WO2005097098A2 - Use of ppr delta agonists for treating demyelinating diseases - Google Patents

Use of ppr delta agonists for treating demyelinating diseases Download PDF

Info

Publication number
WO2005097098A2
WO2005097098A2 PCT/US2005/010371 US2005010371W WO2005097098A2 WO 2005097098 A2 WO2005097098 A2 WO 2005097098A2 US 2005010371 W US2005010371 W US 2005010371W WO 2005097098 A2 WO2005097098 A2 WO 2005097098A2
Authority
WO
WIPO (PCT)
Prior art keywords
cells
compound
ppar
disease
invitrogen
Prior art date
Application number
PCT/US2005/010371
Other languages
French (fr)
Other versions
WO2005097098A3 (en
Inventor
Karen Chandross
Jean Merrill
Anne Minnich
Lan Lee
Olga Khorkova
Yun Liu
Original Assignee
Aventis Pharmaceuticals Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Aventis Pharmaceuticals Inc. filed Critical Aventis Pharmaceuticals Inc.
Priority to AU2005231358A priority Critical patent/AU2005231358A1/en
Priority to CA002561159A priority patent/CA2561159A1/en
Priority to BRPI0509540-9A priority patent/BRPI0509540A/en
Priority to EP05729215A priority patent/EP1737440A2/en
Priority to JP2007506442A priority patent/JP2007530703A/en
Priority to MXPA06011218A priority patent/MXPA06011218A/en
Publication of WO2005097098A2 publication Critical patent/WO2005097098A2/en
Publication of WO2005097098A3 publication Critical patent/WO2005097098A3/en
Priority to IL178165A priority patent/IL178165A0/en
Priority to US11/535,240 priority patent/US20070149580A1/en
Priority to NO20064985A priority patent/NO20064985L/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/425Thiazoles
    • A61K31/4261,3-Thiazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/192Carboxylic acids, e.g. valproic acid having aromatic groups, e.g. sulindac, 2-aryl-propionic acids, ethacrynic acid 
    • 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
    • A61P25/28Drugs 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
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • This invention relates to the use of PPAR delta agonists for the treatment of multiple sclerosis (MS) and other demyelinating diseases.
  • This invention also relates to the use of certain compounds that are selective PPAR delta agonists for the treatment of MS and other demyelinating diseases.
  • PPARs peroxisome proliferator-activated receptors
  • Three closely related isoforms have been identified and cloned and are commonly known as PPAR alpha, PPAR gamma and PPAR delta.
  • Each receptor subtype has a signature DNA binding domain (DBD) and a ligand-binding domain (LBD), both being necessary for ligand activated gene expression.
  • PPARs bind as heterodimers with a retinoid X receptor. See J. Berger and D. E. Miller, Ann. Rev. Med., 2002, 53, 409-435.
  • PPAR delta also known as PPAR beta
  • PPAR beta is expressed in a broad range of mammalian tissue, but little information regarding its biological functions or the full array of genes regulated by the receptor have been elucidated.
  • agonists may be useful to treat conditions such as dyslipedemia and certain dermatological conditions, while antagonists may be useful to treat osteoporosis or colorectal cancer (D. Sternbach, in Annual Reports in Medicinal Chemistry, Volume 38, A. M. Doherty, ed., Elsevier Academic Press, 2003 pp. 71-80).
  • PPAR delta appears to be significantly expressed in the CNS; however much of its function there still remains undiscovered. Of singular interest however, is the discovery that PPAR delta was expressed in rodent oligodendrocytes, the major lipid producing cells of the CNS (J. Granneman, et al., J. Neurosci. Res., 1998, 51, 563-573).
  • a PPAR delta selective agonist was found to significantly increase oligodendroglial myelin gene expression and myelin sheath diameter in mouse cultures (I. Saluja et al., Glia, 2001, 33, 194-204). Demyelinating conditions are manifested in loss of myelin— the multiple dense layers of lipids and protein which cover many nerve fibers. These layers are provided by oligodendroglia in the central nervous system (CNS), and Schwann cells in the peripheral nervous system (PNS).
  • CNS central nervous system
  • PNS peripheral nervous system
  • oligodendrocytes the myelin forming cells in the CNS, are destroyed and axons are damaged, resulting in severely impaired neuronal activity and functional deficits, including palegia.
  • demyelination may be irreversible; it is usually accompanied or followed by axonal degeneration, and often by cellular degeneration. Demyelination can occur as a result of neuronal damage or damage to the myelin itself-whether due to aberrant immune responses, local injury, ischemia, metabolic disorders, toxic agents, or viral infections (Prineas and McDonald, Demyelinating Diseases. In Greenfield's Neuropathology, ⁇ .sup.th ed.
  • oligodendrocyte progenitor cells are present throughout areas of demyelination, suggesting the possibility of self -repair if these progenitor cells can be induced to undergo differentiation to mature oligodendrocytes.
  • Central demyelination occurs in several conditions, often of uncertain etiology, that have come to be known as the primary demyelinating diseases. Of these, multiple sclerosis is the most prevalent. Other primary demyelinating diseases include adrenoleukodystrophy (ALD), adrenomyeloneuropathy, AIDS-vacuolar myelopathy, HTLV- associated myelopathy, Leber's hereditary optic atrophy, progressive multifocal leukoencephalopathy (PML), subacute sclerosing panencephalitis, and tropical spastic paraparesis.
  • ALD adrenoleukodystrophy
  • adrenomyeloneuropathy AIDS-vacuolar myelopathy
  • HTLV- associated myelopathy HTLV- associated myelopathy
  • Leber's hereditary optic atrophy progressive multifocal leukoencephalopathy (PML), subacute sclerosing panencephalitis,
  • MS is a chronic, devastating, neurological disease that affects mostly young adults.
  • the pathogenesis of MS is a complex process that leads to destruction of myelin and oligodendroglia, as well as axonal damage, in the brain and spinal cord (Prineas and
  • MS may follow a relapsing-remitting, or it may take a chronically progressive course with increasing physical disability (Gold et al., Mol. Med. Today, 6:88-91, 2000).
  • the symptoms of MS include lack of co-ordination, paresthesias, speech and visual disturbances, and weakness.
  • Corticosteroids oral prednisone at 60-100 mg/day, tapered over 2-3 weeks, or intravenous methylprednisolone at 500-1000 mg/day, for 3-5 days
  • MS oral prednisone
  • intravenous methylprednisolone at 500-1000 mg/day, for 3-5 days
  • Long-term corticosteroid treatment is rarely justified, and can cause numerous medical complications, including osteoporosis, ulcers, and diabetes (Beers and Berkow, eds., The Merck Manual of Diagnosis and Therapy, 17.sup.th ed.
  • Immunosuppressive drugs azathioprine, cladribine, cyclophosphamide, and methotrexate
  • azathioprine, cladribine, cyclophosphamide, and methotrexate are used for more severe progressive forms. However, they are not uniformly beneficial, and have significant toxic side effects.
  • Several drugs e.g., baclofen at 30-60 mg/day in divided doses may reduce spasticity by inhibiting the spinal cord reflexes.
  • ALD Alzheimer's disease
  • Symptoms of ALD may include cortical blindness, corticospinal tract dysfunction, mental deterioration, and spasticity.
  • Therapy to control the course of ALD may include bone marrow transplantation and dietary treatment (DiBiase et al., Ann. 1st. Super Sanita, 35: 185-92, 1999), but inexorable neurological deterioration invariably occurs, ultimately leading to death [Krivit et al., Curr. Opin.
  • the present invention entails the use of a small molecule-activated regenerative approach to significantly augment current immunomodulatory therapies for the treatment of demyelinating disorders.
  • Compounds that are known to be selective PPAR delta are known in the art, in particular, compound of formula (1) generally known as GW 501516 described in WO 01/00603.
  • PPAR delta Peroxisome Proliferator Activated Receptor Delta
  • the disease conditions that can be treated in accordance with the practice of this invention include but not limited to multiple sclerosis, Charcot-Marie-Tooth disease, Pelizaeus-Merzbacher disease, encephalomyelitis, neuromyelitis optica, adrenoleukodystrophy, Guillian-Barrte syndrome and disorders in which myelin forming glial cells are damaged including spinal cord injuries, neuropathies and nerve injury.
  • the diseases as disclosed herein can be treated by administering to a patient in need of such treatment a therapeutically effective amount of a PPAR delta agonist.
  • the present invention is also directed to the use of compounds of formula (I) and formula (II) for the treatment of demyelinating diseases, and in particular multiple sclerosis.
  • the present invention also comprises a method of treating multiple sclerosis in patients by administering a combination of a compound of formula (1) or formula (2) or pharmaceutically acceptable salt thereof, with another compound known to be effective for the treatment of multiple sclerosis in therapeutically effective amounts.
  • Compounds that are currently used to treat the disease are the disease-modifying agents such as the interferons (interferon beta 1-a, beta 1-b and alpha 2), glatiramer acetate or corticosteroids such as methylprednisolone and prednisone.
  • chemotherapeutic agents such as methotrexate, azathioprine, cladribine cyclophosphamide and cyclosporine.
  • the expression "pharmaceutically acceptable carrier” means a non- toxic solvent, dispersant, excipient, adjuvant, or other material which is mixed with the compound of the present invention in order to permit the formation of a pharmaceutical composition, i.e., a dosage form capable of administration to the patient.
  • a pharmaceutical composition i.e., a dosage form capable of administration to the patient.
  • a pharmaceutically acceptable oil typically used for parenteral administration.
  • pharmaceutically acceptable salts as used herein means that the salts of the compounds of the present invention can be used in medicinal preparations. Other salts may, however, be useful in the preparation of the compounds according to the invention or of their pharmaceutically acceptable salts.
  • Suitable pharmaceutically acceptable salts of the compounds of this invention include acid addition salts which may, for example, be formed by mixing a solution of the compound according to the invention with a solution of a pharmaceutically acceptable acid such as hydrochloric acid, hydrobromic acid, sulfuric acid, methanesulfonic acid, 2-hydroxyethanesulfonic acid, p-toluenesulfonic acid, fumaric acid, maleic acid, hydroxymaleic acid, malic acid, ascorbic acid, succinic acid, glutaric acid, acetic acid, salicylic acid, cinnamic acid, 2-phenoxybenzoic acid, hydroxybenzoic acid, phenylacetic acid, benzoic acid, oxalic acid, citric acid, tartaric acid, glycolic acid, lactic acid, pyruvic acid, malonic acid, carbonic acid or phosphoric acid.
  • a pharmaceutically acceptable acid such as hydrochloric acid, hydrobromic acid, sulfuric acid, methanesulfonic
  • the acid metal salts such as sodium monohydrogen orthophosphate and potassium hydrogen sulfate can also be formed.
  • the salts so formed may present either as mono- or di- acid salts and can exist either as hydrated or can be substantially anhydrous.
  • suitable pharmaceutically acceptable salts thereof may include alkali metal salts, e.g. sodium or potassium salts; alkaline earth metal salts, e.g. calcium or magnesium salts; and salts formed with suitable organic ligands, e.g. quaternary ammonium salts.
  • suitable pharmaceutically acceptable salts thereof may include alkali metal salts, e.g. sodium or potassium salts; alkaline earth metal salts, e.g. calcium or magnesium salts; and salts formed with suitable organic ligands, e.g. quaternary ammonium salts.
  • the term "therapeutically effective amount” as used herein means an amount of the compound, which is effective in treating the named disorder or condition.
  • the expression "pharmaceutically acceptable carrier” means a non- toxic solvent, dispersant, excipient, adjuvant, or other material which is mixed with the compound of the present invention in order to permit the formation of a pharmaceutical composition, i.e., a dosage form capable of administration to the patient.
  • a pharmaceutical composition i.e., a dosage form capable of administration to the patient.
  • a pharmaceutically acceptable oil typically used for parenteral administration.
  • the invention also provides pharmaceutical compositions comprising one or more of the compounds according to this invention in association with a pharmaceutically acceptable carrier.
  • compositions are in unit dosage forms such as tablets, pills, capsules, powders, granules, sterile parenteral solutions or suspensions, metered aerosol or liquid sprays, drops, ampoules, auto-injector devices or suppositories; for oral, parenteral, intranasal, sublingual or rectal administration, or for administration by inhalation or insufflation.
  • the compositions may be presented in a form suitable for once-weekly or once- monthly administration; for example, an insoluble salt of the active compound, such as the decanoate salt, may be adapted to provide a depot preparation for intramuscular injection.
  • An erodible polymer containing the active ingredient may be envisaged.
  • the principal active ingredient is mixed with a pharmaceutical carrier, e.g. conventional tableting ingredients such as corn starch, lactose, sucrose, sorbitol, talc, stearic acid, magnesium stearate, dicalcium phosphate or gums, and other pharmaceutical diluents, e.g. water, to form a solid preformulation composition containing a homogeneous mixture of a compound of the present invention, or a pharmaceutically acceptable salt thereof.
  • a pharmaceutical carrier e.g. conventional tableting ingredients such as corn starch, lactose, sucrose, sorbitol, talc, stearic acid, magnesium stearate, dicalcium phosphate or gums, and other pharmaceutical diluents, e.g. water
  • a pharmaceutical carrier e.g. conventional tableting ingredients such as corn starch, lactose, sucrose, sorbitol, talc, stearic acid, magnesium stearate, dicalcium phosphate
  • This solid preformulation composition is then subdivided into unit dosage forms of the type described above containing from 0.1 to about 500 mg of the active ingredient of the present invention.
  • Flavored unit dosage forms contain from 1 to 100 mg, for example 1, 2, 5, 10, 25, 50 or 100 mg, of the active ingredient.
  • the tablets or pills of the novel composition can be coated or otherwise compounded to provide a dosage form affording the advantage of prolonged action.
  • the tablet or pill can comprise an inner dosage and an outer dosage component, the latter being in the form of an envelope over the former.
  • the two components can be separated by an enteric layer which serves to resist disintegration in the stomach and permits the inner component to pass intact into the duodenum or to be delayed in release.
  • a variety of materials can be used for such enteric layers or coatings, such materials including a number of polymeric acids and mixtures of polymeric acids with such materials as shellac, cetyl alcohol and cellulose acetate.
  • the liquid forms in which the novel compositions of the present invention may be incorporated for administration orally or by injection include aqueous solutions, suitably flavored syrups, aqueous or oil suspensions, and flavored emulsions with edible oils such as cottonseed oil, sesame oil, coconut oil or peanut oil, as well as elixirs and similar pharmaceutical vehicles.
  • Suitable dispersing or suspending agents for aqueous suspensions include synthetic and natural gums such as tragacanth, acacia, alginate, dextran, sodium carboxymethylcellulose, methylcellulose, polyvinyl-pyrrolidone or gelatin.
  • a suitable dosage level is about 0.01 to 250 mg/kg per day, preferably about 0.05 to 100 mg/kg per day, and especially about 0.05 to 20 mg/kg per day.
  • the compounds may be administered on a regimen of 1 to 4 times per day.
  • a method for treating demyelinating diseases in a patient comprising administration of a therapeutically effective amount of a hPPAR delta agonist.
  • the hPPAR delta agonist is a selective agonist.
  • the demylenating disease is selected from the group consisting of multiple sclerosis, Charcot-Marie-Tooth disease, Pelizaeus-Merzbacher disease, encephalomyelitis, neuromyelitis optica, adrenoleukodystrophy, Guillian-Barre syndrome and disorders in which myelin forming glial cells are damaged including spinal cord injuries, neuropathies and nerve injury.
  • the demylenating disease is multiple sclerosis.
  • the agonist is selected from group consisting of compound of formula (1) and formula (2)
  • a pharmaceutical composition comprising a compound selected from the group consisting of compound of formula (1) and formula (2) in an amount effective for treating multiple sclerosis, Charcot- Marie-Tooth disease, Pelizaeus-Merzbacher disease, encephalomyelitis, neuromyelitis optica, adrenoleukodystrophy, Guillian-Barre syndrome and disorders in which myelin forming glial cells are damaged including spinal cord injuries, neuropathies and nerve injury in combination with at least on pharmaceutically acceptable carrier (1)
  • composition comprising an amount effective in treating multiple sclerosis.
  • FIG. 1 This graph shows the enhancement of MBP mRNA in cultured rat oligodendrocytes by compound 1.
  • FIG. 3 This graph shows the enhancement of MBP mRNA in cultured rat oligodendrocytes by compound 2.
  • Figure 4A Illustrates the effect of compound 1 on transcriptional markers that confirm PPAR delta agonist pathway activation in cultured rat oligodendrocytes.
  • Figure 4B Further illustrates the effect of compound 1 on transcriptional markers that confirm PPAR delta agonist pathway activation in cultured rat oligodendrocytes, showing that ADRP mRNA is upregulated in cultured rat oligodendrocytes.
  • Figure 5 Shows the increase in the number of O4 immunopositive cells in mixed cultures of human oligodendrocytes effected by compound 1.
  • Figure 6 Shows the increase in the number of O4 immunopositive cells in mixed cultures of human oligodendrocytes effected by compound 2.
  • Compound of formula (1) (GW501516) can be prepared as published in WO 01/00603.
  • Compound of formula (2) (L165,041 ) can be prepared as described in W0 97/28149.
  • the following test protocols are used to ascertain the biological properties of the compounds of this invention.
  • the following examples are being presented to further illustrate the invention. However, they should not be construed as limiting the invention in any manner.
  • the PPAR delta agonists of the present invention are evaluated in in vitro and in vivo models for their ability to promote myelin expression and enhance regenerative processes.
  • the optimum nuclear receptor selectivity profile is determined by the GAL4/luciferase reporter assays.
  • a rodent cellular assay shows the compound's ability to direct / accelerate differentiation of cultured oligodendrocyte progenitor cells to mature oligodendrocytes.
  • Specific biological assays suggesting efficacy for the treatment of MS are lysolecithin induced demyelination and experimental allergic encephalomyelitis performed in rodents.
  • the PPAR delta reporter cell line contains two genetic elements, a luciferase reporter element (pdeltaM-GAL4-Luc- Zeo) and a PPAR delta fusion protein (GR-GAL4-humanPPAR delta-LBD), which mediates expression of the luciferase reporter element depending on a PPAR delta ligand.
  • the stably and constitutively expressed fusion protein GR-GAL4-humanPPAR delta-LBD binds in the cell nucleus of the PPAR delta reporter cell line via the GAL4 protein portion to the GAL4 DNA binding motifs 5 '-upstream of the luciferase reporter element which is stably integrated in the genome of the cell line.
  • cs-FCS fatty acid-depleted fetal calf serum
  • PPAR delta ligands bind and activate the PPAR delta fusion protein and thereby stimulate expression of the luciferase reporter gene.
  • the luciferase, which is formed can be detected by means of chemiluminescence via an appropriate substrate.
  • the production of the stable PPAR delta reporter cell line is based on a stable HEK- cell clone which was stably transfected with a luciferase reporter element. This step was already described above in the section "construction of the PPAR alpha reporter cell line”.
  • the PPAR delta fusion protein GR-GAL4-humanPPAR delta-LBD was stably introduced into this cell clone.
  • Glucocorticoid receptor accesion # P04150
  • GAL4 yeast transcription factor
  • the cDNA of the ligand-binding domain of the human PPAR delta receptor was cloned in at the 3'-end of this GR-GAL4 construct.
  • the fusion construct prepared in this way (GR-GAL4-humanPPAR delta-LBD) was recloned into the plasmid pcDNA3 (Invitrogen) in order to enable constitutive expression by the cytomegalovirus promoter.
  • This plasmid was linearized with a restriction endonuclease and stably transfected into the previously described cell clone containing the luciferase reporter element.
  • the resulting PPAR delta reporter cell line which contains a luciferase reporter element and constitutively expresses the PPAR delta fusion protein (GR-GAL4- human PPAR delta-LBD) was isolated by selection with zeocin (0.5 mg/ml) and G418 (0.5 mg/ml).
  • the activity of PPAR delta agonists is determined in a 3-day assay, which is described below:
  • the PPAR delta reporter cell line is cultivated to 80% confluence in DMEM (# 41965- 039, Invitrogen) which is mixed with the following additions: 10% cs-FCS (fetal calf serum; #SH-30068.03, Hyclone), 0.5 mg/ml zeocin (#R250-01, Invitrogen), 0.5 mg/ml G418 (#10131-027, Invitrogen), 1% penicillin-streptomycin solution (#15140-122, Invitrogen) and 2 mM L-glutamine (#25030-024, Invitrogen).
  • 10% cs-FCS fetal calf serum
  • #SH-30068.03, Hyclone fetal calf serum
  • zeocin fetal calf serum
  • G418 0.5 mg/ml G418
  • penicillin-streptomycin solution #15140-122, Invitrogen
  • the cultivation takes place in standard cell culture bottles (# 353112, Becton Dickinson) in a cell culture incubator at 37°C in the presence of 5% CO 2 .
  • the 80%-confluent cells are washed once with 15 ml of PBS (#14190-094, Invitrogen), treated with 3 ml of trypsin solution (#25300-054, Invitrogen) at 37°C for 2 min, taken up in 5 ml of the DMEM described and counted in a cell counter. After dilution to 500.000 cells/ml, 35,000 cells are seeded in each well of a 96 well microtiter plate with a clear plastic base
  • the plates are incubated in the cell culture incubator at 37°C and 5% CO 2 for 24 h.
  • Day 2 PPAR delta agonists to be tested are dissolved in DMSO in a concentration of 10 mM.
  • This stock solution is diluted in DMEM (#41965-039, Invitrogen) which is mixed with 5% cs- FCS (#SH-30068.03, Hyclone), 2 mM L-glutamine (#25030-024, Invitrogen) and the previously described antibiotics (zeocin, G418, penicillin and streptomycin).
  • Test substances are tested in 11 different concentrations in the range from 10 ⁇ M to 100 pM. More potent compounds are tested in concentration ranges from 1 ⁇ M to 10 pM or between 100 nM and 1 pM.
  • the medium of the PPAR delta reporter cell line seeded on day 1 is completely removed by aspiration, and the test substances diluted in medium are immediately added to the cells.
  • the dilution and addition of the substances is carried out by a robot (Beckman FX).
  • the final volume of the test substances diluted in medium is 100 ⁇ l per well of a 96 well microtiter plate.
  • the DMSO concentration in the assay is less than 0.1 % v/v in order to avoid cytotoxic effects of the solvent.
  • Each plate was charged with a standard PPAR delta agonist, which was likewise diluted in 11 different concentrations, in order to demonstrate the functioning of the assay in each individual plate.
  • the assay plates are incubated in an incubator at 37°C and 5% CO 2 for 24 h.
  • the PPAR delta reporter cells treated with the test substances are removed from the incubator, and the medium is aspirated off.
  • the cells are lyzed by pipetting 50 ⁇ l of Bright Glo reagent (from Promega) into each well of a 96 well microtiter plate. After incubation at room temperature in the dark for 10 minutes, the microtiter plates are measured in the luminometer (Trilux from Wallac). The measuring time for each well of a microtiter plate is 1 sec. Evaluation:
  • the raw data from the luminometer are transferred into a Microsoft Excel file.
  • Dose- effect plots and EC50 values of PPAR agonists are calculated using the XL.Fit program as specified by the manufacturer (IDBS).
  • HEK human embryo kidney
  • pdeltaM-GAL4-Luc-Zeo a luciferase reporter element
  • GR- GAL4-humanPPAR alpha-LBD PPAR alpha fusion protein
  • the stably and constitutively expressed fusion protein GR-GAL4-humanPPAR alpha-LBD binds in the cell nucleus of the PPAR alpha reporter cell line via the GAL4 protein portion to the GAL4 DNA binding motifs 5 '-upstream of the luciferase reporter element which is stably integrated in the genome of the cell line.
  • cs-FCS fatty acid-depleted fetal calf serum
  • PPAR alpha ligands bind and activate the PPAR alpha fusion protein and thereby stimulate the expression of the luciferase reporter gene.
  • the luciferase which is formed can be detected by means of chemiluminescence via an appropriate substrate.
  • the PPAR alpha reporter cell line was prepared in two stages. Firstly, the luciferase reporter element was constructed and stably transfected into HEK cells. For this purpose, five binding sites of the yeast transcription factor GAL4 (Accession # AF264724) were cloned in 5 '-upstream of a 68 bp-long minimal MMTV promoter (Accession # V01175). The minimal MMTV promoter section contains a CCAAT box and a TATA element in order to enable efficient transcription by RNA polymerase II. The cloning and sequencing of the GAL4- MMTN construct took place in analogy to the description of Sambrook J. et. al. (Molecular cloning, Cold Spring Harbor Laboratory Press, 1989).
  • the complete Photinus pyralis gene (Accession # Ml 5077) was cloned in 3 '-downstream of the GAL4-MMTN element.
  • the luciferase reporter element consisting of five GAL4 binding sites, MMTV promoter and luciferase gene was recloned into a plasmid which confers zeocin resistance in order to obtain the plasmid pdeltaM-GAL4-Luc-Zeo.
  • This vector was transfected into HEK cells in accordance with the statements in Ausubel, F.M. et al. (Current protocols in molecular biology, Vol. 1-3, John Wiley & Sons, Inc., 1995).
  • zeocin-containing medium 0.5 mg/ml
  • zeocin-containing medium 0.5 mg/ml
  • PPAR alpha fusion protein GR-GAL4-humanPPAR alpha-LBD
  • the cD ⁇ A coding for the ⁇ -terminal 76 amino acids of the glucocorticoid receptor accesion # P04150
  • the cD ⁇ A section coding for amino acids 1-147 of the yeast transcription factor GAL4 accesion # P04386.
  • the cD ⁇ A of the ligand-binding domain of the human PPAR alpha receptor was cloned in at the 3'-end of this GR-GAL4 construct.
  • the fusion construct prepared in this way (GR-GAL4-humanPPAR alpha-LBD) was recloned into the plasmid pcD ⁇ A3 (Invitrogen) in order to enable constitutive expression therein by the cytomegalovirus promoter.
  • This plasmid was linearized with a restriction endonuclease and stably transfected into the previously described cell clone containing the luciferase reporter element.
  • the finished PPAR alpha reporter cell line which contains a luciferase reporter element and constitutively expresses the PPAR alpha fusion protein (GR-GAL4-human PPAR alpha-LBD) was isolated by selection with zeocin (0.5 mg/ml) and G418 (0.5 mg/ml).
  • the activity of PPAR alpha agonists is determined in a 3-day assay, which is described below:
  • the PPAR alpha reporter cell line is cultivated to 80% confluence in DMEM (# 41965-039, Invitrogen) which is mixed with the following additions: 10% cs-FCS (fetal calf serum; #SH-30068.03, Hyclone), 0.5 mg/ml zeocin (#R250-01, Invitrogen), 0.5 mg/ml G418 (#10131-027, Invitrogen), 1% penicillin-streptomycin solution (#15140-122, Invitrogen) and 2 mM L-glutamine (#25030-024, Invitrogen).
  • 10% cs-FCS fetal calf serum
  • #SH-30068.03, Hyclone fetal calf serum
  • zeocin fetal calf serum
  • G418 0.5 mg/ml G418
  • penicillin-streptomycin solution #15140-122, Invitrogen
  • the cultivation takes place in standard cell culture bottles (# 353112, Becton Dickinson) in a cell culture incubator at 37°C in the presence of 5% CO 2 .
  • the 80%-confluent cells are washed once with 15 ml of PBS (#14190-094, Invitrogen), treated with 3 ml of trypsin solution (#25300-054, Invitrogen) at 37°C for 2 min, taken up in 5 ml of the DMEM described and counted in a cell counter. After dilution to 500.000 cells/ml, 35,000 cells are seeded in each well of a 96 well microtiter plate with a clear plastic base (#3610, Corning Costar). The plates are incubated in the cell culture incubator at 37°C and 5% CO 2 for 24 h.
  • Day 2 PPAR alpha agonists to be tested are dissolved in DMSO in a concentration of 10 mM. This stock solution is diluted in DMEM (#41965-039, Invitrogen) which is mixed with 5% cs- FCS (#SH-30068.03, Hyclone), 2 mM L-glutamine (#25030-024, Invitrogen) and the previously described antibiotics (zeocin, G418, penicillin and streptomycin).
  • Test substances are tested in 11 different concentrations in the range from 10 ⁇ M to 100 pM. More potent compounds are tested in concentration ranges from 1 ⁇ M to 10 pM or between 100 nM and 1 pM.
  • the medium of the PPAR alpha reporter cell line seeded on day 1 is completely removed by aspiration, and the test substances diluted in medium are immediately added to the cells. The dilution and addition of the substances is carried out by a robot (Beckman FX).
  • the final volume of the test substances diluted in medium is 100 ⁇ l per well of a 96 well microtiter plate.
  • the DMSO concentration in the assay is less than 0.1 % v/v in order to avoid cytotoxic effects of the solvent.
  • Each plate was charged with a standard PPAR alpha agonist, which was likewise diluted in 11 different concentrations, in order to demonstrate the functioning of the assay in each individual plate.
  • the assay plates are incubated in an incubator at 37°C and 5% CO 2 for 24 h.
  • the PPAR alpha reporter cells treated with the test substances are removed from the incubator, and the medium is aspirated off.
  • the cells are lyzed by pipetting 50 ⁇ l of Bright Glo reagent (from Promega) into each well of a 96 well microtiter plate. After incubation at room temperature in the dark for 10 minutes, the microtiter plates are measured in the luminometer (Trilux from Wallac). The measuring time for each well of a microtiter plate is 1 sec.
  • luciferase assay is performed in 96 well plates as follows:
  • Dilution steps and addition of diluted compounds is done using a Beckman Biomek 2000 or Beckman FX robot.
  • Determination of EC50 values is done with Microsoft Exel in combination with XLFit (develop by IDBS) using the fitting algorithm #205. Determination of EC50 values in the cellular human RXR receptor assay Cell based RXR Assay protocol
  • luciferase assay is performed in 96 well plates as follows: Day 1: Plating of cells
  • Table 1 shows the results if the reporter assays. The results show that compounds 1 and 2 are selective PPAR delta activators with low PPAR alpha, gamma and RXR activity.
  • Primary rat oligodendrocyte progenitor cells are obtained from the neocortex of newborn (postnatal days 2-3) rats or mice and are enriched, after removal of microglia, by mechanical separation from the astrocytic monolayer using a modification of the technique originally described by McCarthy and de Vellis (1980). 2. Remove the meninges from neonatal rat brain and mechanically dissociate tissue. Plate cells on T75 flasks and feed cells with DMEM/F12 + 10% FBS. 3. Collect oligodendrocytes growing on the astrocyte bed layer by shaking-off method fourteen days after the original prep date.
  • Progenitor cell passaging to increase cell numbers for screening assays 1. When the culture are confluent, rinse the culture with PBS, add trypsin and incubate for -2-3 min at 37°C. 2. Neutralize and centrifuge the cell suspension at 900g for 5 min. 3. Resuspend the cell pellet in SFM + PDGF/FGF. 4. Feed the cells with fresh growth factors every 48 hrs to keep enrich for rapidly dividing progenitor cells. 5. Cells are passaged no more than 4-5 times prior to experimental assays. 6. All experiments involving oligodendrocyte progenitor cells were done using cells that were continuously maintained under these conditions.
  • the enriched rat Central Glia-4 (CG4) progenitor cell line may be used, which is maintained in base media (DMEM, with 2 mM glutamine, lmM sodium pyruvate, biotin (40 nM), insulin (1 ⁇ M) and NI) supplemented with 30% conditioned media from the B-104 neuroblastoma cell line.
  • base media DMEM, with 2 mM glutamine, lmM sodium pyruvate, biotin (40 nM), insulin (1 ⁇ M) and NI
  • base media fetal calf serum
  • insulin 500 nM
  • A2B5 and MBP immunoreactivity is used to confirm >95% enrichment in immature and mature cultures, respectively.
  • Compound dose response evaluations are performed at 6 concentrations (10 ⁇ M, 1 ⁇ M, 100 nM, 10 nM, 1 nM, and 0.1 nM); c. Triplicates wells are run for each compound concentration. 4. Culture the treated cells for 7 d prior to using in experimental assays.
  • HUMAN Oligodendrocyte cultures Preparation of cells: 1. Human neurospheres collected from E19.5 - E22 human embryo cortex) are cultured for 2 weeks in progenitor media: DMEM/F12 containing 100 ⁇ g/ml transferring, 30 nM triiodothyronine, 20 nM hydrocortisone, 20 nM progesterone, 10 nM biotin, lx trace elements, 30 nM selenium, 60 uM putrescine, 0.1% BSA, 5 U/ml PenStrep, 25 ⁇ g/ml insulin) supplemented with PDGF and FGF. 2. Neurospheres are dissociated with 20 U/ml papain at 37°C for 30-50 min. 3. Cells are plated onto PDL coated dishes at density of 50,000-100,000 cell/well in progenitor media containing PDGF/FGF and incubated at 37°C with 5-6% CO2.
  • oligodendrocyte-specific antibodies are used to assess ability of compound to accelerate/promote oligodendrocyte differentiation (for example, O4, OI, or myelin basic protein immunoreactivity is over time between compound treated and untreated cultures). 1. Cells are plated onto poly-D-lysine treated 4-well chamber slides at 5xl0 3 to 20xl0 3 cells/well and grown as described above. Sequential staining is performed on oligodendrocyte populations with increasing degrees of cellular differentiation, as determined by days in vitro without PDGF and FGF. 2.
  • Live staining for 30 min at 37°C is used to detect oligodendrocyte stage specific cell surface marker expression (including A2B5, O4, and OI). 3. Subsequently, cells are fixed with 4% paraformaldehyde, 10 min, room temperature. 4. Fixed staining procedures are used to detect oligodendrocyte stage specific marker expression (including myelin basic protein, MBP). 5. Rinse with PBS. 6. Permeabilize with 0.1% Triton/0.01% NaAz diluted in IX PBS for 10 min, room temperature. 7. Block with 5-10% goat serum in antibody dilution buffer (0.1% Triton-X 100 and 1% IgG-free bovine serum albumin; also used to dilute antibodies), 15 min, room temperature. 8.
  • RAT/MOUSE/HUMAN Bromodeoxyuridine (BrdU) immunostaining To confirm that compounds do not promote cell proliferation.
  • Oligodendrocyte progenitor cells are labeled with 10 ⁇ M BrdU for 20 hr and then fixed with either 70% ethanol or 4% paraformaldehyde. 2. The cells are incubated successively with biotinylated mouse anti-BrdU and Streptavidin-Peroxidase, with three intervening washes with PBS. 3. Colormetric visualization of the BrdU immunoreactivity is developed with DAB and total cell numbers are assessed using the counter-stain hematoxylin. 4. BrdU immunopositive cells are counted by two independent observers.
  • RAT/MOUSE/HUMAN Culture Image analysis Fluorescent microscopy is used to quantitate the extent of oligodendrocyte differentiation after compound exposure. This assay demonstrates that selective agonists accelerate/promote oligodendrocytes differentiation.
  • Manual Cell Counting Four fields are randomly selected for each experimental condition and 500-600 cells are counted in each field. The percentage of MBP (or O4) immunpositive cells (mature process bearing cells with or without myelin sheets) versus DAPI positive cells (total cell number) cells are compared in the control and drug-treated groups.
  • Automated Cell Counting Fluorescent microscopy was used to quantitate the extent of oligodendrocyte differentiation after compound exposure.
  • Immunofluorescence images are obtained using a Zeiss AxioVision digital imaging system, with a Zeiss AxioCam HRc cooled CCD camera connected to the same microscope. All microscopic imaging parameters are set for acquiring images for the analysis of cellular immunofluorescence intensity. The percentage of MBP positive (differentiated) cells versus total cells (DAPI nuclear stained) is compared in the control versus drug-treated groups. Cellular autofluorescence was undetectable under the imaging conditions, a) 3.
  • Human oligodendrocyte differentiation assay manually count total number of O4 immunopositive cells/well (bipolar and multipolar). The results using rat oligodendrocyte cultures are shown in Figure 1 and the results using human oligodendrocyte mixed cultures are shown in Figures 5 and 6. As the results show, PPAR delta agonists enhance or accelerate rat and human oligodendrocyte differentiation, as measured by increased myelin basic protein expression compared to untreated controls. This novel finding would suggest that compound 1 and compound 2 and selective PPAR delta agonists in general would be enhance, accelerate, or stimulate oligodendrocyte differentiation and myelin formation in vivo, in the diseased or injured CNS, including MS and other demyelinating disorders.
  • PCR RAT/MOUSE/HUMAN Quantitative Polvmerase Chain Reaction
  • the 18S ribosomal RNA primer/probe mix (186 bp product), suspended in Taqman 2X PCR Master Mix is used as an internal control. 5. Quantitative PCR is carried out using real-time TaqmanTM technology (Gibson, et al., 1996) with a model 7700 Sequence Detector System (Applied Biosystems, Foster City, CA). 6. The results are analyzed using Sequence Detection Systems software version 1.91. Results for these assays are shown in Figures 2, 3, 4A, and 4B. These results suggest that PPAR delta selective agonists bind the PPAR delta receptor and directly activate the PPAR delta pathway in oligodendrocytes and should act similarly in vivo.
  • RAT ELISA Assay To evaluate compound induced PPAR delta pathway activation and the extent of oligodendrocyte maturation (changes in protein levels).
  • mice The bone is removed and rats are injected with 2 ⁇ l ethidium bromide, lysolecithin, or SIN-1 into the right caudal cerebellar peduncle, DV -7.1 mm, over a 2 min period by means of a Hamilton ⁇ l syringe and needle. Alternatively injections are made into the spinal cord, corpus callosum, or cortex. 7. The needle is left in position for the subsequent 2 min. 8. After withdrawal of the needle the incision is sutured. 9. Each rat receives an i.m. injection of 0.003mg buprenorphine into a hind leg. 10. The rat is placed in a warming cupboard until it regains consciousness. At which time it is returned to its home cage. Do not allow more than 2 rats per cage, as they will pull each other's suture out. 11. Similar procedures are also done using mice.
  • Rat Experimental Allergic Encephalomyelitis (Rat EAE) Disease Model Experimental allergic encephalomyelitis (EAE) is a T-cell-mediated autoimmune disease of the nervous system that develops in susceptible animals following sensitization with either whole spinal cord homogenate or a component (myelin basic protein).
  • the EAE rodent model is an appropriate tool for studying the inflammation of the brain and spinal cord observed in MS patients.
  • injection of whole spinal cord or spinal cord components such as myelin basic protein induces an autoimmune response based on the activation of T- lymphocytes.
  • Clinical disease typically becomes manifest around day 8-10 after inoculation, observed as a broad spectrum of behavioral anomalies ranging from mild gait disturbances and tail atony to complete paralysis and death.
  • EAE acute EAE
  • chronic relapsing EAE chronic relapsing EAE
  • treatment paradigms may be used: the drug or treatment of choice may be administered before immunization, during the nonsymptomatic period or during the clinical disease.
  • Antigen preparation (for approximately 720 animals): 1. Weigh 5 grams of frozen guinea pig spinal cord.
  • mice Female Lewis rats (Charles River) are given free access to food and water and should be acclimated a minimum of 3 days before use in experiments.
  • Rats weighing 160 and 220 grams are initially induced with 5% isoflurane (Aerrane, Fort Dodge), 30% O 2 , 70% N 2 O for 2-5 minutes.
  • the rat is then placed onto a circulating water heating blanket (Gaymar) (dorsal surface up) and into the nose cone for spontaneous respiration of anesthetic gases.
  • the isoflurane is reduced to 2%.
  • the animals are removed from the nose cone, weighed and numbered.
  • the rats are allowed to awake from anesthesia and are placed into individual cages.
  • STAGE 1 Abnormal gate and tail atony
  • STAGE 2 Mild but definite weakness of one or both hind legs
  • STAGE: 3 Severe weakness of one or both hind legs or mild ataxia
  • STAGE: 4 Severe paraparesis and minimal hind leg movement
  • STAGE: 5 No hind leg movement and paraplegia
  • STAGE 6 Moribund state with no spontaneous movement and impaired respiration. Increasing degree of front leg involvement and urinary and fecal incontinence may also occur STAGE:7 DEATH Treatment is begun on day 10 after immunization. Since the disease symptoms in this model typically appear 10-11 days after inoculation, this time point may be considered to represent the initial phase of an acute episode of MS. It is judged that this delay of the start of treatment mimics the clinical situation more closely than the traditionally used protocols where drugs are administered at the time of, or even before, inoculation (Teitelbaum D. et al., Proc Natl Acad Sci USA 1999; 96: 3842-3847 and Brod S. A., et al., Ann Neurol 2000; 47: 127-131).

Abstract

A method for treating demyelinating diseases in a patient in need thereof by treatment with an effective amount of a PPAR delta agonist is disclosed. Demyelinating diseases that may be effectively treated by this method include but are not limited to multiple sclerosis, Charcot-Marie-Tooth disease, Pelizaeus-Merzbacher disease, encephalomyelitis, neuromyelitis optica, adrenoleukodystrophy, Guillian-Barre syndrome and disorders in which myelin forming glial cells are damaged including spinal cord injuries, neuropathies and nerve injury.

Description

USE OF PEROXISOME PROLIFERATOR ACTIVATED RECEPTOR DELTA AGONISTS FOR THE TREATMENT OF MS AND OTHER DEMYELINATING DISEASES
FIELD OF THE INVENTION This invention relates to the use of PPAR delta agonists for the treatment of multiple sclerosis (MS) and other demyelinating diseases. This invention also relates to the use of certain compounds that are selective PPAR delta agonists for the treatment of MS and other demyelinating diseases. BACKGROUND OF THE INVENTION The peroxisome proliferator-activated receptors (PPARs) comprise a subfamily of the nuclear receptor superfamily. Three closely related isoforms have been identified and cloned and are commonly known as PPAR alpha, PPAR gamma and PPAR delta. Each receptor subtype has a signature DNA binding domain (DBD) and a ligand-binding domain (LBD), both being necessary for ligand activated gene expression. PPARs bind as heterodimers with a retinoid X receptor. See J. Berger and D. E. Miller, Ann. Rev. Med., 2002, 53, 409-435. PPAR delta (also known as PPAR beta) is expressed in a broad range of mammalian tissue, but little information regarding its biological functions or the full array of genes regulated by the receptor have been elucidated. However, it has recently been found that agonists may be useful to treat conditions such as dyslipedemia and certain dermatological conditions, while antagonists may be useful to treat osteoporosis or colorectal cancer (D. Sternbach, in Annual Reports in Medicinal Chemistry, Volume 38, A. M. Doherty, ed., Elsevier Academic Press, 2003 pp. 71-80). PPAR delta appears to be significantly expressed in the CNS; however much of its function there still remains undiscovered. Of singular interest however, is the discovery that PPAR delta was expressed in rodent oligodendrocytes, the major lipid producing cells of the CNS (J. Granneman, et al., J. Neurosci. Res., 1998, 51, 563-573). Moreover, it was also found that a PPAR delta selective agonist was found to significantly increase oligodendroglial myelin gene expression and myelin sheath diameter in mouse cultures (I. Saluja et al., Glia, 2001, 33, 194-204). Demyelinating conditions are manifested in loss of myelin— the multiple dense layers of lipids and protein which cover many nerve fibers. These layers are provided by oligodendroglia in the central nervous system (CNS), and Schwann cells in the peripheral nervous system (PNS). In multiple sclerosis (MS), oligodendrocytes, the myelin forming cells in the CNS, are destroyed and axons are damaged, resulting in severely impaired neuronal activity and functional deficits, including palegia. In patients with demyelinating conditions, demyelination may be irreversible; it is usually accompanied or followed by axonal degeneration, and often by cellular degeneration. Demyelination can occur as a result of neuronal damage or damage to the myelin itself-whether due to aberrant immune responses, local injury, ischemia, metabolic disorders, toxic agents, or viral infections (Prineas and McDonald, Demyelinating Diseases. In Greenfield's Neuropathology, ό.sup.th ed. (Edward Arnold: New York, 1997) 813-811, Beers and Berkow, eds., The Merck Manual of Diagnosis and Therapy, 17.sup.th ed. (Whitehouse Station, N.J.: Merck Research Laboratories, 1999) 1299, 1437, 1473-76, 1483). However, newly formed oligodendrocyte progenitor cells are present throughout areas of demyelination, suggesting the possibility of self -repair if these progenitor cells can be induced to undergo differentiation to mature oligodendrocytes. Central demyelination (demyelination of the CNS) occurs in several conditions, often of uncertain etiology, that have come to be known as the primary demyelinating diseases. Of these, multiple sclerosis is the most prevalent. Other primary demyelinating diseases include adrenoleukodystrophy (ALD), adrenomyeloneuropathy, AIDS-vacuolar myelopathy, HTLV- associated myelopathy, Leber's hereditary optic atrophy, progressive multifocal leukoencephalopathy (PML), subacute sclerosing panencephalitis, and tropical spastic paraparesis. In addition, there are acute conditions in which demyelination can occur in the CNS, e.g., acute disseminated encephalomyelitis (ADEM) and acute viral encephalitis. Furthermore, acute transverse myelitis, a syndrome in which an acute spinal cord transection of unknown cause affects both gray and white matter in one or more adjacent thoracic segments, can also result in demyelination. MS is a chronic, devastating, neurological disease that affects mostly young adults. The pathogenesis of MS is a complex process that leads to destruction of myelin and oligodendroglia, as well as axonal damage, in the brain and spinal cord (Prineas and
McDonald, Demyelinating Diseases. In Greenfield's Neuropathology, ό.sup.th ed. (Edward Arnold: New York, 1997) 813-811, Trapp et al., N. Engl. J. Med, 338:278-85, 1998). Histopathologically, MS is characterized by inflammation, plaques of demyelination infiltrating cells in the CΝS tissue, loss of oligodendroglia, and focal axonal injury (Prineas and McDonald, Demyelinating Diseases. In Greenfield's Neuropathology, ό.sup.th ed.
(Edward Arnold: New York, 1997) 813-811). The disease is thought to result from aberrant immune responses to myelin, and possibly non-myelin, self-antigens (Bar-Or et al., J. Neuroimmunol. 100:252-59, 1999, Hartung, H.-P., Current Opinion in Neurology, 8:191-99, 1995). Clinically, MS may follow a relapsing-remitting, or it may take a chronically progressive course with increasing physical disability (Gold et al., Mol. Med. Today, 6:88-91, 2000). Typically, the symptoms of MS include lack of co-ordination, paresthesias, speech and visual disturbances, and weakness. Current treatments for the various demyelinating conditions are often expensive, symptomatic, and only partially effective, and may cause undesirable secondary effects. Corticosteroids (oral prednisone at 60-100 mg/day, tapered over 2-3 weeks, or intravenous methylprednisolone at 500-1000 mg/day, for 3-5 days) represent the main form of therapy for MS. While these may shorten the symptomatic period during attacks, they may not affect eventual long-term disability. Long-term corticosteroid treatment is rarely justified, and can cause numerous medical complications, including osteoporosis, ulcers, and diabetes (Beers and Berkow, eds., The Merck Manual of Diagnosis and Therapy, 17.sup.th ed. (Whitehouse Station, N.J.: Merck Research Laboratories, 1999) 1299, 1437, 1473-76, 1483). Immunomodulatory therapy with recombinant human interferon-.beta. (Betaseron and Avonex) and with co-polymer (Copaxon) slightly reduces the frequency of relapses in MS, and may help delay eventual disability (Beers and Berkow, eds., The Merck Manual of Diagnosis and Therapy, 17.sup.th ed. (Whitehouse Station, N.J.: Merck Research
Laboratories, 1999) 1299, 1437, 1473-76, 1483). Both forms of interferon-.beta. and co- polymer are currently used as treatment modalities for MS, but all are exceedingly expensive. Immunosuppressive drugs (azathioprine, cladribine, cyclophosphamide, and methotrexate) are used for more severe progressive forms. However, they are not uniformly beneficial, and have significant toxic side effects. Several drugs (e.g., baclofen at 30-60 mg/day in divided doses) may reduce spasticity by inhibiting the spinal cord reflexes. Cautious and judicious use is required, though, because the drug-induced reduction in spasticity in MS patients often exacerbates weakness, thereby further incapacitating the patient. Similarly, current treatment for ALD, another devastating demyelinating disease, is relatively ineffective. Symptoms of ALD may include cortical blindness, corticospinal tract dysfunction, mental deterioration, and spasticity. Therapy to control the course of ALD may include bone marrow transplantation and dietary treatment (DiBiase et al., Ann. 1st. Super Sanita, 35: 185-92, 1999), but inexorable neurological deterioration invariably occurs, ultimately leading to death [Krivit et al., Curr. Opin. Hematol., 6:377-82, 1999, (Beers and Berkow, eds., The Merck Manual of Diagnosis and Therapy, 17.sup.th ed. (Whitehouse Station, N.J.: Merck Research Laboratories, 1999) 1299, 1437, 1473-76, 1483). Some progress has been realized in the treatment of animals with EAE and EAN, by using glial cell transplants and growth factors, and by inhibiting adhesion molecules, autoantibodies, and cytokines (Njenga and Rodriguez, Current Opinion in Neurology, 9:159-64, 1996. However, none of these treatments has been shown to be beneficial in humans, and some require extensive neurosurgical intervention. Thus, it is clear from the foregoing that there exists a need for more effective, and less expensive and invasive, methods to treat the varied array of demyelinating conditions, without producing undesirable secondary effects. The present invention entails the use of a small molecule-activated regenerative approach to significantly augment current immunomodulatory therapies for the treatment of demyelinating disorders. Compounds that are known to be selective PPAR delta are known in the art, in particular, compound of formula (1) generally known as GW 501516 described in WO 01/00603.
Figure imgf000006_0001
(1) Compound of formula (2) also known as L165,041 and has been disclosed in European Patent Application 28063 and in W0 97/28149 wherein it was identified as a selective PPAR delta agonist.
Figure imgf000007_0001
(2) Due to the potential ability of Peroxisome Proliferator Activated Receptor Delta (PPAR delta) agonists to accelerate the differentiation of acutely isolated oligodendrocyte progenitor cells from rodent cerebrum and to significantly increase both myelin sheath diameter and myelin gene expression, there exists the potential for PPAR delta agonists to activate the PPAR delta pathway in oligodendrocyte progenitor cells and enhance neuronal repair by restoring the myelin sheath to demyelinated axons in demyelinating disease, particularly MS.
SUMMARY OF THE INVENTION Thus in accordance with the practice of this invention there is provided a method of treating a variety of demyelinating disease conditions with PPAR delta agonists, and in particular multiple sclerosis. In general, the disease conditions that can be treated in accordance with the practice of this invention include but not limited to multiple sclerosis, Charcot-Marie-Tooth disease, Pelizaeus-Merzbacher disease, encephalomyelitis, neuromyelitis optica, adrenoleukodystrophy, Guillian-Barrte syndrome and disorders in which myelin forming glial cells are damaged including spinal cord injuries, neuropathies and nerve injury. The diseases as disclosed herein can be treated by administering to a patient in need of such treatment a therapeutically effective amount of a PPAR delta agonist. The present invention is also directed to the use of compounds of formula (I) and formula (II) for the treatment of demyelinating diseases, and in particular multiple sclerosis.
Figure imgf000008_0001
(1)
Figure imgf000008_0002
(2)
The present invention also comprises a method of treating multiple sclerosis in patients by administering a combination of a compound of formula (1) or formula (2) or pharmaceutically acceptable salt thereof, with another compound known to be effective for the treatment of multiple sclerosis in therapeutically effective amounts. Compounds that are currently used to treat the disease are the disease-modifying agents such as the interferons (interferon beta 1-a, beta 1-b and alpha 2), glatiramer acetate or corticosteroids such as methylprednisolone and prednisone. Also, chemotherapeutic agents such as methotrexate, azathioprine, cladribine cyclophosphamide and cyclosporine. DETAILED DESCRIPTION OF THE INVENTION As used herein, the expression "pharmaceutically acceptable carrier" means a non- toxic solvent, dispersant, excipient, adjuvant, or other material which is mixed with the compound of the present invention in order to permit the formation of a pharmaceutical composition, i.e., a dosage form capable of administration to the patient. One example of such a carrier is a pharmaceutically acceptable oil typically used for parenteral administration. The term "pharmaceutically acceptable salts" as used herein means that the salts of the compounds of the present invention can be used in medicinal preparations. Other salts may, however, be useful in the preparation of the compounds according to the invention or of their pharmaceutically acceptable salts. Suitable pharmaceutically acceptable salts of the compounds of this invention include acid addition salts which may, for example, be formed by mixing a solution of the compound according to the invention with a solution of a pharmaceutically acceptable acid such as hydrochloric acid, hydrobromic acid, sulfuric acid, methanesulfonic acid, 2-hydroxyethanesulfonic acid, p-toluenesulfonic acid, fumaric acid, maleic acid, hydroxymaleic acid, malic acid, ascorbic acid, succinic acid, glutaric acid, acetic acid, salicylic acid, cinnamic acid, 2-phenoxybenzoic acid, hydroxybenzoic acid, phenylacetic acid, benzoic acid, oxalic acid, citric acid, tartaric acid, glycolic acid, lactic acid, pyruvic acid, malonic acid, carbonic acid or phosphoric acid. The acid metal salts such as sodium monohydrogen orthophosphate and potassium hydrogen sulfate can also be formed. Also, the salts so formed may present either as mono- or di- acid salts and can exist either as hydrated or can be substantially anhydrous. Furthermore, where the compounds of the invention carry an acidic moiety, suitable pharmaceutically acceptable salts thereof may include alkali metal salts, e.g. sodium or potassium salts; alkaline earth metal salts, e.g. calcium or magnesium salts; and salts formed with suitable organic ligands, e.g. quaternary ammonium salts. The term "therapeutically effective amount" as used herein means an amount of the compound, which is effective in treating the named disorder or condition. As used herein, the expression "pharmaceutically acceptable carrier" means a non- toxic solvent, dispersant, excipient, adjuvant, or other material which is mixed with the compound of the present invention in order to permit the formation of a pharmaceutical composition, i.e., a dosage form capable of administration to the patient. One example of such a carrier is a pharmaceutically acceptable oil typically used for parenteral administration. The invention also provides pharmaceutical compositions comprising one or more of the compounds according to this invention in association with a pharmaceutically acceptable carrier. Preferably these compositions are in unit dosage forms such as tablets, pills, capsules, powders, granules, sterile parenteral solutions or suspensions, metered aerosol or liquid sprays, drops, ampoules, auto-injector devices or suppositories; for oral, parenteral, intranasal, sublingual or rectal administration, or for administration by inhalation or insufflation. Alternatively, the compositions may be presented in a form suitable for once-weekly or once- monthly administration; for example, an insoluble salt of the active compound, such as the decanoate salt, may be adapted to provide a depot preparation for intramuscular injection. An erodible polymer containing the active ingredient may be envisaged. For preparing solid compositions such as tablets, the principal active ingredient is mixed with a pharmaceutical carrier, e.g. conventional tableting ingredients such as corn starch, lactose, sucrose, sorbitol, talc, stearic acid, magnesium stearate, dicalcium phosphate or gums, and other pharmaceutical diluents, e.g. water, to form a solid preformulation composition containing a homogeneous mixture of a compound of the present invention, or a pharmaceutically acceptable salt thereof. When referring to these preformulation compositions as homogeneous, it is meant that the active ingredient is dispersed evenly throughout the composition so that the composition may be readily subdivided into equally effective unit dosage forms such as tablets, pills and capsules. This solid preformulation composition is then subdivided into unit dosage forms of the type described above containing from 0.1 to about 500 mg of the active ingredient of the present invention. Flavored unit dosage forms contain from 1 to 100 mg, for example 1, 2, 5, 10, 25, 50 or 100 mg, of the active ingredient. The tablets or pills of the novel composition can be coated or otherwise compounded to provide a dosage form affording the advantage of prolonged action. For example, the tablet or pill can comprise an inner dosage and an outer dosage component, the latter being in the form of an envelope over the former. The two components can be separated by an enteric layer which serves to resist disintegration in the stomach and permits the inner component to pass intact into the duodenum or to be delayed in release. A variety of materials can be used for such enteric layers or coatings, such materials including a number of polymeric acids and mixtures of polymeric acids with such materials as shellac, cetyl alcohol and cellulose acetate. The liquid forms in which the novel compositions of the present invention may be incorporated for administration orally or by injection include aqueous solutions, suitably flavored syrups, aqueous or oil suspensions, and flavored emulsions with edible oils such as cottonseed oil, sesame oil, coconut oil or peanut oil, as well as elixirs and similar pharmaceutical vehicles. Suitable dispersing or suspending agents for aqueous suspensions include synthetic and natural gums such as tragacanth, acacia, alginate, dextran, sodium carboxymethylcellulose, methylcellulose, polyvinyl-pyrrolidone or gelatin. In the treatment of various disease states as described herein, a suitable dosage level is about 0.01 to 250 mg/kg per day, preferably about 0.05 to 100 mg/kg per day, and especially about 0.05 to 20 mg/kg per day. The compounds may be administered on a regimen of 1 to 4 times per day. In one aspect of this invention there is disclosed a method for treating demyelinating diseases in a patient comprising administration of a therapeutically effective amount of a hPPAR delta agonist. In a further aspect of this embodiment, the hPPAR delta agonist is a selective agonist. In another aspect of this embodiment is disclosed a method wherein the demylenating disease is selected from the group consisting of multiple sclerosis, Charcot-Marie-Tooth disease, Pelizaeus-Merzbacher disease, encephalomyelitis, neuromyelitis optica, adrenoleukodystrophy, Guillian-Barre syndrome and disorders in which myelin forming glial cells are damaged including spinal cord injuries, neuropathies and nerve injury. In a further aspect of this embodiment is disclosed the method wherein the demylenating disease is multiple sclerosis. In yet another aspect of this embodiment is disclosed the method wherein the agonist is selected from group consisting of compound of formula (1) and formula (2)
Figure imgf000011_0001
(1)
Figure imgf000011_0002
(2) In another embodiment disclosed in the present invention is a pharmaceutical composition comprising a compound selected from the group consisting of compound of formula (1) and formula (2) in an amount effective for treating multiple sclerosis, Charcot- Marie-Tooth disease, Pelizaeus-Merzbacher disease, encephalomyelitis, neuromyelitis optica, adrenoleukodystrophy, Guillian-Barre syndrome and disorders in which myelin forming glial cells are damaged including spinal cord injuries, neuropathies and nerve injury in combination with at least on pharmaceutically acceptable carrier
Figure imgf000012_0001
(1)
Figure imgf000012_0002
(2)
In a further aspect of this embodiment is disclosed a pharmaceutical composition comprising an amount effective in treating multiple sclerosis.
BRIEF DESCRIPTION OF THE DRAWINGS Figure 1. Illustrates the enhancement of myelin basic protein (MBP) immunoreactivity in cultured rat oligodendrocytes by PPAR delta agonists.
Figure 2. This graph shows the enhancement of MBP mRNA in cultured rat oligodendrocytes by compound 1.
Figure 3. This graph shows the enhancement of MBP mRNA in cultured rat oligodendrocytes by compound 2.
Figure 4A. Illustrates the effect of compound 1 on transcriptional markers that confirm PPAR delta agonist pathway activation in cultured rat oligodendrocytes.
Figure 4B. Further illustrates the effect of compound 1 on transcriptional markers that confirm PPAR delta agonist pathway activation in cultured rat oligodendrocytes, showing that ADRP mRNA is upregulated in cultured rat oligodendrocytes.
Figure 5. Shows the increase in the number of O4 immunopositive cells in mixed cultures of human oligodendrocytes effected by compound 1.
Figure 6. Shows the increase in the number of O4 immunopositive cells in mixed cultures of human oligodendrocytes effected by compound 2. Compound Examples:
Compound of formula (1) (GW501516) can be prepared as published in WO 01/00603. Compound of formula (2) (L165,041 ) can be prepared as described in W0 97/28149.
Biological Examples: The following test protocols are used to ascertain the biological properties of the compounds of this invention. The following examples are being presented to further illustrate the invention. However, they should not be construed as limiting the invention in any manner. The PPAR delta agonists of the present invention are evaluated in in vitro and in vivo models for their ability to promote myelin expression and enhance regenerative processes. The optimum nuclear receptor selectivity profile is determined by the GAL4/luciferase reporter assays. A rodent cellular assay shows the compound's ability to direct / accelerate differentiation of cultured oligodendrocyte progenitor cells to mature oligodendrocytes. Specific biological assays suggesting efficacy for the treatment of MS are lysolecithin induced demyelination and experimental allergic encephalomyelitis performed in rodents.
Determination of EC50 values of PPAR agonists in the cellular PPAR delta assay
Principle The potency of substances, which bind to human PPAR delta and activate it in an agonistic manner, is analyzed using a stably transfected HEK cell line (HEK== human embryo kidney) which is referred to here as PPAR delta reporter cell line. The PPAR delta reporter cell line contains two genetic elements, a luciferase reporter element (pdeltaM-GAL4-Luc- Zeo) and a PPAR delta fusion protein (GR-GAL4-humanPPAR delta-LBD), which mediates expression of the luciferase reporter element depending on a PPAR delta ligand. The stably and constitutively expressed fusion protein GR-GAL4-humanPPAR delta-LBD binds in the cell nucleus of the PPAR delta reporter cell line via the GAL4 protein portion to the GAL4 DNA binding motifs 5 '-upstream of the luciferase reporter element which is stably integrated in the genome of the cell line. There is only little expression of the luciferase reporter gene in the absence of a PPAR delta ligand if fatty acid-depleted fetal calf serum (cs-FCS) is used in the assay. PPAR delta ligands bind and activate the PPAR delta fusion protein and thereby stimulate expression of the luciferase reporter gene. The luciferase, which is formed can be detected by means of chemiluminescence via an appropriate substrate.
Construction of the PPAR delta reporter cell line:
The production of the stable PPAR delta reporter cell line is based on a stable HEK- cell clone which was stably transfected with a luciferase reporter element. This step was already described above in the section "construction of the PPAR alpha reporter cell line". In a second step, the PPAR delta fusion protein (GR-GAL4-humanPPAR delta-LBD was stably introduced into this cell clone. For this purpose, the cDNA coding for the N-terminal
76 amino acids of the glucocorticoid receptor (Accession # P04150) was linked to the cDNA section coding for amino acids 1-147 of the yeast transcription factor GAL4 (Accession # P04386). The cDNA of the ligand-binding domain of the human PPAR delta receptor (amino acids S139-Y441; Accession # L07592) was cloned in at the 3'-end of this GR-GAL4 construct. The fusion construct prepared in this way (GR-GAL4-humanPPAR delta-LBD) was recloned into the plasmid pcDNA3 (Invitrogen) in order to enable constitutive expression by the cytomegalovirus promoter. This plasmid was linearized with a restriction endonuclease and stably transfected into the previously described cell clone containing the luciferase reporter element. The resulting PPAR delta reporter cell line which contains a luciferase reporter element and constitutively expresses the PPAR delta fusion protein (GR-GAL4- human PPAR delta-LBD) was isolated by selection with zeocin (0.5 mg/ml) and G418 (0.5 mg/ml).
Assay procedure and evaluation:
The activity of PPAR delta agonists is determined in a 3-day assay, which is described below:
Day 1 The PPAR delta reporter cell line is cultivated to 80% confluence in DMEM (# 41965- 039, Invitrogen) which is mixed with the following additions: 10% cs-FCS (fetal calf serum; #SH-30068.03, Hyclone), 0.5 mg/ml zeocin (#R250-01, Invitrogen), 0.5 mg/ml G418 (#10131-027, Invitrogen), 1% penicillin-streptomycin solution (#15140-122, Invitrogen) and 2 mM L-glutamine (#25030-024, Invitrogen). The cultivation takes place in standard cell culture bottles (# 353112, Becton Dickinson) in a cell culture incubator at 37°C in the presence of 5% CO2. The 80%-confluent cells are washed once with 15 ml of PBS (#14190-094, Invitrogen), treated with 3 ml of trypsin solution (#25300-054, Invitrogen) at 37°C for 2 min, taken up in 5 ml of the DMEM described and counted in a cell counter. After dilution to 500.000 cells/ml, 35,000 cells are seeded in each well of a 96 well microtiter plate with a clear plastic base
(#3610, Corning Costar). The plates are incubated in the cell culture incubator at 37°C and 5% CO2 for 24 h.
Day 2 PPAR delta agonists to be tested are dissolved in DMSO in a concentration of 10 mM.
This stock solution is diluted in DMEM (#41965-039, Invitrogen) which is mixed with 5% cs- FCS (#SH-30068.03, Hyclone), 2 mM L-glutamine (#25030-024, Invitrogen) and the previously described antibiotics (zeocin, G418, penicillin and streptomycin). Test substances are tested in 11 different concentrations in the range from 10 μM to 100 pM. More potent compounds are tested in concentration ranges from 1 μM to 10 pM or between 100 nM and 1 pM. The medium of the PPAR delta reporter cell line seeded on day 1 is completely removed by aspiration, and the test substances diluted in medium are immediately added to the cells. The dilution and addition of the substances is carried out by a robot (Beckman FX). The final volume of the test substances diluted in medium is 100 μl per well of a 96 well microtiter plate. The DMSO concentration in the assay is less than 0.1 % v/v in order to avoid cytotoxic effects of the solvent.
Each plate was charged with a standard PPAR delta agonist, which was likewise diluted in 11 different concentrations, in order to demonstrate the functioning of the assay in each individual plate. The assay plates are incubated in an incubator at 37°C and 5% CO2 for 24 h.
Day 3 The PPAR delta reporter cells treated with the test substances are removed from the incubator, and the medium is aspirated off. The cells are lyzed by pipetting 50 μl of Bright Glo reagent (from Promega) into each well of a 96 well microtiter plate. After incubation at room temperature in the dark for 10 minutes, the microtiter plates are measured in the luminometer (Trilux from Wallac). The measuring time for each well of a microtiter plate is 1 sec. Evaluation:
The raw data from the luminometer are transferred into a Microsoft Excel file. Dose- effect plots and EC50 values of PPAR agonists are calculated using the XL.Fit program as specified by the manufacturer (IDBS).
Determination of EC n values of PPAR agonists in the cellular PPAR alpha assay: Principle The potency of substances which bind to human PPAR alpha and activate it in an agonistic manner is analyzed using a stably transfected HEK cell line (HEK= human embryo kidney) which is referred to here as PPAR alpha reporter cell line. It contains two genetic elements, a luciferase reporter element (pdeltaM-GAL4-Luc-Zeo) and a PPAR alpha fusion protein (GR- GAL4-humanPPAR alpha-LBD) which mediates expression of the luciferase reporter element depending on a PPAR alpha ligand. The stably and constitutively expressed fusion protein GR-GAL4-humanPPAR alpha-LBD binds in the cell nucleus of the PPAR alpha reporter cell line via the GAL4 protein portion to the GAL4 DNA binding motifs 5 '-upstream of the luciferase reporter element which is stably integrated in the genome of the cell line. There is only weak expression of the luciferase reporter gene in the absence of a PPAR alpha ligand if fatty acid-depleted fetal calf serum (cs-FCS) is used in the assay. PPAR alpha ligands bind and activate the PPAR alpha fusion protein and thereby stimulate the expression of the luciferase reporter gene. The luciferase which is formed can be detected by means of chemiluminescence via an appropriate substrate.
Construction of the PPAR alpha reporter cell line:
The PPAR alpha reporter cell line was prepared in two stages. Firstly, the luciferase reporter element was constructed and stably transfected into HEK cells. For this purpose, five binding sites of the yeast transcription factor GAL4 (Accession # AF264724) were cloned in 5 '-upstream of a 68 bp-long minimal MMTV promoter (Accession # V01175). The minimal MMTV promoter section contains a CCAAT box and a TATA element in order to enable efficient transcription by RNA polymerase II. The cloning and sequencing of the GAL4- MMTN construct took place in analogy to the description of Sambrook J. et. al. (Molecular cloning, Cold Spring Harbor Laboratory Press, 1989). Then the complete Photinus pyralis gene (Accession # Ml 5077) was cloned in 3 '-downstream of the GAL4-MMTN element. After sequencing, the luciferase reporter element consisting of five GAL4 binding sites, MMTV promoter and luciferase gene was recloned into a plasmid which confers zeocin resistance in order to obtain the plasmid pdeltaM-GAL4-Luc-Zeo. This vector was transfected into HEK cells in accordance with the statements in Ausubel, F.M. et al. (Current protocols in molecular biology, Vol. 1-3, John Wiley & Sons, Inc., 1995). Then zeocin-containing medium (0.5 mg/ml) was used to select a suitable stable cell clone which showed very low basal expression of the luceriferase gene. In a second step, the PPAR alpha fusion protein (GR-GAL4-humanPPAR alpha-LBD was introduced into the stable cell clone described. For this purpose, initially the cDΝA coding for the Ν-terminal 76 amino acids of the glucocorticoid receptor (Accession # P04150) was linked to the cDΝA section coding for amino acids 1-147 of the yeast transcription factor GAL4 (Accession # P04386). The cDΝA of the ligand-binding domain of the human PPAR alpha receptor (amino acids S167-Y468; Accession # S74349) was cloned in at the 3'-end of this GR-GAL4 construct. The fusion construct prepared in this way (GR-GAL4-humanPPAR alpha-LBD) was recloned into the plasmid pcDΝA3 (Invitrogen) in order to enable constitutive expression therein by the cytomegalovirus promoter. This plasmid was linearized with a restriction endonuclease and stably transfected into the previously described cell clone containing the luciferase reporter element. The finished PPAR alpha reporter cell line which contains a luciferase reporter element and constitutively expresses the PPAR alpha fusion protein (GR-GAL4-human PPAR alpha-LBD) was isolated by selection with zeocin (0.5 mg/ml) and G418 (0.5 mg/ml).
Assay procedure:
The activity of PPAR alpha agonists is determined in a 3-day assay, which is described below:
Day 1 The PPAR alpha reporter cell line is cultivated to 80% confluence in DMEM (# 41965-039, Invitrogen) which is mixed with the following additions: 10% cs-FCS (fetal calf serum; #SH-30068.03, Hyclone), 0.5 mg/ml zeocin (#R250-01, Invitrogen), 0.5 mg/ml G418 (#10131-027, Invitrogen), 1% penicillin-streptomycin solution (#15140-122, Invitrogen) and 2 mM L-glutamine (#25030-024, Invitrogen). The cultivation takes place in standard cell culture bottles (# 353112, Becton Dickinson) in a cell culture incubator at 37°C in the presence of 5% CO2. The 80%-confluent cells are washed once with 15 ml of PBS (#14190-094, Invitrogen), treated with 3 ml of trypsin solution (#25300-054, Invitrogen) at 37°C for 2 min, taken up in 5 ml of the DMEM described and counted in a cell counter. After dilution to 500.000 cells/ml, 35,000 cells are seeded in each well of a 96 well microtiter plate with a clear plastic base (#3610, Corning Costar). The plates are incubated in the cell culture incubator at 37°C and 5% CO2 for 24 h.
Day 2 PPAR alpha agonists to be tested are dissolved in DMSO in a concentration of 10 mM. This stock solution is diluted in DMEM (#41965-039, Invitrogen) which is mixed with 5% cs- FCS (#SH-30068.03, Hyclone), 2 mM L-glutamine (#25030-024, Invitrogen) and the previously described antibiotics (zeocin, G418, penicillin and streptomycin).
Test substances are tested in 11 different concentrations in the range from 10 μM to 100 pM. More potent compounds are tested in concentration ranges from 1 μM to 10 pM or between 100 nM and 1 pM. The medium of the PPAR alpha reporter cell line seeded on day 1 is completely removed by aspiration, and the test substances diluted in medium are immediately added to the cells. The dilution and addition of the substances is carried out by a robot (Beckman FX). The final volume of the test substances diluted in medium is 100 μl per well of a 96 well microtiter plate. The DMSO concentration in the assay is less than 0.1 % v/v in order to avoid cytotoxic effects of the solvent. Each plate was charged with a standard PPAR alpha agonist, which was likewise diluted in 11 different concentrations, in order to demonstrate the functioning of the assay in each individual plate. The assay plates are incubated in an incubator at 37°C and 5% CO2 for 24 h.
Day 3 The PPAR alpha reporter cells treated with the test substances are removed from the incubator, and the medium is aspirated off. The cells are lyzed by pipetting 50 μl of Bright Glo reagent (from Promega) into each well of a 96 well microtiter plate. After incubation at room temperature in the dark for 10 minutes, the microtiter plates are measured in the luminometer (Trilux from Wallac). The measuring time for each well of a microtiter plate is 1 sec.
Evaluation:
The raw data from the luminometer are transferred into a Microsoft Excel file. Dose- effect plots and EC50 values of PPAR agonists are calculated using the XL.Fit program as specified by the manufacturer (IDBS). Determination of EC50 values of PPAR agonists in the cellular PPAR gamma assay
Cell based PPAR gamma Assay protocol
To perform cell based assays a luciferase assay is performed in 96 well plates as follows:
Day 1: Plating of cells:
• Wash cells grown to 80-90% confluency once in PBS
• Trypsinize for 2 min • Add 15ml assay medium (DMEM, Invitrogen, Cat.No. 41965-039; 5% Charcoal/Dextran Treated FBS, Hyclone, Cat.No. SH30068; 0.5 mg/ml Zeocin, Invitrogen, Cat.No. 46-0072; 0.5mg/ml Geneticin, Invitrogen, Cat.No. 10131-027; 1% Penicillin/Streptomycin, Invitrogen, Cat.No. 15140-122; 2 mM L-Glutamine, Invitrogen, Cat.No. 25030-024; 7.5 μg/ml Blasticidin S HC1, Invitrogen, Cat.No. R210-01; 1 μg/ml Doxycycline, Clontech, Cat.No. 8634-1)
• Count cells
• Dilute cells in assay medium to 500.000 cells/ml
• Dispense 100 μl of cell suspension per well in clear bottom Corning plates (makes 50.000 cells/well) • Incubate for 24 h at 37°C, 5% CO2
Day 2: Dosing with test compounds: • Solve test compounds in DMSO to make a 10 mM stock solution
• Dilute compound to appropriate concentration in assay medium (DMEM, Invitrogen, Cat.No. 41965-039; 5% Charcoal/Dextran Treated FBS, Hyclone, Cat.No. SH30068; 0.5mg/ml Zeocin, Invitrogen, Cat.No. 46-0072; 0.5mg/ml Geneticin, Invitrogen, Cat.No. 10131-027; 1% Penicillin/Streptomycin, Invitrogen, Cat.No. 15140-122; 2mM L- Glutamin, Invitrogen, Cat.No. 25030-024; 7.5μg/ml Blasticidin S HCl, Invitrogen, Cat.No. R210-01; lμg/ml Doxycycline, Clontech, Cat.No. 8634-1) (regular FCS is harboring\ free fatty acids interfering with the PPAR ligand binding domains).
• Aspirate medium (cells are quite sensitive at this step; make sure that cells are no longer than 1 min without being covered by medium)
• Transfer diluted compounds to 96 wells (100 μl medium including compound)
• Make controls with standard compound (e.g. Rosiglitazon) as well as a DMSO control (0,1 % DMSO)
• Incubate cells for 24 h at 37°C at 5% CO2
Dilution steps and addition of diluted compounds is done using a Beckman Biomek 2000 or Beckman FX robot.
Day 3: Cell Lysis and measurement of luciferase activity:
• Aspirate medium from cells
• Freeze plates at -20°C (optional)
• Thaw plates for 30 min (if necessary)
• Add 50 μl Bright-Glo-Luciferase Assay Reagent (Promega, Cat.No. E2650) • Incubate for 10 min in the dark
• Measure luminescence 2 sec per well (Wallac Microbeta)
Data analysis:
Determination of EC50 values is done with Microsoft Exel in combination with XLFit (develop by IDBS) using the fitting algorithm #205. Determination of EC50 values in the cellular human RXR receptor assay Cell based RXR Assay protocol
To perform cell based assays a luciferase assay is performed in 96 well plates as follows: Day 1: Plating of cells
• Wash cells grown to 80-90% confluency once in PBS
• Trypsinize for 2 min
• Add 15ml culture medium (DMEM, Invitrogen, Cat.No. 41965-039; 10% Charcoal/Dextran Treated FBS, Hyclone, Cat.No. SH30068; 0.5mg/ml Zeocin, Invitrogen, Cat.No. 46-0072; 0.5mg/ml Geneticin, Invitrogen, Cat.No. 10131-027; 1% Penicillin/Streptomycin, Invitrogen, Cat.No. 15140-122; 2mM L-Glutamin, Invitrogen, Cat.No. 25030-024)
• Count cells • Dilute cells in culture medium to 175.000 cells/ml
• Dispense 200 μl of cell suspension per well in clear bottom Corning plates (makes 35.000 cells/well)
• Incubate for 24 h at 37°C, 5% CO2 Day 2: Dosing with test compounds
• Solve test compounds in DMSO to make a 10 mM stock solution
• Dilute compound to appropriate concentration in assay medium (DMEM w/o phenol-red, Invitrogen, Cat.No. 21063-029; 5% Charcoal/Dextran Treated FBS, Hyclone, Cat.No. SH30068; 0.5mg/ml Zeocin, Invitrogen, Cat.No. 46-0072; 0.5mg/ml Geneticin, Invitrogen, Cat.No. 10131-027; 1% Penicillin/Streptomycin, Invitrogen, Cat.No. 15140- 122; 2mM L-Glutamin, Invitrogen, Cat.No. 25030-024) (regular FCS is harboring free fatty acids interfering with the PPAR ligand binding domains).
• Aspirate medium (cells are quite sensitive at this step; make sure that cells are no longer than 1 min without being covered by medium) • Transfer diluted compounds to 96 wells (100 μl medium including compound)
• Make controls with standard compound (e.g. RPR258134) as well as a DMSO control (0,1 % DMSO)
• Incubate cells for 24 h at 37°C at 5% CO2 Dilution steps and addition of diluted compounds is done using a Beckman Biomek 2000 or Beckman FX robot.
Day 3: Cell Lysis and measurement of luciferase activity
• Aspirate medium from cells
• Freeze plates at -20°C (optional)
• Thaw plates for 30 min (if necessary)
• Add 50 μl Bright-Glo-Luciferase Assay Reagent (Promega, Cat.No. E2650) • Incubate for 10 min in the dark
• Measure luminescence 2 sec per well (Wallac Microbeta)
Data analysis
Determination of EC50 values is done with Microsoft Exel in combination with XLFit (develop by IDBS) using the fitting algorithm #205.
Table 1 shows the results if the reporter assays. The results show that compounds 1 and 2 are selective PPAR delta activators with low PPAR alpha, gamma and RXR activity.
Table 1 Reporter Assavs
Human Mouse Human Human Human PPAR PPAR delta PPAR delta PPAR PPAR RXR1
Compound EC5o (μM) ECso (μM) alpha gamma EC50 (μM) EC50 (μM) EC50 (μM) 1 < 0.00457 7.8 (4x)*, 1.3 3.97 (3. lx)* No increase (29x)* 6 (2x)*, 4(2x)* 2 0.039 1 (2x)* 1.1 1.1 (4.3x)* No increase (27x)*
*Value represents the fold increase over baseline luciferase activity. Retinoid X receptor RAT/MICE Oligodendrocyte cultures
Preparation of cells: 1. Primary rat oligodendrocyte progenitor cells are obtained from the neocortex of newborn (postnatal days 2-3) rats or mice and are enriched, after removal of microglia, by mechanical separation from the astrocytic monolayer using a modification of the technique originally described by McCarthy and de Vellis (1980). 2. Remove the meninges from neonatal rat brain and mechanically dissociate tissue. Plate cells on T75 flasks and feed cells with DMEM/F12 + 10% FBS. 3. Collect oligodendrocytes growing on the astrocyte bed layer by shaking-off method fourteen days after the original prep date. Centrifuge the suspension and resuspend the cell pellet in serum free media (SFM; DMEM combined with 25 μg/ml transferring, 30 nM triiodothyronine, 20 nM hydrocortisone, 20 nM progesterone, 10 nM biotin, lx trace elements, 30 nM selenium, 1 μg/ml putrescine, 0.1% BSA, 5 U/ml PenStrep, 10 μg/ml insulin) supplemented with the following growth factors: Platelet derived growth factor-AA (PDGF) and fibroblast growth factor-2 (FGF). 4. Plate the cells on PDL-coated dishes and incubate at 37°C with 6-7% CO2.
5. Media components are replaced every 48 hr to keep the cells in a progenitor state.
Progenitor cell passaging to increase cell numbers for screening assays: 1. When the culture are confluent, rinse the culture with PBS, add trypsin and incubate for -2-3 min at 37°C. 2. Neutralize and centrifuge the cell suspension at 900g for 5 min. 3. Resuspend the cell pellet in SFM + PDGF/FGF. 4. Feed the cells with fresh growth factors every 48 hrs to keep enrich for rapidly dividing progenitor cells. 5. Cells are passaged no more than 4-5 times prior to experimental assays. 6. All experiments involving oligodendrocyte progenitor cells were done using cells that were continuously maintained under these conditions. Greater than 95% of all cells were A2B5 immunopositive and expressed 2' 3' -cyclic nucleotide 3' - phosphodiesterase II mRNA. 7. To generate mature oligodendrocytes, 24 h after plating progenitor cells were switched to SFM supplemented with or without IGF-I and grown under these conditions for 7 d prior to experimental assays. 8. Alternatively, the enriched rat Central Glia-4 (CG4) progenitor cell line may be used, which is maintained in base media (DMEM, with 2 mM glutamine, lmM sodium pyruvate, biotin (40 nM), insulin (1 μM) and NI) supplemented with 30% conditioned media from the B-104 neuroblastoma cell line. To induce differentiation, CG4 cells are switched to base media with 1% fetal calf serum (removed after 2 days) and insulin (500 nM). A2B5 and MBP immunoreactivity is used to confirm >95% enrichment in immature and mature cultures, respectively. Rat/Mouse Culture Compound Treatment: 1. Put 10,000 - 15,000 cells /well in 24-well PDL coated plates and culture the cells in presence of mitogen (10 ng/ml) overnight. 2. In the presence of mitogen: a. Next day, remove the old medium and add compounds in fresh medium (with mitogen) b. Compound dose response evaluations are performed at 6 different concentrations (10 μM, 1 μM, 100 nM, 10 nM, 1 nM, and 0.1 nM); c. Triplicates wells are run for each compound concentration. 3. In the absence of mitogen: a. Next day, remove the old medium and add compounds in fresh medium (without mitogen) b. Compound dose response evaluations are performed at 6 concentrations (10 μM, 1 μM, 100 nM, 10 nM, 1 nM, and 0.1 nM); c. Triplicates wells are run for each compound concentration. 4. Culture the treated cells for 7 d prior to using in experimental assays.
HUMAN Oligodendrocyte cultures Preparation of cells: 1. Human neurospheres collected from E19.5 - E22 human embryo cortex) are cultured for 2 weeks in progenitor media: DMEM/F12 containing 100 μg/ml transferring, 30 nM triiodothyronine, 20 nM hydrocortisone, 20 nM progesterone, 10 nM biotin, lx trace elements, 30 nM selenium, 60 uM putrescine, 0.1% BSA, 5 U/ml PenStrep, 25 μg/ml insulin) supplemented with PDGF and FGF. 2. Neurospheres are dissociated with 20 U/ml papain at 37°C for 30-50 min. 3. Cells are plated onto PDL coated dishes at density of 50,000-100,000 cell/well in progenitor media containing PDGF/FGF and incubated at 37°C with 5-6% CO2.
4. Media and growth factors are replenished every 48 hr.
Human Culture Compound Treatment: 1. 24 to 48 hr after plating remove the old medium and add compounds in fresh medium (with mitogen) 2. Compound dose response evaluations are performed at 3-6 different concentrations (10 μM, 1 μM, 100 nM, 10 nM, 1 nM, and 0.1 nM)
3. Triplicates wells are run for each compound concentration. 5. Culture the treated cells for 7 d prior to using in experimental assays.
RAT/MOUSE/HUMAN Oligodendrocyte Specific Immunostaining: Following compound exposure, oligodendrocyte-specific antibodies are used to assess ability of compound to accelerate/promote oligodendrocyte differentiation (for example, O4, OI, or myelin basic protein immunoreactivity is over time between compound treated and untreated cultures). 1. Cells are plated onto poly-D-lysine treated 4-well chamber slides at 5xl03 to 20xl03 cells/well and grown as described above. Sequential staining is performed on oligodendrocyte populations with increasing degrees of cellular differentiation, as determined by days in vitro without PDGF and FGF. 2. Live staining for 30 min at 37°C is used to detect oligodendrocyte stage specific cell surface marker expression (including A2B5, O4, and OI). 3. Subsequently, cells are fixed with 4% paraformaldehyde, 10 min, room temperature. 4. Fixed staining procedures are used to detect oligodendrocyte stage specific marker expression (including myelin basic protein, MBP). 5. Rinse with PBS. 6. Permeabilize with 0.1% Triton/0.01% NaAz diluted in IX PBS for 10 min, room temperature. 7. Block with 5-10% goat serum in antibody dilution buffer (0.1% Triton-X 100 and 1% IgG-free bovine serum albumin; also used to dilute antibodies), 15 min, room temperature. 8. Add primary antibody diluted in antibody dilution buffer. 9. Incubate overnight, gently rocking, 4° C. 10. Next day, rinse with PBS IX 5 min, followed by 3X 15 min each, room temperature. 11. Incubate with appropriate secondary antibodies, 45 min, room temperature. 12. Cell nuclei are stained with 4,6-diamidino-2-phenylindole (DAPI), 15min, room temperature. 13. Rinse several times with PBS and evaluate using fluorescent microscopy. 14. The following conditions are compared over time and at different compound doses: PDGF/FGF alone, SFM alone, SFM-IGFl alone, PDGF/FGF and compound, SFM and compound.
RAT/MOUSE/HUMAN Bromodeoxyuridine (BrdU) immunostaining: To confirm that compounds do not promote cell proliferation.
1. Oligodendrocyte progenitor cells are labeled with 10 μM BrdU for 20 hr and then fixed with either 70% ethanol or 4% paraformaldehyde. 2. The cells are incubated successively with biotinylated mouse anti-BrdU and Streptavidin-Peroxidase, with three intervening washes with PBS. 3. Colormetric visualization of the BrdU immunoreactivity is developed with DAB and total cell numbers are assessed using the counter-stain hematoxylin. 4. BrdU immunopositive cells are counted by two independent observers.
RAT/MOUSE/HUMAN Culture Image analysis: Fluorescent microscopy is used to quantitate the extent of oligodendrocyte differentiation after compound exposure. This assay demonstrates that selective agonists accelerate/promote oligodendrocytes differentiation. 1. Manual Cell Counting: Four fields are randomly selected for each experimental condition and 500-600 cells are counted in each field. The percentage of MBP (or O4) immunpositive cells (mature process bearing cells with or without myelin sheets) versus DAPI positive cells (total cell number) cells are compared in the control and drug-treated groups. 2. Automated Cell Counting: Fluorescent microscopy was used to quantitate the extent of oligodendrocyte differentiation after compound exposure. Six fields/well are randomly selected to assess the number of differentiating oligodendrocytes among the total population (~8 to 15x10 cells are counted/well). Immunofluorescence images are obtained using a Zeiss AxioVision digital imaging system, with a Zeiss AxioCam HRc cooled CCD camera connected to the same microscope. All microscopic imaging parameters are set for acquiring images for the analysis of cellular immunofluorescence intensity. The percentage of MBP positive (differentiated) cells versus total cells (DAPI nuclear stained) is compared in the control versus drug-treated groups. Cellular autofluorescence was undetectable under the imaging conditions, a) 3. Human oligodendrocyte differentiation assay: manually count total number of O4 immunopositive cells/well (bipolar and multipolar).. The results using rat oligodendrocyte cultures are shown in Figure 1 and the results using human oligodendrocyte mixed cultures are shown in Figures 5 and 6. As the results show, PPAR delta agonists enhance or accelerate rat and human oligodendrocyte differentiation, as measured by increased myelin basic protein expression compared to untreated controls. This novel finding would suggest that compound 1 and compound 2 and selective PPAR delta agonists in general would be enhance, accelerate, or stimulate oligodendrocyte differentiation and myelin formation in vivo, in the diseased or injured CNS, including MS and other demyelinating disorders.
RAT/MOUSE/HUMAN Quantitative Polvmerase Chain Reaction (PCR); To evaluate compound induced PPAR delta pathway activation and the extent of oligodendrocyte maturation (changes in mRNA levels). 1. Total RNA is extracted from cultured oligodendrocytes using TriZol reagent. 2. Subsequently, mRNA is treated with RNase-free DNase, repurified, and then converted to cDNA template using a RT reaction (Clontech Advantage RT for PCR Kit). 3. PPAR delta pathway member transcript expression is quantitated using Sybr Green PCR Master Mix. 4. The 18S ribosomal RNA primer/probe mix (186 bp product), suspended in Taqman 2X PCR Master Mix is used as an internal control. 5. Quantitative PCR is carried out using real-time Taqman™ technology (Gibson, et al., 1996) with a model 7700 Sequence Detector System (Applied Biosystems, Foster City, CA). 6. The results are analyzed using Sequence Detection Systems software version 1.91. Results for these assays are shown in Figures 2, 3, 4A, and 4B. These results suggest that PPAR delta selective agonists bind the PPAR delta receptor and directly activate the PPAR delta pathway in oligodendrocytes and should act similarly in vivo. RAT ELISA Assay: To evaluate compound induced PPAR delta pathway activation and the extent of oligodendrocyte maturation (changes in protein levels).
1. Plates are washed with PBS, and then keep on ice. Add 200 μl ice old lysis buffer (Tris 50mM, pH7.4, MgCI2 2mM, EDTA ImM, β-mercaptoethanol 5mM, Nonidet P-40 1%, Protease inhibitor cocktail (Roche): 1 tablet/50 ml) to each well.
2. Lyse cells by using pipette to up down and spin plates at 2000 rpm at 4°C for 5 min. The supernatant is ready to use.
3. Pipet 50 μl of standard, controls and samples to the wells.
4. Add 50 μl of MBP Assay Buffer to each well.
5. Incubate the well, shaking at 500-700 rpm on orbital microplate shaker for 2 hr at room temperature. 6. Add lOOul of the MBP Antibody-Biotin Conjugate to each well.
7. Incubate the well, shaking at 500-700 rpm on orbital microplate shaker forl hr at room temperature.
8. Wash well 5 times with Wash Solution. Blot dry by inverting the plate on absorbent material. 9. Dilute the streptavi din-enzyme conjugate concentrate 1:50 with MBP Elisa Assay buffer, (must be diluted immediately prior to use in the assay).
10. Add 100 μl streptavidin-enzyme conjugate solutions to each well.
11. Incubate the well, shaking at 500-700 rpm on orbital microplate shaker for 30 min at room temperature. 12. Wash well 5 times with the Wash Solution. Blot dry by inverting the plate on absorbent material.
13. Add 100 μl of TMB Chromogen Solution to each well.
14. Incubate the well, shaking at 500-700 rpm on orbital microplate shaker for 10-20 min at room temperature. Avoid exposure to direct sunlight.
15. Add 100 μl of the Stopping Solution to each well.
16. Read the absorbance of the solution in the wells within 30 min, using a microplate reader set to 450 nM.
The above results taken in general and shown in Figures 1-6 illustrate that PPAR delta agonists promote oligodendrocyte differentiation even in the presence of mitogens, which normally keep cells mitotically active and inhibit cellular differentiation. Thus, it is expected that in the injured or diseased CNS selective PPAR delta agonists will cause dividing oligodendrocyte progenitor cells to express myelin proteins and ensheath demyelinated or hypomyelinated axons.
In Vivo Proof of Concept Models Focal Lesions: (used to assess whether compounds protect myelin integrity or accelerate/enhance the rate of remyelination.) 1. Rats 7 weeks of age are given free access to food and water and acclimatized for a minimum of 4 days before use in experiments. 2. Prior to surgery each animal is weighed. The rat is then anaesthetized with ketamine (100 mg/ml) in combination with xylazine (20 mg/ml) in a ratio of 1.8 : 1. The rats are injected with 0.15ml/180g body weight i.p. of the anaesthetic solution prior to the surgical procedure. The animal is prepared for surgery using aseptic conditions in accordance with the IACUC guidelines. All surgical instruments will be autoclaved. The hair is clipped between the ears and this region will then be scrubbed with Betadine, flushed with sterile saline and finally wiped with a pre-packaged sterile alcohol swab. 3. For the surgical procedure, the rat is placed on its ventral surface in a small animal stereotaxic instrument designed to hold the head steady. The incisor bar is always set at -3.9 mm, since this has been shown to achieve a flat-skull position for SD rats. 4. An incision is made in the previously shaven skin overlying the skull between the ears. 5. A small area of bone (0.75mm in diameter) is drilled at the following coordinates AP - 1.8, ML -3.1 from lambda.
6. The bone is removed and rats are injected with 2μl ethidium bromide, lysolecithin, or SIN-1 into the right caudal cerebellar peduncle, DV -7.1 mm, over a 2 min period by means of a Hamilton μl syringe and needle. Alternatively injections are made into the spinal cord, corpus callosum, or cortex. 7. The needle is left in position for the subsequent 2 min. 8. After withdrawal of the needle the incision is sutured. 9. Each rat receives an i.m. injection of 0.003mg buprenorphine into a hind leg. 10. The rat is placed in a warming cupboard until it regains consciousness. At which time it is returned to its home cage. Do not allow more than 2 rats per cage, as they will pull each other's suture out. 11. Similar procedures are also done using mice.
Rat Experimental Allergic Encephalomyelitis (Rat EAE) Disease Model: Experimental allergic encephalomyelitis (EAE) is a T-cell-mediated autoimmune disease of the nervous system that develops in susceptible animals following sensitization with either whole spinal cord homogenate or a component (myelin basic protein). The EAE rodent model is an appropriate tool for studying the inflammation of the brain and spinal cord observed in MS patients. In rodents, injection of whole spinal cord or spinal cord components such as myelin basic protein induces an autoimmune response based on the activation of T- lymphocytes. Clinical disease typically becomes manifest around day 8-10 after inoculation, observed as a broad spectrum of behavioral anomalies ranging from mild gait disturbances and tail atony to complete paralysis and death. Weight loss typically occurs. In animals that survive, spontaneous recovery occurs, accompanied by variable recovery of most motor function. Depending on the species, allergen, and methodology used, animals tested by the EAE model may experience a single (acute EAE) or several (chronic relapsing EAE) attacks. Several treatment paradigms may be used: the drug or treatment of choice may be administered before immunization, during the nonsymptomatic period or during the clinical disease.
Animals: Female Lewis rats, 160-220g (Charles River) Antigen: Whole Guinea Pig spinal cord (Harlan Biosciences). Complete Freund's adjuvant H37 Ra [lmg/ml Mycobacterium Tuberculosis H37 Ra] (Difco).
Additional antigen: Mycobacterium Tuberculosis (Difco). Bordetella Pertussis [Heat Killed] (Difco). Antigen preparation: (for approximately 720 animals): 1. Weigh 5 grams of frozen guinea pig spinal cord.
2. Add 5g spinal cord to 5ml 0.9% saline (lg/ml) in a round bottom centrifuge tube
3. Homogenize on ice with the Tissue-tech until the tissue is completely disrupted (approximately 5 minutes).
4. Add 10 ml Complete Freund's adjuvant H37 Ra supplemented with 200 mg Mycobacterium Tuberculosis (20 mg / ml Complete Freund's adjuvant H37 Ra).
5. Extract homogenate / adjuvant from tube by sucking it into a 10 ml syringe fitted with an 18 gauge emulsifying needle. 6. Emulsify between two 30 ml glass syringes until it becomes difficult to continue passing the material through the needle. (Approximately 5 minutes {there must be no separation between the oil phase and the aqueous phase}).
7. Use immediately or keep on ice until needed (not more than 30 min) (do not freeze).
Protocol
1. Female Lewis rats (Charles River) are given free access to food and water and should be acclimated a minimum of 3 days before use in experiments.
2. Rats weighing 160 and 220 grams are initially induced with 5% isoflurane (Aerrane, Fort Dodge), 30% O2, 70% N2O for 2-5 minutes.
3. The rat is then placed onto a circulating water heating blanket (Gaymar) (dorsal surface up) and into the nose cone for spontaneous respiration of anesthetic gases. The isoflurane is reduced to 2%.
4. Two subcutaneous injections (0.1 ml each) of either antigen or normal saline are made into ventral surface of the hind paws.
5. The animals are removed from the nose cone, weighed and numbered.
6. The rats are allowed to awake from anesthesia and are placed into individual cages.
7. The animals are observed daily for signs of EAE induction (see criteria below)
STAGE:0 NORMAL
STAGE 1 Abnormal gate and tail atony STAGE 2 Mild but definite weakness of one or both hind legs STAGE: 3 Severe weakness of one or both hind legs or mild ataxia STAGE: 4 Severe paraparesis and minimal hind leg movement STAGE: 5 No hind leg movement and paraplegia
STAGE: 6 Moribund state with no spontaneous movement and impaired respiration. Increasing degree of front leg involvement and urinary and fecal incontinence may also occur STAGE:7 DEATH Treatment is begun on day 10 after immunization. Since the disease symptoms in this model typically appear 10-11 days after inoculation, this time point may be considered to represent the initial phase of an acute episode of MS. It is judged that this delay of the start of treatment mimics the clinical situation more closely than the traditionally used protocols where drugs are administered at the time of, or even before, inoculation (Teitelbaum D. et al., Proc Natl Acad Sci USA 1999; 96: 3842-3847 and Brod S. A., et al., Ann Neurol 2000; 47: 127-131).

Claims

CLAIMSWhat is claimed is:
1. A method for the treatment of demyelinating diseases in a patient comprising the administration of a therapeutically effective amount of a hPPAR delta agonist.
2. The method according to claim 1 wherein the hPPAR delta agonist is a selective agonist.
3. The method according to claim 1 wherein said demyelinating disease is selected form the group consisting of multiple sclerosis, Charcot-Marie-Tooth disease, Pelizaeus- Merzbacher disease, encephalomyelitis, neuromyelitis optica, adrenoleukodystrophy, Guillian-Barre syndrome and disorders in which myelin forming glial cells are damaged including spinal cord injuries, neuropathies and nerve injury.
4. The method according to claim 3 wherein the demyelinating disease is multiple sclerosis.
5. The method according to claim 1 wherein the agonist is selected from the group consisting of compound of formula (1) and formula (2)
Figure imgf000036_0001
(1)
Figure imgf000037_0001
(2).
6. A pharmaceutical composition comprising a compound selected from the group consisting of compound of formula (1) and formula (2) in an amount effective for treating multiple sclerosis, Charcot-Marie-Tooth disease, Pelizaeus-Merzbacher disease, encephalomyelitis, neuromyelitis optica, adrenoleukodystrophy, Guillian-Barre syndrome and disorders in which myelin forming glial cells are damaged including spinal cord injuries, neuropathies and nerve injury in combination with at least on pharmaceutically acceptable carrier
Figure imgf000037_0002
(1)
Figure imgf000037_0003
(2).
7. The pharmaceutical composition according to claim 6 comprising an amount effective for treating multiple sclerosis.
PCT/US2005/010371 2004-04-01 2005-03-29 Use of ppr delta agonists for treating demyelinating diseases WO2005097098A2 (en)

Priority Applications (9)

Application Number Priority Date Filing Date Title
AU2005231358A AU2005231358A1 (en) 2004-04-01 2005-03-29 Use of PPR delta agonists for treating demyelinating diseases
CA002561159A CA2561159A1 (en) 2004-04-01 2005-03-29 Use of peroxisome proliferator activated receptor delta agonists for the treatment of ms and other demyelinating diseases
BRPI0509540-9A BRPI0509540A (en) 2004-04-01 2005-03-29 use of peroxisome proliferator-activated delta receptor agonists for the treatment of ms and other demyelinating diseases
EP05729215A EP1737440A2 (en) 2004-04-01 2005-03-29 Use of ppar delta agonists for treating demyelinating diseases
JP2007506442A JP2007530703A (en) 2004-04-01 2005-03-29 Use of peroxisome proliferator-activated receptor delta agonists to treat MS and other demyelinating diseases
MXPA06011218A MXPA06011218A (en) 2004-04-01 2005-03-29 Use of ppr delta agonists for treating demyelinating diseases.
IL178165A IL178165A0 (en) 2004-04-01 2006-09-18 Use of ppr delta agonists for treating demyelinating diseases
US11/535,240 US20070149580A1 (en) 2004-04-01 2006-09-26 Use of peroxisome proliferator activated receptor delta agonists for the treatment of ms and other demyelinating diseases
NO20064985A NO20064985L (en) 2004-04-01 2006-10-31 Use of peroxisome proliferator-activated receptor delta diagnostics for the treatment of MS and other demyelinating diseases

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US55853304P 2004-04-01 2004-04-01
US60/558,533 2004-04-01

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US11/535,240 Continuation US20070149580A1 (en) 2004-04-01 2006-09-26 Use of peroxisome proliferator activated receptor delta agonists for the treatment of ms and other demyelinating diseases

Publications (2)

Publication Number Publication Date
WO2005097098A2 true WO2005097098A2 (en) 2005-10-20
WO2005097098A3 WO2005097098A3 (en) 2005-12-22

Family

ID=34977094

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2005/010371 WO2005097098A2 (en) 2004-04-01 2005-03-29 Use of ppr delta agonists for treating demyelinating diseases

Country Status (16)

Country Link
US (1) US20070149580A1 (en)
EP (1) EP1737440A2 (en)
JP (1) JP2007530703A (en)
KR (1) KR20060134191A (en)
CN (1) CN1950077A (en)
AU (1) AU2005231358A1 (en)
BR (1) BRPI0509540A (en)
CA (1) CA2561159A1 (en)
IL (1) IL178165A0 (en)
MA (1) MA28561B1 (en)
MX (1) MXPA06011218A (en)
NO (1) NO20064985L (en)
RU (1) RU2006138495A (en)
SG (1) SG138623A1 (en)
WO (1) WO2005097098A2 (en)
ZA (1) ZA200607850B (en)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007003581A1 (en) 2005-06-30 2007-01-11 Novo Nordisk A/S Phenoxy acetic acids as ppar delta activators
US7943613B2 (en) 2005-12-22 2011-05-17 High Point Pharmaceuticals, Llc Compounds, their preparation and use
US7943612B2 (en) 2006-03-09 2011-05-17 High Point Pharmaceuticals, Llc Compounds that modulate PPAR activity, their preparation and use
US7968723B2 (en) 2004-05-05 2011-06-28 High Point Pharmaceuticals, Llc Compounds, their preparation and use
US8053598B2 (en) 2004-05-05 2011-11-08 High Point Pharmaceuticals, Llc Compounds, their preparation and use
US8329725B2 (en) 2008-06-09 2012-12-11 Sanofi Annelated pyrrolidin sulfonamides with oxadiazolone headgroup, processes for their preparation and their use as pharmaceuticals
US8716317B2 (en) 2008-06-09 2014-05-06 Sanofi Sulfonamides with heterocycle and oxadiazolone headgroup, processes for their preparation and their use as pharmaceuticals
US10456406B2 (en) 2013-09-09 2019-10-29 Vtv Therapeutics Llc Use of a PPAR-δ agonist for reducing loss of muscle strength, muscle mass, or type I muscle fibers in an immobilized limb
US11931365B2 (en) 2022-01-25 2024-03-19 Reneo Pharmaceuticals, Inc. Use of PPAR-delta agonists in the treatment of disease

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2630816C (en) * 2005-11-28 2014-08-12 Senju Pharmaceutical Co., Ltd. Pharmaceutical comprising ppar agonist
AU2010300531A1 (en) 2009-09-30 2012-05-24 President And Fellows Of Harvard College Methods for modulation of autophagy through the modulation of autophagy-inhibiting gene products
SG181509A1 (en) 2009-12-17 2012-07-30 Sanofi Sa Animal model expressing luciferase under control of the myelin basic protein promoter (mbp-luci) and use of the model for bioluminescence in vivo imaging
EP2675893B1 (en) * 2011-02-18 2019-01-09 The Scripps Research Institute Directed differentiation of oligodendrocyte precursor cells to a myelinating cell fate

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997028149A1 (en) * 1996-02-02 1997-08-07 Merck & Co., Inc. Method for raising hdl cholesterol levels
WO2001000603A1 (en) * 1999-06-25 2001-01-04 Glaxo Group Limited Thiazole and oxazole derivatives and their pharmaceutical use
WO2002013812A1 (en) * 2000-08-17 2002-02-21 Pershadsingh Harrihar A Methods for treating inflammatory diseases
WO2002028434A2 (en) * 2000-10-05 2002-04-11 Glaxo Group Limited Use of a ppar delta activator for treating inflammatory conditions
WO2002028433A2 (en) * 2000-10-05 2002-04-11 Glaxo Group Limited Use of a ppar delta activator in the treatment of diseases related to no inhibit ion or tnf inhibition
WO2004093910A1 (en) * 2003-04-22 2004-11-04 Astellas Pharma Inc. REMEDY FOR CEREBRAL NEURODEGENERATIVE DISEASES USING PPARδ AGONIST

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050020654A1 (en) * 2003-03-15 2005-01-27 Pershadsingh Harrihar A. Novel PPAR agonists, pharmaceutical compositions and uses thereof

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997028149A1 (en) * 1996-02-02 1997-08-07 Merck & Co., Inc. Method for raising hdl cholesterol levels
WO2001000603A1 (en) * 1999-06-25 2001-01-04 Glaxo Group Limited Thiazole and oxazole derivatives and their pharmaceutical use
WO2002013812A1 (en) * 2000-08-17 2002-02-21 Pershadsingh Harrihar A Methods for treating inflammatory diseases
WO2002028434A2 (en) * 2000-10-05 2002-04-11 Glaxo Group Limited Use of a ppar delta activator for treating inflammatory conditions
WO2002028433A2 (en) * 2000-10-05 2002-04-11 Glaxo Group Limited Use of a ppar delta activator in the treatment of diseases related to no inhibit ion or tnf inhibition
WO2004093910A1 (en) * 2003-04-22 2004-11-04 Astellas Pharma Inc. REMEDY FOR CEREBRAL NEURODEGENERATIVE DISEASES USING PPARδ AGONIST

Cited By (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7968723B2 (en) 2004-05-05 2011-06-28 High Point Pharmaceuticals, Llc Compounds, their preparation and use
US8053598B2 (en) 2004-05-05 2011-11-08 High Point Pharmaceuticals, Llc Compounds, their preparation and use
US8217086B2 (en) 2005-06-30 2012-07-10 High Point Pharmaceuticals, Llc Phenoxy acetic acids as PPAR delta activators
EP2298742A1 (en) 2005-06-30 2011-03-23 High Point Pharmaceuticals, LLC Phenoxy acetic acids as PPAR delta activators
US8426473B2 (en) 2005-06-30 2013-04-23 High Point Pharnaceuticals, LLC Phenoxy acetic acids as PPAR delta activators
WO2007003581A1 (en) 2005-06-30 2007-01-11 Novo Nordisk A/S Phenoxy acetic acids as ppar delta activators
US7943669B2 (en) 2005-06-30 2011-05-17 High Point Pharmaceuticals, Llc Phenoxy acetic acids as PPAR delta activators
US8362016B2 (en) 2005-12-22 2013-01-29 High Point Pharmaceuticals, Llc Phenyl propionic acids as PPAR delta activators
US9663481B2 (en) 2005-12-22 2017-05-30 Vtv Therapeutics Llc Phenoxy acetic acids and phenyl propionic acids as PPARδ agonists
US11420929B2 (en) 2005-12-22 2022-08-23 Vtv Therapeutics Llc Phenoxy acetic acids and phenyl propionic acids as PPAR delta agonists
US10947180B2 (en) 2005-12-22 2021-03-16 Vtv Therapeutics Llc Phenoxy acetic acids and phenyl propionic acids as PPAR delta agonists
US7943613B2 (en) 2005-12-22 2011-05-17 High Point Pharmaceuticals, Llc Compounds, their preparation and use
US9855274B2 (en) 2005-12-22 2018-01-02 Vtv Therapeutics Llc Phenoxy acetic acids and phenyl propionic acids as PPAR delta agonists
EP2386540A1 (en) 2005-12-22 2011-11-16 High Point Pharmaceuticals, LLC Novel compounds, their preparation and use
US7943612B2 (en) 2006-03-09 2011-05-17 High Point Pharmaceuticals, Llc Compounds that modulate PPAR activity, their preparation and use
US8946212B2 (en) 2008-06-09 2015-02-03 Sanofi-Aventis Annelated N-heterocyclic sulfonamides with oxadiazolone headgroup, processes for their preparation and their use as pharmaceuticals
US8716317B2 (en) 2008-06-09 2014-05-06 Sanofi Sulfonamides with heterocycle and oxadiazolone headgroup, processes for their preparation and their use as pharmaceuticals
US8329725B2 (en) 2008-06-09 2012-12-11 Sanofi Annelated pyrrolidin sulfonamides with oxadiazolone headgroup, processes for their preparation and their use as pharmaceuticals
US10456406B2 (en) 2013-09-09 2019-10-29 Vtv Therapeutics Llc Use of a PPAR-δ agonist for reducing loss of muscle strength, muscle mass, or type I muscle fibers in an immobilized limb
EP3756661A1 (en) 2013-09-09 2020-12-30 vTv Therapeutics LLC Use of a ppar-delta agonist for treating muscle atrophy
US11096946B2 (en) 2013-09-09 2021-08-24 Vtv Therapeutics Llc Use of a PPAR-δ agonist for reducing loss of muscle strength, muscle mass, or type I muscle fibers in an immobilized limb
US11931365B2 (en) 2022-01-25 2024-03-19 Reneo Pharmaceuticals, Inc. Use of PPAR-delta agonists in the treatment of disease

Also Published As

Publication number Publication date
MA28561B1 (en) 2007-04-03
SG138623A1 (en) 2008-01-28
NO20064985L (en) 2006-10-31
US20070149580A1 (en) 2007-06-28
ZA200607850B (en) 2008-10-29
WO2005097098A3 (en) 2005-12-22
CA2561159A1 (en) 2005-10-20
IL178165A0 (en) 2008-03-20
RU2006138495A (en) 2008-05-10
JP2007530703A (en) 2007-11-01
EP1737440A2 (en) 2007-01-03
KR20060134191A (en) 2006-12-27
AU2005231358A1 (en) 2005-10-20
BRPI0509540A (en) 2007-09-18
MXPA06011218A (en) 2007-01-16
CN1950077A (en) 2007-04-18

Similar Documents

Publication Publication Date Title
US20070149580A1 (en) Use of peroxisome proliferator activated receptor delta agonists for the treatment of ms and other demyelinating diseases
Granneman et al. Member of the peroxisome proliferator–activated receptor family of transcription factors is differentially expressed by oligodendrocytes
WO2015131788A1 (en) Treatment of neurological conditions
EP2806948B1 (en) Method for enhancing remyelination using gli1 inhibitors
JP2010528016A (en) Methods and compositions for stimulating cells
Jana et al. Gemfibrozil, a lipid-lowering drug, increases myelin genes in human oligodendrocytes via peroxisome proliferator-activated receptor-β
US7132287B2 (en) Method for neural stem cell differentiation using 5HT-1A agonists
US11779565B2 (en) Small organic molecules for use in the treatment of neuroinflammatory disorders
US9028810B2 (en) Composition for inducing migration of neural stem cells containing periostin as effective ingredient
Wang et al. Paired related homeobox protein 1 regulates quiescence in human oligodendrocyte progenitors
Zhang et al. Thymosin beta4 promotes oligodendrogenesis in the demyelinating central nervous system
EP3761982A1 (en) Treatment of demyelinating diseases
US20090155223A1 (en) Cell-based compositions and methods for treating conditions of the nervous system
WO2013021196A2 (en) Oligodendrocyte differentiation
Wu et al. IL-10 protects against OPC ferroptosis by regulating lipid reactive oxygen species levels post stroke
WO2017081033A1 (en) Cell differentiation or reprogramming using fezf2 and lmo4
US11435345B2 (en) Detection and treatment of demyelinating diseases
US20210186903A1 (en) Treatment for demyelinating disease
Meng et al. PPAR-γ Agonist Promotes Proliferation of Neural Stem Cells Via Mitophagy Inhibition
WO2008122087A1 (en) Methods for treating pervasive developmental disorders

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 BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG 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 NA NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SM SY 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): GM KE LS MW MZ NA 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 IS IT LT LU MC NL PL 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
DPEN Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed from 20040101)
WWE Wipo information: entry into national phase

Ref document number: 549822

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 178165

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 200607850

Country of ref document: ZA

Ref document number: DZP2005000463

Country of ref document: DZ

WWE Wipo information: entry into national phase

Ref document number: 12006501857

Country of ref document: PH

WWE Wipo information: entry into national phase

Ref document number: 11535240

Country of ref document: US

Ref document number: 2561159

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2005231358

Country of ref document: AU

Ref document number: 3643/CHENP/2006

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: PA/a/2006/011218

Country of ref document: MX

Ref document number: 2007506442

Country of ref document: JP

Ref document number: 06098506

Country of ref document: CO

NENP Non-entry into the national phase

Ref country code: DE

WWW Wipo information: withdrawn in national office

Ref document number: DE

WWE Wipo information: entry into national phase

Ref document number: 2005729215

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2005231358

Country of ref document: AU

Date of ref document: 20050329

Kind code of ref document: A

WWP Wipo information: published in national office

Ref document number: 2005231358

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2006138495

Country of ref document: RU

Ref document number: 1020067022955

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 200580014850.6

Country of ref document: CN

WWP Wipo information: published in national office

Ref document number: 1020067022955

Country of ref document: KR

WWP Wipo information: published in national office

Ref document number: 2005729215

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 11535240

Country of ref document: US

ENP Entry into the national phase

Ref document number: PI0509540

Country of ref document: BR