WO2011107530A2 - Novel inhibitors - Google Patents

Novel inhibitors Download PDF

Info

Publication number
WO2011107530A2
WO2011107530A2 PCT/EP2011/053154 EP2011053154W WO2011107530A2 WO 2011107530 A2 WO2011107530 A2 WO 2011107530A2 EP 2011053154 W EP2011053154 W EP 2011053154W WO 2011107530 A2 WO2011107530 A2 WO 2011107530A2
Authority
WO
WIPO (PCT)
Prior art keywords
compound
alkyl
formula
mmol
phenyl
Prior art date
Application number
PCT/EP2011/053154
Other languages
French (fr)
Other versions
WO2011107530A3 (en
Inventor
Ulrich Heiser
Daniel Ramsbeck
Torsten Hoffmann
Livia Boehme
Hans-Ulrich Demuth
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 JP2012555421A priority Critical patent/JP6026284B2/en
Priority to EP11707395.7A priority patent/EP2542549B1/en
Priority to ES11707395.7T priority patent/ES2586231T3/en
Publication of WO2011107530A2 publication Critical patent/WO2011107530A2/en
Publication of WO2011107530A3 publication Critical patent/WO2011107530A3/en

Links

Classifications

    • 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
    • C07D417/04Heterocyclic 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 directly linked by a ring-member-to-ring-member bond
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41841,3-Diazoles condensed with carbocyclic rings, e.g. benzimidazoles
    • 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
    • A61P25/18Antipsychotics, i.e. neuroleptics; Drugs for mania or schizophrenia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • 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
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • 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
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings directly linked by a ring-member-to-ring-member bond

Definitions

  • the invention relates to novel heterocyclic 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
  • Inhibitors of QC are described in WO 2004/098625, WO 2004/098591 , WO 2005/039548, WO 2005/075436, WO 2008/055945, WO 2008/055947, WO 2008/055950 and WO2008/065141 .
  • 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 " ⁇ and “K D” 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).
  • POP prolyl endopeptidase
  • POP-activity is defined as the catalytic activity of an endoprotease that is capable to hydrolyze post proline bonds in peptides or proteins where 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 1 10995), 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 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)
  • 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
  • the subject method and medical use utilize an agent with an I C 50 for QC inhibition of 10 ⁇ or less, more preferably of 1 ⁇ or less, even more preferably of 0.1 ⁇ or less or 0.01 ⁇ or less, or most preferably 0.001 ⁇ or less.
  • I ndeed, inhibitors with K, values in the lower micromolar, preferably the nanomolar and even more preferably the picomolar range are contemplated.
  • 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.
  • 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.
  • Ci -4 alkyl group 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).
  • 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
  • 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- i2 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 C2-12 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 C3-1 0 cycloalkyl group (i.e. 3 to 10 ring carbon atoms), more suitably a C 3- 8 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 C5-1 0 cycloalkenyl group (i .e. 5 to 1 0 ring carbon atoms), more suitably a C 5 -s 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.
  • heterocyclyl groups containing one hetero atom include pyrrolidine, tetrahydrofuran and piperidine, and exemplary heterocyclyl groups containing two hetero atoms include morpholine, piperazine, dioxolane and dioxane.
  • 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 -12 aryl group, suitably a C 6- io aryl group, more suitably a C 6- 8 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 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.
  • 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 Ci -4 alkylene moiety.
  • halogen or halo
  • fluorine F
  • chlorine CI
  • bromine Br
  • amino refers to the group -NH 2 .
  • phenyl substituted by phenyl refers to biphenyl.
  • ⁇ wr denotes a single bond where the stereochemistry is not defined.
  • benzimidazol l When benzimidazol l is shown as benzimidazol-5-yl, which is represented as:
  • 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.
  • 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 /V-methyl-D-glucamine.
  • crystalline forms of the compounds may exist as polymorphs and as such are intended to be included in the present invention.
  • 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. Summary of the invention
  • R 1 represents a heteroaryl group optionally substituted by one or more groups selected from Ci -6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, Ci -6 haloalkyl, -Ci -6 thioalkyl, -SOCi -4 alkyl, -S0 2 Ci -4 alkyl, Ci_ 6 alkoxy-, -0-C 3-8 cycloal kyl , C 3-8 cycloalkyl, -S0 2 C 3 - 8 cycloalkyl, -SOC 3-6 cycloalkyl, C 3- 6 alkenyloxy-, C 3-6 alkynyloxy-, -C(0)Ci -6 alkyl, -C(0)OCi -6 alkyl, Ci- 6 alkoxy-Ci- 6 alkyl-, nitro, halogen, cyano, -C(0)OH, -NHCi -4 alkyl, -N(C 1 _ 4 alkyl
  • carbocyclyl groups may optionally be substituted by one or more groups selected from Ci -4 alkyl, oxo, halogen and Ci -4 alkoxy;
  • Y represents a 5 membered heteroaryl group selected from triazolyl, oxadiazolyl, thiadiazolyl, thiazolyl, triazol-one or triazol-thione;
  • X represents a linker selected from -(CH 2 ) m -R 2 , -(CH 2 ) n -S-R 2 , -(CH 2 ) p -0-R 2 or -(CH 2 ) p -S0 2 - R 2 ;
  • n represents an integer selected from 1 to 4.
  • n an integer selected from 1 or 2;
  • p represents an integer selected from 0 to 2;
  • R 2 represents aryl, heteroaryl, carbocyclyl or heterocyclyl
  • any of aforesaid aryl and heteroaryl groups may optionally be substituted by one or more groups selected from Ci -6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, Ci -6 haloalkyl, -Ci_ 6thioalkyl, -SOCi -4 alkyl, -S0 2 Ci -4 alkyl , Ci -6 alkoxy-, -0-C 3-8 cycloalkyl, C 3-8 cycloalkyl, - S0 2 C 3-8 cycloalkyl, -SOC 3-6 cycloalkyl, C 3-6 alkenyloxy-, C 3-6 alkynyloxy-, -C(0)Ci -6 alkyl, - C(0)OCi -6 alkyl, Ci- 6 alkoxy-Ci- 6 alkyl-, nitro, halogen, haloCi -6 alkyl, haloCi -6 alkoxy, cyano,
  • any of aforesaid carbocyclyl and heterocyclyl groups may optionally be substituted by one or more groups selected from Ci -4 alkyl, oxo, halogen and Ci -4 alkoxy; or R 2 represents phenyl substituted by phenyl, phenyl substituted by a monocyclic heteroaryl group, phenyl substituted by phenoxy, phenyl substituted by heterocyclyl, phenyl substituted by -0-Ci -4 alkyl-heterocyclyl, phenyl substituted by benzyloxy, phenyl fused to carbocyclyl or phenyl fused to heterocyclyl, such that when Y represents oxadiazolyl and X represents CH 2 , R 2 represents a group other than phenyl substituted by phenoxy;
  • any of aforesaid phenyl, benzyloxy and heteroaryl groups may optionally be substituted by one or more groups selected from Ci -4 alkyl, halogen and Ci -4 alkoxy, and in which any of aforesaid carbocyclyl and heterocyclyl groups may optionally be substituted by one or more groups selected from methyl, phenyl, oxo, halogen and Ci_ 4alkoxy; with the proviso that the compound of formula (I) is a compound other than:
  • R 1 represents a heteroaryl group optionally substituted by one or more groups selected from
  • Ci -6 alkyl C 2-6 alkenyl, C 2-6 alkynyl, Ci -6 haloalkyl, -Ci -6 thioalkyl, -SOCi -4 alkyl, -S0 2 Ci -4 alkyl, Ci_ 6 alkoxy-, -0-C 3-8 cycloal kyl , C 3-8 cycloalkyl, -S0 2 C 3 - 8 cycloalkyl, -SOC 3-6 cycloalkyl, C 3-
  • carbocyclyl groups may optionally be substituted by one or more groups selected from Ci -4 alkyl, oxo, halogen and Ci -4 alkoxy;
  • Y represents a 5 membered heteroaryl group selected from triazolyl, oxadiazolyl, thiadiazolyl, thiazolyl, triazol-one or triazol-thione;
  • X represents a linker selected from -(CH 2 ) m -, -(CH 2 ) n -S-, -(CH 2 ) p -0- or -(CH 2 ) p -S0 2 -;
  • n represents an integer selected from 1 to 4.
  • n an integer selected from 1 or 2;
  • p represents an integer selected from 0 to 2;
  • R 2 represents aryl, heteroaryl, carbocyclyl or heterocyclyl
  • any of aforesaid aryl and heteroaryl groups may optionally be substituted by one or more groups selected from Ci -6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, Ci -6 haloalkyl, -Ci_ 6thioalkyl, -SOCi -4 alkyl, -S0 2 Ci -4 alkyl, Ci -6 alkoxy-, -0-C 3-8 cycloalkyl, C 3-8 cycloalkyl, -
  • any of aforesaid carbocyclyl and heterocyclyl groups may optionally be substituted by one or more groups selected from Ci -4 alkyl, oxo, halogen and Ci -4 alkoxy; or R 2 represents phenyl substituted by phenyl, phenyl substituted by a monocyclic heteroaryl group, phenyl substituted by phenoxy, phenyl substituted by heterocyclyl, phenyl substituted by -0-Ci -4 alkyl-heterocyclyl, phenyl substituted by benzyloxy, phenyl fused to carbocyclyl or phenyl fused to heterocyclyl, such that when Y represents oxadiazolyl and X represents CH 2 , R 2 represents a group other than phenyl substituted by phenoxy;
  • any of aforesaid phenyl, benzyloxy and heteroaryl groups may optionally be substituted by one or more groups selected from Ci -4 alkyl, halogen and Ci -4 alkoxy, and in which any of aforesaid carbocyclyl and heterocyclyl groups may optionally be substituted by one or more groups selected from methyl, phenyl, oxo, halogen and Ci_ 4alkoxy;
  • 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. 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.
  • -S0 2 cyclohexyl cyclohexyl
  • -SOC 3- 6 cycloalkyl e.g. -SOcyclopropyl
  • C 3-6 alkenyloxy- e.g. -O-buten-2-yl
  • C 3-6 alkynyloxy- e.g. - O-buten-2-yl
  • -C(0)Ci -6 alkyl e.g. -C(O)ethyl
  • -C(0)OCi -6 alkyl e.g. -C(O)O-methyl
  • Ci_ 6 alkoxy-Ci- 6 alkyl- e.g. methoxy-ethyl-
  • nitro, halogen e.g.
  • 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 heteroaryl
  • examples include monocyclic (e.g. 5 and 6 membered) and bicyclic (e.g. 9 and 10 membered, particularly 9 membered) heteroaryl rings, especially rings containing nitrogen atoms (e.g. 1 or 2 nitrogen atoms).
  • a suitable bicyclic heteroaryl ring is a 9-membered heteroaryl ring containing 1 or 2 nitrogen atoms, especially a benzene ring fused to a 5-membered ring containing one or two nitrogen atoms (e.g. 1 H-benzoimidazolyl). Most suitably the point of attachment is through a benzene ring, e.g. the group is 1 H- benzoimidazol-5-yl.
  • heteroaryl groups may either be unsubstituted (which is more typical) or may suitably be substituted by one or more (e.g. 1 or 2) substituents selected from alkyl (e.g. Ci- alkyl such as Me), alkoxy- (e.g. Ci- alkoxy- such as OMe) and halogen (e.g. F).
  • alkyl e.g. Ci- alkyl such as Me
  • alkoxy- e.g. Ci- alkoxy- such as OMe
  • halogen e.g. F
  • R 1 heteroaryl groups include a 5-membered ring containing 2 or 3 nitrogen atoms, which ring may optionally be substituted (e.g. in particular by one or two groups, such as methyl, for example:
  • R 1 heteroaryl groups include a 9-membered bicyclic ring containing 2 nitrogen atoms, which ring may optionally be substituted, for example:
  • examples include:
  • examples include:
  • an example includes:
  • examples include:
  • aryl may typically represent phenyl.
  • exemplary substituted phenyl groups include 3-methylphenyl-, 2,3-dichlorophenyl-, 2,3- difluorophenyl-, 2,4-dichlorophenyl-, 2,4-difluororophenyl-, 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-, 2,6-difluoro-4- (methoxy)phenyl-, 2-isopropyl-6-methylphenyl-, 3-(cyclopentyloxy)-4-methoxyphenyl-, 3,4,5- trimethoxyphenyl-, 3,
  • R 2 may represent unsubstituted phenyl-.
  • substituted phenyl groups include 2,3,4- trifluorophenyl, 2,3-difluoro-4-methylphenyl, 2-bromo-4-fluorophenyl-, 2-bromo-5- fluorophenyl-, 2-chlorophenyl-, 2-fluorophenyl-, 2-fluoro-5-(trifluoromethyl)phenyl-, 2-hydroxy-
  • R 2 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-
  • R 2 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. pyridin-2-yl and pyridin-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-yk
  • 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.
  • 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 2 represents carbocyclyl
  • examples include cycloalkyi and cycloalkenyl.
  • Examples of cycloalkyi 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, 3-methyl-cyclohex-3-enyl, 3-methyl-cyclohex-3-enyl, 3-methyl-cyclohex-3-enyl.
  • R 2 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 2 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.
  • R 2 represents phenyl substituted by phenyl in which any of aforesaid phenyl groups may optionally be substituted, examples include -biphenyl-4-yl.
  • R 2 represents phenyl substituted by a monocyclic heteroaryl group, in which any of aforesaid phenyl and heteroaryl groups may optionally be substituted, examples include 4- (oxazol-5-yl)phenyk
  • R 2 represents phenyl substituted by benzyloxy in which any of aforesaid phenyl and benzyloxy groups may optionally be substituted
  • examples include 4-benzyloxy-phenyl-, 4-(3- methylbenzyloxy)phenyl- and 4-(4-methylbenzyloxy)phenyk
  • R 2 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 2 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-yk
  • R 1 represents bicyclic heteroaryl, especially 9-membered bicyclic heteroaryl. More suitably, R 1 represents a bicyclic heteroaryl ring system and in particular a phenyl ring fused with a 5 membered heteroaryl ring containing one or more (e.g. one or two, suitably one, more suitably two) nitrogen atoms or a pyridine ring fused with a 5-membered heteroaryl ring containing one or more (e.g. one or two, suitably one, more suitably two) nitrogen atoms.
  • R 1 represents bicyclic heteroaryl, preferably the heteroaryl group does not contain S atoms.
  • R 1 represents a phenyl ring fused to a 5-membered heteroaryl ring, preferably R 1 is linked to the core of formula (I) through the phenyl ring.
  • R 1 represents a pyridine ring fused to a 5-membered heteroaryl ring, preferably R 1 is linked to the core of formula (I) through the pyridine ring.
  • R 1 represents unsubstituted heteroaryl.
  • R 1 suitably represents 1 H-benzoimidazolyl or imidazo[1 ,2-a]pyridine, particularly 1 H- benzoimidazolyl, especially 1 H-benzoimidazol-5-yl.
  • R 1 represents
  • A represents a bond and R 11 , R 12 and R 13 independently represent H or Ci -2 alkyl.
  • R 1 represents
  • R 14 and R 15 independently represent H or Ci -2 alkyl.
  • R 1 represents
  • C represents a bond and R 16 and R 17 independently represent H or Ci -2 alkyl.
  • R 1 represents
  • R 14 represents H and R 15 represents H .
  • R 14 represents H and R 15 represents Ci -2 alkyl.
  • R 14 represents Ci -2 alkyl and R 15 represents H.
  • R 14 represents methyl and R 15 represents H.
  • B represents a bond
  • R 11 suitably represents H
  • R 12 suitably represents H or methyl.
  • R 13 suitably represents H or methyl.
  • R 12 represents H and R 13 represents methyl. In another embodiment, R 12 represents methyl and R 13 represents H . I n a third embodiment, R 12 represents H and R 13 represents H.
  • A represents a bond
  • R 1 represents
  • R 16 represents H and R 17 represents H . In another embodiment R 16 represents H and R 17 represents Ci -2 alkyl. In a third embodiment R 16 represents Ci -2 alkyl and R 17 represents H.
  • C represents a bond
  • R 18 represents H and R 19 represents H .
  • R 18 represents H and R 19 represents Ci -2 alkyl.
  • R 18 represents Ci -2 alkyl and R 19 represents H.
  • D represents a bond
  • R 1 represents
  • Y represents triazolyl, oxadiazolyl or thiadiazolyl. More suitably, Y represents triazolyl or thiadiazolyl. In a further embodiment, Y is attached to R 1 via a carbon atom.
  • Y represents triazolyl
  • Y represents oxadiazolyl.
  • Y represents oxadiazolyl, such 1 ,3,4-oxadiazolyl, i.e.
  • Y represents thiadiazolyl.
  • Y represents thiadiazolyl, such as: 1 ,3,4-thiadiazolyl, i.e.
  • Y represents thiazolyl.
  • Y represents thiazolyl, such as:
  • Y represents triazol-one, such as:
  • Y represents triazol-thione, such as:
  • X represents a linker selected from -(CH 2 ) m -R 2 , -(CH 2 ) P -S-R 2 , -(CH 2 ) P -0-R 2 or - (CH 2 ) p -S0 2 -R 2 .
  • X represents a linker selected from -(CH 2 ) m -R 2 and -(CH 2 ) P -S-R 2 (i.e. -CH 2 - R 2 , (CH 2 ) 2 -R 2 or -CH 2 -S-R 2 ).
  • X represents -(CH 2 ) m -R 2 .
  • X represents -CH 2 -R 2 , -(CH 2 ) 2 -R 2 or -(CH 2 ) 3 -R 2 .
  • X represents -CH 2 -R 2 or -(CH 2 ) 2 -R 2 .
  • X represents -CH 2 -R 2 .
  • X represents -(CH 2 ) 2 -R 2 .
  • X represents -(CH 2 ) n -S-R 2 .
  • X represents -CH 2 -S-R 2 .
  • X represents -(CH 2 ) p -0-R 2 .
  • X represents -CH 2 -0-R 2 .
  • X represents -(CH 2 ) p -S0 2 -R 2 .
  • X represents -CH 2 -S0 2 -R 2 .
  • m represents an integer selected from 1 to 3. In a further embodiment, m represents an integer selected from 1 or 2.
  • n represents 1 .
  • p represents an integer selected from 0 or 1 . In a further embodiment, p represents 1 .
  • R 2 represents aryl, heteroaryl, carbocyclyl, heterocyclyl, phenyl substituted by phenoxy or phenyl fused to heterocyclyl. More suitably, R 2 represents aryl, heteroaryl, phenyl substituted by phenoxy or phenyl fused to heterocyclyl. Yet more suitably, R 2 represents optionally substituted aryl or phenyl substituted by phenoxy. Still yet more suitably, R 2 represents optionally substituted aryl.
  • R 2 represents carbocyclyl.
  • R 2 suitably represents cyclohexyl.
  • R 2 represents optionally substituted aryl.
  • R 2 suitably represents optionally substituted phenyl or napthyl. More suitably, R 2 represents optionally substituted phenyl.
  • R 2 represents phenyl optionally substituted by one or more groups selected from Ci -6 alkyl (e.g. methyl), Ci -6 alkoxy (e.g. methoxy or ethoxy), -Ci -6 thioalkyl (e.g. methylthio), haloCi -6 alkyl (e.g. trifluoromethyl), haloCi -6 alkoxy (e.g. trifluoromethoxy) or halogen (e.g. chlorine or fluorine).
  • R 2 represents phenyl optionally substituted by one or more groups selected from Ci -6 alkoxy (e.g. methoxy) or halogen (e.g. chlorine).
  • R 2 represents optionally substituted phenyl
  • R 2 suitably represents 2-methylphenyl, 4- methylphenyl, 4-trifluoromethylphenyl, 5-trifluoromethylphenyl, 2,5-bis(trifluoromethyl), 4-methylthio(phenyl), 2-methoxyphenyl, 3-methoxyphenyl, 4-methoxyphenyl, 2,3- dimethoxyphenyl, 2,4-dimethoxyphenyl, 2,5-dimethoxyphenyl, 3,4-dimethoxyphenyl, 3,4,5- trimethoxyphenyl, 4-ethoxyphenyl, 2-fluorophenyl, 3-fluorophenyl, 4-fluorophenyl, 2,3- difluorophenyl, 2,4-difluorophenyl, 3,4-difluorophenyl, 2-chlorophenyl, 3-chlorophenyl, 4- chlorophenyl, 2,3-dichlorophenyl, 2,4-
  • R 2 represents optionally substituted heteroaryl.
  • R 2 suitably represents optionally substituted pyridyl.
  • R 2 represents unsubstituted pyridyl, e.g. pyridin-4-yl.
  • R 2 represents phenyl substituted by phenyl, the aforesaid phenyl groups optionally being substituted.
  • R 2 suitably represents phenyl substituted by 3- phenyl, phenyl substituted by 4-phenyl, phenyl substituted by 3-(3-chlorophenyl), phenyl substituted by 4-(3-chlorophenyl), phenyl substituted by 4-(3,4-dichlorophenyl) or 3- fluorophenyl substituted by 4-phenyl.
  • R 2 represents optionally substituted phenyl substituted by optionally su bstituted phenoxy.
  • R 2 represents optionally substituted phenyl substituted by optionally substituted phenoxy
  • R 2 suitably represents phenyl substituted by 4-phenoxy.
  • R 2 represents optionally substituted phenyl substituted by optionally substituted heterocyclyl.
  • R 2 suitably represents 3-chlorophenyl substituted by 4- morpholinyl, phenyl substituted by 4-piperazinyl substituted by 4N-methyl, phenyl substituted by 4-piperazinyl substituted by 4N-phenyl, phenyl substituted by 3-piperazinyl substituted by 4N-phenyl or 2-chlorophenyl substituted by 6-piperazinyl substituted by 4N-ethyl.
  • R 2 represents optionally substituted phenyl substituted by -0-C-i -4 alkyl- heterocyclyl.
  • R 2 represents optionally substituted phenyl substituted by -0-Ci -4 alkyl- heterocyclyl
  • R 2 suitably represents phenyl substituted by 4-0-(CH 2 )2-morpholinyl, 4-0- (CH 2 )3-morpholinyl, 2-0-(CH 2 )2-morpholinyl or 4-0-(CH 2 )2-piperazinyl.
  • R 2 represents optionally substituted phenyl fused to optionally substituted heterocyclyl.
  • R 2 when R 2 represents optionally substituted phenyl fused to optionally substituted heterocyclyl, R 2 suitably represents benzo- 1 ,3-dioxolanyl.
  • Y represents triazolyl or thiadiazolyl
  • X represents -(CH 2 ) m - and -(CH 2 ) P -S- (i.e. -CH 2 -, (CH 2 )2- or -CH 2 -S-)
  • R 2 represents phenyl optionally substituted by one or more groups selected from Ci -6 alkoxy (e.g. methoxy) or halogen (e.g. chlorine) or R 2 represents phenyl substituted by phenoxy.
  • Process (a) typically comprises dissolving a compound of formula (II) in a suitable solvent, such as formic acid and formic acid orthoethylester, followed by treatment with sodium formate and Pd/C and heating to 1 1 0°C for 24 h.
  • a suitable solvent such as formic acid and formic acid orthoethylester
  • Process (b) typically comprises heating to 1 50 °C overnight.
  • a non-limiting example of the methodology of process (b) is described in Section A, Method 1 herein.
  • Process (d) typically comprises dissolving the compound of formula X-COOH in DMF, followed by treatment with carbonyldiimidazole and stirred at room temperature for 1 hour. The compound of formula (V) is then added followed by heating at 1 10 °C overnight.
  • Process (d) is described in Section B herein.
  • Process (f) typically comprises reaction in the presence of acetonitrile, Lawesson's reagent and POCI 3 followed by heating to reflux overnight.
  • acetonitrile Lawesson's reagent and POCI 3 followed by heating to reflux overnight.
  • a non-limiting example of the methodology of process (f) is described in Section C, Method 3 herein.
  • Process (g) typically comprises reaction in the presence of DMF and carbonyldiimidazole followed by heating to 1 10 °C for 24 hours.
  • a non-limiting example of the methodology of process (g) is described in Section D herein.
  • Process (h) preparing a compound of formula (I) wherein Y represents thiazolyl by reacting a compound of formula R 1 -COOH with a compound of formula X-CO-CH 2 -NH 2 , wherein R 1 and X are as defined above for compounds of formula (I ).
  • Process (h) typically comprises reaction in the presence of acetonitrile, Lawesson's reagent, TEA and POCI 3 followed by heating to reflux overnight.
  • acetonitrile, Lawesson's reagent, TEA and POCI 3 followed by heating to reflux overnight.
  • Process (i) typically comprises the use of 5% Na 2 C0 3 followed by heating to reflux for 3 hours.
  • a non-limiting example of the methodology of process (i) is described in Section G herein. interconversion of compounds of formula (I); and deprotecting a compound of formula (I) which is protected.
  • Novel intermediates are claimed as an aspect of the present invention.
  • Physiological substrates of QC (EC) in mammals are, e.g. amyloid beta-peptides (3-40), (3- 42), (1 1 -40 and (1 1 -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 Amino acid sequences of physiological active peptides with an N-terminal glutamine residue, which are prone to be cyclized to final pGlu
  • Gly-Tyr-Glu-Val-His-His-Gln-Lys- neurodegeneration e.g. in
  • Gly-Tyr-Glu-Val-His-His-Gln-Lys- neurodegeneration e.g. in
  • Phe-Phe-Ala-Glu-Asp-Val-Gly-Ser- neurodegeneration e.g. in Asn-Lys-Gly-Ala-lle-lle-Gly-Leu-Met- Alzheimer's Disease, Familial Val-Gly-Gly-Val-Val-lle-Ala British Dementia, Familial
  • Phe-Phe-Ala-Glu-Asp-Val-Gly-Ser- neurodegeneration e.g. in Asn-Lys-Gly-Ala-lle-lle-Gly-Leu-Met- Alzheimer's Disease, Familial Val-Gly-Gly-Val-Val British Dementia, Familial
  • ADan EASNCFA IRHFENKFAV ETLIC Pyroglutamated form plays a
  • FPP QEP amide 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 i m porta nt role i n regu lati ng sperm fertility.
  • TRH QHP amide TRH functions as a regulator of the biosynthesis of TSH in the
  • neuromodulator in the central and peripheral nervous systems.
  • GnRH QHWSYGL RP(G) amide Stimulates the secretion of gonadotropins; it stimulates the
  • CCL16 small QPKVPEW VNTPSTCCLK Shows chemotactic activity for inducible cytokine YYEKVLPRRL WGYRKALNC lymphocytes and monocytes
  • SCYA16 shows chemotactic activity for monocytes
  • THP-1 monocytes but not for resting lymphocytes and neutrophils. Induces a calcium flux in THP-1 cells that were desensitized by prior expression to RANTES.
  • CCL8 small QPDSVSI PITCCFNVIN Chemotactic factor that attracts inducible cytokine RKIPIQRLES YTRITNIQCP monocytes, lymphocytes,
  • RWVRDSMKHL DQIFQNLKP play a role in neoplasia
  • This protein can bind heparin.
  • CCL2 (MCP-1 , small QPDAINA PVTCCYNFTN Chemotactic factor that attracts inducible cytokine RKISVQRLAS YRRITSSKCP monocytes and basophils but A2) KEAVIFKTIV AKEICADPKQ not neutrophils or eosinophils.
  • CCL18 small QVGTNKELC CLVYTSWQIP Chemotactic factor that attracts inducible cytokine QKFIVDYSET SPQCPKPGVI lymphocytes but not monocytes A18
  • LLTKRGRQIC ADPNKKWVQK or granulocytes. May be
  • naive T lymphocytes toward dendritic cells and activated macrophages in lymph nodes, has chemotactic activity for naive T cells, CD4+ and CD8+ T cells and thus may play a role in both humoral and cell-mediated immunity responses.
  • VGTELFRVPP VSTAATWQSS migration processes at the
  • APHQPGPSLW AEAKTSEAPS endothelium binds to CX3CR1 .
  • CCL7 small QPVGINT STTCCYRFIN Chemotactic factor that attracts inducible cytokine KKIPKQRLES YRRTTSSHCP monocytes and eosinophils, but
  • Orexin A (Hypocretin- QPLPDCCRQK TCSCRLYELL Neuropeptide that plays a 1 ) HGAGNHAAGI LTL significant role in the regulation of food intake and sleep-
  • Orexin-A binds to both 0X1 R and OX2R with a high affinity.
  • Substance P RPK PQQFFGLM Belongs to the tachykinins.
  • Tachykinins are active peptides which excite neurons, evoke behavioral responses, are potent vasodilators and secretagogues, and contract (directly or indirectly) many smooth muscles.
  • QYNAD Gln-Tyr-Asn-Ala-Asp Acts on voltage-gated sodium channels.
  • Glutamate is found in positions 3, 1 1 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(1 1 -40) and Abeta (1 1 -42) can be generated by the ⁇ -secretase enzyme ⁇ -site amyloid precursor protein- cleaving enzyme (BACE) at different sites (Huse J.T. 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.
  • BACE ⁇ -secretase enzyme ⁇ -site amyloid precursor protein- cleaving enzyme
  • 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.
  • 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, G.J. 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. 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, T.J. and Chen, D. 2000 Regul Pept 9337- 44).
  • 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 anti psychotic 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,
  • pancreatitis Bossetia, M., et al., (2005) Am.J Physiol Gastrointest.Liver Physiol 288, G1259-G1265); Alzheimer's disease (Yamamoto, M., et al., (2005) Am.J Pathol. 166, 1475-1485); lung fibrosis (Inoshima, I ., et al., (2004) Am.J Physiol Lung Cell Mol. Physiol 286, L1038-L1044); renal fibrosis (Wada, T., et al., (2004) J Am. Soc. Nephrol.
  • 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) Int.J 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.
  • 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-wake
  • 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 H u ntington , comprising the ad m in istration 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), TN Falpha inhibitors, muscarinic M1 receptor antagonists, NMDA receptor antagonists, sigma-1 receptor inh ibitors, h istamine H3 antagonists, immunomodulatory agents, immunosuppressive agents, MCP-1 antagonists or an agent selected from the group consisting of antegren (natalizumab), Neurelan (fampridine
  • 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
  • Tricyclic antidepressants e.g. amitryptiline, clomipramine, desipramine, doxepin, imipramine
  • Azapirones e.g. buspirone, tandopsirone
  • Serotonin-norepinephrine reuptake inhibitors e.g. venlafaxine, duloxetine
  • 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-1279 (syn. to HMR-1715, MNA-279),
  • autoimmune suppressant e.g. laquinimod
  • 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,
  • AGT-1 anti-granulocyte-macrophage colony-stimulating factor (GM- CSF) monoclonal antibody
  • Nogo receptor modulators ABT-874, alemtuzumab (CAMPATH), anti-OX40 antibody
  • PNA peptide nucleic acid
  • interferon alpha e.g. Alfaferone, human alpha interferon (syn. to Omniferon, Alpha Leukoferon),
  • interferon beta e.g. Frone, interferon beta-1 a like Avonex, Betron (Rebif), interferon beta analogs, interferon beta-transferrin fusion protein, recombinant interferon beta- l b like Betaseron,
  • peptides e.g. AT-008, AnergiX.MS, Immunokine (alpha-lmmunokine-NNS03), cyclic peptides like ZD-7349,
  • sCD8 soluble CD8
  • TNF-alpha e.g. BLX-1002, thalidomide, SH-636,
  • 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
  • CCR1 chemokine receptor-1
  • AMPA receptor antagonists e.g. ER-167288-01 and ER-099487, E-2007, talampanel
  • potassium channel blockers e.g. fampridine
  • IgLC immunoglobulin light chain
  • apoptosis inducing antigens e.g. Apogen MS
  • bb alpha-2 adrenoceptor agonist
  • tizanidine e.g. Zanaflex, Ternelin, Sirdalvo, Sirdalud, Mionidine
  • topoisomerase II modulators e.g. mitoxantrone hydrochloride
  • adenosine deaminase inhibitor e.g. cladribine (syn. to Leustatin, Mylinax, RWJ- 26251 ),
  • 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, PNU-156804, alpha-fetoprotein peptide (AFP), IPDS,
  • retinoid receptor agonists e.g. adapalene (syn. to Differin, CD-271 ),
  • TGF-beta e.g. GDF-1 (growth and differentiation factor 1 ),
  • TGF-beta-2 e.g. BetaKine
  • MMP inhibitors e.g. glycomed
  • nn) phosphodiesterase 4 (PDE4) inhibitors e.g. RPR-122818,
  • purine nucleoside phosphorylase inhibitors e.g. 9-(3-pyridylmethyl)-9-deazaguanine, peldesine (syn. to BCX-34, TO-200),
  • alpha-4/beta-1 integrin antagonists e.g. ISIS-104278,
  • CD49d alpha4 integrin
  • cytokine-inducing agents e.g. nucleosides, ICN-17261 ,
  • heat shock protein vaccines e.g. HSPPC-96
  • neuregulin growth factors e.g. GGF-2 (syn. to neuregulin, glial growth factor 2), vv) cathepsin S - inhibitors,
  • bropirimine analogs e.g. PNU-56169, PNU-63693,
  • Monocyte chemoattractant protein-1 inhibitors e.g. benzimidazoles like MCP-1 inhibitors, LKS-1456, PD-064036, PD-064126, PD-084486, PD-172084, PD-172386.
  • 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 2005/02561 6 , WO 2005/02551 6 , WO 2005/023858 , WO 2005/018424, WO 2005
  • 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 (Eli Lilly & Co.); AAB- 02 (Elan); bapineuzumab (Elan); BAN-2401 (Bioarctic Neuroscience AB); ABP-102 (Abiogen Pharma SpA); BA-27, BC-05 (Takeda); R-14
  • antibodies which recognize the N-terminus of the ⁇ peptide.
  • a suitable antibody, which recognizes the ⁇ - ⁇ -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 2008/01 1348.
  • a method for producing a vaccine composition for treating an amyloid-associated disease is disclosed in WO 2007/06841 1.
  • 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 2005/0071 99 , 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 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, WO03/09920
  • 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 (Eli 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- 21 166 (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 , WO2005/01 4553 , U S 6 , 890 , 956 , E P 1 334085 , E P 1 263774 , WO2004/101538, WO2004/00958, WO2004/
  • 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 (Eli 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 US6,01 1 ,155; US6, 107,317; US6,1 10,949; US6, 124,305; US6, 172,081 ; W099/61431 , W099/67278, W099/67279, DE19834591 , WO97/40832, WO95/15309, W098/19998, WO00/07617, WO99/38501 , W099/46272, WO99/38501 , WO01/68603, WO01/40180, WO01/81337, WO01 /81 304, WO01 /55105, WO02/02560, WO01 /34594, WO02/38541 , WO02/083128, WO03/072556, WO03/002593, WO03/000250, WO03/000180, WO03/000181 , EP
  • 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-1 149 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 Ingelheim 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;
  • 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)1 1 ) (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 (Eli Lilly & Co.); EGb-761 (Dr.
  • 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.
  • 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, W099/571 19, W099/13878, WO98/40102, WO98/01 157, WO96/20946, WO94/07890 and W092/21333.
  • 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), 7S,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 ,31 1 , 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 5,1 12,847, US 5,100,904, US 5,254,550, US 5,262,431 , US 5,340,832, US 4,956,380, EP 0303434, J P 03056486,
  • 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 97/1991 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 and WO 98/07420; WO 00/30674, US patents Nos.
  • 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
  • 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).
  • M 1 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 MS); LY-593093 (Eli 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.
  • 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-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 ) (P
  • 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 I I 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
  • CCB calcium channel blockers
  • beta-blockers beta-blockers
  • Angiotensin II receptor blockers are understood to be those active agents that bind to the AT1 -receptor subtype of angiotensin II 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 II receptor blockers which may be employed in the combination of the present invention include AT-i receptor antagonists having differing structural features, preferred are those with non-peptidic structures.
  • 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
  • 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 II 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.
  • 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.
  • metoprolol is suitably administered as its tartrate salt
  • propranolol is suitably administered as the hydrochloride salt
  • 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. 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.
  • 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 JTT705 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 (N F-KB) inhibitors/antibodies, ⁇ kinase (IKK) inhibitors/antibodies, activator protein-1 (AP-1 ) inhibitors/antibodies, STAT transcription factors inhibitors/antibodies, altered IL-6, partial
  • 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 2006/1 16435.
  • 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, US2006/31 6449, WO2004/056727, WO03/053368, WO00/1 98289, 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-1 1006 (Celgene Corp.); SSR-150106 (Sanofi-Aventis); MLN-1202 (Millenium Pharmaceuticals Inc.); AGI-1067, AGIX-4207, AGI-1096 (AtherioGenics Inc.); PRS-21 1095, PRS-21 1092 (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
  • 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.
  • 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, Ci
  • a QC inhibitor preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1 -89, 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 -89, i n com bi nation 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 -89, i n com bi nation 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 1-89, 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 -89, 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 -89, 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 -89, 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 -89, i n com bi nation 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 -89, i n com bi nation 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 -89, i n com bi nation 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 -89, in combination with HMG-Co-A- reductase inhibitors, for the prevention and/or treatment of restenosis, wherein the HMG-Co-A-reductase in h i bitor is selected from atorvastati n , cerivastati n , 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 -89, in combination with HMG
  • a QC inhibitor preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1 -89, 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 -89, 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 -89, 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 -89, in combination with beta- secretase in hibitors for the prevention and/or treatment of m ild cognitive impairment, wherein the beta-secretase inhibitor is selected from WY-25105, GW- 840736X and CTS-21 166,
  • a QC inhibitor preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1 -89, 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-21 166,
  • a QC inhibitor preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1 -89, 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-21 166,
  • a QC inhibitor preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1 -89, in combination with gamma- secretase in hibitors for the prevention and/or treatment of m ild 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 -89, 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 -89, in combination with gamma- secretase inhibitors for the prevention and/or treatment of neurodegeneration in
  • the gamma-secretase inhibitor is selected from LY- 450139, LY-41 1575 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 - 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 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 insolu ble salt of the active compound, such as the decanoate salt, may be adapted to provide a depot preparation for intramuscular injection.
  • 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 gu ms, and other pharmaceutical d il uents , e.g . water, to form a solid preformulation composition containing a homogeneous mixture of a compound of the present invention , or a pharmaceutically acceptable salt thereof.
  • a pharmaceutical carrier e.g. conventional tableting ingredients such as corn starch, lactose, sucrose, sorbitol, talc, stearic acid, magnesium stearate, dicalcium phosphate or gu ms, and other pharmaceutical d il uents , e.g . water
  • preformulation compositions 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.
  • 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 liq u id forms, such as solutions, syru ps , el ixi rs, em ulsions, 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.
  • 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, 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.
  • 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 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 31 st Edition page 341 and pages cited therein) or the above mentioned publications.
  • Step 2 N-Hydroxy-1 H-benzofdlimidazole-5-carboxamidine
  • a solution of 5-cyanobenzimidazole (1.43 g; 10 mmol; 1 eq.) in EtOH (30 ml) was treated with K 2 C0 3 (2.76 g; 20 mmol; 2 eq.) and hydroxylaminehydrochloride (765 mg; 1 1 mmol; 1 .1 eq.) and heated to reflux over night. After cooling to room temperature, the mixture was diltuted with Et 2 0. The resulting solid was collected by filtration, washed with Et 2 0 and water and used without further purification. Yield: 1 .28 g (72.7 %); MS m/z: 177.3 [M+H] +
  • Benzimidazol-5-carbohydrazide (1 eq.) was suspended in dry THF (10 ml). After addition of TEA (1 .1 eq.) the respective acid chloride (1.1 eq.) was added dropwise and the reaction was stirred at room temperature for 3 h. The mixture was then quenched with water and extracted with EtOAc (3x25 ml). The combined organic layers were dried over Na 2 S0 4 and evaporated. The remains were redissolved in acetonitrile (10 ml), treated with Lawesson ' s reagent (1 .5 eq.) and heated to reflux for 1 h.
  • Benzimidazol-5-carbohydrazide (1 eq.) and the respective carboxylic acid were suspended in acetonitrile (10 ml). Lawesson ' s reagent (1 .5 eq.) and POCI 3 (1 .5 eq.) were added and the mixture was heated to reflux over night. After cooling to room temperature, the mixture was basified by means of sat. NaHC0 3 -solution and extracted with EtOAc (3x25 ml). The combined organic layers were dried over Na 2 S0 4 , evaporated and the residue was purified by semi-preparative HPLC. -1 ,2,4-Triazoles
  • Step 1 The residue of Step 1 was suspended in dry EtOH (20 ml) and treated with hydrazine- monohydrate (1 .5 g; 30 mmol). The mixture was stirred at room temperature for 3 h. The precipitated solid was collected by filtration and washed with a small amount of water. The product was used without further purification. Yield: 0.75 g (51.2 %); MS m/z: 176.4 [M+H] +
  • Benzimidazol-5-carboxylic acid (1.62 g; 10 mmol; 1 eq.) was suspended in toluol (50 ml), treated with SOCI 2 (3.63 ml; 50 mmol; 5 eq.) and heated to reflux over night. The volatiles were evaporated and the remains were taken up with THF (50 ml). After cooling to 0°C 1 ,8- diazabicyclo[5.4.0]undec-7-en (2.23 ml; 15 mmol; 1 .5 eq.) was added carefully.
  • Phenylpropionic acid (0.451 g; 3 mmol; 1 eq.) was dissolved in THF (10 ml), treated with Carbonyldiimidazole (0.486 g; 3 mmol; 1 eq.) and stirred at room temperature for 1 h. The mixture was cooled to 0°C and a solution of isocyanoethylacetate (0.393 ml; 3.6 mmol; 1 .2 eq.) and 1 ,8-diazabicyclo[5.4.0]undec-7-ene (1 .34 ml; 9 mmol; 3 eq.) in TH F (10 ml) was added dropwise.
  • 3-(2,3-Dimethoxyphenyl propionic acid (0.630 g; 3 mmol; 1 eq.) was dissolved in THF (10 ml), treated with carbonyldiimidazole (0.486 g; 3 mmol; 1 eq.) and stirred at room temperature for 1 h.
  • the mixture was cooled to 0°C and a solution of isocyanoethylacetate (0.393 ml; 3.6 mmol; 1 .2 eq.) and 1 ,8-diazabicyclo[5.4.0]undec-7-ene (1 .34 ml; 9 mmol; 3 eq.) in THF (10 ml) was added dropwise.
  • Step 1 4-(2-(Trimethylsilyl)ethvnviy2-nitrobenzenamine
  • Benzimidazole-5-carbohydrazide (1 eq.) was suspended in THF (10 ml), treated with the respective isocyanate or isothiocyanate (1 eq.) and heated to reflux for 1 hour. The solvent was removed in vacuo, the remains were taken up with Na 2 C0 3 (5% in H 2 0, 20 ml) and heated to reflux for 3 hours. After cooling to room temperature, the mixture was neutralized by means of 4 N HCI and extracted with EtOAc (3x25 ml). The combined organic layers were dried over Na 2 S0 4 , evaporated and the residue was purified by flash chromatography on silica using a CHCI 3 /MeOH Gradient.
  • the compound was synthesized starting from H-benzimidazole-5-carboxylic acid dimethylaminomethylene-hydrazide (300mg, 1 .29mmol), 2, 4-dichloro benzyl amine (0.35mL, 2.60mmol), acetic acid (5ml_) as described above. 45mg (16.60%)
  • Example 13 5-(4-(2-Chloro-6-fluorobenzyl)-4H-1 ,2,4-triazol-3-yl)-1 H-benzordlimidazole
  • the compound was synthesized starting from H-benzimidazole-5-carboxylic acid dimethylaminomethylene-hydrazide (300mg, 1 .29mmol), 2-chloro-6-flouro benzyl amine (0.35mL, 2.60mmol), acetic acid (5mL) as described above. 45mg (10.60%)
  • Example 20 5-(4-(4-(Trifluoromethoxy)benzyl)-4H-1 ,2,4-triazol-3-yl)-1 H-benzo[dlimidazole
  • the compound was synthesized starting from H-benzimidazole-5-carboxylic acid dimethylaminomethylene-hydrazide (300mg, 1 .29mmol), 4-triflouromethoxy benzyl amine (0.4ml_, 2.60mmol), acetic acid (5ml_) as described above. Yield: 40mg (8.5%)
  • the compound was synthesized starting from H-benzimidazole-5-carboxylic acid dimethylaminomethylene-hydrazide (300mg, 1.29mmol), 2-chloro-5-(trifluoro methyl)-benzyl amine (0.51 mL, 3.25mmol), acetic acid (5mL) as described above.
  • Example 24 5-(4-(3-Chloro-4-methoxybenzyl)-4H-1 ,2,4-triazol-3-yl)-1 H-benzo[dlimidazole
  • the compound was synthesized starting from H-benzimidazole-5-carboxylic acid dimethylaminomethylene-hydrazide (300mg, 1.29mmol), 3-chloro-4-methoxy-benzyl amine (445mg, 2.60mmol), acetic acid (5ml_) as described above. Yield: 45mg (10%)
  • Example 29 5-(5-(3,4-Dimethoxyphenethyl)-1 ,3,4-oxadiazol-2-yl)-1 H-benzo[dlimidazole
  • Example 30 5-(5-(2,4-Dimethoxyphenethyl)-1 ,3,4-oxadiazol-2-yl)-1 H-benzo[dlimidazole
  • the compound was synthesized starting from benzimidazol-5-carbohydrazide (176 mg, 1 mmol), TEA (0.153 ml; 1 .1 mmol), 2,4-dimethoxyphenylpropionylchloride (228 mg, 1.1 mmol) and POCI 3 (0.5 ml; 5.5 mmol) as described in method 1 ; yield: 0.013 mg (3.7%); MS m/z: 351 .3[M+H] + ; 1 H-NMR (DMSO d 6 , 400 MHz): ⁇ 2.97-3.00 (m, 2H); 3.1 1 -3.13 (m, 2H); 3.71 (s, 6H); 6.41 -6.43 (m, 1 H); 6.51 (br s, 1 H); 7.0
  • Example 34 5-(5-(4-(Trifluoromethyl)phenethyl)-1 ,3,4-oxadiazol-2-yl)-1 H-benzo[dlimidazole
  • Example 38 5-(5-(4-(Methylthio)phenethyl)-1 ,3,4-oxadiazol-2-yl)-1 H-benzo[dlimidazole
  • Example 47 5-(5-(3,4-Dimethoxyphenethyl)-1 ,3,4-thiadiazol-2-yl)-1 H-benzofdlimidazole
  • Example 48 5-(5-(2,4-Dimethoxyphenethyl)-1 ,3,4-thiadiazol-2-yl)-1 H-benzo[dlimidazole
  • Example 49 5-(5-(2,3-Dimethoxyphenethyl)-1 ,3,4-thiadiazol-2-yl)-1 H-benzo[dlimidazole
  • Example 50 5-(5-(4-(Methylthio)phenethyl)-1 ,3,4-thiadiazol-2-yl)-1 H-benzo[dlimidazole
  • Example 53 5-(5-(4-(Trifluoromethyl)phenethyl)-1 ,3,4-thiadiazol-2-yl)-1 H-benzo[dlimidazole
  • Example 55 5-(5-(3,4,5-Trimethoxyphenethyl)-1 ,3,4-thiadiazol-2-yl)-1 H-benzo[dlimidazole
  • Example 62 5-(5-(2,5-Dimethoxyphenethyl)-1 ,3,4-thiadiazol-2-yl)-1 H-benzo[dlimidazole
  • Example 70 7-(5-(2-(Benzordin ,3ldioxol-5-yl)ethyl)-1 ,3,4-thiadiazol-2-yl)H-imidazori ,2- alpyridine
  • Example 74 5-(5-(3,4-Dimethoxyphenethyl)-4H-1 ,2,4-triazol-3-yl)-1 H-benzo[dlimidazole
  • the compound was synthesized starting from 3-(3,4-dimethoxyphenyl)propionic acid (210 mg; 1 mmol), carbonyldiimidazole (162 mg; 1 mmol) and benzimidazol-5-carboxamidrazone (175 mg; 1 mmol) as described above; yield: 0.078 g (22.3%); MS m/z: 350.4 [M+H] + ; 1 H- NMR (DMSO d 6 , 400 MHz): ⁇ 2.92-3.02 (m, 2H); 3.68 (s, 6H); 6.72-7.00 (m, 3H); 7.57-7.87 (m, 2H); 8.14-8.30 (m, 2H); HPLC (METHOD [A]): rt 9.29 min (100%)
  • Example 84 5-(1 -((Phenylthio)methyl)-1 H-1 ,2,3-triazol-4-yl)-1 H-benzordlimidazole
  • Example 85 5-( 1 -(3,4-Dimethoxyphenethyl)-1 H-1 ,2,3-triazol-4-yl)-1 H-benzofdlimidazole
  • 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 ⁇ pGlu- /3NA from H-Gln-/3NA per minute under the described conditions.
  • QC was activity determined using H-Gln-AMC as substrate. Reactions were carried out at 30 °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 m M EDTA and an appropriately diluted aliquot of QC in a final volume of 250 ⁇ ⁇ . 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 a-Ketoglutaric acid and 30 U/ml glutamate dehydrogenase in a final volume of 250 ⁇ . 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 °C, using either the SPECTRAFIuor Plus or the Sunrise (both from TECAN) reader for microplates. Kinetic data was evaluated using GraFit software.
  • the sample composition was the same as described above, except of the putative inhibitory compound added.
  • samples contained 4 m M of the respective inhibitor and a substrate concentration at 1 K M .
  • 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:: 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 - 60 % (A), 15 min - 20 min - ⁇ 60 - 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 compounds were analyzed using a gradient at a flow rate of 0.6 mL/min; whereby eluent (A) was acetonitrile, eluent (B) was water and eluent (C) 2% formic acid in acetonitrile applying the following gradient:
  • 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 ⁇ ) 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 ⁇ ) 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
  • ⁇ -derived peptides were incubated in 100 ⁇ 0.1 M sodium acetate buffer, pH 5.2 or 0.1 M Bis-Tris buffer, pH 6.5 at 30 °C. Peptides were applied in 0.5 mM [Ap(3-1 1 )a] or 0.1 5 mM [Ap(3-21 )a] concentrations, and 0.2 U QC is added all 24 hours. I n case of Ap(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).
  • 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 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.
  • the invention embraces all combinations of preferred and more preferred groups and embodiments of groups recited above.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Immunology (AREA)
  • Neurosurgery (AREA)
  • Neurology (AREA)
  • Biomedical Technology (AREA)
  • Epidemiology (AREA)
  • Diabetes (AREA)
  • Rheumatology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Hematology (AREA)
  • Reproductive Health (AREA)
  • Cardiology (AREA)
  • Psychiatry (AREA)
  • Hospice & Palliative Care (AREA)
  • Pregnancy & Childbirth (AREA)
  • Obesity (AREA)
  • Pain & Pain Management (AREA)
  • Urology & Nephrology (AREA)
  • Vascular Medicine (AREA)
  • Oncology (AREA)
  • Dermatology (AREA)
  • Gynecology & Obstetrics (AREA)
  • Child & Adolescent Psychology (AREA)
  • Psychology (AREA)
  • Endocrinology (AREA)
  • Transplantation (AREA)

Abstract

The invention relates to novel heterocyclic 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.

Description

Novel Inhibitors
Field of the invention
The invention relates to novel heterocyclic 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 Mol 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, 1 1246-1 1250), 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 Glu-i-conversion is favored around pH 6.0 while Gln-i-conversion to pGlu-derivatives occurs with a pH-optimum of around 8.0. Since the formation of pGlu-A3-related peptides can be suppressed by inhibition of recombinant human QC a n d QC-activity from pig pituitary extracts, the enzyme QC is a target in drug development for treatment of Alzheimer's disease. Inhibitors of QC are described in WO 2004/098625, WO 2004/098591 , WO 2005/039548, WO 2005/075436, WO 2008/055945, WO 2008/055947, WO 2008/055950 and WO2008/065141 .
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 " Κ and "KD" 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 where 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 1 10995), 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 imgf000005_0001
Scheme 2: Cyclization of L-homoglutamine
peptide
Figure imgf000005_0002
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_0003
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 I C50 for QC inhibition of 10 μΜ or less, more preferably of 1 μΜ or less, even more preferably of 0.1 μΜ or less or 0.01 μΜ or less, or most preferably 0.001 μΜ or less. I ndeed, 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 C1-12 alkyl group, suitably a Ci-8 alkyl group, e.g. 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 C2-12 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 C3-10 cycloalkyl group (i.e. 3 to 10 ring carbon atoms), more suitably a C3-8 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 C5-10 cycloalkenyl group (i .e. 5 to 1 0 ring carbon atoms), more suitably a C5-s 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, piperazine, dioxolane and dioxane. 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 C6-12 aryl group, suitably a C6-io aryl group, more suitably a C6-8 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 Ci-4alkylene moiety.
The term "halogen" or "halo" comprises fluorine (F), chlorine (CI) and bromine (Br). The term "amino" refers to the group -NH2.
The term "phenyl substituted by phenyl" refers to biphenyl.
The term " ^Λ wr " denotes a single bond where the stereochemistry is not defined.
When benzimidazol l is shown as benzimidazol-5-yl, which is represented as:
Figure imgf000009_0001
the person skilled in the art will appreciate that benzimidazol-6-yl, which is represented as:
Figure imgf000010_0001
is an equivalent structure. As employed herein, the two forms of benzimidazolyl are covered by the term "benzimidazol-5-yl". 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 naphthalenes- acrylic), benzoic, 4-methoxybenzoic, 2- or 4-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 /V-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 is provided a compound of formula (I):
Figure imgf000013_0001
(I)
or a pharmaceutically acceptable salt, solvate or polymorph thereof, including all tautomers and stereoisomers thereof wherein:
R1 represents a heteroaryl group optionally substituted by one or more groups selected from Ci-6alkyl, C2-6alkenyl, C2-6alkynyl, Ci-6haloalkyl, -Ci-6thioalkyl, -SOCi-4alkyl, -S02Ci-4alkyl, Ci_ 6alkoxy-, -0-C3-8cycloal kyl , C3-8cycloalkyl, -S02C3-8cycloalkyl, -SOC3-6cycloalkyl, C3- 6alkenyloxy-, C3-6alkynyloxy-, -C(0)Ci-6alkyl, -C(0)OCi-6alkyl, Ci-6alkoxy-Ci-6alkyl-, nitro, halogen, cyano, -C(0)OH, -NHCi-4alkyl, -N(C1_4alkyl)(C1-4alkyl), -C(0)N(Ci-4alkyl)(Ci-4alkyl), - C(0)NH2, -C(0)NH(Ci-4alkyl) and -C(O)NH(C3-i0cycloalkyl);
and in which any of aforesaid carbocyclyl groups may optionally be substituted by one or more groups selected from Ci-4alkyl, oxo, halogen and Ci-4alkoxy;
Y represents a 5 membered heteroaryl group selected from triazolyl, oxadiazolyl, thiadiazolyl, thiazolyl, triazol-one or triazol-thione;
X represents a linker selected from -(CH2)m-R2, -(CH2)n-S-R2, -(CH2)p-0-R2 or -(CH2)p-S02- R2;
m represents an integer selected from 1 to 4;
n represents an integer selected from 1 or 2;
p represents an integer selected from 0 to 2;
R2 represents aryl, heteroaryl, carbocyclyl or heterocyclyl;
in which any of aforesaid aryl and heteroaryl groups may optionally be substituted by one or more groups selected from Ci-6alkyl, C2-6alkenyl, C2-6alkynyl, Ci-6haloalkyl, -Ci_ 6thioalkyl, -SOCi-4alkyl, -S02Ci-4alkyl , Ci-6alkoxy-, -0-C3-8cycloalkyl, C3-8cycloalkyl, - S02C3-8cycloalkyl, -SOC3-6cycloalkyl, C3-6alkenyloxy-, C3-6alkynyloxy-, -C(0)Ci-6alkyl, - C(0)OCi-6alkyl, Ci-6alkoxy-Ci-6alkyl-, nitro, halogen, haloCi-6alkyl, haloCi-6alkoxy, cyano, hydroxyl, -C(0)OH, -NH2, -NHCi-4alkyl, -N(Ci-4alkyl)(Ci-4alkyl), -C(0)N(d.
4alkyl)(Ci-4alkyl), -C(0)NH2, -C(0)NH(Ci-4alkyl) and -C(O)NH(C3-i0cycloalkyl);
and in which any of aforesaid carbocyclyl and heterocyclyl groups may optionally be substituted by one or more groups selected from Ci-4alkyl, oxo, halogen and Ci-4alkoxy; or R2 represents phenyl substituted by phenyl, phenyl substituted by a monocyclic heteroaryl group, phenyl substituted by phenoxy, phenyl substituted by heterocyclyl, phenyl substituted by -0-Ci-4alkyl-heterocyclyl, phenyl substituted by benzyloxy, phenyl fused to carbocyclyl or phenyl fused to heterocyclyl, such that when Y represents oxadiazolyl and X represents CH2, R2 represents a group other than phenyl substituted by phenoxy;
in which any of aforesaid phenyl, benzyloxy and heteroaryl groups may optionally be substituted by one or more groups selected from Ci-4alkyl, halogen and Ci-4alkoxy, and in which any of aforesaid carbocyclyl and heterocyclyl groups may optionally be substituted by one or more groups selected from methyl, phenyl, oxo, halogen and Ci_ 4alkoxy; with the proviso that the compound of formula (I) is a compound other than:
6-(5-(2,3-dimethoxyphenethyl)-1 ,3,4-thiadiazol-2-yl)H-imidazo[1 ,2-a]pyridine;
6-(5-(4-(methylthio)phenethyl)-1 ,3,4-thiadiazol-2-yl)H-imidazo[1 ,2-a]pyridine; or
6-(5-(2-chloro-3-methoxyphenethyl)-1 ,3,4-thiadiazol-2-yl)H-imidazo[1 ,2-a]pyridine.
Detailed description of the invention
According to one particular aspect of the invention which may be mentioned there is provided a compound of formula (I):
Figure imgf000015_0001
(I)
or a pharmaceutically acceptable salt, solvate or polymorph thereof, including all tautomers and stereoisomers thereof wherein:
R1 represents a heteroaryl group optionally substituted by one or more groups selected from
Ci-6alkyl, C2-6alkenyl, C2-6alkynyl, Ci-6haloalkyl, -Ci-6thioalkyl, -SOCi-4alkyl, -S02Ci-4alkyl, Ci_ 6alkoxy-, -0-C3-8cycloal kyl , C3-8cycloalkyl, -S02C3-8cycloalkyl, -SOC3-6cycloalkyl, C3-
6alkenyloxy-, C3-6alkynyloxy-, -C(0)Ci-6alkyl, -C(0)OCi-6alkyl, Ci-6alkoxy-Ci-6alkyl-, nitro, halogen, cyano, hydroxyl, -C(0)OH, -NH2, -NHCi-4alkyl, -N(C1_4alkyl)(C1-4alkyl), -C(0)N(d.
4alkyl)(Ci-4alkyl), -C(0)NH2, -C(0)NH(Ci-4alkyl) and -C(O)NH(C3-i0cycloalkyl);
and in which any of aforesaid carbocyclyl groups may optionally be substituted by one or more groups selected from Ci-4alkyl, oxo, halogen and Ci-4alkoxy;
Y represents a 5 membered heteroaryl group selected from triazolyl, oxadiazolyl, thiadiazolyl, thiazolyl, triazol-one or triazol-thione;
X represents a linker selected from -(CH2)m-, -(CH2)n-S-, -(CH2)p-0- or -(CH2)p-S02-;
m represents an integer selected from 1 to 4;
n represents an integer selected from 1 or 2;
p represents an integer selected from 0 to 2;
R2 represents aryl, heteroaryl, carbocyclyl or heterocyclyl;
in which any of aforesaid aryl and heteroaryl groups may optionally be substituted by one or more groups selected from Ci-6alkyl, C2-6alkenyl, C2-6alkynyl, Ci-6haloalkyl, -Ci_ 6thioalkyl, -SOCi-4alkyl, -S02Ci-4alkyl, Ci-6alkoxy-, -0-C3-8cycloalkyl, C3-8cycloalkyl, -
S02C3-8cycloalkyl, -SOC3-6cycloalkyl, C3-6alkenyloxy-, C3-6alkynyloxy-, -C(0)Ci-6alkyl, - C(0)OCi-6alkyl, Ci-6alkoxy-Ci-6alkyl-, nitro, halogen, haloCi-6alkyl, haloCi-6alkoxy, cyano, hydroxyl, -C(0)OH, -NH2, -NHCi-4alkyl, -N(Ci-4alkyl)(Ci-4alkyl), -C(0)N(d. 4alkyl)(Ci-4alkyl), -C(0)NH2, -C(0)NH(Ci-4alkyl) and -C(O)NH(C3-i0cycloalkyl);
and in which any of aforesaid carbocyclyl and heterocyclyl groups may optionally be substituted by one or more groups selected from Ci-4alkyl, oxo, halogen and Ci-4alkoxy; or R2 represents phenyl substituted by phenyl, phenyl substituted by a monocyclic heteroaryl group, phenyl substituted by phenoxy, phenyl substituted by heterocyclyl, phenyl substituted by -0-Ci-4alkyl-heterocyclyl, phenyl substituted by benzyloxy, phenyl fused to carbocyclyl or phenyl fused to heterocyclyl, such that when Y represents oxadiazolyl and X represents CH2, R2 represents a group other than phenyl substituted by phenoxy;
in which any of aforesaid phenyl, benzyloxy and heteroaryl groups may optionally be substituted by one or more groups selected from Ci-4alkyl, halogen and Ci-4alkoxy, and in which any of aforesaid carbocyclyl and heterocyclyl groups may optionally be substituted by one or more groups selected from methyl, phenyl, oxo, halogen and Ci_ 4alkoxy;
with the proviso that the compound of formula (I) is a compound other than:
6-(5-(2,3-dimethoxyphenethyl)-1 ,3,4-thiadiazol-2-yl)H-imidazo[1 ,2-a]pyridine;
6-(5-(4-(methylthio)phenethyl)-1 ,3,4-thiadiazol-2-yl)H-imidazo[1 ,2-a]pyridine; or
6-(5-(2-chloro-3-methoxyphenethyl)-1 ,3,4-thiadiazol-2-yl)H-imidazo[1 ,2-a]pyridine.
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), -C1-6thioalkyl (e.g. -S-methyl), -SOCi-4alkyl (e.g. -SOmethyl), -S02Ci-4alkyl (e.g. -S02methyl), Ci-6alkoxy- (e.g. methoxy, ethoxy), -0-C3-8cycloalkyl (e.g. -O-cyclopentyl), C3-8cycloalkyl (e.g. cyclopropyl, cyclohexyl), -S02C3-8cycloalkyl (e.g. -S02cyclohexyl), -SOC3- 6cycloalkyl (e.g. -SOcyclopropyl), C3-6alkenyloxy- (e.g. -O-buten-2-yl), C3-6alkynyloxy- (e.g. - O-buten-2-yl), -C(0)Ci-6alkyl (e.g. -C(O)ethyl), -C(0)OCi-6alkyl (e.g. -C(O)O-methyl), Ci_ 6alkoxy-Ci-6alkyl- (e.g. methoxy-ethyl-), nitro, halogen (e.g. fluoro, chloro, bromo), cyano, hydroxyl, -C(0)OH, -NH2, -NHCi-4alkyl (e.g. -NHmethyl), -N(C1-4alkyl)(C1-4alkyl) (e.g. - N(methyl)2), -C(0)N(Ci-4alkyl)(Ci-4alkyl) (e.g. -C(0)N(methyl)2), -C(0)NH2, -C(0)NH(Ci-4alkyl) (e.g. -C(O)NHmethyl), -C(O)NH(C3-i0cycloalkyl) (e.g. -C(O)NHcyclopropyl). 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 and hydroxy.
When R1 represents heteroaryl, examples include monocyclic (e.g. 5 and 6 membered) and bicyclic (e.g. 9 and 10 membered, particularly 9 membered) heteroaryl rings, especially rings containing nitrogen atoms (e.g. 1 or 2 nitrogen atoms). A suitable bicyclic heteroaryl ring is a 9-membered heteroaryl ring containing 1 or 2 nitrogen atoms, especially a benzene ring fused to a 5-membered ring containing one or two nitrogen atoms (e.g. 1 H-benzoimidazolyl). Most suitably the point of attachment is through a benzene ring, e.g. the group is 1 H- benzoimidazol-5-yl. Aforementioned heteroaryl groups may either be unsubstituted (which is more typical) or may suitably be substituted by one or more (e.g. 1 or 2) substituents selected from alkyl (e.g. Ci- alkyl such as Me), alkoxy- (e.g. Ci- alkoxy- such as OMe) and halogen (e.g. F).
Particular examples of R1 heteroaryl groups include a 5-membered ring containing 2 or 3 nitrogen atoms, which ring may optionally be substituted (e.g. in particular by one or two groups, such as methyl, for example:
Figure imgf000017_0001
Other examples of R1 heteroaryl groups include a 9-membered bicyclic ring containing 2 nitrogen atoms, which ring may optionally be substituted, for example:
Figure imgf000017_0002
Figure imgf000018_0001
When Y represents triazolyl, examples include:
Figure imgf000018_0002
When Y represents oxadiazolyl, examples include:
Figure imgf000018_0003
When Y represents thiadiazolyl, an example includes:
Figure imgf000018_0004
When Y represents thiazolyl, examples include:
Figure imgf000018_0005
When Y represents triazol-one, an example includes
Figure imgf000018_0006
When Y represents triazol-thione, an example includes: H
Figure imgf000019_0001
When R2 represents optionally substituted aryl, aryl may typically represent phenyl. Exemplary substituted phenyl groups include 3-methylphenyl-, 2,3-dichlorophenyl-, 2,3- difluorophenyl-, 2,4-dichlorophenyl-, 2,4-difluororophenyl-, 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-, 2,6-difluoro-4- (methoxy)phenyl-, 2-isopropyl-6-methylphenyl-, 3-(cyclopentyloxy)-4-methoxyphenyl-, 3,4,5- trimethoxyphenyl-, 3,4-dimethoxyphenyl-, 3,4-dichlorophenyl-, 3,4-difluorophenyl-, 3,4- dimethylphenyl-, 3,4,5-trifluorophenyl-, 3,5-bis(trifluororomethyl)phenyl-, 3,5- dimethoxyphenyl-, 2-methoxyphenyl-, 3-methoxyphenyl-, 4-(trifluoromethyl)phenyl-, 4-bromo-
2- (trifluoromethyl)phenyl-, 4-bromophenyl-, 4-chloro-3-(trifluoromethyl)phenyl-, 4- chlorophenyl-, 4-cyanophenyl-, 4-ethoxyphenyl-, 4-ethylphenyl-, 4-fluorophenyl-, 4- isopropylphenyl-, 4-methoxyphenyl-, 4-ethoxyphenyl-, 4-propoxyphenyl-, 4-butoxyphenyl-, 4- pentoxyphenyl-, 4-isopropyloxyphenyl-, 4-tetrafluoroethyloxyphenyl-. Alternatively, R2 may represent unsubstituted phenyl-. Further exemplary substituted phenyl groups include 2,3,4- trifluorophenyl, 2,3-difluoro-4-methylphenyl, 2-bromo-4-fluorophenyl-, 2-bromo-5- fluorophenyl-, 2-chlorophenyl-, 2-fluorophenyl-, 2-fluoro-5-(trifluoromethyl)phenyl-, 2-hydroxy-
3- methoxyphenyl-, 2-hydroxy-5-methylphenyl-, 3-chlorophenyl-, 3-fluorophenyl-, 3-fluoro-4- (trifluoromethyl)phenyl-, 3-fluoro-5-(trifluoromethyl)phenyl-, 2-fluoro-4-(trifluoromethyl)phenyl- , 3-fluoro-4-(methoxy)phenyl-, 3-hydroxy-4-methoxyphenyl-, 4-bromo-2-fluorophenyl, 4- chloro-3-(trifluoromethyl)phenyl-, 4-chloro-3-methylphenyl, 4-chlorophenyl-, 4-fluorophenyl- and 4-propoxyphenyl-.
When R2 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 R2 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. pyridin-2-yl and pyridin-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-yk 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, benzo[1 ,2,5]thiadiazol-6-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 R2 represents carbocyclyl, examples include cycloalkyi and cycloalkenyl. Examples of cycloalkyi 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, 3-methyl-cyclohex-3-enyl, 3-methyl-cyclohex-3-enyl.
When R2 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 R2 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. When R2 represents phenyl substituted by phenyl in which any of aforesaid phenyl groups may optionally be substituted, examples include -biphenyl-4-yl.
When R2 represents phenyl substituted by a monocyclic heteroaryl group, in which any of aforesaid phenyl and heteroaryl groups may optionally be substituted, examples include 4- (oxazol-5-yl)phenyk
When R2 represents phenyl substituted by benzyloxy in which any of aforesaid phenyl and benzyloxy groups may optionally be substituted, examples include 4-benzyloxy-phenyl-, 4-(3- methylbenzyloxy)phenyl- and 4-(4-methylbenzyloxy)phenyk
When R2 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 R2 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-yk
Suitably R1 represents bicyclic heteroaryl, especially 9-membered bicyclic heteroaryl. More suitably, R1 represents a bicyclic heteroaryl ring system and in particular a phenyl ring fused with a 5 membered heteroaryl ring containing one or more (e.g. one or two, suitably one, more suitably two) nitrogen atoms or a pyridine ring fused with a 5-membered heteroaryl ring containing one or more (e.g. one or two, suitably one, more suitably two) nitrogen atoms. When R1 represents bicyclic heteroaryl, preferably the heteroaryl group does not contain S atoms. When R1 represents a phenyl ring fused to a 5-membered heteroaryl ring, preferably R1 is linked to the core of formula (I) through the phenyl ring. When R1 represents a pyridine ring fused to a 5-membered heteroaryl ring, preferably R1 is linked to the core of formula (I) through the pyridine ring. Suitably R1 represents unsubstituted heteroaryl. In particular, R1 suitably represents 1 H-benzoimidazolyl or imidazo[1 ,2-a]pyridine, particularly 1 H- benzoimidazolyl, especially 1 H-benzoimidazol-5-yl. In one embodiment R1 represents
Figure imgf000022_0001
wherein A represents a bond and R11, R12 and R13 independently represent H or Ci-2alkyl.
In a second embodiment R1 represents
Figure imgf000022_0002
wherein B represents a bond and R14 and R15 independently represent H or Ci-2alkyl.
In a third embodiment R1 represents
Figure imgf000022_0003
wherein C represents a bond and R16 and R17 independently represent H or Ci-2alkyl.
In a fourth embodiment R1 represents
Figure imgf000022_0004
wherein D represents a bond and R18 and R19 independently represent H or Ci-2alkyl. Suitabl R1 represents
Figure imgf000023_0001
In one embodiment R14 represents H and R15 represents H . In another embodiment R14 represents H and R15 represents Ci-2alkyl. In a third embodiment R14 represents Ci-2alkyl and R15 represents H. In a fourth embodiment R14 represents methyl and R15 represents H.
Suitably B represents a bond.
Figure imgf000023_0002
R11 suitably represents H,
R12 suitably represents H or methyl.
R13 suitably represents H or methyl.
In one embodiment of the invention, R12 represents H and R13 represents methyl. In another embodiment, R12 represents methyl and R13 represents H . I n a third embodiment, R12 represents H and R13 represents H.
Suitably A represents a bond.
Alternatively R1 represents
Figure imgf000024_0001
In one embodiment R16 represents H and R17 represents H . In another embodiment R16 represents H and R17 represents Ci-2alkyl. In a third embodiment R16 represents Ci-2alkyl and R17 represents H.
Suitably C represents a bond.
Alternativel R1 represents
Figure imgf000024_0002
In one embodiment R18 represents H and R19 represents H . In another embodiment R18 represents H and R19 represents Ci-2alkyl. In a third embodiment R18 represents Ci-2alkyl and R19 represents H.
Suitably D represents a bond.
More suitabl R1 represents
Figure imgf000024_0003
Most suitably R1 represents
Figure imgf000025_0001
Suitably, Y represents triazolyl, oxadiazolyl or thiadiazolyl. More suitably, Y represents triazolyl or thiadiazolyl. In a further embodiment, Y is attached to R1 via a carbon atom.
Suitably, Y represents triazolyl, such
4H-1 ,2,4-triazolyl, i.e.
Figure imgf000025_0002
or
1 H-1 ,2,3-triazolyl,
Figure imgf000025_0003
Suitably, Y represents oxadiazolyl. In one embodiment, Y represents oxadiazolyl, such 1 ,3,4-oxadiazolyl, i.e.
Figure imgf000025_0004
Suitably, Y represents thiadiazolyl. In one embodiment, Y represents thiadiazolyl, such as: 1 ,3,4-thiadiazolyl, i.e.
Figure imgf000025_0005
Suitably, Y represents thiazolyl. In one embodiment, Y represents thiazolyl, such as:
Figure imgf000026_0001
Suitably, Y represents triazol-one, such as:
2H-1 ,2,4-triazol-3-one, i.e.
Figure imgf000026_0002
Suitably, Y represents triazol-thione, such as:
4H-1 ,2,4-triazol-3-thione, i.e.
Figure imgf000026_0003
Suitably, X represents a linker selected from -(CH2)m-R2, -(CH2)P-S-R2, -(CH2)P-0-R2 or - (CH2)p-S02-R2.
More suitably, X represents a linker selected from -(CH2)m-R2 and -(CH2)P-S-R2 (i.e. -CH2- R2, (CH2)2-R2 or -CH2-S-R2).
Suitably, X represents -(CH2)m-R2. In one embodiment, X represents -CH2-R2, -(CH2)2-R2 or -(CH2)3-R2. In a further embodiment, X represents -CH2-R2 or -(CH2)2-R2. In a yet further embodiment, X represents -CH2-R2. In an alternative embodiment, X represents -(CH2)2-R2.
Suitably, X represents -(CH2)n-S-R2. In one embodiment, X represents -CH2-S-R2.
Suitably, X represents -(CH2)p-0-R2. In one embodiment, X represents -CH2-0-R2.
Suitably, X represents -(CH2)p-S02-R2. In one embodiment, X represents -CH2-S02-R2.
Suitably, m represents an integer selected from 1 to 3. In a further embodiment, m represents an integer selected from 1 or 2. Suitably, n represents 1 . Suitably, p represents an integer selected from 0 or 1 . In a further embodiment, p represents 1 .
Suitably, R2 represents aryl, heteroaryl, carbocyclyl, heterocyclyl, phenyl substituted by phenoxy or phenyl fused to heterocyclyl. More suitably, R2 represents aryl, heteroaryl, phenyl substituted by phenoxy or phenyl fused to heterocyclyl. Yet more suitably, R2 represents optionally substituted aryl or phenyl substituted by phenoxy. Still yet more suitably, R2 represents optionally substituted aryl.
In one embodiment, R2 represents carbocyclyl. When R2 represents carbocyclyl, R2 suitably represents cyclohexyl.
In one embodiment, R2 represents optionally substituted aryl. When R2 represents optionally substituted aryl, R2 suitably represents optionally substituted phenyl or napthyl. More suitably, R2 represents optionally substituted phenyl.
In one embodiment, R2 represents phenyl optionally substituted by one or more groups selected from Ci-6 alkyl (e.g. methyl), Ci-6 alkoxy (e.g. methoxy or ethoxy), -Ci-6thioalkyl (e.g. methylthio), haloCi-6 alkyl (e.g. trifluoromethyl), haloCi-6 alkoxy (e.g. trifluoromethoxy) or halogen (e.g. chlorine or fluorine). In a further embodiment, R2 represents phenyl optionally substituted by one or more groups selected from Ci-6 alkoxy (e.g. methoxy) or halogen (e.g. chlorine).
When R2 represents optionally substituted phenyl, R2 suitably represents 2-methylphenyl, 4- methylphenyl, 4-trifluoromethylphenyl, 5-trifluoromethylphenyl, 2,5-bis(trifluoromethyl), 4-methylthio(phenyl), 2-methoxyphenyl, 3-methoxyphenyl, 4-methoxyphenyl, 2,3- dimethoxyphenyl, 2,4-dimethoxyphenyl, 2,5-dimethoxyphenyl, 3,4-dimethoxyphenyl, 3,4,5- trimethoxyphenyl, 4-ethoxyphenyl, 2-fluorophenyl, 3-fluorophenyl, 4-fluorophenyl, 2,3- difluorophenyl, 2,4-difluorophenyl, 3,4-difluorophenyl, 2-chlorophenyl, 3-chlorophenyl, 4- chlorophenyl, 2,3-dichlorophenyl, 2,4-dichlorophenyl, 2,5-dichlorophenyl, 2,6-dichlorophenyl, 3,4-dichlorophenyl, 3,5-dichlorophenyl, 3-chloro-4-fluorophenyl, 2-chloro-6-fluorophenyl, 4- trifluoromethoxyphenyl, 3-chloro-4-methoxyphenyl, 2-chloro-3-methoxyphenyl or 2-chloro-5- trifluoromethylphenyl.
I n one embodiment, R2 represents optionally substituted heteroaryl. When R2 represents optionally substituted heteroaryl, R2 suitably represents optionally substituted pyridyl. In an alternative embodiment, R2 represents unsubstituted pyridyl, e.g. pyridin-4-yl.
In one embodiment, R2 represents phenyl substituted by phenyl, the aforesaid phenyl groups optionally being substituted. When R2 represents phenyl substituted by phenyl, the aforesaid phenyl groups optionally being substituted, R2 suitably represents phenyl substituted by 3- phenyl, phenyl substituted by 4-phenyl, phenyl substituted by 3-(3-chlorophenyl), phenyl substituted by 4-(3-chlorophenyl), phenyl substituted by 4-(3,4-dichlorophenyl) or 3- fluorophenyl substituted by 4-phenyl.
I n one embod iment, R2 represents optionally substituted phenyl substituted by optionally su bstituted phenoxy. When R2 represents optionally substituted phenyl substituted by optionally substituted phenoxy, R2 suitably represents phenyl substituted by 4-phenoxy.
I n one embod iment, R2 represents optionally substituted phenyl substituted by optionally substituted heterocyclyl. When R2 represents optionally substituted phenyl substituted by optionally substituted heterocyclyl, R2 suitably represents 3-chlorophenyl substituted by 4- morpholinyl, phenyl substituted by 4-piperazinyl substituted by 4N-methyl, phenyl substituted by 4-piperazinyl substituted by 4N-phenyl, phenyl substituted by 3-piperazinyl substituted by 4N-phenyl or 2-chlorophenyl substituted by 6-piperazinyl substituted by 4N-ethyl.
In one embodiment, R2 represents optionally substituted phenyl substituted by -0-C-i-4alkyl- heterocyclyl. When R2 represents optionally substituted phenyl substituted by -0-Ci-4alkyl- heterocyclyl, R2 suitably represents phenyl substituted by 4-0-(CH2)2-morpholinyl, 4-0- (CH2)3-morpholinyl, 2-0-(CH2)2-morpholinyl or 4-0-(CH2)2-piperazinyl.
I n on e em bod i m ent, R2 represents optionally substituted phenyl fused to optionally substituted heterocyclyl. Wh e n R2 represents optionally substituted phenyl fused to optionally substituted heterocyclyl, R2 suitably represents benzo-1 ,3-dioxolanyl, 4- methoxy(benzo-1 ,3-dioxolanyl), 6-methoxy(benzo-1 ,3-dioxolanyl), 2,2-difluoro(benzo-1 ,3- dioxolanyl) or benzo-1 ,4-dioxanyl. In a further embodiment, when R2 represents optionally substituted phenyl fused to optionally substituted heterocyclyl, R2 suitably represents benzo- 1 ,3-dioxolanyl.
One particular subset of compounds of formula (I) which may be mentioned are those wherein R1 re resents
Figure imgf000029_0001
Y represents triazolyl or thiadiazolyl, X represents -(CH2)m- and -(CH2)P-S- (i.e. -CH2-, (CH2)2- or -CH2-S-) and R2 represents phenyl optionally substituted by one or more groups selected from Ci-6 alkoxy (e.g. methoxy) or halogen (e.g. chlorine) or R2 represents phenyl substituted by phenoxy.
Processes
According to a further aspect of the invention there is provided a process for preparing a compound of formula (I) which comprises:
(a) preparing a compound of formula (I) wherein R1 represents 1 H-benzo[d]imidazolyl from a compound of formula (II):
Figure imgf000029_0002
NH2
(II)
wherein X and Y are as defined above for compounds of formula (I). Process (a) typically comprises dissolving a compound of formula (II) in a suitable solvent, such as formic acid and formic acid orthoethylester, followed by treatment with sodium formate and Pd/C and heating to 1 1 0°C for 24 h. A non-limiting example of the methodology of process (a) described in Section F herein.
(b) preparing a compound of formula (I) wherein Y represents 1 ,2,4-triazolyl by reacting compound of formula (III):
Figure imgf000030_0001
R1 (III)
wherein R1 is as defined above for compounds of formula (I), with a compound of formula X- NH2, wherein X is as defined above for compounds of formula (I). Process (b) typically comprises heating to 1 50 °C overnight. A non-limiting example of the methodology of process (b) is described in Section A, Method 1 herein.
(c) preparing a compound of formula (I) wherein Y represents 1 ,2,4-triazolyl by reacting a compound of formula (IV):
Figure imgf000030_0002
(IV)
wherein R1 is as defined above for compounds of formula (I), with a compound of formula X- NH2, wherein X is as defined above for compounds of formula (I). Process (c) typically comprises reaction in the presence of glacial acetic acid followed by heating at 130-135 °C for 48 hours. A non-limiting example of the methodology of process (c) is described in Section A, Method 2 herein. (d) preparing a compound of formula (I) wherein Y represents 1 ,2,4-oxadiazolyl by reacting a compound of formula (V):
OH
Figure imgf000031_0001
R
(V)
wherein R1 is as defined above for compounds of formula (I), with a compound of formula X- COOH, wherein X is as defined above for compounds of formula (I). Process (d) typically comprises dissolving the compound of formula X-COOH in DMF, followed by treatment with carbonyldiimidazole and stirred at room temperature for 1 hour. The compound of formula (V) is then added followed by heating at 1 10 °C overnight. A non-limiting example of the methodology of process (d) is described in Section B herein.
(e) preparing a compound of formula (I) wherein Y represents 1 ,3,4-thiadiazolyl from a compound of formula (VI):
X
O H
°γ ΝΗ
R1 (VI)
wherein R1 and X are as defined above for compounds of formula (I). Process (e) typically comprises the use of with Lawesson's reagent and heating to reflux. A non-limiting example of the methodology of process (e) is described in Section C, Methods 1 and 2 herein. (f) preparing a compound of formula (I) wherein Y represents 1 ,3,4-thiadiazolyl by reacting a compound of formula (VII):
NH2
Figure imgf000032_0001
(VII)
wherein R1 is as defined above for compounds of formula (I), with a compound of formula X- COOH, wherein X is as defined above for compounds of formula (I). Process (f) typically comprises reaction in the presence of acetonitrile, Lawesson's reagent and POCI3 followed by heating to reflux overnight. A non-limiting example of the methodology of process (f) is described in Section C, Method 3 herein.
(g) preparing a compound of formula (I) wherein Y represents 4H-1 ,2,4-triazolyl by reacting a compound of formula (VIII):
NH2
Figure imgf000032_0002
R
(VIM)
wherein R1 is as defined above for compounds of formula (I), with a compound of formula X- COOH, wherein X is as defined above for compounds of formula (I). Process (g) typically comprises reaction in the presence of DMF and carbonyldiimidazole followed by heating to 1 10 °C for 24 hours. A non-limiting example of the methodology of process (g) is described in Section D herein.
(h) preparing a compound of formula (I) wherein Y represents thiazolyl by reacting a compound of formula R1-COOH with a compound of formula X-CO-CH2-NH2, wherein R1 and X are as defined above for compounds of formula (I ). Process (h) typically comprises reaction in the presence of acetonitrile, Lawesson's reagent, TEA and POCI3 followed by heating to reflux overnight. A non-limiting example of the methodology of process (h) is described in Section E herein.
(i) preparing a compound of formula (I) wherein Y represents 1 ,2,4-triazol-one or 1 ,2,4- triazol-thione from a compound of formula (IX):
Figure imgf000033_0001
(IX)
wherein R1 and X are as defined above for compounds of formula (I) and W represents O or S. Process (i) typically comprises the use of 5% Na2C03 followed by heating to reflux for 3 hours. A non-limiting example of the methodology of process (i) is described in Section G herein. interconversion of compounds of formula (I); and deprotecting a compound of formula (I) which is protected.
Compounds of formula (I) and intermediate compounds may also be prepared using techniques analogous to those known to a skilled person, or described herein.
Novel intermediates are claimed as an aspect of the present invention.
Therapeutic uses
Physiological substrates of QC (EC) in mammals are, e.g. amyloid beta-peptides (3-40), (3- 42), (1 1 -40 and (1 1 -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
Peptide Amino acid sequence Function
Abeta(1 -42) Asp-Ala-Glu-Phe-Arg-His-Asp-Ser- Plays a role in
Gly-Tyr-Glu-Val-His-His-Gln-Lys- neurodegeneration, e.g. in
Leu-Val-Phe-Phe-Ala-Glu-Asp-Val- Alzheimer's Disease, Familial
Gly-Ser-Asn-Lys-Gly-Ala-lle-lle-Gly- British Dementia, Familial
Leu-Met-Val-Gly-Gly-Val-Val-lle-Ala Danish Dementia, Down
Syndrome
Abeta(1 -40) Asp-Ala-Glu-Phe-Arg-His-Asp-Ser- Plays a role in
Gly-Tyr-Glu-Val-His-His-Gln-Lys- neurodegeneration, e.g. in
Leu-Val-Phe-Phe-Ala-Glu-Asp-Val- Alzheimer's Disease, Familial
Gly-Ser-Asn-Lys-Gly-Ala-lle-lle-Gly- British Dementia, Familial
Leu-Met-Val-Gly-Gly-Val-Val Danish Dementia, Down
Syndrome
Abeta(3-42) Glu-Phe-Arg-His-Asp-Ser-Gly-Tyr- Plays a role in
Glu-Val-His-His-Gln-Lys-Leu-Val- neurodegeneration, e.g. in
Phe-Phe-Ala-Glu-Asp-Val-Gly-Ser- Alzheimer's Disease, Familial
Asn-Lys-Gly-Ala-lle-lle-Gly-Leu-Met- British Dementia, Familial
Val-Gly-Gly-Val-Val-lle-Ala Danish Dementia, Down
Syndrome
Abeta(3-40) Glu-Phe-Arg-His-Asp-Ser-Gly-Tyr- Plays a role in
Glu-Val-His-His-Gln-Lys-Leu-Val- neurodegeneration, e.g. in
Phe-Phe-Ala-Glu-Asp-Val-Gly-Ser- Alzheimer's Disease, Familial
Asn-Lys-Gly-Ala-lle-lle-Gly-Leu-Met- British Dementia, Familial
Val-Gly-Gly-Val-Val Danish Dementia, Down
Syndrome Peptide Amino acid sequence Function
Abeta(1 1 -42) Glu-Val-His-His-Gln-Lys-Leu-Val- Plays a role in
Phe-Phe-Ala-Glu-Asp-Val-Gly-Ser- neurodegeneration, e.g. in Asn-Lys-Gly-Ala-lle-lle-Gly-Leu-Met- Alzheimer's Disease, Familial Val-Gly-Gly-Val-Val-lle-Ala British Dementia, Familial
Danish Dementia, Down Syndrome
Abeta(1 1 -40) Glu-Val-His-His-Gln-Lys-Leu-Val- Plays a role in
Phe-Phe-Ala-Glu-Asp-Val-Gly-Ser- neurodegeneration, e.g. in Asn-Lys-Gly-Ala-lle-lle-Gly-Leu-Met- Alzheimer's Disease, Familial Val-Gly-Gly-Val-Val British Dementia, Familial
Danish Dementia, Down Syndrome
ABri EASNCFA IRHFENKFAV ETLIC Pyroglutamated form plays a
SRTVKKNIIEEN role in Familial British Dementia
ADan EASNCFA IRHFENKFAV ETLIC Pyroglutamated form plays a
FNLFLNSQEKHY role in Familial Danish
Dementia
Gastrin 17 QGPWL EEEEEAYGWM DF Gastrin stimulates the stomach
(amide) mucosa to produce and secrete Swiss-Prot: P01350 hydrochloric acid and the
pancreas to secrete its digestive enzymes. It also stimulates smooth muscle contraction and increases blood circulation and water secretion in the stomach and intestine.
Neurotensin QLYENKPRRP YIL Neurotensin plays an endocrine or paracrine role in the
Swiss-Prot: P30990 regulation of fat metabolism. It causes contraction of smooth muscle. Peptide Amino acid sequence Function
FPP QEP amide 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 i m porta nt role i n regu lati ng sperm fertility.
TRH QHP amide TRH functions as a regulator of the biosynthesis of TSH in the
Swiss-Prot: P20396 anterior pituitary gland and as a neurotransmitter/
neuromodulator in the central and peripheral nervous systems.
GnRH QHWSYGL RP(G) amide Stimulates the secretion of gonadotropins; it stimulates the
Swiss-Prot: P01 148 secretion of both luteinizing and follicle-stimulating hormones.
Peptide Amino acid sequence Function
CCL16 (small QPKVPEW VNTPSTCCLK Shows chemotactic activity for inducible cytokine YYEKVLPRRL WGYRKALNC lymphocytes and monocytes
A16) HLPAIIFVTK RNREVCTNPN but not neutrophils. Also shows
DDWVQEYIKD PNLPLLPTRN potent myelosuppressive
Swiss-Prot: 015467 LSTVKIITAK NGQPQLLNSQ activity, suppresses
proliferation of myeloid progenitor cells. Recombinant
SCYA16 shows chemotactic activity for monocytes and
THP-1 monocytes, but not for resting lymphocytes and neutrophils. Induces a calcium flux in THP-1 cells that were desensitized by prior expression to RANTES.
CCL8 (small QPDSVSI PITCCFNVIN Chemotactic factor that attracts inducible cytokine RKIPIQRLES YTRITNIQCP monocytes, lymphocytes,
A8) KEAVIFKTKR GKEVCADPKE basophils and eosinophils. May
RWVRDSMKHL DQIFQNLKP play a role in neoplasia and
Swiss-Prot: P80075 inflammatory host responses.
This protein can bind heparin.
Peptide Amino acid sequence Function
CCL2 (MCP-1 , small QPDAINA PVTCCYNFTN Chemotactic factor that attracts inducible cytokine RKISVQRLAS YRRITSSKCP monocytes and basophils but A2) KEAVIFKTIV AKEICADPKQ not neutrophils or eosinophils.
KWVQDSMDHL DKQTQTPKT Augments monocyte anti-tumor
Swiss-Prot: P13500 activity. Has been implicated in the pathogenesis of diseases characterized by monocytic infiltrates, like psoriasis, rheumatoid arthritis or atherosclerosis. May be involved in the recruitment of monocytes into the arterial wall during the disease process of atherosclerosis. Binds to CCR2 and CCR4.
CCL18 (small QVGTNKELC CLVYTSWQIP Chemotactic factor that attracts inducible cytokine QKFIVDYSET SPQCPKPGVI lymphocytes but not monocytes A18) LLTKRGRQIC ADPNKKWVQK or granulocytes. May be
YISDLKLNA involved in B cell migration into
Swiss-Prot: P55774 B cell follicles in lymph nodes.
Attracts naive T lymphocytes toward dendritic cells and activated macrophages in lymph nodes, has chemotactic activity for naive T cells, CD4+ and CD8+ T cells and thus may play a role in both humoral and cell-mediated immunity responses. Peptide Amino acid sequence Function
Fractalkine QHHGVT KCNITCSKMT The soluble form is chemotactic
(neurotactin) SKIPVALLIH YQQNQASCGK for T cells and monocytes, but
RAIILETRQH RLFCADPKEQ not for neutrophils. The
Swiss-Prot: P78423 WVKDAMQHLD RQAAALTRNG membrane-bound form
GTFEKQIGEV KPRTTPAAGG promotes adhesion of those
MDESVVLEPE ATGESSSLEP leukocytes to endothelial cells.
TPSSQEAQRA LGTSPELPTG May play a role in regulating
VTGSSGTRLP PTPKAQDGGP leukocyte adhesion and
VGTELFRVPP VSTAATWQSS migration processes at the
APHQPGPSLW AEAKTSEAPS endothelium binds to CX3CR1 .
TQDPSTQAST ASSPAPEENA
PSEGQRVWGQ GQSPRPENSL
EREEMGPVPA HTDAFQDWGP
GSMAHVSWP VSSEGTPSRE
PVASGSWTPK AEEPIHATMD
PQRLGVLITP VPDAQAATRR
QAVGLLAFLG LLFCLGVAMF
TYQSLQGCPR KMAGEMAEGL
RYIPRSCGSN SYVLVPV
CCL7 (small QPVGINT STTCCYRFIN Chemotactic factor that attracts inducible cytokine KKIPKQRLES YRRTTSSHCP monocytes and eosinophils, but
A7) REAVIFKTKL DKEICADPTQ not neutrophils. Augments
KWVQDFMKHL DKKTQTPKL monocyte anti-tumor activity.
Swiss-Prot: P80098 Also induces the release of gelatinase B. This protein can bind heparin. Binds to CCR1 ,
CCR2 and CCR3. Peptide Amino acid sequence Function
Orexin A (Hypocretin- QPLPDCCRQK TCSCRLYELL Neuropeptide that plays a 1 ) HGAGNHAAGI LTL significant role in the regulation of food intake and sleep-
Swiss-Prot 043612 wakefulness, possibly by
coordinating the complex behavioral and physiologic responses of these
complementary homeostatic functions. It plays also a broader role in the homeostatic regulation of energy metabolism, autonomic function, hormonal balance and the regulation of body fluids. Orexin-A binds to both 0X1 R and OX2R with a high affinity.
Substance P RPK PQQFFGLM Belongs to the tachykinins.
Tachykinins are active peptides which excite neurons, evoke behavioral responses, are potent vasodilators and secretagogues, and contract (directly or indirectly) many smooth muscles.
QYNAD Gln-Tyr-Asn-Ala-Asp Acts on voltage-gated sodium channels.
Glutamate is found in positions 3, 1 1 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(1 1 -40) and Abeta (1 1 -42) can be generated by the β-secretase enzyme β-site amyloid precursor protein- cleaving enzyme (BACE) at different sites (Huse J.T. 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, G.J. 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, T.J. and Chen, D. 2000 Regul Pept 9337- 44).
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 anti psychotic 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 II 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 Clin. Invest s, 773-778); rheumatoid arthritis (Gong, J. H., et al., (1997) J Exp. Med 186, 131 -137; Ogata, H., et al., (1997) J Pathol. 182, 106-1 14); pancreatitis (Bhatia, M., et al., (2005) Am.J Physiol Gastrointest.Liver Physiol 288, G1259-G1265); Alzheimer's disease (Yamamoto, M., et al., (2005) Am.J Pathol. 166, 1475-1485); lung fibrosis (Inoshima, I ., et al., (2004) Am.J 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) Int.J 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, 808-81 1 ). 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 H u ntington , comprising the ad m in istration 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), TN Falpha inhibitors, muscarinic M1 receptor antagonists, NMDA receptor antagonists, sigma-1 receptor inh ibitors, h istamine H3 antagonists, immunomodulatory agents, immunosuppressive agents, MCP-1 antagonists or an agent selected from the group consisting of antegren (natalizumab), Neurelan (fampridine-S R), cam path (alemtuzu mab), I R 208, N B I 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- l b like Betaseron,
h) interferon tau,
i) peptides, e.g. AT-008, AnergiX.MS, Immunokine (alpha-lmmunokine-NNS03), 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 II 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 2005/02561 6 , WO 2005/02551 6 , WO 2005/023858 , WO 2005/018424, WO 2005/01 1599, WO 2005/000193, WO 2004/108895, WO 2004/098631 , WO 2004/08041 9 , WO 2004/071 408 , WO 2004/0691 82 , 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 (Eli 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 Αβ peptide. A suitable antibody, which recognizes the Αβ-Ν-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 2008/01 1348. A method for producing a vaccine composition for treating an amyloid-associated disease is disclosed in WO 2007/06841 1.
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 2005/0071 99 , 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 (Eli 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- 21 166 (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 , WO2005/01 4553 , U S 6 , 890 , 956 , E P 1 334085 , E P 1 263774 , WO2004/101538, WO2004/00958, WO2004/08991 1 , WO2004/073630, WO2004/069826, WO2004/039370, WO2004/031 139, WO2004/031 137, 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 (Eli 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 US6,01 1 ,155; US6, 107,317; US6,1 10,949; US6, 124,305; US6, 172,081 ; W099/61431 , W099/67278, W099/67279, DE19834591 , WO97/40832, WO95/15309, W098/19998, WO00/07617, WO99/38501 , W099/46272, WO99/38501 , WO01/68603, WO01/40180, WO01/81337, WO01 /81 304, 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 , WO 03/037327 , WO03/0401 74 , WO03/045977 , WO 03/055881 , 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, WO2004/1 10436, WO2004/1 1 1041 , WO2004/1 12701 , WO2005/000846, WO2005/000848, WO2005/01 1581 , 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, WO2005/1 13510, WO2005/1 16014, WO2005/1 16029, WO2005/1 18555, WO2005/120494, WO2005/121089, WO2005/121 131 , 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-1 149 (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 Ingelheim 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)1 1 ) (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 (Eli 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-81 12 (Sanofi-Aventis); CR-3465 (Rottapharm SpA.); GP-0203, NCS- 613 (Centre National de la Recherche Scientifique); KF-19514 (Kyowa Hakko Kogyo Co. Ltd.); ONO-6126 (Ono Pharmaceutical Co. Ltd.); OS-0217 (Dainippon Pharmaceutical Co. Ltd.); IBFB-13001 1 , 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, W099/571 19, W099/13878, WO98/40102, WO98/01 157, WO96/20946, WO94/07890 and W092/21333.
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), 7S,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 ,31 1 , 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 5,1 12,847, US 5,100,904, US 5,254,550, US 5,262,431 , US 5,340,832, US 4,956,380, EP 0303434, J P 03056486, J P 01 143897, 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 931 3065, J P 05201 970, WO 9412474, EP 0670309, EP 0451 547, J P 06339390, US 5,073,549, US 4,999,349, EP 0268281 , US 4,743,616, EP 0232849, EP 0224272, JP 621 14978, JP 621 14957, 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 0421 1648, 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 97/1991 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 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 J P 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 061 16284 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). M 1 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 MS); LY-593093 (Eli 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, W099/471 31 , WO99/07359, WO98/30243, W097/38993, W097/13754, W094/29255, WO94/20476, W094/19356, WO93/03034 and W092/19238. 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, W099/53922, W099/48891 , W099/45963, WO99/01416, WO99/07413, WO99/0141 6, WO98/50075, WO98/50044, WO98/10757, WO98/05337, W097/32873, W097/23216, W097/23215, W097/23214, W096/14318, WO96/08485, W095/31986, W095/26352, WO95/26350, W095/26349, W095/26342, W095/12594, WO95/02602, WO95/02601 , WO94/20109, W094/13641 , WO94/09016 and W093/25534. 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 I I 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 II receptor blockers are understood to be those active agents that bind to the AT1 -receptor subtype of angiotensin II 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 II receptor blockers which may be employed in the combination of the present invention include AT-i 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 imgf000060_0001
the compound with the designation SC-52458 of the following formula
Figure imgf000060_0002
and the compound with the designation the compound ZD-8731 of the formula
Figure imgf000060_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 II 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 JTT705 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 (N F-KB) inhibitors/antibodies, ΙκΒ 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 2006/1 16435. 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, US2006/31 6449, WO2004/056727, WO03/053368, WO00/1 98289, 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-1 1006 (Celgene Corp.); SSR-150106 (Sanofi-Aventis); MLN-1202 (Millenium Pharmaceuticals Inc.); AGI-1067, AGIX-4207, AGI-1096 (AtherioGenics Inc.); PRS-21 1095, PRS-21 1092 (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 -89, 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 -89, i n com bi nation 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 -89, i n com bi nation 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 1-89, 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 -89, 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 -89, 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 -89, 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 -89, 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 -89, i n com bi nation 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 -89, i n com bi nation 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 -89, i n com bi nation 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 -89, in combination with HMG-Co-A- reductase inhibitors, for the prevention and/or treatment of restenosis, wherein the HMG-Co-A-reductase in h i bitor is selected from atorvastati n , cerivastati n , 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 -89, 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 -89, 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 -89, 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 -89, 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 -89, 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 -89, in combination with beta- secretase in hibitors for the prevention and/or treatment of m ild cognitive impairment, wherein the beta-secretase inhibitor is selected from WY-25105, GW- 840736X and CTS-21 166,
a QC inhibitor, preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1 -89, 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-21 166,
a QC inhibitor, preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1 -89, 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-21 166,
a QC inhibitor, preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1 -89, in combination with gamma- secretase in hibitors for the prevention and/or treatment of m ild 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 -89, 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 -89, 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-41 1575 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 insolu ble 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 gu ms, and other pharmaceutical d il uents , 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 liq u id forms, such as solutions, syru ps , el ixi rs, em ulsions, 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 31 st Edition page 341 and pages cited therein) or the above mentioned publications.
xamples
Figure imgf000074_0001
Figure imgf000075_0001
Figure imgf000076_0001
Figure imgf000077_0001
Figure imgf000078_0001
Figure imgf000079_0001
Figure imgf000080_0001
Figure imgf000081_0001
Figure imgf000082_0001
Figure imgf000083_0001
Figure imgf000084_0001
Figure imgf000085_0001
Figure imgf000086_0001
Figure imgf000087_0001
Figure imgf000088_0001
Figure imgf000089_0001
Figure imgf000090_0001
Figure imgf000091_0001
General synthesis description: -Benzyl-1 ,2,4-Triazoles
Figure imgf000092_0001
5-(1 ,3,4-oxadiazol-2-yl)-1 H-benzo[dlimidazole
Benzimidazol-5-ca rbohyd razi d e ( 1 .77 g ; 1 0 m m ol ) was su spend ed i n form ic aci d orthoethylester (20 ml) and heated to reflux for 5 h. The solvent was evaporated and the residue was used without further purification. MS m/z: 187.3 [M+H]+
General procedure
5-(1 ,3,4-Oxadiazol-2-yl)-1 H-benzo[d]imidazole (1 eq . ) was treated with the respective benzylamine (0.5 ml) and heated to 150°C over night. After cooling the mixture was directly purified by semi-preparative HPLC.
Method 2
Figure imgf000092_0002
1 H-Benzimidazole-5-carboxylic acid dimethylaminomethylene-hydrazide
Dimethylformamide dimethyl acetal (19g, 15.90mmol) was added to a suspension of 1 H- benzo[d]imidazole-5-carbohydrazide (3.50g, 19.88mmol) in 1 :1 toluene; methanol (60ml_) and refluxed overnight. The volatiles were evaporated in vacuo to afford crude compound which was triturated with 6% methanol in chloroform (10ml_), precipitated solid was filtered and dried under reduced pressure to afford 3.20g (69.56%%) 1 H-benzimidazole-5- carboxylic acid dimethylaminomethylene-hydrazide as brown solid.
General procedure
A mixture of 1 H-benzimidazole-5-carboxylic acid dimethylaminomethylene-hydrazide (1 eq) and substituted benzyl amine (2eq) in glacial acetic acid (5ml_) was heated in a sealed tube at 130-135°C for 48h. The solvent was evaporated in vacuo to afford crude compound. The crude compound was purified by conducting two successive preparative TLC procedures a) 7% methanol in chloroform followed by, b) 7% methanol in ethyl acetate +2% ammonia solution as eluent to afford the product.
Figure imgf000093_0001
N-Hydroxy-1 H-benzofdlimidazole-5-carboxamidine
Step 1 : 1 H-Benzo[dlimidazole-5-carbonitrile
3,4-Diaminobenzonitrile (5.0 g; 37 mmol) was dissolved in 5 N HCI (200 ml). After addition of formic acid (20 ml) the mixture was heated to reflux for 3 h. After cooling to room temperature, the mixture was basified by means of aqueous NH3-solution and put into a fridge. The precipitated solid was collected by filtration, washed with water and used without further purification. Yield: 3.6 g (67.6%) MS m/z: 144.2 [M+H]+
Step 2: N-Hydroxy-1 H-benzofdlimidazole-5-carboxamidine A solution of 5-cyanobenzimidazole (1.43 g; 10 mmol; 1 eq.) in EtOH (30 ml) was treated with K2C03 (2.76 g; 20 mmol; 2 eq.) and hydroxylaminehydrochloride (765 mg; 1 1 mmol; 1 .1 eq.) and heated to reflux over night. After cooling to room temperature, the mixture was diltuted with Et20. The resulting solid was collected by filtration, washed with Et20 and water and used without further purification. Yield: 1 .28 g (72.7 %); MS m/z: 177.3 [M+H]+
General procedure
The respective carboxylic acid (1 eq.) was dissolved in DMF (5 ml), treated with carbonyldiimidazole (1 eq.) and stirred at room temperature for 1 h. N-Hydroxy-benzimidazol- 5-carboxamidine (1 eq.) was added and the mixture was heated to 1 10°C over night. After cooling to room temperature, the mixture was diluted with water until a precipitate was formed, basified by means of sat. NaHC03-solution and extracted with EtOAc (3x25 ml). The combined organic layers were dried over Na2S04, evaporated and the residue was purified by flash chromatography on silica using a CHCI3/MeOH gradient.
(C) 1 ,3,4-Thiadiazoles
Figure imgf000095_0001
General procedures
Method 1
Benzimidazol-5-carbohydrazide (1 eq.) was suspended in dry THF (10 ml). After addition of TEA (1 .1 eq.) the respective acid chloride (1.1 eq.) was added dropwise and the reaction was stirred at room temperature for 3 h. The mixture was then quenched with water and extracted with EtOAc (3x25 ml). The combined organic layers were dried over Na2S04 and evaporated. The remains were redissolved in acetonitrile (10 ml), treated with Lawesson's reagent (1 .5 eq.) and heated to reflux for 1 h. After cooling to roomtemperature the mixture was quenched with water, basified by means of 2 N NaOH and extracted with EtOAc (3x25 ml). The combined organic layers were dried over Na2S04, evaporated and the residue was purified by flash chromatography on silica using a CHCI3/MeOH gradient. Method 2
/V,/V-Dicyclohexylcarbodiimide (1 eq.) and the respective carboxylic acid (1 eq.) were dissolved in THF (10 ml) and stirred at room temperature for 1 h. Benzimidazol-5- carbohydrazide (1 eq.) was added and the mixture was stirred at 50°C over night. After cooling to room temperature, Lawesson's reagent (1 .5 eq.) was added and the mixture was refluxed for 5 h. After cooling, the mixture was quenched by means of sat. NaHC03-solution and extracted with EtOAc (3x25 ml). The combined organic layers were dried over Na2S04, evaporated and the residue was purified by semi-preparative HPLC. Method 3
Benzimidazol-5-carbohydrazide (1 eq.) and the respective carboxylic acid were suspended in acetonitrile (10 ml). Lawesson's reagent (1 .5 eq.) and POCI3 (1 .5 eq.) were added and the mixture was heated to reflux over night. After cooling to room temperature, the mixture was basified by means of sat. NaHC03-solution and extracted with EtOAc (3x25 ml). The combined organic layers were dried over Na2S04, evaporated and the residue was purified by semi-preparative HPLC. -1 ,2,4-Triazoles
Figure imgf000096_0001
Benzimidazol-5-carboxamidrazone Step 1 : Benzimidazol-5-(ethylcarboxiimidate)-dihydrochloride
5-Cyanobenzimidazole (1 .43 g; 10 mmol) was suspended in dry EtOH (20 ml). HCI-gas was bubbled through the ice-cooled suspension for 15 min. The reaction flask was sealed and the mixture was stirred at room temperature for 48 h. Et20 was added and the solid was collected by filtration, washed with Et20 and EtOH and was used without further purification. Yield: 2.19 g (83.6 %); MS m/z: 190.3 [M+H]+
Step 2: Benzimidazol-5-carboxamidrazone
The residue of Step 1 was suspended in dry EtOH (20 ml) and treated with hydrazine- monohydrate (1 .5 g; 30 mmol). The mixture was stirred at room temperature for 3 h. The precipitated solid was collected by filtration and washed with a small amount of water. The product was used without further purification. Yield: 0.75 g (51.2 %); MS m/z: 176.4 [M+H]+
General procedure
A solution of the respective carboxylic acid (1 eq . ) in DM F (5 ml) was treated with carbonyldiimidazole (1 eq.) and stirred at room temperature for 1 h. After the addition of benzimidazol-5-carboxamidrazone (175 mg; 1 mmol), the mixture was heated to 1 10°C and stirred for 24 h. After cooling to room temperature, water was added until a precipitate was formed. The mixture was basified by means of sat. NaHC03-solution and extracted with EtOAc (3x25 ml). The combined organic layers were dried over Na2S04, evaporated and the residue was purified by flash chromatography using a CHCI3/MeOH gradient.
(E) Thiazoles
Figure imgf000097_0001
Benzimidazol-5-aminomethylketon-dihvdrochloride Step 1 : Ethyl 5-(Benzimidazol-5-yl)oxazol-4-carboxylate
Benzimidazol-5-carboxylic acid (1.62 g; 10 mmol; 1 eq.) was suspended in toluol (50 ml), treated with SOCI2 (3.63 ml; 50 mmol; 5 eq.) and heated to reflux over night. The volatiles were evaporated and the remains were taken up with THF (50 ml). After cooling to 0°C 1 ,8- diazabicyclo[5.4.0]undec-7-en (2.23 ml; 15 mmol; 1 .5 eq.) was added carefully. A mixture of isocyanoacetate (1 .32 ml; 12 mmol; 1 .2 eq.) and 1 ,8-diazabicyclo[5.4.0]undec-7-ene (4.46 ml; 30 mmol; 3 eq.) in THF (50 ml) was added dropwise. After complete addition, the reaction was stirred at room temperature for 24 h. The reaction was diluted with water and extracted with EtOAc (3x100 ml). The combined organic layers were dried over Na2S04, evaporated and the residue was purified by flash chromatography on silica using a CHCI3/MeOH gradient. Yield: 2.0 g (78.0 %); MS m/z: 258.3 [M+H]+
Step 2: Benzimidazol-5-aminomethylketone-dihvdrochloride
Ethyl 5-(benzimidazol-5-yl)oxazol-4-carboxylate (2.0 g; 7.8 mmol) was dissolved in MeOH (20 ml) and treated with cone, aqueous HCI (40 ml). The mixture was refluxed over night. The solvent was removed in vacuo and the residue was used without further purification. MS m/z: 176.3 [M+H]+
Amino-4-phenylbutan-2-one-hydrochloride
Phenylpropionic acid (0.451 g; 3 mmol; 1 eq.) was dissolved in THF (10 ml), treated with Carbonyldiimidazole (0.486 g; 3 mmol; 1 eq.) and stirred at room temperature for 1 h. The mixture was cooled to 0°C and a solution of isocyanoethylacetate (0.393 ml; 3.6 mmol; 1 .2 eq.) and 1 ,8-diazabicyclo[5.4.0]undec-7-ene (1 .34 ml; 9 mmol; 3 eq.) in TH F (10 ml) was added dropwise. After complete addition, the mixture was stirred at room temperature for 24 h, then diluted with water and extracted with EtOAc (3x25 ml). The combined organic layers were dried over Na2S04, evaporated and the residue was purified by flash chromatography on silica using a CHCI3/MeOH gradient. Yield: 0.319 g (43.4%); MS m/z: 246.3 [M+H]+.
The remains were dissolved in MeOH (5 ml), treated with cone, aqueous HCI (15 ml) and refluxed over night. The solvent was removed in vacuo and the residue was used without further purification. MS m/z: 164.4 [M+H]+
Amino-4-(2,3-dimethoxyphenyl)butan-2-one-hvdrochloride
3-(2,3-Dimethoxyphenyl propionic acid (0.630 g; 3 mmol; 1 eq.) was dissolved in THF (10 ml), treated with carbonyldiimidazole (0.486 g; 3 mmol; 1 eq.) and stirred at room temperature for 1 h. The mixture was cooled to 0°C and a solution of isocyanoethylacetate (0.393 ml; 3.6 mmol; 1 .2 eq.) and 1 ,8-diazabicyclo[5.4.0]undec-7-ene (1 .34 ml; 9 mmol; 3 eq.) in THF (10 ml) was added dropwise. After complete addition the mixture was stirred at room temperature for 24 h, then diluted with water and extracted with EtOAc (3x25 ml). The combined organic layers were dried over Na2S04, evaporated and the residue was purified by flash chromatography on silica using a CHCb/MeOH gradient. Yield: 0.517 g (56.5%); MS m/z: 306.1 [M+H]+. The remains were dissolved in MeOH (5 ml), treated with cone, aqueous HCI (15 ml) and refluxed over night. The solvent was removed in vacuo and the residue was used without further purification. MS m/z: 224.3 [M+H]+
Lawesson
Figure imgf000099_0001
reflux
General procedure
The respective aminomethylketon-hydrochloride (1 eq.) and the respective carboxylic acid (1 eq.) were suspended in acetonitrile (10 ml). Lawesson's reagent (1 .5 eq.), TEA (3 eq.) and POCI3 (1 .5 eq.) were added and the mixture was heated to reflux over night. After cooling to room temperature the mixture was carfully basified by means of sat. NaHCC¾-solution and extracted with EtOAc (3x25 ml). The combined organic layers were dried over Ν¾304, evaporated and the residue was purified by semi-preparative HPLC.
Figure imgf000099_0002
4-Ethynyl-2-nitrobenzenamine
Step 1 : 4-(2-(Trimethylsilyl)ethvnviy2-nitrobenzenamine
4-lodo-2-nitroaniline (5.32 g, 20 mmol; 1 eq.) was dissolved in TH F (20 ml). PdCb(PPh3)2 (0.154 g; 0.22 mmol; 0.01 1 eq.), copper(l)-iodide (0.084 g; 0.44 mmol; 0.022 eq.) and DIPEA (10.5 ml; 60 mmol; 3 eq.) were added. To that mixture was added dropwise tnmethylsilylacetylene (3.1 ml; 22 mmol, 1 .1 eq.) at room temperature. After 3 h, the mixture was diluted with water and extracted with EtOAc (3x100 ml). The combined organic layers were dried over Na2S04, evaporated and purified by flash chromatography on silica using a CHCIs/MeOH gradient. Yield: 3.96 g (84.6%); MS m/z: 235.3 [M+H]+; 469.4 [2M+H]+
Step 2: 4-Ethynyl-2-nitrobenzenamine
2-Nitro-4-(trimethylsilylethinyl)aniline (3.96 g; 16.9 mmol; 1 eq.) was dissolved in MeOH (50 ml) and CH2CI2 (50 ml) and treated with K2C03 (9.33 g; 67.6 mmol; 4 eq.). After stirring at room temperature for 5 h, the mixture was diluted with water. The organic layer was separated and the aqueous layer was extracted with CH2CI2 (3x100 ml). The combined organic layers were dried over Na2S04, evaporated and the residue was purified by flash chromatography on silica using a CHCI3/MeOH gradient. Yield: 2.06 g (75.1 %); MS m/z: 163.4 [M+H]+
Figure imgf000100_0001
HCOOH
NaCHOO CH(OEt)3 Pd/C 110 °C
Figure imgf000100_0002
General procedure Step 1
A so l u ti o n of N a N3 (1 eq.) in DMSO (2 ml) was treated with the respective phenylethylbromide (1 eq.) and stirred at room temperature for 24 h. After that time, the reaction was diluted with water (2 ml). Sodium ascorbate (0.1 eq.), 4-ethinyl-2-nitroaniline (1 eq.) and CuS04 x 5 H20 (1 M solution in water; 0.2 eq.) were added sequentially and the mixture was stirred for a further 3 h. Water was added until a precipitate was formed. The solid was collected by filtration, washed with a small amount of water and was used without further purification.
Step 2
A solution of the respective azide (1 eq.) in DMSO (2 ml) was diluted with water (2 ml). Sodium ascorbate (0.1 eq.), 4-ethinyl-2-nitroaniline (1 eq.) and CuS04 x 5 H20 (1 M solution in water; 0.2 eq.) were added sequentially and the mixture was stirred for a further 3 h. Water was added until a precipitate was formed. The solid was collected by filtration, washed with a small amount of water and was used without further purification.
Step 3
The respective 4-(1 -alkyl-1 ,2,3-triazol-4-yl)-2-nitroaniline was dissolved in formic acid (5 ml) and formic acid orthoethylester (5 ml), treated with sodium formate (2.04 g; 30 mmol) and Pd/C and heated in a sealed tube to 1 10°C for 24 h. After cooling to room temperature, the mixture was filtred through Celite and the volatiles were removed in vacuo. The remains were taken up with a small amount of water, basified by means of sat. NaHC03-solution and extracted with EtOAc (3x25 ml) The combined organic layers were dried over Na2S04, evaporated and purified by flash chromatography on silica using a CHCI3/MeOH gradient.
(G) 1 ,2,4-Triazolones/-thiones
Figure imgf000102_0001
x = o,s
General procedure
Benzimidazole-5-carbohydrazide (1 eq.) was suspended in THF (10 ml), treated with the respective isocyanate or isothiocyanate (1 eq.) and heated to reflux for 1 hour. The solvent was removed in vacuo, the remains were taken up with Na2C03 (5% in H20, 20 ml) and heated to reflux for 3 hours. After cooling to room temperature, the mixture was neutralized by means of 4 N HCI and extracted with EtOAc (3x25 ml). The combined organic layers were dried over Na2S04, evaporated and the residue was purified by flash chromatography on silica using a CHCI3/MeOH Gradient.
Synthesis of the examples
Example 1 : 5-(4-Benzyl-4H-1 ,2,4-triazol-3-yl)-1 H-benzo[dlimidazole
The compound was synthesized starting from 5-(1 ,3,4-oxadiazol-2-yl)benzimidazole (186 mg, 1 mmol) and benzylamine (0.5 ml) as described above; yield: 0.072 g (26.2%); MS m/z: 276.5 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 5.39 (s, 2H); 6.99-7.01 (m, 2H); 7.20-7.29 (m, 3H); 7.63 (dd, 1 H, 4J=1 .7 Hz, 3J=8.7 Hz); 7.83 (d, 1 H, 3J=8.7 Hz); 7.93 (br s, 1 H); 8.79 (s, 1 H); 9.09 (s, 1 H); HPLC (METHOD [A]): rt 7.81 min (100%) Example 2: 5-(4-(3,4-Dimethoxybenzyl)-4H-1 ,2,4-triazol-3-yl)-1 H-benzoidlimidazole
The compound was synthesized starting from 5-(1 ,3,4-oxadiazol-2-yl)benzimidazole (186 mg, 1 mmol) and 3,4-Dimethoxybenzylamine (0.5 ml) as described above; yield: 0.087 g (25.9%); MS m/z: 336.3 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 3.55 (s, 3H); 3.65 (s, 3H); 5.27 (s, 2H); 6.50 (dd, 1 H, 4J=2.1 Hz, 3J=8.3 Hz); 6.64 (d, 1 H, 4J=2.1 Hz); 6.81 (d, 1 H, 3J=8.3 Hz), 7.62 (dd, 1 H, 4J=1 .2 Hz, 3J=8.7 Hz); 7.83 (d, 1 H, 3J=8.7 Hz); 7.92 (br s, 1 H); 8.76 (s, 1 H); 8.98 (s, 1 H); HPLC (METHOD [A]): rt 7.36 min (100%)
Example 3: 5-(4-(2-Fluorobenzyl)-4H-1 ,2,4-triazol-3-yl)-1 H-benzo[dlimidazole
The compound was synthesized starting from 5-(1 ,3,4-oxadiazol-2-yl)benzimidazole (186 mg, 1 mmol) and 2-fluorobenzylamine (0.5 ml) as described above; yield: 0.044 g (15.0%); MS m/z: 294.2 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 5.43 (s, 2H); 6.92-6.95 (m, 1 H); 7.05-7.14 (m, 2H); 7.22-7.32 (m, 1 H); 7.62 (dd, 1 H , 4J = 1 .7 Hz, 3J=8.7 Hz); 7.83 (d, 1 H , 3J=8.7 Hz); 7.94 (d, 1 H, 4J=1.7 Hz); 8.73 (s, 1 H); 9.09 (s, 1 H); HPLC (METHOD [A]): rt 8.36 min (98.9%)
Example 4: 5-(4-(4-Fluorobenzyl)-4H-1 ,2,4-triazol-3-yl)-1 H-benzo[dlimidazole
The compound was synthesized starting from 5-(1 ,3,4-oxadiazol-2-yl)benzimidazole (186 mg, 1 mmol) and 4-fluorobenzylamine (0.5 ml) as described above; yield: 0.085 g (29.0%); MS m/z: 294.3 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 5.36 (s, 2H); 7.03-7.1 1 (m, 4H); 7.58 (dd, 1 H, 4J=1 .7 Hz, 3J=8.3 Hz); 7,80 (d, 1 H, 3J=8.3 Hz); 7.88 (d, 1 H, 4J=1 .7 Hz); 8.76 (s, 1 H); 8.94 (s, 1 H); HPLC (METHOD [A]): rt 8.69 min (99.2%)
Example 5: 5-(4-(4-Chlorobenzyl)-4H-1 ,2,4-triazol-3-yl)-1 H-benzo[dlimidazole
The compound was synthesized starting from 5-(1 ,3,4-oxadiazol-2-yl)benzimidazole (186 mg, 1 mmol) and 4-chlorobenzylamine (0.5 ml) as described above; yield: 0.063 g (20.4%); MS m/z: 310.4 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 5.38 (s, 2H); 7.01 -7.03 (m, 2H); 7.32-7.37 (m, 2H); 7.57 (dd, 1 H, 4J=1 .7 Hz, 3J=8.3 Hz); 7.79 (d, 1 H, 3J=8.3 Hz); 7.87 (br s, 1 H); 8.76 (s, 1 H); 8.94 (s, 1 H); HPLC (METHOD [A]): rt 9.94 min (98.2%)
Example 6: 5-(4-(3,4-Difluorobenzyl)-4H-1 ,2,4-triazol-3-yl)-1 H-benzordlimidazole
The compound was synthesized starting from 5-(1 ,3,4-oxadiazol-2-yl)benzimidazole (186 mg, 1 mmol) and 3,4-difluorobenzylamine (0.5 ml) as described above; yield: 0.049 g (15.8%); MS m/z: 312.2 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 5.36 (s, 2H); 6.81 -6.84 (m, 1 H); 7.13-7.19 (m, 1 H); 7.29-7.36 (m, 1 H); 7.59 (dd, 1 H, 4J=1 .7 Hz, 3J=8.7 Hz); 7.82 (d, 1 H, 3J=8.7 Hz); 7.88 (d, 1 H, 4J=1.7 Hz); 8.77 (s, 1 H); 9,03 (s, 1 H); HPLC (METHOD [A]): rt 8.82 min (95.6%) Example 7: 5-(4-(4-Methylbenzyl)-4H-1 ,2,4-triazol-3-yl)-1 H-benzordlimidazole The compound was synthesized starting from 5-(1 ,3,4-oxadiazol-2-yl)benzimidazole (186 mg, 1 mmol) and 4-methylbenzylamine (0.5 ml) as described above; yield: 0.071 g (24.6%); MS m/z: 290.1 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 2.20 (s, 3H); 5.32 (s, 2H); 6.89-6.91 (m, 2H); 7.06-7.08 (m, 2H); 7.58 (dd, 1 H, 4J=1 .7 Hz, 3J=8.7 Hz); 7.78-7.80 (m, 1 H); 7.88-7.89 (m, 1 H); 8.74 (s,1 H); 8.89 (s, 1 H); HPLC (METHOD [A]): rt 9.29 min (97.4%)
Example 8: 5-(4-(4-Methoxybenzyl)-4H-1 ,2,4-triazol-3-yl)-1 H-benzo[dlimidazole
The compound was synthesized starting from 5-(1 ,3,4-oxadiazol-2-yl)benzimidazole (186 mg, 1 mmol) and 4-methoxybenzylamine (0.5 ml) as described above; yield: 0.083 g (27.2%); MS m/z: 306.4 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 3.66 (s, 3H); 5.29 (s, 2H); 6.80-6.83 (m, 2H); 6.95-6.97 (m, 2H); 7.61 (dd, 1 H , 4J=1 .7 Hz, 3J=8.7 Hz); 7.82 (d, 1 H , 3J=8.7 Hz); 7.91 (d, 1 H, 4J=1.7 Hz); 8.73 (s, 1 H); 8.96 (s, 1 H); HPLC (METHOD [A]): rt 8.18 min (99.6%)
Example 9: 5-(4-(2-Chlorobenzyl)-4H-1 ,2,4-triazol-3-yl)-1 H-benzordlimidazole
The compound was synthesized starting from 5-(1 ,3,4-oxadiazol-2-yl)benzimidazole (186 mg, 1 mmol) and 2-chlorobenzylamine (0.5 ml) as described above; yield: 0.089 g (28.8%); MS m/z: 310.3 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 5.44 (s, 2H); 6.83-6.85 (m, 1 H); 7.22-7.31 (m, 2H); 7.39-7.43 (m, 1 H); 7.59 (dd, 1 H , 4J=1 .7 Hz, 3J=8.3 Hz); 7.81 (d, 1 H , 3J=8.3 Hz); 7.91 (br s, 1 H); 8.68 (s, 1 H); 9.03 (s, 1 H); H PLC (METHOD [A]): rt 8.77 min (98.9%)
Example 10: 5-(4-(4-Phenoxybenzyl)-4H-1 ,2,4-triazol-3-yl)-1 H-benzofdlimidazole
The compound was synthesized starting from H-benzimidazole-5-carboxylic acid dimethylaminomethylene-hydrazide (203mg, .88mmol), 4-phenoxy benzyl amine (350mg, 1 .76mmol), acetic acid (5mL) as described above. Yield: 50mg (15.52%)
MS m/z: 368.2 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 5.36 (s, 2H); 6.91 -6.95 (br m, 4H); 7.04-7.07 (m, 2H); 7.1 1 -7.15 (m, 1 H); 7.35-7.43 (br m, 3H); 7.68-7.70 (br m, 1 H); 7.78 (s,1 H); 8.31 (s, 1 H); 8.68 (s, 1 H); HPLC (METHOD [A]): rt 1 1.12 min (97.3%) Example 1 1 : 5-(4-(3-Chlorobenzyl)-4H-1 ,2,4-triazol-3-yl)-1 H-benzordlimidazole The compound was synthesized starting from H-benzimidazole-5-carboxylic acid dimethylaminomethylene-hydrazide (200mg, 0.86mmol), 3-chloro benzyl amine (250mg, 1 .73mmol), Acetic acid (5mL) as described above. Yield: 45mg (16.60%)
MS m/z: 310.1 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 5.40 (s, 2H); 6.94 (br s, 1 H); 7.10 (s, 1 H); 7.27-7.33 (m, 2H); 7.39-7.41 (m, 1 H); 7.68-7.70 (m, 1 H); 7.77 (s, 1 H); 8.32 (s, 1 H); 8.73 (s, 1 H); 12.75 (br s, 1 H); HPLC (METHOD [A]): rt 7.95 min (98.6%)
Example 12: 5-(4-(2.4-Dichlorobenzyl)-4H-1 ,2,4-triazol-3-yl)-1 H-benzordlimidazole
The compound was synthesized starting from H-benzimidazole-5-carboxylic acid dimethylaminomethylene-hydrazide (300mg, 1 .29mmol), 2, 4-dichloro benzyl amine (0.35mL, 2.60mmol), acetic acid (5ml_) as described above. 45mg (16.60%)
MS m/z: 344.1 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 5.43 (s, 2H); 6.87-6.89 (m, 1 H); 7.38-7.42 (m, 2H); 7.65-7.69 (m, 2H); 7.76 (s, 1 H); 8.33 (s, 1 H); 8.64 (s, 1 H); 12.72 (br s, 1 H); HPLC (METHOD [A]): rt 9.15 min (100%)
Example 13: 5-(4-(2-Chloro-6-fluorobenzyl)-4H-1 ,2,4-triazol-3-yl)-1 H-benzordlimidazole The compound was synthesized starting from H-benzimidazole-5-carboxylic acid dimethylaminomethylene-hydrazide (300mg, 1 .29mmol), 2-chloro-6-flouro benzyl amine (0.35mL, 2.60mmol), acetic acid (5mL) as described above. 45mg (10.60%)
MS m/z: 328.1 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 5.44 (s, 2H); 7.16-7.30 (br m, 2H); 7.37-7.44 (br m, 2H); 7.70-7.71 (m, 1 H); 7.87 (s, 1 H); 8.35 (s, 1 H); 8.41 (s, 1 H); 12.70 (br s, 1 H); HPLC (METHOD [A]): rt 7.12 min (100%) Example 14: 5-(4-(3,5-Dichlorobenzyl)-4H-1 ,2,4-triazol-3-yl)-1 H-benzordlimidazole
The compound was synthesized starting from H-benzimidazole-5-carboxylic acid dimethylaminomethylene-hydrazide (300mg, 1 .29mmol), 3, 5-dichloro benzyl amine (0.35mL, 2.60mmol), acetic acid (5mL) as described above. Yield: 50mg (1 1 .60%)
MS m/z: 344.1 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 5.41 (s, 2H); 7.03-7.04 (m, 2H); 7.38-7.40 (m, 1 H); 7.53 (s, 1 H); 7.69-7.71 (m, 1 H); 7.76 (s, 1 H); 8.35 (s, 1 H); 8.75 (s, 1 H); 12.79 (br s, 1 H); HPLC (METHOD [A]): rt 9.20 min (95.2%) Example 15: 5-(4-(3-Chloro-4-fluorobenzyl)-4H-1 ,2,4-triazol-3-yl)-1 H-benzo[dlimidazole The compound was synthesized starting from H-benzimidazole-5-carboxylic acid dimethylaminomethylene-hydrazide (300mg, 1 .29mmol), 3-chloro-4-flouro benzyl amine (420mg, 2.60mmol), acetic acid (5mL). as described above. Yield: 50mg (1 1 .65%)
MS m/z: 328.1 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 5.37 (s, 2H); 7.00 (br s, 1 H); 7.26- 7.41 (br m, 3H); 7.69-7.71 (m, 1 H); 7.76 (s, 1 H); 8.34 (s, 1 H); 8.72 (s, 1 H); 12.72 (br s, 1 H); HPLC (METHOD [A]): rt 8.29 min (99.1 %)
Example 16: 5-(4-(2.5-Dichlorobenzyl)-4H-1 ,2,4-triazol-3-yl)-1 H-benzoidlimidazole
The compound was synthesized starting from H-benzimidazole-5-carboxylic acid dimethylaminomethylene-hydrazide (300mg, 1.29mmol), 2,5-dichloro benzyl amine (0.35ml_, 2.60mmol), acetic acid (5mL) as described above. Yield: 40mg (8.92%)
MS m/z: 344.1 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 5.43 (s, 2H); 6.88 (s, 1 H); 7.39-7.42 (m, 2H); 7.49-7.50 (m, 1 H); 7.63-7.81 (br m, 2H); 8.33 (s, 1 H); 8.66 (s, 1 H); 12.71 (br s, 1 H); HPLC (METHOD [A]): rt 10.29 min (100%)
Example 17: 5-(4-(3,4-Dichlorobenzyl)-4H-1 ,2,4-triazol-3-yl)-1 H-benzordlimidazole
The compound was synthesized starting from H-benzimidazole-5-carboxylic acid dimethylaminomethylene-hydrazide (300mg, 1 .29mmol), 3,4-dichloro benzyl amine (.35ml_, 2.60mmol), Acetic acid (5mL) as described above. Yield: 45mg (9%)
MS m/z: 344.1 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 5.40 (s, 2H); 6.95-6.98 (m, 1 H); 7.31 -7.32 (m, 1 H); 7.39-7.41 (m, 1 H); 7.56-7.58 (m, 1 H); 7.68-7.71 (m, 1 H); 7.76 (s, 1 H); 8.34 (s, 1 H); 8.74 (s, 1 H); 12.72 (br s, 1 H); HPLC (METHOD [A]): rt 9.65 min (98.4%) Example 18: 5-(4-(2-Methoxybenzyl)-4H-1 ,2,4-triazol-3-yl)-1 H-benzoidlimidazole
The compound was synthesized starting from H-benzimidazole-5-carboxylic acid dimethylaminomethylene-hydrazide (300mg, 1 .29mmol), 2-methoxy benzyl amine (0.4mL, 2.60mmol), Acetic acid (5mL) as described above. Yield: 40mg (10%)
MS m/z: 306.1 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): 1 H-NMR (400 MHz, DMSO-d6): δ 12.76 (br s, H); 8.57 (s, H); 8.34 (s, H); 7.79 (s, H); 7.71 -7.69 (m, H); 7.45-7.42 (br m, H); 7.31 -7.26 (m, H); 7.01 -6.99 (m, H); 6.88-6.85 (m, H); 6.76-6.74 (m, H); 5.27(s, 2H); 3.70 (s, 3H), HPLC (METHOD [A]): rt 7.04 min (97.63%) Example 19: 5-(4-(2,6-Dichlorobenzyl)-4H-1 ,2,4-triazol-3-yl)-1 H-benzordlimidazole
The compound was synthesized starting from H-benzimidazole-5-carboxylic acid dimethylaminomethylene-hydrazide (300mg, 1.29mmol), 2,6-dichloro benzyl amine (0.35ml_, 2.60mmol), Acetic acid (5ml_) as described above. Yield: 50mg (1 1.15%)
MS m/z: 344.1 [M+H]+; 1 H-NMR (400 MHz, DMSO-d6): δ 12.79 (br s, H); 8.37 (s, H); 8.19 (s, H); 7.95 (s, H); 7.75- 7.73 (m, H); 7.53-7.50 (br m, 3H); 7.44-7.40 (br m, H); 5.50 (s, 2H), HPLC (METHOD [A]): rt 7.95 min (100%)
Example 20: 5-(4-(4-(Trifluoromethoxy)benzyl)-4H-1 ,2,4-triazol-3-yl)-1 H-benzo[dlimidazole The compound was synthesized starting from H-benzimidazole-5-carboxylic acid dimethylaminomethylene-hydrazide (300mg, 1 .29mmol), 4-triflouromethoxy benzyl amine (0.4ml_, 2.60mmol), acetic acid (5ml_) as described above. Yield: 40mg (8.5%)
MS m/z: 360.2 [M+H]+; 1H-NMR (400 MHz, DMSO-d6): δ 12.85 (br s, H); 8.74 (s, H); 8.37 (s, H); 7.79 (s, H); 7.70-7.68 (m, H); 7.44-7.42 (m, H); 7.32-7.30 (m, 2H); 7.18-7.14 (m, 2H); 5.43 (s, 2H), HPLC (METHOD [A]): rt 9.68 min (100%)
Example 21 : 5-(4-(2,3-Difluorobenzyl)-4H-1 ,2,4-triazol-3-yl)-1 H-benzordlimidazole
The compound was synthesized starting from H-benzimidazole-5-carboxylic acid dimethylaminomethylene-hydrazide (300mg, 1 .29mmol), 2,3-diflouro benzyl amine (0.3ml_, 2.60mmol), acetic acid (5ml_) as described above. Yield: 45mg (1 1.25%)
MS m/z: 312.1 [M+H]+; 1H-NMR (400 MHz, DMSO-d6): δ 12.81 (br s, H), 8.70 (s, H); 8.36 (s, H); 7.80 (s, H); 7.70-7.68 (m, H); 7.43-7.33 (br m, 2H); 7.27-7.23 (br m, H); 7.18-7.10 (br m, H); 6.76-6.73 (m, H); 5.49 (s, 2H), HPLC (METHOD [A]): rt 6.88 min (100%) Example 22: 5-(4-(2,3-Dichlorobenzyl)-4H-1 ,2,4-triazol-3-yl)-1 H-benzordlimidazole
The compound was synthesized starting from H-benzimidazole-5-carboxylic acid dimethylaminomethylene-hydrazide (300mg, 1.29mmol), 2,3-dichloro benzyl amine (0.35mL, 2.60mmol), acetic acid (5mL) as described above. Yield: 55mg (12%)
MS m/z: 344.0 [M+H]+; 1H-NMR (400 MHz, DMSO-d6): δ 12.79 (br, H); 8.66 (s, H); 8.34-8.33 (m, H); 7.77 (s, H); 7.69-7.67 (m, H); 7.61 -7.59(br m, H); 7.43-7.41 (m, H); 7.34-7.30 (m, H); 6.79-6.77 (m, H); 5.49(s, 2H), HPLC (METHOD [A]): rt 8.85 min (100%) Example 23: 5-(4-(2-Chloro-5-(trifluoromethyl)benzyl)-4H-1 ,2,4-triazol-3-yl)-1 H- benzofdlimidazole
The compound was synthesized starting from H-benzimidazole-5-carboxylic acid dimethylaminomethylene-hydrazide (300mg, 1.29mmol), 2-chloro-5-(trifluoro methyl)-benzyl amine (0.51 mL, 3.25mmol), acetic acid (5mL) as described above. Yield: 45mg (9.10%) MS m/z: 378.2 [M+H]+; 1H-NMR (400 MHz, DMSO-d6): δ 8.68 (s, H); 8.34 (s, H); 7.76 (s, H); 7.66-7.65 (m, 3H); 7.37-7.34 (br m, H); 7.12-7.1 1 (m, H); 5.55 (s, 2H), HPLC (METHOD [A]): rt 9.12 min (100%)
Example 24: 5-(4-(3-Chloro-4-methoxybenzyl)-4H-1 ,2,4-triazol-3-yl)-1 H-benzo[dlimidazole The compound was synthesized starting from H-benzimidazole-5-carboxylic acid dimethylaminomethylene-hydrazide (300mg, 1.29mmol), 3-chloro-4-methoxy-benzyl amine (445mg, 2.60mmol), acetic acid (5ml_) as described above. Yield: 45mg (10%)
MS m/z: 340.2 [M+H]+; 1H-NMR (400 MHz, DMSO-d6): δ 12.79 (br s, H); 8.70 (s, H); 8.33 (s, H); 7.79 (s, H); 7.71 -7.69 (m, H); 7.43-7.7.41 (m, H); 7.13-7.12 (m, H); 7.07-7.05 (m, H); 6.94-6.91 (m, H); 5.31 (s, 2H); 3.79 (s, 3H), HPLC (METHOD [A]): rt 8.32 min (98.43%)
Example 25: 5-(4-(4-Ethoxybenzyl)-4H-1 ,2,4-triazol-3-yl)-1 H-benzo[dlimidazole
The compound was synthesized starting from H-benzimidazole-5-carboxylic acid dimethylaminomethylene-hydrazide (350mg, 1 .51 mmol), 4-ethoxy benzyl amine (.570mg, 3.79mmol), acetic acid (5ml_) as described above. Yield: 70mg (14.4%)
MS m/z: 320.2 [M+H]+; 1H-NMR (400 MHz, DMSO-d6): δ 12.54 (br s, H); 8.64 (s, H); 8.26 (s, H); 8.16 (s, H); 7.87-7.84 (m, H); 7.63-7.61 (m, H); 7.33-7.30 (m, 2H); 6.93-6.91 (m, H); 5.36 (s, 2H); 4.03-3.96 (m, 2H); 1 .32-1 .28 (m, 3H), HPLC (METHOD [A]): rt 10.88 min (100%)
Example 26: 5-(5-Benzyl-1 ,3,4-oxadiazol-2-yl)-1 H-benzo[dlimidazole
The compound was synthesized starting from benzimidazol-5-carbohydrazide (176 mg, 1 mmol), TEA (0.153 ml; 1 .1 mmol), phenylacetylchloride (170 mg, 0.145 ml, 1 .1 mmol) and POCI3 (0.5 ml; 5.5 mmol) as described in method 1 ; yield: 0.026 mg (9.4%); MS m/z: 277.4 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 4.34 (s, 2H); 7.25-7.28 (m, 1 H); 7.29-7.38 (m, 4H); 7.83 (d, 1 H, 3J=8.7 Hz); 7.91 (dd, 1 H, 4J=1 .7 Hz, 3J=8.7 Hz); 8.19-8.20 (m, 1 H); 8.85 (s, 1 H); HPLC (METHOD [A]): rt 1 1 .17 min (100%) Example 27: 5-(5-Phenethyl-1 ,3,4-oxadiazol-2-yl)-1 H-benzo[dlimidazole
The compound was synthesized starting from benzimidazol-5-carbohydrazide (176 mg, 1 mmol), TEA (0.153 ml; 1.1 mmol), phenylpropionylchloride (185 mg, 0.163 ml, 1 .1 mmol) and POCIs (0.5 ml; 5.5 mmol) as described in method 1 ; yield: 0.031 mg (10.7%); MS m/z: 291 .3 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 3.09 (t, 2H, 3J=7.7 Hz); 3.25 (t, 2H, 3J=7.5 Hz); 7.15-7.19 (m, 1 H); 7.23-7.27 (m, 4H); 7.84 (d, 1 H , 3J=8.7 Hz); 7.92 (dd, 1 H , 4J=1 .7 Hz, 3J=8.7 Hz); 8.20 (br s, 1 H); 8.87 (s, 1 H); HPLC (METHOD [A]): rt 1 1.82 min (100%)
Example 28: 5-(5-(2-Methylphenethyl)-1 ,3,4-oxadiazol-2-yl)-1 H-benzo[dlimidazole
The compound was synthesized starting from benzimidazol-5-carbohydrazide (176 mg, 1 mmol) and 3-(4-methylphenyl)propionic acid (164 mg; 1 mmol) as described in method 2; yield: 0.025 g (8.2%); MS m/z: 305.3 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 2.31 (s, 3H); 3.1 1 (t, 2H, 3J=7.9 Hz); 3.23 (t, 2H, 3J=7.9 Hz), 7.10-7.21 (m, 4H); 7.87 (d, 1 H, 3J=8.7 Hz); 7.95 (dd, 1 H, 4J=1 .7 Hz, 3J=8.7 Hz); 8.23 (d, 1 H, 4J=1 .7 Hz); 8.90 (s, 1 H); HPLC (METHOD [A]): rt 12.32 min (96.5%)
Example 29: 5-(5-(3,4-Dimethoxyphenethyl)-1 ,3,4-oxadiazol-2-yl)-1 H-benzo[dlimidazole The compound was synthesized starting from benzimidazol-5-carbohydrazide (176 mg, 1 mmol), TEA (0.153 ml; 1 .1 mmol), 3,4-dimethoxyphenylpropionylchloride (228 mg, 1.1 mmol) and POCI3 (0.5 ml; 5.5 mmol) as described in method 1 ; yield: 0.019 mg (5.4%); MS m/z: 351 .3 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 3.04 (t, 2H, 3J=7.5 Hz); 3.23 (t, 2H, 3J=7.5 Hz); 3.69 (s, 6H); 6.78 (dd, 1 H, 4J=1 .7 Hz, 3J=8.3 Hz); 6.84 (d, 1 H, 3J=8.3 Hz); 6.90 (d, 1 H, 4J=1 .7 Hz); 7.76 (d, 1 H, 3J=8.3 Hz); 7.83 (dd, 1 H, 4J=1 .7 Hz, 3J=8.3 Hz); 8.17 (br s, 1 H), 8.40 (s, 1 H); HPLC (METHOD [A]): rt 10.29 min (94.2%)
Example 30: 5-(5-(2,4-Dimethoxyphenethyl)-1 ,3,4-oxadiazol-2-yl)-1 H-benzo[dlimidazole The compound was synthesized starting from benzimidazol-5-carbohydrazide (176 mg, 1 mmol), TEA (0.153 ml; 1 .1 mmol), 2,4-dimethoxyphenylpropionylchloride (228 mg, 1.1 mmol) and POCI3 (0.5 ml; 5.5 mmol) as described in method 1 ; yield: 0.013 mg (3.7%); MS m/z: 351 .3[M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 2.97-3.00 (m, 2H); 3.1 1 -3.13 (m, 2H); 3.71 (s, 6H); 6.41 -6.43 (m, 1 H); 6.51 (br s, 1 H); 7.06-7.08 (m, 1 H); 7.75-7.77 (m, 1 H); 7.81 -7.83 (m, 1 H); 8.15 (br s, 1 H); 8.43 (s, 1 H); HPLC (METHOD [A]): rt 12.06 min (100%) Example 31 : 5-(5-(2,3-Dimethoxyphenethyl)-1 ,3,4-oxadiazol-2-yl)-1 H-benzo[dlimidazole The compound was synthesized starting from benzimidazol-5-carbohydrazide (176 mg, 1 mmol), TEA (0.153 ml; 1 .1 mmol), 2,3-dimethoxyphenylpropionylchloride (228 mg, 1.1 mmol) and POCI3 (0.5 ml; 5.5 mmol) as described in method 1 ; yield: 0.016 mg (4.6%); MS m/z: 351 .3 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 3.06-3.10 (m, 2H); 3.17-3.21 (m, 2H); 3.73 (s, 3H); 3.79 (s, 3H); 6.82-6.84 (m, 1 H); 6.90-6.92 (m, 1 H); 6.95-6.99 (m, 1 H); 7.75-7.77 (m, 1 H); 7.80-7.82 (m, 1 H); 8.14 (br s, 1 H); 8.41 (s, 1 H); HPLC (METHOD [A]): rt 1 1 .49 min (94.5%) Example 32: 5-(5-(2-(Benzordin ,3ldioxol-5-yl)ethyl)-1 ,3,4-oxadiazol-2-yl)-1 H- benzofdlimidazole
The compound was synthesized starting from benzimidazol-5-carbohydrazide (176 mg, 1 mmol), TEA (0.153 ml; 1 .1 mmol), 2,3-dimethoxyphenylpropionylchloride (228 mg, 1.1 mmol) and POCI3 (0.5 ml; 5.5 mmol) as described in method 1 ; yield: 0.016 mg (4.6%); MS m/z: 351 .3 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 3.06-3.10 (m, 2H); 3.17-3.21 (m, 2H); 3.73 (s, 3H); 3.79 (s, 3H); 6.82-6.84 (m, 1 H); 6.90-6.92 (m, 1 H); 6.95-6.99 (m, 1 H); 7.75-7.77 (m, 1 H); 7.80-7.82 (m, 1 H); 8.14 (br s, 1 H); 8.41 (s, 1 H); HPLC (METHOD [A]): rt 1 1 .49 min (94.5%) Example 33: 5-(5-(2,4-Dichlorophenethyl)-1 ,3,4-oxadiazol-2-yl)-1 H-benzo[dlimidazole
The compound was synthesized starting from benzimidazol-5-carbohydrazide (176 mg, 1 mmol) and 3-(2,4-dichlorophenyl)propionic acid (220 mg; 1 mmol) as described in method 2; yield: 0.019 g (5.3%); MS m/z: 359.2 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 3.20-3.24 (m, 2H); 3.26-3.30 (m, 2H); 7.38 (dd, 1 H, 4J=2.1 Hz, 3J=8.3 Hz); 7.47 (d, 1 H, 3J=8.3 Hz); 7.59 (d, 1 H, 4J=2.1 Hz); 7.87 (d, 1 H, 3J=8.3 Hz); 7.93 (dd, 1 H, 4J=1 .7 Hz, 3J=8.3 Hz); 8.23 (br s, 1 H); 8.87 (s, 1 H); HPLC (METHOD [A]): rt 14.21 min (97.6%)
Example 34: 5-(5-(4-(Trifluoromethyl)phenethyl)-1 ,3,4-oxadiazol-2-yl)-1 H-benzo[dlimidazole The compound was synthesized starting from benzimidazol-5-carbohydrazide (176 mg, 1 mmol) and 3-(4-trifluoromethylphenyl)propionic acid (21 8 mg; 1 mmol) as described in method 2; yield: 0.043 mg (12.0%); MS m/z: 359.2 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 3.21 -3.25 (m, 2H); 3.31 -3.35 (m, 2H); 7.54-7.56 (m, 2H); 7.64-7.67 (m, 2H); 7.86 (d, 1 H , 3J=8.7 Hz); 7.92 (dd, 1 H, 4J=1 .7 Hz, 3J=8.7 Hz); 8.24 (d, 1 H, 4J=1.7 Hz); 8.87 (s, 1 H); HPLC (METHOD [A]): rt 13.82 min (95.9%) Example 35: 5-(5-(3,4,5-Trimethoxyphenethyl)-1 ,3,4-oxadiazol-2-yl)-1 H-benzo[dlimidazole The compound was synthesized starting from benzimidazol-5-carbohydrazide (176 mg, 1 mmol) and 3-(3,4,5-trimethoxyphenyl)propionic acid (241 mg; 1 mmol) as described in method 2; yield: 0.016 g (4.2%); MS m/z: 381 .4 [M+H]+; 1H-NMR (CD3OD, 400 MHz): δ 3.12- 3.16 (m, 2H); 3.31 -3.33 (m, 2H); 3.70 (s, 3H), 3.77 (s, 6H); 6.56 (s, 2H); 7.94 (d, 1 H, 3J=8.7 Hz); 8.15 (dd, 1 H, 4J=1.7 Hz, 3J=8.7 Hz); 8.381 -8.382 (m, 1 H); 9.12 (s, 1 H); HPLC (METHOD [A]): rt 10.41 min (89.6%) Example 36: 5-(5-(2,4-Difluorophenethyl)-1 ,3,4-oxadiazol-2-yl)-1 H-benzordlimidazole
The compound was synthesized starting from benzimidazol-5-carbohydrazide (176 mg, 1 mmol) and 3-(2,4-difluorphenyl)propionic acid (186 mg, 1 mmol) as described in method 2; yield: 0.018 g (5.5%); MS m/z: 327.2 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 3.1 1 -3.15 (m, 2H); 3.24-3.27 (m, 2H); 7.00-7.05 (m, 1 H); 7.16-7.21 (m, 1 H); 7.41 -7.47 (m, 1 H); 7.86 (d, 1 H, 3J=8.3 Hz); 7.93 (dd, 1 H , 4J=1 .7 Hz, 3J=8.3 Hz); 8.22 (br s, 1 H); 8.86 (s, 1 H ); H PLC (METHOD [A]): rt 12.06 min (100%)
Example 37: 5-(5-(2,5-Bis(trifluoromethyl)phenethyl)-1 ,3,4-oxadiazol-2-yl)-1 H- benzofdlimidazole
The compound was synthesized starting from benzimidazol-5-carbohydrazide (176 mg, 1 mmol) and 3-(2,5-trifluoromethylphenyl)propionic acid (287 mg; 1 mmol) as described in method 2; yield: 0.009 g (2.1 %); MS m/z: 427.3 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 3.35-3.38 (m, 4H); 7.83-7.87 (m, 2H); 7.90 (dd, 1 H , 4J=1 .7 Hz, 3J=8.7 Hz); 7.96 (d, 1 H , 3J=8.3 Hz); 8.04 (br s, 1 H); 8.20 (br s, 1 H); 8.81 (s, 1 H); HPLC (METHOD [A]): rt 15.36 min (95.5%)
Example 38: 5-(5-(4-(Methylthio)phenethyl)-1 ,3,4-oxadiazol-2-yl)-1 H-benzo[dlimidazole The compound was synthesized starting from benzimidazol-5-carbohydrazide (176 mg, 1 mmol) and 3-(4-methylthiophenyl)propionic acid (197 mg; 1 mmol) as described in method 2; yield: 0.057 g (16.9%); MS m/z: 337.4 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 2.43 (s, 3H); 3.08 (t, 2H, 3J=7.5 Hz); 3.25 (t, 2H, 3J=7.5 Hz); 7.17-7.19 (m, 2H); 7.23-7.25 (m, 2H); 7.86 (d, 1 H, 3J=8.3 Hz); 7.93 (dd, 1 H, 4J=1 .7 Hz, 3J=8.3 Hz); 8.24 (d, 1 H, 4J=1.7 Hz); 8.85 (s, 1 H); HPLC (METHOD [A]): rt 12.85 min (99.2%) Example 39: 5-(5-(3-Chloro-4-methoxyphenethyl)-1 ,3,4-oxadiazol-2-yl)-1 H- benzofdlimidazole
The compound was synthesized starting from benzimidazol-5-carbohydrazide (176 mg, 1 mmol) and 3-(3-chloro-4-methoxyphenyl)propionic acid (215 mg; 1 mmol) as described in method 2; yield: 0.071 g (20.1 %); MS m/z: 355.4/357.1 [M+H]+; 1H-N MR (DMSO d6, 400 MHz): δ 3.06-3.08 (m, 2H); 3.24-3.27 (m, 2H); 3.80 (s, 3H); 7.04-7.06 (m, 1 H); 7.21 -7.23 (m, 1 H); 7.39 (br s, 1 H); 7.87-7.89 (m, 1 H); 7.94-7.96 (m, 1 H); 8.25 (br s, 1 H); 8.92 (s, 1 H); HPLC (METHOD [A]): rt 12.55 min (95.7%)
Example 40: 5-(5-(Phenoxymethyl)-1 ,3,4-oxadiazol-2-yl)-1 H-benzo[dlimidazole
The compound was synthesized starting from Benzimidazol-5-carbohydrazide (176 mg, 1 mmol) and Phenoxyacetic acid (153 mg; 1 mmol) as described in method 2 but purified by flashchromatography on silica using a CHCI3/MeOH gradient; yield: 0.049 g (16.8%); MS m/z: 293.2 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): □ 5.48 (s, 2H); 7.00-7.03 (m, 1 H); 7.1 1 - 7.13 (m, 2H); 7.32-7.36 (m, 2H); 7.78 (d, 1 H, 3J=8.3 Hz); 7.86 (dd, 1 H, 4J=1.7 Hz, 3J=8.3 Hz); 8.20 (br s, 1 H); 8.40 (s, 1 H); 12.82 (br s, 1 H); HPLC (METHOD [A]): rt 1 1 .29 min (100%)
Example 41 : 5-(5-((Phenylthio)methyl)-1 ,3,4-oxadiazol-2-yl)-1 H-benzo[dlimidazole
The compound was synthesized starting from benzimidazol-5-carbohydrazide (176 mg, 1 mmol) and phenylthioacetic acid (169 mg; 1 mmol) as described in method 2 but purified by flash chromatography on silica using a CHCI3/MeOH gradient; yield: 0.052 g (16.9%); MS m/z: 309.3 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 4.59 (s, 2H); 7.25-7.30 (m, 1 H); 7.34- 7.38 (m, 2H); 7.47-7.49 (m, 2H); 7.72-7.78 (m, 2H); 8.07 (br s, 1 H); 8.39 (s, 1 H); 12.79 (br s, 1 H); HPLC (METHOD [A]): rt 1 1 .59 min (100%)
Example 42: 5-(5-((Phenylsulfonyl)methyl)-1 ,3,4-oxadiazol-2-yl)-1 H-benzo[dlimidazole
The compound was synthesized starting from benzimidazol-5-carbohydrazide (176 mg, 1 mmol) and 4-phenylsulfonyacetic acid (201 mg; 1 mmol) as described in method 2; yield: 0.066 g (19.4%); MS m/z: 341 .1 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 5.43 (s, 2H); 7.66- 7.70 (m, 2H); 7.79-7.84 (m, 2H); 7.87-7.91 (m, 3H); 8.09 (br s, 1 H); 8.89 (s, 1 H); HPLC (METHOD [A]): rt 9.48 min (98.8%) Example 43: 5-(5-((Pyridin-4-ylthio)methyl)-1 ,3,4-oxadiazol-2-yl)-1 H-benzo[dlimidazole
The compound was synthesized starting from benzimidazol-5-carbohydrazide (176 mg, 1 mmol) and (4-pyridyl)thioacetic acid (170 mg; 1 mmol) as described in method 2 but purified by flash chromatography on silica using a CHCI3/MeOH gradient; yield: 0.026 g (8.4%); MS m/z: 310.1 [M+H]+; 200.1 ([M-C5H4NS)«]+; 155.7 [M+2H]2+; 1H-NMR (DMSO d6, 400 MHz): δ 4.98 (s, 2H); 7.82-7.86 (m, 3H); 7.89 (dd, 1 H, 4J=1 .7 Hz, 3J=8.7 Hz); 8.20 (br s, 1 H); 8.60- 8.61 (m, 2H); 8.77 (s, 1 H); HPLC (METHOD [A]): rt doublepeak 4.60 min (60.0%), 4.96 min (39.7%)
Example 44: 5-(5-((3,4-Dimethoxyphenylthio)methyl)-1 ,3,4-oxadiazol-2-yl)-1 H- benzofdlimidazole
The compound was synthesized starting from benzimidazol-5-carbohydrazide (176 mg, 1 mmol) and 3,4-dimethoxyphenylthioacetic acid (229 mg; 1 mmol) as described in method 2; yield: 0.040 g (10.9%) ; MS m/z: 369.1 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 3.69 (s, 3H); 3.73 (s, 3H); 4.47 (s, 2H); 6.91 (d, 1 H, 3J=8.3 Hz); 7.00 (dd, 1 H, 4J=2.1 Hz,3J=8.3 Hz); 7.04 (d, 1 H, 4J=2.1 Hz); 7.86-7.87 (m, 2H); 8.17 (br s, 1 H); 8.85 (s, 1 H); HPLC (METHOD [A]): rt 10.99 min (99.1 %)
Example 45: 5-(5-((3,4-Dimethoxyphenylsulfonyl)methyl)-1 ,3,4-oxadiazol-2-yl)-1 H- benzofdlimidazole
The compound was synthesized starting from benzimidazol-5-carbohydrazide (176 mg, 1 mmol) and 3,4-dimethoxyphenylsulfonylthioacetic acid (261 mg; 1 mmol) as described in method 2 but purified by flash chromatography on silica using a CHCI3/MeOH gradient; yield: 0.022 g (5.5%); MS m/z: 401.2 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 3.75 (s, 3H); 3.82 (s, 3H); 5.29 (s, 2H); 7.15 (d, 1 H 3J=8.7 Hz); 7.328-7.333 (m, 1 H); 7.38 (dd, 1 H, 4J=2.1 Hz, 3J=8.7 Hz); 7.68-7.72 (m, 1 H); 7.78-7.82 (m, 1 H); 8.01 (br s, 1 H); 8.38 (s, 1 H); 12.80 (br s, 1 H); HPLC (METHOD [A]): rt 10.19 min (90.6%)
Example 46: 5-(5-Phenethyl-1 ,3,4-thiadiazol-2-yl)-1 H-benzo[dlimidazole
The compound was synthesized starting from benzimidazol-5-carbohydrazide (176 mg; 1 mmol), TEA (0.1 53 ml ; 1 .1 mmol), phenylpropionylchloride (0.1 63 ml ; 1 .1 mmol) and Lawesson's reagent (606 mg; 1 .5 mmol) as described in method 1 ; yield: 0.067 g (21 .9%); MS m/z: 307.2 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 3.10 (t, 2H, 3J=7.9 Hz); 3.45 (t, 2H, 3J=7.9 Hz); 7.18-7.22 (m, 1 H); 7.29-7.30 (m, 4H); 7.71 -7.75 (m, 2H); 8.1 1 (br s, 1 H); 8.35 (s, 1 H); 12.70 (br s, 1 H); HPLC (METHOD [A]): rt 12.49 min 94.7%)
Example 47: 5-(5-(3,4-Dimethoxyphenethyl)-1 ,3,4-thiadiazol-2-yl)-1 H-benzofdlimidazole The compound was synthesized starting from benzimidazol-5-carbohydrazide (176 mg; 1 mmol), TEA (0.153 ml; 1 .1 mmol), 3,4-dimethoxyphenylpropionylchloride (228 mg, 1 .1 mmol) and Lawesson 's reagent (606 mg; 1 .5 mmol) as described in method 1 ; yield: 0.017 g (4.6%); MS m/z: 367.2 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 3.00 (t, 2H, 3J=7.5 Hz); 3.40 (t, 2H, 3J=7.5 Hz); 3.68 (s, 3H); 3.70 (s, 3H); 6.75-6.77 (m, 1 H); 6.81 -6.83 (m, 1 H); 6.89 (br s, 1 H); 7.67-7.69 (m, 1 H); 7.73-7.75 (m, 1 H); 8.09 (br s, 1 H); 8.34 (s, 1 H; HPLC (METHOD [A]): rt 10.82 min (100%)
Example 48: 5-(5-(2,4-Dimethoxyphenethyl)-1 ,3,4-thiadiazol-2-yl)-1 H-benzo[dlimidazole The compound was synthesized starting from benzimidazol-5-carbohydrazide (176 mg; 1 mmol), TEA (0.153 ml; 1 .1 mmol), 2,4-dimethoxyphenylpropionylchloride (228 mg, 1 .1 mmol) and Lawesson 's reagent (606 mg; 1 .5 mmol) as described in method 1 ; yield: 0.022 g (6.0%); MS m/z: 367.2 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 2.94 (t, 2H, 3J=7.9 Hz); 3.30 (t, 2H, 3J=7.9 Hz); 3.70 (s, 3H); 3.74 (s, 3H); 6.41 (dd, 1 H, 4J=2.5 Hz, 3J=8.3 Hz); 6.51 (d, 1 H, 4J=2.5 Hz); 7.05 (d, 1 H, 3J=8.3 Hz); 7.68 (d, 1 H, 3J=8.7 Hz); 7.74 (dd, 1 H, 4J=1 .7 Hz, 3J=8.7 Hz); 8.09 (br s, 1 H); 8.34 (s, 1 H); HPLC (METHOD [A]): rt 12.63 min (97.6%)
Example 49: 5-(5-(2,3-Dimethoxyphenethyl)-1 ,3,4-thiadiazol-2-yl)-1 H-benzo[dlimidazole The compound was synthesized starting from benzimidazol-5-carbohydrazide (176 mg; 1 mmol), TEA (0.153 ml; 1 ,1 mmol), 2,3-dimethoxyphenylpropionylchloride (228 mg, 1 .1 mmol) and Lawesson 's reagent (606 mg; 1 .5 mmol) as described in method 1 ; yield: 0.025 g (6.8%); MS m/z: 367.2 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 3.07 (t, 2H, 3J=7.9 Hz); 3.38 (t, 2H, 3J=7.9 Hz); 3.73 (s, 3H); 3.79 (s, 3H); 6.84 (dd, 1 H, 4J=1 .7 Hz, 3J=8.3 Hz); 6.91 (dd, 1 H, 4J = 1 .7 Hz, 3J=8.3 Hz); 6.98 (t, 1 H, 3J=8.3 Hz); 7.71 (d, 1 H, 3J=8.3 Hz); 7.77 (dd, 1 H, 4J=1 .7 Hz, 3J=8.3 Hz); 8.12 (d, 1 H, 4J=1 .7 Hz); 8.37 (s, 1 H); HPLC (METHOD [A]): rt 12.03 min (95.8%)
Example 50: 5-(5-(4-(Methylthio)phenethyl)-1 ,3,4-thiadiazol-2-yl)-1 H-benzo[dlimidazole The compound was synthesized starting from 3-(4-methylthiophenyl)propionic acid (197 mg; 1 mmol), DCC (206 mg; 1 mmol), benzimidazol-5-carbohydrazide (176 mg; 1 mmol) and Lawesson 's reagent (606 mg; 1 .5 mmol) as described in method 2; yield: 0.029 mg (8.2%); MS m/z: 353.3 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 2.43 (s, 3H); 3.07 (t, 2H, 3J=7.5 Hz); 3.44 (t, 2H, 3J=7.5 Hz); 7.17-7.20 (m, 2H); 7.24-7.26 (m, 2H); 7.83 (d, 1 H, 3J=8.7 Hz); 7.92 (dd, 1 H, 4J=1 .7 Hz, 3J=8.7 Hz); 8.22 (br s, 1 H); 8.93 (s, 1 H); HPLC (METHOD [A]): rt 13.92 min (99.1 %)
Example 51 : 5-(5-(2,4-Dichlorophenethyl)-1 ,3,4-thiadiazol-2-yl)-1 H-benzo[dlimidazole
The compound was synthesized starting from 3-(2,4-dichlorophenyl)propionic acid (220 mg; 1 mmol), benzimidazol-5-carbohydrazide (176 mg; 1 mmol), Lawesson 's reagent (606 mg; 1 .5 mmol) and POCI3 (0.137 ml; 1 .5 mmol) as described in method 3; yield: 0.016 g (4.3%); MS m/z: 375.3 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 3.21 (t, 2H, 3J=7.5 Hz); 3,46 (t, 2H, 3J=7.5 Hz); 7.38 (dd, 1 H, 4J=2.1 Hz, 3J=8.3 Hz); 7.46 (d, 1 H, 3J=8.3 Hz); 7.61 (d, 1 H, 4J=2.1 Hz); 7.83 (d, 1 H, 3J=8.7 Hz); 7.92 (dd, 1 H; 4J=1.7 Hz, 3J=8.7 Hz); 8.23 (d, 1 H, 4J=2.1 Hz); 8.87 (s, 1 H); HPLC (METHOD [A]): rt 15.27 min (90.7%)
Example 52: 5-(5-(3-Chloro-4-methoxyphenethyl)-1 ,3,4-thiadiazol-2-yl)-1 H- benzofdlimidazole
The compound was synthesized starting from 3-(3-chloro-4-methoxyphenyl)propionic acid (215 mg; 1 mmol), DCC (206 mg; 1 mmol), benzimidazol-5-carbohydrazide (176 mg; 1 mmol) and Lawesson's reagent (606 mg; 1.5 mmol) as described in method 2; yield: 0.032 g (8.6%); MS m/z: 371 .1 /373.2 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 3.05 (t, 2H, 3J=7.5 Hz); 3.44 (t, 2H, 3J=7.5 Hz); 3.81 (s, 3H); 7.05 (d, 1 H, 3J=8.7 Hz); 7.22 (dd, 1 H, 4J=2.1 Hz, 3J=8.7 Hz); 7.39 (d, 1 H, 4J=2.1 Hz); 7.83 (d, 1 H, 3J=8.7 Hz); 7.92 (dd, 1 H, 4J=1 .7 Hz, 3J=8.7 Hz); 8.22 (d, 1 H, 4J=1.7 Hz); 8.93 (s, 1 H); HPLC (METHOD [A]): rt 13.65 min (98.9%)
Example 53: 5-(5-(4-(Trifluoromethyl)phenethyl)-1 ,3,4-thiadiazol-2-yl)-1 H-benzo[dlimidazole The compound was synthesized starting from 3-(4-trifluoromethylphenyl)propionic acid (219 mg; 1 mmol), benzimidazol-5-carbohydrazide (176 mg; 1 mmol), Lawesson 's reagent (606 mg; 1 .5 mmol) and POCI3 (0.137 ml; 1 .5 mmol) as described in method 3; yield: 0.017 g (4.5%);; MS m/z: 375.1 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 3.22 (t, 2H, 3J=7.5 Hz); 3.52 (t, 2H, 3J=7.5 Hz); 7.53-7.55 (m, 2H); 7.65-7.67 (m, 2H); 7.84 (d, 1 H, 3J=8.7 Hz); 7.92 (dd, 1 H, 4J=1 .7 Hz, 3J=8.7 Hz); 8.22-8.23 (m, 1 H); 8.93 (s, 1 H); HPLC (METHOD [A]): rt 14.82 min (98.3%) Example 54: 5-(5-(2-(Benzordin ,3ldioxol-5-yl)ethyl)-1 ,3,4-thiadiazol-2-yl)-1 H- benzofdlimidazole
The compound was synthesized starting from 3-benzo(1 ,3)dioxol-3-yl-propionic acid (195 mg; 1 mmol), DCC (206 mg; 1 mmol), benzimidazol-5-carbohydrazide (176 mg; 1 mmol) and Lawesson 's reagent (606 mg; 1 .5 mmol) as described in method 2; yield: 0.019 g (5.4%); MS m/z: 351 .3 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 3.02 (t, 2H, 3J=7.5 Hz); 3.42 (t, 2H, 3J=7.5 Hz); 5.96 (s, 2H); 6.73 (dd, 1 H, 4J=1 .7 Hz, 3J=7.9 Hz); 6.81 (d, 1 H, 3J=7.9 Hz); 6.92 (d, 1 H, 4J=1 .7 Hz); 7.83 (d, 1 H, 3J=8.3 Hz); 7.92 (dd, 1 H, 4J=1 .7 Hz, 3J=8.3 Hz); 8.22 (d, 1 H, 4J=1.7 Hz); 8.93 (s, 1 H); HPLC (METHOD [A]): rt 12.40 min (99.4%)
Example 55: 5-(5-(3,4,5-Trimethoxyphenethyl)-1 ,3,4-thiadiazol-2-yl)-1 H-benzo[dlimidazole The compound was synthesized starting from 3-(3,4,5-trimethoxyphenyl)propionic acid (241 mg; 1 mmol), DCC (206 mg; 1 mmol), benzimidazol-5-carbohydrazide (176 mg; 1 mmol) and Lawesson 's reagent (606 mg; 1 .5 mmol) as described in method 2; yield: 0.033 g (8.3%); MS m/z: 397.3 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 3.04 (t, 2H , 3J=7.9 Hz); 3.47 (t, 2H, 3J=7.9 Hz); 3.62 (s, 3H); 3.74 (s, 6H); 6.62 (s, 2H); 7.83 (d, 1 H , 3J=8.7 Hz); 7.93 (dd, 1 H, 4J=1 .7 Hz, 3J=8.7 Hz); 8.22 (d, 1 H, 4J=1 .7 Hz); 8.91 (s, 1 H); HPLC (METHOD [A]): rt 1 1 .54 min (98.2%) Example 56: 5-(5-(2,5-Bis(trifluoromethyl)phenethyl)-1 ,3,4-thiadiazol-2-yl)-1 H- benzofdlimidazole
The compound was synthesized starting from 3-(2,5-bis(trifluoromethyl)phenyl)propionic acid (287 mg; 1 mmol), benzimidazol-5-carbohydrazide (176 mg; 1 mmol), Lawesson 's reagent (606 mg; 1 .5 mmol) and POCI3 (0.137 ml; 1.5 mmol) as described in method 3; yield: 0.013 g (2.9%); MS m/z: 443.4 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 3.36-3.40 (m, 2H); 3.51 - 3.55 (m, 2H); 7.82-7.85 (m, 2H); 7.92 (dd, 1 H, 4J=1 .7 Hz, 3J=8.3 Hz); 7.97 (d, 1 H, 3J=8.3 Hz); 8.06 (br s, 1 H); 8.23-8.24 (m, 1 H); 8.88 (s, 1 H); HPLC (METHOD [A]): rt 16.42 min (96.7%)
Example 57: 5-(5-(2,4-Difluorophenethyl)-1 ,3,4-thiadiazol-2-yl)-1 H-benzo[dlimidazole
The compound was synthesized starting from 3-(2,4-difluorophenyl)propionic acid (187 mg; 1 mmol), benzimidazol-5-carbohydrazide (176 mg; 1 mmol), Lawesson 's reagent (606 mg; 1 .5 mmol) and POCI3 (0.137 ml; 1 .5 mmol) as described in method 3; yield: 0.009 g (2.6%); MS m/z: 342.2 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 3.12 (t, 2H, 3J=7.5 Hz); 3.44 (t, 2H , 3J=7.5 Hz); 7.02-7.05 (m, 1 H); 7.17-7.22 (m, 1 H); 7.40-7.46 (m, 1 H); 7.79 (d, 1 H, 3J=8.7 Hz); 7.87 (dd, 1 H, 4J=1 .7 Hz, 3J=8.7 Hz); 8.19 (d, 1 H, 4J=1 .7 Hz); 8.72 (s, 1 H); HPLC (METHOD [A]): rt 12.69 min (85.5%)
Example 58: 5-(5-(3,4-Difluorophenethyl)-1 ,3,4-thiadiazol-2-yl)-1 H-benzo[dlimidazole
The compound was synthesized starting from 3-(3,4-difluorophenyl)propionic acid (186 mg; 1 mmol), benzimidazol-5-carbohydrazide (176 mg; 1 mmol), Lawesson 's reagent (606 mg; 1 .5 mmol) and POCI3 (0.137 ml; 1 .5 mmol) as described in method 3; yield: 0.02 g (5.8%); MS m/z: 343.3 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 3.1 1 (t, 2H, 3J=7.5 Hz); 3.47 (t, 2H, 3J=7.5 Hz); 6.97-6.99 (m, 1 H); 7.32-7.35 (m, 1 H); 7.40-7.46 (m, 1 H); 7.82 (d, 1 H, 3J=8.7 Hz); 7.91 (dd, 1 H, 4J=1 .7 Hz, 3J=8.7 Hz); 8.22 (d, 1 H, 4J=1 .7 Hz), 8.87 (s, 1 H); HPLC (METHOD [A]): rt 13.09 min (85.7%)
Example 59: 5-(5-(3-Fluorophenethyl)-1 ,3,4-thiadiazol-2-yl)-1 H-benzo[dlimidazole
The compound was synthesized starting from 3-(3-fluorophenyl)propionic acid (169 mg; 1 mmol), benzimidazol-5-carbohydrazide (176 mg; 1 mmol), Lawesson 's reagent (606 mg; 1 .5 mmol) and POCI3 (0.137 ml; 1 .5 mmol) as described in method 3; yield: 0.015 g (4.6%); MS m/z: 325.3 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 3.14 (t, 2H, 3J=7.5 Hz); 3.48 (t, 2H, 3J=7.5 Hz); 6.96-7.05 (m, 2H); 7.13-7.17 (m, 1 H); 7.30-7.36 (m, 1 H); 7.81 (d, 1 H, 3J=8.3 Hz); 7.89 (dd, 1 H, 4J=1 .7 Hz, 3J=8.3 Hz); 8.20-8.21 (m, 1 H); 8.84 (s, 1 H); HPLC (METHOD [A]): rt 12.67 min (97.1 %)
Example 60: 5-(5-(3-Methoxyphenethyl)-1 ,3,4-thiadiazol-2-yl)-1 H-benzo[dlimidazole
The compound was synthesized starting from 3-(3-methoxyphenyl)propionic acid (181 mg; 1 mmol), DCC (206 mg; 1 mmol), benzimidazol-5-carbohydrazide (176 mg; 1 mmol) and Lawesson 's reagent (606 mg; 1 .5 mmol) as described in method 2; yield: 0.019 g (5.7%); MS m/z: 337.3 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 3.08 (t, 2H, 3J=7.5 Hz); 3.46 (t, 2H, 3J=7.5 Hz); 3.72 (s, 3H); 6.76-6.78 (m, 1 H); 6.85-6.88 (m, 2H); 7.20 (t, 1 H, 3J=7.9 Hz); 7.81 (d, 1 H, 3J=8.7 Hz); 7.89 (dd, 1 H, 4J=1 .7 Hz, 3J=8.7 Hz); 8.20 (d, 1 H, 4J=1 .7 Hz); 8.83 (s, 1 H); HPLC (METHOD [A]): rt 12.24 min (95.9%)
Example 61 : 5-(5-(2-Methoxyphenethyl)-1 ,3,4-thiadiazol-2-yl)-1 H-benzo[dlimidazole
The compound was synthesized starting from 3-(2-methoxyphenyl)propionic acid (181 mg; 1 mmol), DCC (206 mg; 1 mmol), benzimidazol-5-carbohydrazide (176 mg; 1 mmol) and Lawesson 's reagent (606 mg; 1 .5 mmol) as described in method 2; yield: 0.012 g (3.6%); MS m/z: 337.4 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 3.06 (t, 2H, 3J=7.5 Hz); 3.39 (t, 2H, 3J=7.5 Hz); 3.79 (s, 3H); 6.84-6.88 (m, 1 H); 6.97-6.99 (m, 2H); 7.18-7.20 (m, 1 H); 7.80 (d, 1 H, 3J=8.7 Hz); 7.88 (dd, 1 H, 4J=1 .7 Hz, 3J=8.7 Hz); 8.19-8.20 (m, 1 H); 8.78 (s, 1 H); HPLC (METHOD [A]): rt 12.78 min (92.7%)
Example 62: 5-(5-(2,5-Dimethoxyphenethyl)-1 ,3,4-thiadiazol-2-yl)-1 H-benzo[dlimidazole The compound was synthesized starting from 3-(2,5-dimethoxyphenyl)propionic acid (210 mg; 1 mmol), DCC (206 mg; 1 mmol), benzimidazol-5-carbohydrazide (176 mg; 1 mmol) and Lawesson 's reagent (606 mg; 1 .5 mmol) as described in method 2; yield: 0.04 g (10.9%); MS m/z: 367.3 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 3.03 (t, 2H, 3J=7.5 Hz); 3.38 (t, 2H, 3J=7.5 Hz); 3.66 (s, 3H); 3.73 (s, 3H); 6.76 (dd, 1 H, 4J=2.9 Hz, 3J=9.1 Hz); 6.82 (d, 1 H , 4J=2.9 Hz); 6.89 (d, 1 H, 3J=9.1 Hz); 7.81 (d, 1 H, 3J=8.7 Hz); 7.89 (dd, 1 H, 4J=1 .7 Hz, 3J=8.7 Hz); 8;21 (d, 1 H, 4J=1.7 Hz); 8.82 (s, 1 H); HPLC (METHOD [A]): rt 12.62 min (93.1 %) Example 63: 5-(5-(2-Chloro-3-methoxyphenethyl)-1 ,3,4-thiadiazol-2-yl)-1 H- benzofdlimidazole
The compound was synthesized starting from 3-(2-Chlor-3-methoxyphenyl)propionic acid (215 mg; 1 mmol), DCC (206 mg; 1 mmol), Benzimidazol-5-carbohydrazide (176 mg; 1 mmol) and Lawesson's reagent (606 mg; 1 .5 mmol) as described in method 2; yield: 0.015 g (4.1 %); MS m/z: 371.3 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 3.20-3.24 (m, 2H); 3.42- 3.46 (m, 2H); 3.84 (s, 3H); 6.98-7.04 (m, 2H); 7.22-7.26 (m, 1 H); 7.78-7.80 (m, 1 H); 7.86- 7.89 (m, 1 H); 8.20 (br s, 1 H); 8.72 (s, 1 H); HPLC (METHOD [A]): rt 13.13 min (97.8%)
Example 64: 5-(5-(Phenoxymethyl)-1 ,3,4-thiadiazol-2-yl)-1 H-benzo[dlimidazole
The compound was synthesized starting from phenoxyacetic acid (153 mg; 1 mmol), benzimidazol-5-carbohydrazide (176 mg; 1 mmol), Lawesson's reagent (606 mg; 1 .5 mmol) and POCIs (0.137 ml; 1 .5 mmol) as described in method 3; yield: 0.031 g (10.1 %); MS m/z: 309.0 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 5.64 (s, 2H); 6.99-7.03 (m, 1 H); 7.09-7.12 (m, 2H); 7.32-7.36 (m, 2H); 7.84 (d, 1 H, 3J=8.3 Hz); 7.98 (dd, 1 H, 4J=1.7 Hz, 3J=8.3 Hz); 8.31 (d, 1 H, 4J=1.7 Hz); 8.90 (s, 1 H); HPLC (METHOD [A]): rt 12.07 min (95.9%)
Example 65: 5-(5-((Phenylthio)methyl)-1 ,3,4-thiadiazol-2-yl)-1 H-benzo[dlimidazole
The compound was synthesized starting from (phenylthio)acetic acid (169 mg; 1 mmol), benzimidazol-5-carbohydrazide (176 mg; 1 mmol), Lawesson's reagent (606 mg; 1 .5 mmol) and POCI3 (0.137 ml; 1 .5 mmol) as described in method 3; yield: 0.019 g (5.9%); MS m/z: 325.7 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 4.77 (s, 2H); 7.21 -7.25 (m, 1 H); 7.31 -7.35 (m, 2H); 7.43-7.45 (m, 2H); 7.79 (d, 1 H, 3J=8.3 Hz); 7.89 (dd, 1 H, 4J=1.7 Hz, 3J=8.3 Hz); 8.21 (br s, 1 H); 8.83 (s, 1 H); HPLC (METHOD [A]): rt 12.30 min (95.2%)
Example 66: 5-(5-((Phenylsulfonyl)methyl)-1 ,3,4-thiadiazol-2-yl)-1 H-benzo[dlimidazole
The compound was synthesized starting from phenylsulfonylacetic acid (201 mg; 1 mmol), benzimidazol-5-carbohydrazide (176 mg; 1 mmol), Lawesson's reagent (606 mg; 1 .5 mmol) and POCI3 (0.137 ml; 1 .5 mmol) as described in method 3; yield: 0.051 G (14.3%); MS m/z: 357.1 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 5.51 (s, 2H); 7.63-7.67 (m, 2H); 7.75-7.77 (m, 1 H); 7.84-7.87 (m, 3H); 7.98 (dd, 1 H, 4J=1.7 Hz, 3J=8.3 Hz); 8.30 (d, 1 H, 4J=1 .7 Hz); 8.95 (s, 1 H); HPLC (METHOD [A]): rt 10.49 min (98.3%)
Example 67: 5-(5-((3,4-Dimethoxyphenylthio)methyl)-1 ,3,4-thiadiazol-2-yl)-1 H- benzofdlimidazole
The compound was synthesized starting from (3,4-dimethoxyphenylthio)acetic acid (229 mg; 1 mmol), benzimidazol-5-carbohydrazide (176 mg; 1 mmol), Lawesson's reagent (606 mg; 1 .5 mmol) and POCI3 (0.137 ml; 1 .5 mmol) as described in method 3; yield: 0.042 g (10.9%); MS m/z: 385.3 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 3.71 (br s, 6H); 4.66 (s, 2H); 6.89 (d, 1 H, 3J=8.3 Hz); 6.98 (dd, 1 H, 4J=2.1 Hz, 3J=8.3 Hz); 7.03 (d, 1 H, 4J=2.1 Hz); 7.81 (d, 1 H, 3J=8.3 Hz); 7.90 (dd, 1 H, 4J=1 .7 Hz, 3J=8.3 Hz); 8.22 (d, 1 H, 4J=1 .7 Hz); 8.88 (s, 1 H); HPLC (METHOD [A]): rt 1 1 .72 min (95.9%)
Example 68: 5-(5-((3,4-Dimethoxyphenylsulfonyl)methyl)-1 ,3,4-thiadiazol-2-yl)-1 H- benzofdlimidazole
The compound was synthesized starting from (3,4-dimethoxyphenylsulfonyl)acetic acid (261 mg; 1 mmol), benzimidazol-5-carbohydrazide (176 mg; 1 mmol), Lawesson's reagent (606 mg; 1.5 mmol) and POCI3 (0.137 ml; 1.5 mmol) as described in method 3 but purified by flash chromatography on silica using a CHCI3/MeOH gradient; yield: 0.026 g (6.3%); MS m/z: 417.2 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 3.76 (s, 3H); 3.81 (s, 3H); 5.38 (s, 2H); 7.13 (d, 1 H 3J=8.7 Hz); 7.30 (d, 1 H, 4J=2.1 Hz); 7.36 (dd, 1 H, 4J=2.1 Hz, 3J=8.7 Hz); 7.69-7.71 (m, 1 H); 7.78-7.80 (m, 1 H); 8.16 (br s, 1 H); 8.35 (s, 1 H); 12.76 (br s, 1 H); HPLC (METHOD [A]): rt 10.63 min (97.8%) Example 69: 7-(5-(2,3-Dimethoxyphenethyl)-1 ,3,4-thiadiazol-2-yl)H-imidazo[1 ,2-alpyridine The compound was synthesized starting from 3-(2,3-dimethoxyphenyl)propionic acid (210 mg; 1 mmol), imidazo[1 ,2-a]pyridine-7-carbohydrazide (176 mg; 1 mmol), Lawesson's reagent (606 mg; 1 .5 mmol) and POCI3 (0.137 ml; 1 .5 mmol) as described in method 3; yield: 0.026 g (7%); MS m/z: 367.2 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 3.09 (t, 2H, 3J=7.5 Hz); 3.46 (t, 2H, 3J=7.5 Hz); 3.73 (s, 3H); 3.78 (s, 3H); 6.84 (dd, 1 H, 4J=1 .7 Hz, 3J=7.5 Hz); 6.92 (dd, 1 H, 4J=1 .7 Hz, 3J=7.9 Hz); 6.96-7.00 (m, 1 H); 7.85 (dd, 1 H, 4J=1 .7 Hz, 3J=7.1 Hz); 8.09 (d, 1 H, 4J=1 .7 Hz); 8.32-8.33 (m, 2H); 8.89 (d, 1 H, 3J=7.1 Hz); HPLC (METHOD [A]): rt 1 1.55 min (97.8%)
Example 70: 7-(5-(2-(Benzordin ,3ldioxol-5-yl)ethyl)-1 ,3,4-thiadiazol-2-yl)H-imidazori ,2- alpyridine
The compound was synthesized starting from 3-benzo(1 ,3)dioxol-3-yl-propionic acid (195 mg; 1 mmol), imidazo[1 ,2-a]pyridine-7-carbohydrazide (176 mg; 1 mmol), Lawesson's reagent (606 mg; 1 .5 mmol) and POCI3 (0.137 ml; 1 .5 mmol) as described in method 3; yield: 0.014 g (4%); MS m/z: 351 .3 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 3.04 (t, 2H, 3J=7.5 Hz); 3.48 (t, 2H, 3J=7.5 Hz); 5.96 (s, 2H); 6.73 (dd, 1 H, 4J=1 .7 Hz, 3J=7.9 Hz); 6.81 (d, 1 H, 3J=7.9 Hz); 6.92 (d, 1 H, 4J=1 .7 Hz); 7.84 (dd, 1 H, 4J=1 .7 Hz, 3J=7.1 Hz); 8.09 (d, 1 H, 4J=1 .7 Hz); 8.32 (m, 2H); 8.88-8.90 (m, 1 H); HPLC (METHOD [A]): rt 1 1 .12 min (95.2%)
Example 71 : 7-(5-(2-Chloro-3-methoxyphenethyl)-1 ,3,4-thiadiazol-2-yl)H-imidazo[1 ,2- alpyridine
The compound was synthesized starting from 3-(2-chloro-3-methoxyphenyl)propionic acid (215 mg; 1 mmol), imidazo[1 ,2-a]pyridine-7-carbohydrazide (176 mg; 1 mmol), Lawesson's reagent (606 mg; 1 .5 mmol) and POCI3 (0.137 ml; 1 .5 mmol) as described in method 3; yield: 0.030 g (8%); MS m/z: 371 .3 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 3.24 (t, 2H, 3J=7.5 Hz); 3.51 (t, 2H, 3J=7.5 Hz); 3.84 (s, 3H); 6.99 (dd, 1 H, 4J=1 .2 Hz, 3J=7.5 Hz); 7.03 (dd, 1 H, 4J=1 .2 Hz, 3J=8.3 Hz); 7.22-7.23 (m, 1 H); 8.86 (dd, 1 H, 4J=1 .7 Hz, 3J=7.1 Hz); 8.10 (d, 1 H, 4J = 1 .7 Hz); 8.33-8.35 (m, 2H); 8.89- 8.91 (m, 1 H); H PLC (M ETHOD [A]): rt 12.21 min (97.9%) Example 72: 7-(5-(4-(Methylthio)phenethyl)-1 ,3,4-thiadiazol-2-yl)H-imidazo[1 ,2-alpyridine The compound was synthesized starting from 3-(4-methylthiophenyl)propionic acid (197 mg; 1 mmol), imidazo[1 ,2-a]pyridine-7-carbohydrazide (176 mg; 1 mmol), Lawesson's reagent (606 mg; 1 .5 mmol) and POCI3 (0.137 ml; 1.5 mmol) as described in method 3; yield: 0.021 g (6%); MS m/z: 353.4 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 2.43 (s, 3H); 3.09 (t, 2H , 3J=7.5 Hz); 3.50 (t, 2H, 3J=7.5 Hz); 7.17-7.20 (m, 2H); 7.24-7.26 (m, 2H); 7.82 (dd, H, 4J=1.7 Hz, 3J =7.1 Hz); 8.07 (d , H , 4J=1 .7 Hz); 8.30-8.31 (m, 2H); 8.87-8.89 (m, 1 H); HPLC (METHOD [A]): rt 12.77 min (97.7%)
Example 73: 5-(5-Phenethyl-4H-1 ,2,4-triazol-3-yl)-1 H-benzordlimidazole
The compound was synthesized starting from 3-phenylpropionic acid (150 mg; 1 mmol), carbonyldiimidazole (162 mg; 1 mmol) and benzimidazol-5-carboxamidrazone (175 mg; 1 mmol) as described above; yield: 0.062 g (21 .5%); MS m/z: 290.1 [M+H]+; 145.8 [M+2H]2+; 1H-NMR (DMSO d6, 400 MHz): δ 2.94 (br s, 2H); 3.30 (br s); 7.00 (s, 1 H); 7.25-7.28 (m, 4H); 7.59-7.65 (m, 1 H); 7.85-7.87 (m, 1 H); 8.16-8.30 (m, 1 H); HPLC (METHOD [A]): rt 10.06 min (100%)
Example 74: 5-(5-(3,4-Dimethoxyphenethyl)-4H-1 ,2,4-triazol-3-yl)-1 H-benzo[dlimidazole The compound was synthesized starting from 3-(3,4-dimethoxyphenyl)propionic acid (210 mg; 1 mmol), carbonyldiimidazole (162 mg; 1 mmol) and benzimidazol-5-carboxamidrazone (175 mg; 1 mmol) as described above; yield: 0.078 g (22.3%); MS m/z: 350.4 [M+H]+; 1H- NMR (DMSO d6, 400 MHz): δ 2.92-3.02 (m, 2H); 3.68 (s, 6H); 6.72-7.00 (m, 3H); 7.57-7.87 (m, 2H); 8.14-8.30 (m, 2H); HPLC (METHOD [A]): rt 9.29 min (100%)
Example 75: 5-(5-(2-Chloro-3-methoxyphenethyl)-4H-1 ,2,4-triazol-3-yl)-1 H- benzofdlimidazole
The compound was synthesized starting from 3-(2-chloro-3-methoxyphenyl)propionic acid (215 mg; 1 mmol), carbonyldiimidazole (162 mg; 1 mmol) and benzimidazol-5- carboxamidrazone (175 mg; 1 mmol) as described above; yield: 0.067 g (19.0%); MS m/z: 354.3 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 2.93-3.03 (m, 2H); 3.14-3.15 (m, 2H); 3.82 (s, 3H); 6.92 (br s, 1 H); 6.98-6.99 (m, 1 H); 7.18-7.21 (m, 1 H); 7.53-7.64 (m, 1 H); 7.83-7.87 (m, 1 H); 8.12-8.29 (m, 2H); 12.48 (br s, 0.5H); 12.63-12.68 (m, 0.5H); 13.60 (br s, 0.5H); 13.93-13.98 (m, 0.5H); HPLC (METHOD [A]): rt 1 1 .03 min (100%) Example 76: 5-(2-(3,4-Dimethoxyphenethyl)thiazol-5-yl)-1 H-benzo[dlimidazole The compound was synthesized starting from benzimidazol-5-aminomethylketon- dihydrochloride (248 mg; 1 mmol), 3-(3,4-dimethoxyphenyl)propionic acid (210 mg; 1 mmol), Lawesson's reagent (606 mg; 1 .5 mmol), TEA (0.22 ml, 3 mmol) and POCI3 (0.137 ml; 1 .5 mmol) as described above; yield: 0.023 g (6.3%); MS m/z: 366.4 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 3.01 (t, 2H, 3J=7.5 Hz); 3.30 (t, 2H, 3J=7.5 Hz); 3.70 (s, 3H); 3.72 (s, 3H); 6.77 (dd, 1 H, 4J=1.7 Hz, 3J=8.3 Hz); 6.84 (d, 1 H, 3J=8.3 Hz); 6.88 (d, 1 H, 4J=1.7 Hz); 7.76 (dd, 1 H, 4J=1 .2 Hz, 3J=8.3 Hz); 7.85 (d, 1 H, 3J=8.3 Hz); 7.96 (br s, 1 H); 8.16 (s, 1 H); 9.34 (s, 1 H); HPLC (METHOD [A]): rt 1 1 .66 min (100%)
Example 77: 5-(2-(2-Chloro-3-methoxyphenethyl)thiazol-5-yl)-1 H-benzo[dlimidazole
The compound was synthesized starting from benzimidazol-5-aminomethylketon- dihydrochloride (248 mg; 1 mmol), 3-(2-chloro-3-methoxyphenyl)propionic acid (248 mg; 1 mmol), Lawesson's reagent (606 mg; 1 .5 mmol), TEA (0.22 ml, 3 mmol) and POCI3 (0.137 ml; 1 .5 mmol) as described above; yield: 0.015 g (4.1 %); MS m/z: 370.1 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 3.17-3.21 (m, 2H); 3.28-3.30 (m, 2H); 3.84 (s, 3H); 6.97-7.02 (m, 2H); 7.23 (t, 1 H, 3J=7.9 Hz); 7.74 (dd, 1 H, 4J=1 .2 Hz, 3J=8.3 Hz); 7.82 (d, 1 H, 3J=8.3 Hz); 7.95 (br s, 1 H); 8.16 (s, 1 H); 9.23 (s, 1 H); HPLC (METHOD [A]): rt 13.71 min (95.7%)
Example 78: 5-(2-(2,3-Dimethoxyphenethyl)thiazol-5-yl)-1 H-benzo[dlimidazole
The compound was synthesized starting from benzimidazol-5-aminomethylketon- dihydrochloride (248 mg; 1 mmol), 3-(2,3-dimethoxyphenyl)propionic acid (210 mg; 1 mmol), Lawesson's reagent (606 mg; 1 .5 mmol), TEA (0.22 ml, 3 mmol) and POCI3 (0.137 ml; 1 .5 mmol) as described above; yield: 0.017 g (4.7%); MS m/z: 366.2 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 3.03-3.07 (m, 2H); 3.24-3.27 (m, 2H); 3.73 (s, 3H); 3.78 (s, 3H); 6.83 (dd, 1 H, 4J=1 .7 Hz, 3J=7.9 Hz); 6.90 (dd, 1 H, 4J=1 .7 Hz, 3J=7.9 Hz); 6.97 (t, 1 H, 3J=7.9 Hz); 7.73 (dd, 1 H, 4J=1.7 Hz, 3J=8.3 Hz); 7.82 (d, 1 H, 3J=8.7 Hz); 7.94 (d, 1 H, 4J=1 .7 Hz); 8.14 (s, 1 H); 9.22 (s, 1 H); HPLC (METHOD [A]): rt 12.80 min (95.9%)
Example 79: 5-(2-(2,5-Dimethoxyphenethyl)thiazol-5-yl)-1 H-benzo[dlimidazole
The compound was synthesized starting from benzimidazol-5-aminomethylketon- dihydrochloride (248 mg; 1 mmol), 3-(2,5-dimethoxyphenyl)propionic acid (210 mg; 1 mmol), Lawesson's reagent (606 mg; 1 .5 mmol), TEA (0.22 ml, 3 mmol) and POCI3 (0.137 ml; 1 .5 mmol) as described above; yield: 0.026 g (7.1 %); MS m/z: 366.2 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 3.01 (t, 2H, 3J=7.5 Hz); 3.25 (t, 2H, 3J=7.5 Hz); 3.66 (s, 3H); 3.74 (s, 3H); 6.75 (dd, 1 H, 4J=2.9 Hz, 3J=8.7 Hz); 6.79 (d, 1 H, 4J=2.9 Hz); 6.89 (d, 1 H, 3J=8.7 Hz); 7.73-7.75 (m , 1 H); 7.83 (d , 1 H , 3J=8.7 Hz); 7.95 (br s, 1 H); 8.14 (s, 1 H); 9.26 (br s, 1 H ); HPLC (METHOD [A]): rt 13.1 1 min (98.6%)
Example 80: 5-(2-Phenethylthiazol-5-yl)-1 H-benzo[dlimidazole
The compound was synthesized starting from benzimidazol-5-aminomethylketon- dihydrochloride (248 mg; 1 mmol), 3-phenylpropionic acid (150 mg; 1 mmol), Lawesson's reagent (606 mg; 1 .5 mmol), TEA (0.22 ml, 3 mmol) and POCI3 (0.137 ml; 1 .5 mmol) as described above; yield: 0.025 g (8.2%); MS m/z: 306.2 [M+H]+; 1H-N MR (DMSO d6, 400 MHz): δ 3.09 (t, 2H, 3J=7.5 Hz); 3.33 (t, 2H, 3J=7.5 Hz); 7.18-7.21 (m, 1 H); 7.28-7.29 (m, 4H); 7.71 -7.73 (m, 1 H); 7.80-7.82 (m, 1 H); 7.93 (br s, 1 H); 8.15 (s, 1 H); 9.22 (br s, 1 H); HPLC (METHOD [A]): rt 12.95 min (94.3%)
Example 81 : 2-(1 H-Benzo[dlimidazol-5-yl)-5-phenethylthiazole
The compound was synthesized starting from amino-4-phenylbutan-2-on-hydrochloride (163 mg; 1 mmol), benzimidazol-5-carboxylic acid (248 mg; 1 mmol), Lawesson's reagent (606 mg; 1 .5 mmol), TEA (0.22 ml, 3 mmol) and POCI3 (0.137 ml; 1 .5 mmol) as described above; yield: 0.042 g (13.8%); MS m/z: 306.1 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 2.98 (t, 2H, 3J=7.5 Hz); 3.20 (t, 2H, 3J=7.5 Hz); 7.17-7.22 (m, 1 H); 7.25-7.31 (m, 4H); 7.62 (s, 1 H); 7.80 (d, 1 H, 3J=8.7 Hz); 7.90 (dd, 1 H, 4J=1 .7 Hz, 3J=8.3 Hz); 8.16 (d, 1 H, 4J=1 .7 Hz); 9.02 (s, 1 H); HPLC (METHOD [A]): rt 13.73 min (97.4%) Example 82: 2-(1 H-Benzo[dlimidazol-5-yl)-5-(2,3-dimethoxyphenethyl)thiazole
The compound was synthesized starting from amino-4-(2,3-dimethoxyphenyl)butan-2-on- hydrochloride (223 mg; 1 mmol), benzimidazol-5-carboxylic acid (248 mg; 1 mmol), Lawesson's reagent (606 mg; 1 .5 mmol), TEA (0.22 ml, 3 mmol) and POCI3 (0.137 ml; 1 .5 mmol) as described above; yield: 0.019 g (5.2%); MS m/z: 366.2 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 2.94 (t, 2H, 3J=7.5 Hz); 3.14 (t, 2H, 3J=7.5 Hz); 3.72 (s, 3H); 3.78 (s, 3H); 6.81 - 6.83 (m, 1 H); 6.89-6.91 (m, 1 H); 6.96-7.00 (m, 1 H); 7.62 (s, 1 H); 7.79 (d, 1 H, 3J=8.7 Hz); 7.90 (dd, 1 H, 4J=1 .7 Hz, 3J=8.7 Hz); 8.16 (br s, 1 H); 8.97 (s, 1 H); HPLC (METHOD [A]): rt 13.59 min (97.7%) Example 83: 5-(1 -Phenethyl-1 H-1 ,2,3-triazol-4-yl)-1 H-benzordlimidazole
The compound was synthesized starting from the respective 4-(1 -alkyl-1 ,2,3-triazol-4-yl)-2- nitroaniline described above according to method 3; yield: 0.035 g (15.6%); MS m/z: 290.0 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 3.23 (t, 2H, 3J=7.5 Hz); 4.67 (t, 2H, 3J=7.5 Hz); 7.20-7.29 (m, 5H); 7.84 (d, 1 H, 3J=8.7 Hz); 7.92 (dd, 1 H, 4J=1 .7 Hz, 3J=8.7 Hz); 8.16 (d, 1 H, 4J=1.7 Hz); 8.64 (s, 1 H); 9.22 (s, 1 H); HPLC (METHOD [A]): rt 10.78 min (98.3%)
Example 84: 5-(1 -((Phenylthio)methyl)-1 H-1 ,2,3-triazol-4-yl)-1 H-benzordlimidazole
The compound was synthesized starting from the respective 4-(1 -alkyl-1 ,2, 3-triazol-4-yl)-2- nitroaniline described above according to method 3; yield: 0.053 g (25.3%); MS m/z: 308.2 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 5.97 (s, 2H); 7.27-7.37 (m, 3H); 7.44-7.46 (m, 2H); 7.59-7.67 (m, 2H); 8.01 (br s, 1 H); 8.22 (s, 1 H); 8.51 (s, 1 H); 12.48 (br s, 1 H); HPLC (METHOD [A]): rt 1 1 .23 min (97.2%)
Example 85: 5-( 1 -(3,4-Dimethoxyphenethyl)-1 H-1 ,2,3-triazol-4-yl)-1 H-benzofdlimidazole The compound was synthesized starting from the respective 4-(1 -alkyl-1 ,2, 3-triazol-4-yl)-2- nitroaniline described above according to method 3; yield: 0.097 g (37.0%); MS m/z: 350.1 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 3.14 (t, 2H, 3J=7.5 Hz); 3.67 (s, 3H); 3.68 (s, 3H); 4.61 (t, 2H, 3J=7.5 Hz); 6.72 (dd, 1 H, 4J=1 .7 Hz, 3J=8.3 Hz); 6.78 (d, 1 H, 4J=1 .7 Hz); 6.83 (d, 1 H, 3J=8.3 Hz); 7.61 -7.67 (m, 2H); 7.99 (br s, 1 H), 8.24 (s, 1 H); 8.48 (s, 1 H); 12.56 (br s, 1 H); HPLC (METHOD [A]): rt 9.79 min (98.5%)
Example 86: 5-( 1 -(4-Methoxyphenethyl)-1 H-1 ,2,3-triazol-4-yl)-1 H-benzofdlimidazole
The compound was synthesized starting from the respective 4-(1 -alkyl-1 ,2, 3-triazol-4-yl)-2- nitroaniline described above according to method 3; yield: 0.058 g (29.9%); MS m/z: 320.1 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 3.15 (t, 2H, 3J=7.1 Hz); 3.69 (s, 3H); 4.59 (t, 2H, 3J=7.1 Hz); 6.82-6.84 (m, 2H); 7.12-7.14 (m, 2H); 7.55-7.61 (m, 1 H); 7.66-7.71 (m, 1 H); 7.94- 8.04 (m, 1 H); 8.22 (s, 1 H); 8.48-8.49 (m, 1 H); 12.47-12.49 (m, 1 H); HPLC (METHOD [A]): rt 10.85 min (99.8%)
Example 87: 5-( 1 -(3-Methoxyphenethyl)-1 H-1 ,2,3-triazol-4-yl)-1 H-benzofdlimidazole
The compound was synthesized starting from the respective 4-(1 -alkyl-1 ,2, 3-triazol-4-yl)-2- nitroaniline described above according to method 3; yield: 0.033 g (13.1 %); MS m/z: 320.1 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 3.20 (t, 2H, 3J=7.2 Hz); 3.69 (s, 3H); 4.64 (t, 2H, 3J=7.3 Hz); 6.76-6.80 (m, 3H); 7.18 (t, 1 H); 7.55-7.61 (m, 1 H); 7.66-7.71 (m, 1 H); 7.94-8.04 (m, 1 H); 8.22 (s, 1 H); 8.48-8.49 (m, 1 H); 12.49 (br s, 1 H); HPLC (METHOD [A]): rt 10.85 min (99.7%)
Example 88: 5-(1 H-Benzordlimidazol-5-yl)-4-benzyl-2H-1 ,2,4-triazol-3(4H)-one
The compound was synthesized starting from benzimidazol-5-carbohydrazide (176 mg; 1 mmol) and benzylisocyanate (0,133 g; 0,123 ml; 1 mmol); Yield: 0,077 (26%); MS m/z: 292.4 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 4.92 (s, 2H); 7.02-7.04 (m, 2H); 7.23-7.27 (m, 3H); 7.52 (dd, 1 H, 4J=1.7 Hz, 3J=8.7 Hz); 7.76 (d, 1 H, 3J=8.7 Hz); 7.81 (m, 1 H); 8.99 (s, 1 H), 12.08 (s, 1 H); HPLC (METHOD [A]): rt 8.28 min (100%)
Example 89: 5-(1 H-Benzordlimidazol-5-yl)-4-benzyl-2H-1 ,2,4-triazole-3(4H)-thione
The compound was synthesized starting from benzimidazol-5-carbohydrazide (176 mg; 1 mmol) and benzylisothiocyanate (0,149 g; 0,13 ml; 1 mmol); Yield: 0,052 g (17%); MS m/z: 308.3 [M+H]+; 1H-NMR (DMSO d6, 400 MHz): δ 5.33 (s, 2H); 6.97-6.99 (m, 2H); 7.15-7.22 (m, 3H); 7.44 (dd, 1 H, 4J=1 .7 Hz, 3J=8.7 Hz); 7.71 (d, 1 H, 3J=8.7 Hz), 7.81 (br s, 1 H); 8.79 (s, 1 H); 14.08 (s, 1 H); HPLC (METHOD [A]): rt 10.47 min (100%) Activity screening
Fluorometric assays
All measurements were performed with a BioAssay Reader HTS-7000Plus for microplates (Perkin Elmer) at 30 °C. QC activity was evaluated fluorometrically using H-Gln-/3NA. The samples consisted of 0.2 mM fluorogenic substrate, 0.25 U pyroglutamyl aminopeptidase (Unizyme, H0rsholm, Denmark) in 0.2 M Tris/HCI, pH 8.0 containing 20 mM EDTA and an appropriately d iluted al iquot of QC i n a final vol ume of 250 μΙ . 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 -naphthylamine under assay conditions. One unit is defined as the amount of QC catalyzing the formation of 1 μηηοΙ pGlu- /3NA from H-Gln-/3NA per minute under the described conditions.
In a second fluorometric assay, QC was activity determined using H-Gln-AMC as substrate. Reactions were carried out at 30 °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 m M EDTA and an appropriately diluted aliquot of QC in a final volume of 250 μ Ι . 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 a-Ketoglutaric acid and 30 U/ml glutamate dehydrogenase in a final volume of 250 μΙ. 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 °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 m M of the respective inhibitor and a substrate concentration at 1 KM. For detailed investigations of the inhibition and determination of Krvalues, 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.
Results Examples 1 to 18, 20 to 36, 38 to 68, 74 to 77, 81 to 87 and 89 were tested and gave hQC IC50 values of less than 10μΜ. Certain specific values are given in the table below : hQC ICso [μΜ] hQC K| [μΜ]
Example no.
8 0.64 0.1 1
9 0.76 0.20
10 0.48 0.08
22 0.52 0.12
24 0.85 0.23
39 0.80 0.13
46 0.64 0.18
47 0.40 0.06
49 0.36 0.09
50 0.76 0.08
52 0.68 0.05
54 0.59 0.06
55 0.49 0.08
57 0.83 0.13
58 0.72 0.14
59 0.56 0.12
60 0.45 0.04
61 0.76 0.17
62 0.48 0.08
63 0.22 0.10
65 0.53 0.12
67 0.08 0.01
68 0.41 0.15
83 0.57 0.14
84 0.57 0.12
85 0.47 0.10
86 0.29 0.08
87 0.55 0.12 Analytical methods
HPLC:
Method [A]: The analytical HPLC-system consisted of a Merck-Hitachi device (model LaChrom®) utilizing a LUNA® RP 18 (5 μηι), 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:: 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 - 60 % (A), 15 min - 20 min -^ 60 - 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).
Method [B]: The analytical HPLC-system consisted of a Agilent MSD 1 100 utilizing a Waters SunFire RP 18 (2,5 μηη), analytical column (length: 50 mm, diameter: 2.1 mm), and a diode array detector (DAD) with λ = 254 nm as the reporting wavelength. The compounds were analyzed using a gradient at a flow rate of 0.6 mL/min; whereby eluent (A) was acetonitrile, eluent (B) was water and eluent (C) 2% formic acid in acetonitrile applying the following gradient:
Figure imgf000128_0001
The purities of all reported compounds were determined by the percentage of the peak area at 214 nm.
Mass-spectrometry, NMR-spectroscopy:
ESI-Mass spectra were obtained with a SCI EX API 365 spectrometer (Perkin Elmer) utilizing the positive ionization mode.
The 1H N MR-Spectra (500 MHz) were recorded at a BRU KER 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 μΙ) 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 μΙ) 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, Αβ-derived peptides were incubated in 100μΙ 0.1 M sodium acetate buffer, pH 5.2 or 0.1 M Bis-Tris buffer, pH 6.5 at 30 °C. Peptides were applied in 0.5 mM [Ap(3-1 1 )a] or 0.1 5 mM [Ap(3-21 )a] concentrations, and 0.2 U QC is added all 24 hours. I n case of Ap(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).
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.
Abbreviations
(DHQ)2PHAL hydroquinine 1 ,4-phthalazinediyl diether
AcOH acetic acid
DAD diode array detector
DCC dicyclohexyl carbodiimide
DEA Diethylamine
DHAP/DAHC dihydroxyacetone phosphate/dihydro-5-azacytidine
DMF dimethylformamide
DMSO dimethylsulfoxide
EDTA ethylenediamine-N,N,N',N'-tetraacetic acid
EtOAc ethyl acetate
EtOH ethanol
FPLC fast performance liquid chromatography
HPLC high performance liquid chromatography
IPA isopropanole
LD-TOF laser-desorption time-of-flight mass spectrometry
ML mother lye
MS mass spectromtry
NMR nuclear magnetic resonance
Pd2dba3 tris(dibenzylideneacetone)dipalladium
TEA triethyl amine
TFA trifluoroacetic acid
THF tetrahydrofuran thin layer chromatography trimethylsilyl cyanide

Claims

Claims
1 . A compound of formula (I):
Figure imgf000132_0001
(I)
or a pharmaceutically acceptable salt, solvate or polymorph thereof, including all tautomers and stereoisomers thereof wherein:
R1 represents a heteroaryl group optionally substituted by one or more groups selected from
Ci-6alkyl, C2-6alkenyl, C2-6alkynyl, Ci-6haloalkyl, -Ci-6thioalkyl, -SOCi-4alkyl, -S02Ci-4alkyl, Ci_ 6alkoxy-, -0-C3-8cycloal kyl , C3-8cycloalkyl, -S02C3-8cycloalkyl, -SOC3-6cycloalkyl, C3-
6alkenyloxy-, C3-6alkynyloxy-, -C(0)Ci-6alkyl, -C(0)OCi-6alkyl, Ci-6alkoxy-Ci-6alkyl-, nitro, halogen, cyano, -C(0)OH, -NHCi-4alkyl, -N(C1_4alkyl)(C1-4alkyl), -C(0)N(Ci-4alkyl)(Ci-4alkyl), -
C(0)NH2, -C(0)NH(Ci-4alkyl) and -C(O)NH(C3-i0cycloalkyl);
and in which any of aforesaid carbocyclyl groups may optionally be substituted by one or more groups selected from Ci-4alkyl, oxo, halogen and Ci-4alkoxy;
Y represents a 5 membered heteroaryl group selected from triazolyl, oxadiazolyl, thiadiazolyl, thiazolyl, triazol-one or triazol-thione;
X represents a linker selected from -(CH2)m-R2, -(CH2)n-S-R2, -(CH2)p-0-R2 or -(CH2)p-S02- R2;
m represents an integer selected from 1 to 4;
n represents an integer selected from 1 or 2;
p represents an integer selected from 0 to 2;
R2 represents aryl, heteroaryl, carbocyclyl or heterocyclyl;
in which any of aforesaid aryl and heteroaryl groups may optionally be substituted by one or more groups selected from Ci-6alkyl, C2-6alkenyl, C2-6alkynyl, Ci-6haloalkyl, -Ci_
6thioalkyl, -SOCi-4alkyl, -S02Ci-4alkyl , Ci-6alkoxy-, -0-C3-8cycloalkyl, C3-8cycloalkyl, - S02C3-8cycloalkyl, -SOC3-6cycloalkyl, C3-6alkenyloxy-, C3-6alkynyloxy-, -C(0)Ci-6alkyl, - C(0)OCi-6alkyl, Ci-6alkoxy-Ci-6alkyl-, nitro, halogen, haloCi-6alkyl, haloCi-6alkoxy, cyano, hydroxyl, -C(0)OH, -NH2, -NHCi-4alkyl, -N(Ci-4alkyl)(Ci-4alkyl), -C(0)N(d. 4alkyl)(Ci-4alkyl), -C(0)NH2, -C(0)NH(Ci-4alkyl) and -C(O)NH(C3-i0cycloalkyl);
and in which any of aforesaid carbocyclyl and heterocyclyl groups may optionally be substituted by one or more groups selected from Ci-4alkyl, oxo, halogen and Ci-4alkoxy; or R2 represents phenyl substituted by phenyl, phenyl substituted by a monocyclic heteroaryl group, phenyl substituted by phenoxy, phenyl substituted by heterocyclyl, phenyl substituted by -0-Ci-4alkyl-heterocyclyl, phenyl substituted by benzyloxy, phenyl fused to carbocyclyl or phenyl fused to heterocyclyl, such that when Y represents oxadiazolyl and X represents CH2, R2 represents a group other than phenyl substituted by phenoxy;
in which any of aforesaid phenyl, benzyloxy and heteroaryl groups may optionally be substituted by one or more groups selected from Ci-4alkyl, halogen and Ci-4alkoxy, and in which any of aforesaid carbocyclyl and heterocyclyl groups may optionally be substituted by one or more groups selected from methyl, phenyl, oxo, halogen and Ci_
4alkoxy;
with the proviso that the compound of formula (I) is a compound other than:
6-(5-(2,3-dimethoxyphenethyl)-1 ,3,4-thiadiazol-2-yl)H-imidazo[1 ,2-a]pyridine;
6-(5-(4-(methylthio)phenethyl)-1 ,3,4-thiadiazol-2-yl)H-imidazo[1 ,2-a]pyridine; or
6-(5-(2-chloro-3-methoxyphenethyl)-1 ,3,4-thiadiazol-2-yl)H-imidazo[1 ,2-a]pyridine.
2. A compound according to claim 1 , wherein R1 represents a phenyl ring fused to a 5- membered heteroaryl ring wherein R1 is linked to the core of formula (I) through the phenyl ring.
3. A com ound according to claim 1 wherein R1 represents
Figure imgf000133_0001
wherein B represents a bond and R14 and R15 independently represent H or Ci-2alkyl.
4. A compound according to claim 3 wherein R1 represents
Figure imgf000134_0001
5. A compound according to any preceding claims, wherein Y represents triazolyl, oxadiazolyl or thiadiazolyl, such as triazolyl or thiadiazolyl.
6. A compound according to any one of claims 1 to 5, wherein X represents a linker selected from -(CH2)m-R2, -(CH2)P-S-R2, -(CH2)p-0-R2 or -(CH2)p-S02-R2, such as -(CH2)m-R2
Figure imgf000134_0002
7. A compound according to claim 6, wherein m represents an integer selected from 1 to
3.
8. A compound according to claim 7, wherein m represents an integer selected from 1 or 2.
9. A compound according to claim 6, wherein n represents 1.
10. A compound according to claim 6, wherein p represents 1.
1 1 . A compound according to any one of claims 1 to 10 wherein R2 represents aryl, heteroaryl, carbocyclyl, heterocyclyl, phenyl substituted by phenoxy or phenyl fused to heterocyclyl; the aforesaid aryl, heteroaryl, phenyl, heterocyclyl and carbocyclyl optionally being substituted.
12. A compound according to claim 1 1 wherein R2 represents aryl, heteroaryl, phenyl substituted by phenoxy or phenyl fused to heterocyclyl; the aforesaid aryl, heteroaryl, phenyl and heterocyclyl optionally being substituted.
13. A compound according to claim 12 wherein R2 represents optionally substituted aryl or phenyl substituted by phenoxy.
14. A compou nd accord ing to claim 1 3 wherei n R2 represents phenyl optionally substituted by one or more groups selected from Ci-6 alkyl, Ci-6 alkoxy, -Ci-6thioalkyl, haloCi-6 alkyl, haloCi-6 alkoxy or halogen.
15. A com pou n d accord i n g to clai m 1 4 wh erei n R2 represents phenyl optionally substituted by one or more groups selected from methyl, methoxy, ethoxy, methylthio, trifluoromethyl, trifluoromethoxy, chlorine or fluorine.
16. A compound according to claim 12 wherein R2 represents optionally substituted heteroaryl, such as pyridyl, e.g. pyridin-4-yl.
17. A compound according to claim 12 wherein R2 represents optionally substituted phenyl substituted by optionally substituted phenoxy, such as phenyl substituted by 4- phenoxy.
18. A compound according to claim 12 wherein R2 represents optionally substituted phenyl fused to optionally substituted heterocyclyl, such as benzo-1 ,3-dioxolanyl, 4- methoxy(benzo-1 ,3-dioxolanyl), 6-methoxy(benzo-1 ,3-dioxolanyl), 2,2-difluoro(benzo-1 ,3- dioxolanyl) or benzo-1 ,4-dioxanyl, e.g. benzo-1 ,3-dioxolanyl.
19. A compound according to any one of examples 1 to 89 or a pharmaceutically acceptable salt, solvate or polymorph thereof, including all tautomers and stereoisomers.
20. A compound according to claims 1 to 19 for use as a medicament.
21 . A pharmaceutical composition comprising a compound according to any one of claims 1 to 19 optionally in combination with one or more therapeutically acceptable diluents or carriers.
22. The pharmaceutical composition of claim 21 , 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.
23. The pharmaceutical composition of claim 21 or 22, 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.
24. A compound according to any one of claims 1 to 19 or a pharmaceutical composition according to any one of claims 21 to 23 for use in the treatment of a disease selected from the group consisting of Kennedy's disease, duodenal cancer with or without 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.
25. A compound according to any one of claims 1 to 19 or a pharmaceutical composition according to any one of claims 21 to 23 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 , neurodegeneratio n i n Down Syn d ro m e a n d Huntington's disease.
26. A compound according to any one of claims 1 to 19 or a pharmaceutical composition according to any one of claims 21 to 23 for use in the treatment of a disease selected from the group consisiting of rheumatoid arthritis, atherosclerosis, pancreatitis and restenosis.
27. A method of treatment or prevention of a disease selected from the group consisting of Kennedy's disease, ulcer disease, duodenal cancer with or without 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 19 or a pharmaceutical composition according to any one of claims 21 to 23.
28. 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 Synd rome and H untington's disease, which comprises administering to a subject an effective amount of a compound according to any one of claims 1 to 19 or a pharmaceutical composition according to any one of claims 21 to 23.
29. 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 19 or a pharmaceutical composition according to any one of claims 21 to 23.
30. A process for preparation of a compound of formula (I) according to any one of claims 1 to 19, which comprises: (a) preparing a compound of formula (I) wherein R1 represents 1 H-benzo[d]imidazolyl from a compound of formula (II):
Figure imgf000138_0001
NH2
(II)
wherein X and Y are as defined in claim 1 ;
(b) preparing a compound of formula (I) wherein Y represents 1 ,2,4-triazolyl by reacting compound of formula (III):
Figure imgf000138_0002
R1 (III)
wherein R1 is as defined in claim 1 , with a compound of formula X-NH2, wherein X is as defined in claim 1 ;
(c) preparing a compound of formula (I) wherein Y represents 1 ,2,4-triazolyl by reacting a compound of formula (IV):
O
NH
Figure imgf000138_0003
(IV)
wherein R1 is as defined in claim 1 , with a compound of formula X-NH2, wherein X is as defined in claim 1 ; (d) preparing a compound of formula (I) wherein Y represents 1 ,2,4-oxadiazolyl by reacting a compound of formula (V):
OH
HN ^ NH
R1
(V)
wherein R1 is as defined in claim 1 , with a compound of formula X-COOH, wherein defined in claim 1 ;
(e) preparing a compound of formula (I) wherein Y represents 1 ,3,4-thiadiazolyl from a compound of formula (VI):
X
O NH
Ck. . NH
R1
(VI)
wherein R1 and X are as defined in claim 1 ;
(f) preparing a compound of formula (I) wherein Y represents 1 ,3,4-thiadiazolyl by reacting a compound of formula (VII):
NH2
O^ NH
R1 (VII)
wherein R1 is as defined in claim 1 , with a compound of formula X-COOH, wherein X is as defined in claim 1 ; (g) preparing a compound of formula (I) wherein Y represents 4H-1 ,2,4-triazolyl by reacting a compound of formula (VIII):
NH2
Figure imgf000140_0001
R
(VIM)
wherein R1 is as defined in claim 1 , with a compound of formula X-COOH, wherein X is as defined in claim 1 ;
(h) preparing a compound of formula (I) wherein Y represents thiazolyl by reacting a compound of formula R1-COOH with a compound of formula X-CO-CH2-NH2, wherein R1 and X are as defined in claim 1 ;
(i) preparing a compound of formula (I) wherein Y represents 1 ,2,4-triazol-one or 1 ,2,4- triazol-thione from a compound of formula (IX):
W NH
O^ . NH
R1 (IX)
wherein R1 and X are as defined in claim 1 ;
(j) interconversion of compounds of formula (I); or (k) deprotecting a compound of formula (I) which is protected.
PCT/EP2011/053154 2010-03-03 2011-03-03 Novel inhibitors WO2011107530A2 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
JP2012555421A JP6026284B2 (en) 2010-03-03 2011-03-03 Inhibitors of glutaminyl cyclase
EP11707395.7A EP2542549B1 (en) 2010-03-03 2011-03-03 Inhibitors of glutaminyl cyclase
ES11707395.7T ES2586231T3 (en) 2010-03-03 2011-03-03 Glutaminyl cyclase inhibitors

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US30995110P 2010-03-03 2010-03-03
US61/309,951 2010-03-03

Publications (2)

Publication Number Publication Date
WO2011107530A2 true WO2011107530A2 (en) 2011-09-09
WO2011107530A3 WO2011107530A3 (en) 2011-11-10

Family

ID=43798321

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2011/053154 WO2011107530A2 (en) 2010-03-03 2011-03-03 Novel inhibitors

Country Status (5)

Country Link
US (1) US9181233B2 (en)
EP (1) EP2542549B1 (en)
JP (1) JP6026284B2 (en)
ES (1) ES2586231T3 (en)
WO (1) WO2011107530A2 (en)

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8729263B2 (en) 2012-08-13 2014-05-20 Novartis Ag 1,4-disubstituted pyridazine analogs there of and methods for treating SMN-deficiency-related conditions
US9040712B2 (en) 2013-01-23 2015-05-26 Novartis Ag Thiadiazole analogs thereof and methods for treating SMN-deficiency-related-conditions
WO2016097305A1 (en) 2014-12-19 2016-06-23 Probiodrug Ag Novel method for the detection of pglu-abeta peptides
US9505728B2 (en) 2012-03-09 2016-11-29 Inception 2, Inc. Triazolone compounds and uses thereof
DE102015011780A1 (en) 2015-09-16 2017-03-16 Hochschule Anhalt New glutaminyl cyclase inhibitors
US9676754B2 (en) 2012-12-20 2017-06-13 Inception 2, Inc. Triazolone compounds and uses thereof
US9776976B2 (en) 2013-09-06 2017-10-03 Inception 2, Inc. Triazolone compounds and uses thereof
CN108250196A (en) * 2016-12-29 2018-07-06 上海汇伦生命科技有限公司 (Piperazine -1- bases)The preparation method of -1 hydrogen-imidazo heteroaromatic ring compounds
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
US11220492B2 (en) 2017-05-17 2022-01-11 Arcus Biosciences, Inc. Quinazoline-pyrazole derivatives for the treatment of cancer-related disorders
US11672799B2 (en) 2013-07-31 2023-06-13 Novartis Ag 1,4-disubstituted pyridazine quinolne analogs there of and methods for treating SMN-deficiency-related conditions

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20230135378A (en) * 2022-03-16 2023-09-25 재단법인 대구경북첨단의료산업진흥재단 Thiadiazole compounds and pharmaceutical composition for preventing or treating inflammatory diseases comprising the same

Citations (547)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0172458A2 (en) 1984-07-31 1986-02-26 Suntory Limited Novel biologically active compound having anti-prolyl endopeptidase activity
JPS6137764B2 (en) 1980-08-14 1986-08-26 Tokyo Shibaura Electric Co
EP0201742A2 (en) 1985-04-16 1986-11-20 Suntory Limited N-Acylpyrrolidine derivatives, processes for preparing them, pharmaceutical composition and use
EP0201743A2 (en) 1985-04-16 1986-11-20 Suntory Limited Dipeptide derivatives of fatty acids, process for preparing them, pharmaceutical composition and use
EP0201741A2 (en) 1985-04-16 1986-11-20 Suntory Limited Dipeptide derivatives, processes for preparing them, pharmaceutical composition and use
JPS62114978A (en) 1985-11-14 1987-05-26 Suntory Ltd Novel pyrrolidinylimide having prolyl-endopeptidase inhibiting activity and production and use thereof
JPS62114957A (en) 1985-11-13 1987-05-26 Suntory Ltd Novel pyrrolidine derivative having prolylendopeptidase inhibiting action its production and use thereof
EP0224272A1 (en) 1985-11-29 1987-06-03 Suntory Limited Pyrrolidinylamide ester derivative having anti-prolyl endopeptidase activity, its synthesis and use and a pharmaceutical composition containing it
EP0232849A2 (en) 1986-02-04 1987-08-19 Suntory Limited Pyrrolidineamide derivative of acylamino acid, pharmaceutical composition and use
EP0253310A2 (en) 1986-07-11 1988-01-20 E.I. Du Pont De Nemours And Company Angiotensin II receptor blocking imidazoles
EP0268281A2 (en) 1986-11-18 1988-05-25 Suntory Limited Pyrrolidineamide derivative having anti-prolyl endopeptidase activity, compositions comprising the same, processes for the preparation of the same, and the use of the same for the preparation of a medicament of therapeutic value
EP0268190A1 (en) 1986-11-20 1988-05-25 Ono Pharmaceutical Co., Ltd. Prolinal derivatives
JPS63162672A (en) 1986-12-25 1988-07-06 Ono Pharmaceut Co Ltd Novel prolinal derivative, its production and antiamnestic agent containing said derivative
EP0275482A2 (en) 1986-12-29 1988-07-27 Ono Pharmaceutical Co., Ltd. Proline derivatives
EP0277588A1 (en) 1987-02-04 1988-08-10 Ono Pharmaceutical Co., Ltd. Prolinal derivatives
EP0280956A1 (en) 1987-02-23 1988-09-07 Ono Pharmaceutical Co., Ltd. Thiazolidine derivatives
JPS63264454A (en) 1986-12-29 1988-11-01 Ono Pharmaceut Co Ltd Novel prolinal derivative, production thereof and antiamnesic agent containing said derivative
JPS6469A (en) 1987-02-04 1989-01-05 Ono Pharmaceut Co Ltd Novel prolinal derivative, production thereof and drug for alleviating amnesia containing said derivative
JPS6442465A (en) 1987-08-07 1989-02-14 Wakunaga Pharma Co Ltd N-acylprolylpyrrolidine derivative, production and use thereof
EP0303434A1 (en) 1987-08-08 1989-02-15 Kissei Pharmaceutical Co. Ltd. Thiazolidine compounds
JPS6468396A (en) 1987-09-10 1989-03-14 Yakult Honsha Kk Novel peptide and amnesia alleviating drug comprising said peptide as active ingredient
JPH01143897A (en) 1987-11-30 1989-06-06 Kissei Pharmaceut Co Ltd Thiazolidine derivative
WO1989006242A1 (en) 1987-10-08 1989-07-13 The Mclean Hospital Corporation Antibodies to a4 amyloid peptide
JPH01226880A (en) 1988-03-07 1989-09-11 Kissei Pharmaceut Co Ltd Thiazolidine derivative
JPH01250370A (en) 1987-12-23 1989-10-05 Zeria Pharmaceut Co Ltd Novel amino acid imide derivative, its production and use thereof
EP0345428A1 (en) 1988-04-08 1989-12-13 Ono Pharmaceutical Co., Ltd. Anti-amnesic pyrrolidine derivatives
EP0355794A2 (en) 1988-08-26 1990-02-28 Merrell Dow Pharmaceuticals Inc. Neuropeptide Y antagonists
EP0355793A2 (en) 1988-08-26 1990-02-28 Merrell Pharmaceuticals Inc. Neuropeptide Y agonists and partial agonists
EP0372484A2 (en) 1988-12-08 1990-06-13 Zeria Pharmaceutical Co., Ltd. Condensed benzene derivative
JPH02207070A (en) 1989-02-07 1990-08-16 Zeria Pharmaceut Co Ltd Amide acid imide derivative, drug containing same and intermediate for producing the compound
WO1990012870A1 (en) 1989-04-14 1990-11-01 Research Foundation For Mental Hygiene, Inc. Monoclonal antibody to amyloid peptide
WO1990012871A1 (en) 1989-04-14 1990-11-01 Research Foundation For Mental Hygiene, Inc. Cerebrovascular amyloid protein-specific monoclonal antibody sv17-6e10
JPH02275858A (en) 1988-09-14 1990-11-09 Yoshitomi Pharmaceut Ind Ltd Pyridine compound
WO1990014840A1 (en) 1989-06-06 1990-12-13 California Biotechnology Inc. Recombinant alzheimer's amyloid protein
EP0403159A2 (en) 1989-06-14 1990-12-19 Smithkline Beecham Corporation Imidazolyl-alkenoic acids
US4999349A (en) 1990-03-22 1991-03-12 Bristol-Myers Squibb Co. BU-4164E - A and B, prolyl endopeptidase inhibitors
JPH0331298B2 (en) 1984-09-18 1991-05-02 Sanyo Electric Co
DE3939797A1 (en) 1989-12-01 1991-06-06 Basf Ag NEW PEPTIDES DERIVED FROM NEUROPEPTIDE Y
WO1991010664A1 (en) 1990-01-12 1991-07-25 American Home Products Corporation PYRIDAZINO[4,5-b]INDOLIZINES
JPH0356486B2 (en) 1984-08-22 1991-08-28
EP0443983A1 (en) 1990-02-19 1991-08-28 Ciba-Geigy Ag Acyl compounds
EP0454511A1 (en) 1990-03-20 1991-10-30 Sanofi N-substituted heterocycle derivatives, their preparation, compositions containing them
JPH03255080A (en) 1990-03-05 1991-11-13 Yoshitomi Pharmaceut Ind Ltd Benzene compound
EP0459136A1 (en) 1990-04-27 1991-12-04 Takeda Chemical Industries, Ltd. Benzimidazole derivatives, their production and use
WO1991018877A1 (en) 1990-06-07 1991-12-12 Zeria Pharmaceutical Co., Ltd. Novel arylalkanoylamine derivative and drug containing the same
US5073549A (en) 1990-03-22 1991-12-17 Bristol-Myers Squibb Company BU-4164E - A and B, prolyl endopeptidase inhibitors and their methods of use
EP0468339A2 (en) 1990-07-27 1992-01-29 Nippon Kayaku Kabushiki Kaisha Alpha-keto-amide derivatives having protease inhibiting activity
EP0468469A2 (en) 1990-07-27 1992-01-29 Japan Tobacco Inc. Proline derivatives
JPH049367B2 (en) 1984-02-08 1992-02-20
US5118811A (en) 1989-04-13 1992-06-02 Japan Tobacco Inc. Amino acid derivatives possessing prolyl endopeptidase-inhibitory activities
JPH04208299A (en) 1990-11-30 1992-07-29 Ajinomoto Co Inc Prolyl endopeptidase-inhibiting peptide
JPH04235162A (en) 1990-08-09 1992-08-24 Zeria Pharmaceut Co Ltd Novel succinamide derivative and drug containing the same
EP0503785A1 (en) 1991-02-21 1992-09-16 Sankyo Company Limited 1-Biphenylimidazole derivatives, their preparation and their therapeutic use
WO1992019238A1 (en) 1991-05-01 1992-11-12 Mayo Foundation For Medical Education And Research Huperzine a analogs
WO1992021333A2 (en) 1991-05-24 1992-12-10 Pharmavene, Inc. Treatment of drug withdrawal symptoms and drug craving with type b monoamine oxidase inhibitors
WO1993000361A1 (en) 1991-06-20 1993-01-07 Snow Brand Milk Products Co., Ltd. Novel prolyl endopeptidase inhibitors sna-115 and sna-115t, production thereof, and strain which produces said inhibitors
EP0522314A1 (en) 1991-07-03 1993-01-13 BASF Aktiengesellschaft Thermoplastic moulding composition based on polycarbonates, styrene/acrylanitrile-polymerisates and polyolefins
WO1993003034A1 (en) 1991-07-29 1993-02-18 Warner-Lambert Company Quinazoline derivatives as acetylcholinesterase inhibitors
JPH0515314B2 (en) 1986-09-30 1993-03-01 Kogyo Gijutsuin
US5198458A (en) 1986-02-04 1993-03-30 Suntory Limited Pyrrolidineamide derivatives of acylamino acid and pharmaceutical composition containing the same
JPH0525125B2 (en) 1984-07-16 1993-04-12 Shin Meiwa Ind Co Ltd
EP0539086A2 (en) 1991-10-24 1993-04-28 American Home Products Corporation Condensed pyrimidine derivatives and their use as angiotensine II antagonists
WO1993012139A1 (en) 1991-12-19 1993-06-24 Garvan Institute Of Medical Research A novel molecule which inhibits neuropeptide tyrosine biological function
WO1993013065A1 (en) 1991-12-27 1993-07-08 Japan Tobacco Inc. Proline derivative
JPH05201970A (en) 1992-01-24 1993-08-10 Japan Tobacco Inc New proline derivative
EP0556482A2 (en) 1991-12-24 1993-08-25 Snow Brand Milk Products Co., Ltd. Propyl endopeptidase inhibitor
US5254550A (en) 1986-11-20 1993-10-19 Ono Pharmaceutical Co., Ltd. Prolinal derivatives and pharmaceutical compositions thereof
US5262431A (en) 1986-12-29 1993-11-16 Ono Pharmaceutical Co., Ltd. Prolinal derivatives and pharmaceutical compositions thereof
JPH05331072A (en) 1992-05-27 1993-12-14 Agency Of Ind Science & Technol Prolyl endopeptidase inhibitor
WO1993025534A1 (en) 1992-06-10 1993-12-23 Zeneca Limited 2,5-DIOXO-2,5-DIHYDRO-1H-BENZ[b]AZEPINES AS NMDA RECEPTOR ANTAGONISTS
WO1994000486A1 (en) 1992-06-20 1994-01-06 The Wellcome Foundation Limited Neuropeptide y antagonists
WO1994007890A1 (en) 1992-10-05 1994-04-14 Ube Industries, Ltd. Pyrimidine compound
JPH06116284A (en) 1992-10-02 1994-04-26 Yamanouchi Pharmaceut Co Ltd New peptide
WO1994009016A1 (en) 1992-10-16 1994-04-28 Sri International 2-piperidinecarboxylic acid derivatives useful as nmda receptor antagonists
WO1994012474A1 (en) 1992-11-20 1994-06-09 Japan Tobacco Inc. Compound with prolyl endopeptidase inhibitor activity and pharmaceutical use thereof
WO1994013641A1 (en) 1992-12-16 1994-06-23 Japan Tobacco Inc. Benzomorphan useful as nmda receptor antagonist
US5328899A (en) 1988-07-15 1994-07-12 The Salk Institute For Biological Studies NPY peptide analogs
JPH06192298A (en) 1992-12-24 1994-07-12 Mitsui Toatsu Chem Inc Novel function peptide
WO1994017197A1 (en) 1993-01-25 1994-08-04 Takeda Chemical Industries, Ltd. ANTIBODY AGAINST β-AMYLOID OR DERIVATIVE THEREOF AND USE THEREOF
WO1994017035A1 (en) 1993-01-20 1994-08-04 Dr. Karl Thomae Gmbh Aminoacid derivates, medicaments containing these compounds and process for preparing the same
JPH06234693A (en) 1993-02-09 1994-08-23 Snow Brand Milk Prod Co Ltd New isotetracenone-based substance and its production
WO1994019356A1 (en) 1993-02-16 1994-09-01 Merrell Dow Pharmaceuticals Inc. Silylated acetylcholinesterase inhibitors
EP0614911A1 (en) 1993-02-15 1994-09-14 Sanofi Compounds having a sulfonamide and an amidinegroup, process for preparation and pharmaceutical compositions comprising them
WO1994020109A1 (en) 1993-03-03 1994-09-15 Rhone-Poulenc Rorer S.A. Use of 2h-1,2,4-benzothiadiazine 3(4h)-one 1,1 dioxide derivatives as non-competitive nmda receptor antagonists
WO1994020476A1 (en) 1993-03-02 1994-09-15 Fujisawa Pharmaceutical Co., Ltd. Novel heterocyclic compound
EP0618193A1 (en) 1993-03-24 1994-10-05 Adir Et Compagnie Nitrogen containing bicyclic derivatives as prolyl-endopeptidase inhibitors
WO1994029255A1 (en) 1993-06-04 1994-12-22 Merrell Pharmaceuticals Inc. Aromatic acetylcholinesterase inhibitors
WO1995000161A1 (en) 1993-06-18 1995-01-05 University Of Cincinnati Neuropeptide y antagonists and agonists
WO1995001352A1 (en) 1993-06-30 1995-01-12 Zeria Pharmaceutical Co., Ltd. Thiazolidine derivative and medicine containing the same
WO1995002602A1 (en) 1993-07-16 1995-01-26 Rhone-Poulenc Rorer S.A. Imidazo(1,2-a)pyrazin-4-one derivatives for use as ampa and nmda receptor antagonists
WO1995002601A1 (en) 1993-07-16 1995-01-26 Rhone-Poulenc Rorer S.A. IMIDAZO[1,2-a]PYRAZINE-4-ONE, PREPARATION THEREOF AND DRUGS CONTAINING SAME
WO1995012594A1 (en) 1993-11-05 1995-05-11 Rhone-Poulenc Rorer S.A. 7H-IMIDAZO(1,2-a)PYRAZINE-8-ONE NMDA RECEPTOR ANTAGONISTS
WO1995015309A1 (en) 1993-12-03 1995-06-08 Ferring B.V. Dp-iv-serine protease inhibitors
WO1995015310A1 (en) 1993-12-02 1995-06-08 Merrell Pharmaceuticals Inc. Prolyl endopeptidase inhibitors
WO1995026349A1 (en) 1994-03-28 1995-10-05 Rhone-Poulenc Rorer S.A. INDENO[1,2-e]PYRAZINE-4-ONES, PREPARATION THEREOF AND DRUGS CONTAINING SAME
WO1995026342A1 (en) 1994-03-28 1995-10-05 Rhone-Poulenc Rorer S.A. 5H-INDENO[1,2-b]PYRAZINE-2,3-DIONE DERIVATIVES, PREPARATION THEREOF AND DRUGS CONTAINING SAME
WO1995026350A1 (en) 1994-03-28 1995-10-05 Rhone-Poulenc Rorer S.A. Imidazo(1,2-a)indeno(1,2-e)pyrazine-4-one derivatives and pharmaceutical compositions containing same
WO1995026352A1 (en) 1994-03-28 1995-10-05 Rhone-Poulenc Rorer S.A. IMIDAZO[1,2-a]PYRAZINE-4-ONE DERIVATIVES, PREPARATION THEREOF AND DRUGS CONTAINING SAME
JPH07267988A (en) 1994-03-31 1995-10-17 Yamanouchi Pharmaceut Co Ltd New peptide
WO1995031986A1 (en) 1994-05-24 1995-11-30 Astra Aktiebolag Combination product of dichloroacetic acid and an nmda antagonist
WO1996008485A1 (en) 1994-09-13 1996-03-21 Pfizer Limited Quinoxalinedione nmda receptor antagonists
US5506256A (en) 1990-07-27 1996-04-09 Yoshitomi Pharmaceutical Industries, Ltd. Proline derivatives possessing prolyl endopeptidase-inhibitory activity
EP0709373A1 (en) 1993-07-23 1996-05-01 Zaidan Hojin Biseibutsu Kagaku Kenkyu Kai Novel pyrrolidine derivative
WO1996012490A1 (en) 1994-10-20 1996-05-02 Eli Lilly And Company Methods of inhibiting conditions associated with neuropeptide y
WO1996012489A1 (en) 1994-10-20 1996-05-02 Eli Lilly And Company Bicyclic neuropeptide y receptor antagonists
WO1996014318A1 (en) 1994-11-02 1996-05-17 Rhone-Poulenc Rorer S.A. SPIRO[HETEROCYCLE-IMIDAZO[1,2-a]INDENO[1,2-e]PYRAZINE]-4'-ONES, PREPARATION THEREOF AND DRUGS CONTAINING SAME
WO1996020946A1 (en) 1994-12-29 1996-07-11 Research Development Foundation Flavin adenine dinucleotide analogues, their pharmaceutical compositions, and their activity as monoamine oxidase inhibitors
WO1996021655A2 (en) 1995-01-11 1996-07-18 Hoechst Marion Roussel, Inc. Substituted oxazolidine calpain and/or cathepsin b inhibitors
WO1996022305A2 (en) 1995-01-12 1996-07-25 The Wellcome Foundation Limited Modified peptides
WO1996025435A1 (en) 1995-02-14 1996-08-22 Bayer Corporation MONOCLONAL ANTIBODY SPECIFIC FOR βA4 PEPTIDE
US5552411A (en) 1995-05-26 1996-09-03 Warner-Lambert Company Sulfonylquinolines as central nervous system and cardiovascular agents
EP0747378A1 (en) 1995-06-07 1996-12-11 Bristol-Myers Squibb Company Dihydropyridine NPY antagonists
EP0747356A1 (en) 1995-06-07 1996-12-11 Bristol-Myers Squibb Company Dihydropyridine NPY antagonists: piperazine derivatives
EP0747357A2 (en) 1995-06-07 1996-12-11 Bristol-Myers Squibb Company Dihydropyridine NPY antagonists: piperidine derivatives
WO1996040660A1 (en) 1995-06-07 1996-12-19 Pfizer Inc. Certain substituted benzylamine derivatives: a new class of neuropeptide y1 specific ligands
JPH0940693A (en) 1995-07-31 1997-02-10 Gekkeikan Sake Co Ltd Inhibitor of prolyl endopeptidase
WO1997009308A1 (en) 1995-09-01 1997-03-13 Eli Lilly And Company Indolyl neuropeptide y receptor antagonists
WO1997012615A1 (en) 1995-10-05 1997-04-10 Warner-Lambert Company Benzimidazole derivatives as 15-lo inhibitors
WO1997013754A1 (en) 1995-10-11 1997-04-17 Pelayo Camps Garcia Novel polycyclic aminopyridine compounds as acetylcholinesterase inhibitors, preparation process and use thereof
WO1997019682A1 (en) 1995-12-01 1997-06-05 Synaptic Pharmaceutical Corporation Aryl sulfonamide and sulfamide derivatives and uses thereof
WO1997019914A1 (en) 1995-11-30 1997-06-05 Dr. Karl Thomae Gmbh Amino acid derivatives, medicaments containing said compounds and methods of producing said compounds
WO1997019913A1 (en) 1995-11-30 1997-06-05 Dr. Karl Thomae Gmbh Amino acid derivatives, medicaments containing said compounds and methods of producing them
WO1997019911A1 (en) 1995-11-30 1997-06-05 Dr. Karl Thomae Gmbh Amino acid derivatives, pharmaceutical compositions containing these compounds and processes for preparing them
WO1997020820A1 (en) 1995-12-01 1997-06-12 Novartis Ag Heteroaryl compounds
WO1997020823A2 (en) 1995-12-01 1997-06-12 Novartis Ag 2-amino quinazoline derivatives as npy receptor antagonists
WO1997020822A1 (en) 1995-12-01 1997-06-12 Novartis Ag Quinazolin-2,4-diazirines as npy receptor antagonist
WO1997020821A1 (en) 1995-12-01 1997-06-12 Novartis Ag Heteroaryl derivatives
JPH09157253A (en) 1995-12-12 1997-06-17 Yamanouchi Pharmaceut Co Ltd New amino acid derivative
WO1997023214A1 (en) 1995-12-22 1997-07-03 Warner-Lambert Company 4-substituted piperidine analogs and their use as subtype selective nmda receptor antagonists
WO1997023215A1 (en) 1995-12-22 1997-07-03 Warner-Lambert Company 2-substituted piperidine analogs and their use as subtype-selective nmda receptor antagonists
WO1997023216A1 (en) 1995-12-22 1997-07-03 Warner-Lambert Company 4-substituted piperidine analogs and their use as subtype selective nmda receptor antagonists
WO1997025041A1 (en) 1996-01-09 1997-07-17 Eli Lilly And Company Benzimidzolyl neuropeptide y receptor antagonists
US5663192A (en) 1994-10-20 1997-09-02 Eli Lilly And Company Heterocyclic neuropeptide Y receptor antagonists
WO1997032873A1 (en) 1996-03-09 1997-09-12 Pfizer Research And Development Company, N.V./S.A. Quinoxalinediones
WO1997034843A1 (en) 1996-03-22 1997-09-25 Libbey Glass Inc. Apparatus and method for forming a decorative pattern on glassware having an edge
WO1997038993A1 (en) 1996-04-12 1997-10-23 Hoechst Marion Roussel, Inc. Isatin derivatives as acetylcholinesterase inhibitors and analgesics
WO1997040832A1 (en) 1996-04-25 1997-11-06 Probiodrug Gesellschaft für Arzneimittelforschung mbH Use of dipeptidyl peptidase iv effectors for lowering the blood glucose level in mammals
WO1997046250A1 (en) 1996-06-04 1997-12-11 Synaptic Pharmaceutical Corporation Methods of modifying feeding behavior, compounds useful in such methods, and dna encoding a hypothalamic atypical neuropeptide y/peptide yy receptor (y5)
WO1998001157A1 (en) 1996-07-05 1998-01-15 The Wwk Trust Compositions for the treatment of peripheral neuropathies containing antidepressants and/or monoamine oxidase inhibitors and/or vitamin b12 and/or precursors or inducers of a neurotransmitter
WO1998003493A1 (en) 1996-07-23 1998-01-29 Neurogen Corporation Certain substituted benzylamine derivatives; a new class of neuropeptide-y1 specific ligands
WO1998003492A1 (en) 1996-07-23 1998-01-29 Neurogen Corporation Certain substituted benzylamine derivatives; a new class of neuropeptide y1 specific ligands
WO1998003494A1 (en) 1996-07-23 1998-01-29 Neurogen Corporation Certain amido- and amino-substituted benzylamine derivatives; a new class of neuropeptite y1 specific ligands
WO1998005337A1 (en) 1996-08-01 1998-02-12 Cocensys, Inc. Use of gaba and nmda receptor ligands for the treatment of migraine headache
WO1998006703A1 (en) 1996-08-14 1998-02-19 Warner-Lambert Company 2-phenyl benzimidazole derivatives as mcp-1 antagonists
WO1998007420A1 (en) 1996-08-23 1998-02-26 Agouron Acquisition Corp. Neuropeptide-y ligands
WO1998010757A2 (en) 1996-09-11 1998-03-19 The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services The use of functional n-methyl-d-aspartate antagonists to ameliorate or prevent aminoglycoside-induced ototoxicity
JPH1077300A (en) 1996-09-04 1998-03-24 Gekkeikan Sake Co Ltd Prolyl endopeptidase-inhibiting peptide
WO1998015647A1 (en) 1996-10-08 1998-04-16 Novartis Ag Modulation of apoptosis
WO1998019998A2 (en) 1996-11-07 1998-05-14 Novartis Ag N-substituted 2-cyanopyrrolidines
WO1998030243A1 (en) 1997-01-08 1998-07-16 Warner-Lambert Company Acetylcholinesterase inhibitors in combination with muscarinic agonists for the treatment of alzheimer's disease
WO1998040102A1 (en) 1997-03-13 1998-09-17 The Provost, Fellows And Scholars Of The College Of The Holy And Undivided Trinity Of Queen Elizabeth Near Dublin Cytoprotective agents comprising monoamine oxidase inhibitors
WO1998044955A1 (en) 1997-04-09 1998-10-15 Mindset Ltd. Recombinant antibodies specific for beta-amyloid ends, dna encoding and methods of use thereof
WO1998046559A1 (en) 1997-04-16 1998-10-22 Arqule, Inc. SYNTHESIS AND USE OF α-KETOAMIDE DERIVATIVES AND ARRAYS
WO1998050044A1 (en) 1997-05-07 1998-11-12 Algos Pharmaceutical Corporation Composition and method combining an antidepressant with an nmda receptor antagonist, for treating neuropathic pain
WO1998050075A1 (en) 1997-05-07 1998-11-12 Algos Pharmaceutical Corporation Cox-2 inhibitors in combination with nmda-blockers for treating pain
US5847155A (en) 1990-06-04 1998-12-08 Pfizer Inc Aromatic pyrrolidine amide prolyl endopeptidase inhibitors
WO1999001416A2 (en) 1997-06-30 1999-01-14 Merz + Co. Gmbh & Co. 1-amino-alkylcyclohexane nmda receptor antagonists
WO1999007413A1 (en) 1997-08-11 1999-02-18 Algos Pharmaceutical Corporation Substance p inhibitors in combination with nmda-blockers for treating pain
WO1999007359A1 (en) 1997-08-08 1999-02-18 Shire International Licensing B.V. Use of cholinesterase inhibitors for treating attention deficit disorders
WO1999007351A2 (en) 1997-08-07 1999-02-18 Zeneca Limited Indole derivatives and their use as mcp-1 antagonists
WO1999013878A1 (en) 1997-09-18 1999-03-25 Astrazeneca Ab A COMBINATION OF A MONOAMINE OXIDASE INHIBITOR AND A h5-HT1B ANTAGONIST OR PARTIAL AGONIST
WO1999015498A1 (en) 1997-09-23 1999-04-01 Astrazeneca Ab New npy antagonists
WO1999027944A1 (en) 1997-12-02 1999-06-10 Neuralab Limited Prevention and treatment of amyloidogenic disease
WO1999038501A2 (en) 1998-02-02 1999-08-05 Trustees Of Tufts College Method of regulating glucose metabolism, and reagents related thereto
WO1999046272A1 (en) 1998-03-09 1999-09-16 Fondatech Benelux N.V. Serine peptidase modulators
WO1999045963A1 (en) 1998-03-13 1999-09-16 Algos Pharmaceutical Corporation Analgesic combination comprising nmda receptor antagonists and narcotic analgesics
WO1999046991A1 (en) 1998-03-17 1999-09-23 University Of Maryland Biotechnology Institute Methods for treating hiv-associated dementia
WO1999047131A2 (en) 1998-03-16 1999-09-23 Merck Sharp & Dohme Limited Combination of a gaba-a alpha 5 inverse agonist and an acetylcholinesterase inhibitor
WO1999048891A1 (en) 1998-03-20 1999-09-30 Merck Patent Gmbh 1-(3-heteroarylpropyl- or -prop-2-enyl)-4-benzylpiperidines used as nmda receptor antagonists
WO1999053922A1 (en) 1998-04-21 1999-10-28 Algos Pharmaceutical Corporation Analgesic compositions comprising nmda-antagonists and abt-594
WO1999057119A1 (en) 1998-05-04 1999-11-11 Neotherapeutics, Inc. Novel dopamine-like 9-substituted hypoxanthine and methods of use
WO1999057120A1 (en) 1998-05-04 1999-11-11 Neotherapeutics, Inc. Novel serotonin-like 9-substituted hypoxanthine and methods of use
WO1999060024A1 (en) 1998-05-21 1999-11-25 The University Of Tennessee Research Corporation Methods for amyloid removal using anti-amyloid antibodies
WO1999061431A1 (en) 1998-05-28 1999-12-02 Probiodrug Gesellschaft für Arzneimittelforschung mbH New dipeptidyl peptidase iv effectors
WO1999067279A1 (en) 1998-06-24 1999-12-29 Probiodrug Gesellschaft für Arzneimittelforschung mbH Compounds of unstable dipeptidyl peptidase iv inhibitors
WO1999067278A1 (en) 1998-06-24 1999-12-29 Probiodrug Gesellschaft für Arzneimittelforschung mbH Prodrugs of dipeptidyl peptidase iv inhibitors
US6011155A (en) 1996-11-07 2000-01-04 Novartis Ag N-(substituted glycyl)-2-cyanopyrrolidines, pharmaceutical compositions containing them and their use in inhibiting dipeptidyl peptidase-IV
WO2000000197A1 (en) 1998-06-26 2000-01-06 Warner-Lambert Company 4-benzyl piperidine alkylsulfoxide heterocycles and their use as subtype-selective nmda receptor antagonists
WO2000002549A2 (en) 1998-07-10 2000-01-20 Du Pont Pharmaceuticals Company Composition for and method of treating neurological disorders
DE19834591A1 (en) 1998-07-31 2000-02-03 Probiodrug Ges Fuer Arzneim Use of substances that decrease the activity of dipeptidyl peptidase IV to increase blood sugar levels, e.g. for treating hypoglycemia
WO2000007600A1 (en) 1998-08-07 2000-02-17 R.J. Reynolds Tobacco Company Pharmaceutical compositions for the prevention and treatment of central nervous system disorders
WO2000009483A2 (en) 1998-08-17 2000-02-24 Finetech Ltd. Process and intermediates for production of donepezil and related compounds
WO2000015205A2 (en) 1998-09-11 2000-03-23 Eisai Co., Ltd. Use of acetylcholinesterase inhibitors for the preparation of pharmaceutical compositions for the treatment of functional and/or organic pain syndromes
WO2000023057A2 (en) 1998-10-16 2000-04-27 Janssen Pharmaceutica N.V. Atypical antiphsychotic in combination with acetylcholinesterase inhibitor for improving cognition
WO2000029023A1 (en) 1998-11-12 2000-05-25 Algos Pharmaceutical Corporation Cox-2 inhibitors in combination with nmda-blockers for treating pain
WO2000030446A1 (en) 1998-11-23 2000-06-02 Bonnie Davis Dosage formulations for acetylcholinesterase inhibitors
WO2000030674A1 (en) 1998-11-26 2000-06-02 Ferring Bv Neuropeptide y y4 agents in the treatment of reproductive disorders
WO2000033840A1 (en) 1998-12-11 2000-06-15 Bonnie Davis Use of acetylcholinesterase inhibitors on the modulation of the hypothalamic-pituitary-gonadal axis
WO2000046195A1 (en) 1999-02-05 2000-08-10 Astrazeneca Ab Anti-inflammatory indole derivatives
WO2000046199A2 (en) 1999-02-05 2000-08-10 Astrazeneca Ab Indole derivatives as anti-inflammation agents
WO2000046198A1 (en) 1999-02-05 2000-08-10 Astrazeneca Ab Anti-inflammatory indole derivatives
WO2000046196A1 (en) 1999-02-05 2000-08-10 Astrazeneca Ab Indole derivatives and their use as mcp-1 antagonists
WO2000046197A1 (en) 1999-02-05 2000-08-10 Astrazeneca Ab Indole derivatives and their use as mcp-1 receptor antagonists
US6107317A (en) 1999-06-24 2000-08-22 Novartis Ag N-(substituted glycyl)-thiazolidines, pharmaceutical compositions containing them and their use in inhibiting dipeptidyl peptidase-IV
US6110949A (en) 1999-06-24 2000-08-29 Novartis Ag N-(substituted glycyl)-4-cyanothiazolidines, pharmaceutical compositions containing them and their use in inhibiting dipeptidyl peptidase-IV
US6114336A (en) 1996-10-23 2000-09-05 Sanofi Cosmetic composition containing a neuropeptide Y receptor antagonist
US6121311A (en) 1999-04-28 2000-09-19 Japan Tobacco Inc. Method for treating cocainism
WO2000056711A1 (en) 1999-03-23 2000-09-28 Sumitomo Pharmaceuticals Co., Ltd. Tricyclic indole-2-carboxylic acid compound used as nmda receptor antagonist
WO2000063250A1 (en) 1999-04-15 2000-10-26 Merck Frosst Canada & Co. Antibodies that recognize app cleaved by caspases and methods of use
WO2000068197A1 (en) 1999-05-05 2000-11-16 Ortho-Mcneil Pharmaceutical, Inc. 3a,4,5,9b-TETRAHYDRO-1H-BENZ[e]INDOL-2-YL AMINE-DERIVED NEUROPEPTIDE Y RECEPTORS LIGANDS USEFUL IN THE TREATMENT OF OBESITY AND OTHER DISORDERS
WO2000071144A1 (en) 1999-05-19 2000-11-30 Domer, Inc. Prolyl endopeptidase inhibitor
WO2000072880A2 (en) 1999-05-28 2000-12-07 Neuralab Limited Prevention and treatment of amyloidogenic disease
WO2000077178A1 (en) 1999-06-16 2000-12-21 Boston Biomedical Research Institute IMMUNOLOGICAL CONTROL OF β-AMYLOID LEVELS IN VIVO
WO2001000215A1 (en) 1999-06-25 2001-01-04 Wake Forest University Compositions for treating or preventing neurodegeneration and cognitive decline
US6172081B1 (en) 1999-06-24 2001-01-09 Novartis Ag Tetrahydroisoquinoline 3-carboxamide derivatives
WO2001010831A1 (en) 1999-08-06 2001-02-15 Grünenthal GmbH Substituted pyrrolidin-2,3,4-trion-3-oxime derivatives useful as nmda-receptor antagonists
WO2001010833A1 (en) 1999-08-06 2001-02-15 Grünenthal GmbH Substituted 1,5-dihydropyrrol-2-on derivatives as nmda receptor antagonists for the treatment of pain
WO2001012176A2 (en) 1999-08-16 2001-02-22 Sanofi-Synthelabo Use of monoamine oxidase inhibitors for the manufacture of drugs intended for the treatment of obesity
WO2001012598A2 (en) 1999-08-13 2001-02-22 The Trustees Of Columbia University In The City Of New York METHODS OF INHIBITING BINDING OF β-SHEET FIBRIL TO RAGE AND CONSEQUENCES THEREOF
WO2001014318A2 (en) 1999-08-24 2001-03-01 Probiodrug Ag New effectors of dipeptidyl peptidase iv for topical use
WO2001026656A2 (en) 1999-10-11 2001-04-19 Societe De Conseils De Recherches Et D'applications Scientifiques (S.C.R.A.S.) 5-membered heterocycle derivatives and use thereof as monoamine oxidase inhibitors
WO2001032640A1 (en) 1999-11-01 2001-05-10 Merz Pharmaceuticals Gmbh 1-cyclic amino-alkylcyclohexane compounds, pharmaceutical compositions thereof, and their use as anticonvulsants
WO2001034594A1 (en) 1999-11-12 2001-05-17 Guilford Pharmaceuticals, Inc. Dipeptidyl peptidase iv inhibitors and methods of making and using dipeptidyl peptidase iv inhibitors
WO2001040180A2 (en) 1999-11-30 2001-06-07 Ferring Bv N-substituted 2-cyanopyrrolidines and their use as antidiabetic agents
WO2001053255A1 (en) 2000-01-24 2001-07-26 Merck Sharp & Dohme Limited Gamma-secretase inhibitors
WO2001055105A1 (en) 2000-01-24 2001-08-02 Novo Nordisk A/S N-substituted 2-cyanopyroles and -pyrrolines which are inhibitors of the enzyme dpp-iv
WO2001057226A1 (en) 2000-02-03 2001-08-09 Millennium Pharmaceuticals, Inc. Humanized anti-ccr2 antibodies and methods of use therefor
WO2001062801A2 (en) 2000-02-24 2001-08-30 Washington University Humanized antibodies that sequester amyloid beta peptide
WO2001066096A2 (en) 2000-03-06 2001-09-13 Immune Network Ltd. Compositions for prevention and treatment of dementia
WO2001066564A2 (en) 2000-03-03 2001-09-13 Merck Sharp & Dohme Limited Gamma-secretase inhibitors
WO2001068603A2 (en) 2000-03-10 2001-09-20 Bristol-Myers Squibb Co. Cyclopropyl-fused pyrrolidine-based inhibitors of dipeptidyl iv, processes for their preparation, and their use
WO2001077144A1 (en) 2000-04-07 2001-10-18 Merck Sharp & Dohme Limited Gamma-secretase inhibitors
WO2001078728A1 (en) 2000-04-13 2001-10-25 Eisai Co., Ltd. Acetylcholinesterase inhibitors containing 1-benzyl- pyridinium salts
WO2001081295A1 (en) 2000-04-26 2001-11-01 Warner-Lambert Company Cyclohexylamine derivative as subtype selective nmda receptor antagonists
WO2001081337A1 (en) 2000-04-26 2001-11-01 Ferring B.V. Inhibitors of dipeptidyl peptidase iv
WO2001081304A1 (en) 2000-04-26 2001-11-01 Ferring Bv Inhibitors of dipeptidyl peptidase iv
WO2001085145A2 (en) 2000-05-09 2001-11-15 Pfizer Products Inc. A pharmaceutical composition and method of treatment of diseases of cognitive dysfunction in a mammal
WO2001092204A1 (en) 2000-06-01 2001-12-06 Warner-Lambert Company Cyclohexylamine derivatives as subtype selective nmda receptor antagonists
WO2001094321A1 (en) 2000-06-06 2001-12-13 Warner-Lambert Company Bicyclic cyclohexylamines and their use as nmda receptor antagonists
WO2001098289A1 (en) 2000-06-22 2001-12-27 Pharmos Corporation Novel non-psychotropic cannabinoids
WO2001098262A1 (en) 2000-06-23 2001-12-27 Merck Sharp & Dohme Limited Amidine derivatives as selective antagonists of nmda receptors
WO2002002560A2 (en) 2000-07-04 2002-01-10 Novo Nordisk A/S Purine-2,6-diones which are inhibitors of the enzyme dipeptidyl peptidase iv (dpp-iv)
WO2002021509A1 (en) 2000-09-01 2002-03-14 Snap-On Technologies, Inc. Computer-implemented speech recognition system training
WO2002027418A2 (en) 2000-09-25 2002-04-04 Motorwiz, Inc. Model-based machine diagnostics and prognostics using theory of noise and communications
WO2002032412A2 (en) 2000-10-17 2002-04-25 Pfizer Products Inc. Combination use of acetylcholinesterase inhibitors and gabaa inverse agonists for the treatment of cognitive disorders
WO2002034718A1 (en) 2000-10-24 2002-05-02 Richter Gedeon Vegyeszeti Gyar Rt. Amide derivatives as nmda receptor antagonists
WO2002036555A1 (en) 2000-11-02 2002-05-10 Merck Sharp & Dohme Limited Sulfamides as gamma-secretase inhibitors
WO2002038541A1 (en) 2000-11-10 2002-05-16 Taisho Pharmaceutical Co., Ltd. Cyanopyrrolidine derivatives
WO2002041842A2 (en) 2000-11-03 2002-05-30 Proteotech. Inc. Antibody pti-hs7 for treatment of alzheimer's disease and other amyloidoses and parkinson's disease
WO2002046222A2 (en) 2000-12-07 2002-06-13 Mario Chojkier Compositions and methods for diagnosing alzheimer's disease
WO2002046237A2 (en) 2000-12-06 2002-06-13 Neuralab Limited Humanized antibodies that recognize beta amyloid peptide
US6426365B1 (en) 1997-02-12 2002-07-30 Japan Tobacco Inc. CETP activity inhibitors
WO2002060900A2 (en) 2001-01-31 2002-08-08 Telik, Inc. Antagonists of mcp-1 function and methods of use thereof
US6448229B2 (en) 2000-07-06 2002-09-10 Merck Sharp & Dohme Ltd. Gamma secretase inhibitors
WO2002070509A2 (en) 2001-03-01 2002-09-12 Telik, Inc. Antagonists of mcp-1 function and methods of use thereof
WO2002072542A2 (en) 2001-03-08 2002-09-19 Emory University Ph-dependent nmda receptor antagonists
WO2002074240A2 (en) 2001-03-16 2002-09-26 Cornell Research Foundation, Inc. Anti-amyloid antibody based diagnosis and treatment of a neurological disease or disorder
WO2002081463A1 (en) 2001-04-03 2002-10-17 Telik, Inc. Antagonists of mcp-1 function and methods of use thereof
WO2002083128A1 (en) 2001-04-12 2002-10-24 Bristol-Myers Squibb Company 2,1-oxazoline and 1,2-pyrazoline-based inhibitors of dipeptidyl peptidase iv and method
WO2002088306A2 (en) 2001-04-30 2002-11-07 Eli Lilly And Company Humanized antibodies
WO2002088307A2 (en) 2001-04-30 2002-11-07 Eli Lilly And Company Humanized antibodies
EP1258476A1 (en) 2001-05-15 2002-11-20 Les Laboratoires Servier Alpha-amino acid derivatives, method for their preparation and their use as dipeptidyl-peptidase IV inhibitors (DPP IV)
WO2002094881A2 (en) 2001-05-18 2002-11-28 Krka Tovarna Zdravil, D.D., Novo Mesto Monoclonal antibody neutralising cathepsin b activity and uses thereof
WO2002096897A1 (en) 2001-05-30 2002-12-05 Neurologic, Inc. PHOSPHINYLMETHYL AND PHOSPHORYLMETHYL SUCCINIC AND GLUTARIC ACID ANALOGS AS β-SECRETASE INHIBITORS
WO2002098849A2 (en) 2001-06-01 2002-12-12 Elan Pharmaceuticals, Inc. Hydroxy alkyl amine derivatives as beta-secretase inhibitors and their use for the treatment of alzheimer’s disease and similar diseases
WO2003000181A2 (en) 2001-06-20 2003-01-03 Merck & Co., Inc. Dipeptidyl peptidase inhibitors for the treatment of diabetes
WO2003000250A1 (en) 2001-06-25 2003-01-03 Ferring Bv 3-fluoro-pyrrolidines as antidiabetic agents
WO2003000180A2 (en) 2001-06-20 2003-01-03 Merck & Co., Inc. Dipeptidyl peptidase inhibitors for the treatment of diabetes
WO2003002531A2 (en) 2001-06-27 2003-01-09 Smithkline Beecham Corporation Fluoropyrrolidines as dipeptidyl peptidase inhibitors
WO2003002593A2 (en) 2001-06-27 2003-01-09 Probiodrug Ag Peptide structures useful for competitive modulation of dipeptidyl peptidase iv catalysis
WO2003002553A2 (en) 2001-06-27 2003-01-09 Smithkline Beecham Corporation Fluoropyrrolidines as dipeptidyl peptidase inhibitors
WO2003002530A2 (en) 2001-06-27 2003-01-09 Smithkline Beecham Corporation Pyrrolidines as dipeptidyl peptidase inhibitors
WO2003004498A1 (en) 2001-07-06 2003-01-16 Merck & Co., Inc. Beta-amino tetrahydroimidazo (1, 2-a) pyrazines and tetrahydrotrioazolo (4, 3-a) pyrazines as dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
WO2003004496A1 (en) 2001-07-03 2003-01-16 Novo Nordisk A/S Dpp-iv-inhibiting purine derivatives for the treatment of diabetes
WO2003010159A1 (en) 2001-07-24 2003-02-06 Richter Gedeon Vegyészeti Gyár Rt. Piperidine derivatives as nmda receptor antagonists
WO2003012141A1 (en) 2001-07-31 2003-02-13 Takeda Chemical Industries, Ltd. Method of screening alzheimer’s disease-associated gene
WO2003014162A1 (en) 2001-08-03 2003-02-20 Medical & Biological Laboratories Co., Ltd. ANTIBODY RECOGNIZING GM1 GANGLIOSIDE-BOUND AMYLOID β-PROTEIN AND DNA ENCODING THE ANTIBODY
WO2003013527A1 (en) 2001-08-03 2003-02-20 Schering Corporation Sulfonamide derivatives as gamma secretase inhibitors
WO2003014075A2 (en) 2001-08-03 2003-02-20 Schering Corporation Novel gamma secretase inhibitors
WO2003015691A2 (en) 2001-08-17 2003-02-27 Eli Lilly And Company RAPID IMPROVEMENT OF COGNITION IN CONDITIONS RELATED TO A$g(b)
WO2003016466A2 (en) 2001-08-17 2003-02-27 Eli Lilly And Company ANTI-Aβ ANTIBODIES
WO2003020289A1 (en) 2001-08-30 2003-03-13 Ortho-Mcneil Pharmaceutical, Inc. Treatment of dementia and memory disorders with anticonvulsants and acetylcholinesterase inhibitors
WO2003024965A2 (en) 2001-09-19 2003-03-27 Novo Nordisk A/S Heterocyclic compounds that are inhibitors of the enzyme dpp-iv
WO2003024942A1 (en) 2001-09-14 2003-03-27 Mitsubishi Pharma Corporation Thiazolidine derivative and medicinal use thereof
WO2003033524A2 (en) 2001-10-12 2003-04-24 Probiodrug Ag Peptidyl ketones as inhibitors of dpiv
WO2003035057A1 (en) 2001-10-23 2003-05-01 Ferring B.V. Inhibitors of dipeptidyl peptidase iv
WO2003035067A1 (en) 2001-10-23 2003-05-01 Ferring B.V. Novel dipeptidyl peptidase iv (dp-iv) inhibitors as anti-diabetic agents
WO2003037376A1 (en) 2001-11-02 2003-05-08 Kensuke Egashira Preventives and/or remedies for post-transplant arteriosclerosis as rejection of organ transplant
WO2003037327A1 (en) 2001-10-26 2003-05-08 F. Hoffmann-La-Roche Ag N-substituted pyrrolidin derivatives as dipeptidyl peptidase iv inhibitors
WO2003040183A2 (en) 2001-11-09 2003-05-15 The Genetics Company, Inc Compounds for the diagnosis/prevention/treatment of alzheimer's disease
WO2003039467A2 (en) 2001-11-02 2003-05-15 Diagenics International Corporation Monoclonal antibodies specific for beta-amyloid.
WO2003040174A2 (en) 2001-11-09 2003-05-15 Probiodrug Ag Substituted amino ketone compounds
WO2003039454A2 (en) 2001-10-23 2003-05-15 Oklahoma Medical Research Foundation Beta-secretase inhibitors and methods of use
WO2003043987A2 (en) 2001-11-19 2003-05-30 Elan Pharmaceuticals, Inc. (4-phenyl) piperidin-3-yl-phenylcarboxylate derivatives and related compounds as beta-secretase inhibitors for the treatment of alzheimer's disease
WO2003045128A2 (en) 2001-11-21 2003-06-05 New York University SYNTHETIC IMMUNOGENIC BUT NON-DEPOSIT-FORMING POLYPEPTIDES AND PEPTIDES HOMOLOGOUS TO AMYLOID β, PRION PROTEIN, AMYLIN, α-SYNUCLEIN, OR POLYGLUTAMINE REPEATS FOR INDUCTION OF AN IMMUNE RESPONSE THERETO
WO2003045977A2 (en) 2001-11-26 2003-06-05 Trustees Of Tufts College Peptidomimetic Inhibitors of Post-Proline Cleaving Enzymes
WO2003048204A1 (en) 2001-12-06 2003-06-12 Takeda Chemical Industries, Ltd. Alzheimer’s disease-associated gene and protein and use thereof
WO2003051374A2 (en) 2001-12-17 2003-06-26 New York State Office Of Mental Health SEQUESTRATION OF Aβ IN THE PERIPHERY IN THE ABSENCE OF IMMUNOMODULATING AGENT AS A THERAPEUTIC APPROACH FOR THE TREATMENT OR PREVENTION OF BETA-AMYLOID RELATED DISEASES
WO2003053368A2 (en) 2001-12-19 2003-07-03 Atherogenics, Inc. Chalcone derivatives and their use to treat diseases
WO2003055881A1 (en) 2001-12-27 2003-07-10 F. Hoffmann-La Roche Ag Pyrido(2,1-a)isoquinoline derivatives as dpp-iv inhibitors
WO2003055514A1 (en) 2001-12-21 2003-07-10 Antigenics Inc. Compositions comprising immunoreactive reagents and saponins, and methods of use thereof
WO2003057666A2 (en) 2001-12-26 2003-07-17 Guilford Pharmaceuticals Inhibitors of dipeptidyl peptidase iv
WO2003057204A2 (en) 2002-01-08 2003-07-17 Nordic Bioscience A/S Modulation of iamt (pimt or pcmt) in immune system
WO2003059346A1 (en) 2002-01-18 2003-07-24 The Genetics Company Inc. Beta-secretase inhibitors
WO2003063760A2 (en) 2002-01-31 2003-08-07 Tel Aviv University Future Technology Development L.P. Peptides antibodies directed thereagainst and methods using same for diagnosing and treating amyloid-associated diseases
WO2003066592A1 (en) 2002-02-06 2003-08-14 Schering Corporation Gamma secretase inhibitors
WO2003068748A1 (en) 2002-02-13 2003-08-21 F. Hoffmann-La Roche Ag Novel pyridine- and quinoline-derivatives
WO2003068757A1 (en) 2002-02-13 2003-08-21 F. Hoffmann-La Roche Ag Novel pyridin- and pyrimidin-derivatives
WO2003070760A2 (en) 2002-02-20 2003-08-28 F. Hoffmann-La Roche Ag Anti-amyloid beta antibodies and their use
WO2003072556A1 (en) 2001-06-27 2003-09-04 Prosidion Ltd. Glutaminyl based dpiv inhibitors
WO2003074081A1 (en) 2002-02-28 2003-09-12 Mindset Biopharmaceuticals Usa Inc. SPECIFIC ANTIBODIES TO AMYLOID β PEPTIDE, PHARMACEUTICAL COMPOSITIONS AND METHODS OF USE THEREOF
WO2003074500A2 (en) 2002-03-06 2003-09-12 Sanofi-Aventis N-aminoacetyl-pyrrolidine-2-carbonitriles and their use as ddp-iv inhibitors
WO2003077858A2 (en) 2002-03-12 2003-09-25 Neuralab Limited Humanized antibodies that recognize beta amyloid peptide
WO2003082820A1 (en) 2002-03-29 2003-10-09 Eisai Co., Ltd. (1-indanone)-(1,2,3,6-tetrahydropyridine) derivative
WO2003082817A2 (en) 2002-03-25 2003-10-09 Merck & Co., Inc. Beta-amino heterocyclic dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
WO2003086310A2 (en) 2002-04-12 2003-10-23 Ramot At Tel Aviv University Ltd. Prevention of brain inflammation as a result of induced autoimmune response
WO2003089460A1 (en) 2002-04-19 2003-10-30 The Governing Council Of The University Of Toronto Immunological methods and compositions for the treatment of alzheimer's disease
WO2003091278A1 (en) 2002-04-24 2003-11-06 Hiroshi Mori Gamma-secretase inhibitors
WO2003091220A1 (en) 2002-04-26 2003-11-06 Schering Corporation Muscarinic antagonists
US6649196B2 (en) 2001-03-12 2003-11-18 Mayo Foundation For Medical Education And Research Methods of reducing β-amyloid polypeptides
WO2003099202A2 (en) 2002-05-17 2003-12-04 Merck & Co., Inc. Beta-secretase inhibitors
WO2003101958A2 (en) 2002-06-04 2003-12-11 Pfizer Products Inc. Flourinated cyclic amides as dipeptidyl peptidase iv inhibitors
WO2003101458A1 (en) 2002-05-31 2003-12-11 H. Lundbeck A/S A combination of an nmda-antagonist and acetylcholine esterase inhibitors for the treatment of alzheimer's disease
WO2003101449A2 (en) 2002-06-04 2003-12-11 Pfizer Products Inc. Process for the preparation of 3,3,4,4-tetrafluoropyrrolidine and derivatives thereof
WO2003104229A1 (en) 2002-06-06 2003-12-18 エーザイ株式会社 Novel fused imidazole derivative
WO2003104437A2 (en) 2002-06-11 2003-12-18 Northwestern University Anti-addl antibodies and uses thereof
WO2004000958A1 (en) 2002-06-19 2003-12-31 Surface Specialties, S.A. Semi-gloss powder coating compositions
WO2004007468A1 (en) 2002-07-15 2004-01-22 Merck & Co., Inc. Piperidino pyrimidine dipeptidyl peptidase inhibitors for the treatment of diabetes
WO2004007446A1 (en) 2002-07-10 2004-01-22 Yamanouchi Pharmaceutical Co., Ltd. Novel azetidine derivative or salt thereof
WO2004009062A2 (en) 2002-07-19 2004-01-29 Khalid Iqbal NMDA RECEPTOR ANTAGONISTS AND THEIR USE IN INHIBITING ABNORMAL HYPERPHOSPHORYLATION OF MICROTUBULE ASSOCIATED PROTEIN tau
US6686449B2 (en) 2000-06-30 2004-02-03 Pharmacia & Upjohn Company Mutant presenilin 1 polypeptides
WO2004013098A1 (en) 2002-08-05 2004-02-12 Bristol-Myers Squibb Company Novel gamma-lactams as beta-secretase inhibitors
WO2004018468A2 (en) 2002-08-21 2004-03-04 Boehringer Ingelheim Pharma Gmbh & Co. Kg 8-[3-amino-piperidin-1-yl]-xanthines, the production thereof and the use of the same as medicaments
WO2004018469A1 (en) 2002-08-22 2004-03-04 Boehringer Ingelheim Pharma Gmbh & Co. Kg Novel purine derivatives, production and use thereof as medicaments
WO2004018467A2 (en) 2002-08-22 2004-03-04 Boehringer Ingelheim Pharma Gmbh & Co. Kg Phenacyl xanthine derivatives as dpp-iv inhibitor
WO2004024090A2 (en) 2002-09-12 2004-03-25 The Regents Of The University Of California Immunogens and corresponding antibodies specific for high molecular weight aggregation intermediates common to amyloids formed from proteins of differing sequence
WO2004024921A1 (en) 2002-09-12 2004-03-25 Juridical Foundation The Chemo-Sero-Therapeutic Research Institute Human antihuman mcp-1 antibody and antibody fragment thereof
WO2004024770A1 (en) 2002-09-12 2004-03-25 Universidad De Zaragoza Polyclonal antibodies, preparation method thereof and use of same
US6713276B2 (en) 2000-06-28 2004-03-30 Scios, Inc. Modulation of Aβ levels by β-secretase BACE2
WO2004026822A2 (en) 2002-09-19 2004-04-01 Abbott Laboratories Pharmaceutical compositions as inhibitors of dipeptidyl peptidase-iv (dpp-iv)
WO2004026851A1 (en) 2002-09-20 2004-04-01 Axys Pharmaceuticals, Inc. 3-(3,5-disubstituted-4-hydroxyphenyl)propionamide derivatives as cathepsin b inhibitors
WO2004029630A1 (en) 2002-09-27 2004-04-08 Janssen Pharmaceutica N.V. N-11 truncated amyloid-beta monoclonal antibodies, compositions, methods and uses
WO2004028522A1 (en) 2002-09-25 2004-04-08 Innovative Drug Delivery Systems, Inc. Analgesic compositions comprising nmda receptor antagonists and benzalkonium chloride
WO2004031137A1 (en) 2002-10-04 2004-04-15 Merck Sharp & Dohme Limited Cyclohexyl sulphone derivatives as gamma-secretase inhibitors
WO2004031400A2 (en) 2002-10-01 2004-04-15 Northwestern University Amyloid beta-derived diffusible ligands (addls), addl-surrogates, addl-binding molecules, and uses thereof
WO2004031139A1 (en) 2002-10-04 2004-04-15 Merck Sharp & Dohme Limited Cyclohexyl sulphones as gamma-secretase inhibitors
WO2004033455A2 (en) 2002-10-08 2004-04-22 Novo Nordisk A/S Hemisuccinate salts of heterocyclic dpp-iv inhibitors
WO2004032868A2 (en) 2002-10-09 2004-04-22 Rinat Neuroscience Corp. Methods of treating alzheimer's disease using antibodies directed against amyloid beta peptide and compositions thereof
WO2004032836A2 (en) 2002-10-07 2004-04-22 Merck & Co., Inc. Antidiabetic beta-amino heterocylcic dipeptidyl peptidase inhibitors
WO2004032929A2 (en) 2002-10-09 2004-04-22 Neuropharma, S.A. Dual binding site acetylcholinesterase inhibitors for the treatment of alzheimer’s disease
WO2004037234A2 (en) 2002-10-24 2004-05-06 Merz Pharma Gmbh & Co. Kgaa Combination therapy using 1-aminocyclohexane derivatives and acetylcholinesterase inhibitors
WO2004037169A2 (en) 2002-10-18 2004-05-06 Merck & Co., Inc. Beta-amino heterocyclic dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
WO2004039773A2 (en) 2002-10-30 2004-05-13 Nordic Bioscience A/S Coumpounds modulating the activity of gapdh and/or iamt
WO2004039371A2 (en) 2002-10-29 2004-05-13 Pharmacia Corporation Compositions of cyclooxygenase-2 selective inhibitors and nmda receptor antagonists for the treatment or prevention of neuropathic pain
WO2004039370A1 (en) 2002-11-01 2004-05-13 Merck Sharp & Dohme Limited Sulfonamides, sulfamates and sulfamides as gamma-secretase inhibitors
WO2004041795A1 (en) 2002-10-30 2004-05-21 Guilford Pharmaceuticals Inc. Novel inhibitors of dipeptidyl peptidase iv
WO2004043940A1 (en) 2002-11-07 2004-05-27 Merck & Co., Inc. Phenylalanine derivatives as dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
WO2004043916A1 (en) 2002-11-12 2004-05-27 Merck & Co., Inc. Phenylcarboxamide beta-secretase inhibitors for the treatment of alzheimer's disease
WO2004044204A2 (en) 2002-11-06 2004-05-27 Institut Pasteur Variable fragments of single-chain camelide antibodies and uses thereof for diagnosing and treating various pathologies
WO2004048352A2 (en) 2002-11-27 2004-06-10 Fujisawa Pharmaceutical Co., Ltd. 2-cyanopyrrolidines and their analogues as dpp-iv inhibitors
WO2004050022A2 (en) 2002-12-04 2004-06-17 Merck & Co., Inc. Phenylalanine derivatives as dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
WO2004052850A2 (en) 2002-12-09 2004-06-24 Bristol-Myers Squibb Company Methods and compounds producing dipeptidyl peptidase iv inhibitors and intermediates thereof
WO2004056727A2 (en) 2002-12-19 2004-07-08 Atherogenics, Inc. Process of making chalcone derivatives
WO2004058266A1 (en) 2002-12-20 2004-07-15 Merck & Co., Inc. 3-amino-4-phenylbutanoic acid derivatives as dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
WO2004062625A2 (en) 2003-01-07 2004-07-29 Merck & Co., Inc. Macrocyclic beta-secretase inhibitors for treatment of alzheimer's disease
WO2004064778A2 (en) 2003-01-17 2004-08-05 Merck & Co. Inc. 3-amino-4-phenylbutanoic acid derivatives as dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
WO2004067561A1 (en) 2003-01-31 2004-08-12 Abbott Gmbh & Co. Kg Amyloid-$g(b)(1-42) oligomers, derivatives thereof, antibodies for the same, method for production and use therof
WO2004069182A2 (en) 2003-02-01 2004-08-19 Neuralab Limited Active immunization to generate antibodies to soluble a-beta
WO2004069826A1 (en) 2003-02-04 2004-08-19 F. Hoffmann-La Roche Ag Malonamide derivatives as gamma-secretase inhibitors
WO2004069162A2 (en) 2003-01-31 2004-08-19 Merck & Co., Inc. 3-amino-4-phenylbutanoic acid derivatives as dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
WO2004071408A2 (en) 2003-02-10 2004-08-26 Applied Molecular Evolution, Inc. Aβ BINDING MOLECULES
WO2004071454A2 (en) 2003-02-13 2004-08-26 Guilford Pharmaceuticals Inc. Substituted azetidine compounds as inhibitors of dipeptidyl peptidase iv
WO2004073630A2 (en) 2003-02-18 2004-09-02 Roskamp Research Llc Anti-angiogenic and anti-tumoral properties of beta and gamma secretase inhibitors
WO2004076434A1 (en) 2003-02-28 2004-09-10 Aic Dipeptidyl peptidase inhibitors
WO2004076433A1 (en) 2003-02-28 2004-09-10 Aic Dipeptidyl peptidase inhibitors
WO2004078908A2 (en) 2003-03-06 2004-09-16 Santhera Pharmaceuticals (Schweiz) Gmbh Alpha-keto carbonyl calpain inhibitors
WO2004080419A2 (en) 2003-03-12 2004-09-23 Neuralab Limited Humanized antibodies that recognize beta amyloid peptide
WO2004084884A1 (en) 2003-03-24 2004-10-07 The Chinese University Of Hong Kong Use of tanshinone derivates as cholinesterase inhibitors in treating related diseases
WO2004084830A2 (en) 2003-03-21 2004-10-07 Buck Institute Method for treating alzheimer’s dementia
WO2004087053A2 (en) 2003-03-25 2004-10-14 Syrrx, Inc. Dipeptidyl peptidase inhibitors
WO2004087158A2 (en) 2003-03-28 2004-10-14 Acadia Pharmaceuticals Inc. Muscarinic m1 receptor agonists for pain management
WO2004089351A2 (en) 2003-04-10 2004-10-21 Cambridge University Technical Services Ltd Treatment of progressive neurological disorders comprising a mao inhibitor (miclobemid) and an acetylcholinesterase inhibitor (tacrin)
WO2004089911A1 (en) 2003-04-10 2004-10-21 Merck Sharp & Dohme Limited Pyrazole derivatives as gamma-secretase inhibitors useful in the treatment of alzheimer’s disease
WO2004089362A1 (en) 2003-04-11 2004-10-21 Novo Nordisk A/S 2-cyanopyrroles and their analogues as ddp-iv inhibitors
WO2004092189A1 (en) 2003-04-09 2004-10-28 Wyeth Derivatives of 2-(8,9-dioxo-2,6-diazabicyclo(5.2.0)non-1(7)-en-2-yl)alkyl phosphonic acid and their use as n-methyl-d-aspartate (nmda) recetor antagonists
WO2004098625A2 (en) 2003-05-05 2004-11-18 Probiodrug Ag Medical use of inhibitors of glutaminyl and glutamate cyclases
WO2004099185A1 (en) 2003-05-09 2004-11-18 Fujisawa Pharmaceutical Co. Ltd. 2-cyanopyrrolidine derivatives and their use as dpp-iv inhibitors
WO2004098631A1 (en) 2003-05-08 2004-11-18 Universidad De Zaragoza Alzheimer's disease treatment method
WO2004098591A2 (en) 2003-05-05 2004-11-18 Probiodrug Ag Inhibitors of glutaminyl cyclase and their use in the treatment of neurological diseases
WO2004099134A2 (en) 2003-05-05 2004-11-18 Prosidion Ltd. Glutaminyl based dp iv-inhibitors
WO2004101562A2 (en) 2003-05-13 2004-11-25 Schering Corporation Bridged n-arylsulfonylpiperidines as gamma-secretase inhibitors
WO2004101538A1 (en) 2003-05-16 2004-11-25 Merck Sharp & Dohme Limited Cyclohexyl sulphones as gamma-secretase inhibitors
WO2004103276A2 (en) 2003-05-14 2004-12-02 Merck & Co., Inc. 3-amino-4-phenylbutanoic acid derivatives as dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
WO2004103993A1 (en) 2003-05-14 2004-12-02 Syrrx, Inc. Dipeptidyl peptidase inhibitors
WO2004108895A2 (en) 2003-05-30 2004-12-16 Neuralab Limited Humanized antibodies that recognize beta amyloid peptide
WO2004108730A1 (en) 2003-06-05 2004-12-16 Fujisawa Pharmaceutical Co., Ltd. 1h-imidazo`4,5-d!pyridazines as dpp-iv inhibitors for the treatment of niddm
WO2004111041A1 (en) 2003-06-12 2004-12-23 Fujisawa Pharmaceutical Co., Ltd. Pyrrolidine, thiazolidine and oxazolidine compounds which inhibit dipeptidyl peptidase-iv (dpp)
WO2004110436A1 (en) 2003-06-06 2004-12-23 Merck & Co., Inc. Fused indoles as dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
WO2004112701A2 (en) 2003-06-17 2004-12-29 Merck & Co., Inc. Cyclohexylglycine derivatives as dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
WO2005000193A2 (en) 2003-06-30 2005-01-06 Tel Aviv University Future Technology Development L.P. Peptides antibodies directed thereagainst and methods using same for diagnosing and treating amyloid-associated diseases
WO2005000846A1 (en) 2003-06-20 2005-01-06 F.Hoffmann-La Roche Ag Hexahydropyridoisoqinolines as dpp-iv inhibitors
WO2005000848A1 (en) 2003-06-20 2005-01-06 F. Hoffmann-La Roche Ag Pyrido` 2, 1-a - isoquinoline derivatives as dpp-iv inhibitors
WO2005000216A2 (en) 2003-05-27 2005-01-06 Forest Laboratories, Inc. Combination of an nmda receptor antagonist and a selective serotonin reuptake inhibitor for the treatment of depression and other mood disorders
WO2005004802A2 (en) 2003-06-30 2005-01-20 Merck & Co., Inc. N-alkyl phenylcarboxamide beta-secretase inhibitors for the treatment of alzheimer's disease
WO2005004803A2 (en) 2003-07-01 2005-01-20 Merck & Co., Inc. Phenylcarboxylate beta-secretase inhibitors for the treatment of alzheimer's disease
WO2005008250A1 (en) 2003-07-21 2005-01-27 Angiogenetics Sweden Ab Compounds and methods for promoting angiogenesis by using a gamma-secretase inhibitor or inhibiting the gamma-secretase pathway
WO2005007614A1 (en) 2003-07-03 2005-01-27 The Government Of The United States Of America As Represented By The Secretary, Department Of Health And Human Services 6011 Monoamine oxidase inhibitors
WO2005007199A1 (en) 2003-07-16 2005-01-27 Rvx Therapeutics, Inc. COMPOUNDS AND METHODS FOR DOWNREGULATING THE EFFECTS OF TGF-β
WO2005009421A2 (en) 2003-07-28 2005-02-03 Merz Pharma Gmbh & Co. Kgaa The use of 1-amino-alkylcyclohexane compounds in the treatment of pain hypersensitivity
WO2005011599A2 (en) 2003-08-01 2005-02-10 Northwestern University Antibodies specific for toxic amyloid beta protein oligomers
WO2005011581A2 (en) 2003-07-31 2005-02-10 Merck & Co., Inc. Hexahydrodiazepinones as dipeptidyl peptidase-iv inhibitors for the treatment or prevention of diabetes
WO2005014553A1 (en) 2003-08-05 2005-02-17 Merck Sharp & Dohme Limited Novel gamma-secretase inhibitors
WO2005016911A1 (en) 2003-08-13 2005-02-24 Takeda Pharmaceutical Company Limited 4-pyrimidone derivatives and their use as peptidyl peptidase inhibitors
WO2005018424A2 (en) 2003-08-18 2005-03-03 Research Foundation For Mental Hygiene, Inc. Antibodies specific for fibrillar amyloid and a procedure to detect fibrillar amyloid deposits
WO2005018545A2 (en) 2003-08-14 2005-03-03 Merck & Co., Inc. Macrocyclic beta-secretase inhibitors for the treatment of alzheimer's disease
WO2005023762A1 (en) 2003-09-04 2005-03-17 Abbott Laboratories Pyrrolidine-2-carbonitrile derivatives and their use as inhibitors of dipeptidyl peptidase-iv (dpp-iv)
WO2005023858A1 (en) 2003-09-05 2005-03-17 Cellzome Ag Protein complexes associated with app-processing
WO2005025554A2 (en) 2003-09-09 2005-03-24 Japan Tobacco Inc. Dipeptidyl peptidase iv inhibitor
WO2005025616A1 (en) 2003-09-09 2005-03-24 Takeda Pharmaceutical Company Limited Use of antibody
WO2005026148A1 (en) 2003-09-08 2005-03-24 Takeda San Diego, Inc. Dipeptidyl peptidase inhibitors
WO2005025516A2 (en) 2003-09-12 2005-03-24 The Regents Of The University Of California Monoclonal antibodies specific for conformational epitopes of prefibrillar aggregates
WO2005027975A1 (en) 2003-09-22 2005-03-31 Pfizer Limited Combinations of alpha-2-delta ligands and acetylcholinesterase inhibitors
WO2005028440A1 (en) 2003-09-16 2005-03-31 Schering Corporation Novel gamma secretase inhibitors
WO2005028511A2 (en) 2003-03-28 2005-03-31 Centocor, Inc. Anti-amyloid antibodies, compositions, methods and uses
WO2005030731A1 (en) 2003-09-24 2005-04-07 Merck Sharp & Dohme Limited Gamma-secretase inhibitors
WO2005030751A2 (en) 2003-09-08 2005-04-07 Takeda Pharmaceutical Company Limited Dipeptidyl peptidase inhibitors
WO2005033106A1 (en) 2003-10-06 2005-04-14 Alangudi Sankaranarayanan Azolidinecarbonitriles and their use as dpp-iv inhibitors
WO2005032471A2 (en) 2003-10-03 2005-04-14 Merck & Co., Inc. Benzylether and benzylamino beta-secretase inhibitors for the treatment of alzheimer's disease
WO2005035522A1 (en) 2003-10-08 2005-04-21 Pfizer Japan, Inc. 1-‘2-(4-hydroxyphenyl)-2-hydroxyethyl!-piperidin-4-ol compounds as nmda receptor antagonists
WO2005037828A1 (en) 2003-10-20 2005-04-28 Lg Life Sciences Ltd. Novel inhibitors of dpp-iv, methods of preparing the same, and pharmaceutical compositions containing the same as an active agent
WO2005040095A1 (en) 2003-10-16 2005-05-06 Astrazeneca Ab Inhibitors of dipeptidyl peptidase iv
WO2005040126A1 (en) 2003-10-06 2005-05-06 F. Hoffman-La Roche Ag Substituted dibenzo-azepine and benzo-diazepine derivatives useful as gamma-secretase inhibitors
WO2005039580A1 (en) 2003-10-16 2005-05-06 Boehringer Ingelheim International Gmbh Pharmaceutical composition comprising a monoamine neurotransmitter re-uptake inhibitor and an acetylcholinesterase inhibitor
WO2005039548A2 (en) 2003-10-15 2005-05-06 Probiodrug Ag Use of effectors of glutaminyl and glutamate cyclases
WO2005044830A1 (en) 2003-11-11 2005-05-19 F. Hoffmann-La Roche Ag Phosphinic acids derivatives, beta-secretase inhibitors for the treatment of alzheimer’s disease
WO2005044195A2 (en) 2003-11-04 2005-05-19 Merck & Co., Inc. Fused phenylalanine derivatives as dipeptidyl peptidase-iv inhibitors for the treatment or prevention of diabetes
WO2005047297A1 (en) 2003-11-12 2005-05-26 Phenomix Corporation Heterocyclic boronic acid compounds
WO2005051914A1 (en) 2003-11-24 2005-06-09 Merck & Co., Inc. Benzylether and benzylamino beta-secretase inhibitors for the treatment of alzheimer's disease
WO2005051950A1 (en) 2003-11-27 2005-06-09 Boehringer Ingelheim International Gmbh Novel 8-(piperazine-1-yl)- and 8-([1,4]diazepan-1-yl)-xanthine, the production and use thereof in the from of a drug
WO2005055996A1 (en) 2003-12-05 2005-06-23 Forest Laboratories, Inc. Memantine for the prevention or reduction of suicidality and for treatment of major depression associated with suicidality
WO2005056003A1 (en) 2003-12-09 2005-06-23 Santhera Pharmaceuticals (Schweiz) Gmbh Dpp-iv inhibitors
WO2005056013A1 (en) 2003-12-09 2005-06-23 Santhera Pharmaceuticals (Schweiz) Gmbh Dpp-iv inhibitors
WO2005058849A1 (en) 2003-12-15 2005-06-30 Glenmark Pharmaceuticals Ltd. New dipeptidyl peptidase in inhibitors; process for their preparation and compositions containing them
WO2005065195A2 (en) 2003-12-19 2005-07-21 Merck & Co., Inc. Phenylamide and pyridylamide beta-secretase inhibitors for the treatment of alzheimer's disease
WO2005070429A1 (en) 2004-01-22 2005-08-04 Neurosearch A/S Pharmaceutical composition comprising a monoamine neurotransmitter re-uptake inhibitor and an n-methyl-d-aspartate (nmda) receptors antagonist
WO2005072705A1 (en) 2004-01-29 2005-08-11 Neuromolecular, Inc. Combination of a nmda receptor antagonist and a mao-inhibitor or a gadpf-inhibitor for the treatment of central nervous system-related conditions
WO2005075436A2 (en) 2004-02-05 2005-08-18 Probiodrug Ag Novel inhibitors of glutaminyl cyclase
WO2005075426A1 (en) 2004-02-03 2005-08-18 Glenmark Pharmaceuticals Ltd. Novel dipeptidyl peptidase iv inhibitors; processes for their preparation and compositions thereof
WO2005079779A1 (en) 2003-10-22 2005-09-01 Merz Pharma Gmbh & Co. Kgaa THE USE OF 1-AMINOCYCLOHEXANE DERIVATIVES TO MODIFY DEPOSITION OF FIBRILLOGENIC Aß PEPTIDES IN AMYLOIDOPATHIES
WO2005079789A1 (en) 2004-02-17 2005-09-01 Axonyx, Inc. Combinatorial therapy with an acetylcholinesterase inhibitor and (3ar)-1,3a,8-trimethyl-1,2,3,3a,8,8a-hexahydropyrrolo[2,3-b]indol-5-yl phenylcarbamate
WO2005079756A2 (en) 2004-02-13 2005-09-01 Neuromolecular, Inc. Combination of a nmda receptor antagonist and an anti-depressive drug mao-inhibitor or a gadph-inhibitor for the treatment of psychiatric conditions
WO2005080435A1 (en) 2004-02-20 2005-09-01 Immuno-Biological Laboratories Co., Ltd. Monoclonal antibody and use thereof
WO2005081872A2 (en) 2004-02-20 2005-09-09 Ac Immune Sa Methods and compositions comprising supramolecular constructs
WO2005082348A2 (en) 2004-02-23 2005-09-09 Trustees Of Tufts College Inhibitors of dipeptidylpeptidase iv for regulating glucose metabolism
WO2005085246A1 (en) 2004-02-18 2005-09-15 Boehringer Ingelheim International Gmbh 8-[3-amino-piperidin-1-yl]-xanthine, the production thereof and the use in the form of a dpp inhibitor
WO2005087235A1 (en) 2004-03-09 2005-09-22 National Health Research Institutes Pyrrolidine compounds
WO2005092009A2 (en) 2004-03-19 2005-10-06 Axonyx, Inc. Acetylcholinesterase inhibitors and n-methyl-d-aspartate antagonists useful in the treatment of cognitive disorders
WO2005095381A1 (en) 2004-03-15 2005-10-13 Takeda Pharmaceutical Company Limited Dipeptidyl peptidase inhibitors
WO2005095343A1 (en) 2004-03-05 2005-10-13 Santhera Pharmaceuticals (Schweiz) Gmbh Dpp-iv inhibitors
WO2005095339A1 (en) 2004-03-31 2005-10-13 Pfizer Products Inc. Dicyanopyrrolidines as dipeptidyl peptidase iv inhibitors
WO2005097103A2 (en) 2004-04-01 2005-10-20 Axys Pharmaceuticals, Inc. Diabetes and metabolic syndrome therapy utilizing cathepsin b inhibitors
WO2005097768A2 (en) 2004-04-05 2005-10-20 Schering Corporation Novel gamma secretase inhibitors
WO2005103020A1 (en) 2004-04-20 2005-11-03 Merck & Co., Inc. 1,3,5-substituted phenyl derivative compounds useful as beta-secretase inhibitors for the treatment of alzheimer's disease
WO2005102390A2 (en) 2004-04-22 2005-11-03 Pfizer Japan, Inc. Combinations comprising alpha-2-delta ligands and nmda receptor antagonists
WO2005103043A1 (en) 2004-04-20 2005-11-03 Merck & Co., Inc. 2, 4, 6-substituted pyridyl derivative compounds useful as beta-secretase inhibitors for the treatment of alzheimer’s disease
WO2005105133A2 (en) 2004-04-23 2005-11-10 Massachusetts Eye And Ear Infirmary Methods and compositions for preserving the viability of photoreceptor cells
WO2005105998A1 (en) 2004-04-27 2005-11-10 Juridical Foundation The Chemo-Sero-Therapeutic Research Institute HUMAN ANTIAMYLOID β PEPTIDE ANTIBODY AND ANTIBODY FRAGMENT THEREOF
WO2005108382A1 (en) 2004-05-04 2005-11-17 Merck & Co., Inc. 1,2,4-oxadiazole derivatives as dipeptidyl peptidase-iv inhibitors for the treatment or prevention of diabetes
WO2005113510A1 (en) 2004-05-21 2005-12-01 Santhera Pharmaceuticals (Schweiz) Ag Dpp-iv inhibitors
WO2005113484A1 (en) 2004-05-13 2005-12-01 Merck & Co., Inc. Phenyl carboxamide compounds useful as beta-secretase inhibitors for the treatment of alzheimer's disease
WO2005116029A1 (en) 2004-05-18 2005-12-08 Merck & Co., Inc. Cyclohexylalanine derivatives as dipeptidyl peptidase-iv inhibitors for the treatment or prevention of diabetes
WO2005116014A1 (en) 2004-05-12 2005-12-08 Pfizer Products Inc. Proline derivatives and their use as dipeptidyl peptidase iv inhibitors
WO2005118555A1 (en) 2004-06-04 2005-12-15 Takeda Pharmaceutical Company Limited Dipeptidyl peptidase inhibitors
WO2005120494A1 (en) 2004-06-08 2005-12-22 Santhera Pharmaceuticals (Schweiz) Ag 1-`(3r)-amino-4-(2-fluoro-phenyl)-butyl !-pyrrolidine-(2r)-carboxilic acid-benzyl amine derivatives and related compounds as dipeptidyl-peptidase iv (dpp-iv) inhibitors for the treatment of type 2 diabetes mellitus
WO2005121131A1 (en) 2004-06-08 2005-12-22 Santhera Pharmaceuticals (Schweiz) Ag Dpp-iv inhibitors
WO2005121089A1 (en) 2004-06-08 2005-12-22 Santhera Pharmaceuticals (Schweiz) Ag Dpp-iv inhibitors
WO2005120571A2 (en) 2004-06-07 2005-12-22 Ramot At Tel Aviv University Ltd. Method of passive immunization against disease or disorder characterized by amyloid aggregation with diminished risk of neuroinflammation
WO2005123775A1 (en) 2004-06-21 2005-12-29 Bioarctic Neuroscience Ab Antibodies specific for soluble amyloid beta peptide protofibrils and uses thereof
WO2005123685A1 (en) 2004-06-16 2005-12-29 Astrazeneca Ab Tetrahydroquinolones and aza-analogues thereof for use as dpp-iv inhibitors in the treatement of diabetes
US6982264B2 (en) 2001-06-27 2006-01-03 Elan Pharmaceuticals, Inc. Substituted alcohols useful in treatment of Alzheimer's disease
WO2006002004A1 (en) 2004-06-15 2006-01-05 Merck & Co., Inc. Pyrrolidin-3-yl compounds useful as beta-secretase inhibitors for the treatment of alzheimer's disease
WO2006004880A2 (en) 2004-06-30 2006-01-12 Schering Corporation Substituted n-arylsulfonylheterocyclic amines as gamma-secretase inhibitors
WO2006008661A2 (en) 2004-07-19 2006-01-26 Neurochem (International) Limited Diagnostic methods of multiple organ amyloidosis
WO2006009886A1 (en) 2004-06-21 2006-01-26 Merck & Co., Inc. Aminocyclohexanes as dipeptidyl peptidase-iv inhibitors for the treatment or prevention of diabetes
US6992081B2 (en) 2000-03-23 2006-01-31 Elan Pharmaceuticals, Inc. Compounds to treat Alzheimer's disease
WO2006010965A1 (en) 2004-07-29 2006-02-02 Richter Gedeon Vegyészeti Gyár Rt. Indole-2 -carboxamidine derivatives as nmda receptor antagonists
WO2006014638A2 (en) 2004-07-19 2006-02-09 The General Hospital Corporation ANTIBODIES TO CROSS-LINKED AMYLOID β OLIGOMERS
WO2006014478A1 (en) 2004-07-02 2006-02-09 Northwestern University MONOLOCAL ANTIBODIES THAT TARGET PATHOLOGICAL ASSEMBLIES OF AMYLOID β (ABETA)
WO2006013104A1 (en) 2004-08-05 2006-02-09 Santhera Pharmaceuticals (Schweiz) Ag Heterocyclic compounds useful as dpp- iv inhibitors
WO2006014762A1 (en) 2004-07-22 2006-02-09 Schering Corporation Substituted amide beta secretase inhibitors
WO2006014944A1 (en) 2004-07-28 2006-02-09 Schering Corporation Macrocyclic beta-secretase inhibitors
WO2006017292A1 (en) 2004-07-12 2006-02-16 Phenomix Corporation Constrained cyano compounds
WO2006016644A1 (en) 2004-08-11 2006-02-16 Mitsubishi Chemical Corporation Antibody and utilization of the same
WO2006020017A2 (en) 2004-07-16 2006-02-23 Takeda Pharmaceutical Company Limited Dipeptidyl peptidase inhibitors
WO2006023750A2 (en) 2004-08-23 2006-03-02 Merck & Co., Inc. Fused triazole derivatives as dipeptidyl peptidase-iv inhibitors for the treatment or prevention of diabetes
WO2006021413A1 (en) 2004-08-25 2006-03-02 Santhera Pharmaceuticals (Schweiz) Ag Alpha-keto carbonyl calpain inhibitors
WO2006021409A1 (en) 2004-08-25 2006-03-02 Santhera Pharmaceuticals (Schweiz) Ag Alpha-keto carbonyl calpain inhibitors
WO2006026408A2 (en) 2004-08-27 2006-03-09 Wyeth Research Ireland Limited Production of anti-amyloid beta antibodies
WO2006026204A2 (en) 2004-08-26 2006-03-09 Bristol-Myers Squibb Company Novel gamma-lactams as beta-secretase inhibitors
WO2006029850A1 (en) 2004-09-14 2006-03-23 The Genetics Company, Inc. Hydrazone derivatives and their use as beta secretase inhibitors
WO2006034296A2 (en) 2004-09-17 2006-03-30 Comentis, Inc. Amino-containing compounds which inhibit memapsin 2 beta-secretase activity and methods of use thereof
WO2006034277A1 (en) 2004-09-17 2006-03-30 Comentis, Inc. Bicyclic compounds which inhibit beta-secretase activity and methods of use thereof
WO2006036291A2 (en) 2004-07-30 2006-04-06 Rinat Neuroscience Corp. Antibodies directed against amyloid-beta peptide and methods using same
WO2006039325A2 (en) 2004-10-01 2006-04-13 Merck & Co., Inc. Aminopiperidines as dipeptidyl peptidase-iv inhibitors for the treatment or prevention of diabetes
WO2006039470A2 (en) 2004-09-29 2006-04-13 Centocor, Inc. Anti- amyloid antibodies, compositions, methods and uses
US7030239B2 (en) 2000-03-23 2006-04-18 Elan Pharmaceuticals, Inc. Compounds to treat Alzheimer's disease
WO2006040688A2 (en) 2004-10-12 2006-04-20 Ernir Snorrason Inhibitors of acetylcholinesterase for treating skin diseases
WO2006042103A2 (en) 2004-10-05 2006-04-20 Axys Pharmaceuticals, Inc. Reversible inhibitors of cathepsin b
WO2006039807A1 (en) 2004-10-15 2006-04-20 Biopharmacopae Design International Inc. Methods and therapeutic compositions comprising plant extracts for the treatment of cancer
WO2006041976A1 (en) 2004-10-08 2006-04-20 Novartis Ag Combination of organic compounds
US7034182B2 (en) 2000-06-30 2006-04-25 Elan Pharmaceuticals, Inc. Compounds to treat Alzheimer's disease
WO2006044497A2 (en) 2004-10-13 2006-04-27 Merck & Co., Inc. Spiropiperidine compounds useful as beta-secretase inhibitors for the treatment of alzhermer’s disease
WO2006046644A1 (en) 2004-10-28 2006-05-04 Sanko Junyaku Co., Ltd. Method of examining alzheimer’s disease and diagnostic reagent
WO2006047248A1 (en) 2004-10-25 2006-05-04 Novartis Ag Combination of dpp-iv inhibitor, ppar antidiabetic and metformin
WO2006055434A2 (en) 2004-11-17 2006-05-26 Merck & Co., Inc. Macrocyclic tertiary amine beta-secretase inhibitors for the treatment of alzheimer's disease
WO2006055178A2 (en) 2004-10-25 2006-05-26 Merck & Co., Inc. Anti-addl antibodies and uses thereof
WO2006058059A2 (en) 2004-11-23 2006-06-01 Neuromolecular Pharmaceuticals, Inc. Composition comprising a sustained release coating or matrix and an nmda receptor antagonist, method for administration such nmda antagonist to a subject
WO2006058236A2 (en) 2004-11-24 2006-06-01 Neuromolecular Pharmaceuticals, Inc. Composition comprising an nmda receptor antagonist and levodopa and use thereof for treating neurological disease
WO2006057983A1 (en) 2004-11-23 2006-06-01 Merck & Co., Inc. Macrocyclic aminopyridyl beta-secretase inhibitors for the treatment of alzheimer's disease
WO2006058064A2 (en) 2004-11-29 2006-06-01 Merck & Co., Inc. Fused aminopiperidines as dipeptidyl peptidase-iv inhibitors for the treatment or prevention of diabetes
WO2006057945A2 (en) 2004-11-23 2006-06-01 Merck & Co., Inc. 2,3,4,6-substituted pyridyl derivative compounds useful as beta-secretase inhibitors for the treatment of alzheimer's disease
WO2006058720A2 (en) 2003-11-03 2006-06-08 Probiodrug Ag Novel compounds for the treatment of neurological disorders
WO2006060473A2 (en) 2004-12-03 2006-06-08 Mucosal Therapeutics Llc Methods of treatment of injured or diseased joints with lubricin compositions
WO2006058628A2 (en) 2004-11-30 2006-06-08 F.Hoffmann-La Roche Ag Substituted benzoquinolizines as dpp-iv inhibitors for the treatment of diabetes
WO2006060109A1 (en) 2004-10-29 2006-06-08 Merck & Co., Inc. 2-aminopyridine compounds useful as beta-secretase inhibitors for the treatment of alzheimer's disease
WO2006066233A1 (en) 2004-12-15 2006-06-22 Neuralab Limited An immunoprecipitation-based assay for predicting in vivo efficacy of beta-amyloid antibodies
WO2006066171A1 (en) 2004-12-15 2006-06-22 Neuralab Limited Amyloid βετα antibodies for use in improving cognition
WO2006066089A1 (en) 2004-12-15 2006-06-22 Neuralab Limited Humanized amyloid beta antibodies for use in improving cognition
WO2006066049A2 (en) 2004-12-15 2006-06-22 Neuralab Limited Humanized antibodies that recognize beta amyloid peptide
WO2006068978A2 (en) 2004-12-21 2006-06-29 Takeda Pharmaceutial Company Limited Dipeptidyl peptidase inhibitors
WO2006066747A1 (en) 2004-12-20 2006-06-29 F. Hoffmann-La Roche Ag 4-aminopiperidine derivatives
WO2006066770A1 (en) 2004-12-20 2006-06-29 F.Hoffmann-La Roche Ag Cycloalkylamine derivatives
WO2006070394A1 (en) 2004-12-28 2006-07-06 Council Of Scientific And Industrial Research Substituted carbamic acid quinolin-6-yl esters useful as acetylcholinesterase inhibitors
WO2006071274A2 (en) 2004-12-23 2006-07-06 Voyager Pharmaceutical Corporation Leuprolide acetate and acetylcholinesterase inhibitors or nmda receptor antagonists for the treatment of alzheimer’s disease
WO2006074265A2 (en) 2005-01-06 2006-07-13 Merck & Co., Inc. Drug combination therapy and pharmaceutical compositions for treating inflammatory disorders
WO2006078576A2 (en) 2005-01-19 2006-07-27 Merck & Co., Inc. Aminomethyl beta-secretase inhibitors for the treatment of alzheimer's disease
WO2006081171A1 (en) 2005-01-24 2006-08-03 Amgen Inc. Humanized anti-amyloid antibody
WO2006085961A2 (en) 2004-06-30 2006-08-17 Centocor, Inc. Anti-mcp-1 antibodies, compositions, methods and uses
WO2006091988A1 (en) 2005-02-28 2006-08-31 Thomas Christian Lines Composition for treating mental health disorders
WO2006094674A1 (en) 2005-03-07 2006-09-14 Michael Hermanussen Nmda receptor antagonists in the medical intervention of metabolic disorders
WO2006095041A1 (en) 2005-03-09 2006-09-14 Consejo Superior De Investigaciones Científicas Method for the in vitro diagnosis of alzheimer's disease using a monoclonal antibody
WO2006095613A1 (en) 2005-03-09 2006-09-14 Konica Minolta Medical & Graphic, Inc. Rare earth activated alkaline earth metal fluorohalide photostimulable phosphor and radiation image conversion panel using the same
WO2006099352A1 (en) 2005-03-10 2006-09-21 Bristol-Myers Squibb Company Novel isophthalates as beta-secretase inhibitors
WO2006097624A1 (en) 2005-03-17 2006-09-21 Sanofi-Aventis 7-(2-(4-(3-trifluoromethyl-phenyl)-1,2,3,6-tetrahydro-pyrid-1-yl)ethyl) isoquinoline besylate salt, preparation and therapeutic use thereof
WO2006103116A1 (en) 2005-04-01 2006-10-05 Biotherapix Molecular Medicines S.L.U. Human antibodies with beta-amyloid peptide-binding capacity and their applications
WO2006116435A2 (en) 2005-04-27 2006-11-02 Novartis Ag Methods of treating atherosclerosis
WO2006118959A2 (en) 2005-04-29 2006-11-09 Rinat Neuroscience Corp. Antibodies directed against amyloid-beta peptide and methods using same
WO2006125202A2 (en) 2005-05-19 2006-11-23 Centocor, Inc. Anti- mcp-1 antibodies, compositions, methods and uses
WO2006137354A1 (en) 2005-06-21 2006-12-28 Medical & Biological Laboratories Co., Ltd. Antibody having inhibitory effect on amyloid fibril formation
WO2007068412A2 (en) 2005-12-12 2007-06-21 Ac Immune Sa A beta 1-42 specific monoclonal antibodies with therapeutic properties
WO2007068411A2 (en) 2005-12-12 2007-06-21 Ac Immune S.A. Therapeutic vaccine
WO2008011348A2 (en) 2006-07-14 2008-01-24 Ac Immune S.A. Humanized antibody against amyloid beta
WO2008055947A1 (en) 2006-11-09 2008-05-15 Probiodrug Ag Novel inhibitors of glutaminyl cyclase
WO2008055950A1 (en) 2006-11-09 2008-05-15 Probiodrug Ag Novel inhibitors of glutaminyl cyclase
WO2008055945A1 (en) 2006-11-09 2008-05-15 Probiodrug Ag 3-hydr0xy-1,5-dihydr0-pyrr0l-2-one derivatives as inhibitors of glutaminyl cyclase for the treatment of ulcer, cancer and other diseases
WO2008065141A1 (en) 2006-11-30 2008-06-05 Probiodrug Ag Novel inhibitors of glutaminyl cyclase
EP2011349A2 (en) 2006-03-17 2009-01-07 Vodafone Group PLC Improvements in an ehspa architecture

Family Cites Families (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4746669A (en) * 1985-12-23 1988-05-24 Merck & Co., Inc. Substituted thiazoles as immunoregulants
KR20030078958A (en) 2001-03-01 2003-10-08 시오노기세이야쿠가부시키가이샤 Nitrogen-containing heteroaryl compounds having hiv integrase inhibitory activity
WO2004005283A1 (en) 2002-07-09 2004-01-15 Vertex Pharmaceuticals Incorporated Imidazoles, oxazoles and thiazoles with protein kinase inhibiting activities
TWI341314B (en) 2002-08-09 2011-05-01 Astrazeneca Ab New compounds
WO2004031177A1 (en) 2002-09-30 2004-04-15 Banyu Pharmaceutical Co., Ltd. 2-aminobenzimidazole derivative
US7732162B2 (en) * 2003-05-05 2010-06-08 Probiodrug Ag Inhibitors of glutaminyl cyclase for treating neurodegenerative diseases
WO2005121132A1 (en) 2004-06-11 2005-12-22 Shionogi & Co., Ltd. Fused heterocyclic compound having anti-hcv effect
US20090221644A1 (en) 2005-06-30 2009-09-03 Stuart Edward Bradley Gpcr Agonists
US8492378B2 (en) 2006-08-03 2013-07-23 Takeda Pharmaceutical Company Limited GSK-3β inhibitor
JP5275236B2 (en) * 2006-09-29 2013-08-28 スミスクライン ビーチャム コーポレーション Substituted indole compounds
CN101790526A (en) * 2007-06-08 2010-07-28 雅培制药有限公司 5-heteroaryl substituted indazoles as kinase inhibitors
WO2008157273A1 (en) * 2007-06-14 2008-12-24 Smithkline Beecham Corporation Chemical compounds
EP2211619A1 (en) 2007-10-18 2010-08-04 Merck Sharp & Dohme Corp. Substituted 1,2,4-oxadiazoles and analogs thereof as cb2 receptor modulators, useful in the treatment of pain, respiratory and non-respiratory diseases
US8415353B2 (en) * 2008-03-04 2013-04-09 Merck Sharp & Dohme Corp. Amino-quinoxaline and amino-quinoline compounds for use as adenosine A2a receptor antagonists
WO2011038293A1 (en) * 2009-09-28 2011-03-31 Intermune, Inc. Cyclic peptide inhibitors of hepatitis c virus replication

Patent Citations (587)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS6137764B2 (en) 1980-08-14 1986-08-26 Tokyo Shibaura Electric Co
JPH049367B2 (en) 1984-02-08 1992-02-20
JPH0525125B2 (en) 1984-07-16 1993-04-12 Shin Meiwa Ind Co Ltd
EP0172458A2 (en) 1984-07-31 1986-02-26 Suntory Limited Novel biologically active compound having anti-prolyl endopeptidase activity
US4743616A (en) 1984-07-31 1988-05-10 Suntory Limited Novel biologically active compound having anti-prolyl endopeptidase activity
JPH0356486B2 (en) 1984-08-22 1991-08-28
JPH0331298B2 (en) 1984-09-18 1991-05-02 Sanyo Electric Co
US4873342A (en) 1985-04-16 1989-10-10 Suntory Limited Dipeptide derivative and synthesis and use thereof
US4772587A (en) 1985-04-16 1988-09-20 Suntory Limited Dipeptide derivative of fatty acid
EP0201741A2 (en) 1985-04-16 1986-11-20 Suntory Limited Dipeptide derivatives, processes for preparing them, pharmaceutical composition and use
EP0201743A2 (en) 1985-04-16 1986-11-20 Suntory Limited Dipeptide derivatives of fatty acids, process for preparing them, pharmaceutical composition and use
EP0201742A2 (en) 1985-04-16 1986-11-20 Suntory Limited N-Acylpyrrolidine derivatives, processes for preparing them, pharmaceutical composition and use
JPS62114957A (en) 1985-11-13 1987-05-26 Suntory Ltd Novel pyrrolidine derivative having prolylendopeptidase inhibiting action its production and use thereof
JPS62114978A (en) 1985-11-14 1987-05-26 Suntory Ltd Novel pyrrolidinylimide having prolyl-endopeptidase inhibiting activity and production and use thereof
EP0224272A1 (en) 1985-11-29 1987-06-03 Suntory Limited Pyrrolidinylamide ester derivative having anti-prolyl endopeptidase activity, its synthesis and use and a pharmaceutical composition containing it
US4757083A (en) 1985-11-29 1988-07-12 Suntory Limited Novel pyrrolidinylamide ester derivatives having anti-prolyl endopeptidase activity and synthesis and use thereof
US5198458A (en) 1986-02-04 1993-03-30 Suntory Limited Pyrrolidineamide derivatives of acylamino acid and pharmaceutical composition containing the same
US4826870A (en) 1986-02-04 1989-05-02 Suntory Limited Pyrrolidineamide derivative of acylamino acid and pharmaceutical containing the same
EP0232849A2 (en) 1986-02-04 1987-08-19 Suntory Limited Pyrrolidineamide derivative of acylamino acid, pharmaceutical composition and use
EP0253310A2 (en) 1986-07-11 1988-01-20 E.I. Du Pont De Nemours And Company Angiotensin II receptor blocking imidazoles
JPH0515314B2 (en) 1986-09-30 1993-03-01 Kogyo Gijutsuin
US4810721A (en) 1986-11-18 1989-03-07 Suntory Limited Pyrrolidineamide derivative having anti-prolyl endopeptidase
EP0268281A2 (en) 1986-11-18 1988-05-25 Suntory Limited Pyrrolidineamide derivative having anti-prolyl endopeptidase activity, compositions comprising the same, processes for the preparation of the same, and the use of the same for the preparation of a medicament of therapeutic value
US4956380A (en) 1986-11-20 1990-09-11 Ono Pharmaceutical Co., Ltd. Prolinal compounds useful in treating amnesia
EP0268190A1 (en) 1986-11-20 1988-05-25 Ono Pharmaceutical Co., Ltd. Prolinal derivatives
US5254550A (en) 1986-11-20 1993-10-19 Ono Pharmaceutical Co., Ltd. Prolinal derivatives and pharmaceutical compositions thereof
US5100904A (en) 1986-11-20 1992-03-31 Ono Pharmaceutical Co., Ltd. Prolinal compounds which are useful in treating amnesia
US5340832A (en) 1986-11-20 1994-08-23 Ono Pharmaceutical Co., Ltd. Prolinal derivatives useful for treating amnesia
JPS63162672A (en) 1986-12-25 1988-07-06 Ono Pharmaceut Co Ltd Novel prolinal derivative, its production and antiamnestic agent containing said derivative
EP0275482A2 (en) 1986-12-29 1988-07-27 Ono Pharmaceutical Co., Ltd. Proline derivatives
US5112847A (en) 1986-12-29 1992-05-12 Ono Pharmaceutical Co., Ltd. Prolinal derivatives having inhibitory activity on prolyl endopeptidase
US4983624A (en) 1986-12-29 1991-01-08 Ono Pharmaceutical Co., Ltd Novel prolinal derivatives
US5262431A (en) 1986-12-29 1993-11-16 Ono Pharmaceutical Co., Ltd. Prolinal derivatives and pharmaceutical compositions thereof
JPS63264454A (en) 1986-12-29 1988-11-01 Ono Pharmaceut Co Ltd Novel prolinal derivative, production thereof and antiamnesic agent containing said derivative
US4977180A (en) 1987-02-04 1990-12-11 Ono Pharmaceutical Co., Ltd. Novel prolinal derivatives
EP0277588A1 (en) 1987-02-04 1988-08-10 Ono Pharmaceutical Co., Ltd. Prolinal derivatives
US5091406A (en) 1987-02-04 1992-02-25 Ono Pharmaceutical Co., Ltd. Prolinal derivatives
JPS6469A (en) 1987-02-04 1989-01-05 Ono Pharmaceut Co Ltd Novel prolinal derivative, production thereof and drug for alleviating amnesia containing said derivative
US4857537A (en) 1987-02-23 1989-08-15 Ono Pharmaceutical Co., Ltd. Novel thiazolidine derivatives
EP0280956A1 (en) 1987-02-23 1988-09-07 Ono Pharmaceutical Co., Ltd. Thiazolidine derivatives
EP0461677A1 (en) 1987-02-23 1991-12-18 Ono Pharmaceutical Co., Ltd. Novel thiazolidine derivatives
JPS6442465A (en) 1987-08-07 1989-02-14 Wakunaga Pharma Co Ltd N-acylprolylpyrrolidine derivative, production and use thereof
EP0303434A1 (en) 1987-08-08 1989-02-15 Kissei Pharmaceutical Co. Ltd. Thiazolidine compounds
JPS6468396A (en) 1987-09-10 1989-03-14 Yakult Honsha Kk Novel peptide and amnesia alleviating drug comprising said peptide as active ingredient
WO1989006242A1 (en) 1987-10-08 1989-07-13 The Mclean Hospital Corporation Antibodies to a4 amyloid peptide
JPH01143897A (en) 1987-11-30 1989-06-06 Kissei Pharmaceut Co Ltd Thiazolidine derivative
JPH01250370A (en) 1987-12-23 1989-10-05 Zeria Pharmaceut Co Ltd Novel amino acid imide derivative, its production and use thereof
JPH01226880A (en) 1988-03-07 1989-09-11 Kissei Pharmaceut Co Ltd Thiazolidine derivative
EP0345428A1 (en) 1988-04-08 1989-12-13 Ono Pharmaceutical Co., Ltd. Anti-amnesic pyrrolidine derivatives
US5328899A (en) 1988-07-15 1994-07-12 The Salk Institute For Biological Studies NPY peptide analogs
EP0355794A2 (en) 1988-08-26 1990-02-28 Merrell Dow Pharmaceuticals Inc. Neuropeptide Y antagonists
EP0355793A2 (en) 1988-08-26 1990-02-28 Merrell Pharmaceuticals Inc. Neuropeptide Y agonists and partial agonists
JPH02275858A (en) 1988-09-14 1990-11-09 Yoshitomi Pharmaceut Ind Ltd Pyridine compound
US5028604A (en) 1988-12-08 1991-07-02 Zeria Pharmaceutical Co., Ltd. Condensed benzene derivative
EP0372484A2 (en) 1988-12-08 1990-06-13 Zeria Pharmaceutical Co., Ltd. Condensed benzene derivative
JPH02207070A (en) 1989-02-07 1990-08-16 Zeria Pharmaceut Co Ltd Amide acid imide derivative, drug containing same and intermediate for producing the compound
US5118811A (en) 1989-04-13 1992-06-02 Japan Tobacco Inc. Amino acid derivatives possessing prolyl endopeptidase-inhibitory activities
WO1990012870A1 (en) 1989-04-14 1990-11-01 Research Foundation For Mental Hygiene, Inc. Monoclonal antibody to amyloid peptide
WO1990012871A1 (en) 1989-04-14 1990-11-01 Research Foundation For Mental Hygiene, Inc. Cerebrovascular amyloid protein-specific monoclonal antibody sv17-6e10
WO1990014840A1 (en) 1989-06-06 1990-12-13 California Biotechnology Inc. Recombinant alzheimer's amyloid protein
EP0403159A2 (en) 1989-06-14 1990-12-19 Smithkline Beecham Corporation Imidazolyl-alkenoic acids
DE3939797A1 (en) 1989-12-01 1991-06-06 Basf Ag NEW PEPTIDES DERIVED FROM NEUROPEPTIDE Y
WO1991010664A1 (en) 1990-01-12 1991-07-25 American Home Products Corporation PYRIDAZINO[4,5-b]INDOLIZINES
EP0443983A1 (en) 1990-02-19 1991-08-28 Ciba-Geigy Ag Acyl compounds
JPH03255080A (en) 1990-03-05 1991-11-13 Yoshitomi Pharmaceut Ind Ltd Benzene compound
EP0454511A1 (en) 1990-03-20 1991-10-30 Sanofi N-substituted heterocycle derivatives, their preparation, compositions containing them
JPH06339390A (en) 1990-03-22 1994-12-13 Bristol Myers Squibb Co Bu-4164e-a and b and prolyl endopeptidase inhibitor
US5073549A (en) 1990-03-22 1991-12-17 Bristol-Myers Squibb Company BU-4164E - A and B, prolyl endopeptidase inhibitors and their methods of use
EP0451547A1 (en) 1990-03-22 1991-10-16 Bristol-Myers Squibb Company 1,4,8-Triazacyclotridecane derivatives having prolyl endopeptidase inhibiting activity
US4999349A (en) 1990-03-22 1991-03-12 Bristol-Myers Squibb Co. BU-4164E - A and B, prolyl endopeptidase inhibitors
EP0459136A1 (en) 1990-04-27 1991-12-04 Takeda Chemical Industries, Ltd. Benzimidazole derivatives, their production and use
US5847155A (en) 1990-06-04 1998-12-08 Pfizer Inc Aromatic pyrrolidine amide prolyl endopeptidase inhibitors
WO1991018877A1 (en) 1990-06-07 1991-12-12 Zeria Pharmaceutical Co., Ltd. Novel arylalkanoylamine derivative and drug containing the same
US5407950A (en) 1990-06-07 1995-04-18 Zeria Pharmaceutical Co., Ltd. Arylalkanoylamine derivative and drug containing the same
US5221752A (en) 1990-07-27 1993-06-22 Nippon Kayaku Kabushiki Kaisha α-keto amide derivatives
US5506256A (en) 1990-07-27 1996-04-09 Yoshitomi Pharmaceutical Industries, Ltd. Proline derivatives possessing prolyl endopeptidase-inhibitory activity
JPH04211648A (en) 1990-07-27 1992-08-03 Nippon Kayaku Co Ltd Keto-acid amide derivative
EP0468469A2 (en) 1990-07-27 1992-01-29 Japan Tobacco Inc. Proline derivatives
EP0468339A2 (en) 1990-07-27 1992-01-29 Nippon Kayaku Kabushiki Kaisha Alpha-keto-amide derivatives having protease inhibiting activity
JPH04235162A (en) 1990-08-09 1992-08-24 Zeria Pharmaceut Co Ltd Novel succinamide derivative and drug containing the same
JPH04208299A (en) 1990-11-30 1992-07-29 Ajinomoto Co Inc Prolyl endopeptidase-inhibiting peptide
EP0503785A1 (en) 1991-02-21 1992-09-16 Sankyo Company Limited 1-Biphenylimidazole derivatives, their preparation and their therapeutic use
WO1992019238A1 (en) 1991-05-01 1992-11-12 Mayo Foundation For Medical Education And Research Huperzine a analogs
WO1992021333A2 (en) 1991-05-24 1992-12-10 Pharmavene, Inc. Treatment of drug withdrawal symptoms and drug craving with type b monoamine oxidase inhibitors
WO1993000361A1 (en) 1991-06-20 1993-01-07 Snow Brand Milk Products Co., Ltd. Novel prolyl endopeptidase inhibitors sna-115 and sna-115t, production thereof, and strain which produces said inhibitors
EP0522314A1 (en) 1991-07-03 1993-01-13 BASF Aktiengesellschaft Thermoplastic moulding composition based on polycarbonates, styrene/acrylanitrile-polymerisates and polyolefins
WO1993003034A1 (en) 1991-07-29 1993-02-18 Warner-Lambert Company Quinazoline derivatives as acetylcholinesterase inhibitors
EP0539086A2 (en) 1991-10-24 1993-04-28 American Home Products Corporation Condensed pyrimidine derivatives and their use as angiotensine II antagonists
WO1993012139A1 (en) 1991-12-19 1993-06-24 Garvan Institute Of Medical Research A novel molecule which inhibits neuropeptide tyrosine biological function
EP0556482A2 (en) 1991-12-24 1993-08-25 Snow Brand Milk Products Co., Ltd. Propyl endopeptidase inhibitor
WO1993013065A1 (en) 1991-12-27 1993-07-08 Japan Tobacco Inc. Proline derivative
JPH05201970A (en) 1992-01-24 1993-08-10 Japan Tobacco Inc New proline derivative
JPH05331072A (en) 1992-05-27 1993-12-14 Agency Of Ind Science & Technol Prolyl endopeptidase inhibitor
WO1993025534A1 (en) 1992-06-10 1993-12-23 Zeneca Limited 2,5-DIOXO-2,5-DIHYDRO-1H-BENZ[b]AZEPINES AS NMDA RECEPTOR ANTAGONISTS
WO1994000486A1 (en) 1992-06-20 1994-01-06 The Wellcome Foundation Limited Neuropeptide y antagonists
JPH06116284A (en) 1992-10-02 1994-04-26 Yamanouchi Pharmaceut Co Ltd New peptide
WO1994007890A1 (en) 1992-10-05 1994-04-14 Ube Industries, Ltd. Pyrimidine compound
WO1994009016A1 (en) 1992-10-16 1994-04-28 Sri International 2-piperidinecarboxylic acid derivatives useful as nmda receptor antagonists
WO1994012474A1 (en) 1992-11-20 1994-06-09 Japan Tobacco Inc. Compound with prolyl endopeptidase inhibitor activity and pharmaceutical use thereof
EP0670309A1 (en) 1992-11-20 1995-09-06 Japan Tobacco Inc. Compound with prolyl endopeptidase inhibitor activity and pharmaceutical use thereof
WO1994013641A1 (en) 1992-12-16 1994-06-23 Japan Tobacco Inc. Benzomorphan useful as nmda receptor antagonist
JPH06192298A (en) 1992-12-24 1994-07-12 Mitsui Toatsu Chem Inc Novel function peptide
WO1994017035A1 (en) 1993-01-20 1994-08-04 Dr. Karl Thomae Gmbh Aminoacid derivates, medicaments containing these compounds and process for preparing the same
WO1994017197A1 (en) 1993-01-25 1994-08-04 Takeda Chemical Industries, Ltd. ANTIBODY AGAINST β-AMYLOID OR DERIVATIVE THEREOF AND USE THEREOF
JPH06234693A (en) 1993-02-09 1994-08-23 Snow Brand Milk Prod Co Ltd New isotetracenone-based substance and its production
EP0614911A1 (en) 1993-02-15 1994-09-14 Sanofi Compounds having a sulfonamide and an amidinegroup, process for preparation and pharmaceutical compositions comprising them
WO1994019356A1 (en) 1993-02-16 1994-09-01 Merrell Dow Pharmaceuticals Inc. Silylated acetylcholinesterase inhibitors
WO1994020476A1 (en) 1993-03-02 1994-09-15 Fujisawa Pharmaceutical Co., Ltd. Novel heterocyclic compound
WO1994020109A1 (en) 1993-03-03 1994-09-15 Rhone-Poulenc Rorer S.A. Use of 2h-1,2,4-benzothiadiazine 3(4h)-one 1,1 dioxide derivatives as non-competitive nmda receptor antagonists
EP0618193A1 (en) 1993-03-24 1994-10-05 Adir Et Compagnie Nitrogen containing bicyclic derivatives as prolyl-endopeptidase inhibitors
WO1994029255A1 (en) 1993-06-04 1994-12-22 Merrell Pharmaceuticals Inc. Aromatic acetylcholinesterase inhibitors
WO1995000161A1 (en) 1993-06-18 1995-01-05 University Of Cincinnati Neuropeptide y antagonists and agonists
WO1995001352A1 (en) 1993-06-30 1995-01-12 Zeria Pharmaceutical Co., Ltd. Thiazolidine derivative and medicine containing the same
WO1995002602A1 (en) 1993-07-16 1995-01-26 Rhone-Poulenc Rorer S.A. Imidazo(1,2-a)pyrazin-4-one derivatives for use as ampa and nmda receptor antagonists
WO1995002601A1 (en) 1993-07-16 1995-01-26 Rhone-Poulenc Rorer S.A. IMIDAZO[1,2-a]PYRAZINE-4-ONE, PREPARATION THEREOF AND DRUGS CONTAINING SAME
US5965556A (en) 1993-07-23 1999-10-12 Zaidan Hojin Biseibutsu Kagaku Kenkyukai Pyrrolidine derivatives
US5756763A (en) 1993-07-23 1998-05-26 Zaidan Hojin Biseibutsu Kagaku Kenkyukai Pyrrolidine derivatives
EP0709373A1 (en) 1993-07-23 1996-05-01 Zaidan Hojin Biseibutsu Kagaku Kenkyu Kai Novel pyrrolidine derivative
WO1995012594A1 (en) 1993-11-05 1995-05-11 Rhone-Poulenc Rorer S.A. 7H-IMIDAZO(1,2-a)PYRAZINE-8-ONE NMDA RECEPTOR ANTAGONISTS
WO1995015310A1 (en) 1993-12-02 1995-06-08 Merrell Pharmaceuticals Inc. Prolyl endopeptidase inhibitors
WO1995015309A1 (en) 1993-12-03 1995-06-08 Ferring B.V. Dp-iv-serine protease inhibitors
WO1995026342A1 (en) 1994-03-28 1995-10-05 Rhone-Poulenc Rorer S.A. 5H-INDENO[1,2-b]PYRAZINE-2,3-DIONE DERIVATIVES, PREPARATION THEREOF AND DRUGS CONTAINING SAME
WO1995026352A1 (en) 1994-03-28 1995-10-05 Rhone-Poulenc Rorer S.A. IMIDAZO[1,2-a]PYRAZINE-4-ONE DERIVATIVES, PREPARATION THEREOF AND DRUGS CONTAINING SAME
WO1995026350A1 (en) 1994-03-28 1995-10-05 Rhone-Poulenc Rorer S.A. Imidazo(1,2-a)indeno(1,2-e)pyrazine-4-one derivatives and pharmaceutical compositions containing same
WO1995026349A1 (en) 1994-03-28 1995-10-05 Rhone-Poulenc Rorer S.A. INDENO[1,2-e]PYRAZINE-4-ONES, PREPARATION THEREOF AND DRUGS CONTAINING SAME
JPH07267988A (en) 1994-03-31 1995-10-17 Yamanouchi Pharmaceut Co Ltd New peptide
WO1995031986A1 (en) 1994-05-24 1995-11-30 Astra Aktiebolag Combination product of dichloroacetic acid and an nmda antagonist
WO1996008485A1 (en) 1994-09-13 1996-03-21 Pfizer Limited Quinoxalinedione nmda receptor antagonists
US5567714A (en) 1994-10-20 1996-10-22 Eli Lilly And Company Methods of treating obesity by inhibiting physiological conditions associated with an excess of neuropeptide Y
US5663192A (en) 1994-10-20 1997-09-02 Eli Lilly And Company Heterocyclic neuropeptide Y receptor antagonists
WO1996012489A1 (en) 1994-10-20 1996-05-02 Eli Lilly And Company Bicyclic neuropeptide y receptor antagonists
WO1996012490A1 (en) 1994-10-20 1996-05-02 Eli Lilly And Company Methods of inhibiting conditions associated with neuropeptide y
WO1996014318A1 (en) 1994-11-02 1996-05-17 Rhone-Poulenc Rorer S.A. SPIRO[HETEROCYCLE-IMIDAZO[1,2-a]INDENO[1,2-e]PYRAZINE]-4'-ONES, PREPARATION THEREOF AND DRUGS CONTAINING SAME
WO1996020946A1 (en) 1994-12-29 1996-07-11 Research Development Foundation Flavin adenine dinucleotide analogues, their pharmaceutical compositions, and their activity as monoamine oxidase inhibitors
WO1996021655A2 (en) 1995-01-11 1996-07-18 Hoechst Marion Roussel, Inc. Substituted oxazolidine calpain and/or cathepsin b inhibitors
WO1996022305A2 (en) 1995-01-12 1996-07-25 The Wellcome Foundation Limited Modified peptides
WO1996025435A1 (en) 1995-02-14 1996-08-22 Bayer Corporation MONOCLONAL ANTIBODY SPECIFIC FOR βA4 PEPTIDE
US5552411A (en) 1995-05-26 1996-09-03 Warner-Lambert Company Sulfonylquinolines as central nervous system and cardiovascular agents
EP0747356A1 (en) 1995-06-07 1996-12-11 Bristol-Myers Squibb Company Dihydropyridine NPY antagonists: piperazine derivatives
EP0747357A2 (en) 1995-06-07 1996-12-11 Bristol-Myers Squibb Company Dihydropyridine NPY antagonists: piperidine derivatives
WO1996040660A1 (en) 1995-06-07 1996-12-19 Pfizer Inc. Certain substituted benzylamine derivatives: a new class of neuropeptide y1 specific ligands
EP0747378A1 (en) 1995-06-07 1996-12-11 Bristol-Myers Squibb Company Dihydropyridine NPY antagonists
JPH0940693A (en) 1995-07-31 1997-02-10 Gekkeikan Sake Co Ltd Inhibitor of prolyl endopeptidase
WO1997009308A1 (en) 1995-09-01 1997-03-13 Eli Lilly And Company Indolyl neuropeptide y receptor antagonists
WO1997012615A1 (en) 1995-10-05 1997-04-10 Warner-Lambert Company Benzimidazole derivatives as 15-lo inhibitors
WO1997013754A1 (en) 1995-10-11 1997-04-17 Pelayo Camps Garcia Novel polycyclic aminopyridine compounds as acetylcholinesterase inhibitors, preparation process and use thereof
WO1997019911A1 (en) 1995-11-30 1997-06-05 Dr. Karl Thomae Gmbh Amino acid derivatives, pharmaceutical compositions containing these compounds and processes for preparing them
WO1997019913A1 (en) 1995-11-30 1997-06-05 Dr. Karl Thomae Gmbh Amino acid derivatives, medicaments containing said compounds and methods of producing them
WO1997019914A1 (en) 1995-11-30 1997-06-05 Dr. Karl Thomae Gmbh Amino acid derivatives, medicaments containing said compounds and methods of producing said compounds
WO1997020820A1 (en) 1995-12-01 1997-06-12 Novartis Ag Heteroaryl compounds
WO1997020823A2 (en) 1995-12-01 1997-06-12 Novartis Ag 2-amino quinazoline derivatives as npy receptor antagonists
WO1997020822A1 (en) 1995-12-01 1997-06-12 Novartis Ag Quinazolin-2,4-diazirines as npy receptor antagonist
WO1997020821A1 (en) 1995-12-01 1997-06-12 Novartis Ag Heteroaryl derivatives
WO1997019682A1 (en) 1995-12-01 1997-06-05 Synaptic Pharmaceutical Corporation Aryl sulfonamide and sulfamide derivatives and uses thereof
JPH09157253A (en) 1995-12-12 1997-06-17 Yamanouchi Pharmaceut Co Ltd New amino acid derivative
WO1997023215A1 (en) 1995-12-22 1997-07-03 Warner-Lambert Company 2-substituted piperidine analogs and their use as subtype-selective nmda receptor antagonists
WO1997023214A1 (en) 1995-12-22 1997-07-03 Warner-Lambert Company 4-substituted piperidine analogs and their use as subtype selective nmda receptor antagonists
WO1997023216A1 (en) 1995-12-22 1997-07-03 Warner-Lambert Company 4-substituted piperidine analogs and their use as subtype selective nmda receptor antagonists
WO1997025041A1 (en) 1996-01-09 1997-07-17 Eli Lilly And Company Benzimidzolyl neuropeptide y receptor antagonists
WO1997032873A1 (en) 1996-03-09 1997-09-12 Pfizer Research And Development Company, N.V./S.A. Quinoxalinediones
WO1997034843A1 (en) 1996-03-22 1997-09-25 Libbey Glass Inc. Apparatus and method for forming a decorative pattern on glassware having an edge
WO1997038993A1 (en) 1996-04-12 1997-10-23 Hoechst Marion Roussel, Inc. Isatin derivatives as acetylcholinesterase inhibitors and analgesics
WO1997040832A1 (en) 1996-04-25 1997-11-06 Probiodrug Gesellschaft für Arzneimittelforschung mbH Use of dipeptidyl peptidase iv effectors for lowering the blood glucose level in mammals
WO1997046250A1 (en) 1996-06-04 1997-12-11 Synaptic Pharmaceutical Corporation Methods of modifying feeding behavior, compounds useful in such methods, and dna encoding a hypothalamic atypical neuropeptide y/peptide yy receptor (y5)
WO1998001157A1 (en) 1996-07-05 1998-01-15 The Wwk Trust Compositions for the treatment of peripheral neuropathies containing antidepressants and/or monoamine oxidase inhibitors and/or vitamin b12 and/or precursors or inducers of a neurotransmitter
WO1998003494A1 (en) 1996-07-23 1998-01-29 Neurogen Corporation Certain amido- and amino-substituted benzylamine derivatives; a new class of neuropeptite y1 specific ligands
WO1998003492A1 (en) 1996-07-23 1998-01-29 Neurogen Corporation Certain substituted benzylamine derivatives; a new class of neuropeptide y1 specific ligands
WO1998003493A1 (en) 1996-07-23 1998-01-29 Neurogen Corporation Certain substituted benzylamine derivatives; a new class of neuropeptide-y1 specific ligands
WO1998005337A1 (en) 1996-08-01 1998-02-12 Cocensys, Inc. Use of gaba and nmda receptor ligands for the treatment of migraine headache
WO1998006703A1 (en) 1996-08-14 1998-02-19 Warner-Lambert Company 2-phenyl benzimidazole derivatives as mcp-1 antagonists
WO1998007420A1 (en) 1996-08-23 1998-02-26 Agouron Acquisition Corp. Neuropeptide-y ligands
JPH1077300A (en) 1996-09-04 1998-03-24 Gekkeikan Sake Co Ltd Prolyl endopeptidase-inhibiting peptide
WO1998010757A2 (en) 1996-09-11 1998-03-19 The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services The use of functional n-methyl-d-aspartate antagonists to ameliorate or prevent aminoglycoside-induced ototoxicity
WO1998015647A1 (en) 1996-10-08 1998-04-16 Novartis Ag Modulation of apoptosis
US6114336A (en) 1996-10-23 2000-09-05 Sanofi Cosmetic composition containing a neuropeptide Y receptor antagonist
US6011155A (en) 1996-11-07 2000-01-04 Novartis Ag N-(substituted glycyl)-2-cyanopyrrolidines, pharmaceutical compositions containing them and their use in inhibiting dipeptidyl peptidase-IV
WO1998019998A2 (en) 1996-11-07 1998-05-14 Novartis Ag N-substituted 2-cyanopyrrolidines
US6124305A (en) 1996-11-07 2000-09-26 Novartis Ag Use of N-(substituted glycyl)-2-cyanopyrrolidines in inhibiting dipeptidyl peptidase-IV
WO1998030243A1 (en) 1997-01-08 1998-07-16 Warner-Lambert Company Acetylcholinesterase inhibitors in combination with muscarinic agonists for the treatment of alzheimer's disease
US6426365B1 (en) 1997-02-12 2002-07-30 Japan Tobacco Inc. CETP activity inhibitors
WO1998040102A1 (en) 1997-03-13 1998-09-17 The Provost, Fellows And Scholars Of The College Of The Holy And Undivided Trinity Of Queen Elizabeth Near Dublin Cytoprotective agents comprising monoamine oxidase inhibitors
WO1998044955A1 (en) 1997-04-09 1998-10-15 Mindset Ltd. Recombinant antibodies specific for beta-amyloid ends, dna encoding and methods of use thereof
WO1998046559A1 (en) 1997-04-16 1998-10-22 Arqule, Inc. SYNTHESIS AND USE OF α-KETOAMIDE DERIVATIVES AND ARRAYS
WO1998050044A1 (en) 1997-05-07 1998-11-12 Algos Pharmaceutical Corporation Composition and method combining an antidepressant with an nmda receptor antagonist, for treating neuropathic pain
WO1998050075A1 (en) 1997-05-07 1998-11-12 Algos Pharmaceutical Corporation Cox-2 inhibitors in combination with nmda-blockers for treating pain
WO1999001416A2 (en) 1997-06-30 1999-01-14 Merz + Co. Gmbh & Co. 1-amino-alkylcyclohexane nmda receptor antagonists
WO1999007351A2 (en) 1997-08-07 1999-02-18 Zeneca Limited Indole derivatives and their use as mcp-1 antagonists
WO1999007359A1 (en) 1997-08-08 1999-02-18 Shire International Licensing B.V. Use of cholinesterase inhibitors for treating attention deficit disorders
WO1999007413A1 (en) 1997-08-11 1999-02-18 Algos Pharmaceutical Corporation Substance p inhibitors in combination with nmda-blockers for treating pain
WO1999013878A1 (en) 1997-09-18 1999-03-25 Astrazeneca Ab A COMBINATION OF A MONOAMINE OXIDASE INHIBITOR AND A h5-HT1B ANTAGONIST OR PARTIAL AGONIST
WO1999015498A1 (en) 1997-09-23 1999-04-01 Astrazeneca Ab New npy antagonists
WO1999027944A1 (en) 1997-12-02 1999-06-10 Neuralab Limited Prevention and treatment of amyloidogenic disease
WO1999038501A2 (en) 1998-02-02 1999-08-05 Trustees Of Tufts College Method of regulating glucose metabolism, and reagents related thereto
WO1999046272A1 (en) 1998-03-09 1999-09-16 Fondatech Benelux N.V. Serine peptidase modulators
WO1999045963A1 (en) 1998-03-13 1999-09-16 Algos Pharmaceutical Corporation Analgesic combination comprising nmda receptor antagonists and narcotic analgesics
WO1999047131A2 (en) 1998-03-16 1999-09-23 Merck Sharp & Dohme Limited Combination of a gaba-a alpha 5 inverse agonist and an acetylcholinesterase inhibitor
WO1999046991A1 (en) 1998-03-17 1999-09-23 University Of Maryland Biotechnology Institute Methods for treating hiv-associated dementia
WO1999048891A1 (en) 1998-03-20 1999-09-30 Merck Patent Gmbh 1-(3-heteroarylpropyl- or -prop-2-enyl)-4-benzylpiperidines used as nmda receptor antagonists
WO1999053922A1 (en) 1998-04-21 1999-10-28 Algos Pharmaceutical Corporation Analgesic compositions comprising nmda-antagonists and abt-594
WO1999057119A1 (en) 1998-05-04 1999-11-11 Neotherapeutics, Inc. Novel dopamine-like 9-substituted hypoxanthine and methods of use
WO1999057120A1 (en) 1998-05-04 1999-11-11 Neotherapeutics, Inc. Novel serotonin-like 9-substituted hypoxanthine and methods of use
WO1999060024A1 (en) 1998-05-21 1999-11-25 The University Of Tennessee Research Corporation Methods for amyloid removal using anti-amyloid antibodies
WO1999061431A1 (en) 1998-05-28 1999-12-02 Probiodrug Gesellschaft für Arzneimittelforschung mbH New dipeptidyl peptidase iv effectors
WO1999067279A1 (en) 1998-06-24 1999-12-29 Probiodrug Gesellschaft für Arzneimittelforschung mbH Compounds of unstable dipeptidyl peptidase iv inhibitors
WO1999067278A1 (en) 1998-06-24 1999-12-29 Probiodrug Gesellschaft für Arzneimittelforschung mbH Prodrugs of dipeptidyl peptidase iv inhibitors
WO2000000197A1 (en) 1998-06-26 2000-01-06 Warner-Lambert Company 4-benzyl piperidine alkylsulfoxide heterocycles and their use as subtype-selective nmda receptor antagonists
WO2000002549A2 (en) 1998-07-10 2000-01-20 Du Pont Pharmaceuticals Company Composition for and method of treating neurological disorders
DE19834591A1 (en) 1998-07-31 2000-02-03 Probiodrug Ges Fuer Arzneim Use of substances that decrease the activity of dipeptidyl peptidase IV to increase blood sugar levels, e.g. for treating hypoglycemia
WO2000007600A1 (en) 1998-08-07 2000-02-17 R.J. Reynolds Tobacco Company Pharmaceutical compositions for the prevention and treatment of central nervous system disorders
WO2000009483A2 (en) 1998-08-17 2000-02-24 Finetech Ltd. Process and intermediates for production of donepezil and related compounds
WO2000015205A2 (en) 1998-09-11 2000-03-23 Eisai Co., Ltd. Use of acetylcholinesterase inhibitors for the preparation of pharmaceutical compositions for the treatment of functional and/or organic pain syndromes
WO2000023057A2 (en) 1998-10-16 2000-04-27 Janssen Pharmaceutica N.V. Atypical antiphsychotic in combination with acetylcholinesterase inhibitor for improving cognition
WO2000029023A1 (en) 1998-11-12 2000-05-25 Algos Pharmaceutical Corporation Cox-2 inhibitors in combination with nmda-blockers for treating pain
WO2000030446A1 (en) 1998-11-23 2000-06-02 Bonnie Davis Dosage formulations for acetylcholinesterase inhibitors
WO2000030674A1 (en) 1998-11-26 2000-06-02 Ferring Bv Neuropeptide y y4 agents in the treatment of reproductive disorders
WO2000033840A1 (en) 1998-12-11 2000-06-15 Bonnie Davis Use of acetylcholinesterase inhibitors on the modulation of the hypothalamic-pituitary-gonadal axis
WO2000046199A2 (en) 1999-02-05 2000-08-10 Astrazeneca Ab Indole derivatives as anti-inflammation agents
WO2000046197A1 (en) 1999-02-05 2000-08-10 Astrazeneca Ab Indole derivatives and their use as mcp-1 receptor antagonists
WO2000046196A1 (en) 1999-02-05 2000-08-10 Astrazeneca Ab Indole derivatives and their use as mcp-1 antagonists
WO2000046198A1 (en) 1999-02-05 2000-08-10 Astrazeneca Ab Anti-inflammatory indole derivatives
WO2000046195A1 (en) 1999-02-05 2000-08-10 Astrazeneca Ab Anti-inflammatory indole derivatives
WO2000056711A1 (en) 1999-03-23 2000-09-28 Sumitomo Pharmaceuticals Co., Ltd. Tricyclic indole-2-carboxylic acid compound used as nmda receptor antagonist
WO2000063250A1 (en) 1999-04-15 2000-10-26 Merck Frosst Canada & Co. Antibodies that recognize app cleaved by caspases and methods of use
US6121311A (en) 1999-04-28 2000-09-19 Japan Tobacco Inc. Method for treating cocainism
WO2000068197A1 (en) 1999-05-05 2000-11-16 Ortho-Mcneil Pharmaceutical, Inc. 3a,4,5,9b-TETRAHYDRO-1H-BENZ[e]INDOL-2-YL AMINE-DERIVED NEUROPEPTIDE Y RECEPTORS LIGANDS USEFUL IN THE TREATMENT OF OBESITY AND OTHER DISORDERS
WO2000071144A1 (en) 1999-05-19 2000-11-30 Domer, Inc. Prolyl endopeptidase inhibitor
WO2000072880A2 (en) 1999-05-28 2000-12-07 Neuralab Limited Prevention and treatment of amyloidogenic disease
WO2000077178A1 (en) 1999-06-16 2000-12-21 Boston Biomedical Research Institute IMMUNOLOGICAL CONTROL OF β-AMYLOID LEVELS IN VIVO
US6110949A (en) 1999-06-24 2000-08-29 Novartis Ag N-(substituted glycyl)-4-cyanothiazolidines, pharmaceutical compositions containing them and their use in inhibiting dipeptidyl peptidase-IV
US6107317A (en) 1999-06-24 2000-08-22 Novartis Ag N-(substituted glycyl)-thiazolidines, pharmaceutical compositions containing them and their use in inhibiting dipeptidyl peptidase-IV
US6172081B1 (en) 1999-06-24 2001-01-09 Novartis Ag Tetrahydroisoquinoline 3-carboxamide derivatives
WO2001000215A1 (en) 1999-06-25 2001-01-04 Wake Forest University Compositions for treating or preventing neurodegeneration and cognitive decline
WO2001010831A1 (en) 1999-08-06 2001-02-15 Grünenthal GmbH Substituted pyrrolidin-2,3,4-trion-3-oxime derivatives useful as nmda-receptor antagonists
WO2001010833A1 (en) 1999-08-06 2001-02-15 Grünenthal GmbH Substituted 1,5-dihydropyrrol-2-on derivatives as nmda receptor antagonists for the treatment of pain
WO2001012598A2 (en) 1999-08-13 2001-02-22 The Trustees Of Columbia University In The City Of New York METHODS OF INHIBITING BINDING OF β-SHEET FIBRIL TO RAGE AND CONSEQUENCES THEREOF
WO2001012176A2 (en) 1999-08-16 2001-02-22 Sanofi-Synthelabo Use of monoamine oxidase inhibitors for the manufacture of drugs intended for the treatment of obesity
WO2001014318A2 (en) 1999-08-24 2001-03-01 Probiodrug Ag New effectors of dipeptidyl peptidase iv for topical use
WO2001026656A2 (en) 1999-10-11 2001-04-19 Societe De Conseils De Recherches Et D'applications Scientifiques (S.C.R.A.S.) 5-membered heterocycle derivatives and use thereof as monoamine oxidase inhibitors
WO2001032640A1 (en) 1999-11-01 2001-05-10 Merz Pharmaceuticals Gmbh 1-cyclic amino-alkylcyclohexane compounds, pharmaceutical compositions thereof, and their use as anticonvulsants
WO2001034594A1 (en) 1999-11-12 2001-05-17 Guilford Pharmaceuticals, Inc. Dipeptidyl peptidase iv inhibitors and methods of making and using dipeptidyl peptidase iv inhibitors
WO2001040180A2 (en) 1999-11-30 2001-06-07 Ferring Bv N-substituted 2-cyanopyrrolidines and their use as antidiabetic agents
WO2001055105A1 (en) 2000-01-24 2001-08-02 Novo Nordisk A/S N-substituted 2-cyanopyroles and -pyrrolines which are inhibitors of the enzyme dpp-iv
WO2001053255A1 (en) 2000-01-24 2001-07-26 Merck Sharp & Dohme Limited Gamma-secretase inhibitors
US6756511B2 (en) 2000-01-24 2004-06-29 Merck Sharp & Dohme Limited Gamma-secretase inhibitors
WO2001057226A1 (en) 2000-02-03 2001-08-09 Millennium Pharmaceuticals, Inc. Humanized anti-ccr2 antibodies and methods of use therefor
WO2001062801A2 (en) 2000-02-24 2001-08-30 Washington University Humanized antibodies that sequester amyloid beta peptide
WO2001066564A2 (en) 2000-03-03 2001-09-13 Merck Sharp & Dohme Limited Gamma-secretase inhibitors
US7049296B2 (en) 2000-03-03 2006-05-23 Merck Sharp & Dohme Ltd. Gamma-secretase inhibitors
EP1263774A2 (en) 2000-03-03 2002-12-11 MERCK SHARP & DOHME LTD. Gamma-secretase inhibitors
WO2001066096A2 (en) 2000-03-06 2001-09-13 Immune Network Ltd. Compositions for prevention and treatment of dementia
WO2001068603A2 (en) 2000-03-10 2001-09-20 Bristol-Myers Squibb Co. Cyclopropyl-fused pyrrolidine-based inhibitors of dipeptidyl iv, processes for their preparation, and their use
US7030239B2 (en) 2000-03-23 2006-04-18 Elan Pharmaceuticals, Inc. Compounds to treat Alzheimer's disease
US6992081B2 (en) 2000-03-23 2006-01-31 Elan Pharmaceuticals, Inc. Compounds to treat Alzheimer's disease
WO2001077144A1 (en) 2000-04-07 2001-10-18 Merck Sharp & Dohme Limited Gamma-secretase inhibitors
US6984626B2 (en) 2000-04-07 2006-01-10 Merck, Sharp & Dohme Ltd. Gamma-secretase inhibitors
WO2001078728A1 (en) 2000-04-13 2001-10-25 Eisai Co., Ltd. Acetylcholinesterase inhibitors containing 1-benzyl- pyridinium salts
WO2001081337A1 (en) 2000-04-26 2001-11-01 Ferring B.V. Inhibitors of dipeptidyl peptidase iv
WO2001081304A1 (en) 2000-04-26 2001-11-01 Ferring Bv Inhibitors of dipeptidyl peptidase iv
WO2001081295A1 (en) 2000-04-26 2001-11-01 Warner-Lambert Company Cyclohexylamine derivative as subtype selective nmda receptor antagonists
WO2001085145A2 (en) 2000-05-09 2001-11-15 Pfizer Products Inc. A pharmaceutical composition and method of treatment of diseases of cognitive dysfunction in a mammal
WO2001092204A1 (en) 2000-06-01 2001-12-06 Warner-Lambert Company Cyclohexylamine derivatives as subtype selective nmda receptor antagonists
WO2001094321A1 (en) 2000-06-06 2001-12-13 Warner-Lambert Company Bicyclic cyclohexylamines and their use as nmda receptor antagonists
WO2001098289A1 (en) 2000-06-22 2001-12-27 Pharmos Corporation Novel non-psychotropic cannabinoids
WO2001098262A1 (en) 2000-06-23 2001-12-27 Merck Sharp & Dohme Limited Amidine derivatives as selective antagonists of nmda receptors
US6713276B2 (en) 2000-06-28 2004-03-30 Scios, Inc. Modulation of Aβ levels by β-secretase BACE2
US6686449B2 (en) 2000-06-30 2004-02-03 Pharmacia & Upjohn Company Mutant presenilin 1 polypeptides
US7034182B2 (en) 2000-06-30 2006-04-25 Elan Pharmaceuticals, Inc. Compounds to treat Alzheimer's disease
WO2002002560A2 (en) 2000-07-04 2002-01-10 Novo Nordisk A/S Purine-2,6-diones which are inhibitors of the enzyme dipeptidyl peptidase iv (dpp-iv)
US6448229B2 (en) 2000-07-06 2002-09-10 Merck Sharp & Dohme Ltd. Gamma secretase inhibitors
WO2002021509A1 (en) 2000-09-01 2002-03-14 Snap-On Technologies, Inc. Computer-implemented speech recognition system training
WO2002027418A2 (en) 2000-09-25 2002-04-04 Motorwiz, Inc. Model-based machine diagnostics and prognostics using theory of noise and communications
WO2002032412A2 (en) 2000-10-17 2002-04-25 Pfizer Products Inc. Combination use of acetylcholinesterase inhibitors and gabaa inverse agonists for the treatment of cognitive disorders
WO2002034718A1 (en) 2000-10-24 2002-05-02 Richter Gedeon Vegyeszeti Gyar Rt. Amide derivatives as nmda receptor antagonists
EP1334085A1 (en) 2000-11-02 2003-08-13 Merck Sharp & Dohme Ltd. Sulfamides as gamma-secretase inhibitors
WO2002036555A1 (en) 2000-11-02 2002-05-10 Merck Sharp & Dohme Limited Sulfamides as gamma-secretase inhibitors
WO2002041842A2 (en) 2000-11-03 2002-05-30 Proteotech. Inc. Antibody pti-hs7 for treatment of alzheimer's disease and other amyloidoses and parkinson's disease
WO2002038541A1 (en) 2000-11-10 2002-05-16 Taisho Pharmaceutical Co., Ltd. Cyanopyrrolidine derivatives
WO2002046237A2 (en) 2000-12-06 2002-06-13 Neuralab Limited Humanized antibodies that recognize beta amyloid peptide
WO2002046222A2 (en) 2000-12-07 2002-06-13 Mario Chojkier Compositions and methods for diagnosing alzheimer's disease
WO2002060900A2 (en) 2001-01-31 2002-08-08 Telik, Inc. Antagonists of mcp-1 function and methods of use thereof
WO2002070509A2 (en) 2001-03-01 2002-09-12 Telik, Inc. Antagonists of mcp-1 function and methods of use thereof
WO2002072542A2 (en) 2001-03-08 2002-09-19 Emory University Ph-dependent nmda receptor antagonists
US6649196B2 (en) 2001-03-12 2003-11-18 Mayo Foundation For Medical Education And Research Methods of reducing β-amyloid polypeptides
WO2002074240A2 (en) 2001-03-16 2002-09-26 Cornell Research Foundation, Inc. Anti-amyloid antibody based diagnosis and treatment of a neurological disease or disorder
WO2002081463A1 (en) 2001-04-03 2002-10-17 Telik, Inc. Antagonists of mcp-1 function and methods of use thereof
WO2002083128A1 (en) 2001-04-12 2002-10-24 Bristol-Myers Squibb Company 2,1-oxazoline and 1,2-pyrazoline-based inhibitors of dipeptidyl peptidase iv and method
WO2002088307A2 (en) 2001-04-30 2002-11-07 Eli Lilly And Company Humanized antibodies
WO2002088306A2 (en) 2001-04-30 2002-11-07 Eli Lilly And Company Humanized antibodies
EP1258476A1 (en) 2001-05-15 2002-11-20 Les Laboratoires Servier Alpha-amino acid derivatives, method for their preparation and their use as dipeptidyl-peptidase IV inhibitors (DPP IV)
WO2002094881A2 (en) 2001-05-18 2002-11-28 Krka Tovarna Zdravil, D.D., Novo Mesto Monoclonal antibody neutralising cathepsin b activity and uses thereof
WO2002096897A1 (en) 2001-05-30 2002-12-05 Neurologic, Inc. PHOSPHINYLMETHYL AND PHOSPHORYLMETHYL SUCCINIC AND GLUTARIC ACID ANALOGS AS β-SECRETASE INHIBITORS
US6562783B2 (en) 2001-05-30 2003-05-13 Neurologic, Inc. Phosphinylmethyl and phosphorylmethyl succinic and glutauric acid analogs as β-secretase inhibitors
WO2002098849A2 (en) 2001-06-01 2002-12-12 Elan Pharmaceuticals, Inc. Hydroxy alkyl amine derivatives as beta-secretase inhibitors and their use for the treatment of alzheimer’s disease and similar diseases
WO2003000180A2 (en) 2001-06-20 2003-01-03 Merck & Co., Inc. Dipeptidyl peptidase inhibitors for the treatment of diabetes
WO2003000181A2 (en) 2001-06-20 2003-01-03 Merck & Co., Inc. Dipeptidyl peptidase inhibitors for the treatment of diabetes
WO2003000250A1 (en) 2001-06-25 2003-01-03 Ferring Bv 3-fluoro-pyrrolidines as antidiabetic agents
WO2003002593A2 (en) 2001-06-27 2003-01-09 Probiodrug Ag Peptide structures useful for competitive modulation of dipeptidyl peptidase iv catalysis
WO2003002531A2 (en) 2001-06-27 2003-01-09 Smithkline Beecham Corporation Fluoropyrrolidines as dipeptidyl peptidase inhibitors
US6982264B2 (en) 2001-06-27 2006-01-03 Elan Pharmaceuticals, Inc. Substituted alcohols useful in treatment of Alzheimer's disease
WO2003002553A2 (en) 2001-06-27 2003-01-09 Smithkline Beecham Corporation Fluoropyrrolidines as dipeptidyl peptidase inhibitors
WO2003002530A2 (en) 2001-06-27 2003-01-09 Smithkline Beecham Corporation Pyrrolidines as dipeptidyl peptidase inhibitors
WO2003072556A1 (en) 2001-06-27 2003-09-04 Prosidion Ltd. Glutaminyl based dpiv inhibitors
WO2003004496A1 (en) 2001-07-03 2003-01-16 Novo Nordisk A/S Dpp-iv-inhibiting purine derivatives for the treatment of diabetes
WO2003004498A1 (en) 2001-07-06 2003-01-16 Merck & Co., Inc. Beta-amino tetrahydroimidazo (1, 2-a) pyrazines and tetrahydrotrioazolo (4, 3-a) pyrazines as dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
WO2003010159A1 (en) 2001-07-24 2003-02-06 Richter Gedeon Vegyészeti Gyár Rt. Piperidine derivatives as nmda receptor antagonists
WO2003012141A1 (en) 2001-07-31 2003-02-13 Takeda Chemical Industries, Ltd. Method of screening alzheimer’s disease-associated gene
US7122675B2 (en) 2001-08-03 2006-10-17 Schering Corporation Gamma secretase inhibitors
US6683091B2 (en) 2001-08-03 2004-01-27 Schering Corporation Gamma Secretase inhibitors
WO2003014162A1 (en) 2001-08-03 2003-02-20 Medical & Biological Laboratories Co., Ltd. ANTIBODY RECOGNIZING GM1 GANGLIOSIDE-BOUND AMYLOID β-PROTEIN AND DNA ENCODING THE ANTIBODY
WO2003013527A1 (en) 2001-08-03 2003-02-20 Schering Corporation Sulfonamide derivatives as gamma secretase inhibitors
WO2003014075A2 (en) 2001-08-03 2003-02-20 Schering Corporation Novel gamma secretase inhibitors
WO2003015691A2 (en) 2001-08-17 2003-02-27 Eli Lilly And Company RAPID IMPROVEMENT OF COGNITION IN CONDITIONS RELATED TO A$g(b)
WO2003016466A2 (en) 2001-08-17 2003-02-27 Eli Lilly And Company ANTI-Aβ ANTIBODIES
WO2003020289A1 (en) 2001-08-30 2003-03-13 Ortho-Mcneil Pharmaceutical, Inc. Treatment of dementia and memory disorders with anticonvulsants and acetylcholinesterase inhibitors
WO2003024942A1 (en) 2001-09-14 2003-03-27 Mitsubishi Pharma Corporation Thiazolidine derivative and medicinal use thereof
WO2003024965A2 (en) 2001-09-19 2003-03-27 Novo Nordisk A/S Heterocyclic compounds that are inhibitors of the enzyme dpp-iv
WO2003033524A2 (en) 2001-10-12 2003-04-24 Probiodrug Ag Peptidyl ketones as inhibitors of dpiv
WO2003035057A1 (en) 2001-10-23 2003-05-01 Ferring B.V. Inhibitors of dipeptidyl peptidase iv
WO2003039454A2 (en) 2001-10-23 2003-05-15 Oklahoma Medical Research Foundation Beta-secretase inhibitors and methods of use
WO2003035067A1 (en) 2001-10-23 2003-05-01 Ferring B.V. Novel dipeptidyl peptidase iv (dp-iv) inhibitors as anti-diabetic agents
WO2003037327A1 (en) 2001-10-26 2003-05-08 F. Hoffmann-La-Roche Ag N-substituted pyrrolidin derivatives as dipeptidyl peptidase iv inhibitors
WO2003037376A1 (en) 2001-11-02 2003-05-08 Kensuke Egashira Preventives and/or remedies for post-transplant arteriosclerosis as rejection of organ transplant
WO2003039467A2 (en) 2001-11-02 2003-05-15 Diagenics International Corporation Monoclonal antibodies specific for beta-amyloid.
WO2003040183A2 (en) 2001-11-09 2003-05-15 The Genetics Company, Inc Compounds for the diagnosis/prevention/treatment of alzheimer's disease
WO2003040174A2 (en) 2001-11-09 2003-05-15 Probiodrug Ag Substituted amino ketone compounds
WO2003043987A2 (en) 2001-11-19 2003-05-30 Elan Pharmaceuticals, Inc. (4-phenyl) piperidin-3-yl-phenylcarboxylate derivatives and related compounds as beta-secretase inhibitors for the treatment of alzheimer's disease
WO2003045128A2 (en) 2001-11-21 2003-06-05 New York University SYNTHETIC IMMUNOGENIC BUT NON-DEPOSIT-FORMING POLYPEPTIDES AND PEPTIDES HOMOLOGOUS TO AMYLOID β, PRION PROTEIN, AMYLIN, α-SYNUCLEIN, OR POLYGLUTAMINE REPEATS FOR INDUCTION OF AN IMMUNE RESPONSE THERETO
WO2003045977A2 (en) 2001-11-26 2003-06-05 Trustees Of Tufts College Peptidomimetic Inhibitors of Post-Proline Cleaving Enzymes
WO2003048204A1 (en) 2001-12-06 2003-06-12 Takeda Chemical Industries, Ltd. Alzheimer’s disease-associated gene and protein and use thereof
WO2003051374A2 (en) 2001-12-17 2003-06-26 New York State Office Of Mental Health SEQUESTRATION OF Aβ IN THE PERIPHERY IN THE ABSENCE OF IMMUNOMODULATING AGENT AS A THERAPEUTIC APPROACH FOR THE TREATMENT OR PREVENTION OF BETA-AMYLOID RELATED DISEASES
WO2003053368A2 (en) 2001-12-19 2003-07-03 Atherogenics, Inc. Chalcone derivatives and their use to treat diseases
WO2003055514A1 (en) 2001-12-21 2003-07-10 Antigenics Inc. Compositions comprising immunoreactive reagents and saponins, and methods of use thereof
WO2003057666A2 (en) 2001-12-26 2003-07-17 Guilford Pharmaceuticals Inhibitors of dipeptidyl peptidase iv
WO2003057144A2 (en) 2001-12-26 2003-07-17 Guilford Pharmaceuticals Change inhibitors of dipeptidyl peptidase iv
WO2003055881A1 (en) 2001-12-27 2003-07-10 F. Hoffmann-La Roche Ag Pyrido(2,1-a)isoquinoline derivatives as dpp-iv inhibitors
WO2003057204A2 (en) 2002-01-08 2003-07-17 Nordic Bioscience A/S Modulation of iamt (pimt or pcmt) in immune system
WO2003059346A1 (en) 2002-01-18 2003-07-24 The Genetics Company Inc. Beta-secretase inhibitors
WO2003063760A2 (en) 2002-01-31 2003-08-07 Tel Aviv University Future Technology Development L.P. Peptides antibodies directed thereagainst and methods using same for diagnosing and treating amyloid-associated diseases
WO2003066592A1 (en) 2002-02-06 2003-08-14 Schering Corporation Gamma secretase inhibitors
WO2003068757A1 (en) 2002-02-13 2003-08-21 F. Hoffmann-La Roche Ag Novel pyridin- and pyrimidin-derivatives
WO2003068748A1 (en) 2002-02-13 2003-08-21 F. Hoffmann-La Roche Ag Novel pyridine- and quinoline-derivatives
WO2003070760A2 (en) 2002-02-20 2003-08-28 F. Hoffmann-La Roche Ag Anti-amyloid beta antibodies and their use
WO2003074081A1 (en) 2002-02-28 2003-09-12 Mindset Biopharmaceuticals Usa Inc. SPECIFIC ANTIBODIES TO AMYLOID β PEPTIDE, PHARMACEUTICAL COMPOSITIONS AND METHODS OF USE THEREOF
WO2003074500A2 (en) 2002-03-06 2003-09-12 Sanofi-Aventis N-aminoacetyl-pyrrolidine-2-carbonitriles and their use as ddp-iv inhibitors
WO2003077858A2 (en) 2002-03-12 2003-09-25 Neuralab Limited Humanized antibodies that recognize beta amyloid peptide
WO2003082817A2 (en) 2002-03-25 2003-10-09 Merck & Co., Inc. Beta-amino heterocyclic dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
WO2003082820A1 (en) 2002-03-29 2003-10-09 Eisai Co., Ltd. (1-indanone)-(1,2,3,6-tetrahydropyridine) derivative
WO2003086310A2 (en) 2002-04-12 2003-10-23 Ramot At Tel Aviv University Ltd. Prevention of brain inflammation as a result of induced autoimmune response
WO2003089460A1 (en) 2002-04-19 2003-10-30 The Governing Council Of The University Of Toronto Immunological methods and compositions for the treatment of alzheimer's disease
WO2003091278A1 (en) 2002-04-24 2003-11-06 Hiroshi Mori Gamma-secretase inhibitors
WO2003091220A1 (en) 2002-04-26 2003-11-06 Schering Corporation Muscarinic antagonists
WO2003099202A2 (en) 2002-05-17 2003-12-04 Merck & Co., Inc. Beta-secretase inhibitors
WO2003101458A1 (en) 2002-05-31 2003-12-11 H. Lundbeck A/S A combination of an nmda-antagonist and acetylcholine esterase inhibitors for the treatment of alzheimer's disease
WO2003101958A2 (en) 2002-06-04 2003-12-11 Pfizer Products Inc. Flourinated cyclic amides as dipeptidyl peptidase iv inhibitors
WO2003101449A2 (en) 2002-06-04 2003-12-11 Pfizer Products Inc. Process for the preparation of 3,3,4,4-tetrafluoropyrrolidine and derivatives thereof
WO2003104229A1 (en) 2002-06-06 2003-12-18 エーザイ株式会社 Novel fused imidazole derivative
WO2003104437A2 (en) 2002-06-11 2003-12-18 Northwestern University Anti-addl antibodies and uses thereof
WO2004000958A1 (en) 2002-06-19 2003-12-31 Surface Specialties, S.A. Semi-gloss powder coating compositions
WO2004007446A1 (en) 2002-07-10 2004-01-22 Yamanouchi Pharmaceutical Co., Ltd. Novel azetidine derivative or salt thereof
WO2004007468A1 (en) 2002-07-15 2004-01-22 Merck & Co., Inc. Piperidino pyrimidine dipeptidyl peptidase inhibitors for the treatment of diabetes
WO2004009062A2 (en) 2002-07-19 2004-01-29 Khalid Iqbal NMDA RECEPTOR ANTAGONISTS AND THEIR USE IN INHIBITING ABNORMAL HYPERPHOSPHORYLATION OF MICROTUBULE ASSOCIATED PROTEIN tau
WO2004013098A1 (en) 2002-08-05 2004-02-12 Bristol-Myers Squibb Company Novel gamma-lactams as beta-secretase inhibitors
WO2004018468A2 (en) 2002-08-21 2004-03-04 Boehringer Ingelheim Pharma Gmbh & Co. Kg 8-[3-amino-piperidin-1-yl]-xanthines, the production thereof and the use of the same as medicaments
WO2004018467A2 (en) 2002-08-22 2004-03-04 Boehringer Ingelheim Pharma Gmbh & Co. Kg Phenacyl xanthine derivatives as dpp-iv inhibitor
WO2004018469A1 (en) 2002-08-22 2004-03-04 Boehringer Ingelheim Pharma Gmbh & Co. Kg Novel purine derivatives, production and use thereof as medicaments
WO2004024921A1 (en) 2002-09-12 2004-03-25 Juridical Foundation The Chemo-Sero-Therapeutic Research Institute Human antihuman mcp-1 antibody and antibody fragment thereof
WO2004024090A2 (en) 2002-09-12 2004-03-25 The Regents Of The University Of California Immunogens and corresponding antibodies specific for high molecular weight aggregation intermediates common to amyloids formed from proteins of differing sequence
WO2004024770A1 (en) 2002-09-12 2004-03-25 Universidad De Zaragoza Polyclonal antibodies, preparation method thereof and use of same
WO2004026822A2 (en) 2002-09-19 2004-04-01 Abbott Laboratories Pharmaceutical compositions as inhibitors of dipeptidyl peptidase-iv (dpp-iv)
WO2004026851A1 (en) 2002-09-20 2004-04-01 Axys Pharmaceuticals, Inc. 3-(3,5-disubstituted-4-hydroxyphenyl)propionamide derivatives as cathepsin b inhibitors
WO2004028522A1 (en) 2002-09-25 2004-04-08 Innovative Drug Delivery Systems, Inc. Analgesic compositions comprising nmda receptor antagonists and benzalkonium chloride
WO2004029629A1 (en) 2002-09-27 2004-04-08 Janssen Pharmaceutica N.V. N-11 truncated amyloid-beta nomoclonal antibodies, compositions, methods and uses
WO2004029630A1 (en) 2002-09-27 2004-04-08 Janssen Pharmaceutica N.V. N-11 truncated amyloid-beta monoclonal antibodies, compositions, methods and uses
WO2004031400A2 (en) 2002-10-01 2004-04-15 Northwestern University Amyloid beta-derived diffusible ligands (addls), addl-surrogates, addl-binding molecules, and uses thereof
US7101895B2 (en) 2002-10-04 2006-09-05 Merck Sharp & Dohme Limited Cyclohexyl sulphone derivatives as gamma-secretase inhibitors
US6890956B2 (en) 2002-10-04 2005-05-10 Merck Sharp & Dohme Limited Cyclohexyl sulphones as gamma-secretase inhibitors
WO2004031139A1 (en) 2002-10-04 2004-04-15 Merck Sharp & Dohme Limited Cyclohexyl sulphones as gamma-secretase inhibitors
WO2004031137A1 (en) 2002-10-04 2004-04-15 Merck Sharp & Dohme Limited Cyclohexyl sulphone derivatives as gamma-secretase inhibitors
WO2004032836A2 (en) 2002-10-07 2004-04-22 Merck & Co., Inc. Antidiabetic beta-amino heterocylcic dipeptidyl peptidase inhibitors
WO2004033455A2 (en) 2002-10-08 2004-04-22 Novo Nordisk A/S Hemisuccinate salts of heterocyclic dpp-iv inhibitors
WO2004032929A2 (en) 2002-10-09 2004-04-22 Neuropharma, S.A. Dual binding site acetylcholinesterase inhibitors for the treatment of alzheimer’s disease
WO2004032868A2 (en) 2002-10-09 2004-04-22 Rinat Neuroscience Corp. Methods of treating alzheimer's disease using antibodies directed against amyloid beta peptide and compositions thereof
WO2004037169A2 (en) 2002-10-18 2004-05-06 Merck & Co., Inc. Beta-amino heterocyclic dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
WO2004037234A2 (en) 2002-10-24 2004-05-06 Merz Pharma Gmbh & Co. Kgaa Combination therapy using 1-aminocyclohexane derivatives and acetylcholinesterase inhibitors
WO2004039371A2 (en) 2002-10-29 2004-05-13 Pharmacia Corporation Compositions of cyclooxygenase-2 selective inhibitors and nmda receptor antagonists for the treatment or prevention of neuropathic pain
WO2004041795A1 (en) 2002-10-30 2004-05-21 Guilford Pharmaceuticals Inc. Novel inhibitors of dipeptidyl peptidase iv
WO2004039773A2 (en) 2002-10-30 2004-05-13 Nordic Bioscience A/S Coumpounds modulating the activity of gapdh and/or iamt
WO2004039370A1 (en) 2002-11-01 2004-05-13 Merck Sharp & Dohme Limited Sulfonamides, sulfamates and sulfamides as gamma-secretase inhibitors
WO2004044204A2 (en) 2002-11-06 2004-05-27 Institut Pasteur Variable fragments of single-chain camelide antibodies and uses thereof for diagnosing and treating various pathologies
WO2004043940A1 (en) 2002-11-07 2004-05-27 Merck & Co., Inc. Phenylalanine derivatives as dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
WO2004043916A1 (en) 2002-11-12 2004-05-27 Merck & Co., Inc. Phenylcarboxamide beta-secretase inhibitors for the treatment of alzheimer's disease
US7109217B2 (en) 2002-11-12 2006-09-19 Merck & Co., Inc. Phenylcarboxamide beta-secretase inhibitors for the treatment of Alzheimer's disease
WO2004048352A2 (en) 2002-11-27 2004-06-10 Fujisawa Pharmaceutical Co., Ltd. 2-cyanopyrrolidines and their analogues as dpp-iv inhibitors
WO2004050022A2 (en) 2002-12-04 2004-06-17 Merck & Co., Inc. Phenylalanine derivatives as dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
WO2004052850A2 (en) 2002-12-09 2004-06-24 Bristol-Myers Squibb Company Methods and compounds producing dipeptidyl peptidase iv inhibitors and intermediates thereof
WO2004056727A2 (en) 2002-12-19 2004-07-08 Atherogenics, Inc. Process of making chalcone derivatives
WO2004058266A1 (en) 2002-12-20 2004-07-15 Merck & Co., Inc. 3-amino-4-phenylbutanoic acid derivatives as dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
WO2004062625A2 (en) 2003-01-07 2004-07-29 Merck & Co., Inc. Macrocyclic beta-secretase inhibitors for treatment of alzheimer's disease
WO2004064778A2 (en) 2003-01-17 2004-08-05 Merck & Co. Inc. 3-amino-4-phenylbutanoic acid derivatives as dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
WO2004067561A1 (en) 2003-01-31 2004-08-12 Abbott Gmbh & Co. Kg Amyloid-$g(b)(1-42) oligomers, derivatives thereof, antibodies for the same, method for production and use therof
WO2004069162A2 (en) 2003-01-31 2004-08-19 Merck & Co., Inc. 3-amino-4-phenylbutanoic acid derivatives as dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
WO2004069182A2 (en) 2003-02-01 2004-08-19 Neuralab Limited Active immunization to generate antibodies to soluble a-beta
WO2004069826A1 (en) 2003-02-04 2004-08-19 F. Hoffmann-La Roche Ag Malonamide derivatives as gamma-secretase inhibitors
WO2004071408A2 (en) 2003-02-10 2004-08-26 Applied Molecular Evolution, Inc. Aβ BINDING MOLECULES
WO2004071454A2 (en) 2003-02-13 2004-08-26 Guilford Pharmaceuticals Inc. Substituted azetidine compounds as inhibitors of dipeptidyl peptidase iv
WO2004073630A2 (en) 2003-02-18 2004-09-02 Roskamp Research Llc Anti-angiogenic and anti-tumoral properties of beta and gamma secretase inhibitors
WO2004076433A1 (en) 2003-02-28 2004-09-10 Aic Dipeptidyl peptidase inhibitors
WO2004076434A1 (en) 2003-02-28 2004-09-10 Aic Dipeptidyl peptidase inhibitors
WO2004078908A2 (en) 2003-03-06 2004-09-16 Santhera Pharmaceuticals (Schweiz) Gmbh Alpha-keto carbonyl calpain inhibitors
WO2004080419A2 (en) 2003-03-12 2004-09-23 Neuralab Limited Humanized antibodies that recognize beta amyloid peptide
WO2004084830A2 (en) 2003-03-21 2004-10-07 Buck Institute Method for treating alzheimer’s dementia
WO2004084884A1 (en) 2003-03-24 2004-10-07 The Chinese University Of Hong Kong Use of tanshinone derivates as cholinesterase inhibitors in treating related diseases
WO2004087053A2 (en) 2003-03-25 2004-10-14 Syrrx, Inc. Dipeptidyl peptidase inhibitors
WO2005028511A2 (en) 2003-03-28 2005-03-31 Centocor, Inc. Anti-amyloid antibodies, compositions, methods and uses
WO2004087158A2 (en) 2003-03-28 2004-10-14 Acadia Pharmaceuticals Inc. Muscarinic m1 receptor agonists for pain management
WO2004092189A1 (en) 2003-04-09 2004-10-28 Wyeth Derivatives of 2-(8,9-dioxo-2,6-diazabicyclo(5.2.0)non-1(7)-en-2-yl)alkyl phosphonic acid and their use as n-methyl-d-aspartate (nmda) recetor antagonists
WO2004089911A1 (en) 2003-04-10 2004-10-21 Merck Sharp & Dohme Limited Pyrazole derivatives as gamma-secretase inhibitors useful in the treatment of alzheimer’s disease
WO2004089351A2 (en) 2003-04-10 2004-10-21 Cambridge University Technical Services Ltd Treatment of progressive neurological disorders comprising a mao inhibitor (miclobemid) and an acetylcholinesterase inhibitor (tacrin)
WO2004089362A1 (en) 2003-04-11 2004-10-21 Novo Nordisk A/S 2-cyanopyrroles and their analogues as ddp-iv inhibitors
WO2004098591A2 (en) 2003-05-05 2004-11-18 Probiodrug Ag Inhibitors of glutaminyl cyclase and their use in the treatment of neurological diseases
WO2004098625A2 (en) 2003-05-05 2004-11-18 Probiodrug Ag Medical use of inhibitors of glutaminyl and glutamate cyclases
WO2004099134A2 (en) 2003-05-05 2004-11-18 Prosidion Ltd. Glutaminyl based dp iv-inhibitors
WO2004098631A1 (en) 2003-05-08 2004-11-18 Universidad De Zaragoza Alzheimer's disease treatment method
WO2004099185A1 (en) 2003-05-09 2004-11-18 Fujisawa Pharmaceutical Co. Ltd. 2-cyanopyrrolidine derivatives and their use as dpp-iv inhibitors
WO2004101562A2 (en) 2003-05-13 2004-11-25 Schering Corporation Bridged n-arylsulfonylpiperidines as gamma-secretase inhibitors
WO2004103993A1 (en) 2003-05-14 2004-12-02 Syrrx, Inc. Dipeptidyl peptidase inhibitors
WO2004103276A2 (en) 2003-05-14 2004-12-02 Merck & Co., Inc. 3-amino-4-phenylbutanoic acid derivatives as dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
WO2004101538A1 (en) 2003-05-16 2004-11-25 Merck Sharp & Dohme Limited Cyclohexyl sulphones as gamma-secretase inhibitors
WO2005000216A2 (en) 2003-05-27 2005-01-06 Forest Laboratories, Inc. Combination of an nmda receptor antagonist and a selective serotonin reuptake inhibitor for the treatment of depression and other mood disorders
WO2004108895A2 (en) 2003-05-30 2004-12-16 Neuralab Limited Humanized antibodies that recognize beta amyloid peptide
WO2004108730A1 (en) 2003-06-05 2004-12-16 Fujisawa Pharmaceutical Co., Ltd. 1h-imidazo`4,5-d!pyridazines as dpp-iv inhibitors for the treatment of niddm
WO2004110436A1 (en) 2003-06-06 2004-12-23 Merck & Co., Inc. Fused indoles as dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
WO2004111041A1 (en) 2003-06-12 2004-12-23 Fujisawa Pharmaceutical Co., Ltd. Pyrrolidine, thiazolidine and oxazolidine compounds which inhibit dipeptidyl peptidase-iv (dpp)
WO2004112701A2 (en) 2003-06-17 2004-12-29 Merck & Co., Inc. Cyclohexylglycine derivatives as dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
WO2005000848A1 (en) 2003-06-20 2005-01-06 F. Hoffmann-La Roche Ag Pyrido` 2, 1-a - isoquinoline derivatives as dpp-iv inhibitors
WO2005000846A1 (en) 2003-06-20 2005-01-06 F.Hoffmann-La Roche Ag Hexahydropyridoisoqinolines as dpp-iv inhibitors
WO2005000193A2 (en) 2003-06-30 2005-01-06 Tel Aviv University Future Technology Development L.P. Peptides antibodies directed thereagainst and methods using same for diagnosing and treating amyloid-associated diseases
WO2005004802A2 (en) 2003-06-30 2005-01-20 Merck & Co., Inc. N-alkyl phenylcarboxamide beta-secretase inhibitors for the treatment of alzheimer's disease
WO2005004803A2 (en) 2003-07-01 2005-01-20 Merck & Co., Inc. Phenylcarboxylate beta-secretase inhibitors for the treatment of alzheimer's disease
WO2005007614A1 (en) 2003-07-03 2005-01-27 The Government Of The United States Of America As Represented By The Secretary, Department Of Health And Human Services 6011 Monoamine oxidase inhibitors
WO2005007199A1 (en) 2003-07-16 2005-01-27 Rvx Therapeutics, Inc. COMPOUNDS AND METHODS FOR DOWNREGULATING THE EFFECTS OF TGF-β
WO2005008250A1 (en) 2003-07-21 2005-01-27 Angiogenetics Sweden Ab Compounds and methods for promoting angiogenesis by using a gamma-secretase inhibitor or inhibiting the gamma-secretase pathway
WO2005009421A2 (en) 2003-07-28 2005-02-03 Merz Pharma Gmbh & Co. Kgaa The use of 1-amino-alkylcyclohexane compounds in the treatment of pain hypersensitivity
WO2005011581A2 (en) 2003-07-31 2005-02-10 Merck & Co., Inc. Hexahydrodiazepinones as dipeptidyl peptidase-iv inhibitors for the treatment or prevention of diabetes
WO2005011599A2 (en) 2003-08-01 2005-02-10 Northwestern University Antibodies specific for toxic amyloid beta protein oligomers
WO2005014553A1 (en) 2003-08-05 2005-02-17 Merck Sharp & Dohme Limited Novel gamma-secretase inhibitors
WO2005016911A1 (en) 2003-08-13 2005-02-24 Takeda Pharmaceutical Company Limited 4-pyrimidone derivatives and their use as peptidyl peptidase inhibitors
WO2005018545A2 (en) 2003-08-14 2005-03-03 Merck & Co., Inc. Macrocyclic beta-secretase inhibitors for the treatment of alzheimer's disease
WO2005018424A2 (en) 2003-08-18 2005-03-03 Research Foundation For Mental Hygiene, Inc. Antibodies specific for fibrillar amyloid and a procedure to detect fibrillar amyloid deposits
WO2005023762A1 (en) 2003-09-04 2005-03-17 Abbott Laboratories Pyrrolidine-2-carbonitrile derivatives and their use as inhibitors of dipeptidyl peptidase-iv (dpp-iv)
WO2005023858A1 (en) 2003-09-05 2005-03-17 Cellzome Ag Protein complexes associated with app-processing
WO2005030751A2 (en) 2003-09-08 2005-04-07 Takeda Pharmaceutical Company Limited Dipeptidyl peptidase inhibitors
WO2005026148A1 (en) 2003-09-08 2005-03-24 Takeda San Diego, Inc. Dipeptidyl peptidase inhibitors
WO2005025616A1 (en) 2003-09-09 2005-03-24 Takeda Pharmaceutical Company Limited Use of antibody
WO2005025554A2 (en) 2003-09-09 2005-03-24 Japan Tobacco Inc. Dipeptidyl peptidase iv inhibitor
WO2005025516A2 (en) 2003-09-12 2005-03-24 The Regents Of The University Of California Monoclonal antibodies specific for conformational epitopes of prefibrillar aggregates
WO2005028440A1 (en) 2003-09-16 2005-03-31 Schering Corporation Novel gamma secretase inhibitors
WO2005027975A1 (en) 2003-09-22 2005-03-31 Pfizer Limited Combinations of alpha-2-delta ligands and acetylcholinesterase inhibitors
WO2005030731A1 (en) 2003-09-24 2005-04-07 Merck Sharp & Dohme Limited Gamma-secretase inhibitors
WO2005032471A2 (en) 2003-10-03 2005-04-14 Merck & Co., Inc. Benzylether and benzylamino beta-secretase inhibitors for the treatment of alzheimer's disease
WO2005040126A1 (en) 2003-10-06 2005-05-06 F. Hoffman-La Roche Ag Substituted dibenzo-azepine and benzo-diazepine derivatives useful as gamma-secretase inhibitors
WO2005033106A1 (en) 2003-10-06 2005-04-14 Alangudi Sankaranarayanan Azolidinecarbonitriles and their use as dpp-iv inhibitors
WO2005035522A1 (en) 2003-10-08 2005-04-21 Pfizer Japan, Inc. 1-‘2-(4-hydroxyphenyl)-2-hydroxyethyl!-piperidin-4-ol compounds as nmda receptor antagonists
WO2005039548A2 (en) 2003-10-15 2005-05-06 Probiodrug Ag Use of effectors of glutaminyl and glutamate cyclases
WO2005040095A1 (en) 2003-10-16 2005-05-06 Astrazeneca Ab Inhibitors of dipeptidyl peptidase iv
WO2005039580A1 (en) 2003-10-16 2005-05-06 Boehringer Ingelheim International Gmbh Pharmaceutical composition comprising a monoamine neurotransmitter re-uptake inhibitor and an acetylcholinesterase inhibitor
WO2005037828A1 (en) 2003-10-20 2005-04-28 Lg Life Sciences Ltd. Novel inhibitors of dpp-iv, methods of preparing the same, and pharmaceutical compositions containing the same as an active agent
WO2005079779A1 (en) 2003-10-22 2005-09-01 Merz Pharma Gmbh & Co. Kgaa THE USE OF 1-AMINOCYCLOHEXANE DERIVATIVES TO MODIFY DEPOSITION OF FIBRILLOGENIC Aß PEPTIDES IN AMYLOIDOPATHIES
WO2006058720A2 (en) 2003-11-03 2006-06-08 Probiodrug Ag Novel compounds for the treatment of neurological disorders
WO2005044195A2 (en) 2003-11-04 2005-05-19 Merck & Co., Inc. Fused phenylalanine derivatives as dipeptidyl peptidase-iv inhibitors for the treatment or prevention of diabetes
WO2005044830A1 (en) 2003-11-11 2005-05-19 F. Hoffmann-La Roche Ag Phosphinic acids derivatives, beta-secretase inhibitors for the treatment of alzheimer’s disease
WO2005047297A1 (en) 2003-11-12 2005-05-26 Phenomix Corporation Heterocyclic boronic acid compounds
WO2005051914A1 (en) 2003-11-24 2005-06-09 Merck & Co., Inc. Benzylether and benzylamino beta-secretase inhibitors for the treatment of alzheimer's disease
WO2005051950A1 (en) 2003-11-27 2005-06-09 Boehringer Ingelheim International Gmbh Novel 8-(piperazine-1-yl)- and 8-([1,4]diazepan-1-yl)-xanthine, the production and use thereof in the from of a drug
WO2005055996A1 (en) 2003-12-05 2005-06-23 Forest Laboratories, Inc. Memantine for the prevention or reduction of suicidality and for treatment of major depression associated with suicidality
WO2005056003A1 (en) 2003-12-09 2005-06-23 Santhera Pharmaceuticals (Schweiz) Gmbh Dpp-iv inhibitors
WO2005056013A1 (en) 2003-12-09 2005-06-23 Santhera Pharmaceuticals (Schweiz) Gmbh Dpp-iv inhibitors
WO2005058849A1 (en) 2003-12-15 2005-06-30 Glenmark Pharmaceuticals Ltd. New dipeptidyl peptidase in inhibitors; process for their preparation and compositions containing them
WO2005065195A2 (en) 2003-12-19 2005-07-21 Merck & Co., Inc. Phenylamide and pyridylamide beta-secretase inhibitors for the treatment of alzheimer's disease
WO2005070429A1 (en) 2004-01-22 2005-08-04 Neurosearch A/S Pharmaceutical composition comprising a monoamine neurotransmitter re-uptake inhibitor and an n-methyl-d-aspartate (nmda) receptors antagonist
WO2005072705A1 (en) 2004-01-29 2005-08-11 Neuromolecular, Inc. Combination of a nmda receptor antagonist and a mao-inhibitor or a gadpf-inhibitor for the treatment of central nervous system-related conditions
WO2005075426A1 (en) 2004-02-03 2005-08-18 Glenmark Pharmaceuticals Ltd. Novel dipeptidyl peptidase iv inhibitors; processes for their preparation and compositions thereof
WO2005075436A2 (en) 2004-02-05 2005-08-18 Probiodrug Ag Novel inhibitors of glutaminyl cyclase
WO2005079756A2 (en) 2004-02-13 2005-09-01 Neuromolecular, Inc. Combination of a nmda receptor antagonist and an anti-depressive drug mao-inhibitor or a gadph-inhibitor for the treatment of psychiatric conditions
WO2005079789A1 (en) 2004-02-17 2005-09-01 Axonyx, Inc. Combinatorial therapy with an acetylcholinesterase inhibitor and (3ar)-1,3a,8-trimethyl-1,2,3,3a,8,8a-hexahydropyrrolo[2,3-b]indol-5-yl phenylcarbamate
WO2005085246A1 (en) 2004-02-18 2005-09-15 Boehringer Ingelheim International Gmbh 8-[3-amino-piperidin-1-yl]-xanthine, the production thereof and the use in the form of a dpp inhibitor
WO2005080435A1 (en) 2004-02-20 2005-09-01 Immuno-Biological Laboratories Co., Ltd. Monoclonal antibody and use thereof
WO2005081872A2 (en) 2004-02-20 2005-09-09 Ac Immune Sa Methods and compositions comprising supramolecular constructs
WO2005082348A2 (en) 2004-02-23 2005-09-09 Trustees Of Tufts College Inhibitors of dipeptidylpeptidase iv for regulating glucose metabolism
WO2005095343A1 (en) 2004-03-05 2005-10-13 Santhera Pharmaceuticals (Schweiz) Gmbh Dpp-iv inhibitors
WO2005087235A1 (en) 2004-03-09 2005-09-22 National Health Research Institutes Pyrrolidine compounds
WO2005095381A1 (en) 2004-03-15 2005-10-13 Takeda Pharmaceutical Company Limited Dipeptidyl peptidase inhibitors
WO2005092009A2 (en) 2004-03-19 2005-10-06 Axonyx, Inc. Acetylcholinesterase inhibitors and n-methyl-d-aspartate antagonists useful in the treatment of cognitive disorders
WO2005095339A1 (en) 2004-03-31 2005-10-13 Pfizer Products Inc. Dicyanopyrrolidines as dipeptidyl peptidase iv inhibitors
WO2005097103A2 (en) 2004-04-01 2005-10-20 Axys Pharmaceuticals, Inc. Diabetes and metabolic syndrome therapy utilizing cathepsin b inhibitors
WO2005097768A2 (en) 2004-04-05 2005-10-20 Schering Corporation Novel gamma secretase inhibitors
WO2005103020A1 (en) 2004-04-20 2005-11-03 Merck & Co., Inc. 1,3,5-substituted phenyl derivative compounds useful as beta-secretase inhibitors for the treatment of alzheimer's disease
WO2005103043A1 (en) 2004-04-20 2005-11-03 Merck & Co., Inc. 2, 4, 6-substituted pyridyl derivative compounds useful as beta-secretase inhibitors for the treatment of alzheimer’s disease
WO2005102390A2 (en) 2004-04-22 2005-11-03 Pfizer Japan, Inc. Combinations comprising alpha-2-delta ligands and nmda receptor antagonists
WO2005105133A2 (en) 2004-04-23 2005-11-10 Massachusetts Eye And Ear Infirmary Methods and compositions for preserving the viability of photoreceptor cells
WO2005105998A1 (en) 2004-04-27 2005-11-10 Juridical Foundation The Chemo-Sero-Therapeutic Research Institute HUMAN ANTIAMYLOID β PEPTIDE ANTIBODY AND ANTIBODY FRAGMENT THEREOF
WO2005108382A1 (en) 2004-05-04 2005-11-17 Merck & Co., Inc. 1,2,4-oxadiazole derivatives as dipeptidyl peptidase-iv inhibitors for the treatment or prevention of diabetes
WO2005116014A1 (en) 2004-05-12 2005-12-08 Pfizer Products Inc. Proline derivatives and their use as dipeptidyl peptidase iv inhibitors
WO2005113484A1 (en) 2004-05-13 2005-12-01 Merck & Co., Inc. Phenyl carboxamide compounds useful as beta-secretase inhibitors for the treatment of alzheimer's disease
WO2005116029A1 (en) 2004-05-18 2005-12-08 Merck & Co., Inc. Cyclohexylalanine derivatives as dipeptidyl peptidase-iv inhibitors for the treatment or prevention of diabetes
WO2005113510A1 (en) 2004-05-21 2005-12-01 Santhera Pharmaceuticals (Schweiz) Ag Dpp-iv inhibitors
WO2005118555A1 (en) 2004-06-04 2005-12-15 Takeda Pharmaceutical Company Limited Dipeptidyl peptidase inhibitors
WO2005120571A2 (en) 2004-06-07 2005-12-22 Ramot At Tel Aviv University Ltd. Method of passive immunization against disease or disorder characterized by amyloid aggregation with diminished risk of neuroinflammation
WO2005121089A1 (en) 2004-06-08 2005-12-22 Santhera Pharmaceuticals (Schweiz) Ag Dpp-iv inhibitors
WO2005120494A1 (en) 2004-06-08 2005-12-22 Santhera Pharmaceuticals (Schweiz) Ag 1-`(3r)-amino-4-(2-fluoro-phenyl)-butyl !-pyrrolidine-(2r)-carboxilic acid-benzyl amine derivatives and related compounds as dipeptidyl-peptidase iv (dpp-iv) inhibitors for the treatment of type 2 diabetes mellitus
WO2005121131A1 (en) 2004-06-08 2005-12-22 Santhera Pharmaceuticals (Schweiz) Ag Dpp-iv inhibitors
WO2006002004A1 (en) 2004-06-15 2006-01-05 Merck & Co., Inc. Pyrrolidin-3-yl compounds useful as beta-secretase inhibitors for the treatment of alzheimer's disease
WO2005123685A1 (en) 2004-06-16 2005-12-29 Astrazeneca Ab Tetrahydroquinolones and aza-analogues thereof for use as dpp-iv inhibitors in the treatement of diabetes
WO2006009886A1 (en) 2004-06-21 2006-01-26 Merck & Co., Inc. Aminocyclohexanes as dipeptidyl peptidase-iv inhibitors for the treatment or prevention of diabetes
WO2005123775A1 (en) 2004-06-21 2005-12-29 Bioarctic Neuroscience Ab Antibodies specific for soluble amyloid beta peptide protofibrils and uses thereof
WO2006085961A2 (en) 2004-06-30 2006-08-17 Centocor, Inc. Anti-mcp-1 antibodies, compositions, methods and uses
WO2006004880A2 (en) 2004-06-30 2006-01-12 Schering Corporation Substituted n-arylsulfonylheterocyclic amines as gamma-secretase inhibitors
WO2006014478A1 (en) 2004-07-02 2006-02-09 Northwestern University MONOLOCAL ANTIBODIES THAT TARGET PATHOLOGICAL ASSEMBLIES OF AMYLOID β (ABETA)
WO2006017292A1 (en) 2004-07-12 2006-02-16 Phenomix Corporation Constrained cyano compounds
WO2006020017A2 (en) 2004-07-16 2006-02-23 Takeda Pharmaceutical Company Limited Dipeptidyl peptidase inhibitors
WO2006019965A2 (en) 2004-07-16 2006-02-23 Takeda San Diego, Inc. Dipeptidyl peptidase inhibitors
WO2006014638A2 (en) 2004-07-19 2006-02-09 The General Hospital Corporation ANTIBODIES TO CROSS-LINKED AMYLOID β OLIGOMERS
WO2006008661A2 (en) 2004-07-19 2006-01-26 Neurochem (International) Limited Diagnostic methods of multiple organ amyloidosis
WO2006014762A1 (en) 2004-07-22 2006-02-09 Schering Corporation Substituted amide beta secretase inhibitors
WO2006014944A1 (en) 2004-07-28 2006-02-09 Schering Corporation Macrocyclic beta-secretase inhibitors
WO2006010965A1 (en) 2004-07-29 2006-02-02 Richter Gedeon Vegyészeti Gyár Rt. Indole-2 -carboxamidine derivatives as nmda receptor antagonists
WO2006036291A2 (en) 2004-07-30 2006-04-06 Rinat Neuroscience Corp. Antibodies directed against amyloid-beta peptide and methods using same
WO2006013104A1 (en) 2004-08-05 2006-02-09 Santhera Pharmaceuticals (Schweiz) Ag Heterocyclic compounds useful as dpp- iv inhibitors
WO2006016644A1 (en) 2004-08-11 2006-02-16 Mitsubishi Chemical Corporation Antibody and utilization of the same
WO2006023750A2 (en) 2004-08-23 2006-03-02 Merck & Co., Inc. Fused triazole derivatives as dipeptidyl peptidase-iv inhibitors for the treatment or prevention of diabetes
WO2006021413A1 (en) 2004-08-25 2006-03-02 Santhera Pharmaceuticals (Schweiz) Ag Alpha-keto carbonyl calpain inhibitors
WO2006021409A1 (en) 2004-08-25 2006-03-02 Santhera Pharmaceuticals (Schweiz) Ag Alpha-keto carbonyl calpain inhibitors
WO2006026204A2 (en) 2004-08-26 2006-03-09 Bristol-Myers Squibb Company Novel gamma-lactams as beta-secretase inhibitors
WO2006026408A2 (en) 2004-08-27 2006-03-09 Wyeth Research Ireland Limited Production of anti-amyloid beta antibodies
WO2006029850A1 (en) 2004-09-14 2006-03-23 The Genetics Company, Inc. Hydrazone derivatives and their use as beta secretase inhibitors
WO2006034277A1 (en) 2004-09-17 2006-03-30 Comentis, Inc. Bicyclic compounds which inhibit beta-secretase activity and methods of use thereof
WO2006034296A2 (en) 2004-09-17 2006-03-30 Comentis, Inc. Amino-containing compounds which inhibit memapsin 2 beta-secretase activity and methods of use thereof
WO2006039470A2 (en) 2004-09-29 2006-04-13 Centocor, Inc. Anti- amyloid antibodies, compositions, methods and uses
WO2006039325A2 (en) 2004-10-01 2006-04-13 Merck & Co., Inc. Aminopiperidines as dipeptidyl peptidase-iv inhibitors for the treatment or prevention of diabetes
WO2006042103A2 (en) 2004-10-05 2006-04-20 Axys Pharmaceuticals, Inc. Reversible inhibitors of cathepsin b
WO2006041976A1 (en) 2004-10-08 2006-04-20 Novartis Ag Combination of organic compounds
WO2006040688A2 (en) 2004-10-12 2006-04-20 Ernir Snorrason Inhibitors of acetylcholinesterase for treating skin diseases
WO2006044497A2 (en) 2004-10-13 2006-04-27 Merck & Co., Inc. Spiropiperidine compounds useful as beta-secretase inhibitors for the treatment of alzhermer’s disease
WO2006039807A1 (en) 2004-10-15 2006-04-20 Biopharmacopae Design International Inc. Methods and therapeutic compositions comprising plant extracts for the treatment of cancer
WO2006055178A2 (en) 2004-10-25 2006-05-26 Merck & Co., Inc. Anti-addl antibodies and uses thereof
WO2006047248A1 (en) 2004-10-25 2006-05-04 Novartis Ag Combination of dpp-iv inhibitor, ppar antidiabetic and metformin
WO2006046644A1 (en) 2004-10-28 2006-05-04 Sanko Junyaku Co., Ltd. Method of examining alzheimer’s disease and diagnostic reagent
WO2006060109A1 (en) 2004-10-29 2006-06-08 Merck & Co., Inc. 2-aminopyridine compounds useful as beta-secretase inhibitors for the treatment of alzheimer's disease
WO2006055434A2 (en) 2004-11-17 2006-05-26 Merck & Co., Inc. Macrocyclic tertiary amine beta-secretase inhibitors for the treatment of alzheimer's disease
WO2006057945A2 (en) 2004-11-23 2006-06-01 Merck & Co., Inc. 2,3,4,6-substituted pyridyl derivative compounds useful as beta-secretase inhibitors for the treatment of alzheimer's disease
WO2006058059A2 (en) 2004-11-23 2006-06-01 Neuromolecular Pharmaceuticals, Inc. Composition comprising a sustained release coating or matrix and an nmda receptor antagonist, method for administration such nmda antagonist to a subject
WO2006057983A1 (en) 2004-11-23 2006-06-01 Merck & Co., Inc. Macrocyclic aminopyridyl beta-secretase inhibitors for the treatment of alzheimer's disease
WO2006058236A2 (en) 2004-11-24 2006-06-01 Neuromolecular Pharmaceuticals, Inc. Composition comprising an nmda receptor antagonist and levodopa and use thereof for treating neurological disease
WO2006058064A2 (en) 2004-11-29 2006-06-01 Merck & Co., Inc. Fused aminopiperidines as dipeptidyl peptidase-iv inhibitors for the treatment or prevention of diabetes
WO2006058628A2 (en) 2004-11-30 2006-06-08 F.Hoffmann-La Roche Ag Substituted benzoquinolizines as dpp-iv inhibitors for the treatment of diabetes
WO2006060473A2 (en) 2004-12-03 2006-06-08 Mucosal Therapeutics Llc Methods of treatment of injured or diseased joints with lubricin compositions
WO2006066171A1 (en) 2004-12-15 2006-06-22 Neuralab Limited Amyloid βετα antibodies for use in improving cognition
WO2006066049A2 (en) 2004-12-15 2006-06-22 Neuralab Limited Humanized antibodies that recognize beta amyloid peptide
WO2006066089A1 (en) 2004-12-15 2006-06-22 Neuralab Limited Humanized amyloid beta antibodies for use in improving cognition
WO2006066233A1 (en) 2004-12-15 2006-06-22 Neuralab Limited An immunoprecipitation-based assay for predicting in vivo efficacy of beta-amyloid antibodies
WO2006066747A1 (en) 2004-12-20 2006-06-29 F. Hoffmann-La Roche Ag 4-aminopiperidine derivatives
WO2006066770A1 (en) 2004-12-20 2006-06-29 F.Hoffmann-La Roche Ag Cycloalkylamine derivatives
WO2006068978A2 (en) 2004-12-21 2006-06-29 Takeda Pharmaceutial Company Limited Dipeptidyl peptidase inhibitors
WO2006071274A2 (en) 2004-12-23 2006-07-06 Voyager Pharmaceutical Corporation Leuprolide acetate and acetylcholinesterase inhibitors or nmda receptor antagonists for the treatment of alzheimer’s disease
WO2006070394A1 (en) 2004-12-28 2006-07-06 Council Of Scientific And Industrial Research Substituted carbamic acid quinolin-6-yl esters useful as acetylcholinesterase inhibitors
WO2006074265A2 (en) 2005-01-06 2006-07-13 Merck & Co., Inc. Drug combination therapy and pharmaceutical compositions for treating inflammatory disorders
WO2006078576A2 (en) 2005-01-19 2006-07-27 Merck & Co., Inc. Aminomethyl beta-secretase inhibitors for the treatment of alzheimer's disease
WO2006081171A1 (en) 2005-01-24 2006-08-03 Amgen Inc. Humanized anti-amyloid antibody
WO2006091988A1 (en) 2005-02-28 2006-08-31 Thomas Christian Lines Composition for treating mental health disorders
WO2006094674A1 (en) 2005-03-07 2006-09-14 Michael Hermanussen Nmda receptor antagonists in the medical intervention of metabolic disorders
WO2006095613A1 (en) 2005-03-09 2006-09-14 Konica Minolta Medical & Graphic, Inc. Rare earth activated alkaline earth metal fluorohalide photostimulable phosphor and radiation image conversion panel using the same
WO2006095041A1 (en) 2005-03-09 2006-09-14 Consejo Superior De Investigaciones Científicas Method for the in vitro diagnosis of alzheimer's disease using a monoclonal antibody
WO2006099352A1 (en) 2005-03-10 2006-09-21 Bristol-Myers Squibb Company Novel isophthalates as beta-secretase inhibitors
WO2006097624A1 (en) 2005-03-17 2006-09-21 Sanofi-Aventis 7-(2-(4-(3-trifluoromethyl-phenyl)-1,2,3,6-tetrahydro-pyrid-1-yl)ethyl) isoquinoline besylate salt, preparation and therapeutic use thereof
WO2006103116A1 (en) 2005-04-01 2006-10-05 Biotherapix Molecular Medicines S.L.U. Human antibodies with beta-amyloid peptide-binding capacity and their applications
WO2006116435A2 (en) 2005-04-27 2006-11-02 Novartis Ag Methods of treating atherosclerosis
WO2006118959A2 (en) 2005-04-29 2006-11-09 Rinat Neuroscience Corp. Antibodies directed against amyloid-beta peptide and methods using same
WO2006125202A2 (en) 2005-05-19 2006-11-23 Centocor, Inc. Anti- mcp-1 antibodies, compositions, methods and uses
WO2006137354A1 (en) 2005-06-21 2006-12-28 Medical & Biological Laboratories Co., Ltd. Antibody having inhibitory effect on amyloid fibril formation
WO2007068412A2 (en) 2005-12-12 2007-06-21 Ac Immune Sa A beta 1-42 specific monoclonal antibodies with therapeutic properties
WO2007068411A2 (en) 2005-12-12 2007-06-21 Ac Immune S.A. Therapeutic vaccine
EP2011349A2 (en) 2006-03-17 2009-01-07 Vodafone Group PLC Improvements in an ehspa architecture
WO2008011348A2 (en) 2006-07-14 2008-01-24 Ac Immune S.A. Humanized antibody against amyloid beta
WO2008055947A1 (en) 2006-11-09 2008-05-15 Probiodrug Ag Novel inhibitors of glutaminyl cyclase
WO2008055950A1 (en) 2006-11-09 2008-05-15 Probiodrug Ag Novel inhibitors of glutaminyl cyclase
WO2008055945A1 (en) 2006-11-09 2008-05-15 Probiodrug Ag 3-hydr0xy-1,5-dihydr0-pyrr0l-2-one derivatives as inhibitors of glutaminyl cyclase for the treatment of ulcer, cancer and other diseases
WO2008065141A1 (en) 2006-11-30 2008-06-05 Probiodrug Ag Novel inhibitors of glutaminyl cyclase

Non-Patent Citations (41)

* Cited by examiner, † Cited by third party
Title
"Remington's Pharmaceutical Sciences", MACK PUBLISHING CO., article "British and US Pharmacopoeias"
BATEMAN, R. C. ET AL., BIOCHEMISTRY, vol. 40, 2001, pages 11246 - 11250
BATEMAN, R. C. J., J NEUROSCI METHODS, vol. 30, 1989, pages 23 - 28
BECK A. ET AL., J PEPT RES, vol. 57, no. 6, 2001, pages 528 - 38
BHATIA, M., AM.J PHYSIOL GASTROINTEST.LIVER PHYSIOL, vol. 288, 2005, pages G1259 - G1265
BINDER, E. B. ET AL., BIOL PSYCHIATRY, vol. 50, 2001, pages 856 - 872
BOCKERS, T. M. ET AL., J NEUROENDOCRINOL, vol. 7, 1995, pages 445 - 453
BRINKMEIER H. ET AL., NATURE MEDICINE, vol. 6, 2000, pages 808 - 811
BUSBY, W. H. J. ET AL., J BIOL CHEM, vol. 262, 1987, pages 8532 - 8536
CLERICI, F. ET AL., NEUROBIOL.AGING, vol. 27, 2006, pages 1763 - 1768
COLL, B. ET AL., CYTOKINE, vol. 34, 2006, pages 51 - 55
CONSALVO, A. P. ET AL., ANAL BIOCHEM, vol. 175, 1988, pages 131 - 138
DAHL, S. W. ET AL., PROTEIN EXPR PURIF, vol. 20, 2000, pages 27 - 36
DOCKRAY, G.J., J PHYSIOL, vol. 15, 1999, pages 315 - 324
EI MOUSSAOUI, A. ET AL., CELL MOL LIFE SCI, vol. 58, 2001, pages 556 - 570
FISCHER, W. H.; SPIESS, J., PROC NATL ACAD SCI U S A, vol. 84, 1987, pages 3628 - 3632
FRASER, L.R.; ADEOYA-OSIGUWA, S. A., VITAM HORM, vol. 63, 2001, pages 1 - 28
GALIMBERTI, D. ET AL., ARCH.NEUROL., vol. 63, 2006, pages 538 - 543
GOLOLOBOV, M. Y. ET AL., BIOL CHEM HOPPE SEYLER, vol. 377, 1996, pages 395 - 398
GONG, J. H. ET AL., J EXP.MED, vol. 186, 1997, pages 131 - 137
GOSLING, J. ET AL., J CLIN.LNVEST, vol. 103, 1999, pages 773 - 778
H. BUNDGAARD: "Design of Prodrugs", 1985, ELSEVIER
HUSE J.T. ET AL., J. BIOL. CHEM., vol. 277, no. 18, 2002, pages 16278 - 16284
INOSHIMA, I. ET AL., AM.J PHYSIOL LUNG CELL MOL.PHYSIOL, vol. 286, 2004, pages L1038 - L1044
J.F.W. MCOMIE: "Protective Groups in Organic Chemistry", 1973, PLENUM PRESS
KATABUCHI, H. ET AL., MED ELECTRON MICROSC., vol. 36, 2003, pages 253 - 262
KOH, T.J.; CHEN, D., REGUL PEPT, 2000, pages 9337 - 44
LI, S. ET AL., J EXP.MED, vol. 202, 2005, pages 617 - 624
MARTINDALE: "The Extra Pharmacopoeia", THE PHARMACEUTICAL PRESS
MESSER, M., NATURE, vol. 4874, 1963, pages 1299
MOL. CELL, vol. 2, 1998, pages 275 - 281
OGATA, H. ET AL., J PATHOL., vol. 182, 1997, pages 106 - 114
OHTA, M. ET AL., INT.J ONCOL., vol. 22, 2003, pages 773 - 778
PARK, I. W.; WANG, J. F.; GROOPMAN, J. E., BLOOD, vol. 97, 2001, pages 352 - 358
POHL, T. ET AL., PROC NATL ACAD SCI U S A, vol. 88, 1991, pages 10059 - 10063
SAIDO T.C., MEDICAL HYPOTHESES, vol. 54, no. 3, 2000, pages 427 - 429
SAIURA, A. ET AL., ARTERIOSCLER. THROMB. VASC. BIOL., vol. 24, 2004, pages 1886 - 1890
T.W. GREENE; P.G.M. WUTS: "Protective Groups in Organic Synthesis", 1991, JOHN WILEY & SONS
WADA, T. ET AL., JAM.SOC.NEPHROL., vol. 15, 2004, pages 940 - 948
WHITE, F. A. ET AL., PROC. NATL. ACAD.SCI.U.S.A, 2005
YAMAMOTO, M. ET AL., AM.J PATHOL., vol. 166, 2005, pages 1475 - 1485

Cited By (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9505728B2 (en) 2012-03-09 2016-11-29 Inception 2, Inc. Triazolone compounds and uses thereof
US10195196B2 (en) 2012-08-13 2019-02-05 Novartis Ag 1,4-disubstituted pyridazine analogs there of and methods for treating SMN-deficiency-related conditions
US9545404B2 (en) 2012-08-13 2017-01-17 Novartis Ag 1,4-disubstituted pyridazine analogs there of and methods for treating SMN-deficiency-related conditions
US11229648B2 (en) 2012-08-13 2022-01-25 Novartis Ag 1,4-disubstituted pyridazine analogs thereof and methods for treating SMN-deficiency-related conditions
US10758533B2 (en) 2012-08-13 2020-09-01 Novartis Ag 1,4-disubstituted pyridazine analogs there of and methods for treating SMN-deficiency-related conditions
US8729263B2 (en) 2012-08-13 2014-05-20 Novartis Ag 1,4-disubstituted pyridazine analogs there of and methods for treating SMN-deficiency-related conditions
US9676754B2 (en) 2012-12-20 2017-06-13 Inception 2, Inc. Triazolone compounds and uses thereof
US11666557B2 (en) 2012-12-20 2023-06-06 Tempest Therapeutics, Inc. Triazolone compounds and uses thereof
US10568871B2 (en) 2012-12-20 2020-02-25 Tempest Therapeutics, Inc. Triazolone compounds and uses thereof
US9040712B2 (en) 2013-01-23 2015-05-26 Novartis Ag Thiadiazole analogs thereof and methods for treating SMN-deficiency-related-conditions
US11672799B2 (en) 2013-07-31 2023-06-13 Novartis Ag 1,4-disubstituted pyridazine quinolne analogs there of and methods for treating SMN-deficiency-related conditions
US9776976B2 (en) 2013-09-06 2017-10-03 Inception 2, Inc. Triazolone compounds and uses thereof
WO2016097305A1 (en) 2014-12-19 2016-06-23 Probiodrug Ag Novel method for the detection of pglu-abeta peptides
DE102015011780A1 (en) 2015-09-16 2017-03-16 Hochschule Anhalt New glutaminyl cyclase inhibitors
CN108250196A (en) * 2016-12-29 2018-07-06 上海汇伦生命科技有限公司 (Piperazine -1- bases)The preparation method of -1 hydrogen-imidazo heteroaromatic ring compounds
CN108250196B (en) * 2016-12-29 2022-12-30 上海汇伦医药股份有限公司 Process for the preparation of (piperazin-1-yl) -1-hydro-imidazoheteroaromatic compounds
US11220492B2 (en) 2017-05-17 2022-01-11 Arcus Biosciences, Inc. Quinazoline-pyrazole derivatives for the treatment of cancer-related disorders
EP3461819A1 (en) 2017-09-29 2019-04-03 Probiodrug AG Inhibitors of glutaminyl cyclase
WO2019149689A1 (en) 2018-01-31 2019-08-08 Probiodrug Ag Humanized and de-immunized antibodies
EP3521308A1 (en) 2018-01-31 2019-08-07 Probiodrug AG Humanized and de-immunized antibodies

Also Published As

Publication number Publication date
US9181233B2 (en) 2015-11-10
ES2586231T3 (en) 2016-10-13
WO2011107530A3 (en) 2011-11-10
JP2013521260A (en) 2013-06-10
US20110224259A1 (en) 2011-09-15
EP2542549A2 (en) 2013-01-09
JP6026284B2 (en) 2016-11-16
EP2542549B1 (en) 2016-05-11

Similar Documents

Publication Publication Date Title
EP2118101B1 (en) Imidazo [1,5-a] pyridine derivatives as inhibitors of glutaminyl cyclase
EP2865670B1 (en) Thiourea derivatives as glutaminyl cyclase inhibitors
EP2142513B1 (en) Nitrovinyl-diamine derivatives as glutaminyl cyclase inhibitors
EP2542549B1 (en) Inhibitors of glutaminyl cyclase
EP2545047B1 (en) Heterocyclic inhibitors of glutaminyl cyclase (qc, ec 2.3.2.5)
AU2009289238B2 (en) Novel inhibitors
EP2142515B1 (en) Nitrovinyl-diamine derivatives as glutaminyl cyclase inhibitors
EP2686313B1 (en) Benzimidazole derivatives as inhibitors of glutaminyl cyclase
WO2008128983A1 (en) Cyano-guanidine derivatives as glutaminyl cyclase inhibitors
EP2146968A1 (en) Urea derivatives as glutaminyl cyclase inhibitors
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: 11707395

Country of ref document: EP

Kind code of ref document: A2

WWE Wipo information: entry into national phase

Ref document number: 2012555421

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2011707395

Country of ref document: EP