EP1418936A2 - Compositions pharmaceutiques destines a prevenir ou traiter les maladies associees aux cellules th1 et th2 par modulation du rapport th1/th2 - Google Patents

Compositions pharmaceutiques destines a prevenir ou traiter les maladies associees aux cellules th1 et th2 par modulation du rapport th1/th2

Info

Publication number
EP1418936A2
EP1418936A2 EP02724144A EP02724144A EP1418936A2 EP 1418936 A2 EP1418936 A2 EP 1418936A2 EP 02724144 A EP02724144 A EP 02724144A EP 02724144 A EP02724144 A EP 02724144A EP 1418936 A2 EP1418936 A2 EP 1418936A2
Authority
EP
European Patent Office
Prior art keywords
sdf
stimulant
zap
syk
cell
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP02724144A
Other languages
German (de)
English (en)
Inventor
Tan Jinquan
Lars Kaergaard Poulsen
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.)
ALK Abello AS
Original Assignee
ALK Abello AS
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 ALK Abello AS filed Critical ALK Abello AS
Publication of EP1418936A2 publication Critical patent/EP1418936A2/fr
Withdrawn legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/24Heavy metals; Compounds thereof
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/48Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving transferase
    • C12Q1/485Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving transferase involving kinase
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/7125Nucleic acids or oligonucleotides having modified internucleoside linkage, i.e. other than 3'-5' phosphodiesters
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/06Aluminium, calcium or magnesium; Compounds thereof, e.g. clay
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/42Phosphorus; Compounds thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/1703Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • A61K38/1709Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/177Receptors; Cell surface antigens; Cell surface determinants
    • A61K38/1793Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/195Chemokines, e.g. RANTES
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/20Interleukins [IL]
    • A61K38/2013IL-2
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/20Interleukins [IL]
    • A61K38/2026IL-4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/45Transferases (2)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/39Medicinal preparations containing antigens or antibodies characterised by the immunostimulating additives, e.g. chemical adjuvants
    • 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
    • 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
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0636T lymphocytes
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5044Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics involving specific cell types
    • G01N33/5047Cells of the immune system
    • G01N33/505Cells of the immune system involving T-cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/515Animal cells
    • A61K2039/5158Antigen-pulsed cells, e.g. T-cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55516Proteins; Peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55522Cytokines; Lymphokines; Interferons
    • A61K2039/55527Interleukins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55522Cytokines; Lymphokines; Interferons
    • A61K2039/55527Interleukins
    • A61K2039/55533IL-2
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/57Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/23Interleukins [IL]

Definitions

  • the present invention relates to a pharmaceutical composition for preventing or treating a Th1 cell-related disease in a human or an animal by reducing the Th1/Th2 ratio.
  • the present invention relates to pharmaceutical composition for preventing or treating a Th2 cell-related disease in a human or an animal by increasing the Th1/Th2 ratio.
  • T helper cells differentiate into either Th1 or Th2 T cells depending upon antigen stimulation and cytokine environment. So far, it is not very clear whether instructive or/and selective differentiation may contribute to Th cell formation in vivo. T helper cells cells first activated by antigen in the presence of IL-12 develop predominantly into Th1 cells, whereas those activated in the presence of IL-4 develop predominantly into Th2 cells (Coffman et al., 1999). It is believed that progenitor Th cells require individual cellular divisions before becoming competent to synthesize cytokines indicative of either the Th1 or Th2 pathway (Bird et al., 1998).
  • Syk-family including Syk and ZAP-70 tyrosine kinases are essential for lymphocyte development and activation.
  • Syk and ZAP-70 tyrosine kinases are essential for lymphocyte development and activation.
  • these two kinases are differentially regulated, both in terms of expression and activity (Chu et al., 1998).
  • Th1 and Th2 cell phenotypes are different from each other in early activation signal transduction pathways, especially in the role of TCR related protein tyrosine kinases (Tamura et al., 1995).
  • TCR and its downstream protein tyrosine kinases such as Fyn, p56(lck), and ZAP-70 are involved in the development and differentiation of Th1 and Th2 cells (Swith et al., 1998; Faith et al., 1997; al-Ramadi et al., 1996; Deckert et al., 1998).
  • Cbl is an adaptor protein that functions as a negative regulator of many signaling pathways such as Syk family that start from receptors at the cell surface (Meng et al., 1999; Ota et al., 1997).
  • Cbl-b a homologue of Cbl, has a positive role in T-cell signaling via a direct interaction with the upstream kinase ZAP-70 (Zhang et al., 1999).
  • CXCR4 the only known receptor for SDF-1 ⁇ , can cross-talk with TCR to affect anti-CD3-stimulated phosphorylation of critical downstream substrates of TCR signaling including ZAP-70 (Peacock et al., 1999).
  • ZAP-70 the only known receptor for SDF-1 ⁇
  • Jourdan et al., 1998 discloses the finding that IL-4 specifically enhances cell surface expression of CXCR4 in Th2 cells and also in Th1 cells.
  • the CXCR4 ligand SDF-1 ⁇ activates the p42 MAP-kinase ERK-2. Such activation renders T cells susceptible to HIV infection and promotes SDF-1 ⁇ induced migration of the cells.
  • the activity of p42 MAP-kinase was measured in order to examine whether the IL-4 induced CXCR4 was functional.
  • the activity of ZAP-70 tyrosine kinase was measured as a measure of the global level of signalling proteins.
  • Th2 responses are enhanced in NFAT1 knock-out mice (Hodge et al. 1996; Viola et al. 1998) and Th2 responses are impaired in NFAT2 knock-out mice (Ranger et al. 1998; Yoshida et al. 1998).
  • NFATp NFAT1
  • NFAT4 in combination act as selective repressors of Th2 cells.
  • the document describes a) mice deficient in both NFATp and NFAT4 that exhibit a phenotype characteristic of increased Th2 cell activity, b) methods of identifying modulators of Th2 cell activity, using either cells deficient in both
  • NFAT p and NFAT4 mice deficient in both NFATp and NFAT4 or indicator compositions containing both NFATp and NFAT4, c) methods of regulating Th2 cell activity using agents that modulate the activity of NFATp and NFAT4, and d) methods of diagnosing disorders associated with aberrant Th2 cell activity by assessing changes in NFATp and/or NFAT4 expression.
  • US-A-5 958 671 discloses methods of modulating production of a Th2- associated cytokine, in particular interleukin-4 (IL-4), by modulating the activity of c-Maf and methods of modulating development of Th1 and Th2 subsets in a subject using agents that modulate transcription factor activity.
  • IL-4 interleukin-4
  • the document describes a method of stimulating IL-4 production by using a first agent to stimulate the expression or activity of c-Maf in combination with a second agent that stimulates the expression or activity of any member of the NFAT family, preferably NFATp.
  • the document describes a method of inhibiting IL-4 production by using a first agent to inhibit the expression or activity of a maf family protein in combination with a second agent that inhibits the expression or activity of any member of the NFAT family.
  • the objective of the invention is to provide therapeutic substances for treating Th1 and Th2 cell-related diseases.
  • the objective of the invention is obtained with the first aspect of the invention, which relates to a pharmaceutical composition for preventing or treating a Th1 cell-related disease in a human or an animal by reducing the Th1/Th2 ratio comprising an active substance selected from the group consisting of a) IL-4 and SDF-1 ⁇ , b) a stimulant of IL-4 and a stimulant of SDF-1 ⁇ , c) an antagonist to IL-2 and an antagonist to SDF-1 ⁇ , d) an inhibitor of Syk or NFAT1 , e) a stimulant of ZAP-70 or NFAT2, f) an IL-4 stimulating adjuvant and SDF-1 ⁇ , g) a functional derivative, analogue or part of any of the substances a)-f) or h) a combination of any of the substances a)-g).
  • an active substance selected from the group consisting of a) IL-4 and SDF-1 ⁇ , b) a stimulant of IL-4 and a stimulant of SDF-1 ⁇ ,
  • the object of the invention is obtained with the second aspect of the invention, which relates to pharmaceutical composition for preventing or treating a Th2 cell-related disease in a human or an animal by increasing the Th1/Th2 ratio comprising an active substance selected from the group consisting of o) IL-2 and SDF-1 ⁇ , p) a stimulant of IL-2 and a stimulant of SDF-1 ⁇ , q) an antagonist to IL-4 and an antagonist to SDF-1 ⁇ , r) an inhibitor of ZAP-70 or NFAT2, s) a stimulant of Syk or NFAT1 , t) an IL-2 stimulating adjuvant and SDF-1 ⁇ , u) a functional derivative, analogue or part of any of the substances o)-t) or v) a combination of any of the substances o)-u).
  • an active substance selected from the group consisting of o) IL-2 and SDF-1 ⁇ , p) a stimulant of IL-2 and a stimulant of SDF-1 ⁇ , q) an
  • the present invention is based on the discovery of several steps in the mechanisms, which determine the differentiation of progenitor T cells into Th1 and Th2 cells, both at the extracellular and the infracellular level. Firstly, the present invention is based on the discovery that stimulation of a progenitor T cell with IL-2 or an IL-2 stimulating adjuvant in conjunction with SDF-1 ⁇ causes the CD4 + T cell to develop into a Th1 cell.
  • IL-2, IL-4, adjuvants and SDF-1 ⁇ bind to their respective receptors on the surface of the progenitor T cells and trigger specific infracellular regulatory pathways leading to the said differentiation into either a Th1 or a Th2 cell.
  • the present invention is based on the discovery that the pathway leading to differentiation of a T cell into a Th1 cell involves the activation of the tyrosine kinase Syk as well as the transcription factor NFAT1 , whereas the other NFAT transcription factors are not activated.
  • the pathway leading to differentiation of a T cell into a Th2 cell involves the activation of the tyrosine kinase ZAP-70 as well as the transcription factor NFAT2, whereas the other NFAT transcription factors are not activated.
  • the present invention is further based on the recognition that any substance capable of interfering with the function of any of substances found to be involved in the said T cell differentiation may be used as an active substance for preventing or treating diseases, which are mediated by either Th1 or Th2 cells.
  • a Th1 cell-related disease i.e. a disease in which Th1 cells support the disease process
  • Th2 cell-related disease i.e. a disease in which Th2 cells support the disease process
  • Th2 cell-related disease i.e. a disease in which Th2 cells support the disease process, by either inhibiting the differentiation of T cells into Th2 cells, or by stimulating the differentiation of T cells into counteracting Th1 cells.
  • the present invention relates further to the following:
  • a pharmaceutical composition comprising an active substance selected from the group consisting of a) IL-4 and SDF-1 ⁇ , b) a stimulant of IL-4 and a stimulant of SDF-1 ⁇ , c) an antagonist to IL-2 and an antagonist to SDF-1 ⁇ , d) an inhibitor of Syk or NFAT1 , e) a stimulant of ZAP-70 or NFAT2, f) an IL-4 stimulating adjuvant and SDF-1 ⁇ , g) a functional derivative, analogue or part of any of the substances a)-f) or h) a combination of any of the substances a)-g).
  • a pharmaceutical for preventing or treating a Th1 cell-related disease by reducing the Th1/Th2 ratio of a) IL-4 and SDF-1 ⁇ , b) a stimulant of IL-4 and a stimulant of SDF-1 ⁇ , c) an antagonist to IL-2 and an antagonist to SDF-1 ⁇ , d) an inhibitor of Syk or NFAT1 , e) a stimulant of ZAP- 70 or NFAT2, f) an IL-4 stimulating adjuvant and SDF-1 ⁇ , g) a functional derivative, analogue or part of any of the substances a)-f) or h) a combination of any of the substances a)-g).
  • a method of preventing or treating a Th1 cell-related disease by reducing the Th1/Th2 ratio comprising administering to a subject an effective dose of an active substance selected from the group consisting of a) IL-4 and SDF-1 ⁇ , b) a stimulant of IL-4 and a stimulant of SDF-1 ⁇ , c) an antagonist to IL-2 and an antagonist to SDF-1 ⁇ , d) an inhibitor of Syk or NFAT1 , e) a stimulant of ZAP-70 or NFAT2, f) an IL-4 stimulating adjuvant and SDF-1 ⁇ , g) a functional derivative, analogue or part of any of the substances a)-f) or h) a combination of any of the substances a)-g).
  • a method of preventing or treating a Th1 cell-related disease by reducing the Th1/Th2 ratio comprising removing T helper cells from a subject, contacting ex vivo the cells with an effective dose of an active substance selected from the group consisting of a) IL-4 and SDF-1 ⁇ , b) a stimulant of IL-4 and a stimulant of SDF-1 ⁇ , c) an antagonist to IL-2 and an antagonist to SDF-1 ⁇ , d) an inhibitor of Syk or NFAT1 , e) a stimulant of ZAP-70 or NFAT2, f) an IL-4 stimulating adjuvant and SDF-1 ⁇ , g) a functional derivative, analogue or part of any of the substances a)-f) or h) a combination of any of the substances a)-g).
  • an active substance selected from the group consisting of a) IL-4 and SDF-1 ⁇ , b) a stimulant of IL-4 and a stimulant of SDF-1 ⁇ , c) an antagonist
  • a method as mentioned in one of the two previous paragraphs comprising subjecting the subject or recipient to be treated to a second treatment involving the manipulation of the immune system.
  • the said second treatment involving the manipulation of the immune system amy be selected from the group consisting of a vaccination, antigen specific immunotherapy, allergen specific immunotherapy, nonspecific immunotherapy and an organ transplantation.
  • a pharmaceutical composition comprising an active substance selected from the group consisting of o) IL-2 and SDF-1 ⁇ , p) a stimulant of IL-2 and a stimulant of SDF-1 ⁇ , q) an antagonist to IL-4 and an antagonist to SDF-1 ⁇ , r) an inhibitor of ZAP-70 or NFAT2, s) a stimulant of Syk or NFAT1 , t) an IL-2 stimulating adjuvant and SDF-1 ⁇ , u) a functional derivative, analogue or part of any of the substances o)-t) or v) a combination of any of the substances o)-u).
  • a pharmaceutical for preventing or treating a Th2 cell-related disease by increasing the Th1/Th2 ratio of o) IL-2 and SDF-1 ⁇ , p) a stimulant of IL-2 and a stimulant of SDF-1 ⁇ , q) an antagonist to IL-4 and an antagonist to SDF-1 ⁇ , r) an inhibitor of ZAP-70 or NFAT2, s) a stimulant of Syk or NFAT1 , t) an IL-2 stimulating adjuvant and SDF-1 ⁇ , u) a functional derivative, analogue or part of any of the substances o)-t) or v) a combination of any of the substances o)-u).
  • a method of preventing or treating a Th2 cell-related disease by increasing the Th1/Th2 ratio comprising administering to a subject an effective dose of an active substance selected from the group consisting of o) IL-2 and SDF- l ⁇ , p) a stimulant of IL-2 and a stimulant of SDF-1 ⁇ , q) an antagonist to IL-4 and an antagonist to SDF-1 ⁇ , r) an inhibitor of ZAP-70 or NFAT2, s) a stimulant of Syk or NFAT1 , t) an IL-2 stimulating adjuvant and SDF-1 ⁇ , u) a functional derivative, analogue or part of any of the substances o)-t) or v) a combination of any of the substances o)-u).
  • an active substance selected from the group consisting of o) IL-2 and SDF- l ⁇ , p) a stimulant of IL-2 and a stimulant of SDF-1 ⁇ , q) an antagonist to IL-4 and an antagonist to SDF-1
  • a method of preventing or treating a Th2 cell-related disease by increasing the Th1/Th2 ratio comprising removing T helper cells from a subject, contacting ex vivo the cells with an effective dose of an active substance selected from the group consisting of o) IL-2 and SDF-1 ⁇ , p) a stimulant of IL-2 and a stimulant of SDF-1 ⁇ , q) an antagonist to IL-4 and an antagonist to SDF-1 ⁇ , r) an inhibitor of ZAP-70 or NFAT2, s) a stimulant of Syk or NFAT1 , t) an IL-2 stimulating adjuvant and SDF-1 ⁇ , u) a functional derivative, analogue or part of any of the substances o)-t) or v) a combination of any of the substances o)-u).
  • an active substance selected from the group consisting of o) IL-2 and SDF-1 ⁇ , p) a stimulant of IL-2 and a stimulant of SDF-1 ⁇ , q) an antagonist
  • a method as mentioned in one of the two previous paragraphs comprising subjecting the subject or recipient to be treated to a second treatment involving the manipulation of the immune system.
  • the said second treatment involving the manipulation of the immune system may be selected from the group consisting of a vaccination, antigen specific immunotherapy, allergen specific immunotherapy, nonspecific immunotherapy and organ transplantation.
  • An antisense peptide nucleic acid that is complementary to a DNA molecule encoding the tyrosine kinase Syk or a part thereof.
  • the Syk PNA preferably comprises 5-25, more preferablyl 0-20, most preferably 13-18 bases.
  • the Syk PNA has the sequence of SEQ ID NO. 01.
  • An antisense peptide nucleic acid that is complementary to a DNA molecule encoding the tyrosine kinase Syk or a part thereof for preventing or treating a Th1 cell-related disease by reducing the Th1/Th2 ratio.
  • PNA antisense peptide nucleic acid
  • a method of preventing or treating a Th1 cell-related disease by reducing the Th1/Th2 ratio comprising administering to a subject an effective dose of an antisense peptide nucleic acid (PNA) that is complementary to a DNA molecule encoding the tyrosine kinase Syk or a part thereof.
  • PNA antisense peptide nucleic acid
  • An antisense peptide nucleic acid that is complementary to a DNA molecule encoding the tyrosine kinase ZAP-70 or a part thereof.
  • the ZAP-70 PNA preferably comprises 5-25, more preferablyl 0-20, most preferably 13- 18 bases.
  • the ZAP-70 PNA has the sequence of SEQ ID NO. 02.
  • An antisense peptide nucleic acid that is complementary to a DNA molecule encoding the tyrosine kinase ZAP-70 or a part thereof for preventing or treating a Th2 cell-related disease by increasing the Th1/Th2 ratio.
  • PNA antisense peptide nucleic acid
  • a method of preventing or treating a Th2 cell-related disease by increasing the Th1/Th2 ratio comprising administering to a subject an effective dose of an antisense peptide nucleic acid (PNA) that is complementary to a DNA molecule encoding the tyrosine kinase ZAP-70 or a part thereof.
  • PNA antisense peptide nucleic acid
  • An in vitro diagnostic method of evaluating the T helper cell profile of a subject comprising obtaining a T helper cell containing sample from the subject, measuring the level of phosphorylated Syk, phosphorylated ZAP-70, intranucleic NFAT1 and/or intranucleic NFAT2 in the sample and using the measuring results obtained to assess the Th1/Th2 level.
  • An in vitro method of testing the effect of a product or a method on the Th1/Th2 ratio comprising obtaining a T helper cell containing culture with a known Th1/Th2 ratio, subjecting the T helper cells to the product or method, measuring the level of phosphorylated Syk, phosphorylated ZAP-70, intranucleic NFAT1 and/or intranucleic NFAT2 in the sample and using the measuring results obtained to assess the change in Th1/Th2 level.
  • a diagnostic test kit comprising one or more probes specific for binding to phosphorylated Syk, phosphorylated ZAP-70, intranucleic NFAT1 and/or intranucleic NFAT2, and optionally a detection system.
  • a method of producing a culture enriched in Th1 cells comprising obtaining a T helper cell containing sample, subjecting the sample to an active substance selected from the group consisting of a) IL-2 and SDF-1 ⁇ , b) a stimulant of IL-2 and a stimulant of SDF-1 ⁇ , c) an antagonist to IL-4 and an antagonist to SDF-1 ⁇ , d) an inhibitor of ZAP-70 or NFAT2, e) a stimulant of Syk or NFAT1 , f) a functional derivative, analogue or part of any of the substances a)-e) or g) a combination of any of the substances a)-f) to increase the Th1/Th2 ratio.
  • a method of producing a culture enriched in Th2 cells comprising obtaining a T helper cell containing sample, subjecting the sample to an active substance selected from the group consisting of a) IL-4 and SDF-1 ⁇ , b) a stimulant of IL-4 and a stimulant of SDF-1 ⁇ , c) an antagonist to IL-2 and an antagonist to SDF-1 ⁇ , d) an inhibitor of Syk or NFAT1 , e) a stimulant of ZAP-70 or NFAT2, f) a functional derivative, analogue or part of any of the substances a)-e) or g) a combination of any of the substances a)-f) to decrease the Th1/Th2 ratio.
  • Fig. 1A shows the activation of Syk in CB T cells either freshly isolated or stimulated with different combinations among IL-2 (10 ng/ml), IL-4
  • FIG. 1 B shows the activation of ZAP-70 in CB T cells either freshly isolated or stimulated with different combinations among IL-2 (10 ng/ml), IL- 4 (10 ng/ml), and SDF-1 ⁇ (100 ng/ml) as indicated using an immune complex kinase assay (KA) and immunoblotting (IB).
  • Fig. 1 C shows the activation of Cbl in CB T cells either freshly isolated or stimulated with different combinations among IL-2 (10 ng/ml), IL-4
  • Fig. 1 D shows the activation of Cbl-b in CB T cells either freshly isolated or stimulated with different combinations among IL-2 (10 ng/ml), IL-4
  • Fig. 2A shows the blocking effect of PNA Syk antisense on the activation of
  • Fig. 2B shows the blocking effect of PNA ZAP-70 antisense on the activation of ZAP-70 kinase using an immune complex kinase assay (KA) and immunoblotting (IB).
  • Fig. 3A-D show the activation and identification of NFAT in CB T cells upon stimulation with different combinations among IL-2 (10 ng/ml), IL-4
  • Fig. 4 shows a schematic representation of regulatory pathways leading to Th1 and Th2 differentiation.
  • CB switch cord blood
  • IL-2 or IL-4 in combination with SDF-1 ⁇ induce a strong and persistent activation of Syk and ZAP-70 kinase within 8 days in CB CD4 + T cells, respectively.
  • An attenuated pattern or an increased pattern of activity of Cbl kinase has been seen within 8 days in the cells stimulated with IL-2 and SDF-1 ⁇ or with IL-4 and SDF-1 ⁇ .
  • Syk or ZAP-70 PNA antisense selectively inhibit IFN- ⁇ or IL-4 mRNA expression in CB CD4 ⁇ T cells induced by IL-2 or IL-4 combination with SDF-1 ⁇ , implying that Syk and ZAP-70 kinase activation is essential for the Th1 or Th2 cell on-switch by IL-2 or IL-4 in combination with SDF-1 ⁇ .
  • the experimental findings of the present invention is based on the use of cord blood CD4 + T cells, which were considered to be naive cells. It is believed that the findings for naive T cells are also valid for on-going immune responses, i.e. that it is possible to initiate an additional immune response, which is superimposed on the on-going response.
  • Th1 -related diseases Th1 -related diseases
  • the Th1 -related diseases comprise the following groups of diseases: infectious disesases, autoimmune diseases, delayed type hypersensitivity diseases and cancer.
  • the group of infectious diseases includes diseases caused by parasites and viruses, such as HIV.
  • the group of autoimmune diseases include encephalomyelopathic, demyelinating and other autoimune diseases.
  • encephalomyelopathic diseases include, but are not limited to, multiple sclerosis (MS); disseminated sclerosis; focal sclerosis; insular sclerosis; tabes dorsalis (posterior sclerosis); acute and chronic experimental allergic (or autoimmune) encephalomyelitis (EAE), which is an animal model of MS; Guillain-Barre syndrome; experimental allergic neuritis (an animal model of Guillain-Barre syndrome); acute disseminated encephalomyelitis; myalgic encephalomyelitis (benign and epidemic); viral encephalomyelitis; granulomatous encephalomyelitis; etc.
  • MS multiple sclerosis
  • disseminated sclerosis focal sclerosis
  • insular sclerosis tabes dorsalis (posterior sclerosis)
  • EAE acute and chronic experimental allergic (or autoimmune) encephalomyelitis
  • EAE acute and chronic experimental allergic (or autoimmune)
  • animal diseases such as but not limited to canine distemper; feline distemper; equine encephalomyelitis (eastern, Venezuelan, and western); avian encephalomyelitis; porcine encephalomyelitis; bovine encephalomyelitis; mouse encephalomyelitis; etc.
  • demyelinating diseases include, but are not limited to, multiple sclerosis (MS), disseminated sclerosis (DS), acute disseminated encephalomyelitis, progressive multifocal leukoencephalopati (PML), and subacute sclerotic panencephalitis (SSPE).
  • MS multiple sclerosis
  • DS disseminated sclerosis
  • PML progressive multifocal leukoencephalopati
  • SSPE subacute sclerotic panencephalitis
  • MS mutiple sclerosis
  • polyartheritis nodosaasthma hypersensitivity pneumonitis
  • interstitial lung disease sarcoidosis
  • idiopathic pulmonary fibrosis interstitial lung disease associated with Crohn ' s Disease or ulcerative colitis or Whipple's disease
  • interstitial lung disease associated with Wegeners granulomatosis or hypersensitivity vasculitis
  • vasculitis syndromes Hennoch-Sch ⁇ nleins purpura, Goodpastures syndrome, Wegeners granulomatosis,
  • renal diseases such as antibody mediated glomerulopathia as in acute glomerulonephritis, nephritis associated with systemic lupus erythematosus (SLE), nephritis associated with other systemic diseases such as Wegeners granulomatosis and Goodpastures syndrome and mixed connective tissue disease, chronic interstitial nephritis, chronic glomerulonephritis,
  • gastrointestinal diseases such as Crohn ' s Disease, Ulcerative colitis, coeliac disease, Whipple's disease, collagenous colitis, eosinophillic colitis, lymphatic colitis,
  • hepatobilliary diseases such as auto-immune hepatitis, alcohol induced hepatitis, periportal fibrosis, primary billiary cirrhosis, sclerosing colangitis,
  • skin diseases such as psoriasis, atopic dermatitis, eczema, allergic skin disease, progressive systemic sclerosis (scleroderma), exfoliating dermatitis, pemphigus vulgaris,
  • RA rheumatoid arthritis
  • ankylosing spondylitis arthritis associated with psoriasis or inflammatory bowel disease
  • muscoloskelletal diseases such as myastenia gravis (MG), polymyositis, endocrine diseases such as insulin dependent diabetes mellitus (IDDM), auto-immune thyroiditis (Hashimoto), thyreotoxicosis, hyperthyroidism (Graves disease),
  • cardiovascular diseases such as cardiomyopathia, vasculitis, cardiovascular disease associated with systemic diseases as systemic lupus erythematosus, l o polyarthritis nodosa, rheumatoid arthritis, scleroderma, sarcoidosis.
  • a noticeable characteristic of autoimmune diseases is their familial clustering and association with the expression of particular genes, in particular genes of class I and class II major histocompatibility complex (MHC).
  • MHC major histocompatibility complex
  • MS appears to be more common in subjects who live in temperate
  • autoimmune diseases may be an infectious agent such as a virus.
  • infectious agent such as a virus.
  • the etilogy of several human and animal diseases can be attributed to viral infection. For example,
  • Theiler's murine encephalomyelitis is a demyelinating disease with clinical and pathological signs similar to EAE. Although antibodies are involved in some effector responses of autoimmune diseases, the triggering event in most cases begins with the activation of CD4 T-cells that are required for B cell maturation and clonal expansion.
  • the group of delayed type hypersensitivity include contact hypersensitivity to low molecular weight substances. Th2-related diseases
  • the Th2-related diseases comprise the following groups of diseases: Allergic diseases and cancer.
  • the group of allergic diseases includes hay fever, rhinoconjunctivitis, rhinitis and asthma.
  • allergens to which allergic reactions occur, include inhalation allergens originating i.a. from trees, grasses, herbs, fungi, house dust mites, storage mites, cockroaches and animal hair, feathers, and dandruff.
  • Important pollen allergens from trees, grasses and herbs are such originating from the taxonomic orders of Fagales, Oleales and Pinales including i.a. birch (Betula), alder (Alnus), hazel (Corylus), hornbeam ⁇ Carpinus) and olive (Olea), the order of Poales including i.a.
  • Important inhalation allergens from fungi are i.a. such originating from the genera Alterna a and Cladospo ⁇ um.
  • Other important inhalation allergens are those from house dust mites of the genus Dermatophagoides, storage mites from the genus Lepidoglyphys destructor, those from cockroaches and those from mammals such as cat, dog, horse, cow, and bird.
  • allergen components include e.g. Bet v 1 (B. verrucosa, birch), Aln g 1 [Alnus glutinosa, alder), Cor a 1 (Corylus avelana, hazel) and Car b 1 (Carpinus betulus, hornbeam) of the Fagales order.
  • cancer cells may raise an immune response to antigens specifically expressed by the cancer cell. Since both Th1 and Th2 immune responses may drive strong inflammatory responses leading to cytotoxic eradication of tissue, modulation of Th1 or Th2 immune responses may be used in the treatment of cancer.
  • Cancers which can be treated with the composition according to the invention can be histogenetically classified as malignant epithelial tumours, including carcinomas and adenocarcinomas, and as malignant non-epithelial tumours, including liposarcomas, fibrosarcomas, chondrosarcomas, osteosarcomas, leiomyosarcomas, rhabdomyosarcomas, gliomas, neuroblastomas, medulloblastomas, malignant melanoma, malignant meningioma, various leukemias, various myeloproliferative disorders, various lymphomas (Hodgkin's lymphoma and non-Hodgkin lymphoma), haemangiosarcoma, Kaposi's sarcoma, lymphangiosarcoma, malignant teratoma, dysgerminoma, seminoma, and choricarcinoma.
  • malignant epithelial tumours including
  • Carcinomas and adenocarcinomas are the most abundant (accounting for approximately 90% of deaths from cancer) and are therefore interesting target diseases to treat/prevent according to the invention.
  • the most important carcinomas and adenocarcinomas are those of the airways (espially of bronchial origin), of the breast, of the colorectum and of the stomach.
  • carcinomas and adenocarcinomas of the prostate, the ovary, of the lymphoid tissue and bone marrow, of the uterus, of the pancreas, of the esophagus, the urinary bladder, and the kidney cause a significant number of deaths and are therefore of interest.
  • the group of cancer diseases further includes Sezary's syndrome, cutaneous T-cell lymphoma, hepatic carcinoma and lung cancer.
  • the pharmaceutical composition of the invention in addition to the active substance contains a pathogenic agent eliciting the disease to be treated.
  • a pathogenic agent eliciting the disease to be treated.
  • the pharmaceutical composition of the invention may further contain a pathogenic substance eliciting the Th1 -related disease to be treated, in particular an infectious agent eliciting an infectious disease or an antigen.
  • the said antigen may be an autoantigen eliciting an autoimmune disease, a hapten or an allergen eliciting a delayed type hypersensitivity.
  • the pharmaceutical composition of the invention may further contain a pathogenic substance eliciting the Th2-related disease to be treated, in particular a parasite organism or part therof or an antigen.
  • the antigen may be an allergen eliciting an allergic disease.
  • the active substances according to the present invention i.e. substances a)- g) of claim 1 and o)-u) of claim 24 comprise both substances having an extracellular site of action, e.g. IL-4 and SDF-1 ⁇ , and substances having an infracellular site of action, e.g. modulators of Syk, ZAP-70, NFAT1 and NFAT2.
  • the substances having an extracellular site of action typically bind to a receptor on the extracellular side of the cell membrane in a receptor-ligand system.
  • a receptor-ligand system may be inhibited by any substance binding to the receptor or ligand in a manner so that the binding between the receptor and ligand is impaired or prevented.
  • inhibitors are antibodies, low molecular organic substances and free receptors.
  • a receptor- ligand system may be stimulated by any substance binding to the receptor and ligand in a manner so as to facilitate the binding between the receptor and ligand.
  • the receptor-ligand system may be stimulated by increasing the number of ligand molecules by addition thereof or the number of receptor molecules by adding a substance stimulating the infracellular expression of the receptor.
  • the substances having an infracellular site of action may be any substance capable of modulating the activity or expression of the substance.
  • IL-2, IL-4 and SDF-1 ⁇ may be obtained by purification of the subsances from a biological material or by producing the substances using conventional recombinant techniques.
  • stimulants of IL-2, IL-4 or SDF-1 ⁇ are substances stimulating the infracellular activation and extracellular expression of IL-2, IL-4 or SDF- 1 ⁇ receptor.
  • stimulants of IL-2, IL-4 or SDF-1 ⁇ receptor are substances stimulating the infracellular activation and extracellular expression of IL-2, IL-4 or SDF- 1 ⁇ receptor.
  • substances capable of stimulating the expression of IL-2, IL-4 or SDF-1 ⁇ receptor reference is made to the below section with the heading "e) and s) Stimulants of Syk, ZAP-70, NFAT1 or NFAT2", the content of which apply equally to this section.
  • Antagonists to IL-2, IL-4 or SDF-1 ⁇ include monoclonal and polyclonal antibodies to the IL-2, IL-4 or SDF-1 ⁇ , both antibodies which bind to the cell receptor binding site of IL-2, IL-4 or SDF-1 ⁇ and antibodies which bind to other parts of the IL-2, IL-4 or SDF-1 ⁇ molecules in such a manner that the binding of the molecule to its receptor is reduced or hindered.
  • the antagonists to the ligands IL-2, IL-4 or SDF-1 ⁇ include monoclonal and polyclonal antibodies to the receptors of IL-2, IL-4 or SDF-1 ⁇ (CXCR4).
  • the antibodies to the receptors may both be antibodies, which bind to the ligand binding site of the receptor and antibodies which bind to other parts of the receptor thereby reducing or preventing the binding of the ligand to the receptor.
  • the antagonists to IL-2, IL-4 or SDF-1 ⁇ further includes peptides having a binding affinity for either the ligand IL-2, IL-4 or SDF-1 ⁇ or their receptors, e.g. peptides comprising the binding site of one of the said molecules or a part thereof.
  • the antagonists of IL-2, IL-4 or SDF-1 ⁇ may be low molecular compounds, e.g. organic compounds.
  • the antagonists of IL-2, IL-4 or SDF-1 ⁇ may be added as free receptors, i.e. receptors not bound to a T ceil, and free ligands, which are able to bind to its receptor but unable to exert its effector function on T cells or only able to exert a reduced effector function.
  • antagonists of IL-2, IL-4 or SDF-1 ⁇ are substances inhibiting the intracellular expression of IL-2, IL-4 or SDF-1 ⁇ receptor.
  • substances capable of inhibiting the expression of IL-2, IL-4 or SDF-1 ⁇ receptor reference is made to the below section with the heading "d) and r) Inhibitors of Syk, ZAP-70, NFAT1 or NFAT2", the content of which apply equally to this section.
  • Inhibitory compounds of the invention can be, for example, intracellular binding molecules that act to specifically inhibit the expression or activity of Syk, ZAP-70, NFAT1 or NFAT2.
  • intracellular binding molecule is intended to include molecules that act intracellularly to inhibit the expression or activity of a protein by binding to the protein or to a nucleic acid (e.g., an mRNA molecule) that encodes the protein.
  • intracellular binding molecules examples include antisense nucleic acids, intracellular antibodies, peptidic compounds that inhibit the interaction of Syk, ZAP-70, NFAT1 or NFAT2 with a target molecule (e.g., calcineurin) and chemical agents that specifically inhibit Syk, ZAP-70, NFAT1 or NFAT2 activity.
  • a target molecule e.g., calcineurin
  • inhibitory compounds include phosphatases and other enzymes and compounds capable of dephosphorylation.
  • an inhibitory compound of the invention is an antisense nucleic acid molecule that is complementary to a gene encoding Syk, ZAP- 70, NFAT1 or NFAT2, or to a portion of said gene, or a recombinant expression vector encoding said antisense nucleic acid molecule.
  • the antisense nucleic acid molecule may be a DNA, a RNA, a PNA, a LNA or a phosphorothioate.
  • the use of antisense nucleic acids to downregulate the expression of a particular protein in a cell is well known in the art.
  • An antisense nucleic acid molecule comprises a nucleotide sequence that is complementary to the coding strand of another nucleic acid molecule (e.g., an mRNA sequence) and accordingly is capable of hydrogen bonding to the coding strand of the other nucleic acid molecule.
  • Antisense sequences complementary to a sequence of an mRNA can be complementary to a sequence found in the coding region of the mRNA, the 5' or 3' untranslated region of the mRNA or a region bridging the coding region and an untranslated region (e.g., at the junction of the 5' untranslated region and the coding region).
  • an antisense nucleic acid can be complementary in sequence to a regulatory region of the gene encoding the mRNA, for instance a transcription initiation sequence or regulatory element.
  • an antisense nucleic acid is designed so as to be complementary to a region preceding or spanning the initiation codon on the coding strand or in the 3' untranslated region of an mRNA.
  • antisense nucleic acids of the invention can be designed according to the rules of Watson and Crick base pairing.
  • the antisense nucleic acid molecule can be complementary to the entire coding region of a Syk, ZAP-70, NFAT1 or NFAT2 mRNA, but more preferably is an oligonucleotide which is antisense to only a portion of the coding or noncoding region of a Syk, ZAP-70, NFAT1 or NFAT2 mRNA.
  • the antisense oligonucleotide can be complementary to the region surrounding the translation start site of a Syk, ZAP-70, NFAT1 or NFAT2 mRNA.
  • An antisense oligonucleotide can be, for example, about 5, 10, 15, 20, 25, 30, 35, 40, 45 or 50 nucleotides in length.
  • An antisense nucleic acid of the invention can be constructed using chemical synthesis and enzymatic ligation reactions using procedures known in the art.
  • an antisense nucleic acid e.g.
  • an antisense oligonucleotide can be chemically synthesized using naturally occurring nucleotides or variously modified nucleotides designed to increase the biological stability of the molecules or to increase the physical stability of the duplex formed between the antisense and sense nucleic acids, e.g., phosphorothioate derivatives and acridine substituted nucleotides an be used.
  • modified nucleotides which can be used to generate the antisense nucleic acid include 5-fluorouracil, 5 bromouracil, 5 chlorouracil, 5-iodouracil, hypoxanthine, xanthine, 4- acetylcytosine, 5 (carboxyhydroxylmethyl) uracil, 5-carboxymethylamino- methyl-2-thiouridine, 5 carboxymethylaminomethyl-uracil, dihydrouracil, beta-D-galactosylqueosine, inosine, N6-isopentenyladenine, I -methyl- guanine, I -methylinosine, 2,2 dimethylguanine, 2 methyladenine, 2- methylguanine,,3-methylcytosine, 5-methylcytosine, N6 adenine, 7 methyl- guanine, 5-methylaminomethyluracil, 5-methoxyaminomethyl-2 thiouracil, beta D-mannosyl,
  • an antisense nucleic acid can be produced biologically using an expression vector into which all or a portion of Syk, ZAP-70, NFAT1 or NFAT2 cDNA has been subcloned in an antisense orientation (i. e., nucleic acid transcribed from the inserted nucleic acid will be of an antisense orientation to a target nucleic acid of interest).
  • Regulatory sequences operatively linked to a nucleic acid cloned in the antisense orientation can be chosen which direct the expression of the antisense RNA molecule in a cell of interest, for instance promoters and/or enhancers or other regulatory sequences can be chosen which direct constitutive, tissue specific or inducible expression of antisense RNA.
  • the antisense expression vector is prepared according to standard recombinant DNA methods for constructing recombinant expression vectors, except that the Syk, ZAP-70, NFAT1 or NFAT2 cDNA (or portion thereof) is cloned into the vector in the antisense orientation.
  • the antisense expression vector can be in the form of, for example, a recombinant plasmid, phagemid or attenuated virus.
  • the antisense expression vector is introduced into cells using a standard transfection technique.
  • the antisense nucleic acid molecules of the invention are typically administered to a subject or generated in situ such that they hybridize with or bind to cellular mRNA and/or genomic DNA encoding a Syk, ZAP-70, NFAT1 or NFAT2 protein to thereby inhibit expression of the protein, e.g., by inhibiting transcription and/or translation.
  • the hybridization can be by conventional nucleotide complementarity to form a stable duplex, or, for example, in the case of an antisense nucleic acid molecule which binds to DNA duplexes, through specific interactions in the major groove of the double helix.
  • An example of a route of administration of an antisense nucleic acid molecule of the invention includes direct injection at a tissue site.
  • an antisense nucleic acid molecule can be modified to target selected cells and then administered systemically.
  • an antisense molecule can be modified such that it specifically binds to a receptor or an antigen expressed on a selected cell surface, e.g., by linking the antisense nucleic acid molecule to a peptide or an antibody which binds to a cell surface receptor or antigen.
  • the antisense nucleic acid molecule can also be delivered to cells using the vectors described herein. To achieve sufficient intracellular concentrations of the antisense molecules, vector constructs in which the antisense nucleic acid molecule is placed under the control of a strong pol 11 or pol III promoter are preferred.
  • the antisense nucleic acid molecule of the invention is an ⁇ -anomeric nucleic acid molecule.
  • An ⁇ -anomeric nucleic acid molecule forms specific double-stranded hybrids with complementary RNA in which, contrary to the usual P-units, the strands run parallel to each other.
  • the antisense nucleic acid molecule can also comprise a 2'-o methylribonucleotide or a chimeric RNA-DNA analogue.
  • an antisense nucleic acid of the invention is a ribozyme.
  • Ribozymes are catalytic RNA molecules with ribonuclease activity which are capable of cleaving a single-stranded nucleic acid, such as an mRNA, to which they have a complementary region.
  • ribozymes e.g., hammerhead ribozymes
  • a ribozyme having specificity for a Syk-, ZAP-70-, NFAT1 - or NFAT2-encoding nucleic acid can be designed based upon the nucleotide sequence of the Syk, ZAP-70, NFAT1 or NFAT2 cDNA.
  • a derivative of a Tetrahymena L-19 IVS RNA can be constructed in which the nucleotide sequence of the active site is complementary to the nucleotide sequence to be cleaved in a Syk-, ZAP-70-, NFAT1 - or NFAT2-encoding mRNA.
  • Syk, ZAP-70, NFAT1 and NFAT2 gene expression can be inhibited by targeting nucleotide sequences complementary to a regulatory region of a Syk, ZAP-70, NFAT1 or NFAT2 gene (e.g., a Syk, ZAP-70, NFAT1 or NFAT2 promoter and/or enhancer) to form triple helical structures that prevent transcription of an NFAT1 gene in target cells.
  • a Syk, ZAP-70, NFAT1 or NFAT2 gene e.g., a Syk, ZAP-70, NFAT1 or NFAT2 promoter and/or enhancer
  • Syk, ZAP-70, NFAT1 and NFAT2 can be inhibited by a nucleic acid encoding a catalytically inactive mutant of Syk, ZAP-70, NFAT1 and NFAT2.
  • a recombinant expression vector is prepared which encodes the antibody chains in a form such that, upon introduction of the vector into a cell, the antibody chains are expressed as a functional antibody in an intracellular compartment of the cell.
  • an intracellular antibody that specifically binds the transcription factor is expressed within the nucleus of the cell.
  • Nuclear expression of an intracellular antibody can be accomplished by removing from the antibody light and heavy chain genes those nucleotide sequences that encode the N terminal hydrophobic leader sequences and adding nucleotide sequences encoding a nuclear localization signal at either the N- or C-terminus of the light and heavy chain genes.
  • a preferred nuclear localization signal to be used for nuclear targeting of the intracellular antibody chains is the nuclear localization signal of SV40 Large T antigen.
  • antibody light and heavy chain cDNAs encoding antibody chains specific for the target protein of interest e.g. Syk, ZAP-70, NFAT1 or NFAT2 protein
  • the target protein of interest e.g. Syk, ZAP-70, NFAT1 or NFAT2 protein
  • antibody light and heavy chain cDNAs encoding antibody chains specific for the target protein of interest is isolated, typically from a hybridoma that secretes a monoclonal antibody specific for Syk, ZAP- 70, NFAT1 or NFAT2 protein.
  • Preparation of antisera against Syk, ZAP-70, NFAT1 or NFAT2 protein has been described in the art.
  • Anti-Syk, -ZAP-70, - NFAT1 or -NFAT2 antibodies can be prepared by immunizing a suitable subject, (e.g., rabbit, goat, mouse or other mammal) with a Syk, ZAP-70, NFAT1 or NFAT2 immunogen, respectively.
  • a suitable subject e.g., rabbit, goat, mouse or other mammal
  • An appropriate immunogenic preparation can contain, for examples, recombinantly expressed Syk, ZAP- 70, NFAT1 or NFAT2 protein or a chemically synthesized Syk, ZAP-70, NFAT1 or NFAT2 peptide.
  • the preparation can further include an adjuvant, such as Freund's complete or incomplete adjuvant, or similar immunostimulatory compound.
  • Antibody-producing cells can be obtained from the subject and used to prepare monoclonal antibodies by standard techniques, such as the hybridoma technique.
  • the technology for producing monoclonal antibody hybridomas is well known. Briefly, an immortal cell line (typically a myeloma) is fused to lymphocytes (typically splenocytes) from a mammal immunized with a Syk, ZAP-70, NFAT1 or NFAT2 protein immunogen as described above, and the culture supernatants of the resulting hybridoma cells are screened to identify a hybridoma producing a monoclonal antibody that binds specifically to the Syk, ZAP-70, NFAT1 or NFAT2 protein.
  • lymphocytes typically splenocytes
  • the immortal cell line e.g., a myeloma cell line
  • murine hybridomas can be made by fusing lymphocytes from a mouse immunized with an immunogenic preparation of the present invention with an immortalized mouse cell line.
  • Preferred immortal cell lines are mouse myeloma cell lines that are sensitive to culture medium containing hypoxanthine, aminopterin and thymidine ("HAT medium"). Any of a number of myeloma cell lines may be used as a fusion partner according to standard techniques, e.g., the P3- NSI/1-Ag4-1 , P3-x63-Ag8.653 or Sp2/0-Agl4 myeloma lines. These myeloma lines are available from the American Type Culture Collection (ATCC), Rockville, Md. Typically, HAT-sensitive mouse myeloma cells are fused to mouse splenocytes using polyethylene glycol ("PEG").
  • PEG polyethylene glycol
  • Hybridoma cells resulting from the fusion are then selected using HAT medium, which kills unfused and unproductively fused myeloma cells (unfused splenocytes die after several days because they are not transformed).
  • Hybridoma cells producing a monoclonal antibody that specifically binds the maf protein are identified by screening the hybridoma culture supernatants for such antibodies, e.g., using a standard ELISA assay.
  • a monoclonal antibody that binds to Syk, ZAP-70, NFAT1 or NFAT2 can be identified and isolated by screening a recombinant combinatorial immunoglobulin library (e.g., an antibody phage display library) with the protein, or a peptide thereof, to thereby isolate immunoglobulin library members that bind specifically to the protein.
  • Kits for generating and screening phage display libraries are commercially available (e.g., the Pharmacia Recombinant Phage Antibody System, Catalog No. 27-9400-0 1 ; and the Stratagene SurfZ4pTM Phage Display Kit, Catalog No. 240612). Additionally, examples of methods and compounds particularly amenable for use in generating and screening antibody display library can be found in the literature.
  • DNAs encoding the light and heavy chains of the monoclonal antibody are isolated by standard molecular biology techniques.
  • light and heavy chain cDNAs can be obtained, for example, by PCR amplification or cDNA library screening.
  • cDNA encoding the light and heavy chains can be recovered from the display package (e.g., phage) isolated during the library screening process.
  • Nucleotide sequences of antibody light and heavy chain genes from which PCR primers or cDNA library probes can be prepared are known in the art. For example, many such sequences are disclosed in Kabat, E.A., et al. (1991 ) Sequences of Proteins oflmmunological Interest, Fifth Edition, U. S. Department of Health and Human Services, NIH Publication No. 91 -3242 and in the "Vbase" human germline sequence database.
  • the antibody light and heavy chain sequences are cloned into a recombinant expression vector using standard methods.
  • sequences encoding the hydrophobic leaders of the light and heavy chains are removed and sequences encoding a nuclear localization signal (e.g., from SV40 Large T antigen) are linked in-frame to sequences encoding either the amino- or carboxy terminus of both the light and heavy chains.
  • the expression vector can encode an intracellular antibody in one of several different forms. For example, in one embodiment, the vector encodes full-length antibody light and heavy chains such that a full-length antibody is expressed intracellularly.
  • the vector encodes a full- length light chain but only the VH/CHI region of the heavy chain such that a Fab fragment is expressed intracellularly.
  • the vector encodes a single chain antibody (scFv) wherein the variable regions of the light and heavy chains are linked by a flexible peptide linker (e.g., (GIY4Ser)3) and expressed as a single chain molecule.
  • scFv single chain antibody
  • GIY4Ser flexible peptide linker
  • the expression vector encoding the Syk, ZAP-70, NFAT1 or NFAT2-specific intracellular antibody is introduced into the cell by standard transfection methods as described hereinbefore.
  • an inhibitory compound of the invention is a peptidic compound derived from the Syk, ZAP-70, NFAT1 or NFAT2 amino acid sequence.
  • the inhibitory compound(s) comprises a portion of Syk, ZAP-70, NFAT1 or NFAT2 (or a mimetic thereof) that mediates interaction of Syk, ZAP-70, NFAT1 or NFAT2, respectively, with a target molecule such that contact of Syk, ' ZAP-70, NFAT1 or NFAT2 with this peptidic compound competitively inhibits the interaction of Syk, ZAP-70, NFAT1 or NFAT2, respectively, with the target molecule.
  • the peptide compound is designed based on the region of NFAT1 that mediates interaction of NFAT1 with calcineurin.
  • NFAT1 that mediates interaction of NFAT1 with calcineurin.
  • a conserved region in the amino terminus of NFAT proteins mediates interaction of the NFAT proteins with calcineurin and peptides spanning the region inhibit the ability of calcineurin to bind to and phosphorylate NFAT proteins, without affecting the phosphatase activity of calcineurin against other substrates.
  • a NFAT inhibitory compound is a peptidic compound, which is prepared based on a calcineurin-interacting region of NFAT1.
  • a peptide can be derived from the calcineurin-interacting region of NFAT1 having an amino acid sequence that comprises the above-mentioned amino acid motif 1.
  • longer regions of human NFAT1 can be used such as a peptide, which spans the above-mentioned amino acid motif 1.
  • the peptidic compounds of the invention can be made intracellularly in cells (e.g., lymphoid cells) by introducing into the cells an expression vector encoding the peptide(s).
  • expression vectors can be made by standard techniques, using, for example, oligonucleotides that encode one of the above discussed amino acid motifs.
  • the peptide(s) can be expressed in intracellularly as a fusion with another protein or peptide (e.g., a GST fusion).
  • the peptides can be made by chemical synthesis using standard peptide synthesis techniques.
  • Synthesized peptides can then be introduced into cells by a variety of means known in the art for introducing peptides into cells (e.g., liposome and the like). Recombinant methods of making NFAT inhibitory peptides, and methods using them to inhibit NFAT activity in cells, are described further in Avramburu et al., (1998) Mol. Cell. 1 :627-637.
  • NFAT1 activity can be inhibited by mutating the calcineurin-binding region in the amino terminus, comprising the above mentioned motif 1.
  • the wildtype NFAT1 amino acid can be modified to the mutated sequence to create a mutated form of NFAT1 with reduced activity.
  • inhibitory agents that can be used to specifically inhibit the activity of Syk, ZAP-70, NFAT1 or NFAT2 proteins are chemical compounds that directly inhibit Syk, ZAP-70, NFAT1 or NFAT2 activity or inhibit the interaction between Syk, ZAP-70, NFAT1 or NFAT2 and target molecules. Such compounds can be identified using screening assays that select for such compounds.
  • ZAP-70 inhibitory chemical compounds are a 1 ,2,4-oxadiazole analog derived from L-glutamine, L-alanine, L-homo-Phenylalanine or L- serine (Vu, 2000); a mimetic of the bidentate ⁇ -ITAM peptide (Vu, 2000); a monodentate compound (Vu, 2000); a peptoid (Vu, 2000); a isothiazolone compound (Trevillyan et al., 1999); nocodazole (Huby et al., 1998); methyl-3- (N-isothiazolone)-2-thiophenecarboxylate (Trevillyan et al., 1999).
  • stimulatory compounds include active Syk, ZAP-70, NFAT1 or NFAT2 protein, expression vectors encoding Syk, ZAP-70, NFAT1 or NFAT2 and chemical agents that specifically stimulate Syk, ZAP-70, NFAT1 or NFAT2 activity.
  • a preferred stimulatory compound is at least one nucleic acid molecule encoding Syk, ZAP-70, NFAT1 or NFAT2, wherein the nucleic acid molecule(s) is introduced into the subject in a form suitable for expression of the Syk, ZAP-70, NFAT1 or NFAT2 proteins in the cells of the subject.
  • Syk, ZAP-70, NFAT1 or NFAT2 cDNAs full length or partial Syk, ZAP-70, NFAT1 or NFAT2 cDNA sequence
  • the vector is transfected into cells using standard molecular biology techniques.
  • the Syk, ZAP-70, NFAT1 or NFAT2 cDNAs can be obtained, for example, by amplification using the polymerase chain reaction (PCR) or by screening an appropriate cDNA library.
  • the nucleotide sequences of Syk, ZAP-70, NFAT1 or NFAT2 cDNAs are known in the art and can be used for the design of PCR primers that allow for amplification of the cDNAs by standard PCR methods or for the design of a hybridization probe that can be used to screen a cDNA library using standard hybridization methods.
  • the DNA fragments are introduced into one or more suitable expression vector, as described above.
  • a single expression vector that carries both Syk and NFAT1 or ZAP-70 and NFAT2 coding sequences can be used, or two separate vectors can be used.
  • Nucleic acid molecules encoding Syk, ZAP-70, NFAT1 or NFAT2 in the form suitable for expression of the Syk, ZAP-70, NFAT1 or NFAT2 in a host cell can be prepared as described above using nucleotide sequences known in the art.
  • the nucleotide sequences can be used for the design of PCR primers that allow for amplification of a cDNA by standard PCR methods or for the design of a hybridization probe that can be used to screen a cDNA library using standard hybridization methods.
  • a stimulatory compound for stimulating expression of Syk, ZAP-70, NFAT1 or NFAT2 in a cell is a chemical compound that specifically stimulates the expression or activity of endogenous Syk, ZAP-70, NFAT1 or NFAT2 in the cell.
  • Bisperoxovanadium (bpV) is an example of a stimulant of Zap-70.
  • Stimulating chemical compounds can be identified using screening assays that select for compounds that stimulate the expression or activity of Syk, ZAP-70, NFAT1 or NFAT2.
  • the IL-4 stimulating adjuvant according to the present invention may be selected from Group A consisting of
  • the Group A adjuvant is Alhydrogel or Calcium phosphate.
  • the said Group A adjuvants are also referred to as Th2 cell stimulating adjuvants.
  • the IL-2 stimulating adjuvant according to the present invention may be selected from Group B consisting of
  • Murametide MPL monophosphoryl lipid A
  • the Group B adjuvant is CpG molecules or MPL (monophosphoryl lipid A).
  • the said Group B adjuvants are also referred to as Th1 cell stimulating adjuvants.
  • the IL-4 stimulating adjuvant according to the invention may be any adjuvant, which gives rise to a cytokine response which is more Th2 cell skewed than the cytokine response of any of the Group B adjuvants mentioned above.
  • the IL-2 stimulating adjuvant according to the invention may be any adjuvant, which gives rise to a cytokine response which is more Th1 cell skewed than the cytokine response of any of the Group A adjuvants mentioned above.
  • the derivatives, analogues or parts of substances a)-f) and o)-t) may be any derivative, analogue or part known in the prior art.
  • the active substance is a protein
  • a part of the protein having at least a partly conserved functionality may be used.
  • the active substance is an antibody
  • a part of the antibody molecule comprising at least a part of its specificity may be used.
  • the active substance used in the present invention may i.a. be an organic substance, a peptide, a protein and a nucleic acid.
  • the active substance may be formulated in any pharmaceutical composition known in the prior art, including pharmaceutical compositions for injection and for oral, parenteral, pulmonary and nasal administration.
  • Oral compositions include tablets, capsules, pills, troches or lozenges, cachets or pellets.
  • the pharmaceutical composition may include any pharmaceutical acceptable excipient known in the prior art, including diluents, preservatives, solubilizers, emulsifiers, colorants, flavoring agents, disintegrants, binders, antifhctional agents, glidants, surfactants, adjuvants and/or carriers, as described e.g. in Remington's Pharmaceutical Sciences, 18th Ed. (1990, Mack Publishing Co., Easton, PA 18042).
  • Method of treatment including diluents, preservatives, solubilizers, emulsifiers, colorants, flavoring agents, disintegrants, binders, antifhctional agents, glidants, surfactants, adjuvants and/or carriers, as described e.g. in Remington's Pharmaceutical Sciences, 18th Ed. (1990, Mack Publishing Co., Easton, PA 18042).
  • the pharmaceutical composition of the invention is administered by parenteral injection or mucosal administration.
  • Local administration is preferred over systemic administration to avoid any undesirable side effects of SDF-1 ⁇ .
  • the expression “local administration” means topical administration or local injection.
  • local injection means injection, wherein the systemic dissemination of the active substance is less than 50 %, preferably 40 %, more preferably 30 %, more preferably 20 % and most preferably 10 %.
  • the method of treatment of the invention can be practiced either in vitro or in vivo (the latter is discussed further in the following subsection).
  • cells can be obtained from a subject by standard methods and incubated (i.e., cultured) in vitro with the pharmaceutical composition of the invention.
  • lymphoid cells can be isolated from a subject, treated in vitro using a modulatory agent of the invention and then readministered to the same subject, or another subject tissue compatible with the donor of the cells.
  • the modulatory method of the invention comprises culturing cells in vitro with an active substance (modulator) and further comprises administering the cells to a subject to thereby modulate Th1/Th2 cell ratio in a subject.
  • an active substance modulator
  • an active substancend is administered to a subject in vivo.
  • the compounds can be introduced into cells of a subject using methods known in the art for introducing nucleic acid (e.g., DNA) into cells in vivo. 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. 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 which naturally disrupt endosomes, thereby releasing material into the cytoplasm can be used to avoid degradation of the complex by intracellular lysosomes.
  • Retroviruses Defective retroviruses are well characterized for use in gene transfer for gene therapy purposes (for a review see Miller, A.D. (1990) Blood 76:27 1 ).
  • a recombinant retrovirus can be constructed having a nucleotide sequence 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 which 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 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. 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.
  • Suitable adenoviral vectors derived from the adenovirus strain Ad type 5 d1324 or other strains of adenovirus (e.g., Ad2, Ad3, Ad7 etc.) are well known 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, endothelial cells, hepatocytes and muscle cells.
  • 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
  • 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. 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. 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.
  • 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 5 detected by a filter hybridization technique (e.g., Southern blotting) and RNA produced by transcription of introduced DNA can be detected, for example, by Northern blotting, RNase protection or reverse transcriptase-polymerase chain reaction (RT-PCR).
  • a filter hybridization technique e.g., Southern blotting
  • RNA produced by transcription of introduced DNA can be detected, for example, by Northern blotting, RNase protection or reverse transcriptase-polymerase chain reaction (RT-PCR).
  • the gene product can be detected by an appropriate assay, for example by immunological detection of a produced l o protein, such as with a specific antibody, or by a functional assay to detect a functional activity of the gene product, such as an enzymatic assay.
  • an appropriate assay for example by immunological detection of a produced l o protein, such as with a specific antibody, or by a functional assay to detect a functional activity of the gene product, such as an enzymatic assay.
  • the diagnostic test kit comprises one or more probes for each compound to be measured.
  • any known probe suitable of binding to the compound to be measured i.e. phosphorylated Syk, phosphorylated ZAP-70, intranucleic NFAT1 or intranucleic NFAT2, may be used, e.g. antibodies, nucleic acids, fragments therof etc.
  • the probe may be labeled, e.g. with a
  • the kit may comprise a detection system.
  • detection system any known detection system may be used.
  • the detection system comprises a detecting reagent, which is capable of binding to the probe, and 25 which comprises a label compound.
  • An example of such a detection system is a labelled antibody to the probe.
  • a stimulant of X means any agent stimulating the expression and/or the activity of X directly or indirectly regardless of the mechanism involved.
  • an inhibitor of X means any agent inhibiting the expression and/or the activity of X directly or indirectly regardless of the mechanism involved.
  • Th1 cell means any CD4 + T helper 1 cell capable of secreting IL-2 and IFN ⁇ .
  • Th2 cell means any CD4 + T helper 2 cell capable of secreting IL-4 and IL-5.
  • Th1 cell-related disease means any disease, in which Th1 cells support, cause or mediate the disease process or in which Th1 cells are involved in curing or alleviating the symptoms of the disease.
  • Th2 cell-related disease means any disease, in which Th2 cells support, cause or mediate the disease process or in which Th2 cells are involved in curing or alleviating the symptoms of the disease.
  • pathogenic substance means any substance, which elicits a disease in the subject.
  • antigen means any substance, which elicits an immunological disease in the subject.
  • immunological disease means any disease, which is supported by the immunological response system.
  • intranucleic NFAT1 and “intranucleic NFAT2” mean the NFAT1 and NFAT2, respectively, present in the nucleus of the T lymphocyte.
  • antisense nucleic acid molecule means any nucleic acid molecule in the form of a DNA, a RNA, a PNA, a LNA or a phosphorothioate or derivatives, analogs or fragments thereof capable of down-regulating the expression of a particular protein encoded by a nucleic acid complementary of the antisense nucleic acid.
  • the expression “reducing the Th1/Th2 ratio” means that the level of Th1 cells is reduced and/or that the level of Th2 cells is increased.
  • the expression “increasing the Th1/Th2 ratio” means that the level of Th1 cells is increased and/or that the level of Th2 cells is reduced.
  • IL-2 Interleukin 2
  • IL-4 Interleukin 4
  • ZAP-70 ZAP-70 tyrosine kinase ( ⁇ chain associated protein 70 kD)
  • Cb1 casitas B-lineage lymphoma
  • Cb1 -b casitas B-lineage lymphoma-bregion
  • NFAT Nuclear factor of activated T cells.
  • TCR T cell receptor
  • RNA Ribonucleic acid
  • PNA Peptide nucleic acid
  • CD4 + T cells from umbilical cord blood of uncomplicated births were purified as described elsewhere (Jinquan et al., 1997). Briefly, mononuclear cells were separated from heparinized cord blood using Ficoll-Hypaque (Nycomed, Oslo, Norway). CD4 + T cells were purified using positive selection of Dynabeads (Dynal A/S, Norway) according to the manufacturer's instructions. The purity of CD4 + T cells was > 96% measured by flow cytometry. All serum IgM were under detectable level.
  • the cells were washed twice in PBS and then fixed and permeabilized using IntraPrep® (Coulter- ImmunoTech, Miami, Florida, USA) according to manufacture's instructions.
  • IntraPrep® Coulter- ImmunoTech, Miami, Florida, USA
  • the cells were then incubated with the primary mouse anti human IL-4 mAb (10 ⁇ g /ml, R&D Systems, Oxon, UK) for 15 min at room temperature. Cells were washed twice and stained with FITC- conjugated goat anti mouse antibodies for 15 min at room temperature.
  • the stained cells were incubated with mouse IgG (300 ⁇ g/ml) for 30 min at room temperature to block free binding sites of the goat anti mouse antibodies.
  • the cells were then incubated with the primary mouse anti human INF- ⁇ mAb (10 ⁇ g /ml, R&D Systems,) for 15 min at room temperature. Cells were washed twice and stained with PE-conjugated goat anti mouse antibodies for 15 min at room temperature. Cells were washed and resuspended in PBS containing 0.5% formaldehyde for FACS analysis.
  • the cells (5 x 10 6 ) were solubilized in 1 ml of cold TNE buffer consisting of 50 mM Tris-HCL (pH 8.0), 150 mM NaCl, 1 % (v/v) Nonidet P-40 containing 20 mM EDTA, 10 ⁇ g/ml aprotinin, 0.4 mM sodium vanadate, and 10 mM sodium pyrophosphate.
  • the cell lysates were centrifuged at 10,000 x g for 5 min and the supernatants were precleared with protein G-Sepharose.
  • the lysates were then incubated with 5 ⁇ g of rabbit anti-Syk (c-20), rabbit anti-ZAP-70 (LR), goat anti-Cbl (C- 15), or goat anti-Cbl-b (C-20) (all from Santa Cruz BioTech. Inc., Santa Cruz, CA, USA), at 4°C for 1 h and the immune complexes were precipitated with protein G-Sepharose. For blotting, the immune complexes were washes five times with TNE buffer.
  • the immune complex precipitated with protein G-Sepharose was washed four times with TNE buffer and four times with kinase buffer (50 mM HEPES-NaOH, pH 7.4, and 10 mM MnCI 2 ).
  • the immunoprecipitates were suspended in 20 ⁇ l of kinase buffer containing 10 ⁇ Ci of [ ⁇ - 32 P] ATP and incubated at 30°C for 30 min.
  • the reaction was stopped by the addition of 15 ⁇ l of 3X sample buffer (195 mM Tris-HCl, pH 6.8, 9% SDS, 15% 2-ME, and 30% glycerol). Then the mixture was boiled for 5 min and subjected to 8% SDS-PAGE under reducing conditions, followed by autoradiography.
  • proteins in immunoprecipitates were resolved by SDS-PAGE under reducing conditions and then transferred to polyvinylidene difluoride microporous membrane (Schleicher & Schuell Life Science, Dassel, Germany).
  • the membrane was blocked in 5% BSA-TBS (20 mM Tris-HCL, pH 7.5, and 150 mM NaCl), and then incubated with anti-Syk, anti-ZAP-70, anti-Cbl, or anti-Cbl-b.
  • Immunoblots were incubated with [ ⁇ - 125 l] protein A (NEN ® Life Science Products, Inc., Boston, MA, USA). After the incubation the membrane was washed with TBS and followed by autoradiography.
  • PNA Peptide nucleic acid
  • purified CB T cells were permeabilized with a buffered solution containing a relatively low concentration of detergent (20 mM Tris-HCL, pH 8.3, 1.5 mM MgCL2, 68 mM KCL, 0.05% Tween 20, 1 mM ethylenebis (oxyethlene-nithlo)tetraacetic acid, 5.0% glycerol, and 0.1 mM 4-(2-aminoethyl)benzenesulfonyl fluoride), The cells were then cultured in RPMI 1640 with 10% FCS in the presence of antisense PNA (PE Applied Biosystemss, Foster City, USA) at 2 ⁇ M with or without stimuli in period of time indicated. PNA sequences used were as follow:
  • antisense Syk (333) 5'-ATTTTTTGACATGGGA-3' (918) .
  • antisense ZAP-70 (738) 5'-GTTTGCGCTCGGCCTC-3' (723) .
  • SEQ ID NO. 02 (SEQ ID NO.
  • RNA from CB T cells (2 x 10 6 ) was prepared by using Quick Prep® Total RNA Extraction Kit (Pharmacia Biotech, USA) and any potential contaminating chromosomal DNA was digested with DNAse I according to the manufacturer's instructions.
  • DNAse I DNAse I
  • reverse transcription the RNA was reverse transcribed by using oligo (dT)12-18 and Superscript II reverse transcriptase (Life Technologies, Grand Island, USA), according to the manufacturer's instructions.
  • Reverse transcription was performed for 60 min at 37° C, and any potential contaminating protein was denatured by incubation for 10 min at 95° C.
  • Quantitative PCR was performed in special optical tubes in a 96 well microtiter plate (PE Applied Biosystems, Foster City, USA) format on an ABI PRISM® 7700 Sequence Detector Systems (PE Applied Biosystems), according to the manufacturer's instructions.
  • ⁇ IFN sense 5'-TGTAAGCCCCCAGAAACAGAAAG-3'
  • ⁇ IFN antisense 5'-TTGCCCATCAAGAAACAGCAG-3'
  • IL-4 sense 5'-TCACTCTTCACTCTTTTCTTCCCC-3'
  • IL-4 antisense 5'-TCTTCCCACTTTGCTGTTCCTC-3'.
  • oligonucleotide sequences were designed by using software Primer ExpressTM 1.0 (PE Applied Biosystems). They span exon junctions in order to prevent from amplification of genomic DNA. Taqman® universal PCR master mix (PE Applied Biosystems) containing Passive Reference 1 (ROX) was used to normalize for non-PCR-related fluctuations in fluorescence signal.
  • the standard DNA template with known amounts of molecules (1.0 x 10 3 , 2.0 x 10 3 , 4.0 x 10 3 , 1.0 x 10 4 , 2.0 x 10 4 , 1.0 x 10 5 per well) and "no template" controls were used to create standard curves. All unknown cDNAs were diluted to contain equal amounts of ⁇ -actin cDNA.
  • PCR retain conditions were 2 min at 50°C, 10 min at 95°C, 40 cycles with 15 s at 95°C, 60 s at 60°C for each amplification.
  • Potential PCR product contamination was digested by uracil-N-glycosylase (UNG) since dTTP is substituted by dUTP (Kruse et al., 1997). All PCR experiments were performed with a hot start. In the reaction system, UNG and AmpliTaq Gold
  • Electrophoretic mobility shift assay (EMSA)
  • Nuclear extracts were prepared as described by McCaffrey (McCaffrey et al., 1992; Aramburu et al., 1995). Briefly, 400 ⁇ l ice-cold Dignam buffer A and then 25 ⁇ l 10% NP-40 were added into the cells (5 x 10 6 ). The cells were vortexed and centrifuged (9000 rpm, 30 s, 4°C). Pelleted nuclei were lysed in 50 ⁇ l Dignam buffer C, centrifuged (12,000 rpm, 10 min, 4°C), and the resulting supernatants were diluted (1 :1 ) with Dignam buffer D.
  • Double- stranded synthetic oligonucleotide DNA probes were end-labeled with [ ⁇ -32P] ATP (5.000 Ci/mmol) and T4 polynucleotide kinase (Amersham Pharmacia Biotech Inc., UK).
  • the sequences of the oligonucleotide probes used (5' to 3', one strand) were (Jain et al., 1993; McCaffrey et al., 1993): human IL-2 distal NFAT site (NFAT hulL-2) GGAGGAAAAACTGTTTCATACAGAAGG (binding sequences was underlined).
  • EMSA reactions were performed at room temperature in a final volume of 25 ⁇ l.
  • Nuclear extracts (3 ⁇ g protein per reaction volume) were incubated for 15 min in binding buffer (Aramburu et al., 1995), followed by addition of 0.5 ng 32P-labeled probes to react for 15 min, and the samples were electrophoresed on nondenatured 5% polyacrylamide gels in 0.25 TBE buffer.
  • the mAbs or pAbs used for supershift (all at 10 ⁇ g/ml) were incubated with the nuclear extracts on ice for 30 min.
  • Th1 and Th2 cytokine detection by flow cytometry are administered by different combinations among IL-2 (10 ng/ml), IL-4 (10 ng/ml), and SDF-1 ⁇ (100 ng/ml) as indicated before intracellular cytokine assay as as described in Materials and Methods, b: The numbers listed in the table are in percentage detected with intracellular cytokine assay as described in Materials and Methods.
  • Th1 and Th2 cytokines are IFN- ⁇ , IL-4 or IFN- ⁇ and IL-4.
  • c CXCR4 mouse mAb at 5 ⁇ g/ml.
  • d Isotype Ig matched with CXCR4 mouse mAb at 5 ⁇ g/ml.
  • e N.D. no determination. The listed data are from a single representative experiment of five performed.
  • CD4 + T cells from normal CB seem to be "undifferentiated and unprimed" showing naive Th pattern.
  • IFN- ⁇ and IL-4 double positive are 9.7%, whereas IFN- ⁇ or IL-4 single positive are 8.5% or 12.1 %, respectively.
  • the cells After 8 days of stimulation with IL-2 and SDF-1 ⁇ , the cells have been switched to Th1 pattern in terms of expression of IFN- ⁇ (84%), whereas the stimulation with IL-4 and SDF-1 ⁇ leads the CB T cells to express Th2 pattern (90.3%). None of IL-2, IL-4 and SDF-1 ⁇ alone nor combination of IL-2 and IL-4 has shown such function (data not shown).
  • Table 2 Real time detection and amplification of cDNA of IFN- ⁇ and IL-4 in CB T cells.
  • IL-2 and IL-4 were used at 10 ng/ml and SDF-1 ⁇ was used at 100 ng/ml. The shown values are representatives of four similar experiments performed.
  • the mRNA expression of IFN- ⁇ or IL-4 in CB T cells by real time quantitative RT-PCR has also been examined.
  • Fig. 1 shows the activation of Syk (A), ZAP-70 (B), Cbl (C), and Cbl-b (D) kinase in the CB T cells.
  • the cells were either freshly isolated or stimulated with different combinations among IL-2 (10 ng/ml), IL-4 (10 ng/ml), and SDF-1 ⁇ (100 ng/ml) as indicated.
  • KA represents immune complex kinase assay and IB represents immunoblotting.
  • the cells were stimulated for different time intervals indicated, lysed, and immunoprecipitated with rabbit anti-Syk pAb, rabbit anti-ZAP-70 pAb, goat anti-Cbl pAb or goat anti-Cbl-b pAb as described in Materials and Methods.
  • the immunoprecipitates were subjected to kinase reactions or immunoblotted with each antibody indicated as described in Materials and Methods. The leftmost lanes were where antibodies have been used alone.
  • IL-2 and SDF-1 ⁇ together has induced an activation of Syk kinase within 30 min.
  • a strong and persistent phosphorylation of Syk kinase has been seen in 4d and 8d stimulation with IL-2 and SDF-1 ⁇ , whereas neither combination of IL-2 and IL-4 nor combination of IL-4 and SDF-1 ⁇ has induced Syk kinase activation compared with the level in freshly isolated CB T cells (Fig. 1A).
  • IL-4 and SDF-1 ⁇ together has induced a phosphorylation of ZAP-70 kinase within 30 min.
  • IL-2 and SDF-1 ⁇ together has induced an weak activation of Cbl-b kinase within 30 min.
  • a strong and persistent activation of Cbl-b kinase has been seen in 4d and 8d stimulation with either IL-2 and SDF-1 ⁇ or IL-4 and SDF-1 ⁇ (Fig. 1 D). Since they were reported to have contrary effect on ZAP-70 kinase, the final outcome of the regulatory effect of Cbl and Cbl-b depends upon the balance between the activity of the two kinases. No band has been observed in lanes where goat serum has been used in immunoprecipitation assays (data not shown).
  • Fig. 2 shows the blocking effects of PNA Syk and PNA ZAP-70 antisenses on the activation of Syk kinase and ZAP-70 kinase.
  • the cells were purified CB T cells, which were permeabilized prior to further assays, and cultured in the presence or absence of PNA antisense indicated as described in Materials and Methods and with other stimuli indicated for 8 days, As described in connection with Fig. 1 , the cells were then lysed, immunoprecipitated, and subjected to kinase reactions (KA) or immunoblotted (IB). The leftmost lanes were where antibodies have been used alone. In order to confirm the above observation, PNA antisense assays have been conducted.
  • KA kinase reactions
  • IB immunoblotted
  • Syk PNA antisense and ZAP-70 PNA antisense also significantly inhibit kinase activity and protein expression of Syk and ZAP-70 in non- stimulated CB T cells in culture within 8 days, respectively (Fig. 2).
  • Syk PNA antisense can significantly reduce Syk kinase activity within one day (data not shown), and completely abolish Syk kinase activity within 8 days in (IL-2 + SDF-1 ⁇ )- and (IL-4 + SDF-1 ⁇ )-stimulated CB T cells (Fig. 2A). The total amount of Syk protein is also severely reduced.
  • ZAP-70 PNA antisense can significantly reduce ZAP-70 kinase activity within one day (data not shown), and completely abolish ZAP-70 kinase activity within 8 days in (IL-2 + SDF-1 ⁇ )- and (IL-4 + SDF-1 ⁇ )-stimulated CB T cells (Fig. 2B).
  • the ZAP-70 protein is also completely eliminated. No band has been observed in lanes where rabbit serum has been used in immunoprecipitation assays (data not shown).
  • Table 3 Real time detection and amplification of cDNA of IFN- ⁇ and IL-4 in CB T cells.
  • IL-2 and IL-4 were used at 10 ng/ml and SDF-1 ⁇ was used at 100 ng/ml. The shown values are representatives of four similar experiments performed.
  • Syk PNA antisense has dramatically inhibited IFN- ⁇ mRNA expression (4.5 10 2 copies) induced by IL-2 and SDF-1 ⁇ (Table 3), whereas it has not significantly changed IFN- ⁇ mRNA expression (1.2 * 10 3 copies) induced by IL-4 and SDF-1 ⁇ .
  • ZAP-70 PNA antisense has not such ability to inhibit IFN- ⁇ mRNA expression (1.1 10 5 copies) induced by IL-2 and SDF-1 ⁇ , whereas it has not significantly changed IFN- ⁇ mRNA expression (1.4 * 10 2 copies) induced by IL-4 and SDF-1 ⁇ either.
  • ZAP-70 PNA antisense has dramatically inhibited IL-4 mRNA expression (6.1 10 1 copies) induced by IL-4 and SDF-1 ⁇ (Table 3), whereas it has not significantly changed IL-4 mRNA expression (1.8 10 2 copies) induced by IL-2 and SDF-1 ⁇ .
  • Syk PNA antisense has no such ability to inhibit IL-4 mRNA expression (1.9 10 4 copies) induced by IL-4 and SDF-1 ⁇ , whereas it has not significantly changed IL-4 mRNA expression (8.5 x 10 2 copies) induced by IL-2 and SDF-1 ⁇ either.
  • Syk PNA antisense and ZAP-70 PNA antisense also significantly inhibit mRNA expression of IFN- ⁇ and IL-4 in non-stimulated CB T cells in culture within 8 days, respectively (data not shown). The correlation coefficients in experiments for real time mRNA quantification are > 0.97.
  • Fig. 3 shows activation and identification of NFAT in CB T cells upon stimulation with different combinations among IL-2 (10 ng/ml), IL-4 (10 ng/ml), and SDF-1 ⁇ (100 ng/ml).
  • the tested materials were either nuclei extracts (A, C and D) or whole cell extracts (B).
  • the cells were freshly isolated or stimulated with different stimuli for each time intervals as indicated.
  • NFAT activation was assessed by EMSA using a 32 P-labeled NFAT hulL-2 probe as described in
  • IL-2 + IL-4 only induce weak activation of NFAT in terms of translocation of NFAT from cytoplasm to nuclear.
  • (IL-2 + SDF-1 ⁇ ) or (IL-4 + SDF-1 ⁇ ) stimulations have induced strong and persistent NFAT activations in CB T cells in 1d, 4d and 8d, whereas this phenomenon has not been seen in CB T cells stimulated with (IL-2 + IL-4) (Fig. 3A).
  • IL-2 + SDF-1 ⁇ , IL-4 + SDF-1 ⁇ , or IL-2 + IL-4) Fig.
  • NFAT complex has been detected with NFAT hulL-2 probe in nuclear extracts of CB T cells after stimulation with IL-2 and SDF-1 ⁇ for 8d. This complex has been supershifted by the anti-NFAT1 mAb. Neither isotype mouse antibody nor anti-NFAT2 mAb can affect the relative mobility of the NFAT complex in nuclear extracts of (IL-2 + SDF-1 ⁇ )- stimulated CB T cells. In contrast, NFAT complex has been supershifted by the anti-NFAT2 mAb in nuclear extracts of CB T cells after stimulation with IL-4 and SDF-1 ⁇ .
  • Neither isotype mouse antibody nor anti-NFAT1 mAb can affect the relative mobility of the NFAT complex in nuclear extracts of (IL-4 + SDF- 1 ⁇ )-stimulated CB T cells. These results confirm that only NFAT1 , but not NFAT2, is present in nuclear extracts of (IL-2 + SDF-1 ⁇ )-stimulated CB T cells. These results also confirm that only NFAT2, but not NFAT1 , is present in nuclear extracts of (IL-4 + SDF-1 ⁇ )-stimulated CB T cells. In Fig.
  • Th1 and Th2 cytokine profiles could modify the immune response at sites of inflammation (Coffman et al., 1999).
  • allergic inflammation is a Th2-associated disease (Casolaro et al, 1996).
  • Th2 cytokines play an essential role in the initiation, development, progression and termination of allergic inflammatory process.
  • the determination of the expression and dynamics of Th1 and Th2 cytokine will be critical for the diagnosis and prognosis of Th1- and Th2-associated diseases.
  • Th1 and Th2 clones were first isolated from hyperimmunized mice (O ' Garra et al., 1998) or during chronic diseases in humans (Romagnani et al., 1994; Sher et al., 1992).
  • Th1 and Th2 T cells have been isolated from peripheral blood, draining lymph nodes and affected tissues during chronic infectious diseases and allergy (Romagnani et al., 1994). Although their ability to influence chronic disease or pathological process by their production of high levels of regulatory cytokines is not in doubt, to what numerical extent Th1 and Th2 cells dominate such in vivo responses is as yet not clear (O ' Garra et al., 1998). Th1 and Th2 subsets develop from the same T cell precursor, which is a mature, naive CD4 + T cell producing mainly IL-2 upon antigen- specific stimulation (O ' Garra et al., 1998).
  • Th1 and Th2 differentiation from this precursor are cytokines present at the initiation of the immune response at the stage of ligation of the TCR (Abbas et al., 1996).
  • the present data showing that IL-2 or IL-4 in combination with SDF-1 ⁇ can switch non-antigen-specific CB CD4 + T cells to Th1 or Th2 cells certainly raises the question whether SDF-1 ⁇ has a mitogen-like function in terms of direction of Th1 and Th2 cell formation. If not, the question can be extended to whether the stage of antigen specifying in Th1 and Th2 cell formation is absolutely necessary.
  • TCR-mediated signal transduction is critical for T cell development.
  • Syk and ZAP-70 belonging to the Syk kinase family and associated with TCR, are different both in terms of expression and activity despite their clear structural resemblance (Chu et al., 1998).
  • the kinases in the Syk family are selectively involved in activation of Th1 and Th2 cells (Tamura et al., 1995; Faith et al., 1999), the signaling pathways leading to polarization of Th1 and Th2 cells are not very clear until now.
  • Th1 and Th2 cells are interconvertible, but with repeated stimulation both populations become irreversible.
  • the biochemical basis of this reversibility or stability may be the regulated transcription of cytokine genes or the expression of cytokine receptors (Abbas et al., 1996).
  • Some clinical symptoms seem not to sustain such opinion. For example, allergic asthma in which Th2 cytokines play a crucial and predominate role, could be onset within seconds, but during the remission, the balance between Th1 and Th2 cytokines could be upheld reasonably.
  • Th cells could be entirely and quickly switched to Th1 or Th2 cells upon antigen stimulation and cytokine stimulation in vivo, which occurs locally in an early phase and systematically in a late phase.
  • Newborn children have a low level of trigger-able Th1 and Th2 cells. These cells become memory Th1 and Th2 cells during development of naive Th cells.
  • Memory Th1 and Th2 cells which are in balance in normal condition in term of number and ability of synthesis and secretion of cytokines, may exist in vivo after exposure of antigen.
  • Th1 and Th2 cells perhaps genetic "ill” Th1 and Th2 cells, together with other immune cells such as macrophages, provide an autocrine cytokine environment in situ, where Th1 - or Th2- polarized diseases occur. LIST OF REFERENCES

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Immunology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Epidemiology (AREA)
  • Organic Chemistry (AREA)
  • Zoology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Biomedical Technology (AREA)
  • Cell Biology (AREA)
  • Wood Science & Technology (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • Biochemistry (AREA)
  • Inorganic Chemistry (AREA)
  • Molecular Biology (AREA)
  • Hematology (AREA)
  • Genetics & Genomics (AREA)
  • General Engineering & Computer Science (AREA)
  • Oncology (AREA)
  • Analytical Chemistry (AREA)
  • Physics & Mathematics (AREA)
  • Urology & Nephrology (AREA)
  • Communicable Diseases (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • General Physics & Mathematics (AREA)
  • Toxicology (AREA)
  • Biophysics (AREA)

Abstract

L'invention concerne une composition pharmaceutique destinée à prévenir ou traiter une maladie associée aux cellules Th1 et Th2 chez un humain ou un animal par modulation du rapport Th1/Th2. Cette composition comprend une substance active constituée par (i) IL-4 et SDF-1α ou IL-2 et SDF-1α ainsi que leurs modulateurs, (ii) un adjuvant stimulateur d'IL-4 et SDF-1α ou un adjuvant stimulateur d'IL-2 et SDF-1α, (iii) un modulateur des tyrosine kinases Syk ou ZAP-70, ou (iv) un modulateur des facteurs nucléaires des cellules T activées NFAT1 ou NFAT2.
EP02724144A 2001-05-09 2002-05-07 Compositions pharmaceutiques destines a prevenir ou traiter les maladies associees aux cellules th1 et th2 par modulation du rapport th1/th2 Withdrawn EP1418936A2 (fr)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US28971101P 2001-05-09 2001-05-09
DKPA200100726 2001-05-09
US289711P 2001-05-09
DK200100726 2001-05-09
PCT/DK2002/000295 WO2002089832A2 (fr) 2001-05-09 2002-05-07 Compositions pharmaceutiques destines a prevenir ou traiter les maladies associees aux cellules th1 et th2 par modulation du rapport th1/th2

Publications (1)

Publication Number Publication Date
EP1418936A2 true EP1418936A2 (fr) 2004-05-19

Family

ID=26069017

Family Applications (1)

Application Number Title Priority Date Filing Date
EP02724144A Withdrawn EP1418936A2 (fr) 2001-05-09 2002-05-07 Compositions pharmaceutiques destines a prevenir ou traiter les maladies associees aux cellules th1 et th2 par modulation du rapport th1/th2

Country Status (5)

Country Link
EP (1) EP1418936A2 (fr)
JP (1) JP2004536800A (fr)
CN (1) CN1531440A (fr)
CA (1) CA2447360A1 (fr)
WO (1) WO2002089832A2 (fr)

Families Citing this family (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1444989A1 (fr) 2003-02-07 2004-08-11 Giorgio Dr. Stassi Sensibilisation de cellules à l'apoptose par bloquage selective de cytokines
WO2005007623A2 (fr) * 2003-07-03 2005-01-27 The Trustees Of The University Of Pennsylvania Inhibition de l'expression d'une syk kinase
US20050191283A1 (en) * 2003-10-16 2005-09-01 Suzanne Kadereit Methods of treating NFAT-related disorders
JP2009155204A (ja) * 2005-12-20 2009-07-16 Locomogene Inc アレルギー性疾患用医薬組成物
TWI426918B (zh) * 2007-02-12 2014-02-21 Merck Sharp & Dohme Il-23拮抗劑於治療感染之用途
US9695397B2 (en) * 2008-10-01 2017-07-04 Immunovative Therapies Ltd. Th1 vaccination priming for active immunotherapy
WO2014172805A1 (fr) * 2013-04-26 2014-10-30 Lee Der-Tsai Composition à activité de cytokine améliorée et son application
JP6671298B2 (ja) 2014-05-16 2020-03-25 アムジェン インコーポレイテッド Th1及びth2細胞集団を検出するためのアッセイ
CN104357392A (zh) * 2014-11-07 2015-02-18 广州洁汉贸易有限公司 极化cd4+t细胞的方法
CN104694631A (zh) * 2015-02-05 2015-06-10 华中农业大学 一种西瓜基因表达的实时荧光定量pcr分析方法
CN108088998B (zh) * 2016-11-23 2019-12-27 四川大学华西医院 一种肺癌筛查试剂盒
EP4053267A4 (fr) * 2019-11-01 2024-03-20 Kyoto University Procédé de production de lymphocyte t
CN115927169B (zh) * 2022-10-11 2023-08-11 再造再生医学科技(杭州)有限公司 用于扩增cd34+造血干细胞的培养液和体外扩增cd34+造血干细胞的方法

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ATE145556T1 (de) * 1990-09-06 1996-12-15 Schering Corp Verwendung von interleukin-4 zur behandlung der festen tumoren
CA2104958A1 (fr) * 1991-03-07 1992-09-08 Jean C. Nichols Utilisation de molecules ciblees sur les recepteurs de la surface cellulaire pour le traitement des maladies virales
AU723595B2 (en) * 1995-06-07 2000-08-31 University Of Pennsylvania Methods of inhibiting phagocytosis
AU1230600A (en) * 1998-10-28 2000-05-15 President And Fellows Of Harvard College Regulation of th2 cell activity by modulation of nfatp and nfat4 activity

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO02089832A2 *

Also Published As

Publication number Publication date
WO2002089832A2 (fr) 2002-11-14
JP2004536800A (ja) 2004-12-09
CN1531440A (zh) 2004-09-22
WO2002089832A3 (fr) 2004-03-25
CA2447360A1 (fr) 2002-11-14

Similar Documents

Publication Publication Date Title
US20030103938A1 (en) Pharmaceutical compositions for preventing or treating Th1 and Th2 cell related diseases by modulating the Th1/Th2 ratio
Mogilenko et al. Immune ageing at single-cell resolution
US5298396A (en) Method for identifying T cells disease involved in autoimmune disease
CN101248180B (zh) 作为检测日光照射、***癌和其它癌症的诊断工具的线粒体突变及重排
JP2005520479A (ja) 乾癬の確認、評価、予防および治療のための組成物、キットおよび方法
JP2011509676A (ja) ヒトTh17細胞の選択的分化、同定および調節
US20190178888A1 (en) Methods of determining prognosis of sepsis and treating same
KR20110015409A (ko) 염증성 장 질환에 대한 유전자 발현 마커
CN113260633A (zh) 用于癌症免疫疗法的诊断方法和组合物
EP1418936A2 (fr) Compositions pharmaceutiques destines a prevenir ou traiter les maladies associees aux cellules th1 et th2 par modulation du rapport th1/th2
JP6710004B2 (ja) 免疫療法のためのモニタリングまたは診断ならびに治療剤の設計
CN101111768A (zh) 肺癌预后
CN111295454A (zh) 靶向染色体不稳定性和下游胞质dna信号传导以治疗癌症
Peters et al. Protective intravenous BCG vaccination induces enhanced immune signaling in the airways
US20090131346A1 (en) Antibody and method for identification of dendritic cells
CN109963865A (zh) 用于检测和治疗肥大细胞活性相关病症的生物标记
WO2019068087A1 (fr) Système de prédiction de réponse à une thérapie anticancéreuse et ses méthodes d'utilisation
CN109061191A (zh) S100p蛋白作为标志物在诊断活动性结核病中的应用
AU2002254873A1 (en) Pharmaceutical compositions for preventing or treating TH1 and TH2 cell related diseases by modulating the TH1/TH2 ratio.
Tackenberg et al. CD45 isoform expression in autoimmune myasthenia gravis
AU728909B2 (en) Immunological method
JP2021136934A (ja) 癌患者への免疫チェックポイント阻害薬の投与の有効性を予測するためのデータを収集する方法
US20050019764A1 (en) Method for analyzing t lymphocytes with the aid of t lymphocyte receptors of an organism
KR102202120B1 (ko) 알츠하이머 질환의 진단 또는 치료를 위한 Ube2h의 용도
US20230287390A1 (en) Compositions and methods for identifying epitopes

Legal Events

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

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20031118

AK Designated contracting states

Kind code of ref document: A2

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

AX Request for extension of the european patent

Extension state: AL LT LV MK RO SI

17Q First examination report despatched

Effective date: 20040720

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

Free format text: STATUS: THE APPLICATION HAS BEEN WITHDRAWN

18W Application withdrawn

Effective date: 20070103