WO2008128983A1 - Cyano-guanidine derivatives as glutaminyl cyclase inhibitors - Google Patents

Cyano-guanidine derivatives as glutaminyl cyclase inhibitors Download PDF

Info

Publication number
WO2008128983A1
WO2008128983A1 PCT/EP2008/054712 EP2008054712W WO2008128983A1 WO 2008128983 A1 WO2008128983 A1 WO 2008128983A1 EP 2008054712 W EP2008054712 W EP 2008054712W WO 2008128983 A1 WO2008128983 A1 WO 2008128983A1
Authority
WO
WIPO (PCT)
Prior art keywords
methyl
alkyl
imidazol
cyano
propyl
Prior art date
Application number
PCT/EP2008/054712
Other languages
French (fr)
Inventor
Mirko Buchholz
Ulrich Heiser
Torsten Hoffmann
Livia BÖHME
Original Assignee
Probiodrug Ag
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 Probiodrug Ag filed Critical Probiodrug Ag
Priority to JP2010503512A priority Critical patent/JP5675342B2/en
Priority to DK08736361.0T priority patent/DK2160380T3/en
Priority to ES08736361.0T priority patent/ES2474693T3/en
Priority to EP08736361.0A priority patent/EP2160380B1/en
Publication of WO2008128983A1 publication Critical patent/WO2008128983A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D233/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings
    • C07D233/54Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members
    • C07D233/56Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members with only hydrogen atoms or radicals containing only hydrogen and carbon atoms, attached to ring carbon atoms
    • C07D233/61Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members with only hydrogen atoms or radicals containing only hydrogen and carbon atoms, attached to ring carbon atoms with hydrocarbon radicals, substituted by nitrogen atoms not forming part of a nitro radical, attached to ring nitrogen atoms
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/14Prodigestives, e.g. acids, enzymes, appetite stimulants, antidyspeptics, tonics, antiflatulents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/18Drugs for disorders of the alimentary tract or the digestive system for pancreatic disorders, e.g. pancreatic enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/08Drugs for genital or sexual disorders; Contraceptives for gonadal disorders or for enhancing fertility, e.g. inducers of ovulation or of spermatogenesis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/02Drugs for disorders of the nervous system for peripheral neuropathies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/18Antipsychotics, i.e. neuroleptics; Drugs for mania or schizophrenia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/20Hypnotics; Sedatives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/22Anxiolytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/24Antidepressants
    • 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
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D233/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings
    • C07D233/54Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members
    • C07D233/64Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members with substituted hydrocarbon radicals attached to ring carbon atoms, e.g. histidine
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D235/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings
    • C07D235/02Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings condensed with carbocyclic rings or ring systems
    • C07D235/04Benzimidazoles; Hydrogenated benzimidazoles
    • C07D235/06Benzimidazoles; Hydrogenated benzimidazoles with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached in position 2
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/12Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a chain containing hetero atoms as chain links

Definitions

  • the invention relates to novel cyanoguanidine derivatives as inhibitors of glutaminyl cyclase (QC, EC 2.3.2.5).
  • QC catalyzes the intramolecular cyclization of N-terminal glutamine residues into pyroglutamic acid (5-oxo-prolyl, pGlu * ) under liberation of ammonia and the intramolecular cyclization of N-terminal glutamate residues into pyroglutamic acid under liberation of water.
  • Glutaminyl cyclase catalyzes the intramolecular cyclization of N-terminal glutamine residues into pyroglutamic acid (pGlu * ) liberating ammonia.
  • pGlu * pyroglutamic acid
  • EP 02 01 1 349.4 discloses polynucleotides encoding insect glutaminyl cyclase, as well as polypeptides encoded thereby and their use in methods of screening for agents that reduce glutaminyl cyclase activity. Such agents are useful as pesticides.
  • k or “Kf and "K 0” are binding constants, which describe the binding of an inhibitor to and the subsequent release from an enzyme. Another measure is the “IC 50 " value, which reflects the inhibitor concentration, which at a given substrate concentration results in 50 % enzyme activity.
  • DP IV-inhibitor or "dipeptidyl peptidase IV inhibitor” is generally known to a person skilled in the art and means enzyme inhibitors, which inhibit the catalytic activity of DP IV or DP IV-like enzymes.
  • DP IV-activity is defined as the catalytic activity of dipeptidyl peptidase IV (DP IV) and DP IV- like enzymes. These enzymes are post-proline (to a lesser extent post-alanine, post-serine or post-glycine) cleaving serine proteases found in various tissues of the body of a mammal including kidney, liver, and intestine, where they remove dipeptides from the N-terminus of biologically active peptides with a high specificity when proline or alanine form the residues that are adjacent to the N-terminal amino acid in their sequence.
  • PEP-inhibitor or "prolyl endopeptidase inhibitor” is generally known to a person skilled in the art and means enzyme inhibitors, which inhibit the catalytic activity of prolyl endopeptidase (PEP, prolyl oligopeptidase, POP).
  • PEP-activity is defined as the catalytic activity of an endoprotease that is capable to hydrolyze post proline bonds in peptides or proteins were the proline is in amino acid position 3 or higher counted from the N-terminus of a peptide or protein substrate.
  • QC as used herein comprises glutaminyl cyclase (QC) and QC-like enzymes.
  • QC and QC-like enzymes have identical or similar enzymatic activity, further defined as QC activity.
  • QC-like enzymes can fundamentally differ in their molecular structure from QC.
  • QC-like enzymes are the glutaminyl-peptide cyclotransferase-like proteins (QPCTLs) from human (GenBank NM_017659), mouse (GenBank BC058181 ), Macaca fascicularis (GenBank AB168255), Macaca mulatta (GenBank XM_001 110995), Canis familiaris (GenBank XM_541552), Rattus norvegicus (GenBank XM_001066591 ), Mus musculus (GenBank BC058181 ) and Bos taurus (GenBank BT026254).
  • QPCTLs glutaminyl-peptide cyclotransferase-like proteins
  • QC activity is defined as intramolecular cyclization of N-terminal glutamine residues into pyroglutamic acid (pGlu * ) or of N-terminal L-homoglutamine or L- ⁇ - homoglutamine to a cyclic pyro-homoglutamine derivative under liberation of ammonia. See therefore schemes 1 and 2.
  • EC as used herein comprises the activity of QC and QC-like enzymes as glutamate cyclase (EC), further defined as EC activity.
  • EC activity as used herein is defined as intramolecular cyclization of N-terminal glutamate residues into pyroglutamic acid (pGlu * ) by QC. See therefore scheme 3.
  • QC-inhibitor "glutaminyl cyclase inhibitor” is generally known to a person skilled in the art and means enzyme inhibitors, which inhibit the catalytic activity of glutaminyl cyclase (QC) or its glutamyl cyclase (EC) activity.
  • the subject method and medical use utilize an agent with an IC 5 O for QC inhibition of 10 ⁇ M or less, more preferably of 1 ⁇ M or less, even more preferably of 0.1 ⁇ M or less or 0.01 ⁇ M or less, or most preferably 0.001 ⁇ M or less.
  • an agent with an IC 5 O for QC inhibition of 10 ⁇ M or less, more preferably of 1 ⁇ M or less, even more preferably of 0.1 ⁇ M or less or 0.01 ⁇ M or less, or most preferably 0.001 ⁇ M or less.
  • inhibitors with K, values in the lower micromolar, preferably the nanomolar and even more preferably the picomolar range are contemplated.
  • active agents are described herein, for convenience, as "QC inhibitors", it will be understood that such nomenclature is not intending to limit the subject of the invention to a particular mechanism of action.
  • the QC inhibitors of the subject method or medical use will be small molecules, e.g., with molecular weights of 500 g/mole or less, 400 g/mole or less, preferably of 350 g/mole or less, and even more preferably of 300 g/mole or less and even of 250 g/mole or less.
  • subject refers to an animal, preferably a mammal, most preferably a human, who has been the object of treatment, observation or experiment.
  • terapéuticaally effective amount means that amount of active compound or pharmaceutical agent that elicits the biological or medicinal response in a tissue system, animal or human being sought by a researcher, veterinarian, medical doctor or other clinician, which includes alleviation of the symptoms of the disease or disorder being treated.
  • the term “pharmaceutically acceptable” embraces both human and veterinary use:
  • pharmaceutically acceptable embraces a veterinarily acceptable compound or a compound acceptable in human medicine and health care.
  • alkyl denotes a C M2 alkyl group, suitably a Ci -6 alkyl group, e.g. Ci -4 alkyl group.
  • Alkyl groups may be straight chain or branched. Suitable alkyl groups include, for example, methyl, ethyl, propyl (e.g. n-propyl and isopropyl), butyl (e.g n-butyl, iso-butyl, sec-butyl and tert-butyl), pentyl (e.g. n- pentyl), hexyl (e.g. n-hexyl), heptyl (e.g.
  • alk for example in the expressions "alkoxy”, “haloalkyl” and “thioalkyl” should be interpreted in accordance with the definition of "alkyl".
  • alkoxy groups include methoxy, ethoxy, propoxy (e.g. n-propoxy), butoxy (e.g. n-butoxy), pentoxy (e.g. n-pentoxy), hexoxy (e.g. n- hexoxy), heptoxy (e.g. n-heptoxy) and octoxy (e.g. n-octoxy).
  • exemplary thioalkyl groups include methylthio-.
  • haloalkyl groups include fluoroalkyl e.g. CF 3 .
  • alkenyl denotes a C 2- i 2 alkenyl group, suitably a C 2 -6 alkenyl group, e.g. a C 2-4 alkenyl group, which contains at least one double bond at any desired location and which does not contain any triple bonds.
  • Alkenyl groups may be straight chain or branched.
  • Exemplary alkenyl groups including one double bond include propenyl and butenyl.
  • Exemplary alkenyl groups including two double bonds include pentadienyl, e.g. (1 E, 3E)-pentadienyl.
  • alkynyl denotes a C 2- i 2 alkynyl group, suitably a C 2 -6 alkynyl group, e.g. a C 2-4 alkynyl group, which contains at least one triple bond at any desired location and may or may not also contain one or more double bonds.
  • Alkynyl groups may be straight chain or branched.
  • Exemplary alkynyl groups include propynyl and butynyl.
  • alkylene denotes a chain of formula -(CH 2 ) n - wherein n is an integer e.g. 2-5, unless specifically limited.
  • cycloalkyl denotes a C 3- i 0 cycloalkyl group (i.e. 3 to 10 ring carbon atoms), more suitably a C 3- S cycloalkyl group, e.g. a C 3- 6 cycloalkyl group.
  • exemplary cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and cyclooctyl.
  • a most suitable number of ring carbon atoms is three to six.
  • cycloalkenyl denotes a C 5- io cycloalkenyl group (i.e. 5 to 10 ring carbon atoms), more suitably a C 5-8 cycloalkenyl group e.g. a C 5-6 cycloalkenyl group.
  • exemplary cycloalkenyl groups include cyclopropenyl, cyclohexenyl, cycloheptenyl and cyclooctenyl.
  • a most suitable number of ring carbon atoms is five to six.
  • Carbocyclyl denotes any ring system in which all the ring atoms are carbon and which contains between three and twelve ring carbon atoms, suitably between three and ten carbon atoms and more suitably between three and eight carbon atoms.
  • Carbocyclyl groups may be saturated or partially unsaturated, but do not include aromatic rings. Examples of carbocyclyl groups include monocyclic, bicyclic, and tricyclic ring systems, in particular monocyclic and bicyclic ring systems. Other carbocylcyl groups include bridged ring systems (e.g. bicyclo[2.2.1]heptenyl).
  • a specific example of a carbocyclyl group is a cycloalkyl group. A further example of a carbocyclyl group is a cycloalkenyl group.
  • heterocyclyl refers to a carbocyclyl group wherein one or more (e.g. 1 , 2 or 3) ring atoms are replaced by heteroatoms selected from N, S and O.
  • a specific example of a heterocyclyl group is a cycloalkyl group (e.g. cyclopentyl or more particularly cyclohexyl) wherein one or more (e.g. 1 , 2 or 3, particularly 1 or 2, especially 1 ) ring atoms are replaced by heteroatoms selected from N, S or O.
  • exemplary heterocyclyl groups containing one hetero atom include pyrrolidine, tetrahydrofuran and piperidine, and exemplary heterocyclyl groups containing two hetero atoms include morpholine and piperazine.
  • a further specific example of a heterocyclyl group is a cycloalkenyl group (e.g.
  • a cyclohexenyl group wherein one or more (e.g. 1 , 2 or 3, particularly 1 or 2, especially 1 ) ring atoms are replaced by heteroatoms selected from N, S and O.
  • An example of such a group is dihydropyranyl (e.g. 3,4- dihydro-2H-pyran-2-yl-).
  • aryl denotes a C 6- i 2 aryl group, suitably a C 6- io aryl group, more suitably a C 6- S aryl group.
  • Aryl groups will contain at least one aromatic ring (e.g. one, two or three rings).
  • An example of a typical aryl group with one aromatic ring is phenyl.
  • An example of a typical aryl group with two aromatic rings is naphthyl.
  • heteroaryl unless specifically limited, denotes an aryl residue, wherein one or more (e.g. 1 , 2, 3, or 4, suitably 1 , 2 or 3) ring atoms are replaced by heteroatoms selected from
  • Exemplary monocyclic heteroaryl groups having one heteroatom include: five membered rings (e.g. pyrrole, furan, thiophene); and six membered rings (e.g. pyridine, such as pyridin-2-yl, pyridin-3-yl and pyridin-4-yl).
  • Exemplary monocyclic heteroaryl groups having two heteroatoms include: five membered rings
  • pyrazole e.g. pyrazole, oxazole, isoxazole, thiazole, isothiazole, imidazole, such as imidazol-1-yl, imidazol-2-yl imidazol-4-yl
  • six membered rings e.g. pyridazine, pyrimidine, pyrazine.
  • Exemplary monocyclic heteroaryl groups having three heteroatoms include: 1 ,2,3-triazole and
  • Exemplary monocyclic heteroaryl groups having four heteroatoms include tetrazole.
  • Exemplary bicyclic heteroaryl groups include: indole (e.g. indol-6-yl), benzofuran, benzthiophene, quinoline, isoquinoline, indazole, benzimidazole, benzthiazole, quinazoline and purine.
  • alkylaryl unless specifically limited, denotes an aryl residue which is connected via an alkylene moiety e.g. a Ci -4 alkylene moiety.
  • alkylheteroaryl unless specifically limited, denotes a heteroaryl residue which is connected via an alkylene moiety e.g. a C-i ⁇ alkylene moiety.
  • halogen or “halo” comprises fluorine (F), chlorine (Cl) and bromine (Br).
  • amino refers to the group -NH 2 .
  • phenyl substituted by phenyl refers to biphenyl.
  • the compounds according to this invention may accordingly exist as enantiomers. Where the compounds possess two or more chiral centers, they may additionally exist as diastereomers. It is to be understood that all such isomers and mixtures thereof are encompassed within the scope of the present invention.
  • stereoisomers Where the processes for the preparation of the compounds according to the invention give rise to a mixture of stereoisomers, these isomers may be separated by conventional techniques such as preparative chromatography.
  • the compounds may be prepared in racemic form, or individual enantiomers may be prepared either by enantiospecific synthesis or by resolution.
  • the compounds may, for example, be resolved into their components enantiomers by standard techniques, such as the formation of diastereomeric pairs by salt formation with an optically active acid, such as (-)-di-p-toluoyl-d-tartaric acid and/or (+)-di-p-toluoyl-l-tartaric acid followed by fractional crystallization and regeneration of the free base.
  • the compounds may also be resolved by formation of diastereomeric esters or amides, followed by chromatographic separation and removal of the chiral auxiliary. Alternatively, the compounds may be resolved using a chiral HPLC column.
  • Salts and solvates of the compounds of formula (I) and physiologically functional derivatives thereof which are suitable for use in medicine are those wherein the counter-ion or associated solvent is pharmaceutically acceptable.
  • salts and solvates having non- pharmaceutically acceptable counter-ions or associated solvents are within the scope of the present invention, for example, for use as intermediates in the preparation of other compounds and their pharmaceutically acceptable salts and solvates.
  • Suitable salts according to the invention include those formed with both organic and inorganic acids or bases.
  • Pharmaceutically acceptable acid addition salts include those formed from hydrochloric, hydrobromic, sulfuric, nitric, citric, tartaric, phosphoric, lactic, pyruvic, acetic, trifluoroacetic, triphenylacetic, sulfamic, sulfanilic, succinic, oxalic, fumaric, maleic, malic, mandelic, glutamic, aspartic, oxaloacetic, methanesulfonic, ethanesulfonic, arylsulfonic (for example p-toluenesulfonic, benzenesulfonic, naphthalenesulfonic or naphthalenedisulfonic), salicylic, glutaric, gluconic, tricarballylic, cinnamic, substituted cinnamic (for example, phenyl
  • Pharmaceutically acceptable base salts include ammonium salts, alkali metal salts such as those of sodium and potassium, alkaline earth metal salts such as those of calcium and magnesium and salts with organic bases such as dicyclohexylamine and ⁇ /-methyl-D-glucamine.
  • Polymorph crystal forms Furthermore, some of the crystalline forms of the compounds may exist as polymorphs and as such are intended to be included in the present invention. In addition, some of the compounds may form solvates with water (i.e. hydrates) or common organic solvents, and such solvates are also intended to be encompassed within the scope of this invention.
  • the compounds, including their salts, can also be obtained in the form of their hydrates, or include other solvents used for their crystallization.
  • the present invention further includes within its scope prodrugs of the compounds of this invention.
  • prodrugs will be functional derivatives of the compounds which are readily convertible in vivo into the desired therapeutically active compound.
  • the term “administering” shall encompass the treatment of the various disorders described with prodrug versions of one or more of the claimed compounds, but which converts to the above specified compound in vivo after administration to the subject. Conventional procedures for the selection and preparation of suitable prodrug derivatives are described, for example, in "Design of Prodrugs", ed. H. Bundgaard, Elsevier, 1985.
  • any of the processes for preparation of the compounds of the present invention it may be necessary and/or desirable to protect sensitive or reactive groups on any of the molecules concerned. This may be achieved by means of conventional protecting groups, such as those described in Protective Groups in Organic Chemistry, ed. J.F.W. McOmie, Plenum Press, 1973; and T.W. Greene & P. G. M. Wuts, Protective Groups in Organic Synthesis, John Wiley & Sons, 1991 , fully incorporated herein by reference.
  • the protecting groups may be removed at a convenient subsequent stage using methods known from the art.
  • composition is intended to encompass a product comprising the claimed compounds in the therapeutically effective amounts, as well as any product which results, directly or indirectly, from combinations of the claimed compounds.
  • suitable carriers and additives may advantageously include water, glycols, oils, alcohols, flavoring agents, preservatives, coloring agents and the like;
  • suitable carriers and additives include starches, sugars, diluents, granulating agents, lubricants, binders, disintegrating agents and the like.
  • Carriers which can be added to the mixture, include necessary and inert pharmaceutical excipients, including, but not limited to, suitable binders, suspending agents, lubricants, flavorants, sweeteners, preservatives, coatings, disintegrating agents, dyes and coloring agents.
  • Soluble polymers as targetable drug carriers can include polyvinylpyrrolidone, pyran copolymer, polyhydroxypropylmethacrylamidephenol, polyhydroxyethylaspartamide-phenol, or polyethyleneoxidepolyllysine substituted with palmitoyl residue.
  • the compounds of the present invention may be coupled to a class of biodegradable polymers useful in achieving controlled release of a drug, for example, polyactic acid, polyepsilon caprolactone, polyhydroxy butyeric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacrylates and cross- linked or amphipathic block copolymers of hydrogels.
  • Suitable binders include, without limitation, starch, gelatin, natural sugars such as glucose or betalactose, corn sweeteners, natural and synthetic gums such as acacia, tragacanth or sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride and the like.
  • Disintegrators include, without limitation, starch, methyl cellulose, agar, bentonite, xanthan gum and the like.
  • R 1 represents alkyl; alkenyl, wherein the double bond is not adjacent to the nitrogen; carbocyclyl; -Ci -6 alkyl-carbocyclyl; heterocyclyl; -Ci -6 alkyl-heterocyclyl; aryl; heteroaryl; -Ci- 6 alkylaryl; -Ci -6 alkylheteroaryl; -phenyl fused to carbocyclyl or -phenyl fused to heterocyclyl; in which any of the aforesaid carbocyclyl and heterocyclyl groups may optionally be substituted by one or more groups selected from methyl and oxo; and in which any of the aforesaid phenyl, aryl and heteroaryl groups may optionally be substituted by one or more substituents selected from Ci -6 alkyl, C 2- 6al
  • R 1 represents phenyl substituted by phenyl, or phenyl substituted by an optionally substituted monocyclic heteroaryl group in which any of the aforesaid phenyl and monocyclic heteroaryl groups may optionally be substituted by one or more groups selected from Ci -4 alkyl, halogen and Ci -4 alkoxy; or R 1 represents phenyl substituted by benzyloxy- in which any of the aforesaid phenyl or benzyloxy groups may optionally be substituted on the ring by one or more groups selected from Ci -4 alkyl, halogen and Ci -4 alkoxy; and
  • A represents
  • Y represents a C 2-5 alkylene chain, which may optionally be substituted by one or two methyl groups or may optionally be substituted by two alkylene substituents at the same position wherein the two alkylene substituents are joined to each other to form a C 3-5 spiro- cycloalkyl group and
  • R 2 , R 3 and R 4 independently represent H or Ci -2 alkyl, provided that R 2 and R 3 and R 4 do not all represent H; or A represents
  • Z represents a bond, -CH 2 -, -CH 2 -CH 2 -, -CH(Me)-, -CH(Me)-CH 2 - Or -CH 2 -CH(Me)- and
  • R 5 and R 6 independently represent H or Ci -2 alkyl
  • R 1 represents alkyl; alkenyl, wherein the double bond is not adjacent to the nitrogen; carbocyclyl; -Ci -6 alkyl-carbocyclyl; heterocyclyl; -Ci -6 alkyl-heterocyclyl; aryl; heteroaryl; -Ci- 6 alkylaryl; -Ci -6 alkylheteroaryl; -phenyl fused to carbocyclyl or -phenyl fused to heterocyclyl; in which any of the aforesaid carbocyclyl and heterocyclyl groups may optionally be substituted by one or more groups selected from methyl and oxo; and in which any of the aforesaid phenyl, aryl and heteroaryl groups may optionally be substituted by one or more substituents selected from Ci -6 alkyl, C 2- 6alkenyl, C 2- 6alkynyl, Ci- 6 haloalkyl
  • R 1 represents phenyl substituted by phenyl, or phenyl substituted by an optionally substituted monocyclic heteroaryl group in which any of the aforesaid phenyl and monocyclic heteroaryl groups may optionally be substituted by one or more groups selected from Ci -4 alkyl, halogen and Ci -4 alkoxy.
  • carbocyclyl and heterocyclyl When carbocyclyl and heterocyclyl are substituted, they are typically substituted by 1 or 2 substituents (e.g. 1 substitent). Typically the substituent is methyl. More typically carbocyclyl and heterocyclyl groups are unsubstituted.
  • aryl and heteroaryl When aryl and heteroaryl are substituted, they are typically substituted by 1 , 2 or 3 (e.g. 1 or 2) substituents.
  • Substituents for aryl and heteroaryl are selected from Ci -6 alkyl (e.g. methyl), C 2- 6 alkenyl (e.g. buten-3-yl), C 2-6 alkynyl (e.g. butyn-3-yl), Ci -6 haloalkyl (e.g. fluoromethyl, trifluoromethyl), -Ci -6 thioalkyl (e.g. -S-methyl), -SO 2 Ci -4 alkyl (e.g. -SO 2 methyl), Ci -6 alkoxy- (e.g.
  • -O-C 3-8 cycloalkyl e.g. -O-cyclopentyl
  • C 3-8 cycloalkyl e.g. cyclopropyl, cyclohexyl
  • -SO 2 C 3-8 cycloalkyl e.g. -SO 2 cyclohexyl
  • C 3-6 alkenyloxy- e.g. -O-buten-2-yl
  • C 3- 6 alkynyloxy- e.g. -O-buten-2-yl
  • -C(O)d -6 alkyl e.g.
  • -C(O)ethyl -C(O)OCi -6 alkyl (e.g. -C(O)O- methyl), d-ealkoxy-d-ealkyl- (e.g. methoxy-ethyl-), nitro, halogen (e.g. fluoro, chloro, bromo), cyano, hydroxyl, -C(O)OH, -NH 2 , -NHCi -4 alkyl (e.g. -NHmethyl), -N(C 1-4 alkyl)(C 1-4 alkyl) (e.g.
  • Ci -6 haloalkyl e.g. Ci -6 fluoroalkyl, e.g. CF 3
  • Ci -6 alkoxy e.g. OMe
  • halogen hydroxy and cyano.
  • substituents will be selected from Ci -6 alkyl (e.g. methyl), Ci -6 haloalkyl (e.g. Ci -6 fluoroalkyl, e.g. CF 3 ), Ci -6 alkoxy (e.g. OMe), halogen and hydroxy.
  • R 1 represents alkyl
  • examples include propyl (e.g. n-propyl, isopropyl), butyl (e.g. n-butyl- sec-butyl, isobutyl and tert-butyl), pentyl (e.g. n-pentyl, 3,3,-dimethylpropyl), hexyl, heptyl and octyl.
  • R 1 represents alkenyl
  • R 1 represents carbocyclyl (which may optionally be substituted)
  • examples include cycloalkyl and cycloalkenyl.
  • examples of cycloalkyl include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and cycloheptyl.
  • Examples of cycloalkenyl include cyclohexenyl (e.g. cyclohex-2- enyl, cyclohex-3-enyl).
  • substituted carbocyclyl examples include 2-methyl-cyclohexyl-, 3- methyl-cyclohexyl-, 4-methyl-cyclohexyl-, 2-methyl-cyclohex-2-enyl, 2-methyl-cyclohex-3-enyl, S-methyl-cyclohex-S-enyl, 3-methyl-cyclohex-3-enyl.
  • R 1 represents -C-i- ⁇ alkyl-carbocyclyl (which may optionally be substituted)
  • examples include -methyl-cyclopentyl, -methyl-cyclohexyl, -ethyl-cyclohexyl, -propyl-cyclohexyl, -methyl- cyclohexenyl, -ethyl-cyclohexenyl, -methyl(4-methylcyclohexyl) and -propyl (3- methylcyclyohexyl).
  • R 1 represents heterocyclyl (which may optionally be substituted)
  • examples include tetrahydrofuranyl, morpholinyl, piperdinyl, 3,4-dihydro-2H-pyranyl, pyrrolidinyl, methyltetrahydrofuranyl- (e.g. 5-methyltetrahydrofuran-2-yl).
  • R 1 represents -Ci- ⁇ alkyl-heterocyclyl (which may optionally be substituted)
  • examples include -methyl-tetrahydrofuranyl (e.g. -methyl-tetrahydrofuran-2-yl, -methyl-tetrahydrofuran-3- yl), -ethyl-tetrahydrofuranyl, -methyl-piperidinyl.
  • R 1 represents optionally substituted aryl
  • aryl may typically represent phenyl.
  • Aryl may most typically represent substituted phenyl.
  • Exemplary substituted phenyl groups include 2,4- dichlorophenyl-, 2,4-diflurorophenyl-, 2,4-dimethoxyphenyl-, 2,4-dimethylphenyl-2,4- bis(trifluoromethyl)phenyl-, 2,4,6-trifluorophenyl-, 2,4,6-trimethylphenyl-, 2,6-dichlorophenyl-, 2,6-difluorophenyl-, 2,6-dimethoxyphenyl-, 2-isopropyl-6-methylphenyl-, 3-(cyclopentyloxy)-4- methoxyphenyl-, 3,4,5-trimethoxyphenyl-, 3,4-dimethoxyphenyl-, 3,4-dichlorophenyl-, 3,4- dimethylphenyl-, 3,4,5-trifluorophenyl-, 3,5-bis(trifluroromethyl)phenyl-, 3,5-dimethoxyphenyl-, 3- me
  • R 1 represents optionally substituted aryl and aryl represents naphthyl
  • examples include unsubstituted naphthyl (e.g. naphthalen-1-yl, naphthalen-2-yl, naphthalen-3-yl) as well as substituted naphthyl (e.g. 4-methyl-naphthalen-2-yl-, 5-methyl-naphthalen-3-yl-, 7-methyl- naphthalen-3-y- and 4-fluoro-naphthalen-2-yl-).
  • unsubstituted naphthyl e.g. naphthalen-1-yl, naphthalen-2-yl, naphthalen-3-yl
  • substituted naphthyl e.g. 4-methyl-naphthalen-2-yl-, 5-methyl-naphthalen-3-yl-, 7-methyl- naphthalen-3-y- and 4-fluoro-naphthalen-2-yl
  • R 1 represents optionally substituted heteroaryl
  • examples include monocyclic rings (e.g. 5 or 6 membered rings) and bicyclic rings (e.g. 9 or 10 membered rings) which may optionally be substituted.
  • Example 5 membered rings include pyrrolyl (e.g. pyrrol-2-yl) and imidazolyl (e.g. 1 H-imidazol-2-yl or 1 H-imidazol-4-yl), pyrazolyl (e.g. 1 H-pyrazol-3-yl), furanyl (e.g. furan-2-yl), thiazolyl (e.g. thiazol-2-yl), thiophenyl (e.g.
  • Example 6 membered rings include pyridinyl (e.g. pyridine-2-yl and pyridine-4-yl). Specific substituents that may be mentioned are one or more e.g. 1 , 2 or 3 groups selected from halogen, hydroxyl, alkyl (e.g. methyl) and alkoxy- (e.g. methoxy-).
  • Example substituted 5 membered rings include 4,5- dimethyl-furan-2-yl-, 5-hydroxymethyl-furan-2-yl-, 5-methyl-furan-2-yl- and 6-methyl-pyridin-2-yl- .
  • An example substituted 6-membered ring is 1-oxy-pyridin-4-yk
  • Example 9 membered rings include 1 H indolyl (e.g. 1 H-indol-3-yl, 1 H-indol-5-yl), benzothiophenyl (e.g. benzo[b]thiophen-3- yl, particularly 2-benzo[b]thiophen-3-yl), benzo[1 ,2,5]-oxadiazolyl (e.g. benzo[1 ,2,5]-oxadiazol-5- yl), benzo[1 ,2,5]-thiadiazolyl (e.g. benzo[1 ,2,5]-thiadiazol-5-yl).
  • 1 H indolyl e.g. 1 H-indol-3-yl, 1 H-indol-5-yl
  • benzothiophenyl e.g. benzo[b]thiophen-3- yl, particularly 2-benz
  • Example 10 membered rings include quinolinyl (e.g.quinolin-3-yl, quinolin-4-yl, quinolin-8-yl). Specific substituents that may be mentioned are one or more e.g. 1 , 2 or 3 groups selected from halogen, hydroxyl, alkyl (e.g. methyl) and alkoxy- (e.g. methoxy-).
  • Example substituted 9-membered rings include 1-methyl- 1 H-indol-3-yl, 2-methyl-1 H-indol-3-yl, 6-methyl-1 H-indol-3-yl.
  • Example substituted 10 membered rings include 2-chloro-quinolin-3-yl, 8-hydroxy-quinolin-2-yl, oxo-chromenyl (e.g.4-oxo-4H- chromen-3-yl) and 6-methyl-4-oxo-4H-chromen-3-yl.
  • R 1 represents -alkylaryl in which aryl is optionally substituted
  • examples include -Ci- 4 alkylaryl.
  • Another specific group is -alkyl(substituted phenyl) e.g. in which phenyl is substituted by one or more groups selected from alkyl, fluoroalkyl, halogen and alkoxy (e.g. methyl, trifluoromethyl, tert-butyl, chloro, fluoro and methoxy) and, for example, alkyl is Ci -4 alkyl.
  • Another specific group is -alkyl(bicyclic aryl) e.g. wherein bicyclic aryl is optionally substituted naphthyl.
  • a further specific group is benzyl.
  • R 1 represents -alkylheteroaryl in which heteroaryl is optionally substituted
  • examples include -Ci -4 alkylheteroaryl e.g. -methylheteroaryl and -ethylheteroaryl (e.g. 1-heteroarylethyl- and 2-heteroarylethyl-), -propylheteroaryl and -butylheteroaryl in which heteroaryl is optionally substituted.
  • -alkylheteroaryl groups include pyridinylmethyl-, N-methyl- pyrrol-2-methyl- N-methyl-pyrrol-2-ethyl-, N-methyl-pyrrol-3-methyl-, N-methyl-pyrrol-3-ethyl-, 2- methyl-pyrrol-1 -methyl-, 2-methyl-pyrrol-1 -ethyl-, 3-methyl-pyrrol-1 -methyl-, 3-methyl-pyrrol-1 - ethyl-, 4-pyridino-methyl-, 4-pyridino-ethyl-, 2-(thiazol-2-yl)-ethyl-, 2-ethyl-indol-1 -methyl-, 2- ethyl-indol-1 -ethyl-, 3-ethyl-indol-1 -methyl-, 3-ethyl-indol-1 -ethyl-, 4-methyl-pyridin-2-methyl-, 4- methyl-pyridin-2-methyl-
  • R 1 represents optionally substituted -phenyl fused to optionally substituted carbocyclyl
  • examples include indanyl (e.g. indan-4-yl-, 2-methyl-indan-4-yl-), indenyl and tetralinyl.
  • R 1 represents optionally substituted -phenyl fused to optionally substituted heterocyclyl
  • examples include benzo[1 ,3]dioxo-4-yl- and 2,3-dihydro-benzo[1 ,4]dioxin-4-yl-.
  • R 1 represents phenyl substituted by phenyl, or phenyl substituted by a monocyclic heteroaryl group, in which any of aforesaid phenyl and heteroaryl groups may optionally be substituted, typically the phenyl ring connected directly to the nitrogen atom is unsubstituted and the terminal phenyl ring or the monocyclic heteroaryl ring is optionally substituted by one, two or three substitutents (e.g. one or two, e.g. one). Typically the terminal phenyl or monocyclic heteroaryl group is unsubstituted. Typically the terminal phenyl or monocyclic heteroaryl group substitutes the other phenyl group at the 4-position.
  • Examples include -biphenyl-4-yl and 4- (oxazol-5-yl)phenyk
  • R 1 represents phenyl substituted by benzyloxy- in which any of the aforesaid phenyl or benzyloxy groups may optionally be substituted on the ring by one or more groups selected from Ci ⁇ alkyl, halogen and C-i ⁇ alkoxy
  • examples include 4-(benzyloxy)phenyk
  • Further exampes include 4-((4-fluoro-benzyl)oxy)phenyl-, 4-((4-chloro-benzyl)oxy)phenyl- and 4-((4- methoxy-benzyl)oxy)phenyk
  • the terminal benzyloxy group substitutes the phenyl group at the 4-position.
  • the phenyl ring connected directly to the nitrogen atom is unsubstituted.
  • R 2 examples include H, methyl and ethyl.
  • R 3 examples include H, methyl and ethyl.
  • R 4 examples include H, methyl and ethyl.
  • group Y examples include -(CHz) 2 -, -(CHz) 3 -, -(CHz) 4 -. -(CHz) 5 -, -CH 2 CH(Me)CH 2 -, -CH 2 CH(Me)CH 2 CH 2 -, -CH 2 CH 2 CH(Me)CH 2 - and
  • R 5 examples include H, methyl and ethyl.
  • R 6 examples include H, methyl and ethyl.
  • R 1 represents alkyl, aryl; carbocyclyl; heteroaryl; -phenyl fused to carbocyclyl; phenyl fused to heterocyclyl; or R 1 represents phenyl substituted by optionally substituted phenyl or phenyl substituted by a monocyclic heteroaryl group; any of which aforesaid aryl, carbocyclyl, heteroaryl, phenyl and heterocyclyl may optionally be substituted.
  • R 1 When R 1 represents optionally substituted aryl, R 1 suitably represents optionally substituted phenyl, especially substituted phenyl.
  • Example substituents are selected from Ci -6 alkyl (e.g. methyl, ethyl, isopropyl), Ci -6 alkoxy (e.g. methoxy), -O-C 3-8 cycloalkyl (e.g. -O-cyclopentyl), Ci- 6 haloalkyl (e.g. trifluoromethyl) and halogen (e.g. chloro).
  • R 1 represents aryl; heteroaryl; -phenyl fused to carbocyclyl; phenyl fused to monocyclic heterocyclyl; or R 1 represents phenyl substituted by phenyl, or phenyl substituted by a monocyclic heteroaryl group; in which any of the aforesaid phenyl, heteroaryl, carbocyclyl and heterocyclyl may optionally be substituted.
  • R 1 represents optionally substituted aryl. In another embodiment R 1 represents optionally substituted heteroaryl. In a third embodiment R 1 represents optionally substituted phenyl fused to optionally substituted carbocyclyl. In another embodiment R 1 represents optionally substituted phenyl fused to optionally substituted heterocyclyl. In a further embodiment R 1 represents optionally substituted phenyl substituted by optionally substituted phenyl or optionally substituted phenyl substituted by an optionally substituted monocyclic heteroaryl group. When R 1 represents optionally substituted aryl, R 1 suitably represents optionally substituted phenyl, especially substituted phenyl.
  • phenyl suitably has one, two or three substituents (e.g. one or two substituents, e.g. one substituent, e.g. one substituent ).
  • substituents are for example at the 2, 4 and 6 positions of the phenyl ring or alternatively at the 3, 4, and 5 positions of the phenyl ring.
  • substituents are for example at the 3 and 4 positions of the phenyl ring or at the 3 and 5 positions of the phenyl ring.
  • the substituent is at the 2, 3 or 4 position of the phenyl ring, most suitably at the 4-position of the phenyl ring.
  • Example substituents are selected from Ci -6 alkyl (e.g. methyl, ethyl, isopropyl), Ci -6 alkoxy (e.g. methoxy), C-i- ⁇ haloalkyl (e.g. trifluoromethyl) and halogen (e.g. chloro). Another example substituent is cyano. Further example substituents are selected from Ci -3 alkyl (e.g. methyl, ethyl, isopropyl), Ci -3 alkoxy (e.g. methoxy), Ci -3 haloalkyl (e.g. trifluoromethyl), halogen (e.g. chloro) and cyano.
  • Ci -6 alkyl e.g. methyl, ethyl, isopropyl
  • Ci -3 alkoxy e.g. methoxy
  • Ci -3 haloalkyl e.g. trifluoromethyl
  • halogen e.g. chloro
  • Specific examples include 2,4,6-trimethylphenyl-, 3,4,5-trimethoxyphenyl-, 3,4-dichlorophenyl-, 3,4-dimethylphenyl-, 3,5-dimethoxyphenyl-, 4-methoxyphenyl-, 4- cyanophenyl-, 4-ethoxyphenyl-, 4-ethylphenyl-, 4-isopropylphenyl- or 4-methoxyphenyl-.
  • R 1 represents unsubstituted aryl
  • R 1 suitably represents phenyl, naphthalen-1-yl or naphthalen-2-yl.
  • R 1 represents optionally substituted heteroaryl
  • R 1 suitably represents benzo[c][1 ,2,5]thiadiazol-6-yl.
  • R 1 represents optionally substituted phenyl fused to optionally substituted heterocyclyl
  • R 1 suitably represents 2,3-dihydrobenzo[b][1 ,4]dioxin-7-yl or benzo[d][1 ,3]dioxol-6-yl.
  • R 1 represents phenyl substituted by phenyl, or phenyl substituted by a monocyclic heteroaryl group in which any of aforesaid phenyl and/or monocyclic heteroaryl may optionally be substituted.
  • R 1 suitably represents biphenyl-4-yl.
  • R 1 represents aryl; heteroaryl; phenyl fused to heterocyclyl; or R 1 represents phenyl substituted by phenyl, or phenyl substituted by a monocyclic heteroaryl group group in which any of the aforementioned phenyl, aryl, heteroaryl and heterocyclyl may optionally be substituted. Most suitably R 1 represents substituted phenyl or R 1 represents phenyl fused to heterocyclyl.
  • R 1 represents substituted phenyl.
  • R 1 represents phenyl fused to heterocyclyl.
  • A represents
  • R 2 suitably represents H
  • R 3 suitably represents H or methyl.
  • R 4 suitably represents H or methyl.
  • R 3 represents H and R 4 represents methyl. In another embodiment, R 3 represents methyl and R 4 represents H. Most suitably R 2 represents H, R 3 represents H and R 4 represents methyl.
  • Y represents an unsubstituted C 2-5 alkylene chain. More suitably Y represents -(CH 2 ) 3 - or -(CH 2 ) 4 -. In one embodiment, Y represents -(CH 2 )3-- In another embodiment, Y represents
  • the spiro-cycloalkyl group is suitably C 3 spiro-cycloalkyl.
  • R 5 represents H and R 6 represents H. In another embodiment R 5 represents H and R 6 represents Ci -2 alkyl. In a third embodiment R 5 represents Ci -2 alkyl and R 6 represents
  • Z represents a bond, -CH 2 - or -CH 2 CH 2 -. In one embodiment Z represents a bond. In another embodiment, Z represents -CH 2 -. In a third embodiment, Z represents -CH 2 CH 2 -.
  • A represents
  • B suitably represents H.
  • benzimidazolyl is shown as benzimidazol-5-yl, which is represented as:
  • the reaction is typically performed in the presence of a reagent that removes hydrogen disulfide (e.g. a carbodiimide such as N 1 -((ethylimino)methylen)-N 3 ,N 3 -dimethylpropan-1 ,3-diamine hydrochloride).
  • a reagent that removes hydrogen disulfide e.g. a carbodiimide such as N 1 -((ethylimino)methylen)-N 3 ,N 3 -dimethylpropan-1 ,3-diamine hydrochloride.
  • the reaction may be carried out in a polar organic solvent or a mixture thereof (e.g. a mixture of ethanol and dimethylformamide).
  • a compound of formula (II) may be prepared by reaction of a compound of formula (IV)
  • M + represents a metal ion (e.g. sodium ion, Na + ) with a compound of formula (V)
  • the reaction may suitably be carried out in a polar protic organic solvent (e.g. an alcohol such as ethanol) at elevated temperature.
  • a polar protic organic solvent e.g. an alcohol such as ethanol
  • R 7 represents Ci ⁇ alkyl e.g. methyl with a compound of formula (III).
  • the reaction may suitably be carried out in a polar protic organic solvent (e.g. an alcohol such as ethanol) at elevated temperature.
  • a polar protic organic solvent e.g. an alcohol such as ethanol
  • a compound of formula (Vl) may be prepared by reaction of a compound of formula (VII)
  • R 8 represents Ci -6 alkyl e.g. methyl with a compound of formula (VIII)
  • the reaction may suitably be carried out in a polar protic organic solvent (e.g. an alcohol such as ethanol) at elevated temperature.
  • a polar protic organic solvent e.g. an alcohol such as ethanol
  • Compounds of formula (Vl) may be formed in situ i.e. the compounds of formula (Vl) need not be isolated from the reaction mixture after reaction of compounds of formula (VII) and (VIII) before onwards reaction to compounds of formula (I).
  • R , R , R and Y are defined as above and B represents H, a compound of formula (Ilia)
  • a compound of formula (IX) may be prepared by deprotection of a compound of formula (X)
  • PG represents a protecting group (e.g. trityl) and L " represents a suitable counterion such as Br " .
  • suitable deprotecting conditions include use of trifluoroacetic acid when PG repesents trityl).
  • a compound of formula (X) may be prepared by reaction of a compound of formula (Xl)
  • L represents a suitable leaving group e.g. Br.
  • a compound of formula (Xl) may be prepared from a compound of formula (XIII)
  • a base e.g. triethylamine
  • a suitable protecting reagent e.g. chlorotriphenylmethane
  • a polar organic solvent e.g. dimethylformamide
  • Physiological substrates of QC (EC) in mammals are, e.g. amyloid beta-peptides (3-40), (3-42), (1 1-40 and (11-42), ABri, ADan, Gastrin, Neurotensin, FPP, CCL 2, CCL 7, CCL 8, CCL 16, CCL 18, Fractalkine, Orexin A, [Gln 3 ]-glucagon(3-29), [Gln 5 ]-substance P(5-1 1 ) and the peptide QYNAD.
  • Table 1 The compounds and/or combinations according to the present invention and pharmaceutical compositions comprising at least one inhibitor of QC (EC) are useful for the treatment of conditions that can be treated by modulation of QC activity.
  • Glutamate is found in positions 3, 11 and 22 of the amyloid ⁇ -peptide.
  • the mutation from glutamic acid (E) to glutamine (Q) in position 22 has been described as the so called Dutch type cerebroarterial amyloidosis mutation.
  • the ⁇ -amyloid peptides with a pyroglutamic acid residue in position 3, 1 1 and/or 22 have been described to be more cytotoxic and hydrophobic than the amyloid ⁇ -peptides 1-40(42/43) (Saido T.C. 2000 Medical Hypotheses 54(3): 427-429).
  • the multiple N-terminal variations, e.g. Abeta(3-40), Abeta(3-42), Abeta(11-40) and Abeta (11- 42) can be generated by the ⁇ -secretase enzyme ⁇ -site amyloid precursor protein-cleaving enzyme (BACE) at different sites (Huse JT. et al. 2002 J. Biol. Chem.
  • Transepithelial transducing cells particularly the gastrin (G) cell, co-ordinate gastric acid secretion with the arrival of food in the stomach.
  • G gastrin
  • Biosynthetic precursors and intermediates are putative growth factors; their products, the amidated gastrins, regulate epithelial cell proliferation, the differentiation of acid-producing parietal cells and histamine-secreting enterochromaffin-like (ECL) cells, and the expression of genes associated with histamine synthesis and storage in ECL cells, as well as acutely stimulating acid secretion.
  • Gastrin also stimulates the production of members of the epidermal growth factor (EGF) family, which in turn inhibit parietal cell function but stimulate the growth of surface epithelial cells.
  • EGF epidermal growth factor
  • Plasma gastrin concentrations are elevated in subjects with Helicobacter pylori, who are known to have increased risk of duodenal ulcer disease and gastric cancer (Dockray, GJ. 1999 J Physiol 15 315-324).
  • the peptide hormone gastrin released from antral G cells, is known to stimulate the synthesis and release of histamine from ECL cells in the oxyntic mucosa via CCK-2 receptors.
  • the mobilized histamine induces acid secretion by binding to the H(2) receptors located on parietal cells.
  • gastrin in both its fully amidated and less processed forms (progastrin and glycine-extended gastrin), is also a growth factor for the gastrointestinal tract. It has been established that the major trophic effect of amidated gastrin is for the oxyntic mucosa of stomach, where it causes increased proliferation of gastric stem cells and ECL cells, resulting in increased parietal and ECL cell mass.
  • the major trophic target of the less processed gastrin e.g. glycine-extended gastrin
  • the colonic mucosa Koh, TJ. and Chen, D. 2000 Regul Pept 9337 ⁇ 4.
  • Neurotensin is a neuropeptide implicated in the pathophysiology of schizophrenia that specifically modulates neurotransmitter systems previously demonstrated to be misregulated in this disorder.
  • Clinical studies in which cerebrospinal fluid (CSF) NT concentrations have been measured revealed a subset of schizophrenic patients with decreased CSF NT concentrations that are restored by effective antipsychotic drug treatment.
  • CSF cerebrospinal fluid
  • Considerable evidence also exists concordant with the involvement of NT systems in the mechanism of action of antipsychotic drugs.
  • the behavioral and biochemical effects of centrally administered NT remarkably resemble those of systemically administered antipsychotic drugs, and antipsychotic drugs increase NT neurotransmission. This concatenation of findings led to the hypothesis that NT functions as an endogenous antipsychotic.
  • FPP Fertilization promoting peptide
  • TRH thyrotrophin releasing hormone
  • FPP and adenosine have been shown to stimulate cAMP production in uncapacitated cells but inhibit it in capacitated cells, with FPP receptors somehow interacting with adenosine receptors and G proteins to achieve regulation of AC. These events affect the tyrosine phosphorylation state of various proteins, some being important in the initial "switching on” others possibly being involved in the acrosome reaction itself.
  • Calcitonin and angiotensin II also found in seminal plasma, have similar effects in vitro on uncapacitated spermatozoa and can augment responses to FPP. These molecules have similar effects in vivo, affecting fertility by stimulating and then maintaining fertilizing potential.
  • CCL2 (MCP-1 ), CCL7, CCL8, CCL16, CCL18 and fractalkine play an important role in pathophysiological conditions, such as suppression of proliferation of myeloid progenitor cells, neoplasia, inflammatory host responses, cancer, psoriasis, rheumatoid arthritis, atherosclerosis, vasculitis, humoral and cell-mediated immunity responses, leukocyte adhesion and migration processes at the endothelium, inflammatory bowel disease, restenosis, pulmonary fibrosis, pulmonary hypertention, liver fibrosis, liver cirrhosis, nephrosclerosis, ventricular remodeling, heart failure, arteriopathy after organ transplantations and failure of vein grafts.
  • pathophysiological conditions such as suppression of proliferation of myeloid progenitor cells, neoplasia, inflammatory host responses, cancer, psoriasis, rheumatoid arthritis, atherosclerosis, vasculitis,
  • MCP-1 might also play a role in gestosis (Katabuchi, H., et al., (2003) Med Electron Microsc. 36, 253-262), as a paracrine factor in tumor development (Ohta, M., et al., (2003) InU Oncol. 22, 773-778; Li, S., et al., (2005) J Exp.Med 202, 617-624), neuropathic pain (White, F. A., et al., (2005) Proc. Natl. Acad.Sci.U.S.A) and AIDS (Park, I. W., Wang, J. F., and Groopman, J. E. (2001 ) Blood 97, 352-358; Coll, B., et al., (2006) Cytokine 34, 51-55).
  • MCP-1 levels are increased in CSF of AD patients and patients showing mild cognitive impairment (MCI) (Galimberti, D., et al., (2006) Arch. Neurol. 63, 538-543). Furthermore, MCP-1 shows an increased level in serum of patients with MCI and early AD (Clerici, F., et al., (2006) Neurobiol. Aging 27, 1763-1768).
  • MCI mild cognitive impairment
  • cytotoxic T lymphocyte peptide-based vaccines against hepatitis B, human immunodeficiency virus and melanoma were recently studied in clinical trials.
  • This peptide is a Melan-A/MART-1 antigen immunodominant peptide analog, with an N-terminal glutamic acid. It has been reported that the amino group and gamma- carboxylic group of glutamic acids, as well as the amino group and gamma-carboxamide group of glutamines, condense easily to form pyroglutamic derivatives.
  • Orexin A is a neuropeptide that plays a significant role in the regulation of food intake and sleep-wakefulness, possibly by coordinating the complex behavioral and physiologic responses of these complementary homeostatic functions. It plays also a role in the homeostatic regulation of energy metabolism, autonomic function, hormonal balance and the regulation of body fluids.
  • QYNAD is a substrate of the enzyme glutaminyl cyclase (QC, EC 2.3.2.5), which is also present in the brain of mammals, especially in human brain. Glutaminyl cyclase catalyzes effectively the formation of pEYNAD from its precursor QYNAD.
  • the present invention provides the use of the compounds of formula (I) for the preparation of a medicament for the prevention or alleviation or treatment of a disease selected from the group consisting of mild cognitive impairment, Alzheimer's disease, Familial British Dementia, Familial Danish Dementia, neurodegeneration in Down Syndrome, Huntington's disease, Kennedy's disease, ulcer disease, duodenal cancer with or w/o Helicobacter pylori infections, colorectal cancer, Zolliger-Ellison syndrome, gastric cancer with or without Helicobacter pylori infections, pathogenic psychotic conditions, schizophrenia, infertility, neoplasia, inflammatory host responses, cancer, malign metastasis, melanoma, psoriasis, rheumatoid arthritis, atherosclerosis, pancreatitis, restenosis, impaired humoral and cell- mediated immune responses, leukocyte adhesion and migration processes in the endothelium, impaired food intake, impaired sleep-waken,
  • a QC inhibitor according to the present invention can lead to suppression of male fertility.
  • the present invention provides the use of inhibitors of QC (EC) activity in combination with other agents, especially for the treatment of neuronal diseases, artherosclerosis and multiple sclerosis.
  • the present invention also provides a method of treatment of the aforementioned diseases comprising the administration of a therapeutically active amount of at least one compound of formula (I) to a mammal, preferably a human.
  • said method and corresponding uses are for the treatment of a disease selected from the group consisting of mild cognitive impairment, Alzheimer's disease, Familial British Dementia, Familial Danish Dementia, neurodegeneration in Down Syndrome, Parkinson's disease and Chorea Huntington, comprising the administration of a therapeutically active amount of at least one compound of formula (I) to a mammal, preferably a human.
  • a disease selected from the group consisting of mild cognitive impairment, Alzheimer's disease, Familial British Dementia, Familial Danish Dementia, neurodegeneration in Down Syndrome, Parkinson's disease and Chorea Huntington, comprising the administration of a therapeutically active amount of at least one compound of formula (I) to a mammal, preferably a human.
  • the present invention provides a method of treatment and corresponding uses for the treatment of rheumatoid arthritis, atherosclerosis, pancreatitis and restenosis.
  • the present invention provides a composition, preferably a pharmaceutical composition, comprising at least one QC inhibitor optionally in combination with at least one other agent selected from the group consisting of nootropic agents, neuroprotectants, antiparkinsonian drugs, amyloid protein deposition inhibitors, beta amyloid synthesis inhibitors, antidepressants, anxiolytic drugs, antipsychotic drugs and anti-multiple sclerosis drugs.
  • at least one QC inhibitor optionally in combination with at least one other agent selected from the group consisting of nootropic agents, neuroprotectants, antiparkinsonian drugs, amyloid protein deposition inhibitors, beta amyloid synthesis inhibitors, antidepressants, anxiolytic drugs, antipsychotic drugs and anti-multiple sclerosis drugs.
  • said QC inhibitor is a compound of formula (I) of the present invention.
  • the aforementioned other agent is selected from the group consisting of beta- amyloid antibodies, cysteine protease inhibitors, PEP-inhibitors, LiCI, acetylcholinesterase (AChE) inhibitors, PIMT enhancers, inhibitors of beta secretases, inhibitors of gamma secretases, inhibitors of aminopeptidases, preferably inhibitors of dipeptidyl peptidases, most preferably DP IV inhibitors; inhibitors of neutral endopeptidase, inhibitors of Phosphodiesterase- 4 (PDE-4), TNFalpha inhibitors, muscarinic M1 receptor antagonists, NMDA receptor antagonists, sigma-1 receptor inhibitors, histamine H3 antagonists, immunomodulatory agents, immunosuppressive agents, MCP-1 antagonists or an agent selected from the group consisting of antegren (natalizumab), Neurelan (fampridine-SR), campath (alemtuzumab), IR 208, NBI 57
  • the other agent may be, for example, an anti-anxiety drug or antidepressant selected from the group consisting of
  • Benzodiazepines e.g. alprazolam, chlordiazepoxide, clobazam, clonazepam, clorazepate, diazepam, fludiazepam, loflazepate, lorazepam, methaqualone, oxazepam, prazepam, tranxene,
  • alprazolam chlordiazepoxide
  • clobazam clonazepam
  • clorazepate diazepam
  • fludiazepam fludiazepam
  • loflazepate lorazepam
  • methaqualone oxazepam
  • prazepam tranxene
  • SSRI's Selective serotonin re-uptake inhibitors
  • SSRI's e.g. citalopram, fluoxetine, fluvoxamine, escitalopram, sertraline, paroxetine
  • Tricyclic antidepressants e.g. amitryptiline, clomipramine, desipramine, doxepin, imipramine
  • Azapirones e.g. buspirone, tandopsirone
  • SNRI's Serotonin-norepinephrine reuptake inhibitors
  • NRI's Norepinephrine reuptake inhibitors (NRI's), e.g. reboxetine,
  • NPY-receptor ligands NPY agonists or antagonists.
  • the other agent may be, for example, an anti-multiple sclerosis drug selected from the group consisting of
  • dihydroorotate dehydrogenase inhibitors e.g. SC-12267, teriflunomide, MNA-715, HMR-
  • autoimmune suppressant e.g. laquinimod
  • paclitaxel e.g. paclitaxel
  • antibodies e.g. AGT-1 , anti-granulocyte-macrophage colony-stimulating factor (GM- CSF) monoclonal antibody, Nogo receptor modulators, ABT-874, alemtuzumab
  • interferon beta e.g. Frone, interferon beta-1 a like Avonex, Betron (Rebif), interferon beta analogs, interferon beta-transferrin fusion protein, recombinant interferon beta-1 b like Betaseron, h) interferon tau, i) peptides, e.g. AT-008, AnergiX.MS, lmmunokine (alpha-lmmunokine-NNSO3), cyclic peptides like ZD-7349, j) therapeutic enzymes, e.g. soluble CD8 (sCD8), k) multiple sclerosis-specific autoantigen-encoding plasmid and cytokine-encoding plasmid, e.g. BHT-3009;
  • interferon beta e.g. Frone, interferon beta-1 a like Avonex, Betron (Rebif), interferon beta analogs, interferon beta-transferrin fusion protein, re
  • TNF-alpha e.g. BLX-1002, thalidomide, SH-636, m
  • TNF antagonists e.g. solimastat, lenercept (syn. to RO-45-2081 , Tenefuse), onercept
  • TNF alpha e.g. etanercept (syn. to Enbrel, TNR-001 )
  • CD28 antagonists e.g. abatacept
  • p) Lck tyrosine kinase inhibitors q) cathepsin K inhibitors
  • analogs of the neuron-targeting membrane transporter protein taurine and the plant- derived calpain inhibitor leupeptin e.g. Neurodur
  • chemokine receptor-1 (CCR1 ) antagonist e.g. BX-471 , t) CCR2 antagonists
  • AMPA receptor antagonists e.g.
  • IgLC free immunoglobulin light chain
  • Apogen MS bb) alpha-2 adrenoceptor agonist, e.g. tizanidine (syn. to Zanaflex, Ternelin, Sirdalvo, Sirdalud, Mionidine), cc) copolymer of L-tyrosine, L-lysine, L-glutamic acid and L-alanine, e.g. glatiramer acetate (syn. to Copaxone, COP-1 , copolymer-1 ), dd) topoisomerase Il modulators, e.g. mitoxantrone hydrochloride, ee) adenosine deaminase inhibitor, e.g.
  • interleukin-10 e.g. ilodecakin (syn. to Tenovil, Sch-52000, CSIF)
  • interleukin-12 antagonists e.g. lisofylline (syn. to CT-1501 R, LSF, lysofylline), hh) Ethanaminum, e.g. SRI-62-834 (syn. to CRC-8605, NSC-614383),
  • immunomodulators e.g.
  • SAIK-MS alpha-fetoprotein peptide (AFP), IPDS, jj) retinoid receptor agonists, e.g. adapalene (syn. to Differin, CD-271 ), kk) TGF-beta, e.g. GDF-1 (growth and differentiation factor 1 ),
  • TGF-beta-2 e.g. BetaKine, mm
  • MMP inhibitors e.g. glycomed, nn) phosphodiesterase 4 (PDE4) inhibitors, e.g. RPR-122818, oo) purine nucleoside phosphorylase inhibitors, e.g. 9-(3-pyridylmethyl)-9-deazaguanine, peldesine (syn. to BCX-34, TO-200), pp) alpha-4/beta-1 integrin antagonists, e.g. ISIS-104278, qq) antisense alpha4 integrin (CD49d), e.g.
  • cytokine-inducing agents e.g. nucleosides, ICN-17261 ,
  • compositions e.g. for parenteral, enteral or oral administration, comprising at least one QC inhibitor, optionally in combination with at least one of the other aforementioned agents.
  • the invention provides a method for the treatment of these conditions.
  • the method comprises either co-administration of at least one QC inhibitor and at least one of the other agents or the sequential administration thereof.
  • Co-administration includes administration of a formulation, which comprises at least one QC inhibitor and at least one of the other agents or the essentially simultaneous administration of separate formulations of each agent.
  • Beta-amyloid antibodies and compositions containing the same are described, e.g. in WO 2006/137354, WO 2006/1 18959, WO 2006/1031 16, WO 2006/095041 , WO 2006/081 171 , WO 2006/066233, WO 2006/066171 , WO 2006/066089, WO 2006/066049, WO 2006/055178, WO 2006/046644, WO 2006/039470, WO 2006/036291 , WO 2006/026408, WO 2006/016644, WO 2006/014638, WO 2006/014478, WO 2006/008661 , WO 2005/123775, WO 2005/120571 , WO 2005/105998, WO 2005/081872, WO 2005/080435, WO 2005/02851 1 , WO 2005/025616, WO 2005/025516, WO 2005/023858, WO 2005/018424, WO 2005/011599,
  • the beta-amyloid antibodies may be selected from, for example, polyclonal, monoclonal, chimenic or humanized antibodies. Furthermore, said antibodies may be useful to develop active and passive immune therapies, i.e. vaccines and monoclonal antibodies.
  • Suitable examples of beta-amyloid antibodies are ACU-5A5, huC091 (Acumen/Merck); PF- 4360365, RI-1014, RI-1219, RI-409, RN-1219 (Rinat Neuroscience Corp (Pfizer Inc)); the nanobody therapeutics of Ablynx/Boehringer Ingelheim; beta-amyloid-specific humanized monoclonal antibodies of Intellect Neurosciences/IBL; m266, m266.2 (EIi Lilly & Co.); AAB-02 (Elan); bapineuzumab (Elan); BAN-2401 (Bioarctic Neuroscience AB); ABP-102 (Abiogen Pharma SpA); BA-27, BC-05 (Takeda); R
  • antibodies which recognize the N-terminus of the A ⁇ peptide.
  • a suitable antibody which recognizes the A ⁇ -N-Terminus is, for example Acl-24 (AC Immune SA).
  • a monoclonal antibody against beta-amyloid peptide is disclosed in WO 2007/068412.
  • Respective chimeric and humanized antibodies are disclosed in WO 2008/011348.
  • a method for producing a vaccine composition for treating an amyloid-associated disease is disclosed in WO 2007/068411.
  • Suitable cysteine protease inhibitors are inhibitors of cathepsin B.
  • Inhibitors of cathepsin B and compositions containing such inhibitors are described, e.g. in WO 2006/060473, WO 2006/042103, WO 2006/039807, WO 2006/021413, WO 2006/021409, WO 2005/097103, WO 2005/007199, WO2004/084830, WO 2004/078908, WO 2004/026851 , WO 2002/094881 , WO 2002/027418, WO 2002/021509, WO 1998/046559, WO 1996/021655.
  • PIMT enhancers are 10-aminoaliphatyl-dibenz[b, f] oxepines described in WO 98/15647 and WO 03/057204, respectively. Further useful according to the present invention are modulators of PIMT activity described in WO 2004/039773.
  • Inhibitors of beta secretase and compositions containing such inhibitors are described, e.g. in
  • beta secretase inhibitors for the purpose of the present invention are WY- 25105 (Wyeth); Posiphen, (+)-phenserine (TorreyPines / NIH); LSN-2434074, LY-2070275, LY- 2070273, LY-2070102 (EIi Lilly & Co.); PNU-159775A, PNU-178025A, PNU-17820A, PNU- 33312, PNU-38773, PNU-90530 (Elan / Pfizer); KMI-370, KMI-358, kmi-008 (Kyoto University); OM-99-2, OM-003 (Athenagen Inc.); AZ-12304146 (AstraZeneca / Astex); GW-840736X (GlaxoSmithKline pic), DNP-004089 (De Novo Pharmaceuticals Ltd.) and CT-21166 (CoMentis Inc.).
  • Inhibitors of gamma secretase and compositions containing such inhibitors are described, e.g. in WO2005/008250, WO2006/004880, US 7,122,675, US 7,030,239, US 6,992,081 , US 6,982,264, WO2005/097768, WO2005/028440, WO2004/101562, US 6,756,51 1 , US 6,683,091 , WO03/066592, WO03/014075, WO03/013527, WO02/36555, WO01/53255, US 7,109,217, US 7,101 ,895, US 7,049,296, US 7,034,182, US 6,984,626, WO2005/040126, WO2005/030731 , WO2005/014553, US 6,890,956, EP 1334085, EP 1263774, WO2004/101538, WO2004/00958, WO2004/08991 1 , WO2004/0736
  • Suitable gamma secretase inhibitors for the purpose of the present invention are GSI-953, WAY-GSI-A, WAY-GSI-B (Wyeth); MK-0752, MRK-560, L-852505, L-685-458, L-852631 , L- 852646 (Merck & Co. Inc.); LY-450139, LY-41 1575, AN-37124 (EIi Lilly & Co.); BMS-299897, BMS-433796 (Bristol-Myers Squibb Co.); E-2012 (Eisai Co. Ltd.); EHT-0206, EHT-206 (ExonHit Therapeutics SA); and NGX-555 (TorreyPines Therapeutics Inc.).
  • DP IV-inhibitors and compositions containing such inhibitors are described, e.g. in US6,011 ,155; US6,107,317; US6, 1 10,949; US6, 124,305; US6, 172,081 ; WO99/61431 , WO99/67278, WO99/67279, DE19834591 , WO97/40832, WO95/15309, WO98/19998, WO00/07617, WO99/38501 , WO99/46272, WO99/38501 , WO01/68603, WO01/40180, WO01/81337, WO01/81304, WO01/55105, WO02/02560, WO01/34594, WO02/38541 , WO02/083128, WO03/072556, WO03/002593, WO03/000250, WO03/000180, WO03/000181 , EP1258476,
  • Suitable DP IV-inhibitors for the purpose of the present invention are for example Sitagliptin, des-fluoro-sitagliptin (Merck & Co. Inc.); vildagliptin, DPP-728, SDZ-272-070 (Novartis) ; ABT- 279, ABT-341 (Abbott Laboratories); denagliptin, TA-6666 (GlaxoSmithKline pic); SYR-322 (Takeda San Diego Inc.); talabostat (Point Therapeutics Inc.); Ro-0730699, R-1499, R-1438 (Roche Holding AG); FE-99901 1 (Ferring Pharmaceuticals); TS-021 (Taisho Pharmaceutical Co.
  • GRC-8200 (Glenmark Pharmaceuticals Ltd.); ALS-2-0426 (Alantos Pharmaceuticals Holding Inc.); ARI-2243 (Arisaph Pharmaceuticals Inc.); SSR-162369 (Sanofi-Synthelabo); MP- 513 (Mitsubishi Pharma Corp.); DP-893, CP-867534-01 (Pfizer Inc.); TSL-225, TMC-2A (Tanabe Seiyaku Co.
  • PHX-1149 Phenomenix Corp.
  • saxagliptin Bristol-Myers Squibb Co.
  • PSN-9301 ((OSI) Prosidion), S-40755 (Servier); KRP-104 (ActivX Biosciences Inc.); sulphostin (Zaidan Hojin); KR-62436 (Korea Research Institute of Chemical Technology); P32/98 (Probiodrug AG); Bl-A, Bl-B (Boehringer lngelheim Corp.); SK-0403 (Sanwa Kagaku Kenkyusho Co. Ltd.); and NNC-72-2138 (Novo Nordisk A/S).
  • Other preferred DP IV-inhibitors are
  • dipeptide-like compounds disclosed in WO 99/61431 , e.g. N-valyl prolyl, O-benzoyl hydroxylamine, alanyl pyrrolidine, isoleucyl thiazolidine like L-allo-isoleucyl thiazolidine, L-threo- isoleucyl pyrrolidine and salts thereof, especially the fumaric salts, and L-allo-isoleucyl pyrrolidine and salts thereof;
  • peptide structures disclosed in WO 03/002593, e.g. tripeptides; (iii) peptidylketones, disclosed in WO 03/033524; (vi) substituted aminoketones, disclosed in WO 03/040174; (v) topically active DP IV-inhibitors, disclosed in WO 01/14318; (vi) prodrugs of DP IV-inhibitors, disclosed in WO 99/67278 and WO 99/67279; and
  • Suitable beta amyloid synthesis inhibitors for the purpose of the present invention are for example Bisnorcymserine (Axonyx Inc.); (R)-flurbiprofen (MCP-7869; Flurizan) (Myriad Genetics); nitroflurbiprofen (NicOx); BGC-20-0406 (Sankyo Co. Ltd.) and BGC-20-0466 (BTG pic).
  • Suitable amyloid protein deposition inhibitors for the purpose of the present invention are for example SP-233 (Samaritan Pharmaceuticals); AZD-103 (Ellipsis Neurotherapeutics Inc.); AAB- 001 (Bapineuzumab), AAB-002, ACC-001 (Elan Corp pic); Colostrinin (ReGen Therapeutics pic); Tramiprosate (Neurochem); AdPEDI-(amyloid-beta1-6)11 ) (Vaxin Inc.); MPI-127585, MPI-
  • Transthyretin (State University of New York); PTI-777, DP-74, DP 68, Exebryl (ProteoTech Inc.); m266 (EIi Lilly & Co.); EGb-761 (Dr. Willmar Schwabe GmbH); SPI-014 (Satori Pharmaceuticals Inc.); ALS-633, ALS-499 (Advanced Life Sciences Inc.); AGT-160 (ArmaGen Technologies
  • Suitable PDE-4 inhibitors for the purpose of the present invention are for example Doxofylline (Instituto Biologico Chemioterapica ABC SpA.); idudilast eye drops, tipelukast, ibudilast (Kyorin
  • a preferred PDE-4-inhibitor is Rolipram.
  • MAO inhibitors and compositions containing such inhibitors are described, e.g. in WO2006/091988, WO2005/007614, WO2004/089351 , WO01/26656, WO01/12176, WO99/57120, WO99/571 19, WO99/13878, WO98/40102, WO98/01157, WO96/20946, WO94/07890 and WO92/21333.
  • Suitable MAO-inhibitors for the purpose of the present invention are for example Linezolid (Pharmacia Corp.); RWJ-416457 (RW Johnson Pharmaceutical Research Institute); budipine (Altana AG); GPX-325 (BioResearch Ireland); isocarboxazid; phenelzine; tranylcypromine; indantadol (Chiesi Farmaceutici SpA.); moclobemide (Roche Holding AG); SL-25.1 131 (Sanofi- Synthelabo); CX-1370 (Burroughs Wellcome Co.); CX-157 (Krenitsky Pharmaceuticals Inc.); desoxypeganine (HF Arzneiffenforschung GmbH & Co.
  • Linezolid Pharmacia Corp.
  • RWJ-416457 RW Johnson Pharmaceutical Research Institute
  • budipine Altana AG
  • GPX-325 BioResearch Ireland
  • isocarboxazid phenelzine
  • Suitable histamine H3 antagonists for the purpose of the present invention are, e.g. ABT-239, ABT-834 (Abbott Laboratories); 3874-H1 (Aventis Pharma); UCL-2173 (Berlin Free University), UCL-1470 (BioProjet, Societe Civile de für); DWP-302 (Daewoong Pharmaceutical Co Ltd); GSK-189254A, GSK-207040A (GlaxoSmithKline Inc.); cipralisant, GT-2203 (Gliatech Inc.); Ciproxifan (INSERM), • /S,2S-2-(2-Aminoethyl)-1-(1 H-imidazol-4-yl)cyclopropane (Hokkaido University); JNJ-17216498, JNJ-5207852 (Johnson & Johnson); NNC-0038-0000-1049 (Novo Nordisk A/S); and Sch-79687 (Schering-Plough).
  • PEP inhibitors and compositions containing such inhibitors are described, e.g. in JP 01042465,
  • Suitable prolyl endopeptidase inhibitors for the purpose of the present invention are, e.g. Fmoc- Ala-Pyrr-CN, Z-Phe-Pro-Benzothiazole (Probiodrug), Z-321 (Zeria Pharmaceutical Co Ltd.); ONO-1603 (Ono Pharmaceutical Co Ltd); JTP-4819 (Japan Tobacco Inc.) and S-17092 (Servier).
  • NPY neuropeptide Y
  • NPY mimetic NPY mimetic
  • NPY agonist or antagonist NPY ligand of the NPY receptors.
  • Preferred according to the present invention are antagonists of the NPY receptors.
  • Suitable ligands or antagonists of the NPY receptors are 3a, 4,5,9b-tetrahydro-1 h-benz[e]indol- 2-yl amine-derived compounds as disclosed in WO 00/68197.
  • NPY receptor antagonists which may be mentioned include those disclosed in European patent applications EP 0 614 91 1 , EP 0 747 357, EP 0 747 356 and EP 0 747 378; international patent applications WO 94/17035, WO 97/19911 , WO 97/19913, WO 96/12489, WO 97/19914, WO
  • NPY antagonists include those compounds that are specifically disclosed in these patent documents.
  • More preferred compounds include amino acid and non- peptide-based NPY antagonists.
  • Amino acid and non-peptide-based NPY antagonists which may be mentioned include those disclosed in European patent applications EP 0 614 91 1 , EP 0 747 357, EP 0 747 356 and EP 0 747 378; international patent applications WO 94/17035, WO 97/1991 1 , WO 97/19913, WO 96/12489, WO 97/19914, WO 96/22305, WO 96/40660, WO 96/12490, WO 97/09308, WO 97/20820, WO 97/20821 , WO 97/20822, WO 97/20823, WO 97/19682, WO 97/25041 , WO 97/34843, WO 97/46250, WO 98/03492, WO 98/03493, WO 98/03494, WO 98/07
  • Particularly preferred compounds include amino acid-based NPY antagonists.
  • Amino acid-based compounds which may be mentioned include those disclosed in international patent applications WO 94/17035, WO 97/1991 1 , WO 97/19913, WO 97/19914 or, preferably, WO 99/15498.
  • Preferred amino acid-based NPY antagonists include those that are specifically disclosed in these patent documents, for example BIBP3226 and, especially, (R)-N2- (diphenylacetyl)-(R)-N-[1-(4-hydroxy- phenyl) ethyl] arginine amide (Example 4 of international patent application WO 99/15498).
  • M1 receptor agonists and compositions containing such inhibitors are described, e.g. in WO2004/087158, WO91/10664.
  • Suitable M1 receptor antagonists for the purpose of the present invention are for example CDD- 0102 (Cognitive Pharmaceuticals); Cevimeline (Evoxac) (Snow Brand Milk Products Co. Ltd.); NGX-267 (TorreyPines Therapeutics); sabcomeline (GlaxoSmithKline); alvameline (H Lundbeck A/S); LY-593093 (EIi Lilly & Co.); VRTX-3 (Vertex Pharmaceuticals Inc.); WAY-132983 (Wyeth) and CI-101 7/ (PD-151832) (Pfizer Inc.).
  • Acetylcholinesterase inhibitors and compositions containing such inhibitors are described, e.g. in WO2006/071274, WO2006/070394, WO2006/040688, WO2005/092009, WO2005/079789, WO2005/039580, WO2005/027975, WO2004/084884, WO2004/037234, WO2004/032929, WO03/101458, WO03/091220, WO03/082820, WO03/020289, WO02/32412, WO01/85145, WO01/78728, WO01/66096, WO00/02549, WO01/00215, WO00/15205, WO00/23057, WO00/33840, WO00/30446, WO00/23057, WO00/15205, WO00/09483, WO00/07600, WO00/02549, WO99/47131 , WO99/073
  • Suitable acetylcholinesterase inhibitors for the purpose of the present invention are for example Donepezil (Eisai Co. Ltd.); rivastigmine (Novartis AG); (-)-phenserine (TorreyPines Therapeutics); ladostigil (Hebrew University of Jerusalem); huperzine A (Mayo Foundation); galantamine (Johnson & Johnson); Memoquin (Universita di Bologna); SP-004 (Samaritan Pharmaceuticals Inc.); BGC-20-1259 (Sankyo Co.
  • NMDA receptor antagonists and compositions containing such inhibitors are described, e.g. in WO2006/094674, WO2006/058236, WO2006/058059, WO2006/010965, WO2005/000216, WO2005/102390, WO2005/079779, WO2005/079756, WO2005/072705, WO2005/070429, WO2005/055996, WO2005/035522, WO2005/009421 , WO2005/000216, WO2004/092189, WO2004/039371 , WO2004/028522, WO2004/009062, WO03/010159, WO02/072542, WO02/34718, WO01/98262, WO01/94321 , WO01/92204, WO01/81295, WO01/32640, WO01/10833, WO01/10831 , WO00/5671 1 , WO00/290
  • Suitable NMDA receptor antagonists for the purpose of the present invention are for example Memantine (Merz & Co. GmbH); topiramate (Johnson & Johnson); AVP-923 (Neurodex) (Center for Neurologic Study); EN-3231 (Endo Pharmaceuticals Holdings Inc.); neramexane (MRZ-
  • CNP-3381 (Vernalis); perzinfotel (EAA-090, WAY-126090, EAA-129) (Wyeth); RGH-896 (Gedeon Richter Ltd.); traxoprodil (CP-101606), besonprodil (PD-196860, CI-1041 ) (Pfizer Inc.); CGX-1007 (Cognetix Inc.); delucemine (NPS-1506) (NPS Pharmaceuticals Inc.); EVT-101 (Roche Holding AG); acamprosate (Synchroneuron LLC); CR-3991 , CR-2249, CR-3394 (Rottapharm SpA.); AV-101 (4-CI-kynurenine (4-CI-KYN)), 7-chloro-kynurenic acid (7-CI-KYNA) (VistaGen); NPS-1407 (NPS Pharmaceuticals Inc.); YT-1006 (Yaupon Therapeutics Inc.); ED- 1812 (Sose
  • the present invention relates to combination therapies useful for the treatment of atherosclerosis, restenosis or arthritis, administering a QC inhibitor in combination with another therapeutic agent selected from the group consisting of inhibitors of the angiotensin converting enzyme (ACE); angiotensin Il receptor blockers; diuretics; calcium channel blockers (CCB); beta-blockers; platelet aggregation inhibitors; cholesterol absorption modulators; HMG-Co-A reductase inhibitors; high density lipoprotein (HDL) increasing compounds; renin inhibitors; IL-6 inhibitors; antiinflammatory corticosteroids; antiproliferative agents; nitric oxide donors; inhibitors of extracellular matrix synthesis; growth factor or cytokine signal transduction inhibitors; MCP-1 antagonists and tyrosine kinase inhibitors providing beneficial or synergistic therapeutic effects over each monotherapy component alone.
  • ACE angiotensin converting enzyme
  • CB calcium channel blockers
  • beta-blockers beta-blockers
  • Angiotensin Il receptor blockers are understood to be those active agents that bind to the AT1 - receptor subtype of angiotensin Il receptor but do not result in activation of the receptor. As a consequence of the blockade of the AT1 receptor, these antagonists can, e.g. be employed as antihypertensive agents.
  • Suitable angiotensin Il receptor blockers which may be employed in the combination of the present invention include AT 1 receptor antagonists having differing structural features, preferred are those with non-peptidic structures.
  • AT 1 receptor antagonists having differing structural features, preferred are those with non-peptidic structures.
  • Preferred AT1 -receptor antagonists are those agents that have been approved and reached the market, most preferred is valsartan, or a pharmaceutically acceptable salt thereof.
  • a suitable ACE inhibitor to be employed in the combination of the present invention is, e.g.
  • Preferred ACE inhibitors are those agents that have been marketed, most preferred are benazepril and enalapril.
  • a diuretic is, for example, a thiazide derivative selected from the group consisting of chlorothiazide, hydrochlorothiazide, methylclothiazide, and chlorothalidon.
  • the most preferred diuretic is hydrochlorothiazide.
  • a diuretic furthermore comprises a potassium sparing diuretic such as amiloride or triameterine, or a pharmaceutically acceptable salt thereof.
  • the class of CCBs essentially comprises dihydropyridines (DHPs) and non-DHPs, such as diltiazem-type and verapamil-type CCBs.
  • DHPs dihydropyridines
  • non-DHPs such as diltiazem-type and verapamil-type CCBs.
  • a CCB useful in said combination is preferably a DHP representative selected from the group consisting of amlodipine, felodipine, ryosidine, isradipine, lacidipine, nicardipine, nifedipine, niguldipine, niludipine, nimodipine, nisoldipine, nitrendipine and nivaldipine, and is preferably a non-DHP representative selected from the group consisting of flunarizine, prenylamine, diltiazem, fendiline, gallopamil, mibefradil, anipamil, tiapamil and verapamil, and in each case, a pharmaceutically acceptable salt thereof. All these CCBs are therapeutically used, e.g. as anti- hypertensive, anti-angina pectoris or anti-arrhythmic drugs.
  • Preferred CCBs comprise amlodipine, diltiazem, isradipine, nicardipine, nifedipine, nimodipine, nisoldipine, nitrendipine and verapamil or, e.g. dependent on the specific CCB, a pharmaceutically acceptable salt thereof.
  • DHP is amlodipine or a pharmaceutically acceptable salt thereof, especially the besylate.
  • An especially preferred representative of non-DHPs is verapamil or a pharmaceutically acceptable salt, especially the hydrochloride, thereof.
  • Beta-blockers suitable for use in the present invention include beta-adrenergic blocking agents (beta-blockers), which compete with epinephrine for beta-adrenergic receptors and interfere with the action of epinephrine.
  • beta-blockers are selective for the beta- adrenergic receptor as compared to the alpha-adrenergic receptors, and so do not have a significant alpha-blocking effect.
  • Suitable beta-blockers include compounds selected from acebutolol, atenolol, betaxolol, bisoprolol, carteolol, carvedilol, esmolol, labetalol, metoprolol, nadolol, oxprenolol, penbutolol, pindolol, propranolol, sotalol and timolol.
  • beta- blocker is an acid or base or otherwise capable of forming pharmaceutically acceptable salts or prodrugs
  • these forms are considered to be encompassed herein, and it is understood that the compounds may be administered in free form or in the form of a pharmaceutically acceptable salt or a prodrug, such as a physiologically hydrolyzable and acceptable ester.
  • a pharmaceutically acceptable salt or a prodrug such as a physiologically hydrolyzable and acceptable ester.
  • metoprolol is suitably administered as its tartrate salt
  • propranolol is suitably administered as the hydrochloride salt, and so forth.
  • Platelet aggregation inhibitors include PLAVIX® (clopidogrel bisulfate), PLETAL® (cilostazol) and aspirin.
  • Cholesterol absorption modulators include ZETIA® (ezetimibe) and KT6-971 (Kotobuki Pharmaceutical Co. Japan).
  • HMG-Co-A reductase inhibitors also called beta-hydroxy-beta-methylglutaryl-co-enzyme-A reductase inhibitors or statins
  • statins are understood to be those active agents which may be used to lower lipid levels including cholesterol in blood.
  • the class of HMG-Co-A reductase inhibitors comprises compounds having differing structural features.
  • the compounds which are selected from the group consisting of atorvastatin, cerivastatin, fluvastatin, lovastatin, pitavastatin, pravastatin, rosuvastatin and simvastatin, or in each case, a pharmaceutically acceptable salt thereof.
  • HMG-Co-A reductase inhibitors are those agents, which have been marketed, most preferred is atorvastatin, pitavastatin or simvastatin, or a pharmaceutically acceptable salt thereof.
  • HDL-increasing compounds include, but are not limited to, cholesterol ester transfer protein (CETP) inhibitors.
  • CETP inhibitors include JTT7O5 disclosed in Example 26 of U.S. Patent No. 6,426,365 issued July 30, 2002, and pharmaceutically acceptable salts thereof.
  • Interleukin 6 mediated inflammation may be achieved indirectly through regulation of endogenous cholesterol synthesis and isoprenoid depletion or by direct inhibition of the signal transduction pathway utilizing interleukin-6 inhibitor/antibody, interleukin-6 receptor inhibitor/antibody, interleukin-6 antisense oligonucleotide (ASON), gp130 protein inhibitor/antibody, tyrosine kinase inhibitors/antibodies, serine/threonine kinase inhibitors/antibodies, mitogen-activated protein (MAP) kinase inhibitors/antibodies, phosphatidylinositol 3-kinase (PI3K) inhibitors/antibodies, Nuclear factor kappaB (NF- ⁇ B) inhibitors/antibodies, IKB kinase (IKK) inhibitors/antibodies, activator protein-1 (AP-1 ) inhibitors/antibodies, STAT transcription factors inhibitors/antibodies, altered IL-6, partial peptid
  • a suitable antiinflammatory corticosteroid is dexamethasone.
  • Suitable antiproliferative agents are cladribine, rapamycin, vincristine and taxol.
  • a suitable inhibitor of extracellular matrix synthesis is halofuginone.
  • a suitable growth factor or cytokine signal transduction inhibitor is, e.g. the ras inhibitor R1 15777.
  • a suitable tyrosine kinase inhibitor is tyrphostin.
  • Suitable renin inhibitors are described, e.g. in WO 2006/116435.
  • a preferred renin inhibitor is aliskiren, preferably in the form of the hemi-fumarate salt thereof.
  • MCP-1 antagonists may, e.g. be selected from anti-MCP-1 antibodies, preferably monoclonal or humanized monoclonal antibodies, MCP-1 expression inhibitors, CCR2-antagonists, TNF-alpha inhibitors, VCAM-1 gene expression inhibitors and anti-C5a monoclonal antibodies.
  • MCP-1 antagonists and compositions containing such inhibitors are described, e.g. in WO02/070509, WO02/081463, WO02/060900, US2006/670364, US2006/677365, WO2006/097624, US2006/316449, WO2004/056727, WO03/053368, WO00/198289, WO00/157226, WO00/046195, WO00/046196, WO00/046199, WO00/046198, WO00/046197, WO99/046991 , WO99/007351 , WO98/006703, WO97/012615, WO2005/105133, WO03/037376, WO2006/125202, WO2006/085961 , WO2004/024921 , WO2006/074265.
  • Suitable MCP-1 antagonists are, for instance, C-243 (Telik Inc.); NOX-E36 (Noxxon Pharma AG); AP-761 (Actimis Pharmaceuticals Inc.); ABN-912, NIBR-177 (Novartis AG); CC-11006
  • anti-C5a monoclonal antibodies e.g. neutrazumab (G2 Therapies Ltd.); AZD-6942
  • Combinations of QC-inhibitors with MCP-1 antagonists may be useful for the treatment of inflammatory diseases in general, including neurodegenerative diseases.
  • Combinations of QC-inhibitors with MCP-1 antagonists are preferred for the treatment of Alzheimer's disease.
  • the QC inhibitor is combined with one or more compounds selected from the following group:
  • PF-4360365 m266, bapineuzumab, R-1450, Posiphen, (+)-phenserine, MK-0752, LY-450139, E-2012, (R)-flurbiprofen, AZD-103, AAB-001 (Bapineuzumab), Tramiprosate, EGb-761 , TAK- 070, Doxofylline, theophylline, cilomilast, tofimilast, roflumilast, tetomilast, tipelukast, ibudilast, HT-0712, MEM-1414, oglemilast, Linezolid, budipine, isocarboxazid, phenelzine, tranylcypromine, indantadol, moclobemide, rasagiline, ladostigil, safinamide, ABT-239, ABT- 834, GSK-189254A, Ciprox
  • a QC inhibitor preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1-30, in combination with Atorvastatin for the treatment and/or prevention of artherosclerosis, a QC inhibitor, preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1-30, in combination with immunosuppressive agents, preferably rapamycin for the prevention and/or treatment of restenosis, a QC inhibitor, preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1-30, in combination with immunosuppressive agents, preferably paclitaxel for the prevention and/or treatment of restenosis, - a QC inhibitor, preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1-30, in combination with AChE inhibitors, preferably Donepezil, for the prevention and/or treatment of Alzheimer's disease, a QC inhibitor,
  • a QC inhibitor for the prevention and/or treatment of multiple sclerosis a QC inhibitor, preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1-30, in combination with Copaxone, for the prevention and/or treatment of multiple sclerosis, - a QC inhibitor, preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1-30, in combination with dexamethasone, for the prevention and/or treatment of restenosis, a QC inhibitor, preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1-30, in combination with dexamethasone, for the prevention and/or treatment of atherosclerosis, a QC inhibitor, preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1-30, in combination with dexamethasone, for the prevention and/or treatment of rheumatid arthritis, a
  • the gamma-secretase inhibitor is selected from LY-450139, LY- 411575 and AN-37124.
  • Such a combination therapy is in particular useful for AD, FAD, FDD and neurodegeneration in Down syndrome as well as atherosclerosis, rheumatoid arthritis, restenosis and pancreatitis.
  • Such combination therapies might result in a better therapeutic effect (less proliferation as well as less inflammation, a stimulus for proliferation) than would occur with either agent alone.
  • At least one compound of formula (I) optionally in combination with at least one of the other aforementioned agents can be used as the active ingredient(s).
  • the active ingredient(s) is intimately admixed with a pharmaceutical carrier according to conventional pharmaceutical compounding techniques, which carrier may take a wide variety of forms depending of the form of preparation desired for administration, e.g., oral or parenteral such as intramuscular.
  • a pharmaceutical carrier may take a wide variety of forms depending of the form of preparation desired for administration, e.g., oral or parenteral such as intramuscular.
  • any of the usual pharmaceutical media may be employed.
  • suitable carriers and additives include water, glycols, oils, alcohols, flavoring agents, preservatives, coloring agents and the like;
  • suitable carriers and additives include starches, sugars, diluents, granulating agents, lubricants, binders, disintegrating agents and the like. Because of their ease in administration, tablets and capsules represent the most advantageous oral dosage unit form, in which case solid pharmaceutical carriers are obviously employed. If desired, tablets may be sugar coated or enteric coated by standard techniques.
  • the carrier will usually comprise sterile water, though other ingredients, for example, for purposes such as aiding solubility or for preservation, may be included.
  • Injectable suspensions may also prepared, in which case appropriate liquid carriers, suspending agents and the like may be employed.
  • the pharmaceutical compositions herein will contain, per dosage unit, e.g., tablet, capsule, powder, injection, teaspoonful and the like, an amount of the active ingredient(s) necessary to deliver an effective dose as described above.
  • the pharmaceutical compositions herein will contain, per dosage unit, e.g., tablet, capsule, powder, injection, suppository, teaspoonful and the like, from about 0.03 mg to 100 mg/kg (preferred 0.1
  • each active ingredient or combination thereof may be administered in any order.
  • the dosages may be varied depending upon the requirement of the patients, the severity of the condition being treated and the compound being employed. The use of either daily administration or post- periodic dosing may be employed.
  • compositions are in unit dosage forms from such as tablets, pills, capsules, powders, granules, sterile parenteral solutions or suspensions, metered aerosol or liquid sprays, drops, ampoules, autoinjector devices or suppositories; for oral parenteral, intranasal, sublingual or rectal administration, or for administration by inhalation or insufflation.
  • the composition may be presented in a form suitable for once-weekly or once-monthly administration; for example, an insoluble salt of the active compound, such as the decanoate salt, may be adapted to provide a depot preparation for intramuscular injection.
  • a pharmaceutical carrier e.g.
  • a solid preformulation composition containing a homogeneous mixture of a compound of the present invention, or a pharmaceutically acceptable salt thereof.
  • preformulation compositions as homogeneous, it is meant that the active ingredient is dispersed evenly throughout the composition so that the composition may be readily subdivided into equally effective dosage forms such as tablets, pills and capsules.
  • This solid preformulation composition is then subdivided into unit dosage forms of the type described above containing from 0.1 to about 500 mg of each active ingredient or combinations thereof of the present invention.
  • the tablets or pills of the compositions of the present invention can be coated or otherwise compounded to provide a dosage form affording the advantage of prolonged action.
  • the tablet or pill can comprise an inner dosage and an outer dosage component, the latter being in the form of an envelope over the former.
  • the two components can be separated by an enteric layer which serves to resist disintegration in the stomach and permits the inner component to pass intact into the duodenum or to be delayed in release.
  • enteric layers or coatings such materials including a number of polymeric acids with such materials as shellac, cetyl alcohol and cellulose acetate.
  • Suitable dispersing or suspending agents for aqueous suspensions include synthetic and natural gums such as tragacanth, acacia, alginate, dextran, sodium carboxymethylcellulose, methylcellulose, polyvinylpyrrolidone or gelatin.
  • the pharmaceutical composition may contain between about 0.01 mg and 100 mg, preferably about 5 to 50 mg, of each compound, and may be constituted into any form suitable for the mode of administration selected.
  • Carriers include necessary and inert pharmaceutical excipients, including, but not limited to, binders, suspending agents, lubricants, flavorants, sweeteners, preservatives, dyes, and coatings.
  • Compositions suitable for oral administration include solid forms, such as pills, tablets, caplets, capsules (each including immediate release, timed release and sustained release formulations), granules, and powders, and liquid forms, such as solutions, syrups, elixirs, emulsions, and suspensions.
  • Forms useful for parenteral administration include sterile solutions, emulsions and suspensions.
  • compounds of the present invention may be administered in a single daily dose, or the total daily dosage may be administered in divided doses of two, three or four times daily.
  • compounds for the present invention can be administered in intranasal form via topical use of suitable intranasal vehicles, or via transdermal skin patches well known to those of ordinary skill in that art.
  • the dosage administration will, of course, be continuous rather than intermittent throughout the dosage regimen.
  • the active drug component can be combined with an oral, non-toxic pharmaceutically acceptable inert carrier such as ethanol, glycerol, water and the like.
  • suitable binders include, without limitation, starch, gelatin, natural sugars such as glucose or betalactose, corn sweeteners, natural and synthetic gums such as acacia, tragacanth or sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride and the like.
  • Disintegrators include, without limitation, starch, methyl cellulose, agar, bentonite, xanthan gum and the like.
  • liquid forms in suitable flavored suspending or dispersing agents such as the synthetic and natural gums, for example, tragacanth, acacia, methyl-cellulose and the like.
  • suitable suspending or dispersing agents such as the synthetic and natural gums, for example, tragacanth, acacia, methyl-cellulose and the like.
  • tragacanth for example, tragacanth, acacia, methyl-cellulose and the like.
  • methyl-cellulose methyl-cellulose and the like.
  • suitable suspending or dispersing agents such as the synthetic and natural gums, for example, tragacanth, acacia, methyl-cellulose and the like.
  • sterile suspensions and solutions are desired.
  • Isotonic preparations which generally contain suitable preservatives are employed when intravenous administration is desired.
  • the compounds or combinations of the present invention can also be administered in the form of liposome delivery systems, such as small unilamellar vesicles, large unilamellar vesicles, and multilamellar vesicles.
  • Liposomes can be formed from a variety of phospholipids, such as cholesterol, stearylamine or phosphatidylcholines.
  • Compounds or combinations of the present invention may also be delivered by the use of monoclonal antibodies as individual carriers to which the compound molecules are coupled.
  • the compounds of the present invention may also be coupled with soluble polymers as targetable drug carriers.
  • Such polymers can include polyvinylpyrrolidone, pyran copolymer, polyhydroxypropylmethacrylamidephenol, polyhydroxyethylaspartamid-ephenol, or polyethyl eneoxidepolyllysine substituted with palmitoyl residue.
  • the compounds of the present invention may be coupled to a class of biodegradable polymers useful in achieving controlled release of a drug, for example, polyactic acid, polyepsilon caprolactone, polyhydroxy butyeric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacrylates and cross- linked or amphipathic block copolymers of hydrogels.
  • biodegradable polymers useful in achieving controlled release of a drug, for example, polyactic acid, polyepsilon caprolactone, polyhydroxy butyeric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacrylates and cross- linked or amphipathic block copolymers of hydrogels.
  • Compounds or combinations of this invention may be administered in any of the foregoing compositions and according to dosage regimens established in the art whenever treatment of the addressed disorders is required.
  • the daily dosage of the products may be varied over a wide range from 0.01 to 1.000 mg per mammal per day.
  • the compositions are preferably provided in the form of tablets containing, 0.01 , 0.05, 0.1 , 0.5, 1.0, 2.5, 5.0, 10.0, 15.0, 25.0, 50.0, 100, 150, 200,
  • an effective amount of the drug is ordinarily supplied at a dosage level of from about 0.1 mg/kg to about 300 mg/kg of body weight per day. Preferably, the range is from about 1 to about 50 mg/kg of body weight per day.
  • the compounds or combinations may be administered on a regimen of 1 to 4 times per day.
  • Optimal dosages to be administered may be readily determined by those skilled in the art, and will vary with the particular compound used, the mode of administration, the strength of the preparation, the mode of administration, and the advancement of disease condition. In addition, factors associated with the particular patient being treated, including patient age, weight, diet and time of administration, will result in the need to adjust dosages.
  • the invention also provides a process for preparing a pharmaceutical composition
  • a pharmaceutical composition comprising at least one compound of formula (I), optionally in combination with at least one of the other aforementioned agents and a pharmaceutically acceptable carrier.
  • compositions are preferably in a unit dosage form in an amount appropriate for the relevant daily dosage.
  • suitable dosages, including especially unit dosages, of the the compounds of the present invention include the known dosages including unit doses for these compounds as described or referred to in reference text such as the British and US Pharmacopoeias, Remington's Pharmaceutical Sciences (Mack Publishing Co.), Martindale The Extra Pharmacopoeia (London, The Pharmaceutical Press) (for example see the 31st Edition page 341 and pages cited therein) or the above mentioned publications. Examples
  • the filtrate was cooled down to 0 0 C and solid NaOH was added until a final pH-value of 10-12 is reached.
  • the aqueous solution was extracted by means of CHCI 3 (3x50 ml_). The combined organic layers were dried over Na 2 SO 4 , filtered and the solvent was removed.
  • the product was purified by means of flash-chromatography using silica gel and a CHCI 3 /MeOH-gradient containing aqueous ammonia (2% v/v). Yield: 0.31 g (65.1 %).
  • the system consisted of Merck-Hitachi device (model LaChrom) equipped with a SP250/21 Luna ® 100-7 C18 semi-preparative column (Phenomenex. length: 250 mm, diameter: 21 mm).
  • the compounds were purified using a gradient at a flow rate of 6 ml_/min; whereby eluent (A) was acetonitrile, eluent (B) was water, both containing 0.1 % (v/v) trifluoro acetic acid applying the following gradient: 0 min - 40 min. 40 -95 % (A)
  • the compound was synthesized starting from 1-ethyl-4-isothiocyanatobenzene (0.24 g, 1.5 mmol) and 3-(5-Methyl-1 H-imidazol-1-yl)propan-1 -amine (0.20 g, 1.5 mmol) as described above.
  • the compound was synthesized starting from 1-isopropyl-4-isothiocyanatobenzene (0.27 g, 1.5 mmol) and 3-(5-Methyl-1 H-imidazol-1 -yl)propan-1 -amine (0.20 g, 1.5 mmol) as described above.
  • the compound was synthesized starting from 2,3-dihydro-6-isothiocyanato-benzo[b][1 ,4]dioxine (0.29 g, 1.5 mmol) and 3-(5-Methyl-1 H-imidazol-1 -yl)propan-1 -amine (0.20 g, 1.5 mmol) as described above.
  • Example 4 2-Cyano(4-cyanophenyl)-3-(3-(5-methyl-1 H-imidazol-1 -yl)propyl)-guanidine
  • the compound was synthesized starting from 4-isothiocyanatobenzonitrile (0.24 g, 1.5 mmol) and 3-(5-Methyl-1 H-imidazol-1 -yl)propan-1 -amine (0.20 g, 1.5 mmol) as described above.
  • the compound was synthesized starting from 5-isothiocyanato-1 ,2,3-trimethoxybenzene
  • the compound was synthesized starting from 1-isothiocyanato-4-methoxybenzene (0.25 g, 1.5 mmol) and 3-(5-Methyl-1 H-imidazol-1 -yl)propan-1 -amine (0.20 g, 1.5 mmol) as described above.
  • the compound was synthesized starting from 4-isothiocyanato-1 ,2-dimethylbenzene (0.36 g, 2.2 mmol) and 3-(5-Methyl-1 H-imidazol-1 -yl)propan-1 -amine (0.20 g, 1.5 mmol) as described above.
  • Example 8 (3-(5-Methyl-1 H-imidazol-1 -yl)propyl)-2-cyano-3-mesitylguanidine
  • the compound was synthesized starting from 2-isothiocyanato-1 ,3,5-trimethylbenzene (0.266 g, 1.5 mmol) and 3-(5-Methyl-1 H-imidazol-1 -yl)propan-1 -amine (0.20 g, 1.5 mmol) as described above.
  • the compound was synthesized starting from 1-isothiocyanato-4-phenylbenzene (0.152 g, 0.72 mmol) and 3-(5-Methyl-1 H-imidazol-1 -yl)propan-1 -amine (0.10 g, 0.72 mmol) as described above.
  • the compound was synthesized starting from 2-isothiocyanatonaphthalene (0.185 g, 1.0 mmol) and 3-(5-Methyl-1 H-imidazol-1 -yl)propan-1 -amine (0.139 g, 1.0 mmol) as described above.
  • the compound was synthesized starting from 1-isothiocyanatonaphthalene (0.278 g, 1.5 mmol) and 3-(5-Methyl-1 H-imidazol-1 -yl)propan-1 -amine (0.20 g, 1.5 mmol) as described above.
  • the compound was synthesized starting from 5-isothiocyanatobenzo[c][1 ,2,5]thiadiazole (0.290 g, 1.5 mmol) and 3-(5-Methyl-1 H-imidazol-1-yl)propan-1 -amine (0.20 g, 1.5 mmol) as described above.
  • the compound was synthesized starting from 1 ,2-dichloro-4-isothiocyanatobenzene (0.16 g, 0.8 mmol) and 3-(5-Methyl-1 H-imidazol-1-yl)propan-1 -amine (0.14 g, 1.0 mmol) as described above. Yield: 0.22 g (78.5%).
  • the compound was synthesized starting from 5-isothiocyanatobenzo[d][1 ,3]dioxole (0.14 g, 0.80 mmol) and 3-(5-Methyl-1 H-imidazol-1-yl)propan-1 -amine (0.14 g, 1.0 mmol) as described above.
  • Example 15 2-Cyano(4-methoxyphenyl)-3-(3-(5-methyl-1 H-imidazol-1 - yl)propyl)guanidine
  • the compound was synthesized starting from 1-isothiocyanato-4-methoxybenzene (0.17 g, 1.0 mmol) and 3-(5-Methyl-1 H-imidazol-1-yl)propan-1 -amine (0.17 g, 1.2 mmol) as described above.
  • the compound was synthesized starting from 1-isothiocyanato-3,5-dimethoxybenzene (0.293 g, 1.5 mmol) and 3-(5-Methyl-1 H-imidazol-1-yl)propan-1 -amine (0.20 g, 1.5 mmol) as described above.
  • the compound was synthesized starting from 1-ethoxy-4-isothiocyanatobenzene (0.108 g, 0.60 mmol) and 3-(4-Methyl-1 H-imidazol-1 -yl)propan-1 -amine (0.084 g, 0.60 mmol) as described above.
  • the compound was synthesized starting from 1-isothiocyanato-3,5-dimethoxybenzene (0.117 g, 0.60 mmol) and 3-(4-Methyl-1 H-imidazol-1 -yl)propan-1 -amine (0.084 g, 0.60 mmol) as described above.
  • the compound was synthesized starting from 2-isothiocyanato-1 ,3,5-trimethylbenzene (0.106 g, 0.60 mmol) and 3-(4-Methyl-1 H-imidazol-1-yl)propan-1 -amine (0.084 g, 0.60 mmol) as described above.
  • Example 21 2-cyano(4-isopropylphenyl)-3-(3-(4-methyl-1 H-imidazol-1 -yl)propyl)- guanidine
  • the compound was synthesized starting from 1-isopropyl-4-isothiocyanatobenzene (0.106 g, 0.60 mmol) and 3-(4-Methyl-1 H-imidazol-1 -yl)propan-1 -amine (0.084 g, 0.60 mmol) as described above.
  • the compound was synthesized starting from 1-ethyl-4-isothiocyanatobenzene (0.100 g, 0.60 mmol) and 3-(4-Methyl-1 H-imidazol-1 -yl)propan-1 -amine (0.084 g, 0.60 mmol) as described above.
  • the compound was synthesized starting from 1-isothiocyanatonaphthalene (0.11 1 g, 0.60 mmol) and 3-(4-Methyl-1 H-imidazol-1 -yl)propan-1 -amine (0.084 g, 0.60 mmol) as described above. Yield: 0.15 g (75.2%).
  • the compound was synthesized starting from 5-isothiocyanatobenzo[c][1 ,2,5]thiadiazole (0.139 g, 0.72 mmol) and 3-(4-Methyl-1 H-imidazol-1 -yl)propan-1 -amine (0.10 g, 0.72 mmol) as described above.
  • the compound was synthesized starting from 5-isothiocyanato-1 ,2,3-trimethoxybenzene
  • the compound was synthesized starting from 1 ,2-dichloro-4-isothiocyanatobenzene (0.147 g, 0.72 mmol) and 3-(4-Methyl-1 H-imidazol-1 -yl)propan-1 -amine (0.10 g, 0.72 mmol) as described above.
  • the compound was synthesized starting from 1-isothiocyanato-4-methoxybenzene (0.119 g, 0.72 mmol) and 3-(4-Methyl-1 H-imidazol-1 -yl)propan-1 -amine (0.10 g, 0.72 mmol) as described above. Yield: 0.090 g (40.0%).
  • the compound was synthesized starting from 1-isothiocyanato-4-phenylbenzene (0.38O g, 1.18 mmol) and 3-(4-Methyl-1 H-imidazol-1 -yl)propan-1 -amine (0.164 g, 1.18 mmol) as described above.
  • Example 30 2-cyano(3-(4-methyl-1 H-imidazol-1 -yl)propyl)-3-(naphthalen-2-yl)-guanidine
  • the compound was synthesized starting from 2-isothiocyanatonaphthalene (0.167 g, 0.90 mmol) and 3-(4-Methyl-1 H-imidazol-1 -yl)propan-1 -amine (0.125 g, 0.90 mmol) as described above.
  • QC activity was determined from a standard curve of ⁇ -naphthylamine under assay conditions. One unit is defined as the amount of QC catalyzing the formation of 1 ⁇ mol pGlu- ⁇ NA from H-GIn- ⁇ NA per minute under the described conditions.
  • QC was activity determined using H-GIn-AMC as substrate. Reactions were carried out at 30 0 C utilizing the NOVOStar reader for microplates (BMG labtechnologies). The samples consisted of varying concentrations of the fluorogenic substrate, 0.1 U pyroglutamyl aminopeptidase (Qiagen) in 0.05 M Tris/HCI, pH 8.0 containing 5 mM EDTA and an appropriately diluted aliquot of QC in a final volume of 250 ⁇ l. Excitation/emission wavelengths were 380/460 nm. The assay reactions were initiated by addition of glutaminyl cyclase. QC activity was determined from a standard curve of 7-amino-4-methylcoumarin under assay conditions. The kinetic data were evaluated using GraFit sofware.
  • This novel assay was used to determine the kinetic parameters for most of the QC substrates.
  • QC activity was analyzed spectrophotometrically using a continuous method, that was derived by adapting a previous discontinuous assay (Bateman, R. C. J. 1989 J Neurosci Methods 30, 23-28) utilizing glutamate dehydrogenase as auxiliary enzyme.
  • Samples consisted of the respective QC substrate, 0.3 mM NADH, 14 mM ⁇ -Ketoglutaric acid and 30 U/ml glutamate dehydrogenase in a final volume of 250 ⁇ l. Reactions were started by addition of QC and persued by monitoring of the decrease in absorbance at 340 nm for 8-15 min.
  • the initial velocities were evaluated and the enzymatic activity was determined from a standard curve of ammonia under assay conditions. All samples were measured at 30 0 C, using either the SPECTRAFIuor Plus or the Sunrise (both from TECAN) reader for microplates. Kinetic data was evaluated using GraFit software.
  • Inhibitor assay For inhibitor testing, the sample composition was the same as described above, except of the putative inhibitory compound added. For a rapid test of QC-inhibition, samples contained 4 mM of the respective inhibitor and a substrate concentration at 1 K M . For detailed investigations of the inhibition and determination of K-values, influence of the inhibitor on the auxiliary enzymes was investigated first. In every case, there was no influence on either enzyme detected, thus enabling the reliable determination of the QC inhibition. The inhibitory constant was evaluated by fitting the set of progress curves to the general equation for competitive inhibition using GraFit software.
  • the compounds were analyzed using a gradient at a flow rate of 1 mL/min; whereby eluent (A) was acetonitrile, eluent (B) was water, both containing 0.1 % (v/v) trifluoro acetic acid applying the following gradient: Method [A]: 0 min - 5 min -» 5% (A), 5 min - 17 min -» 5 - 15% (A), 15 min - 27 min -» 15 - 95% (A) 27 min - 30 min -> 95% (A).
  • the high resolution positive ion ESI mass spectra were obtained from a Bruker Apex III 7Oe Fourier transform ion cyclotron resonance mass spectrometer (Bruker Daltonics, Billerica, USA) equipped with an InfinityTM cell, a 7.0 Tesla superconducting magnet (Bruker, Düsseldorf, Germany), an RF-only hexapole ion guide and an external electrospray ion source (API Apollo, voltages: endplate, -3.700V; capillary, -4.400V; capillary exit, 100V; skimmer 1.15 V; skimmer 2.6 V). Nitrogen was used as drying gas at 15O 0 C.
  • the sample solutions were introduced continuously via a syringe pump with a flow rate of 120 ⁇ l h "1 . All data were acquired with 256 k data points and zero filled to 1024 k by averaging 32 scans. The melting points were detected utilizing a Kofler melting point device. They are not corrected The 1 H NMR-Spectra (500 MHz) were recorded at a BRUKER AC 500. The solvent was DMSO- D 6 , unless otherwise specified. Chemial shifts are expressed as parts per million (ppm) downfiled from tetramethylsilan. Splitting patterns have been designated as follows: s (singulet), d (doublet), dd (doublet of doublet), t (triplet), m (multiplet) and br (broad signal).
  • Matrix-assisted laser desorption/ionization mass spectrometry was carried out using the Hewlett-Packard G2025 LD-TOF System with a linear time of flight analyzer.
  • the instrument was equipped with a 337 nm nitrogen laser, a potential acceleration source (5 kV) and a 1.0 m flight tube.
  • Detector operation was in the positive-ion mode and signals are recorded and filtered using LeCroy 9350M digital storage oscilloscope linked to a personal computer. Samples (5 ⁇ l) were mixed with equal volumes of the matrix solution.
  • DHAP/DAHC For matrix solution DHAP/DAHC was used, prepared by solving 30 mg 2 ' ,6 ' -dihydroxyacetophenone (Aldrich) and 44 mg diammonium hydrogen citrate (Fluka) in 1 ml acetonitrile/0.1% TFA in water (1/1 , v/v). A small volume ( ⁇ 1 ⁇ l) of the matrix-analyte-mixture was transferred to a probe tip and immediately evaporated in a vacuum chamber (Hewlett-Packard G2024A sample prep accessory) to ensure rapid and homogeneous sample crystallization.
  • a vacuum chamber Hewlett-Packard G2024A sample prep accessory
  • a ⁇ -derived peptides were incubated in 10O ⁇ l 0.1 M sodium acetate buffer, pH 5.2 or 0.1 M Bis-Tris buffer, pH 6.5 at 30 0 C. Peptides were applied in 0.5 mM [A ⁇ (3-1 1 )a] or 0.15 mM [A ⁇ (3-21 )a] concentrations, and 0.2 U QC is added all 24 hours. In case of A ⁇ (3-21 )a, the assays contained 1 % DMSO.
  • samples are removed from the assay tube, peptides extracted using ZipTips (Millipore) according to the manufacturer ' s recommendations, mixed with matrix solution (1 :1 v/v) and subsequently the mass spectra recorded. Negative controls either contain no QC or heat deactivated enzyme.
  • the sample composition was the same as described above, with exception of the inhibitory compound added (5 mM or 2 mM of a test compound of the invention).
  • the first QC inhibitors were disclosed in WO 2004/098591 and WO 2005/075436. There are no other potent QC inhibitors known in the art. The same holds true for combinations and compositions for the treatment of neuronal diseases comprising QC inhibitors.
  • Compounds and combinations of the invention may have the advantage that they are, for example, more potent, more selective, have fewer side-effects, have better formulation and stability properties, have better pharmacokinetic properties, be more bioavailable, be able to cross blood brain barrier and are more effective in the brain of mammals, are more compatible or effective in combination with other drugs or be more readily synthesized than other compounds of the prior art.
  • the invention embraces all combinations of preferred and more preferred groups and embodiments of groups recited above.

Abstract

The present invention relates to compounds of formula (I), combinations and uses thereof for disease therapy, or a pharmaceutically acceptable salt, solvate or polymorph thereof, including all tautomers and stereoisomers thereof wherein: A represents formula (A) or formula (B) and B, R1, R2, R3, R4, R5, R6 and Z are as defined throughout the description and the claims.

Description

CYANO-GUANIDINE DERIVATIVES AS GLUTAMINYL CYCLASE INHIBITORS
Field of the invention
The invention relates to novel cyanoguanidine derivatives as inhibitors of glutaminyl cyclase (QC, EC 2.3.2.5). QC catalyzes the intramolecular cyclization of N-terminal glutamine residues into pyroglutamic acid (5-oxo-prolyl, pGlu*) under liberation of ammonia and the intramolecular cyclization of N-terminal glutamate residues into pyroglutamic acid under liberation of water.
Background of the invention Glutaminyl cyclase (QC, EC 2.3.2.5) catalyzes the intramolecular cyclization of N-terminal glutamine residues into pyroglutamic acid (pGlu*) liberating ammonia. A QC was first isolated by Messer from the latex of the tropical plant Carica papaya in 1963 (Messer, M. 1963 Nature 4874, 1299). 24 years later, a corresponding enzymatic activity was discovered in animal pituitary (Busby, W. H. J. et al. 1987 J Biol Chem 262, 8532-8536; Fischer, W. H. and Spiess, J. 1987 Proc Natl Acad Sci U S A 84, 3628-3632). For the mammalian QC, the conversion of GIn into pGlu by QC could be shown for the precursors of TRH and GnRH (Busby, W. H. J. et al. 1987 J Biol Chem 262, 8532-8536; Fischer, W. H. and Spiess, J. 1987 Proc Natl Acad Sci U S A 84, 3628-3632). In addition, initial localization experiments of QC revealed a co-localization with its putative products of catalysis in bovine pituitary, further improving the suggested function in peptide hormone synthesis (Bockers, T. M. et al. 1995 J Neuroendocrinol 7, 445- 453). In contrast, the physiological function of the plant QC is less clear. In the case of the enzyme from C. papaya, a role in the plant defense against pathogenic microorganisms was suggested (El Moussaoui, A. et al.2001 Cell MoI Life Sci 58, 556-570). Putative QCs from other plants were identified by sequence comparisons recently (Dahl, S. W. et al.2000 Protein Expr Purif 20, 27-36). The physiological function of these enzymes, however, is still ambiguous.
The QCs known from plants and animals show a strict specificity for L-Glutamine in the N- terminal position of the substrates and their kinetic behavior was found to obey the Michaelis- Menten equation (Pohl, T. et al. 1991 Proc Natl Acad Sci U S A 88, 10059-10063; Consalvo, A. P. et al. 1988 Anal Biochem 175, 131-138; Gololobov, M. Y. et al. 1996 Biol Chem Hoppe Seyler 377, 395-398). A comparison of the primary structures of the QCs from C. papaya and that of the highly conserved QC from mammals, however, did not reveal any sequence homology (Dahl, S. W. et al. 2000 Protein Expr Purif 20, 27-36). Whereas the plant QCs appear to belong to a new enzyme family (Dahl, S. W. et al. 2000 Protein Expr Purif 20, 27-36), the mammalian QCs were found to have a pronounced sequence homology to bacterial aminopeptidases (Bateman, R. C. et al. 2001 Biochemistry 40, 11246-11250), leading to the conclusion that the QCs from plants and animals have different evolutionary origins.
Recently, it was shown that recombinant human QC as well as QC-activity from brain extracts catalyze both, the N-terminal glutaminyl as well as glutamate cyclization. Most striking is the finding, that cyclase-catalyzed Glurconversion is favored around pH 6.0 while Glnrconversion to pGlu-derivatives occurs with a pH-optimum of around 8.0. Since the formation of pGlu-Aβ- related peptides can be suppressed by inhibition of recombinant human QC and QC-activity from pig pituitary extracts, the enzyme QC is a target in drug development for treatment of Alzheimer's disease.
First inhibitors of QC are described in WO 2004/098625, WO 2004/098591 , WO 2005/039548 and WO 2005/075436.
EP 02 01 1 349.4 discloses polynucleotides encoding insect glutaminyl cyclase, as well as polypeptides encoded thereby and their use in methods of screening for agents that reduce glutaminyl cyclase activity. Such agents are useful as pesticides.
Definitions
The terms "k," or "Kf and "K0" are binding constants, which describe the binding of an inhibitor to and the subsequent release from an enzyme. Another measure is the "IC50" value, which reflects the inhibitor concentration, which at a given substrate concentration results in 50 % enzyme activity.
The term "DP IV-inhibitor" or "dipeptidyl peptidase IV inhibitor" is generally known to a person skilled in the art and means enzyme inhibitors, which inhibit the catalytic activity of DP IV or DP IV-like enzymes.
"DP IV-activity" is defined as the catalytic activity of dipeptidyl peptidase IV (DP IV) and DP IV- like enzymes. These enzymes are post-proline (to a lesser extent post-alanine, post-serine or post-glycine) cleaving serine proteases found in various tissues of the body of a mammal including kidney, liver, and intestine, where they remove dipeptides from the N-terminus of biologically active peptides with a high specificity when proline or alanine form the residues that are adjacent to the N-terminal amino acid in their sequence. The term "PEP-inhibitor" or "prolyl endopeptidase inhibitor" is generally known to a person skilled in the art and means enzyme inhibitors, which inhibit the catalytic activity of prolyl endopeptidase (PEP, prolyl oligopeptidase, POP).
"PEP-activity" is defined as the catalytic activity of an endoprotease that is capable to hydrolyze post proline bonds in peptides or proteins were the proline is in amino acid position 3 or higher counted from the N-terminus of a peptide or protein substrate.
The term "QC" as used herein comprises glutaminyl cyclase (QC) and QC-like enzymes. QC and QC-like enzymes have identical or similar enzymatic activity, further defined as QC activity. In this regard, QC-like enzymes can fundamentally differ in their molecular structure from QC. Examples of QC-like enzymes are the glutaminyl-peptide cyclotransferase-like proteins (QPCTLs) from human (GenBank NM_017659), mouse (GenBank BC058181 ), Macaca fascicularis (GenBank AB168255), Macaca mulatta (GenBank XM_001 110995), Canis familiaris (GenBank XM_541552), Rattus norvegicus (GenBank XM_001066591 ), Mus musculus (GenBank BC058181 ) and Bos taurus (GenBank BT026254).
The term "QC activity" as used herein is defined as intramolecular cyclization of N-terminal glutamine residues into pyroglutamic acid (pGlu*) or of N-terminal L-homoglutamine or L-β- homoglutamine to a cyclic pyro-homoglutamine derivative under liberation of ammonia. See therefore schemes 1 and 2.
Scheme 1 : Cyclization of glutamine by QC
Figure imgf000004_0001
Scheme 2: Cyclization of L-homoglutamine by QC
Figure imgf000005_0001
The term "EC" as used herein comprises the activity of QC and QC-like enzymes as glutamate cyclase (EC), further defined as EC activity.
The term "EC activity" as used herein is defined as intramolecular cyclization of N-terminal glutamate residues into pyroglutamic acid (pGlu*) by QC. See therefore scheme 3.
Scheme 3: N-terminal cyclization of uncharged glutamyl peptides by QC (EC)
Figure imgf000005_0002
The term "QC-inhibitor" "glutaminyl cyclase inhibitor" is generally known to a person skilled in the art and means enzyme inhibitors, which inhibit the catalytic activity of glutaminyl cyclase (QC) or its glutamyl cyclase (EC) activity.
Potency of QC inhibition
In light of the correlation with QC inhibition, in preferred embodiments, the subject method and medical use utilize an agent with an IC5O for QC inhibition of 10 μM or less, more preferably of 1 μM or less, even more preferably of 0.1 μM or less or 0.01 μM or less, or most preferably 0.001 μM or less. Indeed, inhibitors with K, values in the lower micromolar, preferably the nanomolar and even more preferably the picomolar range are contemplated. Thus, while the active agents are described herein, for convenience, as "QC inhibitors", it will be understood that such nomenclature is not intending to limit the subject of the invention to a particular mechanism of action.
Molecular weight of QC inhibitors
In general, the QC inhibitors of the subject method or medical use will be small molecules, e.g., with molecular weights of 500 g/mole or less, 400 g/mole or less, preferably of 350 g/mole or less, and even more preferably of 300 g/mole or less and even of 250 g/mole or less.
The term "subject" as used herein, refers to an animal, preferably a mammal, most preferably a human, who has been the object of treatment, observation or experiment.
The term "therapeutically effective amount" as used herein, means that amount of active compound or pharmaceutical agent that elicits the biological or medicinal response in a tissue system, animal or human being sought by a researcher, veterinarian, medical doctor or other clinician, which includes alleviation of the symptoms of the disease or disorder being treated.
As used herein, the term "pharmaceutically acceptable" embraces both human and veterinary use: For example the term "pharmaceutically acceptable" embraces a veterinarily acceptable compound or a compound acceptable in human medicine and health care.
Throughout the description and the claims the expression "alkyl", unless specifically limited, denotes a CM2 alkyl group, suitably a Ci-6 alkyl group, e.g. Ci-4 alkyl group. Alkyl groups may be straight chain or branched. Suitable alkyl groups include, for example, methyl, ethyl, propyl (e.g. n-propyl and isopropyl), butyl (e.g n-butyl, iso-butyl, sec-butyl and tert-butyl), pentyl (e.g. n- pentyl), hexyl (e.g. n-hexyl), heptyl (e.g. n-heptyl) and octyl (e.g. n-octyl). The expression "alk", for example in the expressions "alkoxy", "haloalkyl" and "thioalkyl" should be interpreted in accordance with the definition of "alkyl". Exemplary alkoxy groups include methoxy, ethoxy, propoxy (e.g. n-propoxy), butoxy (e.g. n-butoxy), pentoxy (e.g. n-pentoxy), hexoxy (e.g. n- hexoxy), heptoxy (e.g. n-heptoxy) and octoxy (e.g. n-octoxy). Exemplary thioalkyl groups include methylthio-. Exemplary haloalkyl groups include fluoroalkyl e.g. CF3.
The expression "alkenyl", unless specifically limited, denotes a C2-i2 alkenyl group, suitably a C2-6 alkenyl group, e.g. a C2-4 alkenyl group, which contains at least one double bond at any desired location and which does not contain any triple bonds. Alkenyl groups may be straight chain or branched. Exemplary alkenyl groups including one double bond include propenyl and butenyl. Exemplary alkenyl groups including two double bonds include pentadienyl, e.g. (1 E, 3E)-pentadienyl.
The expression "alkynyl", unless specifically limited, denotes a C2-i2 alkynyl group, suitably a C2-6 alkynyl group, e.g. a C2-4 alkynyl group, which contains at least one triple bond at any desired location and may or may not also contain one or more double bonds. Alkynyl groups may be straight chain or branched. Exemplary alkynyl groups include propynyl and butynyl.
The expression "alkylene" denotes a chain of formula -(CH2)n- wherein n is an integer e.g. 2-5, unless specifically limited.
The expression "cycloalkyl", unless specifically limited, denotes a C3-i0 cycloalkyl group (i.e. 3 to 10 ring carbon atoms), more suitably a C3-S cycloalkyl group, e.g. a C3-6 cycloalkyl group. Exemplary cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and cyclooctyl. A most suitable number of ring carbon atoms is three to six.
The expression "cycloalkenyl", unless specifically limited, denotes a C5-io cycloalkenyl group (i.e. 5 to 10 ring carbon atoms), more suitably a C5-8 cycloalkenyl group e.g. a C5-6 cycloalkenyl group. Exemplary cycloalkenyl groups include cyclopropenyl, cyclohexenyl, cycloheptenyl and cyclooctenyl. A most suitable number of ring carbon atoms is five to six.
The expression "carbocyclyl", unless specifically limited, denotes any ring system in which all the ring atoms are carbon and which contains between three and twelve ring carbon atoms, suitably between three and ten carbon atoms and more suitably between three and eight carbon atoms. Carbocyclyl groups may be saturated or partially unsaturated, but do not include aromatic rings. Examples of carbocyclyl groups include monocyclic, bicyclic, and tricyclic ring systems, in particular monocyclic and bicyclic ring systems. Other carbocylcyl groups include bridged ring systems (e.g. bicyclo[2.2.1]heptenyl). A specific example of a carbocyclyl group is a cycloalkyl group. A further example of a carbocyclyl group is a cycloalkenyl group.
The expression "heterocyclyl", unless specifically limited, refers to a carbocyclyl group wherein one or more (e.g. 1 , 2 or 3) ring atoms are replaced by heteroatoms selected from N, S and O.
A specific example of a heterocyclyl group is a cycloalkyl group (e.g. cyclopentyl or more particularly cyclohexyl) wherein one or more (e.g. 1 , 2 or 3, particularly 1 or 2, especially 1 ) ring atoms are replaced by heteroatoms selected from N, S or O. Exemplary heterocyclyl groups containing one hetero atom include pyrrolidine, tetrahydrofuran and piperidine, and exemplary heterocyclyl groups containing two hetero atoms include morpholine and piperazine. A further specific example of a heterocyclyl group is a cycloalkenyl group (e.g. a cyclohexenyl group) wherein one or more (e.g. 1 , 2 or 3, particularly 1 or 2, especially 1 ) ring atoms are replaced by heteroatoms selected from N, S and O. An example of such a group is dihydropyranyl (e.g. 3,4- dihydro-2H-pyran-2-yl-).
The expression "aryl", unless specifically limited, denotes a C6-i2 aryl group, suitably a C6-io aryl group, more suitably a C6-S aryl group. Aryl groups will contain at least one aromatic ring (e.g. one, two or three rings). An example of a typical aryl group with one aromatic ring is phenyl. An example of a typical aryl group with two aromatic rings is naphthyl.
The expression "heteroaryl", unless specifically limited, denotes an aryl residue, wherein one or more (e.g. 1 , 2, 3, or 4, suitably 1 , 2 or 3) ring atoms are replaced by heteroatoms selected from
N, S and O, or else a 5-membered aromatic ring containing one or more (e.g. 1 , 2, 3, or 4, suitably 1 , 2 or 3) ring atoms selected from N, S and O. Exemplary monocyclic heteroaryl groups having one heteroatom include: five membered rings (e.g. pyrrole, furan, thiophene); and six membered rings (e.g. pyridine, such as pyridin-2-yl, pyridin-3-yl and pyridin-4-yl). Exemplary monocyclic heteroaryl groups having two heteroatoms include: five membered rings
(e.g. pyrazole, oxazole, isoxazole, thiazole, isothiazole, imidazole, such as imidazol-1-yl, imidazol-2-yl imidazol-4-yl); six membered rings (e.g. pyridazine, pyrimidine, pyrazine).
Exemplary monocyclic heteroaryl groups having three heteroatoms include: 1 ,2,3-triazole and
1 ,2,4-triazole. Exemplary monocyclic heteroaryl groups having four heteroatoms include tetrazole. Exemplary bicyclic heteroaryl groups include: indole (e.g. indol-6-yl), benzofuran, benzthiophene, quinoline, isoquinoline, indazole, benzimidazole, benzthiazole, quinazoline and purine.
The expression "-alkylaryl", unless specifically limited, denotes an aryl residue which is connected via an alkylene moiety e.g. a Ci-4alkylene moiety.
The expression "-alkylheteroaryl", unless specifically limited, denotes a heteroaryl residue which is connected via an alkylene moiety e.g. a C-i^alkylene moiety.
The term "halogen" or "halo" comprises fluorine (F), chlorine (Cl) and bromine (Br). The term "amino" refers to the group -NH2.
The term "phenyl substituted by phenyl" refers to biphenyl.
Stereoisomers:
All possible stereoisomers of the claimed compounds are included in the present invention.
Where the compounds according to this invention have at least one chiral center, they may accordingly exist as enantiomers. Where the compounds possess two or more chiral centers, they may additionally exist as diastereomers. It is to be understood that all such isomers and mixtures thereof are encompassed within the scope of the present invention.
Preparation and isolation of stereoisomers: Where the processes for the preparation of the compounds according to the invention give rise to a mixture of stereoisomers, these isomers may be separated by conventional techniques such as preparative chromatography. The compounds may be prepared in racemic form, or individual enantiomers may be prepared either by enantiospecific synthesis or by resolution. The compounds may, for example, be resolved into their components enantiomers by standard techniques, such as the formation of diastereomeric pairs by salt formation with an optically active acid, such as (-)-di-p-toluoyl-d-tartaric acid and/or (+)-di-p-toluoyl-l-tartaric acid followed by fractional crystallization and regeneration of the free base. The compounds may also be resolved by formation of diastereomeric esters or amides, followed by chromatographic separation and removal of the chiral auxiliary. Alternatively, the compounds may be resolved using a chiral HPLC column.
Pharmaceutically acceptable salts:
In view of the close relationship between the free compounds and the compounds in the form of their salts or solvates, whenever a compound is referred to in this context, a corresponding salt, solvate or polymorph is also intended, provided such is possible or appropriate under the circumstances.
Salts and solvates of the compounds of formula (I) and physiologically functional derivatives thereof which are suitable for use in medicine are those wherein the counter-ion or associated solvent is pharmaceutically acceptable. However, salts and solvates having non- pharmaceutically acceptable counter-ions or associated solvents are within the scope of the present invention, for example, for use as intermediates in the preparation of other compounds and their pharmaceutically acceptable salts and solvates.
Suitable salts according to the invention include those formed with both organic and inorganic acids or bases. Pharmaceutically acceptable acid addition salts include those formed from hydrochloric, hydrobromic, sulfuric, nitric, citric, tartaric, phosphoric, lactic, pyruvic, acetic, trifluoroacetic, triphenylacetic, sulfamic, sulfanilic, succinic, oxalic, fumaric, maleic, malic, mandelic, glutamic, aspartic, oxaloacetic, methanesulfonic, ethanesulfonic, arylsulfonic (for example p-toluenesulfonic, benzenesulfonic, naphthalenesulfonic or naphthalenedisulfonic), salicylic, glutaric, gluconic, tricarballylic, cinnamic, substituted cinnamic (for example, phenyl, methyl, methoxy or halo substituted cinnamic, including 4-methyl and 4-methoxycinnamic acid), ascorbic, oleic, naphthoic, hydroxynaphthoic (for example 1- or 3-hydroxy-2-naphthoic), naphthaleneacrylic (for example naphthalene-2-acrylic), benzoic, 4-methoxybenzoic, 2- or A- hydroxybenzoic, 4-chlorobenzoic, 4-phenylbenzoic, benzeneacrylic (for example 1 ,4- benzenediacrylic), isethionic acids, perchloric, propionic, glycolic, hydroxyethanesulfonic, pamoic, cyclohexanesulfamic, salicylic, saccharinic and trifluoroacetic acid. Pharmaceutically acceptable base salts include ammonium salts, alkali metal salts such as those of sodium and potassium, alkaline earth metal salts such as those of calcium and magnesium and salts with organic bases such as dicyclohexylamine and Λ/-methyl-D-glucamine.
All pharmaceutically acceptable acid addition salt forms of the compounds of the present invention are intended to be embraced by the scope of this invention.
Polymorph crystal forms: Furthermore, some of the crystalline forms of the compounds may exist as polymorphs and as such are intended to be included in the present invention. In addition, some of the compounds may form solvates with water (i.e. hydrates) or common organic solvents, and such solvates are also intended to be encompassed within the scope of this invention. The compounds, including their salts, can also be obtained in the form of their hydrates, or include other solvents used for their crystallization.
Prodrugs:
The present invention further includes within its scope prodrugs of the compounds of this invention. In general, such prodrugs will be functional derivatives of the compounds which are readily convertible in vivo into the desired therapeutically active compound. Thus, in these cases, the methods of treatment of the present invention, the term "administering" shall encompass the treatment of the various disorders described with prodrug versions of one or more of the claimed compounds, but which converts to the above specified compound in vivo after administration to the subject. Conventional procedures for the selection and preparation of suitable prodrug derivatives are described, for example, in "Design of Prodrugs", ed. H. Bundgaard, Elsevier, 1985.
Protective Groups:
During any of the processes for preparation of the compounds of the present invention, it may be necessary and/or desirable to protect sensitive or reactive groups on any of the molecules concerned. This may be achieved by means of conventional protecting groups, such as those described in Protective Groups in Organic Chemistry, ed. J.F.W. McOmie, Plenum Press, 1973; and T.W. Greene & P. G. M. Wuts, Protective Groups in Organic Synthesis, John Wiley & Sons, 1991 , fully incorporated herein by reference. The protecting groups may be removed at a convenient subsequent stage using methods known from the art.
As used herein, the term "composition" is intended to encompass a product comprising the claimed compounds in the therapeutically effective amounts, as well as any product which results, directly or indirectly, from combinations of the claimed compounds.
Carriers and Additives for galenic formulations:
Thus, for liquid oral preparations, such as for example, suspensions, elixirs and solutions, suitable carriers and additives may advantageously include water, glycols, oils, alcohols, flavoring agents, preservatives, coloring agents and the like; for solid oral preparations such as, for example, powders, capsules, gelcaps and tablets, suitable carriers and additives include starches, sugars, diluents, granulating agents, lubricants, binders, disintegrating agents and the like.
Carriers, which can be added to the mixture, include necessary and inert pharmaceutical excipients, including, but not limited to, suitable binders, suspending agents, lubricants, flavorants, sweeteners, preservatives, coatings, disintegrating agents, dyes and coloring agents.
Soluble polymers as targetable drug carriers can include polyvinylpyrrolidone, pyran copolymer, polyhydroxypropylmethacrylamidephenol, polyhydroxyethylaspartamide-phenol, or polyethyleneoxidepolyllysine substituted with palmitoyl residue. Furthermore, the compounds of the present invention may be coupled to a class of biodegradable polymers useful in achieving controlled release of a drug, for example, polyactic acid, polyepsilon caprolactone, polyhydroxy butyeric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacrylates and cross- linked or amphipathic block copolymers of hydrogels.
Suitable binders include, without limitation, starch, gelatin, natural sugars such as glucose or betalactose, corn sweeteners, natural and synthetic gums such as acacia, tragacanth or sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride and the like.
Disintegrators include, without limitation, starch, methyl cellulose, agar, bentonite, xanthan gum and the like.
Summary of the invention
According to the invention there are provided compounds of formula (I),
Figure imgf000012_0001
or a pharmaceutically acceptable salt, solvate or polymorph thereof, including all tautomers and stereoisomers thereof wherein: R1 represents alkyl; alkenyl, wherein the double bond is not adjacent to the nitrogen; carbocyclyl; -Ci-6alkyl-carbocyclyl; heterocyclyl; -Ci-6alkyl-heterocyclyl; aryl; heteroaryl; -Ci- 6alkylaryl; -Ci-6alkylheteroaryl; -phenyl fused to carbocyclyl or -phenyl fused to heterocyclyl; in which any of the aforesaid carbocyclyl and heterocyclyl groups may optionally be substituted by one or more groups selected from methyl and oxo; and in which any of the aforesaid phenyl, aryl and heteroaryl groups may optionally be substituted by one or more substituents selected from Ci-6alkyl, C2-6alkenyl, C2-6alkynyl, Ci- 6haloalkyl, - Ci-6thioalkyl, -SO2Ci-4alkyl, Ci-6alkoxy-, -O-C3-8cycloalkyl, C3-8cycloalkyl, -SO2C3- βcycloalkyl, C3-6alkenyloxy-, C3-6alkynyloxy-, -C(O)Ci-6alkyl, Ci-6alkoxy-Ci.6alkyl-, nitro, halogen, cyano, hydroxyl, -C(O)OH, -NH2, -NHC^alkyl, -N(C1-4alkyl)(C1-4alkyl), -C(O)N(Ci-4alkyl)(d. 4alkyl), -C(O)NH2, -C(O)NH(d-4alkyl), C(O)OCi-6alkyl, -SOCi-4alkyl and -SOCs-ecycloalkyl; or R1 represents phenyl substituted by phenyl, or phenyl substituted by an optionally substituted monocyclic heteroaryl group in which any of the aforesaid phenyl and monocyclic heteroaryl groups may optionally be substituted by one or more groups selected from Ci-4alkyl, halogen and Ci-4alkoxy; or R1 represents phenyl substituted by benzyloxy- in which any of the aforesaid phenyl or benzyloxy groups may optionally be substituted on the ring by one or more groups selected from Ci-4alkyl, halogen and Ci-4alkoxy; and
A represents
Figure imgf000013_0001
wherein Y represents a C2-5 alkylene chain, which may optionally be substituted by one or two methyl groups or may optionally be substituted by two alkylene substituents at the same position wherein the two alkylene substituents are joined to each other to form a C3-5spiro- cycloalkyl group and
R2, R3 and R4 independently represent H or Ci-2alkyl, provided that R2 and R3 and R4 do not all represent H; or A represents
Figure imgf000013_0002
wherein Z represents a bond, -CH2-, -CH2-CH2-, -CH(Me)-, -CH(Me)-CH2- Or -CH2-CH(Me)- and
R5 and R6 independently represent H or Ci-2alkyl and
B represents H or methyl. Typically R1 represents alkyl; alkenyl, wherein the double bond is not adjacent to the nitrogen; carbocyclyl; -Ci-6alkyl-carbocyclyl; heterocyclyl; -Ci-6alkyl-heterocyclyl; aryl; heteroaryl; -Ci- 6alkylaryl; -Ci-6alkylheteroaryl; -phenyl fused to carbocyclyl or -phenyl fused to heterocyclyl; in which any of the aforesaid carbocyclyl and heterocyclyl groups may optionally be substituted by one or more groups selected from methyl and oxo; and in which any of the aforesaid phenyl, aryl and heteroaryl groups may optionally be substituted by one or more substituents selected from Ci-6alkyl, C2-6alkenyl, C2-6alkynyl, Ci- 6haloalkyl, - Ci-6thioalkyl, -SO2Ci-4alkyl, Ci-6alkoxy-, -O-Cs-βcycloalkyl, C3-8cycloalkyl, -SO2C3- βcycloalkyl, C3-6alkenyloxy-, C3-6alkynyloxy-, -C(O)Ci-6alkyl, d-ealkoxy-d-ealkyl-, nitro, halogen, cyano, hydroxyl, -C(O)OH, -NH2, -NHCi-4alkyl, -N(C1-4alkyl)(C1-4alkyl), -C(O)N(Ci-4alkyl)(Ci. 4alkyl), -C(O)NH2 and -C(O)NH(Ci-4alkyl); or R1 represents phenyl substituted by phenyl, or phenyl substituted by an optionally substituted monocyclic heteroaryl group in which any of the aforesaid phenyl and monocyclic heteroaryl groups may optionally be substituted by one or more groups selected from Ci-4alkyl, halogen and Ci-4alkoxy.
Compounds of formula (I) are provided as either the (£) or (Z) isomer at the double bond substituted by cyano or as a mixture thereof. The coverage of the (£) or (Z) isomer or mixture thereof is denoted by ^A/WP
Detailed description of the invention
When carbocyclyl and heterocyclyl are substituted, they are typically substituted by 1 or 2 substituents (e.g. 1 substitent). Typically the substituent is methyl. More typically carbocyclyl and heterocyclyl groups are unsubstituted.
When aryl and heteroaryl are substituted, they are typically substituted by 1 , 2 or 3 (e.g. 1 or 2) substituents. Substituents for aryl and heteroaryl are selected from Ci-6alkyl (e.g. methyl), C2- 6alkenyl (e.g. buten-3-yl), C2-6alkynyl (e.g. butyn-3-yl), Ci-6haloalkyl (e.g. fluoromethyl, trifluoromethyl), -Ci-6thioalkyl (e.g. -S-methyl), -SO2Ci-4alkyl (e.g. -SO2methyl), Ci-6alkoxy- (e.g. methoxy, ethoxy), -O-C3-8cycloalkyl (e.g. -O-cyclopentyl), C3-8cycloalkyl (e.g. cyclopropyl, cyclohexyl), -SO2C3-8cycloalkyl (e.g. -SO2cyclohexyl), C3-6alkenyloxy- (e.g. -O-buten-2-yl), C3- 6alkynyloxy- (e.g. -O-buten-2-yl), -C(O)d-6alkyl (e.g. -C(O)ethyl), -C(O)OCi-6alkyl (e.g. -C(O)O- methyl), d-ealkoxy-d-ealkyl- (e.g. methoxy-ethyl-), nitro, halogen (e.g. fluoro, chloro, bromo), cyano, hydroxyl, -C(O)OH, -NH2, -NHCi-4alkyl (e.g. -NHmethyl), -N(C1-4alkyl)(C1-4alkyl) (e.g. - N(methyl)2), -C(O)N(d-4alkyl)(d-4alkyl) (e.g. -C(O)N(methyl)2), -C(O)NH2 and -C(O)NH(Ci- 4alkyl) (e.g. -C(O)NHmethyl). Further suitable examples are -C(O)OCi-6alkyl (e.g. C(O)OMe), - SOCi-4alkyl (e.g. SOMe) and -SOCs-ecycloalkyl (e.g. -SO-cyclopropyl). More typically, substituents will be selected from Ci-6alkyl (e.g. methyl), Ci-6haloalkyl (e.g. Ci-6fluoroalkyl, e.g. CF3), Ci-6alkoxy (e.g. OMe), halogen, hydroxy and cyano. Most typically, substituents will be selected from Ci-6alkyl (e.g. methyl), Ci-6haloalkyl (e.g. Ci-6fluoroalkyl, e.g. CF3), Ci-6alkoxy (e.g. OMe), halogen and hydroxy.
When R1 represents alkyl, examples include propyl (e.g. n-propyl, isopropyl), butyl (e.g. n-butyl- sec-butyl, isobutyl and tert-butyl), pentyl (e.g. n-pentyl, 3,3,-dimethylpropyl), hexyl, heptyl and octyl.
When R1 represents alkenyl, examples include propen-2-yl (i.e. -CH2-CH=CH2), buten-2-yl, buten-3-yl and penten-3-yl.
When R1 represents carbocyclyl (which may optionally be substituted), examples include cycloalkyl and cycloalkenyl. Examples of cycloalkyl include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and cycloheptyl. Examples of cycloalkenyl include cyclohexenyl (e.g. cyclohex-2- enyl, cyclohex-3-enyl). Examples of substituted carbocyclyl include 2-methyl-cyclohexyl-, 3- methyl-cyclohexyl-, 4-methyl-cyclohexyl-, 2-methyl-cyclohex-2-enyl, 2-methyl-cyclohex-3-enyl, S-methyl-cyclohex-S-enyl, 3-methyl-cyclohex-3-enyl.
When R1 represents -C-i-βalkyl-carbocyclyl (which may optionally be substituted), examples include -methyl-cyclopentyl, -methyl-cyclohexyl, -ethyl-cyclohexyl, -propyl-cyclohexyl, -methyl- cyclohexenyl, -ethyl-cyclohexenyl, -methyl(4-methylcyclohexyl) and -propyl (3- methylcyclyohexyl).
When R1 represents heterocyclyl (which may optionally be substituted), examples include tetrahydrofuranyl, morpholinyl, piperdinyl, 3,4-dihydro-2H-pyranyl, pyrrolidinyl, methyltetrahydrofuranyl- (e.g. 5-methyltetrahydrofuran-2-yl).
When R1 represents -Ci-βalkyl-heterocyclyl (which may optionally be substituted), examples include -methyl-tetrahydrofuranyl (e.g. -methyl-tetrahydrofuran-2-yl, -methyl-tetrahydrofuran-3- yl), -ethyl-tetrahydrofuranyl, -methyl-piperidinyl. When R1 represents optionally substituted aryl, aryl may typically represent phenyl. Aryl may most typically represent substituted phenyl. Exemplary substituted phenyl groups include 2,4- dichlorophenyl-, 2,4-diflurorophenyl-, 2,4-dimethoxyphenyl-, 2,4-dimethylphenyl-2,4- bis(trifluoromethyl)phenyl-, 2,4,6-trifluorophenyl-, 2,4,6-trimethylphenyl-, 2,6-dichlorophenyl-, 2,6-difluorophenyl-, 2,6-dimethoxyphenyl-, 2-isopropyl-6-methylphenyl-, 3-(cyclopentyloxy)-4- methoxyphenyl-, 3,4,5-trimethoxyphenyl-, 3,4-dimethoxyphenyl-, 3,4-dichlorophenyl-, 3,4- dimethylphenyl-, 3,4,5-trifluorophenyl-, 3,5-bis(trifluroromethyl)phenyl-, 3,5-dimethoxyphenyl-, 3- methoxyphenyl-, 4-(trifluoromethyl)phenyl-, 4-bromo-2-(trifluoromethyl)phenyl-, 4-bromophenyl-, 4-chloro-3-(trifluoromethyl)phenyl-, 4-chlorophenyl-, 4-cyanophenyl-, 4-ethoxyphenyl-, A- ethylphenyl-, 4-fluorophenyl-, 4-isopropylphenyl-, 4-methoxyphenyl-. Alternatively, R1 may represents unsubstituted phenyl-.
When R1 represents optionally substituted aryl and aryl represents naphthyl, examples include unsubstituted naphthyl (e.g. naphthalen-1-yl, naphthalen-2-yl, naphthalen-3-yl) as well as substituted naphthyl (e.g. 4-methyl-naphthalen-2-yl-, 5-methyl-naphthalen-3-yl-, 7-methyl- naphthalen-3-y- and 4-fluoro-naphthalen-2-yl-).
When R1 represents optionally substituted heteroaryl, examples include monocyclic rings (e.g. 5 or 6 membered rings) and bicyclic rings (e.g. 9 or 10 membered rings) which may optionally be substituted. Example 5 membered rings include pyrrolyl (e.g. pyrrol-2-yl) and imidazolyl (e.g. 1 H-imidazol-2-yl or 1 H-imidazol-4-yl), pyrazolyl (e.g. 1 H-pyrazol-3-yl), furanyl (e.g. furan-2-yl), thiazolyl (e.g. thiazol-2-yl), thiophenyl (e.g. thiophen-2-yl, thiophen-3-yl). Example 6 membered rings include pyridinyl (e.g. pyridine-2-yl and pyridine-4-yl). Specific substituents that may be mentioned are one or more e.g. 1 , 2 or 3 groups selected from halogen, hydroxyl, alkyl (e.g. methyl) and alkoxy- (e.g. methoxy-). Example substituted 5 membered rings include 4,5- dimethyl-furan-2-yl-, 5-hydroxymethyl-furan-2-yl-, 5-methyl-furan-2-yl- and 6-methyl-pyridin-2-yl- . An example substituted 6-membered ring is 1-oxy-pyridin-4-yk Example 9 membered rings include 1 H indolyl (e.g. 1 H-indol-3-yl, 1 H-indol-5-yl), benzothiophenyl (e.g. benzo[b]thiophen-3- yl, particularly 2-benzo[b]thiophen-3-yl), benzo[1 ,2,5]-oxadiazolyl (e.g. benzo[1 ,2,5]-oxadiazol-5- yl), benzo[1 ,2,5]-thiadiazolyl (e.g. benzo[1 ,2,5]-thiadiazol-5-yl). Example 10 membered rings include quinolinyl (e.g.quinolin-3-yl, quinolin-4-yl, quinolin-8-yl). Specific substituents that may be mentioned are one or more e.g. 1 , 2 or 3 groups selected from halogen, hydroxyl, alkyl (e.g. methyl) and alkoxy- (e.g. methoxy-). Example substituted 9-membered rings include 1-methyl- 1 H-indol-3-yl, 2-methyl-1 H-indol-3-yl, 6-methyl-1 H-indol-3-yl. Example substituted 10 membered rings include 2-chloro-quinolin-3-yl, 8-hydroxy-quinolin-2-yl, oxo-chromenyl (e.g.4-oxo-4H- chromen-3-yl) and 6-methyl-4-oxo-4H-chromen-3-yl.
When R1 represents -alkylaryl in which aryl is optionally substituted, examples include -Ci- 4alkylaryl. Another specific group is -alkyl(substituted phenyl) e.g. in which phenyl is substituted by one or more groups selected from alkyl, fluoroalkyl, halogen and alkoxy (e.g. methyl, trifluoromethyl, tert-butyl, chloro, fluoro and methoxy) and, for example, alkyl is Ci-4 alkyl. Another specific group is -alkyl(bicyclic aryl) e.g. wherein bicyclic aryl is optionally substituted naphthyl. A further specific group is benzyl.
When R1 represents -alkylheteroaryl in which heteroaryl is optionally substituted, examples include -Ci-4alkylheteroaryl e.g. -methylheteroaryl and -ethylheteroaryl (e.g. 1-heteroarylethyl- and 2-heteroarylethyl-), -propylheteroaryl and -butylheteroaryl in which heteroaryl is optionally substituted. Specific examples of -alkylheteroaryl groups include pyridinylmethyl-, N-methyl- pyrrol-2-methyl- N-methyl-pyrrol-2-ethyl-, N-methyl-pyrrol-3-methyl-, N-methyl-pyrrol-3-ethyl-, 2- methyl-pyrrol-1 -methyl-, 2-methyl-pyrrol-1 -ethyl-, 3-methyl-pyrrol-1 -methyl-, 3-methyl-pyrrol-1 - ethyl-, 4-pyridino-methyl-, 4-pyridino-ethyl-, 2-(thiazol-2-yl)-ethyl-, 2-ethyl-indol-1 -methyl-, 2- ethyl-indol-1 -ethyl-, 3-ethyl-indol-1 -methyl-, 3-ethyl-indol-1 -ethyl-, 4-methyl-pyridin-2-methyl-, 4- methyl-pyridin-2-yl-ethyl-, 4-methyl-pyridin-3-methyl-, 4-methyl-pyridin-3-ethyl-.
When R1 represents optionally substituted -phenyl fused to optionally substituted carbocyclyl, examples include indanyl (e.g. indan-4-yl-, 2-methyl-indan-4-yl-), indenyl and tetralinyl.
When R1 represents optionally substituted -phenyl fused to optionally substituted heterocyclyl, examples include benzo[1 ,3]dioxo-4-yl- and 2,3-dihydro-benzo[1 ,4]dioxin-4-yl-.
When R1 represents phenyl substituted by phenyl, or phenyl substituted by a monocyclic heteroaryl group, in which any of aforesaid phenyl and heteroaryl groups may optionally be substituted, typically the phenyl ring connected directly to the nitrogen atom is unsubstituted and the terminal phenyl ring or the monocyclic heteroaryl ring is optionally substituted by one, two or three substitutents (e.g. one or two, e.g. one). Typically the terminal phenyl or monocyclic heteroaryl group is unsubstituted. Typically the terminal phenyl or monocyclic heteroaryl group substitutes the other phenyl group at the 4-position. Examples include -biphenyl-4-yl and 4- (oxazol-5-yl)phenyk When R1 represents phenyl substituted by benzyloxy- in which any of the aforesaid phenyl or benzyloxy groups may optionally be substituted on the ring by one or more groups selected from Ci^alkyl, halogen and C-i^alkoxy, examples include 4-(benzyloxy)phenyk Further exampes include 4-((4-fluoro-benzyl)oxy)phenyl-, 4-((4-chloro-benzyl)oxy)phenyl- and 4-((4- methoxy-benzyl)oxy)phenyk Typically the terminal benzyloxy group substitutes the phenyl group at the 4-position. Typically the phenyl ring connected directly to the nitrogen atom is unsubstituted.
When A represents
Figure imgf000018_0001
Examples of R2 include H, methyl and ethyl.
Examples of R3 include H, methyl and ethyl.
Examples of R4 include H, methyl and ethyl.
Examples of group Y include -(CHz)2-, -(CHz)3-, -(CHz)4-. -(CHz)5-, -CH2CH(Me)CH2-, -CH2CH(Me)CH2CH2-, -CH2CH2CH(Me)CH2- and
Figure imgf000018_0002
wherein the imidazole ring is on the left hand side
When A represents
Figure imgf000019_0001
Examples of R5 include H, methyl and ethyl.
Examples of R6 include H, methyl and ethyl.
Suitably R1 represents alkyl, aryl; carbocyclyl; heteroaryl; -phenyl fused to carbocyclyl; phenyl fused to heterocyclyl; or R1 represents phenyl substituted by optionally substituted phenyl or phenyl substituted by a monocyclic heteroaryl group; any of which aforesaid aryl, carbocyclyl, heteroaryl, phenyl and heterocyclyl may optionally be substituted.
When R1 represents optionally substituted aryl, R1 suitably represents optionally substituted phenyl, especially substituted phenyl. Example substituents are selected from Ci-6alkyl (e.g. methyl, ethyl, isopropyl), Ci-6alkoxy (e.g. methoxy), -O-C3-8cycloalkyl (e.g. -O-cyclopentyl), Ci- 6haloalkyl (e.g. trifluoromethyl) and halogen (e.g. chloro).
More suitably R1 represents aryl; heteroaryl; -phenyl fused to carbocyclyl; phenyl fused to monocyclic heterocyclyl; or R1 represents phenyl substituted by phenyl, or phenyl substituted by a monocyclic heteroaryl group; in which any of the aforesaid phenyl, heteroaryl, carbocyclyl and heterocyclyl may optionally be substituted.
In one embodiment R1 represents optionally substituted aryl. In another embodiment R1 represents optionally substituted heteroaryl. In a third embodiment R1 represents optionally substituted phenyl fused to optionally substituted carbocyclyl. In another embodiment R1 represents optionally substituted phenyl fused to optionally substituted heterocyclyl. In a further embodiment R1 represents optionally substituted phenyl substituted by optionally substituted phenyl or optionally substituted phenyl substituted by an optionally substituted monocyclic heteroaryl group. When R1 represents optionally substituted aryl, R1 suitably represents optionally substituted phenyl, especially substituted phenyl.
When R1 represents substituted phenyl, phenyl suitably has one, two or three substituents (e.g. one or two substituents, e.g. one substituent, e.g. one substituent ). When phenyl has three substituents, the substituents are for example at the 2, 4 and 6 positions of the phenyl ring or alternatively at the 3, 4, and 5 positions of the phenyl ring. When phenyl has two substituents, the substituents are for example at the 3 and 4 positions of the phenyl ring or at the 3 and 5 positions of the phenyl ring. When phenyl has one substituent, the substituent is at the 2, 3 or 4 position of the phenyl ring, most suitably at the 4-position of the phenyl ring.
Example substituents are selected from Ci-6alkyl (e.g. methyl, ethyl, isopropyl), Ci-6alkoxy (e.g. methoxy), C-i-βhaloalkyl (e.g. trifluoromethyl) and halogen (e.g. chloro). Another example substituent is cyano. Further example substituents are selected from Ci-3alkyl (e.g. methyl, ethyl, isopropyl), Ci-3alkoxy (e.g. methoxy), Ci-3haloalkyl (e.g. trifluoromethyl), halogen (e.g. chloro) and cyano. Specific examples include 2,4,6-trimethylphenyl-, 3,4,5-trimethoxyphenyl-, 3,4-dichlorophenyl-, 3,4-dimethylphenyl-, 3,5-dimethoxyphenyl-, 4-methoxyphenyl-, 4- cyanophenyl-, 4-ethoxyphenyl-, 4-ethylphenyl-, 4-isopropylphenyl- or 4-methoxyphenyl-.
When R1 represents unsubstituted aryl, R1 suitably represents phenyl, naphthalen-1-yl or naphthalen-2-yl.
When R1 represents optionally substituted heteroaryl, R1 suitably represents benzo[c][1 ,2,5]thiadiazol-6-yl.
When R1 represents optionally substituted phenyl fused to optionally substituted heterocyclyl, R1 suitably represents 2,3-dihydrobenzo[b][1 ,4]dioxin-7-yl or benzo[d][1 ,3]dioxol-6-yl.
When R1 represents phenyl substituted by phenyl, or phenyl substituted by a monocyclic heteroaryl group in which any of aforesaid phenyl and/or monocyclic heteroaryl may optionally be substituted. R1 suitably represents biphenyl-4-yl.
More suitably R1 represents aryl; heteroaryl; phenyl fused to heterocyclyl; or R1 represents phenyl substituted by phenyl, or phenyl substituted by a monocyclic heteroaryl group group in which any of the aforementioned phenyl, aryl, heteroaryl and heterocyclyl may optionally be substituted. Most suitably R1 represents substituted phenyl or R1 represents phenyl fused to heterocyclyl.
Most suitably R1 represents substituted phenyl. Alternatively, most suitably R1 represents phenyl fused to heterocyclyl.
Suitably A represents
Figure imgf000021_0001
R2 suitably represents H,
R3 suitably represents H or methyl.
R4 suitably represents H or methyl.
In one embodiment of the invention, R3 represents H and R4 represents methyl. In another embodiment, R3 represents methyl and R4 represents H. Most suitably R2 represents H, R3 represents H and R4 represents methyl.
Suitably Y represents an unsubstituted C2-5 alkylene chain. More suitably Y represents -(CH2)3- or -(CH2)4-. In one embodiment, Y represents -(CH2)3-- In another embodiment, Y represents
-(CHz)4-.
When Y represents a C2-5 alkylene chain, which is substituted by two alkylene substituents at the same position wherein the two alkylene substituents are joined to each other to form a C3- 5spiro-cycloalkyl group, the spiro-cycloalkyl group is suitably C3spiro-cycloalkyl.
Alternatively A represents
Figure imgf000022_0001
In one embodiment R5 represents H and R6 represents H. In another embodiment R5 represents H and R6 represents Ci-2alkyl. In a third embodiment R5 represents Ci-2alkyl and R6 represents
H.
Suitably Z represents a bond, -CH2- or -CH2CH2-. In one embodiment Z represents a bond. In another embodiment, Z represents -CH2-. In a third embodiment, Z represents -CH2CH2-.
More suitably A represents
Figure imgf000022_0002
Most suitably A represents
Figure imgf000022_0003
particularly
Figure imgf000023_0001
B suitably represents H.
When benzimidazolyl is shown as benzimidazol-5-yl, which is represented as:
Figure imgf000023_0002
the person skilled in the art will appreciate that benzimidazol-6-yl, which is represented as:
Figure imgf000023_0003
is an equivalent structure. As employed herein, the two forms of benzimidazolyl are covered by the term "benzimidazol-5-yl".
Processes
A process for preparation of a compound of formula (I)
Figure imgf000023_0004
wherein R1, A and B are defined as above. comprises reaction of a compound of formula (II)
Figure imgf000024_0001
with a compound of formula (III).
Figure imgf000024_0003
The reaction is typically performed in the presence of a reagent that removes hydrogen disulfide (e.g. a carbodiimide such as N1-((ethylimino)methylen)-N3,N3-dimethylpropan-1 ,3-diamine hydrochloride).
The reaction may be carried out in a polar organic solvent or a mixture thereof (e.g. a mixture of ethanol and dimethylformamide).
A compound of formula (II) may be prepared by reaction of a compound of formula (IV)
Figure imgf000024_0002
wherein M+ represents a metal ion (e.g. sodium ion, Na+) with a compound of formula (V)
Figure imgf000024_0004
The reaction may suitably be carried out in a polar protic organic solvent (e.g. an alcohol such as ethanol) at elevated temperature.
Compounds of formulae (III). (IV) and (V) are either known or may be prepared by conventional methods known per se. Alternatively a compound of formula (I) may be prepared by reaction of a compound of formula (Vl)
Figure imgf000025_0001
wherein R7 represents Ci^alkyl e.g. methyl with a compound of formula (III).
Figure imgf000025_0002
The reaction may suitably be carried out in a polar protic organic solvent (e.g. an alcohol such as ethanol) at elevated temperature.
A compound of formula (Vl) may be prepared by reaction of a compound of formula (VII)
Figure imgf000025_0003
wherein R8 represents Ci-6alkyl e.g. methyl with a compound of formula (VIII)
Figure imgf000025_0004
The reaction may suitably be carried out in a polar protic organic solvent (e.g. an alcohol such as ethanol) at elevated temperature. Compounds of formula (Vl) may be formed in situ i.e. the compounds of formula (Vl) need not be isolated from the reaction mixture after reaction of compounds of formula (VII) and (VIII) before onwards reaction to compounds of formula (I).
Compounds of formulae (III), (VII) and (VIII) are either known or may be prepared by conventional methods known per se. See for example Buchholz et al, J. Med. Chem., 2006, 49(2), p664-677.
For example, when A represents
Figure imgf000026_0001
wherein R , R , R and Y are defined as above and B represents H, a compound of formula (Ilia)
Figure imgf000026_0002
may be prepared from a compound of formula (IX)
Figure imgf000026_0003
by cleavage of the isoindolin-1 ,3-dione group (e.g. by use of hydrazine). A compound of formula (IX) may be prepared by deprotection of a compound of formula (X)
Figure imgf000027_0001
wherein PG represents a protecting group (e.g. trityl) and L" represents a suitable counterion such as Br". (Suitable deprotecting conditions include use of trifluoroacetic acid when PG repesents trityl).
A compound of formula (X) may be prepared by reaction of a compound of formula (Xl)
Figure imgf000027_0002
with a compound of formula (XII)
Figure imgf000027_0003
wherein L represents a suitable leaving group e.g. Br.
The reaction may typically be carried out at elevated temperature in an organic solvent (e.g. acetonitrile). A compound of formula (Xl) may be prepared from a compound of formula (XIII)
Figure imgf000028_0001
in the presence of a base (e.g. triethylamine) and a suitable protecting reagent (e.g. chlorotriphenylmethane) in a polar organic solvent (e.g. dimethylformamide).
Compounds of formula (XII) and (XIII) are either known or may be prepared by conventional methods known per se.
Therapeutic uses
Physiological substrates of QC (EC) in mammals are, e.g. amyloid beta-peptides (3-40), (3-42), (1 1-40 and (11-42), ABri, ADan, Gastrin, Neurotensin, FPP, CCL 2, CCL 7, CCL 8, CCL 16, CCL 18, Fractalkine, Orexin A, [Gln3]-glucagon(3-29), [Gln5]-substance P(5-1 1 ) and the peptide QYNAD. For further details see table 1. The compounds and/or combinations according to the present invention and pharmaceutical compositions comprising at least one inhibitor of QC (EC) are useful for the treatment of conditions that can be treated by modulation of QC activity.
Table 1 : Amino acid sequences of physiological active peptides with an N-terminal glutamine residue, which are prone to be cyclized to final pGlu
Figure imgf000028_0002
Figure imgf000029_0001
Figure imgf000030_0001
Figure imgf000031_0001
Figure imgf000032_0001
Figure imgf000033_0001
Figure imgf000034_0001
Glutamate is found in positions 3, 11 and 22 of the amyloid β-peptide. Among them the mutation from glutamic acid (E) to glutamine (Q) in position 22 (corresponding to amyloid precursor protein APP 693, Swissprot P05067) has been described as the so called Dutch type cerebroarterial amyloidosis mutation.
The β-amyloid peptides with a pyroglutamic acid residue in position 3, 1 1 and/or 22 have been described to be more cytotoxic and hydrophobic than the amyloid β-peptides 1-40(42/43) (Saido T.C. 2000 Medical Hypotheses 54(3): 427-429). The multiple N-terminal variations, e.g. Abeta(3-40), Abeta(3-42), Abeta(11-40) and Abeta (11- 42) can be generated by the β-secretase enzyme β-site amyloid precursor protein-cleaving enzyme (BACE) at different sites (Huse JT. et al. 2002 J. Biol. Chem. 277 (18): 16278-16284), and/or by aminopeptidase or dipeptidylaminopeptidase processing from the full lenght peptides Abeta(1-40) and Abeta(1-42). In all cases, cyclization of the then N-terminal occuring glutamic acid residue is catalyzed by QC.
Transepithelial transducing cells, particularly the gastrin (G) cell, co-ordinate gastric acid secretion with the arrival of food in the stomach. Recent work showed that multiple active products are generated from the gastrin precursor, and that there are multiple control points in gastrin biosynthesis. Biosynthetic precursors and intermediates (progastrin and Gly-gastrins) are putative growth factors; their products, the amidated gastrins, regulate epithelial cell proliferation, the differentiation of acid-producing parietal cells and histamine-secreting enterochromaffin-like (ECL) cells, and the expression of genes associated with histamine synthesis and storage in ECL cells, as well as acutely stimulating acid secretion. Gastrin also stimulates the production of members of the epidermal growth factor (EGF) family, which in turn inhibit parietal cell function but stimulate the growth of surface epithelial cells. Plasma gastrin concentrations are elevated in subjects with Helicobacter pylori, who are known to have increased risk of duodenal ulcer disease and gastric cancer (Dockray, GJ. 1999 J Physiol 15 315-324).
The peptide hormone gastrin, released from antral G cells, is known to stimulate the synthesis and release of histamine from ECL cells in the oxyntic mucosa via CCK-2 receptors. The mobilized histamine induces acid secretion by binding to the H(2) receptors located on parietal cells. Recent studies suggest that gastrin, in both its fully amidated and less processed forms (progastrin and glycine-extended gastrin), is also a growth factor for the gastrointestinal tract. It has been established that the major trophic effect of amidated gastrin is for the oxyntic mucosa of stomach, where it causes increased proliferation of gastric stem cells and ECL cells, resulting in increased parietal and ECL cell mass. On the other hand, the major trophic target of the less processed gastrin (e.g. glycine-extended gastrin) appears to be the colonic mucosa (Koh, TJ. and Chen, D. 2000 Regul Pept 9337^4).
Neurotensin (NT) is a neuropeptide implicated in the pathophysiology of schizophrenia that specifically modulates neurotransmitter systems previously demonstrated to be misregulated in this disorder. Clinical studies in which cerebrospinal fluid (CSF) NT concentrations have been measured revealed a subset of schizophrenic patients with decreased CSF NT concentrations that are restored by effective antipsychotic drug treatment. Considerable evidence also exists concordant with the involvement of NT systems in the mechanism of action of antipsychotic drugs. The behavioral and biochemical effects of centrally administered NT remarkably resemble those of systemically administered antipsychotic drugs, and antipsychotic drugs increase NT neurotransmission. This concatenation of findings led to the hypothesis that NT functions as an endogenous antipsychotic. Moreover, typical and atypical antipsychotic drugs differentially alter NT neurotransmission in nigrostriatal and mesolimbic dopamine terminal regions, and these effects are predictive of side effect liability and efficacy, respectively (Binder, E. B. et al. 2001 Biol Psychiatry 50 856-872).
Fertilization promoting peptide (FPP), a tripeptide related to thyrotrophin releasing hormone (TRH), is found in seminal plasma. Recent evidence obtained in vitro and in vivo showed that FPP plays an important role in regulating sperm fertility. Specifically, FPP initially stimulates nonfertilizing (uncapacitated) spermatozoa to "switch on" and become fertile more quickly, but then arrests capacitation so that spermatozoa do not undergo spontaneous acrosome loss and therefore do not lose fertilizing potential. These responses are mimicked, and indeed augmented, by adenosine, known to regulate the adenylyl cyclase (AC)/cAMP signal transduction pathway. Both FPP and adenosine have been shown to stimulate cAMP production in uncapacitated cells but inhibit it in capacitated cells, with FPP receptors somehow interacting with adenosine receptors and G proteins to achieve regulation of AC. These events affect the tyrosine phosphorylation state of various proteins, some being important in the initial "switching on" others possibly being involved in the acrosome reaction itself. Calcitonin and angiotensin II, also found in seminal plasma, have similar effects in vitro on uncapacitated spermatozoa and can augment responses to FPP. These molecules have similar effects in vivo, affecting fertility by stimulating and then maintaining fertilizing potential. Either reductions in the availability of FPP, adenosine, calcitonin, and angiotensin Il or defects in their receptors contribute to male infertility (Fraser, L. R. and Adeoya-Osiguwa, S. A. 2001 Vitam Horm 63, 1- 28).
CCL2 (MCP-1 ), CCL7, CCL8, CCL16, CCL18 and fractalkine play an important role in pathophysiological conditions, such as suppression of proliferation of myeloid progenitor cells, neoplasia, inflammatory host responses, cancer, psoriasis, rheumatoid arthritis, atherosclerosis, vasculitis, humoral and cell-mediated immunity responses, leukocyte adhesion and migration processes at the endothelium, inflammatory bowel disease, restenosis, pulmonary fibrosis, pulmonary hypertention, liver fibrosis, liver cirrhosis, nephrosclerosis, ventricular remodeling, heart failure, arteriopathy after organ transplantations and failure of vein grafts.
A number of studies have underlined in particular the crucial role of MCP-1 for the development of atherosclerosis (Gu, L., et al., (1998) Mol.Cell 2, 275-281 ; Gosling, J., et al., (1999) J CHn. Invest 103, 773-778); rheumatoid arthritis (Gong, J. H., et al., (1997) J Exp. Med 186, 131- 137; Ogata, H., et al., (1997) J Pathol. 182, 106-114); pancreatitis (Bhatia, M., et al., (2005) AmJ Physiol Gastrointest. Liver Physiol 288, G1259-G1265); Alzheimer's disease (Yamamoto, M., et al., (2005) AmJ Pathol. 166, 1475-1485); lung fibrosis (Inoshima, I., et al., (2004) AmJ Physiol Lung Cell Mol.Physiol 286, L1038-L1044); renal fibrosis (Wada, T., et al., (2004) J Am.Soc.Nephrol. 15, 940-948), and graft rejection (Saiura, A., et al., (2004) Arterioscler. Thromb. Vase. Biol. 24, 1886-1890). Furthermore, MCP-1 might also play a role in gestosis (Katabuchi, H., et al., (2003) Med Electron Microsc. 36, 253-262), as a paracrine factor in tumor development (Ohta, M., et al., (2003) InU Oncol. 22, 773-778; Li, S., et al., (2005) J Exp.Med 202, 617-624), neuropathic pain (White, F. A., et al., (2005) Proc. Natl. Acad.Sci.U.S.A) and AIDS (Park, I. W., Wang, J. F., and Groopman, J. E. (2001 ) Blood 97, 352-358; Coll, B., et al., (2006) Cytokine 34, 51-55).
MCP-1 levels are increased in CSF of AD patients and patients showing mild cognitive impairment (MCI) (Galimberti, D., et al., (2006) Arch. Neurol. 63, 538-543). Furthermore, MCP-1 shows an increased level in serum of patients with MCI and early AD (Clerici, F., et al., (2006) Neurobiol. Aging 27, 1763-1768).
Several cytotoxic T lymphocyte peptide-based vaccines against hepatitis B, human immunodeficiency virus and melanoma were recently studied in clinical trials. One interesting melanoma vaccine candidate alone or in combination with other tumor antigens, is the decapeptide ELA. This peptide is a Melan-A/MART-1 antigen immunodominant peptide analog, with an N-terminal glutamic acid. It has been reported that the amino group and gamma- carboxylic group of glutamic acids, as well as the amino group and gamma-carboxamide group of glutamines, condense easily to form pyroglutamic derivatives. To overcome this stability problem, several peptides of pharmaceutical interest have been developed with a pyroglutamic acid instead of N-terminal glutamine or glutamic acid, without loss of pharmacological properties. Unfortunately compared with ELA, the pyroglutamic acid derivative (PyrELA) and also the N-terminal acetyl-capped derivative (AcELA) failed to elicit cytotoxic T lymphocyte (CTL) activity. Despite the apparent minor modifications introduced in PyrELA and AcELA, these two derivatives probably have lower affinity than ELA for the specific class I major histocompatibility complex. Consequently, in order to conserve full activity of ELA, the formation of PyrELA must be avoided (Beck A. et al. 2001 , J Pept Res 57(6):528-38.).
Orexin A is a neuropeptide that plays a significant role in the regulation of food intake and sleep-wakefulness, possibly by coordinating the complex behavioral and physiologic responses of these complementary homeostatic functions. It plays also a role in the homeostatic regulation of energy metabolism, autonomic function, hormonal balance and the regulation of body fluids.
Recently, increased levels of the pentapeptide QYNAD were identified in the cerebrospinal fluid (CSF) of patients suffering from multiple sclerosis or Guillain-Barre syndrome compared to healthy individuals (Brinkmeier H. et al. 2000, Nature Medicine 6, 808-811 ). There is a big controversy in the literature about the mechanism of action of the pentapeptide Gln-Tyr-Asn- Ala-Asp (QYNAD), especially its efficacy to interact with and block sodium channels resulting in the promotion of axonal dysfunction, which are involved in inflammatory autoimmune diseases of the central nervous system. But recently, it could be demonstrated that not QYNAD, but its cyclized, pyroglutamated form, pEYNAD, is the active form, which blocks sodium channels resulting in the promotion of axonal dysfunction. Sodium channels are expressed at high density in myelinated axons and play an obligatory role in conducting action potentials along axons within the mammalian brain and spinal cord. Therefore, it is speculated that they are involved in several aspects of the pathophysiology of inflammatory autoimmune diseases, especially multiple sclerosis, the Guillain-Barre syndrome and chronic inflammatory demyelinizing polyradiculoneuropathy. Furthermore, QYNAD is a substrate of the enzyme glutaminyl cyclase (QC, EC 2.3.2.5), which is also present in the brain of mammals, especially in human brain. Glutaminyl cyclase catalyzes effectively the formation of pEYNAD from its precursor QYNAD.
Accordingly, the present invention provides the use of the compounds of formula (I) for the preparation of a medicament for the prevention or alleviation or treatment of a disease selected from the group consisting of mild cognitive impairment, Alzheimer's disease, Familial British Dementia, Familial Danish Dementia, neurodegeneration in Down Syndrome, Huntington's disease, Kennedy's disease, ulcer disease, duodenal cancer with or w/o Helicobacter pylori infections, colorectal cancer, Zolliger-Ellison syndrome, gastric cancer with or without Helicobacter pylori infections, pathogenic psychotic conditions, schizophrenia, infertility, neoplasia, inflammatory host responses, cancer, malign metastasis, melanoma, psoriasis, rheumatoid arthritis, atherosclerosis, pancreatitis, restenosis, impaired humoral and cell- mediated immune responses, leukocyte adhesion and migration processes in the endothelium, impaired food intake, impaired sleep-wakefulness, impaired homeostatic regulation of energy metabolism, impaired autonomic function, impaired hormonal balance or impaired regulation of body fluids, multiple sclerosis, the Guillain-Barre syndrome and chronic inflammatory demyelinizing polyradiculoneuropathy.
Furthermore, by administration of a compound according to the present invention to a mammal it can be possible to stimulate the proliferation of myeloid progenitor cells.
In addition, the administration of a QC inhibitor according to the present invention can lead to suppression of male fertility.
In a preferred embodiment, the present invention provides the use of inhibitors of QC (EC) activity in combination with other agents, especially for the treatment of neuronal diseases, artherosclerosis and multiple sclerosis.
The present invention also provides a method of treatment of the aforementioned diseases comprising the administration of a therapeutically active amount of at least one compound of formula (I) to a mammal, preferably a human.
Most preferably, said method and corresponding uses are for the treatment of a disease selected from the group consisting of mild cognitive impairment, Alzheimer's disease, Familial British Dementia, Familial Danish Dementia, neurodegeneration in Down Syndrome, Parkinson's disease and Chorea Huntington, comprising the administration of a therapeutically active amount of at least one compound of formula (I) to a mammal, preferably a human.
Even preferably, the present invention provides a method of treatment and corresponding uses for the treatment of rheumatoid arthritis, atherosclerosis, pancreatitis and restenosis.
Pharmaceutical combinations
In a preferred embodiment, the present invention provides a composition, preferably a pharmaceutical composition, comprising at least one QC inhibitor optionally in combination with at least one other agent selected from the group consisting of nootropic agents, neuroprotectants, antiparkinsonian drugs, amyloid protein deposition inhibitors, beta amyloid synthesis inhibitors, antidepressants, anxiolytic drugs, antipsychotic drugs and anti-multiple sclerosis drugs.
Most preferably, said QC inhibitor is a compound of formula (I) of the present invention.
More specifically, the aforementioned other agent is selected from the group consisting of beta- amyloid antibodies, cysteine protease inhibitors, PEP-inhibitors, LiCI, acetylcholinesterase (AChE) inhibitors, PIMT enhancers, inhibitors of beta secretases, inhibitors of gamma secretases, inhibitors of aminopeptidases, preferably inhibitors of dipeptidyl peptidases, most preferably DP IV inhibitors; inhibitors of neutral endopeptidase, inhibitors of Phosphodiesterase- 4 (PDE-4), TNFalpha inhibitors, muscarinic M1 receptor antagonists, NMDA receptor antagonists, sigma-1 receptor inhibitors, histamine H3 antagonists, immunomodulatory agents, immunosuppressive agents, MCP-1 antagonists or an agent selected from the group consisting of antegren (natalizumab), Neurelan (fampridine-SR), campath (alemtuzumab), IR 208, NBI 5788/MSP 771 (tiplimotide), paclitaxel, Anergix.MS (AG 284), SH636, Differin (CD 271 , adapalene), BAY 361677 (interleukin-4), matrix-metalloproteinase-inhibitors (e.g. BB 76163), interferon-tau (trophoblastin) and SAIK-MS.
Furthermore, the other agent may be, for example, an anti-anxiety drug or antidepressant selected from the group consisting of
(a) Benzodiazepines, e.g. alprazolam, chlordiazepoxide, clobazam, clonazepam, clorazepate, diazepam, fludiazepam, loflazepate, lorazepam, methaqualone, oxazepam, prazepam, tranxene,
(b) Selective serotonin re-uptake inhibitors (SSRI's), e.g. citalopram, fluoxetine, fluvoxamine, escitalopram, sertraline, paroxetine,
(c) Tricyclic antidepressants, e.g. amitryptiline, clomipramine, desipramine, doxepin, imipramine
(d) Monoamine oxidase (MAO) inhibitors,
(e) Azapirones, e.g. buspirone, tandopsirone, (f) Serotonin-norepinephrine reuptake inhibitors (SNRI's), e.g. venlafaxine, duloxetine,
(g) Mirtazapine,
(h) Norepinephrine reuptake inhibitors (NRI's), e.g. reboxetine,
(i) Bupropione,
(j) Nefazodone, (k) beta-blockers, (I) NPY-receptor ligands: NPY agonists or antagonists.
In a further embodiment, the other agent may be, for example, an anti-multiple sclerosis drug selected from the group consisting of
a) dihydroorotate dehydrogenase inhibitors, e.g. SC-12267, teriflunomide, MNA-715, HMR-
1279 (syn. to HMR-1715, MNA-279), b) autoimmune suppressant, e.g. laquinimod, c) paclitaxel, d) antibodies, e.g. AGT-1 , anti-granulocyte-macrophage colony-stimulating factor (GM- CSF) monoclonal antibody, Nogo receptor modulators, ABT-874, alemtuzumab
(CAMPATH), anti-OX40 antibody, CNTO-1275, DN-1921 , natalizumab (syn. to AN- 100226, Antegren, VLA-4 Mab), daclizumab (syn. to Zenepax, Ro-34-7375, SMART anti-Tac), J-695, priliximab (syn. to Centara, CEN-000029, cM-T412), MRA, Dantes, anti-IL-12-antibody, e) peptide nucleic acid (PNA) preparations, e.g. reticulose, f) interferon alpha, e.g. Alfaferone, human alpha interferon (syn. to Omniferon, Alpha Leukoferon), g) interferon beta, e.g. Frone, interferon beta-1 a like Avonex, Betron (Rebif), interferon beta analogs, interferon beta-transferrin fusion protein, recombinant interferon beta-1 b like Betaseron, h) interferon tau, i) peptides, e.g. AT-008, AnergiX.MS, lmmunokine (alpha-lmmunokine-NNSO3), cyclic peptides like ZD-7349, j) therapeutic enzymes, e.g. soluble CD8 (sCD8), k) multiple sclerosis-specific autoantigen-encoding plasmid and cytokine-encoding plasmid, e.g. BHT-3009;
I) inhibitor of TNF-alpha, e.g. BLX-1002, thalidomide, SH-636, m) TNF antagonists, e.g. solimastat, lenercept (syn. to RO-45-2081 , Tenefuse), onercept
(STNFR1 ), CC-1069, n) TNF alpha, e.g. etanercept (syn. to Enbrel, TNR-001 ) o) CD28 antagonists, e.g. abatacept, p) Lck tyrosine kinase inhibitors, q) cathepsin K inhibitors, r) analogs of the neuron-targeting membrane transporter protein taurine and the plant- derived calpain inhibitor leupeptin, e.g. Neurodur, s) chemokine receptor-1 (CCR1 ) antagonist, e.g. BX-471 , t) CCR2 antagonists, u) AMPA receptor antagonists, e.g. ER- 167288-01 and ER-099487, E-2007, talampanel, v) potassium channel blockers, e.g. fampridine, w) tosyl-proline-phenylalanine small-molecule antagonists of the VLA-4A/CAM interaction, e.g. TBC-3342, x) cell adhesion molecule inhibitors, e.g. TBC-772, y) antisense oligonucleotides, e.g. EN-101 , z) antagonists of free immunoglobulin light chain (IgLC) binding to mast cell receptors, e.g. F-991 , aa) apoptosis inducing antigens, e.g. Apogen MS, bb) alpha-2 adrenoceptor agonist, e.g. tizanidine (syn. to Zanaflex, Ternelin, Sirdalvo, Sirdalud, Mionidine), cc) copolymer of L-tyrosine, L-lysine, L-glutamic acid and L-alanine, e.g. glatiramer acetate (syn. to Copaxone, COP-1 , copolymer-1 ), dd) topoisomerase Il modulators, e.g. mitoxantrone hydrochloride, ee) adenosine deaminase inhibitor, e.g. cladribine (syn. to Leustatin, Mylinax, RWJ-26251 ), ff) interleukin-10, e.g. ilodecakin (syn. to Tenovil, Sch-52000, CSIF), gg) interleukin-12 antagonists, e.g. lisofylline (syn. to CT-1501 R, LSF, lysofylline), hh) Ethanaminum, e.g. SRI-62-834 (syn. to CRC-8605, NSC-614383), ii) immunomodulators, e.g. SAIK-MS, PNU-156804, alpha-fetoprotein peptide (AFP), IPDS, jj) retinoid receptor agonists, e.g. adapalene (syn. to Differin, CD-271 ), kk) TGF-beta, e.g. GDF-1 (growth and differentiation factor 1 ),
II) TGF-beta-2, e.g. BetaKine, mm) MMP inhibitors, e.g. glycomed, nn) phosphodiesterase 4 (PDE4) inhibitors, e.g. RPR-122818, oo) purine nucleoside phosphorylase inhibitors, e.g. 9-(3-pyridylmethyl)-9-deazaguanine, peldesine (syn. to BCX-34, TO-200), pp) alpha-4/beta-1 integrin antagonists, e.g. ISIS-104278, qq) antisense alpha4 integrin (CD49d), e.g. ISIS-17044, ISIS-27104, rr) cytokine-inducing agents, e.g. nucleosides, ICN-17261 , ss) cytokine inhibitors, tt) heat shock protein vaccines, e.g. HSPPC-96, uu) neuregulin growth factors, e.g. GGF-2 (syn. to neuregulin, glial growth factor 2), vv) cathepsin S - inhibitors, ww) bropirimine analogs, e.g. PNU-56169, PNU-63693, xx) Monocyte chemoattractant protein-1 inhibitors, e.g. benzimidazoles like MCP-1 inhibitors, LKS-1456, PD-064036, PD-064126, PD-084486, PD-172084, PD-172386.
Further, the present invention provides pharmaceutical compositions e.g. for parenteral, enteral or oral administration, comprising at least one QC inhibitor, optionally in combination with at least one of the other aforementioned agents.
These combinations provide a particularly beneficial effect. Such combinations are therefore shown to be effective and useful for the treatment of the aforementioned diseases. Accordingly, the invention provides a method for the treatment of these conditions.
The method comprises either co-administration of at least one QC inhibitor and at least one of the other agents or the sequential administration thereof.
Co-administration includes administration of a formulation, which comprises at least one QC inhibitor and at least one of the other agents or the essentially simultaneous administration of separate formulations of each agent.
Beta-amyloid antibodies and compositions containing the same are described, e.g. in WO 2006/137354, WO 2006/1 18959, WO 2006/1031 16, WO 2006/095041 , WO 2006/081 171 , WO 2006/066233, WO 2006/066171 , WO 2006/066089, WO 2006/066049, WO 2006/055178, WO 2006/046644, WO 2006/039470, WO 2006/036291 , WO 2006/026408, WO 2006/016644, WO 2006/014638, WO 2006/014478, WO 2006/008661 , WO 2005/123775, WO 2005/120571 , WO 2005/105998, WO 2005/081872, WO 2005/080435, WO 2005/02851 1 , WO 2005/025616, WO 2005/025516, WO 2005/023858, WO 2005/018424, WO 2005/011599, WO 2005/000193, WO 2004/108895, WO 2004/098631 , WO 2004/080419, WO 2004/071408, WO 2004/069182, WO 2004/067561 , WO 2004/044204, WO 2004/032868, WO 2004/031400, WO 2004/029630, WO 2004/029629, WO 2004/024770, WO 2004/024090, WO 2003/104437, WO 2003/089460, WO 2003/086310, WO 2003/077858, WO 2003/074081 , WO 2003/070760, WO 2003/063760, WO 2003/055514, WO 2003/051374, WO 2003/048204, WO 2003/045128, WO 2003/040183, WO 2003/039467, WO 2003/016466, WO 2003/015691 , WO 2003/014162, WO 2003/012141 , WO 2002/088307, WO 2002/088306, WO 2002/074240, WO 2002/046237, WO 2002/046222, WO 2002/041842, WO 2001/062801 , WO 2001/012598, WO 2000/077178, WO 2000/072880, WO 2000/063250, WO 1999/060024, WO 1999/027944, WO 1998/044955, WO 1996/025435, WO 1994/017197, WO 1990/014840, WO 1990/012871 , WO 1990/012870, WO 1989/006242.
The beta-amyloid antibodies may be selected from, for example, polyclonal, monoclonal, chimenic or humanized antibodies. Furthermore, said antibodies may be useful to develop active and passive immune therapies, i.e. vaccines and monoclonal antibodies. Suitable examples of beta-amyloid antibodies are ACU-5A5, huC091 (Acumen/Merck); PF- 4360365, RI-1014, RI-1219, RI-409, RN-1219 (Rinat Neuroscience Corp (Pfizer Inc)); the nanobody therapeutics of Ablynx/Boehringer Ingelheim; beta-amyloid-specific humanized monoclonal antibodies of Intellect Neurosciences/IBL; m266, m266.2 (EIi Lilly & Co.); AAB-02 (Elan); bapineuzumab (Elan); BAN-2401 (Bioarctic Neuroscience AB); ABP-102 (Abiogen Pharma SpA); BA-27, BC-05 (Takeda); R-1450 (Roche); ESBA-212 (ESBATech AG); AZD- 3102 (AstraZeneca) and beta-amyloid antibodies of Mindset BioPharmaceuticals Inc. Especially preferred are antibodies, which recognize the N-terminus of the Aβ peptide. A suitable antibody, which recognizes the Aβ-N-Terminus is, for example Acl-24 (AC Immune SA). A monoclonal antibody against beta-amyloid peptide is disclosed in WO 2007/068412. Respective chimeric and humanized antibodies are disclosed in WO 2008/011348. A method for producing a vaccine composition for treating an amyloid-associated disease is disclosed in WO 2007/068411.
Suitable cysteine protease inhibitors are inhibitors of cathepsin B. Inhibitors of cathepsin B and compositions containing such inhibitors are described, e.g. in WO 2006/060473, WO 2006/042103, WO 2006/039807, WO 2006/021413, WO 2006/021409, WO 2005/097103, WO 2005/007199, WO2004/084830, WO 2004/078908, WO 2004/026851 , WO 2002/094881 , WO 2002/027418, WO 2002/021509, WO 1998/046559, WO 1996/021655.
Examples of suitable PIMT enhancers are 10-aminoaliphatyl-dibenz[b, f] oxepines described in WO 98/15647 and WO 03/057204, respectively. Further useful according to the present invention are modulators of PIMT activity described in WO 2004/039773.
Inhibitors of beta secretase and compositions containing such inhibitors are described, e.g. in
WO03/059346, WO2006/099352, WO2006/078576, WO2006/060109, WO2006/057983,
WO2006/057945, WO2006/055434, WO2006/044497, WO2006/034296, WO2006/034277, WO2006/029850, WO2006/026204, WO2006/014944, WO2006/014762, WO2006/002004, US 7,109,217, WO2005/1 13484, WO2005/103043, WO2005/103020, WO2005/065195, WO2005/051914, WO2005/044830, WO2005/032471 , WO2005/018545, WO2005/004803, WO2005/004802, WO2004/062625, WO2004/043916, WO2004/013098, WO03/099202, WO03/043987, WO03/039454, US 6,562,783, WO02/098849 and WO02/096897.
Suitable examples of beta secretase inhibitors for the purpose of the present invention are WY- 25105 (Wyeth); Posiphen, (+)-phenserine (TorreyPines / NIH); LSN-2434074, LY-2070275, LY- 2070273, LY-2070102 (EIi Lilly & Co.); PNU-159775A, PNU-178025A, PNU-17820A, PNU- 33312, PNU-38773, PNU-90530 (Elan / Pfizer); KMI-370, KMI-358, kmi-008 (Kyoto University); OM-99-2, OM-003 (Athenagen Inc.); AZ-12304146 (AstraZeneca / Astex); GW-840736X (GlaxoSmithKline pic), DNP-004089 (De Novo Pharmaceuticals Ltd.) and CT-21166 (CoMentis Inc.).
Inhibitors of gamma secretase and compositions containing such inhibitors are described, e.g. in WO2005/008250, WO2006/004880, US 7,122,675, US 7,030,239, US 6,992,081 , US 6,982,264, WO2005/097768, WO2005/028440, WO2004/101562, US 6,756,51 1 , US 6,683,091 , WO03/066592, WO03/014075, WO03/013527, WO02/36555, WO01/53255, US 7,109,217, US 7,101 ,895, US 7,049,296, US 7,034,182, US 6,984,626, WO2005/040126, WO2005/030731 , WO2005/014553, US 6,890,956, EP 1334085, EP 1263774, WO2004/101538, WO2004/00958, WO2004/08991 1 , WO2004/073630, WO2004/069826, WO2004/039370, WO2004/031 139, WO2004/031137, US 6,713,276, US 6,686,449, WO03/091278, US 6,649,196, US 6,448,229, WO01/77144 and WO01/66564.
Suitable gamma secretase inhibitors for the purpose of the present invention are GSI-953, WAY-GSI-A, WAY-GSI-B (Wyeth); MK-0752, MRK-560, L-852505, L-685-458, L-852631 , L- 852646 (Merck & Co. Inc.); LY-450139, LY-41 1575, AN-37124 (EIi Lilly & Co.); BMS-299897, BMS-433796 (Bristol-Myers Squibb Co.); E-2012 (Eisai Co. Ltd.); EHT-0206, EHT-206 (ExonHit Therapeutics SA); and NGX-555 (TorreyPines Therapeutics Inc.).
DP IV-inhibitors and compositions containing such inhibitors are described, e.g. in US6,011 ,155; US6,107,317; US6, 1 10,949; US6, 124,305; US6, 172,081 ; WO99/61431 , WO99/67278, WO99/67279, DE19834591 , WO97/40832, WO95/15309, WO98/19998, WO00/07617, WO99/38501 , WO99/46272, WO99/38501 , WO01/68603, WO01/40180, WO01/81337, WO01/81304, WO01/55105, WO02/02560, WO01/34594, WO02/38541 , WO02/083128, WO03/072556, WO03/002593, WO03/000250, WO03/000180, WO03/000181 , EP1258476, WO03/002553, WO03/002531 , WO03/002530, WO03/004496, WO03/004498, WO03/024942, WO03/024965, WO03/033524, WO03/035057, WO03/035067, WO03/037327, WO03/040174, WO03/045977, WO03/055881 , WO03/057144, WO03/057666, WO03/068748, WO03/068757, WO03/082817, WO03/101449, WO03/101958, WO03/104229, WO03/74500, WO2004/007446, WO2004/007468, WO2004/018467, WO2004/018468, WO2004/018469, WO2004/026822, WO2004/032836, WO2004/033455, WO2004/037169, WO2004/041795, WO2004/043940, WO2004/048352, WO2004/050022, WO2004/052850, WO2004/058266, WO2004/064778, WO2004/069162, WO2004/071454, WO2004/076433, WO2004/076434, WO2004/087053, WO2004/089362, WO2004/099185, WO2004/103276, WO2004/103993, WO2004/108730, WO2004/110436, WO2004/1 11041 , WO2004/1 12701 , WO2005/000846, WO2005/000848, WO2005/011581 , WO2005/01691 1 , WO2005/023762, WO2005/025554, WO2005/026148, WO2005/030751 , WO2005/033106, WO2005/037828, WO2005/040095, WO2005/044195, WO2005/047297, WO2005/051950, WO2005/056003, WO2005/056013, WO2005/058849, WO2005/075426, WO2005/082348, WO2005/085246, WO2005/087235, WO2005/095339, WO2005/095343, WO2005/095381 , WO2005/108382, WO2005/113510, WO2005/116014, WO2005/116029, WO2005/1 18555, WO2005/120494, WO2005/121089, WO2005/121131 , WO2005/123685, WO2006/995613; WO2006/009886; WO2006/013104; WO2006/017292; WO2006/019965; WO2006/020017; WO2006/023750; WO2006/039325; WO2006/041976; WO2006/047248; WO2006/058064; WO2006/058628; WO2006/066747; WO2006/066770 and WO2006/068978.
Suitable DP IV-inhibitors for the purpose of the present invention are for example Sitagliptin, des-fluoro-sitagliptin (Merck & Co. Inc.); vildagliptin, DPP-728, SDZ-272-070 (Novartis) ; ABT- 279, ABT-341 (Abbott Laboratories); denagliptin, TA-6666 (GlaxoSmithKline pic); SYR-322 (Takeda San Diego Inc.); talabostat (Point Therapeutics Inc.); Ro-0730699, R-1499, R-1438 (Roche Holding AG); FE-99901 1 (Ferring Pharmaceuticals); TS-021 (Taisho Pharmaceutical Co. Ltd.); GRC-8200 (Glenmark Pharmaceuticals Ltd.); ALS-2-0426 (Alantos Pharmaceuticals Holding Inc.); ARI-2243 (Arisaph Pharmaceuticals Inc.); SSR-162369 (Sanofi-Synthelabo); MP- 513 (Mitsubishi Pharma Corp.); DP-893, CP-867534-01 (Pfizer Inc.); TSL-225, TMC-2A (Tanabe Seiyaku Co. Ltd.); PHX-1149 (Phenomenix Corp.); saxagliptin (Bristol-Myers Squibb Co.); PSN-9301 ((OSI) Prosidion), S-40755 (Servier); KRP-104 (ActivX Biosciences Inc.); sulphostin (Zaidan Hojin); KR-62436 (Korea Research Institute of Chemical Technology); P32/98 (Probiodrug AG); Bl-A, Bl-B (Boehringer lngelheim Corp.); SK-0403 (Sanwa Kagaku Kenkyusho Co. Ltd.); and NNC-72-2138 (Novo Nordisk A/S). Other preferred DP IV-inhibitors are
(i) dipeptide-like compounds, disclosed in WO 99/61431 , e.g. N-valyl prolyl, O-benzoyl hydroxylamine, alanyl pyrrolidine, isoleucyl thiazolidine like L-allo-isoleucyl thiazolidine, L-threo- isoleucyl pyrrolidine and salts thereof, especially the fumaric salts, and L-allo-isoleucyl pyrrolidine and salts thereof;
(ii) peptide structures, disclosed in WO 03/002593, e.g. tripeptides; (iii) peptidylketones, disclosed in WO 03/033524; (vi) substituted aminoketones, disclosed in WO 03/040174; (v) topically active DP IV-inhibitors, disclosed in WO 01/14318; (vi) prodrugs of DP IV-inhibitors, disclosed in WO 99/67278 and WO 99/67279; and
(v) glutaminyl based DP IV-inhibitors, disclosed in WO 03/072556 and WO 2004/099134.
Suitable beta amyloid synthesis inhibitors for the purpose of the present invention are for example Bisnorcymserine (Axonyx Inc.); (R)-flurbiprofen (MCP-7869; Flurizan) (Myriad Genetics); nitroflurbiprofen (NicOx); BGC-20-0406 (Sankyo Co. Ltd.) and BGC-20-0466 (BTG pic).
Suitable amyloid protein deposition inhibitors for the purpose of the present invention are for example SP-233 (Samaritan Pharmaceuticals); AZD-103 (Ellipsis Neurotherapeutics Inc.); AAB- 001 (Bapineuzumab), AAB-002, ACC-001 (Elan Corp pic); Colostrinin (ReGen Therapeutics pic); Tramiprosate (Neurochem); AdPEDI-(amyloid-beta1-6)11 ) (Vaxin Inc.); MPI-127585, MPI-
423948 (Mayo Foundation); SP-08 (Georgetown University); ACU-5A5 (Acumen / Merck);
Transthyretin (State University of New York); PTI-777, DP-74, DP 68, Exebryl (ProteoTech Inc.); m266 (EIi Lilly & Co.); EGb-761 (Dr. Willmar Schwabe GmbH); SPI-014 (Satori Pharmaceuticals Inc.); ALS-633, ALS-499 (Advanced Life Sciences Inc.); AGT-160 (ArmaGen Technologies
Inc.); TAK-070 (Takeda Pharmaceutical Co. Ltd.); CHF-5022, CHF-5074, CHF-5096 and CHF-
5105 (Chiesi Farmaceutici SpA.).
Suitable PDE-4 inhibitors for the purpose of the present invention are for example Doxofylline (Instituto Biologico Chemioterapica ABC SpA.); idudilast eye drops, tipelukast, ibudilast (Kyorin
Pharmaceutical Co. Ltd.); theophylline (Elan Corp.); cilomilast (GlaxoSmithKline pic); Atopik
(Barrier Therapeutics Inc.); tofimilast, CI-1044, PD-189659, CP-220629, PDE 4d inhibitor BHN
(Pfizer Inc.); arofylline, LAS-37779 (Almirall Prodesfarma SA.); roflumilast, hydroxypumafentrine
(Altana AG), tetomilast (Otska Pharmaceutical Co. Ltd.); tipelukast, ibudilast (Kyorin Pharmaceutical), CC-10004 (Celgene Corp.); HT-0712, IPL-4088 (Inflazyme Pharmaceuticals Ltd.); MEM-1414, MEM-1917 (Memory Pharmaceuticals Corp.); oglemilast, GRC-4039 (Glenmark Pharmaceuticals Ltd.); AWD-12-281 , ELB-353, ELB-526 (Elbion AG); EHT-0202 (ExonHit Therapeutics SA.); ND-1251 (Neuro3d SA.); 4AZA-PDE4 (4 AZA Bioscience NV.); AVE-8112 (Sanofi-Aventis); CR-3465 (Rottapharm SpA.); GP-0203, NCS-613 (Centre National de Ia Recherche Scientifique); KF-19514 (Kyowa Hakko Kogyo Co. Ltd.); ONO-6126 (Ono Pharmaceutical Co. Ltd.); OS-0217 (Dainippon Pharmaceutical Co. Ltd.); IBFB-130011 , IBFB- 150007, IBFB-130020, IBFB-140301 (IBFB Pharma GmbH); IC-485 (ICOS Corp.); RBx-14016 and RBx-1 1082 (Ranbaxy Laboratories Ltd.). A preferred PDE-4-inhibitor is Rolipram.
MAO inhibitors and compositions containing such inhibitors are described, e.g. in WO2006/091988, WO2005/007614, WO2004/089351 , WO01/26656, WO01/12176, WO99/57120, WO99/571 19, WO99/13878, WO98/40102, WO98/01157, WO96/20946, WO94/07890 and WO92/21333.
Suitable MAO-inhibitors for the purpose of the present invention are for example Linezolid (Pharmacia Corp.); RWJ-416457 (RW Johnson Pharmaceutical Research Institute); budipine (Altana AG); GPX-325 (BioResearch Ireland); isocarboxazid; phenelzine; tranylcypromine; indantadol (Chiesi Farmaceutici SpA.); moclobemide (Roche Holding AG); SL-25.1 131 (Sanofi- Synthelabo); CX-1370 (Burroughs Wellcome Co.); CX-157 (Krenitsky Pharmaceuticals Inc.); desoxypeganine (HF Arzneimittelforschung GmbH & Co. KG); bifemelane (Mitsubishi-Tokyo Pharmaceuticals Inc.); RS-1636 (Sankyo Co. Ltd.); esuprone (BASF AG); rasagiline (Teva Pharmaceutical Industries Ltd.); ladostigil (Hebrew University of Jerusalem); safinamide (Pfizer) and NW-1048 (Newron Pharmaceuticals SpA.).
Suitable histamine H3 antagonists for the purpose of the present invention are, e.g. ABT-239, ABT-834 (Abbott Laboratories); 3874-H1 (Aventis Pharma); UCL-2173 (Berlin Free University), UCL-1470 (BioProjet, Societe Civile de Recherche); DWP-302 (Daewoong Pharmaceutical Co Ltd); GSK-189254A, GSK-207040A (GlaxoSmithKline Inc.); cipralisant, GT-2203 (Gliatech Inc.); Ciproxifan (INSERM), /S,2S-2-(2-Aminoethyl)-1-(1 H-imidazol-4-yl)cyclopropane (Hokkaido University); JNJ-17216498, JNJ-5207852 (Johnson & Johnson); NNC-0038-0000-1049 (Novo Nordisk A/S); and Sch-79687 (Schering-Plough).
PEP inhibitors and compositions containing such inhibitors are described, e.g. in JP 01042465,
JP 03031298, JP 04208299, WO 00/71 144, US 5,847,155; JP 09040693, JP 10077300, JP 05331072, JP 05015314, WO 95/15310, WO 93/00361 , EP 0556482, JP 06234693, JP 01068396, EP 0709373, US 5,965,556, US 5,756,763, US 6,121 ,311 , JP 63264454, JP 64000069, JP 63162672, EP 0268190, EP 0277588, EP 0275482, US 4,977,180, US 5,091 ,406, US 4,983,624, US 5,112,847, US 5,100,904, US 5,254,550, US 5,262,431 , US 5,340,832, US 4,956,380, EP 0303434, JP 03056486, JP 01143897, JP 1226880, EP 0280956, US 4,857,537, EP 0461677, EP 0345428, JP 02275858, US 5,506,256, JP 06192298, EP 0618193, JP 03255080, EP 0468469, US 5,1 18,81 1 , JP 05025125, WO 9313065, JP 05201970, WO 9412474, EP 0670309, EP 0451547, JP 06339390, US 5,073,549, US 4,999,349, EP 0268281 , US 4,743,616, EP 0232849, EP 0224272, JP 621 14978, JP 62114957, US 4,757,083, US 4,810,721 , US 5,198,458, US 4,826,870, EP 0201742, EP 0201741 , US 4,873,342, EP 0172458, JP 61037764, EP 0201743, US 4,772,587, EP 0372484, US 5,028,604, WO 91/18877, JP 04009367, JP 04235162, US 5,407,950, WO 95/01352, JP 01250370, JP 02207070, US 5,221 ,752, EP 0468339, JP 04211648, WO 99/46272, WO 2006/058720 and PCT/EP2006/061428.
Suitable prolyl endopeptidase inhibitors for the purpose of the present invention are, e.g. Fmoc- Ala-Pyrr-CN, Z-Phe-Pro-Benzothiazole (Probiodrug), Z-321 (Zeria Pharmaceutical Co Ltd.); ONO-1603 (Ono Pharmaceutical Co Ltd); JTP-4819 (Japan Tobacco Inc.) and S-17092 (Servier).
Other suitable compounds that can be used according to the present invention in combination with QC-inhibitors are NPY, an NPY mimetic or an NPY agonist or antagonist or a ligand of the NPY receptors.
Preferred according to the present invention are antagonists of the NPY receptors.
Suitable ligands or antagonists of the NPY receptors are 3a, 4,5,9b-tetrahydro-1 h-benz[e]indol- 2-yl amine-derived compounds as disclosed in WO 00/68197.
NPY receptor antagonists which may be mentioned include those disclosed in European patent applications EP 0 614 91 1 , EP 0 747 357, EP 0 747 356 and EP 0 747 378; international patent applications WO 94/17035, WO 97/19911 , WO 97/19913, WO 96/12489, WO 97/19914, WO
96/22305, WO 96/40660, WO 96/12490, WO 97/09308, WO 97/20820, WO 97/20821 , WO
97/20822, WO 97/20823, WO 97/19682, WO 97/25041 , WO 97/34843, WO 97/46250, WO
98/03492, WO 98/03493, WO 98/03494 and WO 98/07420; WO 00/30674, US patents Nos. 5,552,41 1 , 5,663,192 and 5,567,714; 6,1 14,336, Japanese patent application JP 09157253; international patent applications WO 94/00486, WO 93/12139, WO 95/00161 and WO 99/15498; US Patent No. 5,328,899; German patent application DE 393 97 97; European patent applications EP 355 794 and EP 355 793; and Japanese patent applications JP 06116284 and JP 07267988. Preferred NPY antagonists include those compounds that are specifically disclosed in these patent documents. More preferred compounds include amino acid and non- peptide-based NPY antagonists. Amino acid and non-peptide-based NPY antagonists which may be mentioned include those disclosed in European patent applications EP 0 614 91 1 , EP 0 747 357, EP 0 747 356 and EP 0 747 378; international patent applications WO 94/17035, WO 97/1991 1 , WO 97/19913, WO 96/12489, WO 97/19914, WO 96/22305, WO 96/40660, WO 96/12490, WO 97/09308, WO 97/20820, WO 97/20821 , WO 97/20822, WO 97/20823, WO 97/19682, WO 97/25041 , WO 97/34843, WO 97/46250, WO 98/03492, WO 98/03493, WO 98/03494, WO 98/07420 and WO 99/15498 ; US patents Nos. 5,552,41 1 , 5,663,192 and 5,567,714; and Japanese patent application JP 09157253. Preferred amino acid and non- peptide-based NPY antagonists include those compounds that are specifically disclosed in these patent documents.
Particularly preferred compounds include amino acid-based NPY antagonists. Amino acid- based compounds, which may be mentioned include those disclosed in international patent applications WO 94/17035, WO 97/1991 1 , WO 97/19913, WO 97/19914 or, preferably, WO 99/15498. Preferred amino acid-based NPY antagonists include those that are specifically disclosed in these patent documents, for example BIBP3226 and, especially, (R)-N2- (diphenylacetyl)-(R)-N-[1-(4-hydroxy- phenyl) ethyl] arginine amide (Example 4 of international patent application WO 99/15498).
M1 receptor agonists and compositions containing such inhibitors are described, e.g. in WO2004/087158, WO91/10664.
Suitable M1 receptor antagonists for the purpose of the present invention are for example CDD- 0102 (Cognitive Pharmaceuticals); Cevimeline (Evoxac) (Snow Brand Milk Products Co. Ltd.); NGX-267 (TorreyPines Therapeutics); sabcomeline (GlaxoSmithKline); alvameline (H Lundbeck A/S); LY-593093 (EIi Lilly & Co.); VRTX-3 (Vertex Pharmaceuticals Inc.); WAY-132983 (Wyeth) and CI-101 7/ (PD-151832) (Pfizer Inc.).
Acetylcholinesterase inhibitors and compositions containing such inhibitors are described, e.g. in WO2006/071274, WO2006/070394, WO2006/040688, WO2005/092009, WO2005/079789, WO2005/039580, WO2005/027975, WO2004/084884, WO2004/037234, WO2004/032929, WO03/101458, WO03/091220, WO03/082820, WO03/020289, WO02/32412, WO01/85145, WO01/78728, WO01/66096, WO00/02549, WO01/00215, WO00/15205, WO00/23057, WO00/33840, WO00/30446, WO00/23057, WO00/15205, WO00/09483, WO00/07600, WO00/02549, WO99/47131 , WO99/07359, WO98/30243, WO97/38993, WO97/13754, WO94/29255, WO94/20476, WO94/19356, WO93/03034 and WO92/19238.
Suitable acetylcholinesterase inhibitors for the purpose of the present invention are for example Donepezil (Eisai Co. Ltd.); rivastigmine (Novartis AG); (-)-phenserine (TorreyPines Therapeutics); ladostigil (Hebrew University of Jerusalem); huperzine A (Mayo Foundation); galantamine (Johnson & Johnson); Memoquin (Universita di Bologna); SP-004 (Samaritan Pharmaceuticals Inc.); BGC-20-1259 (Sankyo Co. Ltd.); physostigmine (Forest Laboratories Inc.); NP-0361 (Neuropharma SA); ZT-1 (Debiopharm); tacrine (Warner-Lambert Co.); metrifonate (Bayer Corp.) and INM-176 (Whanln).
NMDA receptor antagonists and compositions containing such inhibitors are described, e.g. in WO2006/094674, WO2006/058236, WO2006/058059, WO2006/010965, WO2005/000216, WO2005/102390, WO2005/079779, WO2005/079756, WO2005/072705, WO2005/070429, WO2005/055996, WO2005/035522, WO2005/009421 , WO2005/000216, WO2004/092189, WO2004/039371 , WO2004/028522, WO2004/009062, WO03/010159, WO02/072542, WO02/34718, WO01/98262, WO01/94321 , WO01/92204, WO01/81295, WO01/32640, WO01/10833, WO01/10831 , WO00/5671 1 , WO00/29023, WO00/00197, WO99/53922, WO99/48891 , WO99/45963, WO99/01416, WO99/07413, WO99/01416, WO98/50075, WO98/50044, WO98/10757, WO98/05337, WO97/32873, WO97/23216, WO97/23215, WO97/23214, WO96/14318, WO96/08485, WO95/31986, WO95/26352, WO95/26350, WO95/26349, WO95/26342, WO95/12594, WO95/02602, WO95/02601 , WO94/20109, WO94/13641 , WO94/09016 and WO93/25534.
Suitable NMDA receptor antagonists for the purpose of the present invention are for example Memantine (Merz & Co. GmbH); topiramate (Johnson & Johnson); AVP-923 (Neurodex) (Center for Neurologic Study); EN-3231 (Endo Pharmaceuticals Holdings Inc.); neramexane (MRZ-
2/579) (Merz and Forest); CNS-5161 (CeNeS Pharmaceuticals Inc.); dexanabinol (HU-21 1 ;
Sinnabidol; PA-5021 1 ) (Pharmos); EpiCept NP-1 (Dalhousie University); indantadol (V-3381 ;
CNP-3381 ) (Vernalis); perzinfotel (EAA-090, WAY-126090, EAA-129) (Wyeth); RGH-896 (Gedeon Richter Ltd.); traxoprodil (CP-101606), besonprodil (PD-196860, CI-1041 ) (Pfizer Inc.); CGX-1007 (Cognetix Inc.); delucemine (NPS-1506) (NPS Pharmaceuticals Inc.); EVT-101 (Roche Holding AG); acamprosate (Synchroneuron LLC); CR-3991 , CR-2249, CR-3394 (Rottapharm SpA.); AV-101 (4-CI-kynurenine (4-CI-KYN)), 7-chloro-kynurenic acid (7-CI-KYNA) (VistaGen); NPS-1407 (NPS Pharmaceuticals Inc.); YT-1006 (Yaupon Therapeutics Inc.); ED- 1812 (Sosei R&D Ltd.); himantane (hydrochloride N-2-(adamantly)-hexamethylen-imine) (RAMS); Lancicemine (AR-R-15896) (AstraZeneca); EVT-102, Ro-25-6981 and Ro-63-1908 (Hoffmann-La Roche AG / Evotec).
Furthermore, the present invention relates to combination therapies useful for the treatment of atherosclerosis, restenosis or arthritis, administering a QC inhibitor in combination with another therapeutic agent selected from the group consisting of inhibitors of the angiotensin converting enzyme (ACE); angiotensin Il receptor blockers; diuretics; calcium channel blockers (CCB); beta-blockers; platelet aggregation inhibitors; cholesterol absorption modulators; HMG-Co-A reductase inhibitors; high density lipoprotein (HDL) increasing compounds; renin inhibitors; IL-6 inhibitors; antiinflammatory corticosteroids; antiproliferative agents; nitric oxide donors; inhibitors of extracellular matrix synthesis; growth factor or cytokine signal transduction inhibitors; MCP-1 antagonists and tyrosine kinase inhibitors providing beneficial or synergistic therapeutic effects over each monotherapy component alone.
Angiotensin Il receptor blockers are understood to be those active agents that bind to the AT1 - receptor subtype of angiotensin Il receptor but do not result in activation of the receptor. As a consequence of the blockade of the AT1 receptor, these antagonists can, e.g. be employed as antihypertensive agents.
Suitable angiotensin Il receptor blockers which may be employed in the combination of the present invention include AT1 receptor antagonists having differing structural features, preferred are those with non-peptidic structures. For example, mention may be made of the compounds that are selected from the group consisting of valsartan (EP 443983), losartan (EP 253310), candesartan (EP 459136), eprosartan (EP 403159), irbesartan (EP 45451 1 ), olmesartan (EP 503785), tasosartan (EP 539086), telmisartan (EP 522314), the compound with the designation E-41 77 of the formula
Figure imgf000053_0001
the compound with the designation SC-52458 of the following formula
Figure imgf000053_0002
and the compound with the designation the compound ZD-8731 of the formula
Figure imgf000053_0003
or, in each case, a pharmaceutically acceptable salt thereof.
Preferred AT1 -receptor antagonists are those agents that have been approved and reached the market, most preferred is valsartan, or a pharmaceutically acceptable salt thereof.
The interruption of the enzymatic degradation of angiotensin to angiotensin Il with ACE inhibitors is a successful variant for the regulation of blood pressure and thus also makes available a therapeutic method for the treatment of hypertension. A suitable ACE inhibitor to be employed in the combination of the present invention is, e.g. a compound selected from the group consisting alacepril, benazepril, benazeprilat; captopril, ceronapril, cilazapril, delapril, enalapril, enaprilat, fosinopril, imidapril, lisinopril, moveltopril, perindopril, quinapril, ramipril, spirapril, temocapril and trandolapril, or in each case, a pharmaceutically acceptable salt thereof.
Preferred ACE inhibitors are those agents that have been marketed, most preferred are benazepril and enalapril.
A diuretic is, for example, a thiazide derivative selected from the group consisting of chlorothiazide, hydrochlorothiazide, methylclothiazide, and chlorothalidon. The most preferred diuretic is hydrochlorothiazide. A diuretic furthermore comprises a potassium sparing diuretic such as amiloride or triameterine, or a pharmaceutically acceptable salt thereof.
The class of CCBs essentially comprises dihydropyridines (DHPs) and non-DHPs, such as diltiazem-type and verapamil-type CCBs.
A CCB useful in said combination is preferably a DHP representative selected from the group consisting of amlodipine, felodipine, ryosidine, isradipine, lacidipine, nicardipine, nifedipine, niguldipine, niludipine, nimodipine, nisoldipine, nitrendipine and nivaldipine, and is preferably a non-DHP representative selected from the group consisting of flunarizine, prenylamine, diltiazem, fendiline, gallopamil, mibefradil, anipamil, tiapamil and verapamil, and in each case, a pharmaceutically acceptable salt thereof. All these CCBs are therapeutically used, e.g. as anti- hypertensive, anti-angina pectoris or anti-arrhythmic drugs.
Preferred CCBs comprise amlodipine, diltiazem, isradipine, nicardipine, nifedipine, nimodipine, nisoldipine, nitrendipine and verapamil or, e.g. dependent on the specific CCB, a pharmaceutically acceptable salt thereof. Especially preferred as DHP is amlodipine or a pharmaceutically acceptable salt thereof, especially the besylate. An especially preferred representative of non-DHPs is verapamil or a pharmaceutically acceptable salt, especially the hydrochloride, thereof.
Beta-blockers suitable for use in the present invention include beta-adrenergic blocking agents (beta-blockers), which compete with epinephrine for beta-adrenergic receptors and interfere with the action of epinephrine. Preferably, the beta-blockers are selective for the beta- adrenergic receptor as compared to the alpha-adrenergic receptors, and so do not have a significant alpha-blocking effect. Suitable beta-blockers include compounds selected from acebutolol, atenolol, betaxolol, bisoprolol, carteolol, carvedilol, esmolol, labetalol, metoprolol, nadolol, oxprenolol, penbutolol, pindolol, propranolol, sotalol and timolol. Where the beta- blocker is an acid or base or otherwise capable of forming pharmaceutically acceptable salts or prodrugs, these forms are considered to be encompassed herein, and it is understood that the compounds may be administered in free form or in the form of a pharmaceutically acceptable salt or a prodrug, such as a physiologically hydrolyzable and acceptable ester. For example, metoprolol is suitably administered as its tartrate salt, propranolol is suitably administered as the hydrochloride salt, and so forth.
Platelet aggregation inhibitors include PLAVIX® (clopidogrel bisulfate), PLETAL® (cilostazol) and aspirin.
Cholesterol absorption modulators include ZETIA® (ezetimibe) and KT6-971 (Kotobuki Pharmaceutical Co. Japan).
HMG-Co-A reductase inhibitors (also called beta-hydroxy-beta-methylglutaryl-co-enzyme-A reductase inhibitors or statins) are understood to be those active agents which may be used to lower lipid levels including cholesterol in blood.
The class of HMG-Co-A reductase inhibitors comprises compounds having differing structural features. For example, mention may be made of the compounds, which are selected from the group consisting of atorvastatin, cerivastatin, fluvastatin, lovastatin, pitavastatin, pravastatin, rosuvastatin and simvastatin, or in each case, a pharmaceutically acceptable salt thereof.
Preferred HMG-Co-A reductase inhibitors are those agents, which have been marketed, most preferred is atorvastatin, pitavastatin or simvastatin, or a pharmaceutically acceptable salt thereof.
HDL-increasing compounds include, but are not limited to, cholesterol ester transfer protein (CETP) inhibitors. Examples of CETP inhibitors include JTT7O5 disclosed in Example 26 of U.S. Patent No. 6,426,365 issued July 30, 2002, and pharmaceutically acceptable salts thereof. Inhibition of interleukin 6 mediated inflammation may be achieved indirectly through regulation of endogenous cholesterol synthesis and isoprenoid depletion or by direct inhibition of the signal transduction pathway utilizing interleukin-6 inhibitor/antibody, interleukin-6 receptor inhibitor/antibody, interleukin-6 antisense oligonucleotide (ASON), gp130 protein inhibitor/antibody, tyrosine kinase inhibitors/antibodies, serine/threonine kinase inhibitors/antibodies, mitogen-activated protein (MAP) kinase inhibitors/antibodies, phosphatidylinositol 3-kinase (PI3K) inhibitors/antibodies, Nuclear factor kappaB (NF-κB) inhibitors/antibodies, IKB kinase (IKK) inhibitors/antibodies, activator protein-1 (AP-1 ) inhibitors/antibodies, STAT transcription factors inhibitors/antibodies, altered IL-6, partial peptides of IL-6 or IL-6 receptor, or SOCS (suppressors of cytokine signaling) protein, PPAR gamma and/or PPAR beta/delta activators/ligands or a functional fragment thereof.
A suitable antiinflammatory corticosteroid is dexamethasone.
Suitable antiproliferative agents are cladribine, rapamycin, vincristine and taxol.
A suitable inhibitor of extracellular matrix synthesis is halofuginone.
A suitable growth factor or cytokine signal transduction inhibitor is, e.g. the ras inhibitor R1 15777.
A suitable tyrosine kinase inhibitor is tyrphostin.
Suitable renin inhibitors are described, e.g. in WO 2006/116435. A preferred renin inhibitor is aliskiren, preferably in the form of the hemi-fumarate salt thereof.
MCP-1 antagonists may, e.g. be selected from anti-MCP-1 antibodies, preferably monoclonal or humanized monoclonal antibodies, MCP-1 expression inhibitors, CCR2-antagonists, TNF-alpha inhibitors, VCAM-1 gene expression inhibitors and anti-C5a monoclonal antibodies.
MCP-1 antagonists and compositions containing such inhibitors are described, e.g. in WO02/070509, WO02/081463, WO02/060900, US2006/670364, US2006/677365, WO2006/097624, US2006/316449, WO2004/056727, WO03/053368, WO00/198289, WO00/157226, WO00/046195, WO00/046196, WO00/046199, WO00/046198, WO00/046197, WO99/046991 , WO99/007351 , WO98/006703, WO97/012615, WO2005/105133, WO03/037376, WO2006/125202, WO2006/085961 , WO2004/024921 , WO2006/074265.
Suitable MCP-1 antagonists are, for instance, C-243 (Telik Inc.); NOX-E36 (Noxxon Pharma AG); AP-761 (Actimis Pharmaceuticals Inc.); ABN-912, NIBR-177 (Novartis AG); CC-11006
(Celgene Corp.); SSR-150106 (Sanofi-Aventis); MLN-1202 (Millenium Pharmaceuticals Inc.);
AGM 067, AGIX-4207, AGM 096 (AtherioGenics Inc.); PRS-211095, PRS-211092 (Pharmos
Corp.); anti-C5a monoclonal antibodies, e.g. neutrazumab (G2 Therapies Ltd.); AZD-6942
(AstraZeneca pic); 2-mercaptoimidazoles (Johnson & Johnson); TEI-E00526, TEI-6122 (Deltagen); RS-504393 (Roche Holding AG); SB-282241 , SB-380732, ADR-7
(GlaxoSmithKline); anti-MCP-1 monoclonal antibodies (Johnson & Johnson).
Combinations of QC-inhibitors with MCP-1 antagonists may be useful for the treatment of inflammatory diseases in general, including neurodegenerative diseases.
Combinations of QC-inhibitors with MCP-1 antagonists are preferred for the treatment of Alzheimer's disease.
Most preferably the QC inhibitor is combined with one or more compounds selected from the following group:
PF-4360365, m266, bapineuzumab, R-1450, Posiphen, (+)-phenserine, MK-0752, LY-450139, E-2012, (R)-flurbiprofen, AZD-103, AAB-001 (Bapineuzumab), Tramiprosate, EGb-761 , TAK- 070, Doxofylline, theophylline, cilomilast, tofimilast, roflumilast, tetomilast, tipelukast, ibudilast, HT-0712, MEM-1414, oglemilast, Linezolid, budipine, isocarboxazid, phenelzine, tranylcypromine, indantadol, moclobemide, rasagiline, ladostigil, safinamide, ABT-239, ABT- 834, GSK-189254A, Ciproxifan, JNJ-17216498, Fmoc-Ala-Pyrr-CN, Z-Phe-Pro-Benzothiazole, Z-321 , ONO-1603, JTP-4819, S-17092, BIBP3226; (R)-N2-(diphenylacetyl)-(R)-N-[1-(4- hydroxyphenyl) ethyl] arginine amide, Cevimeline, sabcomeline, (PD-151832), Donepezil, rivastigmine, (-)-phenserine, ladostigil, galantamine, tacrine, metrifonate, Memantine, topiramate, AVP-923, EN-3231 , neramexane, valsartan, benazepril, enalapril, hydrochlorothiazide, amlodipine, diltiazem, isradipine, nicardipine, nifedipine, nimodipine, nisoldipine, nitrendipine, verapamil, amlodipine, acebutolol, atenolol, betaxolol, bisoprolol, carteolol, carvedilol, esmolol, labetalol, metoprolol, nadolol, oxprenolol, penbutolol, pindolol, propranolol, sotalol, timolol, PLAVIX® (clopidogrel bisulfate), PLETAL® (cilostazol), aspirin, ZETIA® (ezetimibe) and KT6-971 , statins, atorvastatin, pitavastatin or simvastatin; dexamethasone, cladribine, rapamycin, vincristine, taxol, aliskiren, C-243, ABN-912, SSR- 150106, MLN-1202 and betaferon.
In particular, the following combinations are considered:
a QC inhibitor, preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1-30, in combination with Atorvastatin for the treatment and/or prevention of artherosclerosis, a QC inhibitor, preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1-30, in combination with immunosuppressive agents, preferably rapamycin for the prevention and/or treatment of restenosis, a QC inhibitor, preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1-30, in combination with immunosuppressive agents, preferably paclitaxel for the prevention and/or treatment of restenosis, - a QC inhibitor, preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1-30, in combination with AChE inhibitors, preferably Donepezil, for the prevention and/or treatment of Alzheimer's disease, a QC inhibitor, preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1-30, in combination with interferones, preferably Aronex, for the prevention and/or treatment of multiple sclerosis, a QC inhibitor, preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1-30, in combination with interferones, preferably betaferon, for the prevention and/or treatment of multiple sclerosis, a QC inhibitor, preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1-30, in combination with interferones, preferably
Rebif, for the prevention and/or treatment of multiple sclerosis a QC inhibitor, preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1-30, in combination with Copaxone, for the prevention and/or treatment of multiple sclerosis, - a QC inhibitor, preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1-30, in combination with dexamethasone, for the prevention and/or treatment of restenosis, a QC inhibitor, preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1-30, in combination with dexamethasone, for the prevention and/or treatment of atherosclerosis, a QC inhibitor, preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1-30, in combination with dexamethasone, for the prevention and/or treatment of rheumatid arthritis, a QC inhibitor, preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1-30, in combination with HMG-Co-A-reductase inhibitors, for the prevention and/or treatment of restenosis, wherein the HMG-Co-A- reductase inhibitor is selected from atorvastatin, cerivastatin, fluvastatin, lovastatin, pitavastatin, pravastatin, rosuvastatin and simvastatin, a QC inhibitor, preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1-30, in combination with HMG-Co-A reductase inhibitors, for the prevention and/or treatment of atherosclerosis wherein the HMG-Co- A-reductase inhibitor is selected from atorvastatin, cerivastatin, fluvastatin, lovastatin, pitavastatin, pravastatin, rosuvastatin and simvastatin, a QC inhibitor, preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1-30, in combination with HMG-Co-A reductase inhibitors, for the prevention and/or treatment of rheumatoid arthritis wherein the HMG-Co-A-reductase inhibitor is selected from atorvastatin, cerivastatin, fluvastatin, lovastatin, pitavastatin, pravastatin, rosuvastatin and simvastatin, a QC inhibitor, preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1-30, in combination with amyloid-beta antibodies for the prevention and/or treatment of mild cognitive impairment, wherein the amyloid- beta antibody is Acl-24, a QC inhibitor, preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1-30, in combination with amyloid-beta antibodies for the prevention and/or treatment of Alzheimer's disease, wherein the amyloid-beta antibody is Acl-24, a QC inhibitor, preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1-30, in combination with amyloid-beta antibodies for the prevention and/or treatment of neurodegeneration in Down Syndrome, wherein the amyloid-beta antibody is Acl-24, a QC inhibitor, preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1-30, in combination with beta-secretase inhibitors for the prevention and/or treatment of mild cognitive impairment, wherein the beta- secretase inhibitor is selected from WY-25105, GW-840736X and CTS-21166, a QC inhibitor, preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1-30, in combination with beta-secretase inhibitors for the prevention and/or treatment of Alzheimer's disease, wherein the beta- secretase inhibitor is selected from WY-25105, GW-840736X and CTS-21166, - a QC inhibitor, preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1-30, in combination with beta-secretase inhibitors for the prevention and/or treatment of neurodegeneration in Down Syndrome, wherein the beta-secretase inhibitor is selected from WY-25105, GW-840736X and CTS- 21166, - a QC inhibitor, preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1-30, in combination with gamma-secretase inhibitors for the prevention and/or treatment of mild cognitive impairment, wherein the gamma-secretase inhibitor is selected from LY-450139, LY-41 1575 and AN-37124, a QC inhibitor, preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1-30, in combination with gamma-secretase inhibitors for the prevention and/or treatment of Alzheimer's disease, wherein the gamma-secretase inhibitor is selected from LY-450139, LY-41 1575 and AN-37124, a QC inhibitor, preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1-30, in combination with gamma-secretase inhibitors for the prevention and/or treatment of neurodegeneration in Down
Syndrome, wherein the gamma-secretase inhibitor is selected from LY-450139, LY- 411575 and AN-37124.
Such a combination therapy is in particular useful for AD, FAD, FDD and neurodegeneration in Down syndrome as well as atherosclerosis, rheumatoid arthritis, restenosis and pancreatitis.
Such combination therapies might result in a better therapeutic effect (less proliferation as well as less inflammation, a stimulus for proliferation) than would occur with either agent alone.
With regard to the specific combination of inhibitors of QC and further compounds it is referred in particular to WO 2004/098625 in this regard, which is incorporated herein by reference.
Pharmaceutical compositions
To prepare the pharmaceutical compositions of this invention, at least one compound of formula (I) optionally in combination with at least one of the other aforementioned agents can be used as the active ingredient(s). The active ingredient(s) is intimately admixed with a pharmaceutical carrier according to conventional pharmaceutical compounding techniques, which carrier may take a wide variety of forms depending of the form of preparation desired for administration, e.g., oral or parenteral such as intramuscular. In preparing the compositions in oral dosage form, any of the usual pharmaceutical media may be employed. Thus, for liquid oral preparations, such as for example, suspensions, elixirs and solutions, suitable carriers and additives include water, glycols, oils, alcohols, flavoring agents, preservatives, coloring agents and the like; for solid oral preparations such as, for example, powders, capsules, gelcaps and tablets, suitable carriers and additives include starches, sugars, diluents, granulating agents, lubricants, binders, disintegrating agents and the like. Because of their ease in administration, tablets and capsules represent the most advantageous oral dosage unit form, in which case solid pharmaceutical carriers are obviously employed. If desired, tablets may be sugar coated or enteric coated by standard techniques. For parenterals, the carrier will usually comprise sterile water, though other ingredients, for example, for purposes such as aiding solubility or for preservation, may be included.
Injectable suspensions may also prepared, in which case appropriate liquid carriers, suspending agents and the like may be employed. The pharmaceutical compositions herein will contain, per dosage unit, e.g., tablet, capsule, powder, injection, teaspoonful and the like, an amount of the active ingredient(s) necessary to deliver an effective dose as described above. The pharmaceutical compositions herein will contain, per dosage unit, e.g., tablet, capsule, powder, injection, suppository, teaspoonful and the like, from about 0.03 mg to 100 mg/kg (preferred 0.1
- 30 mg/kg) and may be given at a dosage of from about 0.1 - 300 mg/kg per day (preferred 1
- 50 mg/kg per day) of each active ingredient or combination thereof. The dosages, however, may be varied depending upon the requirement of the patients, the severity of the condition being treated and the compound being employed. The use of either daily administration or post- periodic dosing may be employed.
Preferably these compositions are in unit dosage forms from such as tablets, pills, capsules, powders, granules, sterile parenteral solutions or suspensions, metered aerosol or liquid sprays, drops, ampoules, autoinjector devices or suppositories; for oral parenteral, intranasal, sublingual or rectal administration, or for administration by inhalation or insufflation. Alternatively, the composition may be presented in a form suitable for once-weekly or once-monthly administration; for example, an insoluble salt of the active compound, such as the decanoate salt, may be adapted to provide a depot preparation for intramuscular injection. For preparing solid compositions such as tablets, the principal active ingredient is mixed with a pharmaceutical carrier, e.g. conventional tableting ingredients such as corn starch, lactose, sucrose, sorbitol, talc, stearic acid, magnesium stearate, dicalcium phosphate or gums, and other pharmaceutical diluents, e.g. water, to form a solid preformulation composition containing a homogeneous mixture of a compound of the present invention, or a pharmaceutically acceptable salt thereof. When referring to these preformulation compositions as homogeneous, it is meant that the active ingredient is dispersed evenly throughout the composition so that the composition may be readily subdivided into equally effective dosage forms such as tablets, pills and capsules. This solid preformulation composition is then subdivided into unit dosage forms of the type described above containing from 0.1 to about 500 mg of each active ingredient or combinations thereof of the present invention.
The tablets or pills of the compositions of the present invention can be coated or otherwise compounded to provide a dosage form affording the advantage of prolonged action. For example, the tablet or pill can comprise an inner dosage and an outer dosage component, the latter being in the form of an envelope over the former. The two components can be separated by an enteric layer which serves to resist disintegration in the stomach and permits the inner component to pass intact into the duodenum or to be delayed in release. A variety of material can be used for such enteric layers or coatings, such materials including a number of polymeric acids with such materials as shellac, cetyl alcohol and cellulose acetate.
This liquid forms in which the compositions of the present invention may be incorporated for administration orally or by injection include, aqueous solutions, suitably flavoured syrups, aqueous or oil suspensions, and flavoured emulsions with edible oils such as cottonseed oil, sesame oil, coconut oil or peanut oil, as well as elixirs and similar pharmaceutical vehicles. Suitable dispersing or suspending agents for aqueous suspensions, include synthetic and natural gums such as tragacanth, acacia, alginate, dextran, sodium carboxymethylcellulose, methylcellulose, polyvinylpyrrolidone or gelatin.
The pharmaceutical composition may contain between about 0.01 mg and 100 mg, preferably about 5 to 50 mg, of each compound, and may be constituted into any form suitable for the mode of administration selected. Carriers include necessary and inert pharmaceutical excipients, including, but not limited to, binders, suspending agents, lubricants, flavorants, sweeteners, preservatives, dyes, and coatings. Compositions suitable for oral administration include solid forms, such as pills, tablets, caplets, capsules (each including immediate release, timed release and sustained release formulations), granules, and powders, and liquid forms, such as solutions, syrups, elixirs, emulsions, and suspensions. Forms useful for parenteral administration include sterile solutions, emulsions and suspensions.
Advantageously, compounds of the present invention may be administered in a single daily dose, or the total daily dosage may be administered in divided doses of two, three or four times daily. Furthermore, compounds for the present invention can be administered in intranasal form via topical use of suitable intranasal vehicles, or via transdermal skin patches well known to those of ordinary skill in that art. To be administered in the form of transdermal delivery system, the dosage administration will, of course, be continuous rather than intermittent throughout the dosage regimen.
For instance, for oral administration in the form of a tablet or capsule, the active drug component can be combined with an oral, non-toxic pharmaceutically acceptable inert carrier such as ethanol, glycerol, water and the like. Moreover, when desired or necessary, suitable binders; lubricants, disintegrating agents and coloring agents can also be incorporated into the mixture. Suitable binders include, without limitation, starch, gelatin, natural sugars such as glucose or betalactose, corn sweeteners, natural and synthetic gums such as acacia, tragacanth or sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride and the like. Disintegrators include, without limitation, starch, methyl cellulose, agar, bentonite, xanthan gum and the like.
The liquid forms in suitable flavored suspending or dispersing agents such as the synthetic and natural gums, for example, tragacanth, acacia, methyl-cellulose and the like. For parenteral administration, sterile suspensions and solutions are desired. Isotonic preparations which generally contain suitable preservatives are employed when intravenous administration is desired.
The compounds or combinations of the present invention can also be administered in the form of liposome delivery systems, such as small unilamellar vesicles, large unilamellar vesicles, and multilamellar vesicles. Liposomes can be formed from a variety of phospholipids, such as cholesterol, stearylamine or phosphatidylcholines.
Compounds or combinations of the present invention may also be delivered by the use of monoclonal antibodies as individual carriers to which the compound molecules are coupled. The compounds of the present invention may also be coupled with soluble polymers as targetable drug carriers. Such polymers can include polyvinylpyrrolidone, pyran copolymer, polyhydroxypropylmethacrylamidephenol, polyhydroxyethylaspartamid-ephenol, or polyethyl eneoxidepolyllysine substituted with palmitoyl residue. Furthermore, the compounds of the present invention may be coupled to a class of biodegradable polymers useful in achieving controlled release of a drug, for example, polyactic acid, polyepsilon caprolactone, polyhydroxy butyeric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacrylates and cross- linked or amphipathic block copolymers of hydrogels.
Compounds or combinations of this invention may be administered in any of the foregoing compositions and according to dosage regimens established in the art whenever treatment of the addressed disorders is required.
The daily dosage of the products may be varied over a wide range from 0.01 to 1.000 mg per mammal per day. For oral administration, the compositions are preferably provided in the form of tablets containing, 0.01 , 0.05, 0.1 , 0.5, 1.0, 2.5, 5.0, 10.0, 15.0, 25.0, 50.0, 100, 150, 200,
250 and 500 milligrams of each active ingredient or combinations thereof for the symptomatic adjustment of the dosage to the patient to be treated. An effective amount of the drug is ordinarily supplied at a dosage level of from about 0.1 mg/kg to about 300 mg/kg of body weight per day. Preferably, the range is from about 1 to about 50 mg/kg of body weight per day. The compounds or combinations may be administered on a regimen of 1 to 4 times per day.
Optimal dosages to be administered may be readily determined by those skilled in the art, and will vary with the particular compound used, the mode of administration, the strength of the preparation, the mode of administration, and the advancement of disease condition. In addition, factors associated with the particular patient being treated, including patient age, weight, diet and time of administration, will result in the need to adjust dosages.
In a further aspect, the invention also provides a process for preparing a pharmaceutical composition comprising at least one compound of formula (I), optionally in combination with at least one of the other aforementioned agents and a pharmaceutically acceptable carrier.
The compositions are preferably in a unit dosage form in an amount appropriate for the relevant daily dosage. Suitable dosages, including especially unit dosages, of the the compounds of the present invention include the known dosages including unit doses for these compounds as described or referred to in reference text such as the British and US Pharmacopoeias, Remington's Pharmaceutical Sciences (Mack Publishing Co.), Martindale The Extra Pharmacopoeia (London, The Pharmaceutical Press) (for example see the 31st Edition page 341 and pages cited therein) or the above mentioned publications. Examples
Figure imgf000065_0001
Figure imgf000066_0001
Figure imgf000067_0001
Figure imgf000068_0001
Figure imgf000069_0002
General synthesis description
Figure imgf000069_0001
Sodium cyanamide (1.0 eq.) and the corresponding isothiocyanate (1.0 eq.) were dissolved in 10 mL of dry ethanol. The mixture was stirred under reflux for 3 h. After cooling down to room temperature 5 or 9 (1.0 eq.), and N1-((ethylimino)methylen)-N3,N3-dimethylpropan-1 ,3-diamine hydrochloride (1.2 eq.) and 5 mL of dimethylformamide were added and the mixture was stirred at room temperature for 3 h. The solvent was removed and the remaining residue was taken up in 30 mL of chloroform. The organic layer was washed one times by means of water, dried over Na2SC>4, filtered and than the solvent was removed. Purification of the products was done by means of flash-chromatography utilizing basic aluminum oxide and a gradient consisting of chloroform and methanol or was purified by means of semi-preparative HPLC. Detailed synthesis description
Synthesis of 3-(5-methyl-1 H-imidazol-1 -yl)propan-1 -amine (5)
Figure imgf000070_0001
Scheme 1 : Synthesis of the 3-(5-methyl-1 H-imidazol-1 -yl)alkyl-1 -amines (5)
3-(5-meth yl- 1 H-imidazol- 1-yl)propan- 1 -amine (5) 4-methyl-1 -trityl-1 H-imidazole (2)
4-methyl-1 H-imidazole (1 ) (36.53 mmol, 1 eq) of was dissolved in 120 ml. of dimethylformamide, triethylamine (73.06 mmol, 2eq.) and chlorotriphenylmethane (40.1 mmol, 1.1 eq) where added. The mixture was stirred for 3.5 h. The precipitate filtered off and was washed by means of ice-cooled dimethylformamide (2x50 ml.) and water (2x50 ml_). After removal of the solvent the remaining product was dried over P4O10.
Yield: 10.65 g (98.2%). The product was used without further purification.
1 -Trityl-3-[3-(1 ,3-dioxo-1 ,3-dihydro-2H-isoindol-2-yl)propyl]-1 ,4-dimethyl-1 H-imidazol-3- ium bromide (3) 4-Methyl-1 -trityl-1 H-imidazole (i.e 32.85 mmol, 1 eq.) was suspended in acetonitrile (10 ml.) and 2-(3-bromopropyl)isoindoline-1 ,3-dione (32.85 mmol, 1eq.) was added. The mixture was kept under reflux over night. The organic solvent was removed
Purification was done by flash-chromatography using silica gel and a CHCIs/MeOH-gradient. Yield: 10.65g (63.44%).
2-(3-(5-methyl-1 H-imidazol-1 -yl)propyl)isoindoline-1 ,3-dione (4) 1 -Trityl-3-[3-(1 ,3-dioxo-1 ,3-dihydro-2H-isoindol-2-yl)propyl]-1 ,4-dimethyl-1 H-imidazol-3-ium bromide (i.e. 7.86 mmol) was dissolved in a stirred solution containing methanol (20 ml.) and trifluoracetic acid (4 ml_). The mixture was kept under reflux over night. After that the solvent was removed by means of reduced pressure and the remaining oil was purified by flash- chromatography using silica gel and a CHCI3/MeOH-gradient. Yield: 2.05g (97.0%).
3-(5-methyl-1 H-imidazol-1 -yl)propan-1 -amine (5)
2-(3-(5-methyl-1 H-imidazol-1-yl)propyl)isoindoline-1 ,3-dione (i.e. 8.92 mmol, 1 eq.) and hydzine monohydrate (17.84 mmol, 2eq.) were dissolved in dry EtOH (50 ml_). The mixture was kept under reflux over night, then mixture was concentrated down to a volume of 25 ml_. After that hydrochloric acid (cone, 55 ml.) was added and the mixture was heated up to 50 0C for and kept at this temperature for 30 min. The formed precipitate was filtered off. The filtrate was cooled down to 0 0C and solid NaOH was added until a final pH-value of 10-12 is reached. The aqueous solution was extracted by means of CHCI3 (3x50 ml_). The combined organic layers were dried over Na2SO4, filtered and the solvent was removed. The product was purified by means of flash-chromatography using silica gel and a CHCIs/MeOH-gradient. Yield: 0.74 g (60%), viscous oil Yield over all steps: 36.3 %
1H-NMR (CDCI3, 499,78 MHz): δ 1.79-1.847 (m, 2H); 2.179 (s, 3H); 2.694-2.721 (m, 2H); 3.891- 3.920 (m, 2H); 6.731 (s, H); 7.240 (s, solv.); 7.380 (s; H); ESI-MS m/z: 140.3 (M+H)+, 279.4 (2M+H)+; HPLC (λ = 214 nm) rt: dead time (100%)
3-(4-methyl-1 H-imidazol-1 -yl)propan-1 -amine (9)
Figure imgf000071_0001
Scheme 2: Synthesis of 3-(4-methyl-1 H-imidazol-1 -yl)propan-1 -amine (9) 2-(3-(4/5-methyl- 1 H-imidazol- 1 -yl)propyl)isoindoline- 1, 3-dione (7)
4-methyl-1 H-imidazole (6) (36.53mmol, 1 eq) and sodium hydride (60% in mineral oil, 36.53 mmol, 1.0 eq.) were dissolved in 80 ml. of dimethylformamide. The mixture was stirred at room temperature for 2 h until the formation of hydrogen gas deceased. 2-(3- bromopropyl)isoindoline-1 , 3-dione (34.70 mmol, 0.95 eq.) was added and the mixture was stirred at 90 0C overnight. The solvent was removed and the remaining residue was purified by means of flash-chromatography using silica gel and a CHCIs/MeOH-gradient. Yield: 6.1 g (62.0 %) of a mixture of 2-(3-(4-methyl-1 H-imidazol-1-yl)propyl)isoindoline-1 , 3-dione and 2-(3-(5-methyl-1 H-imidazol-1 -yl)propyl)isoindoline-1 , 3-dione
2-(3-(4-methyl- 1 H-imidazol- 1 -yl)propyl)isoindoline-1 , 3-dione (8)
A mixture consisting of 2-(3-(4-methyl-1 H-imidazol-1 -yl)propyl)isoindoline-1 , 3-dione and 2-(3-(5- methyl-1 H-imidazol-1 -yl)propyl)isoindoline-1 , 3-dione (7) (22.65 mmol, 1 eq.) and tritylchlorid (13.6 mmol, 0.6 eq.) were dissolved in 40 ml. of dichloromethane and kept at a temperature of 0 0C for 10 min and 1.5 h at room temperature. The solvent was removed and pressure the remaining solid was purified by means of flash-chromatography using silica gel and a CHCIs/MeOH-gradient. Yield: 0.92 g (15.1 %)
3-(4-methyl-1 H-imidazol-1 -yl)propan-1 -amine (9)
2-(3-(4-methyl-1 H-imidazol-1 -yl)propyl)isoindoline-1 , 3-dione (3.42 mmol, 1 eq.) and hydrazine monohydrate (6.84 mmol, 2 eq.) were dissolved in 20 ml. of ethanol and the mixture was stirred for 12 h under reflux. The mixture was kept under reflux over night, then mixture was concentrated down to a volume of 25 ml_. After that hydrochloric acid (cone, 55 ml.) was added and the mixture was heated up to 50 0C for and kept at this temperature for 30 min. The formed precipitate was filtered off. The filtrate was cooled down to 0 0C and solid NaOH was added until a final pH-value of 10-12 is reached. The aqueous solution was extracted by means of CHCI3 (3x50 ml_). The combined organic layers were dried over Na2SO4, filtered and the solvent was removed. The product was purified by means of flash-chromatography using silica gel and a CHCI3/MeOH-gradient containing aqueous ammonia (2% v/v). Yield: 0.31 g (65.1 %).
1H-NMR (CDCI3, 499,78 MHz): δ 1.819-1.874 (m, 2H); 2.188 (s, 3H); 2.699-2.712 (m, 2H); 3.910-3.948 (m, 2H); 6.594 (s, H); 7.240 (s, solv.); 7.328 (s; H); ESI-MS m/z: 140.3 (M+H)+, 279.4 (2M+H)+; HPLC (λ = 214 nm) rt: dead time (100%) Semi-preparative HPLC-method
The system consisted of Merck-Hitachi device (model LaChrom) equipped with a SP250/21 Luna® 100-7 C18 semi-preparative column (Phenomenex. length: 250 mm, diameter: 21 mm). The compounds were purified using a gradient at a flow rate of 6 ml_/min; whereby eluent (A) was acetonitrile, eluent (B) was water, both containing 0.1 % (v/v) trifluoro acetic acid applying the following gradient: 0 min - 40 min. 40 -95 % (A)
Synthesis of examples Example 1 : 2-Cyano(4-ethylphenyl)-3-(3-(5-methyl-1 H-imidazol-1 -yl)propyl)guanidine
The compound was synthesized starting from 1-ethyl-4-isothiocyanatobenzene (0.24 g, 1.5 mmol) and 3-(5-Methyl-1 H-imidazol-1-yl)propan-1 -amine (0.20 g, 1.5 mmol) as described above.
Yield: 0.45 g (96.7%). 1H NMR: (CDCI3) δ 1.16-1.20 (m, 3H); 2.08-2.13 (br m, 2H); 2.27 (s, 3H); 2.55-2.60 (m, 2H); 3.39-3.44 (m, 2H); 4.18-4.22 (m, 2H); 6.54-6.57 (m, H); 6.93 (s, H); 7.11-7.16 (br m, 4H); 8.06 (s, H); 9.13 (s, H); MS m/z 311.2 (M+H)+; HPLC (λ = 214 nm, [C]): rt 9.07 min (99.6%).
Example 2: 2-Cyano(4-isopropylphenyl)-3-(3-(5-methyl-1 H-imidazol-1 -yl)propyl)- guanidine
The compound was synthesized starting from 1-isopropyl-4-isothiocyanatobenzene (0.27 g, 1.5 mmol) and 3-(5-Methyl-1 H-imidazol-1 -yl)propan-1 -amine (0.20 g, 1.5 mmol) as described above.
Yield: 0.44 g (90.0%). 1H NMR: (CDCI3) δ 1.18 (s, 3H); 1.20 (s, 3H); 2.1 1-2.13 (m, 2H); 2.29 (s, 3H); 2.81-2.88 (br m; H); 3.42-3.47 (m, 2H); 4.23-4.27 (m, 2H); 6.59-6.62 (m, H); 6.97 (s, 1 H); 7.14-7.19 (m, 4H); 8.09 (s, H); 9.39 (s, H); MS m/z 325.4 (M+H)+; HPLC (λ = 214 nm, [C]): rt 10.31 min (99.9%).
Example 3: 2-Cyano(2,3-dihydrobenzo[b][1,4]dioxin-7-yl)-3-(3-(5-methyl-1 H-imidazol-1 - yl)propyl)guanidine
The compound was synthesized starting from 2,3-dihydro-6-isothiocyanato-benzo[b][1 ,4]dioxine (0.29 g, 1.5 mmol) and 3-(5-Methyl-1 H-imidazol-1 -yl)propan-1 -amine (0.20 g, 1.5 mmol) as described above.
Yield: 0.50 g (99.0%). 1H NMR: (DMSOd6) δ 1.80-1.87 (br m, 2H); 2.13 (s, 3H); 3.1 1-3.15 (m, 2H); 3.82-3.86 (m, 2H); 4.22 (s, 4H); 6.59 (s, H); 6.65-6.66 (m, H); 6.67-6.68 (m, H); 6.72-6.73 (m, H); 6.81-6.83 (m, H); 8.30 (s, H); 8.79 (s,H); MS m/z 341.1 (M+H)+; HPLC (λ = 214 nm, [C]): rt 8.30 min (99.4%).
Example 4: 2-Cyano(4-cyanophenyl)-3-(3-(5-methyl-1 H-imidazol-1 -yl)propyl)-guanidine The compound was synthesized starting from 4-isothiocyanatobenzonitrile (0.24 g, 1.5 mmol) and 3-(5-Methyl-1 H-imidazol-1 -yl)propan-1 -amine (0.20 g, 1.5 mmol) as described above.
Yield: 0.29 g (63.0%). 1H NMR: (DMSOd6) δ 1.87-1.94 (m, 2H); 2.14 (s, 3H);3.21-3.25 (m, 2H);
3.88-3.91 (m, 2H); 6.60 (s, H), 7.38-7.40 (m, 2H); 7.51 (s, 1 H); 7.75-7.78 (m, 2H); 8.30 (s, H);
8.45 (s, H)MS m/z 308.1 (M+H)+; HPLC (λ = 214 nm, [C]): rt 7.75 min (99.15%).
Example 5: 2-Cyano(3,4,5-trimethoxyphenyl)-3-(3-(5-methyl-1 H-imidazol-1 -yl)propyl)- guanidine
The compound was synthesized starting from 5-isothiocyanato-1 ,2,3-trimethoxybenzene
(0.099 g, 0.44 mmol) and 3-(5-Methyl-1 H-imidazol-1 -yl)propan-1 -amine (0.20 g, 1.5 mmol) as described above.
Yield: 0.13 g (80.0%). 1H NMR: (CDCI3) δ 2.15-2.19 (m, 2H); 2.32 (s, 3H); 3.44-3.48 (m, 2H);
3.75 (s, 3H); 3.82 (s, 6H); 4.26-4.30 (m, 2H); 6.53 (s, H); 7.01 (s, H); 7.08-7.13 (m, H); 7.97 (s,
H); 9.54 (s, H);MS m/z 373.1 (M+H)+; HPLC (λ = 214 nm, [C]): rt 7.69 min (99.05%).
Example 6: 2-Cyano(4-ethoxyphenyl)-3-(3-(5-methyl-1 H-imidazol-1 -yl)propyl)-guanidine
The compound was synthesized starting from 1-isothiocyanato-4-methoxybenzene (0.25 g, 1.5 mmol) and 3-(5-Methyl-1 H-imidazol-1 -yl)propan-1 -amine (0.20 g, 1.5 mmol) as described above.
Yield: 0.49 g (99.0%).1H NMR: (CDCI3) δ 1.34-1.38 (m, 3H); 2.09-2.12 (m, 2H); 2.29 (s, 3H);3.37-3.42 (br m, 2H); 3.93-3.98 (m, 2H); 4.19-4.22 (m, 2H); 6.51-6.53 (m, H); 6.80-6.83 (m, 2H); 6.96 (s, H); 7.13-7.15 (m, 2H); 7.81 (s, H); 9.24 (s, H). MS m/z 327.4 (M+H)+; HPLC (λ = 214 nm, [C]): rt 8.15 min (100%).
Example 7: 2-Cyano(3-(5-methyl-1 H-imidazol-1 -yl)propyl)-3-(3.4-dimethylphenyl)- guanidine
The compound was synthesized starting from 4-isothiocyanato-1 ,2-dimethylbenzene (0.36 g, 2.2 mmol) and 3-(5-Methyl-1 H-imidazol-1 -yl)propan-1 -amine (0.20 g, 1.5 mmol) as described above.
Yield: 0.042 g (7.5%); mp: 154.0-156.0 0C; 1H NMR: δ (CD3DO) δ 2.05-2.12 (m; 2H); 2.26 (s; 3H); 2.27 (s; 3H); 2.36-2.37 (m; 3H); 3.30-3.34 (m; 2H); 4.16-4.20 (m; 2H); 6.95-6.96 (m; H); 6.97 (s; H); 7.17-7.19 (m; H); 7.31 (s; H); 8.86 (s; H); MS m/z 311.4 (M+H)+; ESI-FTICR-MS: m/z 31 1.19760 ([M+H]+; calc. for Ci7H23N6 + 311.19787); (λ = 214 nm, [A]): rt 22.93 min (99.3%)
Example 8: (3-(5-Methyl-1 H-imidazol-1 -yl)propyl)-2-cyano-3-mesitylguanidine The compound was synthesized starting from 2-isothiocyanato-1 ,3,5-trimethylbenzene (0.266 g, 1.5 mmol) and 3-(5-Methyl-1 H-imidazol-1 -yl)propan-1 -amine (0.20 g, 1.5 mmol) as described above.
Yield: 0.36 g (73.3%); 1H NMR: δ (CDCI3) δ 1.86-1.93 (m; 2H); 2.12 (s; 3H); 2.18 (s, 6H); 2.28 (s; 3H); 3.19-3.24 (m; 2H); 3.78-3.82 (m; 2H); 4.43 (br s, H); 6.67 (s; H); 6.74 (s; H); 6.95 (s, 2H); 7.26 (s; H); MS m/z 325.3 (M+H)+; HPLC (λ = 214 nm, [B]): rt 9.41 min (100%)
Example 9: (3-(5-methyl-1 H-imidazol-1 -yl)propyl)-2-cyano-3-(biphenyl-4-yl)guanidine
The compound was synthesized starting from 1-isothiocyanato-4-phenylbenzene (0.152 g, 0.72 mmol) and 3-(5-Methyl-1 H-imidazol-1 -yl)propan-1 -amine (0.10 g, 0.72 mmol) as described above.
Yield: 0.09O g (40.0%); 1H NMR: (DMSOd6) δ 1.83-1.90 (m, 2H); 2.12 (s, 3H); 3.15-3.20 (m, 2H); 3.84-3.88 (m, 2H); 6.58 (s, H); 7.28-7.33 (br m, 4H); 7.40-7.44 (m, 2H); 7.50 (s, H); 7.61- 7.64 (m, 4H); 9.07 (s, H). MS m/z 359.4 (M+H)+; HPLC (λ = 214 nm, [C]): rt 14.19 min (99.3%)
Example 10: (3-(5-methyl-1 H-imidazol-1 -yl)propyl)-2-cyano-3-(naphthalen-2-yl)guanidine
The compound was synthesized starting from 2-isothiocyanatonaphthalene (0.185 g, 1.0 mmol) and 3-(5-Methyl-1 H-imidazol-1 -yl)propan-1 -amine (0.139 g, 1.0 mmol) as described above. Yield: 0.17 g (41.0%); 1H NMR: (DMSOd6) δ 1.84-1.91 (br m, 2H); 2.12 (s, 3H); 3.13-3.21 (m, 2H); 3.85-3.89 (m, 2H); 6.60 (s, H); 7.29-7.30 (m, H); 7.35-7.38 (m, 2H); 7.41-7.49 (br m, 2H); 7.54 (s, H); 7.68 (s, H); 7.81-7.87 (br m, 3H); 9.20 (s, H). MS m/z 333.5 (M+H)+; HPLC (λ = 214 nm, [C]): rt 12.03 min (97.7%)
Example 11 : (3-(5-Methyl-1 H-imidazol-1 -yl)propyl)-2-cyano-3-(naphthalen-1 -yl)guanidine
The compound was synthesized starting from 1-isothiocyanatonaphthalene (0.278 g, 1.5 mmol) and 3-(5-Methyl-1 H-imidazol-1 -yl)propan-1 -amine (0.20 g, 1.5 mmol) as described above.
Yield: 0.33 g (66.7%).1H NMR: (500 MHz, DMSOd6) δ 1.81-1.83 (m, 2H); 2.09 (s, 3H); 3.13- 3.14 (m, 2H); 3.78-3.81 (m, 2H); 6.59 (s, H); 6.86 (br s, H); 7.38-7.40 (m, H); 7.47 (s, H); 7.51- 7.60 (br m, 3H); 7.84-7.86 (m, H); 7.90-7.91 (m, H); 7.97-7.99 (m, H); 9.23 (s, H). MS m/z 333.4 (M+H)+; HPLC (λ = 214 nm, [B]): rt 8.78 min (99.1%) Example 12: (3-(5-methyl-1 H-imidazol-1 -yl)propyl)-3-(benzo[c][1 ,2,5]thiadiazol-6-yl)-2- cyanoguanidine
The compound was synthesized starting from 5-isothiocyanatobenzo[c][1 ,2,5]thiadiazole (0.290 g, 1.5 mmol) and 3-(5-Methyl-1 H-imidazol-1-yl)propan-1 -amine (0.20 g, 1.5 mmol) as described above.
Yield: 0.27 g (52.9%). 1H NMR: (DMSOd6) δ 1.86-1.93 (m, 2H); 2.13 (s, 3H); 3.20-3.24 (m, 2H); 3.86-3.90 (m, 2H); 6.58 (s, H); 7.50 (s, H); 7.61-7.63 (m, H); 7.65-7.67 (m, H); 7.79 (s, H); 7.99- 8.01 (m, H); 9.47 (br s, H). MS m/z 341.1 (M+H)+; HPLC (λ = 214 nm, [C]): rt 8.88 min (100%)
Example 13: (3-(5-Methyl-1 H-imidazol-1 -yl)propyl)-3-(3,4-dichlorophenyl)-2- cyanoguanidine
The compound was synthesized starting from 1 ,2-dichloro-4-isothiocyanatobenzene (0.16 g, 0.8 mmol) and 3-(5-Methyl-1 H-imidazol-1-yl)propan-1 -amine (0.14 g, 1.0 mmol) as described above. Yield: 0.22 g (78.5%). 1H NMR: (DMSO-d6) δ 1.83-1.87 (m, 2H); 2.12 (s, 3H); 3.14-3.18 (m, 2H); 3.83-3.87 (m, 2H); 6.58 (s, H); 7.20-7.23 (m, H); 7.46-7.49 (m, 3H); 7.54-7.56 (m, H); 9.17 (s, H). MS m/z 351.5 (M+H)+; HPLC (λ = 214 nm, [B]): rt 10.67 min (97.8%)
Example 14: (Benzo[d][1,3]dioxol-6-yl)-2-cyano-3-(3-(5-methyl-1 H-imidazol-1 - yl)propyl)guanidine
The compound was synthesized starting from 5-isothiocyanatobenzo[d][1 ,3]dioxole (0.14 g, 0.80 mmol) and 3-(5-Methyl-1 H-imidazol-1-yl)propan-1 -amine (0.14 g, 1.0 mmol) as described above.
Yield: 0.21 g (81.2%). 1H NMR: δ (CDCI3) δ 1.91-1.98 (m, 2H); 2.15 (s, 3H); 3.22-3.27 (m, 2H); 3.82-3.85 (m, 2H); 4.92 (br s, H); 6.01 (s, 2H); 6.63-6.68 (br m, 2H); 6.79-6.81 (m, H); 7.32 (s, H); 7.38 (s, H). MS m/z 327.1 (M+H)+; HPLC (λ = 214 nm, [B]): rt 7.42 min (98.5%)
Example 15: 2-Cyano(4-methoxyphenyl)-3-(3-(5-methyl-1 H-imidazol-1 - yl)propyl)guanidine The compound was synthesized starting from 1-isothiocyanato-4-methoxybenzene (0.17 g, 1.0 mmol) and 3-(5-Methyl-1 H-imidazol-1-yl)propan-1 -amine (0.17 g, 1.2 mmol) as described above.
Yield: 0.30 g (96.3%). 1H NMR: (CDCI3) δ 1.90-1.99 (br m, 2H); 2.14 (s, 3H); 3.23-3.27 (m, 2H); 3.80 (s, 3H); 3.82-3.87 (m, 2H); 4.75 (br s, H); 6.67 (s, H), 6.90-6.94 (m, 2H); 7.08-7.1 1 (m, 2H); 7.22-7.26 (m, H + Sol); 7.29 (s, H). MS m/z 313.2 (M+H)+; HPLC (λ = 214 nm, [B]): rt 7.58 min (99.0%)
Example 16: 2-Cyano-(3,5-dimethoxyphenyl)-3-(3-(5-methyl-1 H-imidazol-1 - yl)propyl)guanidine
The compound was synthesized starting from 1-isothiocyanato-3,5-dimethoxybenzene (0.293 g, 1.5 mmol) and 3-(5-Methyl-1 H-imidazol-1-yl)propan-1 -amine (0.20 g, 1.5 mmol) as described above.
Yield: 0.35 g (68.0%); 1H NMR: (CDCI3) δ 1.92-1.99 (m; 2H); 2.15 (s; 3H); 3.25-3.30 (m, 2H); 3.76 (s, 6H); 3.81-3.86 (m; 2H); 5.20 (br s, H); 6.30 (s; 2H); 6.37 (s; H); 6.68 (s, H); 7.31 (s, H); 7.46 (s; H); MS m/z 343.0 (M+H)+; HPLC (λ = 214 nm, [B]): rt 9.87 min (99.0%)
Example 17: 2-cyano(4-ethoxyphenyl)-3-(3-(4-methyl-1 H-imidazol-1 -yl)propyl)-guanidine
The compound was synthesized starting from 1-ethoxy-4-isothiocyanatobenzene (0.108 g, 0.60 mmol) and 3-(4-Methyl-1 H-imidazol-1 -yl)propan-1 -amine (0.084 g, 0.60 mmol) as described above.
Yield: 0.15 g (76.7%). 1H NMR (DMSO-d6): δ 1.29-1.33 (m, 3H); 1.83-1.90 (m, 2H); 2.05 (s,
3H); 3.08-3.13 (m, 2H); 3.84-3.87 (m; 2H); 3.97-4.03 (m; 2H); 6.83 (s, H); 6.87-6.91 (m; 3H);
7.08-7.12 (m; 2H); 7.45 (s; H); 8.77 (s, H). MS m/z 327.4 (M+H)+; HPLC (λ = 214 nm, [C]): rt 8.44 min (96.9%)
Example 18: 2-cyano(3,5-dimethoxyphenyl)-3-(3-(4-methyl-1 H-imidazol-1 -yl)propyl)- guanidine
The compound was synthesized starting from 1-isothiocyanato-3,5-dimethoxybenzene (0.117 g, 0.60 mmol) and 3-(4-Methyl-1 H-imidazol-1 -yl)propan-1 -amine (0.084 g, 0.60 mmol) as described above.
Yield: 0.18 g (87.6%). 1H NMR (DMSOd6): δ 1.96-2.02 (m, 2H); 2.15 (s, 3H); 3.22-3.29 (m, 2H);
3.77 (s, 6H); 3.83-3.90 (m, 2H); 5.16 (br s, H); 6.30-6.31 (m, 2H); 6.37-6.38 (m, H); 6.57 (s, H);
7.30 (s, H); 7.41 (s, H) . MS m/z 343.3 (M+H)+; HPLC (λ = 214 nm, [C]): rt 8.22 min (99.3%)
Example 19: 2-cyano(2,3-dihydrobenzo[b][1,4]dioxin-7-yl)-3-(3-(4-methyl-1 H-imidazol-1 - yl)propyl)guanidine
The compound was synthesized starting from 2,3-dihydro-6-isothiocyanato-benzo[b][1 ,4]dioxine
(0.116 g, 0.60 mmol) and 3-(4-Methyl-1 H-imidazol-1 -yl)propan-1 -amine (0.084 g, 0.60 mmol) as described above. Yield: 0.14 g (72.8%). 1H NMR (DMSO-d6): δ 1.82-1.89 (m, 2H); 2.05 (s, 3H); 3.07-3.12 (m, 2H); 3.84-3.87 (m, 2H); 4.22 (s, 4H); 6.64-6.66 (m, H); 6.67 (s, H); 6.72-6.73 (m, 2H); 6.81-6.83 (m, H); 7.45 (s, H)); 8.77 (s, H). MS m/z 341.2 (M+H)+; HPLC (λ = 214 nm, [C]): rt 8.64 min (100%)
Example 20: 2-cyano(mesityl)-3-(3-(4-methyl-1 H-imidazol-1 -yl)propyl)guanidine
The compound was synthesized starting from 2-isothiocyanato-1 ,3,5-trimethylbenzene (0.106 g, 0.60 mmol) and 3-(4-Methyl-1 H-imidazol-1-yl)propan-1 -amine (0.084 g, 0.60 mmol) as described above.
Yield: 0.088 g (45.2%).1H NMR (DMSOd6): δ 1.80-1.84 (m, 2H); 2.04 (s, 3H); 2.09 (s, 6H); 2.23 (s, 3H); 3.04-3.06 (m, 2H); 3.80-3.84 (m, 2H); 6.59 (br s, H); 6.80 (s, H); 6.91 (s, 2H); 7.42 (s, H); 8.42 (s, H). MS m/z 325.2 (M+H)+; HPLC (λ = 214 nm, [C]): rt 10.05 min (98.6%)
Example 21 : 2-cyano(4-isopropylphenyl)-3-(3-(4-methyl-1 H-imidazol-1 -yl)propyl)- guanidine The compound was synthesized starting from 1-isopropyl-4-isothiocyanatobenzene (0.106 g, 0.60 mmol) and 3-(4-Methyl-1 H-imidazol-1 -yl)propan-1 -amine (0.084 g, 0.60 mmol) as described above.
Yield: 0.17 g (87.3%). 1H NMR (CDCI3): δ 1.23 (s, 3H); 1.25 (s, 3H); 1.95-2.00 (m, 2H); 2.16 (s, 3H); 2.88-2.95 (m, H); 3.21-3.26 (m, 2H); 3.86-3.89 (m, 2H); 4.89 (br s, H); 6.57 (s, H); 7.07- 7.09 (m, 2H); 7.26-7.32 (br m, 3H). MS m/z 325.3 (M+H)+; HPLC (λ = 214 nm, [C]): rt 12.75 min (100%)
Example 22: 2-cyano(4-ethylphenyl)-3-(3-(4-methyl-1 H-imidazol-1 -yl)propyl)guanidine
The compound was synthesized starting from 1-ethyl-4-isothiocyanatobenzene (0.100 g, 0.60 mmol) and 3-(4-Methyl-1 H-imidazol-1 -yl)propan-1 -amine (0.084 g, 0.60 mmol) as described above.
Yield: 0.17 g (91.2%). 1H NMR (CDCI3): δ 1.21-1.25 (m, 3H); 1.94-2.01 (m, 2H); 2.15 (s, 3H);
2.62-2.68 (m, 2H); 3.21-3.26 (m, 2H); 3.85-3.89 (m, 2H); 4.91 (br s, H); 6.56 (s, H); 7.06-7.09
(m, 2H); 7.23 (s, H); 7.25 (s, H); 7.31 (s, H); 7.39 (s, H). MS m/z 31 1.2 (M+H)+; HPLC (λ = 214 nm, [C]): rt 11.23 min (100%)
Example 23: 2-cyano(3-(4-methyl-1 H-imidazol-1 -yl)propyl)-3-(naphthalen-1 -yl)-guanidine
The compound was synthesized starting from 1-isothiocyanatonaphthalene (0.11 1 g, 0.60 mmol) and 3-(4-Methyl-1 H-imidazol-1 -yl)propan-1 -amine (0.084 g, 0.60 mmol) as described above. Yield: 0.15 g (75.2%). 1H NMR (DMSO-d6): δ 1.82-1.86 (m, 2H); 2.04 (s, 3H); 3.09-3.1 1 (m, 2H); 3.79-3.83 (m, 2H); 6.79 (s, H); 6.86 (br s, H); 7.37-7.40 (m, 2H); 7.51-7.60 (br m, 3H); 7.83-7.85 (m; 2H); 7.89-7.99 (m, H); 9.20 (s, H). MS m/z 333.4 (M+H)+; HPLC (λ = 214 nm, [C]): rt 10.72 min (100%)
Example 24: (benzo[c][1 ,2,5]thiadiazol-6-yl)-2-cyano-3-(3-(4-methyl-1 H-imidazol-1 -yl)- propyl)-guanidine
The compound was synthesized starting from 5-isothiocyanatobenzo[c][1 ,2,5]thiadiazole (0.139 g, 0.72 mmol) and 3-(4-Methyl-1 H-imidazol-1 -yl)propan-1 -amine (0.10 g, 0.72 mmol) as described above.
Yield: 0.060 g (24.5%). 1H NMR (DMSOd6, 500 MHz): δ 1.93-1.95 (m, 2H); 2.05 (s, 3H); 3.19- 3.23 (m, 2H); 3.91-3.94 (m, 2H); 6.86 (s; H); 7.49 (s, H); 7.63-7.66 (m, H); 7.67-7.69 (m, H);
7.81 (s, H); 8.03-8.05 (m, H); 9.48 (s, H). MS m/z 341.3 (M+H)+; HPLC (λ = 214 nm, [C]): rt 8.77 min (97.7%)
Example 25: 2-cyano(3,4,5-trimethoxyphenyl)-3-(3-(4-methyl-1 H-imidazol-1 -yl)propyl)- guanidine
The compound was synthesized starting from 5-isothiocyanato-1 ,2,3-trimethoxybenzene
(0.162 g, 0.72 mmol) and 3-(4-Methyl-1 H-imidazol-1 -yl)propan-1 -amine (0.10 g, 0.72 mmol) as described above.
Yield: 0.110 g (41.0%). 1H NMR (CDCI3): δ 1.95-2.01 (m, 2H); 2.14 (s, 3H); 3.22-3.27 (m, 2H);
3.82 (s, 9 H); 3.86-3.88 (m, 2H); 5.03 (br s, H); 6.41 (s, 2H); 6.56 (s, H); 7.28 (s, H); 7.46 (s, H). MS m/z 373.3 (M+H)+; HPLC (λ = 214 nm, [C]): rt 8.64 min (100%)
Example 26: 2-cyano(4-cyanophenyl)-3-(3-(4-methyl-1 H-imidazol-1 -yl)propyl)guanidine
The compound was synthesized starting from 4-isothiocyanatobenzonitrile (0.115 g, 0.72 mmol) and 3-(4-Methyl-1 H-imidazol-1 -yl)propan-1 -amine (0.10 g, 0.72 mmol) as described above. Yield: 0.045 g (20.3%). 1H NMR (DMSO-d6): δ 1.86-1.93 (m, 2H); 3.03 (s, 3H); 3.14-3.19 (m, 2H); 3.86-3.90 (m, 2H); 6.82 (s, H); 7.34-7.36 (m, 2H); 7.44 (s, H); 7.73-7.76 (m, 3H); 9.41 (s, H). MS m/z 308.4 (M+H)+; HPLC (λ = 214 nm, [C]): rt 8.13 min (100%) Example 27: (3,4-dichlorophenyl)-2-cyano-3-(3-(4-methyl-1 H-imidazol-1 - yl)propyl)guanidine
The compound was synthesized starting from 1 ,2-dichloro-4-isothiocyanatobenzene (0.147 g, 0.72 mmol) and 3-(4-Methyl-1 H-imidazol-1 -yl)propan-1 -amine (0.10 g, 0.72 mmol) as described above.
Yield: 0.07O g (27.6%). 1H NMR (DMSO-d6): δ 1.84-1.91 (m, 2H); 2.03 (s, 3H); 3.10-3.15 (m, 2H); 3.84-3.88 (m, 2H); 6.81 (s, H); 7.19-7.22 (m, H); 7.43-7.50 (br m, 3H); 7.54-7.56 (m, H); 9.14 (s, H). MS m/z 351.3 (M+H)+; HPLC (λ = 214 nm, [C]): rt 12.85 min (100%)
Example 28: 2-cyano(4-methoxyphenyl)-3-(3-(4-methyl-1 H-imidazol-1 -yl)propyl)- guanidine
The compound was synthesized starting from 1-isothiocyanato-4-methoxybenzene (0.119 g, 0.72 mmol) and 3-(4-Methyl-1 H-imidazol-1 -yl)propan-1 -amine (0.10 g, 0.72 mmol) as described above. Yield: 0.090 g (40.0%). 1H NMR (CDCI3): δ 1.92-1.98 (m, 2H); 2.14 (s, 3H); 3.19-3.24 (m, 2H); 3.80 (s, 3H); 3.83-3.87 (m, 2H); 4.73 (br s, H); 6.54 (s, H); 6.91-6.94 (m, 2H); 7.07-7.1 1 (m, 2H), 7.23-7.29 (m, 2H). MS m/z 313.2 (M+H)+; HPLC (λ = 214 nm, [C]): rt 8.35 min (100%)
Example 29: 2-cyano-1 -[3-(4-methyl-1 H-imidazol-1 -yl)propyl]-4-phenylbenzene-1 - guanidine
The compound was synthesized starting from 1-isothiocyanato-4-phenylbenzene (0.38O g, 1.18 mmol) and 3-(4-Methyl-1 H-imidazol-1 -yl)propan-1 -amine (0.164 g, 1.18 mmol) as described above.
Yield: 0.25 g (59.1 %). 1H NMR (DMSO-d6, 500 MHz): δ 1.89-1.95 (m, 2H); 2.06 (s, 3H); 3.16- 3.19 (m, 2H); 3.89-3.92 (m, 2H); 6.85 (s, H); 7.31-7.36 (br m, 4H); 7.43-7.48 (br m, 3H); 7.64- 7.66 (m, 4H); 9.08 (br s, H). MS m/z 359.4 (M+H)+; HPLC (λ = 214 nm, [C]): rt 14.19 min (99.3%)
Example 30: 2-cyano(3-(4-methyl-1 H-imidazol-1 -yl)propyl)-3-(naphthalen-2-yl)-guanidine The compound was synthesized starting from 2-isothiocyanatonaphthalene (0.167 g, 0.90 mmol) and 3-(4-Methyl-1 H-imidazol-1 -yl)propan-1 -amine (0.125 g, 0.90 mmol) as described above.
Yield: 0.093 g (31.1%). 1H NMR (DMSO-d6, 500 MHz): δ 1.89-1.95 (m, 2H); 2.06 (s, 3H); 3.16- 3.20 (m, 2H); 3.90-3.93 (m, 2H); 6.87 (s, H); 7.31-7.33 (m, H); 7.38-7.40 (m, H); 7.44-7.57 (br m, 3H); 7.70 (s, H); 7.84-7.90 (br m, 3H); 9.21 (br s, H). MS m/z 333.5 (M+H)+; HPLC (λ = 214 nm, [C]): rt 1 1.73 min (97.14%)
Activity screening Fluorometric assays
All measurements were performed with a BioAssay Reader HTS-7000Plus for microplates (Perkin Elmer) at 30 0C. QC activity was evaluated fluorometrically using H-Gln-βNA. The samples consisted of 0.2 mM fluorogenic substrate, 0.25 U pyroglutamyl aminopeptidase (Unizyme, Hørsholm, Denmark) in 0.2 M Tris/HCI, pH 8.0 containing 20 mM EDTA and an appropriately diluted aliquot of QC in a final volume of 250 μl. Excitation/emission wavelengths were 320/410 nm. The assay reactions were initiated by addition of glutaminyl cyclase. QC activity was determined from a standard curve of β-naphthylamine under assay conditions. One unit is defined as the amount of QC catalyzing the formation of 1 μmol pGlu-βNA from H-GIn- βNA per minute under the described conditions.
In a second fluorometric assay, QC was activity determined using H-GIn-AMC as substrate. Reactions were carried out at 30 0C utilizing the NOVOStar reader for microplates (BMG labtechnologies). The samples consisted of varying concentrations of the fluorogenic substrate, 0.1 U pyroglutamyl aminopeptidase (Qiagen) in 0.05 M Tris/HCI, pH 8.0 containing 5 mM EDTA and an appropriately diluted aliquot of QC in a final volume of 250 μl. Excitation/emission wavelengths were 380/460 nm. The assay reactions were initiated by addition of glutaminyl cyclase. QC activity was determined from a standard curve of 7-amino-4-methylcoumarin under assay conditions. The kinetic data were evaluated using GraFit sofware.
Spectrophotometric assay of QC
This novel assay was used to determine the kinetic parameters for most of the QC substrates. QC activity was analyzed spectrophotometrically using a continuous method, that was derived by adapting a previous discontinuous assay (Bateman, R. C. J. 1989 J Neurosci Methods 30, 23-28) utilizing glutamate dehydrogenase as auxiliary enzyme. Samples consisted of the respective QC substrate, 0.3 mM NADH, 14 mM α-Ketoglutaric acid and 30 U/ml glutamate dehydrogenase in a final volume of 250 μl. Reactions were started by addition of QC and persued by monitoring of the decrease in absorbance at 340 nm for 8-15 min.
The initial velocities were evaluated and the enzymatic activity was determined from a standard curve of ammonia under assay conditions. All samples were measured at 30 0C, using either the SPECTRAFIuor Plus or the Sunrise (both from TECAN) reader for microplates. Kinetic data was evaluated using GraFit software.
Inhibitor assay For inhibitor testing, the sample composition was the same as described above, except of the putative inhibitory compound added. For a rapid test of QC-inhibition, samples contained 4 mM of the respective inhibitor and a substrate concentration at 1 KM. For detailed investigations of the inhibition and determination of K-values, influence of the inhibitor on the auxiliary enzymes was investigated first. In every case, there was no influence on either enzyme detected, thus enabling the reliable determination of the QC inhibition. The inhibitory constant was evaluated by fitting the set of progress curves to the general equation for competitive inhibition using GraFit software.
Analytical methods The analytical HPLC-system consisted of a Merck-Hitachi device (model LaChrom®) utilizing a Li-Chrospher® 100 RP 18 (5 μm), analytical column (length: 125 mm, diameter: 4 mm), and a diode array detector (DAD) with λ = 214 nm as the reporting wavelength. The compounds were analyzed using a gradient at a flow rate of 1 mL/min; whereby eluent (A) was acetonitrile, eluent (B) was water, both containing 0.1 % (v/v) trifluoro acetic acid applying the following gradient: Method [A]: 0 min - 5 min -» 5% (A), 5 min - 17 min -» 5 - 15% (A), 15 min - 27 min -» 15 - 95% (A) 27 min - 30 min -> 95% (A). Method [B]: 0 min - 15 min -> 5 - 50 % (A), 15 min - 20 min -> 50 - 95 % (A), 20 min - 23 min -» 95 % (A). Method [C]: 0 min - 20 min ^ 5 - 60 % (A), 20 min - 25 min -> 60 - 95 % (A), 25 min - 30 min -> 95 % (A). The purities of all reported compounds were determined by the percentage of the peak area at 214 nm. ESI-Mass spectra were obtained with a SCIEX API 365 spectrometer (Perkin Elmer) utilizing the positive ionization mode.
The high resolution positive ion ESI mass spectra were obtained from a Bruker Apex III 7Oe Fourier transform ion cyclotron resonance mass spectrometer (Bruker Daltonics, Billerica, USA) equipped with an Infinity™ cell, a 7.0 Tesla superconducting magnet (Bruker, Karlsruhe, Germany), an RF-only hexapole ion guide and an external electrospray ion source (API Apollo, voltages: endplate, -3.700V; capillary, -4.400V; capillary exit, 100V; skimmer 1.15 V; skimmer 2.6 V). Nitrogen was used as drying gas at 15O0C. The sample solutions were introduced continuously via a syringe pump with a flow rate of 120μl h"1. All data were acquired with 256 k data points and zero filled to 1024 k by averaging 32 scans. The melting points were detected utilizing a Kofler melting point device. They are not corrected The 1H NMR-Spectra (500 MHz) were recorded at a BRUKER AC 500. The solvent was DMSO- D6, unless otherwise specified. Chemial shifts are expressed as parts per million (ppm) downfiled from tetramethylsilan. Splitting patterns have been designated as follows: s (singulet), d (doublet), dd (doublet of doublet), t (triplet), m (multiplet) and br (broad signal).
MALDI-TOF mass spectrometry
Matrix-assisted laser desorption/ionization mass spectrometry was carried out using the Hewlett-Packard G2025 LD-TOF System with a linear time of flight analyzer. The instrument was equipped with a 337 nm nitrogen laser, a potential acceleration source (5 kV) and a 1.0 m flight tube. Detector operation was in the positive-ion mode and signals are recorded and filtered using LeCroy 9350M digital storage oscilloscope linked to a personal computer. Samples (5 μl) were mixed with equal volumes of the matrix solution. For matrix solution DHAP/DAHC was used, prepared by solving 30 mg 2',6'-dihydroxyacetophenone (Aldrich) and 44 mg diammonium hydrogen citrate (Fluka) in 1 ml acetonitrile/0.1% TFA in water (1/1 , v/v). A small volume (~ 1 μl) of the matrix-analyte-mixture was transferred to a probe tip and immediately evaporated in a vacuum chamber (Hewlett-Packard G2024A sample prep accessory) to ensure rapid and homogeneous sample crystallization. For long-term testing of Glu1-cyclization, Aβ-derived peptides were incubated in 10Oμl 0.1 M sodium acetate buffer, pH 5.2 or 0.1 M Bis-Tris buffer, pH 6.5 at 30 0C. Peptides were applied in 0.5 mM [Aβ(3-1 1 )a] or 0.15 mM [Aβ(3-21 )a] concentrations, and 0.2 U QC is added all 24 hours. In case of Aβ(3-21 )a, the assays contained 1 % DMSO. At different times, samples are removed from the assay tube, peptides extracted using ZipTips (Millipore) according to the manufacturer's recommendations, mixed with matrix solution (1 :1 v/v) and subsequently the mass spectra recorded. Negative controls either contain no QC or heat deactivated enzyme. For the inhibitor studies the sample composition was the same as described above, with exception of the inhibitory compound added (5 mM or 2 mM of a test compound of the invention).
The first QC inhibitors were disclosed in WO 2004/098591 and WO 2005/075436. There are no other potent QC inhibitors known in the art. The same holds true for combinations and compositions for the treatment of neuronal diseases comprising QC inhibitors. Compounds and combinations of the invention may have the advantage that they are, for example, more potent, more selective, have fewer side-effects, have better formulation and stability properties, have better pharmacokinetic properties, be more bioavailable, be able to cross blood brain barrier and are more effective in the brain of mammals, are more compatible or effective in combination with other drugs or be more readily synthesized than other compounds of the prior art.
Throughout the specification and the claims which follow, unless the context requires otherwise, the word 'comprise', and variations such as 'comprises' and 'comprising', will be understood to imply the inclusion of a stated integer, step, group of integers or group of steps but not to the exclusion of any other integer, step, group of integers or group of steps.
All patents and patent applications mentioned throughout the specification of the present invention are herein incorporated in their entirety by reference.
The invention embraces all combinations of preferred and more preferred groups and embodiments of groups recited above.

Claims

Claims
1. A compound of formula (I),
Figure imgf000085_0001
or a pharmaceutically acceptable salt, solvate or polymorph thereof, including all tautomers and stereoisomers thereof wherein:
R1 represents alkyl; alkenyl, wherein the double bond is not adjacent to the nitrogen; carbocyclyl; -Ci-6alkyl-carbocyclyl; heterocyclyl; -Ci-6alkyl-heterocyclyl; aryl; heteroaryl; -Ci-6alkylaryl; -Ci-6alkylheteroaryl; -phenyl fused to carbocyclyl or -phenyl fused to heterocyclyl; in which any of the aforesaid carbocyclyl and heterocyclyl groups may optionally be substituted by one or more groups selected from methyl and oxo; and in which any of the aforesaid phenyl, aryl and heteroaryl groups may optionally be substituted by one or more substituents selected from Ci-6alkyl, C2-6alkenyl, C2- 6alkynyl, Ci-6haloalkyl, -Ci-6thioalkyl, -SO2Ci.4alkyl, Ci-6alkoxy-, -O-Cs-scycloalkyl, C3- βcycloalkyl, -SO2C3-8cycloalkyl, C3-6alkenyloxy-, C3-6alkynyloxy-, -C(O)Ci-6alkyl, Ci- 6alkoxy-Ci-6alkyl-, nitro, halogen, cyano, hydroxyl, -C(O)OH, -NH2, -NHCi-4alkyl, -
N(Ci-4alkyl)(Ci-4alkyl), -C(O)N(Ci-4alkyl)(Ci-4alkyl), -C(O)NH2, -C(O)NH(Ci-4alkyl), -
C(O)OCi-6alkyl, -SOCi-4alkyl and -SOCs-ecycloalkyl; or R1 represents phenyl substituted by phenyl, or phenyl substituted by an optionally substituted monocyclic heteroaryl group in which any of the aforesaid phenyl and monocyclic heteroaryl groups may optionally be substituted by one or more groups selected from Ci-4alkyl, halogen and Ci-4alkoxy; or R1 represents phenyl substituted by benzyloxy- in which any of the aforesaid phenyl or benzyloxy groups may optionally be substituted on the ring by one or more groups selected from Ci-4alkyl, halogen and Ci-4alkoxy; and
A represents
Figure imgf000086_0001
wherein Y represents a C2-5 alkylene chain, which may optionally be substituted by one or two methyl groups or may optionally be substituted by two alkylene substituents at the same position wherein the two alkylene substituents are joined to each other to form a C3-5spiro-cycloalkyl group and
R2, R3 and R4 independently represent H or Ci-2alkyl, provided that R2 and R3 and R4 do not all represent H; or A represents
Figure imgf000086_0002
wherein Z represents a bond, -CH2-, -CH2-CH2-, -CH(Me)-, -CH(Me)-CH2- or -CH2- CH(Me)- and
R5 and R6 independently represent H or Ci-2alkyl and
B represents H or methyl.
2. A compound according to claim 1 wherein R1 represents alkyl; alkenyl, wherein the double bond is not adjacent to the nitrogen; carbocyclyl; -Ci-6alkyl-carbocyclyl; heterocyclyl; -Ci-6alkyl-heterocyclyl; aryl; heteroaryl; -Ci-6alkylaryl; -Ci- 6alkylheteroaryl; -phenyl fused to carbocyclyl or -phenyl fused to heterocyclyl; in which any of the aforesaid carbocyclyl and heterocyclyl groups may optionally be substituted by one or more groups selected from methyl and oxo; and in which any of the aforesaid phenyl, aryl and heteroaryl groups may optionally be substituted by one or more substituents selected from Ci-6alkyl, C2-6alkenyl, C2- 6alkynyl, Ci-6haloalkyl, -Ci-6thioalkyl, -SO2Ci-4alkyl, Ci-6alkoxy-, -O-C3-8cycloalkyl, C3- βcycloalkyl, -SO2C3-8cycloalkyl, C3-6alkenyloxy-, C3-6alkynyloxy-, -C(O)Ci-6alkyl, Ci- 6alkoxy-Ci-6alkyl-, nitro, halogen, cyano, hydroxyl, -C(O)OH, -NH2, -NHCi-4alkyl, - N(Ci-4alkyl)(Ci-4alkyl), -C(O)N(Ci-4alkyl)(Ci-4alkyl), -C(O)NH2 and -C(O)NH(Ci-4alkyl); or R1 represents phenyl substituted by phenyl, or phenyl substituted by an optionally substituted monocyclic heteroaryl group in which any of the aforesaid phenyl and monocyclic heteroaryl groups may optionally be substituted by one or more groups selected from Ci-4alkyl, halogen and Ci-4alkoxy.
3. A compound according to claim 1 wherein R1 represents aryl; heteroaryl; -phenyl fused to carbocyclyl; phenyl fused to heterocyclyl; or R1 represents phenyl substituted by phenyl, or phenyl substituted by a monocyclic heteroaryl group; in which any of aforementioned aryl, phenyl, heteroaryl, carbocyclyl and heterocyclyl may optionally be substituted.
4. A compound according to claim 1 wherein R1 represents optionally substituted aryl.
5. A compound according to claim 4 wherein R1 is phenyl substituted by one, two or three substituents selected from Ci-6alkyl, Ci-6alkoxy, Ci-6haloalkyl, halogen and cyano.
6. A compound according to claim 4 wherein R1 represents unsubstituted aryl selected from phenyl, naphthalen-1-yl and naphthalen-2-yl.
7. A compound according to claim 3 wherein R1 represents optionally substituted phenyl fused to optionally substituted heterocyclyl.
8. A compound according to claim 3 wherein R1 represents optionally substituted heteroaryl.
9. A compound according to any one of claims 1 to 8 wherein A represents
Figure imgf000087_0001
wherein Y, R3 and R4 are as defined in claim 1.
10. A compound according to claim 9 wherein R2 represents H,
R3 represents H and R4 represents methyl.
1 1. A compound according to claim 9 or claim 10 wherein Y represents an unsubstituted C2-5alkylene chain.
12. A compound according to claim 11 wherein Y represents -(CH2)3- or -(CH2),-.
13. A compound according to claim 12 wherein A represents
Figure imgf000088_0001
14. A compound according to any one of claims 1 to 8 wherein A represents
Figure imgf000088_0002
wherein R , R and Z are as defined in claim 1.
15. A compound according to claim 14 wherein R5 represents H and
R6 represents H.
16. A compound according to claim 14 claim 15 wherein Z represents a bond, -CH2- or -CH2-CH2-.
17. A compound according to any one of claims 1 to 16 wherein B represents H.
18. A compound according to claim 1 as defined in any one of examples 1 to 30:
(1 ) 2-cyano(4-ethylphenyl)-3-(3-(5-methyl-1 H-imidazol-1 -yl)propyl)guanidine,
(2) 2-cyano(4-isopropylphenyl)-3-(3-(5-methyl-1 H-imidazol-1 -yl)propyl)guanidine,
(3) 2-cyano(2,3-dihydrobenzo[b][1 ,4]dioxin-7-yl)-3-(3-(5-methyl-1 H-imidazol-1 - yl)propyl)guanidine,
(4) 2-cyano(4-cyanophenyl)-3-(3-(5-methyl-1 H-imidazol-1 -yl)propyl)guanidine,
(5) 2-cyano(3,4,5-trimethoxyphenyl)-3-(3-(5-methyl-1 H-imidazol-1 - yl)propyl)guanidine,
(6) 2-cyano(4-ethoxyphenyl)-3-(3-(5-methyl-1 H-imidazol-1 -yl)propyl)guanidine,
(7) 2-cyano(3-(5-methyl-1 H-imidazol-1 -yl)propyl)-3-(3,4-dimethylphenyl)guanidine,
(8) (3-(5-methyl-1 H-imidazol-1 -yl)propyl)-2-cyano-3-mesitylguanidine,
(9) (3-(5-methyl-1 H-imidazol-1 -yl)propyl)-2-cyano-3-(biphenyl-4-yl)guanidine,
(10) (3-(5-methyl-1 H-imidazol-1 -yl)propyl)-2-cyano-3-(naphthalen-2-yl)guanidine,
(1 1 ) (3-(5-methyl-1 H-imidazol-1 -yl)propyl)-2-cyano-3-(naphthalen-1 -yl)guanidine,
(12) (3-(5-methyl-1 H-imidazol-1 -yl)propyl)-3-(benzo[c][1 ,2,5]thiadiazol-6-yl)-2- cyanoguanidine, (13) (3-(5-methyl-1 H-imidazol-1 -yl)propyl)-3-(3,4-dichlorophenyl)-2-cyanoguanidine,
(14) (benzo[d][1 ,3]dioxol-6-yl)-2-cyano-3-(3-(5-methyl-1 H-imidazol-1 - yl)propyl)guanidine,
(15) 2-cyano(4-methoxyphenyl)-3-(3-(5-methyl-1 H-imidazol-1 -yl)propyl)guanidine,
(16) 2-cyano(3,5-dimethoxyphenyl)-3-(3-(5-methyl-1 H-imidazol-1 -yl)propyl)guanidine,
(17) 2-cyano(4-ethoxyphenyl)-3-(3-(4-methyl-1 H-imidazol-1 -yl)propyl)guanidine,
(18) 2-cyano(3,5-dimethoxyphenyl)-3-(3-(4-methyl-1 H-imidazol-1 -yl)propyl)guanidine,
(19) 2-cyano(2,3-dihydrobenzo[b][1 ,4]dioxin-7-yl)-3-(3-(4-methyl-1 H-imidazol-1 - yl)propyl)guanidine,
(20) 2-cyano(mesityl)-3-(3-(4-methyl-1 H-imidazol-1 -yl)propyl)guanidine,
(21 ) 2-cyano(4-isopropylphenyl)-3-(3-(4-methyl-1 H-imidazol-1 -yl)propyl)guanidine,
(22) 2-cyano(4-ethylphenyl)-3-(3-(4-methyl-1 H-imidazol-1 -yl)propyl)guanidine,
(23) 2-cyano(3-(4-methyl-1 H-imidazol-1 -yl)propyl)-3-(naphthalen-1 -yl)guanidine,
(24) (benzo[c][1 ,2,5]thiadiazol-6-yl)-2-cyano-3-(3-(4-methyl-1 H-imidazol-1 - yl)propyl)guanidine,
(25) 2-cyano(3,4,5-trimethoxyphenyl)-3-(3-(4-methyl-1 H-imidazol-1 -yl)propyl) guanidine,
(26) 2-cyano(4-cyanophenyl)-3-(3-(4-methyl-1 H-imidazol-1 -yl)propyl) guanidine,
(27) (3,4-dichlorophenyl)-2-cyano-3-(3-(4-methyl-1 H-imidazol-1 -yl)propyl) guanidine,
(28) 2-cyano(4-methoxyphenyl)-3-(3-(4-methyl-1 H-imidazol-1 -yl)propyl) guanidine,
(29) 2-cyano-1 -[3-(4-methyl-1 H-imidazol-1 -yl)propyl]-4-phenylbenzene-1 -guanidine,
(30) 2-cyano(3-(4-methyl-1 H-imidazol-1 -yl)propyl)-3-(naphthalen-2-yl)guanidine or a pharmaceutically acceptable salt, solvate or polymorph of any one thereof.
19. A compound according to claims 1 to 18 for use as a medicament.
20. A pharmaceutical composition comprising a compound according to any one of claims 1 to 18 optionally in combination with one or more therapeutically acceptable diluents or carriers.
21. The pharmaceutical composition of claim 20, which comprises additionally at least one compound, selected from the group consisting of neuroprotectants, antiparkinsonian drugs, amyloid protein deposition inhibitors, beta amyloid synthesis inhibitors, antidepressants, anxiolytic drugs, antipsychotic drugs and anti-multiple sclerosis drugs.
22. The pharmaceutical composition of claim 20 or 21 , which comprises additionally at least one compound, selected from the group consisting of PEP-inhibitors, LiCI, inhibitors of inhibitors of DP IV or DP IV-like enzymes, acetylcholinesterase (ACE) inhibitors, PIMT enhancers, inhibitors of beta secretases, inhibitors of gamma secretases, inhibitors of neutral endopeptidase, inhibitors of Phosphodiesterase-4 (PDE-4), TNFalpha inhibitors, muscarinic M1 receptor antagonists, NMDA receptor antagonists, sigma-1 receptor inhibitors, histamine H3 antagonists, immunomodulatory agents, immunosuppressive agents or an agent selected from the group consisting of antegren (natalizumab), Neurelan (fampridine-SR), campath (alemtuzumab), IR 208, NBI 5788/MSP 771 (tiplimotide), paclitaxel, Anergix.MS (AG 284), SH636, Differin (CD 271 , adapalene), BAY 361677 (interleukin-4), matrix- metalloproteinase-inhibitors, interferon-tau (trophoblastin) and SAIK-MS.
23. A compound according to any one of claims 1 to 18 or a pharmaceutical composition according to any one of claims 20 to 22 for use in the treatment of a disease selected from the group consisting of Kennedy's disease, duodenal cancer with or w/o Helicobacter pylori infections, colorectal cancer, Zolliger-Ellison syndrome, gastric cancer with or without Helicobacter pylori infections, pathogenic psychotic conditions, schizophrenia, infertility, neoplasia, inflammatory host responses, cancer, malign metastasis, melanoma, psoriasis, impaired humoral and cell-mediated immune responses, leukocyte adhesion and migration processes in the endothelium, impaired food intake, impaired sleep-wakefulness, impaired homeostatic regulation of energy metabolism, impaired autonomic function, impaired hormonal balance or impaired regulation of body fluids, multiple sclerosis, the Guillain-Barre syndrome and chronic inflammatory demyelinizing polyradiculoneuropathy.
24. A compound according to any one of claims 1 to18 or a pharmaceutical composition according to any one of claims 20 to 22 for use in the treatment of a disease selected from the group consisting of mild cognitive impairment, Alzheimer's disease, Familial British Dementia, Familial Danish Dementia, neurodegeneration in Down Syndrome, Parkinson's disease and Huntington's disease.
25. A compound according to any one of claims 1 to 18 or a pharmaceutical composition according to any one of claims 20 to 22 for use in the treatment of a disease selected from the group consisiting of rheumatoid arthritis, atherosclerosis, pancreatitis and restenosis.
26. A method of treatment or prevention of a disease selected from the group consisting of Kennedy's disease, ulcer disease, duodenal cancer with or w/o Helicobacter pylori infections, colorectal cancer, Zolliger-Ellison syndrome, gastric cancer with or without Helicobacter pylori infections, pathogenic psychotic conditions, schizophrenia, infertility, neoplasia, inflammatory host responses, cancer, malign metastasis, melanoma, psoriasis, impaired humoral and cell-mediated immune responses, leukocyte adhesion and migration processes in the endothelium, impaired food intake, impaired sleep-wakefulness, impaired homeostatic regulation of energy metabolism, impaired autonomic function, impaired hormonal balance or impaired regulation of body fluids, multiple sclerosis, the Guillain-Barre syndrome and chronic inflammatory demyelinizing polyradiculoneuropathy, which comprises administering to a subject an effective amount of a compound according to any one of claims 1 to 18 or a pharmaceutical composition according to any one of claims 20 to 22.
27. A method of treatment or prevention of a disease selected from the group consisting of mild cognitive impairment, Alzheimer's disease, Familial British Dementia, Familial Danish Dementia, neurodegeneration in Down Syndrome, Parkinson's disease and Huntington's disease, which comprises administering to a subject an effective amount of a compound according to any one of claims 1 to 18 or a pharmaceutical composition according to any one of claims 20 to 22.
28. A method of treatment or prevention of a disease selected from the group consisting of rheumatoid arthritis, atherosclerosis, pancreatitis and restenosis, which comprises administering to a subject an effective amount of a compound according to any one of claims 1 to 18 or a pharmaceutical composition according to any one of claims 20 to 22.
29. Use of a compound according to any one of claims 1 to 18 in the manufacture of a medicament for the treatment of a disease selected from the group consisting of Kennedy's disease, ulcer disease, duodenal cancer with or w/o Helicobacter pylori infections, colorectal cancer, Zolliger-Ellison syndrome, gastric cancer with or without Helicobacter pylori infections, pathogenic psychotic conditions, schizophrenia, infertility, neoplasia, inflammatory host responses, cancer, malign metastasis, melanoma, psoriasis, impaired humoral and cell-mediated immune responses, leukocyte adhesion and migration processes in the endothelium, impaired food intake, impaired sleep-wakefulness, impaired homeostatic regulation of energy metabolism, impaired autonomic function, impaired hormonal balance or impaired regulation of body fluids, multiple sclerosis, the Guillain-Barre syndrome and chronic inflammatory demyelinizing polyradiculoneuropathy.
30. Use of a compound according to any one of claims 1 to 18 in the manufacture of a medicament for the treatment of a disease selected from the group consisting of Alzheimer's disease, Familial British Dementia, Familial Danish Dementia, neurodegeneration in Down Syndrome, Parkinson's disease and Huntington's disease.
31. Use of a compound according to any one of claims 1 to 18 in the manufacture of a medicament for the treatment of a disease selected from the group consisting of rheumatoid arthritis, atherosclerosis, pancreatitis and restenosis.
32. A process for preparation of a compound of formula (I) according to any one of claims 1 to 18, which comprises reaction of a compound of formula (II)
Figure imgf000092_0001
with a compound of formula (III).
Figure imgf000092_0003
wherein R1 , A and B are as defined in any one of claims 1 to 18.
33. A process for preparation of a compound of formula (I) according to any one of claims 1 to 18, which comprises reaction of a compound of formula (Vl)
Figure imgf000092_0002
wherein R7 represents Ci-6alkyl and R1 is as defined in any one of claims 1 to 18 with a compound of formula (III).
Figure imgf000092_0004
wherein A and B are as defined in any one of claims 1 to 18.
PCT/EP2008/054712 2007-04-18 2008-04-18 Cyano-guanidine derivatives as glutaminyl cyclase inhibitors WO2008128983A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
JP2010503512A JP5675342B2 (en) 2007-04-18 2008-04-18 Cyanoguanidine derivatives as glutaminyl cyclase inhibitors
DK08736361.0T DK2160380T3 (en) 2007-04-18 2008-04-18 Cyano-guanidine derivatives as glutaminyl cyclase inhibitors
ES08736361.0T ES2474693T3 (en) 2007-04-18 2008-04-18 Cyano-guanidine derivatives as glutaminyl cyclase inhibitors
EP08736361.0A EP2160380B1 (en) 2007-04-18 2008-04-18 Cyano-guanidine derivatives as glutaminyl cyclase inhibitors

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US91252807P 2007-04-18 2007-04-18
US60/912,528 2007-04-18

Publications (1)

Publication Number Publication Date
WO2008128983A1 true WO2008128983A1 (en) 2008-10-30

Family

ID=39639502

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2008/054712 WO2008128983A1 (en) 2007-04-18 2008-04-18 Cyano-guanidine derivatives as glutaminyl cyclase inhibitors

Country Status (6)

Country Link
US (1) US8227498B2 (en)
EP (1) EP2160380B1 (en)
JP (1) JP5675342B2 (en)
DK (1) DK2160380T3 (en)
ES (1) ES2474693T3 (en)
WO (1) WO2008128983A1 (en)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011029920A1 (en) 2009-09-11 2011-03-17 Probiodrug Ag Heterocylcic derivatives as inhibitors of glutaminyl cyclase
WO2011076854A1 (en) 2009-12-22 2011-06-30 Probiodrug Ag CLEAVAGE OF β-AMYLOID PRECURSOR PROTEIN
WO2011101433A1 (en) 2010-02-18 2011-08-25 Probiodrug Ag Methods of diagnosing inflammatory diseases by determining pyroglutamate-modified mcp-1 and screening methods for inhibitors of glutaminyl cyclase
WO2012123562A1 (en) 2011-03-16 2012-09-20 Probiodrug Ag Diagnostic antibody assay
WO2013024043A3 (en) * 2011-08-12 2013-06-06 Probiodrug Ag In vivo screening models for treatment of qc-related disorders
WO2016097305A1 (en) 2014-12-19 2016-06-23 Probiodrug Ag Novel method for the detection of pglu-abeta peptides
EP3047854A1 (en) 2008-07-21 2016-07-27 Probiodrug AG Diagnostic antibody assay
WO2019022600A1 (en) 2017-07-24 2019-01-31 Stichting Het Nederlands Kanker Instituut-Antoni van Leeuwenhoek Ziekenhuis Treating pathological conditions by direct and indirect targeting of sirpa - cd47 interaction
EP3461819A1 (en) 2017-09-29 2019-04-03 Probiodrug AG Inhibitors of glutaminyl cyclase
EP3521308A1 (en) 2018-01-31 2019-08-07 Probiodrug AG Humanized and de-immunized antibodies

Families Citing this family (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7732162B2 (en) * 2003-05-05 2010-06-08 Probiodrug Ag Inhibitors of glutaminyl cyclase for treating neurodegenerative diseases
JP5676249B2 (en) * 2007-04-20 2015-02-25 プロビオドルグ エージー Aminopyridine derivatives as glutaminyl cyclase inhibitors
CN101802000A (en) * 2007-09-12 2010-08-11 前体生物药物股份公司 transgenic mice
CA2710494A1 (en) * 2008-01-14 2009-07-23 Probiodrug Ag Mouse models carrying a knock-out mutation of the glutaminyl cyclase gene
WO2014015157A2 (en) 2012-07-19 2014-01-23 Philadelphia Health & Education Corporation Novel sigma receptor ligands and methods of modulating cellular protein homeostasis using same
WO2019089902A1 (en) 2017-11-01 2019-05-09 Drexel University Compounds, compositions, and methods for treating diseases
JPWO2022210189A1 (en) * 2021-03-30 2022-10-06

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998050344A1 (en) * 1997-05-07 1998-11-12 Novo Nordisk A/S Substituted 3,3-diamino-2-propenenitriles, their preparation and use
WO2004098591A2 (en) * 2003-05-05 2004-11-18 Probiodrug Ag Inhibitors of glutaminyl cyclase and their use in the treatment of neurological diseases
WO2005075436A2 (en) * 2004-02-05 2005-08-18 Probiodrug Ag Novel inhibitors of glutaminyl cyclase

Family Cites Families (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE3044566A1 (en) * 1980-11-26 1982-07-15 Basf Ag, 6700 Ludwigshafen N-Substd. imidazole derivs. - selective histamine-H2 antagonists, e.g. for treating stomach and duodenal ulcers
US6948038B2 (en) * 2001-07-24 2005-09-20 Microsoft Corporation System and method for backing up and restoring data
US8338120B2 (en) * 2003-05-05 2012-12-25 Probiodrug Ag Method of treating inflammation with glutaminyl cyclase inhibitors
CN101837127A (en) * 2003-05-05 2010-09-22 前体生物药物股份公司 The application of glutaminyl and glutamate cyclase effector
US7732162B2 (en) * 2003-05-05 2010-06-08 Probiodrug Ag Inhibitors of glutaminyl cyclase for treating neurodegenerative diseases
JP5707014B2 (en) * 2003-10-15 2015-04-22 プロビオドルグ エージー Use of glutaminyl and glutamate cyclase effectors
JP2007509898A (en) * 2003-11-03 2007-04-19 プロビオドルグ エージー Useful combinations for the treatment of neurological disorders
US7667044B2 (en) * 2003-11-03 2010-02-23 Probiodrug Ag Compounds for the treatment of neurological disorders
US20050171112A1 (en) * 2003-11-03 2005-08-04 Probiodrug Ag Combinations useful for the treatment of neuronal disorders
US20080249083A1 (en) * 2006-09-21 2008-10-09 Probiodrug Ag Novel genes related to glutaminyl cyclase
DK2086960T3 (en) * 2006-11-09 2014-06-10 Probiodrug Ag NEW INHIBITORS OF GLUTAMINYLYCYCLASE.
US8420684B2 (en) * 2006-11-09 2013-04-16 Probiodrug Ag Inhibitors of glutaminyl cyclase
US9126987B2 (en) * 2006-11-30 2015-09-08 Probiodrug Ag Inhibitors of glutaminyl cyclase
JP2010516235A (en) * 2007-01-19 2010-05-20 プロビオドルグ エージー In vivo screening model for the treatment of Alzheimer's disease and other QPCT related disorders
EP2118101B1 (en) * 2007-03-09 2012-09-26 Probiodrug AG Imidazo [1,5-a] pyridine derivatives as inhibitors of glutaminyl cyclase

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998050344A1 (en) * 1997-05-07 1998-11-12 Novo Nordisk A/S Substituted 3,3-diamino-2-propenenitriles, their preparation and use
WO2004098591A2 (en) * 2003-05-05 2004-11-18 Probiodrug Ag Inhibitors of glutaminyl cyclase and their use in the treatment of neurological diseases
WO2005075436A2 (en) * 2004-02-05 2005-08-18 Probiodrug Ag Novel inhibitors of glutaminyl cyclase

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
BUCHHOLZ MIRKO ET AL: "The first potent inhibitors for human glutaminyl cyclase: Synthesis and structure-activity relationship", JOURNAL OF MEDICINAL CHEMISTRY, USAMERICAN CHEMICAL SOCIETY. WASHINGTON, vol. 49, no. 2, 1 January 2006 (2006-01-01), pages 664 - 677, XP002462493, ISSN: 0022-2623 *

Cited By (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3338795A1 (en) 2008-07-21 2018-06-27 Probiodrug AG Diagnostic antibody assay
EP3338796A1 (en) 2008-07-21 2018-06-27 Probiodrug AG Diagnostic antibody assay
EP3047854A1 (en) 2008-07-21 2016-07-27 Probiodrug AG Diagnostic antibody assay
WO2011029920A1 (en) 2009-09-11 2011-03-17 Probiodrug Ag Heterocylcic derivatives as inhibitors of glutaminyl cyclase
WO2011076854A1 (en) 2009-12-22 2011-06-30 Probiodrug Ag CLEAVAGE OF β-AMYLOID PRECURSOR PROTEIN
WO2011101433A1 (en) 2010-02-18 2011-08-25 Probiodrug Ag Methods of diagnosing inflammatory diseases by determining pyroglutamate-modified mcp-1 and screening methods for inhibitors of glutaminyl cyclase
WO2012123562A1 (en) 2011-03-16 2012-09-20 Probiodrug Ag Diagnostic antibody assay
US8809508B2 (en) 2011-03-16 2014-08-19 Probiodrug Ag Diagnostic antibody assay
US9657089B2 (en) 2011-03-16 2017-05-23 Probiodrug Ag Diagnostic antibody assay
WO2013024043A3 (en) * 2011-08-12 2013-06-06 Probiodrug Ag In vivo screening models for treatment of qc-related disorders
WO2016097305A1 (en) 2014-12-19 2016-06-23 Probiodrug Ag Novel method for the detection of pglu-abeta peptides
WO2019022600A1 (en) 2017-07-24 2019-01-31 Stichting Het Nederlands Kanker Instituut-Antoni van Leeuwenhoek Ziekenhuis Treating pathological conditions by direct and indirect targeting of sirpa - cd47 interaction
EP3747438A1 (en) 2017-07-24 2020-12-09 Stichting Het Nederlands Kanker Instituut- Antoni van Leeuwenhoek Ziekenhuis Treating pathological conditions by direct and indirect targeting of sirpa - cd47 interaction
EP3747437A1 (en) 2017-07-24 2020-12-09 Stichting Het Nederlands Kanker Instituut- Antoni van Leeuwenhoek Ziekenhuis Treating pathological conditions by direct and indirect targeting of sirpa - cd47 interaction
EP3658141B1 (en) 2017-07-24 2022-11-16 Stichting Het Nederlands Kanker Instituut- Antoni van Leeuwenhoek Ziekenhuis Treating pathological conditions by direct and indirect targeting of sirpalpha - cd47 interaction
EP3461819A1 (en) 2017-09-29 2019-04-03 Probiodrug AG Inhibitors of glutaminyl cyclase
EP3521308A1 (en) 2018-01-31 2019-08-07 Probiodrug AG Humanized and de-immunized antibodies
WO2019149689A1 (en) 2018-01-31 2019-08-08 Probiodrug Ag Humanized and de-immunized antibodies

Also Published As

Publication number Publication date
JP5675342B2 (en) 2015-02-25
DK2160380T3 (en) 2014-06-30
JP2010524895A (en) 2010-07-22
US20080267912A1 (en) 2008-10-30
ES2474693T3 (en) 2014-07-09
EP2160380A1 (en) 2010-03-10
US8227498B2 (en) 2012-07-24
EP2160380B1 (en) 2014-04-02

Similar Documents

Publication Publication Date Title
EP2142514B1 (en) Thiourea derivatives as glutaminyl cyclase inhibitors
EP2142513B1 (en) Nitrovinyl-diamine derivatives as glutaminyl cyclase inhibitors
EP2160380B1 (en) Cyano-guanidine derivatives as glutaminyl cyclase inhibitors
EP2118101B1 (en) Imidazo [1,5-a] pyridine derivatives as inhibitors of glutaminyl cyclase
EP2142515B1 (en) Nitrovinyl-diamine derivatives as glutaminyl cyclase inhibitors
EP2160389B1 (en) Thioxoquinazolinone derivatives as glutaminyl cyclase inhibitors
EP2545047B9 (en) Heterocyclic inhibitors of glutaminyl cyclase (qc, ec 2.3.2.5)
US9512082B2 (en) Inhibitors of glutaminyl cyclase
EP2344157B1 (en) Novel inhibitors
EP2091948A1 (en) Novel inhibitors of glutaminyl cyclase
EP2142536A1 (en) Aminopyrimidine derivatives as glutaminyl cyclase inhibitors
EP2686313B1 (en) Benzimidazole derivatives as inhibitors of glutaminyl cyclase
EP2560953B1 (en) Inhibitors of glutaminyl cyclase

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 08736361

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2010503512

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2008736361

Country of ref document: EP