US20140221400A1 - Use of myeloperoxidase (mpo) inhibitors or pharmaceutically acceptable salts thereof to treat multiple sysyem atrophy (msa) 938 - Google Patents

Use of myeloperoxidase (mpo) inhibitors or pharmaceutically acceptable salts thereof to treat multiple sysyem atrophy (msa) 938 Download PDF

Info

Publication number
US20140221400A1
US20140221400A1 US14/075,790 US201314075790A US2014221400A1 US 20140221400 A1 US20140221400 A1 US 20140221400A1 US 201314075790 A US201314075790 A US 201314075790A US 2014221400 A1 US2014221400 A1 US 2014221400A1
Authority
US
United States
Prior art keywords
thioxo
tetrahydro
methyl
purin
pyrimidin
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US14/075,790
Inventor
Hakan Eriksson
Werner Poewe
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
AstraZeneca AB
Original Assignee
AstraZeneca AB
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 AstraZeneca AB filed Critical AstraZeneca AB
Priority to US14/075,790 priority Critical patent/US20140221400A1/en
Assigned to ASTRAZENECA AB reassignment ASTRAZENECA AB ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ERIKSSON, HAKAN, POEWE, WERNER
Publication of US20140221400A1 publication Critical patent/US20140221400A1/en
Priority to US16/117,020 priority patent/US10772890B2/en
Priority to US17/019,276 priority patent/US20200405724A1/en
Abandoned legal-status Critical Current

Links

Images

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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • 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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • A61K31/52Purines, e.g. adenine
    • A61K31/522Purines, e.g. adenine having oxo groups directly attached to the heterocyclic ring, e.g. hypoxanthine, guanine, acyclovir
    • 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
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • 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
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs 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

  • the present invention relates to the use of Myeloperoxidase (MPO) inhibitors or pharmaceutically acceptable salts thereof for the treatment of multiple system atrophy (MSA).
  • MSA multiple system atrophy
  • the present invention further relates to the use of Myeloperoxidase (MPO) inhibitors or pharmaceutically acceptable salts thereof for the treatment of Huntington's disease (HD).
  • HD Huntington's disease
  • the present invention also relates to the use of Myeloperoxidase (MPO) inhibitors or pharmaceutically acceptable salts thereof for neuroprotection.
  • MPO Myeloperoxidase
  • PMNs polymorphonuclear leukocytes
  • MPO is one member of a diverse protein family of mammalian peroxidases that also includes eosinophil peroxidase, thyroid peroxidase, salivary peroxidase, lactoperoxidase, prostaglandin H synthase, and others.
  • the mature enzyme is a dimer of identical halves. Each half molecule contains a covalently bound heme that exhibits unusual spectral properties responsible for the characteristic green colour of MPO.
  • PMNs are of particular importance for combating infections. These cells contain MPO, with well-documented microbicidal action. PMNs act non-specifically by phagocytosis to engulf microorganisms, incorporate them into vacuoles, termed phagosomes, which fuse with granules containing myeloperoxidase to form phagolysosomes. In phagolysosomes the enzymatic activity of the myeloperoxidase leads to the formation of hypochlorous acid, a potent bactericidal compound.
  • Macrophages are large phagocytic cells, which, like PMNs, are capable of phagocytosing microorganisms. Macrophages can generate hydrogen peroxide and upon activation also produce myeloperoxidase. MPO and hydrogen peroxide can also be released to the outside of the cells where the reaction with chloride can induce damage to adjacent tissue.
  • MPO positive cells are enormous present in the circulation and in tissue undergoing inflammation. More specifically MPO containing macrophages, microglia, astrocytes and/or neurons have been documented in the CNS during disease; multiple sclerosis (Nagra R M, et al. Journal of Neuroimmunology 1997; 78(1-2):97-107; Marik C, et al. Brain. 2007; 130: 2800-15; Gray E, et al. Brain Pathology. 2008; 18: 86-95), Parkinson's disease (Choi D-K. et al. J. Neurosci. 2005; 25(28):6594-600) and Alzheimer's disease (Reynolds W F, et al. Experimental Neurology. 1999; 155:31-41; Green P S. et al. Journal of Neurochemistry. 2004; 90(3):724-33). It is supposed that some aspects of a chronic ongoing inflammation result in an overwhelming destruction where agents from MPO reactions have an important role.
  • the enzyme is released both extracellularly as well as into phagolysosomes in the neutrophils (Hampton M B, Kettle A J, Winterbourn C C. Blood 1998; 92(9):3007-17).
  • a prerequisite for the MPO activity is the presence of hydrogen peroxide, generated by NADPH oxidase and a subsequent superoxide dismutation.
  • the oxidized enzyme is capable to use a plethora of different substrates of which chloride is most recognized. From this reaction the strong non-radical oxidant—hypochlorous acid (HOCl)—is formed. HOCl oxidizes sulphur containing amino acids like cysteine and methionine very efficiently (Peskin A V, Winterbourn C C.
  • MSA Multiple System Atrophy
  • MSA Multiple system atrophy
  • Support for MPO inhibition in an MSA-like pathology can be generated through the use of preclinical disease models for MSA, like transgenic mice with oligodendroglial overexpression of human ⁇ -synuclein with or without a toxin addition like 3-nitropropionic acid.
  • Huntington's disease is a hereditary progressive neurodegenerative disorder characterized clinically by motor and psychiatric disturbances and pathologically by neuronal loss and gliosis (reactive astrocytosis) particularly in the striatum and cerebral cortex.
  • HD is a neurodegenerative disorder caused by expansion of a CAG repeat in the HD gene, coding for polyglutamine in the huntingtin protein.
  • Explanations to the pathological mechanisms include oxidative stress, impaired energy metabolism, and abnormal protein-protein interactions. Such mechanisms are possible to link to MPO activity, which might be manifested through its observed overexpression in pathological HD tissue (Choi D-K. et al. J. Neurosci. 2005; 25(28):6594-600).
  • Support for MPO inhibition in an HD-like pathology can be generated through the use of preclinical disease models for HD.
  • Such models might be mice or rats treated with mitochondrial toxins like 3-nitropropionic acid or malonate (Matthews R T. et al J. Neurosci. 1998; 18:156-63).
  • Useful models might also be transgenic mice expressing mutants of the huntingtin protein with or without a toxin addition like 3-nitropropionic acid (Bogdanov M B. et al. J. Neurochem. 1998; 71:2642-44).
  • FIG. 1 is a graph depicting the mean daily motor score of MSA mice treated with AZD (compound I) and vehicle.
  • FIG. 2 is a graph depicting the rearing and open field activity of MSA mice treated with AZD (compound I) and vehicle.
  • FIG. 3 is a graph depicting stride length of the left and right hindlimbs of MSA mice treated with AZD (compound I) and vehicle.
  • FIG. 4A is a graph depicting the number of TH immunopositive cells in the substantia nigra pars compacta (SNc) of MSA mice treated with AZD (compound I) and vehicle.
  • FIG. 4B is a graph depicting the dopaminergic terminals in striatum of MSA mice treated with AZD (compound I) and vehicle.
  • FIG. 4C is a graph depicting the number of neurons in the striatum of MSA mice treated with AZD (compound I) and vehicle.
  • FIG. 5A is a graph depicting the total number of neurons in the inferior olivary complex in the cerebellum of MSA mice treated with AZD (compound I) and vehicle.
  • FIG. 5B is a graph depicting the total number of neurons in the pontine nuclei in the cerebellum of MSA mice treated with AZD (compound I) and vehicle.
  • FIG. 5C is a graph depicting the neurons per mm 3 in the purkinje cells in the cerebellum of MSA mice treated with AZD (compound I) and vehicle.
  • FIG. 6 is a graph depicting the microglial activation in the substantia nigra and striatum of MSA mice treated with AZD (compound I) and vehicle.
  • MPO inhibitors can be used for the treatment of multiple system atrophy (MSA).
  • the present invention is directed to the use of a MPO inhibitor for the manufacture of a medicament for the treatment of multiple system atrophy (MSA).
  • MSA multiple system atrophy
  • multiple system atrophy means a fatal progressive neurodegenerative disorder. It is defined as a sporadic alpha-synucleinopathy with dysautonomia and Parkinsonian and/or cerebellar motor impairment.
  • MPO inhibitors or pharmaceutically acceptable salts thereof can be used for the treatment of Huntington's disease (HD).
  • the present invention is also directed to the use of a MPO inhibitor or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for the treatment of Huntington's disease.
  • Heuntington's disease is intended to define a hereditary progressive neurodegenerative disorder characterized clinically by motor and psychiatric disturbances and pathologically by neuronal loss and gliosis (reactive astrocytosis) particularly in the striatum and cerebral cortex.
  • the present invention is also related to the use of MPO inhibitors or a pharmaceutically acceptable salt thereof for neuroprotection. Consequently, the present invention is directed to the use of a MPO inhibitor for the manufacture of a medicament for neuroprotection.
  • neuroprotection as used herein is defined as prevention of nerve cell loss and/or sparing of nerve cell fibers.
  • treating refers to reversing, alleviating, delaying or inhibiting the progress of, or preventing the disorder or condition to which such term applies, or one or more symptoms of such disorder or condition.
  • treatment refers to the act of “treating” as defined herein.
  • MPO-inhibitors examples include:
  • At least one of X and Y represents S, and the other represents O or S;
  • L represents a direct bond or C 1-7 alkylene, wherein said C 1-7 alkylene optionally incorporating a heteroatom selected from O, S (O) n and NR 6 , and said C 1-7 alkylene optionally incorporating one or two carbon-carbon double bonds, and said C 1-7 alkylene is optionally substituted by one or more substituents selected independently from OH, halogen, CN and NR 4 R 5 , C 1-6 alkyl and C 1-6 alkoxy, said C 1-6 alkoxy optionally incorporating a carbonyl adjacent to the oxygen;
  • n an integer 0, 1 or 2;
  • R 1 is hydrogen, or
  • R 1 is a saturated or partially unsaturated 3 to 7 membered ring optionally incorporating one or two heteroatoms selected independently from O, N and S, and optionally incorporating a carbonyl group, wherein said ring is optionally substituted by one or more substituents independently selected from halogen, SO 2 R 9 , SO 2 NR 9 R 10 , OH, C 1-7 alkyl, C 1-7 alkoxy, CN, CONR 2 R 3 , NR 2 COR 3 and COR 3 , wherein said C 1-7 alkoxy being optionally further substituted by C 1-6 alkoxy and optionally incorporating a carbonyl adjacent to the oxygen, and said C 1-7 alkyl being optionally further substituted by hydroxy or C 1-6 alkoxy and said C 1-7 alkyl or C 1-6 alkoxy optionally incorporating a carbonyl adjacent to the oxygen or at any position in the C 1-7 alkyl; or
  • R 1 is an aromatic ring system selected from phenyl, biphenyl, naphthyl or a monocyclic or bicyclic heteroaromatic ring structure containing 1 to 3 heteroatoms independently selected from O, N and S, said aromatic ring system being optionally substituted by one or more substituents independently selected from halogen, SO 2 R 9 , SO 2 NR 9 R 10 , OH, C 1-7 alkyl, C 1-7 alkoxy, CN, CONR 2 R 3 , NR 2 COR 3 and COR 3 ; said C 1-7 alkoxy being optionally further substituted by C 1-6 alkoxy and said C 1-6 alkoxy optionally incorporating a carbonyl adjacent to the oxygen, and said C 1-7 alkyl being optionally further substituted by hydroxy or C 1-6 alkoxy and said C 1-7 alkyl or C 1-6 alkoxy optionally incorporating a carbonyl adjacent to the oxygen or at any position in the alkyl;
  • R 12 represents hydrogen or halogen or a carbon optionally substituted with one to three halogen atoms
  • R 2 , R 3 , R 4 , R 5 , R 6 , R 9 and R 10 independently represent hydrogen, C 1-6 alkyl or C 1-6 alkoxy said alkoxy optionally incorporating a carbonyl adjacent to the oxygen, said C 1-6 alkyl being optionally further substituted by halogen, C 1-6 alkoxy, CHO, C 2-6 alkanoyl, OH, CONR 7 R 8 and NR 7 COR 8 ;
  • NR 2 R 3 , NR 4 R 5 and NR 9 R 10 each independently represent a 5 to 7 membered saturated azacyclic ring optionally incorporating one additional heteroatom selected from O, S and NR 11 , said azacyclic ring being optionally further substituted by halogen, C 1-6 alkoxy, CHO, C 2-6 alkanoyl, OH, CONR 7 R 8 and NR 7 COR 8 ;
  • R 7 , R 8 and R 11 independently represent hydrogen or C 1-6 alkyl, or the group NR 7 R 8 represents a 5- to 7-membered saturated azacyclic ring optionally incorporating one additional heteroatom selected from O, S and NR 11 ;
  • one of X and Y represents S, and the other represents O or S;
  • R 13 represents hydrogen or C 1-6 alkyl
  • R 14 represents hydrogen or C 1-6 alkyl; said C 1-6 alkyl group being optionally substituted by:
  • a saturated or partially unsaturated 3- to 7-membered ring optionally incorporating one or two heteroatoms selected independently from O, N and S, and optionally incorporating a carbonyl group; said ring being optionally substituted by one or more substituents selected from halogen, hydroxy, C 1-6 alkoxy and C 1-6 alkyl; said C 1-6 alkyl being optionally further substituted by hydroxy or C 1-6 alkoxy; or
  • an aromatic ring selected from phenyl, furyl or thienyl; said aromatic ring being optionally further substituted by halogen, C 1-6 alkyl or C 1-6 alkoxy;
  • R 15 and R 16 independently represent hydrogen or C 1-6 alkyl
  • said MPO inhibitor is selected from a compound of formula (IIa) or (IIb)
  • X represents S, and Y represents O;
  • R 13 represents hydrogen or C 1-6 alkyl
  • R 14 represents C 1-6 alkyl substituted by a saturated or partially unsaturated 3- to 7-membered ring optionally incorporating one or two heteroatoms selected independently from O, N and S, and optionally incorporating a carbonyl group; said ring being optionally substituted by one or more substituents selected from halogen, hydroxy, C 1-6 alkoxy and C 1-6 alkyl; said alkyl being optionally further substituted by hydroxy or C 1-6 alkoxy;
  • R 15 and R 16 independently represent hydrogen or C 1-6 alkyl
  • the ( ⁇ )-enantiomer of 3-(2-tetrahydrofuryl-methyl)-2-thioxanthine represents 3-(2R-tetrahydrofuryl-methyl)-2-thioxanthine and the (+)-enantiomer of 3-(2-tetrahydrofuryl-methyl)-2-thioxanthine represents 3-(2S-tetrahydrofuryl-methyl)-2-thioxanthine.
  • At least one of X and Y represents S, and the other represents O or S;
  • L represents (R 18 ) p -Q-(CR 19 R 20 ) r ; wherein (R 18 ) p and (CR 19 R 20 ) r each optionally contain one or two double or triple bonds;
  • Q is O, S(O) n , NR 21 , NR 21 C(O), C(O)NR 21 , or a bond;
  • R 18 is selected from C 1-6 alkyl or C 1-6 alkoxy, said C 1-6 alkyl or said C 1-6 alkoxy is optionally substituted with OH, halogen, CF 3 , CHF 2 , CFH 2 , CN, NR 22 R 23 , phenoxy or aryl; and wherein said phenoxy is optionally substituted with C 1-6 alkyl, halogen or C 1-6 alkoxy; and wherein said phenoxy optionally incorporates a carbonyl adjacent to the oxygen and wherein said C 1-6 alkoxy optionally incorporates a carbonyl adjacent to the oxygen; wherein R 19 and R 20 are independently selected from hydrogen, OH, halogen, CF 3 , CHF 2 , CFH 2 , CN, NR 22 R 23 , C1 to 6 alkyl, phenoxy and C 1-6 alkoxy; wherein said phenoxy or C 1-6 alkoxy optionally incorporates a carbonyl adjacent to the oxygen; and wherein said phen
  • p represents an integer 0, 1, 2, 3 or 4 and r represents an integer 0, 1, 2, 3 or 4; and wherein 1 ⁇ p+r ⁇ 7;
  • R 17 represents a mono- or bicyclic heteroaromatic ring system containing one or more heteroatoms selected from N, O and S; wherein said mono- or bicyclic heteroaromatic ring system is optionally fused with one or two 5- or 6-membered saturated or partially saturated ring(s) containing one or more atoms selected from C, N, O and S, wherein said mono- or bicyclic heteroaromatic ring system alone or when fused with one or two 5- or 6-membered saturated or partially saturated ring(s) is optionally substituted with one or more substituents independently selected from halogen, CHF 2 , CH 2 F, CF 3 , SO (n) R 24 , SO (n) NR 24 R 25 , (CH 2 ) n R 26 , NR 22 R 23 , OH, C1 to 7 alkyl, C 1-2 alkoxy, phenoxy, aryl, CN, C(O)NR 27 R 26 , NR 2 C(O)R 26 , C(
  • R 27 , R 26 , R 22 , R 23 , R 21 , R 24 and R 25 are independently selected from hydrogen, C 1-6 alkyl, C 1-6 alkoxy, aryl and phenoxy; said C 1-6 alkoxy or phenoxy is optionally incorporating a carbonyl adjacent to the oxygen; and said C 1-6 alkyl is optionally substituted with halogen, C 1-6 alkoxy, CHO, C 2-6 alkanoyl, OH, C(O)NR 28 R 29 or NR 28 C(O)R 29 ; and said aryl or said phenoxy is optionally substituted with C 1-6 alkyl, halogen or C 1-6 alkoxy;
  • NR 27 R 26 , NR 22 R 23 and NR 24 R 25 each independently represents a 5 to 7 membered saturated azacyclic ring optionally incorporating one additional heteroatom selected from O, S and NR 30 , said ring being optionally further substituted with halogen, C 1-6 alkoxy, CHO, C 2-6 alkanoyl, OH, C(O)NR 28 R 29 or NR 28 C(O)R 29 ;
  • R 28 , R 29 and R 30 independently represent hydrogen or C 1-6 alkyl, or the group NR 28 R 29 represents a 5 to 7 membered saturated azacyclic ring optionally incorporating one additional heteroatom selected from O, S and NR 30 ;
  • n an integer 0, 1 or 2;
  • Suitable pharmaceutically acceptable salts may be useful in the preparation of the compounds according to the present invention.
  • Suitable pharmaceutically acceptable salts of the compounds described herein include acid addition salts which may, for example, be formed by mixing a solution of the compound according to the present invention with a solution of a pharmaceutically acceptable acid such as hydrochloric acid, sulfuric acid, methanesulphonic acid and fumaric acid.
  • 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.
  • pharmaceutically acceptable salts includes both pharmaceutically acceptable acid addition salts and pharmaceutically acceptable cationic salts.
  • pharmaceutically acceptable cationic salts is intended to define but is not limited to such salts as the alkali metal salts, (e.g., sodium and potassium), alkaline earth metal salts (e.g., calcium and magnesium), aluminum salts, ammonium salts, and salts with organic amines such as benzathine (N,N′-dibenzylethylenediamine) and choline.
  • pharmaceutically acceptable acid addition salts is intended to define but is not limited to such salts as the hydrochloride, hydrobromide and sulfate.
  • the pharmaceutically acceptable cationic salts containing free carboxylic acids can be readily prepared by reacting the free acid form of with an appropriate base.
  • Typical bases are sodium hydroxide, sodium methoxide and sodium ethoxide.
  • the pharmaceutically acceptable acid addition salts containing free amine groups can be readily prepared by reacting the free base form with the appropriate acid.
  • MPO inhibitors having an asymmetric carbon atom are chiral compounds, and depending on the presence of asymmetric atoms, the MPO inhibitors may exist in the form of mixtures of isomers, particularly racemates, or in the form of pure isomers such as specific enantiomers.
  • MPO inhibitors or pharmaceutically acceptable salts thereof described herein can be administered in a standard manner such as orally, parenterally, transmucosally (e.g., sublingually or via buccal administration), topically, transdermally, rectally, via inhalation (e.g., nasal or deep lung inhalation).
  • Parenteral administration includes, but is not limited to intravenous, intraarterial, intraperitoneal, subcutaneous, intramuscular, intrathecal or via a high-pressure technique.
  • the MPO inhibitors or pharmaceutically acceptable salts thereof can be in the form of tablets or lozenges formulated in conventional manner.
  • tablets and capsules for oral administration can contain conventional excipients such as binding agents (e.g., syrup, acacia, gelatin, sorbitol, tragacanth, mucilage of starch or polyvinylpyrrolidone), fillers (e.g., lactose, sugar, microcrystalline cellulose, maize-starch, calcium phosphate or sorbitol), lubricants (e.g., magnesium stearate, stearic acid, talc, polyethylene glycol or silica), disintegrants (e.g., potato starch or sodium starch glycollate), or wetting agents (e.g., sodium lauryl sulfate). Tablets may be coated according to methods well known in the art. Such preparations can also be formulated as suppositories for rectal administration, e.g., containing
  • compositions for inhalation comprising MPO inhibitors or pharmaceutically acceptable salts thereof can typically be provided in the form of a solution, suspension, or emulsion that can be administered as a dry powder or in the form of an aerosol using a conventional propellant, such as dichlorodifluoromethane or trichlorofluoromethane.
  • Typical topical and transdermal formulations comprise conventional aqueous or non-aqueous vehicles, such as eye drops, creams, ointments, lotions, and pastes, or are in the form of a medicated plaster, patch, or membrane.
  • MPO inhibitors or pharmaceutically acceptable salts thereof described herein can be formulated for parenteral administration by injection or continuous infusion.
  • Formulations for injection can be in the form of suspensions, solutions, or emulsions in oily or aqueous vehicles, and can contain formulation agents, such as suspending, stabilizing, and/or dispersing agents.
  • the active ingredient can be in powder form for constitution with a suitable vehicle (e.g., sterile, pyrogen-free water) before use.
  • the MPO inhibitors or pharmaceutically acceptable salts thereof in accordance with the present invention also can be formulated as a depot preparation. Such long acting formulations can be administered by implantation (e.g., subcutaneously or intramuscularly) or by intramuscular injection. Accordingly, the compounds of the present invention can be formulated with suitable polymeric or hydrophobic materials (e.g., an emulsion in an acceptable oil), ion exchange resins, or as sparingly soluble derivatives (e.g., a sparingly soluble salt).
  • suitable polymeric or hydrophobic materials e.g., an emulsion in an acceptable oil
  • ion exchange resins e.g., ion exchange resins
  • sparingly soluble derivatives e.g., a sparingly soluble salt
  • a pharmaceutical composition comprising the MPO inhibitors or pharmaceutically acceptable salts thereof according to the present invention can take the form of solutions, suspensions, tablets, pills, capsules, powders, and the like.
  • Tablets containing various excipients such as sodium citrate, calcium carbonate and calcium phosphate are employed along with various disintegrants such as starch and preferably potato or tapioca starch and certain complex silicates, together with binding agents such as polyvinylpyrrolidone, sucrose, gelatin and acacia.
  • binding agents such as polyvinylpyrrolidone, sucrose, gelatin and acacia.
  • lubricating agents such as magnesium stearate, sodium lauryl sulfate and talc may be used to form tablets.
  • Solid compositions of a similar type are also employed as fillers in soft and hard-filled gelatin capsules; preferred materials in this connection also include lactose or milk sugar as well as high molecular weight polyethylene glycols.
  • the MPO inhibitors or pharmaceutically acceptable salts thereof described herein can be incorporated into oral liquid preparations such as aqueous or oily suspensions, solutions, emulsions, syrups, or elixirs, for example.
  • formulations containing these compounds can be presented as a dry product for constitution with water or other suitable vehicle before use.
  • Such liquid preparations can contain conventional additives, such as suspending agents, such as sorbitol syrup, synthetic and natural gums such as tragacanth, acacia, alginate, dextran, sodium carboxymethylcellulose, methylcellulose, polyvinyl-pyrrolidone or gelatin, glucose/sugar syrup, gelatin, hydroxyethylcellulose, hydroxypropylmethylcellulose, aluminum stearate gel, emulsifying agents, such as lecithin, sorbitan monooleate, or acacia; non-aqueous vehicles (which can include edible oils), such as almond oil, fractionated coconut oil, oily esters, propylene glycol, and ethyl alcohol; and preservatives, such as methyl or propyl p-hydroxybenzoate and sorbic acid.
  • suspending agents such as sorbitol syrup, synthetic and natural gums such as tragacanth, acacia, alginate, dextran, sodium carboxymethylcellulose, methyl
  • liquid forms in which the compositions described herein 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.
  • aqueous suspensions and/or elixirs are desired for oral administration
  • the compounds described herein can be combined with various sweetening agents, flavoring agents, coloring agents, emulsifying agents and/or suspending agents, as well as such diluents as water, ethanol, propylene glycol, glycerin and various like combinations thereof.
  • 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.
  • the MPO inhibitors or pharmaceutically acceptable salts thereof described herein can also be administered in a controlled release formulation (definition) such as a slow release or a fast release formulation.
  • a controlled release formulation such as a slow release or a fast release formulation.
  • Such controlled release formulations of the combinations described herein may be prepared using methods well known to those skilled in the art. The method of administration will be determined, by the attendant physician or other person skilled in the art after an evaluation of the patient's condition and requirements.
  • the effective dose of a MPO inhibitor or pharmaceutically acceptable salts thereof according to the present invention may vary, depending upon factors such as the condition of the patient, the severity of the symptoms of the disorder as well as the potency of the selected specific compound, the mode of administration, the age and weight of the patient, and the like. Determining a dose is within the skill of the ordinary artisan. The exact formulation, route of administration, and dosage can be chosen by the individual physician in view of the patient's condition. Dosage amount and interval can be adjusted individually to provide plasma levels of the active moiety, which are sufficient to maintain therapeutic effects.
  • the effective dose of MPO inhibitors or pharmaceutically acceptable salts thereof generally requires administering the compound in a range of from, and including, 1 to 1 000 mg.
  • said range is from, and including, 2 to 800 mg or from, and including, 2 to 400 mg.
  • the amount of MPO inhibitor is selected from about: 5, 10, 50, 100, 150, 200, 250, 300, 350, 400, 500, 550, 600, 700 and 800 mg.
  • transgenic (tg) or wild type mice with 3NP constitutes also the most established models of HD3NP (Brouillet E. et al. Prog. Neurobiol. 1999; 59:427-68). It relies on subacute systemic injection of this mitochondrial-complex II toxin. In mice, this toxin creates HD-like striatal lesions and replicates the metabolic failure occurring in HD. During its extensive use a correlation (Fernagut P O. et al. Neuroscience.
  • a novel mouse model of MSA has been developed by inducing oxidative stress in transgenic mice with oligodendroglial ⁇ -synuclein expression (described herein).
  • This model reproduces the cardinal neuropathological features of the disease including striatonigral degeneration (SND), olivopontocerebellar atrophy (OPCA), astrogliosis and microgliosis combined with oligodendroglial insoluble ⁇ -synuclein inclusions.
  • Mitochondrial inhibition by 3NP in the presence of glial cytoplasmic inclusions in transgenic mice induces a selective neuronal cell death pattern typical for MSA in these animals (Stefanova N. et al. Am. J. Pathol. 2005; 166:869-76).
  • MPO inhibitors have been use to suppress MPO activity in an MSA mouse model consisting of an oligodendroglial ⁇ -SYN overexpression in transgenic mice exposed to mitochondrial inhibition by 3-nitropropionic acid (3NP).
  • the effects were followed by application of established immunohistological and behavioral methods to evaluate the participation of MPO in the pathogenesis of MSA and the possible neuroprotective effects of in an MSA model.
  • Transgenic substantia nigra pars compacta is undergoing early neuronal loss associated with the oligodendroglial ⁇ -synucleinopathy during the time window between two and four months of age. This early neuronal loss was correlated with microglial activation in the SNc. Suppression of microgliosis in the time period between 2 and 4 months of age was found to be neuroprotective for nigral neurons.
  • Microglial activation is a prominent finding in MSA brains. It was shown, in transgenic mice overexpressing human wild type a-synuclein under the control of the proteolipid protein (PLP) promoter, that such mice had intense microglial activation especially in the white matter, which is not the case in wild type C57B1/6 mice (Stefanova N. et al. Am. J. Pathol. 2005; 166:869-76). Further, microglial activation is highly intensified following 3NP exposure and accompanied by MSA-like neuronal degeneration. The correlation of microglial activation with neuronal cell loss suggests that microglial factors might at least partially mediate neurodegeneration by releasing reactive oxygen species, nitrogen oxide (NO), cytokines, or chemokines.
  • NO nitrogen oxide
  • mice were intoxicated chronically with 3NP with slowly increasing doses of toxin according to a previously used scheme (i. e. 4 ⁇ 10 mg/kg, 4 ⁇ 20 mg/kg, 4 ⁇ 40 mg/kg, 4 ⁇ 50 mg/kg intraperitoneal injections every 12th hour for a period of 8 days) to model MSA (Stefanova N. et al. Am. J. Pathol. 2005; 166:869-76)
  • the drug and vehicle (0.1 mol/L meglumine with 20% w/v hydroxypropyl-(3-cyclodextrin, pH 10.8) were stored at 4° C. Mice received the necessary dose of drug/vehicle (10 mL/kg) twice daily by oral gavage during the indicated period.
  • mice To test the locomotor activity of the mice the Flex Field Activity System (San Diego Instruments, CA, USA) was applied, which allows monitoring and real-time counting of horizontal and vertical locomotor activity by 544 photo-beam channels. Mice was placed in the center of the open field (40.5 ⁇ 40.5 ⁇ 36.5 cm) and tested for a 15 min period always at the same time of the day (17.00 h). The tests were performed in a dark room that was completely isolated from external noises and light during the test period.
  • Flex Field Activity System San Diego Instruments, CA, USA
  • the stride length of the forelimbs and hindlimbs of the mice was measured after a habituation to the test for 3 days before its performance according to Fernagut et al. (Fernagut P O. et al. Neuroscience. 2002; 114:1005-17) with slight modification.
  • the limbs of each animal were wetted with a non-toxic food colour and each mouse was let to run on a strip of paper (42 cm long, 4.5 cm wide) down a bright corridor towards a dark goal box. After three runs, the stride length of the hindlimbs on each side was measured, excluding the beginning (7 cm) and the end (7 cm) of the run. The mean stride length for each limb was determined.
  • Nissl staining Coronal sections throughout the whole brain were mounted on slides and processed for standard cresyl violet staining.
  • Immunocytochemistry was performed according to standard protocols (Stefanova N. et al. Am. J. Pathol. 2005; 166:869-76) on free floating sections (40 ⁇ m) to analyze neuronal and glial pathology in MSA mouse model.
  • the following primary antibodies were used: anti-TH tyrosine hydroxylase (Sigma); anti-DARPP-32 (dopamine and cyclic adenosine 3′,5′-monophosphate-regulated phosphoprotein 32); anti-GFAP (glial fibrillary acidic protein, Roche Diagnostics GmbH); anti-CD11b: (Serotec). Secondary antibodies were biotinylated anti-mouse or anti-rat IgG as appropriate.
  • Stereology was applied using a computer-assisted image analysis system (Nikon E-800 microscope, CCD video camera, Optronics MicroFire, Goleta, USA; Stereo Investigator Software, MicroBrightField Europe e.K., Magdeburg, Germany).
  • Optical fractionator was used to count neurons in the striatum, substantia nigra pars compacta, pontine nuclei, and inferior olives.
  • Purkinje cells were counted in a region outlined to include only the Purkinje cell layer as previously reported (German D C. et al. Neuroscience. 2001; 105:999-1005). All data were expressed as mean value ⁇ SEM.
  • Glial activation in substantia nigra and striatum was measure by determining optical density in the target region by delineating its area in serial sections. For all statistical tests performed, a probability level of 5% (p ⁇ 0.05) was considered significant.
  • High dose Compound I (180 ⁇ mol/kg) is neuroprotective regarding striatonigral degeneration in MSA mice ( FIG. 4 ).
  • TH immunopositive cells in the substantia nigra dopaminergic terminals in the striatum as well as the striatal DARPP-32 immunoreactive neurons.
  • High dose Compound I (180 ⁇ mol/kg) is neuroprotective regarding olivopontocerebellar atrophy in MSA mice. Protection of the inferior olivary complex, pontine nuclei and Purkinje cells in the cerebellum ( FIG. 5 ).
  • a high dose Compound I (180 ⁇ mol/kg) was associated with suppression of microglial activation, another marker of neuroinflammation, in MSA mice. This was seen both in the substantia nigra and the striatum ( FIG. 6 ). This suggests that we have pharmacologically corroborated the previously suggested (Stefanova N. et al. Am. J. Pathol. 2005; 166:869-76) link between microglia activation and neurodegeneration.
  • MPO inhibitors have the potential to be neuroprotective also in human neurodegenerative disorders.
  • a neuroprotection of all affected neuronal phenotypes, without any exception, in a model as described herein by MPO inhibitors should offer clear arguments for MPO inhibitors as being neuroprotective, not necessarily only limited to MSA, PD and Huntington's disease.

Abstract

The present invention relates to the use of MPO inhibitors for the treatment of multiple system atrophy. The present invention also relates to the use of MPO inhibitors for the treatment of Huntington's disease. The present invention also relates to the use of MPO inhibitors for neuroprotection.

Description

  • The present invention relates to the use of Myeloperoxidase (MPO) inhibitors or pharmaceutically acceptable salts thereof for the treatment of multiple system atrophy (MSA). The present invention further relates to the use of Myeloperoxidase (MPO) inhibitors or pharmaceutically acceptable salts thereof for the treatment of Huntington's disease (HD). The present invention also relates to the use of Myeloperoxidase (MPO) inhibitors or pharmaceutically acceptable salts thereof for neuroprotection.
  • Myeloperoxidase (MPO) is a heme-containing enzyme found predominantly in polymorphonuclear leukocytes (PMNs). MPO is one member of a diverse protein family of mammalian peroxidases that also includes eosinophil peroxidase, thyroid peroxidase, salivary peroxidase, lactoperoxidase, prostaglandin H synthase, and others. The mature enzyme is a dimer of identical halves. Each half molecule contains a covalently bound heme that exhibits unusual spectral properties responsible for the characteristic green colour of MPO. Cleavage of the disulphide bridge linking the two halves of MPO yields the hemi-enzyme that exhibits spectral and catalytic properties indistinguishable from those of the intact enzyme. The enzyme uses hydrogen peroxide to oxidize chloride to hypochlorous acid. Other halides and pseudohalides (like thiocyanate) are also physiological substrates to MPO.
  • PMNs are of particular importance for combating infections. These cells contain MPO, with well-documented microbicidal action. PMNs act non-specifically by phagocytosis to engulf microorganisms, incorporate them into vacuoles, termed phagosomes, which fuse with granules containing myeloperoxidase to form phagolysosomes. In phagolysosomes the enzymatic activity of the myeloperoxidase leads to the formation of hypochlorous acid, a potent bactericidal compound. Hypochlorous acid is oxidizing in itself, and reacts most avidly with thiols and thioethers, but also converts amines into chloramines, and chlorinates aromatic amino acids. Macrophages are large phagocytic cells, which, like PMNs, are capable of phagocytosing microorganisms. Macrophages can generate hydrogen peroxide and upon activation also produce myeloperoxidase. MPO and hydrogen peroxide can also be released to the outside of the cells where the reaction with chloride can induce damage to adjacent tissue.
  • Linkage of myeloperoxidase activity to disease has been implicated in neurological diseases with a neuroinflammatory response including multiple sclerosis, Alzheimer's disease and Parkinson's disease.
  • MPO positive cells are immensely present in the circulation and in tissue undergoing inflammation. More specifically MPO containing macrophages, microglia, astrocytes and/or neurons have been documented in the CNS during disease; multiple sclerosis (Nagra R M, et al. Journal of Neuroimmunology 1997; 78(1-2):97-107; Marik C, et al. Brain. 2007; 130: 2800-15; Gray E, et al. Brain Pathology. 2008; 18: 86-95), Parkinson's disease (Choi D-K. et al. J. Neurosci. 2005; 25(28):6594-600) and Alzheimer's disease (Reynolds W F, et al. Experimental Neurology. 1999; 155:31-41; Green P S. et al. Journal of Neurochemistry. 2004; 90(3):724-33). It is supposed that some aspects of a chronic ongoing inflammation result in an overwhelming destruction where agents from MPO reactions have an important role.
  • The enzyme is released both extracellularly as well as into phagolysosomes in the neutrophils (Hampton M B, Kettle A J, Winterbourn C C. Blood 1998; 92(9):3007-17). A prerequisite for the MPO activity is the presence of hydrogen peroxide, generated by NADPH oxidase and a subsequent superoxide dismutation. The oxidized enzyme is capable to use a plethora of different substrates of which chloride is most recognized. From this reaction the strong non-radical oxidant—hypochlorous acid (HOCl)—is formed. HOCl oxidizes sulphur containing amino acids like cysteine and methionine very efficiently (Peskin A V, Winterbourn C C. Free Radical Biology and Medicine 2001; 30(5):572-9). It also forms chloramines with amino groups, both in proteins and other biomolecules (Peskin A V. et al. Free Radical Biology and Medicine 2004; 37(10):1622-30). It chlorinates phenols (like tyrosine) (Hazen S L. et al. Mass Free Radical Biology and Medicine 1997; 23(6):909-16) and unsaturated bonds in lipids (Albert C J. et al. J. Biol. Chem. 2001; 276(26):23733-41), oxidizes iron centers (Rosen H, Klebanoff S J. Journal of Biological Chemistry 1982; 257(22):13731-354) and crosslinks proteins (Fu X, Mueller D M, Heinecke J W. Biochemistry 2002; 41(4):1293-301). Various compounds that are MPO inhibitors are disclosed in WO 01/85146, J. Heterocyclic Chemistry, 1992, 29, 343-354, J. Chem. Soc., 1962, 1863, WO03/089430 and WO2006/062465.
  • Multiple System Atrophy (MSA)
  • Multiple system atrophy (MSA) is a neurodegenerative disorder presenting with autonomic failure and with motor impairment resulting from L-dopa-unresponsive parkinsonism, cerebellar ataxia and pyramidal signs. Histologically, there is neuron loss in the striatum, substantia nigra pars compacta, cerebellum, pons, inferior olives and intermediolateral column of the spinal cord. Glial pathology includes astrogliosis, microglial activation and α-synuclein containing oligodendroglial cytoplasmic inclusions. The pronounced neuroinflammation with activated microglia contribution as well as cytoplasmic inclusion bodies, containing aggregated and oxidatively modified proteins, makes it intriguing to consider a significant contribution of MPO activity in the progressive neurodegeneration characterizing the MSA pathology.
  • Support for MPO inhibition in an MSA-like pathology can be generated through the use of preclinical disease models for MSA, like transgenic mice with oligodendroglial overexpression of human α-synuclein with or without a toxin addition like 3-nitropropionic acid.
  • Huntington's Disease (HD)
  • Huntington's disease (HD) is a hereditary progressive neurodegenerative disorder characterized clinically by motor and psychiatric disturbances and pathologically by neuronal loss and gliosis (reactive astrocytosis) particularly in the striatum and cerebral cortex. HD is a neurodegenerative disorder caused by expansion of a CAG repeat in the HD gene, coding for polyglutamine in the huntingtin protein. Explanations to the pathological mechanisms include oxidative stress, impaired energy metabolism, and abnormal protein-protein interactions. Such mechanisms are possible to link to MPO activity, which might be manifested through its observed overexpression in pathological HD tissue (Choi D-K. et al. J. Neurosci. 2005; 25(28):6594-600).
  • Support for MPO inhibition in an HD-like pathology can be generated through the use of preclinical disease models for HD. Such models might be mice or rats treated with mitochondrial toxins like 3-nitropropionic acid or malonate (Matthews R T. et al J. Neurosci. 1998; 18:156-63). Useful models might also be transgenic mice expressing mutants of the huntingtin protein with or without a toxin addition like 3-nitropropionic acid (Bogdanov M B. et al. J. Neurochem. 1998; 71:2642-44).
  • There is a large unmet need for medications that can be used for the treatment of Huntington's disease, for the treatment of multiple system atrophy and/or for neuroprotection.
  • FIG. 1 is a graph depicting the mean daily motor score of MSA mice treated with AZD (compound I) and vehicle.
  • FIG. 2 is a graph depicting the rearing and open field activity of MSA mice treated with AZD (compound I) and vehicle.
  • FIG. 3 is a graph depicting stride length of the left and right hindlimbs of MSA mice treated with AZD (compound I) and vehicle.
  • FIG. 4A is a graph depicting the number of TH immunopositive cells in the substantia nigra pars compacta (SNc) of MSA mice treated with AZD (compound I) and vehicle.
  • FIG. 4B is a graph depicting the dopaminergic terminals in striatum of MSA mice treated with AZD (compound I) and vehicle.
  • FIG. 4C is a graph depicting the number of neurons in the striatum of MSA mice treated with AZD (compound I) and vehicle.
  • FIG. 5A is a graph depicting the total number of neurons in the inferior olivary complex in the cerebellum of MSA mice treated with AZD (compound I) and vehicle.
  • FIG. 5B is a graph depicting the total number of neurons in the pontine nuclei in the cerebellum of MSA mice treated with AZD (compound I) and vehicle.
  • FIG. 5C is a graph depicting the neurons per mm3 in the purkinje cells in the cerebellum of MSA mice treated with AZD (compound I) and vehicle.
  • FIG. 6 is a graph depicting the microglial activation in the substantia nigra and striatum of MSA mice treated with AZD (compound I) and vehicle.
  • It has been found that MPO inhibitors can be used for the treatment of multiple system atrophy (MSA).
  • Consequently, the present invention is directed to the use of a MPO inhibitor for the manufacture of a medicament for the treatment of multiple system atrophy (MSA).
  • The wording “multiple system atrophy” as used herein, means a fatal progressive neurodegenerative disorder. It is defined as a sporadic alpha-synucleinopathy with dysautonomia and Parkinsonian and/or cerebellar motor impairment.
  • It has also been found that MPO inhibitors or pharmaceutically acceptable salts thereof can be used for the treatment of Huntington's disease (HD).
  • Consequently, the present invention is also directed to the use of a MPO inhibitor or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for the treatment of Huntington's disease.
  • The wording “Huntington's disease ” as used herein, is intended to define a hereditary progressive neurodegenerative disorder characterized clinically by motor and psychiatric disturbances and pathologically by neuronal loss and gliosis (reactive astrocytosis) particularly in the striatum and cerebral cortex.
  • Further, the present invention is also related to the use of MPO inhibitors or a pharmaceutically acceptable salt thereof for neuroprotection. Consequently, the present invention is directed to the use of a MPO inhibitor for the manufacture of a medicament for neuroprotection.
  • The term “neuroprotection” as used herein is defined as prevention of nerve cell loss and/or sparing of nerve cell fibers.
  • The term “treating” as used herein, refers to reversing, alleviating, delaying or inhibiting the progress of, or preventing the disorder or condition to which such term applies, or one or more symptoms of such disorder or condition. The term “treatment”, as used herein, refers to the act of “treating” as defined herein.
  • Examples of compounds that can be used as MPO-inhibitors are the following:
  • 1) A compound of formula (I)
  • Figure US20140221400A1-20140807-C00001
  • wherein:
  • At least one of X and Y represents S, and the other represents O or S;
  • L represents a direct bond or C1-7alkylene, wherein said C1-7alkylene optionally incorporating a heteroatom selected from O, S (O)n and NR6, and said C1-7alkylene optionally incorporating one or two carbon-carbon double bonds, and said C1-7alkylene is optionally substituted by one or more substituents selected independently from OH, halogen, CN and NR4R5, C1-6alkyl and C1-6alkoxy, said C1-6alkoxy optionally incorporating a carbonyl adjacent to the oxygen;
  • n represents an integer 0, 1 or 2;
  • R1 is hydrogen, or
  • R1is a saturated or partially unsaturated 3 to 7 membered ring optionally incorporating one or two heteroatoms selected independently from O, N and S, and optionally incorporating a carbonyl group, wherein said ring is optionally substituted by one or more substituents independently selected from halogen, SO2R9, SO2NR9R10, OH, C1-7alkyl, C1-7alkoxy, CN, CONR2R3, NR2COR3 and COR3, wherein said C1-7alkoxy being optionally further substituted by C1-6alkoxy and optionally incorporating a carbonyl adjacent to the oxygen, and said C1-7alkyl being optionally further substituted by hydroxy or C1-6alkoxy and said C1-7alkyl or C1-6alkoxy optionally incorporating a carbonyl adjacent to the oxygen or at any position in the C1-7alkyl; or
  • R1 is an aromatic ring system selected from phenyl, biphenyl, naphthyl or a monocyclic or bicyclic heteroaromatic ring structure containing 1 to 3 heteroatoms independently selected from O, N and S, said aromatic ring system being optionally substituted by one or more substituents independently selected from halogen, SO2R9, SO2NR9R10, OH, C1-7alkyl, C1-7alkoxy, CN, CONR2R3, NR2COR3 and COR3; said C1-7alkoxy being optionally further substituted by C1-6alkoxy and said C1-6alkoxy optionally incorporating a carbonyl adjacent to the oxygen, and said C1-7alkyl being optionally further substituted by hydroxy or C1-6alkoxy and said C1-7alkyl or C1-6alkoxy optionally incorporating a carbonyl adjacent to the oxygen or at any position in the alkyl;
  • R12 represents hydrogen or halogen or a carbon optionally substituted with one to three halogen atoms;
  • at each occurrence, R2, R3, R4, R5, R6, R9 and R10 independently represent hydrogen, C1-6alkyl or C1-6alkoxy said alkoxy optionally incorporating a carbonyl adjacent to the oxygen, said C1-6alkyl being optionally further substituted by halogen, C1-6alkoxy, CHO, C2-6alkanoyl, OH, CONR7R8 and NR7COR8;
  • or the groups NR2R3, NR4R5 and NR9R10 each independently represent a 5 to 7 membered saturated azacyclic ring optionally incorporating one additional heteroatom selected from O, S and NR11, said azacyclic ring being optionally further substituted by halogen, C1-6alkoxy, CHO, C2-6alkanoyl, OH, CONR7R8 and NR7COR8;
  • at each occurrence R7, R8 and R11 independently represent hydrogen or C1-6alkyl, or the group NR7R8 represents a 5- to 7-membered saturated azacyclic ring optionally incorporating one additional heteroatom selected from O, S and NR11;
  • or pharmaceutically acceptable salts, solvates of solvates of salts thereof. These compounds are described in WO 2006/062465.
  • 2) A compound selected from the group consisting of:
  • 1-butyl-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
  • 1-isobutyl-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
  • 1-(pyridin-2-ylmethyl)-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
  • 1-(2-fluoro-benzyl)-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
  • 1-[2-(2-methoxyethoxy)-3-propoxybenzyl]-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
  • 1-(6-ethoxy-pyridin-2-ylmethyl)-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
  • 1-piperidin-3-ylmethyl-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
  • 1-butyl-4-thioxo-1,3,4,5-tetrahydro-2H-pyrrolo[3,2-d]pyrimidin-2-one;
  • 1-(2-isopropoxyethyl)-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
  • 1-(2-methoxy-2-methylpropyl)-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
  • 1-(2-ethoxy-2-methylpropyl)-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
  • 1-(piperidin-4-ylmethyl)-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
  • 1-[(1-methylpiperidin-3-yl)methyl]-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
  • 1-[2-hydroxy-2-(4-methoxyphenyl)ethyl]-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
  • 1-(2-methoxybenzyl)-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
  • 1-(3-methoxybenzyl)-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
  • 1-(2,4-dimethoxybenzyl)-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
  • 1-[(3-chloropyridin-2-yl)methyl]-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
  • 1-{[3-(2-ethoxyethoxy)pyridin-2-yl]methyl}-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
  • 1-[(6-oxo-1,6-dihydropyridin-2-yl)methyl]-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
  • 1-(1H-indol-3-ylmethyl)-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
  • 1-(1H-benzimidazol-2-ylmethyl)-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
  • 1-[(5-chloro-1H-indol-2-yl)methyl]-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
  • 1-[(5-fluoro-1H-indol-2-yl)methyl]-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
  • 1-(1H-indol-6-ylmethyl)-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
  • 1-(1H-indol-5-ylmethyl)-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
  • 1-[(5-fluoro-1H-indol-3-yl)methyl]-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
  • 1-(1H-imidazol-5-ylmethyl)-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
  • 1-(1H-imidazol-2-ylmethyl)-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
  • 1-[(5-chloro-1H-benzimidazol-2-yl)methyl]-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
  • 1-[(4,5-dimethyl-1H-benzimidazol-2-yl)methyl]-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
  • 7-bromo-1-isobutyl-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one; and
  • 1-(3-chlorophenyl)-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
  • or pharmaceutically acceptable salts thereof, solvate or solvate of a salt thereof. These compounds are described in WO 2006/062465.
  • 3) A compound of formula (Ha) or (Hb)
  • Figure US20140221400A1-20140807-C00002
  • wherein:
  • one of X and Y represents S, and the other represents O or S;
  • R13 represents hydrogen or C1-6alkyl;
  • R14 represents hydrogen or C1-6alkyl; said C1-6alkyl group being optionally substituted by:
  • i) a saturated or partially unsaturated 3- to 7-membered ring optionally incorporating one or two heteroatoms selected independently from O, N and S, and optionally incorporating a carbonyl group; said ring being optionally substituted by one or more substituents selected from halogen, hydroxy, C1-6alkoxy and C1-6alkyl; said C1-6alkyl being optionally further substituted by hydroxy or C1-6alkoxy; or
  • ii) C1-6alkoxy; or
  • iii) an aromatic ring selected from phenyl, furyl or thienyl; said aromatic ring being optionally further substituted by halogen, C1-6alkyl or C1-6alkoxy;
  • R15 and R16 independently represent hydrogen or C1-6alkyl;
  • or a pharmaceutically acceptable salt, solvate or solvate of a salt thereof. These compounds are described in WO 2003/089430.
  • According to one aspect of the present invention said MPO inhibitor is selected from a compound of formula (IIa) or (IIb)
  • Figure US20140221400A1-20140807-C00003
  • wherein:
  • X represents S, and Y represents O;
  • R13 represents hydrogen or C1-6alkyl;
  • R14 represents C1-6alkyl substituted by a saturated or partially unsaturated 3- to 7-membered ring optionally incorporating one or two heteroatoms selected independently from O, N and S, and optionally incorporating a carbonyl group; said ring being optionally substituted by one or more substituents selected from halogen, hydroxy, C1-6alkoxy and C1-6alkyl; said alkyl being optionally further substituted by hydroxy or C1-6alkoxy;
  • R15 and R16 independently represent hydrogen or C1-6alkyl;
  • or pharmaceutically acceptable salts, solvates or solvates of a salt thereof. These compounds are described in WO 2003/089430.
  • 4) A compound selected from the group consisting of:
  • 1,3-diisobutyl-8-methyl-6-thioxanthine;
  • 1,3-dibutyl-8-methyl-6-thioxanthine;
  • 3-isobutyl-1,8-dimethyl-6-thioxanthine;
  • 3-(2-methylbutyl)-6-thioxanthine;
  • 3-isobutyl-8-methyl-6-thioxanthine;
  • 3-isobutyl-2-thioxanthine;
  • 3-isobutyl-2,6-dithioxanthine;
  • 3-isobutyl-8-methyl-2-thioxanthine;
  • 3-isobutyl-7-methyl-2-thioxanthine;
  • 3-cyclohexylmethyl-2-thioxanthine;
  • 3-(3-methoxypropyl)-2-thioxanthine;
  • 3-cyclopropylmethyl-2-thioxanthine;
  • 3-isobutyl-1-methyl-2-thioxanthine;
  • 3-(2-tetrahydrofuryl-methyl)-2-thioxanthine;
  • 3-(2-methoxy-ethyl)-2-thioxanthine;
  • 3-(3-(1-morpholinyl)-propyl)-2-thioxanthine;
  • 3-(2-furyl-methyl)-2-thioxanthine;
  • 3-(4-methoxybenzyl)-2-thioxanthine;
  • 3-(4-fluorobenzyl)-2-thioxanthine;
  • 3-phenethyl-2-thioxanthine;
  • (+)-3-(2-tetrahydrofuryl-methyl)-2-thioxanthine;
  • (−)-3-(2-tetrahydrofuryl-methyl)-2-thioxanthine; and
  • 3-n-butyl-2-thioxanthine;
  • or a pharmaceutically acceptable salt, solvate or solvate of a salt thereof. These compounds are described in WO 2003/089430.
  • The (−)-enantiomer of 3-(2-tetrahydrofuryl-methyl)-2-thioxanthine represents 3-(2R-tetrahydrofuryl-methyl)-2-thioxanthine and the (+)-enantiomer of 3-(2-tetrahydrofuryl-methyl)-2-thioxanthine represents 3-(2S-tetrahydrofuryl-methyl)-2-thioxanthine.
  • 5) A compound of formula of Formula (III)
  • Figure US20140221400A1-20140807-C00004
  • wherein
  • at least one of X and Y represents S, and the other represents O or S;
  • L represents (R18)p-Q-(CR19R20)r; wherein (R18)p and (CR19R20)r each optionally contain one or two double or triple bonds;
  • wherein Q is O, S(O)n, NR21, NR21C(O), C(O)NR21, or a bond;
  • wherein R18 is selected from C1-6alkyl or C1-6alkoxy, said C1-6alkyl or said C1-6alkoxy is optionally substituted with OH, halogen, CF3, CHF2, CFH2, CN, NR22R23, phenoxy or aryl; and wherein said phenoxy is optionally substituted with C1-6alkyl, halogen or C1-6alkoxy; and wherein said phenoxy optionally incorporates a carbonyl adjacent to the oxygen and wherein said C1-6alkoxy optionally incorporates a carbonyl adjacent to the oxygen; wherein R19 and R20 are independently selected from hydrogen, OH, halogen, CF3, CHF2, CFH2, CN, NR22R23, C1 to 6 alkyl, phenoxy and C1-6alkoxy; wherein said phenoxy or C1-6alkoxy optionally incorporates a carbonyl adjacent to the oxygen; and wherein said phenoxy is optionally substituted with C1-6alkyl, halogen or C1-6alkoxy;
  • wherein p represents an integer 0, 1, 2, 3 or 4 and r represents an integer 0, 1, 2, 3 or 4; and wherein 1≦p+r≦7;
  • R17 represents a mono- or bicyclic heteroaromatic ring system containing one or more heteroatoms selected from N, O and S; wherein said mono- or bicyclic heteroaromatic ring system is optionally fused with one or two 5- or 6-membered saturated or partially saturated ring(s) containing one or more atoms selected from C, N, O and S, wherein said mono- or bicyclic heteroaromatic ring system alone or when fused with one or two 5- or 6-membered saturated or partially saturated ring(s) is optionally substituted with one or more substituents independently selected from halogen, CHF2, CH2F, CF3, SO(n)R24, SO(n)NR24R25, (CH2)nR26, NR22R23, OH, C1 to 7 alkyl, C1-2alkoxy, phenoxy, aryl, CN, C(O)NR27R26, NR2C(O)R26, C(O)R26, a 5- or 6-membered saturated or partially saturated ring containing one or more atoms selected from C, N, O or S, and a mono- or bicyclic heteroaromatic ring system containing one or more heteroatoms selected from N, S or O; and wherein said C1-7alkoxy is optionally substituted with C1-7alkoxy or aryl; and wherein said C1-7alkoxy or said phenoxy is optionally incorporating a carbonyl adjacent to the oxygen; and wherein said C1-7alkyl is optionally substituted with hydroxy or C1-6alkoxy; and wherein said C1-7alkyl is optionally incorporating a carbonyl at any position in the C C1-7alkyl; and wherein said phenoxy is optionally substituted with C1-6alkyl, halogen or C1-6alkoxy;
  • at each occurrence, R27, R26, R22, R23, R21, R24 and R25 are independently selected from hydrogen, C1-6alkyl, C1-6alkoxy, aryl and phenoxy; said C1-6alkoxy or phenoxy is optionally incorporating a carbonyl adjacent to the oxygen; and said C1-6alkyl is optionally substituted with halogen, C1-6alkoxy, CHO, C2-6alkanoyl, OH, C(O)NR28R29 or NR28C(O)R29; and said aryl or said phenoxy is optionally substituted with C1-6alkyl, halogen or C1-6alkoxy;
  • or the groups NR27R26, NR22R23 and NR24R25 each independently represents a 5 to 7 membered saturated azacyclic ring optionally incorporating one additional heteroatom selected from O, S and NR30, said ring being optionally further substituted with halogen, C1-6alkoxy, CHO, C2-6alkanoyl, OH, C(O)NR28R29 or NR28C(O)R29;
  • at each occurrence R28, R29 and R30 independently represent hydrogen or C1-6alkyl, or the group NR28R29 represents a 5 to 7 membered saturated azacyclic ring optionally incorporating one additional heteroatom selected from O, S and NR30;
  • n represents an integer 0, 1 or 2;
  • with the proviso that for R17 thienyl or furyl is excluded;
  • and with the proviso that when Q is O, S(O)6, NR21, NR21C(O) or C(O)NR21, then p is greater or equal to 1;
  • or a pharmaceutically acceptable salt, solvate or solvate of a salt thereof. These compounds are described in PCT/SE2007/000349.
  • 6) A compound selected from the group consisting of:
  • 3-(pyridin-2-ylmethyl)-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
  • 3-(pyridin-3-ylmethyl)-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
  • 3-(pyridin-4-ylmethyl)-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
  • 3-{[3-ethoxy-4-(2-ethoxyethoxy)pyridin-2-yl]methyl}-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
  • 3-[(5-fluoro-1H-indol-2-yl)methyl]-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
  • 3-[(5-fluoro-1H-indol-2-yl)methyl]-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
  • 3-[(2-butyl-4-chloro-1H-imidazol-5-yl)methyl]-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
  • 3-(1H-benzimidazol-2-ylmethyl)-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
  • 3-[1-(1H-benzimidazol-2-yl)ethyl]-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
  • 3-[(5-chloro-1H-indol-3-yl)methyl]-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one and
  • 3-[(4-fluoro-1H-indol-3-yl)methyl]-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
  • 3-[2-(1H-Benzimidazol-2-yl)ethyl]-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
  • 3-(1H-Pyrazol-3-ylmethyl)-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
  • 3-[(5-Methylpyrazin-2-yl)methyl)-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
  • 3-[(3-Isopropylisoxazol-5-yl)methyl]-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
  • 3-[(4-Methyl-1,2,5-oxadiazol-3-yl)methyl]-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
  • 3-[(6-Butoxypyridin-2-yl)methyl]-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
  • 3-[(4-Butoxypyridin-2-yl)methyl]-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
  • 3-[(3-Butoxypyridin-2-yl)methyl]-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
  • 3-[2-(Pyridin-2-ylmethoxy)propyl]-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
  • 3-[(3,5-Dimethylisoxazol-4-yl)methyl]-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
  • 3-[(1-Methyl-1H-indol-2-yl)methyl]-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
  • 3-(2-Phenyl-2-pyridin-2-ylethyl)-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
  • 3-(Quinolin-4-ylmethyl)-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
  • 3-[(6-Phenoxypyridin-3-yl)methyl]-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
  • 3-{2-[(Quinolin-4-ylmethyl)amino]ethyl}-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
  • 3-(2-{[(1-Methyl-1H-indol-3-yl)methyl]amino}ethyl)-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
  • 3-{2-[Methyl(quinolin-4-ylmethyl)amino]ethyl}-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
  • 3-(2-Aminopropyl)-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one trifluoroacetate;
  • 3-{2-[(Pyridin-2-ylmethyl)amino]propyl}-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one trifluoroacetate;
  • 3-{2-[(Pyridin-3-ylmethyl)amino]propyl}-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
  • 3-{2-[(Pyridin-4-ylmethyl)amino]propyl}-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
  • 3-(2-{[(6-Chloropyridin-3-yl)methyl]amino}propyl)-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one trifluoroacetate;
  • 3-[2-({[6-(Trifluoromethyl)pyridin-3-yl]methyl}amino)propyl]-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one trifluoroacetate;
  • 3-(2-{[(4,6-Dichloropyrimidin-5-yl)methyl]amino}propyl)-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
  • 3-[2-({[2-(Dimethylamino)pyrimidin-5-yl]methyl}amino)propyl]-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
  • 3-{2-[(Quinolin-2-ylmethyl)amino]propyl}-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one trifluoroacetate;
  • 3-{2-[(Quinolin-3-ylmethyl)amino]propyl}-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
  • 3-(2-{[(1-tert-Butyl-3,5-dimethyl-1H-pyrazol-4-yl)methyl]amino}propyl)-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
  • 3-[2-({[1-(1,1-Dioxidotetrahydro-3-thienyl)-3,5-dimethyl-1H-pyrazol-4-yl]methyl}amino)propyl]-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
  • 3-{2-[(1H-Benzoimidazol-2-ylmethyl)amino]propyl}-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
  • 3-[2-({[1-(Phenylsulfonyl)-1H-pyrrol-2-yl]methyl}amino]propyl]-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one trifluoroacetate;
  • 3-{2-[({1-[(4-methylphenyl)sulfonyl]-1H-pyrrol-2-yl}methyl)amino]propyl}-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one trifluoroacetate;
  • 3-(2-{[(1-methyl-1H-pyrrol-2-yl)methyl]amino}propyl)-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
  • 3-[2-({[1-(4-sec-Butylphenyl)-1H-pyrrol-2-yl]methyl}amino)propyl)-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
  • 3-[2-({[1-(3-Methoxyphenyl)-1H-pyrrol-2-yl]methyl}amino)propyl]-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
  • 3-[2-({[2,5-Dimethyl-1-(1,3-thiazol-2-yl)-1H-pyrrol-3-yl]methyl}amino)propyl]-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
  • 3-[2-({[4-(3-Chlorobenzoyl)-1-methyl-1H-pyrrol-2-yl]methyl}amino)propyl]-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
  • 3-{2-[(1H-Imidazol-2-ylmethyl)amino]propyl}-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
  • 3-(2-{[(1-Methyl-1H-imidazol-2-yl)methyl]amino}propyl)-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
  • 3-(2-{[(4-Bromo-1-methyl-1H-imidazol-5-yl)methyl]amino}propyl)-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
  • 3-(2-{[(1-Methyl-1H-indol-3-yl)methyl]amino}propyl)-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
  • 2-Thioxo-3-{2-[(1H-1,2,3-triazol-5-ylmethyl)amino]propyl}-1,2,3,7-tetrahydro-6H-purin-6-one;
  • 3-[2-({[1-(Benzyloxy)-1H-imidazol-2-yl]methyl}amino)propyl]-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
  • 3-(2-{[(6-Bromo-2-methylimidazo[1,2-a]pyridin-3-yl)methyl]amino}propyl}-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
  • 3-{2-[({1-[2-(2-Methoxyphenoxy)ethyl]-1H-pyrrol-2-yl}methyl)amino]propyl]-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
  • N-[1-Methyl-2-(6-oxo-2-thioxo-1,2,6,7-tetrahydro-3H-purin-3-yl)ethyl]pyridine-2-carboxamide;
  • N-[1-Methyl-2-(6-oxo-2-thioxo-1,2,6,7-tetrahydro-3H-purin-3-yl)ethyl]nicotinamide;
  • N-[1-Methyl-2-(6-oxo-2-thioxo-1,2,6,7-tetrahydro-3H-purin-3-yl)-ethyl]isonicotinamide;
  • N-[1-methyl-2-(6-oxo-2-thioxo-1,2,6,7-tetrahydro-3H-purin-3-yl)ethyl]-1,8-naphthyridine-2-carboxamide;
  • N-[1-Methyl-2-(6-oxo-2-thioxo-1,2,6,7-tetrahydro-3H-purin-3-yl)ethyl]quinoline-2-carboxamide;
  • N-[1-Methyl-2-(6-oxo-2-thioxo-1,2,6,7-tetrahydro-3H-purin-3-yl)ethyl]pyrimidine-2-carboxamide; and
  • N-[1-Methyl-2-(6-oxo-2-thioxo-1,2,6,7-tetrahydro-3H-purin-3-yl)ethyl]-1H-imidazole-2-carboxamide trifluroaceate;
  • or a pharmaceutically acceptable salt, solvate or solvate of a salt thereof. These compounds are described in PCT/SE2007/000349.
  • For use in medicine, pharmaceutically acceptable salts may be useful in the preparation of the compounds according to the present invention. Suitable pharmaceutically acceptable salts of the compounds described herein include acid addition salts which may, for example, be formed by mixing a solution of the compound according to the present invention with a solution of a pharmaceutically acceptable acid such as hydrochloric acid, sulfuric acid, methanesulphonic acid and fumaric acid. Furthermore, where the compounds 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 expression “pharmaceutically acceptable salts” includes both pharmaceutically acceptable acid addition salts and pharmaceutically acceptable cationic salts. The expression “pharmaceutically acceptable cationic salts” is intended to define but is not limited to such salts as the alkali metal salts, (e.g., sodium and potassium), alkaline earth metal salts (e.g., calcium and magnesium), aluminum salts, ammonium salts, and salts with organic amines such as benzathine (N,N′-dibenzylethylenediamine) and choline. The expression “pharmaceutically acceptable acid addition salts” is intended to define but is not limited to such salts as the hydrochloride, hydrobromide and sulfate.
  • The pharmaceutically acceptable cationic salts containing free carboxylic acids can be readily prepared by reacting the free acid form of with an appropriate base. Typical bases are sodium hydroxide, sodium methoxide and sodium ethoxide. The pharmaceutically acceptable acid addition salts containing free amine groups can be readily prepared by reacting the free base form with the appropriate acid.
  • The use of optical isomers of MPO inhibitors is also within the scope of the present invention. MPO inhibitors having an asymmetric carbon atom are chiral compounds, and depending on the presence of asymmetric atoms, the MPO inhibitors may exist in the form of mixtures of isomers, particularly racemates, or in the form of pure isomers such as specific enantiomers.
  • Pharmaceutical Formulations
  • The MPO inhibitors or pharmaceutically acceptable salts thereof described herein can be administered in a standard manner such as orally, parenterally, transmucosally (e.g., sublingually or via buccal administration), topically, transdermally, rectally, via inhalation (e.g., nasal or deep lung inhalation). Parenteral administration includes, but is not limited to intravenous, intraarterial, intraperitoneal, subcutaneous, intramuscular, intrathecal or via a high-pressure technique.
  • For buccal administration, the MPO inhibitors or pharmaceutically acceptable salts thereof can be in the form of tablets or lozenges formulated in conventional manner. For example, tablets and capsules for oral administration can contain conventional excipients such as binding agents (e.g., syrup, acacia, gelatin, sorbitol, tragacanth, mucilage of starch or polyvinylpyrrolidone), fillers (e.g., lactose, sugar, microcrystalline cellulose, maize-starch, calcium phosphate or sorbitol), lubricants (e.g., magnesium stearate, stearic acid, talc, polyethylene glycol or silica), disintegrants (e.g., potato starch or sodium starch glycollate), or wetting agents (e.g., sodium lauryl sulfate). Tablets may be coated according to methods well known in the art. Such preparations can also be formulated as suppositories for rectal administration, e.g., containing conventional suppository bases, such as cocoa butter or other glycerides.
  • Compositions for inhalation comprising MPO inhibitors or pharmaceutically acceptable salts thereof can typically be provided in the form of a solution, suspension, or emulsion that can be administered as a dry powder or in the form of an aerosol using a conventional propellant, such as dichlorodifluoromethane or trichlorofluoromethane. Typical topical and transdermal formulations comprise conventional aqueous or non-aqueous vehicles, such as eye drops, creams, ointments, lotions, and pastes, or are in the form of a medicated plaster, patch, or membrane.
  • Additionally, MPO inhibitors or pharmaceutically acceptable salts thereof described herein can be formulated for parenteral administration by injection or continuous infusion. Formulations for injection can be in the form of suspensions, solutions, or emulsions in oily or aqueous vehicles, and can contain formulation agents, such as suspending, stabilizing, and/or dispersing agents. Alternatively, the active ingredient can be in powder form for constitution with a suitable vehicle (e.g., sterile, pyrogen-free water) before use.
  • The MPO inhibitors or pharmaceutically acceptable salts thereof in accordance with the present invention also can be formulated as a depot preparation. Such long acting formulations can be administered by implantation (e.g., subcutaneously or intramuscularly) or by intramuscular injection. Accordingly, the compounds of the present invention can be formulated with suitable polymeric or hydrophobic materials (e.g., an emulsion in an acceptable oil), ion exchange resins, or as sparingly soluble derivatives (e.g., a sparingly soluble salt).
  • For oral administration a pharmaceutical composition comprising the MPO inhibitors or pharmaceutically acceptable salts thereof according to the present invention can take the form of solutions, suspensions, tablets, pills, capsules, powders, and the like. Tablets containing various excipients such as sodium citrate, calcium carbonate and calcium phosphate are employed along with various disintegrants such as starch and preferably potato or tapioca starch and certain complex silicates, together with binding agents such as polyvinylpyrrolidone, sucrose, gelatin and acacia. Additionally, lubricating agents such as magnesium stearate, sodium lauryl sulfate and talc may be used to form tablets. Solid compositions of a similar type are also employed as fillers in soft and hard-filled gelatin capsules; preferred materials in this connection also include lactose or milk sugar as well as high molecular weight polyethylene glycols.
  • Alternatively, the MPO inhibitors or pharmaceutically acceptable salts thereof described herein can be incorporated into oral liquid preparations such as aqueous or oily suspensions, solutions, emulsions, syrups, or elixirs, for example. Moreover, formulations containing these compounds can be presented as a dry product for constitution with water or other suitable vehicle before use. Such liquid preparations can contain conventional additives, such as suspending agents, such as sorbitol syrup, synthetic and natural gums such as tragacanth, acacia, alginate, dextran, sodium carboxymethylcellulose, methylcellulose, polyvinyl-pyrrolidone or gelatin, glucose/sugar syrup, gelatin, hydroxyethylcellulose, hydroxypropylmethylcellulose, aluminum stearate gel, emulsifying agents, such as lecithin, sorbitan monooleate, or acacia; non-aqueous vehicles (which can include edible oils), such as almond oil, fractionated coconut oil, oily esters, propylene glycol, and ethyl alcohol; and preservatives, such as methyl or propyl p-hydroxybenzoate and sorbic acid. The liquid forms in which the compositions described herein 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.
  • When aqueous suspensions and/or elixirs are desired for oral administration, the compounds described herein can be combined with various sweetening agents, flavoring agents, coloring agents, emulsifying agents and/or suspending agents, as well as such diluents as water, ethanol, propylene glycol, glycerin and various like combinations thereof. 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.
  • The MPO inhibitors or pharmaceutically acceptable salts thereof described herein can also be administered in a controlled release formulation (definition) such as a slow release or a fast release formulation. Such controlled release formulations of the combinations described herein may be prepared using methods well known to those skilled in the art. The method of administration will be determined, by the attendant physician or other person skilled in the art after an evaluation of the patient's condition and requirements.
  • Thus, the effective dose of a MPO inhibitor or pharmaceutically acceptable salts thereof according to the present invention may vary, depending upon factors such as the condition of the patient, the severity of the symptoms of the disorder as well as the potency of the selected specific compound, the mode of administration, the age and weight of the patient, and the like. Determining a dose is within the skill of the ordinary artisan. The exact formulation, route of administration, and dosage can be chosen by the individual physician in view of the patient's condition. Dosage amount and interval can be adjusted individually to provide plasma levels of the active moiety, which are sufficient to maintain therapeutic effects.
  • Typically, the effective dose of MPO inhibitors or pharmaceutically acceptable salts thereof generally requires administering the compound in a range of from, and including, 1 to 1 000 mg. According to one embodiment of the present invention, said range is from, and including, 2 to 800 mg or from, and including, 2 to 400 mg. In an alternative embodiment of the present invention the amount of MPO inhibitor is selected from about: 5, 10, 50, 100, 150, 200, 250, 300, 350, 400, 500, 550, 600, 700 and 800 mg.
  • Description of the Methods
  • The treatment of transgenic (tg) or wild type mice with 3NP constitutes also the most established models of HD3NP (Brouillet E. et al. Prog. Neurobiol. 1999; 59:427-68). It relies on subacute systemic injection of this mitochondrial-complex II toxin. In mice, this toxin creates HD-like striatal lesions and replicates the metabolic failure occurring in HD. During its extensive use a correlation (Fernagut P O. et al. Neuroscience. 2002; 114:1005-17) between the time-course and intensity of the motor disorder has been demonstrated, using a semiquantitative scale (rating bradykinesia, truncal dystonia, hindlimb dystonia and clasping and impaired postural control) and the severity of striatal damage (neuronal loss and astrocytic reaction). An impairment of sensorimotor integration has also been demonstrated using quantified tests known to be sensitive to striatonigral dysfunction: general activity, pole test and beam-traversing test. Consequently, several of the important behavioural and histopathological endpoints, of relevance for HD, are the same as in the used MSA model. Thus, the striatal pathology including neuronal loss and parts of the motor behaviour in the MSA model mentioned above also reflect the HD pathology.
  • A novel mouse model of MSA has been developed by inducing oxidative stress in transgenic mice with oligodendroglial α-synuclein expression (described herein). This model reproduces the cardinal neuropathological features of the disease including striatonigral degeneration (SND), olivopontocerebellar atrophy (OPCA), astrogliosis and microgliosis combined with oligodendroglial insoluble α-synuclein inclusions. Mitochondrial inhibition by 3NP in the presence of glial cytoplasmic inclusions in transgenic mice induces a selective neuronal cell death pattern typical for MSA in these animals (Stefanova N. et al. Am. J. Pathol. 2005; 166:869-76).
  • Thus, in the present invention, MPO inhibitors have been use to suppress MPO activity in an MSA mouse model consisting of an oligodendroglial α-SYN overexpression in transgenic mice exposed to mitochondrial inhibition by 3-nitropropionic acid (3NP). The effects were followed by application of established immunohistological and behavioral methods to evaluate the participation of MPO in the pathogenesis of MSA and the possible neuroprotective effects of in an MSA model.
  • Transgenic substantia nigra pars compacta (SNc) is undergoing early neuronal loss associated with the oligodendroglial α-synucleinopathy during the time window between two and four months of age. This early neuronal loss was correlated with microglial activation in the SNc. Suppression of microgliosis in the time period between 2 and 4 months of age was found to be neuroprotective for nigral neurons. The findings suggest that the combined transgenic and neurotoxic MSA mouse model should lend itself as a pre-clinical test for novel therapeutic candidates for MSA, both for early “minimal change” or late progressed “full-blown” MSA paradigms.
  • Microglial activation is a prominent finding in MSA brains. It was shown, in transgenic mice overexpressing human wild type a-synuclein under the control of the proteolipid protein (PLP) promoter, that such mice had intense microglial activation especially in the white matter, which is not the case in wild type C57B1/6 mice (Stefanova N. et al. Am. J. Pathol. 2005; 166:869-76). Further, microglial activation is highly intensified following 3NP exposure and accompanied by MSA-like neuronal degeneration. The correlation of microglial activation with neuronal cell loss suggests that microglial factors might at least partially mediate neurodegeneration by releasing reactive oxygen species, nitrogen oxide (NO), cytokines, or chemokines.
  • Animals
  • A total of 30 (PLP)-α-synuclein transgenic mice were used. Animals were housed at 12/12 hours dark/light cycle with free access to food and water in the animal facility of the Innsbruck Medical University. All experiments were performed in accordance with the Austrian law and after permission for animal experiments of the Federal Ministry for Education, Science, and Culture of Austria.
  • Groups
  • Control group (n=10) MSA mice (tg+3NP), treated with vehicle (Cyclodextrin, prepared by AstraZeneca) p.o. (per oral administration)
  • Low dose group (n=10) MSA mice (tg+3NP) treated with 1-(2-Isopropoxyethyl)-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one (Compound I; prepared by AstraZeneca), 2×60 μmol/kg p.o.
  • High dose group (n=10) MSA mice (tg+3NP) treated with Compound I (prepared by AstraZeneca), 2×180 μmol/kg, p.o.
  • 1-(2-Isopropoxyethyl)-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one (Compound I treatment) was started one week prior to the first 3NP intoxication and stopped three weeks after the first 3NP intoxication (see 3NP intoxication protocol below). Animals underwent behavioral tests during week 3-4 after the beginning of the experiment. On day 28 animals were perfused under deep thiopental anesthesia and the brains were collected for histopathological analysis of neuronal loss and gliosis.
  • 3NP Intoxication
  • Mice were intoxicated chronically with 3NP with slowly increasing doses of toxin according to a previously used scheme (i. e. 4×10 mg/kg, 4×20 mg/kg, 4×40 mg/kg, 4×50 mg/kg intraperitoneal injections every 12th hour for a period of 8 days) to model MSA (Stefanova N. et al. Am. J. Pathol. 2005; 166:869-76)
  • Compound I Treatment
  • The drug and vehicle (0.1 mol/L meglumine with 20% w/v hydroxypropyl-(3-cyclodextrin, pH 10.8) were stored at 4° C. Mice received the necessary dose of drug/vehicle (10 mL/kg) twice daily by oral gavage during the indicated period.
  • Behaviour
  • Behavioural tests were performed blindly to the treatment status according to validated procedures: clinical scale evaluation, pole test and stride length spontaneous locomotor activity test (Stefanova N. et al. Am. J. Pathol. 2005; 166:869-76)
  • Motor Clinical Scale Evaluation.
  • A previously described rating scale for evaluation of hindlimb clasping, general locomotor activity, hindlimb dystonia, truncal dystonia and postural challenge response (0, normal; 1 slightly disturbed, and 2 markedly disabled). (Fernagut P O. et al. Neuroscience. 2002; 114:1005-17)
  • Open Field Activity
  • To test the locomotor activity of the mice the Flex Field Activity System (San Diego Instruments, CA, USA) was applied, which allows monitoring and real-time counting of horizontal and vertical locomotor activity by 544 photo-beam channels. Mice was placed in the center of the open field (40.5×40.5×36.5 cm) and tested for a 15 min period always at the same time of the day (17.00 h). The tests were performed in a dark room that was completely isolated from external noises and light during the test period.
  • Stride Length
  • The stride length of the forelimbs and hindlimbs of the mice was measured after a habituation to the test for 3 days before its performance according to Fernagut et al. (Fernagut P O. et al. Neuroscience. 2002; 114:1005-17) with slight modification. The limbs of each animal were wetted with a non-toxic food colour and each mouse was let to run on a strip of paper (42 cm long, 4.5 cm wide) down a bright corridor towards a dark goal box. After three runs, the stride length of the hindlimbs on each side was measured, excluding the beginning (7 cm) and the end (7 cm) of the run. The mean stride length for each limb was determined.
  • Tissue Preparation
  • Animals were perfused under thiopental overdose with 4% paraformaldehyde (PFA) pH=7.4. Brains were quickly removed and stored for 24 hours in 4% PFA at 4° C. After cryoprotection in a 20% sucrose/0.1M PBS pH 7.4 solution, the brains were frozen and stored at −80° C. Serial sections (total of 7 series) were cut on cryostat (Leica) and collected for histological stainings (one series on slides) and immunohistochemistry (6 series free floating).
  • Nissl staining: Coronal sections throughout the whole brain were mounted on slides and processed for standard cresyl violet staining.
  • Immunocytochemistry was performed according to standard protocols (Stefanova N. et al. Am. J. Pathol. 2005; 166:869-76) on free floating sections (40 μm) to analyze neuronal and glial pathology in MSA mouse model. The following primary antibodies were used: anti-TH tyrosine hydroxylase (Sigma); anti-DARPP-32 (dopamine and cyclic adenosine 3′,5′-monophosphate-regulated phosphoprotein 32); anti-GFAP (glial fibrillary acidic protein, Roche Diagnostics GmbH); anti-CD11b: (Serotec). Secondary antibodies were biotinylated anti-mouse or anti-rat IgG as appropriate. Shortly, after washing in phosphate buffered saline (PBS), sections were incubated in 0.3% H2O2, rinsed again and blocked for 1 hour in 10% normal goat serum in PBS with 0.3% Triton-X100 (PBS-T), followed by overnight incubation in the primary antibody at 4° C. After washing in PBS-T, slices were incubated for 1 hour in the secondary antibody, washed again and incubated for another hour in avidin-biotin complex (Elite Kit, Vector). Finally the reaction was visualized by 3,3′-diaminobenzidine.
  • Stereology was applied using a computer-assisted image analysis system (Nikon E-800 microscope, CCD video camera, Optronics MicroFire, Goleta, USA; Stereo Investigator Software, MicroBrightField Europe e.K., Magdeburg, Germany). Optical fractionator was used to count neurons in the striatum, substantia nigra pars compacta, pontine nuclei, and inferior olives. Purkinje cells were counted in a region outlined to include only the Purkinje cell layer as previously reported (German D C. et al. Neuroscience. 2001; 105:999-1005). All data were expressed as mean value±SEM. Glial activation in substantia nigra and striatum was measure by determining optical density in the target region by delineating its area in serial sections. For all statistical tests performed, a probability level of 5% (p<0.05) was considered significant.
  • Results
  • Effects of Compound I Treatment on Motor Behaviour of MSA Mice
  • There was a significant improvement in the mean daily motor score in MSA mice treated with Compound I compared to vehicle treated mice (FIG. 1). There was also a significant improvement in flex field performance after treatment with high dose Compound I (180 μmol/kg). Both rearing and open field activity was affected (FIG. 2). Similarly, there was a significant improvement in stride length test performance after treatment with high dose Compound I (180 μmol/kg), both left and right hindlimbs were equally affected (FIG. 3).
  • Effects of Compound I Treatment on Neuropathology of MSA Mice
  • High dose Compound I (180 μmol/kg) is neuroprotective regarding striatonigral degeneration in MSA mice (FIG. 4). Evident on TH immunopositive cells in the substantia nigra, dopaminergic terminals in the striatum as well as the striatal DARPP-32 immunoreactive neurons.
  • High dose Compound I (180 μmol/kg) is neuroprotective regarding olivopontocerebellar atrophy in MSA mice. Protection of the inferior olivary complex, pontine nuclei and Purkinje cells in the cerebellum (FIG. 5).
  • A high dose Compound I (180 μmol/kg) was associated with suppression of microglial activation, another marker of neuroinflammation, in MSA mice. This was seen both in the substantia nigra and the striatum (FIG. 6). This suggests that we have pharmacologically corroborated the previously suggested (Stefanova N. et al. Am. J. Pathol. 2005; 166:869-76) link between microglia activation and neurodegeneration.
  • Summary of Findings
  • A significant neuroprotection was demonstrated with Compound I treatment. Neurons were consistently preserved at the level of substantia nigra pars compacta, striatum, cerebellar cortex, pontine nuclei, and inferior olivary complex. This neuroprotection was accompanied by a functional improvement measured by different behavioural tests. The Compound I effects were also related to suppression of microglial activation. The data supports that MPO inhibitors have a potential of being neuroprotective in conditions accompanied by neuroinflammation, including MSA, PD and HD.
  • A widespread neuroprotection, not limited to only a subset of neurons, through a reduced neuronal cell loss and/or reduced loss of neuronal terminals upon treatment in this kind of model with an MPO inhibitor will in addition support that MPO inhibitors have the potential to be neuroprotective also in human neurodegenerative disorders. A neuroprotection of all affected neuronal phenotypes, without any exception, in a model as described herein by MPO inhibitors should offer clear arguments for MPO inhibitors as being neuroprotective, not necessarily only limited to MSA, PD and Huntington's disease.

Claims (8)

1-7. (canceled)
8. A method for treating multiple system atrophy, comprising administering a pharmaceutically and pharmacologically effective amount of a MPO inhibitor or a pharmaceutically acceptable salt thereof to a subject in need of such treatment.
9. A method for treating Huntington's disease, comprising administering a pharmaceutically and pharmacologically effective amount of a MPO inhibitor or a pharmaceutically acceptable salt thereof to a subject in need of such treatment.
10. A method for neuroprotection, comprising administering a pharmaceutically and pharmacologically effective amount of a MPO inhibitor or a pharmaceutically acceptable salt thereof to a subject in need of such treatment.
11. The method according to any one of claims 8 to 10, wherein said MPO inhibitor or pharmaceutically acceptable salt thereof is selected from:
1-butyl-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
1-isobutyl-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
1-(pyridin-2-ylmethyl)-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
1-(2-fluoro-benzyl)-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
1-[2-(2-methoxyethoxy)-3-propoxybenzyl]-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
1-(6-ethoxy-pyridin-2-ylmethyl)-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
1-piperidin-3-ylmethyl-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
1-butyl-4-thioxo-1,3,4,5-tetrahydro-2H-pyrrolo[3,2-d]pyrimidin-2-one;
1-(2-isopropoxyethyl)-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
1-(2-methoxy-2-methylpropyl)-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
1-(2-ethoxy-2-methylpropyl)-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
1-(piperidin-4-ylmethyl)-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
1-[(1-methylpiperidin-3-yl)methyl]-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
1-[2-hydroxy-2-(4-methoxyphenyl)ethyl]-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
1-(2-methoxybenzyl)-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
1-(3-methoxybenzyl)-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
1-(2,4-dimethoxybenzyl)-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
1-[(3-chloropyridin-2-yl)methyl]-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
1-{[3-(2-ethoxyethoxy)pyridin-2-yl]methyl}-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
1-[(6-oxo-1,6-dihydropyridin-2-yl)methyl]-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
1-(1H-indol-3-ylmethyl)-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
1-(1H-benzimidazol-2-ylmethyl)-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
1-[(5-chloro-1H-indol-2-yl)methyl]-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
1-[(5-fluoro-1H-indol-2-yl)methyl]-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
1-(1H-indol-6-ylmethyl)-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
1-(1H-indol-5-ylmethyl)-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
1-[(5-fluoro-1H-indol-3-yl)methyl]-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
1-(1H-imidazol-5-ylmethyl)-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
1-(1H-imidazol-2-ylmethyl)-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
1-[(5-chloro-1H-benzimidazol-2-yl)methyl]-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
1-[(4,5-dimethyl-1H-benzimidazol-2-yl)methyl]-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
7-bromo-1-isobutyl-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one; and
1-(3-chlorophenyl)-2-thioxo-1,2,3,5-tetrahydro-pyrrolo[3,2-c]pyrimidin-4-one.
12. The method according to any one of claims 8 to 10, wherein said MPO inhibitor or pharmaceutically acceptable salt thereof is selected from:
1,3-diisobutyl-8-methyl-6-thioxanthine;
1,3-dibutyl-8-methyl-6-thioxanthine;
3-isobutyl-1,8-dimethyl-6-thioxanthine;
3-(2-methylbutyl)-6-thioxanthine;
3-isobutyl-8-methyl-6-thioxanthine;
3-isobutyl-2-thioxanthine;
3-isobutyl-2,6-dithioxanthine;
3-isobutyl-8-methyl-2-thioxanthine;
3-isobutyl-7-methyl-2-thioxanthine;
3-cyclohexylmethyl-2-thioxanthine;
3-(3-methoxypropyl)-2-thioxanthine;
3-cyclopropylmethyl-2-thioxanthine;
3-isobutyl-1-methyl-2-thioxanthine;
3-(2-tetrahydrofuryl-methyl)-2-thioxanthine;
3-(2-methoxy-ethyl)-2-thioxanthine;
3-(3-(1-morpholinyl)-propyl)-2-thioxanthine;
3-(2-furyl-methyl)-2-thioxanthine;
3-(4-methoxybenzyl)-2-thioxanthine;
3-(4-fluorobenzyl)-2-thioxanthine;
3-phenethyl-2-thioxanthine;
(+)-3-(2-tetrahydrofuryl-methyl)-2-thioxanthine;
(−)-3-(2-tetrahydrofuryl-methyl)-2-thioxanthine; and
3-n-butyl-2-thioxanthine.
13. The method according to any one of claims 8 to 10, wherein said MPO inhibitor or pharmaceutically acceptable salt thereof is selected from:
3-(pyridin-2-ylmethyl)-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
3-(pyridin-3-ylmethyl)-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
3-(pyridin-4-ylmethyl)-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
3-{[3-ethoxy-4-(2-ethoxyethoxy)pyridin-2-yl]methyl}-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
3-[(5-fluoro-1H-indol-2-yl)methyl]-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
3-[(5-fluoro-1H-indol-2-yl)methyl]-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
3-[(2-butyl-4-chloro-1H-imidazol-5-yl)methyl]-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
3-(1H-benzimidazol-2-ylmethyl)-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
3-[1-(1H-benzimidazol-2-yl)ethyl]-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
3-[(5-chloro-1H-indol-3-yl)methyl]-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one and
3-[(4-fluoro-1H-indol-3-yl)methyl]-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
3-[2-(1H-Benzimidazol-2-yl)ethyl]-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
3-(1H-Pyrazol-3-ylmethyl)-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
3-[(5-Methylpyrazin-2-yl)methyl)-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
3-[(3-Isopropylisoxazol-5-yl)methyl]-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
3-[(4-Methyl-1,2,5-oxadiazol-3-yl)methyl]-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
3-[(6-Butoxypyridin-2-yl)methyl]-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
3-[(4-Butoxypyridin-2-yl)methyl]-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
3-[(3-Butoxypyridin-2-yl)methyl]-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
3-[2-(Pyridin-2-ylmethoxy)propyl]-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
3-[(3,5-Dimethylisoxazol-4-yl)methyl]-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
3-[(1-Methyl-1H-indol-2-yl)methyl]-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
3-(2-Phenyl-2-pyridin-2-ylethyl)-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
3-(Quinolin-4-ylmethyl)-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
3-[(6-Phenoxypyridin-3-yl)methyl]-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
3-{2-[(Quinolin-4-ylmethyl)amino]ethyl}-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
3-(2-{[(1-Methyl-1H-indol-3-yl)methyl]amino}ethyl)-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
3-{2-[Methyl(quinolin-4-ylmethyl)amino]ethyl}-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
3-(2-Aminopropyl)-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one trifluoroacetate;
3-{2-[(Pyridin-2-ylmethyl)amino]propyl}-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one trifluoroacetate;
3-{2-[(Pyridin-3-ylmethyl)amino]propyl}-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
3-{2-[(Pyridin-4-ylmethyl)amino]propyl}-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
3-(2-{[(6-Chloropyridin-3-yl)methyl]amino}propyl)-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one trifluoroacetate;
3-[2-({[6-(Trifluoromethyl)pyridin-3-yl]methyl}amino)propyl]-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one trifluoroacetate;
3-(2-{[(4,6-Dichloropyrimidin-5-yl)methyl]amino}propyl)-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
3-[2-({[2-(Dimethylamino)pyrimidin-5-yl]methyl}amino)propyl]-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
3-{2-[(Quinolin-2-ylmethyl)amino]propyl}-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one trifluoroacetate;
3-{2-[(Quinolin-3-ylmethyl)amino]propyl}-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
3-(2-{[(1-tert-Butyl-3,5-dimethyl-1H-pyrazol-4-yl)methyl]amino}propyl)-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
3-[2-({[1-(1,1-Dioxidotetrahydro-3-thienyl)-3,5-dimethyl-1H-pyrazol-4-yl]methyl}amino)propyl]-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
3-{2-[(1H-Benzoimidazol-2-ylmethyl)amino]propyl}-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
3-[2-({[1-(Phenylsulfonyl)-1H-pyrrol-2-yl]methyl}amino]propyl]-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one trifluoroacetate;
3-{2-[({1-[(4-methylphenyl)sulfonyl]-1H-pyrrol-2-yl}methyl)amino]propyl}-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one trifluoroacetate;
3-(2-{[(1-methyl-1H-pyrrol-2-yl)methyl]amino}propyl)-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
3-[2-({[1-(4-sec-Butylphenyl)-1H-pyrrol-2-yl]methyl}amino)propyl)-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
3-[2-({[1-(3-Methoxyphenyl)-1H-pyrrol-2-yl]methyl}amino)propyl]-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
3-[2-({[2,5-Dimethyl-1-(1,3-thiazol-2-yl)-1H-pyrrol-3-yl]methyl}amino)propyl]-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
3-[2-({[4-(3-Chlorobenzoyl)-1-methyl-1H-pyrrol-2-yl]methyl}amino)propyl]-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
3-{2-[(1H-Imidazol-2-ylmethyl)amino]propyl}-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
3-(2-{[(1-Methyl-1H-imidazol-2-yl)methyl]amino}propyl)-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
3-(2-{[(4-Bromo-1-methyl-1H-imidazol-5-yl)methyl]amino}propyl)-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
3-(2-{[(1-Methyl-1H-indol-3-yl)methyl]amino}propyl)-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
2-Thioxo-3-{2-[(1H-1,2,3-triazol-5-ylmethyl)amino]propyl}-1,2,3,7-tetrahydro-6H-purin-6-one;
3-[2-({[1-(Benzyloxy)-1H-imidazol-2-yl]methyl}amino)propyl]-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
3-(2-{[(6-Bromo-2-methylimidazo[1,2-a]pyridin-3-yl)methyl]amino}propyl}-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
3-{2-[({1-[2-(2-Methoxyphenoxy)ethyl]-1H-pyrrol-2-yl}methyl)amino]propyl]-2-thioxo-1,2,3,7-tetrahydro-6H-purin-6-one;
N-[1-Methyl-2-(6-oxo-2-thioxo-1,2,6,7-tetrahydro-3H-purin-3-yl)ethyl]pyridine-2-carboxamide;
N-[1-Methyl-2-(6-oxo-2-thioxo-1,2,6,7-tetrahydro-3H-purin-3-yl)ethyl]nicotinamide;
N-[1-Methyl-2-(6-oxo-2-thioxo-1,2,6,7-tetrahydro-3H-purin-3-yl)-ethyl]isonicotinamide;
N-[1-methyl-2-(6-oxo-2-thioxo-1,2,6,7-tetrahydro-3H-purin-3-yl)ethyl]-1,8-naphthyridine-2-carboxamide;
N-[1-Methyl-2-(6-oxo-2-thioxo-1,2,6,7-tetrahydro-3H-purin-3-yl)ethyl]quinoline-2-carboxamide;
N-[1-Methyl-2-(6-oxo-2-thioxo-1,2,6,7-tetrahydro-3H-purin-3-yl)ethyl]pyrimidine-2-carboxamide; and
N-[1-Methyl-2-(6-oxo-2-thioxo-1,2,6,7-tetrahydro-3H-purin-3-yl)ethyl]-1H-imidazole-2-carboxamide trifluroaceate.
14. A method according to any one of claims 8 to 10, wherein the daily dose of the MPO inhibitor or the pharmaceutically acceptable salt thereof is within the range of from 1 to 1000 mg.
US14/075,790 2007-08-23 2013-11-08 Use of myeloperoxidase (mpo) inhibitors or pharmaceutically acceptable salts thereof to treat multiple sysyem atrophy (msa) 938 Abandoned US20140221400A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US14/075,790 US20140221400A1 (en) 2007-08-23 2013-11-08 Use of myeloperoxidase (mpo) inhibitors or pharmaceutically acceptable salts thereof to treat multiple sysyem atrophy (msa) 938
US16/117,020 US10772890B2 (en) 2007-08-23 2018-08-30 Use of myeloperoxidase (MPO) inhibitors or pharmaceutically acceptable salts thereof to treat multiple system atrophy (MSA) 938
US17/019,276 US20200405724A1 (en) 2007-08-23 2020-09-13 Use of myeloperoxidase (mpo) inhibitors or pharmaceutically acceptable salts thereof to treat multiple system atrophy (msa) 938

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US95752307P 2007-08-23 2007-08-23
US95752507P 2007-08-23 2007-08-23
US12/195,527 US20090054468A1 (en) 2007-08-23 2008-08-21 New Use 938
US13/033,220 US20110207755A1 (en) 2007-08-23 2011-02-23 Use of myeloperoxidase (mpo) inhibitors or pharmaceutically acceptable salts thereof to treat multiple sysyem atrophy (msa) 938
US13/666,504 US20130059870A1 (en) 2007-08-23 2012-11-01 Use of myeloperoxidase (mpo) inhibitors or pharmaceutically acceptable salts thereof to treat multiple sysyem atrophy (msa) 938
US14/075,790 US20140221400A1 (en) 2007-08-23 2013-11-08 Use of myeloperoxidase (mpo) inhibitors or pharmaceutically acceptable salts thereof to treat multiple sysyem atrophy (msa) 938

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US13/666,504 Continuation US20130059870A1 (en) 2007-08-23 2012-11-01 Use of myeloperoxidase (mpo) inhibitors or pharmaceutically acceptable salts thereof to treat multiple sysyem atrophy (msa) 938

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US201715608453A Continuation 2007-08-23 2017-05-30

Publications (1)

Publication Number Publication Date
US20140221400A1 true US20140221400A1 (en) 2014-08-07

Family

ID=40378392

Family Applications (6)

Application Number Title Priority Date Filing Date
US12/195,527 Abandoned US20090054468A1 (en) 2007-08-23 2008-08-21 New Use 938
US13/033,220 Abandoned US20110207755A1 (en) 2007-08-23 2011-02-23 Use of myeloperoxidase (mpo) inhibitors or pharmaceutically acceptable salts thereof to treat multiple sysyem atrophy (msa) 938
US13/666,504 Abandoned US20130059870A1 (en) 2007-08-23 2012-11-01 Use of myeloperoxidase (mpo) inhibitors or pharmaceutically acceptable salts thereof to treat multiple sysyem atrophy (msa) 938
US14/075,790 Abandoned US20140221400A1 (en) 2007-08-23 2013-11-08 Use of myeloperoxidase (mpo) inhibitors or pharmaceutically acceptable salts thereof to treat multiple sysyem atrophy (msa) 938
US16/117,020 Active US10772890B2 (en) 2007-08-23 2018-08-30 Use of myeloperoxidase (MPO) inhibitors or pharmaceutically acceptable salts thereof to treat multiple system atrophy (MSA) 938
US17/019,276 Pending US20200405724A1 (en) 2007-08-23 2020-09-13 Use of myeloperoxidase (mpo) inhibitors or pharmaceutically acceptable salts thereof to treat multiple system atrophy (msa) 938

Family Applications Before (3)

Application Number Title Priority Date Filing Date
US12/195,527 Abandoned US20090054468A1 (en) 2007-08-23 2008-08-21 New Use 938
US13/033,220 Abandoned US20110207755A1 (en) 2007-08-23 2011-02-23 Use of myeloperoxidase (mpo) inhibitors or pharmaceutically acceptable salts thereof to treat multiple sysyem atrophy (msa) 938
US13/666,504 Abandoned US20130059870A1 (en) 2007-08-23 2012-11-01 Use of myeloperoxidase (mpo) inhibitors or pharmaceutically acceptable salts thereof to treat multiple sysyem atrophy (msa) 938

Family Applications After (2)

Application Number Title Priority Date Filing Date
US16/117,020 Active US10772890B2 (en) 2007-08-23 2018-08-30 Use of myeloperoxidase (MPO) inhibitors or pharmaceutically acceptable salts thereof to treat multiple system atrophy (MSA) 938
US17/019,276 Pending US20200405724A1 (en) 2007-08-23 2020-09-13 Use of myeloperoxidase (mpo) inhibitors or pharmaceutically acceptable salts thereof to treat multiple system atrophy (msa) 938

Country Status (26)

Country Link
US (6) US20090054468A1 (en)
EP (1) EP2200616B1 (en)
JP (2) JP5247804B2 (en)
KR (1) KR101588464B1 (en)
CN (4) CN105687199A (en)
AU (1) AU2008289653B2 (en)
BR (1) BRPI0815681A2 (en)
CA (1) CA2698205C (en)
CY (1) CY1113714T1 (en)
DK (1) DK2200616T3 (en)
EA (1) EA017510B1 (en)
ES (1) ES2399846T3 (en)
HK (1) HK1145141A1 (en)
HR (1) HRP20130139T1 (en)
IL (2) IL203738A (en)
ME (1) ME01510B (en)
MX (1) MX2010001983A (en)
MY (1) MY155633A (en)
NZ (1) NZ584135A (en)
PH (1) PH12014501180A1 (en)
PL (1) PL2200616T3 (en)
PT (1) PT2200616E (en)
RS (1) RS52665B (en)
SI (1) SI2200616T1 (en)
WO (1) WO2009025618A1 (en)
ZA (1) ZA201000848B (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9580429B2 (en) 2004-12-06 2017-02-28 Astrazeneca Ab Pyrrolo[3,2-D]pyrimidin-4-one derivatives and their use in therapy

Families Citing this family (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AR039385A1 (en) * 2002-04-19 2005-02-16 Astrazeneca Ab THIOXANTINE DERIVATIVES AS INHIBITORS OF MIELOPEROXIDASA
TW200806667A (en) * 2006-04-13 2008-02-01 Astrazeneca Ab New compounds
TW200804383A (en) * 2006-06-05 2008-01-16 Astrazeneca Ab New compounds
US20090054468A1 (en) * 2007-08-23 2009-02-26 Astrazeneca Ab New Use 938
US8927559B2 (en) 2010-10-11 2015-01-06 Merck Sharp & Dohme Corp. Quinazolinone-type compounds as CRTH2 antagonists
CA2853024C (en) 2011-11-11 2017-08-22 Pfizer Inc. 2-thiopyrimidinones
CN104220440B (en) * 2012-03-29 2017-07-07 巴斯夫欧洲公司 Prevent and treat the Heterobicyclic compounds and derivative I I of the N substitutions of animal pest
US9616063B2 (en) * 2014-12-01 2017-04-11 Astrazeneca Ab 1-[2-(aminomethyl)benzyl]-2-thioxo-1,2,3,5-tetrahydro-4H-pyrrolo[3,2-d]pyrimidin-4-ones as inhibitors of myeloperoxidase
EP3292109A1 (en) 2015-05-05 2018-03-14 Pfizer Inc 2-thiopyrimidinones
BR112019027717A2 (en) * 2017-06-28 2020-07-28 Ptc Therapeutics, Inc. methods to treat huntington's disease
KR20200029513A (en) * 2017-07-17 2020-03-18 아스트라제네카 아베 MPO inhibitors for use in medicine
CN114945573A (en) * 2019-10-10 2022-08-26 拜奥海芬治疗学有限公司 Prodrugs of myeloperoxidase inhibitors
WO2021178734A1 (en) * 2020-03-05 2021-09-10 Biohaven Therapeutics Ltd. Method of treating amyotrophic lateral sclerosis with myeloperoxidase inhibitor
US20230192702A1 (en) * 2020-05-06 2023-06-22 Biohaven Therapeutics Ltd. Process for the preparation of verdiperstat
CN115403584B (en) * 2021-05-26 2024-04-02 长春金赛药业有限责任公司 2-thio-2, 3-dihydropyrimidine-4-one derivatives, pharmaceutical compositions, preparation methods and applications thereof
WO2024038131A1 (en) 2022-08-18 2024-02-22 Astrazeneca Ab Inhibitors of myeloperoxidase

Family Cites Families (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999012546A1 (en) * 1997-09-05 1999-03-18 Kyowa Hakko Kogyo Co., Ltd. Remedial agent for neural degeneration
GB0100620D0 (en) * 2001-01-10 2001-02-21 Vernalis Res Ltd Chemical cokpounds V
GB0100623D0 (en) * 2001-01-10 2001-02-21 Vernalis Res Ltd Chemical compounds IV
EP2942082B1 (en) * 2002-01-28 2019-03-06 Kyowa Hakko Kogyo Co., Ltd A2a receptor antogonists for use in the treatment of movement disorders
AR039385A1 (en) * 2002-04-19 2005-02-16 Astrazeneca Ab THIOXANTINE DERIVATIVES AS INHIBITORS OF MIELOPEROXIDASA
SE0302756D0 (en) * 2003-10-17 2003-10-17 Astrazeneca Ab Novel Compounds
MY140748A (en) * 2004-12-06 2010-01-15 Astrazeneca Ab Novel pyrrolo [3,2-d] pyrimidin-4-one derivatives and their use in therapy
TW200806667A (en) * 2006-04-13 2008-02-01 Astrazeneca Ab New compounds
CN101460501A (en) * 2006-06-05 2009-06-17 阿斯利康(瑞典)有限公司 Pyrrolo[3,2-d]pyrimidin-4-one derivative as myeloperoxidase inhibitor
AR066936A1 (en) * 2007-06-13 2009-09-23 Astrazeneca Ab 3 - (2R - TETRAHYDROFURIL - METHYL) - 2 - THIOXANTINE. PHARMACEUTICAL COMPOSITIONS.
US20090054468A1 (en) * 2007-08-23 2009-02-26 Astrazeneca Ab New Use 938

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Novak et al. "Treatment of Multiple System Atrophy Using Intravenous Immunoglobulin". BMC Neurology. 2012; 12:131-137. *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9580429B2 (en) 2004-12-06 2017-02-28 Astrazeneca Ab Pyrrolo[3,2-D]pyrimidin-4-one derivatives and their use in therapy

Also Published As

Publication number Publication date
AU2008289653A1 (en) 2009-02-26
MY155633A (en) 2015-11-13
EA017510B1 (en) 2013-01-30
NZ584135A (en) 2012-04-27
CN113633641A (en) 2021-11-12
DK2200616T3 (en) 2013-03-04
BRPI0815681A2 (en) 2015-02-18
EP2200616B1 (en) 2012-12-26
CA2698205C (en) 2016-06-21
US20090054468A1 (en) 2009-02-26
MX2010001983A (en) 2010-03-10
WO2009025618A1 (en) 2009-02-26
ME01510B (en) 2014-04-20
KR101588464B1 (en) 2016-01-25
US20190099423A1 (en) 2019-04-04
JP5247804B2 (en) 2013-07-24
PT2200616E (en) 2013-02-27
CN101873857A (en) 2010-10-27
EA201000202A1 (en) 2010-10-29
EP2200616A4 (en) 2010-08-25
JP2010536849A (en) 2010-12-02
AU2008289653B2 (en) 2012-06-28
HK1145141A1 (en) 2011-04-08
US10772890B2 (en) 2020-09-15
US20110207755A1 (en) 2011-08-25
ES2399846T3 (en) 2013-04-03
JP2013151547A (en) 2013-08-08
PL2200616T3 (en) 2013-04-30
HRP20130139T1 (en) 2013-03-31
IL240130A0 (en) 2015-09-24
PH12014501180A1 (en) 2015-08-24
US20130059870A1 (en) 2013-03-07
ZA201000848B (en) 2012-07-25
CN105687199A (en) 2016-06-22
EP2200616A1 (en) 2010-06-30
SI2200616T1 (en) 2013-03-29
CN103638029A (en) 2014-03-19
KR20100055434A (en) 2010-05-26
RS52665B (en) 2013-06-28
CY1113714T1 (en) 2016-06-22
CA2698205A1 (en) 2009-02-26
IL203738A (en) 2015-08-31
US20200405724A1 (en) 2020-12-31

Similar Documents

Publication Publication Date Title
US20200405724A1 (en) Use of myeloperoxidase (mpo) inhibitors or pharmaceutically acceptable salts thereof to treat multiple system atrophy (msa) 938
Liou et al. Mediation of cannabidiol anti-inflammation in the retina by equilibrative nucleoside transporter and A2A adenosine receptor
EP2916868B1 (en) Pharmaceutical compositions containing a pde4 inhibitor and a pi3 delta or dual pi3 delta-gamma kinase inhibitor
US8592420B2 (en) Method of treating an anxiety disorder
US11779565B2 (en) Small organic molecules for use in the treatment of neuroinflammatory disorders
US20090053176A1 (en) New Combination 937
RU2432161C1 (en) New combinations of neramexane for treating neurodegenerative disorders
JP2004505048A (en) Use of selective adenosine A1 receptor agonists, antagonists and allosteric enhancers to treat angiogenesis
US9956202B2 (en) Use of indolyl and indolinyl hydroxamates for treating neurodegenerative disorders or cognitive decicits
US20120094985A1 (en) Pharmaceutical compositions comprising chlorophenyl piperazine derived compounds and use of the compounds in producing medicaments
US20220257629A1 (en) Compositions and methods for upregulation of human fetal hemoglobin
JPWO2020180980A5 (en)
Lapin et al. Antiethanol effects of indol-3-ylpyruvic acid in mice
US20110046141A1 (en) Use of pde5 inhibitors for treating circadian rhythm disorders
KR20010040671A (en) Use of 2-amino-6-trifluoromethoxy-benzothiazole for preventing or treating cerebellum dysfunction

Legal Events

Date Code Title Description
AS Assignment

Owner name: ASTRAZENECA AB, SWEDEN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:ERIKSSON, HAKAN;POEWE, WERNER;SIGNING DATES FROM 20080828 TO 20080901;REEL/FRAME:032498/0704

STCB Information on status: application discontinuation

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