US20100256188A1 - New compounds - Google Patents

New compounds Download PDF

Info

Publication number
US20100256188A1
US20100256188A1 US12/717,407 US71740710A US2010256188A1 US 20100256188 A1 US20100256188 A1 US 20100256188A1 US 71740710 A US71740710 A US 71740710A US 2010256188 A1 US2010256188 A1 US 2010256188A1
Authority
US
United States
Prior art keywords
alkyl
groups
fluoro
optionally substituted
cycloalkyl
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US12/717,407
Other languages
English (en)
Inventor
Roland Pfau
Kirsten Arndt
Henri Doods
Klaus Klinder
Raimund Kuelzer
Dimitrijs LUBRIKS
Juergen Mack
Benjamin Pelcman
Henning Priepke
Robert ROENN
Dirk Stenkamp
Edgars Suna
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Boehringer Ingelheim International GmbH
Original Assignee
Boehringer Ingelheim International GmbH
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=42111729&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20100256188(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Boehringer Ingelheim International GmbH filed Critical Boehringer Ingelheim International GmbH
Assigned to BOEHRINGER INGELHEIM INTERNATIONAL GMBH reassignment BOEHRINGER INGELHEIM INTERNATIONAL GMBH ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: SUNA, EDGARS, KLINDER, KLAUS, LUBRIKS, DIMITRIJS, PELCMAN, BENJAMIM, ARNDT, KIRSTEN, ROENN, ROBERT, KUELZER, RAIMUND, PRIEPKE, HENNING, MACK, JUERGEN, DOODS, HENRI, PFAU, ROLAND, STENKAMP, DIRK
Publication of US20100256188A1 publication Critical patent/US20100256188A1/en
Priority to US13/441,085 priority Critical patent/US20120196897A1/en
Priority to US13/942,077 priority patent/US8921405B2/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D235/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings
    • C07D235/02Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings condensed with carbocyclic rings or ring systems
    • C07D235/04Benzimidazoles; Hydrogenated benzimidazoles
    • C07D235/24Benzimidazoles; Hydrogenated benzimidazoles with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached in position 2
    • C07D235/30Nitrogen atoms not forming part of a nitro radical
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • 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]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • 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
    • C07D403/12Heterocyclic 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 linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/12Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/02Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings
    • C07D409/12Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links

Definitions

  • This invention relates to novel compounds, which are inhibitors of the microsomal prostaglandin E 2 synthase-1 (mPGES-1), pharmaceutical compositions containing them, and their use as medicaments for the treatment and/or prevention of inflammatory diseases and associated conditions.
  • mPGES-1 microsomal prostaglandin E 2 synthase-1
  • rheumatoid diseases e.g. rheumatoid arthritis, osteoarthritis, diseases of the visceral system e.g. inflammatory bowel syndrome, autoimmune diseases, e.g. lupus erythematodes, lung diseases like asthma and COPD.
  • Current treatment with non-steroidal anti-inflammatory drugs (NSAIDs) and Cyclooxygenase (COX)-2 inhibitors are efficacious, but show a prevalence for gastrointestinal and cardiovascular side effects. There is a high need for new treatment options showing equivalent efficacy with an improved side effect profile.
  • COX cyclooxygenase
  • COX-1 cyclooxygenase
  • COX-2 cyclooxygenase
  • COX-1 cyclooxygenase
  • COX-2 cyclooxygenase
  • COX-1 cyclooxygenase
  • COX-2 cyclooxygenase
  • COX-1 cyclooxygenase
  • COX-2 cyclooxygenase
  • COXs metabolise arachidonic acid to the unstable intermediate prostaglandin H 2 (PGH 2 ).
  • PGH 2 is further metabolized to other prostaglandins including PGE 2 , PGF 2 ⁇ , PGD 2 , prostacyclin and thromboxane A 2 .
  • PGE 2 arachidonic acid metabolites are known to have pronounced physiological and pathophysiological activity including pro-inflammatory effects.
  • PGE 2 in particular is known to be a strong pro-inflammatory mediator, and is also known to induce fever, inflammation and pain. Consequently, numerous drugs were developed with a view to inhibiting the formation of PGE 2 , including “NSAIDs” (non-steroidal antiinflammatory drugs) and “coxibs” (selective COX-2 inhibitors). These drugs act predominantly by inhibition of COX-1 and/or COX-2, thereby reducing the formation of PGE 2 .
  • NSAIDs non-steroidal antiinflammatory drugs
  • coxibs selective COX-2 inhibitors
  • the non-selective inhibition of COXs by NSAIDs may give rise to gastrointestinal side-effects and affect platelet and renal function.
  • Even the selective inhibition of COX-2 by coxibs, whilst reducing such gastrointestinal side-effects, is believed to give rise to cardiovascular problems.
  • a drug that preferably selectively inhibits the transformation of PGH 2 to the pro-inflammatory mediator PGE 2 might be expected to reduce the inflammatory response in the absence of a corresponding reduction of the formation of other, beneficial arachidonic acid metabolites. Such inhibition would accordingly be expected to alleviate the undesirable side-effects mentioned above.
  • PGH 2 may be transformed to PGE 2 by prostaglandin E synthases (PGES).
  • PGES prostaglandin E synthases
  • mPGES-1 and mPGES-2 microsomal prostaglandin E synthases
  • cPGES cytosolic prostaglandin E synthase
  • the leukotrienes (LTs) are formed from arachidonic acid by a set of enzymes distinct from those in the COX/PGES pathway.
  • Leukotriene B 4 is known to be a strong proinflammatory mediator, while the cysteinyl-containing leukotrienes C 4 , D 4 and E 4 (CysLTs) are mainly very potent bronchoconstrictors and have thus been implicated in the pathobiology of asthma.
  • LTRas leukotriene receptor antagonists
  • the biological activities of the CysLTs are mediated through two receptors designated CysLT 1 and CysLT 2 .
  • LTRas leukotriene receptor antagonists
  • These drugs may be given orally, but do not control inflammation satisfactorily.
  • the presently used LTRas are highly selective for CysLT 1 . It may be hypothesised that better control of asthma, and possibly also COPD, may be attained if the activity of both of the CysLT receptors could be reduced. This may be achieved by developing unselective LTRas, but also by inhibiting the activity of proteins, e.g. enzymes, involved in the synthesis of the CysLTs. Among these proteins, 5-lipoxygenase, 5-lipoxygenase-activating protein (FLAP), and leukotriene C 4 synthase may be mentioned.
  • FLAP 5-lipoxygenase
  • FLAP 5-lipoxygenase-activating protein
  • mPGES-1, FLAP and leukotriene C 4 synthase belong to the membrane-associated proteins in the eicosanoid and glutathione metabolism (MAPEG) family.
  • Other members of this family include the microsomal glutathione S-transferases (MGST1, MGST2 and MGST3).
  • MGST1, MGST2 and MGST3 microsomal glutathione S-transferases
  • compounds prepared as antagonists to one of the MAPEGs may also exhibit inhibitory activity towards other family members, c.f. J. H Hutchinson et al in J. Med. Chem. 38, 4538 (1995) and D.
  • agents that are capable of inhibiting the action of mPGES-1, and thus reducing the formation of the specific arachidonic acid metabolite PGE 2 are likely to be of benefit in the treatment of inflammation. Further, agents that are capable of inhibiting the action of the proteins involved in the synthesis of the leukotrienes are also likely to be of benefit in the treatment of asthma and COPD.
  • mPGES-1 inhibitors are also known to be of potential use in treating or preventing a neoplasia, for example as described in international patent application WO 2007/124589.
  • the rationale behind this may stem from the fact that the production of PGE2 is believed to promote the formation, growth and/or metastasis of neoplasias.
  • mPGES-1 is often expressed with COX-2 in benign and cancerous neoplasias
  • the inhibition of mPGES-1 (rather than COX-2) may cause the reduction of PGE2 and therefore mPGES-1 inhibitors may be useful for the treatment of benign or malignant neoplasias.
  • a further embodiment of the present invention comprises compounds of the general formula I, namely compounds of formula Ia
  • A, L, M, Q 2 , Q 3 , Q 4 , R 1 , R 6 , R a , R b , R c , W, Z 1 , Z 2 , Z 3 have the same meaning as defined in any of the preceding embodiments, the salts thereof, particularly the physiologically acceptable salts thereof with organic or inorganic acids or bases.
  • a further embodiment of the present invention comprises compounds of the formula I, namely compounds of formula Ib
  • A, L, M, Q 2 , Q 3 , Q 4 , R 1 , R 6 , R 7 , R a , R b , R c , W have the same meaning as defined in any of the preceding embodiments, the salts thereof, particularly the physiologically acceptable salts thereof with organic or inorganic acids or bases.
  • a further embodiment of the present invention comprises compounds of the formula I, namely compounds of formula IC
  • A, L, M, R 1 , R 2 , R 3 , R 4 , R 6 , R a , R b , R c , W, Z 1 , Z 2 , Z 3 have the same meaning as defined in any of the preceding embodiments, the salts thereof, particularly the physiologically acceptable salts thereof with organic or inorganic acids or bases.
  • a further embodiment of the present invention comprises compounds of the formula I, namely compounds of formula Id
  • A, L, M, R 1 , R 2 , R 3 , R 4 , R 6 , R 7 , R a , R b , R c , W have the same meaning as defined in any of the preceding embodiments, the salts thereof, particularly the physiologically acceptable salts thereof with organic or inorganic acids or bases.
  • A, L, M, Q 2 , Q 3 , Q 4 , R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R a , R b , R c , X, Y, Z 1 , Z 2 , Z 3 have the same meaning as defined in any of the preceding embodiments and
  • A, L, Q 2 , Q 3 , Q 4 , R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R a , R b , R c , W, X, Y, Z 1 , Z 2 , Z 3 have the same meaning as defined in any of the preceding embodiments and
  • A, L, M, Q 2 , Q 3 , Q 4 , R 1 , R 2 , R 3 , R 4 , R 5 , R 7 , R a , R b , R c , W, X, Y, Z 1 , Z 2 , Z 3 have the same meaning as defined in any of the preceding embodiments and
  • A, L, M, Q 2 , Q 3 , Q 4 , R 1 , R 5 , R 6 , R 7 , R 9a , R 9b , R a , R b , R c , W, X, Y, Z 1 , Z 2 , Z 3 have the same meaning as defined in any of the preceding embodiments and
  • A, L, M, Q 2 , Q 3 , Q 4 , R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R a , R b , R c , W, X, Y, Z 1 , Z 2 , Z 3 have the same meaning as defined in any of the preceding embodiments and
  • A, L, M, Q 2 , Q 3 , Q 4 , R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R a , R b , R c , W, X, Y, Z 1 , Z 2 , Z 3 have the same meaning as defined in any of the preceding embodiments and
  • A, M, Q 2 , Q 3 , Q 4 , R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R a , R b , R c , W, X, Y, Z 1 , Z 2 , Z 3 have the same meaning as defined in any of the preceding embodiments and
  • A, M, Q 2 , Q 3 , Q 4 , R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R a , R b , R c , W, X, Y, Z 1 , Z 2 , Z 3 have the same meaning as defined in any of the preceding embodiments and
  • L, A, M, Q 2 , Q 3 , Q 4 , R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R a , R b , R c , W, X, Y, Z 1 , Z 2 , Z 3 have the same meaning as defined in any of the preceding embodiments and
  • L, M, Q 2 , Q 3 , Q 4 , R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R a , R b , R c , W, X, Y, Z 1 , Z 2 , Z 3 have the same meaning as defined in any of the preceding embodiments and
  • A, L, Q 2 , Q 3 , Q 4 , R 1 , R 2 , R 3 , R 4 , to R 5 , R 6 , R 7 , R a , R b , R c , W, X, Y, Z 1 , Z 2 , Z 3 have the same meaning as defined in any of the preceding embodiments and
  • A, L, M, Q 2 , Q 3 , Q 4 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R a , R b , R c , W, X, Y, Z 1 , Z 2 , Z 3 have the same meaning as defined in the any of the preceding embodiments and
  • A, L, M, Q 2 , Q 3 , Q 4 , R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , W, X, Y, Z 1 , Z 2 , Z 3 have the same meaning as defined in any of the preceding embodiments and
  • a further embodiment of the present invention comprises compounds of the formula I, namely compounds of formula Ie
  • phrases “pharmaceutically acceptable” is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication commensurate with a reasonable benefit/risk ratio.
  • pharmaceutically acceptable salts refer to derivatives of the disclosed compounds wherein the parent compound is modified by making acid or base salts thereof.
  • examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like.
  • the pharmaceutically acceptable salts include the conventional non-toxic salts or the quaternary ammonium salts of the parent compound formed, for example, from non-toxic inorganic or organic acids.
  • the pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound which contains a basic or acidic moiety by conventional chemical methods.
  • such salts can be prepared by reacting the free acid or base forms of theses compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, nonaqueous media like ether, ethyl acetate, ethanol isopropanol, or acetonitrile are preferred.
  • Lists of suitable salts are found in Remington's Pharmaceutical Sciences, 17th ed., Mack Publishing Company, Easton, Pa., 1985, p. 1418, the disclosure of which is hereby incorporated by reference.
  • Examples of pharmaceutically active salts for each of the compounds which are the subject of this description include, without being restricted thereto, salts which are prepared from pharmaceutically acceptable acids or bases, including organic and inorganic acids and bases. If the preferred compound is basic, salts may be prepared from pharmaceutically acceptable acids. When selecting the most preferred salt, or to clarify whether a salt or the neutral compound is used, properties such as bioavailability, ease of manufacture, workability and shelf life are taken into consideration, inter alia.
  • Suitable pharmaceutically acceptable acids include acetic acid, benzenesulphonic acid (besylate), benzoic acid, p-bromophenylsulphonic acid, camphorsulphonic acid, carbonic acid, citric acid, ethanesulphonic acid, fumaric acid, gluconic acid, glutamic acid, hydrobromic acid, hydrochloric acid, hydriodic acid, isethionic acid, lactic acid, maleic acid, malic acid, mandelic acid, methanesulphonic acid (mesylate), mucinic acid, nitric acid, oxalic acid, pamoic acid, pantothenic acid, phosphoric acid, succinic acid, sulphuric acid, tartaric acid, p-toluenesulphonic acid and the like.
  • Examples of pharmaceutically acceptable salts include, without being restricted thereto, acetate, benzoate, hydroxybutyrate, bisulphate, bisulphite, bromide, butyne-1,4-dioate, caproate, chloride, chlorobenzoate, citrate, dihydrogen phosphate, dinitrobenzoate, fumarate, glutaminate, glycollate, heptanoate, hexyne-1,6-dioate, hydroxybenzoate, iodide, lactate, maleate, malonate, mandelate, metaphosphate, methanesulphonate, methoxybenzoate, methylbenzoate, monohydrogen phosphate, naphthalene-1-sulphonate, naphthalene-2-sulphonate, oxalate, phenylbutyrate, phenylproprionate, phosphate, phthalate, phenylacetate, propanesulphonate, propiolate, propionate,
  • an enantiomer or a diastereomer of a compound of Formula I may display superior activity compared with the other.
  • diastereomers may be separated from diastereomeric mixture of the compounds using conventional, e.g. fractional crystallisation or HPLC techniques.
  • the desired enantiomers may be obtained by methods well known to the skilled man in the art i.e. by reaction of the appropriate optically active starting materials under conditions which will not cause racemisation or epimerisation (i.e. a ‘chiral pool’ method), by reaction of the appropriate starting material with a ‘chiral auxiliary’ which can subsequently be removed at a suitable stage, by derivatisation (i.e.
  • a resolution including a dynamic resolution
  • a resolution for example with a homochiral acid followed by separation of the diastereomeric derivatives by conventional means such as chromatography (see for example Thomas J. Tucker, et al, J. Med. Chem. 1994, 37, 2437-2444), or by reaction with an appropriate chiral reagent or chiral catalyst all under conditions known to the skilled person (e.g. Mark A. Huffman, et al, J. org. Chem. 1995, 60 1590-1594).
  • Compounds of the invention may contain double bonds and may thus exist as E (entadel) and Z (zusammen) geometric isomers about each individual double bond. All such isomers and mixtures thereof are included within the scope of the invention.
  • the subject-matter of this invention also includes deuterated compounds of general formula I, i.e. in which one or more hydrogen atoms, for example one, two, three, four or five hydrogen atoms, are replaced by the hydrogen isotope deuterium.
  • C 1-3 -alkyl (including those which are part of other groups) means alkyl groups with 1 to 3 carbon atoms
  • C 1-4 -alkyl means branched and unbranched alkyl groups with 1 to 4 carbon atoms
  • C 1-6 -alkyl means branched and unbranched alkyl groups with 1 to 6 carbon atoms
  • C 1-8 -alkyl means branched and unbranched alkyl groups with 1 to 8 carbon atoms.
  • Examples include: methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, sec-butyl, tert-butyl, n-pentyl, neopentyl, n-hexyl, n-heptyl and n-octyl.
  • the abbreviations Me, Et, n-Pr, i-Pr, n-Bu, i-Bu, t-Bu, etc. may optionally also be used for the groups mentioned above. Unless stated otherwise, the definitions propyl and butyl include all the possible isomeric forms of the groups in question.
  • propyl includes n-propyl and iso-propyl
  • butyl includes iso-butyl, sec-butyl and tert-butyl
  • alkyl also includes alkyl groups wherein 1-3 hydrogen atoms are replaced by fluorine atoms.
  • C 1-n -alkylene wherein n is an integer 1 to n, either alone or in combination with another radical, denotes an acyclic, straight or branched chain divalent alkyl radical containing from 1 to n carbon atoms.
  • C 1-4 -alkylene includes —(CH 2 )—, —(CH 2 —CH 2 )—, —(CH(CH 3 ))—, —(CH 2 —CH 2 —CH 2 )—, —(C(CH 3 ) 2 )—, —(CH(CH 2 CH 3 ))—, —(CH(CH 3 )—CH 2 )—, —(CH 2 —CH(CH 3 ))—, —(CH 2 —CH 2 —CH 2 —CH 2 )—, —(CH 2 —CH 2 —CH(CH 3 ))—, —(CH(CH 3 )—CH 2 —CH 2 )—, —(CH 2 —CH(CH 3 )—CH 2 —CH 2 )—, —(CH 2 —CH(CH 3 )—CH 2 )—, —(CH 2 —CH(CH 3 )—CH 2 )—, —(CH 2 —CH(
  • C 3-8 -cycloalkyl (including those which are part of other groups) means cyclic alkyl groups with 3 to 8 carbon atoms and the term “C 3-6 -cycloalkyl” means cyclic alkyl groups with 3 to 6 carbon atoms. Examples include: cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl or cyclooctyl and also include the following structures
  • the cyclic alkyl groups may be substituted by one or more groups selected from among methyl, ethyl, iso-propyl, tert-butyl, hydroxy, fluorine, chlorine, bromine and iodine.
  • These cycloalkyls may additionally be annelated (i.e. fused) to a benzene ring, so that nine- to twelve-membered bicyclic heterocycles are formed.
  • C 2-6 -alkenyl (including those which are part of other groups) means branched and unbranched alkenyl groups with 2 to 6 carbon atoms, provided that they have at least one double bond. Alkenyl groups with 2 to 4 carbon atoms are preferred. Examples include: ethenyl or vinyl, propenyl, butenyl, pentenyl, or hexenyl. Unless stated otherwise, the definitions propenyl, butenyl, pentenyl and hexenyl include all the possible isomeric forms of the groups in question.
  • propenyl includes 1-propenyl and 2-propenyl
  • butenyl includes 1-butenyl, 2-butenyl and 3-butenyl, 1-methyl-1-propenyl, 1-methyl-2-propenyl etc.
  • C 2-7 -alkynyl (including those which are part of other groups) means branched and unbranched alkynyl groups with 2 to 7 carbon atoms, provided that they have at least one triple bond. Alkynyl groups with 2 to 4 carbon atoms are preferred. Examples of these include: ethynyl, propynyl, butynyl, pentynyl or hexynyl. Unless stated otherwise, the definitions propynyl, butynyl, pentynyl and hexynyl include all the possible isomeric forms of the groups in question.
  • propynyl includes 1-propynyl and 2-propynyl
  • butynyl includes 1-butynyl, 2-butynyl and 3-butynyl, 1-methyl-1-propynyl, 1-methyl-2-propynyl etc.
  • halo or “halogen”, when used herein, includes fluoro, chloro, bromo and iodo. Unless stated otherwise, fluorine, chlorine and bromine are regarded as preferred halogens.
  • 4-, 5-, 6- or 7-membered heterocycloalkyl means stable 4-, 5-, 6- or 7-membered monocyclic heterocyclic ring systems which may be both saturated and monounsaturated.
  • One or two of the ring carbon atoms may independently be replaced by heteroatoms which are selected from among oxygen, nitrogen and sulphur, the latter two of which heteroatoms may optionally be oxidised (so forming N-oxide, sulfoxide or sulfon).
  • the previously mentioned heterocycles may be linked to the rest of the molecule via a carbon atom or a nitrogen atom.
  • Examples for the 4-, 5-, 6- or 7-membered heterocyclic ring systems include:
  • heterocycloalkyls may additionally be annelated (i.e. fused) to a benzene ring, so that eight- or eleven-membered bicyclic heterocycles are formed.
  • aryl means aromatic ring systems with 6 or 14 carbon atoms (e.g. C 6-10 aryl). Such groups may be monocyclic, bicyclic or tricyclic and have between 6 and 14 ring carbon atoms.
  • Examples include phenyl, 1-naphthyl or 2-naphthyl.
  • the preferred aryl group is phenyl.
  • the aromatic groups may be substituted by one or more groups selected from among methyl, ethyl, n-propyl, iso-propyl, tert-butyl, hydroxy, methoxy, trifluoromethoxy, fluorine, chlorine, bromine and iodine, while the groups may be identical or different.
  • heteroaryl means five- or six-membered heterocyclic aromatic groups which may contain one, two, three or four heteroatoms, selected from among oxygen, sulphur and nitrogen, and which additionally contain a sufficient number of conjugated double bonds to form an aromatic system. These heteroaryls may additionally be annelated (i.e. fused) to a benzene ring, so that nine- or ten-membered bicyclic heteroaryls are formed.
  • Examples for the five- or six-membered heterocyclic aromatic groups include:
  • heteroaryls mentioned previously may be substituted by one or more groups selected from among methyl, ethyl, n-propyl, iso-propyl, tert-butyl, hydroxy, methoxy, trifluoromethoxy, fluorine, chlorine, bromine and iodine, while the groups may be identical or different.
  • a nitrogen atom present in the heteroaryl group may be oxidised, thus forming an N-oxide.
  • the identities of the two —N(C 1-3 alkyl) 2 groups are not to be regarded as being interdependent, i.e. the two —N(C 1-3 alkyl) 2 moieties may be the same or different.
  • the C 1-3 alkyl groups within one particular —N(C 1-3 alkyl) 2 group are independent of one another, i.e. they may be the same or different from each other.
  • compounds of formula I that are the subject of this invention include those that are stable. That is, compounds of the invention include those that are sufficiently robust to survive isolation from e.g. a reaction mixture to a useful degree of purity.
  • the protecting group PG which is introduced in step a) is a standard nitrogen protecting group, well known to those skilled in the art, for example a nitrogen protecting group as described in “ Protective Groups in Organic Synthesis”, 3rd edition, T. W. Greene & P. G. M. Wutz, Wiley-Interscience (1999), for example a tert-butoxycarbonyl-, benzyloxycarbonyl-, ethoxycarbonyl-, methoxycarbonyl-, allyloxycarbonyl- or trifluormethylcarbonyl group.
  • Step a) can be performed according to literature procedures as described in “ Protective Groups in Organic Synthesis”, 3 rd edition, T. W. Greene & P. G. M. Wutz, Wiley-Interscience (1999) for example with reagents like 2-(tert-Butoxycarbonyloxyimino)-2-phenylacetonitrile (BOC-ON), Di-tert-butyl dicarbonate (BOC 2 O), Di-methyl dicarbonate, Di-ethyl dicarbonate, Ethyl chloroformate, Methyl chloroformate, Allyl chloroformate, Benzyl chloroformate or Trifluoroacetic acid chloride under conditions which are known from the literature preferably in presence of a base for example sodium hydroxide, triethylamine, diisopropyl ethyl amine, 4-dimethylamino-pyridine.
  • BOC-ON 2-(tert-Butoxycarbonyloxyimino)-2-phenylaceton
  • Appropriate solvent for this step is for example dichloromethane, tetrahydrofuran (THF), acetonitrile, dimethylformamide (DMF), dimethylacetamide, N methylpyrrolidone or mixtures of the above mentioned solvents.
  • Step b) can be performed with H 2 /Raney-Nickel, H 2 /Palladium on carbon, Fe-powder/aqueous NH 4 Cl, Fe/HCl, Zn/HCl, Na 2 S 2 C 4 , SnCl 2 /HCl, Zn/HCl or NaBH 4 /CuCl or according to procedures described in the literature for example R. Larock, Comprehensive Organic Transformations , VCH Verlagstician, Weinheim (1989).
  • Appropriate solvent for this step is for example dichloromethane, tetrahydrofuran (THF), acetonitrile, dimethylformamide (DMF), dimethylacetamide, N methylpyrrolidone, ethanol, methanol, isopropanol or mixtures of the above mentioned solvents.
  • Step c) can be performed according to standard literature procedures for example with reagents such as 1,1′-thiocarbonyldi-2-pyridone or 1,1′-thiocarbonyldiimidazole or with thiophosgene in a solvent as for example dichloromethane or dimethylformamide and optionally under addition of a base like 4-dimethylamino-pyridine or triethylamine.
  • reagents such as 1,1′-thiocarbonyldi-2-pyridone or 1,1′-thiocarbonyldiimidazole or with thiophosgene in a solvent as for example dichloromethane or dimethylformamide and optionally under addition of a base like 4-dimethylamino-pyridine or triethylamine.
  • Step d) can be performed under standard conditions known to those skilled in the art in presence of a suitable solvent such as diethyl ether, dimethylformamide, dichloromethane, acetononitrile and/or tetrahydrofuran.
  • a suitable solvent such as diethyl ether, dimethylformamide, dichloromethane, acetononitrile and/or tetrahydrofuran.
  • the coupling reaction is preferably performed in the presence of a base such as NaOH, KOH, NaHCO 3 , triethylamine, N-ethyldiisopropylamine, 4-dimethylamino-pyridine or other appropriate bases.
  • Step e) is preferably performed in the presence of a suitable ‘coupling’ reagent.
  • ‘coupling’ reagent for instance a carbodiimide based compound such as dicyclohexylcarbodiimide (DCC), 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide (or salt, e.g. hydrochloride, thereof) or N,N-diisopropylcarbodiimide (DIC) can be used.
  • DCC dicyclohexylcarbodiimide
  • 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide (or salt, e.g. hydrochloride, thereof) or N,N-diisopropylcarbodiimide (DIC) can be used.
  • the reaction may proceed at any suitable temperature (e.g. one between about 0° C.
  • Step d and e can be performed in a step-wise reaction under isolation of the intermediate XV or without isolation of XV.
  • Step f) Protecting groups may be removed in accordance with techniques that are well known to those skilled in the art and as described hereinafter.
  • protected compounds/intermediates described herein may be converted chemically to unprotected compounds using HCl or H 2 SO 4 solutions, trifluoro acetic acid, KOH; Ba(OH) 2 , Pd on carbon, trimethylsilyl iodide or other conditions as described in “ Protective Groups in Organic Synthesis”, 3 rd edition, T. W. Greene & P. G. M. Wutz, Wiley-Interscience (1999).
  • Appropriate co-solvent for this step is for example dichloromethane, tetrahydrofuran (THF), acetonitrile, dimethylformamide (DMF), dimethylacetamide, N methylpyrrolidone or mixtures of the above mentioned solvents.
  • Step g): The coupling of the amine XVII with the acid XXa can be performed with an additional in-situ activating agent like 1-propylphosphonic acid cyclic anhydride, O-(benzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium tetrafluoroborate or hexafluorophosphate, N,N′-dicyclohexylcarbodiimide, 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide, carbonyldiimidazole, oxalyl chloride or other activating agents of the state of the art.
  • an additional in-situ activating agent like 1-propylphosphonic acid cyclic anhydride, O-(benzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium tetrafluoroborate or hexafluorophosphate, N
  • the coupling reaction is preferably performed in the presence of a base such as NaOH, KOH, NaHCO 3 , triethylamine, N-ethyldiisopropylamine, N,N,-dimethylaminopyridine or other appropriate bases of the state of the art and for example described in described in Houben-Weyl, “Methods in Organic Synthesis”, Vol. E22a, p 425ff.
  • a base such as NaOH, KOH, NaHCO 3 , triethylamine, N-ethyldiisopropylamine, N,N,-dimethylaminopyridine or other appropriate bases of the state of the art and for example described in described in Houben-Weyl, “Methods in Organic Synthesis”, Vol. E22a, p 425ff.
  • the coupling of the amine XVII can be performed with an activated acid derivative XXI), where the leaving group LG can be for example, a fluorine, chlorine, bromine, azide or an isopropyloxy-C(O)—O anion.
  • the coupling reaction is preferably performed in the presence of a base such as NaOH, KOH, NaHCO 3 , triethylamine, N-ethyldiisopropylamine, 4-dimethylamino-pyridine or other appropriate bases under conditions which are for example described in Houben-Weyl, “Methods in Organic Synthesis”, Vol. E22a, p 425ff.
  • step g) is carried out in presence of trimethylaluminium or triethylaluminium
  • the leaving group in XXb can also be a methoxy or ethoxy group.
  • the above mentioned coupling reactions are performed in an appropriate solvent for example like dichloromethane, tetrahydrofuran (THF), acetonitrile, dimethylformamide (DMF), dimethylacetamide, N methylpyrrolidone or in mixtures of the above mentioned solvents.
  • an appropriate solvent for example like dichloromethane, tetrahydrofuran (THF), acetonitrile, dimethylformamide (DMF), dimethylacetamide, N methylpyrrolidone or in mixtures of the above mentioned solvents.
  • Step h) The reductive amination is performed according to known procedures for example with sodium triacetoxyborohydride, sodium borohydride or sodium cyanoborhydride in an appropriate solvent like tetrahydrofuran or dichlormethane under addition of acetic acid or trifluoro acetic acid if appropriate, or with Palladium on charcoal under a hydrogen atmosphere in tetrahydrofuran or ethanol or methanol or isopropanol or dimethylformamide, preferably in the presence of acetic acid or trifluoro acetic acid.
  • Step i) can be performed according to standard literature procedures for example with reagents such as thiophosgene in an appropriate solvent as for example dichloromethane or dimethylformamide optionally under addition of a base like 4-dimethylamino-pyridine or triethylamine.
  • reagents such as thiophosgene in an appropriate solvent as for example dichloromethane or dimethylformamide
  • a base like 4-dimethylamino-pyridine or triethylamine.
  • Step j) can be performed under standard conditions known to those skilled in the art in presence of a suitable solvent such as diethyl ether, dimethylformamide, dichloromethane, acetononitrile and/or tetrahydrofuran).
  • a suitable solvent such as diethyl ether, dimethylformamide, dichloromethane, acetononitrile and/or tetrahydrofuran.
  • the coupling reaction is preferably performed in the presence of a base such as NaOH, KOH, NaHCO 3 , triethylamine, N-ethyldiisopropylamine, 4-dimethylamino-pyridine or other appropriate bases.
  • Step k) is preferably performed in the presence of a suitable coupling reagent as for example a carbodiimide based compound such as dicyclohexylcarbodiimide (DCC), 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide (or salt, e.g. hydrochloride, thereof) or N,N-diisopropylcarbodiimide (DIC).
  • DCC dicyclohexylcarbodiimide
  • DCC dicyclohexylcarbodiimide
  • DIC N,N-diisopropylcarbodiimide
  • the reaction may proceed at any suitable temperature (e.g. one between about 0° C. to about 200° C
  • Appropriate co-solvent for this step is for example dichloromethane, tetrahydrofuran, acetonitrile, dimethylformamide, dimethylacetamide, N methylpyrrolidone or mixtures of the above mentioned solvents.
  • Step j and k can be performed in a step-wise reaction under isolation of the intermediate XXI or without isolation of XXI.
  • Step l) Protecting groups may be removed in accordance with techniques that are well known to those skilled in the art and as described hereinafter.
  • protected compounds/intermediates described herein may be converted chemically to unprotected compounds using HCl or H 2 SO 4 solutions, trifluoro acetic acid, KOH; Ba(OH) 2 , Pd on carbon, trimethylsilyl iodide or other conditions as described in “ Protective Groups in Organic Synthesis”, 3 rd edition, T. W. Greene & P. G. M. Wutz, Wiley-Interscience (1999).
  • T Appropriate co-solvent for this step is for example dichloromethane, tetrahydrofuran, acetonitrile, dimethylformamide, dimethylacetamide, N methylpyrrolidone, methanol, ethanol or mixtures of the above mentioned solvents.
  • Step m): The coupling of the amine XXIII with the acid XXa can be performed with an additional in-situ activating agent like 1-propylphosphonic acid cyclic anhydride, O-(benzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium tetrafluoroborate or hexafluorophosphate, N,N′-dicyclohexylcarbodiimide, 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide, carbonyldiimidazole, oxalyl chloride or other activating agents of the state of the art.
  • an additional in-situ activating agent like 1-propylphosphonic acid cyclic anhydride, O-(benzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium tetrafluoroborate or hexafluorophosphate, N
  • the coupling reaction is preferably performed in the presence of a base such as NaOH, KOH, NaHCO 3 , triethylamine, N-ethyldiisopropylamine, N,N,-dimethylaminopyridine or other appropriate bases of the state of the art and for example described in described in Houben-Weyl, “Methods in Organic Synthesis”, Vol. E22a, p 425ff.
  • a base such as NaOH, KOH, NaHCO 3 , triethylamine, N-ethyldiisopropylamine, N,N,-dimethylaminopyridine or other appropriate bases of the state of the art and for example described in described in Houben-Weyl, “Methods in Organic Synthesis”, Vol. E22a, p 425ff.
  • the coupling of the amine XXIII can be performed with an activated acid derivative XXb, where the leaving group LG can be for example, a fluorine, chlorine, bromine, azide or an isopropyloxy-C(O)—O anion.
  • the coupling reaction is preferably performed in the presence of a base such as NaOH, KOH, NaHCO 3 , triethylamine, N-ethyldiisopropylamine, 4-dimethylamino-pyridine or other appropriate bases under conditions which are for example described in Houben-Weyl, “Methods in Organic Synthesis”, Vol. E22a, p 425ff.
  • step g) is carried out in presence of trimethylaluminium or triethylaluminium
  • the leaving group in XXb can also be a methoxy or ethoxy group.
  • Step n) can be performed according to standard literature procedures for example with reagents such as 1,1′-thiocarbonyldi-2-pyridone or 1,1′-thiocarbonyldiimidazole or with thiophosgene in a solvent as for example dichloromethane or dimethylformamide and optionally under addition of a base like 4-dimethylamino-pyridine or triethylamine.
  • reagents such as 1,1′-thiocarbonyldi-2-pyridone or 1,1′-thiocarbonyldiimidazole or with thiophosgene in a solvent as for example dichloromethane or dimethylformamide and optionally under addition of a base like 4-dimethylamino-pyridine or triethylamine.
  • Step o) can be performed under standard conditions known to those skilled in the art in presence of a suitable solvent such as diethyl ether, dimethylformamide, dichloromethane, acetononitrile and/or tetrahydrofuran.
  • a suitable solvent such as diethyl ether, dimethylformamide, dichloromethane, acetononitrile and/or tetrahydrofuran.
  • the coupling reaction is preferably performed in the presence of a base such as NaOH, KOH, NaHCO 3 , triethylamine, N-ethyldiisopropylamine, 4-dimethylamino-pyridine or other appropriate bases.
  • Step p) is preferably performed in the presence of a suitable ‘coupling’ reagent.
  • ‘coupling’ reagent for instance a carbodiimide based compound such as dicyclohexylcarbodiimide (DCC), 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide (or salt, e.g. hydrochloride, thereof) or N,N-diisopropylcarbodiimide (DIC) can be used.
  • DCC dicyclohexylcarbodiimide
  • 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide (or salt, e.g. hydrochloride, thereof) or N,N-diisopropylcarbodiimide (DIC) can be used.
  • the reaction may proceed at any suitable temperature (e.g. one between about 0° C.
  • Step o and p can be performed in a step-wise reaction under isolation of the intermediate XXV or without isolation of XXV.
  • the protecting group PG 1 is a standard nitrogen protecting group, well known to those skilled in the art, for example a nitrogen protecting group as described in “ Protective Groups in Organic Synthesis”, 3 rd edition, T. W. Greene & P. G. M. Wutz, Wiley-Interscience (1999), for example a tert-butoxycarbonyl-, benzyloxycarbonyl-, ethoxycarbonyl-, methoxycarbonyl-, allyloxycarbonyl- or trifluormethylcarbonyl group.
  • Step q) can be performed under standard conditions known to those skilled in the art in presence of a suitable solvent such as diethyl ether, dimethylformamide, dichloromethane, acetononitrile and/or tetrahydrofuran.
  • a suitable solvent such as diethyl ether, dimethylformamide, dichloromethane, acetononitrile and/or tetrahydrofuran.
  • the coupling reaction is preferably performed in the presence of a base such as NaOH, KOH, NaHCO 3 , triethylamine, N-ethyldiisopropylamine, 4-dimethylamino-pyridine or other appropriate bases.
  • Step r) is preferably performed in the presence of a suitable ‘coupling’ reagent.
  • ‘coupling’ reagent for instance a carbodiimide based compound such as dicyclohexylcarbodiimide (DCC), 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide (or salt, e.g. hydrochloride, thereof) or N,N-diisopropylcarbodiimide (DIC) can be used.
  • DCC dicyclohexylcarbodiimide
  • 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide (or salt, e.g. hydrochloride, thereof) or N,N-diisopropylcarbodiimide (DIC) can be used.
  • the reaction may proceed at any suitable temperature (e.g. one between about 0° C.
  • Step q and r can be performed in a step-wise reaction under isolation of the intermediate XXVII or without isolation of XXVII.
  • the protecting group PG1 may be removed in accordance with techniques that are well known to those skilled in the art and as described hereinafter.
  • protected compounds/intermediates described herein may be converted chemically to unprotected compounds using HCl or H 2 SO 4 solutions, trifluoro acetic acid, KOH; Ba(OH) 2 , Pd on carbon, trimethylsilyl iodide or other conditions as described in “ Protective Groups in Organic Synthesis”, 3 rd edition, T. W. Greene & P. G. M. Wutz, Wiley-Interscience (1999).
  • Appropriate co-solvent for this step is for example dichloromethane, tetrahydrofuran (THF), acetonitrile, dimethylformamide (DMF), dimethylacetamide, N methylpyrrolidone, methanol, ethanol or mixtures of the above mentioned solvents.
  • an additional in-situ activating agent like 1-propylphosphonic acid cyclic anhydride, O-(benzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium tetrafluoroborate or hexafluorophosphate, N
  • the coupling reaction is preferably performed in the presence of a base such as NaOH, KOH, NaHCO 3 , triethylamine, N-ethyldiisopropylamine, N,N,-dimethylaminopyridine or other appropriate bases of the state of the art and for example described in described in Houben-Weyl, “Methods in Organic Synthesis”, Vol. E22a, p 425ff.
  • a base such as NaOH, KOH, NaHCO 3 , triethylamine, N-ethyldiisopropylamine, N,N,-dimethylaminopyridine or other appropriate bases of the state of the art and for example described in described in Houben-Weyl, “Methods in Organic Synthesis”, Vol. E22a, p 425ff.
  • the coupling of the amine XXVIII can be performed with an activated acid derivative A-C(O)-LG, where the leaving group LG can be for example, a fluorine, chlorine, bromine, azide or an isopropyloxy-C(O)—O anion or with A-SO 2 —Cl.
  • the coupling reaction is preferably performed in the presence of a base such as NaOH, KOH, NaHCO 3 , triethylamine, N-ethyldiisopropylamine, N,N,-dimethylaminopyridine or other appropriate bases under conditions which are for example described in Houben-Weyl, “Methods in Organic Synthesis”, Vol. E22a, p 425ff.
  • the above mentioned coupling reactions are performed in an appropriate solvent for example like dichloromethan, tetrahydrofurane (THF), acetonitrile, dimethylformamide (DMF), dimethylacetamide, N methylpyrrolidone or in mixtures of the above mentioned solvents.
  • an appropriate solvent for example like dichloromethan, tetrahydrofurane (THF), acetonitrile, dimethylformamide (DMF), dimethylacetamide, N methylpyrrolidone or in mixtures of the above mentioned solvents.
  • Step u) can be performed under standard conditions known to those skilled in the art in presence of a suitable solvent such as diethyl ether, dimethylformamide, dichloromethane, acetononitrile and/or tetrahydrofuran.
  • a suitable solvent such as diethyl ether, dimethylformamide, dichloromethane, acetononitrile and/or tetrahydrofuran.
  • the coupling reaction is preferably performed in the presence of a base such as NaOH, KOH, NaHCO 3 , triethylamine, N-ethyldiisopropylamine, 4-dimethylamino-pyridine or other appropriate bases.
  • Step v) is preferably performed in the presence of a suitable ‘coupling’ reagent.
  • ‘coupling’ reagent for instance a carbodiimide based compound such as dicyclohexylcarbodiimide (DCC), 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide (or salt, e.g. hydrochloride, thereof) or N,N-diisopropylcarbodiimide (DIC) can be used.
  • DCC dicyclohexylcarbodiimide
  • 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide (or salt, e.g. hydrochloride, thereof) or N,N-diisopropylcarbodiimide (DIC) can be used.
  • the reaction may proceed at any suitable temperature (e.g. one between about 0° C.
  • Step u and v can be performed in a step-wise reaction under isolation of the intermediate XXXI or without isolation of XXXI.
  • Step w) can be performed under known saponification methods for example with aqueous NaOH or KOH in ethanol, methanol or dioxane.
  • Step x) The coupling of the amine)(XXIV with the acid XXXIII can be performed with an additional in-situ activating agent like 1-propylphosphonic acid cyclic anhydride, O-(benzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium tetrafluoroborate or hexafluorophosphate, N,N′-dicyclohexylcarbodiimide, 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide, carbonyldiimidazole, oxalyl chloride or other activating agents of the state of the art.
  • an additional in-situ activating agent like 1-propylphosphonic acid cyclic anhydride, O-(benzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium tetrafluoroborate or hexafluorophosphate,
  • the coupling reaction is preferably performed in the presence of a base such as NaOH, KOH, NaHCO 3 , triethylamine, N-ethyldiisopropylamine, N,N,-dimethylaminopyridine or other appropriate bases of the state of the art and for example described in described in Houben-Weyl, “Methods in Organic Synthesis”, Vol. E22a, p 425ff.
  • a base such as NaOH, KOH, NaHCO 3 , triethylamine, N-ethyldiisopropylamine, N,N,-dimethylaminopyridine or other appropriate bases of the state of the art and for example described in described in Houben-Weyl, “Methods in Organic Synthesis”, Vol. E22a, p 425ff.
  • the above mentioned coupling reaction is performed in an appropriate solvent for example dichloromethane, tetrahydrofuran (THF), acetonitrile, dimethylformamide (DMF), dimethylacetamide, N methylpyrrolidone or in mixtures of the above mentioned solvents.
  • an appropriate solvent for example dichloromethane, tetrahydrofuran (THF), acetonitrile, dimethylformamide (DMF), dimethylacetamide, N methylpyrrolidone or in mixtures of the above mentioned solvents.
  • the protection and deprotection of functional groups may take place before or after a reaction in the above-mentioned schemes.
  • Protecting groups may be removed in accordance with techniques that are well known to those skilled in the art and as described hereinafter. For example, protected compounds/intermediates described herein may be converted chemically to unprotected compounds using standard deprotection techniques.
  • the basis of the assay used is to measure the inhibition of microsomal prostaglandin E 2 synthase-1 (mPGES-1) dependent prostaglandin (PG) E 2 formation from PGH 2 by different compounds.
  • mPGES-1 microsomal prostaglandin E 2 synthase-1
  • PG prostaglandin
  • Microsomes from Rosetta E. coli bacteria expressing recombinant human mPGES-1 can be derived as described below:
  • HTRF time resolved fluorescent
  • the extent of the specific HTRF is measured as a ratio of the emission intensity at 665 nm vs. that at 620 nm.
  • a standard curve using synthetic PGE 2 is used to quantify the amount of PGE 2 in unknown samples. The degree of inhibition can be calculated as percent inhibition by dividing the amount of PGE 2 formed in unknown samples by the amount of PGE 2 formed in control samples.
  • Table 1 demonstrates the mPGES-1 inhibitory effect (in %-inhibition) measured for selected compounds at a concentration of 10 ⁇ M (unless otherwise specified) in the HTRF assay as described above.
  • the examples show that 10 ⁇ M of the compound inhibit PGE 2 production to the indicated degree.
  • Table 2 demonstrates the mPGES-1 inhibitory effect (IC 50 measured for selected examples in the HTRF assay as described above. These data reflect a successful mPGES-1 inhibition for the given compound examples.
  • the present invention relates to compounds of formula I which are useful in the prevention and/or treatment of a disease and/or condition in which the inhibition of prostaglandin E synthases, in particular that of the microsomal prostaglandin E 2 synthase-1 (mPGES-1) is of therapeutic benefit, including but not limited to the treatment and/or prevention of inflammatory diseases and/or associated conditions.
  • mPGES-1 microsomal prostaglandin E 2 synthase-1
  • inflammation will be understood to include any inflammatory disease, disorder or condition per se, any condition that has an inflammatory component associated with it, and/or any condition characterised by inflammation as a symptom, including inter alia acute, chronic, ulcerative, specific, allergic and necrotic inflammation, and other forms of inflammation known to those skilled in the art.
  • the term thus also includes, for the purposes of this invention, inflammatory pain, pain generally and/or fever.
  • a condition has an inflammatory component associated with it, or a condition characterised by inflammation as a symptom
  • compounds of the invention may be useful in the treatment of the inflammatory symptoms and/or the inflammation associated with the condition.
  • Compounds of the invention may also have effects that are not linked to inflammatory mechanisms, such as in the reduction of bone loss in a subject.
  • Such conditions include osteoporosis, osteoarthritis, Paget's disease and/or periodontal diseases.
  • a further aspect of the present invention relates to a compound of formula I as a medicament.
  • Another aspect of the present invention is the use of compounds of formula I for the treatment and/or prevention of a disease and/or condition in which the inhibition of the mPGES-1 is of therapeutic benefit.
  • a further aspect of the present invention is the use of a compound of formula I for the treatment and/or prevention of inflammatory diseases and/or associated conditions.
  • the present invention also relates to the use of compounds of formula I for the treatment and/or prevention of the following diseases and conditions:
  • Rheumatic diseases or autoimmune diseases or muscoskeletal diseases all forms of rheumatic diseases including e.g. soft tissue rheumatism, rheumatoid arthritis, polymyalgia rheumatica, reactive arthritis, tenosynovitis, gout or metabolic arthritis, bursitis, tendonitis, juvenile arthritis, spondyloarthropathies like e.g. spondylitis, ankylosing spondylitis, psoriatric arthropathy; sarcoidosis, fibromyalgia, myositis, polymyositis, osteoarthritis, traumatic arthritis, collagenoses of any origin e.g.
  • Headaches such as migraines with and without aura, tension-type headaches, cluster headaches and headaches with different origins;
  • Neuropathic pain such as low back pain, hip pain, leg pain, non-herpetic neuralgia, post herpetic neuralgia, diabetic neuropathy, nerve injury-induced pain, acquired immune deficiency syndrome (AIDS) related neuropathic pain, head trauma, toxin and chemotherapy caused nerve injuries, phantom limb pain, multiple sclerosis, root avulsions, painful traumatic mononeuropathy, painful polyneuropathy, thalamic pain syndrome, post-stroke pain, central nervous system injury, post surgical pain, carpal tunnel syndrome, trigeminal neuralgia, post mastectomy syndrome, postthoracotomy syndrome, stump pain, repetitive motion pain, neuropathic pain associated hyperalgesia and allodynia, alcoholism and other drug-induced pain;
  • AIDS acquired immune deficiency syndrome
  • tumors such as bone tumors, lymphatic leukemia; Hodgkin's disease, malignant lymphoma; lymphogranulomatoses; lymphosarcoma; solid malignant tumors; extensive metastases
  • Visceral disorders such as chronic pelvic pain, pancreatitis, peptic ulcer, interstitial cystitis, cystitis, renal colic, angina, dysmenorrhoea, menstruation, gynaecological pain, irritable bowel disease (IBS), inflammatory bowel disease, Crohn's disease and ulcerative colitis, nephritis, prostatitis, vulvodynia, non-ulcer dyspepsia, non-cardiac chest pain, myocardial ischemia;
  • IBS irritable bowel disease
  • nephritis inflammatory bowel disease
  • Crohn's disease and ulcerative colitis nephritis
  • prostatitis vulvodynia
  • non-ulcer dyspepsia non-cardiac chest pain
  • myocardial ischemia myocardial ischemia
  • Inflammation associated diseases of ear, nose, mouth and throat like influenza and viral/bacterial infections such as the common cold, allergic rhinitis (seasonal and perennial), pharyngitis, tonsillitis, gingivitis, larhyngitis, sinusitis, and vasomotor rhinitis, fever, hay fever, thyroiditis, otitis, dental conditions like toothache, perioperative and post-operative conditions, trigeminal neuralgia, uveitis; ulceris, allergic keratitis, conjunctivitis, blepharitis, neuritis nervi optici, choroiditis, glaucoma and sympathetic opthalmia, as well as pain thereof.
  • influenza and viral/bacterial infections such as the common cold, allergic rhinitis (seasonal and perennial), pharyngitis, tonsillitis, gingivitis, larhyngitis, sinusitis, and vasomotor rhinitis, fever, hay fever, thyroiditis
  • Neurological diseases such as cerebral oedema and angioedema, cerebral dementia like e.g. Parkinson's and Alzheimers disease, senile dementia; multiple sclerosis, stroke, myasthenia gravis, brain and meningeal infections like encephalomyelitis, meningitis, including HIV as well as schizophrenia, delusional disorders, autism, affective disorders and tic disorders;
  • Lung diseases such as asthma including allergic asthma (atopic or non-atopic) as well as exercise-induced bronchoconstriction, occupational asthma, viral- or bacterial exacerbation of asthma, other non-allergic asthmas and “whez-infant syndrome”, Chronic obstructive pulmonary disease (COPD) including emphysema, adult respiratory distress syndrome, bronchitis, pneumonia, adult respiratory distress syndrome (ARDS), pigeon fancier's disease, farmers lung;
  • COPD chronic obstructive pulmonary disease
  • COPD chronic obstructive pulmonary disease
  • ARDS adult respiratory distress syndrome
  • pigeon fancier's disease farmers lung;
  • Skin diseases such as psoriasis and eczema, dermatitis, sunburn, burns as well as aprains and strains and tissue trauma;
  • Vascular and heart diseases which are inflammation-related like artheriosclerosis including cardiac transplant atherosclerosis, panarteritis nodosa, periarteritis nodosa, arteritis temporalis, Wegner granulomatosis, giant cell arthritis, reperfusion injury and erythema nodosum, thrombosis (e.g.
  • deep vein thrombosis renal, hepathic, portal vein thrombosis
  • coronary artery disease aneurysm, vascular rejection, myocardial infarction, embolism, stroke, thrombosis including venous thrombosis, angina including unstable angina, coronary plaque inflammation, bacterial-induced inflammation including Chlamydia -induced inflammation, viral induced inflammation, and inflammation associated with surgical procedures
  • vascular grafting including coronary artery bypass surgery, revascularization procedures including angioplasty, stent placement, endarterectomy, or other invasive procedures involving arteries, veins and capillaries, artery restenosis
  • vascular grafting including coronary artery bypass surgery, revascularization procedures including angioplasty, stent placement, endarterectomy, or other invasive procedures involving arteries, veins and capillaries, artery restenosis
  • revascularization procedures including angioplasty, stent placement, endarterectomy, or other invasive procedures involving
  • Diabetes-associated symptoms such as diabetic vasculopathy, diabetic neuropathy, diabetic retinopathy, post capillary resistance or diabetic symptoms associated with insulitis (e.g. hypergiycemia, diuresis, proteinuria and increased nitrite and kallikrein urinary excretion);
  • Benign and malignant tumors and neoplasia including cancer, such as colorectal cancer, brain cancer, bone cancer, epithelial cell-derived neoplasia (epithelial carcinoma) such as basal cell carcinoma, adenocarcinoma, gastrointestinal cancer such as lip cancer, mouth cancer, esophageal cancer, small bowel cancer, stomach cancer, colon cancer, liver cancer, bladder cancer, pancreas cancer, ovary cancer, cervical cancer, lung cancer, breast cancer, skin cancer such as squamous cell and basal cell cancers, prostate cancer, renal cell carcinoma, and other known cancers effecting epithelial cells throughout the body; neoplasias like gastrointestinal cancer, Barrett's esophagus, liver cancer, bladder cancer, pancreatic cancer, ovarian cancer, prostate cancer, cervical cancer, lung cancer, breast cancer and skin cancer; adenomatous polyps, including familial adenomatous polyposis (FAP) as well preventing polyps from forming in
  • septic shock e.g. as antihypovolemic and/or antihypotensive agents, sepsis, osteoporosis, benign prostatic hyperplasia and hyperactive bladder, nephritis, pruritis, vitiligo, disturbances of visceral motility at respiratory, genitourinary, gastrointestinal or vascular regions, wounds, allergic skin reactions, mixed-vascular and non-vascular syndromes, septic shock associated with bacterial infections or with trauma, central nervous system injury, tissue damage and postoperative fever, syndromes associated with itching.
  • septic shock e.g. as antihypovolemic and/or antihypotensive agents, sepsis, osteoporosis, benign prostatic hyperplasia and hyperactive bladder, nephritis, pruritis, vitiligo, disturbances of visceral motility at respiratory, genitourinary, gastrointestinal or vascular regions, wounds, allergic skin reactions, mixed-vascular and non-vascular syndrome
  • Preferred according to the present invention is the use of a compound of formula I for the treatment and/or prevention of pain; in particular pain that is associated with any one of the diseases or conditions listed above.
  • Another aspect of the present invention is a method for the treatment and/or prevention of above mentioned diseases and conditions, which method comprises the administration of an effective amount of a compound of formula Ito a human being.
  • Compounds of the invention may also have the advantage that they may be more efficacious than, be less toxic than, be longer acting than, be more potent than, produce fewer side effects than, be more easily absorbed than, and/or have a better pharmacokinetic profile (e.g. higher oral bioavailability and/or lower clearance) than, and/or have other useful pharmacological, physical, or chemical properties over, compounds known in the prior art, whether for use in the above-stated indications or otherwise.
  • pharmacokinetic profile e.g. higher oral bioavailability and/or lower clearance
  • the term “effective amount” refers to an amount of a compound, which confers a therapeutic effect on the treated patient.
  • the effect may be objective (i.e. measurable by some test or marker) or subjective (i.e. the subject gives an indication of or feels an effect).
  • Compounds of the invention may be administered at varying doses.
  • Oral, pulmonary and topical dosages may range from between about 0.01 mg/kg of body weight per day (mg/kg/day) to about 100 mg/kg/day, preferably about 0.01 to about 100 mg/kg/day, and more preferably about 0.1 to about 25 mg/kg/day.
  • the compositions typically contain between about 0.01 mg to about 5000 mg, and preferably between about 1 mg to about 2000 mg, of the active ingredient.
  • the most preferred doses will range from about 0.001 to about 10 mg/kg/hour during constant rate infusion.
  • compounds 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.
  • physician or the skilled person, will be able to determine the actual dosage which will be most suitable for an individual patient, which is likely to vary with the route of administration, the type and severity of the condition that is to be treated, as well as the species, age, weight, sex, renal function, hepatic function and response of the particular patient to be treated.
  • the above-mentioned dosages are exemplary of the average case; there can, of course, be individual instances where higher or lower dosage ranges are merited, and such are within the scope of this invention.
  • Suitable preparations for administering the compounds of formula will be apparent to those with ordinary skill in the art and include for example tablets, pills, capsules, suppositories, lozenges, troches, solutions, syrups, elixirs, sachets, injectables, inhalatives and powders etc. Such formulations may be prepared in accordance with standard and/or accepted pharmaceutical practice.
  • the content of the pharmaceutically active compound(s) should be in the range from 1 to 99 wt.-%, preferably 10 to 90 wt.-%, more preferably 20 to 70 wt.-%, of the composition as a whole.
  • Suitable tablets may be obtained, for example, by mixing one or more compounds according to formula I with known excipients, for example inert diluents, carriers, disintegrants, adjuvants, surfactants, binders and/or lubricants.
  • excipients for example inert diluents, carriers, disintegrants, adjuvants, surfactants, binders and/or lubricants.
  • the tablets may also consist of several layers.
  • a further aspect of the invention is a pharmaceutical formulation including a compound of formula I in admixture with a pharmaceutically acceptable adjuvant, diluent or carrier.
  • the compounds according to the present invention can be combined with other treatment options known to be used in the art in connection with a treatment of any of the indications the treatment of which is in the focus of the present invention.
  • treatment options that are considered suitable for combination with the treatment according to the present inventions are:
  • Combination therapy is also possible with new principles for the treatment of pain e.g. P2X3 antagonists, VR1 antagonists, NK1 and NK2 antagonists, NMDA antagonists, mGluR antagonists and the like.
  • the combination of compounds is preferably a synergistic combination.
  • Synergy as described for example by Chou and Talalay, Adv. Enzyme Regul. 22:27-55 (1984), occurs when the effect of the compounds when administered in combination is greater than the additive effect of the compounds when administered alone as a single agent. In general, a synergistic effect is most clearly demonstrated at suboptimal concentrations of the compounds. Synergy can be in terms of lower cytotoxicity, increased pharmacological effect, or some other beneficial effect of the combination compared with the individual components.
  • the TLC data was obtained by using the following tic plates
  • the diode array detection took place in a wavelength range from 210-550 nm
  • Range of mass-spectrometric detection m/z 120 to m/z 1000
  • N-[1-(3-Amino-4-chlorophenyl)ethyl]acetamide (90 mg, 0.2 mmol) was added to a mixture of 1,1′-thiocarbonyldi-2-pyridone (69 mg, 0.2 mmol), DMAP (7 mg, 0.05 mmol) and DCM (2 mL) at rt. The mixture was concentrated and used in the next step without further purification.
  • N,N-Diisopropylcarbodiimide (46 ⁇ L, 0.2 mmol) was added to 3- ⁇ 3-[5-(1-acetamidoethyl)-2-chlorophenyl]thioureido ⁇ -N-(4-bromophenyl)-4-(methylamino)benz-amide (130 mg, 0.2 mmol) in DMF (2 mL) at rt. The mixture was stirred at 80° C. for 2 h, cooled and concentrated. The residue was purified by chromatography to give the title compound. Yield: 30 mg (24%). MS m/z: 540 [M+H] + .
  • the sub-title compound was prepared from tert-butyl 3-amino-4-chlorobenzyl-carbamate, in accordance with Example 1, step (d).
  • the sub-title compound was prepared in two steps from tert-Butyl-4-chloro-3-isothiocyanatobenzylcarbamate and Ethyl-2-chloro-4,5-diamino-benzoic acid in accordance with Example 1, step (g) with additional TEA and Example 1, step (h).
  • the sub-title compound was prepared from 2-chloro-N-cyclopentyl-4-methylamino-5-nitrobenzamide in accordance with Example 2, step (b).
  • the sub-title compound was prepared from N-(4-Chloro-3-aminobenzyl)-1-methylcyclohexylamide analogous to Example 1, step (d) but with 1,1′-thiocarbonyl-diimidazole instead of 1,1′-thiocarbonyldi-2-pyridone.
  • the title compound was prepared from N-(4-chloro-3-isothiocyanatobenzyl)-1-methylcyclohexylamide and 5-amino-2-chloro-N-cyclopentyl-4-methylaminobenzamide analogous to Example 1, steps (g and h), but without isolation of the thioureido derivative and with EDC as coupling reagent instead of N,N-diisopropylcarbodiimide.
  • the sub-title compound was prepared from di-tert-butyl-2,5-dichloro-4-nitro-phenylimidodicarbonate in accordance with Example 3, step (b).
  • the sub-title compound was prepared from tert-butyl 2-chloro-5-(methylamino)-4-nitrophenylcarbamate in accordance with Example 2, step (b).
  • the title compound was prepared from N-(4-chloro-3-isothiocyanatobenzyl)-1-methylcyclohexylamide and tert-butyl 4-amino-2-chloro-5-(methylamino)phenylcarbamate) in accordance to the procedures described above.
  • the sub-title compound was prepared from 4-chloro-N-cyclopentyl-3-nitro-benzenesulfonamide and methylamine in accordance with Example 3, step (b).
  • the sub-title compound was prepared from N-cyclopentyl-4-methylamino-3-nitro-benzenesulfonamide in accordanced with Example 2, step (b).
  • the sub-title compound was prepared from tert-butyl 4-chloro-3-isothiocyanato-benzylcarbamate (see Example 2d) and methyl 3-amino-4-methylaminobenzoate according to Example 3, step (g).
  • the sub-title compound was prepared from methyl 2- ⁇ 5-[(tert-butoxycarbonyl-amino)methyl]-2-chlorophenylamino ⁇ -1-methylbenzimidazole-5-carboxylate in accordance with the procedure in Example 10, step (a).
  • the sub-title compound was prepared from methyl 2-[5-(aminomethyl)-2-chloro-phenylamino]-1-methylbenzo[d]imidazole-5-carboxylate, and 1-trifluoromethylcyclopropylcarboxylic acid and TBTU in accordance with the procedure in Example 3, step (e).
  • the title compound was prepared from 2-(2-chloro-5- ⁇ [(1-trifluoromethyl-cyclopropanecarbonyl)-amino]-methyl ⁇ -phenylamino)-6-methoxy-1-methyl-1H-benzimidazole-5-carboxylic acid (92 mg; 0.19 mmol) and 4-bromoaniline (33 mg; 0.19 mmol) in accordance with Example 59, step (a). Yield: 73 mg (59%).
  • the sub-title compound was prepared in accordance with the procedure in Example 72, step (c) using (4-fluoro-3-nitrophenyl)methylamine hydrochloride (3.0 g; 14.5 mmol), 1-(trifluoromethyl)-cyclopropanecarboxylic acid (2.5 g; 16.0 mmol), HBTU (6.07 g; 16.0 mmol), TEA (5.88 g; 58.1 mmol) and DMF (50 mL). Yield: 3.8 g (85%).
  • the sub-title compound was prepared in accordance with the procedure in Example 2, step (b) using N-(4-fluoro-3-nitrobenzyl)-1-(trifluoromethyl)-cyclopropanecarboxamide (3.77 g; 12.3 mmol), Fe (3.45 g; 61.6 mmol), NH 4 Cl (aq, sat, 30 mL) and EtOH (30 mL). Yield: 3.2 g (76%).
  • the sub-title compound was prepared in accordance with Example 72, step (b) using ethyl 2-(2,2-difluoroethoxy)-4-(methylamino)-5-nitrobenzoate ((950 mg; 3.12 mmol) see Example 59, step (f)), NaOH (3.1 mL; 2 M aq; 6.2 mmol) and 1,4-dioxane (10 mL). Yield: 800 mg (93%).
  • the sub-title compound was prepared in accordance with Example 72, step (c) using 2-(2,2-difluoroethoxy)-4-(methylamino)-5-nitrobenzoic acid (800 mg; 2.90 mmol), 4-bromoaniline (499 mg; 2.90 mmol), HBTU (1.10 g; 2.90 mmol), TEA (587 mg; 5.80 mmol) and DMF (15 mL). Yield: 1.01 g (81%).
  • the sub-title compound was prepared by hydrogenation according to procedure 3d using N-(4-bromophenyl)-2-(2,2-difluoroethoxy)-4-(methylamino)-5-nitrobenzamide (1.01 g; 2.34 mmol), Ra—Ni (14 mg), H 2 (8 atm) and THF (50 mL). Yield: 905 mg (97%).
  • the sub-title compound was prepared in accordance with Example 59 (f) using ethyl 4-amino-2-fluoro-5-nitrobenzoate (4.80 g; 21.0 mmol), 2,2-difluoroethanol (1.73 g; 21.0 mmol), sodium hydride (0.841 g; 60%; 21.0 mmol), THF (100 mL) and DMF (50 mL). Yield: 2.40 g (39%).
  • the sub-title compound was prepared in accordance with Example 59 (g) using ethyl 4-amino-2-(2,2-difluoroethoxy)-5-nitrobenzoate (1.00 g; 3.44 mmol), Ra—Ni (1.0 g) and THF (50 mL). The sub-title compound was used in the next step without further purification.
  • the sub-title compound was prepared in accordance with Example 59 (h) using ethyl 4,5-diamino-2-(2,2-difluoroethoxy)benzoate (0.466 g; 1.79 mmol; crude material from step (c)), N-(4-chloro-3-isothiocyanatobenzyl)-1-(trifluoromethyl)cyclopropane carboxamide (0.600 g; 1.79 mmol), DIC (0.226 g; 1.79 mmol) and THF (40 mL). Yield: 0.620 g (62%).
  • the sub-title compound was prepared in accordance with the procedure in Example 59, step (a) using 4-amino-2-methoxy-5-nitrobenzoic acid (2.0 g; 9.43 mmol), TBTU (3.33 g; 10.4 mmol), TEA (2.9 mL; 20.8 mmol), 4-bromoaniline (1.62 g; 9.43 mmol) and DMF (70 mL). Yield: 3.41 g (99%).
  • the sub-title compound was prepared in accordance with the procedure in Example 2, step (b) using N-(4-bromophenyl)-4-(2-(dimethylamino)ethylamino)-3-nitrobenzamide (560 mg; 1.38 mmol), Fe (384 mg; 6.88 mmol), NH 4 Cl (aq, sat, 10 mL) and EtOH (10 mL). Yield: 475 mg (91%).
  • the sub-title compound was prepared in accordance with the procedure in Example 59, step (g) using tert-butyl N-[5-cyano-2-(trifluoromethyl)phenyl]-carbamate (8.5 g; 29.7 mmol), Ra—Ni (174 mg; 2.97 mmol), H 2 (5 atm) and THF (60 mL). Yield: 8 g (93%).
  • the sub-title compound was prepared in accordance with the procedures in Example 72, step (c) and Example 72, step (d) using tert-butyl N-[5-(aminomethyl)-2-(trifluoromethyl)phenyl]-carbamate (2.09 g; 7.20 mmol), 1-(trifluoromethyl)-cyclopropanecarboxylic acid (1.11 g; 7.20 mmol), HBTU (2.73 g; 7.20 mmol), TEA (2.19 g; 21.6 mmol), DMF (25 mL), TFA (10 mL) and DCM (50 mL). Yield: 1.8 g (77%).
  • the sub-title compound was prepared in accordance with the procedure in Example 72, step (a) using 4-chloro-2-fluoro-5-isothiocyanatobenzonitrile (270 mg; 1.27 mmol), 5-amino-2-(2,2-difluoroethoxy)-4-(methylamino)-N-((trans)-4-(trifluoromethyl)cyclohexyl)benzamide [502 mg; 1.27 mmol, prepared from 4-trans-trifluoromethyl-cyclohexylamine and 2-(2,2-difluoroethoxy)-4-(methylamino)-5-nitrobenzoic acid in analogy to Example 66, step (f+g)], DIC (160 mg; 1.27 mmol) and DMF (10 mL). Yield: 210 mg (29%).
  • the sub-title compound was prepared in accordance with Example 3d, using 2-[(2-chloro-5-cyano-4-fluoro-phenyl)-amino]-6-(2,2-difluoroethoxy)-1-methyl-N-(trans-4-trifluoromethyl-cyclohex-1-yl)-1H-benzo[d]imidazole-5-carboxamide (210 mg; 0.37 mmol), Ra—Ni (3 mg), H 2 (5 atm) and THF (10 mL). Yield: 150 mg (70%).
  • KOtBu (0.26 g, 95%, 2.39 mmol) was added to a mixture of 2,2-difluoroethanol (0.16 ml, 2.48 mmol) and 10 ml THF. It was stirred for 15 min, ethyl 2-fluoro-4-methylamino-5-nitro-benzoate (0.40 g, 1.65 mmol) was added and it was stirred for 3 d. The mixture was diluted with water, concentrated i.vac. and the crude product was collected by filtration.
  • the sub-title compound was prepared from ethyl 2-(2,2-difluoroethoxy)-4-methylamino-5-amino-benzoate and N-(4-chloro-3-isothiocyanatobenzyl)-2,2-dimethylpropionamide with DIC in DMF at r.t. in analogy to example 72a.
  • the crude material was directly used in the next step.
  • the title compound was prepared from 2-(2-chloro-5- ⁇ [(tert.butylcarbonyl)amino]methyl ⁇ -phenylamino)-6-(2,2-difluoroethoxy)-1-methyl-1H-benzo[d]imidazole-5-carboxylic acid and 3-chloro-4-fluoro-aniline with HATU and TEA in THF in analogy to example 3e and purified via flash chromatography.
  • the sub-title compound was prepared from crude 3-nitro-4-trifluoromethyl-benzylamine with di-tertbutyl-dicarbonate in analogy to example 2a.
  • the sub-title compound was prepared from tert.-butyl-N-(3-nitro-4-trifluoromethyl-benzyl)carbamate with powdered iron in analogy to example 2b.
  • the sub-title compound was prepared from tert.-butyl-N-(3-amino-4-trifluoromethyl-benzyl)carbamate with 1,1′-thiocarbonyldi-2-pyridone in analogy to example 1d.
  • the sub-title compound was prepared from N-(trans-4-trifluoromethyl-cyclohex-1-yl)-2-methoxy-4-amino-5-nitro benzoic acid amide with Ra—Ni in analogy to example 59 g.
  • the title compound was prepared from 2-( ⁇ 5-[(tert.butoxycarbonyl)amino]methyl ⁇ -2-trifluoromethyl-phenylamino)-6-methoxy-N-(trans-4-trifluoromethyl-cyclohex-1-yl)-1H-benzo[d]imidazole-5-carboxamide and 6M HCl in THF in analogy to example 2e.
  • the title compound was prepared from 2-(5-aminomethyl-2-trifluoromethyl-phenylamino)-6-methoxy-N-(trans-4-trifluoromethyl-cyclohex-1-yl)-1H-benzo[d]imidazole-5-carboxamide and 2-tert.butoxycarbonylamino-3,3,3-trifluoro-propionic acid with TBTU, and TEA in THF in analogy to example 3e.
  • the title compound was prepared from 2-(5- ⁇ [(2-tert.Butoxycarbonylamino-3,3,3-trifluoro-propionyl)amino]methyl ⁇ -2-trifluoromethyl-phenylamino)-6-methoxy-N-(trans-4-trifluoromethyl-cyclohex-1-yl)-1H-benzo[d]imidazole-5-carboxamide and 6M HCl in THF in analogy to example 2e.
  • the title compound was prepared from tert.-butyl-N-(3-amino-4-trifluoromethyl-benzyl)carbamate and 6M HCl in THF in analogy to example 2e.
  • the sub-title compound was prepared from 3-amino-4-trifluoromethyl-benzylamine and pivaloylchloride with TEA in analogy to example 1b.
  • the sub-title compound was prepared from N-(3-amino-4-trifluoromethyl-benzyl)-pivaloyl amide with 1,1′-thiocarbonyldi-2-pyridone in analogy to example 1d.
  • the sub-title compound was prepared from 4-trans-trifluoromethyl-cyclohexylamine and 2-fluoro-4-(methylamino)-5-nitrobenzoic acid with DIPEA and TBTU in analogy to example 3e.
  • the sub-title compound was prepared from 5-nitro-2-fluoro-4-(methylamino)-N-(trans-4-trifluoromethyl.cyclohex-1-yl)-benzamide and 2,2-difluoroethanol with KOtBu in analogy to example 86b.
  • the sub-title compound was prepared from 5-nitro-2-(2,2-difluoroethoxy)-4-(methylamino)-N-(trans-4-trifluoromethyl-cyclohex-1-yl)-benzamide, hydrogen and Pd/C in analogy to example 86c.
  • the sub-title compound was prepared from 3-amino-4-methoxy-benzylamine and pivaloylchloride with TEA in analogy to example 1b. Yield: 30%; MS m/z: 237 [M+H]+.
  • the sub-title compound was prepared from N-(3-amino-4-methoxy-benzyl)-pivaloyl amide with 1,1′-thiocarbonyldi-2-pyridone in analogy to example 1d.
  • the sub-title compound was prepared from 2-chloro-4-fluoro-5-nitrobenzoic acid—which was converted into the corresponding acid chloride with thionyl chloride—and 4-trans-trifluoromethyl-cyclohexylamine according to example 70a.
  • the compound was prepared from 4,5-diamino-2-chloro-N-(trans-4-trifluoromethyl-cyclohex-1-yl)-benzamide and N-(3-isothiocyanato-4-methoxy-benzyl)-pivaloylamide in analogy to example 72a.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pain & Pain Management (AREA)
  • Biomedical Technology (AREA)
  • Neurosurgery (AREA)
  • Neurology (AREA)
  • Rheumatology (AREA)
  • Pulmonology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Macromolecular Compounds Obtained By Forming Nitrogen-Containing Linkages In General (AREA)
  • Plural Heterocyclic Compounds (AREA)
US12/717,407 2009-03-05 2010-03-04 New compounds Abandoned US20100256188A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US13/441,085 US20120196897A1 (en) 2009-03-05 2012-04-06 New compounds
US13/942,077 US8921405B2 (en) 2009-03-05 2013-07-15 Compounds

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
EP09154414.8 2009-03-05
EP09154414 2009-03-05
EP09179618 2009-12-17
EP09179618.5 2009-12-17

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US13/441,085 Continuation US20120196897A1 (en) 2009-03-05 2012-04-06 New compounds

Publications (1)

Publication Number Publication Date
US20100256188A1 true US20100256188A1 (en) 2010-10-07

Family

ID=42111729

Family Applications (3)

Application Number Title Priority Date Filing Date
US12/717,407 Abandoned US20100256188A1 (en) 2009-03-05 2010-03-04 New compounds
US13/441,085 Abandoned US20120196897A1 (en) 2009-03-05 2012-04-06 New compounds
US13/942,077 Active US8921405B2 (en) 2009-03-05 2013-07-15 Compounds

Family Applications After (2)

Application Number Title Priority Date Filing Date
US13/441,085 Abandoned US20120196897A1 (en) 2009-03-05 2012-04-06 New compounds
US13/942,077 Active US8921405B2 (en) 2009-03-05 2013-07-15 Compounds

Country Status (37)

Country Link
US (3) US20100256188A1 (es)
EP (1) EP2403852B8 (es)
JP (1) JP5433708B2 (es)
KR (1) KR20110123764A (es)
CN (1) CN102341395B (es)
AP (1) AP2806A (es)
AR (1) AR075789A1 (es)
AU (1) AU2010220263B2 (es)
BR (1) BRPI1009118A2 (es)
CA (1) CA2753149A1 (es)
CL (1) CL2011002180A1 (es)
CO (1) CO6450668A2 (es)
CY (1) CY1115959T1 (es)
DK (1) DK2403852T3 (es)
EA (1) EA020282B1 (es)
EC (1) ECSP11011324A (es)
ES (1) ES2528730T3 (es)
GE (1) GEP20146034B (es)
HK (1) HK1163064A1 (es)
HR (1) HRP20150096T1 (es)
IL (1) IL214033A0 (es)
MA (1) MA33239B1 (es)
ME (1) ME02117B (es)
MX (1) MX2011008948A (es)
MY (1) MY161092A (es)
NZ (1) NZ594589A (es)
PE (1) PE20120589A1 (es)
PL (1) PL2403852T3 (es)
PT (1) PT2403852E (es)
RS (1) RS53757B1 (es)
SG (1) SG174250A1 (es)
SI (1) SI2403852T1 (es)
SM (1) SMT201500053B (es)
TN (1) TN2011000446A1 (es)
TW (1) TWI466869B (es)
UY (1) UY32470A (es)
WO (1) WO2010100249A1 (es)

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20120309738A1 (en) * 2010-12-10 2012-12-06 Boehringer Ingelheim International Gmbh Novel compounds
US20120309755A1 (en) * 2010-12-10 2012-12-06 Boehringer Ingelheim International Gmbh Novel compounds
US8586604B2 (en) 2010-08-20 2013-11-19 Boehringer Ingelheim International Gmbh Inhibitors of the microsomal prostaglandin E2 synthase-1
US8598190B2 (en) 2008-09-25 2013-12-03 Boehringer Ingelheim International Gmbh 3H-imidazo [4, 5-C] pyridine-6-carboxamides as anti-inflammatory agents
US8674113B2 (en) 2010-12-10 2014-03-18 Boehringer Ingelheim International Gmbh Compounds
US8759537B2 (en) 2010-08-20 2014-06-24 Boehringer Ingelheim International Gmbh 3H-imidazo [4, 5-C] pyridine-6-carboxamides as anti-inflammatory agents
US8921405B2 (en) 2009-03-05 2014-12-30 Orexo Ab Compounds
US9962375B2 (en) * 2014-10-29 2018-05-08 Eli Lilly And Company Methyl-piperidine compounds useful for inhibiting microsomal prostaglandin E2 synthase-1
US9969714B2 (en) 2014-10-29 2018-05-15 Eli Lilly And Company Carboxylic acid compounds useful for inhibiting microsomal prostaglandin E2 synthase-1
WO2022251384A1 (en) * 2021-05-25 2022-12-01 Louis Habash Modifying the expression level of a gene encoding an cyclooxygenase enzyme by treating a human subject with a nitroxide
US11691963B2 (en) 2020-05-06 2023-07-04 Ajax Therapeutics, Inc. 6-heteroaryloxy benzimidazoles and azabenzimidazoles as JAK2 inhibitors
US11970494B2 (en) 2021-11-09 2024-04-30 Ajax Therapeutics, Inc. 6-heteroaryloxy benzimidazoles and azabenzimidazoles as JAK2 inhibitors
US11993613B2 (en) 2023-03-30 2024-05-28 Abbvie Inc. Thiazolo[5,4-b]pyridine MALT-1 inhibitors

Families Citing this family (53)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TW201305178A (zh) 2010-10-29 2013-02-01 Glenmark Pharmaceuticals Sa 作為mPGES-1抑制物的三環化合物
AR084174A1 (es) 2010-12-21 2013-04-24 Lilly Co Eli Compuestos de imidazol-2-benzamida utiles para el tratamiento de osteoartritis y una composicion farmaceutica
AR086254A1 (es) 2011-05-26 2013-11-27 Lilly Co Eli Derivados de imidazol utiles para el tratamiento de artritis
WO2013038308A1 (en) 2011-09-15 2013-03-21 Glenmark Pharmaceuticals S.A. SUBSTITUTED BICYCLIC HETEROARYL COMPOUNDS AS mPGES-1 INHIBITORS
PL2766359T3 (pl) * 2011-10-14 2016-11-30 Związki heterocykliczne i ich zastosowanie jako modulatorów receptorowych kinaz tyrozynowych typu iii
WO2013072825A1 (en) 2011-11-16 2013-05-23 Glenmark Pharmaceuticals S.A. Phtalazinone derivatives as mpegs -1 inhibitors
WO2013118071A1 (en) 2012-02-09 2013-08-15 Glenmark Pharmaceuticals S.A. BICYCLIC COMPOUNDS AS mPGES-1 INHIBITORS
WO2013152039A1 (en) 2012-04-02 2013-10-10 The Trustees Of Columbia University In The City Of New York Compounds, compositions, and methods for modulating ferroptosis and treating excitotoxic disorders
WO2013153535A1 (en) 2012-04-13 2013-10-17 Glenmark Pharmaceuticals S.A. TRICYCLIC COMPOUNDS AS mPGES-1 INHIBITORS
CN103450329B (zh) * 2012-05-29 2015-05-27 首都医科大学 3h-咪唑并吡啶-6-甲酰氨基酸苄酯、其合成、抗肿瘤活性和应用
TWI568722B (zh) 2012-06-15 2017-02-01 葛蘭馬克製藥公司 作爲mPGES-1抑制劑之三唑酮化合物
WO2014167444A1 (en) 2013-04-08 2014-10-16 Glenmark Pharmaceuticals S.A. SUBSTITUTED BICYCLIC COMPOUNDS AS mPGES-1 INHIBITORS
JP2016522252A (ja) * 2013-06-20 2016-07-28 ノヴァサイド・アクチボラグ Mpges−1阻害剤としての新規なピペリジニルベンゾイミダゾール誘導体
WO2015059618A1 (en) 2013-10-22 2015-04-30 Glenmark Pharmaceuticals S.A. SUBSTITUTED PYRIMIDINE COMPOUNDS AS mPGES-1 INHIBITORS
US9951027B2 (en) 2014-02-11 2018-04-24 Bayer Pharma Aktiengesellschaft Benzimidazol-2-amines as MIDH1 inhibitors
US9957235B2 (en) 2014-02-11 2018-05-01 Bayer Pharma Aktiengesellschaft Benzimidazol-2-amines as mIDH1 inhibitors
CN105294829B (zh) * 2014-06-13 2018-07-27 首都医科大学 咪唑并吡啶-6-甲酰-氨基酸苄酯,其合成,活性及应用
CN105198960B (zh) * 2014-06-13 2018-10-19 首都医科大学 咪唑并吡啶-6-甲酰-Met-AA-OBzl,其合成,活性和应用
CN105198959B (zh) * 2014-06-13 2018-09-07 首都医科大学 咪唑并吡啶-6-甲酰-Met-Glu(OBzl)2,其合成,活性和应用
CN105315326A (zh) * 2014-07-10 2016-02-10 首都医科大学 咪唑并吡啶-6-甲酰-氨基酸苄酯,合成,活性和应用
CN105315325A (zh) * 2014-07-10 2016-02-10 首都医科大学 咪唑并吡啶-6-甲酰-Met-Arg(NO2)-OBzl,其合成,活性和应用
CN105315335A (zh) * 2014-07-10 2016-02-10 首都医科大学 咪唑并吡啶-6-甲酰-Met-Gln-OBzl,其合成,活性和应用
HUE044602T2 (hu) 2014-08-01 2019-11-28 Glenmark Pharmaceuticals Sa Nanorészecskés készítmény, amely egy MPGES-1 inhibitorot tartalmaz
CN107108522B (zh) 2014-10-23 2020-12-01 德国癌症研究中心 作为mIDH1抑制剂的苯并咪唑-2-胺
CA2965213A1 (en) 2014-10-23 2016-04-28 Bayer Pharma Aktiengesellschaft 1-cyclohexyl-2-phenylaminobenzimidazoles as midh1 inhibitors for the treatment of tumors
CA2988356A1 (en) 2015-06-08 2016-12-15 Bayer Pharma Aktiengesellschaft N-menthylbenzimidazoles as midh1 inhibitors
US10414734B2 (en) 2015-07-16 2019-09-17 Bayer Pharma Aktiengesellschaft 5-hydroxyalkylbenzimidazoles as mIDH1 inhibitors
GB201603311D0 (en) * 2016-02-25 2016-04-13 Jakobsson Per Johan New uses and methods
KR20200090818A (ko) 2017-11-22 2020-07-29 콘드리온 아이피 비.브이. Mpges-1 억제제로서의 신규 화합물
PE20220231A1 (es) 2019-06-25 2022-02-07 Gilead Sciences Inc Proteinas de fusion flt3l-fc y metodos de uso
EP4023638A4 (en) * 2019-08-26 2023-10-04 Kukje Pharma Co., Ltd. INDOLE CARBOXAMIDE DERIVATIVE AND PHARMACEUTICAL COMPOSITION CONTAINING SAME
JP7371243B2 (ja) 2019-10-18 2023-10-30 フォーティ セブン, インコーポレイテッド 骨髄異形成症候群及び急性骨髄性白血病を治療するための併用療法
CN114599392A (zh) 2019-10-31 2022-06-07 四十七公司 基于抗cd47和抗cd20的血癌治疗
TWI778443B (zh) 2019-11-12 2022-09-21 美商基利科學股份有限公司 Mcl1抑制劑
IL294032A (en) 2019-12-24 2022-08-01 Carna Biosciences Inc Compounds that regulate diacylglycerol kinase
CA3169451A1 (en) 2020-02-14 2021-08-19 Jounce Therapeutics, Inc. Antibodies and fusion proteins that bind to ccr8 and uses thereof
EP4143194A1 (en) 2020-05-01 2023-03-08 Gilead Sciences, Inc. Cd73 inhibiting 2,4-dioxopyrimidine compounds
TW202302145A (zh) 2021-04-14 2023-01-16 美商基利科學股份有限公司 CD47/SIRPα結合及NEDD8活化酶E1調節次單元之共抑制以用於治療癌症
TW202313094A (zh) 2021-05-18 2023-04-01 美商基利科學股份有限公司 使用FLT3L—Fc融合蛋白之方法
WO2022271677A1 (en) 2021-06-23 2022-12-29 Gilead Sciences, Inc. Diacylglyercol kinase modulating compounds
AU2022299051A1 (en) 2021-06-23 2023-12-07 Gilead Sciences, Inc. Diacylglyercol kinase modulating compounds
WO2022271659A1 (en) 2021-06-23 2022-12-29 Gilead Sciences, Inc. Diacylglyercol kinase modulating compounds
US11926628B2 (en) 2021-06-23 2024-03-12 Gilead Sciences, Inc. Diacylglyercol kinase modulating compounds
AU2022376954A1 (en) 2021-10-29 2024-05-02 Gilead Sciences, Inc. Cd73 compounds
US20230242508A1 (en) 2021-12-22 2023-08-03 Gilead Sciences, Inc. Ikaros zinc finger family degraders and uses thereof
WO2023122581A2 (en) 2021-12-22 2023-06-29 Gilead Sciences, Inc. Ikaros zinc finger family degraders and uses thereof
TW202340168A (zh) 2022-01-28 2023-10-16 美商基利科學股份有限公司 Parp7抑制劑
EP4245756A1 (en) 2022-03-17 2023-09-20 Gilead Sciences, Inc. Ikaros zinc finger family degraders and uses thereof
US20230355796A1 (en) 2022-03-24 2023-11-09 Gilead Sciences, Inc. Combination therapy for treating trop-2 expressing cancers
TW202345901A (zh) 2022-04-05 2023-12-01 美商基利科學股份有限公司 用於治療結腸直腸癌之組合療法
TW202400138A (zh) 2022-04-21 2024-01-01 美商基利科學股份有限公司 Kras g12d調節化合物
WO2024006929A1 (en) 2022-07-01 2024-01-04 Gilead Sciences, Inc. Cd73 compounds
US20240091351A1 (en) 2022-09-21 2024-03-21 Gilead Sciences, Inc. FOCAL IONIZING RADIATION AND CD47/SIRPa DISRUPTION ANTICANCER COMBINATION THERAPY

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6608084B1 (en) * 1998-08-26 2003-08-19 Aventis Pharma Ltd. Aza-bicycles which modulate the inhibition of cell adhesion
US20040198768A1 (en) * 2003-04-03 2004-10-07 Park Choo Hea Young Method for inhibiting 5-lipoxygenase using a benzoxazole derivative or an analogue thereof
US20050267147A1 (en) * 2003-02-26 2005-12-01 Lydie Poitout Novel benzimidazole and imidazopyridine derivatives and use thereof as a medicament
US20060173036A1 (en) * 2003-03-31 2006-08-03 Lydie Poitout Imidazopyridine derivatives as melanocortin receptor agonists
US20060287344A1 (en) * 2005-01-13 2006-12-21 Albers Ronald J Haloaryl substituted aminopurines, compositions thereof, and methods of treatment therewith
US20070060598A1 (en) * 2005-01-13 2007-03-15 Albers Ronald J Haloaryl substituted aminopurines, compositions thereof, and methods of treatment therewith
US20100004301A1 (en) * 2006-12-14 2010-01-07 Benjamin Pelcman Benzoxazoles Useful in the Treatment of Inflammation

Family Cites Families (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE3006671A1 (de) 1980-02-22 1981-08-27 Dr. Karl Thomae Gmbh, 7950 Biberach Neue benzoxazole, deren herstellung und deren verwendung als arzneumittel
FI91859C (fi) 1987-06-17 1994-08-25 Eisai Co Ltd Analogiamenetelmä antiallergisena aineena aktiivisen bentsotiatsolijohdannaisen valmistamiseksi
JPH06759B2 (ja) 1989-09-22 1994-01-05 ファイザー製薬株式会社 新規なベンゾイミダゾール化合物
PT1153017E (pt) * 1999-02-16 2006-07-31 Aventis Pharma Ltd Compostos biciclicos e sua utilizacao como ligandos do receptor de integrina
KR20010110750A (ko) * 1999-04-12 2001-12-13 앤드류 앵뉴 인테그린 길항제로서의 치환된 비사이클릭 헤테로아릴화합물
CA2372840C (en) * 1999-05-05 2008-07-22 Aventis Pharma Limited Substituted bicyclic compounds
US6340681B1 (en) 1999-07-16 2002-01-22 Pfizer Inc 2-benzimidazolylamine compounds as ORL-1-receptor agonists
EP1222187B1 (en) * 1999-10-06 2004-09-22 Boehringer Ingelheim Pharmaceuticals Inc. Heterocyclic compounds useful as inhibitors of tyrosine kinases
FR2833948B1 (fr) * 2001-12-21 2004-02-06 Sod Conseils Rech Applic Nouveaux derives de benzimidazole et leur utilisation en tant que medicament
MXPA04008458A (es) 2002-03-01 2004-12-06 Smithkline Beecham Corp Diamino pirimidinas y su uso como inhibidores de angiogenesis.
EA007987B1 (ru) 2002-03-29 2007-02-27 Чирон Корпорейшн Замещённые бензазолы и их применение в качестве ингибиторов киназы raf
US7037902B2 (en) 2002-07-03 2006-05-02 Receptron, Inc. Affinity small molecules for the EPO receptor
US7122665B2 (en) 2002-10-15 2006-10-17 Synta Pharmaceuticals Corp. Heterocyclic compounds
US7265138B2 (en) 2003-02-10 2007-09-04 Amgen Inc. Vanilloid receptor ligands and their use in treatments
US7531553B2 (en) 2003-03-21 2009-05-12 Amgen Inc. Heterocyclic compounds and methods of use
JP4842829B2 (ja) 2003-10-31 2011-12-21 武田薬品工業株式会社 含窒素縮合複素環化合物
GB0401334D0 (en) * 2004-01-21 2004-02-25 Novartis Ag Organic compounds
US7470712B2 (en) * 2004-01-21 2008-12-30 Bristol-Myers Squibb Company Amino-benzazoles as P2Y1 receptor inhibitors
JP2008502670A (ja) 2004-06-18 2008-01-31 バイオリポックス エービー 炎症の治療に有用なインドール類
BRPI0519774A2 (pt) * 2005-01-19 2009-02-10 Biolipox Ab composto ou um sal farmaceuticamente aceitÁvel do mesmo, formulaÇço farmacÊutica, uso de um composto ou um sal farmaceuticamente aceitÁvel do mesmo, mÉtodo de tratamento de uma doenÇa em que inibiÇço da atividade de um membro da famÍlia mapeg É desejada e/ou necessÁria, produto combinado, e, processo para a preparaÇço de um composto
CA2599320A1 (en) 2005-02-25 2006-08-31 Kudos Pharmaceuticals Limited Hydrazinomethyl, hydr zonomethyl and 5-membered heterocylic compounds which act as mtor inhibitors and their use as anti cancer agents
CN101379060B (zh) * 2006-02-10 2012-05-23 转化技术制药公司 作为Aurora激酶抑制剂的苯并唑系衍生物、组合物和使用方法
WO2007124589A1 (en) 2006-05-02 2007-11-08 Merck Frosst Canada Ltd. Methods for treating or preventing neoplasias
WO2008009924A2 (en) * 2006-07-18 2008-01-24 Biolipox Ab Indoles useful in the treatment of inflammation
WO2008035956A1 (en) 2006-09-22 2008-03-27 Ewha University - Industry Collaboration Foundation New benzoxazole derivative, process for the preparation thereof and pharmaceutical composition comprising the same
WO2008129276A1 (en) 2007-04-19 2008-10-30 Boehringer Ingelheim International Gmbh Disulfonamides useful in the treatment of inflammation
UY32138A (es) * 2008-09-25 2010-04-30 Boehringer Ingelheim Int Amidas sustituidas del ácido 2-(2,6-dicloro-fenilamino)-6-fluoro-1-metil-1h-bencimidazol-5-carboxílico y sus sales farmacéuticamente aceptables
UY32470A (es) 2009-03-05 2010-10-29 Boehringer Ingelheim Int Derivados de 2-{2-cloro-5-[(sustituido) metil]fenilamino} -1-metil]fenilamino}-1-metilbencimidazol-5-carboxamidas-n-(sustituidas) y sus sales fisiológicamente aceptables, composiciones conteniéndolos y aplicaciones
US8759537B2 (en) 2010-08-20 2014-06-24 Boehringer Ingelheim International Gmbh 3H-imidazo [4, 5-C] pyridine-6-carboxamides as anti-inflammatory agents
US8586604B2 (en) 2010-08-20 2013-11-19 Boehringer Ingelheim International Gmbh Inhibitors of the microsomal prostaglandin E2 synthase-1
US8486968B2 (en) 2010-12-10 2013-07-16 Boehringer Ingelheim International Gmbh Compounds
US8466186B2 (en) 2010-12-10 2013-06-18 Boehringer Ingelheim International Gmbh Compounds
UY33779A (es) 2010-12-10 2012-07-31 Boehringer Ingelheim Int ?2-(fenilamino)-1H-bencimidazol-5-carboxamidas novedosas?

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6608084B1 (en) * 1998-08-26 2003-08-19 Aventis Pharma Ltd. Aza-bicycles which modulate the inhibition of cell adhesion
US20050267147A1 (en) * 2003-02-26 2005-12-01 Lydie Poitout Novel benzimidazole and imidazopyridine derivatives and use thereof as a medicament
US20060173036A1 (en) * 2003-03-31 2006-08-03 Lydie Poitout Imidazopyridine derivatives as melanocortin receptor agonists
US20040198768A1 (en) * 2003-04-03 2004-10-07 Park Choo Hea Young Method for inhibiting 5-lipoxygenase using a benzoxazole derivative or an analogue thereof
US20060287344A1 (en) * 2005-01-13 2006-12-21 Albers Ronald J Haloaryl substituted aminopurines, compositions thereof, and methods of treatment therewith
US20070060598A1 (en) * 2005-01-13 2007-03-15 Albers Ronald J Haloaryl substituted aminopurines, compositions thereof, and methods of treatment therewith
US20100004301A1 (en) * 2006-12-14 2010-01-07 Benjamin Pelcman Benzoxazoles Useful in the Treatment of Inflammation

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
no references cited *

Cited By (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8598190B2 (en) 2008-09-25 2013-12-03 Boehringer Ingelheim International Gmbh 3H-imidazo [4, 5-C] pyridine-6-carboxamides as anti-inflammatory agents
US9040565B2 (en) 2008-09-25 2015-05-26 Orexo Ab 1H-benzimidazole-5-carboxamides as anti-inflammatory agents
US8916599B2 (en) 2008-09-25 2014-12-23 Orexo Ab 1H-benz imidazole-5-carboxamides as anti-inflammatory agents
US8703796B2 (en) 2008-09-25 2014-04-22 Boehringer Ingelheim International Gmbh 3H-imidazo [4, 5-B] pyridine-6-carboxamides as anti-inflammatory agents
US8921405B2 (en) 2009-03-05 2014-12-30 Orexo Ab Compounds
US8759537B2 (en) 2010-08-20 2014-06-24 Boehringer Ingelheim International Gmbh 3H-imidazo [4, 5-C] pyridine-6-carboxamides as anti-inflammatory agents
US8586604B2 (en) 2010-08-20 2013-11-19 Boehringer Ingelheim International Gmbh Inhibitors of the microsomal prostaglandin E2 synthase-1
US8674113B2 (en) 2010-12-10 2014-03-18 Boehringer Ingelheim International Gmbh Compounds
US20120309738A1 (en) * 2010-12-10 2012-12-06 Boehringer Ingelheim International Gmbh Novel compounds
US8486968B2 (en) * 2010-12-10 2013-07-16 Boehringer Ingelheim International Gmbh Compounds
US8466186B2 (en) * 2010-12-10 2013-06-18 Boehringer Ingelheim International Gmbh Compounds
US20120309755A1 (en) * 2010-12-10 2012-12-06 Boehringer Ingelheim International Gmbh Novel compounds
US9962375B2 (en) * 2014-10-29 2018-05-08 Eli Lilly And Company Methyl-piperidine compounds useful for inhibiting microsomal prostaglandin E2 synthase-1
US9969714B2 (en) 2014-10-29 2018-05-15 Eli Lilly And Company Carboxylic acid compounds useful for inhibiting microsomal prostaglandin E2 synthase-1
US11691963B2 (en) 2020-05-06 2023-07-04 Ajax Therapeutics, Inc. 6-heteroaryloxy benzimidazoles and azabenzimidazoles as JAK2 inhibitors
WO2022251384A1 (en) * 2021-05-25 2022-12-01 Louis Habash Modifying the expression level of a gene encoding an cyclooxygenase enzyme by treating a human subject with a nitroxide
US11970494B2 (en) 2021-11-09 2024-04-30 Ajax Therapeutics, Inc. 6-heteroaryloxy benzimidazoles and azabenzimidazoles as JAK2 inhibitors
US11993613B2 (en) 2023-03-30 2024-05-28 Abbvie Inc. Thiazolo[5,4-b]pyridine MALT-1 inhibitors

Also Published As

Publication number Publication date
PT2403852E (pt) 2015-02-06
AR075789A1 (es) 2011-04-27
JP5433708B2 (ja) 2014-03-05
RS53757B1 (en) 2015-06-30
CA2753149A1 (en) 2010-09-10
CY1115959T1 (el) 2017-01-25
AU2010220263B2 (en) 2016-07-14
KR20110123764A (ko) 2011-11-15
GEP20146034B (en) 2014-02-10
CN102341395A (zh) 2012-02-01
EA020282B1 (ru) 2014-10-30
AU2010220263A1 (en) 2011-08-11
TWI466869B (zh) 2015-01-01
JP2012519205A (ja) 2012-08-23
EA201101243A1 (ru) 2012-04-30
US20130303571A1 (en) 2013-11-14
AP2806A (en) 2013-11-30
TN2011000446A1 (en) 2013-03-27
IL214033A0 (en) 2011-08-31
MY161092A (en) 2017-04-14
CL2011002180A1 (es) 2012-01-20
UY32470A (es) 2010-10-29
NZ594589A (en) 2014-01-31
SI2403852T1 (sl) 2015-04-30
US20120196897A1 (en) 2012-08-02
BRPI1009118A2 (pt) 2016-12-06
EP2403852B1 (en) 2014-10-29
ES2528730T3 (es) 2015-02-12
HRP20150096T1 (hr) 2015-02-27
CO6450668A2 (es) 2012-05-31
MA33239B1 (fr) 2012-05-02
TW201043604A (en) 2010-12-16
HK1163064A1 (en) 2012-09-07
US8921405B2 (en) 2014-12-30
ECSP11011324A (es) 2011-10-31
EP2403852A1 (en) 2012-01-11
MX2011008948A (es) 2011-09-27
AP2011005807A0 (en) 2011-08-31
ME02117B (me) 2015-10-20
EP2403852B8 (en) 2014-12-17
SG174250A1 (en) 2011-10-28
PE20120589A1 (es) 2012-05-26
WO2010100249A1 (en) 2010-09-10
CN102341395B (zh) 2014-07-30
PL2403852T3 (pl) 2015-04-30
DK2403852T3 (en) 2015-01-26
SMT201500053B (it) 2015-05-05

Similar Documents

Publication Publication Date Title
US8921405B2 (en) Compounds
US8466186B2 (en) Compounds
US8951999B2 (en) Compounds
US8759537B2 (en) 3H-imidazo [4, 5-C] pyridine-6-carboxamides as anti-inflammatory agents
US8586604B2 (en) Inhibitors of the microsomal prostaglandin E2 synthase-1
US20050203158A1 (en) Substituted benzimidazole compounds
US8486968B2 (en) Compounds
WO2012076673A1 (en) 6-amino-2-phenylamino-1h-benzimidazole-5-carboxamide- derivatives and their use as microsomal prostaglandin e2 synthase-1 inhibitors
OA16323A (en) New compounds.

Legal Events

Date Code Title Description
AS Assignment

Owner name: BOEHRINGER INGELHEIM INTERNATIONAL GMBH, GERMANY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:PFAU, ROLAND;ARNDT, KIRSTEN;DOODS, HENRI;AND OTHERS;SIGNING DATES FROM 20100326 TO 20100429;REEL/FRAME:024486/0582

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO PAY ISSUE FEE