WO2003087159A2 - Traitement medical - Google Patents

Traitement medical Download PDF

Info

Publication number
WO2003087159A2
WO2003087159A2 PCT/GB2003/001525 GB0301525W WO03087159A2 WO 2003087159 A2 WO2003087159 A2 WO 2003087159A2 GB 0301525 W GB0301525 W GB 0301525W WO 03087159 A2 WO03087159 A2 WO 03087159A2
Authority
WO
WIPO (PCT)
Prior art keywords
notch
cells
modulators
protein
notch signalling
Prior art date
Application number
PCT/GB2003/001525
Other languages
English (en)
Other versions
WO2003087159A3 (fr
WO2003087159A8 (fr
Inventor
Mark William Bodmer
Emmanuel Cyrille Pascal Briend
Brian Robert Champion
Andrew Christopher Lennard
Grahame James Mckenzie
Tamara Tugal
George Albert Ward
Lesley Lynn Young
Original Assignee
Lorantis Limited
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
Priority claimed from GB0207930A external-priority patent/GB0207930D0/en
Priority claimed from GB0207929A external-priority patent/GB0207929D0/en
Priority claimed from GB0212282A external-priority patent/GB0212282D0/en
Priority claimed from GB0212283A external-priority patent/GB0212283D0/en
Priority claimed from PCT/GB2002/003397 external-priority patent/WO2003012441A1/fr
Priority claimed from GB0220912A external-priority patent/GB0220912D0/en
Priority claimed from GB0220913A external-priority patent/GB0220913D0/en
Priority claimed from GB0300234A external-priority patent/GB0300234D0/en
Priority to AU2003226537A priority Critical patent/AU2003226537A1/en
Priority to EP03746366A priority patent/EP1492816A2/fr
Priority to JP2003584114A priority patent/JP2006506322A/ja
Application filed by Lorantis Limited filed Critical Lorantis Limited
Priority to EP03766445A priority patent/EP1525221A1/fr
Priority to PCT/GB2003/003285 priority patent/WO2004013179A1/fr
Priority to JP2005506075A priority patent/JP2006513260A/ja
Priority to AU2003255735A priority patent/AU2003255735A1/en
Priority to JP2004571915A priority patent/JP2006515177A/ja
Priority to PCT/GB2003/003908 priority patent/WO2004024764A1/fr
Priority to EP03748255A priority patent/EP1537145A1/fr
Priority to CA002497226A priority patent/CA2497226A1/fr
Priority to AU2003267563A priority patent/AU2003267563A1/en
Publication of WO2003087159A2 publication Critical patent/WO2003087159A2/fr
Priority to PCT/GB2004/000046 priority patent/WO2004060262A2/fr
Priority to JP2006500232A priority patent/JP2006517533A/ja
Priority to PCT/GB2004/000263 priority patent/WO2004064863A1/fr
Priority to EP04704657A priority patent/EP1585543A1/fr
Publication of WO2003087159A3 publication Critical patent/WO2003087159A3/fr
Priority to EP04722319A priority patent/EP1646400B1/fr
Priority to PCT/GB2004/001252 priority patent/WO2004082710A1/fr
Priority to AT04722319T priority patent/ATE474593T1/de
Priority to US10/958,784 priority patent/US20050220886A1/en
Priority to US11/050,346 priority patent/US20060002924A1/en
Priority to US11/078,735 priority patent/US20050261477A1/en
Publication of WO2003087159A8 publication Critical patent/WO2003087159A8/fr
Priority to US11/188,417 priority patent/US20060204508A1/en
Priority to US11/231,494 priority patent/US20060205823A1/en
Priority to US12/766,738 priority patent/US20100303867A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/16Agglomerates; Granulates; Microbeadlets ; Microspheres; Pellets; Solid products obtained by spray drying, spray freeze drying, spray congealing,(multiple) emulsion solvent evaporation or extraction
    • A61K9/167Agglomerates; Granulates; Microbeadlets ; Microspheres; Pellets; Solid products obtained by spray drying, spray freeze drying, spray congealing,(multiple) emulsion solvent evaporation or extraction with an outer layer or coating comprising drug; with chemically bound drugs or non-active substances on their surface
    • A61K9/1676Agglomerates; Granulates; Microbeadlets ; Microspheres; Pellets; Solid products obtained by spray drying, spray freeze drying, spray congealing,(multiple) emulsion solvent evaporation or extraction with an outer layer or coating comprising drug; with chemically bound drugs or non-active substances on their surface having a drug-free core with discrete complete coating layer containing drug
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/02Stomatological preparations, e.g. drugs for caries, aphtae, periodontitis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/08Drugs for disorders of the urinary system of the prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/10Drugs for disorders of the urinary system of the bladder
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/06Antiabortive agents; Labour repressants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/08Drugs for genital or sexual disorders; Contraceptives for gonadal disorders or for enhancing fertility, e.g. inducers of ovulation or of spermatogenesis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/04Drugs for disorders of the muscular or neuromuscular system for myasthenia gravis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/02Drugs for disorders of the nervous system for peripheral neuropathies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/18Antipsychotics, i.e. neuroleptics; Drugs for mania or schizophrenia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/16Otologicals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • A61P31/06Antibacterial agents for tuberculosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/16Antivirals for RNA viruses for influenza or rhinoviruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • A61P31/22Antivirals for DNA viruses for herpes viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • A61P33/02Antiprotozoals, e.g. for leishmaniasis, trichomoniasis, toxoplasmosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • A61P33/10Anthelmintics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/38Drugs for disorders of the endocrine system of the suprarenal hormones
    • A61P5/40Mineralocorticosteroids, e.g. aldosterone; Drugs increasing or potentiating the activity of mineralocorticosteroids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/04Antihaemorrhagics; Procoagulants; Haemostatic agents; Antifibrinolytic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/08Vasodilators for multiple indications
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/30Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto

Definitions

  • the present invention relates to therapeutic modulation of the Notch signalling pathway, particularly, but not exclusively, in immune cells.
  • Notch signal transduction plays a critical role in cell fate determination in vertebrate and invertebrate tissues. Notch is expressed at many stages of Drosophila embryonic and larval development and in many different cells implying a wide range of functions including an important role in neurogenesis and in the differentiation of mesodermal and endode mal cells.
  • Notch- 1, Notch-2, Notch-3 and Notch-4 There are at least four mammalian Notch genes (Notch- 1, Notch-2, Notch-3 and Notch-4).
  • Notch- 1 which most closely resembles the proteins of invertebrates and lower vertebrates, is widely expressed and is essential for early development. Recent evidence suggests that Notch signalling contributes to lineage commitment of immature T-cells in the thymus.
  • T-cells During maturation in the thymus, T-cells acquire the ability to distinguish self-antigens from those that are non-self, a process termed "self tolerance". Tolerance to a non-self antigen, however, may be induced by immunisation under specific conditions with a peptide fragment comprising that antigen. In autoimmune diseases such as multiple sclerosis, rheumatoid arthritis or diabetes, there is a failure of the proper regulation of tolerance. Improved treatment methods for re-establishing tolerance are desirable for autoimmune diseases. Similarly in allergic conditions and for transplantation of an organ or tissue from a donor individual, induction of tolerance to particular foreign antigens or profiles of foreign antigens is desirable.
  • Notch and its ligands suggests a role for these proteins in T-cell acquired immunocompetence.
  • T-cells express Notch- 1 mRNA constitutively. Delta expression is limited to only a subset of T-cells in the peripheral lymphoid tissues. Serrate expression is restricted to a subset of antigen presenting cells (APCs).
  • APCs antigen presenting cells
  • manipulation of the Notch signalling pathway can be used in immunotherapy and in the prevention and/or freatment of T-cell mediated diseases.
  • allergy, autoimmunity, graft rejection, tumour induced aberrations to the T-cell system and infectious diseases caused, for example, by Plasmodium species, Microfilariae, Helminths, Mycobacteria, HIV, Cytomegalovirus, Pseudomonas, Toxoplasma, Echinococcus, Haemophilus influenza type B, measles, Hepatitis C or Toxicara, may be targeted.
  • regulatory T cells which are able to transmit antigen-specific tolerance to other T cells, a process termed infectious tolerance (WO98/20142).
  • infectious tolerance WO98/20142
  • the functional activity of these cells can be mimicked by over-expression of a Notch ligand protein on their cell surfaces or on the surface of antigen presenting cells, hi particular, regulatory T cells can be generated by over-expression of a member of the Delta or Serrate family of Notch ligand proteins.
  • Delta or Serrate induced T cells specific to one antigenic epitope are also able to transfer tolerance to T cells recognising other epitopes on the same or related antigens, a phenomenon termed "epitope spreading".
  • Notch ligand expression also plays a role in cancer. Indeed, upregulated Notch ligand expression has been observed in some tumour cells. These tumour cells are capable of rendering T cells unresponsive to restimulation with a specific antigen, thus providing a possible explanation of how tumour cells prevent normal T cell responses. By downregulating Notch signalling in vivo in T cells, it may be possible to prevent tumour cells from inducing immunotolerance in those T cells that recognise tumour-specific antigens, hi turn, this would allow the T cells to mount an immune response against the tumour cells (WOOO/135990).
  • the present invention seeks to provide methods, uses and compositions for modulating the Notch signalling pathway in therapy, and particularly for modulation of immune cell activity in immunotherapy.
  • a pharmaceutical composition comprising a construct which comprises a multiplicity of bound or linked modulators of Notch signalling.
  • the pharmaceutical composition further comprises a pharmaceutically acceptable diluent or carrier.
  • the composition is in sterile form, especially when used in vivo.
  • the modulators of Notch signalling are presented by the construct in an oreientation suitable for activation of a Notch receptor, and are preferably orientated on the surface of the construct.
  • the invention further provides a method for therapeutic modulation of Notch signalling by administering a construct comprising a multiplicity of bound or linked modulators of Notch signalling.
  • the invention further provides a method for generating a regulatory T-cell or increasing regulatory T-cell activity by contacting a construct comprising a multiplicity of bound or linked modulators of Notch signalling with a T-cell.
  • a method for therapeutic modulation of Notch signalling in immune cells by administering a construct comprising a multiplicity of bound or linked modulators of Notch signalling.
  • a method for therapeutic modulation of immune cell activity by administering a construct comprising a multiplicity of bound or linked modulators of Notch signalling.
  • a method for therapeutic modulation of T-cell activity by administering a construct comprising a multiplicity of bound or linked modulators of Notch signalling.
  • a method for treating inflammation, asthma, allergy, graft rejection, graft- versus-host disease or autoimmune disease by administering a construct comprising a multiplicity of bound or linked modulators of Notch signalling.
  • a construct comprising a multiplicity of bound or linked modulators of Notch signalling for use in the treatment of disease.
  • a construct comprising a multiplicity of bound or linked modulators of Notch signalling for use in the treatment of an immune disorder.
  • a particle bearing a multiplicity of bound modulators of Notch signalling for use in the freatment of disease Suitably the modulators of Notch signalling are presented on the particle in an orientation suitable for activation of a Notch receptor, and are preferably orientated on the surface of the particle.
  • a particle bearing a multiplicity of bound modulators of Notch signalling for use in the treatment of an immune disorder.
  • a construct comprising a multiplicity of bound or linked modulators of Notch signalling in the manufacture of a medicament for modulation of immune cell activity.
  • the immune cells are not stem cells.
  • a method for treating an immune disorder by administering a construct comprising a multiplicity of bound or linked modulators of Notch signalling.
  • a substrate bearing a multiplicity of bound modulators of Notch signalling for use in the treatment of disease.
  • a construct comprising a multiplicity of bound or linked modulators of Notch signalling for the manufacture of a medicament for modulation of expression of a cytokine selected from IL- 10, IL-5, IL-2, TNF-alpha, IFN-gamma or IL-13.
  • a construct comprising a multiplicity of bound or linked modulators of Notch signalling for the manufacture of a medicament for increase of IL- 10 expression.
  • a construct comprising a multiplicity of bound or linked modulators of Notch signalling for the manufacture of a medicament for decrease of expression of a cytokine selected from IL-2, IL-5, TNF-alpha, IFN-gamma or IL-13.
  • a construct comprising a multiplicity of bound or linked modulators of Notch signalling for the manufacture of a medicament for generating an immune modulatory cytokine profile with increased IL-10 expression and reduced IL-5 expression.
  • a construct comprising a multiplicity of bound or linked modulators of Notch signalling for the manufacture of a medicament for generating an immune modulatory cytokine profile with increased IL-10 expression and reduced IL-2, IFN-gamma, IL-5, IL-13 and TNF-alpha expression.
  • the construct may be a substantially or partially water-soluble construct.
  • it may be a water insoluble or dispersable construct
  • a particle comprising a modulator of Notch signalling bound to a particulate support matrix.
  • the particulate support matrix is a bead.
  • a plurality of Notch ligands are bound to the particulate support matrix.
  • a particle comprising a modulator of Notch signalling bound to a particulate support matrix.
  • the particulate support matrix is a bead.
  • the modulator of Notch signalling is a Notch ligand or fragment thereof.
  • a plurality of Notch ligands are bound to the particulate support matrix.
  • a method of modifying immune cell (eg T-cell) activity ex-vivo by contacting an immune cell with a surface (eg, bead, well, plate) to which is bound (chemically or by affinity or adsporption) a Notch signalling agonist, such as a Notch ligand proein comprising a DSL domain and at least 2 EGF domains, and administering the cell to a patient.
  • a surface eg, bead, well, plate
  • a Notch signalling agonist such as a Notch ligand proein comprising a DSL domain and at least 2 EGF domains
  • the invention also provides a plate or well which is coated with a plurality of Notch signalling agonists, suitably the the agonists may be coupled to the plate chemically.
  • a pharmaceutically acceptable support matrix suitable for in vivo administration which bears a modulator of Notch signalling.
  • the support matrix may be in the form of an implantable support matrix, for example in the form of a particle.
  • the support matrix bears a Notch ligand, and suitably bears a multiplicity of Notch ligands.
  • a protein or polypeptide consisting essentially of the following components: i) a Notch ligand DSL domain; ii) 1-5 Notch ligand EGF domains; iii) optionally all or part of a Notch ligand N-terminal domain; and iv) optionally one or more heterologous amino acid sequences; and comprising a coupling element suitable for coupling to a support or carrier agent.
  • a protein or polypeptide consisting essentially of the following components: i) a Notch ligand DSL domain; ii) 2-4 Notch ligand EGF domains; iii) optionally all or part of a Notch ligand N-terminal domain; and iv) optionally one or more heterologous amino acid sequences; and comprising a coupling element suitable for coupling to a support or carrier agent.
  • a protein or polypeptide consisting essentially of the following components: i) a Notch ligand DSL domain; ii) 2-3 Notch ligand EGF domains; iii) optionally all or part of a Notch ligand N-terminal domain; and iv) optionally one or more heterologous amino acid sequences; and comprising a coupling element suitable for coupling to a support or carrier agent.
  • a protein or polypeptide consisting essentially of the following components: i) a Notch ligand DSL domain; ii) 3 Notch ligand EGF domains; iii) optionally all or part of a Notch ligand N-terminal domain; and iv) optionally one or more heterologous amino acid sequences; and comprising a coupling element suitable for coupling to a support or carrier agent.
  • a protein or polypeptide comprising: i) a Notch ligand DSL domain; ii) 1-5 and no more than 5 Notch ligand EGF domains; iii) optionally all or part of a Notch ligand N-terminal domain; and iv) optionally one or more heterologous amino acid sequences; and comprising a coupling element suitable for coupling to a support or carrier agent.
  • a protein or polypeptide comprising: i) a Notch ligand DSL domain; ii) 2-4 and no more than 4 Notch ligand EGF domains; iii) optionally all or part of a Notch ligand N-terminal domain; and iv) optionally one or more heterologous amino acid sequences; and comprising a coupling element suitable for coupling to a support or carrier agent.
  • a protein or polypeptide comprising: i) a Notch ligand DSL domain; ii) 2-3 and no more than 3 Notch ligand EGF domains; iii) optionally all or part of a Notch ligand N-terminal domain; and iv) optionally one or more heterologous amino acid sequences; and comprising a coupling element suitable for coupling to a support or carrier agent.
  • a protein or polypeptide comprising: i) a Notch ligand DSL domain; ii) 3 and no more than 3 Notch ligand EGF domains; iii) optionally all or part of a Notch ligand N-terminal domain; and iv) optionally one or more heterologous amino acid sequences; and comprising a coupling element suitable for coupling to a support or carrier agent.
  • the coupling agent is suitable for chemical coupling, such as a chemically reactive element.
  • the coupling agent may be suitable for adsorption coupling, for example suing electrostatic or hydrophobic interactions, or affinity coupling, for example using antibodies.
  • the coupling agent is at the C-terminus of the protein or polypeptide.
  • the coupling agent is a C-terminal cysteine, aspartate or glutamate residue.
  • the protein or polypeptide has at least 50%, preferably at least 70%, preferably at least 90%, for example at least 95% amino acid sequence similarity (or preferably sequence identity) to the following sequence along the entire length of the latter:
  • a pharmaceutically acceptable support matrix bearing a multiplicity of proteins or polypeptides as described above, said proteins being coupled to the support matrix.
  • the coupling may be chemical coupling, affinity coupling or adsorption coupling.
  • the support matrix may be a particulate support matrix, preferably a bead, preferably a microbead or nanobead.
  • a bead coupled to a protein or polypeptide as described above.
  • the bead has a diameter of from about 0.001 to about 1000 micrometres.
  • the bead is a polymeric bead.
  • the bead comprises a biodegradable material.
  • the bead comprises polystyrene, polyacrylamide, latex, cellulose, silica, dextran, agarose, cellulose, polylactide, or poly(methylmethacrylate) (PMMA) optionally in modified, crosslinked or derivatized form.
  • PMMA poly(methylmethacrylate)
  • compositions eg for in vivo use, comprising a particle or bead as described.
  • plurality means a number being at least two, and preferably at least five, suitably at least ten, twenty or more.
  • multiplex means a number being at least three, and preferably at least five, suitably at least ten, for example at least twenty or a hundred or more.
  • the construct used in the various embodiments of the invention comprises at least 3, preferably at least 5, suitably at least 10, suitably at least 20, for example 100 or more modulators of Notch signalling which may be the same or different.
  • the construct comprises a multiplicity of modulators of Notch signalling bound to a substrate.
  • the substrate is a particulate substrate, such as a bead.
  • the substrate may be biodegradable, especially where used in vivo.
  • such a particle or bead has a diameter (or for a collection of particles or beads, an average diameter) of from about 0.001 to about 1000 micrometres, suitably from 0.01 to 100 micrometres.
  • the particle or bead may suitably be a microbead or nanobead or microsphere or nanosphere.
  • the particle or bead is a polymeric particle or bead.
  • the particle or bead may comprise polystyrene, polyacrylamide, latex, cellulose, silica, dextran, agarose, cellulose, polylactide, or poly(methylmethacrylate) (PMMA) optionally in modified, crosslinked or derivatized form.
  • PMMA poly(methylmethacrylate)
  • the particle or bead comprises a biodegradable material.
  • the particle or bead is in sterile form.
  • Modulators of the Notch signalling pathway may be administered therapeutically on pharmaceutically acceptable support matrices.
  • the invention provides a pharmaceutically acceptable support matrix suitable for in vivo administration which bears a modulator of Notch signalling.
  • the support matrix may be in the form of an implantable support matrix.
  • the support matrix may be in the form of a particle or bead.
  • the support matrix may bear a Notch ligand proteins or polypeptides, preferably a multiplicity of Notch ligand proteins or polypeptides.
  • a Notch ligand proteins or polypeptides preferably a multiplicity of Notch ligand proteins or polypeptides.
  • Such support matrices, particles, beads etc may be administered either in vivo or ex-vivo as well known in the art (for example as described herein under the heading "Phannaceutical Compositions") and used to modulate the Notch signalling pathway (for example to treat conditions as described herein under the heading "Therapy").
  • the modulator of Notch signalling is an agent capable of activating Notch signalling.
  • the agent is capable of activating Notch signalling in lymphocytes, preferably T-cells.
  • the agent may act to reduce activity of effector T-cells such as Th or Tc T-cells, and/or increase activity of regulatory T-cells.
  • each individual modulator may be the same or different to each of the others.
  • the cells used in the present invention are not stem cells.
  • the modulator of Notch signalling is an agent capable of activating a Notch receptor, such as a Notch 1, Notch 2, Notch 3 or Notch 4 receptor.
  • the modulator may be a Notch ligand or a biologically active fragment or derivative of a Notch ligand, or a peptidomimetic of such a Notch ligand.
  • the agent is capable of activating Notch receptors in lymphocytes such as T-cells.
  • the modulator of the Notch signalling pathway may comprise or code for a fusion protein.
  • the modulator may comprise or code for a fusion protein comprising a segment of a Notch ligand extracellular domain and an immunoglobulin F c segment.
  • the modulator of the Notch signalling pathway may comprise a fusion protein comprising a segment of a Notch ligand extracellular domain and an immunoglobulin F c segment (eg IgGl Fc or IgG4 Fc) or a polynucleotide coding for such a fusion protein.
  • a fusion protein comprising a segment of a Notch ligand extracellular domain and an immunoglobulin F c segment (eg IgGl Fc or IgG4 Fc) or a polynucleotide coding for such a fusion protein.
  • Suitable such fusion proteins are described, for example in Example 2 of WO 98/20142.
  • IgG fusion proteins may be prepared as well known in the art, for example, as described in US 5428130 (Genentech).
  • the modulator of the Notch signalling pathway comprises or codes for a protein or polypeptide comprising a Notch ligand DSL domain or a fragment, derivative, homologue, analogue or allelic variant thereof.
  • the modulator of the Notch signalling pathway comprises or codes for a Notch ligand DSL domain and at least one EGF-like domain, suitably at least 1 to 20, suitably at least 2 to 16, for example at least 2 to 10, for example from 2 to 5 EGF-like domains.
  • the DSL and EGF sequences are or correspond to mammalian sequences.
  • Preferred sequences include human sequences.
  • the Notch ligand domains are fused to a heterologous amino acid sequence such as an IgFc sequence.
  • the modulator of the Notch signalling pathway may comprise a Notch intracellular domain (Notch IC) or a fragment, derivative, homologue, analogue or allelic variant thereof, or a polynucleotide sequence which codes for Notch intracellular domain or a fragment, derivative, homologue, analogue or allelic variant thereof.
  • Notch IC Notch intracellular domain
  • a fragment, derivative, homologue, analogue or allelic variant thereof or a polynucleotide sequence which codes for Notch intracellular domain or a fragment, derivative, homologue, analogue or allelic variant thereof.
  • a modulator of the Notch signalling pathway comprises Delta or a fragment, derivative, homologue, analogue or allelic variant thereof or a polynucleotide encoding Delta or a fragment, derivative, homologue, analogue or allelic variant thereof.
  • a modulator of the Notch signalling pathway may comprise Senate/Jagged or a fragment, derivative, homologue, analogue or allelic variant thereof or a polynucleotide encoding Senate/Jagged or a fragment, derivative, homologue, analogue or allelic variant thereof.
  • a modulator of the Notch signalling pathway may comprise Notch or a fragment, derivative, homologue, analogue or allelic variant thereof or a polynucleotide encoding Notch or a fragment, derivative, homologue, analogue or allelic variant thereof.
  • a modulator of the Notch signalling pathway may comprise a dominant negative version of a Notch signalling repressor, or a polynucleotide which codes for a dominant negative version of a Notch signalling repressor.
  • a modulator of the Notch signalling pathway may comprise a polypeptide capable of upregulating the expression or activity of a Notch ligand or a downstream component of the Notch signalling pathway, or a polynucleotide which codes for such a polypeptide.
  • the modulator of the Notch signalling pathway may comprise an antibody, antibody fragment or antibody derivative or a polynucleotide which codes for an antibody, antibody fragment or antibody derivative.
  • a support such as a bead, plate, well
  • the modulator of Notch signalling may be administered in a multimerised form.
  • the modulator of Notch signalling may be bound to a membrane or support.
  • a plurality or multiplicity of modulators for example at least 5, 10, 20 or 100
  • the support may be a particulate support matrix.
  • the support may be a bead.
  • the bead may be, for example, a magnetic bead (e.g. as available under the trade name "Dynal").
  • the modulation of the immune system comprises reduction of T cell activity.
  • the modulation of the immune system may comprise reduction of effector T-cell activity, for example reduction of helper (T H ) and/or cytotoxic (Tc) T-cell activity.
  • the modulation of the immune system may comprise reduction of a Thl or Th2 immune response.
  • the modulation of the immune system provides an increase of regulatory T-cell (T reg) activity, such as an increase of Trl or Th3 regulatory T-cell activity.
  • the modulation of the immune system comprises generation of regulatory T cells (Tregs) and/or enhancement of Treg activity.
  • Tregs regulatory T cells
  • the modulation of the immune system comprises treatment of asthma, allergy, graft rejection, graft- versus-host disease or autoimmune disease.
  • a method for producing a lymphocyte or antigen presenting cell (APC) capable of promoting tolerance comprises incubating a lymphocyte or APC obtained from a human or animal patient with a modulator of the Notch signalling pathway as described above.
  • APC antigen presenting cell
  • the method comprises incubating a lymphocyte or APC obtained from a human or animal patient with an APC in the presence of a modulator of the Notch signalling pathway as described above.
  • a method for producing an APC capable of inducing tolerance in a T cell comprises contacting an APC with a modulator of the Notch signalling pathway as described above.
  • a method for producing a lymphocyte or APC capable of promoting tolerance comprises incubating a lymphocyte or APC obtained from a human or animal patient with a lymphocyte or APC produced as described above.
  • the lymphocyte or APC may be incubated either in vivo or ex-vivo.
  • a particle such as a bead, including microbeads and nanobeads
  • a particle comprising a plurality, preferably a multiplicity of Delta proteins or polypeptides bound to a particulate support matrix.
  • the term "Delta protein or polypeptide” as used herein suitably includes a protein or polypeptide which has at least one DSL domain from a Delta Notch ligand such as Deltal, Delta3 or Delta4, and suitably at least one, preferably at least two, for example 2 to 10 EGF-like domains from a Delta Notch ligand.
  • the Delta Notch ligand is or is derived from a verterbrate, preferably a mammalian Notch ligand sequence, for example a Xenopus, mouse or human sequence.
  • Figure 1 shows a schematic representation of the Notch signalling pathway
  • Figure 2 shows schematic representations of the Notch ligands Jagged and Delta
  • Figure 3 shows aligned amino acid sequences of DSL domains from various Drosophila and mammalian Notch ligands
  • Figure 4 shows the amino acid sequences of human Delta-1, Delta-3 and Delta-4;
  • Figure 5 shows the amino acid sequences of human Jagged-1 and Jagged-2
  • Figure 6 shows schematic representations of various Notch ligand domain/IgFc domain fusion proteins which may be used in the present invention
  • Figure 7 shows a reaction scheme for covalently linking modulators of Notch signalling to beads.
  • Figure 8 shows schematic representations of non-covalent linking of modulators of Notch signalling to beads (using a streptavidin/biotin link).
  • Notch signalling is synonymous with the expression “the Notch signalling pathway” and refers to any one or more of the upstream or downstream events that result in, or from, (and including) activation of the Notch receptor.
  • Notch signalling directs binary cell fate decisions in the embryo. Notch was first described in Drosophila as a transmembrane protein that functions as a receptor for two different ligands, Delta and Senate. Vertebrates express multiple Notch receptors and ligands. At least four Notch receptors (Notch- 1, Notch-2, Notch-3 and Notch-4) have been identified to date in human cells.
  • Notch proteins are synthesized as single polypeptide precursors that undergo cleavage via a Furin-like convertase that yields two polypeptide chains that are further processed to form the mature receptor.
  • the Notch receptor present in the plasma membrane comprises a heterodimer of two Notch proteolytic cleavage products, one comprising an N-terminal fragment consisting of a portion of the extracellular domain, the transmembrane domain and the intracellular domain, and the other comprising the majority of the exfracellular domain.
  • the proteolytic cleavage step of Notch to activate the receptor occurs and is mediated by a furin-like convertase.
  • Notch receptors are inserted into the membrane as disulphide-linked heterodimeric molecules consisting of an extracellular domain containing up to 36 epidermal growth factor (EGF)-like repeats and a transmembrane subunit that contains the cytoplasmic domain.
  • the cytoplasmic domain of Notch contains six ankyrin-like repeats, a polyglutamine stretch (OP A) and a PEST sequence.
  • a further domain termed RAM23 lies proximal to the ankyrin repeats and, like the ankyrin-like repeats, is involved in binding to a transcription factor, known as Suppressor of Hairless [Su(H)J in Drosophila and CBF1 in vertebrates (Tamura).
  • the Notch ligands also display multiple EGF-like repeats in their extracellular domains together with a cysteine-rich DSL (Delta-Serrate Lag2) domain that is characteristic of all Notch ligands (Artavanis-Tsakonas).
  • the Notch receptor is activated by binding of exfracellular ligands, such as Delta, Senate and Scabrous, to the EGF-like repeats of Notch's extracellular domain. Delta requires cleavage for activation. It is cleaved by the ADAM disintegrin metalloprotease Kuzbanian at the cell surface, the cleavage event releasing a soluble and active form of Delta.
  • Su(H) is the Drosophila homologue of C-promoter binding factor- 1 [CBF- 1], a mammalian DNA binding protein involved in the Epstein-Barr virus-induced immortalization of B-cells. It has been demonstrated that, at least in cultured cells, Su(H) associates with the cdclO/ankyrin repeats in the cytoplasm and translocates into Hie nucleus upon the interaction of the Notch receptor with its ligand Delta on adjacent cells. Su(H) includes responsive elements found in tlie promoters of several genes and has been found to be a critical downstream protein in the Notch signalling pathway. The involvement of Su(H) in transcription is thought to be modulated by Hairless.
  • NotchIC The intracellular domain of Notch (NotchIC) also has a direct nuclear function (Lieber). Recent studies have indeed shown that Notch activation requires that the six cdcl 0/ankyrin repeats of the Notch intracellular domain reach the nucleus and participate in franscriptional activation.
  • the site of proteolytic cleavage on the intracellular tail of Notch has been identified between glyl743 and vall744 (tenned site 3, or S3) (Schroeter). It is thought that the proteolytic cleavage step that releases the NotchIC for nuclear entry is dependent on Presenilin activity.
  • the intracellular domain has been shown to accumulate in the nucleus where it forms a transcriptional activator complex with the CSL family protein CBF1 (suppressor of hairless, Su(H) in Drosophila, Lag-2 in C. elegans) (Schroeter; Struhl).
  • CSL family protein CBF1 suppressor of hairless, Su(H) in Drosophila, Lag-2 in C. elegans
  • the NotchlC-CBFl complexes then activate target genes, such as the bHLH proteins HES (hairy-enhancer of split like) 1 and 5 (Weinmaster).
  • This nuclear function of Notch has also been shown for the mammalian Notch homologue (Lu).
  • NotchIC processing occurs only in response to binding of Notch ligands Delta or Serrate/ Jagged.
  • the post-translational modification of the nascent Notch receptor in the Golgi appears, at least in part, to confrol which of the two types of ligand it interacts with on a cell surface.
  • the Notch receptor is modified on its exfracellular domain by Fringe, a glycosyl fransferase enzyme that binds to the Notch/Lin motif. Fringe modifies Notch by adding O-linked fucose groups to the EGF-like repeats (Moloney; Bruckner). This modification by Fringe does not prevent ligand binding, but may influence ligand induced conformational changes in Notch. Furthermore, recent studies suggest that the action of
  • Fringe modifies Notch to prevent it from interacting functionally with Senate/ Jagged ligands but allow it to preferentially interact with Delta (Panin; Hicks).
  • Drosophila has a single Fringe gene, vertebrates are known to express multiple genes (Radical, Manic and Lunatic Fringes) (Irvine).
  • NotchIC proteolytic cleavage of the intracellular domain of Notch
  • CSL family protein CBF1 suppressor of hairless, Su(H) in Drosophila, Lag-2 in C. elegans
  • NotchlC-CBFl complexes then activate target genes, such as the bHLH proteins HES (hairy-enhancer of split like) 1 and 5.
  • Notch can also signal in a CBF1- independent manner that involves the cytoplasmic zinc finger containing protein Deltex ( Figure 3).
  • Deltex does not move to the nucleus following Notch activation but instead can interact with Grb2 and modulate the Ras-Jnk signalling pathway.
  • endogenous modulators of Notch include, for example, the Notch ligands Delta and Senate.
  • An aim of the present invention is the detection of novel Notch signalling modulators.
  • Notch ligand means an agent capable of interacting with a Notch receptor to cause a biological effect.
  • the term as used herein therefore includes naturally occurring protein ligands such as Delta and Serrate/ Jagged as well as antibodies to the Notch receptor, peptidomimetics and small molecules which have conesponding biological effects to the natural ligands.
  • the Notch ligand interacts with the Notch receptor by binding.
  • antibody as used herein includes intact molecules as well as fragments thereof, such as Fab, F(ab')2, Fv and scFv which are capable of binding the epitopic detenninant. These antibody fragments retain some ability to selectively bind with its antigen or receptor and include, for example:
  • Fab fragment which contains a monovalent antigen-binding fragment of an antibody molecule can be produced by digestion of whole antibody with the enzyme papain to yield an intact light chain and a portion of one heavy chain;
  • Fab' the fragment of an antibody molecule can be obtained by treating whole antibody with pepsin, followed by reduction, to yield an intact light chain and a portion of the heavy chain; two Fab' fragments are obtained per antibody molecule;
  • F(ab') 2 the fragment of the antibody that can be obtained by treating whole antibody with the enzyme pepsin without subsequent reduction;
  • F(ab') 2 is a dimer of two Fab' fragments held together by two disulfide bonds;
  • scFv including a genetically engineered fragment containing the variable region of a heavy and a light chain as a fused single chain molecule.
  • a homologue is meant a gene product that exhibits sequence homology, either amino acid or nucleic acid sequence homology, to any one of the known Notch ligands, for example as mentioned above.
  • a homologue of a known Notch ligand will be at least 20%, preferably at least 30%, identical at the amino acid level to the conesponding known Notch ligand over a sequence of at least 10, preferably at least 20, preferably at least 50, suitably at least 100 amino acids, or over the entire length of the Notch ligand.
  • Homologues of Notch ligands can be identified in a number of ways, for example by probing genomic or cDNA libraries with probes comprising all or part of a nucleic acid encoding a Notch ligand under conditions of medium to high stringency (for example 0.03M sodium chloride and 0.03M sodium citrate at from about 50°C to about 60°C).
  • medium to high stringency for example 0.03M sodium chloride and 0.03M sodium citrate at from about 50°C to about 60°C.
  • homologues may also be obtained using degenerate PCR which will generally use primers designed to target sequences within the variants and homologues encoding conserved amino acid sequences. The primers will contain one or more degenerate positions and will be used at stringency conditions lower than those used for cloning sequences with single sequence primers against known sequences.
  • a modulator of Notch signalling for use in the present invention will be a Notch receptor agonist such as a Notch ligand capable of binding to and activating a Notch receptor, preferably a human Notch receptor such as Notchl, Notcl ⁇ 2, Notch3 or Notch4.
  • a Notch receptor agonist such as a Notch ligand capable of binding to and activating a Notch receptor, preferably a human Notch receptor such as Notchl, Notcl ⁇ 2, Notch3 or Notch4.
  • binding and activation may be assessed by a variety of techniques known in the art including in vitro binding assays and activity assays for example as described herein.
  • Notch signalling preferably means specific signalling, meaning that the signalling results substantially or at least predominantly from the Notch signalling pathway, and preferably from Notch/Notch ligand interaction, rather than any other significant interfering or competing cause, such as cytokine signalling.
  • Notch signalling excludes cytokine signalling.
  • modulate refers to a change or alteration in the biological activity of the Notch signalling pathway or a target signalling pathway thereof.
  • modulator may refer to antagonists or inhibitors of Notch signalling, i.e. compounds which block, at least to some extent, the normal biological activity of the Notch signalling pathway. Conveniently such compounds may be refened to herein as inhibitors or antagonists.
  • modulator may refer to agonists of Notch signalling, i.e. compounds which stimulate or upregulate, at least to some extent, the normal biological activity of the Notch signalling pathway. Conveniently such compounds may be refened to as upregulators or agonists.
  • candidate modulator is used to describe any one or more molecule(s) which may be, or is suspected of being, capable of functioning as a modulator of Notch signalling.
  • Said molecules may for example be organic "small molecules” or polypeptides.
  • candidate molecules comprise a plurality of, or a library of such molecules or polypeptides. These molecules may be derived from known modulators. "Derived from” means that the candidate modulator molecules preferably comprise polypeptides which have been fully or partially randomised from a starting sequence which is a known modulator of Notch signalling.
  • candidate molecules comprise polypeptides which are at least 40% homologous, more preferably at least 60% homologous, even more preferably at least 75% homologous or even more, for example 85 %, or 90 %, or even more than 95% homologous to one or more known Notch modulator molecules, using the BLAST algorithm with the parameters as defined herein.
  • the candidate modulator of the present invention may be an organic compound or other chemical.
  • the candidate modulator will be an organic compound comprising two or more hydrocarbyl groups.
  • hydrocarbyl group means a group comprising at least C and H and may optionally comprise one or more other suitable substituents. Examples of such substituents may include halo-, alkoxy-, nitro-, an alkyl group, a cyclic group etc.
  • substituents may include halo-, alkoxy-, nitro-, an alkyl group, a cyclic group etc.
  • a combination of substituents may form a cyclic group. If the hydrocarbyl group comprises more than one C then those carbons need not necessarily be linked to each other.
  • the carbons may be linked via a suitable element or group.
  • the hydrocarbyl group may contain hetero atoms. Suitable hetero atoms will be apparent to those skilled in the art and include, for instance, sulphur, nitrogen and oxygen.
  • the candidate modulator may comprise at least one cyclic group.
  • the cyclic group may be a polycyclic group, such as a non-fused polycyclic group.
  • the agent comprises at least the one of said cyclic groups linked to another hydrocarbyl group.
  • the candidate compound will be an amino acid sequence or a chemical derivative thereof, or a combination thereof.
  • the candidate compound will be a nucleotide sequence, which may be a sense sequence or an anti-sense sequence.
  • the candidate modulator may also be an antibody.
  • Candidate modulators may be synthetic compounds or natural isolated compounds. Various examples of such synthetic or natural modulators are listed below.
  • Agonists of Notch signalling will include any molecule which is capable of up-regulating Notch, the Notch signalling pathway or any one or more of the components of the Notch signalling pathway.
  • Candidate modulators for up-regulating the Notch signalling pathway include compounds capable of transducing or activating the Notch signalling pathway.
  • Modulators for Notch signalling transduction will include molecules which participate in signalling through Notch receptors including activation of Notch, the downstream events of the Notch signalling pathway, transcriptional regulation of downstream target genes and other non-transcriptional downstream events (e.g. post-translational modification of existing proteins). More particularly, such modulators will allow activation of target genes of the Notch signalling pathway.
  • the modulator may be the Notch polypeptide or polynucleotide or a fragment, variant, derivative, mimetic or homologue thereof which retains the signalling transduction ability of Notch or an analogue of Notch which has the signalling transduction ability of Notch.
  • Notch we mean Notch- 1, Notch- 2, Notch-3, Notch-4 and any other Notch homologues or analogues.
  • Analogues of Notch include proteins from the Epstein Ba virus (EBV), such as EBNA2, BARFO or LMP2A.
  • the modulator may be the Notch intracellular domain (Notch IC) or a sub-fragment, variant, derivative, mimetic, analogue or homologue thereof.
  • Modulators for Notch signalling activation include molecules which are capable of activating Notch, the Notch signalling pathway or any one or more of the components of the Notch signalling pathway.
  • Such a modulator may be a dominant negative version of a Notch signalling repressor.
  • the modulator will be capable of inhibiting a Notch signalling repressor.
  • the modulator for Notch signalling activation will be a positive activator of Notch signalling.
  • the modulator will be capable of inducing or increasing Notch or Notch ligand expression.
  • a molecule may be a nucleic acid sequence capable of inducing or increasing Notch or Notch ligand expression.
  • the modulator will be capable of up-regulating expression of the endogenous genes encoding Notch or Notch ligands in target cells.
  • the modulator may be an immunosuppressive cytokine capable of up-regulating the expression of endogenous Notch or Notch ligands in target cells, or a polynucleotide which encodes such a cytokine.
  • Immunosuppressive cytokines include IL-10, IL-13, TGF-beta and FLT3 ligand.
  • Candidate modulators will therefore further include fragments, derivatives, variants, mimetics, analogues and homologues of any of the above.
  • Endogenous agonists include Noggin, Chordin, Follistatin, Xnr3, fibroblast growth factors.
  • Candidate modulators may therefore include derivatives, fragments, variants, mimetics, analogues and homologues thereof, or a polynucleotide encoding any one or more of the above.
  • the modulator may be a Notch ligand, or a polynucleotide encoding a Notch ligand.
  • Notch ligands will typically be capable of binding to a Notch receptor polypeptide present in the membrane of a variety of mammalian cells, for example hemapoietic stem cells.
  • Endogenous Notch ligands include polypeptides of the Delta family, for example Delta-1 (Genbank Accession No. AF003522 - Homo sapiens), Delta-3 (Genbank Accession No. AF084576 - Rattus norvegicus), Delta-like 3 (Mus musculus), Delta-4 (Genbank Accession No.
  • Candidate compounds of the present invention include fragments, derivatives, variants, mimetics, analogues and homologues of any of the above.
  • the modulator will be a constitutively active Notch receptor or Notch intracellular domain, or a polynucleotide encoding such a receptor or intracellular domain.
  • the modulator of Notch signalling will act downstream of the Notch receptor.
  • the activator of Notch signalling may be a constitutively active Deltex polypeptide or a polynucleotide encoding such a polypeptide.
  • Notch signalling pathway Other endogenous downstream components of the Notch signalling pathway include Deltex- 1, Deltex-2, Deltex-3, Suppressor of Deltex (SuDx), Numb and isoforms thereof, Numb associated Kinase (NAK), Notchless, Dishevelled (Dsh), emb5, Fringe genes (such as Radical, Lunatic and Manic), PON, LNX, Disabled, Numblike, Nur77, NFkB2, Mirror, Warthog, Engrailed- 1 and Engrailed-2, Lip-1 and homologues thereof, the polypeptides involved in the Ras/MAPK cascade modulated by Deltex, polypeptides involved in the proteolytic cleavage of Notch such as Presenilin and polypeptides involved in the transcriptional regulation of Notch target genes.
  • Candidate modulators of use in the present invention will therefore include constitutively active forms of any of the above, analogues, homologues, derivatives, variants, mimetics and fragments thereof.
  • Modulators for Notch signalling activation may also include any polypeptides expressed as a result of Notch activation and any polypeptides involved in the expression of such polypeptides, or polynucleotides encoding for such polypeptides.
  • Activation of Notch signalling may also be achieved by repressing inhibitors of the Notch signalling pathway.
  • candidate modulators will include molecules capable of repressing any Notch signalling inhibitors.
  • the molecule will be a polypeptide, or a polynucleotide encoding such a polypeptide, that decreases or interferes with the production or activity of compounds that are capable of producing an decrease in the expression or activity of Notch, Notch ligands, or any downstream components of the Notch signalling pathway.
  • the modulators will be capable of repressing polypeptides of the Toll-like receptor protein family, cytokines such as IL-12, LFN- ⁇ , TNF- ⁇ , and growth factors such as the bone morphogenetic protein (BMP), BMP receptors and activins.
  • cytokines such as IL-12, LFN- ⁇ , TNF- ⁇
  • growth factors such as the bone morphogenetic protein (BMP), BMP receptors and activins.
  • BMP bone morphogenetic protein
  • the modulator of the present invention will be a polypeptide or a polynucleotide. Whether or not any given agent acts as a modulator of Notch signalling (and if so whether it is an activator or inhibitor of such signalling) may be readily determined by use of suitable assays or screens, for example, those described in the Examples herein.
  • Notch ligands typically comprise a number of distinctive domains. Some predicted/potential domain locations for various naturally occurring human Notch ligands (based on amino acid numbering in the precursor proteins) are shown below:
  • a typical DSL domain may include most or all of the following consensus amino acid sequence:
  • DSL domain may include most or all of the following consensus amino acid sequence:
  • ARO is an aromatic amino acid residue, such as tyrosine, phenylalanine, tryptophan or histidine;
  • NOP is a non-polar amino acid residue such as glycine, alanine, proline, leucine, isoleucine or valine;
  • BAS is a basic amino acid residue such as arginine or lysine.
  • ACM is an acid or amide amino acid residue such as aspartic acid, glutamic acid, asparagine or glutamine.
  • DSL domain may include most or all of the following consensus amino acid sequence:
  • Xaa may be any amino acid and Asx is either aspartic acid or asparagine).
  • the DSL domain used may be derived from any suitable species, including for example Drosophila, Xenopus, rat, mouse or human.
  • the DSL domain is derived from a vertebrate, preferably a mammalian, preferably a human Notch ligand sequence.
  • a DSL domain for use in the present invention may have at least 30%>, preferably at least 50%, preferably at least 60%, preferably at least 70%, preferably at least 80%), preferably at least 90%, preferably at least 95% amino acid sequence identity to the DSL domain of human Jagged 1.
  • a DSL domain for use in the present invention may, for example, have at least 30%), preferably at least 50%, preferably at least 60%, preferably at least 70%, preferably at least 80%), preferably at least 90%, preferably at least 95% amino acid sequence identity to the DSL domain of human Jagged 2.
  • a DSL domain for use in the present invention may, for example, have at least 30%, preferably at least 50%, preferably at least 60%, preferably at least 70%, preferably at least 80%, preferably at least 90%, preferably at least 95% amino acid sequence identity to the DSL domain of human Delta 1.
  • a DSL domain for use in the present invention may, for example, have at least 30%, preferably at least 50%, preferably at least 60%>, preferably at least 70%, preferably at least 80%, preferably at least 90%, preferably at least 95% amino acid sequence identity to the DSL domain of human Delta 3.
  • a DSL domain for use in the present invention may, for example, have at least 30%, preferably at least 50%, preferably at least 60%, preferably at least 70%>, preferably at least 80%, preferably at least 90%, preferably at least 95%o amino acid sequence identity to the DSL domain of human Delta 4.
  • Notch ligand N-terminal domain means the part of a Notch ligand sequence from the N-terminus to the start of the DSL domain. It will be appreciated that this term includes sequence variants, fragments, derivatives and mimetics having activity conesponding to naturally occurring domains.
  • heterologous amino acid sequence or “heterologous nucleotide sequence” as used herein means a sequence which is not found in the native sequence (e.g. in the case of a Notch ligand sequence is not found in the native Notch ligand sequence) or its coding sequence.
  • any such heterologous amino acid sequence is not a TSST sequence, and preferably it is not a superantigen sequence.
  • the EGF-like motif has been found in a variety of proteins, as well as EGF and Notch and Notch ligands, including those involved in the blood clotting cascade (Furie and Furie, 1988, Cell 53: 505-518).
  • this motif has been found in exfracellular proteins such as the blood clotting factors IX and X (Rees et al., 1988, EMBO J. 7:2053-2061; Furie and Furie, 1988, Cell 53: 505-518), in other Drosophila genes (Knust et al., 1987 EMBO J.
  • EGF-like domain may include six cysteine residues which have been shown (in EGF) to be involved in disulfide bonds.
  • the main structure is proposed, but not necessarily required, to be a two-stranded beta-sheet followed by a loop to a C-terminal short two-stranded sheet.
  • Subdomains between the conserved cysteines strongly vary in length as shown in the following schematic representation of a typical EGF-like domain: I I I I x ( ) -C-x (0 , 8) -C-x (3 , 12) -C-x (1 , 70) -C-x (1 , 6) -C-x (2) -G-a-x (0 , 21) -G-x (2) -C-x I i*****************************
  • 'C conserved cysteine involved in a disulfide bond.
  • 'G' often conserved glycine 'a': often conserved aromatic amino acid '*': position of both patterns, 'x': any residue
  • the region between the 5th and 6th cysteine contains two conserved glycines of which at least one is normally present in most EGF-like domains.
  • the EGF-like domain used may be derived from any suitable species, including for example Drosophila, Xenopus, rat, mouse or human.
  • the EGF-like domain is derived from a vertebrate, preferably a mammalian, preferably a human Notch ligand sequence.
  • an EGF-like domain for use in the present invention may have at least 30%, preferably at least 50%, preferably at least 60%, preferably at least 70%, preferably at least 80%), preferably at least 90%>, preferably at least 95% amino acid sequence identity to an EGF-like domain of human Jagged 1.
  • an EGF-like domain for use in the present invention may, for example, have at least 30%), preferably at least 50%>, preferably at least 60%, preferably at least 70%, preferably at least 80%, preferably at least 90%>, preferably at least 95% amino acid sequence identity to an EGF-like domain of human Jagged 2.
  • an EGF-like domain for use in the present invention may, for example, have at least 30%>, preferably at least 50%>, preferably at least 60%, preferably at least 70%>, preferably at least 80%, preferably at least 90%>, preferably at least 95% amino acid sequence identity to an EGF-like domain of human Delta 1.
  • an EGF-like domain for use in the present invention may, for example, have at least 30%, preferably at least 50%, preferably at least 60%, preferably at least 70%, preferably at least 80%, preferably at least 90%, preferably at least 95% amino acid sequence identity to an EGF-like domain of human Delta 3.
  • an EGF-like domain for use in the present invention may, for example, have at least 30%, preferably at least 50%>, preferably at least 60%, preferably at least 70%, preferably at least 80%, preferably at least 90%>, preferably at least 95% amino acid sequence identity to an EGF-like domain of human Delta 4.
  • the modulator of Notch signalling may be an antibody, derivative or fragment which binds to and activates Notch.
  • the invention also provides a support (eg bead, plate or well, preferably a bead) to which is coupled (eg chemically, by affinity or adsportion) an antibody capable of binding to and activating Notch.
  • Antibodies may be monoclonal or polyclonal but are preferably monoclonal.
  • the binding affinity (equilibrium association constant (Ka)) may be at least about 10 6 M “1 , at least about 10 7 M “1 , at least about 10 8 M “1, or at least about 10 9 M “1 .
  • the antibody, derivative or fragment binds to one or more EGF or Lin Notch (L/N) domains of Notch (for example to EGF repeats 11 and 12 of Notch).
  • L/N Lin Notch
  • Suitable antibodies for use as blocking agents are obtained by immunizing a host animal with peptides comprising all or a portion of Notch.
  • the peptide used may comprise the complete protein or a fragment or derivatives thereof.
  • Prefened immunogens comprise all or a part of the extracellular domain of human Notch (eg Notchl, Notch2, Notch3 or Notch4, preferably Notchl or Notch2), where these residues contain any post-translation modifications, such as glycosylation, found in the native proteins.
  • Immunogens comprising the exfracellular domain may be produced by a number of techniques which are well known in the art such as expression of cloned genes using conventional recombinant methods and or isolation from T cells or cell populations expressing high levels of Notch.
  • Monoclonal antibodies may be produced by means well known in the art.
  • the spleen and/or lymph nodes of an immunized host animal provide a source of plasma cells.
  • the plasma cells are immortalized by fusion with myeloma cells to produce hybridoma cells.
  • Culture supernatant from individual hybridomas is screened using standard techniques to identify those producing antibodies with the desired specificity.
  • the antibody may be purified from the hybridoma cell supernatants or ascites fluid by conventional techniques, such as affinity chromatography using Notch, Notch ligands or fragments thereof bound to an insoluble support, protein A sepharose, or the like.
  • WO 0020576 discloses a monoclonal antibody secreted by a hybridoma designated A6 having the ATCC Accession No. HB 12654, a monoclonal antibody secreted by a hybridoma designated Cll having the ATCC Accession No. HB 12656 and a monoclonal antibody secreted by a hybridoma designated F3 having the ATCC Accession No. HB12655.
  • antibodies for use to treat human patients in vivo will be chimeric or humanised antibodies.
  • Antibody "humanisation” techniques are well known in the art. These techniques typically involve the use of recombinant DNA technology to manipulate DNA sequences encoding the polypeptide chains of the antibody molecule.
  • CDRs complementarity determining regions
  • a mouse MAb is grafted onto the framework regions of the variable domains of a human immunoglobulin by site directed mutagenesis using long oligonucleotides.
  • CDR-grafted humanised antibodies are much less likely to give rise to an anti-antibody response than humanised chimeric antibodies in view of the much lower proportion of non-human amino acid sequence which they contain.
  • the first criterion is to use as the human acceptor the framework from a particular human immunoglobulin that is unusually homologous to the non-human donor immunoglobulin to be humanised, or to use a consensus framework from many human antibodies.
  • the second criterion is to use the donor amino acid rather than the acceptor if the human acceptor residue is unusual and the donor residue is typical for human sequences at a specific residue of the framework.
  • the third criterion is to use the donor framework amino acid residue rather than the acceptor at positions immediately adjacent to the CDRs.
  • the fourth criterion is to use the donor amino acid residue at framework positions at which the amino acid is predicted to have a side chain atom within about 3 A of the CDRs in a three- dimensional immunoglobulin model and to be capable of interacting with the antigen or with the CDRs of the humanised immunoglobulin. It is proposed that criteria two, three or four may be applied in addition or alternatively to criterion one, and may be applied singly or in any combination.
  • isotype will be guided by the desired effector functions, such as complement fixation, or activity in antibody-dependent cellular cytotoxicity.
  • Suitable isotypes include IgG 1, IgG3 and IgG4.
  • IgG 1 IgG3
  • IgG4 IgG4
  • either of the human light chain constant regions, kappa or lambda may be used.
  • the invention may utilise a construct comprising a multiplicity of modulators of Notch signalling in cross-linked form.
  • the modulators of Notch signalling may, for example, be linked to each other directly or indirectly.
  • Linkages may be covalent or non-covalent (e.g., via electrostatic and/or hydrophobic interactions).
  • direct linkage between modulators of Notch signalling is achieved by chemical cross-linking.
  • Suitable chemical cross-linking procedures are well-known in the art; see, for example, Carlsson J. et al., Biochem. J. 173:723-737, 1978; Cumber, J. A. et al. Methods in Enzymology 112:207-224, 1985; Walden, P. et al., J. Mol. Cell Immunol. 2:191- 197, 1986; Gordon, R. D. et al., Proc. Natl. Acad. Sci. (USA) 84:308-312,1987; Avrameas, S.
  • direct linkage may be achieved by the design and expression of a recombinant chimeric gene encoding a multiplicity of modulators of Notch signalling.
  • the chimeric gene is then expressed in a suitable expression system.
  • the spacer may comprise an antibody, which may, for example, be a monofunctional or bifunctional antibody or antibody derivative.
  • the invention may utilise a construct comprising a multiplicity of modulators of Notch signalling bound to a subsfrate.
  • the substrate can take many different forms such as polymers, plastics, porous materials such as resin or modified cellulose, beads (such as microspheres and microbeads, nanospheres and nanoparticles), laboratory plates and wells and liposomes.
  • the support may be a plate such as a microtiter plate, or for example a well or other suitable container. At least part of the surface may be coated with modulators of Notch signalling bound by covalent or non-covalent means.
  • the substrate may be a particulate subsfrate, such as a bead, sphere, particle or carrier, for example having a diameter (or, for example, within a collection of beads, a mean diameter) of from about 0.001 to about 1000 micrometres, for example from about 0.01 to about 100 micrometres, suitably from about 0.1 to 10 micrometres, for example about 1 to 10 micrometres.
  • Particulate materials such as beads have the advantages of being easier to handle in certain situations, and of potentially providing a larger surface area for interaction with cells. They may also be more suitable for in-vivo applications, especially when the substrate comprises a biodegradable material.
  • diameter normally applies to particles having a substantially spherical or other circular form.
  • particles used in the present invention do not need to have such a regular form, and may have a more inegular form, in which case the relevant dimension is suitably the largest linear dimension.
  • a prefened particle or bead size is from 20-1000 nm, for example about lOOnm (0.1 microns).
  • Substrates may, for example, comprise natural or synthetic polymers such as polystyrene, polyethylene glycol (PEG), polyglycollic acid (PGA), polycaprolactide, polyacrylamide, latex, silica, dextran, agarose, starch, cellulose, chitin/chitosan, polylactide, poly(methylmethacrylate) (PMMA); proteins/polypeptides such as albumins, for example human serum albumins; and modified, crosslinked and derivatized embodiments thereof.
  • natural or synthetic polymers such as polystyrene, polyethylene glycol (PEG), polyglycollic acid (PGA), polycaprolactide, polyacrylamide, latex, silica, dextran, agarose, starch, cellulose, chitin/chitosan, polylactide, poly(methylmethacrylate) (PMMA); proteins/polypeptides such as albumins, for example human serum albumins; and modified, crosslinked and derivat
  • Suitable materials include, for example polystyrene, cellulose, dextran crosslinked with epichlorohydrin (Sephadex.TM., Pharmacia, Uppsala, Sweden), polyacrylamide crosslinked with bisacrylamide (Biogel.TM., BioRad, USA), agar, glass beads, polylactide beads and latex beads.
  • Derivatized microparticles include microparticles derivatized with eg maleimide, aldehyde groups, allyl groups, carboxyalkyl groups such as carboxymethyl, phosphoryl and substituted phosphoryl groups, sulfate, sulfhydryl and sulfonyl groups, and amino and substituted amino groups.
  • Beads may have either hydrophilic or hydrophobic properties.
  • the modulators of Notch signalling may be bound to the substrate (eg bead) by any suitable means.
  • binding may be by non-covalent linking such as surface adsorption (eg by hydrophobic and/or electrostatic interactions) or by covalent linking such as chemical linking.
  • reactive groups such as amino, aldehyde, carboxy, epoxy or toluenesulfonyl (tosyl), thiol or maleimide groups
  • modulators of Notch signalling in the form of proteins or polypeptides may be linked to a substrate (such as a bead) by incubation of a surface activated substrate (such as a bead) with the protein or polypeptide, suitably by incubation for at least 12 to 24 hours suitably at neutral or neutral to high pH and suitably at a sufficiently high temperature such that a reactive group on the substrate/bead (for example a tosyl, epoxy, amino, carboxy, aldehyde, thiol or maleimide group) reacts with a reactive group on the protein or polypeptide (for example a free amino or sulfhydryl group on the protein or polypeptide, or an aldehyde group) to form a covalent link.
  • a reactive group on the substrate/bead for example a tosyl, epoxy, amino, carboxy, aldehyde, thiol or maleimide group
  • a reactive group on the protein or polypeptide for example a
  • linker groups may also be used to bind the substrate/particle/bead to the modulators of Notch signalling if required.
  • Suitable linkers are well known in the art and suitably comprise an acid, basic, aldehyde, ether or ester reactive group or a residue thereof.
  • Suitable linker moieties include, for example, succinimidyl propionate, succinimidyl butanoate, N-hydroxysuccinimide, benzotriazole carbonate, propionaldehyde, maleimide or forked maleimide, biotin, vinyl derivative and phospholipids.
  • modulators of Notch signalling such as Notch ligand proteins and polypeptides may be chemically coupled to beads using a coupling agent such as sulpho-SMCC
  • Antibodies may also be used to bind modulators of Notch signalling to a subsfrate (such as a bead).
  • a subsfrate such as a bead
  • beads coated with antibody-binding materials such as sfreptavidin may be used to bind a biotinylated anti-IgG antibody, which in turn may be bound to a modulator of Notch signalling in the form of an IgG fusion protein.
  • suitably functionalised agents such as biotinylated agents/ligands may be directly coupled to beads such as sfreptavidin beads.
  • particles or beads may be magnetic particles such as magnetic beads, such as for example those available under the trade name DYNABEAD TM from Dynal Biotech, Oslo, Norway. These have the advantage of being easy to separate from fluids magnetically, which can be particularly advantageous for use ex-vivo uses.
  • Such particles or beads may be prepared, for example, by incorporating a magnetic or paramagnetic material such as iron oxide into a polymeric matrix such as polystyrene.
  • DynabeadsTM are superparamagnetic polymer spheres, magnetic only when placed in a magnetic field and with no residual magnetism when the magnetic field is removed. They are composed of highly cross-linked polystyrene with magnetic material precipitated in pores evenly distributed throughout each bead. The pores are preferably filled with an additional polymer layer, which seals the iron material inside the beads. Chemical groups (such as amino, carboxy, epoxy or toluenesulfonyl (tosyl) groups) may then be introduced on the bead surface if appropriate.
  • Particles or beads may be either hydrophilic or hydrophobic. Hydrophilic particles or beads
  • Hydrophobic particles or beads are well suited to coupling of antibodies.
  • the hydrophobic Fc region of the antibody may be adsorbed to the hydrophobic particle or bead surface, followed by a rapid covalent bond formation.
  • the orientation of the antibody is thereby generally optimal with the Fab regions facing outwards.
  • particles or beads may also need to be taken into consideration.
  • Hydrophobic particles or beads facilitate hydrophobic-hydrophobic interactions between the particles or beads and the protein's hydrophobic parts, whereas the hydrophilic beads are suited when hydrophilic-hydrophilic interactions between the particles or beads and the protein's hydrophilic parts are desired.
  • the particles or beads may for example be latex microspheres such as those available from Interfacial Dynamics Corporation (Portland, US). As disclosed on the Interfacial Dynamics web-site (www.idclatex.com) latex microspheres/beads are available with either anionic (negative) or cationic (positive) surface charges. Anionic latexes - such as those with sulfate, carboxyl, or carboxylate modified surface groups - are less likely to bind to negatively-charged cell surfaces and are therefore used frequently in biological applications.
  • Particles or beads may suitably be sterilized before use, for example by pasteurization, suitably for about 24 hours at 78-80° C; or by gamma irradiation, for example at 0.03 megarads suitably for about 24 hours.
  • the particles or beads may be coated with various proteins or polysaccharides that will greatly reduce their capacity to absorb biomolecules non-specifically.
  • Specific irreversible adsorption of protein molecules such as avidin, sfreptavidin, and antibodies may be accomplished by simply mixing the latex and protein together for a specified period of time, then separating the bound from the unbound protein through centrifugation and removal of the supernatant.
  • particles or beads may be coated, for example with BSA or dextrans.
  • proteins, nucleic acids, and other biomolecules may if desired be covalently coupled to the particles or beads. Covalent coupling may require more effort than passive adsorption, but can result in conjugates with greater specificity that remain active longer.
  • Carbodiimide-mediated coupling to CML latexes is a suitable method for conjugating low molecular weight peptides and oligonucleotides.
  • a wide range of suitable beads, micro/nanobeads and micro/nanospheres is also available for example from Polysciences, Inc. 400 Valley Road, Warrington, PA, US.
  • modulators of Notch signalling may be conjugated to the linear or cross-linked backbone of a liposome using conventional techniques (see, e.g. Ostro, M. J. (Ed.), Liposomes: from Biophysics to Therapeutics (Marcel Dekker, New York, 1987)).
  • Ostro M. J. (Ed.), Liposomes: from Biophysics to Therapeutics (Marcel Dekker, New York, 1987)
  • One prefened method of preparing liposomes and conjugating immunoglobulins to their surface is described by Ishimoto, Y. et al., J. Immunol. Met. 75, 351-360 (1984). For example, multilamillar liposomes composed of dipalmitoylphosphatidylcholine, cholesterol and phosphotidylethanolamine are prepared.
  • Modulators of Notch signalling may then be coupled to the phosphatidylethanolamine by the cross-linking agent N-hydroxysuccinimidyl 3-(2-pyridyldithio) propionate.
  • the coupling of the fragment to the liposome can be demonstrated by the release of a pre-trapped marker, e.g., carboxyfluorescence, from the liposomes upon the treatment of secondary antibody against the conjugated fragment and complement.
  • the modulator of Notch signalling comprises an IgFc domain it may, for example, be coupled to a liposome or another carrier of the invention via carbohydrate moieties on the Fc domain.
  • Dextran In vitro biocompatibility of biodegradable dextran-based hydrogels tested with human fibroblasts, Biomaterials 22 (2001 ) 1197- 1203 De Groot et al.. Methacrylate- derivatized dextran is biocompatible and a promising system for drug delivery.
  • PEG Copolymer with styrene; Polystyrene-poly (ethylene glycol) (PS-PEG2000) particles as model systems for site specific drug delivery. 2. The effect of PEG surface density on the in vitro cell interaction and in vivo biodistribution. Pharm. Res. 11 (1994) 1016-1022.
  • Step nanospheres Prolonging the circulation time and modifying the body distribution of intravenously injected polystyrene nanospheres by prior intravenous administration of poloxamine-908.
  • polypeptide is synonymous with the term “amino acid sequence” and/or the term “protein”. In some instances, the term “polypeptide” is synonymous with the term “peptide”.
  • Protein usually refers to a short amino acid sequence that is 10 to 40 amino acids long, preferably 10 to 35 amino acids.
  • polypeptide sequence may be prepared and isolated from a suitable source, or it may be made synthetically or it may be prepared by use of recombinant DNA techniques.
  • polynucleotide sequence is synonymous with the tenn “polynucleotide” and/or the term “nucleotide sequence”.
  • the polynucleotide sequence may be DNA or RNA of genomic or synthetic or of recombinant origin. They may also be cloned by standard techniques. The polynucleotide sequence may be double-stranded or single-stranded whether representing the sense or antisense strand or combinations thereof.
  • Polynucleotide refers to a polymeric form of nucleotides of at least 10 bases in length and up to 1,000 bases or even more. Longer polynucleotide sequences will generally be produced using recombinant means, for example using a PCR (polymerase chain reaction) cloning techniques. This will involve making apair of primers (e.g. of about 15 to 30 nucleotides) flanking a region of the targeting sequence which it is desired to clone, bringing the primers into contact with mRNA or cDNA obtained from an animal or human cell, performing a polymerase chain reaction (PCR) under conditions which bring about amplification of the desired region, isolating the amplified fragment (e.g. by purifying the reaction mixture on an agarose gel) and recovering the amplified DNA.
  • the primers may be designed to contain suitable restriction enzyme recognition sites so that the amplified DNA can be cloned into a suitable cloning vector.
  • the nucleic acid may be RNA or DNA and is preferably DNA. Where it is RNA, manipulations may be performed via cDNA intermediates. Generally, a nucleic acid sequence encoding the first region will be prepared and suitable restriction sites provided at the 5' and/or 3' ends. Conveniently the sequence is manipulated in a standard laboratory vector, such as a plasmid vector based on pBR322 or pUC19 (see below). Reference may be made to Molecular Cloning by Sambrook et al. (Cold Spring Harbor, 1989) or similar standard reference books for exact details of the appropriate techniques. Sources of nucleic acid may be ascertained by reference to published literature or databanks such as GenBank.
  • Nucleic acid encoding the desired first or second sequences may be obtained from academic or commercial sources where such sources are willing to provide the material or by synthesising or cloning the appropriate sequence where only the sequence data are available. Generally this may be done by reference to literature sources which describe the cloning of the gene in question.
  • nucleic acids can be characterised as those nucleotide sequences which hybridise to the nucleic acid sequences known in the art.
  • the polynucleotide sequence may comprise, for example, a protein-encoding domain, an antisense sequence or a functional motif such as a protein-binding domain and includes variants, derivatives, analogues and fragments thereof.
  • the term also refers to polypeptides encoded by the nucleotide sequence.
  • the present invention also encompasses the use of variants, derivatives, analogues, homologues, mimetics and fragments thereof.
  • a variant of any given sequence is a sequence in which the specific sequence of residues (whether amino acid or nucleic acid residues) has been modified in such a manner that the polypeptide or polynucleotide in question retains at least one of its endogenous functions.
  • a variant sequence can be modified by addition, deletion, substitution modification replacement and or variation of at least one residue present in the naturally-occurring protein.
  • derivative as used herein, in relation to proteins or polypeptides of the present invention includes any substitution of, variation of, modification of, replacement of, deletion of and/or addition of one (or more) amino acid residues from or to the sequence providing that the resultant protein or polypeptide retains at least one of its endogenous functions.
  • analogue in relation to polypeptides or polynucleotides, includes any polypeptide or polynucleotide which retains at least one of the functions of the endogenous polypeptide or polynucleotide but generally has a different evolutionary origin thereto.
  • mimetic in relation to polypeptides or polynucleotides, refers to a chemical compound that possesses at least one of the endogenous functions of the polypeptide or polynucleotide which it mimics.
  • amino acid substitutions may be made, for example from 1, 2 or 3 to 10 or 20 substitutions provided that the modified sequence retains the required transport activity or ability to modulate Notch signalling.
  • Amino acid substitutions may include the use of non- naturally occurring analogues.
  • Proteins of use in the present invention may also have deletions, insertions or substitutions of amino acid residues which produce a silent change and result in a functionally equivalent protein.
  • Deliberate amino acid substitutions may be made on the basis of similarity in polarity, charge, solubility, hydrophobicity, hydrophilicity, and/or the amphipathic nature of the residues as long as the transport or modulation function is retained.
  • negatively charged amino acids include aspartic acid and glutamic acid; positively charged amino acids include lysine and arginine; and amino acids with uncharged polar head groups having similar hydrophilicity values include leucine, isoleucine, valine, glycine, alanine, asparagine, glutamine, serine, threonine, phenylalanine, and tyrosine.
  • protein includes single-chain polypeptide molecules as well as multiple-polypeptide complexes where individual constituent polypeptides are linked by covalent or non-covalent means.
  • polypeptide and peptide refer to a polymer in which the monomers are amino acids and are joined together through peptide or disulfide bonds.
  • subunit and domain may also refer to polypeptides and peptides having biological function.
  • “Fragments” are also variants and the term typically refers to a selected region of the polypeptide or polynucleotide that is of interest either functionally or, for example, in an assay. “Fragment” thus refers to an amino acid or nucleic acid sequence that is a portion of a full-length polypeptide or polynucleodtide.
  • Such variants may be prepared using standard recombinant DNA techniques such as site- directed mutagenesis. Where insertions are to be made, synthetic DNA encoding the insertion together with 5' and 3' flanking regions conesponding to the naturally-occurring sequence either side of the insertion site. The flanking regions will contain convenient restriction sites conesponding to sites in the naturally-occurring sequence so that the sequence may be cut with the appropriate enzyme(s) and the synthetic DNA ligated into the cut. The DNA is then expressed in accordance with the invention to make the encoded protein. These methods are only illustrative of the numerous standard techniques known in the art for manipulation of DNA sequences and other known techniques may also be used.
  • Polynucleotide variants will preferably comprise codon optimised sequences.
  • Codon optimisation is known in the art as a method of enhancing RNA stability and therefor gene expression.
  • the redundancy of the genetic code means that several different codons may encode the same amino-acid.
  • Leucine, Arginine and Serine are each encoded by six different codons.
  • Different organisms show preferences in their use of the different codons.
  • Viruses such as HIV, for instance, use a large number of rare codons.
  • Codon usage tables are known in the art for mammalian cells, as well as for a variety of other organisms.
  • at least part of the sequence is codon optimised. Even more preferably, the sequence is codon optimised in its entirety.
  • homologous sequence can be taken to include an amino acid sequence which may be at least 75, 85 or 90% identical, preferably at least 95 or 98% identical.
  • homology should typically be considered with respect to those regions of the sequence (such as amino acids at positions 51, 56 and 57) known to be essential for an activity.
  • homology can also be considered in terms of similarity (i.e. amino acid residues having similar chemical properties/functions), in the context of the present invention it is prefened to express homology in terms of sequence identity.
  • Homology comparisons can be conducted by eye, or more usually, with the aid of readily available sequence comparison programs. These commercially available computer programs can calculate % homology between two or more sequences.
  • Percent homology may be calculated over contiguous sequences, i.e. one sequence is aligned with the other sequence and each amino acid in one sequence is directly compared with the conesponding amino acid in the other sequence, one residue at a time. This is called an "ungapped" alignment. Typically, such ungapped alignments are performed only over a relatively short number of residues.
  • Calculation of maximum %> homology therefor firstly requires the production of an optimal alignment, taking into consideration gap penalties.
  • a suitable computer program for carrying out such an alignment is the GCG Wisconsin Bestfit package (Devereux). Examples of other software than can perform sequence comparisons include, but are not limited to, the BLAST package, FASTA (Atschul) and the GENEWORKS suite of comparison tools. Both BLAST and FASTA are available for offline and online searching. However it is preferred to use the GCG Bestfit program.
  • the final % homology can be measured in terms of identity, the alignment process itself is typically not based on an all-or-nothing pair comparison. Instead, a scaled similarity score matrix is generally used that assigns scores to each pairwise comparison based on chemical similarity or evolutionary distance.
  • BLOSUM62 the default matrix for the BLAST suite of programs.
  • GCG Wisconsin programs generally use either the public default values or a custom symbol comparison table if supplied (see user manual for further details). It is prefened to use the public default values for the GCG package, or in the case of other software, the default matrix, such as BLOSUM62.
  • % homology preferably % sequence identity.
  • the software typically does this as part of the sequence comparison and generates a numerical result.
  • Nucleotide sequences which are homologous to or variants of sequences of use in the present invention can be obtained in a number of ways, for example by probing DNA libraries made from a range of sources.
  • other viral/bacterial, or cellular homologues particularly cellular homologues found in mammalian cells (e.g. rat, mouse, bovine and primate cells) may be obtained and such homologues and fragments thereof in general will be capable of selectively hybridising to the sequences shown in the sequence listing herein.
  • Such sequences may be obtained by probing cDNA libraries made from or genomic DNA libraries from other animal species, and probing such libraries with probes comprising all or part of the reference nucleotide sequence under conditions of medium to high stringency. Similar considerations apply to obtaining species homologues and allelic variants of the amino acid and/or nucleotide sequences useful in the present invention.
  • Variants and strain/species homologues may also be obtained using degenerate PCR which will use primers designed to target sequences within the variants and homologues encoding conserved amino acid sequences within the sequences of use in the present invention.
  • conserveed sequences can be predicted, for example, by aligning the amino acid sequences from several variants/homologues. Sequence alignments can be performed using computer software known in the art. For example the GCG Wisconsin PileUp program is widely used. The primers used in degenerate PCR will contain one or more degenerate positions and will be used at stringency conditions lower than those used for cloning sequences with single sequence primers against known sequences.
  • nucleotide sequences may be obtained by site directed mutagenesis of characterised sequences. This may be useful where for example silent codon changes are required to sequences to optimise codon preferences for a particular host cell in which the nucleotide sequences are being expressed. Other sequence changes may be desired in order to introduce restriction enzyme recognition sites, or to alter the activity of the polynucleotide or encoded polypeptide.
  • primers will be produced by synthetic means, involving a step wise manufacture of the desired nucleic acid sequence one nucleotide at a time. Techniques for accomplishing this using automated techniques are readily available in the art.
  • PCR polymerase chain reaction
  • This will involve making a pair of primers (e.g. of about 15 to 30 nucleotides) flanking a region of the targeting sequence which it is desired to clone, bringing the primers into contact with mRNA or cDNA obtained from an animal or human cell, performing a polymerase chain reaction (PCR) under conditions which bring about amplification of the desired region, isolating the amplified fragment (e.g. by purifying the reaction mixture on an agarose gel) and recovering the amplified DNA.
  • the primers may be designed to contain suitable restriction enzyme recognition sites so that the amplified DNA can be cloned into a suitable cloning vector
  • host cells can be genetically engineered to incorporate expression systems or polynucleotides of the invention.
  • Introduction of a polynucleotide into the host cell can be effected by methods described in many standard laboratory manuals, such as Davis et al and Sambrook et al, such as calcium phosphate transfection, DEAE- dextran mediated transfection, transfection, microinjection, cationic lipid-mediated transfection, electroporation, transduction, scrape loading, ballistic introduction and infection. It will be appreciated that such methods can be employed in vitro or in vivo as drug delivery systems.
  • bacterial cells such as streptococci, staphylococci, E. coli, streptomyces and Bacillus subtilis cells
  • fungal cells such as yeast cells and Aspergillus cells
  • insect cells such as Drosophila S2 and Spodoptera Sf9 cells
  • animal cells such as CHO, COS, NSO, HeLa, C127, 3T3, BHK, 293 and Bowes melanoma cells
  • plant cells include bacterial cells, such as streptococci, staphylococci, E. coli, streptomyces and Bacillus subtilis cells
  • fungal cells such as yeast cells and Aspergillus cells
  • insect cells such as Drosophila S2 and Spodoptera Sf9 cells
  • animal cells such as CHO, COS, NSO, HeLa, C127, 3T3, BHK, 293 and Bowes melanoma cells
  • vectors include, among others, chromosomal, episomal and virus- derived vectors, e.g., vectors derived from bacterial plasmids, from bacteriophage, from transposons, from yeast episomes, from insertion elements, from yeast chromosomal elements, from viruses such as baculoviruses, papova viruses, such as SV40, vaccinia viruses, adenoviruses, fowl pox viruses, pseudorabies viruses and refroviruses, and vectors derived from combinations thereof, such as those derived from plasmid and bacteriophage genetic elements, such as cosmids and phagemids.
  • chromosomal, episomal and virus- derived vectors e.g., vectors derived from bacterial plasmids, from bacteriophage, from transposons, from yeast episomes, from insertion elements, from yeast chromosomal elements, from viruses such as baculoviruses, papova
  • the expression system constructs may contain control regions that regulate as well as engender expression.
  • any system or vector suitable to maintain, propagate or express polynucleotides and/or to express a polypeptide in a host may be used for expression in this regard.
  • the appropriate DNA sequence may be inserted into the expression system by any of a variety of well-known and routine techniques, such as, for example, those set forth in Sambrook et al
  • secretion signals may be incorporated into the expressed polypeptide. These signals may be endogenous to the polypeptide or they may be heterologous signals.
  • Active agents for use in the invention can be recovered and purified from recombinant cell cultures by well-known methods including ammonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, hydroxylapatite chromatography and lectin chromatography. Most preferably, high performance liquid chromatography is employed for purification. Well known techniques for refolding protein may be employed to regenerate active conformation when the polypeptide is denatured during isolation and or purification.
  • any one or more of the above candidate modulators is brought into contact with a cell of the immune system.
  • Cells of the immune system of use in the present invention are described below.
  • Cells of use in the present invention are cells of the immune system capable of transducing the Notch signalling pathway.
  • the cells of use in the present invention are T-cells. These include, but are not limited to, CD4 + and CD8 + mature T cells, immature T cells of peripheral or thymic origin and NK-T cells.
  • the cells will be antigen-presenting cells (APCs).
  • APCs include dendritic cells (DCs) such as interdigitating DCs or follicular DCs, Langerhans cells, PBMCs, macrophages, B-lymphocytes, T-lymphocytes, or other cell types such as epithelial cells, fibroblasts or endothelial cells, constitutively expressing or activated to express a MHC Class II molecules on their surfaces.
  • Precursors of APCs include CD34 cells, monocytes, fibroblasts and endothelial cells.
  • the APCs or precursors may be modified by the culture conditions or may be genetically modified, for instance by transfection of one or more genes.
  • the T cells or APCs may be isolated from a patient, or from a donor individual or another individual.
  • the cells are preferably mammalian cells such as human or mouse cells. Preferably the cells are of human origin.
  • the APC or precursor APC may be provided by a cell proliferating in culture such as an established cell line or a primary cell culture. Examples include hybridoma cell lines, L-cells and human fibroblasts such as MRC-5.
  • Preferred cell lines for use in the present invention include Jurkat, H9, CEM and EL4 T- cells; long-term T-cell clones such as human HA1.7 or mouse D10 cells; T-cell hybridomas such as DO11.10 cells; macrophage-like cells such as U937 or THP1 cells; B-cell lines such as EBV-transformed cells such as Raji, A20 and Ml cells.
  • Dendritic cells can be isolated prepared by a number of means, for example they can either be purified directly from peripheral blood, or generated from CD34 precursor cells for example after mobilisation into peripheral blood by treatment with GM-CSF, or directly from bone marrow. From peripheral blood, adherent precursors can be treated with a GM-CSF/IL-4 mixture (Inaba et al), or from bone marrow, non-adherent CD34 + cells can be treated with GM-CSF and TNF- ⁇ (Caux et al).
  • GM-CSF/IL-4 mixture Inaba et al
  • non-adherent CD34 + cells can be treated with GM-CSF and TNF- ⁇ (Caux et al).
  • DCs can also be routinely prepared from the peripheral blood of human volunteers, similarly to the method of Sallusto and Lanzavecchia J Exp Med (1994) 179(4) 1109-18 using purified peripheral blood mononucleocytes (PBMCs) and treating 2 hour adherent cells with GM-CSF and IL-4. If required, these may be depleted of CD 19 + B cells and CD3 + , CD2 + T cells using magnetic beads (Coffin et al) . Culture conditions may include other cytokines such as GM-CSF or IL-4 for the maintenance and, or activity of the dendritic cells or other antigen presenting cells.
  • PBMCs peripheral blood mononucleocytes
  • T cells and B cells for use in the invention are preferably obtained from cell lines such as lymphoma or leukemia cell lines, T cell hybridomas or B cell hybridomas but may also be isolated from an individual suffering from a disease of the immune system or a recipient for a transplant operation or from a related or unrelated donor individual.
  • T cells and B cells may be obtained from blood or another source (such as lymph nodes, spleen, or bone manow) and may be enriched or purified by standard procedures. Alternatively whole blood may be used or leukocyte enriched blood or purified white blood cells as a source of T cells and other cell types. It is particularly prefened to use helper T cells (CD4 ). Alternatively other T cells such as CD8 + cells may be used.
  • Candidate modulators of use in the present invention are brought into contact with a cell of the immune system as described above.
  • modulation of Notch signalling by a candidate modulator is detected.
  • Assays for detecting modulation of Notch signalling will be described below. Many of these assays will involve monitoring the expression of a "target gene”.
  • Endogenous target genes of the Notch signalling pathway include Deltex, genes of the Hes family (Hes-1 in particular), Enhancer of Split [E(spl)] complex genes, 11-10, CD-23, Dlx-1, CTLA4, CD-4, DIM, Numb, Mastermind and Dsh. Although all genes the expression of which is modulated by Notch activation may be used for the purpose of the present invention, prefened endogenous target genes are described below.
  • Deltex an intracellular docking protein, replaces Su(H) as it leaves its site of interaction with the intracellular tail of Notch, as shown in Figure 1.
  • Deltex is a cytoplasmic protem containing a zinc-finger (Artavanis-Tsakonas; Osborne). It interacts with the ankyrin repeats of the Notch intracellular domain. Studies indicate that Deltex promotes Notch pathway activation by interacting with Grb2 and modulating the Ras-JNK signalling pathway (Matsuno).
  • Deltex also acts as a docking protein which prevents Su(H) from binding to the intracellular tail of Notch (Matsuno). Thus, Su(H) is released into the nucleus where it acts as a transcriptional modulator.
  • Notch the intracellular tail of Notch
  • DTX1 Homo sapiens Deltex
  • Hes-1 (Hairy-enhancer of Split-1) (Takebayasbi) is a transcriptional factor with a basic helix- loop-helix structure. It binds to an important functional site in the CD4 silencer leading to repression of CD4 gene expression. Thus, Hes-1 is strongly involved in the determination of T- cell fate.
  • Other genes from the Hes family include Hes-5 (mammalian Enhancer of Split homologue), the expression of which is also upregulated by Notch activation, and Hes-3. Expression of Hes-1 is upregulated as a result of Notch activation.
  • the sequence of human Hes-1 can be found in GenBank Accession Nos. AK000415 and AF264785.
  • E(s ⁇ l) gene complex [E(spl)-C] (Leimeister) comprises seven genes of which only E(spl) and Groucho show visible phenotypes when mutant.
  • E(spl) was named after its ability to enhance Split mutations, Split being another name for Notch.
  • E(spl)-C genes repress Delta through regulation of achaete-scute complex gene expression. Expression of E(spl) is upregulated as a result of Notch activation.
  • IL- 10 (interleukin- 10) is a factor produced by Th2 helper T-cells . It is a co-regulator of mast cell growth and shows extensive homology with the Epstein-Barr bcrfi gene. Although it is not known to be a direct downstream target of the Notch signalling pathway, its expression has been found to be strongly upregulated coincident with Notch activation.
  • the mRNA sequence of IL-10 may be found in GenBank ref. No. GI1041812.
  • CD-23 is the human leukocyte differentiation antigen CD23 (FCE2) which is a key molecule for B-cell activation and growth. It is the low-affinity receptor for IgE. Furthermore, the truncated molecule can be secreted, then functioning as a potent mitogenic growth factor. Although it is not thought to be a direct downstream target of the Notch signalling pathway, its expression has been found to be strongly upregulated coincident with Notch activation.
  • FCE2 human leukocyte differentiation antigen CD23
  • Dlx-1 (distalless-1) expression is downregulated as a result of Notch activation.
  • Sequences for Dlx genes may be found in GenBank Accession Nos. U51000-3.
  • CTLA4 cytotoxic T-lymphocyte activated protein 4
  • the promoter region of the gene encoding CTLA4 has CBFl response elements and its expression is upregulated as a result of Notch activation.
  • the sequence of CTLA4 can be found in GenBank Accession No. L15006.
  • CD-4 expression is downregulated as a result of Notch activation.
  • a sequence for the CD-4 antigen may be found in GenBank Accession No. XM006966.
  • the assay of the present invention is set up to detect either inhibition or enhancement of Notch signalling in cells of tl e immune system by candidate modulators.
  • the method comprises mixing cells of the immune system, where necessary transformed or transfected, etc. with a synthetic reporter gene, in an appropriate buffer, with a sufficient amount of candidate modulator and monitoring Notch signalling.
  • the modulators may be small molecules, proteins, antibodies or other ligands as described above. Amounts or activity of the target gene (also described above) will be measured for each compound tested using standard assay techniques and appropriate controls.
  • the detected signal is compared with a reference signal and any modulation with respect to the reference signal measured.
  • the assay may also be run in the presence of a known antagonist of the Notch signalling pathway in order to identify compounds capable of rescuing the Notch signal.
  • a known antagonist of the Notch signalling pathway Any one or more of appropriate targets - such as an amino acid sequence and/or nucleotide sequence - may be used for identifying a compound capable of modulating the Notch signalling pathway in cells of the immune system in any of a variety of drug screening techniques.
  • the target employed in such a test may be free in solution, affixed to a solid support, borne on a cell surface, or located infracellularly.
  • the assay of the present invention is a cell based assay.
  • the assay of the present invention may be a screen, whereby a number of agents are tested.
  • the assay method of the present invention is a high through put screen.
  • This invention also contemplates the use of competitive drug screening assays in which neutralising antibodies capable of binding a target specifically compete with a test compound for binding to a target.
  • nucleic acid assays are also known. Any conventional technique which is known or which is subsequently disclosed may be employed. Examples of suitable nucleic acid assay are mentioned below and include amplification, PCR, RT-PCR, RNase protection, blotting, spectrometry, reporter gene assays, gene chip arrays and other hybridization methods.
  • Target gene presence, amplification and/or expression may be measured in a sample directly, for example, by conventional Southern blotting, Northern blotting to quantitate the transcription of target mRNA, dot blotting (DNA or RNA analysis), or in situ hybridisation, using an appropriately labelled probe.
  • Southern blotting Northern blotting to quantitate the transcription of target mRNA
  • dot blotting DNA or RNA analysis
  • in situ hybridisation using an appropriately labelled probe.
  • nucleic acids for analysis from samples generally requires nucleic acid amplification.
  • Many amplification methods rely on an enzymatic chain reaction (such as a polymerase chain reaction, a ligase chain reaction, or a self-sustained sequence replication) or from the replication of all or part of the vector into which it has been cloned.
  • the amplification according to the invention is an exponential amplification, as exhibited by for example the polymerase chain reaction.
  • amplification methods have been described in the literature, for example, general reviews of these methods in Landegren, U., et al., Science 242:229-237 (1988) and Lewis, R., Genetic Engineering News 10:1, 54-55 (1990).
  • amplification methods may be used in the methods of our invention, and include polymerase chain reaction (PCR), PCR in situ, ligase amplification reaction (LAR), ligase hybridisation, Qbeta bacteriophage replicase, transcription-based amplification system (TAS), genomic amplification with transcript sequencing (GAWTS), nucleic acid sequence-based amplification (NASBA) and in situ hybridisation.
  • Primers suitable for use in various amplification techniques can be prepared according to methods known in the art.
  • PCR is a nucleic acid amplification method described ter alia in U.S. Pat. Nos. 4,683,195 and 4,683,202.
  • PCR consists of repeated cycles of DNA polymerase generated primer extension reactions.
  • PCR was originally developed as a means of amplifying DNA from an impure sample. The teclinique is based on a temperature cycle which repeatedly heats and cools the reaction solution allowing primers to anneal to target sequences and extension of those primers for the formation of duplicate daughter strands.
  • RT-PCR uses an RNA template for generation of a first strand cDNA with a reverse franscriptase. The cDNA is then amplified according to standard PCR protocol.
  • PCR can be used to amplify any known nucleic acid in a diagnostic context (Mok et al., (1994), Gynaecologic Oncology, 52: 247-252).
  • Self-sustained sequence replication is a variation of TAS, which involves the isothermal amplification of a nucleic acid template via sequential rounds of reverse franscriptase (RT), polymerase and nuclease activities that are mediated by an enzyme cocktail and appropriate oligonucleotide primers (Guatelli et al. (1990) Proc. Natl. Acad. Sci.
  • RNA of the RNA DNA heteroduplex is used instead of heat denaturation.
  • RNase H and all other enzymes are added to the reaction and all steps occur at the same temperature and without further reagent additions. Following this process, amplifications of 10 6 to 109 have been achieved in one hour at 42 °C.
  • Ligation amplification reaction or ligation amplification system uses DNA ligase and four oligonucleotides, two per target strand. This technique is described by Wu, D. Y. and Wallace, R. B. (1989) Genomics 4:560. The oligonucleotides hybridise to adjacent sequences on the target DNA and are joined by the ligase. The reaction is heat denatured and the cycle repeated.
  • rolling circle amplification (Lizardi et al, (1998) Nat Genet 19:225) is an amplification technology available commercially (RCATTM) which is driven by DNA polymerase and can replicate circular oligonucleotide probes with either linear or geometric kinetics under isothermal conditions.
  • RCATTM rolling circle amplification
  • a geometric amplification occurs via DNA strand displacement and hyperbranchmg to generate 10 or more copies of each circle in 1 hour.
  • RCAT generates in a few minutes a linear chain of thousands of tandemly linked DNA copies of a target covalently linked to that target.
  • SDA strand displacement amplification
  • SDA comprises both a target generation phase and an exponential amplification phase.
  • amplification primers for copying the base sequence and bumper primers for displacing the newly created strands to form altered targets capable of exponential amplification.
  • the exponential amplification process begins with altered targets (single-stranded partial DNA strands with restricted enzyme recognition sites) from the target generation phase.
  • An amplification primer is bound to each strand at its complementary DNA sequence.
  • DNA polymerase then uses the primer to identify a location to extend the primer from its 3' end, using the altered target as a template for adding individual nucleotides.
  • the extended primer thus forms a double-stranded DNA segment containing a complete restriction enzyme recognition site at each end.
  • a restriction enzyme is then bound to the double stranded DNA segment at its recognition site.
  • the restriction enzyme dissociates from the recognition site after having cleaved only one strand of the double-sided segment, forming a nick.
  • DNA polymerase recognises the nick and extends the strand from the site, displacing the previously created strand.
  • the recognition site is thus repeatedly nicked and restored by the restriction enzyme and DNA polymerase with continuous displacement of DNA strands containing the target segment.
  • Each displaced strand is then available to anneal with amplification primers as above. The process continues with repeated nicking, extension and displacement of new DNA strands, resulting in exponential amplification of the original DNA target.
  • the present invention provides for the detection of gene expression at the RNA level.
  • Typical assay formats utilising ribonucleic acid hybridisation include nuclear run-on assays, RT-PCR and RNase protection assays (Melton et al., Nuc. Acids Res. 12:7035. Methods for detection which can be employed include radioactive labels, enzyme labels, chemiluminescent labels, fluorescent labels and other suitable labels.
  • Real-time PCR uses probes labeled with a fluorescent tag or fluorescent dyes and differs from end-point PCR for quantitative assays in that it is used to detect PCR products as they accumulate rather than for the measurement of product accumulation after a fixed number of cycles.
  • the reactions are characterized by the point in time during cycling when amplification of a target sequence is first detected through a significant increase in fluorescence.
  • the ribonuclease protection (RNase protection) assay is an exfelely sensitive technique for the quantitation of specific RNAs in solution.
  • the ribonuclease protection assay can be performed on total cellular RNA or poly(A)-selected mRNA as a target.
  • the sensitivity of the ribonuclease protection assay derives from the use of a complementary in vitro transcript probe which is radiolabeled to high specific activity.
  • the probe and target RNA are hybridized in solution, after which the mixture is diluted and treated with ribonuclease (RNase) to degrade all remaining single-stranded RNA.
  • RNase ribonuclease
  • the hybridized portion of the probe will be protected from digestion and can be visualized via electrophoresis of the mixture on a denaturing polyacrylamide gel followed by autoradiography. Since the protected fragments are analyzed by high resolution polyacrylamide gel electrophoresis, the ribonuclease protection assay can be employed to accurately map mRNA features. If the probe is hybridized at a molar excess with respect to the target RNA, then the resulting signal will be directly proportional to the amount of complementary RNA in the sample.
  • PCR technology as described e.g. in section 14 of Sambrook et al., 1989, requires the use of oligonucleotide probes that will hybridise to target nucleic acid sequences. Strategies for selection of oligonucleotides are described below.
  • a probe is e.g. a single-stranded DNA or RNA that has a sequence of nucleotides that includes between 10 and 50, preferably between 15 and 30 and most preferably at least about 20 contiguous bases that are the same as (or the complement of) an equivalent or greater number of contiguous bases.
  • the nucleic acid sequences selected as probes should be of sufficient length and sufficiently unambiguous so that false positive results are minimised.
  • the nucleotide sequences are usually based on conserved or highly homologous nucleotide sequences or regions of polypeptides.
  • the nucleic acids used as probes may be degenerate at one or more positions.
  • nucleic acid probes of the invention are labelled with suitable label means for ready detection upon hybridisation.
  • suitable label means is a radiolabel.
  • the prefened method of labelling a DNA fragment is by incorporating 32 P dATP with the Klenow fragment of DNA polymerase in a random priming reaction, as is well known in the art.
  • Oligonucleotides are usually end-labelled with 32 P-labelled ATP and polynucleotide kinase.
  • other methods e.g. non-radioactive
  • Prefened are such sequences, probes which hybridise under high-stringency conditions.
  • Stringency of hybridisation refers to conditions under which polynucleic acids hybrids are stable. Such conditions are evident to those of ordinary skill in the field. As known to those of skill in the art, the stability of hybrids is reflected in the melting temperature (Tm) of the hybrid which decreases approximately 1 to 1.5°C with every 1% decrease in sequence homology. In general, the stability of a hybrid is a function of sodium ion concentration and temperature. Typically, the hybridisation reaction is performed under conditions of higher stringency, followed by washes of varying stringency.
  • high stringency refers to conditions that permit hybridisation of only those nucleic acid sequences that form stable hybrids in 1 M Na+ at 65-68 °C.
  • High stringency conditions can be provided, for example, by hybridisation in an aqueous solution containing 6x SSC, 5x Denhardt's, 1 % SDS (sodium dodecyl sulphate), 0.1 Na+ pyrophosphate and 0.1 mg/ml denatured salmon sperm DNA as non specific competitor.
  • high stringency washing may be done in several steps, with a final wash (about 30 min) at the hybridisation temperature in 0.2 - 0. lx SSC, 0.1 % SDS.
  • Gene expression may also be detected using a reporter system.
  • a reporter system may comprise a readily identifiable marker under the control of an expression system, e.g. of the gene being monitored.
  • Fluorescent markers which can be detected and sorted by FACS, are prefened. Especially prefened are GFP and luciferase.
  • Another type of prefened reporter is cell surface markers, i.e. proteins expressed on the cell surface and therefor easily identifiable.
  • cell-based screening assays can be designed by constructing cell lines in which the expression of a reporter protein, i.e.
  • an easily assayable protein such as ⁇ - galactosidase, chloramphenicol acetyltransferase (CAT) or luciferase
  • CAT chloramphenicol acetyltransferase
  • luciferase is dependent on the activation of a Notch.
  • a reporter gene encoding one of the above polypeptides may be placed under the control of an response element which is specifically activated by Notch signalling.
  • Alternative assay formats include assays which directly assess responses in a biological system. If a cell-based assay system is employed, the test compound(s) indentified may then be subjected to in vivo testing to determine their effect on Notch signalling pathway.
  • reporter constructs useful for detecting Notch signalling by expression of a reporter gene may be constructed according to the general teaching of Sambrook et al
  • constructs according to the invention comprise a promoter of the gene of interest (i.e. of an endogenous target gene), and a coding sequence encoding the desired reporter constructs, for example of GFP or luciferase.
  • a promoter of the gene of interest i.e. of an endogenous target gene
  • a coding sequence encoding the desired reporter constructs for example of GFP or luciferase.
  • Vectors encoding GFP and luciferase are known in the art and available commercially.
  • Sorting of cells may be performed by any technique known in the art, as exemplified above.
  • cells may be sorted by flow cytometry or FACS.
  • flow cytometry FACS
  • FACS Fluorescence Activated Cell Sorting
  • F.A.C.S. Fluorescence Activated Cell Sorting
  • flow cytometry Fluorescence Activated Cell Sorting
  • FACS can be used to measure target gene expression in cells transfected with recombinant DNA encoding polypeptides. This can be achieved directly, by labelling of the protein product, or indirectly by using a reporter gene in the construct.
  • reporter genes are ⁇ -galactosidase and Green Fluorescent Protein (GFP).
  • GFP Green Fluorescent Protein
  • ⁇ -galactosidase activity can be detected by FACS using fluorogenic substrates such as fluorescein digalactoside (FDG).
  • FDG is introduced into cells by hypotonic shock, and is cleaved by the enzyme to generate a fluorescent product, which is trapped within the cell.
  • One enzyme can therefor generate a large amount of fluorescent product.
  • Cells expressing GFP constructs will fluoresce without the addition of a subsfrate. Mutants of GFP are available which have different excitation frequencies, but which emit fluorescence in the same channel, h a two-laser FACS machine, it is possible to distinguish cells which are excited by the different lasers and therefor assay two transfections at the same time.
  • the invention comprises the use of nucleic acid probes complementary to mRNA.
  • Such probes can be used to identify cells expressing polypeptides individually, such that they may subsequently be sorted either manually, or using FACS sorting.
  • Nucleic acid probes complementary to mRNA may be prepared according to the teaching set forth above, using the general procedures as described by Sambrook et al (1989).
  • the invention comprises the use of an antisense nucleic acid molecule, complementary to a target mRNA, conjugated to a fluorophore which may be used in FACS cell sorting.
  • the present invention also provides a method of detection of polypeptides.
  • the advantage of using a protein assay is that Notch activation can be directly measured.
  • Assay techniques that can be used to determine levels of a polypeptide are well known to those skilled in the art. Such assay methods include radioimmunoassays, competitive-binding assays, protein gel assay, Western Blot analysis, antibody sandwich assays, antibody detection, FACS and ELISA assays.
  • polypeptides can be detected by differential mobility on protein gels, or by other size analysis techniques, such as mass specfrometry.
  • the detection means may be sequence-specific.
  • polypeptide or RNA molecules can be developed which specifically recognise polypeptides in vivo or in vitro.
  • RNA aptamers can be produced by SELEX.
  • SELEX is a method for the in vitro evolution of nucleic acid molecules with highly specific binding to target molecules. It is described, for example, in U.S. patents 5654151, 5503978, 5567588 and 5270163, as well as PCT publication WO 96/38579
  • the invention in certain embodiments, includes antibodies specifically recognising and binding to polypeptides.
  • Antibodies may be recovered from the serum of immunised animals. Monoclonal antibodies may be prepared from cells from immunised animals in the conventional manner.
  • the antibodies of the invention are useful for identifying cells expressing the genes being monitored.
  • Antibodies according to the invention may be whole antibodies of natural classes, such as IgE and IgM antibodies, but are preferably IgG antibodies. Moreover, the invention includes antibody fragments, such as Fab, F(ab')2, Fv and ScFv. Small fragments, such Fv and ScFv, possess advantageous properties for diagnostic and therapeutic applications on account of their small size and consequent superior tissue distribution.
  • the antibodies may comprise a label.
  • labels which allow the imaging of the antibody in neural cells in vivo.
  • Such labels may be radioactive labels or radioopaque labels, such as metal particles, which are readily visuahsable within tissues.
  • radioactive labels such as metal particles, which are readily visuahsable within tissues.
  • radioopaque labels such as metal particles, which are readily visuahsable within tissues.
  • fluorescent labels or other labels which are visuahsable in tissues and which may be used for cell sorting.
  • antibodies as used herein can be altered antibodies comprising an effector protein such as a label.
  • labels which allow the imaging of the distribution of the antibody in vivo.
  • Such labels can be radioactive labels or radioopaque labels, such as metal particles, which are readily visuahsable within the body of a patient.
  • radioactive labels or radioopaque labels, such as metal particles, which are readily visuahsable within the body of a patient.
  • they can be fluorescent labels or other labels which are visuahsable on tissue
  • Antibodies as described herein can be produced in cell culture. Recombinant DNA technology can be used to produce the antibodies according to established procedure, in bacterial or preferably mammalian cell culture. The selected cell culture system optionally secretes the antibody product, although antibody products can be isolated from non-secreting cells.
  • Multiplication of hybridoma cells or mammalian host cells in vitro is carried out in suitable culture media, which are the customary standard culture media, for example Dulbecco's Modified Eagle Medium (DMEM) or RPMI 1640 medium, optionally replenished by a mammalian serum, e.g. foetal calf serum, or trace elements and growth sustaining supplements, e.g. feeder cells such as normal mouse peritoneal exudate cells, spleen cells, bone marrow macrophages, 2-aminoethanol, insulin, transferrin, low density lipoprotein, oleic acid, or the like.
  • suitable culture media which are the customary standard culture media, for example Dulbecco's Modified Eagle Medium (DMEM) or RPMI 1640 medium
  • a mammalian serum e.g. foetal calf serum
  • trace elements and growth sustaining supplements e.g. feeder cells
  • feeder cells such as normal mouse peritoneal exudate cells, sple
  • Multiplication of host cells which are bacterial cells or yeast cells is likewise carried out in suitable culture media known in the art, for example for bacteria in medium LB, NZCYM, NZYM, NZM, Terrific Broth, SOB, SOC, 2 x YT, or M9 Minimal Medium, and for yeast in medium YPD, YEPD, Minimal Medium, or Complete Minimal Dropout Medium.
  • In vitro production provides relatively pure antibody preparations and allows scale-up to give large amounts of the desired antibodies.
  • Techniques for bacterial cell, yeast or mammalian cell cultivation are known in the art and include homogeneous suspension culture, e.g. in an airlift reactor or in a continuous stiner reactor, or immobilised or entrapped cell culture, e.g. in hollow fibres, microcapsules, on agarose microbeads or ceramic cartridges.
  • the desired antibodies can also be obtained by multiplying mammalian cells in vivo.
  • hybridoma cells producing the desired antibodies are injected into histocompatible mammals to cause growth of antibody-producing tumours.
  • the animals are primed with a hydrocarbon, especially mineral oils such as pristane (tetramethyl-pentadecane), prior to the injection.
  • pristane tetramethyl-pentadecane
  • hybridoma cells obtained by fusion of suitable myeloma cells with antibody-producing spleen cells from Balb/c mice, or transfected cells derived from hybridoma cell line Sp2/0 that produce the desired antibodies are injected intraperitoneally into Balb/c mice optionally pre-treated with pristane, and, after one to two weeks, ascitic fluid is taken from the animals.
  • the cell culture supernatants are screened for the desired antibodies, preferentially by an enzyme immunoassay, e.g. a sandwich assay or a dot-assay, or a radioimmunoassay.
  • an enzyme immunoassay e.g. a sandwich assay or a dot-assay, or a radioimmunoassay.
  • the immunoglobulins in the culture supernatants or in the ascitic fluid can be concentrated, e.g. by precipitation with ammonium sulphate, dialysis against hygroscopic material such as polyethylene glycol, filfration through selective membranes, or the like.
  • the antibodies are purified by the customary chromatography methods, for example gel filtration, ion-exchange chromatography, chromatography over DEAE-cellulose and/or (immuno-) affinity chromatography, e.g. affinity chromatography with the target antigen, or with Protein- A.
  • customary chromatography methods for example gel filtration, ion-exchange chromatography, chromatography over DEAE-cellulose and/or (immuno-) affinity chromatography, e.g. affinity chromatography with the target antigen, or with Protein- A.
  • the antibody is preferably provided together with means for detecting the antibody, which can be enzymatic, fluorescent, radioisotopic or other means.
  • the antibody and the detection means can be provided for simultaneous, simultaneous separate or sequential use, in a kit.
  • the antibodies of the invention are assayed for immunospecific binding by any method known in the art.
  • the immunoassays which can be used include but are not limited to competitive and non-competitive assay systems using techniques such as western blots, radioimmunoassays, ELISA, sandwich immunoassays, immunoprecipitation assays, precipitin reactions, gel diffusion precipitin reactions, immunodiffusion assays, agglutination assays, complement-fixation assays, immunoradiometric assays, fluorescent immunoassays and protein A immunoassays.
  • Such assays are routine in the art (see, for example, Ausubel et al, eds, 1994, Cunent Protocols in Molecular Biology, Vol. 1, John Wiley & Sons, Inc., New York, which is inco ⁇ orated by reference herein in its entirety). Exemplary immunoassays are described briefly below.
  • Immunoprecipitation protocols generally comprise lysing a population of cells in a lysis buffer such as RIPA buffer (1% NP-40 or Triton X-100, 1% sodium deoxycholate, 0.1% SDS, 0.15 M NaCl, 0.01 M sodium phosphate at pH 7.2,1% Trasylol) supplemented with protein phosphatase and/or protease inhibitors (e. g., EDTA, PMSF, aprotinin, sodium vanadate), adding the antibody of interest to the cell lysate, incubating for a period of time (e.
  • a lysis buffer such as RIPA buffer (1% NP-40 or Triton X-100, 1% sodium deoxycholate, 0.1% SDS, 0.15 M NaCl, 0.01 M sodium phosphate at pH 7.2,1% Trasylol
  • protein phosphatase and/or protease inhibitors e. g., EDTA, PMSF, aprotin
  • Western blot analysis generally comprises preparing protein samples, electrophoresis of the protein samples in a polyacrylamide gel (e. g., 8%-20% SDS-PAGE depending on the molecular weight of the antigen), transferring the protein sample from the polyacrylamide gel to a membrane such as nitrocellulose, PVDF or nylon, blocking the membrane in blocking solution (e. g., PBS with 3% BSA or non-fat milk), washing the membrane in washing buffer (e. g., PBS-Tween 20), exposing the membrane to a primary antibody (the antibody of interest) diluted in blocking buffer, washing the membrane in washing buffer, exposing the membrane to a secondary antibody (which recognises the primary antibody, e.
  • a polyacrylamide gel e. g., 8%-20% SDS-PAGE depending on the molecular weight of the antigen
  • a membrane such as nitrocellulose, PVDF or nylon
  • blocking solution e. g., PBS with 3% BSA or non-fat milk
  • washing buffer
  • an antihuman antibody conjugated to an enzymatic substrate (e. g., horseradish peroxidase or alkaline phosphatase) or radioactive molecule (e. g., 32 P or 125 I) diluted in blocking buffer, washing the membrane in wash buffer, and detecting the presence of the antigen.
  • an enzymatic substrate e. g., horseradish peroxidase or alkaline phosphatase
  • radioactive molecule e. g., 32 P or 125 I
  • ELISAs generally comprise preparing antigen, coating the well of a 96 well microtitre plate with the antigen, adding the antibody of interest conjugated to a detectable compound such as an enzymatic subsfrate (e. g., horseradish peroxidase or alkaline phosphatase) to the well and incubating for a period of time, and detecting the presence of the antigen.
  • a detectable compound such as an enzymatic subsfrate (e. g., horseradish peroxidase or alkaline phosphatase)
  • a detectable compound such as an enzymatic subsfrate (e. g., horseradish peroxidase or alkaline phosphatase)
  • Immobilisation approaches include covalent immobilisation, such as using amine coupling, surface thiol coupling, ligand thiol coupling and aldehyde coupling, and high affinity capture which relies on high affinity binding of a ligand to an immobilised capturing molecule.
  • capturing molecules include: sfreptavidin, anti-mouse Ig antibodies, ligand-specific antibodies, protian A, protein G and Tag-specific capture.
  • immobilisation is achieved through binding to a support, particularly a particulate support which is preferably in the form of a bead.
  • the assay will generally involve removal of a sample from a patient prior to the step of detecting a signal resulting from cleavage of the intracellular domain.
  • the invention additionally provides a method of screening for a candidate modulator of Notch signalling, the method comprising mixing in a buffer an appropriate amount of Notch, wherein Notch is suitably labelled with detection means for monitoring cleavage of Notch; and a sample of a candidate ligand; and monitoring any cleavage of Notch.
  • sample refers to a collection of inorganic, organic or biochemical molecules which is either found in nature (e.g., in a biological- or other specimen) or in an artificially-constructed grouping, such as agents which may be found and/or mixed in a laboratory.
  • the biological sample may refer to a whole organism, but more usually to a subset of its tissues, cells or component parts (e.g. body fluids, including but not limited to blood, mucus, saliva and urine).
  • the present invention provides a method of detecting novel modulators of Notch signalling.
  • the modulators identified may be used as therapeutic agents - i.e. in therapy applications.
  • the term "therapy” includes curative effects, alleviation effects, and prophylactic effects.
  • the therapy may be on humans or animals.
  • Modulators identified by the assay method of the present invention may be used to treat disorders and/or conditions of the immune system.
  • the compounds can be used in the treatment of T cell mediated diseases or disorders.
  • a detailed description of the conditions affected by the Notch signalling pathway may be found in our WO98/20142, WO00/36089 and WO/00135990.
  • T cells Diseased or infectious states that may be described as being mediated by T cells include, but are not limited to, any one or more of asthma, allergy, tumour induced abenations to the T cell system and infectious diseases such as those caused by Plasmodium species, Microfilariae, Helminths, Mycobacteria, HTV, Cytomegaloviras, Pseudomonas, Toxoplasma, Echinococcus, Haemophilus influenza type B, measles, Hepatitis C or Toxicara.
  • infectious diseases such as those caused by Plasmodium species, Microfilariae, Helminths, Mycobacteria, HTV, Cytomegaloviras, Pseudomonas, Toxoplasma, Echinococcus, Haemophilus influenza type B, measles, Hepatitis C or Toxicara.
  • infectious diseases such as those caused by Plasmodium species, Microfilariae, Helminths, Mycobacteria,
  • autoimmune disorders range from organ specific diseases (such as thyroiditis, insulitis, multiple sclerosis, iridocyclitis, uveitis, orchitis, hepatitis, Addison's disease, myasthenia gravis) to systemic illnesses such as rheumatoid arthritis or lupus erythematosus.
  • organ specific diseases such as thyroiditis, insulitis, multiple sclerosis, iridocyclitis, uveitis, orchitis, hepatitis, Addison's disease, myasthenia gravis
  • systemic illnesses such as rheumatoid arthritis or lupus erythematosus.
  • Other disorders include immune hypeneactivity, such as allergic reactions.
  • organ-specific autoimmune diseases include multiple sclerosis, insulin dependent diabetes mellitus, several forms of anemia (aplastic, hemolytic), autoimmune hepatitis, thyroiditis, insulitis, iridocyclitis, skleritis, uveitis, orchitis, myasthenia gravis, idiopathic thrombocytopenic purpura, inflammatory bowel diseases (Crohn's disease, ulcerative colitis).
  • Systemic autoimmune diseases include: rheumatoid arthritis, juvenile arthritis, scleroderma and systemic sclerosis, sjogren's syndrom, undifferentiated connective tissue syndrome, antiphospholipid syndrome, different forms of vasculitis (polyarteritis nodosa, allergic granulomatosis and angiitis, Wegner's granulomatosis, Kawasaki disease, hypersensitivity vasculitis, Henoch-Schoenlein purpura, Behcet's Syndrome, Takayasu arteritis, Giant cell arteritis, Thrombangiitis obliterans), lupus erythematosus, polymyalgia rheumatica, correspondingl (mixed) cryoglobulinemia, Psoriasis vulgaris and psoriatic arthritis, diffus fasciitis with or without eosinophilia, polymyositis and other idiopathic inflammatory myopathie
  • a more extensive list of disorders includes: unwanted immune reactions and inflammation including arthritis, including rheumatoid arthritis, inflammation associated with hypersensitivity, allergic reactions, asthma, systemic lupus erythematosus, collagen diseases and other autoimmune diseases, inflammation associated with atherosclerosis, arteriosclerosis, atherosclerotic heart disease, reperfusion injury, cardiac anest, myocardial infarction, vascular inflammatory disorders, respiratory distress syndrome or other cardiopulmonary diseases, inflammation associated with peptic ulcer, ulcerative colitis and other diseases of the gastrointestinal tract, hepatic fibrosis, liver cirrhosis or other hepatic diseases, thyroiditis or other glandular diseases, glomerulonephritis or other renal and urologic diseases, otitis or other oto-rhino-laryngological diseases, dermatitis or other dermal diseases, periodontal diseases or other dental diseases, orchitis or epididimo-orchitis, infertility, orchidal trauma or other immune-related testicular
  • retinitis or cystoid macular oedema retinitis or cystoid macular oedema, sympathetic ophthalmia, scleritis, retinitis pigmentosa, immune and inflammatory components of degenerative fondus disease, inflammatory components of ocular trauma, ocular inflammation caused by infection, proliferative vifreo-retinopathies, acute ischaemic optic neuropathy, excessive scarring, e.g.
  • autoimmune diseases or conditions or disorders where, both in the central nervous system (CNS) or in any other organ, immune and/or inflammation suppression would be beneficial, Parkinson's disease, complication and or side effects from treatment of Parkinson's disease, AIDS-related dementia complex HIV-related encephalopathy, Devic's disease, Sydenham chorea, Alzheimer's disease and other degenerative diseases, conditions or disorders of the CNS, inflammatory components of stokes, post-polio syndrome, immune and inflammatory components of psychiatric disorders, myelitis, encephalitis, subacute sclerosing pan-encephalitis, encephalomyelitis, acute neuropathy, subacute neuropathy, chronic neuropathy, Guillaim-Barre syndrome, Sydenham chora, myasthenia gravis, pseudo- tumour cerebri, Down's Syndrome, Huntington's disease, amyotroph
  • monocyte or leukocyte proliferative diseases e.g. leukaemia
  • monocytes or lymphocytes for the prevention and/or treatment of graft rejection in cases of transplantation of natural or artificial cells, tissue and organs such as cornea, bone manow, organs, lenses, pacemakers, natural or artificial skin tissue.
  • the present invention is also useful in methods for altering the fate of a cell, tissue or organ type by altering Notch pathway function in the cell.
  • the present application has application in the treatement of malignant and pre-neoplastic disorders.
  • the present invention is especially useful in relation to adenocarcinomas such as: small cell lung cancer, and cancer of the kidney, uterus, prostrate, bladder, ovary, colon and breast.
  • malignancies which may be treatable according to the present invention include acute and chronic leukemias, lymphomas, myelomas, sarcomas such as fibrosarcoma, myxosarcoma, liposarcoma, lymphangioendotheliosarcoma, angiosarcoma, endotheliosarcoma, chondrosarcoma, osteogenic sarcoma, chordoma, lymphangiosarcoma, synovioma, mesothelioma, leimyosarcoma, rhabdomyosarcoma, colon carcinoma, ovarian cancer, prostate cancer, pancreatic cancer, breasy cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sewat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinomas, cystadenocarcinoma, medullary carcinoma, bronchogenic carcinoma, choriocarcino
  • Nervous system disorders which may be treated according to the present invention include neurological lesions including traumatic lesions resulting from physical injuries; ischaemic lesions; malignant lesions; infectious lesions such as those caused by HIV, herpes zoster or herpes simplex virus, Lyme disease, tuberculosis or syphilis; degenerative lesions and diseases and demyelinated lesions.
  • the present invention may be used to treat, for example, diabetes (including diabetic neuropathy, Bell's palsy), systemic lupus erythematosus, sarcoidosis, multiple sclerosis, human immunodeficiency virus-associated myelopathy, transverse myelopathy or various etiologies, progressive multifocal leukoencephalopathy, central pontine myelinolysis, Parkinson's disease, Alzheimer's disease, Huntington's chorea, amyotrophic lateral sclerosis, cerebral infarction or ischemia, spinal cord infarction or ischemia, progressive spinal muscular atrophy, progressive bulbar palsy, primary lateral sclerosis, infantile and juvenile muscular atrophy, progressive bulbar paralysis of childhood (Fazio-Londe syndrome), poliomyelitis and the post polio syndrome, and Hereditary Motorsensory Neuropathy (Charcot-Marie-Tooth Disease).
  • diabetes including diabetic neuropathy, Bell's palsy
  • the present invention may further be useful in the promotion of tissue regeneration and repair.
  • the present invention may also be used to treat diseases associated with defective tissue repair and regeneration such as, for example, cinhosis of the liver, hypertrophic scar formation and psoriasis.
  • the invention may also be useful in the treatment of vomropenia or anemia and in techniques of organ regeneration and tissue engineering.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising administering a therapeutically effective amount of at least one compound identified by the method of the present invention and a pharmaceutically acceptable carrier, diluent or excipients (including combinations thereof).
  • the pharmaceutical compositions may be for human or animal usage in human and veterinary medicine and will typically comprise any one or more of a pharmaceutically acceptable diluent, carrier, or excipient.
  • Acceptable carriers or diluents for therapeutic use are well known in the pharmaceutical art, and are described, for example, in Remington's Phannaceutical Sciences, Mack Publishing Co. (A. R. Gennaro edit. 1985).
  • the choice of pharmaceutical carrier, excipient or diluent can be selected with regard to the intended route of administration and standard pharmaceutical practice.
  • the pharmaceutical compositions may comprise as - or in addition to - the carrier, excipient or diluent any suitable binder(s), lubricant(s), suspending agent(s), coating agent(s), solubilising agent(s).
  • Preservatives, stabilizers, dyes and even flavoring agents may be provided in the pharmaceutical composition.
  • preservatives include sodium benzoate, sorbic acid and esters of p-hydroxybenzoic acid.
  • Antioxidants and suspending agents may be also used.
  • composition formulation requirements dependent on the different delivery systems.
  • the pharmaceutical composition of the present invention may be formulated to be delivered using a mini-pump or by a mucosal route, for example, as a nasal spray or aerosol for inhalation or ingestable solution, or parenterally in which the composition is formulated by an injectable form, for delivery, by, for example, an intravenous, intramuscular or subcutaneous route.
  • the formulation may be designed to be delivered by both routes.
  • the compound is to be delivered mucosally through the gastrointestinal mucosa, it should be able to remain stable during transit though the gastrointestinal tract; for example, it should be resistant to proteolytic degradation, stable at acid pH and resistant to the detergent effects of bile.
  • compositions can be administered by inhalation, in the form of a suppository or pessary, topically in the form of a lotion, solution, cream, ointment or dusting powder, by use of a skin patch, orally in the form of tablets containing excipients such as starch or lactose, or in capsules or ovules either alone or in admixture with excipients, or in the form of elixirs, solutions or suspensions containing flavouring or colouring agents, or they can be injected parenterally, for example intravenously, intramuscularly or subcutaneously.
  • compositions may be best used in the form of a sterile aqueous solution which may contain other substances, for example enough salts or monosaccharides to make the solution isotonic with blood.
  • a sterile aqueous solution which may contain other substances, for example enough salts or monosaccharides to make the solution isotonic with blood.
  • buccal or sublingual administration the compositions may be administered in the form of tablets or lozenges which can be formulated in a conventional manner.
  • a physician will determine the actual dosage which will be most suitable for an individual subject and it will vary with the age, weight and response of the particular patient.
  • the dosages below are exemplary of the average case. There can, of course, be individual instances where higher or lower dosage ranges are merited.
  • compositions of the present invention may be administered by direct injection.
  • the composition may be formulated for parenteral, mucosal, intramuscular, intravenous, subcutaneous, intraocular or transdermal administration.
  • the term "administered” includes delivery by viral or non- viral techniques.
  • Viral delivery mechanisms include but are not limited to adenoviral vectors, adeno-associated viral (AAV) vectors, herpes viral vectors, retroviral vectors, lentiviral vectors, and baculoviral vectors.
  • Non- viral delivery mechanisms include lipid mediated transfection, liposomes, immunoliposomes, lipofectin, cationic facial amphiphiles (CFAs) and combinations thereof.
  • the routes for such delivery mechanisms include but are not limited to mucosal, nasal, oral, parenteral, gastrointestinal, topical, or sublingual routes.
  • administered includes but is not limited to delivery by a mucosal route, for example, as a nasal spray or aerosol for inhalation or as an ingestable solution; a parenteral route where delivery is by an injectable form, such as, for example, an intravenous, intramuscular, intradermal, infra-articular, infrathecal, infra-peritoneal or subcutaneous route, or via the alimentary tract (for example, via the Peyers patches).
  • an injectable form such as, for example, an intravenous, intramuscular, intradermal, infra-articular, infrathecal, infra-peritoneal or subcutaneous route, or via the alimentary tract (for example, via the Peyers patches).
  • the routes of administration and dosages described are intended only as a guide since a skilled practitioner will be able to determine readily the optimum route of adminisfration and dosage for any particular patient depending on, for example, the age, weight and condition of the patient.
  • the pharmaceutical compositions are in unit dosage form.
  • the present invention includes both human and veterinary applications.
  • antigen-presenting cells may be "professional" antigen presenting cells or may be another cell that may be induced to present antigen to T cells.
  • APC precursor may be used which differentiates or is activated under the conditions of culture to produce an APC.
  • APCs include dendritic cells (DCs) such as interdigitating DCs or folhcular DCs, Langerhans cells, PBMCs, macrophages, B-lymphocytes, or other cell types such as epithelial cells, fibroblasts or endothelial cells, activated or engineered by transfection to express a MHC molecule (Class I or II) on their surfaces.
  • DCs dendritic cells
  • PBMCs macrophages
  • B-lymphocytes or other cell types
  • Precursors of APCs include CD34 + cells, monocytes, fibroblasts and endothelial cells.
  • the APCs or precursors may be modified by the culture conditions or may be genetically modified, for instance by transfection of one or more genes encoding proteins which play a role in antigen presentation and/or in combination of selected cytokine genes which would promote to immune potentiation (for example IL-2, IL-12, IFN- ⁇ , TNF- ⁇ , IL-18 etc.).
  • proteins include MHC molecules (Class I or Class II), CD80, CD86, or CD40.
  • DCs or DC- precursors are included as a source of APCs.
  • Dendritic cells can be isolated/prepared by a number of means, for example they can either be purified directly from peripheral blood, or generated from CD34 precursor cells for example after mobilisation into peripheral blood by freatment with GM-CSF, or directly from bone marrow. From peripheral blood, adherent precursors can be treated with a GM-CSF/IL-4 mixture (Inaba K, et al. (1992) J. Exp. Med. 175: 1157-1167 (Inaba)), or from bone marrow, non-adherent CD34 + cells can be treated with GM-CSF and TNF-a (Caux C, et al. (1992) Nature 360: 258-261 (Caux)).
  • GM-CSF/IL-4 mixture Inaba K, et al. (1992) J. Exp. Med. 175: 1157-1167 (Inaba)
  • non-adherent CD34 + cells can be treated with GM-CSF and TNF-a (Caux C, et al. (19
  • DCs can also be routinely prepared from the peripheral blood of human volunteers, similarly to the method of Sallusto and Lanzavecchia (Sallusto F and Lanzavecchia A (1994) J. Exp. Med. 179: 1109-1118) using purified peripheral blood mononucleocytes (PBMCs) and treating 2 hour adherent cells with GM- CSF and IL-4. If required, these may be depleted of CD19 + B cells and CD3 + , CD2 + T cells using magnetic beads (Coffin RS, et al. (1998) Gene Therapy 5: 718-722 (Coffin)). Culture conditions may include other cytokines such as GM-CSF or IL-4 for the maintenance and, or activity of the dendritic cells or other antigen presenting cells.
  • PBMCs peripheral blood mononucleocytes
  • the term "antigen presenting cell or the like" are used herein is not intended to be limited to APCs.
  • APCs any vehicle capable of presenting to the T cell population may be used, for the sake of convenience the term APCs is used to refer to all these.
  • prefened examples of suitable APCs include dendritic cells, L cells, hybridomas, fibroblasts, lymphomas, macrophages, B cells or synthetic APCs such as lipid membranes.
  • T cells from any suitable source such as a healthy patient, may be used and may be obtained from blood or another source (such as lymph nodes, spleen, or bone marrow). They may optionally be enriched or purified by standard procedures.
  • the T cells may be used in combination with other immune cells, obtained from the same or a different individual.
  • whole blood may be used or leukocyte enriched blood or purified white blood cells as a source of T cells and other cell types. It is particularly preferred to use helper T cells (CD4 + ).
  • other T cells such as CD8 + cells may be used. It may also be convenient to use cell lines such as T cell hybridomas.
  • T cells/APCs may be cultured as described above. For example, they may be prepared for administration to a patient or incubated with T cells in vitro (ex vivo).
  • modified cells of the present invention are preferably administered to a host by direct injection into the lymph nodes of the patient.
  • a host by direct injection into the lymph nodes of the patient.
  • the cells will be taken from an enriched cell population.
  • the term "enriched" as applied to the cell populations of the invention refers to a more homogeneous population of cells which have fewer other cells with which they are naturally associated.
  • An enriched population of cells can be achieved by several methods known in the art. For example, an enriched population of T-cells can be obtained using immunoaffinity chromatography using monoclonal antibodies specific for determinants found only on T-cells. Enriched populations can also be obtained from mixed cell suspensions by positive selection (collecting only the desired cells) or negative selection (removing the undesirable cells). The technology for capturing specific cells on affinity materials is well known in the art (Wigzel, et al., J. Exp. Med., 128:23, 1969; Mage, et al., J.
  • Monoclonal antibodies against antigens specific for mature, differentiated cells have been used in a variety of negative selection strategies to remove undesired cells, for example, to deplete T-cells or malignant cells from allogeneic or autologous marrow grafts, respectively (Gee, et al., J.N.C.I. 80:154, 1988).
  • Purification of human hematopoietic cells by negative selection with monoclonal antibodies and immunomagnetic microspheres can be accomplished using multiple monoclonal antibodies (Griffin, et al., Blood, 63:904, 1984).
  • Procedures for separation of cells may include magnetic separation, using antibody coated magnetic beads, affinity chromatography, cytotoxic agents joined to a monoclonal antibody or used in conjunction with a monoclonal antibody, for example, complement and cytotoxins, and "panning" with antibodies attached to a solid matrix, for example, plate, or other convenient teclinique.
  • Techniques providing accurate separation include fluorescence activated cell sorters, which can have varying degrees of sophistication, for example, a plurality of color channels, low angle and obtuse light scattering detecting channels, impedance channels, etc.
  • the therapeutic agents used in the present invention may be administered directly to patients in vivo.
  • the agents may be administered to cells such as T cells and/or APCs in an ex vivo manner.
  • leukocytes such as T cells or APCs may be obtained from a patient or donor in known manner, treated/incubated ex vivo in the manner of the present invention, and then administered to a patient.
  • routes of adminisfration may be employed if desired. For example, where appropriate one component (such as the modulator of Notch signalling) may be administered ex-vivo and the other may be administered in vivo, or vice versa.
  • T-cells and APCs as described above are cultured in a suitable culture medium such as DMEM or other defined media, optionally in the presence of fetal calf serum.
  • a suitable culture medium such as DMEM or other defined media, optionally in the presence of fetal calf serum.
  • Polypeptide substances may be administered to T-cells and/or APCs by infroducing nucleic acid constructs/viral vectors encoding the polypeptide into cells under conditions that allow for expression of the polypeptide in the T-cell and/or APC.
  • nucleic acid constructs encoding antisense constructs may be introduced into the T-cells and/or APCs by transfection, viral infection or viral transduction.
  • nucleotide sequences will be operably linked to confrol sequences, including promoters/enhancers and other expression regulation signals.
  • operably linked means that the components described are in a relationship permitting them to function in their intended manner.
  • a regulatory sequence "operably linked" to a coding sequence is peferably ligated in such a way that expression of the coding sequence is achieved under condition compatible with the control sequences.
  • the promoter is typically selected from promoters which are functional in mammalian cells, although prokaryotic promoters and promoters functional in other eukaryotic cells may be used.
  • the promoter is typically derived from promoter sequences of viral or eukaryotic genes. For example, it may be a promoter derived from the genome of a cell in which expression is to occur. With respect to eukaryotic promoters, they may be promoters that function in a ubiquitous manner (such as promoters of a-actin, b-actin, tubulin) or, alternatively, a tissue-specific manner (such as promoters of the genes for pyruvate kinase).
  • Tissue-specific promoters specific for lymphocytes, dendritic cells, skin, brain cells and epithelial cells within the eye are particularly prefened, for example the CD2, CD1 lc, keratin 14, Wnt-1 and Rhodopsin promoters respectively.
  • the epithelial cell promoter SPC is used. They may also be promoters that respond to specific stimuli, for example promoters that bind steroid hormone receptors.
  • Viral promoters may also be used, for example the Moloney murine leukaemia virus long terminal repeat (MMLV LTR) promoter, the rous sarcoma virus (RS V) LTR promoter or the human cytomegalovirus (CMV) IE promoter.
  • MMLV LTR Moloney murine leukaemia virus long terminal repeat
  • RS V rous sarcoma virus
  • CMV human cytomegalovirus
  • the promoters may also be advantageous for the promoters to be inducible so that the levels of expression of the heterologous gene can be regulated during the life-time of the cell. Inducible means that the levels of expression obtained using the promoter can be regulated.
  • any of the above promoters may be modified by the addition of further regulatory sequences, for example enhancer sequences.
  • Chimeric promoters may also be used comprising sequence elements from two or more different promoters.
  • the regulatory sequences may be cell specific such that the gene of interest is only expressed in cells of use in the present invention.
  • Such cells include, for example, APCs and T-cells.
  • a small aliquot of cells may be tested for up-regulation of Notch signalling activity as described above.
  • the cells may be prepared for administration to a patient or incubated with T-cells in vitro (ex vivo).
  • any of the assays described above can be adapted to monitor or to detect reduced reactivity and tolerisation in immune cells for use in clinical applications.
  • Such assays will involve, for example, detecting increased Notch-ligand expression or activity in host cells or monitoring Notch cleavage in donor cells. Further methods of monitoring immune cell activity are set out below.
  • Immune cell activity may be monitored by any suitable method known to those skilled in the art. For example, cytotoxic activity may be monitored. Natural killer (NK) cells will demonstrate enhanced cytotoxic activity after activation. Therefore any drop in or stabilisation of cytotoxicity will be an indication of reduced reactivity.
  • NK Natural killer
  • leukocytes Once activated, leukocytes express a variety of new cell surface antigens.
  • NK cells for example, will express transferrin receptor, HLA-DR and the CD25 IL-2 receptor after activation. Reduced reactivity may therefore be assayed by monitoring expression of these antigens.
  • EA-1 and MLR3 are glycoproteins having major components of 28kD and 32kD.
  • EA-1 and MLR3 are not HLA class II antigens and an MLR3 Mab will block IL-1 binding. These antigens appear on activated T- cells within 18 hours and can therefore be used to monitor immune cell reactivity.
  • leukocyte reactivity may be monitored as described in EP 0325489, which is incorporated herein by reference. Briefly this is accomplished using a monoclonal antibody ("Anti-Leu23”) which interacts with a cellular antigen recognised by the monoclonal antibody produced by the hybridoma designated as ATCC No. HB-9627.
  • Anti-Leu23 a monoclonal antibody
  • ATCC No. HB-9627 a monoclonal antibody
  • Anti-Leu 23 recognises a cell surface antigen on activated and antigen stimulated leukocytes. On activated NK cells, the antigen, Leu 23, is expressed within 4 hours after activation and continues to be expressed as late as 72 hours after activation. Leu 23 is a disulfide-linked homodimer composed of 24 kD subunits with at least two N-linked carbohydrates. Because the appearance of Leu 23 on NK cells conelates with the development of cytotoxicity and because the appearance of Leu 23 on certain T-cells conelates with stimulation of the T-cell antigen receptor complex, Anti-Leu 23 is useful in monitoring the reactivity of leukocytes.
  • immune cells may be used to present antigens or allergens and/or may be treated to modulate expression or interaction of Notch, a Notch ligand or the Notch signalling pathway.
  • APCs Antigen Presenting Cells
  • DMEM fetal calf serum
  • Optimum cytokine concenfrations may be determined by tifration.
  • One or more modulators of Notch signalling are then typically added to the culture medium together with the antigen of interest.
  • the antigen may be added before, after or at substantially the same time as the substance(s).
  • Cells are typically incubated with the substance(s) and antigen for at least one hour, preferably at least 3 hours, suitably at least 9, 12, 24, 48 or 36 or more hours at 37°C. If required, a small aliquot of cells may be tested for modulated target gene expression as described above. Alternatively, cell activity may be measured by the inhibition of T cell activation by monitoring surface markers, cytokine secretion or proliferation as described in
  • polypeptide substances may in one embodiment be administered to APCs by introducing nucleic acid consfructs/viral vectors encoding the polypeptide into cells under conditions that allow for expression of the polypeptide in the APC.
  • nucleic acid constructs encoding antigens may be introduced into the APCs by transfection, viral infection or viral transduction.
  • T cells are generally co-cultured with the APCs.
  • it may be prefened to prepare primed APCs first and then incubate them with T cells.
  • primed APCs once the primed APCs have been prepared, they may be pelleted and washed with PBS before being resuspended in fresh culture medium. This has the advantage that if, for example, it is desired to treat the T cells with a different substance(s), then the T cell will not be brought into contact with the different substance(s) used with the APC.
  • primed APCs Once primed APCs have been prepared, it is not always necessary to administer any substances to the T cell since the primed APC is itself capable of inducing immunotolerance leading to increased Notch or Notch ligand expression in the T cell, presumably via Notch/Notch ligand interactions between the primed APC and T cell.
  • Incubations will typically be for at least 1 hour, preferably at least 3, 6 , 12, 24, 48 or 36 or more hours, in suitable culture medium at 37°C.
  • the progress of Notch signalling may be determined for a small aliquot of cells using the methods described above.
  • T cells fransfected with a nucleic acid constract directing the expression of, for example Delta, may be used as a confrol.
  • Induction of immunotolerance may be determined, for example, by subsequently challenging T cells with antigen and measuring IL-2 production compared with confrol cells not exposed to APCs.
  • Primed T cells or B cells may also be used to induce immunotolerance in other T cells or B cells in the absence of APCs using similar culture techniques and incubation times.
  • T cells may be cultured and primed in the absence of APCs by use of APC substitutes such as anti-TCR antibodies (e.g. anti-CD3) with or without antibodies to costimulatory molecules (e.g. anti-CD28) or alternatively T cells may be activated with MHC- peptide complexes (e.g. tetramers).
  • APC substitutes such as anti-TCR antibodies (e.g. anti-CD3) with or without antibodies to costimulatory molecules (e.g. anti-CD28) or alternatively T cells may be activated with MHC- peptide complexes (e.g. tetramers).
  • Induction of immunotolerance may be determined by subsequently challenging T cells with antigen and measuring IL-2 production compared with control cells not exposed to APCs.
  • T cells or B cells which have been primed in this way may be used according to the invention to promote or increase immunotolerance in other T cells or B cells.
  • CD4+ cell purification Spleens were removed from mice (variously Balb/c females, 8-10 weeks, C57B/6 females, 8- 10 weeks, CARDl females, 8-10 weeks (DO 11.10 transgenic, CAR transgenic)) and passed through a 0.2 ⁇ M cell strainer into 20ml R10F medium (R10F-RPMI 1640 media (Gibco Cat No 22409) plus 2mM L-glutamine, 50 ⁇ g/ml Penicillin, 50 ⁇ g/ml Streptomycin, 5 x 10 "5 M ⁇ -mercapto-ethanol in 10%o fetal calf serum). The cell suspension was spun (1150 ⁇ m 5min) and the media removed.
  • R10F medium R10F-RPMI 1640 media (Gibco Cat No 22409) plus 2mM L-glutamine, 50 ⁇ g/ml Penicillin, 50 ⁇ g/ml Streptomycin, 5 x 10 "5 M ⁇ -mercapto-ethanol in 10%o fetal calf
  • CD4+ cells were purified from the suspensions by positive selection on a Magnetic Associated Cell Sorter (MACS) column (Miltenyi Biotec, Bisley, UK: Cat No 130-042-401) using CD4 (L3T4) beads (Miltenyi Biotec Cat No 130-049-201), according to the manufacturer's directions.
  • MCS Magnetic Associated Cell Sorter
  • DPBS Dulbecco's Phosphate Buffered Saline
  • 1 ⁇ g/ml anti-CD3 antibody Pharmingen, San Diego, US: Cat No 553058, Clone No 145-2C11
  • l ⁇ g/ml anti-IgG4 antibody Pharmingen Cat No 555878
  • lOO ⁇ l of coating mixture was used per well. Plates were incubated overnight at 4°C then washed with DPBS. Each well then received either lOO ⁇ l DPBS or lOO ⁇ l DPBS plus lO ⁇ g/ml Notch ligand (mouse Delta 1 extracellular domain/Ig4Fc fusion protein; Fc-delta).
  • the plates were incubated for 2-3 hours at 37°C then washed again with DPBS before cells (prepared as in Example 1) were added.
  • the plates were coated with DPBS plus l ⁇ g/ml anti-hamsterlgG antibody (Pharmingen Cat No 554007) plus l ⁇ g/ml anti-IgG4 antibody. lOO ⁇ l of coating mixture was added per well. Plates were incubated overnight at 4°C then washed with DPBS. Each well then received either lOO ⁇ l DPBS plus anti-CD3 antibody (l ⁇ g/ml) or, lOO ⁇ l DPBS plus anti-CD3 antibody (l ⁇ g/ml) plus Fc-delta (lO ⁇ g/ml). The plates were incubated for 2-3 hours at 37°C then washed again with DPBS before cells (prepared as in Example 1) were added.
  • CD4+ cells were cultured in 96 well, flat-bottomed plates pre-coated according to Example 2 (A) or 2 (B). Cells were re-suspended, following counting, at 2 x 10 6 /ml in R10F medium plus 4 ⁇ g/ml anti-CD28 antibody (Pharmingen, Cat No 553294, Clone No 37.51). lOO ⁇ l cell suspension was added per well. lOO ⁇ l of R10F medium was then added to each well to give a final volume of 200 ⁇ l (2 x 10 5 cells/well, anti-CD28 final concentration 2 ⁇ g/ml) The plates were then incubated at 37°C for 72 hours.
  • 125 ⁇ l supernatant was then removed from each well and stored at -20°C until tested by ELISA for IL-10, IFNg and IL-13using antibody pairs from R & D Systems (Abingdon, UK).
  • the cells were then split 1 in 3 into new wells (not coated) and fed with R10F medium plus recombinant human IL-2 (2.5ng/ml, PeproTech Inc, London, UK: Cat No 200-02).
  • Murine (Balb/c) stimulated CD4 + T-cells from Example 3 were harvested at 4, 16 and 24 hours.
  • Total cellular RNA was isolated using the RNeasyTM RNA isolation kit (Qiagen, Crawley, UK) according to the manufacturer's guidelines.
  • RNA was reverse transcribed using SuperscriptTM II Reverse Transcriptase (Invitrogen, Paisley, UK) using Oligo dT (12 . 18) or a random decamer mix according to the manufacturer's guidelines.
  • Oligo dT( 12 -i8)- and random decamer-primed cDNAs were mixed in equal proportions to provide the working cDNA sample for real-time quantitative PCR analysis.
  • mice Hes-1 Forward GGTGCTGATAACAGCGGAAT Reverse ATTTTTGGAATCCTTCACGC
  • C p The endpoint used in real-time PCR quantification, the Crossing Point (C p ), is defined as the PCR cycle number that crosses an algorithm-defined signal threshold.
  • Quantitative analysis of gene-specific cDNA was achieved firstly by generating a set of standards using the C p s from a set of serially-diluted gene-specific amplicons which had been previously cloned into a plasmid vector (pCR2.1, Invitrogen). These serial dilutions fall into a standard curve against which the C p s from the cDNA samples were compared. Using this system, expression levels of the 18S rRNA house-keeping gene were generated for each cDNA sample.
  • Hes-1 was then analysed by the same method using serially-diluted Hes-1 -specific standards, and the Hes-1 value divided by the 18S rRNA value to generate a value, which represents the relative expression of Hes-1 in each cDNA sample. All Cp analysis was performed using the Second Derivative Maximum algorithm within the Lightcycler system software.
  • Example 3 The procedure of Example 3 was then followed, except that instead of adding lOO ⁇ l R10F medium per well as in Example 3, lOO ⁇ l of polarising cocktail was added per well as follows:
  • Un-polarised cells R10F medium.
  • Thl polarised cells R10F medium plus anti-IL-4 antibody (lO ⁇ g/ml, Pharmingen Cat No
  • Th2 polarised cells RlOFmedium plus anti-IL-12 antibody (lO ⁇ g/ml, Pharmingen Cat No 554475) plus anti-IFNg antibody (l ⁇ g/ml, Pharmingen Cat No 554408) plus IL-4 (lOng/ml, Peprotech Cat No 214-14).
  • Example 2(A) (with the modification that ligand was not added to the plate) and Example 3 (with the modification that soluble Fc-delta was added with the RIOF medium) was used to compare soluble Fc-delta with plate-bound Fc-delta against controls. Results are shown in Figure 12.
  • 96-well flat-bottomed plates were coated with PBS plus l ⁇ g/ml anti-CD3 antibody. The plates were incubated overnight at 4°C then washed with DPBS.
  • the cells were re-suspended at 2 x 10 6 /ml in R10F medium plus anti-CD28 antibody (4 ⁇ g/ml). lOO ⁇ l cell suspension was added per well. lOO ⁇ l of R10F medium was then added per well to give a final volume of 200 ⁇ l. (2 x 10 5 cells/well, anti-CD28 final concentration 2 ⁇ g/ml). The plates were then incubated at 37°C for 72 hours. After 72 hours supernatants were removed for ELISA as described in Example 3 (primary stimulation).
  • Example 3 Primary stimulated cells from Example 3 were harvested after 7 days and restimulated with APCs of the same strain (2 x 10 4 per well) plus anti-CD3 antibody.
  • Mouse spleen cells were isolated as described in Example 1 up to the counting step. Thy-1.2 antibody-binding cells were then removed on a MACS column and the flowthrough was recovered and treated with mitomycin-C for 45 minutes then added to a 96 well plate in lOO ⁇ l RIOF medium with equal numbers of cells from Example 3 and 0.5 ⁇ g/ml anti-CD3 antibody.
  • Cell proliferation was measured using a kit from Roche Molecular Biochemicals, Cell Proliferation ELISA, BrdU (chemiluminescent) 1 669 915, according to the manufacturer's instructions. Plates were pulsed at 72 hours and read on a luminometer.
  • Cytokines (IL-10 and IFN- ⁇ ) were measured as described in Example 3. Results are shown in Figure 13.
  • TPl TPl; equivalent to 2 CBFl repeats
  • BamHI and Bglll cohesive ends was generated as follows:
  • This sequence was pentamerised by repeated insertion into a Bglll site and the resulting TPl pentamer (equivalent to 10 CBFl repeats) was inserted into pGL3-AdTATA at the Bglll site to generate plasmid pLOR91.
  • a cDNA clone spanning the complete coding sequence of the human Notch2 gene was constructed as follows. A 3' cDNA fragment encoding the entire intracellular domain and a portion of the extracellular domain was isolated from a human placental cDNA library (OriGene Technologies Ltd., USA) using a PCR-based screening strategy. The remaining 5' coding sequence was isolated using a RACE (Rapid Amplification of cDNA Ends) strategy and ligated onto the existing 3' fragment using a unique restriction site common to both fragments (Cla I).
  • RACE Rapid Amplification of cDNA Ends
  • pLOR92 When expressed in mammalian cells, pLOR92 thus expresses the full-length human Notch2 protein with V5 and His tags at the 3' end of the intracellular domain.
  • Wild-type CHO-K1 cells (eg see ATCC No CCL 61) were transfected with pLOR92 (pcDNA3.1-FLNotch2-V5-His) using Lipfectamine 2000TM (Invitrogen) to generate a stable CHO cell clone expressing full length human Notch2 (N2).
  • Transfectant clones were selected in Dulbecco's Modified Eagle Medium (DMEM) plus 10%o heat inactivated fetal calf serum ((HI)FCS) plus glutamine plus Penicillin-Streptomycin (P/S) plus 1 mg/ml G418 (GeneticinTM - Invitrogen) in 96-well plates using limiting dilution.
  • DMEM Dulbecco's Modified Eagle Medium
  • (HI)FCS) 10%o heat inactivated fetal calf serum
  • P/S Penicillin-Streptomycin
  • G418 GeneticinTM - Invitrogen
  • CHO-N2 stable clone N27 was found to give high levels of induction when transiently transfected with pLOR91 (lOxCBFl-Luc) and co-cultured with the stable CHO cell clone expressing full length human DLL1 (CHO-Deltal).
  • a hygromycin gene cassette (obtainable from pcDNA3.1/hygro, Invitrogen) was inserted into pLOR91 (lOxCBFl-Luc) using BamHI and Sail and this vector (lOxCBFl-Luc-hygro) was fransfected into the CHO-N2 stable clone (N27) using Lipfectamine 2000 (Invitrogen).
  • Transfectant clones were selected in DMEM plus 10%(HI)FCS plus glutamine plus P/S plus 0.4 mg/ml hygromycin B (Invitrogen) plus 0.5 mg/ml G418 (Invitrogen) in 96-well plates using limiting dilution. Individual colonies were expanded in DMEM plus 10%(HI)FCS plus glutamine plus P/S + 0.2 mg/ml hygromycin B plus 0.5 mg/ml G418 (Invitrogen).
  • Clones were tested by co-culture with a stable CHO cell clone expressing FL human DLL1.
  • Three stable reporter cell lines were produced N27#ll, N27#17 and N27#36.
  • N27#ll was selected for further use because of its low background signal in the absence of Notch signalling, and hence high fold induction when signalling is initiated.
  • Assays were set up in 96-well plates with 2 x 10 4 N27#ll cells per well in 100 ⁇ l per well of DMEM plus 10%(HI)FCS plus glutamine plus P/S.
  • CHO-N2 (Clone N27) cells were maintained in DMEM plus 10%(HI)FCS plus glutamine plus P/S plus 0.5 mg/ml G418 and a T 80 flask of the CHO-N2 cells was fransfected as follows. The medium on the cells was replaced with 8 ml of fresh in DMEM plus 10%(HI)FCS plus glutamine plus P/S. In a sterile bijou 10 ⁇ g of pLOR91 (lOxCBFl-Luc) was added to OptiMem (Invitrogen) to give a final volume of 1 ml and mixed. In a second sterile bijou 20 ⁇ l of Lipofectamine 2000 reagent was added to 980 ⁇ l of OptiMem and mixed.
  • each bijou were mixed and left at room temperature for 20 minutes.
  • the 2 ml of transfection mixture was added to the flask of cells containing 8 ml of medium and the resulting mixture was left in a CO incubator overnight before removing the fransfected cells and adding to the 96-well plate containing the immobilised Notch ligand protein.
  • transfected CHO-N2 cells were removed using 0.02% ⁇ EDTA solution (Sigma), spun down and resuspended in 10 ml DMEM plus 10%(HI)FCS plus glutamine plus P/S. 10 ⁇ l of cells were counted and the cell density was adjusted to 2.0 x 10 5 cells/ml with fresh DMEM plus 10%(HI)FCS plus glutamine plus P/S. 100 ⁇ l per well was added to a 96-well tissue culture plate (flat bottom), i.e. 2.0 x 10 4 fransfected cells per well, using a multi-channel pipette and the plate was then incubated overnight.
  • the lid of the plate was sealed with parafilm and the plate was left at 4 °C overnight or at 37 °C for 2 hours.
  • the protein was then removed and the plate was washed with 200 ⁇ l of PBS.
  • the IVS, IRES, Neo and pA elements were removed from plasmid pIRESneo2 (Clontech, USA) and inserted into a pUC cloning vector downstream of a chicken beta-actin promoter (eg see GenBank Accession No E02199).
  • Mouse Delta-1 eg see GenBank Accession No NM_007865 was inserted between the actin promoter and IVS elements and a sequence with multiple stop codons in all three reading frames was inserted between the Delta and IVS elements.
  • the resulting constract was transfected into A20 cells using electroporation and G418 to provide A20 cells expressing mouse Deltal on their surfaces (A20-Delta).
  • CHO cells were maintained in DMEM plus 10%(HI)FCS plus glutamine plus P/S and CHO-hDeltal-V5-His (clone#10) cells were maintained in DMEM plus 10%(HI)FCS plus glutamine plus P/S plus 0.5mg/ml G418.
  • Cells were removed using 0.02% EDTA solution (Sigma), spun down and resuspended in 10 ml DMEM plus 10%(HI)FCS plus glutamine plus P/S. 10 ⁇ l of cells were counted and the cell density was adjusted to 5.0 x 10 5 cells/ml with fresh DMEM plus 10%(HI)FCS plus glutamine plus P/S. 300 ⁇ l of each cell line at 5.0 x 10 5 cells/ml was added into duplicate wells of a 96-well tissue culture plate. 150 ⁇ l of DMEM plus 10%(HI)FCS plus glutamine plus P/S was added in to the next 5 wells below each well.
  • 150 ⁇ l of cells were serially diluted into the next 4 wells giving cell density dilution of 5.0 x 10 5 cells/ml, 2.5 x 10 5 cells/ml, 1.25 x 10 5 cells/ml, 0.625 x 10 5 cells/ml, 0.3125 x 10 5 cells/ml and 0 cells/ml.
  • CHO-N2 cells 5 x 10 4 CHO-N2 cells were plated on a 96 well plate.
  • CHO-Delta or A20-Delta cells were titrated in as required (max ratio CHO-N2: CHO-Delta was 1:1, max ratio CHO-N2: A20- Delta was 1:2). The mixture was incubated overnight before conducting a luciferase assay.
  • Results of sample assays (using the stable CHO-Notch2-10xCBFl-Luc reporter cell line described above with (A) plate-immobilised human Delta- l/Ig4Fc fusion protein, (B) plate- immobilised mouse Delta-l/Ig4Fc fusion protein, (C) CHO / CHO-human Deltal co- cultured cells and (D) A20/ A20-mouse Deltal co-cultured cells as actives against conesponding controls) are shown in Figures 14 A to D.
  • Fc-tagged Notch ligands were immobilised on Streptavidin-Dynabeads (CELLection Biotin Binder Dynabeads [Cat. No. 115.21] at 4.0 x 10 8 beads/ml from Dynal (UK) Ltd; beads) in combination with biotinylated ⁇ -IgG-4 (clone JDC14 at 0.5 mg/ml from Pharmingen [Cat. No. 555879]) as follows:
  • the mixture was then spun down at 13,000 ⁇ m for 1 minute and the beads were resupsended in a 50 ⁇ l PBS per sample.
  • 50 ⁇ l of biotinylated ⁇ -IgG-4 -ooated beads were added to each sample and the mixture was incubated on a rotary shaker at 4 °C overnight.
  • the tube was then spun at 1000 ⁇ m for 5 minutes at room temperature.
  • the beads then were washed with 10 ml of PBS, spun down, resupended in 1 ml of PBS, transfened to a sterile Eppendorf tube, washed with a further 2 x 1 ml of PBS, spun down and resuspended beads in a final volume of 250 ⁇ l of DMEM plus 10%(HI)FCS plus glutamine plus P/S, i.e. at 1.0 x 10 5 beads/ ⁇ l.
  • Stable N27#ll cells from Example 8 were removed using 0.02% EDTA solution (Sigma), spun down and resuspended in 10 ml DMEM plus 10%»(HI)FCS plus glutamine plus P/S. 10 ⁇ l of cells were counted and the cell density was adjusted to 1.0 x 10 5 cells/ml with fresh DMEM plus 10%(HI)FCS plus glutamine plus P/S. 1.0 x 10 5 of the cells were plated out per well of a 24-well plate in a 1 ml volume of DMEM plus 10%(HI)FCS plus glutamine plus P/S and cells were placed in an incubator to settle down for at least 30 minutes.
  • M450 Sfreptavidin Dynabeads were coated with anti-hamster-IgGl biotinylated monoclonal antibody, anti-human-IgG4 biotinylated monoclonal antibody or both antibodies and rotated for 2 hours at room temperature.
  • Beads were washed three times with PBS (1ml).
  • the anti-hamster-IgGl beads were then further incubated with anti-CD3 ⁇ chain monoclonal antibody, the anti-human-IgG4 beads were further incubated with Fc-Delta, and the double coated beads incubated with both anti- CD3 ⁇ chain monoclonal antibody and Fc-Delta.
  • Beads were rotated overnight at 4°C, washed three times with PBS (1ml) and resuspended.
  • T-cell assays were carried out with CD4+ T-cells and the beads. Supernatants were removed after 72 hours and cytokines measured by ELISA as described in Example 3. Results are shown in Figure 15.
  • PBMC Human peripheral blood mononuclear cells
  • Human CD4+ T cells were isolated by positive selection using anti-CD4 microbeads from Miltenyi Biotech according to the manufacturer's instructions.
  • the CD4+ T cells were incubated in triplicates in a 96-well-plate (flat bottom) at 10 5 CD4/well/200 ⁇ l in RPMI medium containing 10% FCS, glutamine, penicillin, streptomycin and ⁇ 2 -mercaptoethanol.
  • Cytokine production was induced by stimulating the cells with anti-CD3/CD28 T cell expander beads from Dynal at a 1:1 ratio (bead/cell) or plate bound anti-CD3 (clone UCHT1, BD Biosciences, 5 ⁇ g/ml) and soluble anti-CD28 (clone CD28.2, BD Biosciences, 2 ⁇ g/ml).
  • Beads coated with mouse DeltalEC domain-hIgG4 fusion protein prepared as described above with the modifications that incubation with human IgG4 was for 30-40 minutes at room temperature and incubation with Delta-Fc was for two hours at room temperature
  • control beads were added in some of the wells at a 10:1 ratio (beads/cell).
  • Example 11 The procedure of Example 11 was repeated except that the ratio of control beads to cells and mouse Deltal -hIgG4 fusion protein coated beads to cells was varied between 16:1 and 0.25:1 (variously 16:1, 8:1, 4:1, 2:1, 1:1, 0.5:1, 0.25:1) and human Deltal-hIgG4 fusion protein coated beads were also used at the same ratios for comparison.
  • Example 11 The procedure of Example 11 was repeated except that prior to the stimulation the human CD4+ were separated into CD45RO+ (memory cells) and CD45RO- (naive cells, data not shown on the slide).
  • the magnetic separation was done using anti-CD4 Multisort microbeads (cat.No. 551-01) and then anti-CD45RO microbeads (cat.No.460-01) supplied by Miltenyi Biotech and following Miltenyi' s protocol.
  • Spleens were removed from mice (variously Balb/c females, 8-10 weeks, C57B/6 females, 8- 10 weeks, CARDl females, 8-10 weeks (DO 11.10 transgenic, CAR transgenic)) and passed through a 0.2 ⁇ M cell strainer into 20ml R10F medium (R10F-RPMI 1640 media (Gibco Cat No 22409) plus 2mM L-glutamine, 50 ⁇ g/ml Penicillin, 50 ⁇ g/ml Streptomycin, 5 x 10 "5 M ⁇ -mercapto-ethanol in 10% fetal calf serum). The cell suspension was spun (1150 ⁇ m 5min) and the media removed.
  • CD4+ cells were purified from the suspensions by positive selection on a Magnetic Associated Cell Sorter (MACS) column (Miltenyi Biotec, Bisley, UK: Cat No 130-042-401) using CD4 (L3T4) beads (Miltenyi Biotec Cat No 130-049-201), according to the manufacturer's directions.
  • ACK lysis buffer 0.15M NH C1, l.OM KHC0 3 , O.lmM Na 2 EDTA in double distilled water
  • spleen to lyse red blood cells.
  • CD4+ cells were purified from the suspensions by positive selection on a Magnetic Associated Cell Sorter (MACS) column (Miltenyi Biotec, Bisley, UK: Cat No 130-042-401) using CD4 (L3T4) beads (Miltenyi Biotec Cat No 130-049-201), according to the manufacturer's directions.
  • MCS Magnetic Associated Cell Sorter
  • 96 well flat-bottomed plates were coated with Dulbecco's Phosphate Buffered Saline (DPBS) plus l ⁇ g/ml anti-CD3 antibody (Pharmingen, San Diego, US: Cat No 553058, Clone No 145-2C11) plus l ⁇ g/ml anti-IgG4 antibody (Pharmingen Cat No 555878). lOO ⁇ l of coating mixture was used per well. Plates were incubated overnight at 4°C then washed with DPBS.
  • DPBS Dulbecco's Phosphate Buffered Saline
  • l ⁇ g/ml anti-CD3 antibody Pharmingen, San Diego, US: Cat No 553058, Clone No 145-2C11
  • l ⁇ g/ml anti-IgG4 antibody Pharmingen Cat No 555878
  • CD4+ cells were cultured in 96 well, flat-bottomed plates pre-coated as in (ii) above. Cells were re-suspended, following counting, at 2 x 10 6 /ml in RIOF medium plus 4 ⁇ g/ml anti- CD28 antibody (Pharmingen, Cat No 553294, Clone No 37.51). lOO ⁇ l cell suspension was added per well. lOO ⁇ l of polarising or control medium was then added to each well to give a final volume of 200 ⁇ l (2 x 10 5 cells/well, anti-CD28 final concentration 2 ⁇ g/ml) as follows:
  • Un-polarised cells RIOF medium.
  • Thl polarised cells RIOF medium plus anti-IL-4 antibody (lO ⁇ g/ml, Pharmingen Cat No
  • IL-12 (lOng/ml, Peprotech 210-12).
  • Th2 polarised cells RlOFmedium plus anti-IL-12 antibody (lO ⁇ g/ml, Pharmingen Cat No 554475) plus anti-IFNg antibody (l ⁇ g/ml, Pharmingen Cat No 554408) plus IL-4 (lOng/ml, Peprotech Cat No 214-14).
  • 125 ⁇ l supernatant was then removed from each well and stored at -20°C until tested by ELISA for IL-10 and TNFa using antibody pairs from R & D Systems (Abingdon, UK). The cells were then split 1 in 3 into new wells (not coated) and fed with R10F medium plus recombinant human IL-2 (2.5ng/ml, PeproTech Inc, London, UK: Cat No 200-02).
  • fusion protein comprising the exfracellular domain of human Deltal fused to the Fc domain of human IgG4 (refened to herein as "hDeltal-IgG4Fc" and also refened to in the accompanying Figures as "D1E8G4" and "DlE8Fc4") was prepared by inserting a nucleotide sequence coding for the extracellular domain of human Deltal (see, eg Genbank Accession No AF003522) into the expression vector pCON ⁇ (Lonza Biologies, Slough, UK) and expressing the resulting constract in CHO cells.
  • a 1622bp extracellular (EC) fragment of human Delta-like ligand 1 (hECDLL-1; see GenBank Accession No AF003522) was gel purified using a Qiagen QIAquickTM Gel Extraction Kit (cat 28706) according to the manufacturer's instractions (Qiagen, Valencia, CA, US). The fragment was then ligated into a pCR Blunt cloning vector (Invitrogen, UK) cut Hindlll - BsiWI, thus eliminating a Hindlll, BsiWI and Apal site.
  • the ligation was transformed into DH5 ⁇ cells, streaked onto LB + Kanamycin (30ug/ml) plates and incubated at 37°C overnight. Colonies were picked from the plates into 3ml LB + Kanamycin ⁇ Ougmi "1 ) and grown up overnight at 37°C. Plasmid DNA was purified from the cultures using a Qiagen Qiaquick Spin Miniprep kit (cat 27106) according to the manufacturer's instractions, then diagnostically digested with Hindlll.
  • a clone was chosen and streaked onto an LB + Kanamycin (30ug/ml) plate with the glycerol stock of modified pCRBlunt-hECDLL-1 and incubated at 37°C overnight. A colony was picked off this plate into 60ml LB + Kanamycin (30ug/ml) and incubated at 37°C overnight.
  • the culture was maxiprepped using a Clontech Nucleobond Maxi Kit (cat K3003-2) according to the manufacturer's instractions (Clontech, PaloAlto, CA, US), and the final DNA pellet was resuspended in 300ul dH 2 O and stored at -20°C.
  • modified pCR Blunt-hECDLL-1 vector was linearised with Hindlll and partially digested with Apal.
  • the 1622bp hECDLL-1 fragment was then gel purified using a Clontech Nucleospin® Extraction Kit (K3051-1) according to the manufacturer's instractions.
  • the DNA was then passed through another Clontech Nucleospin® column and followed the isolation from PCR protocol, concentration of sample was then checked by agarose gel analysis ready for ligation.
  • Plasmid pcon ⁇ (Lonza Biologies, UK) was cut with Hindlll - Apal and the following oligos were ligated in (SEQ ID NO: 1):
  • the ligation was transformed into DH5 ⁇ cells and LB + Amp (lOOug/ml) plates were streaked with 200ul of the transformation and incubated at 37°C overnight. The following day 12 clones were picked into 2 x YT + Ampicillin (lOOugmf 1 ) and grown up at 37°C throughout the day. Plasmid DNA was purified from the cultures using a Qiagen Qiaquick Spin Miniprep kit (cat 27106) and diagnostically digested with Notl. A clone
  • pDev41 (designated "pDev41") was chosen and an LB + Amp (lOOug/ml) plate was streaked with the glycerol stock of pDev41 and incubated at 37°C overnight. The following day a clone was picked from this plate into 60ml LB + Amp (lOOug/ml) and incubated with shaking at 37°C overnight. The clone was maxiprepped using a Clontech Nucleobond Maxi Kit (cat K3003- 2) according to the manufacturer's instractions and stored at -20°C.
  • the pDev41 clone 5 maxiprep was then digested with Apal - EcoRI to generate the IgG4Fc fragment (1624bp).
  • the digest was purified on a 1% agarose gel and the main band was cut out and purified using a Clontech Nucleospin Extraction Kit (K3051-1).
  • the polynucleotide was then cloned into the polylinker region of pEE14.4 (Lonza Biologies, UK) downstream of the strong hCMN promoter enhancer region (hCMN-MIE) and upstream of SN40 polyadenylation signal (encodes the GS gene required for selection in glutamine free media; contains the GS minigene - GS cD ⁇ A which includes the last infron and polylinker adenylation signals of the wild type hamster GS gene) which is under the control of the late SN40 promoter, has the hCMN promoter to drive transcription of the desired gene.
  • 5ug of the maxiprep of pEE14.4 was digested with Hindlll - EcoRI, and the product was gel extracted and treated with alkaline phosphatase.
  • a 3 fragment ligation was set up with pEE14.4 cut Hindlll - EcoRI, ECDLL-1 from modified pCR Blunt (Hindlll - Apal) and the IgG4Fc fragment cut from pDev41 (Apal - EcoRI). This was transformed into DH5 ⁇ cells and LB + Amp (lOOug/ml) plates were streaked with 200ul of the transformation and incubated at 37C overnight. The following day 12 clones were picked into 2 x YT + Amp (lOOug/ml) and minipreps were grown up at 37°C throughout the day.
  • Plasmid DNA was purified from the preps using a Qiagen Qiaquick spin miniprep kit (Cat No 27106), diagnostically digested (with EcoRI and Hindlll) and a clone (clone 8; designated "pDev44") was chosen for maxiprepping.
  • the glycerol stock of pDev44 clone 8 was streaked onto an LB + Amp (lOOugmf 1 ) plate and incubated at 37°C overnight. The following day a colony was picked into 60ml LB + Amp (lOOugmf 1 ) broth and incubated at 37°C overnight.
  • the plasmid DNA was isolated using a Clontech Nucleobond Maxiprep Kit (Cat K3003-2).
  • a Kozak sequence was inserted into the expression construct as follows. Oligonucleotides were kinase treated and annealed to generate the following sequences:
  • pDev44 was digested with Hindlll - BstBI, gel purified and treated with alkaline phosphatase. The digest was ligated with the oligos, transformed into DH5 ⁇ cells by heat -Ill-
  • the resulting DNA preparation was cleaned up using phenol/chloroform/IAA extraction followed by ethanol wash and precipitation.
  • the pellets were resuspended in sterile water and linearisation and quantification was checked by agarose gel electrophoresis and UN spectrophotometry.
  • amino acid sequence of the resulting expressed fusion protein was as follows (SEQ ID NO: 4): MGSRCALALAVLSALLCQVWSSGVFELKLQEFVNKKGLLGNRNCCRGGAGPPPCA CRTFFRVCLKHYQASVSPEPPCTYGSAVTPVLGVDSFSLPDGGGADSAFSNPIRFPFG FTWPGTFSLI ⁇ EALHTDSPDDLATENPERLISRLATQRHLTVGEEWSQDLHSSGRTDL KYSYRFVCDEHYYGEGCSVFCRPRDDAFGHFTCGERGEKVCNPGWKGPYCTEPICL
  • first underlined sequence is the predicted signal peptide (cleaved from the mature protein) and the second underlined sequence is the IgG4 Fc sequence.
  • the protein normally exists as a dimer linked by cysteine disulphide bonds (see eg schematic representation in Figure 6).
  • DLL-1 human Delta 1
  • DlE3cys free cysteine residue
  • a template containing the entire coding sequence for the exfracellular (EC) domain of human DLL-1 (with two silent mutations) was prepared by a PCR cloning strategy from a placental cDNA library made from placental polyA+ RNA (Clontech; cat no 6518- 1 ) and combined with a C-terminal V5HIS tag in a pCDNA3.1 plasmid (Invitrogen, UK)
  • the template was cut Hindlll to Pmel to provide a fragment coding for the EC domain and this was used as a template for PCR using primers as follows:
  • PCR was carried out using Pfu turbo polymerase (Stratagene, La Jolla, CA, US) with cycling conditions as follows: 95C 5min, 95C lmin, 45-69C lmin, 72C lmin for 25 cycles, 72C lOmin.
  • the products at 58C, 62C & 67C were purified from 1% agarose gel in 1 x TAE using a Qiagen gel extraction kit according to the manufacturer's instractions, ligated into pCRIIblunt vector (InVifrogen TOPO-blunt kit) and then transformed into TOP 10 cells (InVifrogen). The resulting clone sequence was verified, and only the original two silent mutations were found to be present in the parental clone.
  • the resulting sequence coding for "DlE3Cys" was excised using Pmel and Hindlll, purified on 1% agarose gel, lx TAE using a Qiagen gel extraction kit and ligated into pCDNA3.1V5HIS (Invitrogen) between the Pmel and Hindlll sites, thereby eliminating the V5HIS sequence.
  • the resulting DNA was transformed into TOP10 cells. The resulting clone sequence was verified at the 3 '-ligation site.
  • the DlE3Cys-coding fragment was excised from the pCDNA3.1 plasmid using Pmel and Hindlll.
  • a pEE14.4 vector plasmid (Lonza Biologies, UK) was then restricted using EcoRI, and the 5 '-overhangs were filled in using Klenow fragment polymerase.
  • the vector DNA was cleaned on a Qiagen PCR purification column, restricted using Hindlll, then treated with Shrimp Alkaline Phosphatase (Roche).
  • the pEE14.4 vector and DlE3cys fragments were purified on 1% agarose gel in 1 x TAE using a Qiagen gel extraction kit prior to ligation (T4 ligase) to give plasmid pEE14.4 DLL ⁇ 4-8cys. The resulting clone sequence was verified.
  • the DlE3Cys coding sequence is as follows (SEQ ID NO: 7):
  • the DNA was prepared for stable cell line fransfection/selection in a Lonza GS system using a Qiagen endofree maxi-prep kit.
  • the pEE14.4 DLL ⁇ 4-8cys plasmid DNA from (i) above was linearised by restriction enzyme digestion with Pvul, and then cleaned up using phenol chloroform isoamyl alcohol (IAA), followed by ethanol precipitation. Plasmid DNA was checked on an agarose gel for linearisation, and spec'd at 260/280nm for quantity and quality of prep. Transfection
  • CHO-Kl cells were seeded into 6 wells at 7.5 x 10 5 cells per well in 3ml media (DMEM 10%) FCS) 24hrs prior to transfection, giving 95% confluency on the day of transfection.
  • Lipofectamine 2000 was used to transfect the cells using 5ug of linearised DNA. The transfection mix was left on the cell sheet for 5 Vz hours before replacing with 3ml semi- selective media (DMEM, 10% dFCS, GS) for overnight incubation.
  • DMEM Dulbecco's Modified Eagle Medium
  • 10%dFCS fetal calf serum
  • GS glutamine synthase
  • 25uM L-MSX methionine sulphoximine
  • T500 flasks were seeded with lx 10 7 cells in 80ml of selective media. After 4 days incubation the media was removed, cell sheet rinsed with DPBS and 150ml of 325 media with GS supplement added to each flask. Flasks were incubated for 7 further days before harvesting. Harvest media was filtered through a 0.65- 0.45um filter to clarify prior to freezing. Frozen harvests were purified by FPLC as follows: Frozen harvest was thawed and filtered. A 17ml Q Sepharose column was equilibrated in 0.1M Tris pH8 buffer, for 10 column volumes.
  • the harvest was loaded onto the column using a PI pump set at 3ml/min, the flowthrough was collected into a separate container (this is a reverse purification - a lot of the BSA contaminant binds to the Q Sepharose FF and our target protein does not and hence remains in the flowthrough).
  • the flowthrough was concenfrated in a TFF rig using a lOkDa cut off filter cartridge, during concentration it was washed 3 x with 0.1M Sodium phosphate pH 7 buffer. The 500ml was concentrated down to 35ml, to a final concentration of 3mg/ml.
  • the amino acid sequence of the resulting expressed DlE3Cys protein was as follows (SEQ ID NO: 8):
  • sequence in italics is the leader peptide
  • the underlined sequence is the DSL domain
  • the bold sequences are the three EGF repeats
  • the terminal Cys residue is shown bold underlined
  • 40 ⁇ g DlE3Cys protein from (ii) above was made up to lOO ⁇ l to include lOOmM sodium phosphate pH 7.0 and 5mM EDTA.
  • 2 volumes of immobilised TCEP tris[2- carboxyethyl]phosphine hydrochloride; Pierce, Rockford, IL, US, Cat No: 77712; previously washed 3 times 1ml lOOmM sodium phosphate pH 7.0) were added and the mixture was incubated for 30 minutes at room temperature, with rotating.
  • the resin was pelleted at room temperature in a microfuge (13,000 revs/min, 5 minutes) and the supernatant was transfened to a clean Eppendorf tube and stored on ice. Protein concenfration was measured by Warburg-Christian method.
  • Dynabeads M-450 Epoxy (Dynal, Cat. no. 140.01; 4.5 ⁇ m average diameter) are supplied by Dynal (Dynal Biotech, Oslo, Norway) as ethanol-washed beads in distilled water at 4 x 10 8 beads/ml. These beads are magnetic polystyrene beads that have a surface epoxy (glycidyl ether) reactive group which does not require further activation.
  • Proteins are adsorbed hydrophobically on initial coupling with covalent coupling of primary amine groups occuning after 24 h. Coupling reactions occur at neutral pH over a 24 h incubation time at a temperature between 4 °C and 37 °C.
  • hDeltal-IgG4Fc fusion protein from Example 15 or DlE3Cys protein from Example 16 above was added ( ⁇ 5 ⁇ g per 10 7 beads typical starting concentration) to beads at a final concenfration of 4 - 8 x 10 8 beads/ml.
  • a blocking protein was added to assist binding orientation as follows: The beads were incubated for 15-30 min at 4 - 37 °C, with shaking. 0.1 % BSA was added as blocking protein (Dynal typically suggest 0.1-0.5%) BSA or HAS for antibody binding) The beads were then left for 16 - 20 h at 4 - 37 °C, depending on stability of protein, with shaking to ensure covalent coupling. Coated beads were washed x 3 with PBS/0.1 % BSA using a magnet. The addition of 0.1% BSA in the wash buffer here ensures complete blocking of the beads after coating. Coated beads were stored at 4 °C.
  • Sulfo-SMCC sulphosuccinimidyl 4-(N-maleimidomethyl) cyclohexane-1-carboxylate; Pierce, Cat No: 22322
  • Pierce, Cat No: 22322 Sulfo-SMCC (sulphosuccinimidyl 4-(N-maleimidomethyl) cyclohexane-1-carboxylate; Pierce, Cat No: 22322) was added to 0.2 mg/ml and the mixture was incubated for 30 minutes at room temperature, with rotating. The beads were washed 3 times with 0.5ml lOOmM sodium phosphate pH 7.0, then made up to 1 x 10 9 /ml in lOOmM sodium phosphate pH 7.
  • Surfactant-free sulphate white polystyrene latex beads (product number 1-5000) were supplied by Interfacial Dynamics Co ⁇ oration (Portland, Oregon, US).
  • the polystyrene microspheres have a mean diameter of 4.9 ⁇ m and were supplied dispersed in distilled de- ionised water at 6.3 x 10 8 beads/ml.
  • the beads are negatively charged and have sulphate groups on the surface - the surface is hydrophobic in nature.
  • CD4+ cell purification Spleens were removed from mice (Balb/c females, 8-10 weeks) and treated with lmg/ml Collagenase D (Boehringer Mannheim) in RPMI medium with no supplements for 40 min. Tissue was passed through a 0.2 ⁇ cell strainer (Falcon) into 20ml RIOF medium (R10F- RPMI 1640 media (Gibco Cat No 22409) plus 2mM L-glutamine, 50 ⁇ g/ml Penicillin, 50 ⁇ g/ml Sfreptomycin, 5 x 10 "5 M ⁇ -mercapto-ethanol in 10% fetal calf serum). The cell suspension was spun (1150 ⁇ m 5min) and the media removed.
  • RIOF medium R10F- RPMI 1640 media (Gibco Cat No 22409) plus 2mM L-glutamine, 50 ⁇ g/ml Penicillin, 50 ⁇ g/ml Sfreptomycin, 5 x 10 "5 M ⁇ -mercap
  • CD4+ cells were purified from the suspensions by positive selection on a Magnetic Associated Cell Sorter (MACS) column (Miltenyi Biotec, Bisley, UK: Cat No 130-042-401) using CD4 (L3T4) beads (Miltenyi Biotec Cat No 130-049-201), according to the manufacturer's directions.
  • ACK lysis buffer 0.15M NH 4 C1, l.OM KHC0 , O.lmM Na 2 EDTA in double distilled water
  • spleen to lyse red blood cells.
  • CD4+ cells were purified from the suspensions by positive selection on a Magnetic Associated Cell Sorter (MACS) column (Miltenyi Biotec, Bisley, UK: Cat No 130-042-401) using CD4 (L3T4) beads (Miltenyi Biotec Cat No 130-049-201), according to the manufacturer's directions.
  • MCS Magnetic Associated Cell Sorter
  • Antibody Coating The following protocol was used for coating 96 well flat-bottomed plates with antibodies.
  • the plates were coated with Dulbecco's Phosphate Buffered Saline (DPBS) plus l ⁇ g/mL anti-hamster IgG antibody (Pharmingen, San Diego, US: Cat No 554007). lOO ⁇ L of coating mixture was used per well. Plates were incubated overnight at 4°C then washed with DPBS. Each well then received lOO ⁇ L DPBS plus 0.1-l ⁇ g/mL anti-CD3 (Pharmingen Cat No 553058, Clone No 145-2C11).
  • DPBS Dulbecco's Phosphate Buffered Saline
  • l ⁇ g/mL anti-hamster IgG antibody Pharmingen, San Diego, US: Cat No 554007
  • the plates were incubated for 2-3 hours at 37°C then washed again with DPBS before cells (prepared as in Example 20) were added.
  • CD4+ cells were cultured in 96 well, flat-bottomed plates pre-coated according to Example 21. Cells were resuspended following counting at 4x10 /mL in RIOF medium and 50 ⁇ L suspension added per well. RIOF medium plus 8 ⁇ g/mL CD28 antibody (Pharmingen, Cat No 553294, Clone No 37.51) was added at 50 ⁇ L per well. Beads coated with Notch ligand were added in appropriate volumes to give final ratios of 0.1-20:1 beads:cell. R10F medium was added to give a final volume of 200 ⁇ L per well (2xl0 5 cells/well, anti-CD28 final concentration 2 ⁇ g/mL). The plates were then incubated at 37°C for 72 hours.
  • MyOne Sfreptavidin beads (l ⁇ m, Dynal 650.01) and CELLection Biotin Binder (4.5 ⁇ m, Dynal 115.21) were coated with anti-hIgG4-Biotin antibodies based on the binding capacity recommended by the supplier.
  • CELLection beads The beads were then washed and further incubated with either 100 ⁇ g of human DeltalEC domain-hIgG4 fusion protein or 100 ⁇ g of hIgG4 (Sigma) for 2 hours at room temperature. After washing, the MyOne beads and the CELLection beads were resuspended in 500 ⁇ l of RPMI/BSA 0.1% and stored at 4°C.
  • PBMC Human peripheral blood mononuclear cells
  • Human CD4+ T cells were isolated by positive selection using anti-CD4 microbeads from Miltenyi Biotech according to the manufacturer's instractions. The CD4+ T cells were incubated in triplicates in a 96-well-plate (flat bottom) at 10 5 CD4/well/200 ⁇ l in RPMI medium containing 10%> FCS, glutamine, penicillin, streptomycin and ⁇ 2 -mercaptoethanol.
  • Cytokine production was induced by stimulating the cells with anti-CD3/CD28 T cell expander beads from Dynal at a 1:1 ratio (bead/cell). 10, 5, 2.5, 1.25, 0.62 ⁇ l of beads coated with human Deltal EC domain-hIgG4 fusion protein (prepared as described above) or control beads were added in some of the wells. The supernatants were removed after 3 days of incubation at 37°C/ 5%oCO 2 /humidified atmosphere and cytokine production was evaluated by ELISA using Pharmingen kits OptEIA Set human IL10 (catalog No. 555157) and OptEIA Set human IL-5 (catalog No. 555202) for IL-10, IL-5 respectively and a human IL-2 DuoSet from R&D Systems (catalog. No DY202) for IL-2 according to the manufacturer's instructions.
  • OptEIA Set human IL10 catalog No. 555157
  • OptEIA Set human IL-5
  • PBMC peripheral blood mononuclear cells
  • Human CD 14+ monocytes and CD4+ T cells were isolated from PBMC from donor A and B respectively by positive selection using anti-CD 14 and anti-CD4 microbeads from Miltenyi
  • the CD 14+ cells were differentiated into dendritic cells (DC) by incubation for 6 days in medium [RPMI / 10%FCS / glutamine / B2-mercaptoethanol / antibiotics] in the presence of hGM-CSF 50 ng/ml and hIL-4 50 ng/ml (both from Peprotech). Dendritic cell maturation was induced by addition into the culture of LPS l ⁇ g/ml (Sigma L-2654) for the last 24 hours.
  • Matured-DC were treated for 1 hour with 50 ⁇ g/ml Mitomycin C (Sigma) in RPMI and washed 4 times. These cells were then plated at 4xl0 4 , lxlO 4 , 2.5xl0 3 , 6.25xl0 2 cells/well in triplicates in a 96-well-plate in RPMI medium containing 10% FCS, glutamine, penicillin, streptomycin and ⁇ 2 -mercaptoethanol. 2x10 5 Allogenic CD4+ T cells (donor B) were added into each well given a final volume of 200 ⁇ l/well.
  • Mitomycin C Sigma

Abstract

L'invention concerne une méthode de modulation thérapeutique de la signalisation Notch. Cette méthode consiste à administrer une construction comprenant une multiplicité de modulateurs de la signalisation Notch, fixés, liés ou immobilisés.
PCT/GB2003/001525 2002-04-05 2003-04-04 Traitement medical WO2003087159A2 (fr)

Priority Applications (25)

Application Number Priority Date Filing Date Title
AU2003226537A AU2003226537A1 (en) 2002-04-05 2003-04-04 Modulators of the notch signalling pathway and uses thereof in medical treatment
EP03746366A EP1492816A2 (fr) 2002-04-05 2003-04-04 Modulateurs de la voie de signalation notch et utilisations associees dans le traitement medical
JP2003584114A JP2006506322A (ja) 2002-04-05 2003-04-04 内科療法
JP2005506075A JP2006513260A (ja) 2002-08-03 2003-08-01 Notchシグナル伝達経路調節因子の複合体およびその薬物治療への使用
AU2003255735A AU2003255735A1 (en) 2002-08-03 2003-08-01 Conjugate of notch signalling pathway modulators and their use in medical treatment
EP03766445A EP1525221A1 (fr) 2002-08-03 2003-08-01 Conjuges de modulateurs de la voie de signalisation notch et leur utilisation dans les traitements medicaux
PCT/GB2003/003285 WO2004013179A1 (fr) 2002-08-03 2003-08-01 Conjuges de modulateurs de la voie de signalisation notch et leur utilisation dans les traitements medicaux
AU2003267563A AU2003267563A1 (en) 2002-09-10 2003-09-09 Pharmaceutical composition and medical treatments comprising notch ligand proteins
CA002497226A CA2497226A1 (fr) 2002-09-10 2003-09-09 Compositions pharmaceutiques et traitements medicaux comprenant des proteines a ligand notch
JP2004571915A JP2006515177A (ja) 2002-09-10 2003-09-09 Notchリガンドタンパク質を含む医薬組成物及び医学的処置
EP03748255A EP1537145A1 (fr) 2002-09-10 2003-09-09 Composition pharmaceutique et traitements medicaux comprenant des proteines a ligand notch
PCT/GB2003/003908 WO2004024764A1 (fr) 2002-09-10 2003-09-09 Composition pharmaceutique et traitements medicaux comprenant des proteines a ligand notch
PCT/GB2004/000046 WO2004060262A2 (fr) 2003-01-07 2004-01-07 Traitement medical
EP04704657A EP1585543A1 (fr) 2003-01-23 2004-01-23 Traitement de maladies autoimmunes au moyen d'un activateur de la voie de signalisation notch
PCT/GB2004/000263 WO2004064863A1 (fr) 2003-01-23 2004-01-23 Traitement de maladies autoimmunes au moyen d'un activateur de la voie de signalisation notch
JP2006500232A JP2006517533A (ja) 2003-01-23 2004-01-23 Notchシグナル伝達経路のアクチベーターを用いる自己免疫疾患の治療
AT04722319T ATE474593T1 (de) 2003-03-21 2004-03-22 Behandlung von allergischen erkrankungen unter verwendung eines modulators des notch signaling pathway
PCT/GB2004/001252 WO2004082710A1 (fr) 2003-03-21 2004-03-22 Traitement de maladies allergiques utilisant un modulateur de la voie de signalisation notch
EP04722319A EP1646400B1 (fr) 2003-03-21 2004-03-22 Traitement de maladies allergiques utilisant un modulateur de la voie de signalisation notch
US10/958,784 US20050220886A1 (en) 2002-04-05 2004-10-05 Modulators of the Notch signalling pathway and uses thereof in medical treatment
US11/050,346 US20060002924A1 (en) 2002-08-03 2005-02-03 Conjugate of notch signalling pathway modulators and their use in medical treatment
US11/078,735 US20050261477A1 (en) 2002-09-10 2005-03-10 Pharmaceutical compositions and medical treatments comprising notch ligand proteins
US11/188,417 US20060204508A1 (en) 2003-01-23 2005-07-25 Treatment of autoimmune diseases using an activator for the notch signalling pathway
US11/231,494 US20060205823A1 (en) 2003-03-21 2005-09-21 Treatment of allergic diseases using a modulator of the Notch signaling pathway
US12/766,738 US20100303867A1 (en) 2003-03-21 2010-04-23 a composition comprising a notch ligand and an allergenb or allergen bystander antigen

Applications Claiming Priority (18)

Application Number Priority Date Filing Date Title
GB0207929.1 2002-04-05
GB0207930A GB0207930D0 (en) 2002-04-05 2002-04-05 A method
GB0207930.9 2002-04-05
GB0207929A GB0207929D0 (en) 2002-04-05 2002-04-05 A method
GB0212282.8 2002-05-28
GB0212282A GB0212282D0 (en) 2002-05-28 2002-05-28 A method
GB0212283.6 2002-05-28
GB0212283A GB0212283D0 (en) 2002-05-28 2002-05-28 A method
PCT/GB2002/003397 WO2003012441A1 (fr) 2001-07-25 2002-07-25 Procede de detection de modulateurs de signalisation de notch
GBPCT/GB02/03397 2002-07-25
PCT/GB2002/003426 WO2003011317A1 (fr) 2001-07-25 2002-07-25 Modulateurs de signalisation de notch utilises en immunotherapie
GBPCT/GB02/03426 2002-07-25
GB0220912.0 2002-09-10
GB0220913.8 2002-09-10
GB0220912A GB0220912D0 (en) 2002-09-10 2002-09-10 Medical treatment
GB0220913A GB0220913D0 (en) 2002-09-10 2002-09-10 Medical treatment
GB0300234A GB0300234D0 (en) 2003-01-07 2003-01-07 Medical treatment
GB0300234.2 2003-01-07

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US10/958,784 Continuation-In-Part US20050220886A1 (en) 2002-04-05 2004-10-05 Modulators of the Notch signalling pathway and uses thereof in medical treatment

Publications (3)

Publication Number Publication Date
WO2003087159A2 true WO2003087159A2 (fr) 2003-10-23
WO2003087159A3 WO2003087159A3 (fr) 2004-02-05
WO2003087159A8 WO2003087159A8 (fr) 2005-05-12

Family

ID=34109266

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2003/001525 WO2003087159A2 (fr) 2002-04-05 2003-04-04 Traitement medical

Country Status (4)

Country Link
EP (1) EP1492816A2 (fr)
JP (1) JP2006506322A (fr)
AU (1) AU2003226537A1 (fr)
WO (1) WO2003087159A2 (fr)

Cited By (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004022730A1 (fr) * 2002-09-05 2004-03-18 Lorantis Limited Immunotherapie utilisant des modulateurs de signalisation notch
WO2004024764A1 (fr) * 2002-09-10 2004-03-25 Lorantis Limited Composition pharmaceutique et traitements medicaux comprenant des proteines a ligand notch
WO2004060262A2 (fr) * 2003-01-07 2004-07-22 Lorantis Limited Traitement medical
WO2004062686A2 (fr) * 2003-01-09 2004-07-29 Lorantis Limited Traitement medical
WO2004064863A1 (fr) * 2003-01-23 2004-08-05 Lorantis Limited Traitement de maladies autoimmunes au moyen d'un activateur de la voie de signalisation notch
WO2004083372A2 (fr) * 2003-03-21 2004-09-30 Lorantis Limited Traitement medical
WO2004087195A2 (fr) * 2003-04-01 2004-10-14 Lorantis Limited Dosages et traitements medicaux
EP1709150A2 (fr) * 2003-11-26 2006-10-11 Health Research, Inc. Utilisation d'agents qui interferent avec la voie de transmission du signal notch pour le traitement des troubles plasmocytaires
WO2007027226A2 (fr) * 2005-04-28 2007-03-08 Board Of Regents, The University Of Texas System Systemes et methodes pour la production de cellules differenciees
US7449303B2 (en) 2003-05-02 2008-11-11 Health Research, Inc. Use of JAG2 expression in diagnosis of plasma cell disorders
EP2193143A2 (fr) * 2007-08-23 2010-06-09 The Trustees of Columbia University in the City of New York Compositions protéines de fusion notch humanisées et procédés de traitement
US7906116B2 (en) 2005-09-01 2011-03-15 Parkash Gill Methods for using and identifying modulators of Delta-like 4
US8993725B2 (en) 2004-04-29 2015-03-31 The Trustees Of Columbia University In The City Of New York Notch-based fusion proteins and uses thereof
US9475855B2 (en) 2008-08-22 2016-10-25 The Trustees Of Columbia University In The City Of New York Human Notch3 based fusion proteins as decoy inhibitors of Notch3 signaling
US9738708B2 (en) 2011-10-04 2017-08-22 The Trustees Of Columbia University In The City Of New York Human Notch1 decoys
WO2019157597A1 (fr) * 2018-02-14 2019-08-22 Sunnybrook Research Institute Procédé de génération de cellules de la lignée des lymphocytes t
WO2020023807A1 (fr) * 2018-07-26 2020-01-30 The Regents Of The University Of California Traitement d'une occlusion vasculaire par activation de la voie de signalisation notch

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020047099A1 (fr) * 2018-08-28 2020-03-05 Fred Hutchinson Cancer Research Center Procédés et compositions pour thérapie adoptive par lymphocytes t comportant une signalisation notch induite

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998020142A1 (fr) * 1996-11-07 1998-05-14 Lorantis Limited Notch
EP0913404A1 (fr) * 1996-07-16 1999-05-06 Asahi Kasei Kogyo Kabushiki Kaisha Inhibiteur de differenciation
WO2000025809A1 (fr) * 1998-10-30 2000-05-11 Smith & Nephew Plc Compositions contenant des agents actifs sur les recepteurs notch
WO2003041735A2 (fr) * 2001-11-14 2003-05-22 Lorantis Limited Traitement medical

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0913404A1 (fr) * 1996-07-16 1999-05-06 Asahi Kasei Kogyo Kabushiki Kaisha Inhibiteur de differenciation
WO1998020142A1 (fr) * 1996-11-07 1998-05-14 Lorantis Limited Notch
WO2000025809A1 (fr) * 1998-10-30 2000-05-11 Smith & Nephew Plc Compositions contenant des agents actifs sur les recepteurs notch
WO2003041735A2 (fr) * 2001-11-14 2003-05-22 Lorantis Limited Traitement medical

Non-Patent Citations (7)

* Cited by examiner, † Cited by third party
Title
GRAY G E ET AL: "HUMAN LIGANDS OF THE NOTCH RECEPTOR" AMERICAN JOURNAL OF PATHOLOGY, PHILADELPHIA, PA, US, vol. 154, no. 3, March 1999 (1999-03), pages 785-794, XP000960906 ISSN: 0002-9440 *
HICKS C ET AL: "A SECRETED DELTA1-FC FUSION PROTEIN FUNCTIONS BOTH AS AN ACTIVATOR AND INHIBITOR OF NOTCH1 SIGNALING" JOURNAL OF NEUROSCIENCE RESEARCH, WILEY-LISS, US, vol. 68, no. 6, 15 June 2002 (2002-06-15), pages 655-667, XP009013890 ISSN: 0360-4012 *
JANG M-S ET AL: "NOTCH SIGNALING AS A TARGET IN MULTIMODALITY CANCER THERAPY" CURRENT OPINION IN MOLECULAR THERAPEUTICS, CURRENT DRUGS, LONDON,, GB, vol. 2, no. 1, February 2000 (2000-02), pages 55-65, XP009013844 ISSN: 1464-8431 *
OSBORNE B ET AL: "Notch and the immune system" IMMUNITY, CELL PRESS, US, vol. 11, no. 6, December 1999 (1999-12), pages 653-663, XP002217474 ISSN: 1074-7613 *
See also references of EP1492816A2 *
SUN X ET AL: "SECRETED FORMS OF DELTA AND SERRATE DEFINE ANTAGONISTS OF NOTCH SIGNALING IN DROSOPHILA" DEVELOPMENT, COMPANY OF BIOLOGISTS, CAMBRIDGE,, GB, vol. 124, no. 17, 1997, pages 3439-3448, XP002925005 ISSN: 0950-1991 *
ZLOBIN A ET AL: "TOWARD THE RATIONAL DESIGN OF CELL FATE MODIFIERS: NOTCH SIGNALING AS A TARGET FOR NOVEL BIOPHARMACEUTICALS" CURRENT PHARMACEUTICAL BIOTECHNOLOGY, BENTHAM SCIENCE PUBLISHERS, BOCA RATON,FL, US, vol. 1, no. 1, July 2000 (2000-07), pages 83-106, XP008004456 ISSN: 1389-2010 *

Cited By (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004022730A1 (fr) * 2002-09-05 2004-03-18 Lorantis Limited Immunotherapie utilisant des modulateurs de signalisation notch
WO2004024764A1 (fr) * 2002-09-10 2004-03-25 Lorantis Limited Composition pharmaceutique et traitements medicaux comprenant des proteines a ligand notch
WO2004060262A2 (fr) * 2003-01-07 2004-07-22 Lorantis Limited Traitement medical
WO2004060262A3 (fr) * 2003-01-07 2004-12-09 Lorantis Ltd Traitement medical
WO2004062686A2 (fr) * 2003-01-09 2004-07-29 Lorantis Limited Traitement medical
WO2004062686A3 (fr) * 2003-01-09 2004-11-18 Lorantis Ltd Traitement medical
WO2004064863A1 (fr) * 2003-01-23 2004-08-05 Lorantis Limited Traitement de maladies autoimmunes au moyen d'un activateur de la voie de signalisation notch
WO2004083372A2 (fr) * 2003-03-21 2004-09-30 Lorantis Limited Traitement medical
WO2004083372A3 (fr) * 2003-03-21 2004-11-04 Lorantis Ltd Traitement medical
WO2004087195A2 (fr) * 2003-04-01 2004-10-14 Lorantis Limited Dosages et traitements medicaux
WO2004087195A3 (fr) * 2003-04-01 2005-02-24 Lorantis Ltd Dosages et traitements medicaux
US7449303B2 (en) 2003-05-02 2008-11-11 Health Research, Inc. Use of JAG2 expression in diagnosis of plasma cell disorders
EP1709150A4 (fr) * 2003-11-26 2007-11-21 Health Research Inc Utilisation d'agents qui interferent avec la voie de transmission du signal notch pour le traitement des troubles plasmocytaires
EP1709150A2 (fr) * 2003-11-26 2006-10-11 Health Research, Inc. Utilisation d'agents qui interferent avec la voie de transmission du signal notch pour le traitement des troubles plasmocytaires
US8993725B2 (en) 2004-04-29 2015-03-31 The Trustees Of Columbia University In The City Of New York Notch-based fusion proteins and uses thereof
WO2007027226A2 (fr) * 2005-04-28 2007-03-08 Board Of Regents, The University Of Texas System Systemes et methodes pour la production de cellules differenciees
WO2007027226A3 (fr) * 2005-04-28 2007-04-26 Univ Texas Systemes et methodes pour la production de cellules differenciees
US7906116B2 (en) 2005-09-01 2011-03-15 Parkash Gill Methods for using and identifying modulators of Delta-like 4
US9127085B2 (en) 2007-08-23 2015-09-08 The Trustees Of Columbia University In The City Of New York Compositions of humanized notch fusion proteins and methods of treatment
EP2193143A4 (fr) * 2007-08-23 2012-01-25 Univ Columbia Compositions protéines de fusion notch humanisées et procédés de traitement
CN101883786A (zh) * 2007-08-23 2010-11-10 纽约哥伦比亚大学理事会 人源化的notch融合蛋白组合物及治疗方法
EP2193143A2 (fr) * 2007-08-23 2010-06-09 The Trustees of Columbia University in the City of New York Compositions protéines de fusion notch humanisées et procédés de traitement
US9475855B2 (en) 2008-08-22 2016-10-25 The Trustees Of Columbia University In The City Of New York Human Notch3 based fusion proteins as decoy inhibitors of Notch3 signaling
US9738708B2 (en) 2011-10-04 2017-08-22 The Trustees Of Columbia University In The City Of New York Human Notch1 decoys
US10227399B2 (en) 2011-10-04 2019-03-12 The Trustees Of Columbia University In The City Of New York Human Notch1 decoys
WO2019157597A1 (fr) * 2018-02-14 2019-08-22 Sunnybrook Research Institute Procédé de génération de cellules de la lignée des lymphocytes t
CN112041431A (zh) * 2018-02-14 2020-12-04 桑尼布鲁克研究所 产生t细胞谱系细胞的方法
WO2020023807A1 (fr) * 2018-07-26 2020-01-30 The Regents Of The University Of California Traitement d'une occlusion vasculaire par activation de la voie de signalisation notch
CN112770754A (zh) * 2018-07-26 2021-05-07 加利福尼亚大学董事会 通过激活Notch信号传导治疗血管阻塞

Also Published As

Publication number Publication date
WO2003087159A3 (fr) 2004-02-05
AU2003226537A8 (en) 2003-10-27
WO2003087159A8 (fr) 2005-05-12
AU2003226537A1 (en) 2003-10-27
EP1492816A2 (fr) 2005-01-05
JP2006506322A (ja) 2006-02-23

Similar Documents

Publication Publication Date Title
EP1492816A2 (fr) Modulateurs de la voie de signalation notch et utilisations associees dans le traitement medical
US20050025751A1 (en) Modulators of Notch signalling for use in immunotherapy
US20060204508A1 (en) Treatment of autoimmune diseases using an activator for the notch signalling pathway
EP1448599A2 (fr) Inhibiteurs de la voie de signalation de notch pour l'utilisation dans le traitement du cancer
US20100303867A1 (en) a composition comprising a notch ligand and an allergenb or allergen bystander antigen
JP2013503205A (ja) 移植拒絶反応を阻害するための方法および組成物
JP4530129B2 (ja) スーパー抗原
EP1537145A1 (fr) Composition pharmaceutique et traitements medicaux comprenant des proteines a ligand notch
US20060140943A1 (en) Immunotherapy using modulators of notch signalling
US20060003927A1 (en) Modulation of immune function
US20050220886A1 (en) Modulators of the Notch signalling pathway and uses thereof in medical treatment
JP2006507240A (ja) 免疫機能の調節
US20060128619A1 (en) Therapeutic use of modulators of notch
WO2004013179A1 (fr) Conjuges de modulateurs de la voie de signalisation notch et leur utilisation dans les traitements medicaux
WO2004060262A2 (fr) Traitement medical
WO2004083372A2 (fr) Traitement medical
WO2004087195A2 (fr) Dosages et traitements medicaux
AU2002355670A1 (en) Modulators of notch signalling for use in immunotherapy

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ OM PH PL PT RO RU SC SD SE SG SK SL TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 10958784

Country of ref document: US

Ref document number: 2003584114

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 2003746366

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 2003746366

Country of ref document: EP

CFP Corrected version of a pamphlet front page
CR1 Correction of entry in section i

Free format text: IN PCT GAZETTE 43/2003 UNDER (30) REPLACE "PCT/GB2/03397 AND PCT/GB2/03426" BY "PCT/GB2002/003397 AND PCT/GB2002/003426"