US20110144309A1 - Manipulation of Cytokine Levels Using CD83 Gene Products - Google Patents

Manipulation of Cytokine Levels Using CD83 Gene Products Download PDF

Info

Publication number
US20110144309A1
US20110144309A1 US12/941,689 US94168910A US2011144309A1 US 20110144309 A1 US20110144309 A1 US 20110144309A1 US 94168910 A US94168910 A US 94168910A US 2011144309 A1 US2011144309 A1 US 2011144309A1
Authority
US
United States
Prior art keywords
seq
antibody
cells
antibodies
variable region
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
US12/941,689
Inventor
Fred Ramsdell
Sean C. Proll
Karen Staehling-Hampton
Mark W. Appleby
Leon Fernando Garcia-Martinez
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.)
Celltech R&D Inc
Original Assignee
Celltech R&D Inc
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 Celltech R&D Inc filed Critical Celltech R&D Inc
Priority to US12/941,689 priority Critical patent/US20110144309A1/en
Publication of US20110144309A1 publication Critical patent/US20110144309A1/en
Assigned to CELLTECH R & D, INC. reassignment CELLTECH R & D, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: PROLL, SEAN, APPLEBY, MARK W, HAMPTON, KAREN STAEHLING, PAEPER, BRYAN
Assigned to CELLTECH R & D, INC. reassignment CELLTECH R & D, INC. CORRECTIVE ASSIGNMENT TO CORRECT THE NAMES OF ASSIGNEES AND EXECUTION DATE OF ASSIGNMENT PREVIOUSLY RECORDED ON REEL 026886, FRAME 0397. Assignors: RAMSDELL, FRED, PROLL, SEAN C, APPLEBY, MARK W, HAMPTON, KAREN STAEHLING, MARTINEZ, LEON FERNANDO GARCIA
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • 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
    • A61P1/16Drugs 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
    • 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
    • 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
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/02Drugs for genital or sexual disorders; Contraceptives for disorders of the vagina
    • 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
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/14Drugs for dermatological disorders for baldness or alopecia
    • 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
    • A61P21/04Drugs 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
    • A61P27/14Decongestants or antiallergics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/16Otologicals
    • 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
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • A61P31/06Antibacterial agents for tuberculosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/16Antivirals for RNA viruses for influenza or rhinoviruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • A61P31/22Antivirals for DNA viruses for herpes viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • A61P33/02Antiprotozoals, e.g. for leishmaniasis, trichomoniasis, toxoplasmosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • A61P33/10Anthelmintics
    • 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
    • A61P37/00Drugs for immunological or allergic disorders
    • 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
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the invention relates to an altered CD83 gene product, and methods of modulating cytokine levels by modulating the expression of mutant and wild type CD83 gene products produced in a mammal.
  • the invention also relates to the regulation of T cell and dendritic cell activity and conditions and treatments related thereto.
  • CD83 is a 45 kilodalton glycoprotein that is predominantly expressed on the surface of dendritic cells and other cells of the immune system. Structural analysis of the predicted amino acid sequence of CD83 indicates that it is a member of the immunoglobulin superfamily. See, Zhou et al., J. Immunol. 149:735 (1992)). U.S. Pat. No. 5,316,920 and WO 95/29236 disclose further information about CD83. While such information suggests that CD83 plays a role in the immune system, that role is undefined, and the interrelationship of CD83 with cellular factors remains unclear.
  • the invention provides a method of modulating cytokine levels by modulating the activity or expression of the CD83 gene products.
  • cytokine levels can be modulated in a mammal or in mammalian cells that are involved in the immune response, for example, antigen presenting cells or T cells.
  • the invention therefore provides a method of modulating cytokine production in a mammal or in an immune cell by modulating the activity or expression of a CD83 polypeptide.
  • the production of a cytokine such as interleukin-2, interleukin-4, or interleukin-10 can be modulated by modulating the activity or expression of a CD83 polypeptide.
  • an antibody is used that can modulate the activity or expression of a CD83 polypeptide.
  • the immune cells are T cells or antigen presenting cells.
  • the immune cells are CD4+ T cells.
  • the invention also provides a method of modulating granulocyte macrophage colony stimulating factor production in a mammal or in an immune cell by modulating the activity or expression of CD83 polypeptides.
  • an antibody is used that can modulate the activity or expression of a CD83 polypeptide.
  • the antibody can be administered to the mammal or the immune cell can be contacted with the antibody.
  • the immune cells are T cells or antigen presenting cells. In other embodiments, the immune cells are CD4+ T cells.
  • the invention also provides a method of modulating tumor necrosis factor production in a mammal or in a mammalian cell by modulating the activity or expression of CD83 polypeptides.
  • an antibody is used that can modulate the activity or expression of a CD83 polypeptide.
  • the antibody can be administered to the mammal or the mammalian cell can be contacted with the antibody.
  • the immune cells are T cells or antigen presenting cells. In other embodiments, the immune cells are CD4+ T cells.
  • the invention further provides a method of inhibiting proliferation of a human peripheral blood mononuclear cell by modulating the activity or expression of CD83 polypeptides.
  • an antibody is used that can modulate the activity or expression of a CD83 polypeptide.
  • the antibody can be administered to the mammal or the human peripheral blood mononuclear cell can be contacted with the antibody.
  • the invention also provides an antibody that can bind to a CD83 polypeptide comprising SEQ ID NO:4, SEQ ID NO:8 or SEQ ID NO:9, wherein activated CD4 + T-cells produce lower levels of interleukin-4 when the T-cells are contacted with the antibody.
  • the invention further provides an antibody that can bind to a CD83 polypeptide comprising SEQ ID NO:4, SEQ ID NO:8 or SEQ ID NO:9, wherein CD4 + T-cells proliferation is decreased when the T-cells are contacted with the antibody.
  • Such an antibody can have an amino acid sequence that includes SEQ ID NO:11, SEQ ID NO:13, SEQ ID NO:15, SEQ ID NO:17, SEQ ID NO:19, SEQ ID NO:21, SEQ ID NO:23, SEQ ID NO:24, SEQ ID NO:25, SEQ ID NO:26, SEQ ID NO:27, SEQ ID NO:28, SEQ ID NO:29, SEQ ID NO:30, SEQ ID NO:31, SEQ ID NO:32, SEQ ID NO:33, SEQ ID NO:34, SEQ ID NO:35, SEQ ID NO:36, SEQ ID NO:37, SEQ ID NO:38, SEQ ID NO:39, SEQ ID NO:40, SEQ ID NO:41, SEQ ID NO:42, SEQ ID NO:43, SEQ ID NO:44, SEQ ID NO:45, SEQ ID NO:46, SEQ ID NO:47, SEQ ID NO:48, SEQ ID NO:52, SEQ ID NO:53, SEQ ID NO:
  • Nucleic acids encoding such an antibody can have, for example, a sequence that includes SEQ ID NO:12, SEQ ID NO:14, SEQ ID NO:16, SEQ ID NO:18, SEQ ID NO:20, SEQ ID NO:22, SEQ ID NO:59, SEQ ID NO:61, SEQ ID NO:63 or SEQ ID NO:65.
  • the invention also provides a method for decreasing the activity of a CD83 gene product, comprising contacting the CD83 gene product with an antibody that comprises SEQ ID NO:11, SEQ ID NO:13, SEQ ID NO:15, SEQ ID NO:17, SEQ ID NO:19, SEQ ID NO:21, SEQ ID NO:23, SEQ ID NO:24, SEQ ID NO:25, SEQ ID NO:26, SEQ ID NO:27, SEQ ID NO:28, SEQ ID NO:29, SEQ ID NO:30, SEQ ID NO:31, SEQ ID NO:32, SEQ ID NO:33, SEQ ID NO:34, SEQ ID NO:35, SEQ ID NO:36, SEQ ID NO:37, SEQ ID NO:38, SEQ ID NO:39, SEQ ID NO:40, SEQ ID NO:41, SEQ ID NO:42, SEQ ID NO:43, SEQ ID NO:44, SEQ ID NO:45, SEQ ID NO:46, SEQ ID NO:47, SEQ ID NO
  • the invention further provides a method for decreasing the translation of a CD83 gene product in a mammalian cell, comprising contacting the mammalian cell with a nucleic acid complementary to a CD83 nucleic acid comprising SEQ ID NO:1, SEQ ID NO:3, SEQ ID NO:5, or SEQ ID NO:10.
  • the invention provides a method for decreasing the translation of a CD83 gene product in a mammal, comprising administering to the mammal a nucleic acid complementary to a CD83 nucleic acid comprising SEQ ID NO:1, SEQ ID NO:3, SEQ ID NO:5, or SEQ ID NO:10.
  • the invention further provides a method for decreasing proliferation of CD4+ T-cells in a mammal comprising administering to the mammal an antibody that can bind to a CD83 gene product, wherein the CD83 gene product comprises SEQ ID NO:2 or SEQ ID NO:9.
  • the antibody can have a sequence comprising SEQ ID NO:11, SEQ ID NO:13, SEQ ID NO:15, SEQ ID NO:17, SEQ ID NO:19, SEQ ID NO:21, SEQ ID NO:23, SEQ ID NO:24, SEQ ID NO:25, SEQ ID NO:26, SEQ ID NO:27, SEQ ID NO:28, SEQ ID NO:29, SEQ ID NO:30, SEQ ID NO:31, SEQ ID NO:32, SEQ ID NO:33, SEQ ID NO:34, SEQ ID NO:35, SEQ ID NO:36, SEQ ID NO:37, SEQ ID NO:38, SEQ ID NO:39, SEQ ID NO:40, SEQ ID NO:41, SEQ ID NO:42, SEQ ID NO:43, SEQ ID NO:44, SEQ ID NO:45, SEQ ID NO:46, SEQ ID NO:47, SEQ ID NO:48, SEQ ID NO:52, SEQ ID NO:53, SEQ ID NO:54,
  • the invention also provides a method for decreasing interleukin-2 levels and increasing interleukin-4 levels in a mammal comprising administering to the mammal an antibody that can bind to a CD83 gene product, wherein the CD83 gene product comprises SEQ ID NO:2 or SEQ ID NO:9.
  • the antibody can have a sequence comprising SEQ ID NO:11, SEQ ID NO:13, SEQ ID NO:15, SEQ ID NO:17, SEQ ID NO:19, SEQ ID NO:21, SEQ ID NO:23, SEQ ID NO:24, SEQ ID NO:25, SEQ ID NO:26, SEQ ID NO:27, SEQ ID NO:28, SEQ ID NO:29, SEQ ID NO:30, SEQ ID NO:31, SEQ ID NO:32, SEQ ID NO:33, SEQ ID NO:34, SEQ ID NO:35, SEQ ID NO:36, SEQ ID NO:37, SEQ ID NO:38, SEQ ID NO:39, SEQ ID NO:40, SEQ ID NO:41, SEQ ID NO:42, SEQ ID NO:43, SEQ ID NO:44, SEQ ID NO:45, SEQ ID NO:46, SEQ ID NO:47, SEQ ID NO:48, SEQ ID NO:52, SEQ ID NO:53, SEQ ID NO:54,
  • the invention further provides a method for decreasing interleukin-2 levels and increasing interleukin-4 levels in a mammal comprising administering to the mammal a nucleic acid complementary to a CD83 nucleic acid comprising SEQ ID NO:1, SEQ ID NO:3, SEQ ID NO:5, or SEQ ID NO:10.
  • the interleukin-2 levels are decreased and the interleukin-4 levels are increased to treat an autoimmune disease.
  • the interleukin-2 levels are decreased and the interleukin-4 levels are increased to stimulate production of Th2-associated cytokines in transplant recipients, for example, to prolong survival of transplanted tissues.
  • the invention also provides a method for increasing interleukin-10 levels in a mammal comprising administering to the mammal an antibody that can bind to a CD83 gene product, wherein the CD83 gene product comprises SEQ ID NO:2 or SEQ ID NO:9.
  • the antibody can have a sequence comprising SEQ ID NO:11, SEQ ID NO:13, SEQ ID NO:15, SEQ ID NO:17, SEQ ID NO:19, SEQ ID NO:21, SEQ ID NO:23, SEQ ID NO:24, SEQ ID NO:25, SEQ ID NO:26, SEQ ID NO:27, SEQ ID NO:28, SEQ ID NO:29, SEQ ID NO:30, SEQ ID NO:31, SEQ ID NO:32, SEQ ID NO:33, SEQ ID NO:34, SEQ ID NO:35, SEQ ID NO:36, SEQ ID NO:37, SEQ ID NO:38, SEQ ID NO:39, SEQ ID NO:40, SEQ ID NO:41, SEQ ID NO:42, SEQ ID NO:43, SEQ ID NO:44, SEQ ID NO:45, SEQ ID NO:46, SEQ ID NO:47, SEQ ID NO:48, SEQ ID NO:52, SEQ ID NO:53, SEQ ID NO:54,
  • the invention further provides a method for increasing interleukin-10 levels in a mammal comprising administering to the mammal a nucleic acid complementary to a CD83 nucleic acid comprising SEQ ID NO:1, SEQ ID NO:3, SEQ ID NO:5, or SEQ ID NO:10.
  • the interleukin-10 levels are increased to treat neoplastic disease. In other embodiments, the interleukin-10 levels are increased to treat a tumor.
  • the invention also provides a method for increasing interleukin-2 levels in a mammal comprising administering to the mammal a functional CD83 polypeptide that comprises SEQ ID NO:9.
  • the invention further provides a method for increasing interleukin-2 levels in a mammal comprising: (a) transforming a T cell from the mammal with a nucleic acid encoding a functional CD83 polypeptide operably linked to a promoter functional in a mammalian cell, to generate a transformed T cell; (b) administering the transformed T cell to the mammal to provide increased levels of interleukin-2.
  • the CD83 polypeptide has a sequence that comprises SEQ ID NO:9 or the nucleic acid has a sequence that comprises SEQ ID NO:1, SEQ ID NO:3, SEQ ID NO:5, or SEQ ID NO:10.
  • Such methods for increasing interleukin-2 levels can be used to treat an allergy or an infectious disease.
  • the invention also provides a method for increasing granulocyte macrophage colony stimulating factor levels in a mammal comprising administering to the mammal an antibody that can bind to a CD83 gene product, wherein the CD83 gene product comprises SEQ ID NO:2 or SEQ ID NO:9.
  • Such an antibody can have a sequence comprising SEQ ID NO:11, SEQ ID NO:13, SEQ ID NO:15, SEQ ID NO:17, SEQ ID NO:19, SEQ ID NO:21, SEQ ID NO:23, SEQ ID NO:24, SEQ ID NO:25, SEQ ID NO:26, SEQ ID NO:27, SEQ ID NO:28, SEQ ID NO:29, SEQ ID NO:30, SEQ ID NO:31, SEQ ID NO:32, SEQ ID NO:33, SEQ ID NO:34, SEQ ID NO:35, SEQ ID NO:36, SEQ ID NO:37, SEQ ID NO:38, SEQ ID NO:39, SEQ ID NO:40, SEQ ID NO:41, SEQ ID NO:42, SEQ ID NO:43, SEQ ID NO:44, SEQ ID NO:45, SEQ ID NO:46, SEQ ID NO:47, SEQ ID NO:48, SEQ ID NO:52, SEQ ID NO:53, SEQ ID NO:54
  • the invention further provides a method for increasing granulocyte macrophage colony stimulating factor levels in a mammal comprising administering to the mammal a nucleic acid complementary to a CD83 nucleic acid comprising SEQ ID NO:1, SEQ ID NO:3, SEQ ID NO:5, or SEQ ID NO:10.
  • the invention also provides a method for increasing tumor necrosis factor levels at a selected site in a mammal comprising administering to the site a functional CD83 polypeptide.
  • the invention provides a method for increasing tumor necrosis factor levels in a selected mammalian cell comprising transforming the cell with a nucleic acid encoding a functional CD83 polypeptide.
  • the CD83 polypeptide employed can, for example, have a sequence comprising SEQ ID NO:9.
  • Mammals and birds may be treated by the methods and compositions described and claimed herein. Such mammals and birds include humans, dogs, cats, and livestock, for example, horses, cattle, sheep, goats, chickens, turkeys and the like.
  • the invention further provides a mutant mouse that can serve as an animal model of diminished T cell activation or altered cytokine levels.
  • the mutant mouse has an altered CD83 gene that produces a larger gene product, having SEQ ID NO:4 or containing SEQ ID NO:8.
  • methods of using the mutant mouse model to study the effects of cytokines on the immune system, inflammation, the function and regulation of CD83, T cell and dendritic cell activity, the immune response and conditions and treatments related thereto.
  • the invention further provides a mutant mouse whose somatic and germ cells comprise a mutant CD83 gene encoding a polypeptide comprising SEQ ID NO:4 or SEQ ID NO:8, wherein expression of the mutant CD83 gene reduces CD4+ T cell activation.
  • the mutant CD83 gene can, for example, comprise SEQ ID NO:3.
  • the invention further provides a method of identifying a compound that can modulate CD4+ T cell activation comprising administering a test compound to a mouse having a mutant or wild type transgenic CD83 gene and observing whether CD4+ T cell activation is decreased or increased.
  • the somatic and/or germ cells of the mutant mouse can comprise a mutant CD83 gene encoding a polypeptide comprising SEQ ID NO:4 or SEQ ID NO:8.
  • the somatic and/or germ cells of the mouse can contain a wild type CD83 gene, for example, SEQ ID NO:1 or SEQ ID NO:9.
  • the invention also provides a mutant CD83 gene encoding a polypeptide comprising SEQ ID NO:4 or SEQ ID NO:8.
  • the invention further provides a mutant CD83 gene comprising nucleotide sequence SEQ ID NO:3.
  • FIG. 1 provides flow cytometry data for G3 animals. As shown, reduced numbers of CD4+ T cells are seen in two animals from Pedigree 9, mouse 9.4.1 and mouse 9.4.9. All other animals analyzed on that day exhibit normal numbers of CD4+ T cells.
  • FIG. 2 provides a graph of flow cytometry data for G3 animals. Each diamond symbol represents an individual animal. As shown, multiple animals from the N2 generation exhibit a reduced percentage of CD4+ T cells.
  • FIG. 3 provides the nucleotide sequence of wild type mouse CD83 (SEQ ID NO:1). The ATG start codon and the TGA stop codon are underlined.
  • FIG. 4A-B provides the nucleotide sequence of the mutant CD83 gene (SEQ ID NO:3) of the invention derived from the mutant LCD4.1 animal.
  • the ATG start codon, the mutation and the TGA stop codon are underlined.
  • FIG. 5 provides the amino acid sequence for wild type (top, SEQ ID NO:2) and mutant (bottom, SEQ ID NO:4) CD83 coding regions.
  • the additional C-terminal sequences arising because of the CD83 mutation are underlined.
  • FIG. 6A illustrates that dendritic cells from wild type ( ⁇ , WT DC) and mutant ( ⁇ , mutant DC) mice are capable of the allogeneic activation of CD4+ T cells.
  • CD4+ T cells were stimulated with 10,000, 1000 or 100 dendritic cells for 5 days and proliferation measured by incorporation of tritiated thymidine.
  • FIG. 6B illustrates that CD4+ T cells from mutant mice ( ⁇ , mutant CD4) fail to respond to allogeneic stimulation with BALBc dendritic cells, although wild type animals ( ⁇ , WT CD4+) respond normally.
  • CD4+ T cells were stimulated with 10,000, 1000 or 100 dendritic cells for 5 days and proliferation measured by incorporation of tritiated thymidine.
  • FIG. 7 provides a bar graph illustrating IL-2, IL-4, IL-5, TNF ⁇ , and IFN ⁇ production from wild type CD4+ T cells (white bar) or CD83 mutant CD4+ T cells (dark bar) that had been stimulated with 1 ⁇ g/ml of anti-CD3 antibodies and 0.2 ⁇ g/ml of anti-CD28 antibodies for 72 hours. As illustrated, IL-2 levels are lower, and IL-4 levels are higher in the CD83 mutant T cells.
  • FIG. 8 provides a bar graph illustrating IL-10 production from wild type CD4+ T cells (white bar) or CD83 mutant CD4+ T cells (dark bar) that had been stimulated with 0.1 ⁇ g/ml of anti-CD28 antibodies and 1 to 10 ⁇ g/ml of anti-CD3 antibodies for 72 hours. As illustrated, IL-10 levels are higher in the CD83 mutant T cells.
  • FIG. 9 provides a bar graph illustrating GM-CSF production from wild type CD4+ T cells (white bar) or CD83 mutant CD4+ T cells (dark bar) that had been stimulated with anti-CD3 and anti-CD28 antibodies. As illustrated, GM-CSF production is higher in the CD83 mutant cells than in wild type cells.
  • FIG. 10A provides a bar graph illustrating IL-4 mRNA levels from wild type CD4+ T cells (white bar) or CD83 mutant CD4+ T cells (dark bar) that had been stimulated with anti-CD3 and anti-CD28 antibodies. As illustrated, the IL-4 mRNA levels are higher in the CD83 mutant cells.
  • FIG. 10B provides a bar graph illustrating IL-10 mRNA levels from wild type CD4+ T cells (white bar) or CD83 mutant CD4+ T cells (dark bar) that had been stimulated with anti-CD3 and anti-CD28 antibodies. As illustrated, the IL-10 mRNA levels are higher in the CD83 mutant cells.
  • FIG. 11 provides a graph illustrating that various preparations of anti-CD83 antibodies inhibit IL-4 production in anti-CD3 and anti-CD28 antibody stimulated T cells.
  • the amount of IL-4 produced by T cells in pg/ml is plotted versus the concentration of different anti-CD83 antibody preparations, including the 20B08 ( ⁇ ) anti-CD83 preparation, the 20D04 ( ⁇ ) anti-CD83 preparation, the 14C12 ( ⁇ ) anti-CD83 preparation and the 11G05 (X) anti-CD83 antibody preparation.
  • FIG. 12 provides a graph illustrating that various preparations of anti-CD83 antibodies inhibit T cell proliferation.
  • the graph plots the incorporation of radioactive thymidine in cpms, which was used as an indicator of the amount of T cell proliferation, versus the concentration of the different anti-CD83 antibody preparations, including the 20D04 ( ⁇ ) anti-CD83 preparation, the 11G05 ( ⁇ ) anti-CD83 antibody preparation, the 14C12 ( ⁇ ) anti-CD83 preparation and the 6G05 anti-CD83 preparation (X).
  • FIG. 13 provides a graph illustrating that transgenic mice that over-express wild type CD83 have increased T cell proliferation.
  • the graph plots the incorporation of radioactive thymidine in cpms, which was used as an indicator of the amount of T cell proliferation, versus the concentration of OVA peptide.
  • the transgenic mice utilized had a T-cell receptor specific for chicken ovalbumin (OVA) 323-339 peptide that can activate T-cells.
  • OVA ovalbumin
  • transgenic CD4+ T cells had increased T-cell proliferation.
  • transgenic dendritic cells could not substantially increase wild type CD4+ T cell proliferation.
  • Transgenic CD83 CD4+ T cells mixed with wild type dendritic cells
  • transgenic CD83 CD4+ T cells mixed with transgenic dendritic cells
  • wild type CD4+ T cells mixed with transgenic dendritic cells
  • wild type CD4+ T cells mixed with wild type dendritic cells X
  • FIG. 14 provides a schematic diagram of the structural elements included in the mouse CD83 protein used for generating antibodies.
  • FIG. 15 provides a graph of ELISA data illustrating the titer obtained for different isolates of polyclonal anti-CD83 anti-sera.
  • the first ( ⁇ ), second ( ⁇ ) and third ( ⁇ ) isolates had similar titers, though the titer of the second isolate ( ⁇ ) was somewhat higher.
  • FIG. 16 illustrates that proliferation of PHA-activated human PBMCs was inhibited by antibodies raised against the external region of the mouse CD83 protein ( ⁇ ). Pre-immune serum ( ⁇ ) had little effect on the proliferation of human PBMCs.
  • FIG. 17A provides a sequence alignment of anti-CD83 heavy chain variable regions isolated by the invention. Sequences for isolates 20B08H (SEQ ID NO:52), 6G05H (SEQ ID NO:53), 20D04H (SEQ ID NO:54), 11G05 (SEQ ID NO:66) and 14C12 (SEQ ID NO:67) are provided. The CDR regions are highlighted in bold.
  • FIG. 17B provides a sequence alignment of anti-CD83 light chain variable regions isolated by the invention. Sequences for isolates 20B08H (SEQ ID NO:55), 6G05H (SEQ ID NO:56), 20D04H (SEQ ID NO:57), 11G05 (SEQ ID NO:68) and 14C12 (SEQ ID NO:69) are provided. The CDR regions are highlighted in bold.
  • the invention provides methods for modulating the immune system by using CD83 proteins, CD83 nucleic acids and factors that modulate CD83 activity or expression.
  • loss or reduction of CD83 activity in vivo results in altered cytokine levels, for example, lower interleukin-2 levels, increased interleukin-4 levels, increased GM-CSF levels and increased interleukin-10 levels. Loss or reduction of CD83 activity in vivo can also result in decreased numbers of T cells.
  • the invention also relates to increased CD83 activity in vivo that can result in altered cytokine levels, for example, higher interleukin-2 levels, decreased interleukin-4 levels, decreased GM-CSF levels and decreased interleukin-10 levels.
  • Increased CD83 expression or activity in vitro and in vivo can also result in increased activation and increased numbers of T cells.
  • the effects of CD83 on the immune system, on GM-CSF and on cytokine levels were analyzed by using mutant and transgenic mice.
  • the mutant mouse has an altered CD83 gene that expresses altered (defective) CD83 gene product.
  • the transgenic mouse overexpresses CD83 gene products.
  • the invention provides mammals such as mice that have a mutant or wild type CD83 gene. These mice are useful for identifying the role that CD83 plays in the immune response. These mutant and transgenic animals are useful for identifying factors for manipulating cytokine levels and T cell activation by testing whether those factors and compositions can modulate, inhibit or replace the activity of CD83 in vivo.
  • CD83 is a lymphocyte and dendritic cell activation antigen that is expressed by activated lymphocytes and dendritic cells.
  • CD83 is also a single-chain cell-surface glycoprotein with a molecular weight of about 45,000 that is believed to be a member of the Ig superfamily.
  • the structure predicted from the CD83 amino acid sequence indicates that CD83 is a membrane glycoprotein with a single extracellular Ig-like domain, a transmembrane domain and cytoplasmic domain of approximately forty amino acids.
  • the mature CD83 protein has about 186 amino acids and is composed of a single extracellular V type immunoglobulin (Ig)-like domain, a transmembrane domain and a thirty nine amino acid cytoplasmic domain.
  • Ig immunoglobulin
  • CD83 is translated from three mRNA transcripts of about 1.7, 2.0 and 2.5 kb that are expressed by lymphoblastoid cell lines. It is likely that CD83 undergoes extensive post-translational processing because CD83 is expressed as a single chain molecule, but the determined molecular weight is twice the predicted size of the core protein. See U.S. Pat. No. 5,766,570.
  • CD83 gene product can be encoded by a number of different nucleic acids.
  • a human CD83 nucleic acid is provided below (SEQ ID NO:10).
  • SEQ ID NO:1 A sequence of a wild type mouse CD83 gene that can be used in the invention is provided herein as SEQ ID NO:1.
  • SEQ ID NO:1 is provided below with the ATG start codon and the TGA stop codon identified by underlining.
  • Nucleic acids having SEQ ID NO:1 encode a mouse polypeptide having SEQ ID NO:2, provided below.
  • loss or reduction of CD83 activity in vivo results in altered cytokine levels, for example, lower interleukin-2 levels, increased interleukin-4 levels, increased GM-CSF levels and increased interleukin-10 levels. Loss or reduction of CD83 activity in vivo can also result in decreased numbers of T cells.
  • increased CD83 activity in vivo can also result in altered cytokine levels, for example, higher interleukin-2 levels, decreased interleukin-4 levels, decreased GM-CSF levels and decreased interleukin-10 levels.
  • Increased CD83 expression or activity in vivo can also result in increased activation or increased numbers of T cells.
  • the effect of CD83 on cytokine levels was ascertained through use of a mutant mouse that encodes a mutant CD83.
  • a mutant mouse has a CD83 gene encoding SEQ ID NO:4, with added C-terminal sequences provided by SEQ ID NO:8.
  • the mutant CD83 gene of the invention has SEQ ID NO:3.
  • SEQ ID NO:3 is provided below with the ATG start codon, the mutation, and the TGA stop codon are identified by underlining.
  • the invention provides mutant CD83 nucleic acids that include SEQ ID NO:5.
  • the invention provides mutant CD83 nucleic acids that include SEQ ID NO:7.
  • the invention also provides a mutant CD83 containing SEQ ID NO:8, provided below.
  • RVGSPLVFTK PRAHQISVPE CHPDKRRMSS ILRWQPFFEV 41 LHLTVGSTLL PDTGS SEQ ID NO:8 contains read through sequences that are not present in the wild type CD83 polypeptide but are present in the mutant CD83 gene product provided by the invention.
  • the invention also provides compositions and methods for increasing interleukin-4 levels, increasing GM-CSF levels, increasing interleukin-10 levels and decreasing interleukin-2 levels in a mammal.
  • Such compositions and methods generally operate by decreasing the expression or function of CD83 gene products in the mammal.
  • Interleukin-4 promotes the differentiation of Th2 cells while decreasing the differentiation of precursor cells into Th1 cells.
  • Th2 cells are involved in helping B lymphocytes and in stimulating production of IgG1 and IgE antibodies. Enhancement of Th2 formation may be useful, for example, in autoimmune diseases and in organ transplantation.
  • the invention provides compositions and methods for decreasing interleukin-4 levels, decreasing interleukin-10 levels and increasing interleukin-2 levels in a mammal.
  • Such compositions and methods generally increase the expression or function of CD83 gene products in the mammal.
  • Interleukin-2 promotes the differentiation of Th1 cells and decreases the differentiation of Th-2 cells.
  • Th1 cells are, for example, involved in inducing autoimmune and delayed type hypersensitivity responses. Inhibition of Th2 formation may be useful in treating allergic diseases, malignancies and infectious diseases.
  • T helper type 1 and Th2 are subsets termed T helper type 1 (Th1) and T helper type 2 (Th2) (see e.g., Mosmann, T. R. et al. (1986) J. Immunol. 136:2348-2357; Paul, W. E. and Seder, R. A. (1994) Cell 76:241-251; Seder, R. A. and Paul, W. E. (1994) Ann. Rev. Immunol. 12:635-673).
  • Th1 T helper type 1
  • Th2 T helper type 2
  • Th1 cells secrete interleukin-2 (IL-2) and interferon- ⁇ (IFN- ⁇ ) while Th2 cells produce interleukin-4 (IL4), interleukin-5 (IL-5), interleukin-10 (IL-10) and interleukin-13 (IL-13). Both subsets produce cytokines such as tumor necrosis factor (TNF) and granulocyte/macrophage-colony stimulating factor (GM-CSF).
  • TNF tumor necrosis factor
  • GM-CSF granulocyte/macrophage-colony stimulating factor
  • Th1 and Th2 cells are thought to have differing functional activities. For example, Th1 cells are involved in inducing delayed type hypersensitivity responses, whereas Th2 cells are involved in providing efficient “help” to B lymphocytes and stimulating production of IgG1 and IgE antibodies.
  • Th1 to Th2 cells are highly relevant to the outcome of a wide array of immunologically-mediated clinical diseases including autoimmune, allergic and infectious diseases.
  • autoimmune allergic and infectious diseases.
  • Th1 response animals that are resistant to infection mount predominantly a Th1 response
  • Th2 response animals that are susceptible to progressive infection mount predominantly a Th2 response
  • Th1 to Th2 switch is observed coincident with the release of eggs into the tissues by female parasites and is associated with a worsening of the disease condition (Pearce, E. J., et al. (1991) J. Exp. Med. 173:159-166; Grzych, J-M., et al. (1991) J. Immunol. 141:1322-1327; Kullberg, M. C., et al. (1992) J. Immunol. 148:3264-3270).
  • Th1 to Th2 switch see e.g., Shearer, G. M. and Clerici, M. (1992) Prog. Chem. Immunol. 54:21-43; Clerici, M. and Shearer, G. M. (1993) Immunology Today 14:107-111; Yamamura, M., et al. (1993) J. Clin. Invest. 91:1005-1010; Pisa, P., et al. (1992) Proc. Natl. Acad. Sci. USA 89:7708-7712; Fauci, A. S. (1988) Science 239:617-623).
  • autoimmune diseases have been shown to be associated with a predominant Th1 response.
  • patients with rheumatoid arthritis have predominantly Th1 cells in synovial tissue (Simon, A. K., et al. (1994) Proc. Natl. Acad. Sci. USA 91:8562-8566) and experimental autoimmune encephalomyelitis (EAE) can be induced by autoreactive Th1 cells (Kuchroo, V. K., et al. (1993) J. Immunol. 151:4371-4381).
  • Thp CD4 T helper precursor cells
  • Th1 and Th2 subsets require an understanding of the mechanisms by which the differentiation of CD4 T helper precursor cells (Thp), which secrete only IL-2, choose to become Th1 or Th2 effector cells.
  • Thp CD4 T helper precursor cells
  • the cytokines themselves are potent Th cell inducers and form an autoregulatory loop (see e.g., Paul, W. E. and Seder, R. A. (1994) Cell 76:241-251; Seder, R. A. and Paul, W. E. (1994) Ann. Rev. Immunol. 12:635-673).
  • IL4 promotes the differentiation of Th2 cells while preventing the differentiation of precursors into Th1 cells, while IL-12 and IFN- ⁇ have the opposite effect.
  • one way to alter Th1:Th2 ratios is to increase or decrease the level of selected cytokines by using CD83.
  • Direct administration of cytokines or antibodies to cytokines has been shown to have an effect on certain diseases mediated by either Th1 or Th2 cells.
  • administration of recombinant IL-4 or antibodies to IL-12 ameliorate EAE, a Th1-driven autoimmune disease (see Racke; M. K. et al. (1994) J. Exp. Med. 180:1961-1966; and Leonard, J. P. et al. (1995) J. Exp. Med. 181:381-386), while anti-IL-4 antibodies can ameliorate the Th2-mediated parasitic disease, Leishmania major (Sadick, M. D. et al. (1990) J. Exp. Med. 171:115-127).
  • IL-4 is produced by Th2 cells, which further stimulates production of IgE antibodies and activation of cells that mediate allergic reactions, i.e., mast cells and basophils. IL-4 also plays an important role in eosinophil mediated inflammatory reactions.
  • the stimulation of CD83 production by use of the compositions and methods of the invention can be used to inhibit the production of Th2-associated cytokines, for example IL-4, in allergic patients as a means to down-regulate production of pathogenic IgE antibodies.
  • a stimulatory agent may be directly administered to the subject mammal.
  • the CD83 stimulatory agent e.g. CD83 expression cassette
  • the CD83 stimulatory agent can be administered to cells (e.g., Thp cells or Th2 cells) that may be obtained from the subject and those modified cells can be readministered to the subject mammal.
  • Such co-administration can inhibit (e.g., desensitize) the allergen-specific response.
  • the treatment may be further enhanced by administering Th1-promoting agents, such as the cytokine IL-12 or antibodies to Th2-associated cytokines (e.g., anti-IL-4 antibodies), to the allergic subject in amounts sufficient to further stimulate a Th1-type response.
  • Th1-promoting agents such as the cytokine IL-12 or antibodies to Th2-associated cytokines (e.g., anti-IL-4 antibodies)
  • the invention also relates to CD83-related methods for increasing interleukin-10 (IL-10) levels to reduce the spread of neoplastic diseases and/or prevent neoplastic diseases and the growth of a tumor.
  • IL-10 interleukin-10
  • decreased CD83 activity can dramatically increase the levels of IL-10 in the body and such increased interleukin-10 can be used to treat neoplastic diseases.
  • the invention provides a method for preventing or treating tumors in a mammal, which involves diminishing CD83 expression or activity in the mammal.
  • the tumor is IL-2-dependent, a plasmacytoma, or a leukemia, including a lymphocytic leukemia such as a B cell lymphocytic leukemia.
  • the invention also provides methods for increasing T cell activation or T cell proliferation by increasing CD83 activity or expression. Such methods can also be used to prevent or treat tumors in a mammal.
  • Th2-promoting cytokines also has been reported to increase during a variety of infectious diseases.
  • infectious diseases For example, HIV infection, tuberculosis, leishmaniasis, schistosomiasis, filarial nematode infection, intestinal nematode infection and other such infectious diseases are associated with a Th1 to Th2 shift in the immune response.
  • the stimulatory CD83-related compositions and methods of the invention can be used to inhibit the production of Th2-cells in subjects with infectious diseases to promote an ongoing Th1 response in the patients and to ameliorate the course of the infection.
  • the treatment may be further enhanced by administering other Th1-promoting agents, such as the cytokine IL-12 or antibodies to Th2-associated cytokines (e.g., anti-IL-4 antibodies), to the recipient in amounts sufficient to further stimulate a Th 1-type response.
  • infections of the following microbial organisms can be treated by the methods of the invention: Aeromonas spp., Bacillus spp., Bacteroides spp., Campylobacter spp., Clostridium spp., Enterobacter spp., Enterococcus spp., Escherichia spp., Gastrospirillum sp., Helicobacter spp., Klebsiella spp., Salmonella spp., Shigella spp., Staphylococcus spp., Pseudomonas spp., Vibrio spp., Yersinia spp., and the like.
  • Infections that can be treated by the methods of the invention include those associated with staph infections ( Staphylococcus aureus ), typhus ( Salmonella typhi ), food poisoning ( Escherichia coli , such as O157:H7), bascillary dysentery ( Shigella dysenteria ), pneumonia ( Psuedomonas aerugenosa and/or Pseudomonas cepacia ), cholera ( Vivrio cholerae ), ulcers ( Helicobacter pylori ) and others.
  • Staphylococcus aureus typhus
  • Salmonella typhi Salmonella typhi
  • food poisoning Escherichia coli , such as O157:H7
  • bascillary dysentery Shigella dysenteria
  • pneumonia Psuedomonas aerugenosa and/or Pseudomonas cepacia
  • cholera Vivrio
  • coli serotype O157:H7 has been implicated in the pathogenesis of diarrhea, hemorrhagic colitis, hemolytic uremic syndrome (HUS) and thrombotic thrombocytopenic purpura (TTP).
  • the methods of the invention are also active against drug-resistant and multiply-drug resistant strains of bacteria, for example, multiply-resistant strains of Staphylococcus aureus and vancomycin-resistant strains of Enterococcus faecium and Enterococcus faecalis.
  • virus refers to DNA and RNA viruses, viroids, and prions.
  • Viruses include both enveloped and non-enveloped viruses, for example, hepatitis A virus, hepatitis B virus, hepatitis C virus, human immunodeficiency virus (HIV), poxviruses, herpes viruses, adenoviruses, papovaviruses, parvoviruses, reoviruses, orbiviruses, picornaviruses, rotaviruses, alphaviruses, rubivirues, influenza virus type A and B, flaviviruses, coronaviruses, paramyxoviruses, morbilliviruses, pneumoviruses, rhabdoviruses, lyssaviruses, orthmyxoviruses, bunyaviruses, phleboviruses, nairoviruses, he
  • the CD83-related compositions and methods of the invention can be used in the treatment of autoimmune diseases that are associated with a Th2-type dysfunction.
  • Many autoimmune disorders are the result of inappropriate activation of T cells that are reactive against “self tissues” and that promote the production of cytokines and autoantibodies involved in the pathology of the diseases.
  • Modulation of T helper-type responses can have an effect on the course of the autoimmune disease. For example, in experimental allergic encephalomyelitis, stimulation of a Th2-type response by administration of IL-4 at the time of the induction of the disease diminishes the intensity of the autoimmune disease (Paul, W. E., et al. (1994) Cell 76:241-251).
  • Th2-specific cytokines Koury, S. J., et al. (1992) J. Exp. Med. 176:1355-1364.
  • T cells that can suppress EAE secrete Th2-specific cytokines (Chen, C., et al. (1994) Immunity 1:147-154). Since stimulation of a Th2-type response in experimental allergic encephalomyelitis has a protective effect against the disease, stimulation of a Th2 response in subjects with multiple sclerosis (for which EAE is a model) is likely to be beneficial therapeutically.
  • IL-4 which promotes a Th2 response
  • inhibition of CD83 production can stimulate IL-4 production and/or a Th2 response in a subject suffering from or susceptible to diabetes may ameliorate the effects of the disease or inhibit the onset of the disease.
  • RA rheumatoid arthritis
  • the detrimental Th1 response can be concomitantly down-modulated to thereby ameliorate the effects of the disease.
  • the CD83-related compositions and methods of the invention can be used to stimulate production of Th2-associated cytokines in subjects suffering from, or susceptible to, an autoimmune disease in which a Th2-type response is beneficial to the course of the disease.
  • Such compositions and methods would modulate CD83 activity.
  • the compositions would decrease CD83 activity and thereby increase the level of certain cytokines, for example, IL-4 levels are increased when CD83 activity is diminished.
  • the treatment may be further enhanced by administering other Th2-promoting agents, such as IL-4 itself or antibodies to Th1-associated cytokines, to the subject in amounts sufficient to further stimulate a Th2-type response.
  • the treatment may be further enhanced by administering a Th1-promoting cytokine (e.g., IFN- ⁇ ) to the subject in amounts sufficient to further stimulate a Th1-type response.
  • a Th1-promoting cytokine e.g., IFN- ⁇
  • the efficacy of CD83-related for treating autoimmune diseases can be tested in the animal models provided herein or other models of human diseases (e.g., EAE as a model of multiple sclerosis and the NOD mice as a model for diabetes).
  • animal models include the mrl/lpr/lpr mouse as a model for lupus erythematosus, murine collagen-induced arthritis as a model for rheumatoid arthritis, and murine experimental myasthenia gravis (see Paul ed., Fundamental Immunology, Raven Press, New York, 1989, pp. 840-856).
  • a CD83-modulatory (i.e., stimulatory or inhibitory) agent of the invention is administered to test animals and the course of the disease in the test animals is then monitored by the standard methods for the particular model being used. Effectiveness of the modulatory agent is evidenced by amelioration of the disease condition in animals treated with the agent as compared to untreated animals (or animals treated with a control agent).
  • Non-limiting examples of autoimmune diseases and disorders having an autoimmune component that may be treated according to the invention include diabetes mellitus, arthritis (including rheumatoid arthritis, juvenile rheumatoid arthritis, osteoarthritis, psoriatic arthritis), multiple sclerosis, myasthenia gravis, systemic lupus erythematosis, autoimmune thyroiditis, dermatitis (including atopic dermatitis and eczematous dermatitis), psoriasis, Sjogren's Syndrome, including keratoconjunctivitis sicca secondary to Sjogren's Syndrome, alopecia areata, allergic responses due to arthropod bite reactions, Crohn's disease, aphthous ulcer, ulceris, conjunctivitis, keratoconjunctivitis, ulcerative colitis, asthma, allergic asthma, cutaneous lupus erythematosus, scleroderma, vaginitis, proctiti
  • graft rejection or graft acceptance may not be attributable exclusively to the action of a particular T cell subset (i.e., Th1 or Th2 cells) in the graft recipient
  • studies have implicated a predominant Th2 response in prolonged graft survival and a predominant Th1 response in graft rejection (for a discussion see Dallman, M. J. (1995) Curr. Opin. Immunol. 7:632-638; Takeuchi, T. et al. (1992) Transplantation 53:1281-1291; Tzakis, A. G. et al. (1994) J. Pediatr. Surg. 29:754-756; That, N. L. et al.
  • loss of CD83 function increases interleukin-4 production, which in turn promotes the differentiation of Th2 cells and depresses the differentiation of precursor cells into Th1 cells.
  • methods of the invention that involve decreasing CD83 function can be used to stimulate production of Th2-associated cytokines in transplant recipients to prolong survival of the graft. These methods can be used both in solid organ transplantation and in bone marrow transplantation (e.g., to inhibit graft-versus-host disease). These methods can involve either direct administration of a CD83 inhibitory agent to the transplant recipient or ex vivo treatment of cells obtained from the subject (e.g., Thp, Th1 cells, B cells, non-lymphoid cells) with an inhibitory agent followed by readministration of the cells to the subject. The treatment may be further enhanced by administering other Th2-promoting agents, such as IL-4 itself or antibodies to Th1-associated cytokines, to the recipient in amounts sufficient to further stimulate a Th2-type response.
  • Th2-promoting agents such as IL-4 itself or antibodies to Th1-associated
  • GM-CSF granulocyte macrophage-colony stimulating factor
  • Granulocyte macrophage colony stimulating factor is a hematopoietic growth factor that promotes the proliferation and differentiation of hematopoietic progenitor cells.
  • GM-CSF is approved for treatment of patients requiring increased proliferation of white blood cells.
  • GM-CSF can also be used to treat patients prone to infection such as those undergoing high risk bowel surgery, trauma victims and individuals with HIV.
  • the invention provides a method of increasing the levels of GM-CSF in a mammal or in a mammalian cell by administering an agent that modulates or inhibits CD83 activity or expression.
  • the invention also provides a method of decreasing the levels of GM-CSF in a mammal or in a mammalian cell by administering an agent that modulates or stimulates CD83 activity or expression.
  • the CD83 inhibitory methods of the invention can be used to stimulate production of IL-4 or IL-10 in vitro for commercial production of these cytokines.
  • CD4+ T cells with a null or other mutation in the CD83 gene can be cultured and then stimulated to produce cytokines, for example, by use of anti-CD3 and/or anti-CD28 antibodies to activate the mutant CD4+ T cells.
  • Significant amounts of IL-4 and IL-10 can then be isolated from the culture media.
  • CD4+ T cells can be contacted with the CD83 inhibitory agent in vitro to stimulate IL-4 or IL-10 production and the IL-4 or IL-10 can be recovered from the culture supernatant.
  • the isolated IL-4 and/or IL-10 can be further purified if necessary, and packaged for commercial use.
  • the methods of the invention can be adapted to vaccinations to promote either a Th1 or a Th2 response to an antigen of interest in a subject. That is, CD83 or CD83 modulators of the invention can serve as adjuvants to direct an immune response to a vaccine either to a Th1 response or a Th2 response.
  • CD83 or CD83 modulators of the invention can serve as adjuvants to direct an immune response to a vaccine either to a Th1 response or a Th2 response.
  • the antigen and a CD83 inhibitory agent of the invention can be coadministered to a subject to promote a Th2 response to the antigen in the subject, since Th2 responses provide efficient B cell help and promote IgG1 production.
  • the antigen and a CD83 stimulating agent of the invention can be coadministered to a subject to promote a Th1 response to the antigen in a subject, since Th1 responses favor the development of cell-mediated immune responses (e.g., delayed hypersensitivity responses).
  • the antigen of interest and the modulatory agent can be formulated together into a single pharmaceutical composition or in separate compositions.
  • the antigen of interest and the modulatory agent are administered simultaneously to the subject.
  • any agent that can modulate CD83 to increase or decrease cytokine levels, increase or decrease T cell levels or produce any other CD83-related response can be used in the compositions and methods of the invention.
  • anti-CD83 antibodies of the invention are used to either activate or inhibit CD83 activity. Activation or inhibition by such antibodies can depend on the epitope to which the antibody binds. Hence, antibodies may play a role in boosting or depressing CD83 activity.
  • any agent that can stimulate CD83 to perform its natural functions can be used in the compositions and methods of the invention as a CD83 stimulatory agent.
  • Indicators that CD83 activity is stimulated include increased IL-2 cytokine levels, increased T cell levels, and increased TNF levels relative to unstimulated levels in wild type CD83 cells.
  • Examples of CD83 stimulatory agents include, for example, the CD83 gene product itself, certain anti-CD83 antibodies, CD83-encoding nucleic acids (DNA or RNA), factors that promote CD83 transcription or translation, organic molecules, peptides and the like.
  • any agent that can inhibit CD83 from performing its natural functions can be used in the compositions and methods of the invention as a CD83 inhibitory agent.
  • Indicators that CD83 activity is inhibited include increased IL-4 cytokine levels, increased IL-10 levels, decreased IL-2 production, decreased T cell levels, and decreased TNF levels relative to uninhibited levels in wild type CD83 cells.
  • CD83 inhibitors include anti-CD83 antibodies, CD83 anti-sense nucleic acids (e.g. nucleic acids that can hybridize to CD83 nucleic acids), organic compounds, peptides and agents that can mutate an endogenous CD83 gene.
  • the CD83 stimulatory or inhibitory agents are proteins, for example, CD83 gene products, anti-CD83 antibody preparations, CD83 inhibitors, peptides and protein factors that can promote CD83 transcription or translation.
  • the CD83 stimulatory or inhibitory agents are peptides or organic molecules. Such proteins, organic molecules and organic molecules can be prepared and/or purified as described herein or by methods available in the art, and administered as provided herein.
  • the CD83 stimulatory or inhibitory agents can be nucleic acids including recombinant expression vectors or expression cassettes encoding CD83 gene products, CD83 transcription factors, CD83 anti-sense nucleic acid, intracellular antibodies capable of binding to CD83 or dominant negative CD83 inhibitors.
  • nucleic acids can be operably linked to a promoter that is functional in a mammalian cell, and then introduced into cells of the subject mammal using methods known in the art for introducing nucleic acid (e.g., DNA) into cells.
  • the “promoter functional in a mammalian cell” or “mammalian promoter” is capable of directing transcription of a polypeptide coding sequence operably linked to the promoter.
  • the promoter should generally be active in T cells and antigen presenting cells and may be obtained from a gene that is expressed in T cells or antigen presenting cells. However, it need not be a T cell-specific or an antigen presenting cell specific-promoter. Instead, the promoter may be selected from any mammalian or viral promoter that can function in a T cell.
  • the promoter may be an actin promoter, an immunoglobulin promoter, a heat-shock promoter, or a viral promoter obtained from the genome of viruses such as adenoviruses, retroviruses, lentiviruses, herpes viruses, including but not limited to, polyoma virus, fowlpox virus, adenovirus 2, bovine papilloma virus, avian sarcoma virus, cytomegalovirus (CMV), hepatitis-B virus, Simian Virus 40 (SV40), Epstein Barr virus (EBV), feline immunodeficiency virus (FIV), and Sr.alpha., or are respiratory synsitial viral promoters (RSV) or long terminal repeats (LTRs) of a retrovirus, i.e., a Moloney Murine Leukemia Virus (MoMuLv) (Cepko et al. (1984) Cell 37:1053-1062).
  • any cloning procedure used by one of skill in the art can be employed to make the expression vectors or expression that comprise a promoter operably linked to a CD83 nucleic acid, CD83 transcription factor or a nucleic acid encoding an anti-CD83 antibody. See, e.g., Sambrook et al., Molecular Cloning, A Laboratory Manual, Cold Spring Harbor Laboratory, N.Y., 1989; Sambrook et al., Molecular Cloning, A Laboratory Manual, Cold Spring Harbor Laboratory, N.Y., 2001.
  • mammalian cells After constructing an expression vector or an expression cassette encoding CD83 gene products, CD83 transcription factors, CD83 anti-sense nucleic acid, intracellular antibodies capable of binding to CD83 or dominant negative CD83 inhibitors, mammalian cells can be transformed with the vector or cassette. Examples of such methods include:
  • Naked DNA can be introduced into cells in vivo by directly injecting the DNA into the cells (see e.g., Acsadi et al. (1991) Nature 332:815-818; Wolff et al. (1990) Science 247:1465-1468).
  • a delivery apparatus e.g., a “gene gun” for injecting DNA into cells in vivo can be used.
  • Such an apparatus is commercially available (e.g., from BioRad).
  • Naked DNA can also be introduced into cells in vivo by complexing the DNA to a cation, such as polylysine, which is coupled to a ligand for a cell-surface receptor (see for example Wu, G. and Wu, C. H. (1988) J. Biol. Chem. 263:14621; Wilson et al. (1992) J. Biol. Chem. 267:963-967; and U.S. Pat. No. 5,166,320). Binding of the DNA-ligand complex to the receptor facilitates uptake of the DNA by receptor-mediated endocytosis.
  • a cation such as polylysine
  • a DNA-ligand complex linked to adenovirus capsids that naturally disrupt endosomes, thereby releasing material into the cytoplasm can be used to avoid degradation of the complex by intracellular lysosomes (see for example Curiel et al. (1991) Proc. Natl. Acad. Sci. USA 88:8850; Cristiano et al. (1993) Proc. Natl. Acad. Sci. USA 90:2122-2126).
  • Retroviruses Defective retroviruses are well characterized for use in gene transfer for gene therapy purposes (for a review see Miller, A. D. (1990) Blood 76:271).
  • a recombinant retrovirus can be constructed having nucleotide sequences of interest incorporated into the retroviral genome. Additionally, portions of the retroviral genome can be removed to render the retrovirus replication defective. The replication defective retrovirus is then packaged into virions that can be used to infect a target cell through the use of a helper virus by standard techniques. Protocols for producing recombinant retroviruses and for infecting cells in vitro or in vivo with such viruses can be found in Current Protocols in Molecular Biology, Ausubel, F. M. et al.
  • retroviruses include pLJ, pZIP, pWE and pEM which are available to those skilled in the art.
  • suitable packaging virus lines include ⁇ Crip, ⁇ Cre, ⁇ 2 and ⁇ Am.
  • Retroviruses have been used to introduce a variety of genes into many different cell types, including epithelial cells, endothelial cells, lymphocytes, myoblasts, hepatocytes, bone marrow cells, in vitro and/or in vivo (see for example Eglitis, et al. (1985) Science 230:1395-1398; Danos and Mulligan (1988) Proc.
  • Retroviral vectors require target cell division in order for the retroviral genome (and foreign nucleic acid inserted into it) to be integrated into the host genome to stably introduce nucleic acid into the cell. Thus, it may be necessary to stimulate replication of the target cell.
  • Adenoviruses The genome of an adenovirus can be manipulated such that it encodes and expresses a gene product of interest but is inactivated in terms of its ability to replicate in a normal lytic viral life cycle. See, for example, Berkner et al. (1988) BioTechniques 6:616; Rosenfeld et al. (1991) Science 252:431-434; and Rosenfeld et al. (1992) Cell 68:143-155.
  • Suitable adenoviral vectors derived from the adenovirus strain Ad type 5 d1324 or other strains of adenovirus are available to those skilled in the art.
  • Recombinant adenoviruses are advantageous in that they do not require dividing cells to be effective gene delivery vehicles and can be used to infect a wide variety of cell types, including airway epithelium (Rosenfeld et al. (1992) cited supra), endothelial cells (Lemarchand et al. (1992) Proc. Natl. Acad. Sci. USA 89:6482-6486), hepatocytes (Herz and Gerard (1993) Proc. Natl. Acad. Sci. USA 90:2812-2816) and muscle cells (Quantin et al. (1992) Proc. Natl. Acad. Sci. USA 89:2581-2584).
  • introduced adenoviral DNA (and foreign DNA contained therein) is not integrated into the genome of a host cell but remains episomal, thereby avoiding potential problems that can occur as a result of insertional mutagenesis in situations where introduced DNA becomes integrated into the host genome (e.g., retroviral DNA).
  • the carrying capacity of the adenoviral genome for foreign DNA is large (up to 8 kilobases) relative to other gene delivery vectors (Berkner et al. cited supra; Haj-Ahmand and Graham (1986) J. Virol. 57:267).
  • Most replication-defective adenoviral vectors currently in use are deleted for all or parts of the viral E1 and E3 genes but retain as much as 80% of the adenoviral genetic material.
  • Adeno-associated virus is a naturally occurring defective virus that requires another virus, such as an adenovirus or a herpes virus, as a helper virus for efficient replication and a productive life cycle.
  • AAV Adeno-associated virus
  • AAV vector such as that described in Tratschin et al. (1985) Mol. Cell. Biol. 5:3251-3260 can be used to introduce DNA into cells.
  • a variety of nucleic acids have been introduced into different cell types using AAV vectors (see for example Hermonat et al. (1984) Proc. Natl. Acad. Sci. USA 81:6466-6470; Tratschin et al. (1985) Mol. Cell. Biol.
  • Transformed mammalian cells can then be identified and administered to the mammal from whence they came to permit expression of a CD83 gene product, CD83 transcription factor, CD83 anti-sense nucleic acid, intracellular antibody capable of binding to CD83 proteins, or dominant negative CD83 inhibitors.
  • the efficacy of a particular expression vector system and method of introducing nucleic acid into a cell can be assessed by standard approaches routinely used in the art.
  • DNA introduced into a cell can be detected by a filter hybridization technique (e.g., Southern blotting).
  • RNA produced by transcription of an introduced DNA can be detected, for example, by Northern blotting, RNase protection or reverse transcriptase-polymerase chain reaction (RT-PCR).
  • the CD83 gene product can be detected by an appropriate assay, for example, by immunological detection of a produced CD83 protein, such as with a CD83-specific antibody.
  • the invention provides antibody preparations directed against the mutant and wild type CD83 polypeptides of the invention, for example, against a polypeptide having SEQ ID NO:2, SEQ ID NO:4, SEQ ID NO:7, SEQ ID NO:8 or SEQ ID NO:9.
  • Other antibodies of interest can bind to the cytoplasmic tail of CD83.
  • the invention provides antibodies that block the function of CD83 polypeptides. Such antibodies may be used as CD83 inhibitory agents in the methods of the invention as described herein. In another embodiment, the antibodies of the invention can activate CD83 activity. Such activating antibodies may be used as CD83 stimulatory agents.
  • immunoglobulins All antibody molecules belong to a family of plasma proteins called immunoglobulins, whose basic building block, the immunoglobulin fold or domain, is used in various forms in many molecules of the immune system and other biological recognition systems.
  • a typical immunoglobulin has four polypeptide chains, containing an antigen binding region known as a variable region and a non-varying region known as the constant region.
  • Native antibodies and immunoglobulins are usually heterotetrameric glycoproteins of about 150,000 daltons, composed of two identical light (L) chains and two identical heavy (H) chains. Each light chain is linked to a heavy chain by one covalent disulfide bond, while the number of disulfide linkages varies between the heavy chains of different immunoglobulin isotypes. Each heavy and light chain also has regularly spaced intrachain disulfide bridges. Each heavy chain has at one end a variable domain (VH) followed by a number of constant domains. Each light chain has a variable domain at one end (VL) and a constant domain at its other end.
  • VH variable domain
  • VL variable domain at one end
  • the constant domain of the light chain is aligned with the first constant domain of the heavy chain, and the light chain variable domain is aligned with the variable domain of the heavy chain.
  • Particular amino acid residues are believed to form an interface between the light and heavy chain variable domains (Clothia et al., J. Mol. Biol. 186, 651-66, 1985); Novotny and Haber, Proc. Natl. Acad. Sci. USA 82, 4592-4596 (1985).
  • immunoglobulins can be assigned to different classes. There are at least five (5) major classes of immunoglobulins: IgA, IgD, IgE, IgG and several of these may be further divided into subclasses (isotypes), e.g. IgG-1, IgG-2, IgG-3 and IgG-4; IgA-1 and IgA-2.
  • the heavy chains constant domains that correspond to the different classes of immunoglobulins are called alpha ( ⁇ ), delta ( ⁇ ), epsilon ( ⁇ ), gamma ( ⁇ ) and mu ( ⁇ ), respectively.
  • the light chains of antibodies can be assigned to one of two clearly distinct types, called kappa ( ⁇ ) and lambda ( ⁇ ), based on the amino sequences of their constant domain.
  • kappa
  • lambda
  • the subunit structures and three-dimensional configurations of different classes of immunoglobulins are well known.
  • variable in the context of variable domain of antibodies, refers to the fact that certain portions of the variable domains differ extensively in sequence among antibodies.
  • the variable domains are for binding and determine the specificity of each particular antibody for its particular antigen.
  • variability is not evenly distributed through the variable domains of antibodies. It is concentrated in three segments called complementarity determining regions (CDRs) also known as hypervariable regions both in the light chain and the heavy chain variable domains.
  • CDRs complementarity determining regions
  • variable domains The more highly conserved portions of variable domains are called the framework (FR).
  • the variable domains of native heavy and light chains each comprise four FR regions, largely adopting a ⁇ -sheet configuration, connected by three CDRs, which form loops connecting, and in some cases forming part of, the ⁇ -sheet structure.
  • the CDRs in each chain are held together in close proximity by the FR regions and, with the CDRs from the other chain, contribute to the formation of the antigen binding site of antibodies.
  • the constant domains are not involved directly in binding an antibody to an antigen, but exhibit various effector function, such as participation of the antibody in antibody-dependent cellular toxicity.
  • an antibody that is contemplated for use in the present invention thus can be in any of a variety of forms, including a whole immunoglobulin, an antibody fragment such as Fv, Fab, and similar fragments, a single chain antibody that includes the variable domain complementarity determining regions (CDR), and the like forms, all of which fall under the broad term “antibody,” as used herein.
  • the present invention contemplates the use of any specificity of an antibody, polyclonal or monoclonal, and is not limited to antibodies that recognize and immunoreact with a specific antigen.
  • an antibody or fragment thereof is used that is immunospecific for an antigen or epitope of the invention.
  • antibody fragment refers to a portion of a full-length antibody, generally the antigen binding or variable region.
  • antibody fragments include Fab, Fab′, F(ab′) 2 and Fv fragments.
  • Papain digestion of antibodies produces two identical antigen binding fragments, called the Fab fragment, each with a single antigen binding site, and a residual “Fc” fragment, so-called for its ability to crystallize readily.
  • Pepsin treatment yields an F(ab′) 2 fragment that has two antigen binding fragments, which are capable of cross-linking antigen, and a residual other fragment (which is termed pFc′).
  • Additional fragments can include diabodies, linear antibodies, single-chain antibody molecules, and multispecific antibodies formed from antibody fragments.
  • “functional fragment” with respect to antibodies refers to Fv, F(ab) and F(ab′) 2 fragments.
  • Antibody fragments retain some ability to selectively bind with its antigen or receptor and are defined as follows:
  • Fab is the fragment that contains a monovalent antigen-binding fragment of an antibody molecule.
  • a Fab fragment can be produced by digestion of whole antibody with the enzyme papain to yield an intact light chain and a portion of one heavy chain.
  • Fab′ is the fragment of an antibody molecule can be obtained by treating whole antibody with pepsin, followed by reduction, to yield an intact light chain and a portion of the heavy chain. Two Fab′ fragments are obtained per antibody molecule. Fab′ fragments differ from Fab fragments by the addition of a few residues at the carboxyl terminus of the heavy chain CH1 domain including one or more cysteines from the antibody hinge region.
  • (Fab′) 2 is the fragment of an antibody that can be obtained by treating whole antibody with the enzyme pepsin without subsequent reduction.
  • F(ab′) 2 is a dimer of two Fab′ fragments held together by two disulfide bonds.
  • Fv is the minimum antibody fragment that contains a complete antigen recognition and binding site. This region consists of a dimer of one heavy and one light chain variable domain in a tight, non-covalent association (V H -V L dimer). It is in this configuration that the three CDRs of each variable domain interact to define an antigen binding site on the surface of the V H -V L dimer. Collectively, the six CDRs confer antigen binding specificity to the antibody. However, even a single variable domain (or half of an Fv comprising only three CDRs specific for an antigen) has the ability to recognize and bind antigen, although at a lower affinity than the entire binding site.
  • Single chain antibody defined as a genetically engineered molecule containing the variable region of the light chain, the variable region of the heavy chain, linked by a suitable polypeptide linker as a genetically fused single chain molecule.
  • Such single chain antibodies are also referred to as “single-chain Fv” or “sFv” antibody fragments.
  • the Fv polypeptide further comprises a polypeptide linker between the VH and VL domains that enables the sFv to form the desired structure for antigen binding.
  • diabodies refers to a small antibody fragments with two antigen-binding sites, which fragments comprise a heavy chain variable domain (VH) connected to a light chain variable domain (VL) in the same polypeptide chain (V H -V L ).
  • VH heavy chain variable domain
  • VL light chain variable domain
  • polyclonal antibodies The preparation of polyclonal antibodies is well-known to those skilled in the art. See, for example, Green, et al., Production of Polyclonal Antisera, in: Immunochemical Protocols (Manson, ed.), pages 1-5 (Humana Press); Coligan, et al., Production of Polyclonal Antisera in Rabbits, Rats Mice and Hamsters, in: Current Protocols in Immunology , section 2.4.1 (1992), which are hereby incorporated by reference.
  • the monoclonal antibodies to be used in accordance with the present invention may be made by the hybridoma method first described by Kohler and Milstein, Nature 256, 495 (1975), or may be made by recombinant methods, e.g., as described in U.S. Pat. No. 4,816,567.
  • the monoclonal antibodies for use with the present invention may also be isolated from antibody libraries using the techniques described in Clackson et al. Nature 352: 624-628 (1991), as well as in Marks et al., J. Mol. Biol. 222: 581-597 (1991).
  • Monoclonal antibodies can be isolated and purified from hybridoma cultures by a variety of well-established techniques. Such isolation techniques include affinity chromatography with Protein-A Sepharose, size-exclusion chromatography, and ion-exchange chromatography. See, e.g., Coligan, et al., sections 2.7.1-2.7.12 and sections 2.9.1-2.9.3; Barnes, et al., Purification of Immunoglobulin G (IgG), in: Methods in Molecular Biology , Vol. 10, pages 79-104 (Humana Press (1992).
  • Selected Lymphocyte Antibody Method Another method for generating antibodies involves a Selected Lymphocyte Antibody Method (SLAM).
  • SLAM Selected Lymphocyte Antibody Method
  • the methodology principally involves the growth of antibody forming cells, the physical selection of specifically selected antibody forming cells, the isolation of the genes encoding the antibody and the subsequent cloning and expression of those genes.
  • an animal is immunized with a source of specific antigen.
  • This immunization may consist of purified protein, in either native or recombinant form, peptides, DNA encoding the protein of interest or cells expressing the protein of interest.
  • blood or other tissue
  • Lymphocytes are isolated from the blood and cultured under specific conditions to generate antibody-forming cells, with antibody being secreted into the culture medium. These cells are detected by any of several means (complement mediated lysis of antigen-bearing cells, fluorescence detection or other) and then isolated using micromanipulation technology.
  • the individual antibody forming cells are then processed for eventual single cell PCR to obtain the expressed Heavy and Light chain genes that encode the specific antibody. Once obtained and sequenced, these genes are cloned into an appropriate expression vector and recombinant, monoclonal antibody produced in a heterologous cell system. These antibodies are then purified via standard methodologies such as the use of protein A affinity columns. These types of methods are further described in Babcook, et al., Proc. Natl. Acad. Sci. (USA) 93: 7843-7848 (1996); U.S. Pat. No. 5,627,052; and PCT WO 92/02551 by Schrader.
  • Another method involves humanizing a monoclonal antibody by recombinant means to generate antibodies containing human specific and recognizable sequences. See, for review, Holmes, et al., J. Immunol., 158:2192-2201 (1997) and Vaswani, et al., Annals Allergy, Asthma & Immunol., 81:105-115 (1998).
  • the term “monoclonal antibody” as used herein refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally occurring mutations that may be present in minor amounts. Monoclonal antibodies are highly specific, being directed against a single antigenic site.
  • each monoclonal antibody is directed against a single determinant on the antigen.
  • the monoclonal antibodies are advantageous in that they are synthesized by the hybridoma culture, uncontaminated by other immunoglobulins.
  • the modifier “monoclonal” indicates the antibody is obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method.
  • the monoclonal antibodies herein specifically include “chimeric” antibodies (immunoglobulins) in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is identical with or homologous to corresponding sequences in antibodies derived from another species or belonging to another antibody class or subclass, as well as fragments of such antibodies, so long as they exhibit the desired biological activity (U.S. Pat. No. 4,816,567); Morrison et al. Proc. Natl. Acad. Sci. 81, 6851-6855 (1984).
  • chimeric antibodies immunoglobulins in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is identical with or homologous to corresponding sequences in antibodies
  • Antibody fragments of the present invention can be prepared by proteolytic hydrolysis of the antibody or by expression in E. coli of DNA encoding the fragment.
  • Antibody fragments can be obtained by pepsin or papain digestion of whole antibodies conventional methods.
  • antibody fragments can be produced by enzymatic cleavage of antibodies with pepsin to provide a 5S fragment denoted F(ab′) 2 .
  • a thiol reducing agent optionally a blocking group for the sulfhydryl groups resulting from cleavage of disulfide linkages
  • an enzymatic cleavage using pepsin produces two monovalent Fab′ fragments and an Fc fragment directly.
  • Fv fragments comprise an association of V H and V L chains. This association may be noncovalent or the variable chains can be linked by an intermolecular disulfide bond or cross-linked by chemicals such as glutaraldehyde.
  • the Fv fragments comprise V H and V L chains connected by a peptide linker.
  • sFv single-chain antigen binding proteins
  • CDR peptides (“minimal recognition units”) can be obtained by constructing genes encoding the CDR of an antibody of interest. Such genes are prepared, for example, by using the polymerase chain reaction to synthesize the variable region from RNA of antibody-producing cells. See, for example, Larrick, et al., Methods: a Companion to Methods in Enzymology , Vol. 2, page 106 (1991).
  • the invention further contemplates human and humanized forms of non-human (e.g. murine) antibodies.
  • humanized antibodies are chimeric immunoglobulins, immunoglobulin chains or fragments thereof (such as Fv, Fab, Fab′, F(ab′) 2 or other antigen-binding subsequences of antibodies) that contain minimal sequence derived from non-human immunoglobulin.
  • humanized antibodies are human immunoglobulins (recipient antibody) in which residues from a complementary determining region (CDR) of the recipient are replaced by residues from a CDR of a nonhuman species (donor antibody) such as mouse, rat or rabbit having the desired specificity, affinity and capacity.
  • CDR complementary determining region
  • humanized antibodies may comprise residues that are found neither in the recipient antibody nor in the imported CDR or framework sequences. These modifications are made to further refine and optimize antibody performance.
  • humanized antibodies can comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin and all or substantially all of the Fv regions are those of a human immunoglobulin consensus sequence.
  • the humanized antibody optimally also will comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin.
  • Fc immunoglobulin constant region
  • mutant antibody refers to an amino acid sequence variant of an antibody.
  • one or more of the amino acid residues in the mutant antibody is different from what is present in the reference antibody.
  • Such mutant antibodies necessarily have less than 100% sequence identity or similarity with the reference amino acid sequence.
  • mutant antibodies have at least 75% amino acid sequence identity or similarity with the amino acid sequence of either the heavy or light chain variable domain of the reference antibody.
  • mutant antibodies have at least 80%, more preferably at least 85%, even more preferably at least 90%, and most preferably at least 95% amino acid sequence identity or similarity with the amino acid sequence of either the heavy or light chain variable domain of the reference antibody.
  • the antibodies of the invention are isolated antibodies.
  • An isolated antibody is one that has been identified and separated and/or recovered from a component of the environment in which it was produced. Contaminant components of its production environment are materials that would interfere with diagnostic or therapeutic uses for the antibody, and may include enzymes, hormones, and other proteinaceous or nonproteinaceous solutes.
  • isolated antibody also includes antibodies within recombinant cells because at least one component of the antibody's natural environment will not be present. Ordinarily, however, isolated antibody will be prepared by at least one purification step.
  • the antibodies of the invention can be purified by any available procedure.
  • the antibodies can be affinity purified by binding an antibody preparation to a solid support to which the antigen used to raise the antibodies is bound. After washing off contaminants, the antibody can be eluted by known procedures.
  • Those of skill in the art will know of various techniques common in the immunology arts for purification and/or concentration of polyclonal antibodies, as well as monoclonal antibodies (see for example, Coligan, et al., Unit 9, Current Protocols in Immunology , Wiley Interscience, 1991, incorporated by reference).
  • the antibody will be purified as measurable by at least three different methods: 1) to greater than 95% by weight of antibody as determined by the Lowry method, and most preferably more than 99% by weight; 2) to a degree sufficient to obtain at least 15 residues of N-terminal or internal amino acid sequence by use of a spinning cup sequentator; or 3) to homogeneity by SDS-PAGE under reducing or non-reducing conditions using Coomasie blue or, preferably, silver stain.
  • the invention also provides antibodies that can bind to CD83 polypeptides. Sequences of complementarity determining regions (CDRs) or hypervariable regions from light and heavy chains of these anti-CD83 antibodies are provided. For example, a heavy chain variable region having a CDR1 sequence of SYDMT (SEQ ID NO:23), SYDMS (SEQ ID NO:24), DYDLS (SEQ ID NO:25) or SYDMS (SEQ ID NO:26) can be used in an antibody or other binding moiety to bind to CD83 gene products.
  • CDRs complementarity determining regions
  • SEQ ID NO:24 SYDMS
  • DYDLS SEQ ID NO:25
  • SYDMS SEQ ID NO:26
  • a heavy chain variable region having a CDR2 sequence of YASGSTYY (SEQ ID NO:27), SSSGTTYY (SEQ ID NO:28), YASGSTYY (SEQ ID NO:29), AIDGNPYY (SEQ ID NO:30) or STAYNSHY (SEQ ID NO:31) can be used in an antibody or other binding moiety to bind to CD83 gene products.
  • a heavy chain variable region having a CDR3 sequence of EHAGYSGDTGH (SEQ ID NO:32), EGAGVSMT (SEQ ID NO:33), EDAGFSNA (SEQ ID NO:34), GAGD (SEQ ID NO:35) or GGSWLD (SEQ ID NO:36) can be used in an antibody or other binding moiety to bind to CD83 gene products.
  • a light chain variable region having a CDR1 sequence of RCAYD (SEQ ID NO:37), RCADVV (SEQ ID NO:38), or RCALV (SEQ ID NO:39) can be used in an antibody or other binding moiety to bind to CD83 gene products.
  • a light chain variable region having a CDR2 sequence of QSISTY (SEQ ID NO:40), QSVSSY (SEQ ID NO:41), ESISNY (SEQ ID NO:42), KNVYNNNW (SEQ ID NO:43), or QSVYDNDE (SEQ ID NO:43) can be used in an antibody or other binding moiety to bind to CD83 gene products.
  • a light chain variable region having a CDR3 sequence of QQGYTHSNVDNV (SEQ ID NO:44), QQGYSISDIDNA (SEQ ID NO:45), QCTSGGKFISDGAA (SEQ ID NO:46), AGDYSSSSDNG (SEQ ID NO:47), or QATHYSSDWLTY (SEQ ID NO:48) can be used in an antibody or other binding moiety to bind to CD83 gene products.
  • Light and heavy chains that can bind CD83 polypeptides are also provided by the invention.
  • the invention provides a 20D04 light chain that can bind to CD83 polypeptides.
  • the amino acid sequence for this 20D04 light chain is provided below (SEQ ID NO:11).
  • a nucleic acid sequence for this 20D04 anti-CD83 light chain is provided below (SEQ ID NO:12).
  • the invention provides a 20D04 heavy chain that can bind to CD83 polypeptides.
  • the amino acid sequence for this 20D04 heavy chain is provided below (SEQ ID NO:13).
  • a nucleic acid sequence for this 20D04 anti-CD83 heavy chain is provided below (SEQ ID NO:14).
  • the invention provides a 11G05 light chain that can bind to CD83 polypeptides.
  • the amino acid sequence for this 11G05 light chain is provided below (SEQ ID NO:15).
  • a nucleic acid sequence for this 11G05 anti-CD83 light chain is provided below (SEQ ID NO:16).
  • the invention provides a 11G05 heavy chain that can bind to CD83 polypeptides.
  • the amino acid sequence for this 11G05 heavy chain is provided below (SEQ ID NO:17).
  • a nucleic acid sequence for this 11G05 anti-CD83 heavy chain is provided below (SEQ ID NO:18).
  • the invention provides a 14C12 light chain that can bind to CD83 polypeptides.
  • the amino acid sequence for this 14C12 light chain is provided below (SEQ ID NO:19).
  • a nucleic acid sequence for this 14C12 anti-CD83 light chain is provided below (SEQ ID NO:20).
  • the invention provides a 14C12 heavy chain that can bind to CD83 polypeptide's.
  • the amino acid sequence for this 14C12 heavy chain is provided below (SEQ ID NO:21).
  • a nucleic acid sequence for this 14C12 anti-CD83 heavy chain is provided below (SEQ ID NO:22).
  • the invention provides a M83 020B08L light chain that can bind to CD83 polypeptides.
  • the amino acid sequence for this M83 020B08L light chain is provided below (SEQ ID NO:58).
  • a nucleic acid sequence for this M83 020B08L anti-CD83 light chain is provided below (SEQ ID NO:59).
  • the invention provides a M83 020B08H heavy chain that can bind to CD83 polypeptides.
  • the amino acid sequence for this M83 020B08H heavy chain is provided below (SEQ ID NO:60).
  • a nucleic acid sequence for this M83 020B08H anti-CD83 heavy chain is provided below (SEQ ID NO:61).
  • the invention provides a M83 006G05L light chain that can bind to CD83 polypeptides.
  • the amino acid sequence for this M83 006G05L light chain is provided below (SEQ ID NO:62).
  • a nucleic acid sequence for M83 006G05L anti-CD83 light chain is provided below (SEQ ID NO:63).
  • the invention provides a M83 006G05L heavy chain that can bind to CD83 polypeptides.
  • the amino acid sequence for this M83 006G05L heavy chain is provided below (SEQ ID NO:64).
  • a nucleic acid sequence for this M83 006G05L anti-CD83 heavy chain is provided below (SEQ ID NO:65).
  • Anti-sense nucleic acids can be used to inhibit the function of CD83.
  • the function of CD83 RNA is inhibited, for example, by administering to a mammal a nucleic acid that can inhibit the functioning of CD83 RNA.
  • Nucleic acids that can inhibit the function of a CD83RNA can be generated from coding and non-coding regions of the CD83 gene.
  • nucleic acids that can inhibit the function of a CD83 RNA are often selected to be complementary to CD83 nucleic acids that are naturally expressed in the mammalian cell to be treated with the methods of the invention.
  • the nucleic acids that can inhibit CD83 RNA functions are complementary to CD83 sequences found near the 5′ end of the CD83 coding region.
  • nucleic acids that can inhibit the function of a CD83 RNA can be complementary to the 5′ region of SEQ ID NO:1, SEQ ID NO:3, SEQ ID NO:5 or SEQ ID NO:10.
  • a nucleic acid that can inhibit the functioning of a CD83 RNA need not be 100% complementary to SEQ ID NO:1, SEQ ID NO:3, SEQ ID NO:5 or SEQ ID NO:10. Instead, some variability the sequence of the nucleic acid that can inhibit the functioning of a CD83 RNA is permitted.
  • a nucleic acid that can inhibit the functioning of a CD83 RNA from a human can be complementary to a nucleic acid encoding either a human or a mouse CD83 gene product.
  • nucleic acids that can hybridize under moderately or highly stringent hybridization conditions to a nucleic acid comprising SEQ ID NO:1, SEQ ID NO:3, SEQ ID NO:5 or SEQ ID NO:10 are sufficiently complementary to inhibit the functioning of a CD83 RNA and can be utilized in the methods of the invention.
  • “Stringent hybridization conditions” and “stringent hybridization wash conditions” in the context of nucleic acid hybridization are somewhat sequence dependent, and may differ depending upon the environmental conditions of the nucleic acid. For example, longer sequences tend to hybridize specifically at higher temperatures.
  • An extensive guide to the hybridization of nucleic acids is found in Tijssen, Laboratory Techniques in Biochemistry and Molecular biology-Hybridization with Nucleic Acid Probes, page 1, chapter 2 “Overview of principles of hybridization and the strategy of nucleic acid probe assays” Elsevier, N.Y. (1993). See also, J. Sambrook et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Press, N.Y., pp 9.31-9.58 (1989); J. Sambrook et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Press, N.Y. (3rd ed. 2001).
  • highly stringent hybridization and wash conditions are selected to be about 5° C. lower than the thermal melting point (T m ) for the specific double-stranded sequence at a defined ionic strength and pH.
  • T m thermal melting point
  • highly stringent hybridization and wash conditions are selected to be about 5° C. lower than the thermal melting point (T m ) for the specific double-stranded sequence at a defined ionic strength and pH.
  • T m thermal melting point
  • T m is the temperature (under defined ionic strength and pH) at which 50% of a complementary target sequence hybridizes to a perfectly matched probe.
  • Very stringent conditions are selected to be equal to the T m , for a particular probe.
  • stringency conditions can be adjusted to allow some mismatching in sequences so that lower degrees of similarity can hybridize.
  • stringent conditions will be those in which the salt concentration is less than about 1.5 M Na ion, typically about 0.01 to 1.0 M Na ion concentration (or other salts) at pH 7.0 to 8.3 and the temperature is at least about 30° C. for short probes (e.g., 10 to 50 nucleotides) and at least about 60° C. for long probes (e.g., greater than 50 nucleotides).
  • Stringent conditions may also be achieved with the addition of destabilizing agents such as formamide.
  • Exemplary moderate stringency conditions include hybridization in 40 to 45% formamide, 1.0 M NaCl, 1% SDS at 37° C., and a wash in 0.5 ⁇ to 1 ⁇ SSC at 55 to 60° C.
  • Exemplary high stringency conditions include hybridization in 50% formamide, 1 M NaCl, 1% SDS at 37° C., and a wash in 0.1 ⁇ SSC at 60 to 65° C.
  • the degree of complementarity or sequence identity of hybrids obtained during hybridization is typically a function of post-hybridization washes, the critical factors being the ionic strength and temperature of the final wash solution.
  • the type and length of hybridizing nucleic acids also affects whether hybridization will occur and whether any hybrids formed will be stable under a given set of hybridization and wash conditions.
  • An example of stringent hybridization conditions for hybridization of complementary nucleic acids that have more than 100 complementary residues on a filter in a Southern or Northern blot is 50% formamide with 1 mg of heparin at 42° C., with the hybridization being carried out overnight.
  • An example of highly stringent conditions is 0.1 5 M NaCl at 72° C. for about 15 minutes.
  • An example of stringent wash conditions is a 0.2 ⁇ SSC wash at 65° C. for 15 minutes (see also, Sambrook, infra). Often, a high stringency wash is preceded by a low stringency wash to remove background probe signal.
  • An example of medium stringency for a duplex of, e.g., more than 100 nucleotides, is 1 ⁇ SSC at 45° C.
  • stringent conditions typically involve salt concentrations of less than about 1.0M Na ion, typically about 0.01 to 1.0 M Na ion concentration (or other salts) at pH 7.0 to 8.3, and the temperature is typically at least about 30° C.
  • Stringent conditions can also be achieved with the addition of destabilizing agents such as formamide.
  • destabilizing agents such as formamide.
  • a signal to noise ratio of 2 ⁇ (or higher) than that observed for an unrelated probe in the particular hybridization assay indicates detection of a specific hybridization.
  • Nucleic acids that do not hybridize to each other under stringent conditions are still substantially identical if the proteins that they encode are substantially identical. This occurs, e.g., when a copy of a nucleic acid is created using the maximum codon degeneracy permitted by the genetic code.
  • a reference nucleotide sequence preferably hybridizes to the reference nucleotide sequence in 7% sodium dodecyl sulfate (SDS), 0.5 M NaPO 4 , 1 mM EDTA at 50° C. with washing in 2 ⁇ SSC, 0.1% SDS at 50° C., more desirably in 7% sodium dodecyl sulfate (SDS), 0.5 M NaPO 4 , 1 mM EDTA at 50° C.
  • SDS sodium dodecyl sulfate
  • T m is reduced by about 1° C. for each 1% of mismatching.
  • T m , hybridization, and/or wash conditions can be adjusted to hybridize to sequences of the desired sequence identity. For example, if sequences with >90% identity are sought, the T m can be decreased 10° C.
  • stringent conditions are selected to be about 5° C. lower than the thermal melting point (T m ) for the specific sequence and its complement at a defined ionic strength and pH.
  • severely stringent conditions can utilize a hybridization and/or wash at 1, 2, 3, or 4° C. lower than the thermal melting point (T m );
  • moderately stringent conditions can utilize a hybridization and/or wash at 6, 7, 8, 9, or 10° C. lower than the thermal melting point (T m ); low stringency conditions can utilize a hybridization and/or wash at 11, 12, 13, 14, 15, or 20° C. lower than the thermal melting point (T m ).
  • Inhibitory nucleic acid molecules that comprise, for example, 2, 3, 4, or 5 or more stretches of contiguous nucleotides that are precisely complementary to a CD83 coding sequence, each separated by a stretch of contiguous nucleotides that are not complementary to adjacent CD83 coding sequences, can inhibit the function of CD83 RNA.
  • each stretch of contiguous nucleotides is at least 4, 5, 6, 7, or 8 or more nucleotides in length.
  • Non-complementary intervening sequences are preferably 1, 2, 3, or 4 nucleotides in length.
  • One skilled in the art can easily use the calculated melting point of an anti-sense nucleic acid hybridized to a sense nucleic acid to determine the degree of mismatching that will be tolerated between a particular anti-sense nucleic acid and a particular CD83 RNA.
  • nucleic acids that complementary a CD83 RNA can be administered to a mammal or to directly to the site of the inappropriate immune system activity.
  • nucleic acids that are complementary to a CD83 RNA can generated by transcription from an expression cassette that has been administered to a mammal.
  • a complementary RNA can be transcribed from a CD83 nucleic acid that has been inserted into an expression cassette in the 3′ to 5′ orientation, that is, opposite to the usual orientation employed to generate sense RNA transcripts.
  • the promoter would be positioned to transcribe from a 3′ site towards the 5′ end of the CD83 coding region.
  • an RNA that can inhibit the function of an endogenous CD83 RNA is an anti-sense oligonucleotide.
  • the anti-sense oligonucleotide is complementary to at least a portion of the coding sequence of a gene comprising SEQ ID NO:1, SEQ ID NO:3, SEQ ID NO:5 or SEQ ID NO:10.
  • Such anti-sense oligonucleotides are generally at least six nucleotides in length, but can be about 8, 12, 15, 20, 25, 30, 35, 40, 45, or 50 nucleotides long. Longer oligonucleotides can also be used.
  • CD83 anti-sense oligonucleotides can be provided in a DNA construct and introduced into cells whose division is to be decreased, for example, into CD4+ T cells, Th-1 cells, Th-2 cells or lymphocyte precursor cells.
  • Anti-sense oligonucleotides can be composed of deoxyribonucleotides, ribonucleotides, or a combination of both. Oligonucleotides can be synthesized endogenously from transgenic expression cassettes or vectors as described herein.
  • oligonucleotides can be synthesized manually or by an automated synthesizer, by covalently linking the 5′ end of one nucleotide with the 3′ end of another nucleotide with non-phosphodiester internucleotide linkages such alkylphosphonates, phosphorothioates, phosphorodithioates, alkylphosphonothioates, alkylphosphonates, phosphoramidates, phosphate esters, carbamates, acetamidate, carboxymethyl esters, carbonates, and phosphate triesters. See Brown, 1994, Meth. Mol. Biol. 20:1-8; Sonveaux, 1994, Meth. Mol. Biol. 26:1-72; Uhlmann et al., 1990, Chem. Rev. 90:543-583.
  • CD83 anti-sense oligonucleotides can be modified without affecting their ability to hybridize to a CD83 RNA. These modifications can be internal or at one or both ends of the anti-sense molecule.
  • internucleoside phosphate linkages can be modified by adding peptidyl, cholesteryl or diamine moieties with varying numbers of carbon residues between these moities and the terminal ribose.
  • Modified bases and/or sugars such as arabinose instead of ribose, or a 3′,5′-substituted oligonucleotide in which the 3′ hydroxyl group or the 5′ phosphate group are substituted, can also be employed in a modified anti-sense oligonucleotide.
  • modified oligonucleotides can be prepared by methods available in the art. Agrawal et al., 1992, Trends Biotechnol. 10:152-158; Uhlmann et al., 1990, Chem. Rev. 90:543-584; Uhlmann et al., 1987, Tetrahedron. Lett. 215:3539-3542.
  • ribozyme is an RNA molecule with catalytic activity. See, e.g., Cech, 1987, Science 236: 1532-1539; Cech, 1990, Ann. Rev. Biochem. 59:543-568; Cech, 1992, Curr. Opin. Struct. Biol. 2: 605-609; Couture and Stinchcomb, 1996, Trends Genet. 12: 510-515. Ribozymes can be used to inhibit gene function by cleaving an RNA sequence, as is known in the art (see, e.g., Haseloff et al., U.S. Pat. No. 5,641,673).
  • CD83 nucleic acids complementary to SEQ ID NO:1, SEQ ID NO:3, SEQ ID NO:5 or SEQ ID NO:10 can be used to generate ribozymes that will specifically bind to mRNA transcribed from a CD83 gene.
  • Methods of designing and constructing ribozymes that can cleave other RNA molecules in trans in a highly sequence specific manner have been developed and described in the art (see Haseloff et al. (1988), Nature 334:585-591).
  • the cleavage activity of ribozymes can be targeted to specific RNAs by engineering a discrete “hybridization” region into the ribozyme.
  • the hybridization region contains a sequence complementary to the target RNA and thus specifically hybridizes with the target (see, for example, Gerlach et al., EP 321,201).
  • the target sequence can be a segment of about 10, 12, 15, 20, or 50 contiguous nucleotides selected from a nucleotide sequence shown in SEQ ID NO:1, SEQ ID NO:3, SEQ ID NO:5 or SEQ ID NO:10. Longer complementary sequences can be used to increase the affinity of the hybridization sequence for the target.
  • the hybridizing and cleavage regions of the ribozyme can be integrally related; thus, upon hybridizing to the target RNA through the complementary regions, the catalytic region of the ribozyme can cleave the target.
  • compositions and methods for modulating CD83 activity or expression can include these molecules as well as other components. Representative examples that are discussed in more detail below include transcription factors, RNA-binding factors, organic molecules, or peptides.
  • RNA binding factors include those described in PCT/EP01/14820 and other sources.
  • the HuR protein (Genbank accession number U38175) has the ability to specifically bind to CD83 RNA at AU-rich elements or sites.
  • AU-rich elements comprise sequences such as AUUUA (SEQ ID NO:49), AUUUUA (SEQ ID NO:50) and AUUUUUA (SEQ ID NO:51). Binding by such HuR proteins to CD83 mRNA is thought to increase the stability, transport and translation of CD83 mRNA, and thereby increase the expression of CD83 polypeptides.
  • CD83 expression may be increase by administering HuR proteins or nucleic acids to a mammal.
  • CD83 expression may be decreased by administering factors that block HuR binding to CD83 mRNA.
  • Factors that block HuR binding include proteins or nucleic acids that can bind to the AU-rich elements normally bound by HuR, for example, nucleic acids or anti-sense nucleic acids that are complementary to AU-rich elements.
  • organic molecules may be used to modulate the immune system. These compounds include any compound that can interact with a component of the immune system. Such compounds may interact directly with CD83, indirectly with CD83 or with some other polypeptide, cell or factor that plays a role in the function of the immune system. In some embodiments, the organic molecule can bind to a CD83 polypeptide or a CD83 nucleic acid.
  • Organic molecules can be tested or assayed for their ability to modulate CD83 activity, CD83 function or for their ability to modulate components of the immune system.
  • suitable organic molecules may be selected either from a chemical library, wherein chemicals are assayed individually, or from combinatorial chemical libraries where multiple compounds are assayed at once, then deconvoluted to determine and isolate the most active compounds.
  • combinatorial chemical libraries include those described by Agrafiotis et al., “System and method of automatically generating chemical compounds with desired properties,” U.S. Pat. No. 5,463,564; Armstrong, R. W., “Synthesis of combinatorial arrays of organic compounds through the use of multiple component combinatorial array syntheses,” WO 95/02566; Baldwin, J. J. et al., “Sulfonamide derivatives and their use,” WO 95/24186; Baldwin, J. J. et al., “Combinatorial dihydrobenzopyran library,” WO 95/30642; Brenner, S “New kit for preparing combinatorial libraries,” WO 95/16918; Chenera, B.
  • Peptide molecules that modulate the immune system may be obtained through the screening of combinatorial peptide libraries.
  • Such libraries may either be prepared by one of skill in the art (see e.g., U.S. Pat. Nos. 4,528,266 and 4,359,535, and Patent Cooperation Treaty Publication Nos. WO 92/15679, WO 92/15677, WO 90/07862, WO 90/02809, or purchased from commercially available sources (e.g., New England Biolabs Ph.D.TM Phage Display Peptide Library Kit).
  • the invention provides a method for identifying ligands, receptors, therapeutic drugs and other molecules that can modulate the phenotype of the mutant CD83 in vivo.
  • This method involves administering a test compound to the mutant CD83 mouse of the invention and observing whether the compound causes a change in the phenotype of the mutant mouse.
  • Changes in phenotype that are of interest include increases or decreases in T cells (especially CD4+ T cells), increases or decreases in GMCSF, IL-2, IL-4 or IL-10 cytokine production, increases or decreases in inflammation, increases or decreases in dendritic cell function and other T cell responses known to one of skill in the art.
  • Test compounds can be screened in vitro to ascertain whether they interact directly with CD83.
  • In vitro screening can, for example, identify whether a test compound or molecule can bind to the cytoplasmic tail or the membrane-associated portions of CD83.
  • Such information combined with observation of the in vivo phenotype before and after administration of the test compound provides further insight into the function of CD83 and provides targets for manipulation T cell activation and other functions modulated by CD83.
  • the invention is not limited to identification of molecules that directly associate with CD83.
  • the in vivo screening methods provided herein can, also identify test compounds that have an indirect effect on CD83, or that partially or completely replace a function of CD83.
  • T cell numbers can be observed in blood samples or in samples obtained from thymus, spleen or lymph node tissues.
  • dendritic cells can be pulsed with antigens ex vivo and then injected into mice to prime CD4+ T cells in draining lymphoid organs. See Inaba et al., J. Exp. Med. 172: 631-640, 1990; Liu, et al., J. Exp. Med. 177: 1299-1307, 1993; Sornasse et al., J. Exp. Med. 175: 15-21, 1992.
  • Antigens can also be deposited intramuscularly and dendritic cells from the corresponding afferent lymphatics can carry that antigen in a form stimulatory for T cells. Bujdoso et al., J. Exp. Med. 170: 1285-1302, 1989. According to the invention, factors stimulating the interaction of dendritic cells with T cells in vivo can be identified by administering antigens in this manner and then observing how T cell respond, e.g. by observing whether T cell activation occurs.
  • the CD83 polypeptides and antibodies of the invention are administered so as to achieve a reduction in at least one symptom associated with an infection, indication or disease.
  • the polypeptide or antibody, a variant thereof or a combination thereof may be administered as single or divided dosages, for example, of at least about 0.01 mg/kg to about 500 to 750 mg/kg, of at least about 0.01 mg/kg to about 300 to 500 mg/kg, at least about 0.1 mg/kg to about 100 to 300 mg/kg or at least about 1 mg/kg to about 50 to 100 mg/kg of body weight, although other dosages may provide beneficial results.
  • the amount administered will vary depending on various factors including, but not limited to, the polypeptide or antibody chosen, the disease, the weight, the physical condition, the health, the age of the mammal, whether prevention or treatment is to be achieved, and if the polypeptide or antibody is chemically modified. Such factors can be readily determined by the clinician employing animal models or other test systems that are available in the art.
  • Administration of the therapeutic agents in accordance with the present invention may be in a single dose, in multiple doses, in a continuous or intermittent manner, depending, for example, upon the recipient's physiological condition, whether the purpose of the administration is therapeutic or prophylactic, and other factors known to skilled practitioners.
  • the administration of the CD83 polypeptides and antibodies of the invention may be essentially continuous over a preselected period of time or may be in a series of spaced doses. Both local and systemic administration is contemplated.
  • CD83 polypeptides and antibodies are synthesized or otherwise obtained, purified as necessary or desired and then lyophilized and stabilized.
  • the polypeptide or antibody can then be adjusted to the appropriate concentration, and optionally combined with other agents.
  • the absolute weight of a given polypeptide or antibody included in a unit dose can vary widely. For example, about 0.01 to about 2 g, or about 0.1 to about 500 mg, of at least one polypeptide or antibody of the invention, or a plurality of CD83 polypeptides and antibodies specific for a particular cell type can be administered.
  • the unit dosage can vary from about 0.01 g to about 50 g, from about 0.01 g to about 35 g, from about 0.1 g to about 25 g, from about 0.5 g to about 12 g, from about 0.5 g to about 8 g, from about 0.5 g to about 4 g, or from about 0.5 g to about 2 g.
  • Daily doses of the CD83 polypeptides or antibodies of the invention can vary as well. Such daily doses can range, for example, from about 0.1 g/day to about 50 g/day, from about 0.1 g/day to about 25 g/day, from about 0.1 g/day to about 12 g/day, from about 0.5 g/day to about 8 g/day, from about 0.5 g/day to about 4 g/day, and from about 0.5 g/day to about 2 g/day.
  • one or more suitable unit dosage forms comprising the therapeutic CD83 polypeptides or antibodies of the invention can be administered by a variety of routes including oral, parenteral (including subcutaneous, intravenous, intramuscular and intraperitoneal), rectal, dermal, transdermal, intrathoracic, intrapulmonary and intranasal (respiratory) routes.
  • the therapeutic CD83 polypeptides or antibodies may also be formulated for sustained release (for example, using microencapsulation, see WO 94/07529, and U.S. Pat. No. 4,962,091).
  • the formulations may, where appropriate, be conveniently presented in discrete unit dosage forms and may be prepared by any of the methods well known to the pharmaceutical arts. Such methods may include the step of mixing the therapeutic agent with liquid carriers, solid matrices, semi-solid carriers, finely divided solid carriers or combinations thereof, and then, if necessary, introducing or shaping the product into the desired delivery system.
  • the therapeutic CD83 polypeptides or antibodies of the invention are prepared for oral administration, they are generally combined with a pharmaceutically acceptable carrier, diluent or excipient to form a pharmaceutical formulation, or unit dosage form.
  • a pharmaceutically acceptable carrier diluent or excipient
  • the CD83 polypeptides or antibodies may be present as a powder, a granular formulation, a solution, a suspension, an emulsion or in a natural or synthetic polymer or resin for ingestion of the active ingredients from a chewing gum.
  • the active CD83 polypeptides or antibodies may also be presented as a bolus, electuary or paste.
  • CD83 polypeptides or antibodies of the invention can also be formulated for sustained release, e.g., the CD83 polypeptides or antibodies can be coated, micro-encapsulated, or otherwise placed within a sustained delivery device.
  • the total active ingredients in such formulations comprise from 0.1 to 99.9% by weight of the formulation.
  • pharmaceutically acceptable it is meant a carrier, diluent, excipient, and/or salt that is compatible with the other ingredients of the formulation, and not deleterious to the recipient thereof.
  • compositions containing the therapeutic CD83 polypeptides or antibodies of the invention can be prepared by procedures known in the art using well-known and readily available ingredients.
  • the polypeptide or antibody can be formulated with common excipients, diluents, or carriers, and formed into tablets, capsules, solutions, suspensions, powders, aerosols and the like.
  • excipients, diluents, and carriers that are suitable for such formulations include buffers, as well as fillers and extenders such as starch, cellulose, sugars, mannitol, and silicic derivatives.
  • Binding agents can also be included such as carboxymethyl cellulose, hydroxymethylcellulose, hydroxypropyl methylcellulose and other cellulose derivatives, alginates, gelatin, and polyvinyl-pyrrolidone.
  • Moisturizing agents can be included such as glycerol, disintegrating agents such as calcium carbonate and sodium bicarbonate.
  • Agents for retarding dissolution can also be included such as paraffin.
  • Resorption accelerators such as quaternary ammonium compounds can also be included.
  • Surface active agents such as cetyl alcohol and glycerol monostearate can be included.
  • Adsorptive carriers such as kaolin and bentonite can be added.
  • Lubricants such as talc, calcium and magnesium stearate, and solid polyethyl glycols can also be included. Preservatives may also be added.
  • the compositions of the invention can also contain thickening agents such as cellulose and/or cellulose derivatives. They may also contain gums such as xanthan, guar or carbo gum or gum arabic, or alternatively polyethylene glycols, bentones and montmorillonites, and the like.
  • tablets or caplets containing the CD83 polypeptides or antibodies of the invention can include buffering agents such as calcium carbonate, magnesium oxide and magnesium carbonate.
  • Caplets and tablets can also include inactive ingredients such as cellulose, pregelatinized starch, silicon dioxide, hydroxy propyl methyl cellulose, magnesium stearate, microcrystalline cellulose, starch, talc, titanium dioxide, benzoic acid, citric acid, corn starch, mineral oil, polypropylene glycol, sodium phosphate, zinc stearate, and the like.
  • Hard or soft gelatin capsules containing at least one polypeptide or antibody of the invention can contain inactive ingredients such as gelatin, microcrystalline cellulose, sodium lauryl sulfate, starch, talc, and titanium dioxide, and the like, as well as liquid vehicles such as polyethylene glycols (PEGs) and vegetable oil.
  • inactive ingredients such as gelatin, microcrystalline cellulose, sodium lauryl sulfate, starch, talc, and titanium dioxide, and the like
  • liquid vehicles such as polyethylene glycols (PEGs) and vegetable oil.
  • enteric-coated caplets or tablets containing one or more CD83 polypeptides or antibodies of the invention are designed to resist disintegration in the stomach and dissolve in the more neutral to alkaline environment of the duodenum.
  • the therapeutic CD83 polypeptides or antibodies of the invention can also be formulated as elixirs or solutions for convenient oral administration or as solutions appropriate for parenteral administration, for instance by intramuscular, subcutaneous, intraperitoneal or intravenous routes.
  • the pharmaceutical formulations of the therapeutic CD83 polypeptides or antibodies of the invention can also take the form of an aqueous or anhydrous solution or dispersion, or alternatively the form of an emulsion or suspension or salve.
  • the therapeutic CD83 polypeptides or antibodies may be formulated for parenteral administration (e.g., by injection, for example, bolus injection or continuous infusion) and may be presented in unit dose form in ampules, pre-filled syringes, small volume infusion containers or in multi-dose containers. As noted above, preservatives can be added to help maintain the shelve life of the dosage form.
  • the active CD83 polypeptides or antibodies and other ingredients may form suspensions, solutions, or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • the active CD83 polypeptides or antibodies and other ingredients may be in powder form, obtained by aseptic isolation of sterile solid or by lyophilization from solution, for constitution with a suitable vehicle, e.g., sterile, pyrogen-free water, before use.
  • a suitable vehicle e.g., sterile, pyrogen-free water
  • formulations can contain pharmaceutically acceptable carriers, vehicles and adjuvants that are well known in the art. It is possible, for example, to prepare solutions using one or more organic solvent(s) that is/are acceptable from the physiological standpoint, chosen, in addition to water, from solvents such as acetone, ethanol, isopropyl alcohol, glycol ethers such as the products sold under the name “Dowanol,” polyglycols and polyethylene glycols, C 1 -C 4 alkyl esters of short-chain acids, ethyl or isopropyl lactate, fatty acid triglycerides such as the products marketed under the name “Miglyol,” isopropyl myristate, animal, mineral and vegetable oils and polysiloxanes.
  • organic solvent(s) that is/are acceptable from the physiological standpoint, chosen, in addition to water, from solvents such as acetone, ethanol, isopropyl alcohol, glycol ethers such as the products sold under the name “Dowanol,” polyg
  • antioxidants chosen from antioxidants, surfactants, other preservatives, film-forming, keratolytic or comedolytic agents, perfumes, flavorings and colorings.
  • Antioxidants such as t-butylhydroquinone, butylated hydroxyanisole, butylated hydroxytoluene and ⁇ -tocopherol and its derivatives can be added.
  • combination products that include one or more CD83 polypeptides or antibodies of the present invention and one or more other anti-microbial agents.
  • antibiotics can be included in the pharmaceutical compositions of the invention, such as aminoglycosides (e.g., streptomycin, gentamicin, sisomicin, tobramycin and amicacin), ansamycins (e.g. rifamycin), antimycotics (e.g. polyenes and benzofuran derivatives), ⁇ -lactams (e.g.
  • penicillins and cephalosporins include chloramphenical (including thiamphenol and azidamphenicol), linosamides (lincomycin, clindamycin), macrolides (erythromycin, oleandomycin, spiramycin), polymyxins, bacitracins, tyrothycin, capreomycin, vancomycin, tetracyclines (including oxytetracycline, minocycline, doxycycline), phosphomycin and fusidic acid.
  • the CD83 polypeptides or antibodies are well suited to to formulation as sustained release dosage forms and the like.
  • the formulations can be so constituted that they release the active polypeptide or antibody, for example, in a particular part of the intestinal or respiratory tract, possibly over a period of time.
  • Coatings, envelopes, and protective matrices may be made, for example, from polymeric substances, such as polylactide-glycolates, liposomes, microemulsions, microparticles, nanoparticles, or waxes. These coatings, envelopes, and protective matrices are useful to coat indwelling devices, e.g., stents, catheters, peritoneal dialysis tubing, draining devices and the like.
  • the therapeutic agents may be formulated as is known in the art for direct application to a target area.
  • Forms chiefly conditioned for topical application take the form, for example, of creams, milks, gels, dispersion or microemulsions, lotions thickened to a greater or lesser extent, impregnated pads, ointments or sticks, aerosol formulations (e.g., sprays or foams), soaps, detergents, lotions or cakes of soap.
  • aerosol formulations e.g., sprays or foams
  • Other conventional forms for this purpose include wound dressings, coated bandages or other polymer coverings, ointments, creams, lotions, pastes, jellies, sprays, and aerosols.
  • the therapeutic CD83 polypeptides or antibodies of the invention can be delivered via patches or bandages for dermal administration.
  • the polypeptide or antibody can be formulated to be part of an adhesive polymer, such as polyacrylate or acrylate/vinyl acetate copolymer.
  • an adhesive polymer such as polyacrylate or acrylate/vinyl acetate copolymer.
  • the backing layer can be any appropriate thickness that will provide the desired protective and support functions. A suitable thickness will generally be from about 10 to about 200 microns.
  • Ointments and creams may, for example, be formulated with an aqueous or oily base with the addition of suitable thickening and/or gelling agents.
  • Lotions may be formulated with an aqueous or oily base and will in general also contain one or more emulsifying agents, stabilizing agents, dispersing agents, suspending agents, thickening agents, or coloring agents.
  • the active CD83 polypeptides or antibodies can also be delivered via iontophoresis, e.g., as disclosed in U.S. Pat. Nos. 4,140,122; 4,383,529; or 4,051,842.
  • the percent by weight of a therapeutic agent of the invention present in a topical formulation will depend on various factors, but generally will be from 0.01% to 95% of the total weight of the formulation, and typically 0.1-85% by weight.
  • Drops such as eye drops or nose drops, may be formulated with one or more of the therapeutic CD83 polypeptides or antibodies in an aqueous or non-aqueous base also comprising one or more dispersing agents, solubilizing agents or suspending agents.
  • Liquid sprays are conveniently delivered from pressurized packs. Drops can be delivered via a simple eye dropper-capped bottle, or via a plastic bottle adapted to deliver liquid contents dropwise, via a specially shaped closure.
  • the therapeutic polypeptide or antibody may further be formulated for topical administration in the mouth or throat.
  • the active ingredients may be formulated as a lozenge further comprising a flavored base, usually sucrose and acacia or tragacanth; pastilles comprising the composition in an inert base such as gelatin and glycerin or sucrose and acacia; and mouthwashes comprising the composition of the present invention in a suitable liquid carrier.
  • the pharmaceutical formulations of the present invention may include, as optional ingredients, pharmaceutically acceptable carriers, diluents, solubilizing or emulsifying agents, and salts of the type that are available in the art.
  • pharmaceutically acceptable carriers such as physiologically buffered saline solutions and water.
  • diluents such as phosphate buffered saline solutions pH 7.0-8.0.
  • the CD83 polypeptides or antibodies of the invention can also be administered to the respiratory tract.
  • the present invention also provides aerosol pharmaceutical formulations and dosage forms for use in the methods of the invention.
  • dosage forms comprise an amount of at least one of the agents of the invention effective to treat or prevent the clinical symptoms of a specific infection, indication or disease. Any statistically significant attenuation of one or more symptoms of an infection, indication or disease that has been treated pursuant to the method of the present invention is considered to be a treatment of such infection, indication or disease within the scope of the invention.
  • the composition may take the form of a dry powder, for example, a powder mix of the therapeutic agent and a suitable powder base such as lactose or starch.
  • the powder composition may be presented in unit dosage form in, for example, capsules or cartridges, or, e.g., gelatin or blister packs from which the powder may be administered with the aid of an inhalator, insufflator, or a metered-dose inhaler (see, for example, the pressurized metered dose inhaler (MDI) and the dry powder inhaler disclosed in Newinan, S. P. in Aerosols and the Lung , Clarke, S. W. and Davia, D. eds., pp. 197-224, Butterworths, London, England, 1984).
  • MDI pressurized metered dose inhaler
  • the dry powder inhaler disclosed in Newinan, S. P. in Aerosols and the Lung , Clarke, S. W. and Davia, D. eds., pp.
  • CD83 polypeptides or antibodies of the present invention can also be administered in an aqueous solution when administered in an aerosol or inhaled form.
  • other aerosol pharmaceutical formulations may comprise, for example, a physiologically acceptable buffered saline solution containing between about 0.1 mg/ml and about 100 mg/ml of one or more of the CD83 polypeptides or antibodies of the present invention specific for the indication or disease to be treated.
  • Dry aerosol in the form of finely divided solid polypeptide or antibody or nucleic acid particles that are not dissolved or suspended in a liquid are also useful in the practice of the present invention.
  • CD83 polypeptides or antibodies of the present invention may be formulated as dusting powders and comprise finely divided particles having an average particle size of between about 1 and 5 ⁇ m, alternatively between 2 and 3 ⁇ m.
  • Finely divided particles may be prepared by pulverization and screen filtration using techniques well known in the art.
  • the particles may be administered by inhaling a predetermined quantity of the finely divided material, which can be in the form of a powder.
  • the unit content of active ingredient or ingredients contained in an individual aerosol dose of each dosage form need not in itself constitute an effective amount for treating the particular infection, indication or disease since the necessary effective amount can be reached by administration of a plurality of dosage units.
  • the effective amount may be achieved using less than the dose in the dosage form, either individually, or in a series of administrations.
  • the therapeutic CD83 polypeptides or antibodies of the invention are conveniently delivered from a nebulizer or a pressurized pack or other convenient means of delivering an aerosol spray.
  • Pressurized packs may comprise a suitable propellant such as dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • the dosage unit may be determined by providing a valve to deliver a metered amount.
  • Nebulizers include, but are not limited to, those described in U.S. Pat. Nos.
  • Aerosol delivery systems of the type disclosed herein are available from numerous commercial sources including Fisons Corporation (Bedford, Mass.), Schering Corp. (Kenilworth, N.J.) and American Pharmoseal Co., (Valencia, Calif.).
  • the therapeutic agent may also be administered via nose drops, a liquid spray, such as via a plastic bottle atomizer or metered-dose inhaler.
  • atomizers are the Mistometer (Wintrop) and the Medihaler (Riker).
  • the active ingredients may also be used in combination with other therapeutic agents, for example, pain relievers, anti-inflammatory agents, antihistamines, bronchodilators and the like, whether for the conditions described or some other condition.
  • other therapeutic agents for example, pain relievers, anti-inflammatory agents, antihistamines, bronchodilators and the like, whether for the conditions described or some other condition.
  • the present invention further pertains to a packaged pharmaceutical composition for controlling microbial infections such as a kit or other container.
  • a packaged pharmaceutical composition for controlling microbial infections such as a kit or other container.
  • the kit or container holds a therapeutically effective amount of a pharmaceutical composition for controlling microbial infections and instructions for using the pharmaceutical composition for control of the microbial infection.
  • the pharmaceutical composition includes at least one polypeptide or antibody of the present invention, in a therapeutically effective amount such that the selected disease or immunological condition is controlled.
  • mice Male C57BL6 mice received 3 weekly injections of N-ethyl-N-nitrosourea (ENU) at a concentration of 100 mg/kg. N-Ethyl-N-nitrosourea was quantified prior to injection by spectrophotometry. Mice that regained fertility after a minimum period of 12 weeks were then used to generate pedigree founder G1 animals. G1 male mice were crossed to C57BL6J females and their female progeny (G2 animals) crossed back to their fathers to generate G3 animals for screening.
  • ENU N-ethyl-N-nitrosourea
  • G3 mice were weaned at 3 weeks of age. Each animal then underwent a series of screens designed to assess a number of parameters, including immune function, inflammatory response and bone development.
  • 150-200 ul of whole blood was collected by retro-orbital bleed into heparinized tubes. Cells were pelleted and red blood cells lysed. Samples were then stained with antibodies to cell surface markers expressed on distinct lymphoid and myeloid sub-populations. These samples were analyzed by flow-cytometry.
  • the number of phenodeviants identified (2 from a litter of 9 animals) was suggestive of a trait controlled by a single gene and inherited in a Mendelian fashion.
  • the phenotype was designated LCD4.1 (Low CD4 Mutant 1) and was used for mapping experiments.
  • DNA samples were prepared from samples of tail tissue collected from these N2 mice and used for a genome scan, using a collection of assembled markers, and performed on the ABI 3100 DNA analyzer.
  • Initial genetic linkage was seen to the tip of chromosome 13, where the closest microsatellite marker was D13Mit139 with a LOD score of 8.2.
  • the mutant gene was located between 13.4 and 29.6 cM on chromosome 13. Through additional genotyping, this region was reduced to an 11 cM interval on chromosome 13. No significant linkage to other chromosomal regions was seen.
  • CD83 A candidate gene, CD83, was identified for gene-testing based upon its reported position within the interval. CD83 has previously been used as a marker of dendritic cell activation, suggesting that it might play a role in dendritic cell function and hence in regulating T cell development and function.
  • Dendritic cells can be differentiated from bone marrow of wild type mice by culture in GM-CSF. These cells can be characterized by the surface expression of dendritic cell markers, including CD86 and CD11c. Both LCD4.1 affected and normal animals were capable of giving rise to CD86+CD11+ cells under these culture conditions. LCD4.1 mutant mice thus were capable of generating dendritic cells under in vitro culture conditions. These data suggest that the phenotype seen in LCD4.1 mice is not due to a failure of dendritic cells to develop but rather may reflect a defect in function.
  • FITC FITC labeled cells expressing CD86 were detected in equal proportions in draining lymph node from normal and affected LCD4.1 mice.
  • Spleens were removed from wild type and mutant mice and digested with collagenase to liberate dendritic cells. Spleens were stained for surface expression of CD4 (helper T cells) and CD11c (dendritic cells). Cells expressing these markers were purified by fluorescence activated cell sorting (FACS sorting). CD11c and CD4+ positive cells were also purified from an allogeneic mouse strain, BALBc.
  • Dendritic cells from BALBc animals were used to stimulate CD4+ T cells from wild type and mutant mice.
  • dendritic cells prepared from wild type and mutant mice were used to stimulate BALBc CD4+ T cells. After 5 days in culture proliferative responses were measured by incorporation of tritiated thymidine.
  • Dendritic cells from wild type and mutant mice were both capable of activating allogeneic T cells, suggesting that dendritic cell function was unimpaired in the mutant animal ( FIG. 6 a ).
  • CD4+ T cells from mutant animals exhibited a diminished response after 5 days of stimulation ( FIG. 6 b ).
  • CD83 may have a novel requirement for CD83 functionality on T cells during allogeneic activation.
  • CD83 may be influencing the extent of CD4+ T cell activation or altering the duration of the CD4+ T cell proliferative response.
  • the therapeutic manipulation of CD83 may thus represent a mechanism for the specific regulation of T cell function in the treatment of T cell mediated diseases, including autoimmune disorders.
  • Antibodies capable of blocking CD83 function may be used as therapeutics in the treatment of immune diseases whilst the activation of CD83 may have utility in enhancing immune responses in cancer and other circumstances.
  • CD83 has been described as a marker of dendritic cell activation there is little data as to its function in vivo.
  • the mutation provided by the invention destabilizes or inactivates the protein and leads to impaired surface expression.
  • CD4+ T cell function is impaired although the development of dendritic cells is not inhibited and mutant dendritic cells retain functionality. This results in the impaired development of CD4+ T cells. This impaired ability to activate T cells is also seen in a slight decrease in contact sensitivity responses in LCD4.1 mutant mice.
  • Spleens cells from 6-8-week-old homozygous CD83 wild type or CD83 mutant (LCD4.1) mice were used to isolate CD4 + T-cells by positive selection using magnetic beads (Miltenyi Biotec).
  • a 96 round bottom plate was coated with 50 ⁇ L, per well of a solution containing either 1 or 10 ⁇ g/mL of anti-CD3 and 0.1 or 0.2 ⁇ g/mL of anti-CD28 antibodies (both from Pharmingen) in PBS overnight. This plate was then washed using 150 ⁇ L of PBS three times.
  • 20,000 CD4 + T-cells (either wild type or CD83 mutant) were added in a 200 ⁇ L final volume of RPMI containing 10% FBS, 55 ⁇ M ⁇ -mercaptoethanol and antibiotics. The plates were then incubated in a CO 2 incubator at 37° C. for 44 to 72 hours.
  • cytokine levels supernatants were harvested and cytokines were measured using either a Cytometric Bead Array system (Pharmingen) or ELISA (R&D).
  • RNA measurements the cells were harvested and RNA was isolated using Tri reagent (Sigma). IL-10 and IL-4 mRNA levels were measured by reverse transcription and TaqMan (Applied Biosystems) analysis.
  • FIG. 7 shows the IL-2, IL-4, IL-5, TNF ⁇ and IFN ⁇ levels produced by either wild type or CD83 mutant CD4 + T-cells.
  • Purified cells were incubated as described above in the presence of 1 ⁇ g/mL of anti-CD3 and 0.2 ⁇ g/mL of anti-CD28 antibodies for 72 hours. The supernatants were then simultaneously analyzed for production of IL-2, IL-4, IL-5, TNF ⁇ and IFN ⁇ using the cytometric bead array system from Pharmingen.
  • FIG. 7 demonstrates that CD4 + T-cells from CD83 mutant animals expressed higher levels of IL-4 and lower levels of IL-2 compared to CD4 + T-cells from CD83 wild type animals.
  • Other cytokines and a new set of stimulation assays were analyzed including the production levels of IL-10 and GMCSF by these cells ( FIGS. 8 and 9 ). In both cases, cells from mutant animals produce larger amounts of IL-10 and GMCSF than did wild type animals.
  • FIG. 10 shows that mRNA levels for both IL-4 and IL-10 were increased in cells from activated mutant CD83, CD4 + T-cells compared with cells from wild type animals.
  • CD4 + T-cells were isolated and activated as mentioned above in the presence of increasing concentrations of anti-CD83 antibodies.
  • CD4 + T-cells were isolated from an OT2tg [transgenic mice with a T-cell receptor specific for chicken ovalbumin (OVA) 323-339 peptide].
  • OT2tg transgenic mice with a T-cell receptor specific for chicken ovalbumin (OVA) 323-339 peptide.
  • Dendritic cells were isolated from a C57BL6 mouse by a negative selection using B220 magnetic beads (Miltenyi Biotec) followed by positive selection using CD11-c magnetic beads (Milteny Biotec).
  • CD4 + T-cells Five thousand CD4 + T-cells were then mixed with five thousand dendritic cells in a 96 well plate in the presences of 1 ⁇ M OVA peptide using RPMI (55 ⁇ M BME, 10% FBS plus antibiotics) in a final 200 uL volume. These cells were then incubated for 48 to 72 hours in a CO 2 incubator at 37° C. and pulsed using [ 3 H] thymidine for 8 hours. Cells were then harvested and [ 3 H] thymidine incorporation was quantified using a top counter.
  • RPMI 55 ⁇ M BME, 10% FBS plus antibiotics
  • anti-CD83 antibodies decreased production of IL-4 by activated CD4 + T-cells in a dose dependent manner. Different antibody preparations did provide somewhat different degrees of inhibition of IL-4 production ( FIG. 11 ). Accordingly, the epitope and/or degree of affinity of the antibodies for the CD83 antigen may influence whether or not IL-4 production is significantly inhibited.
  • CD4 + T-cells derived from these TCR transgenic animals express high levels of a T-cell receptor specific for chicken ovalbumin (OVA) 323-339 peptide and thus have high levels of proliferation when mixed with antigen presenting cells (dendritic cells were used) in the presence of the OVA peptide.
  • OVA ovalbumin
  • anti-CD83 antibodies were able to decrease proliferation of CD4 + T-cells in this system ( FIG. 12 ).
  • different antibody preparations had somewhat different effects on the proliferation of CD4 + T-cells. Accordingly, the CD83 epitope and/or degree of affinity of the antibodies for the CD83 antigen may influence whether or not CD4 + T-cell proliferation is significantly inhibited.
  • This Example illustrates that over expression of CD83 in transgenic mice leads to increased T-cell proliferation.
  • a 34.3 kb fragment of normal mouse genomic DNA, including the ⁇ 18 kb coding region of the CD83 gene, as well as ⁇ 10.6 kb of upstream flanking sequences and ⁇ 5.7 kb of downstream sequences was microinjected into normal mouse one-cell embryos.
  • Four individual founder animals were generated.
  • Transgenic mice were then crossed to a male OT2tg mouse.
  • Male offspring carrying both the CD83 and OT2 transgene were used to analyze peptide specific T-cell proliferation.
  • CD4 + T-cells and dendritic cells were isolated from either OT2tg [transgenic mice with a T-cell receptor specific for chicken ovalbumin (OVA) 323-339 peptide] CD83 wild type or from OT2tg CD83 transgenic mice as described above (Example 4).
  • OT2tg CD4 + T-cells from either wild type or CD83 transgenic animals were then mixed with five thousand wild type dendritic cells or five thousand CD83 transgenic dendritic cells in a 96 well plate in the presence of increasing concentrations of OVA peptide using RPMI (55 ⁇ M BME, 10% FBS plus antibiotics) in a final 200 uL volume.
  • OT2tg CD4 + T-cells derived from CD83 transgenic mice proliferated at higher rates than the same cell population derived from a CD83 wild type animal ( FIG. 13 ). This increased proliferation was seen at all the concentrations of OVA peptide tested. Whereas OT2tg CD4 + T-cells derived from CD83 transgenic animals exhibited increased proliferation, dendritic cells from CD83 transgenic animals did not exhibit a substantial increase in proliferation. Therefore, it appears that transgenic expression in the CD4 + T-cell, and not in dendritic cells is what led to the increased proliferation of CD4 + T-cells.
  • This Example shows that antibodies raised against the mouse CD83 protein can inhibit proliferation of human peripheral blood mononuclear cells.
  • Rabbit polyclonal sera was raised against mouse CD83 protein by immunizing rabbits using a mouse CD83 external domain protein fused to a rabbit Ig domain ( FIG. 14 ).
  • Pre-immune sera and anti-mouse polyclonal sera were then purified using a protein A column (Pharmacia Biotech) as described by the manufacturer, then dialyzed against PBS and stored at 4° C.
  • a titer was obtained using an antigen specific ELISA ( FIG. 15 ). As illustrated by FIG. 15 , a good polyclonal response was obtained against the mouse CD83 protein.
  • PBMCs Human peripheral blood mononuclear cells
  • PBMCs Human peripheral blood mononuclear cells
  • media RPMI, 10% FBS, antibiotics
  • PHA Phaseolus vulgaris leucoagglutinin
  • FIG. 16 illustrates that proliferation of PHA-activated human PBMCs was inhibited by antibodies raised against the external region of the mouse CD83 protein. Proliferation of PHA-activated human PBMCs was not affected by addition of increasing concentrations of protein A purified rabbit pre-immune sera. When increasing concentrations of protein A purified rabbit polyclonal sera raised against the mouse CD83 protein was added, a concentration dependent decrease in proliferation was observed.

Abstract

The invention provides methods for modulating cytokine levels, GM-CSF levels and the immune system using CD83 nucleic acids, CD83 polypeptides, anti-CD83 antibodies and factors that influence CD83 activity or expression. The invention also provides mice having a mutant CD83 gene and mice having a transgenic CD83 gene, which are useful for defining the role of CD83 in the immune system and for identifying compounds that can modulate CD83 and the immune system.

Description

  • This application is related to U.S. Application Ser. No. 60/331,958 filed Nov. 21, 2001.
  • FIELD OF THE INVENTION
  • The invention relates to an altered CD83 gene product, and methods of modulating cytokine levels by modulating the expression of mutant and wild type CD83 gene products produced in a mammal. The invention also relates to the regulation of T cell and dendritic cell activity and conditions and treatments related thereto.
  • BACKGROUND OF THE INVENTION
  • CD83 is a 45 kilodalton glycoprotein that is predominantly expressed on the surface of dendritic cells and other cells of the immune system. Structural analysis of the predicted amino acid sequence of CD83 indicates that it is a member of the immunoglobulin superfamily. See, Zhou et al., J. Immunol. 149:735 (1992)). U.S. Pat. No. 5,316,920 and WO 95/29236 disclose further information about CD83. While such information suggests that CD83 plays a role in the immune system, that role is undefined, and the interrelationship of CD83 with cellular factors remains unclear.
  • Moreover, treatment of many diseases could benefit from more effective methods for increasing or decreasing the immune response. Hence, further information about how to modulate the immune system by using factors such as CD83 are needed.
  • SUMMARY OF THE INVENTION
  • The invention provides a method of modulating cytokine levels by modulating the activity or expression of the CD83 gene products. According to the invention, cytokine levels can be modulated in a mammal or in mammalian cells that are involved in the immune response, for example, antigen presenting cells or T cells.
  • The invention therefore provides a method of modulating cytokine production in a mammal or in an immune cell by modulating the activity or expression of a CD83 polypeptide. According to the invention, the production of a cytokine such as interleukin-2, interleukin-4, or interleukin-10 can be modulated by modulating the activity or expression of a CD83 polypeptide. In some embodiments, an antibody is used that can modulate the activity or expression of a CD83 polypeptide. For example, the antibody can be administered to the mammal or the immune cell can be contacted with the antibody. In some embodiments, the immune cells are T cells or antigen presenting cells. In other embodiments, the immune cells are CD4+ T cells.
  • The invention also provides a method of modulating granulocyte macrophage colony stimulating factor production in a mammal or in an immune cell by modulating the activity or expression of CD83 polypeptides. In some embodiments, an antibody is used that can modulate the activity or expression of a CD83 polypeptide. For example, the antibody can be administered to the mammal or the immune cell can be contacted with the antibody. In some embodiments, the immune cells are T cells or antigen presenting cells. In other embodiments, the immune cells are CD4+ T cells.
  • The invention also provides a method of modulating tumor necrosis factor production in a mammal or in a mammalian cell by modulating the activity or expression of CD83 polypeptides. In some embodiments, an antibody is used that can modulate the activity or expression of a CD83 polypeptide. For example, the antibody can be administered to the mammal or the mammalian cell can be contacted with the antibody. In some embodiments, the immune cells are T cells or antigen presenting cells. In other embodiments, the immune cells are CD4+ T cells.
  • The invention further provides a method of inhibiting proliferation of a human peripheral blood mononuclear cell by modulating the activity or expression of CD83 polypeptides. In some embodiments, an antibody is used that can modulate the activity or expression of a CD83 polypeptide. For example, the antibody can be administered to the mammal or the human peripheral blood mononuclear cell can be contacted with the antibody.
  • The invention also provides an antibody that can bind to a CD83 polypeptide comprising SEQ ID NO:4, SEQ ID NO:8 or SEQ ID NO:9, wherein activated CD4+ T-cells produce lower levels of interleukin-4 when the T-cells are contacted with the antibody. The invention further provides an antibody that can bind to a CD83 polypeptide comprising SEQ ID NO:4, SEQ ID NO:8 or SEQ ID NO:9, wherein CD4+ T-cells proliferation is decreased when the T-cells are contacted with the antibody. Such an antibody can have an amino acid sequence that includes SEQ ID NO:11, SEQ ID NO:13, SEQ ID NO:15, SEQ ID NO:17, SEQ ID NO:19, SEQ ID NO:21, SEQ ID NO:23, SEQ ID NO:24, SEQ ID NO:25, SEQ ID NO:26, SEQ ID NO:27, SEQ ID NO:28, SEQ ID NO:29, SEQ ID NO:30, SEQ ID NO:31, SEQ ID NO:32, SEQ ID NO:33, SEQ ID NO:34, SEQ ID NO:35, SEQ ID NO:36, SEQ ID NO:37, SEQ ID NO:38, SEQ ID NO:39, SEQ ID NO:40, SEQ ID NO:41, SEQ ID NO:42, SEQ ID NO:43, SEQ ID NO:44, SEQ ID NO:45, SEQ ID NO:46, SEQ ID NO:47, SEQ ID NO:48, SEQ ID NO:52, SEQ ID NO:53, SEQ ID NO:54, SEQ ID NO:55, SEQ ID NO:56, SEQ ID NO:57, SEQ ID NO:58, SEQ ID NO:60, SEQ ID NO:62 or SEQ ID NO:64. Nucleic acids encoding such an antibody can have, for example, a sequence that includes SEQ ID NO:12, SEQ ID NO:14, SEQ ID NO:16, SEQ ID NO:18, SEQ ID NO:20, SEQ ID NO:22, SEQ ID NO:59, SEQ ID NO:61, SEQ ID NO:63 or SEQ ID NO:65.
  • The invention also provides a method for decreasing the activity of a CD83 gene product, comprising contacting the CD83 gene product with an antibody that comprises SEQ ID NO:11, SEQ ID NO:13, SEQ ID NO:15, SEQ ID NO:17, SEQ ID NO:19, SEQ ID NO:21, SEQ ID NO:23, SEQ ID NO:24, SEQ ID NO:25, SEQ ID NO:26, SEQ ID NO:27, SEQ ID NO:28, SEQ ID NO:29, SEQ ID NO:30, SEQ ID NO:31, SEQ ID NO:32, SEQ ID NO:33, SEQ ID NO:34, SEQ ID NO:35, SEQ ID NO:36, SEQ ID NO:37, SEQ ID NO:38, SEQ ID NO:39, SEQ ID NO:40, SEQ ID NO:41, SEQ ID NO:42, SEQ ID NO:43, SEQ ID NO:44, SEQ ID NO:45, SEQ ID NO:46, SEQ ID NO:47, SEQ ID NO:48, SEQ ID NO:52, SEQ ID NO:53, SEQ ID NO:54, SEQ ID NO:55, SEQ ID NO:56, SEQ ID NO:57, SEQ ID NO:58, SEQ ID NO:60, SEQ ID NO:62 or SEQ ID NO:64. The activity of a CD83 gene product can be decreased in a mammal or in a cell that is involved in an immune response, for example, a T cell.
  • The invention further provides a method for decreasing the translation of a CD83 gene product in a mammalian cell, comprising contacting the mammalian cell with a nucleic acid complementary to a CD83 nucleic acid comprising SEQ ID NO:1, SEQ ID NO:3, SEQ ID NO:5, or SEQ ID NO:10.
  • In another embodiment, the invention provides a method for decreasing the translation of a CD83 gene product in a mammal, comprising administering to the mammal a nucleic acid complementary to a CD83 nucleic acid comprising SEQ ID NO:1, SEQ ID NO:3, SEQ ID NO:5, or SEQ ID NO:10.
  • The invention further provides a method for decreasing proliferation of CD4+ T-cells in a mammal comprising administering to the mammal an antibody that can bind to a CD83 gene product, wherein the CD83 gene product comprises SEQ ID NO:2 or SEQ ID NO:9. The antibody can have a sequence comprising SEQ ID NO:11, SEQ ID NO:13, SEQ ID NO:15, SEQ ID NO:17, SEQ ID NO:19, SEQ ID NO:21, SEQ ID NO:23, SEQ ID NO:24, SEQ ID NO:25, SEQ ID NO:26, SEQ ID NO:27, SEQ ID NO:28, SEQ ID NO:29, SEQ ID NO:30, SEQ ID NO:31, SEQ ID NO:32, SEQ ID NO:33, SEQ ID NO:34, SEQ ID NO:35, SEQ ID NO:36, SEQ ID NO:37, SEQ ID NO:38, SEQ ID NO:39, SEQ ID NO:40, SEQ ID NO:41, SEQ ID NO:42, SEQ ID NO:43, SEQ ID NO:44, SEQ ID NO:45, SEQ ID NO:46, SEQ ID NO:47, SEQ ID NO:48, SEQ ID NO:52, SEQ ID NO:53, SEQ ID NO:54, SEQ ID NO:55, SEQ ID NO:56, SEQ ID NO:57, SEQ ID NO:58, SEQ ID NO:60, SEQ ID NO:62 or SEQ ID NO:64.
  • The invention also provides a method for decreasing interleukin-2 levels and increasing interleukin-4 levels in a mammal comprising administering to the mammal an antibody that can bind to a CD83 gene product, wherein the CD83 gene product comprises SEQ ID NO:2 or SEQ ID NO:9. The antibody can have a sequence comprising SEQ ID NO:11, SEQ ID NO:13, SEQ ID NO:15, SEQ ID NO:17, SEQ ID NO:19, SEQ ID NO:21, SEQ ID NO:23, SEQ ID NO:24, SEQ ID NO:25, SEQ ID NO:26, SEQ ID NO:27, SEQ ID NO:28, SEQ ID NO:29, SEQ ID NO:30, SEQ ID NO:31, SEQ ID NO:32, SEQ ID NO:33, SEQ ID NO:34, SEQ ID NO:35, SEQ ID NO:36, SEQ ID NO:37, SEQ ID NO:38, SEQ ID NO:39, SEQ ID NO:40, SEQ ID NO:41, SEQ ID NO:42, SEQ ID NO:43, SEQ ID NO:44, SEQ ID NO:45, SEQ ID NO:46, SEQ ID NO:47, SEQ ID NO:48, SEQ ID NO:52, SEQ ID NO:53, SEQ ID NO:54, SEQ ID NO:55, SEQ ID NO:56, SEQ ID NO:57, SEQ ID NO:58, SEQ ID NO:60, SEQ ID NO:62 or SEQ ID NO:64.
  • The invention further provides a method for decreasing interleukin-2 levels and increasing interleukin-4 levels in a mammal comprising administering to the mammal a nucleic acid complementary to a CD83 nucleic acid comprising SEQ ID NO:1, SEQ ID NO:3, SEQ ID NO:5, or SEQ ID NO:10. In some embodiments the interleukin-2 levels are decreased and the interleukin-4 levels are increased to treat an autoimmune disease. In other embodiments, the interleukin-2 levels are decreased and the interleukin-4 levels are increased to stimulate production of Th2-associated cytokines in transplant recipients, for example, to prolong survival of transplanted tissues.
  • The invention also provides a method for increasing interleukin-10 levels in a mammal comprising administering to the mammal an antibody that can bind to a CD83 gene product, wherein the CD83 gene product comprises SEQ ID NO:2 or SEQ ID NO:9. The antibody can have a sequence comprising SEQ ID NO:11, SEQ ID NO:13, SEQ ID NO:15, SEQ ID NO:17, SEQ ID NO:19, SEQ ID NO:21, SEQ ID NO:23, SEQ ID NO:24, SEQ ID NO:25, SEQ ID NO:26, SEQ ID NO:27, SEQ ID NO:28, SEQ ID NO:29, SEQ ID NO:30, SEQ ID NO:31, SEQ ID NO:32, SEQ ID NO:33, SEQ ID NO:34, SEQ ID NO:35, SEQ ID NO:36, SEQ ID NO:37, SEQ ID NO:38, SEQ ID NO:39, SEQ ID NO:40, SEQ ID NO:41, SEQ ID NO:42, SEQ ID NO:43, SEQ ID NO:44, SEQ ID NO:45, SEQ ID NO:46, SEQ ID NO:47, SEQ ID NO:48, SEQ ID NO:52, SEQ ID NO:53, SEQ ID NO:54, SEQ ID NO:55, SEQ ID NO:56, SEQ ID NO:57, SEQ ID NO:58, SEQ ID NO:60, SEQ ID NO:62 or SEQ ID NO:64.
  • The invention further provides a method for increasing interleukin-10 levels in a mammal comprising administering to the mammal a nucleic acid complementary to a CD83 nucleic acid comprising SEQ ID NO:1, SEQ ID NO:3, SEQ ID NO:5, or SEQ ID NO:10. In some embodiments, the interleukin-10 levels are increased to treat neoplastic disease. In other embodiments, the interleukin-10 levels are increased to treat a tumor.
  • The invention also provides a method for increasing interleukin-2 levels in a mammal comprising administering to the mammal a functional CD83 polypeptide that comprises SEQ ID NO:9.
  • The invention further provides a method for increasing interleukin-2 levels in a mammal comprising: (a) transforming a T cell from the mammal with a nucleic acid encoding a functional CD83 polypeptide operably linked to a promoter functional in a mammalian cell, to generate a transformed T cell; (b) administering the transformed T cell to the mammal to provide increased levels of interleukin-2. In some embodiments, the CD83 polypeptide has a sequence that comprises SEQ ID NO:9 or the nucleic acid has a sequence that comprises SEQ ID NO:1, SEQ ID NO:3, SEQ ID NO:5, or SEQ ID NO:10. Such methods for increasing interleukin-2 levels can be used to treat an allergy or an infectious disease.
  • The invention also provides a method for increasing granulocyte macrophage colony stimulating factor levels in a mammal comprising administering to the mammal an antibody that can bind to a CD83 gene product, wherein the CD83 gene product comprises SEQ ID NO:2 or SEQ ID NO:9.
  • Such an antibody can have a sequence comprising SEQ ID NO:11, SEQ ID NO:13, SEQ ID NO:15, SEQ ID NO:17, SEQ ID NO:19, SEQ ID NO:21, SEQ ID NO:23, SEQ ID NO:24, SEQ ID NO:25, SEQ ID NO:26, SEQ ID NO:27, SEQ ID NO:28, SEQ ID NO:29, SEQ ID NO:30, SEQ ID NO:31, SEQ ID NO:32, SEQ ID NO:33, SEQ ID NO:34, SEQ ID NO:35, SEQ ID NO:36, SEQ ID NO:37, SEQ ID NO:38, SEQ ID NO:39, SEQ ID NO:40, SEQ ID NO:41, SEQ ID NO:42, SEQ ID NO:43, SEQ ID NO:44, SEQ ID NO:45, SEQ ID NO:46, SEQ ID NO:47, SEQ ID NO:48, SEQ ID NO:52, SEQ ID NO:53, SEQ ID NO:54, SEQ ID NO:55, SEQ ID NO:56, SEQ ID NO:57, SEQ ID NO:58, SEQ ID NO:60, SEQ ID NO:62 or SEQ ID NO:64.
  • The invention further provides a method for increasing granulocyte macrophage colony stimulating factor levels in a mammal comprising administering to the mammal a nucleic acid complementary to a CD83 nucleic acid comprising SEQ ID NO:1, SEQ ID NO:3, SEQ ID NO:5, or SEQ ID NO:10.
  • The invention also provides a method for increasing tumor necrosis factor levels at a selected site in a mammal comprising administering to the site a functional CD83 polypeptide. In another embodiment, the invention provides a method for increasing tumor necrosis factor levels in a selected mammalian cell comprising transforming the cell with a nucleic acid encoding a functional CD83 polypeptide. The CD83 polypeptide employed can, for example, have a sequence comprising SEQ ID NO:9.
  • Mammals and birds may be treated by the methods and compositions described and claimed herein. Such mammals and birds include humans, dogs, cats, and livestock, for example, horses, cattle, sheep, goats, chickens, turkeys and the like.
  • The invention further provides a mutant mouse that can serve as an animal model of diminished T cell activation or altered cytokine levels. The mutant mouse has an altered CD83 gene that produces a larger gene product, having SEQ ID NO:4 or containing SEQ ID NO:8. Also provided are methods of using the mutant mouse model to study the effects of cytokines on the immune system, inflammation, the function and regulation of CD83, T cell and dendritic cell activity, the immune response and conditions and treatments related thereto. Hence, the invention further provides a mutant mouse whose somatic and germ cells comprise a mutant CD83 gene encoding a polypeptide comprising SEQ ID NO:4 or SEQ ID NO:8, wherein expression of the mutant CD83 gene reduces CD4+ T cell activation. The mutant CD83 gene can, for example, comprise SEQ ID NO:3.
  • The invention further provides a method of identifying a compound that can modulate CD4+ T cell activation comprising administering a test compound to a mouse having a mutant or wild type transgenic CD83 gene and observing whether CD4+ T cell activation is decreased or increased. The somatic and/or germ cells of the mutant mouse can comprise a mutant CD83 gene encoding a polypeptide comprising SEQ ID NO:4 or SEQ ID NO:8. Alternatively, the somatic and/or germ cells of the mouse can contain a wild type CD83 gene, for example, SEQ ID NO:1 or SEQ ID NO:9.
  • The invention also provides a mutant CD83 gene encoding a polypeptide comprising SEQ ID NO:4 or SEQ ID NO:8. The invention further provides a mutant CD83 gene comprising nucleotide sequence SEQ ID NO:3.
  • DESCRIPTION OF THE FIGURES
  • FIG. 1 provides flow cytometry data for G3 animals. As shown, reduced numbers of CD4+ T cells are seen in two animals from Pedigree 9, mouse 9.4.1 and mouse 9.4.9. All other animals analyzed on that day exhibit normal numbers of CD4+ T cells.
  • FIG. 2 provides a graph of flow cytometry data for G3 animals. Each diamond symbol represents an individual animal. As shown, multiple animals from the N2 generation exhibit a reduced percentage of CD4+ T cells.
  • FIG. 3 provides the nucleotide sequence of wild type mouse CD83 (SEQ ID NO:1). The ATG start codon and the TGA stop codon are underlined.
  • FIG. 4A-B provides the nucleotide sequence of the mutant CD83 gene (SEQ ID NO:3) of the invention derived from the mutant LCD4.1 animal. The ATG start codon, the mutation and the TGA stop codon are underlined.
  • FIG. 5 provides the amino acid sequence for wild type (top, SEQ ID NO:2) and mutant (bottom, SEQ ID NO:4) CD83 coding regions. The additional C-terminal sequences arising because of the CD83 mutation are underlined.
  • FIG. 6A illustrates that dendritic cells from wild type (♦, WT DC) and mutant (▪, mutant DC) mice are capable of the allogeneic activation of CD4+ T cells. CD4+ T cells were stimulated with 10,000, 1000 or 100 dendritic cells for 5 days and proliferation measured by incorporation of tritiated thymidine.
  • FIG. 6B illustrates that CD4+ T cells from mutant mice (▪, mutant CD4) fail to respond to allogeneic stimulation with BALBc dendritic cells, although wild type animals (♦, WT CD4+) respond normally. CD4+ T cells were stimulated with 10,000, 1000 or 100 dendritic cells for 5 days and proliferation measured by incorporation of tritiated thymidine.
  • FIG. 7 provides a bar graph illustrating IL-2, IL-4, IL-5, TNFα, and IFNγ production from wild type CD4+ T cells (white bar) or CD83 mutant CD4+ T cells (dark bar) that had been stimulated with 1 μg/ml of anti-CD3 antibodies and 0.2 μg/ml of anti-CD28 antibodies for 72 hours. As illustrated, IL-2 levels are lower, and IL-4 levels are higher in the CD83 mutant T cells.
  • FIG. 8 provides a bar graph illustrating IL-10 production from wild type CD4+ T cells (white bar) or CD83 mutant CD4+ T cells (dark bar) that had been stimulated with 0.1 μg/ml of anti-CD28 antibodies and 1 to 10 μg/ml of anti-CD3 antibodies for 72 hours. As illustrated, IL-10 levels are higher in the CD83 mutant T cells.
  • FIG. 9 provides a bar graph illustrating GM-CSF production from wild type CD4+ T cells (white bar) or CD83 mutant CD4+ T cells (dark bar) that had been stimulated with anti-CD3 and anti-CD28 antibodies. As illustrated, GM-CSF production is higher in the CD83 mutant cells than in wild type cells.
  • FIG. 10A provides a bar graph illustrating IL-4 mRNA levels from wild type CD4+ T cells (white bar) or CD83 mutant CD4+ T cells (dark bar) that had been stimulated with anti-CD3 and anti-CD28 antibodies. As illustrated, the IL-4 mRNA levels are higher in the CD83 mutant cells.
  • FIG. 10B provides a bar graph illustrating IL-10 mRNA levels from wild type CD4+ T cells (white bar) or CD83 mutant CD4+ T cells (dark bar) that had been stimulated with anti-CD3 and anti-CD28 antibodies. As illustrated, the IL-10 mRNA levels are higher in the CD83 mutant cells.
  • FIG. 11 provides a graph illustrating that various preparations of anti-CD83 antibodies inhibit IL-4 production in anti-CD3 and anti-CD28 antibody stimulated T cells. The amount of IL-4 produced by T cells in pg/ml is plotted versus the concentration of different anti-CD83 antibody preparations, including the 20B08 (♦) anti-CD83 preparation, the 20D04 (▪) anti-CD83 preparation, the 14C12 (▴) anti-CD83 preparation and the 11G05 (X) anti-CD83 antibody preparation.
  • FIG. 12 provides a graph illustrating that various preparations of anti-CD83 antibodies inhibit T cell proliferation. The graph plots the incorporation of radioactive thymidine in cpms, which was used as an indicator of the amount of T cell proliferation, versus the concentration of the different anti-CD83 antibody preparations, including the 20D04 (♦) anti-CD83 preparation, the 11G05 (▪) anti-CD83 antibody preparation, the 14C12 (▴) anti-CD83 preparation and the 6G05 anti-CD83 preparation (X).
  • FIG. 13 provides a graph illustrating that transgenic mice that over-express wild type CD83 have increased T cell proliferation. The graph plots the incorporation of radioactive thymidine in cpms, which was used as an indicator of the amount of T cell proliferation, versus the concentration of OVA peptide. The transgenic mice utilized had a T-cell receptor specific for chicken ovalbumin (OVA) 323-339 peptide that can activate T-cells. When mixed with either transgenic or wild type dendritic cells in the presence of OVA peptide, transgenic CD4+ T cells had increased T-cell proliferation. However, transgenic dendritic cells could not substantially increase wild type CD4+ T cell proliferation. Transgenic CD83 CD4+ T cells mixed with wild type dendritic cells (♦); transgenic CD83 CD4+ T cells mixed with transgenic dendritic cells (▪); wild type CD4+ T cells mixed with transgenic dendritic cells (▴); and wild type CD4+ T cells mixed with wild type dendritic cells (X).
  • FIG. 14 provides a schematic diagram of the structural elements included in the mouse CD83 protein used for generating antibodies.
  • FIG. 15 provides a graph of ELISA data illustrating the titer obtained for different isolates of polyclonal anti-CD83 anti-sera. The first (♦), second (▪) and third (▴) isolates had similar titers, though the titer of the second isolate (▪) was somewhat higher.
  • FIG. 16 illustrates that proliferation of PHA-activated human PBMCs was inhibited by antibodies raised against the external region of the mouse CD83 protein (♦). Pre-immune serum (▪) had little effect on the proliferation of human PBMCs.
  • FIG. 17A provides a sequence alignment of anti-CD83 heavy chain variable regions isolated by the invention. Sequences for isolates 20B08H (SEQ ID NO:52), 6G05H (SEQ ID NO:53), 20D04H (SEQ ID NO:54), 11G05 (SEQ ID NO:66) and 14C12 (SEQ ID NO:67) are provided. The CDR regions are highlighted in bold.
  • FIG. 17B provides a sequence alignment of anti-CD83 light chain variable regions isolated by the invention. Sequences for isolates 20B08H (SEQ ID NO:55), 6G05H (SEQ ID NO:56), 20D04H (SEQ ID NO:57), 11G05 (SEQ ID NO:68) and 14C12 (SEQ ID NO:69) are provided. The CDR regions are highlighted in bold.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The invention provides methods for modulating the immune system by using CD83 proteins, CD83 nucleic acids and factors that modulate CD83 activity or expression.
  • According to the invention, loss or reduction of CD83 activity in vivo results in altered cytokine levels, for example, lower interleukin-2 levels, increased interleukin-4 levels, increased GM-CSF levels and increased interleukin-10 levels. Loss or reduction of CD83 activity in vivo can also result in decreased numbers of T cells.
  • Moreover, the invention also relates to increased CD83 activity in vivo that can result in altered cytokine levels, for example, higher interleukin-2 levels, decreased interleukin-4 levels, decreased GM-CSF levels and decreased interleukin-10 levels. Increased CD83 expression or activity in vitro and in vivo can also result in increased activation and increased numbers of T cells.
  • The effects of CD83 on the immune system, on GM-CSF and on cytokine levels were analyzed by using mutant and transgenic mice. The mutant mouse has an altered CD83 gene that expresses altered (defective) CD83 gene product. The transgenic mouse overexpresses CD83 gene products. Accordingly, the invention provides mammals such as mice that have a mutant or wild type CD83 gene. These mice are useful for identifying the role that CD83 plays in the immune response. These mutant and transgenic animals are useful for identifying factors for manipulating cytokine levels and T cell activation by testing whether those factors and compositions can modulate, inhibit or replace the activity of CD83 in vivo.
  • CD83
  • CD83 is a lymphocyte and dendritic cell activation antigen that is expressed by activated lymphocytes and dendritic cells. CD83 is also a single-chain cell-surface glycoprotein with a molecular weight of about 45,000 that is believed to be a member of the Ig superfamily. The structure predicted from the CD83 amino acid sequence indicates that CD83 is a membrane glycoprotein with a single extracellular Ig-like domain, a transmembrane domain and cytoplasmic domain of approximately forty amino acids. The mature CD83 protein has about 186 amino acids and is composed of a single extracellular V type immunoglobulin (Ig)-like domain, a transmembrane domain and a thirty nine amino acid cytoplasmic domain. Northern blot analysis has revealed that CD83 is translated from three mRNA transcripts of about 1.7, 2.0 and 2.5 kb that are expressed by lymphoblastoid cell lines. It is likely that CD83 undergoes extensive post-translational processing because CD83 is expressed as a single chain molecule, but the determined molecular weight is twice the predicted size of the core protein. See U.S. Pat. No. 5,766,570.
  • An example of a human CD83 gene product that can be used in the invention is provided below (SEQ ID NO:9):
  •   1 MSRGLQLLLL SCAYSLAPAT PEVKVACSED VDLPCTAPWD
     41 PQVPYTVSWV KLLEGGEERM ETPQEDHLRG QHYHQKGQNG
     81 SFDAPNERPY SLKIRNTTSC NSGTYRCTLQ DPDGQRNLSG
    121 KVILRVTGCP AQRKEETFKK YRAEIVLLLA LVIFYLTLII
    161 FTCKFARLQS IFPDFSKAGM ERAFLPVTSP NKHLGLVTPH
    201 KTELV

    Such a CD83 gene product can be encoded by a number of different nucleic acids. One example of a human CD83 nucleic acid is provided below (SEQ ID NO:10).
  •    1 CCTGGCGCAG CCGCAGCAGC GACGCGAGCG AACTCGGCCG
      41 GGCCCGGGCG CGCGGGGGCG GGACGCGCAC GCGGCGAGGG
      81 CGGCGGGTGA GCCGGGGGCG GGGACGGGGG CGGGACGGGG
     121 GCGAAGGGGG CGGGGACGGG GGCGCCCGCC GGCCTAACGG
     161 GATTAGGAGG GCGCGCCACC CGCTTCCGCT GCCCGCCGGG
     201 GAATCCCCCG GGTGGCGCCC AGGGAAGTTC CCGAACGGGC
     241 GGGCATAAAA GGGCAGCCGC GCCGGCGCCC CACAGCTCTG
     281 CAGCTCGTGG CAGCGGCGCA GCGCTCCAGC CATGTCGCGC
     321 GGCCTCCAGC TTCTGCTCCT GAGCTGCGCC TACAGCCTGG
     361 CTCCCGCGAC GCCGGAGGTG AAGGTGGCTT GCTCCGAAGA
     401 TGTGGACTTG CCCTGCACCG CCCCCTGGGA TCCGCAGGTT
     441 CCCTACACGG TCTCCTGGGT CAAGTTATTG GAGGGTGGTG
     481 AAGAGAGGAT GGAGACACCC CAGGAAGACC ACCTCAGGGG
     521 ACAGCACTAT CATCAGAAGG GGCAAAATGG TTCTTTCGAC
     561 GCCCCCAATG AAAGGCCCTA TTCCCTGAAG ATCCGAAACA
     601 CTACCAGCTG CAACTCGGGG ACATACAGGT GCACTCTGCA
     641 GGACCCGGAT GGGCAGAGAA ACCTAAGTGG CAAGGTGATC
     681 TTGAGAGTGA CAGGATGCCC TGCACAGCGT AAAGAAGAGA
     721 CTTTTAAGAA ATACAGAGCG GAGATTGTCC TGCTGCTGGC
     761 TCTGGTTATT TTCTACTTAA CACTCATCAT TTTCACTTGT
     801 AAGTTTGCAC GGCTACAGAG TATCTTCCCA GATTTTTCTA
     841 AAGCTGGCAT GGAACGAGCT TTTCTCCCAG TTACCTCCCC
     881 AAATAAGCAT TTAGGGCTAG TGACTCCTCA CAAGACAGAA
     921 CTGGTATGAG CAGGATTTCT GCAGGTTCTT CTTCCTGAAG
     961 CTGAGGCTCA GGGGTGTGCC TGTCTGTTAC ACTGGAGGAG
    1001 AGAAGAATGA GCCTACGCTG AAGATGGCAT CCTGTGAAGT
    1041 CCTTCACCTC ACTGAAAACA TCTGGAAGGG GATCCCACCC
    1081 CATTTTCTGT GGGCAGGCCT CGAAAACCAT CACATGACCA
    1121 CATAGCATGA GGCCACTGCT GCTTCTCCAT GGCCACCTTT
    1161 TCAGCGATGT ATGCAGCTAT CTGGTCAACC TCCTGGACAT
    1201 TTTTTCAGTC ATATAAAAGC TATGGTGAGA TGCAGCTGGA
    1241 AAAGGGTCTT GGGAAATATG AATGCCCCCA GCTGGCCCGT
    1281 GACAGACTCC TGAGGACAGC TGTCCTCTTC TGCATCTTGG
    1321 GGACATCTCT TTGAATTTTC TGTGTTTTGC TGTACCAGCC
    1361 CAGATGTTTT ACGTCTGGGA GAAATTGACA GATCAAGCTG
    1401 TGAGACAGTG GGAAATATTT AGCAAATAAT TTCCTGGTGT
    1441 GAAGGTCCTG CTATTACTAA GGAGTAATCT GTGTACAAAG
    1481 AAATAACAAG TCGATGAACT ATTCCCCAGC AGGGTCTTTT
    1521 CATCTGGGAA AGACATCCAT AAAGAAGCAA TAAAGAAGAG
    1561 TGCCACATTT ATTTTTATAT CTATATGTAC TTGTCAAAGA
    1601 AGGTTTGTGT TTTTCTGCTT TTGAAATCTG TATCTGTAGT
    1641 GAGATAGCAT TGTGAACTGA CAGGCAGCCT GGACATAGAG
    1681 AGGGAGAAGA AGTCAGAGAG GGTGACAAGA TAGAGAGCTA
    1721 TTTAATGGCC GGCTGGAAAT GCTGGGCTGA CGGTGCAGTC
    1761 TGGGTGCTCG CCCACTTGTC CCACTATCTG GGTGCATGAT
    1801 CTTGAGCAAG TTCCTTCTGG TGTCTGCTTT CTCCATTGTA
    1841 AACCACAAGG CTGTTGCATG GGCTAATGAA GATCATATAC
    1881 GTGAAAATTA TTTGAAAACA TATAAAGCAC TATACAGATT
    1921 CGAAACTCCA TTGAGTCATT ATCCTTGCTA TGATGATGGT
    1961 GTTTTGGGGA TGAGAGGGTG CTATCCATTT CTCATGTTTT
    2001 CCATTGTTTG AAACAAAGAA GGTTACCAAG AAGCCTTTCC
    2041 TGTAGCCTTC TGTAGGAATT CTTTTGGGGA AGTGAGGAAG
    2081 CCAGGTCCAC GGTCTGTTCT TGAAGCAGTA GCCTAACACA
    2121 CTCCAAGATA TGGACACACG GGAGCCGCTG GCAGAAGGGA
    2161 CTTCACGAAG TGTTGCATGG ATGTTTTAGC CATTGTTGGC
    2201 TTTCCCTTAT CAAACTTGGG CCCTTCCCTT CTTGGTTTCC
    2241 AAAGGCATTT ATTGCTGAGT TATATGTTCA CTGTCCCCCT
    2281 AATATTAGGG AGTAAAACGG ATACCAAGTT GATTTAGTGT
    2321 TTTTACCTCT GTCTTGGCTT TCATGTTATT AAACGTATGC
    2361 ATGTGAAGAA GGGTGTTTTT CTGTTTTATA TTCAACTCAT
    2401 AAGACTTTGG GATAGGAAAA ATGAGTAATG GTTACTAGGC
    2441 TTAATACCTG GGTGATTACA TAATCTGTAC AACGAACCCC
    2481 CATGATGTAA GTTTACCTAT GTAACAAACC TGCACTTATA
    2521 CCCATGAACT TAAAATGAAA GTTAAAAATA AAAAACATAT
    2561 ACAAATAAAA AAAA
  • A sequence of a wild type mouse CD83 gene that can be used in the invention is provided herein as SEQ ID NO:1. SEQ ID NO:1 is provided below with the ATG start codon and the TGA stop codon identified by underlining.
  •    1 GCGCTCCAGC CGC ATG TCGC AAGGCCTCCA GCTCCTGTTT
      41 CTAGGCTGCG CCTGCAGCCT GGCACCCGCG ATGGCGATGC
      81 GGGAGGTGAC GGTGGCTTGC TCCGAGACCG CCGACTTGCC
     121 TTGCACAGCG CCCTGGGACC CGCAGCTCTC CTATGCAGTG
     161 TCCTGGGCCA AGGTCTCCGA GAGTGGCACT GAGAGTGTGG
     201 AGCTCCCGGA GAGCAAGCAA AACAGCTCCT TCGAGGCCCC
     241 CAGGAGAAGG GCCTATTCCC TGACGATCCA AAACACTACC
     281 ATCTGCAGCT CGGGCACCTA CAGGTGTGCC CTGCAGGAGC
     321 TCGGAGGGCA GCGCAACTTG AGCGGCACCG TGGTTCTGAA
     361 GGTGACAGGA TGCCCCAAGG AAGCTACAGA GTCAACTTTC
     401 AGGAAGTACA GGGCAGAAGC TGTGTTGCTC TTCTCTCTGG
     441 TTGTTTTCTA CCTGACACTC ATCATTTTCA CCTGCAAATT
     481 TGCACGACTA CAAAGCATTT TCCCAGATAT TTCTAAACCT
     521 GGTACGGAAC AAGCTTTTCT TCCAGTCACC TCCCCAAGCA
     561 AACATTTGGG GCCAGTGACC CTTCCTAAGA CAGAAACGGT
     601 A TGA GTAGGA TCTCCACTGG TTTTTACAAA GCCAAGGGCA
     641 CATCAGATCA GTGTGCCTGA ATGCCACCCG GACAAGAGAA
     681 GAATGAGCTC CATCCTCAGA TGGCAACCTT TCTTTGAAGT
     721 CCTTCACCTG ACAGTGGGCT CCACACTACT CCCTGACACA
     761 GGGTCTTGAG CACCATCATA TGATCACGAA GCATGGAGTA
     801 TCACCGCTTC TCTGTGGCTG TCAGCTTAAT GTTTCATGTG
     841 GCTATCTGGT CAACCTCGTG AGTGCTTTTC AGTCATCTAC
     881 AAGCTATGGT GAGATGCAGG TGAAGCAGGG TCATGGGAAA
     921 TTTGAACACT CTGAGCTGGC CCTGTGACAG ACTCCTGAGG
     961 ACAGCTGTCC TCTCCTACAT CTGGGATACA TCTCTTTGAA
    1001 TTTGTCCTGT TTCGTTGCAC CAGCCCAGAT GTCTCACATC
    1041 TGGCGGAAAT TGACAGGCCA AGCTGTGAGC CAGTGGGAAA
    1081 TATTTAGCAA ATAATTTCCC AGTGCGAAGG TCCTGCTATT
    1121 AGTAAGGAGT ATTATGTGTA CATAGAAATG AGAGGTCAGT
    1161 GAACTATTCC CCAGCAGGGC CTTTTCATCT GGAAAAGACA
    1201 TCCACAAAAG CAGCAATACA GAGGGATGCC ACATTTATTT
    1241 TTTTAATCTT CATGTACTTG TCAAAGAAGA ATTTTTCATG
    1281 TTTTTTCAAA GAAGTGTGTT TCTTTCCTTT TTTAAAATAT
    1321 GAAGGTCTAG TTACATAGCA TTGCTAGCTG ACAAGCAGCC
    1361 TGAGAGAAGA TGGAGAATGT TCCTCAAAAT AGGGACAGCA
    1401 AGCTAGAAGC ACTGTACAGT GCCCTGCTGG GAAGGGCAGA
    1441 CAATGGACTG AGAAACCAGA AGTCTGGCCA CAAGATTGTC
    1481 TGTATGATTC TGGACGAGTC ACTTGTGGTT TTCACTCTCT
    1521 GGTTAGTAAA CCAGATAGTT TAGTCTGGGT TGAATACAAT
    1561 GGATGTGAAG TTGCTTGGGG AAAGCTGAAT GTAGTGAATA
    1601 CATTGGCAAC TCTACTGGGC TGTTACCTTG TTGATATCCT
    1641 AGAGTTCTGG AGCTGAGCGA ATGCCTGTCA TATCTCAGCT
    1681 TGCCCATCAA TCCAAACACA GGAGGCTACA AAAAGGACAT
    1721 GAGCATGGTC TTCTGTGTGA ACTCCTCCTG AGAAACGTGG
    1761 AGACTGGCTC AGCGCTTTGC GCTTGAAGGA CTAATCACAA
    1801 GTTCTTGAAG ATATGGACCT AGGGGAGCTA TTGCGCCACG
    1841 ACAGGAGGAA GTTCTCAGAT GTTGCATTGA TGTAACATTG
    1881 TTGCATTTCT TTAATGAGCT GGGCTCCTTC CTCATTTGCT
    1921 TCCCAAAGAG ATTTTGTCCC ACTAATGGTG TGCCCATCAC
    1961 CCACACTATG AAAGTAAAAG GGATGCTGAG CAGATACAGC
    2001 GTGCTTACCT CTCAGCCATG ACTTTCATGC TATTAAAAGA
    2041 ATGCATGTGA A
  • Nucleic acids having SEQ ID NO:1 encode a mouse polypeptide having SEQ ID NO:2, provided below.
  •   1 MSQGLQLLFL GCACSLAPAM AMREVTVACS ETADLPCTAP
     41 WDPQLSYAVS WAKVSESGTE SVELPESKQN SSFEAPRRRA
     81 YSLTIQNTTI CSSGTYRCAL QELGGQRNLS GTVVLKVTGC
    121 PKEATESTFR KYRAEAVLLF SLVVFYLTLI IFTCKFARLQ
    161 SIFPDISKPG TEQAFLPVTS PSKHLGPVTL PKTETV
  • According to the invention, loss or reduction of CD83 activity in vivo results in altered cytokine levels, for example, lower interleukin-2 levels, increased interleukin-4 levels, increased GM-CSF levels and increased interleukin-10 levels. Loss or reduction of CD83 activity in vivo can also result in decreased numbers of T cells.
  • Moreover, increased CD83 activity in vivo can also result in altered cytokine levels, for example, higher interleukin-2 levels, decreased interleukin-4 levels, decreased GM-CSF levels and decreased interleukin-10 levels. Increased CD83 expression or activity in vivo can also result in increased activation or increased numbers of T cells.
  • The effect of CD83 on cytokine levels was ascertained through use of a mutant mouse that encodes a mutant CD83. Such a mutant mouse has a CD83 gene encoding SEQ ID NO:4, with added C-terminal sequences provided by SEQ ID NO:8. In contrast to these wild type CD83 nucleic acids and polypeptides, the mutant CD83 gene of the invention has SEQ ID NO:3. SEQ ID NO:3 is provided below with the ATG start codon, the mutation, and the TGA stop codon are identified by underlining.
  •    1 GCGCTCCAGC CGC ATG TCGC AAGGCCTCCA GCTCCTGTTT
      41 CTAGGCTGCG CCTGCAGCCT GGCACCCGCG ATGGCGATGC
      81 GGGAGGTGAC GGTGGCTTGC TCCGAGACCG CCGACTTGCC
     121 TTGCACAGCG CCCTGGGACC CGCAGCTCTC CTATGCAGTG
     161 TCCTGGGCCA AGGTCTCCGA GAGTGGCACT GAGAGTGTGG
     201 AGCTCCCGGA GAGCAAGCAA AACAGCTCCT TCGAGGCCCC
     241 CAGGAGAAGG GCCTATTCCC TGACGATCCA AAACACTACC
     281 ATCTGCAGCT CGGGCACCTA CAGGTGTGCC CTGCAGGAGC
     321 TCGGAGGGCA GCGCAACTTG AGCGGCACCG TGGTTCTGAA
     361 GGTGACAGGA TGCCCCAAGG AAGCTACAGA GTCAACTTTC
     401 AGGAAGTACA GGGCAGAAGC TGTGTTGCTC TTCTCTCTGG
     441 TTGTTTTCTA CCTGACACTC ATCATTTTCA CCTGCAAATT
     481 TGCACGACTA CAAAGCATTT TCCCAGATAT TTCTAAACCT
     521 GGTACGGAAC AAGCTTTTCT TCCAGTCACC TCCCCAAGCA
     561 AACATTTGGG GCCAGTGACC CTTCCTAAGA CAGAAACGGT
     601 A A GAGTAGGA TCTCCACTGG TTTTTACAAA GCCAAGGGCA
     641 CATCAGATCA GTGTGCCTGA ATGCCACCCG GACAAGAGAA
     681 GAATGAGCTC CATCCTCAGA TGGCAACCTT TCTTTGAAGT
     721 CCTTCACCTG ACAGTGGGCT CCACACTACT CCCTGACACA
     761 GGGTCT TGA G CACCATCATA TGATCACGAA GCATGGAGTA
     801 TCACCGCTTC TCTGTGGCTG TCAGCTTAAT GTTTCATGTG
     841 GCTATCTGGT CAACCTCGTG AGTGCTTTTC AGTCATCTAC
     881 AAGCTATGGT GAGATGCAGG TGAAGCAGGG TCATGGGAAA
     921 TTTGAACACT CTGAGCTGGC CCTGTGACAG ACTCCTGAGG
     961 ACAGCTGTCC TCTCCTACAT CTGGGATACA TCTCTTTGAA
    1001 TTTGTCCTGT TTCGTTGCAC CAGCCCAGAT GTCTCACATC
    1041 TGGCGGAAAT TGACAGGCCA AGCTGTGAGC CAGTGGGAAA
    1081 TATTTAGCAA ATAATTTCCC AGTGCGAAGG TCCTGCTATT
    1121 AGTAAGGAGT ATTATGTGTA CATAGAAATG AGAGGTCAGT
    1161 GAACTATTCC CCAGCAGGGC CTTTTCATCT GGAAAAGACA
    1201 TCCACAAAAG CAGCAATACA GAGGGATGCC ACATTTATTT
    1241 TTTTAATCTT CATGTACTTG TCAAAGAAGA ATTTTTCATG
    1281 TTTTTTCAAA GAAGTGTGTT TCTTTCCTTT TTTAAAATAT
    1321 GAAGGTCTAG TTACATAGCA TTGCTAGCTG ACAAGCAGCC
    1361 TGAGAGAAGA TGGAGAATGT TCCTCAAAAT AGGGACAGCA
    1401 AGCTAGAAGC ACTGTACAGT GCCCTGCTGG GAAGGGCAGA
    1441 CAATGGACTG AGAAACCAGA AGTCTGGCCA CAAGATTGTC
    1481 TGTATGATTC TGGACGAGTC ACTTGTGGTT TTCACTCTCT
    1521 GGTTAGTAAA CCAGATAGTT TAGTCTGGGT TGAATACAAT
    1561 GGATGTGAAG TTGCTTGGGG AAAGCTGAAT GTAGTGAATA
    1601 CATTGGCAAC TCTACTGGGC TGTTACCTTG TTGATATCCT
    1641 AGAGTTCTGG AGCTGAGCGA ATGCCTGTCA TATCTCAGCT
    1681 TGCCCATCAA TCCAAACACA GGAGGCTACA AAAAGGACAT
    1721 GAGCATGGTC TTCTGTGTGA ACTCCTCCTG AGAAACGTGG
    1761 AGACTGGCTC AGCGCTTTGC GCTTGAAGGA CTAATCACAA
    1801 GTTCTTGAAG ATATGGACCT AGGGGAGCTA TTGCGCCACG
    1841 ACAGGAGGAA GTTCTCAGAT GTTGCATTGA TGTAACATTG
    1881 TTGCATTTCT TTAATGAGCT GGGCTCCTTC CTCATTTGCT
    1921 TCCCAAAGAG ATTTTGTCCC ACTAATGGTG TGCCCATCAC
    1961 CCACACTATG AAAGTAAAAG GGATGCTGAG CAGATACAGC
    2001 GTGCTTACCT CTCAGCCATG ACTTTCATGC TATTAAAAGA
    2041 ATGCATGTGA A

    The change from a thymidine in SEQ ID NO:1 to an adenine in SEQ ID NO:3 at the indicated position (602) leads to read-through translation because the stop codon at positions 602-604 in SEQ ID NO:1 is changed to a codon that encodes an arginine. Accordingly, mutant CD83 nucleic acids having SEQ ID NO:3 encode an elongated polypeptide having SEQ ID NO:4, provided below, where the extra amino acids are underlined.
  •   1 MSQGLQLLFL GCACSLAPAM AMREVTVACS ETADLPCTAP
     41 WDPQLSYAVS WAKVSESGTE SVELPESKQN SSFEAPRRRA
     81 YSLTIQNTTI CSSGTYRCAL QELGGQRNLS GTVVLKVTGC
    121 PKEATESTFR KYRAEAVLLF SLVVFYLTLI IFTCKFARLQ
    161 SIFPDISKPG TEQAFLPVTS PSKHLGPVTL PKTETVRVGS
    201 PLVFTKPRAH QISVPECHPD KRRMSSILRW QPFFEVLHLT
    241 VGSTLLPDTG S
  • In another embodiment, the invention provides mutant CD83 nucleic acids that include SEQ ID NO:5.
  •   1 ATG TCGCAAG GCCTCCAGCT CCTGTTTCTA GGCTGCGCCT
     41 GCAGCCTGGC ACCCGCGATG GCGATGCGGG AGGTGACGGT
     81 GGCTTGCTCC GAGACCGCCG ACTTGCCTTG CACAGCGCCC
    121 TGGGACCCGC AGCTCTCCTA TGCAGTGTCC TGGGCCAAGG
    161 TCTCCGAGAG TGGCACTGAG AGTGTGGAGC TCCCGGAGAG
    201 CAAGCAAAAC AGCTCCTTCG AGGCCCCCAG GAGAAGGGCC
    241 TATTCCCTGA CGATCCAAAA CACTACCATC TGCAGCTCGG
    281 GCACCTACAG GTGTGCCCTG CAGGAGCTCG GAGGGCAGCG
    321 CAACTTGAGC GGCACCGTGG TTCTGAAGGT GACAGGATGC
    361 CCCAAGGAAG CTACAGAGTC AACTTTCAGG AAGTACAGGG
    401 CAGAAGCTGT GTTGCTCTTC TCTCTGGTTG TTTTCTACCT
    441 GACACTCATC ATTTTCACCT GCAAATTTGC ACGACTACAA
    481 AGCATTTTCC CAGATATTTC TAAACCTGGT ACGGAACAAG
    521 CTTTTCTTCC AGTCACCTCC CCAAGCAAAC ATTTGGGGCC
    561 AGTGACCCTT CCTAAGACAG AAACGGTAAG AGTAGGATCT
    601 CCACTGGTTT TTACAAAGCC AAGGGCACAT CAGATCAGTG
    641 TGCCTGAATG CCACCCGGAC AAGAGAAGAA TGAGCTCCAT
    681 CCTCAGATGG CAACCTTTCT TTGAAGTCCT TCACCTGACA
    721 GTGGGCTCCA CACTACTCCC TGACACAGGG TCT TGA

    Nucleic acids having SEQ ID NO:5 also encode a polypeptide having SEQ ID NO:4.
  • In another embodiment, the invention provides mutant CD83 nucleic acids that include SEQ ID NO:7.
  •   1 A GAGTAGGAT CTCCACTGGT TTTTACAAAG CCAAGGGCAC
     41 ATCAGATCAG TGTGCCTGAA TGCCACCCGG ACAAGAGAAG
     81 AATGAGCTCC ATCCTCAGAT GGCAACCTTT CTTTGAAGTC
    121 CTTCACCTGA CAGTGGGCTC CACACTACTC CCTGACACAG
    161 GGTCT TGA
  • The invention also provides a mutant CD83 containing SEQ ID NO:8, provided below.
  •  1 RVGSPLVFTK PRAHQISVPE CHPDKRRMSS ILRWQPFFEV
    41 LHLTVGSTLL PDTGS

    SEQ ID NO:8 contains read through sequences that are not present in the wild type CD83 polypeptide but are present in the mutant CD83 gene product provided by the invention.
  • CD83 Modulation of Cytokine Levels
  • The invention also provides compositions and methods for increasing interleukin-4 levels, increasing GM-CSF levels, increasing interleukin-10 levels and decreasing interleukin-2 levels in a mammal. Such compositions and methods generally operate by decreasing the expression or function of CD83 gene products in the mammal. Interleukin-4 promotes the differentiation of Th2 cells while decreasing the differentiation of precursor cells into Th1 cells. Th2 cells are involved in helping B lymphocytes and in stimulating production of IgG1 and IgE antibodies. Enhancement of Th2 formation may be useful, for example, in autoimmune diseases and in organ transplantation.
  • Alternatively, the invention provides compositions and methods for decreasing interleukin-4 levels, decreasing interleukin-10 levels and increasing interleukin-2 levels in a mammal. Such compositions and methods generally increase the expression or function of CD83 gene products in the mammal. Interleukin-2 promotes the differentiation of Th1 cells and decreases the differentiation of Th-2 cells. Th1 cells are, for example, involved in inducing autoimmune and delayed type hypersensitivity responses. Inhibition of Th2 formation may be useful in treating allergic diseases, malignancies and infectious diseases.
  • CD4+ T helper cells are not a homogeneous population but can be divided on the basis of cytokine secretion into at least two subsets termed T helper type 1 (Th1) and T helper type 2 (Th2) (see e.g., Mosmann, T. R. et al. (1986) J. Immunol. 136:2348-2357; Paul, W. E. and Seder, R. A. (1994) Cell 76:241-251; Seder, R. A. and Paul, W. E. (1994) Ann. Rev. Immunol. 12:635-673). Th1 cells secrete interleukin-2 (IL-2) and interferon-γ (IFN-γ) while Th2 cells produce interleukin-4 (IL4), interleukin-5 (IL-5), interleukin-10 (IL-10) and interleukin-13 (IL-13). Both subsets produce cytokines such as tumor necrosis factor (TNF) and granulocyte/macrophage-colony stimulating factor (GM-CSF).
  • In addition to their different pattern of cytokine expression, Th1 and Th2 cells are thought to have differing functional activities. For example, Th1 cells are involved in inducing delayed type hypersensitivity responses, whereas Th2 cells are involved in providing efficient “help” to B lymphocytes and stimulating production of IgG1 and IgE antibodies.
  • The ratio of Th1 to Th2 cells is highly relevant to the outcome of a wide array of immunologically-mediated clinical diseases including autoimmune, allergic and infectious diseases. For example, in experimental leishmania infections in mice, animals that are resistant to infection mount predominantly a Th1 response, whereas animals that are susceptible to progressive infection mount predominantly a Th2 response (Heinzel, F. P., et al. (1989) J. Exp. Med. 169:59-72; Locksley, R. M. and Scott, P. (1992) Immunoparasitology Today 1:A58-A6 1). In murine schistosomiasis, a Th1 to Th2 switch is observed coincident with the release of eggs into the tissues by female parasites and is associated with a worsening of the disease condition (Pearce, E. J., et al. (1991) J. Exp. Med. 173:159-166; Grzych, J-M., et al. (1991) J. Immunol. 141:1322-1327; Kullberg, M. C., et al. (1992) J. Immunol. 148:3264-3270).
  • Many human diseases, including chronic infections (such as with human immunodeficiency virus (HIV) and tuberculosis) and certain metastatic carcinomas, also are characterized by a Th1 to Th2 switch (see e.g., Shearer, G. M. and Clerici, M. (1992) Prog. Chem. Immunol. 54:21-43; Clerici, M. and Shearer, G. M. (1993) Immunology Today 14:107-111; Yamamura, M., et al. (1993) J. Clin. Invest. 91:1005-1010; Pisa, P., et al. (1992) Proc. Natl. Acad. Sci. USA 89:7708-7712; Fauci, A. S. (1988) Science 239:617-623).
  • Certain autoimmune diseases have been shown to be associated with a predominant Th1 response. For example, patients with rheumatoid arthritis have predominantly Th1 cells in synovial tissue (Simon, A. K., et al. (1994) Proc. Natl. Acad. Sci. USA 91:8562-8566) and experimental autoimmune encephalomyelitis (EAE) can be induced by autoreactive Th1 cells (Kuchroo, V. K., et al. (1993) J. Immunol. 151:4371-4381).
  • The ability to alter or manipulate ratios of Th1 and Th2 subsets requires an understanding of the mechanisms by which the differentiation of CD4 T helper precursor cells (Thp), which secrete only IL-2, choose to become Th1 or Th2 effector cells. It is clear that the cytokines themselves are potent Th cell inducers and form an autoregulatory loop (see e.g., Paul, W. E. and Seder, R. A. (1994) Cell 76:241-251; Seder, R. A. and Paul, W. E. (1994) Ann. Rev. Immunol. 12:635-673). Thus, IL4 promotes the differentiation of Th2 cells while preventing the differentiation of precursors into Th1 cells, while IL-12 and IFN-γ have the opposite effect.
  • According to the invention, one way to alter Th1:Th2 ratios is to increase or decrease the level of selected cytokines by using CD83. Direct administration of cytokines or antibodies to cytokines has been shown to have an effect on certain diseases mediated by either Th1 or Th2 cells. For example, administration of recombinant IL-4 or antibodies to IL-12 ameliorate EAE, a Th1-driven autoimmune disease (see Racke; M. K. et al. (1994) J. Exp. Med. 180:1961-1966; and Leonard, J. P. et al. (1995) J. Exp. Med. 181:381-386), while anti-IL-4 antibodies can ameliorate the Th2-mediated parasitic disease, Leishmania major (Sadick, M. D. et al. (1990) J. Exp. Med. 171:115-127).
  • Numerous disease conditions are associated with either a predominant Th1-type response or a predominant Th2-type response and the individuals suffering from such disease conditions could benefit from treatment with the CD83 related compositions and methods of the invention. Application of the immunomodulatory methods of the invention to such diseases is described in further detail below.
  • Allergies
  • Allergies are mediated through IgE antibodies whose production is regulated by the activity of Th2 cells and the cytokines produced thereby. In allergic reactions, IL-4 is produced by Th2 cells, which further stimulates production of IgE antibodies and activation of cells that mediate allergic reactions, i.e., mast cells and basophils. IL-4 also plays an important role in eosinophil mediated inflammatory reactions.
  • Accordingly, the stimulation of CD83 production by use of the compositions and methods of the invention can be used to inhibit the production of Th2-associated cytokines, for example IL-4, in allergic patients as a means to down-regulate production of pathogenic IgE antibodies. A stimulatory agent may be directly administered to the subject mammal. Alternatively, the CD83 stimulatory agent (e.g. CD83 expression cassette) can be administered to cells (e.g., Thp cells or Th2 cells) that may be obtained from the subject and those modified cells can be readministered to the subject mammal. Moreover, in certain situations it may be beneficial to co-administer the allergen together with the stimulatory agent either to the subject or to cells treated with the stimulatory agent. Such co-administration can inhibit (e.g., desensitize) the allergen-specific response. The treatment may be further enhanced by administering Th1-promoting agents, such as the cytokine IL-12 or antibodies to Th2-associated cytokines (e.g., anti-IL-4 antibodies), to the allergic subject in amounts sufficient to further stimulate a Th1-type response.
  • Cancer
  • The invention also relates to CD83-related methods for increasing interleukin-10 (IL-10) levels to reduce the spread of neoplastic diseases and/or prevent neoplastic diseases and the growth of a tumor. According to the invention, decreased CD83 activity can dramatically increase the levels of IL-10 in the body and such increased interleukin-10 can be used to treat neoplastic diseases. Hence, the invention provides a method for preventing or treating tumors in a mammal, which involves diminishing CD83 expression or activity in the mammal. In various embodiments, the tumor is IL-2-dependent, a plasmacytoma, or a leukemia, including a lymphocytic leukemia such as a B cell lymphocytic leukemia.
  • The invention also provides methods for increasing T cell activation or T cell proliferation by increasing CD83 activity or expression. Such methods can also be used to prevent or treat tumors in a mammal.
  • Infectious Diseases
  • The expression of Th2-promoting cytokines also has been reported to increase during a variety of infectious diseases. For example, HIV infection, tuberculosis, leishmaniasis, schistosomiasis, filarial nematode infection, intestinal nematode infection and other such infectious diseases are associated with a Th1 to Th2 shift in the immune response. See e.g., Shearer, G. M. and Clerici, M. (1992) Prog. Chem. Immunol. 54:2143; Clerici, M. and Shearer, G. M. (1993) Immunology Today 14:107-111; Fauci, A. S. (1988) Science 239:617-623; Locksley, R. M. and Scott, P. (1992) Immunoparasitology Today 1:A58-A61; Pearce, E. J., et al. (1991) J. Exp. Med. 173:159-166; Grzych, J-M., et al. (1991) J. Immunol. 141:1322-1327; Kullberg, M. C., et al. (1992) J. Immunol. 148:3264-3270; Bancroft, A. J., et al. (1993) J. Immunol. 150:1395-1402; Pearlman, E., et al. (11993) Infect. Immun. 61:1105-1112; Else, K. J., et al. (1994) J. Exp. Med. 179:347-351.
  • Accordingly, the stimulatory CD83-related compositions and methods of the invention can be used to inhibit the production of Th2-cells in subjects with infectious diseases to promote an ongoing Th1 response in the patients and to ameliorate the course of the infection. The treatment may be further enhanced by administering other Th1-promoting agents, such as the cytokine IL-12 or antibodies to Th2-associated cytokines (e.g., anti-IL-4 antibodies), to the recipient in amounts sufficient to further stimulate a Th 1-type response.
  • Hence, for example, infections of the following microbial organisms can be treated by the methods of the invention: Aeromonas spp., Bacillus spp., Bacteroides spp., Campylobacter spp., Clostridium spp., Enterobacter spp., Enterococcus spp., Escherichia spp., Gastrospirillum sp., Helicobacter spp., Klebsiella spp., Salmonella spp., Shigella spp., Staphylococcus spp., Pseudomonas spp., Vibrio spp., Yersinia spp., and the like. Infections that can be treated by the methods of the invention include those associated with staph infections (Staphylococcus aureus), typhus (Salmonella typhi), food poisoning (Escherichia coli, such as O157:H7), bascillary dysentery (Shigella dysenteria), pneumonia (Psuedomonas aerugenosa and/or Pseudomonas cepacia), cholera (Vivrio cholerae), ulcers (Helicobacter pylori) and others. E. coli serotype O157:H7 has been implicated in the pathogenesis of diarrhea, hemorrhagic colitis, hemolytic uremic syndrome (HUS) and thrombotic thrombocytopenic purpura (TTP). The methods of the invention are also active against drug-resistant and multiply-drug resistant strains of bacteria, for example, multiply-resistant strains of Staphylococcus aureus and vancomycin-resistant strains of Enterococcus faecium and Enterococcus faecalis.
  • The methods of the invention are also effective against viruses. The term “virus” refers to DNA and RNA viruses, viroids, and prions. Viruses include both enveloped and non-enveloped viruses, for example, hepatitis A virus, hepatitis B virus, hepatitis C virus, human immunodeficiency virus (HIV), poxviruses, herpes viruses, adenoviruses, papovaviruses, parvoviruses, reoviruses, orbiviruses, picornaviruses, rotaviruses, alphaviruses, rubivirues, influenza virus type A and B, flaviviruses, coronaviruses, paramyxoviruses, morbilliviruses, pneumoviruses, rhabdoviruses, lyssaviruses, orthmyxoviruses, bunyaviruses, phleboviruses, nairoviruses, hepadnaviruses, arenaviruses, retroviruses, enteroviruses, rhinoviruses and the filovirus.
  • Autoimmune Diseases
  • The CD83-related compositions and methods of the invention can be used in the treatment of autoimmune diseases that are associated with a Th2-type dysfunction. Many autoimmune disorders are the result of inappropriate activation of T cells that are reactive against “self tissues” and that promote the production of cytokines and autoantibodies involved in the pathology of the diseases. Modulation of T helper-type responses can have an effect on the course of the autoimmune disease. For example, in experimental allergic encephalomyelitis, stimulation of a Th2-type response by administration of IL-4 at the time of the induction of the disease diminishes the intensity of the autoimmune disease (Paul, W. E., et al. (1994) Cell 76:241-251). Furthermore, recovery of the animals from the disease has been shown to be associated with an increase in a Th2-type response as evidenced by an increase of Th2-specific cytokines (Koury, S. J., et al. (1992) J. Exp. Med. 176:1355-1364). Moreover, T cells that can suppress EAE secrete Th2-specific cytokines (Chen, C., et al. (1994) Immunity 1:147-154). Since stimulation of a Th2-type response in experimental allergic encephalomyelitis has a protective effect against the disease, stimulation of a Th2 response in subjects with multiple sclerosis (for which EAE is a model) is likely to be beneficial therapeutically.
  • Similarly, stimulation of a Th2-type response in type I diabetes in mice provides a protective effect against the disease. Indeed, treatment of NOD mice with IL-4 (which promotes a Th2 response) prevents or delays onset of type I diabetes that normally develops in these mice (Rapoport, M. J., et al. (1993) J. Exp. Med. 178:87-99). Thus, inhibition of CD83 production can stimulate IL-4 production and/or a Th2 response in a subject suffering from or susceptible to diabetes may ameliorate the effects of the disease or inhibit the onset of the disease.
  • Yet another autoimmune disease in which stimulation of a Th2-type response may be beneficial is rheumatoid arthritis (RA). Studies have shown that patients with rheumatoid arthritis have predominantly Th1 cells in synovial tissue (Simon, A. K., et al., (1994) Proc. Natl. Acad. Sci. USA 91:8562-8566). By stimulating a Th2 response in a subject with rheumatoid arthritis, the detrimental Th1 response can be concomitantly down-modulated to thereby ameliorate the effects of the disease.
  • Accordingly, the CD83-related compositions and methods of the invention can be used to stimulate production of Th2-associated cytokines in subjects suffering from, or susceptible to, an autoimmune disease in which a Th2-type response is beneficial to the course of the disease. Such compositions and methods would modulate CD83 activity. In some embodiments, the compositions would decrease CD83 activity and thereby increase the level of certain cytokines, for example, IL-4 levels are increased when CD83 activity is diminished. The treatment may be further enhanced by administering other Th2-promoting agents, such as IL-4 itself or antibodies to Th1-associated cytokines, to the subject in amounts sufficient to further stimulate a Th2-type response. The treatment may be further enhanced by administering a Th1-promoting cytokine (e.g., IFN-γ) to the subject in amounts sufficient to further stimulate a Th1-type response.
  • The efficacy of CD83-related for treating autoimmune diseases can be tested in the animal models provided herein or other models of human diseases (e.g., EAE as a model of multiple sclerosis and the NOD mice as a model for diabetes). Such animal models include the mrl/lpr/lpr mouse as a model for lupus erythematosus, murine collagen-induced arthritis as a model for rheumatoid arthritis, and murine experimental myasthenia gravis (see Paul ed., Fundamental Immunology, Raven Press, New York, 1989, pp. 840-856). A CD83-modulatory (i.e., stimulatory or inhibitory) agent of the invention is administered to test animals and the course of the disease in the test animals is then monitored by the standard methods for the particular model being used. Effectiveness of the modulatory agent is evidenced by amelioration of the disease condition in animals treated with the agent as compared to untreated animals (or animals treated with a control agent).
  • Non-limiting examples of autoimmune diseases and disorders having an autoimmune component that may be treated according to the invention include diabetes mellitus, arthritis (including rheumatoid arthritis, juvenile rheumatoid arthritis, osteoarthritis, psoriatic arthritis), multiple sclerosis, myasthenia gravis, systemic lupus erythematosis, autoimmune thyroiditis, dermatitis (including atopic dermatitis and eczematous dermatitis), psoriasis, Sjogren's Syndrome, including keratoconjunctivitis sicca secondary to Sjogren's Syndrome, alopecia areata, allergic responses due to arthropod bite reactions, Crohn's disease, aphthous ulcer, iritis, conjunctivitis, keratoconjunctivitis, ulcerative colitis, asthma, allergic asthma, cutaneous lupus erythematosus, scleroderma, vaginitis, proctitis, drug eruptions, leprosy reversal reactions, erythema nodosum leprosum, autoimmune uveitis, allergic encephalomyelitis, acute necrotizing hemorrhagic encephalopathy, idiopathic bilateral progressive sensorineural hearing loss, aplastic anemia, pure red cell anemia, idiopathic thrombocytopenia, polychondritis, Wegener's granulomatosis, chronic active hepatitis, Stevens-Johnson syndrome, idiopathic sprue, lichen planus, Crohn's disease, Graves opthalmopathy, sarcoidosis, primary biliary cirrhosis, uveitis posterior, and interstitial lung fibrosis.
  • Transplantation
  • While graft rejection or graft acceptance may not be attributable exclusively to the action of a particular T cell subset (i.e., Th1 or Th2 cells) in the graft recipient, studies have implicated a predominant Th2 response in prolonged graft survival and a predominant Th1 response in graft rejection (for a discussion see Dallman, M. J. (1995) Curr. Opin. Immunol. 7:632-638; Takeuchi, T. et al. (1992) Transplantation 53:1281-1291; Tzakis, A. G. et al. (1994) J. Pediatr. Surg. 29:754-756; That, N. L. et al. (1995) Transplantation 59:274-281. Additionally, adoptive transfer of cells having a Th2 cytokine phenotype prolongs skin graft survival (Maeda, H. et al. (1994) Int. Immunol. 6:855-862) and reduces graft-versus-host disease (Fowler, D. H. et al. (1994) Blood 84:3540-3549; Fowler, D. H. et al. (1994) Prog. Clin. Biol. Res. 389:533-540). Furthermore, administration of IL-4, which promotes Th2 differentiation, prolongs cardiac allograft survival (Levy, A. E. and Alexander, J. W. (1995) Transplantation 60:405-406), whereas administration of IL-12 in combination with anti-IL-10 antibodies, which promotes Th1 differentiation, enhances skin allograft rejection (Gorczynski, R. M. et al. (1995) Transplantation 60:1337-1341).
  • As provided herein, loss of CD83 function increases interleukin-4 production, which in turn promotes the differentiation of Th2 cells and depresses the differentiation of precursor cells into Th1 cells. Accordingly, methods of the invention that involve decreasing CD83 function can be used to stimulate production of Th2-associated cytokines in transplant recipients to prolong survival of the graft. These methods can be used both in solid organ transplantation and in bone marrow transplantation (e.g., to inhibit graft-versus-host disease). These methods can involve either direct administration of a CD83 inhibitory agent to the transplant recipient or ex vivo treatment of cells obtained from the subject (e.g., Thp, Th1 cells, B cells, non-lymphoid cells) with an inhibitory agent followed by readministration of the cells to the subject. The treatment may be further enhanced by administering other Th2-promoting agents, such as IL-4 itself or antibodies to Th1-associated cytokines, to the recipient in amounts sufficient to further stimulate a Th2-type response.
  • Additional Methods of Using CD83
  • In addition to the foregoing disease situations, the modulatory methods of the invention also are useful for other purposes.
  • For example, inhibition of CD83 activity or function gives rise to increased granulocyte macrophage-colony stimulating factor (GM-CSF). Granulocyte macrophage colony stimulating factor is a hematopoietic growth factor that promotes the proliferation and differentiation of hematopoietic progenitor cells. GM-CSF is approved for treatment of patients requiring increased proliferation of white blood cells. Data indicates that GM-CSP is also useful as a vaccine adjuvant Morrissey, et al., J. Immunology 139, 1113-1119 (1987). GM-CSF can also be used to treat patients prone to infection such as those undergoing high risk bowel surgery, trauma victims and individuals with HIV.
  • Accordingly, the invention provides a method of increasing the levels of GM-CSF in a mammal or in a mammalian cell by administering an agent that modulates or inhibits CD83 activity or expression.
  • The invention also provides a method of decreasing the levels of GM-CSF in a mammal or in a mammalian cell by administering an agent that modulates or stimulates CD83 activity or expression.
  • Moreover, in other embodiments the CD83 inhibitory methods of the invention can be used to stimulate production of IL-4 or IL-10 in vitro for commercial production of these cytokines. For example, CD4+ T cells with a null or other mutation in the CD83 gene can be cultured and then stimulated to produce cytokines, for example, by use of anti-CD3 and/or anti-CD28 antibodies to activate the mutant CD4+ T cells. Significant amounts of IL-4 and IL-10 can then be isolated from the culture media. Alternatively, CD4+ T cells can be contacted with the CD83 inhibitory agent in vitro to stimulate IL-4 or IL-10 production and the IL-4 or IL-10 can be recovered from the culture supernatant. The isolated IL-4 and/or IL-10 can be further purified if necessary, and packaged for commercial use.
  • The methods of the invention can be adapted to vaccinations to promote either a Th1 or a Th2 response to an antigen of interest in a subject. That is, CD83 or CD83 modulators of the invention can serve as adjuvants to direct an immune response to a vaccine either to a Th1 response or a Th2 response. For example, to stimulate an antibody response to an antigen of interest (i.e., for vaccination purposes), the antigen and a CD83 inhibitory agent of the invention can be coadministered to a subject to promote a Th2 response to the antigen in the subject, since Th2 responses provide efficient B cell help and promote IgG1 production.
  • Alternatively, to promote a cellular immune response to an antigen of interest, the antigen and a CD83 stimulating agent of the invention can be coadministered to a subject to promote a Th1 response to the antigen in a subject, since Th1 responses favor the development of cell-mediated immune responses (e.g., delayed hypersensitivity responses).
  • The antigen of interest and the modulatory agent can be formulated together into a single pharmaceutical composition or in separate compositions.
  • Thus, in some embodiments, the antigen of interest and the modulatory agent are administered simultaneously to the subject. Alternatively, in certain situations it may be desirable to administer the antigen first and then the modulatory agent or vice versa. For example, in the case of an antigen that naturally evokes a Th1 response, it may be beneficial to first administer the antigen alone to stimulate a Th1 response and then administer a CD83 inhibitory agent, alone or together with a boost of antigen, to shift the immune response to a Th2 response.
  • According to the invention, any agent that can modulate CD83 to increase or decrease cytokine levels, increase or decrease T cell levels or produce any other CD83-related response can be used in the compositions and methods of the invention. In some embodiments, anti-CD83 antibodies of the invention are used to either activate or inhibit CD83 activity. Activation or inhibition by such antibodies can depend on the epitope to which the antibody binds. Hence, antibodies may play a role in boosting or depressing CD83 activity. These CD83 modulatory agents, including anti-CD83 antibodies, are described in more detail below.
  • Stimulating or Inhibiting CD83
  • According to the invention, any agent that can stimulate CD83 to perform its natural functions can be used in the compositions and methods of the invention as a CD83 stimulatory agent. Indicators that CD83 activity is stimulated include increased IL-2 cytokine levels, increased T cell levels, and increased TNF levels relative to unstimulated levels in wild type CD83 cells. Examples of CD83 stimulatory agents include, for example, the CD83 gene product itself, certain anti-CD83 antibodies, CD83-encoding nucleic acids (DNA or RNA), factors that promote CD83 transcription or translation, organic molecules, peptides and the like.
  • Also, according to the invention, any agent that can inhibit CD83 from performing its natural functions can be used in the compositions and methods of the invention as a CD83 inhibitory agent. Indicators that CD83 activity is inhibited include increased IL-4 cytokine levels, increased IL-10 levels, decreased IL-2 production, decreased T cell levels, and decreased TNF levels relative to uninhibited levels in wild type CD83 cells.
  • Examples of CD83 inhibitors include anti-CD83 antibodies, CD83 anti-sense nucleic acids (e.g. nucleic acids that can hybridize to CD83 nucleic acids), organic compounds, peptides and agents that can mutate an endogenous CD83 gene. In some embodiments, the CD83 stimulatory or inhibitory agents are proteins, for example, CD83 gene products, anti-CD83 antibody preparations, CD83 inhibitors, peptides and protein factors that can promote CD83 transcription or translation. In other embodiments, the CD83 stimulatory or inhibitory agents are peptides or organic molecules. Such proteins, organic molecules and organic molecules can be prepared and/or purified as described herein or by methods available in the art, and administered as provided herein.
  • In other embodiments, the CD83 stimulatory or inhibitory agents can be nucleic acids including recombinant expression vectors or expression cassettes encoding CD83 gene products, CD83 transcription factors, CD83 anti-sense nucleic acid, intracellular antibodies capable of binding to CD83 or dominant negative CD83 inhibitors. Such nucleic acids can be operably linked to a promoter that is functional in a mammalian cell, and then introduced into cells of the subject mammal using methods known in the art for introducing nucleic acid (e.g., DNA) into cells.
  • The “promoter functional in a mammalian cell” or “mammalian promoter” is capable of directing transcription of a polypeptide coding sequence operably linked to the promoter. The promoter should generally be active in T cells and antigen presenting cells and may be obtained from a gene that is expressed in T cells or antigen presenting cells. However, it need not be a T cell-specific or an antigen presenting cell specific-promoter. Instead, the promoter may be selected from any mammalian or viral promoter that can function in a T cell. Hence the promoter may be an actin promoter, an immunoglobulin promoter, a heat-shock promoter, or a viral promoter obtained from the genome of viruses such as adenoviruses, retroviruses, lentiviruses, herpes viruses, including but not limited to, polyoma virus, fowlpox virus, adenovirus 2, bovine papilloma virus, avian sarcoma virus, cytomegalovirus (CMV), hepatitis-B virus, Simian Virus 40 (SV40), Epstein Barr virus (EBV), feline immunodeficiency virus (FIV), and Sr.alpha., or are respiratory synsitial viral promoters (RSV) or long terminal repeats (LTRs) of a retrovirus, i.e., a Moloney Murine Leukemia Virus (MoMuLv) (Cepko et al. (1984) Cell 37:1053-1062). The promoter functional in a mammalian cell can be inducible or constitutive.
  • Any cloning procedure used by one of skill in the art can be employed to make the expression vectors or expression that comprise a promoter operably linked to a CD83 nucleic acid, CD83 transcription factor or a nucleic acid encoding an anti-CD83 antibody. See, e.g., Sambrook et al., Molecular Cloning, A Laboratory Manual, Cold Spring Harbor Laboratory, N.Y., 1989; Sambrook et al., Molecular Cloning, A Laboratory Manual, Cold Spring Harbor Laboratory, N.Y., 2001.
  • After constructing an expression vector or an expression cassette encoding CD83 gene products, CD83 transcription factors, CD83 anti-sense nucleic acid, intracellular antibodies capable of binding to CD83 or dominant negative CD83 inhibitors, mammalian cells can be transformed with the vector or cassette. Examples of such methods include:
  • Direct Injection: Naked DNA can be introduced into cells in vivo by directly injecting the DNA into the cells (see e.g., Acsadi et al. (1991) Nature 332:815-818; Wolff et al. (1990) Science 247:1465-1468). For example, a delivery apparatus (e.g., a “gene gun”) for injecting DNA into cells in vivo can be used. Such an apparatus is commercially available (e.g., from BioRad).
  • Receptor-Mediated DNA Uptake: Naked DNA can also be introduced into cells in vivo by complexing the DNA to a cation, such as polylysine, which is coupled to a ligand for a cell-surface receptor (see for example Wu, G. and Wu, C. H. (1988) J. Biol. Chem. 263:14621; Wilson et al. (1992) J. Biol. Chem. 267:963-967; and U.S. Pat. No. 5,166,320). Binding of the DNA-ligand complex to the receptor facilitates uptake of the DNA by receptor-mediated endocytosis. A DNA-ligand complex linked to adenovirus capsids that naturally disrupt endosomes, thereby releasing material into the cytoplasm can be used to avoid degradation of the complex by intracellular lysosomes (see for example Curiel et al. (1991) Proc. Natl. Acad. Sci. USA 88:8850; Cristiano et al. (1993) Proc. Natl. Acad. Sci. USA 90:2122-2126).
  • Retroviruses: Defective retroviruses are well characterized for use in gene transfer for gene therapy purposes (for a review see Miller, A. D. (1990) Blood 76:271). A recombinant retrovirus can be constructed having nucleotide sequences of interest incorporated into the retroviral genome. Additionally, portions of the retroviral genome can be removed to render the retrovirus replication defective. The replication defective retrovirus is then packaged into virions that can be used to infect a target cell through the use of a helper virus by standard techniques. Protocols for producing recombinant retroviruses and for infecting cells in vitro or in vivo with such viruses can be found in Current Protocols in Molecular Biology, Ausubel, F. M. et al. (eds.) Greene Publishing Associates, (1989), Sections 9.10-9.14 and other standard laboratory manuals. Examples of suitable retroviruses include pLJ, pZIP, pWE and pEM which are available to those skilled in the art. Examples of suitable packaging virus lines include ΨCrip, ΨCre, Ψ2 and ΨAm. Retroviruses have been used to introduce a variety of genes into many different cell types, including epithelial cells, endothelial cells, lymphocytes, myoblasts, hepatocytes, bone marrow cells, in vitro and/or in vivo (see for example Eglitis, et al. (1985) Science 230:1395-1398; Danos and Mulligan (1988) Proc. Natl. Acad. Sci. USA 85:6460-6464; Wilson et al. (1988) Proc. Natl. Acad. Sci. USA 85:3014-3018; Armentano et al. (1990) Proc. Natl. Acad. Sci. USA 87:6141-6145; Huber et al. (1991) Proc. Natl. Acad. Sci. USA 88:8039-8043; Ferry et al. (1991) Proc. Natl. Acad. Sci. USA 88:8377-8381; Chowdhury et al. (1991) Science 254:1802-1805; van Beusechem et al. (1992) Proc. Natl. Acad. Sci. USA 89:7640-7644; Kay et al. (1992) Human Gene Therapy 3:641-647; Dai et al. (1992) Proc. Natl. Acad. Sci. USA 89:10892-10895; Hwu et al. (1993) J. Immunol. 150:4104-4115; U.S. Pat. Nos. 4,868,116; 4,980,286; PCT Application WO 89/07136; PCT Application WO 89/02468; PCT Application WO 89/05345; and PCT Application WO 92/07573). Retroviral vectors require target cell division in order for the retroviral genome (and foreign nucleic acid inserted into it) to be integrated into the host genome to stably introduce nucleic acid into the cell. Thus, it may be necessary to stimulate replication of the target cell.
  • Adenoviruses: The genome of an adenovirus can be manipulated such that it encodes and expresses a gene product of interest but is inactivated in terms of its ability to replicate in a normal lytic viral life cycle. See, for example, Berkner et al. (1988) BioTechniques 6:616; Rosenfeld et al. (1991) Science 252:431-434; and Rosenfeld et al. (1992) Cell 68:143-155. Suitable adenoviral vectors derived from the adenovirus strain Ad type 5 d1324 or other strains of adenovirus (e.g., Ad2, Ad3, Ad7 etc.) are available to those skilled in the art. Recombinant adenoviruses are advantageous in that they do not require dividing cells to be effective gene delivery vehicles and can be used to infect a wide variety of cell types, including airway epithelium (Rosenfeld et al. (1992) cited supra), endothelial cells (Lemarchand et al. (1992) Proc. Natl. Acad. Sci. USA 89:6482-6486), hepatocytes (Herz and Gerard (1993) Proc. Natl. Acad. Sci. USA 90:2812-2816) and muscle cells (Quantin et al. (1992) Proc. Natl. Acad. Sci. USA 89:2581-2584). Additionally, introduced adenoviral DNA (and foreign DNA contained therein) is not integrated into the genome of a host cell but remains episomal, thereby avoiding potential problems that can occur as a result of insertional mutagenesis in situations where introduced DNA becomes integrated into the host genome (e.g., retroviral DNA). Moreover, the carrying capacity of the adenoviral genome for foreign DNA is large (up to 8 kilobases) relative to other gene delivery vectors (Berkner et al. cited supra; Haj-Ahmand and Graham (1986) J. Virol. 57:267). Most replication-defective adenoviral vectors currently in use are deleted for all or parts of the viral E1 and E3 genes but retain as much as 80% of the adenoviral genetic material.
  • Adeno-Associated Viruses: Adeno-associated virus (AAV) is a naturally occurring defective virus that requires another virus, such as an adenovirus or a herpes virus, as a helper virus for efficient replication and a productive life cycle. (For a review see Muzyczka et al. Curr. Topics in Micro. and Immunol. (1992) 158:97-129). It is also one of the few viruses that may integrate its DNA into non-dividing cells, and exhibits a high frequency of stable integration (see for example Flotte et al. (1992) Am. J. Respir. Cell. Mol. Biol. 7:349-356; Samulski et al. (1989) J. Virol. 63:3822-3828; and McLaughlin et al. (1989) J. Virol. 62:1963-1973). Vectors containing as little as 300 base pairs of AAV can be packaged and can integrate. Space for exogenous DNA is limited to about 4.5 kb. An AAV vector such as that described in Tratschin et al. (1985) Mol. Cell. Biol. 5:3251-3260 can be used to introduce DNA into cells. A variety of nucleic acids have been introduced into different cell types using AAV vectors (see for example Hermonat et al. (1984) Proc. Natl. Acad. Sci. USA 81:6466-6470; Tratschin et al. (1985) Mol. Cell. Biol. 4:2072-2081; Wondisford et al. (1988) Mol. Endocrinol. 2:32-39; Tratschin et al. (1984) J. Virol. 51:611-619; and Flotte et al. (1993) J. Biol. Chem. 268:3781-3790).
  • Transformed mammalian cells can then be identified and administered to the mammal from whence they came to permit expression of a CD83 gene product, CD83 transcription factor, CD83 anti-sense nucleic acid, intracellular antibody capable of binding to CD83 proteins, or dominant negative CD83 inhibitors. The efficacy of a particular expression vector system and method of introducing nucleic acid into a cell can be assessed by standard approaches routinely used in the art. For example, DNA introduced into a cell can be detected by a filter hybridization technique (e.g., Southern blotting). RNA produced by transcription of an introduced DNA can be detected, for example, by Northern blotting, RNase protection or reverse transcriptase-polymerase chain reaction (RT-PCR). The CD83 gene product can be detected by an appropriate assay, for example, by immunological detection of a produced CD83 protein, such as with a CD83-specific antibody.
  • CD83 Antibodies
  • The invention provides antibody preparations directed against the mutant and wild type CD83 polypeptides of the invention, for example, against a polypeptide having SEQ ID NO:2, SEQ ID NO:4, SEQ ID NO:7, SEQ ID NO:8 or SEQ ID NO:9. Other antibodies of interest can bind to the cytoplasmic tail of CD83.
  • In one embodiment, the invention provides antibodies that block the function of CD83 polypeptides. Such antibodies may be used as CD83 inhibitory agents in the methods of the invention as described herein. In another embodiment, the antibodies of the invention can activate CD83 activity. Such activating antibodies may be used as CD83 stimulatory agents.
  • All antibody molecules belong to a family of plasma proteins called immunoglobulins, whose basic building block, the immunoglobulin fold or domain, is used in various forms in many molecules of the immune system and other biological recognition systems. A typical immunoglobulin has four polypeptide chains, containing an antigen binding region known as a variable region and a non-varying region known as the constant region.
  • Native antibodies and immunoglobulins are usually heterotetrameric glycoproteins of about 150,000 daltons, composed of two identical light (L) chains and two identical heavy (H) chains. Each light chain is linked to a heavy chain by one covalent disulfide bond, while the number of disulfide linkages varies between the heavy chains of different immunoglobulin isotypes. Each heavy and light chain also has regularly spaced intrachain disulfide bridges. Each heavy chain has at one end a variable domain (VH) followed by a number of constant domains. Each light chain has a variable domain at one end (VL) and a constant domain at its other end. The constant domain of the light chain is aligned with the first constant domain of the heavy chain, and the light chain variable domain is aligned with the variable domain of the heavy chain. Particular amino acid residues are believed to form an interface between the light and heavy chain variable domains (Clothia et al., J. Mol. Biol. 186, 651-66, 1985); Novotny and Haber, Proc. Natl. Acad. Sci. USA 82, 4592-4596 (1985).
  • Depending on the amino acid sequences of the constant domain of their heavy chains, immunoglobulins can be assigned to different classes. There are at least five (5) major classes of immunoglobulins: IgA, IgD, IgE, IgG and several of these may be further divided into subclasses (isotypes), e.g. IgG-1, IgG-2, IgG-3 and IgG-4; IgA-1 and IgA-2. The heavy chains constant domains that correspond to the different classes of immunoglobulins are called alpha (α), delta (δ), epsilon (ε), gamma (γ) and mu (μ), respectively. The light chains of antibodies can be assigned to one of two clearly distinct types, called kappa (κ) and lambda (λ), based on the amino sequences of their constant domain. The subunit structures and three-dimensional configurations of different classes of immunoglobulins are well known.
  • The term “variable” in the context of variable domain of antibodies, refers to the fact that certain portions of the variable domains differ extensively in sequence among antibodies. The variable domains are for binding and determine the specificity of each particular antibody for its particular antigen. However, the variability is not evenly distributed through the variable domains of antibodies. It is concentrated in three segments called complementarity determining regions (CDRs) also known as hypervariable regions both in the light chain and the heavy chain variable domains.
  • The more highly conserved portions of variable domains are called the framework (FR). The variable domains of native heavy and light chains each comprise four FR regions, largely adopting a β-sheet configuration, connected by three CDRs, which form loops connecting, and in some cases forming part of, the β-sheet structure. The CDRs in each chain are held together in close proximity by the FR regions and, with the CDRs from the other chain, contribute to the formation of the antigen binding site of antibodies. The constant domains are not involved directly in binding an antibody to an antigen, but exhibit various effector function, such as participation of the antibody in antibody-dependent cellular toxicity.
  • An antibody that is contemplated for use in the present invention thus can be in any of a variety of forms, including a whole immunoglobulin, an antibody fragment such as Fv, Fab, and similar fragments, a single chain antibody that includes the variable domain complementarity determining regions (CDR), and the like forms, all of which fall under the broad term “antibody,” as used herein. The present invention contemplates the use of any specificity of an antibody, polyclonal or monoclonal, and is not limited to antibodies that recognize and immunoreact with a specific antigen. In preferred embodiments, in the context of both the therapeutic and screening methods described below, an antibody or fragment thereof is used that is immunospecific for an antigen or epitope of the invention.
  • The term “antibody fragment” refers to a portion of a full-length antibody, generally the antigen binding or variable region. Examples of antibody fragments include Fab, Fab′, F(ab′)2 and Fv fragments. Papain digestion of antibodies produces two identical antigen binding fragments, called the Fab fragment, each with a single antigen binding site, and a residual “Fc” fragment, so-called for its ability to crystallize readily. Pepsin treatment yields an F(ab′)2 fragment that has two antigen binding fragments, which are capable of cross-linking antigen, and a residual other fragment (which is termed pFc′). Additional fragments can include diabodies, linear antibodies, single-chain antibody molecules, and multispecific antibodies formed from antibody fragments. As used herein, “functional fragment” with respect to antibodies, refers to Fv, F(ab) and F(ab′)2 fragments.
  • Antibody fragments retain some ability to selectively bind with its antigen or receptor and are defined as follows:
  • (1) Fab is the fragment that contains a monovalent antigen-binding fragment of an antibody molecule. A Fab fragment can be produced by digestion of whole antibody with the enzyme papain to yield an intact light chain and a portion of one heavy chain.
  • (2) Fab′ is the fragment of an antibody molecule can be obtained by treating whole antibody with pepsin, followed by reduction, to yield an intact light chain and a portion of the heavy chain. Two Fab′ fragments are obtained per antibody molecule. Fab′ fragments differ from Fab fragments by the addition of a few residues at the carboxyl terminus of the heavy chain CH1 domain including one or more cysteines from the antibody hinge region.
  • (3) (Fab′)2 is the fragment of an antibody that can be obtained by treating whole antibody with the enzyme pepsin without subsequent reduction. F(ab′)2 is a dimer of two Fab′ fragments held together by two disulfide bonds.
  • (4) Fv is the minimum antibody fragment that contains a complete antigen recognition and binding site. This region consists of a dimer of one heavy and one light chain variable domain in a tight, non-covalent association (VH-VL dimer). It is in this configuration that the three CDRs of each variable domain interact to define an antigen binding site on the surface of the VH-VL dimer. Collectively, the six CDRs confer antigen binding specificity to the antibody. However, even a single variable domain (or half of an Fv comprising only three CDRs specific for an antigen) has the ability to recognize and bind antigen, although at a lower affinity than the entire binding site.
  • (5) Single chain antibody (“SCA”), defined as a genetically engineered molecule containing the variable region of the light chain, the variable region of the heavy chain, linked by a suitable polypeptide linker as a genetically fused single chain molecule. Such single chain antibodies are also referred to as “single-chain Fv” or “sFv” antibody fragments. Generally, the Fv polypeptide further comprises a polypeptide linker between the VH and VL domains that enables the sFv to form the desired structure for antigen binding. For a review of sFv see Pluckthun in The Pharmacology of Monoclonal Antibodies, vol. 113, Rosenburg and Moore eds. Springer-Verlag, N.Y., pp. 269-315 (1994).
  • The term “diabodies” refers to a small antibody fragments with two antigen-binding sites, which fragments comprise a heavy chain variable domain (VH) connected to a light chain variable domain (VL) in the same polypeptide chain (VH-VL). By using a linker that is too short to allow pairing between the two domains on the same chain, the domains are forced to pair with the complementary domains of another chain and create two antigen-binding sites. Diabodies are described more fully in, for example, EP 404,097; WO 93/11161, and Hollinger et al., Proc. Natl. Acad. Sci. USA 90: 6444-6448 (1993).
  • The preparation of polyclonal antibodies is well-known to those skilled in the art. See, for example, Green, et al., Production of Polyclonal Antisera, in: Immunochemical Protocols (Manson, ed.), pages 1-5 (Humana Press); Coligan, et al., Production of Polyclonal Antisera in Rabbits, Rats Mice and Hamsters, in: Current Protocols in Immunology, section 2.4.1 (1992), which are hereby incorporated by reference.
  • The preparation of monoclonal antibodies likewise is conventional. See, for example, Kohler & Milstein, Nature, 256:495 (1975); Coligan, et al., sections 2.5.1-2.6.7; and Harlow, et al., in: Antibodies: A Laboratory Manual, page 726 (Cold Spring Harbor Pub. (1988)), which are hereby incorporated by reference. Methods of in vitro and in vivo manipulation of monoclonal antibodies are also available to those skilled in the art. For example, the monoclonal antibodies to be used in accordance with the present invention may be made by the hybridoma method first described by Kohler and Milstein, Nature 256, 495 (1975), or may be made by recombinant methods, e.g., as described in U.S. Pat. No. 4,816,567. The monoclonal antibodies for use with the present invention may also be isolated from antibody libraries using the techniques described in Clackson et al. Nature 352: 624-628 (1991), as well as in Marks et al., J. Mol. Biol. 222: 581-597 (1991).
  • Monoclonal antibodies can be isolated and purified from hybridoma cultures by a variety of well-established techniques. Such isolation techniques include affinity chromatography with Protein-A Sepharose, size-exclusion chromatography, and ion-exchange chromatography. See, e.g., Coligan, et al., sections 2.7.1-2.7.12 and sections 2.9.1-2.9.3; Barnes, et al., Purification of Immunoglobulin G (IgG), in: Methods in Molecular Biology, Vol. 10, pages 79-104 (Humana Press (1992).
  • Another method for generating antibodies involves a Selected Lymphocyte Antibody Method (SLAM). The SLAM technology permits the generation, isolation and manipulation of monoclonal antibodies without the process of hybridoma generation. The methodology principally involves the growth of antibody forming cells, the physical selection of specifically selected antibody forming cells, the isolation of the genes encoding the antibody and the subsequent cloning and expression of those genes.
  • More specifically, an animal (rabbit, mouse, rat, other) is immunized with a source of specific antigen. This immunization may consist of purified protein, in either native or recombinant form, peptides, DNA encoding the protein of interest or cells expressing the protein of interest. After a suitable period, during which antibodies can be detected in the serum of the animal (usually weeks to months), blood (or other tissue) from the animal is harvested. Lymphocytes are isolated from the blood and cultured under specific conditions to generate antibody-forming cells, with antibody being secreted into the culture medium. These cells are detected by any of several means (complement mediated lysis of antigen-bearing cells, fluorescence detection or other) and then isolated using micromanipulation technology. The individual antibody forming cells are then processed for eventual single cell PCR to obtain the expressed Heavy and Light chain genes that encode the specific antibody. Once obtained and sequenced, these genes are cloned into an appropriate expression vector and recombinant, monoclonal antibody produced in a heterologous cell system. These antibodies are then purified via standard methodologies such as the use of protein A affinity columns. These types of methods are further described in Babcook, et al., Proc. Natl. Acad. Sci. (USA) 93: 7843-7848 (1996); U.S. Pat. No. 5,627,052; and PCT WO 92/02551 by Schrader.
  • Another method involves humanizing a monoclonal antibody by recombinant means to generate antibodies containing human specific and recognizable sequences. See, for review, Holmes, et al., J. Immunol., 158:2192-2201 (1997) and Vaswani, et al., Annals Allergy, Asthma & Immunol., 81:105-115 (1998). The term “monoclonal antibody” as used herein refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally occurring mutations that may be present in minor amounts. Monoclonal antibodies are highly specific, being directed against a single antigenic site. Furthermore, in contrast to conventional polyclonal antibody preparations that typically include different antibodies directed against different determinants (epitopes), each monoclonal antibody is directed against a single determinant on the antigen. In additional to their specificity, the monoclonal antibodies are advantageous in that they are synthesized by the hybridoma culture, uncontaminated by other immunoglobulins. The modifier “monoclonal” indicates the antibody is obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method.
  • The monoclonal antibodies herein specifically include “chimeric” antibodies (immunoglobulins) in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is identical with or homologous to corresponding sequences in antibodies derived from another species or belonging to another antibody class or subclass, as well as fragments of such antibodies, so long as they exhibit the desired biological activity (U.S. Pat. No. 4,816,567); Morrison et al. Proc. Natl. Acad. Sci. 81, 6851-6855 (1984).
  • Methods of making antibody fragments are also known in the art (see for example, Harlow and Lane, Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory, New York, (1988), incorporated herein by reference). Antibody fragments of the present invention can be prepared by proteolytic hydrolysis of the antibody or by expression in E. coli of DNA encoding the fragment. Antibody fragments can be obtained by pepsin or papain digestion of whole antibodies conventional methods. For example, antibody fragments can be produced by enzymatic cleavage of antibodies with pepsin to provide a 5S fragment denoted F(ab′)2. This fragment can be further cleaved using a thiol reducing agent, and optionally a blocking group for the sulfhydryl groups resulting from cleavage of disulfide linkages, to produce 3.5S Fab=monovalent fragments. Alternatively, an enzymatic cleavage using pepsin produces two monovalent Fab′ fragments and an Fc fragment directly. These methods are described, for example, in U.S. Pat. No. 4,036,945 and No. 4,331,647, and references contained therein. These patents are hereby incorporated in their entireties by reference.
  • Other methods of cleaving antibodies, such as separation of heavy chains to form monovalent light-heavy chain fragments, further cleavage of fragments, or other enzymatic, chemical, or genetic techniques may also be used, so long as the fragments bind to the antigen that is recognized by the intact antibody. For example, Fv fragments comprise an association of VH and VL chains. This association may be noncovalent or the variable chains can be linked by an intermolecular disulfide bond or cross-linked by chemicals such as glutaraldehyde. Preferably, the Fv fragments comprise VH and VL chains connected by a peptide linker. These single-chain antigen binding proteins (sFv) are prepared by constructing a structural gene comprising DNA sequences encoding the VH and VL domains connected by an oligonucleotide. The structural gene is inserted into an expression vector, which is subsequently introduced into a host cell such as E. coli. The recombinant host cells synthesize a single polypeptide chain with a linker peptide bridging the two V domains. Methods for producing sFvs are described, for example, by Whitlow, et al., Methods: a Companion to Methods in Enzymology, Vol. 2, page 97 (1991); Bird, et al., Science 242:423-426 (1988); Ladner, et al, U.S. Pat. No. 4,946,778; and Pack, et al., Bio/Technology 11:1271-77 (1993).
  • Another form of an antibody fragment is a peptide coding for a single complementarity-determining region (CDR). CDR peptides (“minimal recognition units”) can be obtained by constructing genes encoding the CDR of an antibody of interest. Such genes are prepared, for example, by using the polymerase chain reaction to synthesize the variable region from RNA of antibody-producing cells. See, for example, Larrick, et al., Methods: a Companion to Methods in Enzymology, Vol. 2, page 106 (1991).
  • The invention further contemplates human and humanized forms of non-human (e.g. murine) antibodies. Such humanized antibodies are chimeric immunoglobulins, immunoglobulin chains or fragments thereof (such as Fv, Fab, Fab′, F(ab′)2 or other antigen-binding subsequences of antibodies) that contain minimal sequence derived from non-human immunoglobulin. For the most part, humanized antibodies are human immunoglobulins (recipient antibody) in which residues from a complementary determining region (CDR) of the recipient are replaced by residues from a CDR of a nonhuman species (donor antibody) such as mouse, rat or rabbit having the desired specificity, affinity and capacity.
  • In some instances, Fv framework residues of the human immunoglobulin are replaced by corresponding non-human residues. Furthermore, humanized antibodies may comprise residues that are found neither in the recipient antibody nor in the imported CDR or framework sequences. These modifications are made to further refine and optimize antibody performance. In general, humanized antibodies can comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin and all or substantially all of the Fv regions are those of a human immunoglobulin consensus sequence. The humanized antibody optimally also will comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin. For further details, see: Jones et al., Nature 321, 522-525 (1986); Reichmann et al., Nature 332, 323-329 (1988); Presta, Curr. Op. Struct. Biol. 2, 593-596 (1992); Holmes, et al., J. Immunol., 158:2192-2201 (1997) and Vaswani, et al., Annals Allergy, Asthma & Immunol., 81:105-115 (1998).
  • The invention also provides methods of mutating antibodies to optimize their affinity, selectivity, binding strength or other desirable property. A mutant antibody refers to an amino acid sequence variant of an antibody. In general, one or more of the amino acid residues in the mutant antibody is different from what is present in the reference antibody. Such mutant antibodies necessarily have less than 100% sequence identity or similarity with the reference amino acid sequence. In general, mutant antibodies have at least 75% amino acid sequence identity or similarity with the amino acid sequence of either the heavy or light chain variable domain of the reference antibody. Preferably, mutant antibodies have at least 80%, more preferably at least 85%, even more preferably at least 90%, and most preferably at least 95% amino acid sequence identity or similarity with the amino acid sequence of either the heavy or light chain variable domain of the reference antibody.
  • The antibodies of the invention are isolated antibodies. An isolated antibody is one that has been identified and separated and/or recovered from a component of the environment in which it was produced. Contaminant components of its production environment are materials that would interfere with diagnostic or therapeutic uses for the antibody, and may include enzymes, hormones, and other proteinaceous or nonproteinaceous solutes. The term “isolated antibody” also includes antibodies within recombinant cells because at least one component of the antibody's natural environment will not be present. Ordinarily, however, isolated antibody will be prepared by at least one purification step.
  • If desired, the antibodies of the invention can be purified by any available procedure. For example, the antibodies can be affinity purified by binding an antibody preparation to a solid support to which the antigen used to raise the antibodies is bound. After washing off contaminants, the antibody can be eluted by known procedures. Those of skill in the art will know of various techniques common in the immunology arts for purification and/or concentration of polyclonal antibodies, as well as monoclonal antibodies (see for example, Coligan, et al., Unit 9, Current Protocols in Immunology, Wiley Interscience, 1991, incorporated by reference).
  • In preferred embodiments, the antibody will be purified as measurable by at least three different methods: 1) to greater than 95% by weight of antibody as determined by the Lowry method, and most preferably more than 99% by weight; 2) to a degree sufficient to obtain at least 15 residues of N-terminal or internal amino acid sequence by use of a spinning cup sequentator; or 3) to homogeneity by SDS-PAGE under reducing or non-reducing conditions using Coomasie blue or, preferably, silver stain.
  • The invention also provides antibodies that can bind to CD83 polypeptides. Sequences of complementarity determining regions (CDRs) or hypervariable regions from light and heavy chains of these anti-CD83 antibodies are provided. For example, a heavy chain variable region having a CDR1 sequence of SYDMT (SEQ ID NO:23), SYDMS (SEQ ID NO:24), DYDLS (SEQ ID NO:25) or SYDMS (SEQ ID NO:26) can be used in an antibody or other binding moiety to bind to CD83 gene products. In other embodiments, a heavy chain variable region having a CDR2 sequence of YASGSTYY (SEQ ID NO:27), SSSGTTYY (SEQ ID NO:28), YASGSTYY (SEQ ID NO:29), AIDGNPYY (SEQ ID NO:30) or STAYNSHY (SEQ ID NO:31) can be used in an antibody or other binding moiety to bind to CD83 gene products. In further embodiments of the invention, a heavy chain variable region having a CDR3 sequence of EHAGYSGDTGH (SEQ ID NO:32), EGAGVSMT (SEQ ID NO:33), EDAGFSNA (SEQ ID NO:34), GAGD (SEQ ID NO:35) or GGSWLD (SEQ ID NO:36) can be used in an antibody or other binding moiety to bind to CD83 gene products.
  • Moreover, a light chain variable region having a CDR1 sequence of RCAYD (SEQ ID NO:37), RCADVV (SEQ ID NO:38), or RCALV (SEQ ID NO:39) can be used in an antibody or other binding moiety to bind to CD83 gene products. In other embodiments, a light chain variable region having a CDR2 sequence of QSISTY (SEQ ID NO:40), QSVSSY (SEQ ID NO:41), ESISNY (SEQ ID NO:42), KNVYNNNW (SEQ ID NO:43), or QSVYDNDE (SEQ ID NO:43) can be used in an antibody or other binding moiety to bind to CD83 gene products. In further embodiments, a light chain variable region having a CDR3 sequence of QQGYTHSNVDNV (SEQ ID NO:44), QQGYSISDIDNA (SEQ ID NO:45), QCTSGGKFISDGAA (SEQ ID NO:46), AGDYSSSSDNG (SEQ ID NO:47), or QATHYSSDWLTY (SEQ ID NO:48) can be used in an antibody or other binding moiety to bind to CD83 gene products.
  • Light and heavy chains that can bind CD83 polypeptides are also provided by the invention. For example, in one embodiment, the invention provides a 20D04 light chain that can bind to CD83 polypeptides. The amino acid sequence for this 20D04 light chain is provided below (SEQ ID NO:11).
  •   1 MDMRAPTQLL GLLLLWLPGA RCADVVMTQT PASVSAAVGG
     41 TVTINCQASE SISNYLSWYQ QKPGQPPKLL IYRTSTLASG
     81 VSSRFKGSGS GTEYTLTISG VQCDDVATYY CQCTSGGKFI
    121 SDGAAFGGGT EVVVKGDPVA PTVLLFPPSS DEVATGTVTI
    161 VCVANKYFPD VTVTWEVDGT TQTTGIENSK TPQNSADCTY
    201 NLSSTLTLTS TQYNSHKEYT CKVTQGTTSV VQSFSRKNC
  • A nucleic acid sequence for this 20D04 anti-CD83 light chain is provided below (SEQ ID NO:12).
  •   1 ATGGACATGA GGGCCCCCAC TCAGCTGCTG GGGCTCCTGC
     41 TGCTCTGGCT CCCAGGTGCC AGATGTGCCG ATGTCGTGAT
     81 GACCCAGACT CCAGCCTCCG TGTCTGCAGC TGTGGGAGGC
    121 ACAGTCACCA TCAATTGCCA GGCCAGTGAA AGCATTAGCA
    161 ACTACTTATC CTGGTATCAG CAGAAACCAG GGCAGCCTCC
    201 CAAGCTCCTG ATCTACAGGA CATCCACTCT GGCATCTGGG
    241 GTCTCATCGC GGTTCAAAGG CAGTGGATCT GGGACAGAGT
    281 ACACTCTCAC CATCAGCGGC GTGCAGTGTG ACGATGTTGC
    321 CACTTACTAC TGTCAATGCA CTTCTGGTGG GAAGTTCATT
    361 AGTGATGGTG CTGCTTTCGG CGGAGGGACC GAGGTGGTGG
    401 TCAAAGGTGA TCCAGTTGCA CCTACTGTCC TCCTCTTCCC
    441 ACCATCTAGC GATGAGGTGG CAACTGGAAC AGTCACCATC
    481 GTGTGTGTGG CGAATAAATA CTTTCCCGAT GTCACCGTCA
    521 CCTGGGAGGT GGATGGCACC ACCCAAACAA CTGGCATCGA
    561 GAACAGTAAA ACACCGCAGA ATTCTGCAGA TTGTACCTAC
    601 AACCTCAGCA GCACTCTGAC ACTGACCAGC ACACAGTACA
    641 ACAGCCACAA AGAGTACACC TGCAAGGTGA CCCAGGGCAC
    681 GACCTCAGTC GTCCAGAGCT TCAGTAGGAA GAACTGTTAA
  • In another embodiment, the invention provides a 20D04 heavy chain that can bind to CD83 polypeptides. The amino acid sequence for this 20D04 heavy chain is provided below (SEQ ID NO:13).
  •   1 METGLRWLLL VAVLKGVQCQ SVEESGGRLV TPGTPLTLTC
     41 TVSGFSLSNN AINWVRQAPG KGLEWIGYIW SGGLTYYANW
     81 AEGRFTISKT STTVDLKMTS PTIEDTATYF CARGINNSAL
    121 WGPGTLVTVS SGQPKAPSVF PLAPCCGDTP SSTVTLGCLV
    161 KGYLPEPVTV TWNSGTLTNG VRTFPSVRQS SGLYSLSSVV
    201 SVTSSSQPVT CNVAHPATNT KVDKTVAPST CSKPTCPPPE
    241 LLGGPSVFIF PPKPKDTLMI SRTPEVTCVV VDVSQDDPEV
    281 QFTWYINNEQ VRTARPPLRE QQFNSTIRVV STLPIAHQDW
    321 LRGKEFKCKV HNKALPAPIE KTISKARGQP LEPKVYTMGP
    361 PREELSSRSV SLTCMINGFY PSDISVEWEK NGKAEDNYKT
    401 TPAVLDSDGS YFLYNKLSVP TSEWQRGDVF TCSVMHEALH
    441 NHYTQKSISR SPGK
  • A nucleic acid sequence for this 20D04 anti-CD83 heavy chain is provided below (SEQ ID NO:14).
  •    1 ATGGAGACAG GCCTGCGCTG GCTTCTCCTG GTCGCTGTGC
      41 TCAAAGGTGT CCAGTGTCAG TCGGTGGAGG AGTCCGGGGG
      81 TCGCCTGGTC ACGCCTGGGA CACCCCTGAC ACTCACCTGC
     121 ACCGTCTCTG GATTCTCCCT CAGTAACAAT GCAATAAACT
     161 GGGTCCGCCA GGCTCCAGGG AAGGGGCTAG AGTGGATCGG
     201 ATACATTTGG AGTGGTGGGC TTACATACTA CGCGAACTGG
     241 GCGGAAGGCC GATTCACCAT CTCCAAAACC TCGACTACGG
     281 TGGATCTGAA GATGACCAGT CCGACAATCG AGGACACGGC
     321 CACCTATTTC TGTGCCAGAG GGATTAATAA CTCCGCTTTG
     361 TGGGGCCCAG GCACCCTGGT CACCGTCTCC TCAGGGCAAC
     401 CTAAGGCTCC ATCAGTCTTC CCACTGGCCC CCTGCTGCGG
     441 GGACACACCC TCTAGCACGG TGACCTTGGG CTGCCTGGTC
     481 AAAGGCTACC TCCCGGAGCC AGTGACCGTG ACCTGGAACT
     521 CGGGCACCCT CACCAATGGG GTACGCACCT TCCCGTCCGT
     561 CCGGCAGTCC TCAGGCCTCT ACTCGCTGAG CAGCGTGGTG
     601 AGCGTGACCT CAAGCAGCCA GCCCGTCACC TGCAACGTGG
     641 CCCACCCAGC CACCAACACC AAAGTGGACA AGACCGTTGC
     681 GCCCTCGACA TGCAGCAAGC CCACGTGCCC ACCCCCTGAA
     721 CTCCTGGGGG GACCGTCTGT CTTCATCTTC CCCCCAAAAC
     761 CCAAGGACAC CCTCATGATC TCACGCACCC CCGAGGTCAC
     801 ATGCGTGGTG GTGGACGTGA GCCAGGATGA CCCCGAGGTG
     841 CAGTTCACAT GGTACATAAA CAACGAGCAG GTGCGCACCG
     881 CCCGGCCGCC GCTACGGGAG CAGCAGTTCA ACAGCACGAT
     921 CCGCGTGGTC AGCACCCTCC CCATCGCGCA CCAGGACTGG
     961 CTGAGGGGCA AGGAGTTCAA GTGCAAAGTC CACAACAAGG
    1001 CACTCCCGGC CCCCATCGAG AAAACCATCT CCAAAGCCAG
    1041 AGGGCAGCCC CTGGAGCCGA AGGTCTACAC CATGGGCCCT
    1081 CCCCGGGAGG AGCTGAGCAG CAGGTCGGTC AGCCTGACCT
    1121 GCATGATCAA CGGCTTCTAC CCTTCCGACA TCTCGGTGGA
    1161 GTGGGAGAAG AACGGGAAGG CAGAGGACAA CTACAAGACC
    1201 ACGCCGGCCG TGCTGGACAG CGACGGCTCC TACTTCCTCT
    1241 ACAACAAGCT CTCAGTGCCC ACGAGTGAGT GGCAGCGGGG
    1281 CGACGTCTTC ACCTGCTCCG TGATGCACGA GGCCTTGCAC
    1321 AACCACTACA CGCAGAAGTC CATCTCCCGC TCTCCGGGTA
    1361 AA
  • In another embodiment, the invention provides a 11G05 light chain that can bind to CD83 polypeptides. The amino acid sequence for this 11G05 light chain is provided below (SEQ ID NO:15).
  •   1 MDTRAPTQLL GLLLLWLPGA RCADVVMTQT PASVSAAVGG
     41 TVTINCQSSK NVYNNNWLSW FQQKPGQPPK LLIYYASTLA
     81 SGVPSRFRGS GSGTQFTLTI SDVQCDDAAT YYCAGDYSSS
    121 SDNGFGGGTE VVVKGDPVAP TVLLFPPSSD EVATGTVTIV
    161 CVANKYFPDV TVTWEVDGTT QTTGIENSKT PQNSADCTYN
    201 LSSTLTLTST QYNSHKEYTC KVTQGTTSVV QSFSRKNC
  • A nucleic acid sequence for this 11G05 anti-CD83 light chain is provided below (SEQ ID NO:16).
  •   1 ATGGACACCA GGGCCCCCAC TCAGCTGCTG GGGCTCCTGC
     41 TGCTCTGGCT CCCAGGTGCC AGATGTGCCG ACGTCGTGAT
     81 GACCCAGACT CCAGCCTCCG TGTCTGCAGC TGTGGGAGGC
    121 ACAGTCACCA TCAATTGCCA GTCCAGTAAG AATGTTTATA
    161 ATAACAACTG GTTATCCTGG TTTCAGCAGA AACCAGGGCA
    201 GCCTCCCAAG CTCCTGATCT ATTATGCATC CACTCTGGCA
    241 TCTGGGGTCC CATCGCGGTT CAGAGGCAGT GGATCTGGGA
    281 CACAGTTCAC TCTCACCATT AGCGACGTGC AGTGTGACGA
    321 TGCTGCCACT TACTACTGTG CAGGCGATTA TAGTAGTAGT
    361 AGTGATAATG GTTTCGGCGG AGGGACCGAG GTGGTGGTCA
    401 AAGGTGATCC AGTTGCACCT ACTGTCCTCC TCTTCCCACC
    441 ATCTAGCGAT GAGGTGGCAA CTGGAACAGT CACCATCGTG
    481 TGTGTGGCGA ATAAATACTT TCCCGATGTC ACCGTCACCT
    521 GGGAGGTGGA TGGCACCACC CAAACAACTG GCATCGAGAA
    561 CAGTAAAACA CCGCAGAATT CTGCAGATTG TACCTACAAC
    601 CTCAGCAGCA CTCTGACACT GACCAGCACA CAGTACAACA
    641 GCCACAAAGA GTACACCTGC AAGGTGACCC AGGGCACGAC
    681 CTCAGTCGTC CAGAGCTTCA GTAGGAAGAA CTGTTAA
  • In another embodiment, the invention provides a 11G05 heavy chain that can bind to CD83 polypeptides. The amino acid sequence for this 11G05 heavy chain is provided below (SEQ ID NO:17).
  •   1 METGLRWLLL VAVLKGVQCQ SVEESGGRLV TPGTPLTLTC
     41 TVSGFTISDY DLSWVRQAPG EGLKYIGFIA IDGNPYYATW
     81 AKGRFTISKT STTVDLKITA PTTEDTATYF CARGAGDLWG
    121 PGTLVTVSSG QPKAPSVFPL APCCGDTPSS TVTLGCLVKG
    161 YLPEPVTVTW NSGTLTNGVR TFPSVRQSSG LYSLSSVVSV
    201 TSSSQPVTCN VAHPATNTKV DKTVAPSTCS KPTCPPPELL
    241 GGPSVFIFPP KPKDTLMISR TPEVTCVVVD VSQDDPEVQF
    281 TWYINNEQVR TARPPLREQQ FNSTIRVVST LPIAHQDWLR
    321 GKEFKCKVHN KALPAPIEKT ISKARGQPLE PKVYTMGPPR
    361 EELSSRSVSL TCMINGFYPS DISVEWEKNG KAEDNYKTTP
    401 AVLDSDGSYF LYNKLSVPTS EWQRGDVFTC SVMHEALHNH
    441 YTQKSISRSP GK
  • A nucleic acid sequence for this 11G05 anti-CD83 heavy chain is provided below (SEQ ID NO:18).
  •    1 ATGGAGACAG GCCTGCGCTG GCTTCTCCTG GTCGCTGTGC
      41 TCAAAGGTGT CCAGTGTCAG TCGGTGGAGG AGTCCGGGGG
      81 TCGCCTGGTC ACGCCTGGGA CACCCCTGAC ACTCACCTGC
     121 ACAGTCTCTG GATTCACCAT CAGTGACTAC GACTTGAGCT
     161 GGGTCCGCCA GGCTCCAGGG GAGGGGCTGA AATACATCGG
     201 ATTCATTGCT ATTGATGGTA ACCCATACTA CGCGACCTGG
     241 GCAAAAGGCC GATTCACCAT CTCCAAAACC TCGACCACGG
     281 TGGATCTGAA AATCACCGCT CCGACAACCG AAGACACGGC
     321 CACGTATTTC TGTGCCAGAG GGGCAGGGGA CCTCTGGGGC
     361 CCAGGGACCC TCGTCACCGT CTCTTCAGGG CAACCTAAGG
     401 CTCCATCAGT CTTCCCACTG GCCCCCTGCT GCGGGGACAC
     441 ACCCTCTAGC ACGGTGACCT TGGGCTGCCT GGTCAAAGGC
     481 TACCTCCCGG AGCCAGTGAC CGTGACCTGG AACTCGGGCA
     521 CCCTCACCAA TGGGGTACGC ACCTTCCCGT CCGTCCGGCA
     561 GTCCTCAGGC CTCTACTCGC TGAGCAGCGT GGTGAGCGTG
     601 ACCTCAAGCA GCCAGCCCGT CACCTGCAAC GTGGCCCACC
     641 CAGCCACCAA CACCAAAGTG GACAAGACCG TTGCGCCCTC
     681 GACATGCAGC AAGCCCACGT GCCCACCCCC TGAACTCCTG
     721 GGGGGACCGT CTGTCTTCAT CTTCCCCCCA AAACCCAAGG
     761 ACACCCTCAT GATCTCACGC ACCCCCGAGG TCACATGCGT
     801 GGTGGTGGAC GTGAGCCAGG ATGACCCCGA GGTGCAGTTC
     841 ACATGGTACA TAAACAACGA GCAGGTGCGC ACCGCCCGGC
     881 CGCCGCTACG GGAGCAGCAG TTCAACAGCA CGATCCGCGT
     921 GGTCAGCACC CTCCCCATCG CGCACCAGGA CTGGCTGAGG
     961 GGCAAGGAGT TCAAGTGCAA AGTCCACAAC AAGGCACTCC
    1001 CGGCCCCCAT CGAGAAAACC ATCTCCAAAG CCAGAGGGCA
    1041 GCCCCTGGAG CCGAAGGTCT ACACCATGGG CCCTCCCCGG
    1081 GAGGAGCTGA GCAGCAGGTC GGTCAGCCTG ACCTGCATGA
    1120 TCAACGGCTT CTACCCTTCC GACATCTCGG TGGAGTGGGA
    1161 GAAGAACGGG AAGGCAGAGG ACAACTACAA GACCACGCCG
    1201 GCCGTGCTGG ACAGCGACGG CTCCTACTTC CTCTACAACA
    1241 AGCTCTCAGT GCCCACGAGT GAGTGGCAGC GGGGCGACGT
    1281 CTTCACCTGC TCCGTGATGC ACGAGGCCTT GCACAACCAC
    1321 TACACGCAGA AGTCCATCTC CCGCTCTCCG GGTAAA
  • In another embodiment, the invention provides a 14C12 light chain that can bind to CD83 polypeptides. The amino acid sequence for this 14C12 light chain is provided below (SEQ ID NO:19).
  •   1 MDXRAPTQLL GLLLLWLPGA RCALVMTQTP ASVSAAVGGT
     41 VTINCQSSQS VYDNDELSWY QQKPGQPPKL LIYLASKLAS
     81 GVPSRFKGSG SGTQFALTIS GVQCDDAATY YCQATHYSSD
    121 WYLTFGGGTE VVVKGDPVAP TVLLFPPSSD EVATGTVTIV
    161 CVANKYFPDV TVTWEVDGTT QTTGIENSKT PQNSADCTYN
    201 LSSTLTLTST QYNSHKEYTC KVTQGTTSVV QSFSRKNC
  • A nucleic acid sequence for this 14C12 anti-CD83 light chain is provided below (SEQ ID NO:20).
  •   1 ATGGACATRA GGGCCCCCAC TCAGCTGCTG GGGCTCCTGC
     41 TGCTCTGGCT CCCAGGTGCC AGATGTGCCC TTGTGATGAC
     81 CCAGACTCCA GCCTCCGTGT CTGCAGCTGT GGGAGGCACA
    121 GTCACCATCA ATTGCCAGTC CAGTCAGAGT GTTTATGATA
    161 ACGACGAATT ATCCTGGTAT CAGCAGAAAC CAGGGCAGCC
    201 TCCCAAGCTC CTGATCTATC TGGCATCCAA GTTGGCATCT
    241 GGGGTCCCAT CCCGATTCAA AGGCAGTGGA TCTGGGACAC
    281 AGTTCGCTCT CACCATCAGC GGCGTGCAGT GTGACGATGC
    321 TGCCACTTAC TACTGTCAAG CCACTCATTA TAGTAGTGAT
    361 TGGTATCTTA CTTTCGGCGG AGGGACCGAG GTGGTGGTCA
    401 AAGGTGATCC AGTTGCACCT ACTGTCCTCC TCTTCCCACC
    441 ATCTAGCGAT GAGGTGGCAA CTGGAACAGT CACCATCGTG
    481 TGTGTGGCGA ATAAATACTT TCCCGATGTC ACCGTCACCT
    521 GGGAGGTGGA TGGCACCACC CAAACAACTG GCATCGAGAA
    561 CAGTAAAACA CCGCAGAATT CTGCAGATTG TACCTACAAC
    601 CTCAGCAGCA CTCTGACACT GACCAGCACA CAGTACAACA
    641 GCCACAAAGA GTACACCTGC AAGGTGACCC AGGGCACGAC
    681 CTCAGTCGTC CAGAGCTTCA GTAGGAAGAA CTGTTAA
  • In another embodiment, the invention provides a 14C12 heavy chain that can bind to CD83 polypeptide's. The amino acid sequence for this 14C12 heavy chain is provided below (SEQ ID NO:21).
  •   1 METGLRWLLL VAVLKGVHCQ SVEESGGRLV TPGTPLTLTC
     41 TASGFSRSSY DMSWVRQAPG KGLEWVGVIS TAYNSHYASW
     81 AKGRFTISRT STTVDLKMTS LTTEDTATYF CARGGSWLDL
    121 WGQGTLVTVS SGQPKAPSVF PLAPCCGDTP SSTVTLGCLV
    161 KGYLPEPVTV TWNSGTLTNG VRTFPSVRQS SGLYSLSSVV
    201 SVTSSSQPVT CNVAHPATNT KVDKTVAPST CSKPTCPPPE
    241 LLGGPSVFIF PPKPKDTLMI SRTPEVTCVV VDVSQDDPEV
    281 QFTWYINNEQ VRTARPPLRE QQFNSTIRVV STLPIAHQDW
    321 LRGKEFKCKV HNKALPAPIE KTISKARGQP LEPKVYTMGP
    361 PREELSSRSV SLTCMINGFY PSDISVEWEK NGKAEDNYKT
    401 TPAVLDSDGS YFLYNKLSVP TSEWQRGDVF TCSVMHEALH
    441 NHYTQKSISR SPGK
  • A nucleic acid sequence for this 14C12 anti-CD83 heavy chain is provided below (SEQ ID NO:22).
  •    1 ATGGAGACAG GCCTGCGCTG GCTTCTCCTG GTCGCTGTGC
      41 TCAAAGGTGT CCACTGTCAG TCGGTGGAGG AGTCCGGGGG
      81 TCGCCTGGTC ACGCCTGGGA CACCCCTGAC ACTCACCTGC
     121 ACAGCCTCTG GATTCTCCCG CAGCAGCTAC GACATGAGCT
     161 GGGTCCGCCA GGCTCCAGGG AAGGGGCTGG AATGGGTCGG
     201 AGTCATTAGT ACTGCTTATA ACTCACACTA CGCGAGCTGG
     241 GCAAAAGGCC GATTCACCAT CTCCAGAACC TCGACCACGG
     281 TGGATCTGAA AATGACCAGT CTGACAACCG AAGACACGGC
     321 CACCTATTTC TGTGCCAGAG GGGGTAGTTG GTTGGATCTC
     361 TGGGGCCAGG GCACCCTGGT CACCGTCTCC TCAGGGCAAC
     401 CTAAGGCTCC ATCAGTCTTC CCACTGGCCC CCTGCTGCGG
     441 GGACACACCC TCTAGCACGG TGACCTTGGG CTGCCTGGTC
     481 AAAGGCTACC TCCCGGAGCC AGTGACCGTG ACCTGGAACT
     521 CGGGCACCCT CACCAATGGG GTACGCACCT TCCCGTCCGT
     561 CCGGCAGTCC TCAGGCCTCT ACTCGCTGAG CAGCGTGGTG
     601 AGCGTGACCT CAAGCAGCCA GCCCGTCACC TGCAACGTGG
     641 CCCACCCAGC CACCAACACC AAAGTGGACA AGACCGTTGC
     681 GCCCTCGACA TGCAGCAAGC CCACGTGCCC ACCCCCTGAA
     721 CTCCTGGGGG GACCGTCTGT CTTCATCTTC CCCCCAAAAC
     761 CCAAGGACAC CCTCATGATC TCACGCACCC CCGAGGTCAC
     801 ATGCGTGGTG GTGGACGTGA GCCAGGATGA CCCCGAGGTG
     841 CAGTTCACAT GGTACATAAA CAACGAGCAG GTGCGCACCG
     881 CCCGGCCGCC GCTACGGGAG CAGCAGTTCA ACAGCACGAT
     921 CCGCGTGGTC AGCACCCTCC CCATCGCGCA CCAGGACTGG
     961 CTGAGGGGCA AGGAGTTCAA GTGCAAAGTC CACAACAAGG
    1001 CACTCCCGGC CCCCATCGAG AAAACCATCT CCAAAGCCAG
    1041 AGGGCAGCCC CTGGAGCCGA AGGTCTACAC CATGGGCCCT
    1081 CCCCGGGAGG AGCTGAGCAG CAGGTCGGTC AGCCTGACCT
    1121 GCATGATCAA CGGCTTCTAC CCTTCCGACA TCTCGGTGGA
    1161 GTGGGAGAAG AACGGGAAGG CAGAGGACAA CTACAAGACC
    1200 ACGCCGGCCG TGCTGGACAG CGACGGCTCC TACTTCCTCT
    1241 ACAACAAGCT CTCAGTGCCC ACGAGTGAGT GGCAGCGGGG
    1281 CGACGTCTTC ACCTGCTCCG TGATGCACGA GGCCTTGCAC
    1321 AACCACTACA CGCAGAAGTC CATCTCCCGC TCTCCGGGTA
    1361 AA
  • In another embodiment, the invention provides a M83 020B08L light chain that can bind to CD83 polypeptides. The amino acid sequence for this M83 020B08L light chain is provided below (SEQ ID NO:58).
  •   1 MDMRAPTQLL GLLLLWLPGA RCAYDMTQTP ASVEVAVGGT
     41 VTIKCQASQS ISTYLDWYQQ KPGQPPKLLI YDASDLASGV
     81 PSRFKGSGSG TQFTLTISDL ECADAATYYC QQGYTHSNVD
    121 NVFGGGTEVV VKGDPVAPTV LLFPPSSDEV ATGTVTIVCV
    161 ANKYFPDVTV TWEVDGTTQT TGIENSKTPQ NSADCTYNLS
    201 STLTLTSTQY NSHKEYTCKV TQGTTSVVQS FSRKNC
  • A nucleic acid sequence for this M83 020B08L anti-CD83 light chain is provided below (SEQ ID NO:59).
  •   1 ATGGACATGA GGGCCCCCAC TCAGCTGCTG GGGCTCCTGC
     41 TGCTCTGGCT CCCAGGTGCC AGATGTGCCT ATGATATGAC
     81 CCAGACTCCA GCCTCTGTGG AGGTAGCTGT GGGAGGCACA
    121 GTCACCATCA AGTGCCAGGC CAGTCAGAGC ATTAGTACCT
    161 ACTTAGACTG GTATCAGCAG AAACCAGGGC AGCCTCCCAA
    201 GCTCCTGATC TATGATGCAT CCGATCTGGC ATCTGGGGTC
    241 CCATCGCGGT TCAAAGGCAG TGGATCTGGG ACACAGTTCA
    281 CTCTCACCAT CAGCGACCTG GAGTGTGCCG ATGCTGCCAC
    321 TTACTACTGT CAACAGGGTT ATACACATAG TAATGTTGAT
    361 AATGTTTTCG GCGGAGGGAC CGAGGTGGTG GTCAAAGGTG
    401 ATCCAGTTGC ACCTACTGTC CTCCTCTTCC CACCATCTAG
    441 CGATGAGGTG GCAACTGGAA CAGTCACCAT CGTGTGTGTG
    481 GCGAATAAAT ACTTTCCCGA TGTCACCGTC ACCTGGGAGG
    521 TGGATGGCAC CACCCAAACA ACTGGCATCG AGAACAGTAA
    561 AACACCGCAG AATTCTGCAG ATTGTACCTA CAACCTCAGC
    601 AGCACTCTGA CACTGACCAG CACACAGTAC AACAGCCACA
    641 AAGAGTACAC CTGCAAGGTG ACCCAGGGCA CGACCTCAGT
    681 CGTCCAGAGC TTCAGTAGGA AGAACTGTTA A
  • In another embodiment, the invention provides a M83 020B08H heavy chain that can bind to CD83 polypeptides. The amino acid sequence for this M83 020B08H heavy chain is provided below (SEQ ID NO:60).
  •   1 METGLRWLLL VAVLKGVQCQ SVEESGGRLV TPGTPLTLTC
     41 TVSGFSLSSY DMTWVRQAPG KGLEWIGIIY ASGTTYYANW
     81 AKGRFTISKT STTVDLKVTS PTIGDTATYF CAREGAGVSM
    121 TLWGPGTLVT VSSGQPKAPS VFPLAPCCGD TPSSTVTLGC
    161 LVKGYLPEPV TVTWNSGTLT NGVRTFPSVR QSSGLYSLSS
    201 VVSVTSSSQP VTCNVAHPAT NTKVDKTVAP STCSKPTCPP
    241 PELLGGPSVF IFPPKPKDTL MISRTPEVTC VVVDVSQDDP
    281 EVQFTWYINN EQVRTARPPL REQQFNSTIR VVSTLPIAHQ
    321 DWLRGKEFKC KVHNKALPAP IEKTISKARG QPLEPKVYTM
    361 GPPREELSSR SVSLTCMING FYPSDISVEW EKNGKAEDNY
    401 KTTPAVLDSD GSYFLYNKLS VPTSEWQRGD VFTCSVMHEA
    441 LHNHYTQKSI SRSPGK
  • A nucleic acid sequence for this M83 020B08H anti-CD83 heavy chain is provided below (SEQ ID NO:61).
  •    1 ATGGAGACAG GCCTGCGCTG GCTTCTCCTG GTCGCTGTGC
      41 TCAAAGGTGT CCAGTGTCAG TCGGTGGAGG AGTCCGGGGG
      81 TCGCCTGGTC ACGCCTGGGA CACCCCTGAC ACTCACCTGC
     121 ACAGTCTCTG GATTCTCCCT CAGCAGCTAC GACATGACCT
     161 GGGTCCGCCA GGCTCCAGGG AAGGGGCTGG AATGGATCGG
     201 AATCATTTAT GCTAGTGGTA CCACATACTA CGCGAACTGG
     241 GCGAAAGGCC GATTCACCAT CTCCAAAACC TCGACCACGG
     281 TGGATCTGAA AGTCACCAGT CCGACAATCG GGGACACGGC
     321 CACCTATTTC TGTGCCAGAG AGGGGGCTGG TGTTAGTATG
     361 ACCTTGTGGG GCCCAGGCAC CCTGGTCACC GTCTCCTCAG
     401 GGCAACCTAA GGCTCCATCA GTCTTCCCAC TGGCCCCCTG
     441 CTGCGGGGAC ACACCCTCTA GCACGGTGAC CTTGGGCTGC
     481 CTGGTCAAAG GCTACCTCCC GGAGCCAGTG ACCGTGACCT
     521 GGAACTCGGG CACCCTCACC AATGGGGTAC GCACCTTCCC
     561 GTCCGTCCGG CAGTCCTCAG GCCTCTACTC GCTGAGCAGC
     601 GTGGTGAGCG TGACCTCAAG CAGCCAGCCC GTCACCTGCA
     641 ACGTGGCCCA CCCAGCCACC AACACCAAAG TGGACAAGAC
     681 CGTTGCGCCC TCGACATGCA GCAAGCCCAC GTGCCCACCC
     721 CCTGAACTCC TGGGGGGACC GTCTGTCTTC ATCTTCCCCC
     761 CAAAACCCAA GGACACCCTC ATGATCTCAC GCACCCCCGA
     801 GGTCACATGC GTGGTGGTGG ACGTGAGCCA GGATGACCCC
     841 GAGGTGCAGT TCACATGGTA CATAAACAAC GAGCAGGTGC
     881 GCACCGCCCG GCCGCCGCTA CGGGAGCAGC AGTTCAACAG
     921 CACGATCCGC GTGGTCAGCA CCCTCCCCAT CGCGCACCAG
     961 GACTGGCTGA GGGGCAAGGA GTTCAAGTGC AAAGTCCACA
    1001 ACAAGGCACT CCCGGCCCCC ATCGAGAAAA CCATCTCCAA
    1041 AGCCAGAGGG CAGCCCCTGG AGCCGAAGGT CTACACCATG
    1081 GGCCCTCCCC GGGAGGAGCT GAGCAGCAGG TCGGTCAGCC
    1121 TGACCTGCAT GATCAACGGC TTCTACCCTT CCGACATCTC
    1161 GGTGGAGTGG GAGAAGAACG GGAAGGCAGA GGACAACTAC
    1201 AAGACCACGC CGGCCGTGCT GGACAGCGAC GGCTCCTACT
    1241 TCCTCTACAA CAAGCTCTCA GTGCCCACGA GTGAGTGGCA
    1281 GCGGGGCGAC GTCTTCACCT GCTCCGTGAT GCACGAGGCC
    1321 TTGCACAACC ACTACACGCA GAAGTCCATC TCCCGCTCTC
    1361 CGGGTAAA
  • In another embodiment, the invention provides a M83 006G05L light chain that can bind to CD83 polypeptides. The amino acid sequence for this M83 006G05L light chain is provided below (SEQ ID NO:62).
  •   1 MDMRAPTQLL GLLLLWLPGA RCAYDMTQTP ASVEVAVGGT
     41 VAIKCQASQS VSSYLAWYQQ KPGQPPKPLI YEASMLAAGV
     81 SSRFKGSGSG TDFTLTISDL ECDDAATYYC QQGYSISDID
    121 NAFGGGTEVV VKGDPVAPTV LLFPPSSDEV ATGTVTIVCV
    161 ANKYFPDVTV TWEVDGTTQT TGIENSKTPQ NSADCTYNLS
    201 STLTLTSTQY NSHKEYTCKV TQGTTSVVQS FSRKNC
  • A nucleic acid sequence for M83 006G05L anti-CD83 light chain is provided below (SEQ ID NO:63).
  •   1 ATGGACATGA GGGCCCCCAC TCAACTGCTG GGGCTCCTGC
     41 TGCTCTGGCT CCCAGGTGCC AGATGTGCCT ATGATATGAC
     81 CCAGACTCCA GCCTCTGTGG AGGTAGCTGT GGGAGGCACA
    121 GTCGCCATCA AGTGCCAGGC CAGTCAGAGC GTTAGTAGTT
    161 ACTTAGCCTG GTATCAGCAG AAACCAGGGC AGCCTCCCAA
    201 GCCCCTGATC TACGAAGCAT CCATGCTGGC GGCTGGGGTC
    241 TCATCGCGGT TCAAAGGCAG TGGATCTGGG ACAGACTTCA
    281 CTCTCACCAT CAGCGACCTG GAGTGTGACG ATGCTGCCAC
    321 TTACTATTGT CAACAGGGTT ATTCTATCAG TGATATTGAT
    361 AATGCTTTCG GCGGAGGGAC CGAGGTGGTG GTCAAAGGTG
    401 ATCCAGTTGC ACCTACTGTC CTCCTCTTCC CACCATCTAG
    441 CGATGAGGTG GCAACTGGAA CAGTCACCAT CGTGTGTGTG
    481 GCGAATAAAT ACTTTCCCGA TGTCACCGTC ACCTGGGAGG
    521 TGGATGGCAC CACCCAAACA ACTGGCATCG AGAACAGTAA
    561 AACACCGCAG AATTCTGCAG ATTGTACCTA CAACCTCAGC
    601 AGCACTCTGA CACTGACCAG CACACAGTAC AACAGCCACA
    641 AAGAGTACAC CTGCAAGGTG ACCCAGGGCA CGACCTCAGT
    681 CGTCCAGAGC TTCAGTAGGA AGAACTGTTA A
  • In another embodiment, the invention provides a M83 006G05L heavy chain that can bind to CD83 polypeptides. The amino acid sequence for this M83 006G05L heavy chain is provided below (SEQ ID NO:64).
  •   1 METGLRWLLL VAVLKGVQCQ SVEESGGRLV SPGTPLTLTC
     41 TASGFSLSSY DMSWVRQAPG KGLEYIGIIS SSGSTYYASW
     81 AKGRFTISKT STTVDLEVTS LTTEDTATYF CSREHAGYSG
    121 DTGHLWGPGT LVTVSSGQPK APSVFPLAPC CGDTPSSTVT
    161 LGCLVKGYLP EPVTVTWNSG TLTNGVRTFP SVRQSSGLYS
    201 LSSVVSVTSS SQPVTCNVAH PATNTKVDKT VAPSTCSKPT
    241 CPPPELLGGP SVFIFPPKPK DTLMISRTPE VTCVVVDVSQ
    281 DDPEVQFTWY INNEQVRTAR PPLREQQFNS TIRVVSTLPI
    321 AHQDWLRGKE FKCKVHNKAL PAPIEKTISK ARGQPLEPKV
    361 YTMGPPREEL SSRSVSLTCM INGFYPSDIS VEWEKNGKAE
    401 DNYKTTPAVL DSDGSYFLYN KLSVPTSEWQ RGDVFTCSVM
    441 HEALHNHYTQ KSISRSPGK
  • A nucleic acid sequence for this M83 006G05L anti-CD83 heavy chain is provided below (SEQ ID NO:65).
  •    1 ATGGAGACAG GCCTGCGCTG GCTTCTCCTG GTCGCTGTGC
      41 TCAAAGGTGT CCAGTGTCAG TCGGTGGAGG AGTCCGGGGG
      81 TCGCCTGGTC TCGCCTGGGA CACCCCTGAC ACTCACCTGC
     121 ACAGCCTCTG GATTCTCCCT CAGTAGCTAC GACATGAGCT
     161 GGGTCCGCCA GGCTCCAGGG AAGGGGCTGG AATACATCGG
     201 AATCATTAGT AGTAGTGGTA GCACATACTA CGCGAGCTGG
     241 GCGAAAGGCC GATTCACCAT CTCCAAAACC TCGACCACGG
     281 TGGATCTGGA AGTGACCAGT CTGACAACCG AGGACACGGC
     321 CACCTATTTC TGTAGTAGAG AACATGCTGG TTATAGTGGT
     361 GATACGGGTC ACTTGTGGGG CCCAGGCACC CTGGTCACCG
     401 TCTCCTCGGG GCAACCTAAG GCTCCATCAG TCTTCCCACT
     441 GGCCCCCTGC TGCGGGGACA CACCCTCTAG CACGGTGACC
     481 TTGGGCTGCC TGGTCAAAGG CTACCTCCCG GAGCCAGTGA
     521 CCGTGACCTG GAACTCGGGC ACCCTCACCA ATGGGGTACG
     561 CACCTTCCCG TCCGTCCGGC AGTCCTCAGG CCTCTACTCG
     601 CTGAGCAGCG TGGTGAGCGT GACCTCAAGC AGCCAGCCCG
     641 TCACCTGCAA CGTGGCCCAC CCAGCCACCA ACACCAAAGT
     681 GGACAAGACC GTTGCGCCCT CGACATGCAG CAAGCCCACG
     721 TGCCCACCCC CTGAACTCCT GGGGGGACCG TCTGTCTTCA
     761 TCTTCCCCCC AAAACCCAAG GACACCCTCA TGATCTCACG
     801 CACCCCCGAG GTCACATGCG TGGTGGTGGA CGTGAGCCAG
     841 GATGACCCCG AGGTGCAGTT CACATGGTAC ATAAACAACG
     881 AGCAGGTGCG CACCGCCCGG CCGCCGCTAC GGGAGCAGCA
     921 GTTCAACAGC ACGATCCGCG TGGTCAGCAC CCTCCCCATC
     961 GCGCACCAGG ACTGGCTGAG GGGCAAGGAG TTCAAGTGCA
    1001 AAGTCCACAA CAAGGCACTC CCGGCCCCCA TCGAGAAAAC
    1041 CATCTCCAAA GCCAGAGGGC AGCCCCTGGA GCCGAAGGTC
    1081 TACACCATGG GCCCTCCCCG GGAGGAGCTG AGCAGCAGGT
    1121 CGGTCAGCCT GACCTGCATG ATCAACGGCT TCTACCCTTC
    1162 CGACATCTCG GTGGAGTGGG AGAAGAACGG GAAGGCAGAG
    1201 GACAACTACA AGACCACGCC GGCCGTGCTG GACAGCGACG
    1241 GCTCCTACTT CCTCTACAAC AAGCTCTCAG TGCCCACGAG
    1281 TGAGTGGCAG CGGGGCGACG TCTTCACCTG CTCCGTGATG
    1321 CACGAGGCCT TGCACAACCA CTACACGCAG AAGTCCATCT
    1361 CCCGCTCTCC GGGTAAA
  • Anti-Sense Nucleic Acids
  • Anti-sense nucleic acids can be used to inhibit the function of CD83. In general, the function of CD83 RNA is inhibited, for example, by administering to a mammal a nucleic acid that can inhibit the functioning of CD83 RNA. Nucleic acids that can inhibit the function of a CD83RNA can be generated from coding and non-coding regions of the CD83 gene. However, nucleic acids that can inhibit the function of a CD83 RNA are often selected to be complementary to CD83 nucleic acids that are naturally expressed in the mammalian cell to be treated with the methods of the invention. In some embodiments, the nucleic acids that can inhibit CD83 RNA functions are complementary to CD83 sequences found near the 5′ end of the CD83 coding region. For example, nucleic acids that can inhibit the function of a CD83 RNA can be complementary to the 5′ region of SEQ ID NO:1, SEQ ID NO:3, SEQ ID NO:5 or SEQ ID NO:10.
  • A nucleic acid that can inhibit the functioning of a CD83 RNA need not be 100% complementary to SEQ ID NO:1, SEQ ID NO:3, SEQ ID NO:5 or SEQ ID NO:10. Instead, some variability the sequence of the nucleic acid that can inhibit the functioning of a CD83 RNA is permitted. For example, a nucleic acid that can inhibit the functioning of a CD83 RNA from a human can be complementary to a nucleic acid encoding either a human or a mouse CD83 gene product.
  • Moreover, nucleic acids that can hybridize under moderately or highly stringent hybridization conditions to a nucleic acid comprising SEQ ID NO:1, SEQ ID NO:3, SEQ ID NO:5 or SEQ ID NO:10 are sufficiently complementary to inhibit the functioning of a CD83 RNA and can be utilized in the methods of the invention.
  • “Stringent hybridization conditions” and “stringent hybridization wash conditions” in the context of nucleic acid hybridization are somewhat sequence dependent, and may differ depending upon the environmental conditions of the nucleic acid. For example, longer sequences tend to hybridize specifically at higher temperatures. An extensive guide to the hybridization of nucleic acids is found in Tijssen, Laboratory Techniques in Biochemistry and Molecular biology-Hybridization with Nucleic Acid Probes, page 1, chapter 2 “Overview of principles of hybridization and the strategy of nucleic acid probe assays” Elsevier, N.Y. (1993). See also, J. Sambrook et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Press, N.Y., pp 9.31-9.58 (1989); J. Sambrook et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Press, N.Y. (3rd ed. 2001).
  • Generally, highly stringent hybridization and wash conditions are selected to be about 5° C. lower than the thermal melting point (Tm) for the specific double-stranded sequence at a defined ionic strength and pH. For example, under “highly stringent conditions” or “highly stringent hybridization conditions” a nucleic acid will hybridize to its complement to a detectably greater degree than to other sequences (e.g., at least 2-fold over background). By controlling the stringency of the hybridization and/or washing conditions nucleic acids that are 100% complementary can be hybridized. For DNA-DNA hybrids, the Tm can be approximated from the equation of Meinkoth and Wahl Anal. Biochem. 138:267-284 (1984):

  • Tm81.5° C.+16.6(log M)+0.41 (% GC)−0.61 (% form)−500/L
  • where M is the molarity of monovalent cations, % GC is the percentage of guanosine and cytosine nucleotides in the DNA, % form is the percentage of formamide in the hybridization solution, and L is the length of the hybrid in base pairs. The Tm is the temperature (under defined ionic strength and pH) at which 50% of a complementary target sequence hybridizes to a perfectly matched probe.
  • Very stringent conditions are selected to be equal to the Tm, for a particular probe.
  • Alternatively, stringency conditions can be adjusted to allow some mismatching in sequences so that lower degrees of similarity can hybridize. Typically, stringent conditions will be those in which the salt concentration is less than about 1.5 M Na ion, typically about 0.01 to 1.0 M Na ion concentration (or other salts) at pH 7.0 to 8.3 and the temperature is at least about 30° C. for short probes (e.g., 10 to 50 nucleotides) and at least about 60° C. for long probes (e.g., greater than 50 nucleotides). Stringent conditions may also be achieved with the addition of destabilizing agents such as formamide.
  • Exemplary low stringency conditions include hybridization with a buffer solution of 30 to 35% formamide, 1 M NaCl, 1% SDS (sodium dodecyl sulphate) at 37° C., and a wash in 1× to 2×SSC (20×SSC=3.0 M NaCl and 0.3 M trisodium citrate) at 50 to 55° C. Exemplary moderate stringency conditions include hybridization in 40 to 45% formamide, 1.0 M NaCl, 1% SDS at 37° C., and a wash in 0.5× to 1×SSC at 55 to 60° C. Exemplary high stringency conditions include hybridization in 50% formamide, 1 M NaCl, 1% SDS at 37° C., and a wash in 0.1×SSC at 60 to 65° C.
  • The degree of complementarity or sequence identity of hybrids obtained during hybridization is typically a function of post-hybridization washes, the critical factors being the ionic strength and temperature of the final wash solution. The type and length of hybridizing nucleic acids also affects whether hybridization will occur and whether any hybrids formed will be stable under a given set of hybridization and wash conditions.
  • An example of stringent hybridization conditions for hybridization of complementary nucleic acids that have more than 100 complementary residues on a filter in a Southern or Northern blot is 50% formamide with 1 mg of heparin at 42° C., with the hybridization being carried out overnight. An example of highly stringent conditions is 0.1 5 M NaCl at 72° C. for about 15 minutes. An example of stringent wash conditions is a 0.2×SSC wash at 65° C. for 15 minutes (see also, Sambrook, infra). Often, a high stringency wash is preceded by a low stringency wash to remove background probe signal. An example of medium stringency for a duplex of, e.g., more than 100 nucleotides, is 1×SSC at 45° C. for 15 minutes. An example low stringency wash for a duplex of, e.g., more than 100 nucleotides, is 4-6×SSC at 40° C. for 15 minutes. For short probes (e.g., about 10 to 50 nucleotides), stringent conditions typically involve salt concentrations of less than about 1.0M Na ion, typically about 0.01 to 1.0 M Na ion concentration (or other salts) at pH 7.0 to 8.3, and the temperature is typically at least about 30° C.
  • Stringent conditions can also be achieved with the addition of destabilizing agents such as formamide. In general, a signal to noise ratio of 2× (or higher) than that observed for an unrelated probe in the particular hybridization assay indicates detection of a specific hybridization. Nucleic acids that do not hybridize to each other under stringent conditions are still substantially identical if the proteins that they encode are substantially identical. This occurs, e.g., when a copy of a nucleic acid is created using the maximum codon degeneracy permitted by the genetic code.
  • The following are examples of sets of hybridization/wash conditions that may be used to detect and isolate homologous nucleic acids that are substantially identical to reference nucleic acids of the present invention: a reference nucleotide sequence preferably hybridizes to the reference nucleotide sequence in 7% sodium dodecyl sulfate (SDS), 0.5 M NaPO4, 1 mM EDTA at 50° C. with washing in 2×SSC, 0.1% SDS at 50° C., more desirably in 7% sodium dodecyl sulfate (SDS), 0.5 M NaPO4, 1 mM EDTA at 50° C. with washing in 1×SSC, 0.1% SDS at 50° C., more desirably still in 7% sodium dodecyl sulfate (SDS), 0.5 M NaPO4, 1 mM EDTA at 50° C. with washing in 0.5×SSC, 0.1% SDS at 50° C., preferably in 7% sodium dodecyl sulfate (SDS), 0.5 M NaPO4, 1 mM EDTA at 50° C. with washing in 0.1×SSC, 0.1% SDS at 50° C., more preferably in 7% sodium dodecyl sulfate (SDS), 0.5 M NaPO4, 1 mM EDTA at 50° C. with washing in 0.1×SSC, 0.1% SDS at 65° C.
  • In general, Tm is reduced by about 1° C. for each 1% of mismatching. Thus, Tm, hybridization, and/or wash conditions can be adjusted to hybridize to sequences of the desired sequence identity. For example, if sequences with >90% identity are sought, the Tm can be decreased 10° C. Generally, stringent conditions are selected to be about 5° C. lower than the thermal melting point (Tm) for the specific sequence and its complement at a defined ionic strength and pH. However, severely stringent conditions can utilize a hybridization and/or wash at 1, 2, 3, or 4° C. lower than the thermal melting point (Tm); moderately stringent conditions can utilize a hybridization and/or wash at 6, 7, 8, 9, or 10° C. lower than the thermal melting point (Tm); low stringency conditions can utilize a hybridization and/or wash at 11, 12, 13, 14, 15, or 20° C. lower than the thermal melting point (Tm).
  • If the desired degree of mismatching results in a Tm of less than 45° C. (aqueous solution) or 32° C. (formamide solution), it is preferred to increase the SSC concentration so that a higher temperature can be used. An extensive guide to the hybridization of nucleic acids is found in Tijssen (1993) Laboratory Techniques in Biochemistry and Molecular Biology-Hybridization with Nucleic Acid Probes, Part 1, Chapter 2 (Elsevier, N.Y.); and Ausubel et al., eds. (1995) Current Protocols in Molecular Biology, Chapter 2 (Greene Publishing and Wiley—Interscience, New York). See Sambrook et al. (1989) Molecular Cloning: A Laboratory Manual (2d ed., Cold Spring Harbor. Laboratory Press, Plainview, N.Y.). Using these references and the teachings herein on the relationship between Tm, mismatch, and hybridization and wash conditions, those of ordinary skill can generate variants of the present homocysteine S-methyltransferase nucleic acids.
  • Precise complementarity is therefore not required for successful duplex formation between a nucleic acid that can inhibit a CD83 RNA and the complementary coding sequence of a CD83 RNA. Inhibitory nucleic acid molecules that comprise, for example, 2, 3, 4, or 5 or more stretches of contiguous nucleotides that are precisely complementary to a CD83 coding sequence, each separated by a stretch of contiguous nucleotides that are not complementary to adjacent CD83 coding sequences, can inhibit the function of CD83 RNA. In general, each stretch of contiguous nucleotides is at least 4, 5, 6, 7, or 8 or more nucleotides in length. Non-complementary intervening sequences are preferably 1, 2, 3, or 4 nucleotides in length. One skilled in the art can easily use the calculated melting point of an anti-sense nucleic acid hybridized to a sense nucleic acid to determine the degree of mismatching that will be tolerated between a particular anti-sense nucleic acid and a particular CD83 RNA.
  • Nucleic acids that complementary a CD83 RNA can be administered to a mammal or to directly to the site of the inappropriate immune system activity. Alternatively, nucleic acids that are complementary to a CD83 RNA can generated by transcription from an expression cassette that has been administered to a mammal. For example, a complementary RNA can be transcribed from a CD83 nucleic acid that has been inserted into an expression cassette in the 3′ to 5′ orientation, that is, opposite to the usual orientation employed to generate sense RNA transcripts. Hence, to generate a complementary RNA that can inhibit the function of an endogenous CD83 RNA, the promoter would be positioned to transcribe from a 3′ site towards the 5′ end of the CD83 coding region.
  • In some embodiments an RNA that can inhibit the function of an endogenous CD83 RNA is an anti-sense oligonucleotide. The anti-sense oligonucleotide is complementary to at least a portion of the coding sequence of a gene comprising SEQ ID NO:1, SEQ ID NO:3, SEQ ID NO:5 or SEQ ID NO:10. Such anti-sense oligonucleotides are generally at least six nucleotides in length, but can be about 8, 12, 15, 20, 25, 30, 35, 40, 45, or 50 nucleotides long. Longer oligonucleotides can also be used. CD83 anti-sense oligonucleotides can be provided in a DNA construct and introduced into cells whose division is to be decreased, for example, into CD4+ T cells, Th-1 cells, Th-2 cells or lymphocyte precursor cells.
  • Anti-sense oligonucleotides can be composed of deoxyribonucleotides, ribonucleotides, or a combination of both. Oligonucleotides can be synthesized endogenously from transgenic expression cassettes or vectors as described herein. Alternatively, such oligonucleotides can be synthesized manually or by an automated synthesizer, by covalently linking the 5′ end of one nucleotide with the 3′ end of another nucleotide with non-phosphodiester internucleotide linkages such alkylphosphonates, phosphorothioates, phosphorodithioates, alkylphosphonothioates, alkylphosphonates, phosphoramidates, phosphate esters, carbamates, acetamidate, carboxymethyl esters, carbonates, and phosphate triesters. See Brown, 1994, Meth. Mol. Biol. 20:1-8; Sonveaux, 1994, Meth. Mol. Biol. 26:1-72; Uhlmann et al., 1990, Chem. Rev. 90:543-583.
  • CD83 anti-sense oligonucleotides can be modified without affecting their ability to hybridize to a CD83 RNA. These modifications can be internal or at one or both ends of the anti-sense molecule. For example, internucleoside phosphate linkages can be modified by adding peptidyl, cholesteryl or diamine moieties with varying numbers of carbon residues between these moities and the terminal ribose. Modified bases and/or sugars, such as arabinose instead of ribose, or a 3′,5′-substituted oligonucleotide in which the 3′ hydroxyl group or the 5′ phosphate group are substituted, can also be employed in a modified anti-sense oligonucleotide. These modified oligonucleotides can be prepared by methods available in the art. Agrawal et al., 1992, Trends Biotechnol. 10:152-158; Uhlmann et al., 1990, Chem. Rev. 90:543-584; Uhlmann et al., 1987, Tetrahedron. Lett. 215:3539-3542.
  • In one embodiment of the invention, expression of a CD83 gene is decreased using a ribozyme. A ribozyme is an RNA molecule with catalytic activity. See, e.g., Cech, 1987, Science 236: 1532-1539; Cech, 1990, Ann. Rev. Biochem. 59:543-568; Cech, 1992, Curr. Opin. Struct. Biol. 2: 605-609; Couture and Stinchcomb, 1996, Trends Genet. 12: 510-515. Ribozymes can be used to inhibit gene function by cleaving an RNA sequence, as is known in the art (see, e.g., Haseloff et al., U.S. Pat. No. 5,641,673).
  • CD83 nucleic acids complementary to SEQ ID NO:1, SEQ ID NO:3, SEQ ID NO:5 or SEQ ID NO:10 can be used to generate ribozymes that will specifically bind to mRNA transcribed from a CD83 gene. Methods of designing and constructing ribozymes that can cleave other RNA molecules in trans in a highly sequence specific manner have been developed and described in the art (see Haseloff et al. (1988), Nature 334:585-591). For example, the cleavage activity of ribozymes can be targeted to specific RNAs by engineering a discrete “hybridization” region into the ribozyme. The hybridization region contains a sequence complementary to the target RNA and thus specifically hybridizes with the target (see, for example, Gerlach et al., EP 321,201). The target sequence can be a segment of about 10, 12, 15, 20, or 50 contiguous nucleotides selected from a nucleotide sequence shown in SEQ ID NO:1, SEQ ID NO:3, SEQ ID NO:5 or SEQ ID NO:10. Longer complementary sequences can be used to increase the affinity of the hybridization sequence for the target. The hybridizing and cleavage regions of the ribozyme can be integrally related; thus, upon hybridizing to the target RNA through the complementary regions, the catalytic region of the ribozyme can cleave the target.
  • Other CD83 Modulating Molecules
  • A wide variety of molecules may be used to modulate CD83 activity or function. Such molecules can also be used to modulate the immune system independent of CD83. Compositions and methods for modulating CD83 activity or expression can include these molecules as well as other components. Representative examples that are discussed in more detail below include transcription factors, RNA-binding factors, organic molecules, or peptides.
  • RNA-Binding Factors:
  • One class of molecules that can be used to modulate cytokine levels or GM-CSF levels by way of the CD83 gene is the RNA binding factors. Such factors include those described in PCT/EP01/14820 and other sources.
  • For example, the HuR protein (Genbank accession number U38175) has the ability to specifically bind to CD83 RNA at AU-rich elements or sites. Such AU-rich elements comprise sequences such as AUUUA (SEQ ID NO:49), AUUUUA (SEQ ID NO:50) and AUUUUUA (SEQ ID NO:51). Binding by such HuR proteins to CD83 mRNA is thought to increase the stability, transport and translation of CD83 mRNA, and thereby increase the expression of CD83 polypeptides. Hence, CD83 expression may be increase by administering HuR proteins or nucleic acids to a mammal.
  • Conversely, CD83 expression may be decreased by administering factors that block HuR binding to CD83 mRNA. Factors that block HuR binding include proteins or nucleic acids that can bind to the AU-rich elements normally bound by HuR, for example, nucleic acids or anti-sense nucleic acids that are complementary to AU-rich elements.
  • Organic Molecules:
  • Numerous organic molecules may be used to modulate the immune system. These compounds include any compound that can interact with a component of the immune system. Such compounds may interact directly with CD83, indirectly with CD83 or with some other polypeptide, cell or factor that plays a role in the function of the immune system. In some embodiments, the organic molecule can bind to a CD83 polypeptide or a CD83 nucleic acid.
  • Organic molecules can be tested or assayed for their ability to modulate CD83 activity, CD83 function or for their ability to modulate components of the immune system. For example, within one embodiment of the invention suitable organic molecules may be selected either from a chemical library, wherein chemicals are assayed individually, or from combinatorial chemical libraries where multiple compounds are assayed at once, then deconvoluted to determine and isolate the most active compounds.
  • Representative examples of such combinatorial chemical libraries include those described by Agrafiotis et al., “System and method of automatically generating chemical compounds with desired properties,” U.S. Pat. No. 5,463,564; Armstrong, R. W., “Synthesis of combinatorial arrays of organic compounds through the use of multiple component combinatorial array syntheses,” WO 95/02566; Baldwin, J. J. et al., “Sulfonamide derivatives and their use,” WO 95/24186; Baldwin, J. J. et al., “Combinatorial dihydrobenzopyran library,” WO 95/30642; Brenner, S “New kit for preparing combinatorial libraries,” WO 95/16918; Chenera, B. et al., “Preparation of library of resin-bound aromatic carbocyclic compounds,” WO 95/16712; Ellman, J. A., “Solid phase and combinatorial synthesis of benzodiazepine compounds on a solid support,” U.S. Pat. No. 5,288,514; Felder, E. et al., “Novel combinatorial compound libraries,” WO 95/16209; Lerner, R. et al., “Encoded combinatorial chemical libraries,” WO 93/20242; Pavia, M. R. et al., “A method for preparing and selecting pharmaceutically useful non-peptide compounds from a structurally diverse universal library,” WO 95/04277; Summerton, J. E. and D. D. Weller, “Morpholino-subunit combinatorial library and method,” U.S. Pat. No. 5,506,337; Holmes, C., “Methods for the Solid Phase Synthesis of Thiazolidinones, Metathiazanones, and Derivatives thereof,” WO 96/00148; Phillips, G. B. and G. P. Wei, “Solid-phase Synthesis of Benzimidazoles,” Tet. Letters 37:4887-90, 1996; Ruhland, B. et al., “Solid-supported Combinatorial Synthesis of Structurally Diverse□□-Lactams,” J. Amer. Chem. Soc. 111:253-4, 1996; Look, G. C. et al., “The Identification of Cyclooxygenase-1 Inhibitors from 4-Thiazolidinone Combinatorial Libraries,” Bioorg and Med. Chem. Letters 6:707-12, 1996.
  • Peptides:
  • Peptide molecules that modulate the immune system may be obtained through the screening of combinatorial peptide libraries. Such libraries may either be prepared by one of skill in the art (see e.g., U.S. Pat. Nos. 4,528,266 and 4,359,535, and Patent Cooperation Treaty Publication Nos. WO 92/15679, WO 92/15677, WO 90/07862, WO 90/02809, or purchased from commercially available sources (e.g., New England Biolabs Ph.D.™ Phage Display Peptide Library Kit).
  • Methods of Using the CD83 Mutant Mouse
  • In one embodiment, the invention provides a method for identifying ligands, receptors, therapeutic drugs and other molecules that can modulate the phenotype of the mutant CD83 in vivo. This method involves administering a test compound to the mutant CD83 mouse of the invention and observing whether the compound causes a change in the phenotype of the mutant mouse. Changes in phenotype that are of interest include increases or decreases in T cells (especially CD4+ T cells), increases or decreases in GMCSF, IL-2, IL-4 or IL-10 cytokine production, increases or decreases in inflammation, increases or decreases in dendritic cell function and other T cell responses known to one of skill in the art.
  • Test compounds can be screened in vitro to ascertain whether they interact directly with CD83. In vitro screening can, for example, identify whether a test compound or molecule can bind to the cytoplasmic tail or the membrane-associated portions of CD83. Such information, combined with observation of the in vivo phenotype before and after administration of the test compound provides further insight into the function of CD83 and provides targets for manipulation T cell activation and other functions modulated by CD83.
  • The invention is not limited to identification of molecules that directly associate with CD83. The in vivo screening methods provided herein can, also identify test compounds that have an indirect effect on CD83, or that partially or completely replace a function of CD83.
  • Increases or decreases in T cell numbers can be observed in blood samples or in samples obtained from thymus, spleen or lymph node tissues. In order to observe the activation of T cells and/or the interaction of T cells and dendritic cells, dendritic cells can be pulsed with antigens ex vivo and then injected into mice to prime CD4+ T cells in draining lymphoid organs. See Inaba et al., J. Exp. Med. 172: 631-640, 1990; Liu, et al., J. Exp. Med. 177: 1299-1307, 1993; Sornasse et al., J. Exp. Med. 175: 15-21, 1992. Antigens can also be deposited intramuscularly and dendritic cells from the corresponding afferent lymphatics can carry that antigen in a form stimulatory for T cells. Bujdoso et al., J. Exp. Med. 170: 1285-1302, 1989. According to the invention, factors stimulating the interaction of dendritic cells with T cells in vivo can be identified by administering antigens in this manner and then observing how T cell respond, e.g. by observing whether T cell activation occurs.
  • Increases or decreases in cytokine levels can be observed by methods provided herein or by other methods available in the art.
  • Compositions
  • The CD83 polypeptides and antibodies of the invention, including their salts, are administered so as to achieve a reduction in at least one symptom associated with an infection, indication or disease.
  • To achieve the desired effect(s), the polypeptide or antibody, a variant thereof or a combination thereof, may be administered as single or divided dosages, for example, of at least about 0.01 mg/kg to about 500 to 750 mg/kg, of at least about 0.01 mg/kg to about 300 to 500 mg/kg, at least about 0.1 mg/kg to about 100 to 300 mg/kg or at least about 1 mg/kg to about 50 to 100 mg/kg of body weight, although other dosages may provide beneficial results. The amount administered will vary depending on various factors including, but not limited to, the polypeptide or antibody chosen, the disease, the weight, the physical condition, the health, the age of the mammal, whether prevention or treatment is to be achieved, and if the polypeptide or antibody is chemically modified. Such factors can be readily determined by the clinician employing animal models or other test systems that are available in the art.
  • Administration of the therapeutic agents in accordance with the present invention may be in a single dose, in multiple doses, in a continuous or intermittent manner, depending, for example, upon the recipient's physiological condition, whether the purpose of the administration is therapeutic or prophylactic, and other factors known to skilled practitioners. The administration of the CD83 polypeptides and antibodies of the invention may be essentially continuous over a preselected period of time or may be in a series of spaced doses. Both local and systemic administration is contemplated.
  • To prepare the composition, CD83 polypeptides and antibodies are synthesized or otherwise obtained, purified as necessary or desired and then lyophilized and stabilized. The polypeptide or antibody can then be adjusted to the appropriate concentration, and optionally combined with other agents. The absolute weight of a given polypeptide or antibody included in a unit dose can vary widely. For example, about 0.01 to about 2 g, or about 0.1 to about 500 mg, of at least one polypeptide or antibody of the invention, or a plurality of CD83 polypeptides and antibodies specific for a particular cell type can be administered. Alternatively, the unit dosage can vary from about 0.01 g to about 50 g, from about 0.01 g to about 35 g, from about 0.1 g to about 25 g, from about 0.5 g to about 12 g, from about 0.5 g to about 8 g, from about 0.5 g to about 4 g, or from about 0.5 g to about 2 g.
  • Daily doses of the CD83 polypeptides or antibodies of the invention can vary as well. Such daily doses can range, for example, from about 0.1 g/day to about 50 g/day, from about 0.1 g/day to about 25 g/day, from about 0.1 g/day to about 12 g/day, from about 0.5 g/day to about 8 g/day, from about 0.5 g/day to about 4 g/day, and from about 0.5 g/day to about 2 g/day.
  • Thus, one or more suitable unit dosage forms comprising the therapeutic CD83 polypeptides or antibodies of the invention can be administered by a variety of routes including oral, parenteral (including subcutaneous, intravenous, intramuscular and intraperitoneal), rectal, dermal, transdermal, intrathoracic, intrapulmonary and intranasal (respiratory) routes. The therapeutic CD83 polypeptides or antibodies may also be formulated for sustained release (for example, using microencapsulation, see WO 94/07529, and U.S. Pat. No. 4,962,091). The formulations may, where appropriate, be conveniently presented in discrete unit dosage forms and may be prepared by any of the methods well known to the pharmaceutical arts. Such methods may include the step of mixing the therapeutic agent with liquid carriers, solid matrices, semi-solid carriers, finely divided solid carriers or combinations thereof, and then, if necessary, introducing or shaping the product into the desired delivery system.
  • When the therapeutic CD83 polypeptides or antibodies of the invention are prepared for oral administration, they are generally combined with a pharmaceutically acceptable carrier, diluent or excipient to form a pharmaceutical formulation, or unit dosage form. For oral administration, the CD83 polypeptides or antibodies may be present as a powder, a granular formulation, a solution, a suspension, an emulsion or in a natural or synthetic polymer or resin for ingestion of the active ingredients from a chewing gum. The active CD83 polypeptides or antibodies may also be presented as a bolus, electuary or paste. Orally administered therapeutic CD83 polypeptides or antibodies of the invention can also be formulated for sustained release, e.g., the CD83 polypeptides or antibodies can be coated, micro-encapsulated, or otherwise placed within a sustained delivery device. The total active ingredients in such formulations comprise from 0.1 to 99.9% by weight of the formulation.
  • By “pharmaceutically acceptable” it is meant a carrier, diluent, excipient, and/or salt that is compatible with the other ingredients of the formulation, and not deleterious to the recipient thereof.
  • Pharmaceutical formulations containing the therapeutic CD83 polypeptides or antibodies of the invention can be prepared by procedures known in the art using well-known and readily available ingredients. For example, the polypeptide or antibody can be formulated with common excipients, diluents, or carriers, and formed into tablets, capsules, solutions, suspensions, powders, aerosols and the like. Examples of excipients, diluents, and carriers that are suitable for such formulations include buffers, as well as fillers and extenders such as starch, cellulose, sugars, mannitol, and silicic derivatives. Binding agents can also be included such as carboxymethyl cellulose, hydroxymethylcellulose, hydroxypropyl methylcellulose and other cellulose derivatives, alginates, gelatin, and polyvinyl-pyrrolidone. Moisturizing agents can be included such as glycerol, disintegrating agents such as calcium carbonate and sodium bicarbonate. Agents for retarding dissolution can also be included such as paraffin. Resorption accelerators such as quaternary ammonium compounds can also be included. Surface active agents such as cetyl alcohol and glycerol monostearate can be included. Adsorptive carriers such as kaolin and bentonite can be added. Lubricants such as talc, calcium and magnesium stearate, and solid polyethyl glycols can also be included. Preservatives may also be added. The compositions of the invention can also contain thickening agents such as cellulose and/or cellulose derivatives. They may also contain gums such as xanthan, guar or carbo gum or gum arabic, or alternatively polyethylene glycols, bentones and montmorillonites, and the like.
  • For example, tablets or caplets containing the CD83 polypeptides or antibodies of the invention can include buffering agents such as calcium carbonate, magnesium oxide and magnesium carbonate. Caplets and tablets can also include inactive ingredients such as cellulose, pregelatinized starch, silicon dioxide, hydroxy propyl methyl cellulose, magnesium stearate, microcrystalline cellulose, starch, talc, titanium dioxide, benzoic acid, citric acid, corn starch, mineral oil, polypropylene glycol, sodium phosphate, zinc stearate, and the like. Hard or soft gelatin capsules containing at least one polypeptide or antibody of the invention can contain inactive ingredients such as gelatin, microcrystalline cellulose, sodium lauryl sulfate, starch, talc, and titanium dioxide, and the like, as well as liquid vehicles such as polyethylene glycols (PEGs) and vegetable oil. Moreover, enteric-coated caplets or tablets containing one or more CD83 polypeptides or antibodies of the invention are designed to resist disintegration in the stomach and dissolve in the more neutral to alkaline environment of the duodenum.
  • The therapeutic CD83 polypeptides or antibodies of the invention can also be formulated as elixirs or solutions for convenient oral administration or as solutions appropriate for parenteral administration, for instance by intramuscular, subcutaneous, intraperitoneal or intravenous routes. The pharmaceutical formulations of the therapeutic CD83 polypeptides or antibodies of the invention can also take the form of an aqueous or anhydrous solution or dispersion, or alternatively the form of an emulsion or suspension or salve.
  • Thus, the therapeutic CD83 polypeptides or antibodies may be formulated for parenteral administration (e.g., by injection, for example, bolus injection or continuous infusion) and may be presented in unit dose form in ampules, pre-filled syringes, small volume infusion containers or in multi-dose containers. As noted above, preservatives can be added to help maintain the shelve life of the dosage form. The active CD83 polypeptides or antibodies and other ingredients may form suspensions, solutions, or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents. Alternatively, the active CD83 polypeptides or antibodies and other ingredients may be in powder form, obtained by aseptic isolation of sterile solid or by lyophilization from solution, for constitution with a suitable vehicle, e.g., sterile, pyrogen-free water, before use.
  • These formulations can contain pharmaceutically acceptable carriers, vehicles and adjuvants that are well known in the art. It is possible, for example, to prepare solutions using one or more organic solvent(s) that is/are acceptable from the physiological standpoint, chosen, in addition to water, from solvents such as acetone, ethanol, isopropyl alcohol, glycol ethers such as the products sold under the name “Dowanol,” polyglycols and polyethylene glycols, C1-C4 alkyl esters of short-chain acids, ethyl or isopropyl lactate, fatty acid triglycerides such as the products marketed under the name “Miglyol,” isopropyl myristate, animal, mineral and vegetable oils and polysiloxanes.
  • It is possible to add, if necessary, an adjuvant chosen from antioxidants, surfactants, other preservatives, film-forming, keratolytic or comedolytic agents, perfumes, flavorings and colorings. Antioxidants such as t-butylhydroquinone, butylated hydroxyanisole, butylated hydroxytoluene and α-tocopherol and its derivatives can be added.
  • Also contemplated are combination products that include one or more CD83 polypeptides or antibodies of the present invention and one or more other anti-microbial agents. For example, a variety of antibiotics can be included in the pharmaceutical compositions of the invention, such as aminoglycosides (e.g., streptomycin, gentamicin, sisomicin, tobramycin and amicacin), ansamycins (e.g. rifamycin), antimycotics (e.g. polyenes and benzofuran derivatives), β-lactams (e.g. penicillins and cephalosporins), chloramphenical (including thiamphenol and azidamphenicol), linosamides (lincomycin, clindamycin), macrolides (erythromycin, oleandomycin, spiramycin), polymyxins, bacitracins, tyrothycin, capreomycin, vancomycin, tetracyclines (including oxytetracycline, minocycline, doxycycline), phosphomycin and fusidic acid.
  • Additionally, the CD83 polypeptides or antibodies are well suited to to formulation as sustained release dosage forms and the like. The formulations can be so constituted that they release the active polypeptide or antibody, for example, in a particular part of the intestinal or respiratory tract, possibly over a period of time. Coatings, envelopes, and protective matrices may be made, for example, from polymeric substances, such as polylactide-glycolates, liposomes, microemulsions, microparticles, nanoparticles, or waxes. These coatings, envelopes, and protective matrices are useful to coat indwelling devices, e.g., stents, catheters, peritoneal dialysis tubing, draining devices and the like.
  • For topical administration, the therapeutic agents may be formulated as is known in the art for direct application to a target area. Forms chiefly conditioned for topical application take the form, for example, of creams, milks, gels, dispersion or microemulsions, lotions thickened to a greater or lesser extent, impregnated pads, ointments or sticks, aerosol formulations (e.g., sprays or foams), soaps, detergents, lotions or cakes of soap. Other conventional forms for this purpose include wound dressings, coated bandages or other polymer coverings, ointments, creams, lotions, pastes, jellies, sprays, and aerosols. Thus, the therapeutic CD83 polypeptides or antibodies of the invention can be delivered via patches or bandages for dermal administration. Alternatively, the polypeptide or antibody can be formulated to be part of an adhesive polymer, such as polyacrylate or acrylate/vinyl acetate copolymer. For long-term applications it might be desirable to use microporous and/or breathable backing laminates, so hydration or maceration of the skin can be minimized. The backing layer can be any appropriate thickness that will provide the desired protective and support functions. A suitable thickness will generally be from about 10 to about 200 microns.
  • Ointments and creams may, for example, be formulated with an aqueous or oily base with the addition of suitable thickening and/or gelling agents. Lotions may be formulated with an aqueous or oily base and will in general also contain one or more emulsifying agents, stabilizing agents, dispersing agents, suspending agents, thickening agents, or coloring agents. The active CD83 polypeptides or antibodies can also be delivered via iontophoresis, e.g., as disclosed in U.S. Pat. Nos. 4,140,122; 4,383,529; or 4,051,842. The percent by weight of a therapeutic agent of the invention present in a topical formulation will depend on various factors, but generally will be from 0.01% to 95% of the total weight of the formulation, and typically 0.1-85% by weight.
  • Drops, such as eye drops or nose drops, may be formulated with one or more of the therapeutic CD83 polypeptides or antibodies in an aqueous or non-aqueous base also comprising one or more dispersing agents, solubilizing agents or suspending agents. Liquid sprays are conveniently delivered from pressurized packs. Drops can be delivered via a simple eye dropper-capped bottle, or via a plastic bottle adapted to deliver liquid contents dropwise, via a specially shaped closure.
  • The therapeutic polypeptide or antibody may further be formulated for topical administration in the mouth or throat. For example, the active ingredients may be formulated as a lozenge further comprising a flavored base, usually sucrose and acacia or tragacanth; pastilles comprising the composition in an inert base such as gelatin and glycerin or sucrose and acacia; and mouthwashes comprising the composition of the present invention in a suitable liquid carrier.
  • The pharmaceutical formulations of the present invention may include, as optional ingredients, pharmaceutically acceptable carriers, diluents, solubilizing or emulsifying agents, and salts of the type that are available in the art. Examples of such substances include normal saline solutions such as physiologically buffered saline solutions and water. Specific non-limiting examples of the carriers and/or diluents that are useful in the pharmaceutical formulations of the present invention include water and physiologically acceptable buffered saline solutions such as phosphate buffered saline solutions pH 7.0-8.0.
  • The CD83 polypeptides or antibodies of the invention can also be administered to the respiratory tract. Thus, the present invention also provides aerosol pharmaceutical formulations and dosage forms for use in the methods of the invention. In general, such dosage forms comprise an amount of at least one of the agents of the invention effective to treat or prevent the clinical symptoms of a specific infection, indication or disease. Any statistically significant attenuation of one or more symptoms of an infection, indication or disease that has been treated pursuant to the method of the present invention is considered to be a treatment of such infection, indication or disease within the scope of the invention.
  • Alternatively, for administration by inhalation or insufflation, the composition may take the form of a dry powder, for example, a powder mix of the therapeutic agent and a suitable powder base such as lactose or starch. The powder composition may be presented in unit dosage form in, for example, capsules or cartridges, or, e.g., gelatin or blister packs from which the powder may be administered with the aid of an inhalator, insufflator, or a metered-dose inhaler (see, for example, the pressurized metered dose inhaler (MDI) and the dry powder inhaler disclosed in Newinan, S. P. in Aerosols and the Lung, Clarke, S. W. and Davia, D. eds., pp. 197-224, Butterworths, London, England, 1984).
  • Therapeutic CD83 polypeptides or antibodies of the present invention can also be administered in an aqueous solution when administered in an aerosol or inhaled form. Thus, other aerosol pharmaceutical formulations may comprise, for example, a physiologically acceptable buffered saline solution containing between about 0.1 mg/ml and about 100 mg/ml of one or more of the CD83 polypeptides or antibodies of the present invention specific for the indication or disease to be treated. Dry aerosol in the form of finely divided solid polypeptide or antibody or nucleic acid particles that are not dissolved or suspended in a liquid are also useful in the practice of the present invention. CD83 polypeptides or antibodies of the present invention may be formulated as dusting powders and comprise finely divided particles having an average particle size of between about 1 and 5 μm, alternatively between 2 and 3 μm. Finely divided particles may be prepared by pulverization and screen filtration using techniques well known in the art. The particles may be administered by inhaling a predetermined quantity of the finely divided material, which can be in the form of a powder. It will be appreciated that the unit content of active ingredient or ingredients contained in an individual aerosol dose of each dosage form need not in itself constitute an effective amount for treating the particular infection, indication or disease since the necessary effective amount can be reached by administration of a plurality of dosage units. Moreover, the effective amount may be achieved using less than the dose in the dosage form, either individually, or in a series of administrations.
  • For administration to the upper (nasal) or lower respiratory tract by inhalation, the therapeutic CD83 polypeptides or antibodies of the invention are conveniently delivered from a nebulizer or a pressurized pack or other convenient means of delivering an aerosol spray. Pressurized packs may comprise a suitable propellant such as dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas. In the case of a pressurized aerosol, the dosage unit may be determined by providing a valve to deliver a metered amount. Nebulizers include, but are not limited to, those described in U.S. Pat. Nos. 4,624,251; 3,703,173; 3,561,444; and 4,635,627. Aerosol delivery systems of the type disclosed herein are available from numerous commercial sources including Fisons Corporation (Bedford, Mass.), Schering Corp. (Kenilworth, N.J.) and American Pharmoseal Co., (Valencia, Calif.). For intra-nasal administration, the therapeutic agent may also be administered via nose drops, a liquid spray, such as via a plastic bottle atomizer or metered-dose inhaler. Typical of atomizers are the Mistometer (Wintrop) and the Medihaler (Riker).
  • Furthermore, the active ingredients may also be used in combination with other therapeutic agents, for example, pain relievers, anti-inflammatory agents, antihistamines, bronchodilators and the like, whether for the conditions described or some other condition.
  • The present invention further pertains to a packaged pharmaceutical composition for controlling microbial infections such as a kit or other container. The kit or container holds a therapeutically effective amount of a pharmaceutical composition for controlling microbial infections and instructions for using the pharmaceutical composition for control of the microbial infection. The pharmaceutical composition includes at least one polypeptide or antibody of the present invention, in a therapeutically effective amount such that the selected disease or immunological condition is controlled.
  • The invention will be further described by reference to the following detailed examples, which are given for illustration of the invention, and are not intended to be limiting thereof.
  • Example 1 Mouse Mutation and Characterization Mutant Generation
  • Male C57BL6 mice received 3 weekly injections of N-ethyl-N-nitrosourea (ENU) at a concentration of 100 mg/kg. N-Ethyl-N-nitrosourea was quantified prior to injection by spectrophotometry. Mice that regained fertility after a minimum period of 12 weeks were then used to generate pedigree founder G1 animals. G1 male mice were crossed to C57BL6J females and their female progeny (G2 animals) crossed back to their fathers to generate G3 animals for screening.
  • G3 mice were weaned at 3 weeks of age. Each animal then underwent a series of screens designed to assess a number of parameters, including immune function, inflammatory response and bone development. In the initial screen, conducted at 6 weeks of age, 150-200 ul of whole blood was collected by retro-orbital bleed into heparinized tubes. Cells were pelleted and red blood cells lysed. Samples were then stained with antibodies to cell surface markers expressed on distinct lymphoid and myeloid sub-populations. These samples were analyzed by flow-cytometry.
  • Mutant Identification
  • A group of 27 G3 mice from 2 different pedigrees, pedigree 9 and pedigree 57 (i.e. derived from 2 distinct G1 fathers) were analyzed in this screen. Two animals from pedigree 9 were identified as having a reduced (>2 standard deviation from normal) percentage of CD4+ T cells in peripheral blood (FIG. 1). Both animals were descended from the same G1 and shared the same mother. All other animals screened on that day had a normal percentage of CD4+ T cells. The number of phenodeviants identified (2 from a litter of 9 animals) was suggestive of a trait controlled by a single gene and inherited in a Mendelian fashion.
  • A second litter generated from Pedigree 9 bred to G2 daughter #4 exhibited an identical phenotype with reduced numbers of CD4+ T cells, further suggesting that the trait had a genetic basis. The phenotype was designated LCD4.1 (Low CD4 Mutant 1) and was used for mapping experiments.
  • Mutation Mapping
  • In order to map the LCD4.1 mutant phenotype, affected G3 male mice (presumptive homozygous for the mutation) were bred to female animals from the C3HeB/FeJ strain to generate F1 progeny. These F1 females (presumptively heterozygous for the mutation) were then mated back to their affected father to generate N2 progeny.
  • Blood was collected from N2 animals and flow cytometric analysis was performed to identify CD4+ T cells. For a phenotype controlled by a single gene, breeding homozygous fathers to heterozygous daughters should yield 50% normal N2 animals and 50% affected N2 animals. This ratio of normal to affected animals was observed in the N2 generation: Multiple N2 animals exhibited a reduced percentage of CD4+ T cells, indicating that the phenotype was heritable (FIG. 2).
  • DNA samples were prepared from samples of tail tissue collected from these N2 mice and used for a genome scan, using a collection of assembled markers, and performed on the ABI 3100 DNA analyzer. Initial genetic linkage was seen to the tip of chromosome 13, where the closest microsatellite marker was D13Mit139 with a LOD score of 8.2. By calculating upper and lower confidence limits, the mutant gene was located between 13.4 and 29.6 cM on chromosome 13. Through additional genotyping, this region was reduced to an 11 cM interval on chromosome 13. No significant linkage to other chromosomal regions was seen.
  • Mutation Identification
  • A candidate gene, CD83, was identified for gene-testing based upon its reported position within the interval. CD83 has previously been used as a marker of dendritic cell activation, suggesting that it might play a role in dendritic cell function and hence in regulating T cell development and function.
  • Sequence analysis of the mutant DNA revealed a mutation in the stop codon of CD83. All affected animals were homozygous for this mutation while non-affected animals carried one wild-type allele and one mutant allele (FIG. 3 and FIG. 4). The mutation destroyed the stop codon and resulted in the addition of a unique 55 amino acid tail to the C-terminus of CD83 (FIG. 5).
  • Additional Functional Data
  • A reduction in CD4+ T cells was seen in peripheral blood, spleen tissues and lymph nodes from homozygous LCD4.1 mice. Although there was a reduced number of CD4+ T cells in the thymus there is no overt block in the developmental process and there was no alteration in B cell development in the bone marrow. Histological evaluation of thymus, spleen and lymph nodes from affected mice revealed no gross alteration in tissue architecture.
  • Dendritic cells can be differentiated from bone marrow of wild type mice by culture in GM-CSF. These cells can be characterized by the surface expression of dendritic cell markers, including CD86 and CD11c. Both LCD4.1 affected and normal animals were capable of giving rise to CD86+CD11+ cells under these culture conditions. LCD4.1 mutant mice thus were capable of generating dendritic cells under in vitro culture conditions. These data suggest that the phenotype seen in LCD4.1 mice is not due to a failure of dendritic cells to develop but rather may reflect a defect in function.
  • To track dendritic cells the sensitizing agent FITC was applied to the dorsal surface of the ears of LCD4.1 affected and wild-type mice. FITC was picked up by dendritic cells that then migrated to the draining auricular lymph nodes, where the presence of the FITC label on the dendritic cell surface permitted detection by flow-cytometry. FITC labeled cells expressing CD86 were detected in equal proportions in draining lymph node from normal and affected LCD4.1 mice. These data indicate that LCD4.1 mutant animals are capable of generating dendritic cells in vivo and that these cells are able to pick up antigen in the ear and travel to the draining lymph node.
  • Example 2 CD83 and CD4+ T Cell Function Materials and Methods
  • Spleens were removed from wild type and mutant mice and digested with collagenase to liberate dendritic cells. Spleens were stained for surface expression of CD4 (helper T cells) and CD11c (dendritic cells). Cells expressing these markers were purified by fluorescence activated cell sorting (FACS sorting). CD11c and CD4+ positive cells were also purified from an allogeneic mouse strain, BALBc.
  • Mixed lymphocyte cultures were set up using purified cell populations. Dendritic cells from BALBc animals were used to stimulate CD4+ T cells from wild type and mutant mice. In a reciprocal experiment dendritic cells prepared from wild type and mutant mice were used to stimulate BALBc CD4+ T cells. After 5 days in culture proliferative responses were measured by incorporation of tritiated thymidine.
  • Dendritic cells from wild type and mutant mice were both capable of activating allogeneic T cells, suggesting that dendritic cell function was unimpaired in the mutant animal (FIG. 6 a). In contrast CD4+ T cells from mutant animals exhibited a diminished response after 5 days of stimulation (FIG. 6 b).
  • These data suggest that the mutation in the CD83 gene has minimal effect on dendritic cells intrinsic function but rather has a profound effect upon T cell activity. The CD4+ T cell therefore may have a novel requirement for CD83 functionality on T cells during allogeneic activation. CD83 may be influencing the extent of CD4+ T cell activation or altering the duration of the CD4+ T cell proliferative response. The therapeutic manipulation of CD83 may thus represent a mechanism for the specific regulation of T cell function in the treatment of T cell mediated diseases, including autoimmune disorders. Antibodies capable of blocking CD83 function may be used as therapeutics in the treatment of immune diseases whilst the activation of CD83 may have utility in enhancing immune responses in cancer and other circumstances.
  • CONCLUSION
  • Although CD83 has been described as a marker of dendritic cell activation there is little data as to its function in vivo. The mutation provided by the invention destabilizes or inactivates the protein and leads to impaired surface expression. As a consequence, CD4+ T cell function is impaired although the development of dendritic cells is not inhibited and mutant dendritic cells retain functionality. This results in the impaired development of CD4+ T cells. This impaired ability to activate T cells is also seen in a slight decrease in contact sensitivity responses in LCD4.1 mutant mice.
  • Example 3 Mutant CD83 have Different Cytokine Levels Than Wild Type Mice
  • This Example demonstrates that CD4+ T-cells from CD83 mutant animals express higher levels of IL-4 and lower levels of IL-2 compared to CD4+ T-cells from CD83 wild type animals.
  • Methods for Cell Activation and Cytokine Measurements:
  • Spleens cells from 6-8-week-old homozygous CD83 wild type or CD83 mutant (LCD4.1) mice were used to isolate CD4+ T-cells by positive selection using magnetic beads (Miltenyi Biotec). A 96 round bottom plate was coated with 50 μL, per well of a solution containing either 1 or 10 μg/mL of anti-CD3 and 0.1 or 0.2 μg/mL of anti-CD28 antibodies (both from Pharmingen) in PBS overnight. This plate was then washed using 150 μL of PBS three times. To this pre-coated plate, 20,000 CD4+ T-cells (either wild type or CD83 mutant) were added in a 200 μL final volume of RPMI containing 10% FBS, 55 μM β-mercaptoethanol and antibiotics. The plates were then incubated in a CO2 incubator at 37° C. for 44 to 72 hours. For determination of cytokine levels, supernatants were harvested and cytokines were measured using either a Cytometric Bead Array system (Pharmingen) or ELISA (R&D). For RNA measurements, the cells were harvested and RNA was isolated using Tri reagent (Sigma). IL-10 and IL-4 mRNA levels were measured by reverse transcription and TaqMan (Applied Biosystems) analysis.
  • Results:
  • FIG. 7 shows the IL-2, IL-4, IL-5, TNFα and IFNγ levels produced by either wild type or CD83 mutant CD4+ T-cells. Purified cells were incubated as described above in the presence of 1 μg/mL of anti-CD3 and 0.2 μg/mL of anti-CD28 antibodies for 72 hours. The supernatants were then simultaneously analyzed for production of IL-2, IL-4, IL-5, TNFα and IFNγ using the cytometric bead array system from Pharmingen.
  • FIG. 7 demonstrates that CD4+ T-cells from CD83 mutant animals expressed higher levels of IL-4 and lower levels of IL-2 compared to CD4+ T-cells from CD83 wild type animals. Other cytokines and a new set of stimulation assays were analyzed including the production levels of IL-10 and GMCSF by these cells (FIGS. 8 and 9). In both cases, cells from mutant animals produce larger amounts of IL-10 and GMCSF than did wild type animals. FIG. 10 shows that mRNA levels for both IL-4 and IL-10 were increased in cells from activated mutant CD83, CD4+ T-cells compared with cells from wild type animals.
  • Example 4 Anti-CD83 Antibodies May Mimic The Effects of the CD83 Mutation Methods for Antibody Testing:
  • For modulation of cytokine production by anti-CD83 antibodies, CD4+ T-cells were isolated and activated as mentioned above in the presence of increasing concentrations of anti-CD83 antibodies. For proliferation assays, CD4+ T-cells were isolated from an OT2tg [transgenic mice with a T-cell receptor specific for chicken ovalbumin (OVA) 323-339 peptide]. Dendritic cells were isolated from a C57BL6 mouse by a negative selection using B220 magnetic beads (Miltenyi Biotec) followed by positive selection using CD11-c magnetic beads (Milteny Biotec). Five thousand CD4+ T-cells were then mixed with five thousand dendritic cells in a 96 well plate in the presences of 1 μM OVA peptide using RPMI (55 μM BME, 10% FBS plus antibiotics) in a final 200 uL volume. These cells were then incubated for 48 to 72 hours in a CO2 incubator at 37° C. and pulsed using [3H] thymidine for 8 hours. Cells were then harvested and [3H] thymidine incorporation was quantified using a top counter.
  • Results:
  • In some assays, anti-CD83 antibodies decreased production of IL-4 by activated CD4+ T-cells in a dose dependent manner. Different antibody preparations did provide somewhat different degrees of inhibition of IL-4 production (FIG. 11). Accordingly, the epitope and/or degree of affinity of the antibodies for the CD83 antigen may influence whether or not IL-4 production is significantly inhibited.
  • The effects of anti CD83 antibodies on proliferation of a peptide specific T-cell proliferation assay using the OT2 T-cell receptor (TCR) transgenic system were also observed. CD4+ T-cells derived from these TCR transgenic animals express high levels of a T-cell receptor specific for chicken ovalbumin (OVA) 323-339 peptide and thus have high levels of proliferation when mixed with antigen presenting cells (dendritic cells were used) in the presence of the OVA peptide. In such assays, anti-CD83 antibodies were able to decrease proliferation of CD4+ T-cells in this system (FIG. 12). However, different antibody preparations had somewhat different effects on the proliferation of CD4+ T-cells. Accordingly, the CD83 epitope and/or degree of affinity of the antibodies for the CD83 antigen may influence whether or not CD4+ T-cell proliferation is significantly inhibited.
  • Example 5 Increased T-Cell Proliferation By Transgenic Expression of CD83
  • This Example illustrates that over expression of CD83 in transgenic mice leads to increased T-cell proliferation.
  • Materials and Methods
  • A 34.3 kb fragment of normal mouse genomic DNA, including the ˜18 kb coding region of the CD83 gene, as well as ˜10.6 kb of upstream flanking sequences and ˜5.7 kb of downstream sequences was microinjected into normal mouse one-cell embryos. Four individual founder animals were generated. Transgenic mice were then crossed to a male OT2tg mouse. Male offspring carrying both the CD83 and OT2 transgene were used to analyze peptide specific T-cell proliferation.
  • For proliferation assays, CD4+ T-cells and dendritic cells were isolated from either OT2tg [transgenic mice with a T-cell receptor specific for chicken ovalbumin (OVA) 323-339 peptide] CD83 wild type or from OT2tg CD83 transgenic mice as described above (Example 4). Five thousand OT2tg CD4+ T-cells from either wild type or CD83 transgenic animals were then mixed with five thousand wild type dendritic cells or five thousand CD83 transgenic dendritic cells in a 96 well plate in the presence of increasing concentrations of OVA peptide using RPMI (55 μM BME, 10% FBS plus antibiotics) in a final 200 uL volume. These cells were then incubated for 48 to 72 hours in a CO2 incubator at 37 C and pulsed using [3H] thymidine for 8 hours. Cells were then harvested and [3H] thymidine incorporation was quantified using a top counter.
  • Results:
  • OT2tg CD4+ T-cells derived from CD83 transgenic mice proliferated at higher rates than the same cell population derived from a CD83 wild type animal (FIG. 13). This increased proliferation was seen at all the concentrations of OVA peptide tested. Whereas OT2tg CD4+ T-cells derived from CD83 transgenic animals exhibited increased proliferation, dendritic cells from CD83 transgenic animals did not exhibit a substantial increase in proliferation. Therefore, it appears that transgenic expression in the CD4+ T-cell, and not in dendritic cells is what led to the increased proliferation of CD4+ T-cells.
  • Example 6 Inhibition of Proliferation of PHA Activated Human PBMCs By Protein A Purified Rabbit Anti Mouse CD83 Polyclonal Sera
  • This Example shows that antibodies raised against the mouse CD83 protein can inhibit proliferation of human peripheral blood mononuclear cells.
  • Materials and Methods
  • Rabbit polyclonal sera was raised against mouse CD83 protein by immunizing rabbits using a mouse CD83 external domain protein fused to a rabbit Ig domain (FIG. 14). Pre-immune sera and anti-mouse polyclonal sera were then purified using a protein A column (Pharmacia Biotech) as described by the manufacturer, then dialyzed against PBS and stored at 4° C. To monitor the recognition of mouse CD83 protein by the polyclonal sera, which was obtained at different dates post immunization, a titer was obtained using an antigen specific ELISA (FIG. 15). As illustrated by FIG. 15, a good polyclonal response was obtained against the mouse CD83 protein.
  • Human peripheral blood mononuclear cells (PBMCs) were isolated using a Ficoll gradient (Ficoll Paque Plus, Pharmacia) and washed with PBS buffer. For activation and proliferation studies, five thousand cells were incubated in 200 μL of media (RPMI, 10% FBS, antibiotics) and 5 ug/mL of Phaseolus vulgaris leucoagglutinin (PHA) in the presence or absence of increasing concentrations of Protein A purified pre-immune sera or with similarly purified anti-CD83 polyclonal antibodies. After 48 hours at 37° C. in a CO2 incubator the cells were pulsed with [3H] thymidine for ˜8 hours and harvested. Thymidine incorporation into the PBMCs was measured using a top counter for analysis.
  • Results
  • FIG. 16 illustrates that proliferation of PHA-activated human PBMCs was inhibited by antibodies raised against the external region of the mouse CD83 protein. Proliferation of PHA-activated human PBMCs was not affected by addition of increasing concentrations of protein A purified rabbit pre-immune sera. When increasing concentrations of protein A purified rabbit polyclonal sera raised against the mouse CD83 protein was added, a concentration dependent decrease in proliferation was observed.
  • These data indicate that antibodies raised against the mouse protein are able to cross-react with the human protein. Moreover, antibodies raised against the mouse protein are able to inhibit proliferation of PHA-activated human PBMCs.
  • All publications, patents and patent applications are incorporated herein by reference. While in the foregoing specification this invention has been described in relation to certain preferred embodiments thereof, and many details have been set forth for purposes of illustration, it will be apparent to those skilled in the art that the invention is susceptible to additional embodiments and that certain of the details described herein may be varied considerably without departing from the basic principles of the invention.

Claims (31)

1-62. (canceled)
63. An antibody that can bind to a CD83 polypeptide of SEQ ID NO:9, wherein activated CD4+ T-cells produce lower levels of interleukin-4 when said T-cells are contacted with the antibody, wherein said antibody comprises a light chain and a heavy chain, and wherein the light chain sequence comprises SEQ ID NO:57.
64. The antibody of claim 63, wherein the heavy chain variable region CDR1 of the antibody comprises SEQ ID NO:38.
65. The antibody of claim 63, wherein the heavy chain variable region CDR2 of the antibody comprises SEQ ID NO:42.
66. The antibody of claim 63, wherein the heavy chain variable region CDR3 of the antibody comprises SEQ ID NO:46.
67. The antibody of claim 63, wherein the heavy chain variable region CDR1 of the antibody comprises SEQ ID NO:38, the heavy chain variable region CDR2 of the antibody comprises SEQ ID NO:42 and the heavy chain variable region CDR3 of the antibody comprises SEQ ID NO:46.
68. An antibody that can bind to a CD83 polypeptide of SEQ ID NO:9, wherein activated CD4+ T-cells produce lower levels of interleukin-4 when said T-cells are contacted with the antibody, wherein said antibody comprises a light chain and a heavy chain, and wherein the heavy chain sequence comprises SEQ ID NO:54.
69. The antibody of claim 68, wherein the light chain variable region CDR1 of the antibody comprises SEQ ID NO:24.
70. The antibody of claim 68, wherein the light chain variable region CDR2 of the antibody comprises SEQ ID NO:27.
71. The antibody of claim 68, wherein the light chain variable region CDR3 of the antibody comprises SEQ ID NO:34.
72. The antibody of claim 68, wherein the light chain variable region CDR1 of the antibody comprises SEQ ID NO:24, the light chain variable region CDR2 of the antibody comprises SEQ ID NO:27 and the light chain variable region CDR3 of the antibody comprises SEQ ID NO:34.
73. An antibody that can bind to a CD83 polypeptide of SEQ ID NO:9, wherein CD4+ T-cell proliferation is decreased when said T-cells are contacted with the antibody, wherein said antibody comprises a light chain and a heavy chain, and wherein the antibody comprises a light chain sequence set forth in SEQ ID NO:57.
74. The antibody of claim 73, wherein the heavy chain variable region CDR1 of the antibody comprises SEQ ID NO:38.
75. The antibody of claim 73, wherein the heavy chain variable region CDR2 of the antibody comprises SEQ ID NO:42.
76. The antibody of claim 73, wherein the heavy chain variable region CDR3 of the antibody comprises SEQ ID NO:46.
77. The antibody of claim 73, wherein the heavy chain variable region CDR1 of the antibody comprises SEQ ID NO:38, the heavy chain variable region CDR2 of the antibody comprises SEQ ID NO:42 and the heavy chain variable region CDR3 of the antibody comprises SEQ ID NO:46.
78. An antibody that can bind to a CD83 polypeptide of SEQ ID NO:9, wherein CD4+ T-cell proliferation is decreased when said T-cells are contacted with the antibody, wherein said antibody comprises a light chain and a heavy chain, and wherein the antibody comprises a heavy chain sequence set forth in SEQ ID NO:54.
79. The antibody of claim 78, wherein the light chain variable region CDR1 of the antibody comprises SEQ ID NO:24.
80. The antibody of claim 78, wherein the light chain variable region CDR2 of the antibody comprises SEQ ID NO:27.
81. The antibody of claim 78, wherein the light chain variable region CDR3 of the antibody comprises SEQ ID NO:34.
82. The antibody of claim 78, wherein the light chain variable region CDR1 of the antibody comprises SEQ ID NO:24, the light chain variable region CDR2 of the antibody comprises SEQ ID NO:27 and the light chain variable region CDR3 of the antibody comprises SEQ ID NO:34.
83. An antibody that decreases the activity or expression of a CD83 polypeptide of SEQ ID NO:9, wherein said antibody comprises a light chain and a heavy chain, and wherein the antibody comprises the light chain sequence set forth in SEQ ID NO:57.
84. The antibody of claim 83, wherein the heavy chain variable region CDR1 of the antibody comprises SEQ ID NO:38.
85. The antibody of claim 83, wherein the heavy chain variable region CDR2 of the antibody comprises SEQ ID NO:42.
86. The antibody of claim 83, wherein the heavy chain variable region CDR3 of the antibody comprises SEQ ID NO:46.
87. The antibody of claim 83, wherein the heavy chain variable region CDR1 of the antibody comprises SEQ ID NO:38, the heavy chain variable region CDR2 of the antibody comprises SEQ ID NO:42 and the heavy chain variable region CDR3 of the antibody comprises SEQ ID NO:46.
88. An antibody that decreases the activity or expression of a CD83 polypeptide of SEQ ID NO:9, wherein said antibody comprises a light chain and a heavy chain, and wherein the antibody comprises the heavy chain sequence set forth in SEQ ID NO:57.
89. The antibody of claim 88, wherein the light chain variable region CDR1 of the antibody comprises SEQ ID NO:24.
90. The antibody of claim 88, wherein the light chain variable region CDR2 of the antibody comprises SEQ ID NO:27.
91. The antibody of claim 88, wherein the light chain variable region CDR3 of the antibody comprises SEQ ID NO:34.
92. The antibody of claim 88, wherein the light chain variable region CDR1 of the antibody comprises SEQ ID NO:24, the light chain variable region CDR2 of the antibody comprises SEQ ID NO:27 and the light chain variable region CDR3 of the antibody comprises SEQ ID NO:34.
US12/941,689 2001-11-21 2010-11-08 Manipulation of Cytokine Levels Using CD83 Gene Products Abandoned US20110144309A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/941,689 US20110144309A1 (en) 2001-11-21 2010-11-08 Manipulation of Cytokine Levels Using CD83 Gene Products

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US33195801P 2001-11-21 2001-11-21
PCT/US2002/037738 WO2003045318A2 (en) 2001-11-21 2002-11-21 Manipulation of cytokine levels using cd83 gene products
US10/496,284 US7618629B2 (en) 2001-11-21 2002-11-21 Manipulation of cytokine levels using CD83 gene products
US12/572,592 US7850968B2 (en) 2001-11-21 2009-10-02 Manipulation of cytokine levels using CD83 gene products
US12/941,689 US20110144309A1 (en) 2001-11-21 2010-11-08 Manipulation of Cytokine Levels Using CD83 Gene Products

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US12/572,592 Continuation US7850968B2 (en) 2001-11-21 2009-10-02 Manipulation of cytokine levels using CD83 gene products

Publications (1)

Publication Number Publication Date
US20110144309A1 true US20110144309A1 (en) 2011-06-16

Family

ID=23296083

Family Applications (3)

Application Number Title Priority Date Filing Date
US10/496,284 Expired - Fee Related US7618629B2 (en) 2001-11-21 2002-11-21 Manipulation of cytokine levels using CD83 gene products
US12/572,592 Expired - Fee Related US7850968B2 (en) 2001-11-21 2009-10-02 Manipulation of cytokine levels using CD83 gene products
US12/941,689 Abandoned US20110144309A1 (en) 2001-11-21 2010-11-08 Manipulation of Cytokine Levels Using CD83 Gene Products

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US10/496,284 Expired - Fee Related US7618629B2 (en) 2001-11-21 2002-11-21 Manipulation of cytokine levels using CD83 gene products
US12/572,592 Expired - Fee Related US7850968B2 (en) 2001-11-21 2009-10-02 Manipulation of cytokine levels using CD83 gene products

Country Status (6)

Country Link
US (3) US7618629B2 (en)
EP (1) EP1480598A4 (en)
JP (1) JP2005519586A (en)
AU (1) AU2002357759A1 (en)
CA (1) CA2466845A1 (en)
WO (1) WO2003045318A2 (en)

Families Citing this family (36)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9012141B2 (en) 2000-03-27 2015-04-21 Advaxis, Inc. Compositions and methods comprising KLK3 of FOLH1 antigen
US20040185040A1 (en) 2001-11-21 2004-09-23 Celltech R & D Limited Modulating immune responses
AU2002950779A0 (en) * 2002-08-15 2002-09-12 The Corporation Of The Trustees Of The Order Of The Sisters Of Mercy In Queensland A method of immunomodulation
EP1422241A1 (en) 2002-11-19 2004-05-26 Alexander Steinkasserer Use of soluble forms of CD83 and nucleic acids encoding them for the treatment or prevention of diseases
AU2003300817A1 (en) * 2002-11-21 2004-06-18 Celltech R And D, Inc. Modulating immune responses
US9102726B2 (en) 2002-12-04 2015-08-11 Argos Therapeutics, Inc. Nucleic acid of recombination expression vector encoding soluble forms of CD83, host cells transformed/transfected therewith and pharmaceutical compositions containing same
US7169898B2 (en) 2002-12-04 2007-01-30 Alexander Steinkasserer Soluble CD83 proteins and use thereof for the treatment or prevention of a disease or medical condition caused by dysfunction or undesired function of a cellular immune response involving T cells
US8227437B2 (en) * 2006-06-22 2012-07-24 Tai June Yoo Restoration of hearing loss
WO2008098787A2 (en) * 2007-02-16 2008-08-21 Helmholtz-Zentrum für Infektionsforschung GmbH Cd83 as a molecular switch for the induction of regulatory (immunosuppressive) t-cells
US7906117B2 (en) 2007-05-21 2011-03-15 Alderbio Holdings Llc Antagonists of IL-6 to prevent or treat cachexia, weakness, fatigue, and/or fever
TW201531484A (en) * 2007-05-21 2015-08-16 Alder Biopharmaceuticals Inc Antibodies to TNF alpha and use thereof
US8062864B2 (en) 2007-05-21 2011-11-22 Alderbio Holdings Llc Nucleic acids encoding antibodies to IL-6, and recombinant production of anti-IL-6 antibodies
TWI561530B (en) 2007-05-21 2016-12-11 Alderbio Holdings Llc Antibodies to il-6 and use thereof
US8178101B2 (en) 2007-05-21 2012-05-15 Alderbio Holdings Inc. Use of anti-IL-6 antibodies having specific binding properties to treat cachexia
US8252286B2 (en) 2007-05-21 2012-08-28 Alderbio Holdings Llc Antagonists of IL-6 to prevent or treat thrombosis
US8404235B2 (en) 2007-05-21 2013-03-26 Alderbio Holdings Llc Antagonists of IL-6 to raise albumin and/or lower CRP
US9701747B2 (en) 2007-05-21 2017-07-11 Alderbio Holdings Llc Method of improving patient survivability and quality of life by anti-IL-6 antibody administration
EP2254998B1 (en) * 2008-02-28 2015-12-23 Argos Therapeutics, Inc. Transient expression of immunomodulatory polypeptides for the prevention and treatment of autoimmune disease, allergy and transplant rejection
US9212223B2 (en) 2008-11-25 2015-12-15 Alderbio Holdings Llc Antagonists of IL-6 to prevent or treat thrombosis
US8420089B2 (en) 2008-11-25 2013-04-16 Alderbio Holdings Llc Antagonists of IL-6 to raise albumin and/or lower CRP
US8992920B2 (en) 2008-11-25 2015-03-31 Alderbio Holdings Llc Anti-IL-6 antibodies for the treatment of arthritis
US9452227B2 (en) 2008-11-25 2016-09-27 Alderbio Holdings Llc Methods of treating or diagnosing conditions associated with elevated IL-6 using anti-IL-6 antibodies or fragments
US8323649B2 (en) 2008-11-25 2012-12-04 Alderbio Holdings Llc Antibodies to IL-6 and use thereof
US8337847B2 (en) 2008-11-25 2012-12-25 Alderbio Holdings Llc Methods of treating anemia using anti-IL-6 antibodies
US10016617B2 (en) 2009-11-11 2018-07-10 The Trustees Of The University Of Pennsylvania Combination immuno therapy and radiotherapy for the treatment of Her-2-positive cancers
EP2504031A4 (en) 2009-11-24 2013-06-26 Alderbio Holdings Llc Antibodies to il-6 and use thereof
US9775921B2 (en) 2009-11-24 2017-10-03 Alderbio Holdings Llc Subcutaneously administrable composition containing anti-IL-6 antibody
US9226958B2 (en) 2010-10-01 2016-01-05 University Of Georgia Research Foundation, Inc. Use of Listeria vaccine vectors to reverse vaccine unresponsiveness in parasitically infected individuals
CA2818813C (en) 2010-11-23 2020-10-06 Alder Biopharmaceuticals, Inc. Anti-il-6 antibodies for the treatment of oral mucositis
AU2012229218B2 (en) 2011-03-11 2017-03-02 Advaxis, Inc. Listeria-based adjuvants
JP2013150592A (en) * 2011-07-01 2013-08-08 Genentech Inc Use of anti-cd83 agonist antibody for treating autoimmune disease
JP2013040160A (en) 2011-07-01 2013-02-28 Genentech Inc Use of anti-cd83 agonist antibody for treating autoimmune disease
KR20140134695A (en) 2012-03-12 2014-11-24 어드박시스, 인크. Suppressor cell function inhibition following listeria vaccine treatment
DK2951208T3 (en) 2013-02-01 2020-01-13 Kira Biotech Pty Ltd ANTI-CD83 ANTIBODIES AND USE THEREOF
WO2015120097A2 (en) * 2014-02-04 2015-08-13 Contrafect Corporation Antibodies useful in passive influenza immunization, and compositions, combinations and methods for use thereof
EP3209695A4 (en) 2014-10-23 2018-05-30 DendroCyte BioTech Pty Ltd Cd83 binding proteins and uses thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5316920A (en) * 1992-04-17 1994-05-31 Dana-Faber Cancer Institute, Inc. Lymphocyte activation antigen HB15, a member of the immunoglobulin superfamily
US5766570A (en) * 1992-04-17 1998-06-16 Dana-Farber Cancer Institute, Inc. Lymphocyte activation antigens and antibodies thereto
US6900016B1 (en) * 2000-09-08 2005-05-31 Applera Corporation Polymorphisms in known genes associated with inflammatory autoimmune disease, methods of detection and uses thereof

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5316320A (en) * 1992-03-18 1994-05-31 Corrosion Control Corp. Isolation gasket for critical service flow line applications
WO1997029781A1 (en) * 1996-02-15 1997-08-21 Immunex Corporation Methods and compositions for modulating an immune response
US20030165479A1 (en) * 2001-03-19 2003-09-04 Velleca Mark A. Methods for isolating proteins expressed by dendritic cells
US20050042751A1 (en) * 2001-10-31 2005-02-24 Michel Goldman Generation and use of new types of dendritic cells
AR039067A1 (en) * 2001-11-09 2005-02-09 Pfizer Prod Inc ANTIBODIES FOR CD40

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5316920A (en) * 1992-04-17 1994-05-31 Dana-Faber Cancer Institute, Inc. Lymphocyte activation antigen HB15, a member of the immunoglobulin superfamily
US5766570A (en) * 1992-04-17 1998-06-16 Dana-Farber Cancer Institute, Inc. Lymphocyte activation antigens and antibodies thereto
US6068984A (en) * 1992-04-17 2000-05-30 Dana-Farber Cancer Institute, Inc. Antibodies to lymphocyte activation antigens and uses therefor
US6900016B1 (en) * 2000-09-08 2005-05-31 Applera Corporation Polymorphisms in known genes associated with inflammatory autoimmune disease, methods of detection and uses thereof

Also Published As

Publication number Publication date
US7618629B2 (en) 2009-11-17
EP1480598A4 (en) 2006-03-22
AU2002357759A8 (en) 2003-06-10
US20060083740A1 (en) 2006-04-20
US20100081797A1 (en) 2010-04-01
US7850968B2 (en) 2010-12-14
CA2466845A1 (en) 2003-06-05
WO2003045318A2 (en) 2003-06-05
WO2003045318A3 (en) 2004-09-16
EP1480598A2 (en) 2004-12-01
AU2002357759A1 (en) 2003-06-10
JP2005519586A (en) 2005-07-07

Similar Documents

Publication Publication Date Title
US7850968B2 (en) Manipulation of cytokine levels using CD83 gene products
US7700740B2 (en) Antibodies to CD83
JP5597793B2 (en) ILT3-binding molecules and uses thereof
EP1572976A2 (en) Modulating immune responses
JP3523245B1 (en) Transgenic chromosome-introduced rodents for the production of human antibodies
KR101700073B1 (en) Method of providing disease-specific binding molecules and targets
KR20220053587A (en) Cell surface receptors responsive to loss of heterozygosity
JP2009530423A (en) Methods for preventing and treating amyloidogenic diseases
HUE028830T2 (en) Human anti-b7rp1 neutralizing antibodies
HU228575B1 (en) Remedies for immunological diseases
WO2013025779A1 (en) Anti-b7-h4 antibodies and their uses
JPH11508764A (en) Monkey monoclonal antibodies specific for human B7.1 and / or B7.2, primatized forms thereof, pharmaceutical compositions
WO2007098283A2 (en) Use of antagonists of maif receptor complex molecules and neurotrophic factors for treatment of neurological diseases and disorders
KR20210142638A (en) CD3 antigen-binding fragment and applications thereof
CN111848809A (en) CAR molecule targeting Claudin18.2, immune cell modified by same and application
US20030215450A1 (en) Anti-rank ligand monoclonal antibodies useful in treatment of rank ligand mediated disorders
WO2019220110A1 (en) A cd79-specific chimeric antigen receptor
KR20230052289A (en) Compositions and methods for treating EGFR positive cancer
US20040213788A1 (en) Anti-rank ligand monoclonal antibodies useful in treatment of rank ligand mediated disorders
JP2005515752A6 (en) Anti-RANK ligand monoclonal antibodies useful in the treatment of RANK ligand-mediated disorders
JP2005515752A (en) Anti-RANK ligand monoclonal antibodies useful in the treatment of RANK ligand-mediated disorders
US20230159651A1 (en) Bcma targetting antibodies, chimeric antigen receptors, and uses thereof
WO2023019393A1 (en) Cd123-targetting antibodies, chimeric antigen receptors, and uses thereof

Legal Events

Date Code Title Description
AS Assignment

Owner name: CELLTECH R & D, INC., UNITED KINGDOM

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:APPLEBY, MARK W;PROLL, SEAN;PAEPER, BRYAN;AND OTHERS;SIGNING DATES FROM 20040621 TO 20040923;REEL/FRAME:026886/0397

AS Assignment

Owner name: CELLTECH R & D, INC., WASHINGTON

Free format text: CORRECTIVE ASSIGNMENT TO CORRECT THE NAMES OF ASSIGNEES AND EXECUTION DATE OF ASSIGNMENT PREVIOUSLY RECORDED ON REEL 026886, FRAME 0397;ASSIGNORS:RAMSDELL, FRED;PROLL, SEAN C;HAMPTON, KAREN STAEHLING;AND OTHERS;SIGNING DATES FROM 20030211 TO 20030218;REEL/FRAME:028126/0043

STCB Information on status: application discontinuation

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