythematosus, Phemphigus vulgaris, Thyreoiditis), other diseases with autoimmune aspects in their pathogenesis such as vitiligo, atopic dermatitis, an allergy (e.g. Allergic rhinitis,), asthma (e.g. allergic asthma), GVHD (graft-versus-host disease), graft rejection.
  • (2.1) Method for isolation of cells P (2.1.1) Isolation of PBMC
  • The isolation procedure starts with PBMC (peripheral blood mononuclear cells) isolated by standard density gradient centrifugation from normal buffy coat preparations of healthy volunteers. Alternatively, PBMC isolated from whole peripheral blood or leukapheresis products can be used.
  • Blood from buffy coats is diluted 1:1 with PBS (phosphate buffered saline) containing 0.2% Liquemine (Sodium-Heparin) and 2 mM EDTA at room temperature. The diluted blood is thoroughly pipetted onto prepared Ficoll layers (30 ml diluted blood per 15 ml Ficoll layer per 50 ml tube) and centrifuged for 15 min. (minute) at 200×g (with brake on) at room temperature. 8-10 ml of the upper fluid are carefully removed and the tubes centrifuged at 450×g for 15 min. at room temperature (with brake off). PBMC are collected from the interphase of each gradient, washed three times with 50 ml PBS/1 mM EDTA separately, than pooled and wash two more times. Finally, PBMC are re-diluted in X-VIVO-15 (Cambrex, Verviers, Belgium) in cell culture medium and counted.
  • (2.1.2) Isolation of CD4+ T helper cells
  • For determining suppressive activity of Treg cells highly purified cell populations are needed. Therefore, antibody-coated magnetic beads are used (Miltenyi, Germany and or Dynal, Norway). Magnetic beads in this context are paramagnetic particles that are coupled to specific monoclonal antibodies. They are used to magnetically label the target cell population. The antibody-coated magnetic beads bind to the target cells. This labeled cell fraction is retained by magnetic force and can be recovered subsequently highly purified (positive selection). Before positive isolation of CD4+CD25 T helper cells PBMC are washed two times with 50 ml washing buffer according to the manufacturer's instructions (Miltenyi Germany). For isolation of CD4+ T cells, CD4 microbeads (Miltenyi Germany, 2-4 μL microbeads/107 PBMC) are used according to the manufacturer's instructions. The CD4+ fraction is isolated using a MACS separator (Miltenyi) according to the instructions of the manufacturer. MACS separator retains magnetic bead-labeled cells by magnetic force. Contaminating CD4+CD25+ Treg cells are depleted in a second step by using CD25 Dynabeads, (Dynal, Norway) according to the instructions of the manufacturer (details see 2.1.4) using (0.5 beads/cell). This depletion procedure results in highly purified CD4+CD25 T helper cells (negative selection).
  • To circumvent CD4 antibodies binding to CD4+ cells (to avoid CD4 dependent pre activation) in the isolation procedure of CD4+ T cells, alternatively untouched CD4+ T helper cells are generated by using the negative isolation kit (Miltenyi Germany) according to the manufacturer's instructions. Before negative isolation of CD4+ T helper cells PBMC are washed two times with 50 ml washing buffer according to the manufacturer's instructions (Miltenyi Germany). For isolation of CD4+ T helper cells, PBMC are incubated with a cocktail of biotinylated CD45RO, CD8, CD14, CD16, CD19, CD56, CD36, CD123, anti-TCRγ/δ, and CD235a antibodies. These cells are subsequently magnetically labeled with Anti-Biotin Microbeads for depletion.
  • The CD4+ fraction is isolated using a MACS separator (Miltenyi) according to the instructions of the manufacturer. MACS separator retains magnetic bead-labeled cells by magnetic force. Contaminating CD4+CD25+ Treg cells are depleted in a second step by using CD25 Dynabeads, (Dynal, Norway) according to the instructions of the manufacturer (details see 2.1.4) using 0.5 beads/cell.
  • (2.1.3) Isolation of CD8+ T effector cells
  • Before positive isolation of CD8+ T effector cells PBMC are washed two times with 50 ml washing buffer according to the manufacturer's instructions (Miltenyi Germany). For isolation of CD8+CD25 T cells, CD8 microbeads (Miltenyi Germany, 2-4 μL microbeads/107 PBMC) are used according to the manufacturer's instructions. The CD8+ fraction is isolated using a MACS separator (Miltenyi) according to the instructions of the manufacturer. Contaminating CD8+CD25+ T cells are depleted in a second step by using CD25 Dynabeads, (Dynal, Norway) according to the instructions of the manufacturer (details see 2.1.4) using (0.5 beads/cell). This depletion procedure results in highly purified CD8+CD25 T effector cells (negative selection).
  • (2.1.4) Isolation of CD4+CD25+ Treg cells
  • For Isolation of CD4+CD25+ Treg cells positive and negative selection is combined. PBMC are washed with washing buffer according to the manufacturer's instructions (Miltenyi Germany) and subsequently incubated with CD25 microbeads (2 μL microbeads/107 PBMC) for 20 min. at 4° C. in isolation buffer (1×108/ml) according to the manufacturer's instructions. Afterwards, the cells are washed two times in PBS. The CD25+ fraction is isolated using a MACS separator (Miltenyi) according to the instructions of the manufacturer. The positively selected CD25+ fraction contains 65-80% CD4+ T cells and 20-35% contaminating CD19+ B cells, CD8+ T cells, and few CD14+ monocytes. The contaminating cells are depleted with Dynabeads (Dynal, Norway). The following amounts of beads are used: CD19 Dynabeads: 2 beads/cell, CD8 Dynabeads: 3 beads/cell, CD14 Dynabeads: 1 bead/cell. Collected Dynabeads are washed two times in 15 ml tubes with depletion buffer using the magnetic particle concentrator (Dynal) according to the manufacturer's instructions. The CD25+ PBMC fraction (5×107/ml) is added in depletion buffer and incubated for 20 min. at 4° C. on a shaker (sample mixer, Dynal). Contaminating cells are depleted according to the manufacturer's instructions by the use of the magnetic particle concentrator. For higher purity of CD4+CD25+ Treg cells Dynabeads depletion is repeated once (>98% after two rounds of depletion).
  • (2.1.5) Generation of monocyte-derived dendritic cells
  • Dendritic cells (DC) are generated from buffy coats of healthy volunteers. PBMC (2.1.6) are plated in 6-well tissue culture plates at a density of 15×106 cells/well in 3 ml X-VIVO-15 (Cambrex, Verviers, Belgium) plus 1.5% heat-inactivated autologous plasma containing 800 U/ml GM-CSF (Leukomax; Novartis, Basel, Switzerland) and 1,000 U/ml IL-4 (Strathmann Biotec, Hamburg, Germany). Cultures are fed every other day (days 2, 4 and 6) by removing 1 ml of the medium and adding back 1 ml fresh medium with cytokines. On day 7, non-adherent cells were harvested and transferred to new 6 well plates and cultured further on in the presence of 10 ng/ml IL-1β, 10 ng/ml TNF-α, 1,000 U/ml IL-6 (all from Strathmann, Biotech, Germany) and 1 μg/ml PGE2 (Pharmacia-Upjohn, Uppsala, Sweden). Mature CD83+ DC are harvested at day 9 of culture.
  • (2.1.6) Isolation of CD3-depleted PBMC
  • T cells are depleted from PBMC with CD3 Dynabeads (Dynal, Norway) by using 0.5 beads/cell. Collected Dynabeads are washed two times in 15 ml tubes with depletion buffer using the magnetic particle concentrator (Dynal) according to the manufacturer's instructions. PBMC (5×107/ml) are added in depletion buffer and incubated for 20 min. at 4° C. on a shaker (sample mixer, Dynal). CD3+ cells are depleted according to the manufacturer's instructions by the use of the magnetic particle concentrator resulting in a purity of >98% CD3 PBMC.
  • (2.1.7) Isolation of human CD25-depleted PBMC
  • PBMC are isolated according to (2.1.1). CD25-expressing regulatory T cells in the PBMC preparation are depleted with CD25 Dynabeads (Dynal, Norway) according to the instructions of the manufacturer (details see 2.1.4) by using 0.5 beads/cell. Collected Dynabeads are washed two times in 15 ml tubes with depletion buffer using the magnetic particle concentrator (Dynal) according to the manufacturer's instructions. PBMC (5×107/ml) are added in depletion buffer and incubated for 20 min. at 4° C. on a shaker (sample mixer, Dynal). CD25+ cells are depleted according to the manufacturer's instructions by the use of the magnetic particle concentrator resulting in a purity of >99% CD25-negative PBMC.
  • (2.2) Method for testing suppressive activity of CD4+CD25+ Treg cells
  • (2.2.1) Coculture suppression assay A: Mixed leukocyte reaction (MLR) Cocultures of CD4+ T helper cells or CD8+ T effector cells with CD4+CD25+ Treg cells and allogeneic DC have to be performed to analyze the suppressive activity of CD4+CD25+ Treg cells on CD4+ T helper cells or CD8+ T effector cells. Therefore, 1×105/well CD4+ T helper cells (2.1.2) or CD8+ T effector cells (2.1.3) are cocultured with different numbers of CD4+CD25+ Treg cells (2.1.4; ratio 1:1 to 1:4) and 1×104/well DC in 96 well flat bottom culture plates in X-VIVO 15 (Cambrex, Verviers, Belgium) in the presence or absence of a CD4-binding compound e.g. HIV-1 gp120 (0.1-10 μg/ml). Mature dendritic cells (DC) generated as described (2.1.5) are from the same donor as CD4+CD25+ Treg cells (syngenic) but allogeneic to the CD4+ T helper cells or CD8+ T effector cells are used for T cell stimulation. In this assay, only CD4+ T helper cells or CD8+ T effector cells are activated by the allogeneic DC (MLR) resulting in a strong proliferation of the T cell subset. Non-activated CD4+CD25+ Treg cells did not suppress this proliferation in absence of a Treg cell activating compound. A functional activation of CD4+CD25+ Treg cells by a CD4 binding compound resulted in a reduced proliferation of CD4+T helper cells or CD8+ T effector cells.
  • Proliferation is determined after 4 days of culture by adding 37 kBq 3H-Thymindine (3H-Tdr) for additional 16 h.
  • (2.2.2) Coculture Suppression Assay B: Stimulation of CD8+ T effector cells with allogeneic PBMC and CD4+CD25+ Treg cells from the same healthy volunteer.
  • In order to study the influence of a substance, e.g. HIV-1 gp120 which can bind at least to the epitope given in SEQ ID NO.:1 exclusively on the suppressive function of CD4+CD25+ Treg cells, we developed a co-culture assay which contained CD8+ T cells as effectors to exclude any influence of this substance on the latter (CD8+ T cells don't express CD4). In this setting, activation of alloreactive CD8+ T effector cells is only suppressed by activated CD4+CD25+ Treg cells such as upon additional anti-CD3 mAb stimulation (positive control). To evaluate the influence of e.g. HIV-1 gp120 on the function of CD4+CD25+ Treg cells, isolated CD4+CD25+ Treg cells (2.1.4) are co-cultured with syngenic, T cell-depleted and irradiated (50 Gy) PBMC 2.1.6) and allogeneic CD8+ T effector cells (2.1.3) in presence of varying concentrations (0.1-10 μg/ml) of different HIV-1 gp 120 preparations. Briefly, 1×105 CD4+CD25+ Treg cells are incubated with 3×105 syngenic T cell-depleted PBMC in the presence or absence of varying amounts of HIV-1 gp120. Stimulation with 0.5 μg/ml anti-CD3 monoclonal antibody (OKT-3, ebioscience, USA) serves as positive control. No additional stimulation represents negative control. Either immediately or 24 h. later, 1×105 allogeneic CD8+ T effector cells are added to the cultures and proliferation is determined after additional 72 h. by 3H-Tdr incorporation (37 kBq/well). Functional activation of CD4+CD25+ Treg cells via interaction with the epitope as set forth in SEQ ID NO:1 result in suppressed proliferation of CD8+ T effector cells and therewith identifies a substance as a Treg cell activator (Data see FIG. 1).
  • To show in parallel whether a substance can interfere with HIV-1 gp120 binding to CD4 epitope of a CD4+CD25+ Treg cell, CD4 is added in different concentrations (0.1-10 μg/ml) to isolated CD4+CD25 Treg cells (2.1.4) cocultured with syngenic T cell-depleted PBMC (2.1.6) and allogeneic CD8+ T effector cells (2.1.3) in the presence of varying amounts of the substance. Enhanced proliferation of CD8+ effector cells resembles competitive binding to CD4 and blocked activation of a Treg cell via interaction with the epitope as set forth in SEQ ID NO:1. All cultures are performed in serum free X-VIVO-15 (Cambrex, Verviers, Belgium).
  • Alternatively to irradiation-induced inactivation and block of proliferation PBMC can be treated with Mitomycin C (Sigma, Germany). Briefly, 3×107 PBMC are incubated in 3 ml MEM/10% FCS/180 μg Mitomycin C for 30 min at 37° C. Afterwards cells are washed 5× using MEM/10% FCS. Subsequently cells are subjected to the assay.
  • (2.2.3) Coculture suppression assay C: Stimulation of T cells in presence of pre-activated CD4+CD25+ Treg cells and allogeneic PBMC
  • To evaluate the direct Treg cell activating potential of a compound in the absence of antigen-presenting cells such as PBMC, isolated CD4+CD25+ Treg cells (according to 2.1.4) are pre-cultured in X-VIVO-15 for 16-48 h. alone, in presence of 0.5 μg/ml anti-CD3 monoclonal antibody (OKT-3) as positive control, or in presence of different concentrations of HIV-1 gp120. Afterwards, cells are washed intensively and added to cocultures of syngenic, irradiated (50 Gy) PBMC and allogeneic CD4+ T helper cells or CD8+ T effector cells. Proliferation is determined after additional 72 h. by 3H-Tdr incorporation (37 kBq/well). Functional activation of CD4+CD25+ Treg cells via interaction with the epitope as set forth in SEQ ID NO.: 1 result in suppressed proliferation of CD4+ T helper cells or CD8+ T effector cells and therewith identifies a substance as a Treg cell activator.
  • A reduced incorporated radioactivity resembles suppressed proliferation and identifies CD8+ inhibition and therewith identifies a substance to activate a Treg cell via interaction with the epitope as set forth in SEQ ID NO.: 1.
  • (2.3) Method for readout of suppressive activity of CD4+CD25+ Treg cells
  • (2.3.1) Analysis of proliferation
  • After 3-4 days of incubation of cells in assays provided under (2.2.1), (2.2.2) and (2.2.3) T cell proliferation is measured. For additional 16 h., cells are pulsed with 3H-Tdr (37 kBq/well, MB Biomedicals), and incorporated radioactivity is measured by using a liquid scintillation counter (Betaplate, Wallac/PerkinElmer). In the absence of a Treg cell activating compound, resting Treg cells cannot inhibit the proliferation of CD4+ T helper cells or CD8+ T effector cells (negative control). In contrast, in presence of an activating CD4-binding compound or anti-CD3 monoclonal antibody (positive control) the proliferation of CD4+ T helper cells or CD8+ T effector cells is suppressed by CD4+CD25+ Treg cells. A reduced incorporated radioactivity resembles suppressed proliferation and identifies CD4+ T helper cell or CD8+ T effector cell inhibition and therewith identifies that a CD4+CD25+ Treg cell has been activated.
  • For some experiments the proliferation of either CD4+CD25+ Treg cells, CD4+ T helper cells or CD8+ T effector cells is selectively followed by flow cytometry after labeling of these population with Vibrant CFDA SE Cell Tracer Kit (Invitrogen Life Technologies, San Diego, USA) according to the manufacturer's protocol. For CFDA labeling, T cells (1×107 cells/ml) are incubated in PBS with 1 μM CFDA Vibrant CFDA SE Cell Tracer Kit (Invitrogen Life Technologies, San Diego, USA) at 37° C. for 30 min. Thereafter, cells are washed with X-VIVO-15 and incubated for 30 min. at 37° C. in the dark. After an additional wash the cells are counted and added to the cocultures. In addition, proliferation of cells is measured by flow cytometry after 4-6 days of culture.
  • (2.3.2) Analysis of cytokine production
  • Activated Treg cells not only inhibit the proliferation of cocultured CD4+ T helper cells or CD8+ T effector cells, they suppress also the cytokine synthesis of these T cells. Therefore, detection of cytokines produced by cocultured T cells is an alternative method to analyze the Treg cell activating potential of a reagent. In this assay, isolated CD4+CD25+ Treg cells (according to 2.1.4), syngenic CD3-depleted PBMC (according to 2.1.6) and allogeneic CD8+ T effector cells (according to 2.1.3) are cocultured as described (2.2) before. After 7 days, alloreactive CD8+ T effector cells are polyclonal restimulated with 2.4 μg/ml phytohemagglutinin (PHA, Sigma, Germany) and 1 ng/ml PMA for 5 h. in presence of monensin (BD GolgiStop™, BD Biosciences Pharmingen 1.3 μM). Thereafter, cells are collected, washed with PBS, fixed and permeabilized according to the manufacturer's instruction (perm/fix solution, BD PharMingen, Germany) and stained with 0.5 μg/test of a cytokine-specific monoclonal antibody (anti-IFN-γ, anti-IL-2, anti-TNF-α, all from BD PharMingen). Subsequently, production of cytokines by CD4+ T helper cells or CD8+ T effector cells is analyzed by flow cytometry. In the absence of a Treg cell activating compound Treg cells cannot inhibit the cytokine production of CD4+ T helper cells or CD8+ T effector cells (negative control). In contrast, in presence of an activating CD4-binding compound or anti-CD3 monoclonal antibodies (positive control) the cytokine production of CD4+ T helper cells or CD8+ T effector cells is suppressed by functional activated Treg cells. A reduced cytokine production identifies inhibition and therewith identifies that a CD4+CD25+ Treg cell has been activated.
  • (2.3.3) Analysis of CD25 expression
  • Activated Treg cells inhibit the ability of CD8+ T effector cells and CD4+ T helper cells to express the α-chain of the IL-2 receptor, CD25. Therefore, the analysis of CD25 expression by flow cytometry is an additional method to evaluate the Treg cell activating potential of a reagent. In this assay, isolated CD4+CD25+ Treg cells (according to 2.1.4) and syngenic CD3-depleted PBMC (according to 2.1.6) are cocultured with allogeneic CD8+ T effector cells or allogeneic CD4+ T helper cells (according to 2.1.3, 2.1.2 and 2.2.1, 2.2.2 and 2.2.3). After 7 days, alloreactive CD8+ T effector cells or allogeneic CD4+ T helper cells are stimulated with allogeneic PBMC from the same donor or DC as used in primary culture and expression of CD25 on alloreactive CD8+ T effector or allogeneic CD4+ T helper cells cells are analyzed 24 h. thereafter by flow cytometry. Activation of Treg cells by the test compound results directly in an inhibited CD25 expression on the re-stimulated CD8+ T effector cells or CD4+ T helper cells. In the absence of a Treg cell activating compound, Treg cells cannot inhibit the CD25 expression on re-stimulated CD8+ T effector cells or CD4+ T helper cells (negative control). In contrast, in presence of an activating reagent or an anti-CD3 monoclonal antibody (positive control) the CD25 expression of CD8+ T effector cells or CD4+ T helper cells is suppressed by Treg cells. A reduced CD25 expression identifies inhibition and therewith identifies that a CD4+CD25+ Treg cell has been activated.
  • (2.3.4) Analysis of cyclic AMP production
  • Stimulation of Treg cells results in strong increase of cytosolic (i.e. intracellular) cAMP. Therefore, the analysis of cAMP in Treg cells is an additional method to determine whether a substance which can interfere with HIV-1 gp120 binding to CD4 epitope of a CD4-positive cell can activate a Treg cell. In this assay freshly isolated CD4+CD25+ Treg cells (according to 2.1.4) (1×105-1×106/well) are incubated with anti-CD3 monoclonal antibody (OKT-3; 0.5 μg/ml) or HIV-1 gp120 (0.1-10 m/ml; Protein Science Corp., Meriden, Conn., USA) or left untreated for 16 hours. To assess cytosolic cAMP-concentrations a cAMP-specific ELISA (Parameter™ Cyclic AMP Assay, Cat. No. KGE002; R&D Systems, Wiesbaden, Germany) is applied. Treg cells are washed three times in ice-cold PBS and subsequently lysed (1×107/ml) using lysis buffer supplied by the manufacturer and applied to the ELISA according to the manufacturer's recommendations.
  • Activation of Treg cells by the test compound results directly in an increase of cytosolic cAMP. In the absence of a Treg cell activating compound no increase of cAMP in CD4+CD25+ Treg cells is detected (negative control). In contrast, in presence of an activating CD4-binding compound or an anti-CD3 monoclonal antibody (positive control) CD4+CD25+ Treg cell show a strong increase of cytosolic cAMP. An increase of cytosolic cAMP identifies that a CD4+CD25+ Treg cell has been activated. Data see FIG. 2.
  • Alternatively, additional analysis of assay supernatant cAMP concentration and use of phoshodiesterase inhibitors e.g. Roflumilast (1-50 μM) enhance the cAMP signal in the assay.
  • (3) In vivo assay for determining whether HIV-1 gp120 or a substance which can interfere with HIV-1 gp120-binding to a CD4+CD25+ Treg can activate a CD4+CD25+ Treg in a disease-related model and therewith can be useful in the treatment of an autoimmune disease (e.g. Inflammatory Bowel Disease, Multiple Sclerosis, Rheumatoid Arthritis, Psoriasis, Diabetes Type I, Lupus Erythematosus, Phemphigus vulgaris, Thyreoiditis), other diseases with autoimmune aspects in their pathogenesis such as vitiligo, atopic dermatitis, an allergy (e.g. Allergic rhinitis,), asthma (e.g. allergic asthma), GVHD (graft-versus-host disease), graft rejection.
  • (3.1) Method for induction of GVHD by transfer of human PBMC into NOD-Scid mice and measurement of disease severity.
  • NOD-Scid mice, non-obese diabetic/severe combined immunodeficient mice (mutant allele: PrkdcScid, strain: NOD.CB17-PrkdcScid, (Shultz et al. 1995)) from the Central Laboratory Animal Facility of the University of Mainz (also purchasable from The Jackson Laboratory via Charles River Laboratories, Germany) are used as an animal model for transfer of peripheral human immune cells and thus for the in vivo analysis of immuno-regulatory interactions (Hesselton et al. 1995). Human peripheral blood mononuclear cells (PBMC) transferred into NOD-Scid mice induce an accelerating age-dependent graft-versus-host disease (GVHD). While the number of cells transferred determines the onset of this disease, the co-transfer of human Tregs can gradually delay or prevent the disease. This model system further allows the study of human Treg function. To induce a GVHD NOD-Scid mice at the age of three to six days after birth are intraperitoneally injected with 1×107 to 3×107 human PBMC (isolation of human PBMC according to 2.1.1). GVHD is induced by immunological (xenogenic) activation of human effector T cells in the PBMC fraction injected into the mice. NOD-Scid mice, however, are not able to react against the grafted human PBMC. The human PBMC-induced disease is characterized by lack of weight increase and weight loss, decreased mobility, hunched posture, ruffled fur and organ inflammation in the treated animals (Kizilisik and Al-Sebayel 1997). Transfer of human PBMC results in growth arrest or weight loss within 30 to 40 days (depending on the number of cells transferred) after transfer compared to untreated mice. Lack of weight increase/weight loss is used as a parameter to score GVHD severity. Control mice did not receive any PBMC. Data see FIG. 3.
  • (3.2) Method for induction of GVHD by transfer of human PBMC into NOD-Scid mice and prevention of disease by additional transfer of human regulatory T cells and measurement of disease severity.
  • To prevent a GVHD disease in NOD-Scid mice at the age of three to six days after birth induced by intraperitoneally injection of 1×107 to 3×107 human PBMC (isolation of human PBMC according to 2.1.1) additional 2.5×106 human regulatory T cells (isolation of human regulatory T cells according to 2.1.4) are injected intraperitoneally (ratio Tregs to PBMC: 1:4). Co-transfer of human regulatory T cells along with human PBMC (enhancement of Treg ratio) results in prevention of development of GVHD and weight loss similar development as untreated mice. Lack of weight increase/weight loss is used as a parameter to score GVHD severity. Control mice did not receive any PBMC. Data see FIG. 3.
  • (3.3) Method for induction of GVHD by transfer of human PBMC into NOD-Scid mice and prevention of disease by additional administration of HIV-1 gp120 or a substance which can interfere with HIV-1 gp120-binding and measurement of disease severity.
  • To prevent a GVHD disease induced in NOD-Scid mice at the age of three to six days after birth induced by intraperitoneally injection of 1×107 to 3×107 human PBMC (isolation of human PBMC according to 2.1.1) HIV-1 gp120 or a substance which can interfere with HIV-1 gp120-binding is administered in addition to the injection of human PBMC. Administration of HIV-1 gp120 or a substance which can interfere with HIV-1 gp120-binding induces an activation of human regulatory T cells in the PBMC fraction resulting in prevention of organ inflammation and prevention of growth arrest/weight loss similar to mice treated with human PBMC and additional human regulatory T cells or untreated mice. Lack of weight increase/weight loss is used as a parameter to score GVHD severity. Control mice did not receive any PBMC. Data see FIG. 4.
  • (3.4) Method for induction of GVHD by transfer of human PBMC into NOD-Scid mice and to demonstrate that prevention of disease by administration of HIV-1 gp120 or a substance which can interfere with HIV-1 gp120-binding is mediated by human regulatory T cells in the PBMC fraction injected into NOD-Scid mice and measurement of disease severity.
  • NOD-Scid mice at the age of three to six days after birth are intraperitoneally injected with 1×107 human CD25-depleted PBMC (isolation of human CD25-depleted PBMC according to 2.1.7). GVHD is induced by 1×107 human CD25-depleted PBMC at the same magnitude as by transfer of 1×107 human non CD25-depleted PBMC into NOD-Scid mice. (Data see FIG. 5 and FIG. 3 and FIG. 4) Mice receiving CD25-depleted PBMC and an additional administration of HIV gp120 are not protected from development of GVHD and weight loss. Prevention of GVHD by HIV gp120 or a substance which can interfere with HIV-1 gp120-binding depends on regulatory T cells. Lack of weight increase/weight loss is used as a parameter to score GVHD severity. Data see FIG. 5.
  • DESCRIPTION OF FIGURES
  • FIG. 1: HIV-1 gp120 treatment activates human CD4+CD25+ Treg cells
  • In order to study the influence of HIV-1 gp120 exclusively on the function of CD4+CD25+ Treg cells, we developed a coculture assay which contained CD8+ T cells as effectors to exclude any influence of HIV-1 gp120 on the latter. Therefore, isolated CD4+CD25+ Treg cells are cocultured with T cell-depleted syngenic PBMC and allogeneic CD8+ T effector cells. In this setting, proliferation of alloreactive CD8+ T effector cells is only suppressed by activated CD4+CD25+ Treg cells such as upon additional anti-CD3 mAb stimulation or an activating HIV-1 gp120-mediated signal. (All HIV-1 gp120 proteins: Protein Sciences Corp. Meriden, USA).
  • 1×105 isolated CD25+ Treg cells were incubated with 3×105 irradiated (50 Gy) syngenic T cell-depleted PBMC and 1×105 allogeneic CD8+ T effector cells in presence/absence of different HIV-1 gp120 preparations, addition of 0.5 μg/ml anti-CD3 (OKT-3) served as positive control. Proliferation was determined by 3H-Tdr-Incorporation 4 days later.
  • Grey bars represent proliferation of CD8+ T cells and inactivated syngenic CD3-depleted PBMC, white bars represent the proliferation of CD4+CD25+ Tregs and the black bars represent the proliferation of CD8+ T cells co-cultured with CD4+CD25+ Tregs cells and inactivated syngenic CD3-depleted PBMC. Due to inactivation PBMC don't contribute to proliferation of the samples.
  • As can be seen by the low white bars, Tregs don't show a significant proliferation under all conditions (without additional stimulus, or anti-CD3, or HIV-1 gp120 MN, or HIV-1 gp120 LAV, or HIV-1 gp120 CM). CD8+ T cells (grey bars) and CD8+ T cells co-cultured with Treg cells (black bars) show the same magnitude of proliferation under the condition without additional stimulus representing the controls. Upon anti-CD3 stimulation CD8+ T cells (grey bars) show strong increased proliferation but CD8+ T cells co-cultured with Treg cells (black bars) show a reduced proliferation indicating suppressed proliferation of CD8+ T cells by anti-CD3 activated and therefore suppressive Tregs. The effect is anti-CD3 dose-dependent. Upon stimulation with HIV-1 gp120 MN, or HIV-1 gp120 LAV, or HIV-1 gp120 CM CD8+ T cells co-cultured with Treg cells (black bars) in contrast to CD8+ T cells without Tregs (grey bars) reduced proliferation indicating HIV-1 gp120 activated Tregs. This demonstrates clearly that HIV-1 gp120 activates Tregs which subsequently exert suppressive activity on CD8+ T cells by reducing their proliferation. The suppressive activity of Treg cells and therefore the reduced proliferation is HIV-1 gp120 dose-dependent.
  • FIG. 2: Cyclic AMP measurement in CD4+CD25+ Treg cells.
  • HIV-1 gp120 treatment augments cytosolic cAMP in human CD4+CD25+ Treg cells.
  • CD4+CD25+ regulatory T cells (CD25) are left untreated (Ø) or stimulated with an anti-CD3 monoclonal Antibody (OKT-3; 1 μg/ml) or HIV-1 gp120 (14 ml). Upon 16 hours of stimulation the CD4+CD25+ regulatory T cells are lysed (1×107/ml) and the cytosolic cAMP concentration of 1×106 T cells is assessed using a cAMP-specific ELISA.
  • CD4+CD25+ regulatory T cells (CD25) are left untreated (Ø) or stimulated with an anti-CD3 monoclonal Antibody (OKT-3; 1 μg/ml) or HIV-1 gp120 (1 μg/ml). Upon 16 hours of stimulation the CD4+CD25+ regulatory T cells are lysed (1×107/ml) and the cytosolic cAMP concentration of 1×106 T cells is assessed using a cAMP-specific ELISA.
  • Bars represent the amount of cytosolic (intracellular) cyclic AMP (cAMP) in untreated Treg cells or Treg cells treated with different stimuli. The upper bar represent Tregs left untreated (Ø) and shows the basal level of intracellular cAMP. Upon anti-CD3 stimulation (OKT-3) Tregs are activated and show an increase of intracellular cAMP compared to the untreated control (upper bar) as demonstrated with the second bar. Treatment of Tregs with HIV-1 gp120 also induces an increase of intracellular cAMP as demonstrated with the lower bar. This demonstrated that activation of Treg cells with different stimuli induces an increase of cytosolic cAMP which can be used as readout for activation.
  • FIG. 3: Induction of GVHD by transfer of PBMC into NOD-Scid mice and prevention of GVHD by additional transfer of regulatory T cells (Tregs).
  • Three to six days old NOD-Scid mice are intraperitoneally injected with 1×107 human PBMC without (circles) or together with 2.5×106 human regulatory T cells (triangles). Control mice (rhombi) did not receive any PBMC. Mice having received PBMC develop a fatal GVHD, do not grow and die. Animals having additionally received regulatory T cells are protected from development of GVHD and develop normally (3 mice per group). Lack of weight increase/weight loss is used as a parameter to score GVHD severity. The diagram shows the relative body weight at different time points after transfer.
  • FIG. 4: Induction of GVHD by transfer of PBMC into NOD-Scid mice and prevention of GVHD by additional injection of HIV gp120.
  • Three to six days old NOD-Scid mice are intraperitoneally injected with 1×107 human PBMC without (circles) or together with 5 μg HIV gp120 (triangles). Control mice (rhombs) did not receive any PBMC. Mice having received PBMC develop a fatal GVHD, do not grow and die. Animals having additionally received HIV gp120 are protected from development of GVHD and develop normally (3 mice per group). Lack of weight increase/weight loss is used as a parameter to score GVHD severity. The diagram shows the relative body weight at different time points after transfer.
  • FIG. 5: Prevention of GVHD in NOD-Scid mice transferred with PBMC by additional injection of HIV gp120 depends on the presence of regulatory T cells.
  • Three to six days old NOD-Scid mice are intraperitoneally injected with 1×107 human CD25-depleted PBMC without (circles) or together with 5 μg HIV gp120 (triangles). Control mice (rhombi) do not receive any PBMC (3 mice per group). Mice having received CD25-depleted PBMC develop a fatal GVHD, do not grow and die. Animals having received 1×107 human CD25-depleted PBMC and additionally HIV gp120 are not protected from development of GVHD, also do not grow and die. Lack of weight increase/weight loss is used as a parameter to score GVHD severity. The diagram shows the relative body weight at different time points after transfer.
  • LITERATURE
    • Arthos J, Deen K C, Chaikin M A, Formwald J A, Sathe G, Sattentau Q J, Clapham P R, Weiss R A, McDougal J S, Pietropaolo C, et al.
    • Identification of the residues in human CD4 critical for the binding of HIV.
    • Cell. 1989 57:469-81
    • Bluestone J A, Tang Q.
    • Therapeutic vaccination using CD4+CD25+ antigen-specific regulatory T cells.
    • Proc Natl Acad Sci USA. 2004 101 Suppl 2:14622-6
    • Carrière, D., Vendrell, J. P., Fontaine, C., Reynes, J., Atoui, N., and Pau, B.
    • CD4 V1 domain masking on lymphocytes from HIV-1-infected patients.
    • In: Leucocyte Typing V: White Cell Differentiation Antigens, Volume 1 pp 475-476, Oxford University Press 1995, Editor: S. F. Schlossman et al.
    • Castagna A, Biswas P, Beretta A, Lazzarin A.
    • The appealing story of HIV entry inhibitors: from discovery of biological mechanisms to drug development.
    • Drugs. 2005 65:879-904
    • Culp J S, Johansen H, Hellmig B, Beck J, Matthews T J, Delers A, Rosenberg M. Regulated expression allows high level production and secretion of HIV-1 gp120 envelope glycoprotein in Drosophila Schneider cells.
    • Biotechnology (N Y). 1991 9:173-7
    • Diamond D C, Sleckman B P, Gregory T, Lasky L A, Greenstein J L, Burakoff S J.
    • Inhibition of CD4+ T cell function by the HIV envelope protein, gp120.
    • J. Immunol. 1988 141:3715-7
    • Fontenot J D, Rudensky A Y.
    • A well adapted regulatory contrivance: regulatory T cell development and the forkhead family transcription factor Foxp3.
    • Nat Immunol. 2005 6:331-7
    • Franke R, Hirsch T, Overwin H, Eichler J.
    • Synthetic Mimetics of the CD4 Binding Site of HIV-1 gp120 for the Design of Immunogens.
    • Angew Chem Int Ed Engl. 2007; January 9; [Epub ahead of print]
    • Hesselton R M, Greiner D L, Mordes J P, Rajan T V, Sullivan J L, Shultz L D.
    • High levels of human peripheral blood mononuclear cell engraftment and enhanced susceptibility to human immunodeficiency virus type I infection in NOD/LtSz-scid/scid mice.
    • J Infect Dis. 1995 172: 974-982.
    • Hoffmann P, Eder R, Kunz-Schughart L A, Andreesen R, Edinger M.
    • Large-scale in vitro expansion of polyclonal human CD4(+)CD25high regulatory T cells.
    • Blood. 2004 104:895-903
    • Horwitz D A, Zheng S G, Gray J D, Wang J H, Ohtsuka K, Yamagiwa S. Regulatory T cells generated ex vivo as an approach for the therapy of autoimmune disease.
    • Semin Immunol. 2004 16:135-43
    • Hunig T, Dennehy K.
    • CD28 superagonists: mode of action and therapeutic potential.
    • Immunol Lett. 2005 100:21-8
    • Jeffs S A, McKeating J, Lewis S, Craft H, Biram D. Stephens P E, Brady R L. Antigenicity of truncated forms of the human immunodeficiency virus type 1 envelope glycoprotein.
    • J Gen Virol. 1996 July; 77 (Pt 7):1403-10
    • Jameson B A, Rao P E, Kong L I, Hahn B H, Shaw G M, Hood L E, Kent S B. Location and chemical synthesis of a binding site for HIV-1 on the CD4 protein.
    • Science. 1988 240:1335-9
    • Kizilisik T A, al-Sebayel M.
    • Diagnosis and classification of the severity of graft versus host disease after experimental small-bowel transplantation in small animal models.
    • Transplant Proc. 1997 29:3030-3
    • Klatzmann D, Champagne E, Chamaret S, Gruest J, Guetard D, Hercend T, Gluckman J C, Montagnier L.
    • T-lymphocyte T4 molecule behaves as the receptor for human retrovirus LAV.
    • Nature. 1984 312:767-8
    • Lasky L A, Groopman J E, Fennie C W, Benz P M, Capon D J, Dowbenko D J, Nakamura G R, Nunes W M, Renz M E, Berman P W.
    • Neutralization of the AIDS retrovirus by antibodies to a recombinant envelope glycoprotein.
    • Science. 1986 233:209-12
    • Leonard C K, Spellman M W, Riddle L, Harris R J, Thomas J N, Gregory T J
    • Assignment of intrachain disulfide bonds and characterization of potential glycosylation sites of the type 1 recombinant human immunodeficiency virus envelope glycoprotein (gp120) expressed in Chinese hamster ovary cells.
    • J Biol Chem. 1990 265:10373-82
    • Lohman K L, Attanasio R, Buck D, Carrillo M A, Allan J S, Kennedy R C.
    • Characteristics of murine monoclonal anti-CD4. Epitope recognition, idiotype expression, and variable region gene sequence.
    • J. Immunol. 1992 149:3247-53
    • Markovic I, Clouse K A.
    • Recent advances in understanding the molecular mechanisms of HIV-1 entry and fusion: revisiting current targets and considering new options for therapeutic intervention.
    • Curr HIV Res. 2004 2: 223-34
    • Mizukami T, Fuerst T R, Berger E A, Moss B.
    • Binding region for human immunodeficiency virus (HIV) and epitopes for HIV-blocking monoclonal antibodies of the CD4 molecule defined by site-directed mutagenesis.
    • Proc Natl Acad Sci USA. 1988; 85:9273
    • Moreland L W, Haverty T P, Wacholtz M C, Knowles R W, Bucy R P, Heck L W Jr, Koopman W J.
    • Nondepleting humanized anti-CD4 monoclonal antibody in patients with refractory rheumatoid arthritis.
    • J. Rheumatol. 1998 25:221-8
    • Muesing M A, Smith D H, Cabradilla C D, Benton C V, Lasky L A, Capon D J.
    • Nucleic acid structure and expression of the human AIDS/lymphadenopathy retrovirus.
    • Nature. 1985 313:450-8
    • Repke H, Gabuzda D, Palu G, Emmrich F, Sodroski J.
    • Effects of CD4 synthetic peptides on HIV type I envelope glycoprotein function.
    • J. Immunol. 1992 149:1809-16
    • Robinson D S.
    • Regulation: the art of control? Regulatory T cells and asthma and allergy.
    • Thorax. 2004 59:640-3
    • Roncarolo, M. G, Bacchetta, R, Bordignon C, Narula, S, Levings, M K
    • Type 1 T regulatory cells.
    • Immunol Rev. 2001 182:68-79
    • Sakaguchi, S.
    • Naturally arising Foxp3-expressing CD25+CD4+ regulatory T cells in immunological tolerance to self and non-self.
    • Nature. 2005 6:345-352
    • Shevach E M.
    • CD4+CD25+ suppressor T cells: more questions than answers.
    • Nat Rev Immunol. 2002 2:389-400.
    • Shultz L D, Schweitzer L, Christianson S W, Gott B, Schweitzer I B, Tennent B, McKenna S, Mobraaten L, Rajan T V, Greiner D L, Leiter E H.
    • Multiple defects in innate and adaptive immunologic function in NOD/LtSz-scid mice.
    • J. Immunol. 1995 154: 180-191.
    • Starcich B R, Hahn B H, Shaw G M, McNeely P D, Modrow S, Wolf H, Parks E S, Parks W P, Josephs S F, Gallo R C, et al.
    • Identification and characterization of conserved and variable regions in the envelope gene of HTLV-III/LAV, the retrovirus of AIDS.
    • Cell. 1986 Jun. 6; 45(5):637-48
    • Tang Q, Henriksen K J, Boden E K, Tooley A J, Ye J, Subudhi S K, Zheng X X, Strom T B, Bluestone J A.
    • Cutting edge: CD28 controls peripheral homeostasis of CD4+CD25+ regulatory T cells.
    • J. Immunol. 2003 171:3348-52
    • Thornton A M, Shevach E M.
    • CD4+CD25+ immunoregulatory T cells suppress polyclonal T cell activation in vitro by inhibiting interleukin 2 production.
    • J Exp Med. 1998 188:287-96
    • Weiner, H. L.
    • Induction and mechanism of action of transforming growth factor-beta-secreting Th3 regulatory cells.
    • Immunol Rev. 2001 182:207-14
    • Yang Q E, Stephen A G, Adelsberger J W, Roberts P E, Zhu W, Currens M J, Feng Y, Crise B J, Gorelick R J, Rein A R, Fisher R J, Shoemaker R H, Sei S.
    • Discovery of small-molecule human immunodeficiency virus type 1 entry inhibitors that target the 120-binding domain of CD4.
    • J. Virol. 2005 79:6122-33
    • Zheng S G, Wang J H, Gray J D, Soucier H, Horwitz D A.
    • CD4+ and CD8+ regulatory T cells generated ex vivo with IL-2 and TGF-beta suppress a stimulatory graft-versus-host disease with a lupus-like syndrome.
    • J. Immunol. 2004 172:5213-21

Claims (16)

What is claimed is:
1. A CD4+CD25+ Treg cell activator which activates CD4+CD25+ Treg cell via interaction with the CD4+CD25+ Treg cell epitope as set forth in SEQ ID NO:1, said CD4+CD25+ Treg cell activator being a peptide, a polypeptide, or an antibody or a binding fragment thereof with the proviso that the antibody or antibody fragment is not OKT4A, OKT4D, OKTcdr4a, MAX.12H5 and not Leu3.
2. The CD4+CD25+ Treg cell activator according to claim 1 wherein said CD4+CD25+ Treg cell activator is a peptide comprising the amino acid sequence as set forth in SEQ ID NO.: 3, SEQ ID NO.: 4, SEQ ID NO.: 5, SEQ ID NO.: 6, SEQ ID NO.: 7, SEQ ID NO.: 8, SEQ ID NO.: 9, or SEQ ID NO.: 10.
3. A method of treating an individual in need thereof comprising administering a CD4+CD25+ Treg cell activator which activates CD4+CD25+ Treg cell via interaction with the CD4+CD25+ Treg cell epitope as set forth in SEQ ID NO:1, wherein said CD4+CD25+ Treg cell activator is a peptide, a polypeptide, or an antibody or a binding fragment thereof with the proviso that the antibody or antibody fragment is not OKT4A, OKT4D, OKTcdr4a, MAX.12H5 and not Leu3.
4. The method of treating an individual in need thereof according to claim 3 wherein said Treg cell activator is a peptide comprising the amino acid sequence as set forth in SEQ ID NO.: 3, SEQ ID NO.: 4, SEQ ID NO.: 5, SEQ ID NO.: 6, SEQ ID NO.: 7, SEQ ID NO.: 8, SEQ ID NO.: 9, or SEQ ID NO.: 10.
5. The method of treating an individual in need thereof according to claim 3 wherein said CD4+CD25+ Treg cell activator is HIV-1 gp120.
6. The method of treating an individual in need thereof according to claim 3 wherein said CD4+CD25+ Treg cell activator is selected from a group consisting of: NSC 13778, peptide2 which presents three HIV-1 gp120 fragments bound together through comformationally flexible scaffolds, monoclonal antibody OKT4A, monoclonal antibody OKT4D, monoclonal antibody OKTcdr4a, monoclonal antibody MAX. 12H5, and monoclonal antibody Leu3.
7. Use of a CD4+CD25+ Treg cell activator which activates CD4+CD25+ Treg cell via interaction with the CD4+CD25+ Treg cell epitope as set forth in SEQ ID NO:1 for the preparation of a medicament for the treatment of a disease selected from a group consisting of a non-autoimmune inflammatory disease, an autoimmune inflammatory disease, an inflammatory disease due to organ transplantation; a bone marrow transplantation, and a disease due to exogenously administered self or exogenously administered non-autologous recombinant polypeptide, wherein said CD4+CD25+ Treg cell activator is an antibody or an antibody fragment capable of binding to the peptide as set forth in SEQ ID NO.:1 or a peptide comprising the amino acid sequence as set forth in SEQ ID NO.: 3, SEQ ID NO.: 4, SEQ ID NO.: 5, SEQ ID NO.: 6, SEQ ID NO.: 7, SEQ ID NO.: 8, SEQ ID NO.: 9, or SEQ ID NO.: 10.
8. The use according to according to claim 7 wherein said non-autoimmune inflammatory disease is selected from a group consisting of: asthma, allergic asthma, respiratory allergy, allergic rhinoconjunctivitis, allergic alveolitis, contact allergy, atopic dermatitis, neurodermatitis, food allergy, graft-versus-host disease, non-autoimmune inflammatory bowel disease, acute respiratory distress syndrome, acute inflammatory pancreatitis, burns, wound healing, skin scarring disorders, sarcoidosis, Behcet's disease, Sweet's syndrome.
9. The use according to according to claim 7 wherein said an autoimmune inflammatory disease is selected from a group consisting of: rheumatoid arthritis, rheumatic fever, systemic lupus erythematosus, ulcerative colitis, Crohn's disease, autoimmune inflammatory bowel disease, diabetes type I, gastritis, autoimmune atrophic gastritis, autoimmune hepatitis, Hashimoto's thyroiditis, thyreoiditis, multiple sclerosis, myasthenia gravis, autoimmune haemolytic anemia, Addison's disease, scleroderma, Goodpasture's syndrome, Guillain-Barre syndrome, Graves' disease, glomerulonephritis, psoriasis, pemphigus vulgaris, pemphigoid, vitiligo, idiopathic leukopenia, Sjogren's syndrome, Wegener's granulomatosis.
10. The use according to according to claim 7 wherein said CD4+CD25+ Treg cell activator is HIV-1 gp120.
11. The use according to according to claim 7 wherein said CD4+CD25+ Treg cell activator is selected from a group consisting of: NSC 13778, peptide2 which presents three HIV-1 gp120 fragments bound together through comformationally flexible scaffolds, monoclonal antibody OKT4A, monoclonal antibody OKT4D, monoclonal antibody OKTcdr4a, monoclonal antibody MAX. 12H5, and monoclonal antibody Leu3.
12. A pharmaceutical composition comprising at least one CD4+CD25+ Treg cell activator which activates CD4+CD25+ Treg cell via interaction with the CD4+CD25+ Treg cell epitope as set forth in SEQ ID NO:1 wherein said CD4+CD25+ Treg cell activator is a peptide, a polypeptide, or an antibody or a binding fragment thereof with the proviso that the antibody or antibody fragment is not OKT4A, OKT4D, OKTcdr4a, MAX.12H5 and not Leu3.
13. The pharmaceutical composition according to claim 12 wherein said CD4+CD25+ Treg cell activator is a peptide comprising the amino acid sequence as set forth in SEQ ID NO.: 3, SEQ ID NO.: 4, SEQ ID NO.: 5, SEQ ID NO.: 6, SEQ ID NO.: 7, SEQ ID NO.: 8, SEQ ID NO.: 9, or SEQ ID NO.: 10.
14. A method for reducing or preventing an unwanted immune reaction due to a exogenously administered self or exogenously administered non-autologous recombinant polypeptide comprising administering to a being in need thereof a suitable amount of a pharmaceutical composition comprising a CD4+CD25+ Treg cell activator which is an antibody or an antibody fragment capable of binding to the peptide as set forth in SEQ ID NO.:1 or is a peptide comprising the amino acid sequence as set forth in SEQ ID NO.: 3, SEQ ID NO.: 4, SEQ ID NO.: 5, SEQ ID NO.: 6, SEQ ID NO.: 7, SEQ ID NO.: 8, SEQ ID NO.: 9, or SEQ ID NO.: 10.
15. The method according to claim 14 wherein said CD4+CD25+ Treg cell activator is HIV-1 gp120.
16. The method according to claim 14 wherein said Treg cell activator is selected from a group consisting of: NSC 13778, peptide 2 which presents three HIV-1 gp120 fragments bound together through comformationally flexible scaffolds, monoclonal antibody OKT4A, monoclonal antibody OKT4D, monoclonal antibody OKTcdr4a, monoclonal antibody MAX. 12H5, and monoclonal antibody Leu3.
US14/031,919 2007-02-01 2013-09-19 Screening Method for the Identification of Agents Capable of Activating CD4+CD25+ Regulatory T-Cells Through Interactions with the HIV-1 GP120 Binding Site on CD4 Abandoned US20140093509A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US14/031,919 US20140093509A1 (en) 2007-02-01 2013-09-19 Screening Method for the Identification of Agents Capable of Activating CD4+CD25+ Regulatory T-Cells Through Interactions with the HIV-1 GP120 Binding Site on CD4
US15/285,561 US10729742B2 (en) 2007-02-01 2016-10-05 Screening method for the identification of agents capable of activating CD4+CD25+ regulatory T-cells through interactions with the HIV-1 GP120 binding site on CD4
US16/912,199 US20200316167A1 (en) 2007-02-01 2020-06-25 Screening method for the identification of agents capable of activating CD4+CD25+ regulatory T-cells through interactions with the HIV-1 GP120 binding site on CD4

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
EP07101604 2007-02-01
EP07101604.2 2007-02-01
EP07122424.0 2007-12-05
EP07122424 2007-12-05
PCT/EP2008/051144 WO2008092905A2 (en) 2007-02-01 2008-01-30 Specific activation of a regulatory t cell and its use for treatment of asthma, allergic disease, autoimmune disease, graft rejection and for tolerance induction
US52514209A 2009-10-07 2009-10-07
US14/031,919 US20140093509A1 (en) 2007-02-01 2013-09-19 Screening Method for the Identification of Agents Capable of Activating CD4+CD25+ Regulatory T-Cells Through Interactions with the HIV-1 GP120 Binding Site on CD4

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
PCT/EP2008/051144 Division WO2008092905A2 (en) 2007-02-01 2008-01-30 Specific activation of a regulatory t cell and its use for treatment of asthma, allergic disease, autoimmune disease, graft rejection and for tolerance induction
US12/525,142 Division US8557533B2 (en) 2007-02-01 2008-01-30 Screening method for the identification of agents capable of activating CD4+CD25+ regulatory T-cells through interactions with the HIV-1 GP120 binding site on CD4

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US15/285,561 Division US10729742B2 (en) 2007-02-01 2016-10-05 Screening method for the identification of agents capable of activating CD4+CD25+ regulatory T-cells through interactions with the HIV-1 GP120 binding site on CD4

Publications (1)

Publication Number Publication Date
US20140093509A1 true US20140093509A1 (en) 2014-04-03

Family

ID=39363828

Family Applications (4)

Application Number Title Priority Date Filing Date
US12/525,142 Active 2030-01-15 US8557533B2 (en) 2007-02-01 2008-01-30 Screening method for the identification of agents capable of activating CD4+CD25+ regulatory T-cells through interactions with the HIV-1 GP120 binding site on CD4
US14/031,919 Abandoned US20140093509A1 (en) 2007-02-01 2013-09-19 Screening Method for the Identification of Agents Capable of Activating CD4+CD25+ Regulatory T-Cells Through Interactions with the HIV-1 GP120 Binding Site on CD4
US15/285,561 Active 2028-06-24 US10729742B2 (en) 2007-02-01 2016-10-05 Screening method for the identification of agents capable of activating CD4+CD25+ regulatory T-cells through interactions with the HIV-1 GP120 binding site on CD4
US16/912,199 Abandoned US20200316167A1 (en) 2007-02-01 2020-06-25 Screening method for the identification of agents capable of activating CD4+CD25+ regulatory T-cells through interactions with the HIV-1 GP120 binding site on CD4

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US12/525,142 Active 2030-01-15 US8557533B2 (en) 2007-02-01 2008-01-30 Screening method for the identification of agents capable of activating CD4+CD25+ regulatory T-cells through interactions with the HIV-1 GP120 binding site on CD4

Family Applications After (2)

Application Number Title Priority Date Filing Date
US15/285,561 Active 2028-06-24 US10729742B2 (en) 2007-02-01 2016-10-05 Screening method for the identification of agents capable of activating CD4+CD25+ regulatory T-cells through interactions with the HIV-1 GP120 binding site on CD4
US16/912,199 Abandoned US20200316167A1 (en) 2007-02-01 2020-06-25 Screening method for the identification of agents capable of activating CD4+CD25+ regulatory T-cells through interactions with the HIV-1 GP120 binding site on CD4

Country Status (6)

Country Link
US (4) US8557533B2 (en)
EP (2) EP3112866B1 (en)
JP (2) JP5867795B2 (en)
CA (1) CA2676170C (en)
ES (2) ES2825718T3 (en)
WO (1) WO2008092905A2 (en)

Families Citing this family (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007127787A2 (en) 2006-04-25 2007-11-08 Joslin Diabetes Center, Inc. Insulin autoantigen-specific regulatory cd4+ t cells
WO2008092905A2 (en) * 2007-02-01 2008-08-07 Boehringer Ingelheim International Gmbh Specific activation of a regulatory t cell and its use for treatment of asthma, allergic disease, autoimmune disease, graft rejection and for tolerance induction
RU2539110C2 (en) 2008-03-13 2015-01-10 Биотест Аг Method of treating autoimmune disease (versions)
SG190627A1 (en) 2008-03-13 2013-06-28 Biotest Ag Agent for treating disease
MX2010010026A (en) 2008-03-13 2011-03-21 Biotest Ag Agent for treating disease.
WO2010000730A1 (en) * 2008-06-30 2010-01-07 Universitätsklinikum Heidelberg Immunosuppressive blood cells and methods of producing same
GB0920944D0 (en) 2009-11-30 2010-01-13 Biotest Ag Agents for treating disease
EP2471543A1 (en) * 2010-12-02 2012-07-04 Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V. Tolerance induction or immunosupression to prevent in particular Graft-versus-Host-Disease (GvHD) by short-term pre-incubation of transplanted cell suspensions, tissues or organs coated with ligands to cell surface molecules
WO2012135842A2 (en) * 2011-03-31 2012-10-04 President And Fellows Of Harvard College A unique population of regulatory t cells that regulate tissue regeneration and wound healing
EP3063271A4 (en) * 2013-10-31 2017-05-03 Sloan-Kettering Institute for Cancer Research Methods and compositions for inducing regulatory t-cell generation
WO2019099788A1 (en) * 2017-11-16 2019-05-23 Benaroya Sesearch Institute At Virginia Mason Antigen-specific t regulatory cell assay

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6585979B1 (en) * 1996-07-08 2003-07-01 Genentech, Inc. HIV envelope polypeptides and immunogenic composition

Family Cites Families (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4695459A (en) * 1984-12-26 1987-09-22 The Board Of Trustees Of Leland Stanford Junior University Method of treating autoimmune diseases that are mediated by Leu3/CD4 phenotype T cells
IE872748L (en) * 1986-10-16 1988-04-16 Arjomari Europ Polypeptides derived from the evvelope gene of human¹immunodeficiency virus in recombinant baculovirus infected¹insect cells
JPH0768273B2 (en) 1988-12-02 1995-07-26 カルピス食品工業株式会社 Anti-HIV peptide
CA2066607A1 (en) * 1989-09-22 1991-03-23 Thomas Kieber-Emmons Peptides associated with the cd4 binding region of gp120 and their methods of use
GB8928874D0 (en) 1989-12-21 1990-02-28 Celltech Ltd Humanised antibodies
EP0789076A3 (en) 1996-02-07 1999-04-14 Takeda Chemical Industries, Ltd. G protein coupled receptor proteins, their production and use
EP1066047A4 (en) * 1998-03-25 2004-10-13 Mayo Foundation For Medicaledu Methods and materials for treating inflammatory diseases
US7608273B2 (en) * 1998-08-12 2009-10-27 University Of Western Ontario Recombinant lentivirus encoding modified GP 120 signal sequences
US6337181B1 (en) * 1998-12-21 2002-01-08 Jeffrey Joseph Stewart Method of specifying vaccine components for viral quasispecies
CA2358385C (en) * 1998-12-31 2013-08-06 Chiron Corporation Polynucleotides encoding antigenic hiv type c polypeptides, polypeptides and uses thereof
SE9904660D0 (en) 1999-12-17 1999-12-17 Astra Pharma Inc Novel assays
EP1241249A1 (en) 2001-03-12 2002-09-18 Gerold Schuler CD4+CD25+regulatory T cells from human blood
AU2003205627A1 (en) 2002-02-01 2003-09-02 Bayer Healthcare Ag Diagnostics and therapeutics for diseases associated with gpr72
US20070009986A1 (en) * 2003-01-30 2007-01-11 Poznansky Mark C Compositions for modulating immune cell activity and methods for detection thereof
EP1460088A1 (en) * 2003-03-21 2004-09-22 Biotest AG Humanized anti-CD4 antibody with immunosuppressive properties
US20050048587A1 (en) * 2003-07-17 2005-03-03 Tolerrx, Inc. Methods for identifying tolerance modulatory compounds and uses therefor
SG153796A1 (en) * 2004-06-08 2009-07-29 Novartis Vaccines & Diagnostic Fusion proteins comprising cd4 minimal modules and methods of use thereof
JP2009509142A (en) 2005-09-19 2009-03-05 エーザイ・アール・アンド・ディー・マネジメント株式会社 Methods for identifying GPR83 agonists and GPR83 antagonists capable of modulating regulatory T cell function
WO2008092905A2 (en) 2007-02-01 2008-08-07 Boehringer Ingelheim International Gmbh Specific activation of a regulatory t cell and its use for treatment of asthma, allergic disease, autoimmune disease, graft rejection and for tolerance induction

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6585979B1 (en) * 1996-07-08 2003-07-01 Genentech, Inc. HIV envelope polypeptides and immunogenic composition

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
Huang, W., et al., June 2008, Correceptor tropism can be influenced by amino acid substitutions in the gp41 transmembrane subunit of human immunodeficiency virus type 1 envelope protein, J. Virol. 82(11):5584-5593. *
Lee, S.-K., et al., 2000, A single point mutation in HIV-1 V3 loop alters the immunogenic properties of rgp120, Arch. Virol. 145:2087-2103. *

Also Published As

Publication number Publication date
EP2109770A2 (en) 2009-10-21
US20170014474A1 (en) 2017-01-19
JP6038070B2 (en) 2016-12-07
WO2008092905A3 (en) 2008-10-09
CA2676170C (en) 2021-01-05
ES2825718T3 (en) 2021-05-17
EP3112866A1 (en) 2017-01-04
ES2593789T3 (en) 2016-12-13
WO2008092905A2 (en) 2008-08-07
JP5867795B2 (en) 2016-02-24
US20100074904A1 (en) 2010-03-25
US10729742B2 (en) 2020-08-04
EP3112866B1 (en) 2020-07-29
US20200316167A1 (en) 2020-10-08
US8557533B2 (en) 2013-10-15
JP2010518810A (en) 2010-06-03
EP2109770B1 (en) 2016-07-06
JP2014156477A (en) 2014-08-28
CA2676170A1 (en) 2008-08-07

Similar Documents

Publication Publication Date Title
US20200316167A1 (en) Screening method for the identification of agents capable of activating CD4+CD25+ regulatory T-cells through interactions with the HIV-1 GP120 binding site on CD4
Lu et al. Major peptide autoepitopes for nucleosome-specific T cells of human lupus
Rininsland et al. Granzyme B ELISPOT assay for ex vivo measurements of T cell immunity
Kaneko et al. Impaired induction of cytotoxic T lymphocytes by antagonism of a weak agonist borne by a variant hepatitis C virus epitope
US20150094213A1 (en) Comparative Ligand Mapping from MHC Positive Cells
Martin et al. HLA-DR, ICAM-1, CD40, CD40L, and CD86 are incorporated to a similar degree into clinical human immunodeficiency virus type 1 variants expanded in natural reservoirs such as peripheral blood mononuclear cells and human lymphoid tissue cultured ex vivo
Herberts et al. Autoreactivity against induced or upregulated abundant self-peptides in HLA-A* 0201 following measles virus infection
Oldstone The role of cytotoxic T lymphocytes in infectious disease: history, criteria, and state of the art
Le et al. Ebola virus protein VP40 stimulates IL-12–and IL-18–dependent activation of human natural killer cells
AU769538B2 (en) Efficient ex vivo expansion of CD4+ and CD8+ T-cells from HIV infected subjects
WO2004084838A2 (en) Identification, quantification, and characterization of t cells and t cell antigens
Sarikonda et al. Immunosuppressive mechanisms during viral infectious diseases
JP5133706B2 (en) Hepatitis C virus-derived peptide
Su et al. Novel simian immunodeficiency virus CTL epitopes restricted by MHC class I molecule Mamu-B* 01 are highly conserved for long term in DNA/MVA-vaccinated, SHIV-challenged rhesus macaques
Tsunetsugu-Yokota et al. Transcriptional regulation of HIV-1 LTR during antigen-dependent activation of primary T cells by dendritic cells
Aucher et al. Could CD4 capture by CD8+ T cells play a role in HIV spreading?
Kawashima et al. Identification and characterization of HIV-1 epitopes presented by HLA-A* 2603: comparison between HIV-1 epitopes presented by A* 2601 and A* 2603
Davey et al. T-cell clones derived by CD3 stimulation from hepatitis C virus explanted liver tissue are not representative of dominant clones present in vivo
Aucher et al. Could CD4 Capture by T Cells Play a Role in HIV Spreading?
Archila Diaz Assessment of allergen specific response in humans
Aucher et al. Research Article Could CD4 Capture by CD8+ T Cells Play a Role in HIV Spreading?
Surface The Protein Tyrosine Kinase p56
Borghan et al. Cytotoxic T Lymphocytes on Selection of Escape Mutant Viruses"
Sekaly et al. Enhancement of HIV-Specific CD8 T Cell
Parsons Lessons learned: natural killer cell education as a determinant of the anti-viral functional potential of natural killer cells

Legal Events

Date Code Title Description
AS Assignment

Owner name: JONULEIT, HELMUT, GERMANY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:BOEHRINGER INGELHEIM INTERNATIONAL GMBH;REEL/FRAME:031511/0342

Effective date: 20121213

Owner name: TCF GMBH, GERMANY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:JONULEIT, HELMUT;REEL/FRAME:031511/0530

Effective date: 20130916

Owner name: BOEHRINGER INGELHEIM INTERNATIONAL GMBH, GERMANY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:SCHNEIDER, FRANZ-JOSEF;BECKER, CHRISTIAN;BOPP, TOBIAS;AND OTHERS;SIGNING DATES FROM 20090809 TO 20090826;REEL/FRAME:031511/0102

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION