WO2006058752A1 - [1,2,4]triazolo[4,3-a]pyridine derivatives for the treatment of hyperproliferative diseases - Google Patents

[1,2,4]triazolo[4,3-a]pyridine derivatives for the treatment of hyperproliferative diseases Download PDF

Info

Publication number
WO2006058752A1
WO2006058752A1 PCT/EP2005/012855 EP2005012855W WO2006058752A1 WO 2006058752 A1 WO2006058752 A1 WO 2006058752A1 EP 2005012855 W EP2005012855 W EP 2005012855W WO 2006058752 A1 WO2006058752 A1 WO 2006058752A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
heteroaryl
substituted
aryl
heterocyclyl
Prior art date
Application number
PCT/EP2005/012855
Other languages
French (fr)
Inventor
Ulrich Abel
Holger Deppe
Achim Feurer
Ulrich GRÄDLER
Inge Ott
Victor Giulio Matassa
Original Assignee
Laboratoires Serono S.A.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Laboratoires Serono S.A. filed Critical Laboratoires Serono S.A.
Priority to CA002586796A priority Critical patent/CA2586796A1/en
Priority to US11/791,799 priority patent/US20070299103A1/en
Priority to AT05826556T priority patent/ATE443063T1/en
Priority to AU2005311451A priority patent/AU2005311451A1/en
Priority to JP2007543779A priority patent/JP2008521858A/en
Priority to EP05826556A priority patent/EP1828184B1/en
Priority to DE602005016718T priority patent/DE602005016718D1/en
Publication of WO2006058752A1 publication Critical patent/WO2006058752A1/en
Priority to IL183340A priority patent/IL183340A0/en
Priority to NO20073393A priority patent/NO20073393L/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/18Drugs for disorders of the alimentary tract or the digestive system for pancreatic disorders, e.g. pancreatic enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/08Drugs for disorders of the urinary system of the prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis

Definitions

  • the invention relates to a series of substituted 7-arylamino[1 ,2,4]triazolo[4,3-a]pyridine derivatives that are useful in the treatment of hyperproliferative diseases, such as cancer and inflammation, in mammals. Also disclosed is the use of such compounds in the treatment of hyperproliferative diseases in mammals, especially humans, and pharmaceutical compositions containing such compounds.
  • the Ras/Raf/MEK/ERK pathway is a central signal transduction pathway, which transmits signals from multiple cell surface receptors to transcription factors in the nucleus, which regulate gene expression.
  • This pathway is frequently referred to as the MAP kinase pathway as MAPK stands for mitoge ⁇ -activated protein kinase indicating that this pathway can be stimulated by mitogens, cytokines and growth factors (Steelman et al., Leukemia 2004, 18, 189-218).
  • this pathway can transmit signals, which result in the prevention or induction of apoptosis or cell cycle progression.
  • the Ras/Raf/MEK/ERK pathway has been shown to play important roles in cell proliferation and the prevention of apoptosis.
  • Ras Aberrant activation of this pathway is commonly observed in malignantly transformed cells. Amplification of ras proto-oncogenes and activating mutations that lead to the expression of constitutively active Ras proteins are observed in approximately 30% of all human cancers (Stirewalt et al., Blood 2001, 97, 3589-95). Mutated, oncogenic forms of Ras are found in 50% of colon and >90% pancreatic cancers as well as many other types of cancers (Kohl et al., Science 1993, 260, 1834-1837). The effects of Ras on proliferation and tumorigenesis have been documented in immortal cell lines (McCubrey et al., lnt J Oncol 1995, 7, 295-310).
  • ⁇ Raf mutations have been identified in more than 60% of malignant melanoma (Davies, H et al., Nature 2002, 417, 949-954). Given the high level of mutations that have been detected at Ras, this pathway has always been considered a key target for therapeutic intervention (Chang et al., Leukemia 2003, 17,1263-93).
  • the Ras/Raf/MEK/ERK signaling pathway can exert proliferative or antiproliferative effects through downstream transcription factor targets including NF- ⁇ B, CREB, Ets-1 , AP-1 and c-Myc.
  • ERKs can directly phosphorylate Ets-1 , AP-1 and c-Myc, which lead to their activation.
  • ERKs can phosphorylate and activate a downstream kinase target RSK, which then phosphorylates and activates transcription factors, such as CREB.
  • These transcription factors induce the expression of genes important for cell cycle progression, for example, Cdks, cyclins, growth factors, and apoptosis prevention, for example, antiapoptotic Bcl-2 and cytokines.
  • treatment of cells with growth factors leads to the activation of ERKs which results in proliferation and, in some cases, differentiation (Lewis et al., Adv. Cancer Res, 1998, 74, 49-139).
  • MEK proteins are the primary downstream targets of Raf.
  • the MEK family of genes consists of five genes: MEK1 , MEK2, MEK3, MEK4 and MEK5. This family of dual- specificity kinases has both serine/threonine and tyrosine kinase activity.
  • the structure of MEK consists of an amino-terminal negative regulatory domain and a carboxy-terminal MAP kinase-binding domain, which is necessary for binding and activation of ERKs. Deletion of the regulatory MEK1 domain results in constitutive MEK1 and ERK activation (Steelman et al., Leukemia 2004, 18, 189-218).
  • MEK1 is a 393-amino-acid protein with a molecular weight of 44 kDa (Crews et al., Science 1992, 258, 478-80). MEK1 is modestly expressed in embryonic development and is elevated in adult tissue with the highest levels detected in brain tissue. MEK1 - requires phosphorylation of S218 and S222 for activation, and substitution of these residues with D or glutamic acid (E) led to an increase in activity and foci formation in NIH3T3 cells (Huang et al., MoI Biol Cell, 1995, 6, 237-45).
  • MEK1 constitutive activity of MEK1 in primary cell culture promotes senescence and induces p53 and p16 INK4a , and the opposite was observed in immortalized cells or cells lacking either p53 or p16 INK4a (Lin et al., Genes Dev, 1998 , 12, 3008-3019).
  • Constitutive activity of MEK1 inhibits NF- KB transcription by negatively regulating p38 MAPK activity (Carter et al., J Biol Chem 2000, 275, 27858-64).
  • the main physiological substrates of MEK are the members of the ERK (extracellular signal-regulated kinase) or MAPK (mitogen activated protein kinase) family of genes. Aberrant expression of MEK1 has been detected in many different types of cancer, and mutated forms of MEK1 will transform fibroblast, hematopoietic and other cell types.
  • MEK1 Constitutive activation of MEK1 results in cellular transformation. It therefore represents a likely target for pharmacological intervention in proliferative and inflammatory diseases (Lee et al., Nature 1994, 372, 739-746; Dudley et al., Proc. Natl. Acad. Sci. U.S.A. 1995, 92, 7686-7689).
  • MEK inhibitors have been developed that show potential therapeutic benefit in several studies. For example, small molecule MEK inhibitors have been shown to inhibit human tumor growth in nude mouse xenografts (Yeh, T. et al, Proceedings of the American Association of Cancer Research 2004, 45, Abs 3889 and Lee, P. et al., Proceedings of the American Association of Cancer Research 2004, 45, Abs 3890). MEK inhibitors also entered clinical trials, i.e. ARRY142886 (Wallace, E.
  • PD-184352 was lacking efficacy in clinical phase Il trials. Tumors were much less responsive, as no partial responses and only a few patients with stable disease were observed. As a result, the clinical trials of this molecule were suspended (Mclnnes C IDDB MEETING REPORT 2003). PD-184352 was limited by poor solubility, high metabolic clearance and low bioavailability. This exemplifies the need for novel MEK inhibitors with superior pharmacological properties.
  • this invention provides novel, substituted 7-arylamino[1 ,2,4]triazolo[4,3- a]pyridine derivatives and pharmaceutically acceptable salts, solvates or prodrugs thereof, that are MEK inhibitors and useful in the treatment of the above mentioned diseases.
  • the compounds are defined by Formula (I): Formula (I)
  • Ri, R2, R 9 , R11 R12, R13 and R 14 are independently selected from hydrogen, halogen, cyano, nitro, azido, -OR 3 , -C(O)R 3 , -C(O)OR 3 , -NR 4 C(O)OR 6 , -OC(O)R 3 , -NR 4 S(O)jR 6 , -S(O) J NR 3 R 4 , -S(O)jNR 4 C(O)R 3 , -C(O)NR 4 S(O)jR 6 , S(O)jR 6 , S(O)jR 6 ,
  • R 10 is selected from hydrogen, -OR 3 , -C(O)R 31 -C(O)OR 3 , -NR 4 C(O)OR 6 , -OC(O)R 3 , -NR 4 S(O)jR 6 , -S(O)jNR 3 R 4 , -S(O)jNR 4 C(O)R 3 , -C(O)NR 4 S(O)jR 6l S(O) J R 6 ,
  • L is selected from a bond, C 1 -C 10 alkyl, C 2 -C 10 alkenyl, C 2 -C 10 alkynyl, C 3 -C 10 cycloalkyl, C 3 -C 10 cycloalkylalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocyclyl, heterocyclylalkyl, where each alkyl, alkenyl, alkynyl, cycloalkyl, aryl, heteroaryl and heterocyclyl is unsubstituted or substituted;
  • R 3 is selected from hydrogen, trifluoromethyl, C 1 -C 10 alkyl, C 2 - 10 alkenyl, C 2 -C 10 alkynyl, C 3 -C 10 cycloalkyl, C 3 -C 10 cycloalkylalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocyclyl
  • R 4 is selected from hydrogen or C 1 -C 6 alkyl whereby alkyl may be substituted or unsubstituted; or R 3 and R 4 can be taken together with the atom to which they are attached to form a 4 to 10 membered heteroaryl or heterocyclic ring, each of which is substituted or unsubstituted;
  • R 5 is selected from hydrogen or CrC 6 alkyl whereby alkyl may be substituted or unsubstituted; or
  • R 4 and R 5 can be taken together with the atom to which they are attached to form a 4 to 10 membered carbocyclic, heteroaryl or heterocyclic ring, each of which is substituted or unsubstituted;
  • R 6 is selected from trifluoromethyl,Ci-Ci 0 alkyl, C 3 -Ci 0 cycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocyclyl, and heterocyclylalkyl, where each alkyl, cycloalkyl, aryl, heteroaryl and heterocyclyl substituted or unsubstituted;
  • W is selected from heteroaryl containing 1-4 heteroatoms or heterocyclyl containing 1-4 heteroatoms each of which is unsubstituted or substituted by 1 to 5 substituents ZR 15 ; or W is -C(O)ORi 5 , -C(O)NR 4 R 15 , -C(O)NR 4 OR 15 , -C(O)(C 3 -C 10 cycloalkyl), -C(O)(C 2 -C 10 alkyl), -C(O)(aryl), -C(O)(heteroaryl), -C(O)(heterocyclyl),
  • R 4 and R 15 are as defined herein or may form together a 3 to 7 membered ring with 1 or 2 N atoms and optionally an O atom,
  • Z is a bond, NR 16 , O, NR 16 SO 2 or S
  • R 15 is independently selected from hydrogen, trifluoromethyl, C 1 -Ci 0 alkyl, C 2 -C 10 alkenyl, C 2 -C 10 alkynyl, C 3 -C 10 cycloalkyl, C 3 -C 10 cycloalkylalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocyclyl, and heterocyclylalkyl, where each alkyl, alkenyl, alkynyl, cycloalkyl, aryl, heteroaryl and heterocyclyl is substituted or unsubstituted;
  • R 16 is selected from hydrogen or C 1 -C 10 alkyl, or R 15 and R 16 form together a 4 to 10 membered cyclic ring with 1 or 2 N atoms and optionally an O atom, said ring being substituted or unsubstituted; m is O, 1 , 2, 3, 4 or 5 ;and j is 1 or 2.
  • variants R 1 -R 16 , L, W and Z are defined as above on pages 4 to 5 but with the proviso that the compounds with the following formula including resolved enantiomers, diastereomers, solvates and pharmaceutically acceptable salts thereof are excluded:
  • R 1 , R 2 , R 7 , R 8 , R 9 and R 10 are independently hydrogen, halogen, cyano, nitro, azido, - OR 3 , -C(O)R 3 , -C(O)OR 3 , -NR 4 C(O)OR 6 , -OC(O)R 3 , -NR 4 SO 2 R 6 , -SO 2 NR 3 R 4 ,
  • R 3 is hydrogen, trifluoromethyl, Ci-Ci 0 alkyl, C 2 -Ci 0 alkenyl, C 2 -Ci 0 alkynyl, C 3 -Ci 0 - cycloalkyl, C 3 -Ci 0 cycloalkylalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocyclyl, heterocyclylalkyl, wherein any of said alkyl, alkenyl, alkynyl, cycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocyclyl and heterocyclylalkyl portions are optionally substituted with one or more groups independently selected from oxo, halogen, cyano, nitro, trifluoromethyl, difluoromethoxy, trifluoromethoxy, azido, -NR SO 2 R " " , -SO 2 NR R " ,
  • R 3 and R 4 together with the atom to which they are attached form a 4 to 10 membered heteroaryl or heterocyclic ring, wherein any of said heteroaryl or heterocyclic rings are optionally substituted with one or more groups independently selected from halogen, cyano, nitro, trifluoromethyl, difluoromethoxy, trifluoromethoxy, azido, NR 1 SO 2 R “' , -SO 2 NR R “ , -C(O)R ' , -C(O)OR ' , -OC(O)R ' , -NR C(O)OR “ “ , -NR C(O)R “ , -C(O)NR R '' -SO 2 R “ ' , -NR R “ , -NRC(O)NR “ R ' , -NR 1 C(NCN)NR “ R ' , -OR 1 aryl, heteroaryl, arylalkyl, heteroarylalkyl
  • R"" is lower alkyl, lower alkenyl, aryl or arylalkyl, or any two of R', R", R'" or R"" together with the atom to which they are attached form a 4 to 10 membered carbocyclic, heteroaryl or heterocyclic ring, wherein any of said alkyl, alkenyl, aryl, arylalkyl carbocyclic rings, heteroaryl rings or heterocyclic rings are optionally substituted with one or more groups independently selected from halo, cyano, nitro, trifluoromethyl, difluoromethoxy, trifluoromethoxy, azido, aryl, heteroaryl, arylalkyl, heteroarylalkyl, heterocyclyl, and heterocyclylalkyl;
  • R 4 and R 5 independently are hydrogen or Ci-C 6 alkyl, or
  • R 4 and R 5 together with the atom to which they are attached form a 4 to 10-membered carbocyclic, heteroaryl or heterocyclic ring, wherein said alkyl or any of said carbocyclic, heteroaryl and heterocyclic rings are optionally substituted with one or more groups independently selected from cyano, halogen, nitro, trifluoromethyl, difluoromethoxy, trifluoromethoxy, azido, -NR SO 2 R “ “ , -SO 2 NR R “ , -C(O)R “ “ , C(O)OR “ , -OC(O)R “ , -NR C(O)OR “ “ , -NRC(O)R “ , -C(O)NR R “ , -SO 2 R “ “ , -NR R “ , -NR C(O)NR 11 R “ , -NR C(NCN)NR 1 R ' , -OR " , aryl, heteroaryl, aryl
  • R 6 is trifluoromethyl, C 1 -C 1 0 alkyl, C 3 -Ci 0 cycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocyclyl or heterocyclylalkyl, wherein any of said alkyl, cycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocyclyl and heterocyclylalkyl portions are optionally substituted with one or more groups independently selected from oxo, halogen, cyano, nitro, trifluoromethyl, difluoromethoxy, trifluoromethoxy, azido, -NR SO 2 R " " , -SO 2 NR 1 R “ , -C(O)R ' , C(O)OR ' , -OC(O)R “ , -NR C(O)OR 1 " , -NR C(O)R “ , -C(O
  • W is heteroaryl, heterocyclyl, -C(O)OR 3 , -C(O)NR 3 R 4 , -C(O)NR 4 OR 3 ,
  • the variants have the following meanings:
  • Ri is as defined above, preferably hydrogen, halo, Ci-C 4 alkyl, C 3 -C 4 cycloalkyl, C 2 -C 4 alkenyl, C 2 -C 4 alkynyl, cyano, nitro, OR 3 or NR 3 R 4 ; more preferably hydrogen, halo or C 1 - C 4 alkyl, still more preferably hydrogen or halo, most preferably hydrogen or F.
  • Ri is hydrogen.
  • R 1 is -S(O)jNR 4 C(O)R 3 , -C(O)NR 4 S(O)jR 6 , or S(O)jR 6 .
  • R 2 is as defined above, preferably hydrogen, halo, Ci-C 4 alkyl, C 3 -C 4 cycloalkyl, C 2 -C 4 alkenyl, C 2 -C 4 alkynyl, cyano, nitro, OR 3 or NR 3 R 4 ; more preferably hydrogen, halo or C 1 - C 2 alkyl, still more preferably halo or methyl, most preferably Cl, F or methyl.
  • R 2 is methyl.
  • methyl is preferably further substituted by 1 , 2 or 3 fluorines, preferably 3 fluorines. Most preferably, R 2 is F.
  • R 2 is -S(O) j NR 4 C(O)R 3 , -C(O)NR 4 S(O) j R 6 , or S(O) j R 6 .
  • R 9 is as defined above, preferably hydrogen, halo, C r C 4 alkyl, C 3 -C 4 cycloalkyl, C 2 -C 4 alkenyl, C 2 -C 4 alkynyl, cyano, nitro, OR 3 or NR 3 R 4 ; more preferably hydrogen, halo or C 1 - C 4 alkyl, still more preferably hydrogen, methyl or halo, most preferably hydrogen, methyl, Cl or F.
  • R 9 is hydrogen.
  • R 9 is -S(O)jNR 4 C(O)R 3 , -C(O)NR 4 S(O) j R 6 , or S(O)jR 6 .
  • R 10 is as defined above, preferably hydrogen, -OR 3 , -C(O)R 31 -C(O)OR 3 , -NR 4 C(O)OR 6 , -OC(O)R 3 , -NR 4 S(O) 2 R 6 , -S(O) 2 NR 3 R 4 , S(O) 2 R 6 , -NR 4 C(O)R 3 , -C(O)NR 3 R 4 , -NR 5 C(O)NR 3 R 4 , -NR 3 R 4 , more preferably hydrogen, -OR 3 , -NR 4 C(O)R 3 , -C(O)NR 3 R 4 , -NR 3 R 4 , still more preferably hydrogen, -OR 3 , -NR 4 C(O)R 3
  • R 3 and R 4 are independently Ci-C 6 alkyl, more preferably C 1 -C 4 alkyl, optionally substituted by 1 or 2 alkyl amino, dialkyl amino, amino, O-alkyl, hydroxy, or R 3 and R 4 form together a cyclic ring with 1 or 2 N atoms and optionally an O atom, said ring being optionally substituted by 1 or 2 alkyl amino, amino, hydroxy or O-alkyl.
  • Ri 0 is -S(O)jNR 4 C(O)R 3 , -C(O)NR 4 S(O) J R 6 , -S(O)jR ⁇ , -S(O) j (Ci-C 6 alkyl), -S(O) j (CR 4 R 5 ) m -aryl, -O(CR 4 R5)m-aryl, -NR 4 (CR 4 R 5 )m-aryl, -O(CR 4 R 5 ) m -heteroaryl, -NR 4 (CR 4 R 5 ) m -heteroaryl, -O(CR 4 R 5 ) m -heterocyclyl, or -NR 4 (CR 4 R 5 ) m -heterocyclyl and -S(Ci-C 2 alkyl) substituted with 1 to 5 F, where each aryl, heteroaryl and heterocyclyl is substituted or unsubstituted
  • L is as defined above, preferably a bond, C r C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, C 3 -Ci 0 cycloalkylalkyl, arylalkyl, heteroarylalkyl, heterocyclyl, heterocyclylalkyl, more preferably a bond, CrC 5 alkyl, most preferably a bond, methylene, ethylene, n- propylene or n-butylene.
  • L is ethylene, n-propylene or n-butylene.
  • L is a bond or methylene.
  • all moieties are divalent so that L serves as a linker between the nitrogen atom and R 10 .
  • R 11 is as defined above, preferably hydrogen, halo, C 1 -C 4 alkyl, C 3 -C 4 cycloalkyl, C 2 -C 4 alkenyl, C 2 -C 4 alkynyl, cyano, nitro, OR 3 or NR 3 R 4 ; more preferably hydrogen, halo or C 1 -C 4 alkyl or 0-Ci-C 4 alkyl, still more preferably hydrogen, methyl, O-methyl or halo, most preferably hydrogen, methyl, Cl, Br or F.
  • R 11 is hydrogen.
  • R 11 is methyl.
  • methyl is preferably further substituted by 1 , 2 or 3 fluorines, preferably 3 fluorines.
  • R 11 is -S(O)jNR 4 C(O)R 3 , -C(O)NR 4 S(O),R 6 , or S(O),R 6 .
  • R 12 is as defined above, preferably hydrogen, halo, C 1 -Ci 0 alkyl, C 3 -C 10 cycloalkyl, C 2 -C 10 alkenyl, C 2 -C 10 alkynyl, cyano, nitro, azido; NR 4 SO 2 R 6 ; SO 2 NR 3 R 4 ; SO 2 R 6 ; C(O)NR 3 R 4 ; C(O)OR 3 ; OR 3 , NR 3 R 4 Or -S(C 1 -C 2 alkyl) substituted with 1 to 5 F, more preferably hydrogen, halo, nitro, Ci-C 4 alkyl, 0-C 1 -C 4 alkyl, SCF 3 , SCHF 2 , SCH 2 F, SO 2 NR 3 R 4 or C(O)NR 3 R 4 , still more preferably hydrogen, F, Cl, Br, I, nitro, methyl, ethyl, n-propyl, i-propyl,
  • R 12 is hydrogen. In another embodiment, R 12 is methyl, SCF 3 , SCHF 2 , SCH 2 F or O-methyl, wherein methyl or O-methyl is preferably unsubstituted or further substituted by 1 , 2 or 3 fluorines, preferably 2 or 3 fluorines.
  • R 12 , R 3 and R 4 are independently C 1 -C 6 alkyl, more preferably Ci-C 4 alkyl, optionally substituted by 1 or 2 alkyl amino, dialkyl amino, amino, O-alkyl, hydroxy, or R 3 and R 4 form together a cyclic ring with 1 or 2 N atoms and optionally an O atom, said ring being optionally substituted by 1 or 2 alkyl amino, amino, hydroxy or O-alkyl.
  • Ri 2 is Br or I.
  • R 12 is -S(O) j NR 4 C(O)R 3 , -C(O)NR 4 S(O) j R 6) or S(O) j R 6 .
  • Ri3 is as defined above, preferably hydrogen, halo, Ci-C 4 alkyl, C 3 -C 4 cycloalkyl, C 2 -C 4 alkenyl or C 2 -C 4 alkynyl, more preferably hydrogen, F, Cl or methyl, most preferably hydrogen or F. in one embodiment, Ri 3 is hydrogen.
  • R 13 is -S(O)jNR 4 C(O)R 3 , -C(O)NR 4 S(O) j R 6 , or S(O) ) R 6 .
  • Ri 4 is as defined above, preferably hydrogen, halo, Ci-C 4 alkyl, C 3 -C 4 cycloalkyl, C 2 -C 4 alkenyl Or C 2 -C 4 alkynyl, more preferably hydrogen, F, Cl or methyl, most preferably hydrogen or F. In one embodiment, Ri 4 is hydrogen.
  • R 14 is -S(O) j NR 4 C(O)R 3 , -C(O)NR 4 S(O) j R 6 , or S(O) j R 6 .
  • At least one of R 1 , R 2 , R9, Rn, R 12 , R 13 and R 14 is selected from -S(O)jNR 4 C(O)R 3 , -C(O)NR 4 S(O) j R 6 , and S(O)jR 6
  • each of R 1 , R 2 , Rg to R 14 and L may be substituted.
  • they can be substituted with 1 to 5, preferably 1 to 3, more preferably 1 or 2 groups independently selected from oxo, halogen, cyano, nitro, CF 3 , CHF 2 , CH 2 F, OCF 3 , OCHF 2 , OCH 2 F, SCF 3 , SCHF 2 , SCH 2 F, azido, NR 4 SO 2 R 6 , SO 2 NR 3 R 4 , C(O)R 3 , C(O)OR 3 , OC(O)R 3 , NR 4 C(O)OR 6 , NR 4 C(O)R 3 , C(O)NR 3 R 4 , NR 3 R 4 , NR 5 C(O)NR 3 R 4 ,
  • NR 5 C(NCN)NR 3 R 4 , OR 3 aryl, heteroaryl, arylalkyl, heteroarylalkyl, heterocyclyl, and heterocyclylalkyl, preferably oxo, halogen, cyano, nitro, CF 3 , CHF 2 , CH 2 F, OCF 3 , OCHF 2 , OCH 2 F, SCF 3 , SCHF 2 , SCH 2 F, azido, NR 4 SO 2 R 6 , SO 2 NR 3 R 4 , C(O)R 3 , C(O)OR 3 , OC(O)R 3 , OR 3 , more preferably oxo, halogen, cyano, nitro, trifluoromethyl, difluoromethoxy, trifluoromethoxy or azido, most preferably halogen, cyano, nitro, CF 3 , CHF 2 , CH 2 F, OCF 3 , OCHF 2 , OCH 2 F
  • alkyl, alkenyl, alkynyl, cycloalkyl, aryl, heteroaryl and heterocyclyl are substituted, this refers to any alkyl, alkenyl, alkynyl, cycloalkyl, aryl, heteroaryl and heterocyclyl as a group or sub-structure such as in cycloalkylalkyl, arylalkyl, heteroarylalkyl, heterocyclylalkyl.
  • R 3 is as defined above, preferably hydrogen, trifluoromethyl, Ci-C 4 alkyl, C 2 -C 4 alkenyl, C 2 -C 4 alkynyl, C 3 -C 6 cycloalkyl, C 3 -C 6 cycloalkylalkyl, more preferably hydrogen or Ci-C 4 alkyl most preferably hydrogen, methyl or ethyl.
  • R 4 is as defined above, preferably hydrogen or Ci-C 4 alkyl, more preferably hydrogen, methyl or ethyl.
  • R 3 and R 4 can be taken together with the atom to which they are attached to form a 4 to 7, preferably 5 or 6, membered heteroaryl or heterocyclic ring.
  • R 5 is as defined above, preferably hydrogen or CrC 4 alkyl, more preferably hydrogen, methyl or ethyl.
  • R 4 and R 5 can be taken together with the atom to which they are attached to form a 4 to 7, preferably 5 or 6, membered carbocyclic, heteroaryl or heterocyclic ring.
  • R 6 is as defined above, preferably trifluoromethyl, Ci-C 4 alkyl, C 2 -C 4 alkenyl, C 2 -C 4 alkynyl, C 3 -C 6 cycloalkyl, C 3 -C 6 cycloalkylalkyl, more preferably d-C 4 alkyl, most preferably methyl or ethyl.
  • each of R 3 , R 4 , R 5 , R 6 or the rings formed by R 3 and R 4 and R 4 and R 5 may be substituted.
  • they can be substituted with 1 to 5, preferably 1 to 3, more preferably 1 or 2 groups independently selected from oxo, halogen, cyano, nitro, CF 3 , CHF 2 , CH 2 F, OCF 3 , OCHF 2 , OCH 2 F, azido, NR 1 SO 2 R"", SO 2 NR", SO 2 NR", SO 2 NR", SO 2 NR", C(O)R', C(O)OR', OC(O)R', NR 1 C(O)OR"", NR 1 C(O)R", C(O)NR 1 R", SR"", S(O)R"", SO 2 R', NR 1 R", NR 1 C(O)NR 11 R 1 ", NR 1 C(NCN)NR 11 R"', OR', aryl, heteroaryl, arylalky
  • R 3 is preferably oxo, halogen, nitro, trifluoromethyl, OH, O-methyl, NH 2 or N(methyl) 2 .
  • alkyl, alkenyl, alkynyl, cycloalkyl, aryl, heteroaryl and heterocyclyl are substituted, this refers to any alkyl, alkenyl, alkynyl, cycloalkyl, aryl, heteroaryl and heterocyclyl as a group or sub-structure such as in cycloalkylalkyl, arylalkyl, heteroarylalkyl, heterocyclylalkyl.
  • R 1 is selected from hydrogen, Ci-C 4 alkyl, C 2 -C 4 alkenyl, aryl and arylalkyl, preferably hydrogen or CrC 4 alkyl, more preferably hydrogen or methyl.
  • R" is selected from hydrogen, Ci-C 4 alkyl, C 2 -C 4 alkenyl, aryl and arylalkyl, preferably hydrogen or CrC 4 alkyl, more preferably hydrogen or methyl.
  • R'" is selected from hydrogen, Ci-C 4 alkyl, C 2 -C 4 alkenyl, aryl and arylalkyl, preferably hydrogen or Ci-C 4 alkyl, more preferably hydrogen or methyl.
  • R 11 " is selected from Ci-C 4 alkyl, Ci-C 4 alkenyl, aryl and arylalkyl, preferably Ci-C 4 alkyl, more preferably methyl.
  • any two of R', R", R"' or R"" can be taken together with the atom to which they are attached to form a 4 to 10 membered carbocyclic, heteroaryl or heterocyclic ring, each of which is optionally substituted with one to three groups independently selected from halogen, cyano; nitro, CF 3 , CHF 2 , CH 2 F, OCF 3 , OCHF 2 , OCH 2 F, azido, aryl, heteroaryl, arylalkyl, heteroarylalkyl, heterocyclyl, and heterocyclylalkyl, preferably halogen, cyano; nitro, trifluoromethyl, difluoromethoxy, trifluoromethoxy and azido.
  • W is as defined above, preferably heteroaryl containing 1 , 2 or 3 heteroatoms, or heterocyclyl containing 1 , 2, or 3 heteroatoms, more preferably heteroaryl, each of which is unsubstituted or substituted by 1 to 5, preferably 1 to 3, more preferably 1 , substituents ZR 15 , or W is -C(O)OR 15 , -C(O)NR 4 R 15 , -C(O)NR 4 OR 15 , -C(O)(C 3 -C 10 cycloalkyl),
  • W is heteroaryl containing 1 , 2, or 3, specifically 2 or 3 N atoms, C(O)NR 4 OR 15 or S(O) 2 NR 4 OR 15 .
  • R 4 and R 15 are as defined herein or may form together a 3 to 7 membered ring with 1 or 2 N atoms and optionally an O atom.
  • W is heteroaryl, it is preferably
  • Z and R 15 are as defined above, preferably Z is a bond, NR 16 , NRi 6 SO 2 or O, more preferably NR 16 , wherein R 16 is as defined above, preferably hydrogen Or C 1 -C 4 alkyl, more preferably hydrogen.
  • R 15 is preferably selected from hydrogen, C 1 -C 4 alkyl, C 1 -C 4 alkenyl, C 4 -C 8 cycloalkylalkyl, each may contain 1 N atom optionally an O atom, where alkyl, alkenyl or cycloalkylalkyl may be further substituted by 1 or 2 of OH, 0-C 1 -C 4 alkyl or NR'R", where R' and R" are independently hydrogen or C 1 -C 4 alkyl where R ' and R" may form a 3 to 7 membered ring with 1 or 2 N atoms and optionally an O atom.
  • R 16 and R 15 may form together a 4 to 10 membered cyclic ring with 1 or 2 N atoms and optionally an O atom, said ring being optionally substituted by 1 or 2 alkyl amino, amino, hydroxy or O-alkyl. More preferably R 15 is C 1 -C 4 alkyl or C 1 -C 4 alkenyl optionally substituted with 1 substituent OH 1 O-Me, NH 2 , NHmethyl, NHethyl, N(methyl) 2 or N(ethyl) 2 .
  • Y is O or NR', preferably O.
  • W is preferably -C(O)ORi 5 , -C(O)NR 4 Ri 5 , -C(O)NR 4 ORi 5 , S(O) 1 NR 4 R 15 or S(O) j NR 4 ORi5, more preferably -C(O)NR 4 ORi 5 or S(O) 2 NR 4 ORi 5 .
  • R 15 is preferably as defined below.
  • W is -C(O)NR 4 S(O) 1 R 6 , or -S(O) j NR 4 C(O)Ri 5 , whereby R 4 and R i5 are as defined herein or may form together a 3 to 7 membered ring with 1 or 2 N atoms and optionally an O atom.
  • Z is as defined above, preferably a bond, NRi 6 , NR 16 SO 2 or O, more preferably NR 16 . In another embodiment, Z is S.
  • Ri 5 is as defined above, preferably hydrogen, C 1 -C 4 alkyl, C 1 -C 4 alkenyl, C 4 -C 6 cycloalkylalkyl, more preferably C 1 -C 4 alkyl or C 1 -C 4 alkenyl, yet more preferably C 1 -C 4 alkyl.
  • Alkyl, alkenyl cycloalkyl, alkynyl, aryl, heteroaryl or hetercyclyl may be further substituted with 1 to 5, preferably 1 , 2 or 3, more preferably 1 or 2, substituents selected from OR 3 or NR'R" wherein R 3 is selected from hydrogen, C 1 -C 4 alkyl or C 1 -C 4 alkenyl, C 4 -C 6 cycloalkylalkyl, more preferably hydrogen, methyl or ethyl, and where R' and R" are independently hydrogen or C 1 -C 4 alkyl, or R ' and R " may form a 3 to 7 membered ring with 1 or 2 N atoms and optionally an O atom, more preferably R 1 and R" are independently hydrogen, methyl or ethyl, still more preferably both R' and R" are methyl. Yet more preferably, R 15 may be substituted by 1 or 2 of OH, 0-C 1 -C 4 alkyl or
  • R 15 is C 1 -C 4 alkyl or C 1 -C 4 alkenyl optionally substituted with 1 substituent OH, 0-Me 1 NH 2 , N(methyl) 2 or N(ethyl) 2
  • R 15 when it is described that alkyl, alkenyl, alkynyl, cycloalkyl, aryl, heteroaryl and heterocyclyl are substituted, this refers to any alkyl, alkenyl, alkynyl, cycloalkyl, aryl, heteroaryl and heterocyclyl as a group or sub-structure such as in cycloalkylalkyl, arylalkyl, heteroarylalkyl, heterocyclylalkyl.
  • R 16 is as defined above, preferably hydrogen or C 1 -C 4 alkyl, more preferably hydrogen.
  • R 16 and R 15 may form together a 4 to 10, preferably 5 to 6, membered cyclic ring with 1 or 2 N atoms and optionally an O atom, said ring being optionally substituted by 1 or 2 alkyl amino, amino, hydroxy or O-alkyl.
  • m is as defined above, preferably 0,1 ,2 or 3, more preferably 0,1 or 2, most preferably 1.
  • j is as defined above, preferably 2.
  • any of the preferred definitions for each variant can be combined with the preferred definition of the other variants.
  • the combinations as set forth in the claims are particularly preferred.
  • Aryl is an aromatic mono- or polycyclic moiety with preferably 6 to 20 carbon atoms which is preferably selected from phenyl, biphenyl, naphthyl, tetrahydronaphthyl, fluorenyl, indenyl or phenanthrenyl, more preferably phenyl or naphthyl.
  • Heteroaryl is an aromatic moiety having 6 to 20 carbon atoms with at least one ring containing a heteroatom selected from O, N and/or S, or heteroaryl is an aromatic ring containing at least one heteroatom selected from O, N and/or S and 1 to 6 carbon atoms.
  • heteroaryl contains 1 to 4, more preferably 1 , 2 or 3 heteroatoms selected from O and/or N and is preferably selected from pyridinyl, imidazolyl, pyrimidinyl, pyrazolyl, triazolyl, pyrazinyl, tetrazolyl, furyl, thienyl, isoxazolyl, thiazolyl, oxazolyl, isothiazolyl, pyrrolyl, quinolinyl, isoquinolinyl, indolyl, benzimidazolyl, benzofuranyl, cinnolinyl, indazolyl, indolizinyl, phthalazinyl, pyridazinyl, triazinyl, isoindolyl, pteridinyl, purinyl, oxadiazolyl, triazolyl, thiadiazolyl, thiadiazolyl, furazanyl,
  • heteroaryl examples include pyridinyl, imidazolyl, pyrimidinyl, pyrazolyl, triazolyl, pyrazinyl, tetrazolyl, isoxazolyl, oxazolyl, isothiazolyl, oxadiazolyl, triazolyl.
  • Heteroaryl groups are optionally mono-, di-, or trisubstituted with, e.g., halogen, lower alkyl, lower alkoxy, haloalkyl, aryl, heteroaryl, and hydroxy.
  • Heterocyclyl is a saturated or unsaturated ring containing at least one heteroatom selected from O, N and/or S and 1 to 6 carbon atoms.
  • heterocyclyl contains 1 to 4, more preferably 1 , 2 or 3 heteroatoms selected from O and/or N and is preferably selected from pyrrolidinyl, tetrahydrofuranyl, dihydrofuranyl, tetrahydrothienyl, tetrahydropyranyl, dihydropyranyl, tetrahydrothiopyranyl, piperidino, morpholino, thiomorpholino, thioxanyl, piperazinyl, homopiperazinyl, azetidinyl, oxetanyl, thietanyl, homopiperidinyl, oxepanyl, thiepanyl, oxazepinyl, diazepinyl, thiazepinyl, 1 ,2,3,
  • Alkyl is a saturated hydrocarbon moiety, namely straight chain or branched alkyl having 1 to 10, preferably 1 to 8 carbon atoms, more preferably 1 to 4 carbon atoms, such as methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, n-pentyl, isopentyl, neopentyl, hexyl or heptyl.
  • Cycloalkyl is an alkyl ring having 3 to 10, preferably 3 to 8 carbon atoms, more preferably 3 to 6 carbon atoms, such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl or cyclooctyl.
  • Alkenyl is an unsaturated hydrocarbon moiety with one or more double bonds, preferably one double bond, namely straight chain or branched alkenyl having 1 to 10, preferably 2 to 8 carbon atoms, more preferably 2 to 4 atoms, such as vinyl, allyl, methallyl, buten-2- yl, buten-3-yl, penten-2-yl, penten-3-yl, penten-4-yl, 3-methyl-but-3-enyl, 2-methyl-but-3- enyl, 1-methyl-but-3-enyl, hexenyl or heptenyl.
  • Alkynyl is an unsaturated hydrocarbon moiety with one or more triple bonds, preferably one triple bond, namely straight chain or branched alkynyl having 1 to 10, preferably 2 to 8 carbon atoms, more preferably 2 to 4 atoms, such as ethynyl, propynyl, butyn-2-yl, butyn-3-yl, pentyn-2-yl, pentyn-3-yl, pentyn-4-yl, 2-methyl-but-3-ynyl, 1-methyl-but-3-ynyl, hexynyl or heptynyl.
  • Halo or halogen is a halogen atom preferably selected from F, Cl 1 Br and I 1 preferably F 1 Cl and Br.
  • cycloalkylalkyl, arylalkyl, heretoarylalkyl and heterocyclylalkyl it is contemplated that cycloalkyl, aryl, heretoaryl and heterocyclyl are bonded via an alkylene moiety.
  • This alkylene moiety may be a straight chain or branched chain group.
  • Said alkylene moiety preferably has 1 to 6 carbon atoms.
  • Examples thereof include methylene, ethylene, n-propylene, n-butylene, n-pentylene, n-hexylene, iso-propylene, sec.-butylene, tert.-butylene, 1 ,1 -dimethyl propylene, 1 ,2-dimethyl propylene, 2,2- dimethyl propylene, 1 ,1 -dimethyl butylene, 1 ,2-dimethyl butylene, 1 ,3-dimethyl butylene, 2,2-dimethyl butylene, 2,3-dimethyl butylene, 3,3-dimethyl butylene, 1 -ethyl butylene, 2- ethyl butylene, 3-ethyl butylene, 1 -n-propyl propylene, 2-n-propyl propylene, 1-iso-propyl propylene, 2-iso-propyl propylene, 1 -methyl pentylene, 2-methyl pentylene, 3-methyl pent
  • said alkylene moiety has 1 to 3 carbon atoms, such as methylene, ethylene, n-propylene and iso-propylene. Most preferred is methylene.
  • Preferred embodiments of the compounds according to present invention are shown in scheme 1.
  • the compounds of the present invention can be in the form of a prodrug compound.
  • Prodrug compound means a derivative that is converted into a compound according to the present invention by a reaction with an enzyme, gastric acid or the like under a physiological condition in the living body, e.g. by oxidation, reduction, hydrolysis or the like, each of which is carried out enzymatically.
  • prodrug examples include compounds, wherein the amino group in a compound of the present invention is acylated, alkylated or phosphorylated to form, e.g., eicosanoylamino, alanylamino, pivaloyloxymethylamino or wherein the hydroxyl group is acylated, alkylated, phosphorylated or converted into the borate, e.g. acetyloxy, palmitoyloxy, pivaloyloxy, succinyloxy, fumaryloxy, alanyloxy or wherein the carboxyl group is esterified or amidated.
  • these compounds can be produced from compounds of the present invention according to well-known methods.
  • prodrug examples include compounds, wherein the carboxylate in a compound of the present invention is for example converted into an alkyl-, aryl-, choline-, amino, acyloxymethylester, linolenoyl-ester. Metabolites of compounds of the present invention are also within the scope of the present invention.
  • isomers can be separated by methods well known in the art, e.g. by liquid chromatography. Same applies for enantiomers by using e.g. chiral stationary phases. Additionally, enantiomers may be isolated by converting them into diastereomers, i.e. coupling with an enantiomerically pure auxiliary compound, subsequent separation of the resulting diastereomers and cleavage of the auxiliary residue. Alternatively, any enantiomer of a compound of the present invention may be obtained from stereoselective synthesis using optically pure starting materials.
  • the compounds of the present invention can be in the form of a pharmaceutically acceptable salt or a solvate.
  • pharmaceutically acceptable salts refers to salts prepared from pharmaceutically acceptable non-toxic bases or acids, including inorganic bases or acids and organic bases or acids.
  • the invention also comprises their corresponding pharmaceutically or toxicologically acceptable salts, in particular their pharmaceutically utilizable salts.
  • the compounds of the of the present invention which contain acidic groups can be present on these groups and can be used according to the invention, for example, as alkali metal salts, alkaline earth metal salts or as ammonium salts.
  • salts include sodium salts, potassium salts, calcium salts, magnesium salts or salts with ammonia or organic amines such as, for example, ethylamine, ethanolamine, triethanolamine or amino acids.
  • Compounds of the present invention which contain one or more basic groups, i.e. groups which can be protonated, can be present and can be used according to the invention in the form of their addition salts with inorganic or organic acids.
  • acids examples include hydrogen chloride, hydrogen bromide, phosphoric acid, sulfuric acid, nitric acid, methanesulfonic acid, p-toluenesulfonic acid, naphthalenedisulfonic acids, oxalic acid, acetic acid, tartaric acid, lactic acid, salicylic acid, benzoic acid, formic acid, propionic acid, pivalic acid, diethylacetic acid, malonic acid, succinic acid, pimelic acid, fumaric acid, maleic acid, malic acid, sulfaminic acid, phenylpropionic acid, gluconic acid, ascorbic acid, isonicotinic acid, citric acid, adipic acid, and other acids known to the person skilled in the art.
  • the invention also includes, in addition to the salt forms mentioned, inner salts or betaines (zwitterions).
  • the respective salts can be obtained by customary methods which are known to the person skilled in the art like, for example by contacting these with an organic or inorganic acid or base in a solvent or dispersant, or by anion exchange or cation exchange with other salts.
  • the present invention also includes all salts of the compounds of the present invention which, owing to low physiological compatibility, are not directly suitable for use in pharmaceuticals but which can be used, for example, as intermediates for chemical reactions or for the preparation of pharmaceutically acceptable salts.
  • the present invention provides pharmaceutical compositions comprising a compound of the present invention, or a prodrug compound thereof, or a pharmaceutically acceptable salt or solvate thereof as active ingredient together with a pharmaceutically acceptable carrier.
  • “Pharmaceutical composition” means one or more active ingredients, and one or more inert ingredients that make up the carrier, as well as any product which results, directly or indirectly, from combination, complexation or aggregation of any two or more of the ingredients, or from dissociation of one or more of the ingredients, or from other types of reactions or interactions of one or more of the ingredients. Accordingly, the pharmaceutical compositions of the present invention encompass any composition made by admixing a compound of the present invention and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition of the present invention may additionally comprise one or more other compounds as active ingredients like one or more additional compounds of the present invention, or a prodrug compound or other MEK inhibitors.
  • the compositions include compositions suitable for oral, rectal, topical, parenteral (including subcutaneous, intramuscular, and intravenous), ocular (ophthalmic), pulmonary (nasal or buccal inhalation), or nasal administration, although the most suitable route in any given case will depend on the nature and severity of the conditions being treated and on the nature of the active ingredient. They may be conveniently presented in unit dosage form and prepared by any of the methods well-known in the art of pharmacy.
  • said compounds and pharmaceutical composition are for the treatment of cancer such as brain, lung, squamous cell, bladder, gastic, pancreatic, breast, head, neck, renal, kidney, ovarian, prostate, colorectal, oesohageal, testicular, gynecological or thyroid cancer.
  • said pharmaceutical composition is for the treatment of a noncancerous hyperproliferative disorder such as benign hyperplasia of the skin (e.g., psoriasis), restenosis, or prostate (e.g. benign prostatic hypertrophy (BPH)).
  • benign hyperplasia of the skin e.g., psoriasis
  • restenosis e.g. benign prostatic hypertrophy (BPH)
  • the invention also relates to a compound or pharmaceutical composition for the treatment of pancreatitis or kidney disease (including proliferative glomerulonephtitis and diabetes induced renal disease) or pain in a mammal which comprises a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt, prodrug or hydrate thereof, and a pharmaceutically acceptable carrier.
  • the invention also relates to a compound or pharmaceutical composition for the prevention of blastocyte implantation in a mammal which comprises a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt, prodrug or hydrate thereof, and a pharmaceutically acceptable carrier.
  • the invention also relates to a compound or pharmaceutical composition for treating a disease related to vasculogenesis or angiogenesis in a mammal which comprises a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt, prodrug or hydrate thereof, and a pharmaceutically acceptable carrier.
  • said compound or pharmaceutical composition is for treating a disease selected from the group consisting of tumor angiogenesis, chronic inflammatory disease such as rheumatoid arthritis, inflammatory bowel disease, atherosclerosis, skin diseases such as psoriasis, eczema, and sclerodema, diabetes, diabetic retinopathy, retinopathy of prematurity, age-related macular degeneration, hemangioma, glioma, melanoma, Kaposi's sarcoma and ovarian, breast, lung, pancreatic, prostate, colon and epidermoid cancer.
  • a disease selected from the group consisting of tumor angiogenesis, chronic inflammatory disease such as rheumatoid arthritis, inflammatory bowel disease, atherosclerosis, skin diseases such as psoriasis, eczema, and sclerodema
  • diabetes diabetic retinopathy
  • retinopathy of prematurity age-related macular degeneration
  • the invention also relates to of the use for treating a hyperproliferative disorder in a mammal that comprises administering to said mammal a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt, prodrug or hydrate thereof.
  • said use relates to the treatment of cancer such as brain, lung, squamous cell, bladder, gastic, pancreatic, breast, head, neck, renal, kidney, ovarian, prostate, colorectal, oesohageal, testicular, gynecological or thyroid cancer.
  • said use relates to the treatment of a non-cancerous hyperproliferative disorder such as benign hyperplasia of the skin (e.g., psoriasis), restenosis, or prostate (e.g., benign prostatic hypertrophy (BPH)).
  • a non-cancerous hyperproliferative disorder such as benign hyperplasia of the skin (e.g., psoriasis), restenosis, or prostate (e.g., benign prostatic hypertrophy (BPH)).
  • the invention also relates to a use for the treatment of a hyperproliferative disorder in a mammal that comprises administering to said mammal a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt, prodrug or hydrate thereof, in combination with an anti-tumor agent selected from the group consisting of mitotic inhibitors, alkylating agents, antimetabolites, intercalating antibiotics, growth factor inhibitors, cell cycle inhibitors, enzyme inhibitors, topoisomerase inhibitors, biological response modifiers, antihormones, angiogenesis inhibitors, and anti- androgens.
  • an anti-tumor agent selected from the group consisting of mitotic inhibitors, alkylating agents, antimetabolites, intercalating antibiotics, growth factor inhibitors, cell cycle inhibitors, enzyme inhibitors, topoisomerase inhibitors, biological response modifiers, antihormones, angiogenesis inhibitors, and anti- androgens.
  • the invention also relates to a use of treating pancreatitis or kidney disease or pain in a mammal that comprises administering to said mammal a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt, prodrug or hydrate thereof.
  • the invention also relates to a use of preventing blastocyte implantation in a mammal that comprises administering to said mammal a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt, prodrug or hydrate thereof.
  • the invention also relates to a use of treating diseases related to vasculogenesis or angiogenesis in a mammal that comprises administering to said mammal a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt, prodrug or hydrate thereof.
  • said method is for treating a disease selected from the group consisting of tumor angiogenesis, chronic inflammatory disease such as rheumatoid arthritis, atherosclerosis, inflammatory bowel disease, skin diseases such as psoriasis, eczema, and scleroderma, diabetes, diabetic retinopathy, retinopathy of prematurity, age-related macular degeneration, hemangioma, glioma, melanoma, Kaposi's sarcoma and ovarian, breast, lung, pancreatic, prostate, colon and epidermoid cancer.
  • a disease selected from the group consisting of tumor angiogenesis, chronic inflammatory disease such as rheumatoid arthritis, atherosclerosis, inflammatory bowel disease, skin diseases such as psoriasis, eczema, and scleroderma
  • diabetes diabetic retinopathy, retinopathy of prematurity, age-related macular degeneration, hemangio
  • Patients that can be treated with compounds of the present invention, or pharmaceutically acceptable salts, prodrugs and hydrates of said compounds, according to the methods of this invention include, for example, patients that have been diagnosed as having psoriasis, restenosis, atherosclerosis, BPH, lung cancer, bone cancer, CMML, pancreatic cancer, skin cancer, cancer of the head and neck, cutaneous or intraocular melanoma, uterine cancer, ovarian cancer, rectal cancer, cancer of the anal region, stomach cancer, colon cancer, breast cancer, testicular, gynecologic tumors (e.g., uterine sarcomas, carcinoma of the fallopian tubes, carcinoma of the endometrium, carcinoma of the cervix, carcinoma of the vagina or carcinoma of the vulva), Hodgkin's disease, cancer of the esophagus, cancer of the small intestine, cancer of the endocrine system (e.g., cancer of the thyroid, parathyroid or adrenal glands
  • This invention also relates to a compound or pharmaceutical composition for inhibiting abnormal cell growth in a mammal which comprises an amount of a compound of the present invention, or a pharmaceutically acceptable salt or solvate or prodrug thereof, in combination with an amount of a chemotherapeutic, wherein the amounts of the compound, salt, solvate, or prodrug, and of the chemotherapeutic are together effective in inhibiting abnormal cell growth.
  • chemotherapeutics are presently known in the art.
  • the chemotherapeutic is selected from the group consisting of mitotic inhibitors, alkylating agents, anti-metabolites, intercalating antibiotics, growth factor inhibitors, cell cycle inhibitors, enzymes, topoisomerase inhibitors, biological response modifiers, anti-hormones, angiogenesis inhibitors, and anti-androgens.
  • This invention further relates to a method for inhibiting abnormal cell growth in a mammal or treating a hyperproliferative disorder which method comprises administering to the mammal an amount of a compound of the present invention, or a pharmaceutically acceptable salt or solvate or prodrug thereof, in combination with radiation therapy, wherein the amounts of the compound, salt, solvate, or prodrug, is in combination with the radiation therapy effective in inhibiting abnormal cell growth or treating the hyperproliferative disorder in the mammal.
  • Techniques for administering radiation therapy are known in the art, and these techniques can be used in the combination therapy described herein.
  • the administration of the compound of the invention in this combination therapy can be determined as described herein.
  • this invention further relates to a method for sensitizing abnormal cells in a mammal to treatment with radiation which comprises administering to the mammal an amount of a compound of the present invention or pharmaceutically acceptable salt or solvate or prodrug thereof, which amount is effective is sensitizing abnormal cells to treatment with radiation.
  • the amount of the compound, salt, or solvate in this method can be determined according to the means for ascertaining effective amounts of such compounds described herein.
  • the invention also relates to a method of and to a pharmaceutical composition of inhibiting abnormal cell growth in a mammal which comprises an amount of a compound of the present invention, or a pharmaceutically acceptable salt or solvate thereof, a prodrug thereof, or an isotopically-labeled derivative thereof, and an amount of one or more substances selected from anti-angiogenesis agents, signal transduction inhibitors, and antiproliferative agents.
  • the compounds of the present invention can be combined as the active ingredient in intimate admixture with a pharmaceutical carrier according to conventional pharmaceutical compounding techniques.
  • the carrier may take a wide variety of forms depending on the form of preparation desired for administration, e.g., oral or parenteral (including intravenous).
  • any of the usual pharmaceutical media may be employed, such as, for example, water, glycols, oils, alcohols, flavoring agents, preservatives, coloring agents and the like in the case of oral liquid preparations, such as, for example, suspensions, elixirs and solutions; or carriers such as starches, sugars, microcrystalline cellulose, diluents, granulating agents, lubricants, binders, disintegrating agents and the like in the case of oral solid preparations such as, for example, powders, hard and soft capsules and tablets, with the solid oral preparations being preferred over the liquid preparations.
  • oral liquid preparations such as, for example, suspensions, elixirs and solutions
  • carriers such as starches, sugars, microcrystalline cellulose, diluents, granulating agents, lubricants, binders, disintegrating agents and the like in the case of oral solid preparations such as, for example, powders, hard and soft capsules and tablets, with the solid oral preparations being preferred over the liquid preparation
  • tablets and capsules represent the most advantageous oral dosage unit form in which case solid pharmaceutical carriers are obviously employed. If desired, tablets may be coated by standard aqueous or nonaqueous techniques. Such compositions and preparations should contain at least 0.1 percent of active compound. The percentage of active compound in these compositions may, of course, be varied and may conveniently be between about 2 percent to about 60 percent of the weight of the unit. The amount of active compound in such therapeutically useful compositions is such that an effective dosage will be obtained.
  • the active compounds can also be administered intranasally as, for example, liquid drops or spray.
  • the tablets, pills, capsules, and the like may also contain a binder such as gum tragacanth, acacia, corn starch or gelatin; excipients such as dicalcium phosphate; a disintegrating agent such as corn starch, potato starch, alginic acid; a lubricant such as magnesium stearate; and a sweetening agent such as sucrose, lactose or saccharin.
  • a dosage unit form is a capsule, it may contain, in addition to materials of the above type, a liquid carrier such as a fatty oil.
  • tablets may be coated with shellac, sugar or both.
  • a syrup or elixir may contain, in addition to the active ingredient, sucrose as a sweetening agent, methyl and propylparabens as preservatives, a dye and a flavoring such as cherry or orange flavor.
  • Compounds of the present invention may also be administered parenterally. Solutions or suspensions of these active compounds can be prepared in water suitably mixed with a surfactant such as hydroxy-propylcellulose. Dispersions can also be prepared in glycerol, liquid polyethylene glycols and mixtures thereof in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
  • the pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions.
  • the form must be sterile and must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi.
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (e.g., glycerol, propylene glycol and liquid polyethylene glycol), suitable mixtures thereof, and vegetable oils.
  • Any suitable route of administration may be employed for providing a mammal, especially a human, with an effective dose of a compound of the present invention.
  • oral, rectal, topical, parenteral, ocular, pulmonary, nasal, and the like may be employed.
  • Dosage forms include tablets, troches, dispersions, suspensions, solutions, capsules, creams, ointments, aerosols, and the like.
  • compounds of the present invention are administered orally.
  • the effective dosage of active ingredient employed may vary depending on the particular compound employed, the mode of administration, the condition being treated and the severity of the condition being treated. Such dosage may be ascertained readily by a person skilled in the art.
  • the compounds of the present invention are administered at a daily dosage of from about 0.1 milligram to about 100 milligram per kilogram of animal body weight, preferably given as a single daily dose or in divided doses two to six times a day, or in sustained release form.
  • the total daily dosage is from about 1.0 milligrams to about 1000 milligrams, preferably from about 1 milligram to about 50 milligrams. In the case of a 70 kg adult human, the total daily dose will generally be from about 7 milligrams to about 350 milligrams. This dosage regimen may be adjusted to provide the optimal therapeutic response.
  • LiHMDS Lithium hexamethyldisilazide
  • the compounds of the present invention can be prepared according to the procedures of the following Schemes and Examples, using appropriate materials and are further exemplified by the following specific examples. Moreover, by utilizing the procedures described herein, in conjunction with ordinary skills in the art, additional compounds of the present invention claimed herein can be readily prepared.
  • the compounds illustrated in the examples are not, however, to be construed as forming the only genus that is considered as the invention.
  • the examples further illustrate details for the preparation of the compounds of the present invention. Those skilled in the art will readily understand that known variations of the conditions and processes of the following preparative procedures can be used to prepare these compounds.
  • the instant compounds are generally isolated in the form of their pharmaceutically acceptable salts, such as those described above.
  • the amine-free bases corresponding to the isolated salts can be generated by neutralization with a suitable base, such as aqueous sodium hydrogencarbonate, sodium carbonate, sodium hydroxide and potassium hydroxide, and extraction of the liberated amine-free base into an organic solvent, followed by evaporation.
  • a suitable base such as aqueous sodium hydrogencarbonate, sodium carbonate, sodium hydroxide and potassium hydroxide
  • the amine-free base, isolated in this manner can be further converted into another pharmaceutically acceptable salt by dissolution in an organic solvent, followed by addition of the appropriate acid and subsequent evaporation, precipitation or crystallisation.
  • ethyl 4,6-dichloronicotinate can be reacted with an appropriately substituted aniline in an inert solvent, preferable THF, by addition of a base, preferably but not limited to LiHMDS to yield substituted ethyl 6-chloro-4arylaminonicotinate 1.
  • a base preferably but not limited to LiHMDS
  • Heating with hydrazine hydrate leads to an intermediate hydrazino compound which is acylated by coupling with an activated carboxylic acid building block. Cyclisation is achieved by heating in acidic solvents such as AcOH or PPA.
  • the resulting triazolopyridine is saponified in the next step by heating with lithium hydroxide in a water- alcohol mixture to give triazolopyridine carboxylic acid 2.
  • the latter can be further derivatised by coupling with a substituted O-alkylhydroxylamine using a coupling reagent such as PyBro, PyBOP or DCC for example to give substituted hydroxa
  • Scheme 3 illustrates the preparation of compounds of the present invention where W is heterocyclic.
  • Triazolopyridine carboxylic acid 2 can be coupled with Boc-hydrazine by using a coupling reagent such as PyBOP or DCC for example and the Boc protecting group is then removed by treatment with an acid such as HCI.
  • the resulting hydrazide 4 is further derivatised by reacting with an appropriately substituted isocyanate. Cyclisation can be achieved by heating with triphenyl phosphine and CCI 4 in a suitable solvent to give oxadiazole compound 6.
  • Suitable anilines, carboxylic acids, O-alkyl hydroxylamines, and isocyanates are commercially available from Sigma-Aldrich Chemie GmbH, Kunststoff, Germany or from Acros Organics, Belgium or from Fisher Scientific GmbH, 58239 Schense, Germany or can be routinely prepared by procedures described in "March's Advanced Organic Chemistry: Reactions, Mechanisms, and Structure", 5th Edition; John Wiley & Sons. O- [(4S)2,2-Dimethyl-[1,3]dioxolan-4-ylmethyl]-hydroxylamine and O-[(4ft)2,2-dimethyl-
  • [1,3]dioxolan-4-ylmethyl]-hydroxylamine are prepared according to a procedure described in WO02/06213 A2.
  • Ethyl 4,6-dichloronicotinate is synthesised in three steps from diethyl acetone dicarboxylate according to a literature procedure (DenHertog, Reel Trav Chim Pays- Bas 1946, 65, 129-140).
  • Analytical LC/MS was performed using Reprosil-Pur ODS3, 5 ⁇ M, 1 x 60 mm columns at a flow rate of 250 ⁇ l/min, sample loop 2.5 ⁇ l; retention times are given in minutes.
  • Methods are: (I) runs on a LC10Advp-Pump (Shimadzu) with SPD- MIOAvp UV/Vis diode array detector and QP2010 MS-detector in ESI+ modus with UV- detection at 214, 254 and 275 nm with a gradient of 15-95% acetonitrile (B) in water (A) (0.1 % formic acid), 5 min.
  • Ethyl 4,6-dichloronicotinate (2.Og, 9.1mmol) and 4-bromo-2-methylaniline (1.7g, 9.1mmol) are dissolved in dry THF (20ml) under argon and the mixture is cooled to -78°C.
  • a solution of LiHMDS (1.0M in THF, 32ml) is slowly added and the reaction mixture is allowed to warm to ambient temperature. After 18h the reaction is quenched by adding dilute hydrochloric acid (1.0M, 20.0ml) and the mixture is extracted with DCM (3x 60ml).
  • step A The crude material from step A is dissolved in MeOH (1.8ml) and water (0.2ml).
  • Dowex50X8 resin (20mg) is added and the mixture is heated to 120 0 C with microwave irradiation for 20min, the solution is filtered and evaporated.
  • the crude material is purified by preparative HPLC to give 18.6mg (41.4 ⁇ mol, 30%yield, two steps) of pure hydroxamate 3b.
  • Ethyl 4,6-dichloronicotinate (1) (3.Og, 14mmol) and 4-iodo-2-methylaniline (3.2g, 14mmol) are dissolved in dry THF (20ml) under argon and the mixture is cooled to -78°C.
  • a solution of LiHMDS (1.0M in THF, 48ml) is slowly added and the reaction mixture is allowed to warm to ambient temperature.
  • the reaction is quenched after 2Oh by adding dilute hydrochloric acid (1.0M, 20.0ml), the volatiles are evaporated and the residue is extracted with DCM (3x 60ml).
  • Ethyl 7-[(4-iodo-2-methylphenyl)amino]-3-methyl[1,2,4]triazolo[4,3-a]pyridine-6-carboxylate (130mg, 0.298mmol) is dissolved in THF (6ml), water (1ml) and LiOH hydrate (68mg, 1.62mmol) are added and the mixture is heated for 30 min at 120 0 C with microwave irradiation. Hydrochloric acid (1 N in dioxane, 4ml) is added and the volatiles are removed under reduced pressure.
  • the activity of the compounds of the present invitation may be determined by the following procedure: Inhibition of human MEK1 kinase activity was monitored with a homogenous, fluorescence based assay.
  • the assay uses time resolved fluorescence resonance energy transfer to probe for phosphorylation of ERK1 by MEK1.
  • the assay is carried out in low volume 96 well microtiterplates. In a total volume of 15 ⁇ l, compounds are incubated with 10OnM MEK1 , 15 ⁇ M ATP, 30OnM ERK2 employing a buffer containing 2OmM TRIS/HCI, 10 mM MgCI2, 100 ⁇ M NaVO4, 1 mM DTT, and 0.005% Tween 20 (pH 7.4).
  • Assay 2 Tumor cell proliferation assays (ATP Lite)
  • Murine colon C26, human melanoma A375 and human melanoma Mel5 cells were plated in 96 well Corning white plates (1500 cells/well for C26, and 2000 cells/well for A375, and MiaPaCa-2) and cultured overnight at 37 0 C in 5% CO2. Inhibitors were serially diluted in 100 % DMSO and subsequently added to cells to reach a final concentration of 0.25% DMSO. The cells were incubated for 4 days in the presence of test compounds in cell growth media (DMEM with 10% fetal bovine serum, 2mM glutamine for C26, and MiaPaCa-2, and RPMI with 10% fetal bovine serum, 2mM glutamine for A375).
  • DMEM fetal bovine serum
  • 2mM glutamine for C26 and MiaPaCa-2
  • RPMI 10% fetal bovine serum, 2mM glutamine for A375
  • Microsomal stability assay Compounds were tested on their stability in human, rat and mouse liver microsomal preparations (HLM, RLM and MLM respectively). At a final concentration of 3 ⁇ M, compounds were incubated at 37°C with 0.5 mg/ml human, rat or mouse liver microsomes in a buffer containing 50 mM phosphate, pH 7.4 and 2 mM NADPH. Pooled human liver microsomes or pooled male rat liver microsomes (Sprague Dawley) were obtained from NatuTec (Frankfurt, Germany). Incubations without NADPH served as negative controls.
  • Caco-2 cells obtained from the ATCC at passage number 27 are used.
  • Cells (passage number 40-60) were seeded on to Millipore Multiscreen Caco-2 plates or Falcon HTS inserts at 1 x 105 cells/cm2.
  • Cells were cultured for 20 days in DMEM and media was changed every two or three days. On day 20 the permeability study was performed.
  • HBSS Hanks Balanced Salt Solution
  • HBSS was then removed from the apical compartment and replaced with test compound dosing solutions.
  • the solutions were made by diluting 1OmM DMSO concentrates with HBSS to give a final test compound concentration of 10 ⁇ M (final DMSO concentration adjusted to 1%).
  • the fluorescent integrity marker lucifer yellow was also included in the dosing solution.
  • Analytical standards were made from dosing solutions. Test compound permeability was assessed in duplicate. On each plate compounds of known permeability characteristics were run as controls.
  • the apical compartment inserts were then placed into 'companion' plates containing fresh HBSS. For basolateral to apical (B-A) experiments the experiment was initiated by replacing buffer in the inserts then placing them in companion plates containing dosing solutions. At 120 minutes the companion plate was removed and apical and basolateral samples diluted for analysis by LC-MS/MS (the donor compartment was also sampled to permit determination of starting concentration after non-specific binding has occurred).
  • Lucifer yellow permeation was low if monolayers have not been damaged.
  • Test and control compounds were quantified by LC-MS/MS cassette analysis using a 5-point calibration with appropriate dilution of the samples. Should lucifer yellow Papps were above QC limits in more than one well per test compound, the compound was re-tested.
  • dQ/dt is the rate of permeation of the drug across the cells
  • C 0 is the donor compartment concentration at time zero and A is the area of the cell monolayer.
  • C 0 is obtained from analysis of the donor compartment at the end of the incubation period. Test compounds were grouped into low, medium or high absorption potential based on comparison with control compounds, which have known human absorption.
  • Results are shown in Table 2. Wherein "+” means a caco A-B and caco B-A value of 1-10 and “++”means a caco A-B and caco B-A value of 11 -100 and "n.d.” means not determined.

Abstract

The invention provides novel, substituted 7-arylamino[1,2,4]triazolo[4,3-a]pyridine compounds Formula (I): pharmaceutically acceptable salts, solvates and prodrug compounds thereof, wherein W, R1, R2, R9, R10 R11, R12, R13, R14 and L are as defined in the specification. Such compounds are MEK inhibitors and useful in the treatment of hyperproliferative diseases, such as cancer, restenosis and inflammation. Also disclosed is the use of such compounds in the treatment of hyperproliferative diseases in mammals, especially humans, and pharmaceutical compositions containing such compounds.

Description

[1,2,4] triazolo [4,3-A] PYRIDINE DERIVATIVES FOR THE TREATMENT OF HYPERPROLIFERATIVE DISEASES
Field of the invention
The invention relates to a series of substituted 7-arylamino[1 ,2,4]triazolo[4,3-a]pyridine derivatives that are useful in the treatment of hyperproliferative diseases, such as cancer and inflammation, in mammals. Also disclosed is the use of such compounds in the treatment of hyperproliferative diseases in mammals, especially humans, and pharmaceutical compositions containing such compounds.
Summary of the related art
The Ras/Raf/MEK/ERK pathway is a central signal transduction pathway, which transmits signals from multiple cell surface receptors to transcription factors in the nucleus, which regulate gene expression. This pathway is frequently referred to as the MAP kinase pathway as MAPK stands for mitogeπ-activated protein kinase indicating that this pathway can be stimulated by mitogens, cytokines and growth factors (Steelman et al., Leukemia 2004, 18, 189-218). Depending upon the stimulus and cell type, this pathway can transmit signals, which result in the prevention or induction of apoptosis or cell cycle progression. The Ras/Raf/MEK/ERK pathway has been shown to play important roles in cell proliferation and the prevention of apoptosis. Aberrant activation of this pathway is commonly observed in malignantly transformed cells. Amplification of ras proto-oncogenes and activating mutations that lead to the expression of constitutively active Ras proteins are observed in approximately 30% of all human cancers (Stirewalt et al., Blood 2001, 97, 3589-95). Mutated, oncogenic forms of Ras are found in 50% of colon and >90% pancreatic cancers as well as many other types of cancers (Kohl et al., Science 1993, 260, 1834-1837). The effects of Ras on proliferation and tumorigenesis have been documented in immortal cell lines (McCubrey et al., lnt J Oncol 1995, 7, 295-310). ύRaf mutations have been identified in more than 60% of malignant melanoma (Davies, H et al., Nature 2002, 417, 949-954). Given the high level of mutations that have been detected at Ras, this pathway has always been considered a key target for therapeutic intervention (Chang et al., Leukemia 2003, 17,1263-93).
The Ras/Raf/MEK/ERK signaling pathway can exert proliferative or antiproliferative effects through downstream transcription factor targets including NF-κB, CREB, Ets-1 , AP-1 and c-Myc. ERKs can directly phosphorylate Ets-1 , AP-1 and c-Myc, which lead to their activation. Alternatively, ERKs can phosphorylate and activate a downstream kinase target RSK, which then phosphorylates and activates transcription factors, such as CREB. These transcription factors induce the expression of genes important for cell cycle progression, for example, Cdks, cyclins, growth factors, and apoptosis prevention, for example, antiapoptotic Bcl-2 and cytokines. Overall, treatment of cells with growth factors leads to the activation of ERKs which results in proliferation and, in some cases, differentiation (Lewis et al., Adv. Cancer Res, 1998, 74, 49-139).
MEK proteins are the primary downstream targets of Raf. The MEK family of genes consists of five genes: MEK1 , MEK2, MEK3, MEK4 and MEK5. This family of dual- specificity kinases has both serine/threonine and tyrosine kinase activity. The structure of MEK consists of an amino-terminal negative regulatory domain and a carboxy-terminal MAP kinase-binding domain, which is necessary for binding and activation of ERKs. Deletion of the regulatory MEK1 domain results in constitutive MEK1 and ERK activation (Steelman et al., Leukemia 2004, 18, 189-218).
MEK1 is a 393-amino-acid protein with a molecular weight of 44 kDa (Crews et al., Science 1992, 258, 478-80). MEK1 is modestly expressed in embryonic development and is elevated in adult tissue with the highest levels detected in brain tissue. MEK1 - requires phosphorylation of S218 and S222 for activation, and substitution of these residues with D or glutamic acid (E) led to an increase in activity and foci formation in NIH3T3 cells (Huang et al., MoI Biol Cell, 1995, 6, 237-45). Constitutive activity of MEK1 in primary cell culture promotes senescence and induces p53 and p16INK4a, and the opposite was observed in immortalized cells or cells lacking either p53 or p16INK4a (Lin et al., Genes Dev, 1998 , 12, 3008-3019). Constitutive activity of MEK1 inhibits NF- KB transcription by negatively regulating p38MAPK activity (Carter et al., J Biol Chem 2000, 275, 27858-64). The main physiological substrates of MEK are the members of the ERK (extracellular signal-regulated kinase) or MAPK (mitogen activated protein kinase) family of genes. Aberrant expression of MEK1 has been detected in many different types of cancer, and mutated forms of MEK1 will transform fibroblast, hematopoietic and other cell types.
Constitutive activation of MEK1 results in cellular transformation. It therefore represents a likely target for pharmacological intervention in proliferative and inflammatory diseases (Lee et al., Nature 1994, 372, 739-746; Dudley et al., Proc. Natl. Acad. Sci. U.S.A. 1995, 92, 7686-7689).
Useful inhibitors of MEK have been developed that show potential therapeutic benefit in several studies. For example, small molecule MEK inhibitors have been shown to inhibit human tumor growth in nude mouse xenografts (Yeh, T. et al, Proceedings of the American Association of Cancer Research 2004, 45, Abs 3889 and Lee, P. et al., Proceedings of the American Association of Cancer Research 2004, 45, Abs 3890). MEK inhibitors also entered clinical trials, i.e. ARRY142886 (Wallace, E. et al, Proceedings of the American Association of Cancer Research 2004, 45, Abs 3891), PD-0325901 (Swanton C, Johnston S IDDB MEETING REPORT 2003, February 13-1 ) and PD- 184352 (Waterhouse et al., Proceedings of the American Society for Clinical Oncology 2003, 22, Abs 816).
Compounds suitable as MEK inhibitors are also disclosed in US 5,525,625; WO 98/43960; WO 99/01421; WO 99/01426; WO 00/41505; WO 00/42002; WO 00/42003; WO 00/41994; WO 00/42022; WO 00/42029; WO 00/68201 ; WO 01/68619; WO 02/06213; WO03/035626; A2; WO 03/077855; WO03/077914; WO2004/005284; WO2004/056789.
However, PD-184352 was lacking efficacy in clinical phase Il trials. Tumors were much less responsive, as no partial responses and only a few patients with stable disease were observed. As a result, the clinical trials of this molecule were suspended (Mclnnes C IDDB MEETING REPORT 2003). PD-184352 was limited by poor solubility, high metabolic clearance and low bioavailability. This exemplifies the need for novel MEK inhibitors with superior pharmacological properties.
Description of the invention In view of the foregoing it is the object of the present invention to provide novel MEK inhibitors useful in the treatment of hyperproliferative diseases related to the hyperactivity of MEK as well as diseases modulated by the MEK cascade, such as cancer and inflammation, in mammals with superior pharmacological properties both with respect to their activities as well as their solubility, metabolic clearance and bioavailability characteristics.
As a result, this invention provides novel, substituted 7-arylamino[1 ,2,4]triazolo[4,3- a]pyridine derivatives and pharmaceutically acceptable salts, solvates or prodrugs thereof, that are MEK inhibitors and useful in the treatment of the above mentioned diseases. The compounds are defined by Formula (I): Formula (I)
Figure imgf000005_0001
a pharmaceutically acceptable salt, solvate or prodrug thereof, wherein:
Ri, R2, R9, R11 R12, R13 and R14 are independently selected from hydrogen, halogen, cyano, nitro, azido, -OR3, -C(O)R3, -C(O)OR3, -NR4C(O)OR6, -OC(O)R3, -NR4S(O)jR6 , -S(O)JNR3R4, -S(O)jNR4C(O)R3, -C(O)NR4S(O)jR6, S(O)jR6,
-NR4C(O)R3, -C(O)NR3R4, -NR5C(O)NR3R4, -NR5C(NCN)NR3R4, -NR3R4 and C1- C10 alkyl, C2-C10 alkenyl, C2-C10 alkynyl, C3-C10 cycloalkyl, C3-C10 cycloalkylalkyl, -S(O)J(C1-C6 alkyl), -S(O)j(CR4R5)m-aryl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocyclyl, heterocyclylalkyl, -O(CR4R5)m-aryl, -NR4(CR4R5)m-aryl, -O(CR4R5)m- heteroaryl, -NR4(CR4R5)m-heteroaryl, -O(CR4R5)m-heterocyclyl, -NR4(CR4R5)m- heterocyclyl, and -S(C1-C2 alkyl) substituted with 1 to 5 F, where each alkyl, alkenyl, alkynyl, cycloalkyl, aryl, heteroaryl and heterocyclyl is substituted or unsubstituted;
R10 is selected from hydrogen, -OR3, -C(O)R31-C(O)OR3, -NR4C(O)OR6, -OC(O)R3, -NR4S(O)jR6 , -S(O)jNR3R4, -S(O)jNR4C(O)R3, -C(O)NR4S(O)jR6l S(O)JR6,
-NR4C(O)R3, -C(O)NR3R4, -NR5C(O)NR3R4, -NR5C(NCN)NR3R4, -NR3R4 ; -S(O)J(C1-C6 alkyl), -S(O)j(CR4R5)m-aryl, -O(CR4R5)m-aryl, -NR4(CR4R5)m-aryl, -O(CR4R5)m-heteroaryl, -NR4(CR4R5)m-heteroaryl, -O(CR4R5)m-heterocyclyl, -NR4(CR4R5)m-heterocyclyl, and -S(C1-C2 alkyl) substituted with 1 to 5 F, where each, aryl, heteroaryl and heterocyclyl is substituted or unsubstituted;
L is selected from a bond, C1-C10 alkyl, C2-C10 alkenyl, C2-C10 alkynyl, C3-C10 cycloalkyl, C3-C10 cycloalkylalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocyclyl, heterocyclylalkyl, where each alkyl, alkenyl, alkynyl, cycloalkyl, aryl, heteroaryl and heterocyclyl is unsubstituted or substituted; R3 is selected from hydrogen, trifluoromethyl, C1-C10 alkyl, C2-10 alkenyl, C2-C10 alkynyl, C3-C10 cycloalkyl, C3-C10 cycloalkylalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocyclyl, and heterocyclylalkyl, where each alkyl, alkenyl, alkynyl, cycloalkyl, aryl, heteroaryl and heterocyclyl is substituted or unsubstituted;
R4 is selected from hydrogen or C1-C6 alkyl whereby alkyl may be substituted or unsubstituted; or R3 and R4 can be taken together with the atom to which they are attached to form a 4 to 10 membered heteroaryl or heterocyclic ring, each of which is substituted or unsubstituted;
R5 is selected from hydrogen or CrC6 alkyl whereby alkyl may be substituted or unsubstituted; or
R4 and R5 can be taken together with the atom to which they are attached to form a 4 to 10 membered carbocyclic, heteroaryl or heterocyclic ring, each of which is substituted or unsubstituted;
R6 is selected from trifluoromethyl,Ci-Ci0 alkyl, C3-Ci0 cycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocyclyl, and heterocyclylalkyl, where each alkyl, cycloalkyl, aryl, heteroaryl and heterocyclyl substituted or unsubstituted;
W is selected from heteroaryl containing 1-4 heteroatoms or heterocyclyl containing 1-4 heteroatoms each of which is unsubstituted or substituted by 1 to 5 substituents ZR15; or W is -C(O)ORi5, -C(O)NR4R15, -C(O)NR4OR15, -C(O)(C3-C10 cycloalkyl), -C(O)(C2-C10 alkyl), -C(O)(aryl), -C(O)(heteroaryl), -C(O)(heterocyclyl),
-S(O)JNR4Ri5, -S(O)jNR4OR15, -S(O)jNR4C(O)Ri5, or -C(O)NR4S(O)jR6, whereby R4 and R15 are as defined herein or may form together a 3 to 7 membered ring with 1 or 2 N atoms and optionally an O atom,
Z is a bond, NR16, O, NR16SO2 or S, R15 is independently selected from hydrogen, trifluoromethyl, C1-Ci0 alkyl, C2-C10 alkenyl, C2-C10 alkynyl, C3-C10 cycloalkyl, C3-C10 cycloalkylalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocyclyl, and heterocyclylalkyl, where each alkyl, alkenyl, alkynyl, cycloalkyl, aryl, heteroaryl and heterocyclyl is substituted or unsubstituted;
R16 is selected from hydrogen or C1-C10 alkyl, or R15 and R16 form together a 4 to 10 membered cyclic ring with 1 or 2 N atoms and optionally an O atom, said ring being substituted or unsubstituted; m is O, 1 , 2, 3, 4 or 5 ;and j is 1 or 2.
In a preferred embodiment, the variants R1-R16, L, W and Z are defined as above on pages 4 to 5 but with the proviso that the compounds with the following formula including resolved enantiomers, diastereomers, solvates and pharmaceutically acceptable salts thereof are excluded:
Figure imgf000007_0001
wherein Y is NH;
R1, R2, R7, R8, R9 and R10 are independently hydrogen, halogen, cyano, nitro, azido, - OR3, -C(O)R3, -C(O)OR3, -NR4C(O)OR6, -OC(O)R3, -NR4SO2R6, -SO2NR3R4,
-NR4C(O)R3, -C(O)NR3R4, -NR5C(O)NR3R4, -NR5C(NCN)NR3R4, -NR3R4, C1-C10 alkyl, C2-Ci0 alkenyl, C2-Ci0 alkynyl, C3-Ci0 cycloalkyl, C3-Ci0 cycloalkylalkyl, -S(O)](Ci-C6 alkyl), -S(O)j(CR4R5)m-aryl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocyclyl, heterocyclylalkyl, -O(CR4R5)m-aryl, -NR5(CR4R5)m-aryl, -O(CR4R5)m- heteroaryl, -NR4(CR4R5)m-heteroaryl, -O(CR4R5)m-heterocyclyl or - NR4(CR4R5)m- heterocyclyl, wherein any of said alkyl, alkenyl, alkynyl, cycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocyclyl and heterocyclylalkyl portions are optionally substituted with one or more groups independently selected from oxo, halogen, cyano, nitro, trifluoromethyl, difluoromethoxy, trifluoromethoxy, azido, -NR4SO2R6, -SO2NR3R4, -C(O)R3, -C(O)OR3, -OC(O)R3, -NR4C(O)OR6, -NR4C(O)R3,
-C(O)NR3R4, -NR3R4, -NR5C(O)NR3R4, -NR5C(NCN)R3R4, -OR3, aryl, heteroaryl, arylalkyl, heteroarylalkyl, heterocyclyl, and heterocyclylalkyl,
R3 is hydrogen, trifluoromethyl, Ci-Ci0 alkyl, C2-Ci0 alkenyl, C2-Ci0 alkynyl, C3-Ci0- cycloalkyl, C3-Ci0 cycloalkylalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocyclyl, heterocyclylalkyl, wherein any of said alkyl, alkenyl, alkynyl, cycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocyclyl and heterocyclylalkyl portions are optionally substituted with one or more groups independently selected from oxo, halogen, cyano, nitro, trifluoromethyl, difluoromethoxy, trifluoromethoxy, azido, -NR SO2R " ", -SO2NR R", -C(O)R', -C(O)OR", -OC(O)R", -NR C(O)OR " ", -NRC(O)R", -C(O)NR R ", -SR", -S(O)R " ", -SO2R " ", -NR R ", -NR C(O)NR1 R"", -NR'C(NCN)N"R ",
-OR , aryl, heteroaryl, arylalkyl, heteroarylalkyl, heterocyclyl, and heterocyclylalkyl, or
R3 and R4 together with the atom to which they are attached form a 4 to 10 membered heteroaryl or heterocyclic ring, wherein any of said heteroaryl or heterocyclic rings are optionally substituted with one or more groups independently selected from halogen, cyano, nitro, trifluoromethyl, difluoromethoxy, trifluoromethoxy, azido, NR1SO2R"', -SO2NR R ", -C(O)R', -C(O)OR', -OC(O)R', -NR C(O)OR " ", -NR C(O)R", -C(O)NR R'' -SO2R " ', -NR R ", -NRC(O)NR "R ', -NR1C(NCN)NR "R ', -OR 1 aryl, heteroaryl, arylalkyl, heteroarylalkyl, heterocyclyl, and heterocyclylalkyl; R", R" and R'" independently are hydrogen, lower alkyl, lower alkenyl, aryl and arylalkyl, and
R"" is lower alkyl, lower alkenyl, aryl or arylalkyl, or any two of R', R", R'" or R"" together with the atom to which they are attached form a 4 to 10 membered carbocyclic, heteroaryl or heterocyclic ring, wherein any of said alkyl, alkenyl, aryl, arylalkyl carbocyclic rings, heteroaryl rings or heterocyclic rings are optionally substituted with one or more groups independently selected from halo, cyano, nitro, trifluoromethyl, difluoromethoxy, trifluoromethoxy, azido, aryl, heteroaryl, arylalkyl, heteroarylalkyl, heterocyclyl, and heterocyclylalkyl;
R4 and R5 independently are hydrogen or Ci-C6 alkyl, or
R4 and R5 together with the atom to which they are attached form a 4 to 10-membered carbocyclic, heteroaryl or heterocyclic ring, wherein said alkyl or any of said carbocyclic, heteroaryl and heterocyclic rings are optionally substituted with one or more groups independently selected from cyano, halogen, nitro, trifluoromethyl, difluoromethoxy, trifluoromethoxy, azido, -NR SO2R " ", -SO2NR R ", -C(O)R " ", C(O)OR", -OC(O)R", -NR C(O)OR" ", -NRC(O)R ", -C(O)NR R ", -SO2R " ", -NR R ", -NR C(O)NR11R ", -NR C(NCN)NR 1R ', -OR", aryl, heteroaryl, arylalkyl, heteroarylalkyl, heterocyclyl, and heterocyclylalkyl;
R6 is trifluoromethyl, C1-C10 alkyl, C3-Ci0 cycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocyclyl or heterocyclylalkyl, wherein any of said alkyl, cycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocyclyl and heterocyclylalkyl portions are optionally substituted with one or more groups independently selected from oxo, halogen, cyano, nitro, trifluoromethyl, difluoromethoxy, trifluoromethoxy, azido, -NR SO2R" ", -SO2NR1R ", -C(O)R', C(O)OR', -OC(O)R", -NR C(O)OR1 ", -NR C(O)R", -C(O)NR1R", -SO2R"", -NR R', -NR1C(O)NR11R 1, -NR1C(NCN)NR 1R ", -OR', aryl, heteroaryl, arylalkyl, heteroarylalkyl, heterocyclyl, and heterocyclylalkyl;
W is heteroaryl, heterocyclyl, -C(O)OR3, -C(O)NR3R4, -C(O)NR4OR3,
-C(O)(C3-C10 cycloalkyl), -C(O)(Ci-Ci0 alkyl), -C(O)(aryl), -C(O)(heteroaryl), -C(O)(heterocyclyl), -C(O)NH(SO2)CH3, wherein any of said heteroaryl, heterocyclyl, -C(O)OR3, -C(O)NR3R4, -C(O)NR4OR3, -C(O)(C3-Ci0 cycloalkyl), -C(0)(Ci-Cio alkyl), -C(O)(aryl), -C(O)(heteroaryl), -C(O)(heterocyclyl), C(O)NH(SO2)CH3 are optionally substituted with one or more groups independently selected from -NR3R4, -OR3, R2, C1-Ci0 alkyl, C2-Ci0 alkenyl, and C2-C10 alkynyl, wherein any of said Ci-Ci0 alkyl, C2-Ci0 alkenyl, and C2-Ci0 alkynyl, are optionally substituted with 1 or more groups independently selected from -NR3R4 and - OR3; m is O, 1 , 2, 3 ,4 or 5; and j is 1 or 2.
In preferred embodiments, the variants have the following meanings:
Ri is as defined above, preferably hydrogen, halo, Ci-C4 alkyl, C3-C4 cycloalkyl, C2-C4 alkenyl, C2-C4 alkynyl, cyano, nitro, OR3 or NR3R4; more preferably hydrogen, halo or C1- C4 alkyl, still more preferably hydrogen or halo, most preferably hydrogen or F. In one embodiment, Ri is hydrogen.
In a further embodiment, R1 is -S(O)jNR4C(O)R3, -C(O)NR4S(O)jR6, or S(O)jR6.
R2 is as defined above, preferably hydrogen, halo, Ci-C4 alkyl, C3-C4 cycloalkyl, C2-C4 alkenyl, C2-C4 alkynyl, cyano, nitro, OR3 or NR3R4; more preferably hydrogen, halo or C1- C2 alkyl, still more preferably halo or methyl, most preferably Cl, F or methyl. In one embodiment, R2 is methyl. In another embodiment, methyl is preferably further substituted by 1 , 2 or 3 fluorines, preferably 3 fluorines. Most preferably, R2 is F.
In still another embodiment, R2 is -S(O)jNR4C(O)R3, -C(O)NR4S(O)jR6, or S(O)jR6. R9 is as defined above, preferably hydrogen, halo, CrC4 alkyl, C3-C4 cycloalkyl, C2-C4 alkenyl, C2-C4 alkynyl, cyano, nitro, OR3 or NR3R4; more preferably hydrogen, halo or C1- C4 alkyl, still more preferably hydrogen, methyl or halo, most preferably hydrogen, methyl, Cl or F. In one embodiment, R9 is hydrogen.
In another embodiment, R9 is -S(O)jNR4C(O)R3, -C(O)NR4S(O)jR6, or S(O)jR6. R10 is as defined above, preferably hydrogen, -OR3, -C(O)R31-C(O)OR3, -NR4C(O)OR6, -OC(O)R3, -NR4S(O)2R6, -S(O)2NR3R4, S(O)2R6, -NR4C(O)R3, -C(O)NR3R4, -NR5C(O)NR3R4, -NR3R4, more preferably hydrogen, -OR3, -NR4C(O)R3, -C(O)NR3R4, -NR3R4, still more preferably hydrogen, -OR3, -NR3R4, most preferably hydrogen. In preferred embodiments R3 and R4 are independently Ci-C6 alkyl, more preferably C1-C4 alkyl, optionally substituted by 1 or 2 alkyl amino, dialkyl amino, amino, O-alkyl, hydroxy, or R3 and R4 form together a cyclic ring with 1 or 2 N atoms and optionally an O atom, said ring being optionally substituted by 1 or 2 alkyl amino, amino, hydroxy or O-alkyl. In a further embodiment, Ri0 is -S(O)jNR4C(O)R3, -C(O)NR4S(O)JR6, -S(O)jRβ, -S(O)j(Ci-C6 alkyl), -S(O)j(CR4R5)m-aryl, -O(CR4R5)m-aryl, -NR4(CR4R5)m-aryl, -O(CR4R5)m-heteroaryl, -NR4(CR4R5)m-heteroaryl, -O(CR4R5)m-heterocyclyl, or -NR4(CR4R5)m-heterocyclyl and -S(Ci-C2alkyl) substituted with 1 to 5 F, where each aryl, heteroaryl and heterocyclyl is substituted or unsubstituted.
L is as defined above, preferably a bond, CrC6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, C3-Ci0 cycloalkylalkyl, arylalkyl, heteroarylalkyl, heterocyclyl, heterocyclylalkyl, more preferably a bond, CrC5 alkyl, most preferably a bond, methylene, ethylene, n- propylene or n-butylene. In one embodiment, L is ethylene, n-propylene or n-butylene. In another embodiment, L is a bond or methylene. In the definition of L all moieties are divalent so that L serves as a linker between the nitrogen atom and R10.
R11 is as defined above, preferably hydrogen, halo, C1-C4 alkyl, C3-C4 cycloalkyl, C2-C4 alkenyl, C2-C4 alkynyl, cyano, nitro, OR3 or NR3R4; more preferably hydrogen, halo or C1-C4 alkyl or 0-Ci-C4 alkyl, still more preferably hydrogen, methyl, O-methyl or halo, most preferably hydrogen, methyl, Cl, Br or F. In one embodiment, R11 is hydrogen. In another embodiment, R11 is methyl. In yet another embodiment, methyl is preferably further substituted by 1 , 2 or 3 fluorines, preferably 3 fluorines.
In another embodiment, R11 is -S(O)jNR4C(O)R3, -C(O)NR4S(O),R6, or S(O),R6.
R12 is as defined above, preferably hydrogen, halo, C1-Ci0 alkyl, C3-C10 cycloalkyl, C2-C10 alkenyl, C2-C10 alkynyl, cyano, nitro, azido; NR4SO2R6; SO2NR3R4; SO2R6; C(O)NR3R4; C(O)OR3; OR3, NR3R4 Or -S(C1-C2 alkyl) substituted with 1 to 5 F, more preferably hydrogen, halo, nitro, Ci-C4 alkyl, 0-C1-C4 alkyl, SCF3, SCHF2, SCH2F, SO2NR3R4 or C(O)NR3R4, still more preferably hydrogen, F, Cl, Br, I, nitro, methyl, ethyl, n-propyl, i-propyl, cyclopropyl, O-methyl, SCF3, SCHF2, SCH2F, SO2NR3R4 Or C(O)NR3R4, most preferably hydrogen I, Cl, Br, SCF3, SCHF2, SCH2F, methyl or O-methyl. In one embodiment R12 is hydrogen. In another embodiment, R12 is methyl, SCF3, SCHF2, SCH2F or O-methyl, wherein methyl or O-methyl is preferably unsubstituted or further substituted by 1 , 2 or 3 fluorines, preferably 2 or 3 fluorines. In preferred embodiments of R12, R3 and R4 are independently C1-C6 alkyl, more preferably Ci-C4 alkyl, optionally substituted by 1 or 2 alkyl amino, dialkyl amino, amino, O-alkyl, hydroxy, or R3 and R4 form together a cyclic ring with 1 or 2 N atoms and optionally an O atom, said ring being optionally substituted by 1 or 2 alkyl amino, amino, hydroxy or O-alkyl. Most preferably, Ri2 is Br or I.
In another embodiment, R12 is -S(O)jNR4C(O)R3, -C(O)NR4S(O)jR6) or S(O)jR6. Ri3 is as defined above, preferably hydrogen, halo, Ci-C4 alkyl, C3-C4 cycloalkyl, C2-C4 alkenyl or C2-C4 alkynyl, more preferably hydrogen, F, Cl or methyl, most preferably hydrogen or F. in one embodiment, Ri3 is hydrogen.
In another embodiment, R13 is -S(O)jNR4C(O)R3, -C(O)NR4S(O)jR6, or S(O))R6. Ri4 is as defined above, preferably hydrogen, halo, Ci-C4 alkyl, C3-C4 cycloalkyl, C2-C4 alkenyl Or C2-C4 alkynyl, more preferably hydrogen, F, Cl or methyl, most preferably hydrogen or F. In one embodiment, Ri4 is hydrogen.
In a further embodiment, R14 is -S(O)jNR4C(O)R3, -C(O)NR4S(O)jR6, or S(O)jR6.
In a particular preferred embodiment, at least one of R1, R2, R9, Rn, R12, R13 and R14 is selected from -S(O)jNR4C(O)R3, -C(O)NR4S(O)jR6, and S(O)jR6
As set forth above, the variants of each of R1, R2, Rg to R14 and L may be substituted. In this case they can be substituted with 1 to 5, preferably 1 to 3, more preferably 1 or 2 groups independently selected from oxo, halogen, cyano, nitro, CF3, CHF2, CH2F, OCF3, OCHF2, OCH2F, SCF3, SCHF2, SCH2F, azido, NR4SO2R6, SO2NR3R4, C(O)R3, C(O)OR3, OC(O)R3, NR4C(O)OR6, NR4C(O)R3, C(O)NR3R4, NR3R4, NR5C(O)NR3R4,
NR5C(NCN)NR3R4, OR3, aryl, heteroaryl, arylalkyl, heteroarylalkyl, heterocyclyl, and heterocyclylalkyl, preferably oxo, halogen, cyano, nitro, CF3, CHF2, CH2F, OCF3, OCHF2, OCH2F, SCF3, SCHF2, SCH2F, azido, NR4SO2R6, SO2NR3R4, C(O)R3, C(O)OR3, OC(O)R3, OR3, more preferably oxo, halogen, cyano, nitro, trifluoromethyl, difluoromethoxy, trifluoromethoxy or azido, most preferably halogen, cyano, nitro, CF3, CHF2, CH2F, OCF3, OCHF2, OCH2F, SCF3, SCHF2, SCH2F, OH, O-methyl, NH2 or N(methyl)2. When it is described that alkyl, alkenyl, alkynyl, cycloalkyl, aryl, heteroaryl and heterocyclyl are substituted, this refers to any alkyl, alkenyl, alkynyl, cycloalkyl, aryl, heteroaryl and heterocyclyl as a group or sub-structure such as in cycloalkylalkyl, arylalkyl, heteroarylalkyl, heterocyclylalkyl.
R3 is as defined above, preferably hydrogen, trifluoromethyl, Ci-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, C3-C6 cycloalkyl, C3-C6 cycloalkylalkyl, more preferably hydrogen or Ci-C4 alkyl most preferably hydrogen, methyl or ethyl.
R4 is as defined above, preferably hydrogen or Ci-C4 alkyl, more preferably hydrogen, methyl or ethyl.
In one preferred embodiment, R3 and R4 can be taken together with the atom to which they are attached to form a 4 to 7, preferably 5 or 6, membered heteroaryl or heterocyclic ring. R5 is as defined above, preferably hydrogen or CrC4 alkyl, more preferably hydrogen, methyl or ethyl.
In one embodiment, R4 and R5 can be taken together with the atom to which they are attached to form a 4 to 7, preferably 5 or 6, membered carbocyclic, heteroaryl or heterocyclic ring.
R6 is as defined above, preferably trifluoromethyl, Ci-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, C3-C6 cycloalkyl, C3-C6 cycloalkylalkyl, more preferably d-C4 alkyl, most preferably methyl or ethyl.
As set forth above, the variants of each of R3, R4, R5, R6 or the rings formed by R3 and R4 and R4 and R5 may be substituted. In this case they can be substituted with 1 to 5, preferably 1 to 3, more preferably 1 or 2 groups independently selected from oxo, halogen, cyano, nitro, CF3, CHF2, CH2F, OCF3, OCHF2, OCH2F, azido, NR1SO2R"", SO2NR", C(O)R', C(O)OR', OC(O)R', NR1C(O)OR"", NR1C(O)R", C(O)NR1R", SR"", S(O)R"", SO2R', NR1R", NR1C(O)NR11R1", NR1C(NCN)NR11R"', OR', aryl, heteroaryl, arylalkyl, heteroarylalkyl, heterocyclyl, and heterocyclylalkyl, preferably oxo, halogen, cyano, nitro, CF3, CHF2, CH2F1 OCF3, OCHF2, OCH2F, azido, NR1SO2R11", SO2NR", C(O)R', C(O)OR1, OC(O)R1, NR1C(O)OR11", NR1C(O)R", C(O)NR1R", SR"", S(O)R"", SO2R1, NR1R", NR1C(O)NR11R'", NR1C(NCN)NR11R"' or OR1, more preferably oxo, halogen, cyano, nitro, CF3, CHF2, CH2F, OCF3, OCHF2, OCH2F, azido, SR"", S(O)R"", SO2R', NR1R" or OR". In one embodiment, R3 is preferably oxo, halogen, nitro, trifluoromethyl, OH, O-methyl, NH2 or N(methyl)2. When it is described that alkyl, alkenyl, alkynyl, cycloalkyl, aryl, heteroaryl and heterocyclyl are substituted, this refers to any alkyl, alkenyl, alkynyl, cycloalkyl, aryl, heteroaryl and heterocyclyl as a group or sub-structure such as in cycloalkylalkyl, arylalkyl, heteroarylalkyl, heterocyclylalkyl. R1 is selected from hydrogen, Ci-C4 alkyl, C2-C4 alkenyl, aryl and arylalkyl, preferably hydrogen or CrC4 alkyl, more preferably hydrogen or methyl.
R" is selected from hydrogen, Ci-C4 alkyl, C2-C4 alkenyl, aryl and arylalkyl, preferably hydrogen or CrC4 alkyl, more preferably hydrogen or methyl.
R'" is selected from hydrogen, Ci-C4 alkyl, C2-C4 alkenyl, aryl and arylalkyl, preferably hydrogen or Ci-C4 alkyl, more preferably hydrogen or methyl.
R11" is selected from Ci-C4 alkyl, Ci-C4 alkenyl, aryl and arylalkyl, preferably Ci-C4 alkyl, more preferably methyl. Alternatively, any two of R', R", R"' or R"" can be taken together with the atom to which they are attached to form a 4 to 10 membered carbocyclic, heteroaryl or heterocyclic ring, each of which is optionally substituted with one to three groups independently selected from halogen, cyano; nitro, CF3, CHF2, CH2F, OCF3, OCHF2, OCH2F, azido, aryl, heteroaryl, arylalkyl, heteroarylalkyl, heterocyclyl, and heterocyclylalkyl, preferably halogen, cyano; nitro, trifluoromethyl, difluoromethoxy, trifluoromethoxy and azido.
W is as defined above, preferably heteroaryl containing 1 , 2 or 3 heteroatoms, or heterocyclyl containing 1 , 2, or 3 heteroatoms, more preferably heteroaryl, each of which is unsubstituted or substituted by 1 to 5, preferably 1 to 3, more preferably 1 , substituents ZR15, or W is -C(O)OR15, -C(O)NR4R15, -C(O)NR4OR15, -C(O)(C3-C10 cycloalkyl),
-C(O)(C2-C10 alkyl), -S(O)jNR4C(O)R15, -C(O)NR4S(O)jR6, S(O)JNR4R15 Or S(O)JNR4OR15, more preferably W is heteroaryl containing 1 , 2, or 3, specifically 2 or 3 N atoms, C(O)NR4OR15 or S(O)2NR4OR15. R4 and R15 are as defined herein or may form together a 3 to 7 membered ring with 1 or 2 N atoms and optionally an O atom. When W is heteroaryl, it is preferably
Figure imgf000013_0001
where Z and R15 are as defined above, preferably Z is a bond, NR16, NRi6SO2 or O, more preferably NR16, wherein R16 is as defined above, preferably hydrogen Or C1-C4 alkyl, more preferably hydrogen. R15 is preferably selected from hydrogen, C1-C4 alkyl, C1-C4 alkenyl, C4-C8 cycloalkylalkyl, each may contain 1 N atom optionally an O atom, where alkyl, alkenyl or cycloalkylalkyl may be further substituted by 1 or 2 of OH, 0-C1-C4 alkyl or NR'R", where R' and R" are independently hydrogen or C1-C4 alkyl where R' and R" may form a 3 to 7 membered ring with 1 or 2 N atoms and optionally an O atom. Alternatively, R16 and R15 may form together a 4 to 10 membered cyclic ring with 1 or 2 N atoms and optionally an O atom, said ring being optionally substituted by 1 or 2 alkyl amino, amino, hydroxy or O-alkyl. More preferably R15 is C1-C4 alkyl or C1-C4 alkenyl optionally substituted with 1 substituent OH1 O-Me, NH2, NHmethyl, NHethyl, N(methyl)2 or N(ethyl)2.
Y is O or NR', preferably O. Alternatively, W is preferably -C(O)ORi5, -C(O)NR4Ri5, -C(O)NR4ORi5, S(O)1NR4R15 or S(O)jNR4ORi5, more preferably -C(O)NR4ORi5 or S(O)2NR4ORi5. In these cases R15 is preferably as defined below.
In yet another preferred embodiment, W is -C(O)NR4S(O)1R6, or -S(O)jNR4C(O)Ri5, whereby R4 and Ri5 are as defined herein or may form together a 3 to 7 membered ring with 1 or 2 N atoms and optionally an O atom.
Z is as defined above, preferably a bond, NRi6, NR16SO2 or O, more preferably NR16. In another embodiment, Z is S.
Ri5 is as defined above, preferably hydrogen, C1-C4 alkyl, C1-C4 alkenyl, C4-C6 cycloalkylalkyl, more preferably C1-C4 alkyl or C1-C4 alkenyl, yet more preferably C1-C4 alkyl. Alkyl, alkenyl cycloalkyl, alkynyl, aryl, heteroaryl or hetercyclyl may be further substituted with 1 to 5, preferably 1 , 2 or 3, more preferably 1 or 2, substituents selected from OR3 or NR'R" wherein R3 is selected from hydrogen, C1-C4 alkyl or C1-C4 alkenyl, C4-C6 cycloalkylalkyl, more preferably hydrogen, methyl or ethyl, and where R' and R" are independently hydrogen or C1-C4 alkyl, or R' and R" may form a 3 to 7 membered ring with 1 or 2 N atoms and optionally an O atom, more preferably R1 and R" are independently hydrogen, methyl or ethyl, still more preferably both R' and R" are methyl. Yet more preferably, R15 may be substituted by 1 or 2 of OH, 0-C1-C4 alkyl or NR'R".
Most preferably, R15 is C1-C4 alkyl or C1-C4 alkenyl optionally substituted with 1 substituent OH, 0-Me1 NH2, N(methyl)2 or N(ethyl)2
Regarding R15, when it is described that alkyl, alkenyl, alkynyl, cycloalkyl, aryl, heteroaryl and heterocyclyl are substituted, this refers to any alkyl, alkenyl, alkynyl, cycloalkyl, aryl, heteroaryl and heterocyclyl as a group or sub-structure such as in cycloalkylalkyl, arylalkyl, heteroarylalkyl, heterocyclylalkyl. R16 is as defined above, preferably hydrogen or C1-C4 alkyl, more preferably hydrogen.
Alternatively, R16 and R15 may form together a 4 to 10, preferably 5 to 6, membered cyclic ring with 1 or 2 N atoms and optionally an O atom, said ring being optionally substituted by 1 or 2 alkyl amino, amino, hydroxy or O-alkyl. m is as defined above, preferably 0,1 ,2 or 3, more preferably 0,1 or 2, most preferably 1. j is as defined above, preferably 2.
In the above, any of the preferred definitions for each variant can be combined with the preferred definition of the other variants. The combinations as set forth in the claims are particularly preferred.
In the above and the following, the employed terms have independently the meaning as described below:
Aryl is an aromatic mono- or polycyclic moiety with preferably 6 to 20 carbon atoms which is preferably selected from phenyl, biphenyl, naphthyl, tetrahydronaphthyl, fluorenyl, indenyl or phenanthrenyl, more preferably phenyl or naphthyl.
Heteroaryl is an aromatic moiety having 6 to 20 carbon atoms with at least one ring containing a heteroatom selected from O, N and/or S, or heteroaryl is an aromatic ring containing at least one heteroatom selected from O, N and/or S and 1 to 6 carbon atoms. Preferably, heteroaryl contains 1 to 4, more preferably 1 , 2 or 3 heteroatoms selected from O and/or N and is preferably selected from pyridinyl, imidazolyl, pyrimidinyl, pyrazolyl, triazolyl, pyrazinyl, tetrazolyl, furyl, thienyl, isoxazolyl, thiazolyl, oxazolyl, isothiazolyl, pyrrolyl, quinolinyl, isoquinolinyl, indolyl, benzimidazolyl, benzofuranyl, cinnolinyl, indazolyl, indolizinyl, phthalazinyl, pyridazinyl, triazinyl, isoindolyl, pteridinyl, purinyl, oxadiazolyl, triazolyl, thiadiazolyl, thiadiazolyl, furazanyl, benzofurazanyl, benzothiophenyl, benzothiazolyl, benzoxazolyl, quinazolinyl, quinoxalinyl, naphthyridinyl, and furopyridinyl. Spiro moieties are also included within the scope of this definition. Preferred heteroaryl include pyridinyl, imidazolyl, pyrimidinyl, pyrazolyl, triazolyl, pyrazinyl, tetrazolyl, isoxazolyl, oxazolyl, isothiazolyl, oxadiazolyl, triazolyl. Heteroaryl groups are optionally mono-, di-, or trisubstituted with, e.g., halogen, lower alkyl, lower alkoxy, haloalkyl, aryl, heteroaryl, and hydroxy.
Heterocyclyl is a saturated or unsaturated ring containing at least one heteroatom selected from O, N and/or S and 1 to 6 carbon atoms. Preferably, heterocyclyl contains 1 to 4, more preferably 1 , 2 or 3 heteroatoms selected from O and/or N and is preferably selected from pyrrolidinyl, tetrahydrofuranyl, dihydrofuranyl, tetrahydrothienyl, tetrahydropyranyl, dihydropyranyl, tetrahydrothiopyranyl, piperidino, morpholino, thiomorpholino, thioxanyl, piperazinyl, homopiperazinyl, azetidinyl, oxetanyl, thietanyl, homopiperidinyl, oxepanyl, thiepanyl, oxazepinyl, diazepinyl, thiazepinyl, 1 ,2,3,6- tetrahydropyridinyl, 2-pyrrolinyl, 3-pyrrolinyl, indolinyl, 2H-pyranyl, 4H-pyranyl, dioxanyl, 1 ,3-dioxolanyl, pyrazolinyl, dithianyl, dithiolanyl, dihydropyranyl, dihydrothienyl, dihydrofuranyl, pyrazolidinylimidazolinyl, imidazolidinyl, azetidin-2-one-1-yl, pyrrolidin-2- one-1-yl, piperid-2-one-1-yl, azepan-2-one-1-yl, 3-azabicyco[3.1.0]hexanyl, 3- azabicyclo[4.1.0]heptanyl, azabicyclo[2.2.2]hexanyl, 3H-indolyl and quinolizinyl. Spiro moieties are also included within the scope of this definition. Carbocyclyl is a monocyclic or polycyclic ring system of 3 to 20 carbon atoms which may be saturated, unsaturated or aromatic.
Alkyl is a saturated hydrocarbon moiety, namely straight chain or branched alkyl having 1 to 10, preferably 1 to 8 carbon atoms, more preferably 1 to 4 carbon atoms, such as methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, n-pentyl, isopentyl, neopentyl, hexyl or heptyl.
Cycloalkyl is an alkyl ring having 3 to 10, preferably 3 to 8 carbon atoms, more preferably 3 to 6 carbon atoms, such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl or cyclooctyl. Alkenyl is an unsaturated hydrocarbon moiety with one or more double bonds, preferably one double bond, namely straight chain or branched alkenyl having 1 to 10, preferably 2 to 8 carbon atoms, more preferably 2 to 4 atoms, such as vinyl, allyl, methallyl, buten-2- yl, buten-3-yl, penten-2-yl, penten-3-yl, penten-4-yl, 3-methyl-but-3-enyl, 2-methyl-but-3- enyl, 1-methyl-but-3-enyl, hexenyl or heptenyl. Alkynyl is an unsaturated hydrocarbon moiety with one or more triple bonds, preferably one triple bond, namely straight chain or branched alkynyl having 1 to 10, preferably 2 to 8 carbon atoms, more preferably 2 to 4 atoms, such as ethynyl, propynyl, butyn-2-yl, butyn-3-yl, pentyn-2-yl, pentyn-3-yl, pentyn-4-yl, 2-methyl-but-3-ynyl, 1-methyl-but-3-ynyl, hexynyl or heptynyl. Halo or halogen is a halogen atom preferably selected from F, Cl1 Br and I1 preferably F1 Cl and Br.
In the definitions cycloalkylalkyl, arylalkyl, heretoarylalkyl and heterocyclylalkyl it is contemplated that cycloalkyl, aryl, heretoaryl and heterocyclyl are bonded via an alkylene moiety. This alkylene moiety may be a straight chain or branched chain group. Said alkylene moiety preferably has 1 to 6 carbon atoms. Examples thereof include methylene, ethylene, n-propylene, n-butylene, n-pentylene, n-hexylene, iso-propylene, sec.-butylene, tert.-butylene, 1 ,1 -dimethyl propylene, 1 ,2-dimethyl propylene, 2,2- dimethyl propylene, 1 ,1 -dimethyl butylene, 1 ,2-dimethyl butylene, 1 ,3-dimethyl butylene, 2,2-dimethyl butylene, 2,3-dimethyl butylene, 3,3-dimethyl butylene, 1 -ethyl butylene, 2- ethyl butylene, 3-ethyl butylene, 1 -n-propyl propylene, 2-n-propyl propylene, 1-iso-propyl propylene, 2-iso-propyl propylene, 1 -methyl pentylene, 2-methyl pentylene, 3-methyl pentylene and 4-methyl pentylene. More preferably, said alkylene moiety has 1 to 3 carbon atoms, such as methylene, ethylene, n-propylene and iso-propylene. Most preferred is methylene. Preferred embodiments of the compounds according to present invention are shown in scheme 1.
Figure imgf000017_0001
Scheme 1
The compounds of the present invention can be in the form of a prodrug compound. "Prodrug compound" means a derivative that is converted into a compound according to the present invention by a reaction with an enzyme, gastric acid or the like under a physiological condition in the living body, e.g. by oxidation, reduction, hydrolysis or the like, each of which is carried out enzymatically. Examples of the prodrug are compounds, wherein the amino group in a compound of the present invention is acylated, alkylated or phosphorylated to form, e.g., eicosanoylamino, alanylamino, pivaloyloxymethylamino or wherein the hydroxyl group is acylated, alkylated, phosphorylated or converted into the borate, e.g. acetyloxy, palmitoyloxy, pivaloyloxy, succinyloxy, fumaryloxy, alanyloxy or wherein the carboxyl group is esterified or amidated. These compounds can be produced from compounds of the present invention according to well-known methods. Other examples of the prodrug are compounds, wherein the carboxylate in a compound of the present invention is for example converted into an alkyl-, aryl-, choline-, amino, acyloxymethylester, linolenoyl-ester. Metabolites of compounds of the present invention are also within the scope of the present invention.
Where tautomerism, like e.g. keto-enol tautomerism, of compounds of the present invention or their prodrugs may occur, the individual forms, like e.g. the keto and enol form, are claimed separately and together as mixtures in any ratio. Same applies for stereoisomers, like e.g. enantiomers, cis/trans isomers, conformers and the like.
If desired, isomers can be separated by methods well known in the art, e.g. by liquid chromatography. Same applies for enantiomers by using e.g. chiral stationary phases. Additionally, enantiomers may be isolated by converting them into diastereomers, i.e. coupling with an enantiomerically pure auxiliary compound, subsequent separation of the resulting diastereomers and cleavage of the auxiliary residue. Alternatively, any enantiomer of a compound of the present invention may be obtained from stereoselective synthesis using optically pure starting materials.
The compounds of the present invention can be in the form of a pharmaceutically acceptable salt or a solvate. The term "pharmaceutically acceptable salts" refers to salts prepared from pharmaceutically acceptable non-toxic bases or acids, including inorganic bases or acids and organic bases or acids. In case the compounds of the present invention contain one or more acidic or basic groups, the invention also comprises their corresponding pharmaceutically or toxicologically acceptable salts, in particular their pharmaceutically utilizable salts. Thus, the compounds of the of the present invention which contain acidic groups can be present on these groups and can be used according to the invention, for example, as alkali metal salts, alkaline earth metal salts or as ammonium salts. More precise examples of such salts include sodium salts, potassium salts, calcium salts, magnesium salts or salts with ammonia or organic amines such as, for example, ethylamine, ethanolamine, triethanolamine or amino acids. Compounds of the present invention which contain one or more basic groups, i.e. groups which can be protonated, can be present and can be used according to the invention in the form of their addition salts with inorganic or organic acids. Examples for suitable acids include hydrogen chloride, hydrogen bromide, phosphoric acid, sulfuric acid, nitric acid, methanesulfonic acid, p-toluenesulfonic acid, naphthalenedisulfonic acids, oxalic acid, acetic acid, tartaric acid, lactic acid, salicylic acid, benzoic acid, formic acid, propionic acid, pivalic acid, diethylacetic acid, malonic acid, succinic acid, pimelic acid, fumaric acid, maleic acid, malic acid, sulfaminic acid, phenylpropionic acid, gluconic acid, ascorbic acid, isonicotinic acid, citric acid, adipic acid, and other acids known to the person skilled in the art. If the compounds of the present invention simultaneously contain acidic and basic groups in the molecule, the invention also includes, in addition to the salt forms mentioned, inner salts or betaines (zwitterions). The respective salts can be obtained by customary methods which are known to the person skilled in the art like, for example by contacting these with an organic or inorganic acid or base in a solvent or dispersant, or by anion exchange or cation exchange with other salts. The present invention also includes all salts of the compounds of the present invention which, owing to low physiological compatibility, are not directly suitable for use in pharmaceuticals but which can be used, for example, as intermediates for chemical reactions or for the preparation of pharmaceutically acceptable salts. Furthermore, the present invention provides pharmaceutical compositions comprising a compound of the present invention, or a prodrug compound thereof, or a pharmaceutically acceptable salt or solvate thereof as active ingredient together with a pharmaceutically acceptable carrier.
"Pharmaceutical composition" means one or more active ingredients, and one or more inert ingredients that make up the carrier, as well as any product which results, directly or indirectly, from combination, complexation or aggregation of any two or more of the ingredients, or from dissociation of one or more of the ingredients, or from other types of reactions or interactions of one or more of the ingredients. Accordingly, the pharmaceutical compositions of the present invention encompass any composition made by admixing a compound of the present invention and a pharmaceutically acceptable carrier.
A pharmaceutical composition of the present invention may additionally comprise one or more other compounds as active ingredients like one or more additional compounds of the present invention, or a prodrug compound or other MEK inhibitors. The compositions include compositions suitable for oral, rectal, topical, parenteral (including subcutaneous, intramuscular, and intravenous), ocular (ophthalmic), pulmonary (nasal or buccal inhalation), or nasal administration, although the most suitable route in any given case will depend on the nature and severity of the conditions being treated and on the nature of the active ingredient. They may be conveniently presented in unit dosage form and prepared by any of the methods well-known in the art of pharmacy.
In one embodiment, said compounds and pharmaceutical composition are for the treatment of cancer such as brain, lung, squamous cell, bladder, gastic, pancreatic, breast, head, neck, renal, kidney, ovarian, prostate, colorectal, oesohageal, testicular, gynecological or thyroid cancer. In another embodiment, said pharmaceutical composition is for the treatment of a noncancerous hyperproliferative disorder such as benign hyperplasia of the skin (e.g., psoriasis), restenosis, or prostate (e.g. benign prostatic hypertrophy (BPH)). The invention also relates to a compound or pharmaceutical composition for the treatment of pancreatitis or kidney disease (including proliferative glomerulonephtitis and diabetes induced renal disease) or pain in a mammal which comprises a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt, prodrug or hydrate thereof, and a pharmaceutically acceptable carrier. The invention also relates to a compound or pharmaceutical composition for the prevention of blastocyte implantation in a mammal which comprises a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt, prodrug or hydrate thereof, and a pharmaceutically acceptable carrier. The invention also relates to a compound or pharmaceutical composition for treating a disease related to vasculogenesis or angiogenesis in a mammal which comprises a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt, prodrug or hydrate thereof, and a pharmaceutically acceptable carrier.
In one embodiment, said compound or pharmaceutical composition is for treating a disease selected from the group consisting of tumor angiogenesis, chronic inflammatory disease such as rheumatoid arthritis, inflammatory bowel disease, atherosclerosis, skin diseases such as psoriasis, eczema, and sclerodema, diabetes, diabetic retinopathy, retinopathy of prematurity, age-related macular degeneration, hemangioma, glioma, melanoma, Kaposi's sarcoma and ovarian, breast, lung, pancreatic, prostate, colon and epidermoid cancer.
The invention also relates to of the use for treating a hyperproliferative disorder in a mammal that comprises administering to said mammal a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt, prodrug or hydrate thereof. In one embodiment, said use relates to the treatment of cancer such as brain, lung, squamous cell, bladder, gastic, pancreatic, breast, head, neck, renal, kidney, ovarian, prostate, colorectal, oesohageal, testicular, gynecological or thyroid cancer. In another embodiment, said use relates to the treatment of a non-cancerous hyperproliferative disorder such as benign hyperplasia of the skin (e.g., psoriasis), restenosis, or prostate (e.g., benign prostatic hypertrophy (BPH)).
The invention also relates to a use for the treatment of a hyperproliferative disorder in a mammal that comprises administering to said mammal a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt, prodrug or hydrate thereof, in combination with an anti-tumor agent selected from the group consisting of mitotic inhibitors, alkylating agents, antimetabolites, intercalating antibiotics, growth factor inhibitors, cell cycle inhibitors, enzyme inhibitors, topoisomerase inhibitors, biological response modifiers, antihormones, angiogenesis inhibitors, and anti- androgens.
The invention also relates to a use of treating pancreatitis or kidney disease or pain in a mammal that comprises administering to said mammal a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt, prodrug or hydrate thereof. The invention also relates to a use of preventing blastocyte implantation in a mammal that comprises administering to said mammal a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt, prodrug or hydrate thereof.
The invention also relates to a use of treating diseases related to vasculogenesis or angiogenesis in a mammal that comprises administering to said mammal a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt, prodrug or hydrate thereof. In one embodiment, said method is for treating a disease selected from the group consisting of tumor angiogenesis, chronic inflammatory disease such as rheumatoid arthritis, atherosclerosis, inflammatory bowel disease, skin diseases such as psoriasis, eczema, and scleroderma, diabetes, diabetic retinopathy, retinopathy of prematurity, age-related macular degeneration, hemangioma, glioma, melanoma, Kaposi's sarcoma and ovarian, breast, lung, pancreatic, prostate, colon and epidermoid cancer. Patients that can be treated with compounds of the present invention, or pharmaceutically acceptable salts, prodrugs and hydrates of said compounds, according to the methods of this invention include, for example, patients that have been diagnosed as having psoriasis, restenosis, atherosclerosis, BPH, lung cancer, bone cancer, CMML, pancreatic cancer, skin cancer, cancer of the head and neck, cutaneous or intraocular melanoma, uterine cancer, ovarian cancer, rectal cancer, cancer of the anal region, stomach cancer, colon cancer, breast cancer, testicular, gynecologic tumors (e.g., uterine sarcomas, carcinoma of the fallopian tubes, carcinoma of the endometrium, carcinoma of the cervix, carcinoma of the vagina or carcinoma of the vulva), Hodgkin's disease, cancer of the esophagus, cancer of the small intestine, cancer of the endocrine system (e.g., cancer of the thyroid, parathyroid or adrenal glands), sarcomas of soft tissues, cancer of the urethra, cancer of the penis, prostate cancer, chronic or acute leukemia, solid tumors of childhood, lymphocytic lymphomas, cancer of the bladder, cancer of the kidney or ureter (e.g., renal cell carcinoma, carcinoma of the renal pelvis), or neoplasms of the central nervous system (e.g., primary CNS lymphona, spinal axis tumors, brain stem gliomas or pituitary adenomas).
This invention also relates to a compound or pharmaceutical composition for inhibiting abnormal cell growth in a mammal which comprises an amount of a compound of the present invention, or a pharmaceutically acceptable salt or solvate or prodrug thereof, in combination with an amount of a chemotherapeutic, wherein the amounts of the compound, salt, solvate, or prodrug, and of the chemotherapeutic are together effective in inhibiting abnormal cell growth. Many chemotherapeutics are presently known in the art. In one embodiment, the chemotherapeutic is selected from the group consisting of mitotic inhibitors, alkylating agents, anti-metabolites, intercalating antibiotics, growth factor inhibitors, cell cycle inhibitors, enzymes, topoisomerase inhibitors, biological response modifiers, anti-hormones, angiogenesis inhibitors, and anti-androgens. This invention further relates to a method for inhibiting abnormal cell growth in a mammal or treating a hyperproliferative disorder which method comprises administering to the mammal an amount of a compound of the present invention, or a pharmaceutically acceptable salt or solvate or prodrug thereof, in combination with radiation therapy, wherein the amounts of the compound, salt, solvate, or prodrug, is in combination with the radiation therapy effective in inhibiting abnormal cell growth or treating the hyperproliferative disorder in the mammal. Techniques for administering radiation therapy are known in the art, and these techniques can be used in the combination therapy described herein. The administration of the compound of the invention in this combination therapy can be determined as described herein. It is believed that the compounds of the present invention can render abnormal cells more sensitive to treatment with radiation for purposes of killing and/or inhibiting the growth of such cells. Accordingly, this invention further relates to a method for sensitizing abnormal cells in a mammal to treatment with radiation which comprises administering to the mammal an amount of a compound of the present invention or pharmaceutically acceptable salt or solvate or prodrug thereof, which amount is effective is sensitizing abnormal cells to treatment with radiation. The amount of the compound, salt, or solvate in this method can be determined according to the means for ascertaining effective amounts of such compounds described herein. The invention also relates to a method of and to a pharmaceutical composition of inhibiting abnormal cell growth in a mammal which comprises an amount of a compound of the present invention, or a pharmaceutically acceptable salt or solvate thereof, a prodrug thereof, or an isotopically-labeled derivative thereof, and an amount of one or more substances selected from anti-angiogenesis agents, signal transduction inhibitors, and antiproliferative agents.
In practical use, the compounds of the present invention can be combined as the active ingredient in intimate admixture with a pharmaceutical carrier according to conventional pharmaceutical compounding techniques. The carrier may take a wide variety of forms depending on the form of preparation desired for administration, e.g., oral or parenteral (including intravenous). In preparing the compositions for oral dosage form, any of the usual pharmaceutical media may be employed, such as, for example, water, glycols, oils, alcohols, flavoring agents, preservatives, coloring agents and the like in the case of oral liquid preparations, such as, for example, suspensions, elixirs and solutions; or carriers such as starches, sugars, microcrystalline cellulose, diluents, granulating agents, lubricants, binders, disintegrating agents and the like in the case of oral solid preparations such as, for example, powders, hard and soft capsules and tablets, with the solid oral preparations being preferred over the liquid preparations.
Because of their ease of administration, tablets and capsules represent the most advantageous oral dosage unit form in which case solid pharmaceutical carriers are obviously employed. If desired, tablets may be coated by standard aqueous or nonaqueous techniques. Such compositions and preparations should contain at least 0.1 percent of active compound. The percentage of active compound in these compositions may, of course, be varied and may conveniently be between about 2 percent to about 60 percent of the weight of the unit. The amount of active compound in such therapeutically useful compositions is such that an effective dosage will be obtained. The active compounds can also be administered intranasally as, for example, liquid drops or spray.
The tablets, pills, capsules, and the like may also contain a binder such as gum tragacanth, acacia, corn starch or gelatin; excipients such as dicalcium phosphate; a disintegrating agent such as corn starch, potato starch, alginic acid; a lubricant such as magnesium stearate; and a sweetening agent such as sucrose, lactose or saccharin. When a dosage unit form is a capsule, it may contain, in addition to materials of the above type, a liquid carrier such as a fatty oil.
Various other materials may be present as coatings or to modify the physical form of the dosage unit. For instance, tablets may be coated with shellac, sugar or both. A syrup or elixir may contain, in addition to the active ingredient, sucrose as a sweetening agent, methyl and propylparabens as preservatives, a dye and a flavoring such as cherry or orange flavor.
Compounds of the present invention may also be administered parenterally. Solutions or suspensions of these active compounds can be prepared in water suitably mixed with a surfactant such as hydroxy-propylcellulose. Dispersions can also be prepared in glycerol, liquid polyethylene glycols and mixtures thereof in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
The pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. In all cases, the form must be sterile and must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (e.g., glycerol, propylene glycol and liquid polyethylene glycol), suitable mixtures thereof, and vegetable oils.
Any suitable route of administration may be employed for providing a mammal, especially a human, with an effective dose of a compound of the present invention. For example, oral, rectal, topical, parenteral, ocular, pulmonary, nasal, and the like may be employed. Dosage forms include tablets, troches, dispersions, suspensions, solutions, capsules, creams, ointments, aerosols, and the like. Preferably compounds of the present invention are administered orally.
The effective dosage of active ingredient employed may vary depending on the particular compound employed, the mode of administration, the condition being treated and the severity of the condition being treated. Such dosage may be ascertained readily by a person skilled in the art.
When treating or preventing cancer, inflammation or other proliferative diseases for which compounds of the present invention are indicated, generally satisfactory results are obtained when the compounds of the present invention are administered at a daily dosage of from about 0.1 milligram to about 100 milligram per kilogram of animal body weight, preferably given as a single daily dose or in divided doses two to six times a day, or in sustained release form. For most large mammals, the total daily dosage is from about 1.0 milligrams to about 1000 milligrams, preferably from about 1 milligram to about 50 milligrams. In the case of a 70 kg adult human, the total daily dose will generally be from about 7 milligrams to about 350 milligrams. This dosage regimen may be adjusted to provide the optimal therapeutic response.
Some abbreviations that may appear in this application are as follows.
Abbreviations
Designation b Broad peak
Boc terf.-Butyloxycarbonyl
CDI N,N-Carbonyldiimidazole d Doublet
DCM Dichloromethane dd double doublet
DIPEA N-Ethyldiisopropylamine
DMF N,N-Dimethylformamide
DMSO Dimethylsulfoxide
1-(3-Dimethylaminopropyl)-3-ethylcarbodiimide
EDC hydrochloride
HPLC High pressure liquid chromatography
LiHMDS. Lithium hexamethyldisilazide
NMR Nuclear Magnetic Resonance
PG Protecting group
PyBroP Bromo-tris-pyrrolidino-phosphonium hexafluorophosphate
Benzotriazole-1-yl-oxy-trispyrrolidinophosphonium
PyBOP hexafluorophosphate PPA Polyphosphoric acid q Quartett rt Retention time s Singlet tert Tertiary-butyl
TFA Trifluoroacetic acid
THF Tetrahydrofurane
TLC Thin Layer Chromatography
The compounds of the present invention can be prepared according to the procedures of the following Schemes and Examples, using appropriate materials and are further exemplified by the following specific examples. Moreover, by utilizing the procedures described herein, in conjunction with ordinary skills in the art, additional compounds of the present invention claimed herein can be readily prepared. The compounds illustrated in the examples are not, however, to be construed as forming the only genus that is considered as the invention. The examples further illustrate details for the preparation of the compounds of the present invention. Those skilled in the art will readily understand that known variations of the conditions and processes of the following preparative procedures can be used to prepare these compounds. The instant compounds are generally isolated in the form of their pharmaceutically acceptable salts, such as those described above. The amine-free bases corresponding to the isolated salts can be generated by neutralization with a suitable base, such as aqueous sodium hydrogencarbonate, sodium carbonate, sodium hydroxide and potassium hydroxide, and extraction of the liberated amine-free base into an organic solvent, followed by evaporation. The amine-free base, isolated in this manner, can be further converted into another pharmaceutically acceptable salt by dissolution in an organic solvent, followed by addition of the appropriate acid and subsequent evaporation, precipitation or crystallisation.
An illustration of the preparation of compounds of the present invention is shown in schemes 2 and 3. Unless otherwise indicated in the schemes, the variables have the same meaning as described above.
The examples presented below are intended to illustrate particular embodiments of the invention.
Figure imgf000027_0001
Scheme 2
Figure imgf000028_0001
Scheme 3
As outlined in scheme2, ethyl 4,6-dichloronicotinate can be reacted with an appropriately substituted aniline in an inert solvent, preferable THF, by addition of a base, preferably but not limited to LiHMDS to yield substituted ethyl 6-chloro-4arylaminonicotinate 1. Heating with hydrazine hydrate leads to an intermediate hydrazino compound which is acylated by coupling with an activated carboxylic acid building block. Cyclisation is achieved by heating in acidic solvents such as AcOH or PPA. The resulting triazolopyridine is saponified in the next step by heating with lithium hydroxide in a water- alcohol mixture to give triazolopyridine carboxylic acid 2. The latter can be further derivatised by coupling with a substituted O-alkylhydroxylamine using a coupling reagent such as PyBro, PyBOP or DCC for example to give substituted hydroxamate 3.
Scheme 3 illustrates the preparation of compounds of the present invention where W is heterocyclic. Triazolopyridine carboxylic acid 2 can be coupled with Boc-hydrazine by using a coupling reagent such as PyBOP or DCC for example and the Boc protecting group is then removed by treatment with an acid such as HCI. The resulting hydrazide 4 is further derivatised by reacting with an appropriately substituted isocyanate. Cyclisation can be achieved by heating with triphenyl phosphine and CCI4 in a suitable solvent to give oxadiazole compound 6. Suitable anilines, carboxylic acids, O-alkyl hydroxylamines, and isocyanates are commercially available from Sigma-Aldrich Chemie GmbH, Munich, Germany or from Acros Organics, Belgium or from Fisher Scientific GmbH, 58239 Schwerte, Germany or can be routinely prepared by procedures described in "March's Advanced Organic Chemistry: Reactions, Mechanisms, and Structure", 5th Edition; John Wiley & Sons. O- [(4S)2,2-Dimethyl-[1,3]dioxolan-4-ylmethyl]-hydroxylamine and O-[(4ft)2,2-dimethyl-
[1,3]dioxolan-4-ylmethyl]-hydroxylamine are prepared according to a procedure described in WO02/06213 A2. Ethyl 4,6-dichloronicotinate is synthesised in three steps from diethyl acetone dicarboxylate according to a literature procedure (DenHertog, Reel Trav Chim Pays- Bas 1946, 65, 129-140).
Compounds with other variants in the position of W can be prepared by derivatizing the COOH group appropriately as known to the person skilled in the art as described in Theophil Eicher, Siegfried Hauptmann "The Chemistry of Heterocycles; Structures, Reactions, Synthesis and Application", 2nd edition, Wiley-VCH 2003. The introduction of alternative heterocyclic or heteroaryl groups is exemplified e.g. in WO 03/077855 and WO 01/05391. Unless otherwise noted, all non-aqueous reactions were carried out either under an argon or nitrogen atmosphere with commercial dry solvents. Compounds were purified using flash column chromatography using Merck silica gel 60 (230-400 mesh), or by reverse phase preparative HPLC using a Reprosil-Pur ODS3, 5 μm, 20 x 125 mm column with Shimadzu LC8A-Pump and SPD-10Avp UV/Vis diode array detector. The 1H-NMR spectra were recorded on a Varian VXR-S (300 MHz for 1H-NMR) using d6- dimethylsulfoxide or d4-methanol as solvent; chemical shifts are reported in ppm relative to tetramethylsilane. Analytical LC/MS was performed using Reprosil-Pur ODS3, 5 μM, 1 x 60 mm columns at a flow rate of 250 μl/min, sample loop 2.5μl; retention times are given in minutes. Methods are: (I) runs on a LC10Advp-Pump (Shimadzu) with SPD- MIOAvp UV/Vis diode array detector and QP2010 MS-detector in ESI+ modus with UV- detection at 214, 254 and 275 nm with a gradient of 15-95% acetonitrile (B) in water (A) (0.1 % formic acid), 5 min. linear gradient; (II) idem but linear gradient 8min 1-30% B; (III) idem but linear gradient 8min 10-60% B; (IV) idem but linear gradient 8min 15-99% B; (V) idem but linear gradient 10min 5-95% B; (Vl) idem but linear gradient 10min 10-95% B; (VII) idem but linear gradient 5min 10-90% B; (VIII) idem but linear gradient 5min 5- 95% B.
Example 1
J-^-Bromo-Σ-methylphenylJaminoJS-methylli^^Jtriazolo^S-aJpyridine-β-carboxylic acid (2a)
Figure imgf000029_0001
Step A
Ethyl 4,6-dichloronicotinate (2.Og, 9.1mmol) and 4-bromo-2-methylaniline (1.7g, 9.1mmol) are dissolved in dry THF (20ml) under argon and the mixture is cooled to -78°C. A solution of LiHMDS (1.0M in THF, 32ml) is slowly added and the reaction mixture is allowed to warm to ambient temperature. After 18h the reaction is quenched by adding dilute hydrochloric acid (1.0M, 20.0ml) and the mixture is extracted with DCM (3x 60ml). The combined organic extracts are concentrated in vacuo and the crude material is purified by flash chromatography using silica gel and a gradient of 0-10% ethylacetate in cyclohexane as eluent to give pure ethyl 4-[(4-bromo-2-methylphenyl)amino]-6-chloronicotinate (1a) (900mg, 27% yield).
LC-MS method (I) rt 5.34 min; m/z 369, 371 [M+H]+, Br1CI pattern.
1H-NMR (300MHz, CDCI3): δ = 1.42 (t, J = 7.3Hz, 3H), 2.21 (s, 3H), 4.39 (q, J = 7.3Hz, 2H), 6.50 (S, 1 H)1 7.11 (d, J = 8.1 Hz, 1 H)1 7.35-7.41 (m, 1 H), 7.44-7.47 (m, 1 H), 8.76 (s, 1 H), 9.59 (b, 1 H) Step B
Compound 1a (500mg, 1.35mmol) is dissolved in dry dioxane (10ml), hydrazine hydrate (985μl, 20.3mmol) is added and the mixture is heated at 1500C with microwave irradiation for 30min. The volatiles are removed in vacuo and the crude ethyl 4-[(4-bromo-2- methylphenyl)amino]-6-hydrazinonicotinate is used in the next step. LC-MS method (I) rt 2.26 min; m/z 365, 367 [M+H]+, Br pattern. Step C
4-[(4-Bromo-2-methylphenyl)amino]-6-hydrazinonicotinate (crude from step B1 ~1.35mmol) is dissolved in dry pyridine (8ml) and acetic anhydride (4ml) and the mixture is heated at 6O0C for 20 min. The volatiles are removed in vacuo, acetic acid (5ml) and acetic anhydride (2.5ml) are added and the mixture is heated for 90min at 17O0C with microwave irradiation. The volatiles are removed in vacuo, the residue is redissolved in THF (8ml), water (0.8ml) and LiOH (2.1mmol, 71 mg) is added and the mixture is heated to 12O0C with microwave irradiation for 20minutes. The volatiles are removed in vacuo, the residual material is adsorbed on flash silica and purified by flash chromatography using silica gel and a gradient of 0-10% methanol in DCM as eluent to give 361 mg (1.0 mmol, 74%yield for steps B and C) of pure 7-[(4-Bromo-2-methylphenyl)amino]-3-rnethyl[1 ,2,4]triazolo[4,3-a]pyridine-6- carboxylic acid 2a.
LC-MS method (V) rt 1.86 min; m/z 361, 363 [M+H]+, Br pattern. 1H-NMR (300MHz, DMSO-d6): δ = 2.22 (s, 3H), 2.60 (s, 3H), 6.59 (s, 1H), 7.40-7.50 (m, 3H), 8.60 (s, 1 H).
Example 2
7'[(4-Bromo-2-methylphenyl)amino]-N-hydroxy-3-methyl[1,2,4]triazolo[4,3-a]pyridine-6- carboxamide (3a)
Figure imgf000031_0001
7-[(4-Bromo-2-methylphenyl)amino]-3-methyl[1 ,2,4]triazolo[4,3-a]pyridine-6-carboxylic acid (2a) (50mg, 0.138mmol) is dissolved in dry DMF and hydroxylamine hydrochloride (19.2mg, 0.277mmol), PyBrOP (83.9mg, 0.180mmol) and DIPEA (96μl, 0.554mmol) is added. The mixture is stirred for 6h, the volatiles are evaporated and the crude material is purified by preparative HPLC to give 30 mg (O.OδOmmol, 58%yield) of pure 3a.
LC-MS method (VII) rt 1.92 min; m/z 376, 378 [M+H]+, Br pattern.
1H-NMR (400MHz, MeOH-d4): δ = 2.28 (s, 3H), 2.75 (s, 3H), .6.42 (s, 1 H), 7.30 (d, J = 8.6Hz, 1 H), 7.50-7.55 (m, 1 H), 7.63 (s, 1 H), 8.79 (s, 1 H).
Example 3
7-[(4-Bromo-2-methylphenyl)amino]-N-{[(2S)-2,3-dihydroxypropyl]oxy}-3- methyl[1 ,2,4]triazolo[4,3-a]pyridine-6-carboxamide (3b)
Figure imgf000031_0002
Step A
7-[(4-Bromo-2-methylphenyl)amino]-3-methyl[1 ,2,4]triazolo[4,3-a]pyridine-6-carboxylic acid (2a) (50mg, 0.138mmol) is dissolved in dry DMF (2.0ml) and DIPEA (96μl, 0.554mmol), PyBroP (84mg, 0.180mmol) and O-{[(4S)-2,2-dimethyl-1,3-dioxolan-4- yl]methyl}hydroxylamine (27mg, 0.18mmol) are added. The mixture is stirred for θh at ambient temperature and the volatiles are evaporated. DCM is added and the mixture is washed with saturated NaHCO3 solution, dried (Na2SO4) and evaporated to give the crude hydroxamate which is used without purification in the next step.
LC-MS method (VII) rt 2.64 min; m/z 490, 492 [M+H]+, Br pattern. Step B
The crude material from step A is dissolved in MeOH (1.8ml) and water (0.2ml). Dowex50X8 resin (20mg) is added and the mixture is heated to 1200C with microwave irradiation for 20min, the solution is filtered and evaporated. The crude material is purified by preparative HPLC to give 18.6mg (41.4μmol, 30%yield, two steps) of pure hydroxamate 3b.
LC-MS method (II) rt 6.01 min; m/z 450, 452 [M+H]+, Br pattern.
1H-NMR (400MHz1 MeOH-d4): δ = 2.27 (s, 3H), 2.70 (s, 3H), 3.62-3.66 (m, 2H), 3.92-3.99 (m, 1 H), 4.01-4.07 (m, 1 H), 4.14-4.19 (m, 1 H), 6.43 (s, 1 H), 7.27 (d, J = 8.6Hz, 1 H), 7.40-7.46 (m, 1 H), 7.52 (s, 1 H), 8.56 (s, 1 H).
Example 4
7-[(4-lodo-2-methylphenyl)amino]-N-{[(2R)-2,3-dihydroxypropyl]oxy}-3- methyl[1 ,2,4]triazolo[4,3-a]pyridine-6-carboxamide (3c)
Figure imgf000032_0001
Step A
Ethyl 4,6-dichloronicotinate (1) (3.Og, 14mmol) and 4-iodo-2-methylaniline (3.2g, 14mmol) are dissolved in dry THF (20ml) under argon and the mixture is cooled to -78°C. A solution of LiHMDS (1.0M in THF, 48ml) is slowly added and the reaction mixture is allowed to warm to ambient temperature. The reaction is quenched after 2Oh by adding dilute hydrochloric acid (1.0M, 20.0ml), the volatiles are evaporated and the residue is extracted with DCM (3x 60ml). The combined organic extracts are concentrated in vacuo and the crude material is purified by flash chromatography using silica gel and a gradient of 0-10% ethylacetate in cyclohexane as eluent to give pure ethyl 4-[(4-iodo-2-methylphenyl)amino]-6-chloronicotinate (700mg, 12% yield).
LC-MS method (VIII) rt 5.64 min; m/z 417 [M+H]+, Cl pattern. 1H-NMR (400MHz, DMSO-d6): δ = 1.33 (t, J = 7.1Hz, 3H), 2.13 (s, 3H), 4.35 (q, J = 7.1 Hz, 2H), 6.41 (s, 1 H), 7.12 (d, J = 8.6Hz, 1 H), 7.63 (d, J = 8.1 Hz, 1 H), 7.76 (s, 1 H), 8.65 (s, 1 H), 9.52 (b, 1 H).
Step B
4-[(4-lodo-2-methylphenyl)amino]-6-chloronicotinate (350mg, 0.84mmol) is dissolved in dry dioxane (7ml), hydrazine hydrate (611 μl, 12.6mmol) is added and the mixture is heated at 15O0C with microwave irradiation for 30min. The volatiles are removed in vacuo and the crude ethyl 4-[(4-lodo-2-methylphenyl)amino]-6-hydrazinonicotinate is directly used in the next step.
LC-MS method (VII) rt 2.68 min; m/z 413 [M+H]+. Step C
4-[(4-lodo-2-methylphenyl)amino]-6-hydrazinonicotinate (crude from step B, ~0.84mmol) is dissolved in dry pyridine (8ml) and acetic anhydride (4ml) and the mixture is heated at 70°C for 20 min with microwave irradiation. The volatiles are removed in vacuo, acetic acid (3ml) and acetic anhydride (1.5ml) are added and the mixture is heated for 90min at 1700C with microwave irradiation. The volatiles are removed in vacuo and the crude material is purified by flash-chomatography using silica gel and 5% methanol in DCM as eluent to give 130mg (0.30mmol, 35%yield for steps B and C) ethyl 7-[(4-iodo-2-methylphenyl)amino]-3- methyl[1 ,2,4]triazolo[4,3-a]pyridine-6-carboxylate.
LC-MS method (VII) rt 2.96 min; m/z 437 [M+H]+. Step D
Ethyl 7-[(4-iodo-2-methylphenyl)amino]-3-methyl[1,2,4]triazolo[4,3-a]pyridine-6-carboxylate (130mg, 0.298mmol) is dissolved in THF (6ml), water (1ml) and LiOH hydrate (68mg, 1.62mmol) are added and the mixture is heated for 30 min at 1200C with microwave irradiation. Hydrochloric acid (1 N in dioxane, 4ml) is added and the volatiles are removed under reduced pressure. The residue is partitioned between DCM and water, the organic phase is dried (Na2SO4) and evaporated to give crude 7-[(4-iodo-2-methylphenyl)amino]-3- methyl[1 ,2,4]triazolo[4,3-a]pyridine-6-carboxylic acid.
LC-MS method (VII) rt 2.60 min; m/z 409 [M+H]+. Step E
The crude carboxylic acid from step D (~0.27mmol) is dissolved in dry DMF, ByBrOP (162mg, 0.348mmol), DIPEA (138mg, 1.07mmol) and O-{[(4R)-2,2-dimethyl-1 ,3-dioxolan-4- yl]methyl}hydroxylamine (51 mg, 0.35mmol) are added and the mixture is stirred at 500C over night. The volatiles are evaporated, DCM is added and washed with pH7-phosphate buffer and brine, dried (Na2SO4) and evaporated. The residue is dissolved in methanol (2.7ml) and water (0.3ml) and Dowex50X8 resin (18mg) is added. The mixture is heated at 12O0C for 40min with microwave irradiation, filtered and evaporated. The crude product is purified by preparative HPLC to yield 30mg of hydroxamate 3c.
Assay
The activity of the compounds of the present invitation may be determined by the following procedure: Inhibition of human MEK1 kinase activity was monitored with a homogenous, fluorescence based assay. The assay uses time resolved fluorescence resonance energy transfer to probe for phosphorylation of ERK1 by MEK1. The assay is carried out in low volume 96 well microtiterplates. In a total volume of 15 μl, compounds are incubated with 10OnM MEK1 , 15 μM ATP, 30OnM ERK2 employing a buffer containing 2OmM TRIS/HCI, 10 mM MgCI2, 100 μM NaVO4, 1 mM DTT, and 0.005% Tween 20 (pH 7.4). After two hours, 5 nM Europium-anti-PY20 (Perkin Elmer) and 5OnM Anti-GST-Allophycocyanin (CisBio) in buffer containing 5OmM EDTA and 0,05% BSA are added and the reaction incubated for one hour in the dark. Time-resolved fluorescence is measured using a LJL-Analyst (Molecular Devices) with an excitation wavelength of 340 nm and an emission wavelength of 665 nm. The final concentration of DMSO is 2 %. To assess the inhibitory potential of the compounds, IC50-values were determined.
In this assay compounds of the invention exhibited IC50s within certain ranges. The following compounds exemplify such activity with "+" meaning 1μM < IC50 < 10μM and "++" IC50 < 1 μM
Table 1 : Inhibition of MEK
Figure imgf000034_0001
Assay 2: Tumor cell proliferation assays (ATP Lite)
Murine colon C26, human melanoma A375 and human melanoma Mel5 cells were plated in 96 well Corning white plates (1500 cells/well for C26, and 2000 cells/well for A375, and MiaPaCa-2) and cultured overnight at 370C in 5% CO2. Inhibitors were serially diluted in 100 % DMSO and subsequently added to cells to reach a final concentration of 0.25% DMSO. The cells were incubated for 4 days in the presence of test compounds in cell growth media (DMEM with 10% fetal bovine serum, 2mM glutamine for C26, and MiaPaCa-2, and RPMI with 10% fetal bovine serum, 2mM glutamine for A375). Cell proliferation was quantitated using the ATP lite cell proliferation kit (Packard). Inhibition of cell proliferation is shown in Table 2. Columns 2-4 show the concentration of compounds required to induce 50% cell death (IC50 in μM) of human endometriotic cells. With "+" meaning 100μM < IC50 < 10μM and "++" IC50 < 1 μM and "n.d." means not determined.
Assay 3: Microsomal stability assay Compounds were tested on their stability in human, rat and mouse liver microsomal preparations (HLM, RLM and MLM respectively). At a final concentration of 3 μM, compounds were incubated at 37°C with 0.5 mg/ml human, rat or mouse liver microsomes in a buffer containing 50 mM phosphate, pH 7.4 and 2 mM NADPH. Pooled human liver microsomes or pooled male rat liver microsomes (Sprague Dawley) were obtained from NatuTec (Frankfurt, Germany). Incubations without NADPH served as negative controls. Reactions were stopped after 0, 15, 30, 45 or 60 min by the addition of acetonitrile and microsomes were pelleted by centrifugation (10 min at 6200 x g). Supernatants were analyzed by HPLC regarding the concentration of mother compound. Finally, the half-life of compounds in the regarding microsomal preparation was calculated. Results are shown in Table 2. Wherein "+" means t1/2 of 1-30 min, "++" means ti/2 of 31 -120 min and "+++" means t1/2 of >120 min and "n.d." means not determined.
Assay 4: Caco-2 permeability assay
Caco-2 cells obtained from the ATCC at passage number 27 are used. Cells (passage number 40-60) were seeded on to Millipore Multiscreen Caco-2 plates or Falcon HTS inserts at 1 x 105 cells/cm2. Cells were cultured for 20 days in DMEM and media was changed every two or three days. On day 20 the permeability study was performed.
Permeability was studied by applying compound to the apical surface of cell monolayers and measuring compound permeation into the basolateral compartment. The experiment was also performed in the reverse direction (B-A) to investigate active transport. Hanks Balanced Salt Solution (HBSS) pH 7.4 buffer with 25mM HEPES and 1OmM glucose at 37°C was used as the medium in permeability studies. Incubations were carried out in an atmosphere of 5% CO2 with a relative humidity of 95%. The monolayers were prepared by rinsing both basolateral and apical surfaces twice with HBSS at 37°C. Cells were then incubated with HBSS in both apical and basolateral compartments for 40 minutes to stabilize physiological parameters.
HBSS was then removed from the apical compartment and replaced with test compound dosing solutions. The solutions were made by diluting 1OmM DMSO concentrates with HBSS to give a final test compound concentration of 10μM (final DMSO concentration adjusted to 1%). The fluorescent integrity marker lucifer yellow was also included in the dosing solution. Analytical standards were made from dosing solutions. Test compound permeability was assessed in duplicate. On each plate compounds of known permeability characteristics were run as controls. The apical compartment inserts were then placed into 'companion' plates containing fresh HBSS. For basolateral to apical (B-A) experiments the experiment was initiated by replacing buffer in the inserts then placing them in companion plates containing dosing solutions. At 120 minutes the companion plate was removed and apical and basolateral samples diluted for analysis by LC-MS/MS (the donor compartment was also sampled to permit determination of starting concentration after non-specific binding has occurred).
Analysis
The integrity of the monolayers throughout the experiment is checked by monitoring lucifer yellow permeation using fluorimethc analysis. Lucifer yellow permeation was low if monolayers have not been damaged. Test and control compounds were quantified by LC-MS/MS cassette analysis using a 5-point calibration with appropriate dilution of the samples. Should lucifer yellow Papps were above QC limits in more than one well per test compound, the compound was re-tested.
The permeability coefficient for each compound (Papp) was calculated from the following equation:
Figure imgf000036_0001
Whereby dQ/dt is the rate of permeation of the drug across the cells, C0 is the donor compartment concentration at time zero and A is the area of the cell monolayer. C0 is obtained from analysis of the donor compartment at the end of the incubation period. Test compounds were grouped into low, medium or high absorption potential based on comparison with control compounds, which have known human absorption.
In addition, permeation was studied in both directions across the cells, and an asymmetry index was reported from mean A-B and B-A data. This was derived from:
Figure imgf000037_0001
(A-B)
Results are shown in Table 2. Wherein "+" means a caco A-B and caco B-A value of 1-10 and "++"means a caco A-B and caco B-A value of 11 -100 and "n.d." means not determined.
Table 2: Results of inhibition of tumor cell proliferation and microsomal stability
Figure imgf000037_0002

Claims

Claims
1. A compound of formula (I),
Formula (I)
Figure imgf000038_0001
a pharmaceutically acceptable salt, solvate or prodrug thereof, wherein:
R1, R2, Rg, Rn Ri2, Ri3 and R14 are independently selected from hydrogen, halogen, cyano, nitro, azido, -OR3, -C(O)R31-C(O)OR3, -NR4C(O)OR6, -OC(O)R3, -NR4S(O)1R6 , -S(O)jNR3R4, -S(O)1NR4C(O)R3, -C(O)NR4S(O)1R6, S(O)1R6, -NR4C(O)R3, -C(O)NR3R4, -NR5C(O)NR3R4, -NR5C(NCN)NR3R4, -NR3R4 and C1-
Cio alkyl, C2-Ci0 alkenyl, C2-Ci0 alkynyl, C3-Ci0 cycloalkyl, C3-C10 cycloalkylalkyl, -S(O)1(CrC6 alkyl), -S(O)j(CR4R5)m-aryl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocyclyl, heterocyclylalkyl, -O(CR4R5)m-aryl, -NR4(CR4R5)m-aryl, -O(CR4R5)m- heteroaryl, -NR4(CR4R5)m-heteroaryl, -O(CR4R5)m-heterocyclyl, -NR4(CR4R5W heterocyclyl, and -S(CrC2 alkyl) substituted with 1 to 5 F, where each alkyl, alkenyl, alkynyl, cycloalkyl, aryl, heteroaryl and heterocyclyl is substituted or unsubstituted;
Rio is selected from hydrogen, -OR3, -C(O)R31-C(O)OR3, -NR4C(O)OR6, -OC(O)R3,
-NR4S(O)1R6 , -S(O)1NR3R4, -S(O)1NR4C(O)R3, -C(O)NR4S(O)1R6, S(O)1R6, -NR4C(O)R3, -C(O)NR3R4, -NR5C(O)NR3R4, -NR5C(NCN)NR3R4, -NR3R4 ;
-S(O)1(CrC6 alkyl), -S(OUCR4R5)m-ary\, -O(CR4R5)m-aryl, -NR4(CR4R5)m-aryl, -O(CR4R5)m-heteroaryl, -NR4(CR4R5)m-heteroaryl, -O(CR4R5)m-heterocyclyl, - NR4(CR4R5)m-heterocyclyl, and -S(C1-C2 alkyl) substituted with 1 to 5 F, where each, aryl, heteroaryl and heterocyclyl is substituted or unsubstituted; L is selected from a bond, CrC10 alkyl, C2-Ci0 alkenyl, C2-Ci0 alkynyl, C3-C10 cycloalkyl, C3-Ci0 cycloalkylalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocyclyl, heterocyclylalkyl, where each alkyl, alkenyl, alkynyl, cycloalkyl, aryl, heteroaryl and heterocyclyl is unsubstituted or substituted; R3 is selected from hydrogen, trifluoromethyl, C1-C10 alkyl, C2-10 alkenyl, C2-Ci0 alkynyl, C3-C10 cycloalkyl, C3-C10 cycloalkylalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocyclyl, and heterocyclylalkyl, where each alkyl, alkenyl, alkynyl, cycloalkyl, aryl, heteroaryl and heterocyclyl is substituted or unsubstituted; R4 is selected from hydrogen or CrC6 alkyl whereby alkyl may be substituted or unsubstituted; or
R3 and R4 can be taken together with the atom to which they are attached to form a 4 to 10 membered heteroaryl or heterocyclic ring, each of which is substituted or unsubstituted; R5 is selected from hydrogen or C1-C6 alkyl whereby alkyl may be substituted or unsubstituted; or
R4 and R5 can be taken together with the atom to which they are attached to form a 4 to 10 membered carbocyclic, heteroaryl or heterocyclic ring, each of which is substituted or unsubstituted; R6 is selected from trifluoromethyl, CrC10 alkyl, C3-C10 cycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocyclyl, and heterocyclylalkyl, where each alkyl, cycloalkyl, aryl, heteroaryl and heterocyclyl substituted or unsubstituted;
W is selected from heteroaryl containing 1-4 heteroatoms or heterocyclyl containing 1-4 heteroatoms each of which is unsubstituted or substituted by 1 to 5 substituents ZR15; or W is -C(O)ORi5, -C(O)NR4R15, -C(O)NR4OR15, -C(O)(C3-C10 cycloalkyl),
-C(O)(C2-Ci0 alkyl), -C(O)(aryl), -C(O)(heteroaryl), -C(O)(heterocyclyl), -S(O)jNR4R15l -S(O)JNR4ORi5, -S(O)JNR4C(O)Ri5, or -C(O)NR4S(O)1R6, whereby R4 and R15 are as defined herein or may form together a 3 to 7 membered ring with 1 or 2 N atoms and optionally an O atom, Z is a bond, NR16, O, NR16SO2 Or S,
R15 is independently selected from hydrogen, trifluoromethyl, C1-C10 alkyl, C2-C10 alkenyl, C2-Ci0 alkynyl, C3-Ci0 cycloalkyl, C3-Ci0 cycloalkylalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocyclyl, and heterocyclylalkyl, where each alkyl, alkenyl, alkynyl, cycloalkyl, aryl, heteroaryl and heterocyclyl is substituted or unsubstituted; R16 is selected from hydrogen or C1-C10 alkyl, or Ri5 and Ri6 form together a 4 to 10 membered cyclic ring with 1 or 2 N atoms and optionally an O atom, said ring being substituted or unsubstituted; m is O, 1 , 2, 3, 4 or 5 ;and j is 1 or 2.
2. The compound of Formula (I) according to claimi wherein
R1, R2, Ra, Rn are selected independently from hydrogen, halo, Ci-C4 alkyl, C3-C4 cycloalkyl, C2-C4 alkenyl, C2-C4 alkynyl, cyano, nitro, OR3 or NR3R4 where each alkyl, alkenyl, alkynyl, cycloalkyl is optionally substituted with one to five halogens;
Rio is selected from hydrogen, -OR3, -NR4C(O)R3, -C(O)NR3R4, -NR3R4; L is selected from a bond or Ci-C5 alkyl; Ri2 is selected independently from hydrogen, halo, C1-C10 alkyl, C3-Ci0 cycloalkyl, C2-Ci0 alkenyl, C2-Ci0 alkynyl, cyano, nitro, azido; NR4SO2R6; SO2NR3R4; SO2R6; C(O)NR3R4; -S(O)JNR4C(O)R3, -C(O)NR4S(O)JR6, OR3, NR3R4 Or -S(C1-C2 alkyl) substituted with 1 to 5 F, where each alkyl, alkenyl, alkynyl, cycloalkyl, aryl, heteroaryl and heterocyclyl is substituted or unsubstituted; R13 and R14 are selected independently from H, F, Cl and C1-C4 alkyl, C3-C4 cycloalkyl, C2-C4 alkenyl, C2-C4 alkynyl where each alkyl, alkenyl, cycloalky, alkynyl is optionally further substituted with one to five halogens;
W is selected from heteroaryl containing 1-4 heteroatoms, heterocyclyl containing 1-4 heteroatoms each of which is unsubstituted or substituted by 1 to 3 substituents ZR15, or W is -C(O)ORi5, -C(O)NR4Ri5, -C(O)NR4OR15, -C(O)(C3-Ci0 cycloalkyl),
-C(O)(C2-Ci0 alkyl), -S(O)jNR4C(O)Ri5, -C(O)NR4S(O)jR6, S(O)jN, NR4R15 Or S(O)JNR4OR15;
Z is selected from NR16, NR16SO2 or O;
Ri5 is selected from hydrogen, C1-C4 alkyl, Ci-C4 alkenyl, C4-C6 cycloalkylalkyl, where alkyl or alkenyl may be further substituted by 1 or 2 of OH, 0-Ci-C4 alkyl or NR'R";
R16 is selected from hydrogen or Ci-C4 alkyl;
R' and R" are each independently selected from hydrogen, C1-C4 alkyl, C2-C4 alkenyl, aryl and arylalkyl.
3. The compound of Formula (I) according to claim 1 or 2 wherein Ri is selected independently from H and F;
R2 is selected independently from F, Cl1 Me, where the methyl group is optionally substituted with one to three fluorines;
R9 is selected independently from H, F, Cl; R10 is selected from hydrogen, -OR3, -NR3R4, wherein R3 and R4 are independently Ci- C6 alkyl, optionally substituted by 1 or 2 alkyl amino or O-alkyl, or R3 and R4 form together a cyclic ring with 1 or 2 N atoms and optionally an O atom, said ring being optionally substituted by 1 or 2 alkyl amino or O-alkyl
L is selected from a bond, methylene, ethylene, n-propylene or n-butylene; R11 is selected independently from H, F, Cl, Br, Me, OMe, where the methyl groups are optionally substituted with one to three fluorines;
R12 is selected independently from H, F, Cl, Br, I, nitro, methyl, ethyl, n-propyl, i-propyl, cyclopropyl, SCF3, SCHF2, SCH2F, SO2NR3R4 or C(O)NR3R4 or OMe, where the methyl groups are optionally substituted with one to three fluorines, wherein R3 and R4 are independently C1-C6 alkyl, optionally substituted by 1 or 2 alkyl amino or O- alkyl, or R3 and R4 form together a cyclic ring with 1 or 2 N atoms and optionally an O atom, said ring being optionally substituted by 1 or 2 alkyl amino or O-alkyl;
R13 is selected independently from H and F; R14 is selected independently from H and F; W is selected from -C(O)NR4ORi5 or SO2NR4ORi5; or W is
Figure imgf000041_0001
wherein Z is NR16; R15 is C1-C4 alkyl or C1-C4 alkenyl optionally substituted with 1 to 3 substituents OH, O- Me, NH2, N(methyl)2, NHmethyl, NHethyl or N(ethyl)2; Ri6 is hydrogen or C1-C4 alkyl, or R16 and R15 form together a 4 to 10 membered ring with 1 or 2 N atoms and optionally an O atom, said ring being optionally substituted by 1 or 2 alkyl amino, amino, hydroxy or O-alkyl.
Y is O, S or NR'.
4. The compound of Formula (I) according to any of claims 1 to 3 wherein
W is selected from -C(O)NR4ORi5 or SO2NR4OR15, or W is
Figure imgf000042_0001
wherein
R4 is hydrogen;
Z is NH,
R15 is selected from Ci-C4 alkyl or C1-C4 alkenyl that may be further substituted by 1 or 2 of OH, 0-Ci-C4 alkyl or NR1R",
R' and R" are independently hydrogen, methyl or ethyl; and
Y is O.
5. The compound of Formula (I) according to any of claims 1 to 4 wherein LR10 are together methyl or hydrogen.
6. The compound of any of claims 1 to 5 for use as a medicament.
7. Use of the compound according to any of claims 1 to 5, for the preparation of a medicament for the treatment of hyperproliferative diseases or disorders mediated by aberrant proliferation, including cancer.
8. Use of the compound of any of claims 1 to 5 for the preparation of a medicament for the treatment of hyperproliferative diseases related to the hyperactivity of MEK as well as diseases modulated by the MEK cascade in mammals.
9. Use according to claim 8 for the treatment of diseases selected from the group consisting of cancer, inflammation, pancreatitis or kidney disease, pain, benign hyperplasia of the skin, restenosis, prostate, diseases related to vasculogenesis or angiogenesis, tumor angiogenesis, skin diseases selected from psoriasis, eczema, and sclerodema, diabetes, diabetic retinopathy, retinopathy of prematurity, age- related macular degeneration, hemangioma, glioma, melanoma and Kaposi's sarcoma.
10. Use according to claim 8 or 9 for the treatment of cancer or inflammation.
11. Use according to any of claims 8 to 10 for the treatment of cancer selected from the group consisting of ovarian, breast, lung, pancreatic, prostate, colon and epidermoid cancer; or inflammation selected from the group consisting of rheumatoid arthritis, inflammatory bowel disease, atherosclerosis.
12. A pharmaceutical composition which comprises a compound of any of claims 1 to 5 and a pharmaceutically acceptable carrier.
PCT/EP2005/012855 2004-12-01 2005-12-01 [1,2,4]triazolo[4,3-a]pyridine derivatives for the treatment of hyperproliferative diseases WO2006058752A1 (en)

Priority Applications (9)

Application Number Priority Date Filing Date Title
CA002586796A CA2586796A1 (en) 2004-12-01 2005-12-01 [1,2,4]triazolo[4,3-a]pyridine derivatives for the treatment of hyperproliferative diseases
US11/791,799 US20070299103A1 (en) 2004-12-01 2005-12-01 [1,2,4]Triazolo[4,3-A]Pyridine Derivatives for the Treatment of Hyperproliferative Diseases
AT05826556T ATE443063T1 (en) 2004-12-01 2005-12-01 A1,2,4ÜTRIAZOLOÄ4,3-AÜPYRIDINE DERIVATIVES FOR THE TREATMENT OF HYPERPROLIFERATIVE DISEASES
AU2005311451A AU2005311451A1 (en) 2004-12-01 2005-12-01 [1,2,4]triazolo[4,3-a]pyridine derivatives for the treatment of hyperproliferative diseases
JP2007543779A JP2008521858A (en) 2004-12-01 2005-12-01 [1,2,4] Triazolo [4,3-a] pyridine derivatives for treating hyperproliferative diseases
EP05826556A EP1828184B1 (en) 2004-12-01 2005-12-01 [1,2,4]triazolo[4,3-a]pyridine derivatives for the treatment of hyperproliferative diseases
DE602005016718T DE602005016718D1 (en) 2004-12-01 2005-12-01 Ä1,2,4ÜTRIAZOLOÄ4,3-AÜPYRIDIN DERIVATIVE FOR THE TREATMENT OF HYPERPROLIFERATIVE DISEASES
IL183340A IL183340A0 (en) 2004-12-01 2007-05-21 [1,2,4] triazolo[4,3-a] pyridine derivatives and pharmaceutical compositions containing them
NO20073393A NO20073393L (en) 2004-12-01 2007-07-02 [1,2,4] tnazolo [4,3-A] pyridine derivatives for the treatment of hyperproliferative diseases

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP04028439 2004-12-01
EP04028439.0 2004-12-01

Publications (1)

Publication Number Publication Date
WO2006058752A1 true WO2006058752A1 (en) 2006-06-08

Family

ID=34927603

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2005/012855 WO2006058752A1 (en) 2004-12-01 2005-12-01 [1,2,4]triazolo[4,3-a]pyridine derivatives for the treatment of hyperproliferative diseases

Country Status (11)

Country Link
US (1) US20070299103A1 (en)
EP (1) EP1828184B1 (en)
JP (1) JP2008521858A (en)
AT (1) ATE443063T1 (en)
AU (1) AU2005311451A1 (en)
CA (1) CA2586796A1 (en)
DE (1) DE602005016718D1 (en)
ES (1) ES2330872T3 (en)
IL (1) IL183340A0 (en)
NO (1) NO20073393L (en)
WO (1) WO2006058752A1 (en)

Cited By (36)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009085980A1 (en) * 2007-12-19 2009-07-09 Genentech, Inc. 8-anilin0imidaz0pyridines and their use as anti-cancer and/or anti-inflammatory agents
US7572807B2 (en) 2005-06-09 2009-08-11 Bristol-Myers Squibb Company Heteroaryl 11-beta-hydroxysteroid dehydrogenase type I inhibitors
US7579360B2 (en) 2005-06-09 2009-08-25 Bristol-Myers Squibb Company Triazolopyridine 11-beta hydroxysteroid dehydrogenase type I inhibitors
WO2010022081A1 (en) * 2008-08-19 2010-02-25 Array Biopharma Inc. Triazolopyridine compounds as pim kinase inhibitors
JP2010516698A (en) * 2007-01-19 2010-05-20 アルデア バイオサイエンシズ,インコーポレイティド MEK inhibitor
WO2010068738A1 (en) 2008-12-10 2010-06-17 Dana-Farber Cancer Institute, Inc. Mek mutations conferring resistance to mek inhibitors
JP2011507889A (en) * 2007-12-19 2011-03-10 ジェネンテック, インコーポレイテッド 5-Anilinoimidazopyridine and method of use
US7943659B2 (en) 2006-10-31 2011-05-17 Takeda Pharmaceutical Company Limited MAPK/ERK kinase inhibitors
WO2011106298A1 (en) 2010-02-25 2011-09-01 Dana-Farber Cancer Institute, Inc. Braf mutations conferring resistance to braf inhibitors
US8022057B2 (en) 2007-11-12 2011-09-20 Takeda Pharmaceutical Company Limited MAPK/ERK kinase inhibitors
US8030317B2 (en) 2006-12-20 2011-10-04 Takeda Pharmaceutical Company Limited MAPK/ERK kinase inhibitors
US8119658B2 (en) 2007-10-01 2012-02-21 Bristol-Myers Squibb Company Triazolopyridine 11-beta hydroxysteroid dehydrogenase type I inhibitors
AU2007272783B2 (en) * 2006-07-14 2012-04-26 Amgen Inc. Fused heterocyclic derivatives and methods of use
US8198448B2 (en) 2006-07-14 2012-06-12 Amgen Inc. Fused heterocyclic derivatives and methods of use
US8217177B2 (en) 2006-07-14 2012-07-10 Amgen Inc. Fused heterocyclic derivatives and methods of use
US8575145B2 (en) 2008-08-19 2013-11-05 Array Biopharma Inc. Triazolopyridine compounds as PIM kinase inhibitors
WO2013169858A1 (en) 2012-05-08 2013-11-14 The Broad Institute, Inc. Diagnostic and treatment methods in patients having or at risk of developing resistance to cancer therapy
US8889704B2 (en) 2011-02-25 2014-11-18 Array Biopharma Inc. Triazolopyridine compounds as PIM kinase inhibitors
US8895550B2 (en) 2008-08-19 2014-11-25 Array Biopharma Inc. Triazolopyridine compounds as PIM kinase inhibitors
US8987251B2 (en) 2008-08-19 2015-03-24 Array Biopharma Inc. Triazolopyridine compounds as PIM kinase inhibitors
US9493450B2 (en) 2014-02-13 2016-11-15 Incyte Corporation Cyclopropylamines as LSD1 inhibitors
US9493442B2 (en) 2014-02-13 2016-11-15 Incyte Corporation Cyclopropylamines as LSD1 inhibitors
US9527835B2 (en) 2014-02-13 2016-12-27 Incyte Corporation Cyclopropylamines as LSD1 inhibitors
US9670210B2 (en) 2014-02-13 2017-06-06 Incyte Corporation Cyclopropylamines as LSD1 inhibitors
US9695167B2 (en) 2014-07-10 2017-07-04 Incyte Corporation Substituted triazolo[1,5-a]pyridines and triazolo[1,5-a]pyrazines as LSD1 inhibitors
US9695168B2 (en) 2014-07-10 2017-07-04 Incyte Corporation Substituted imidazo[1,5-α]pyridines and imidazo[1,5-α]pyrazines as LSD1 inhibitors
US9695180B2 (en) 2014-07-10 2017-07-04 Incyte Corporation Substituted imidazo[1,2-a]pyrazines as LSD1 inhibitors
US9758523B2 (en) 2014-07-10 2017-09-12 Incyte Corporation Triazolopyridines and triazolopyrazines as LSD1 inhibitors
US9944647B2 (en) 2015-04-03 2018-04-17 Incyte Corporation Heterocyclic compounds as LSD1 inhibitors
US10166221B2 (en) 2016-04-22 2019-01-01 Incyte Corporation Formulations of an LSD1 inhibitor
US10329255B2 (en) 2015-08-12 2019-06-25 Incyte Corporation Salts of an LSD1 inhibitor
WO2020188015A1 (en) 2019-03-21 2020-09-24 Onxeo A dbait molecule in combination with kinase inhibitor for the treatment of cancer
US10968200B2 (en) 2018-08-31 2021-04-06 Incyte Corporation Salts of an LSD1 inhibitor and processes for preparing the same
WO2021089791A1 (en) 2019-11-08 2021-05-14 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for the treatment of cancers that have acquired resistance to kinase inhibitors
WO2021148581A1 (en) 2020-01-22 2021-07-29 Onxeo Novel dbait molecule and its use
US11078540B2 (en) 2010-03-09 2021-08-03 Dana-Farber Cancer Institute, Inc. Methods of diagnosing and treating cancer in patients having or developing resistance to a first cancer therapy

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2377821T3 (en) * 2006-10-11 2012-04-02 Amgen Inc. Imidazo- and triazolo-pyridine compounds and methods of use thereof.
WO2015081280A1 (en) * 2013-11-26 2015-06-04 Coferon, Inc. Bromodomain ligands capable of dimerizing in an aqueous solution
US20190008859A1 (en) 2015-08-21 2019-01-10 Acerta Pharma B.V. Therapeutic Combinations of a MEK Inhibitor and a BTK Inhibitor
US11492362B1 (en) 2022-02-17 2022-11-08 King Abdulaziz University Pyridine derivatives for the treatment of hyperproliferative diseases

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999001421A1 (en) * 1997-07-01 1999-01-14 Warner-Lambert Company 2-(4-bromo or 4-iodo phenylamino) benzoic acid derivatives and their use as mek inhibitors
WO2003077855A2 (en) * 2002-03-13 2003-09-25 Array Biopharma, Inc N3 alkylated benzimidazole derivatives as mek inhibitors
WO2003077914A1 (en) * 2002-03-13 2003-09-25 Array Biopharma, Inc N3 alkylated benzimidazole derivatives as mek inhibitors

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050049276A1 (en) * 2003-07-23 2005-03-03 Warner-Lambert Company, Llc Imidazopyridines and triazolopyridines
US7144907B2 (en) * 2003-09-03 2006-12-05 Array Biopharma Inc. Heterocyclic inhibitors of MEK and methods of use thereof

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999001421A1 (en) * 1997-07-01 1999-01-14 Warner-Lambert Company 2-(4-bromo or 4-iodo phenylamino) benzoic acid derivatives and their use as mek inhibitors
WO2003077855A2 (en) * 2002-03-13 2003-09-25 Array Biopharma, Inc N3 alkylated benzimidazole derivatives as mek inhibitors
WO2003077914A1 (en) * 2002-03-13 2003-09-25 Array Biopharma, Inc N3 alkylated benzimidazole derivatives as mek inhibitors

Cited By (74)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8148396B2 (en) 2005-06-09 2012-04-03 Bristol-Myers Squibb Company Triazolopyridine 11-beta hydroxysteroid dehydrogenase type I inhibitors
US7572807B2 (en) 2005-06-09 2009-08-11 Bristol-Myers Squibb Company Heteroaryl 11-beta-hydroxysteroid dehydrogenase type I inhibitors
US7579360B2 (en) 2005-06-09 2009-08-25 Bristol-Myers Squibb Company Triazolopyridine 11-beta hydroxysteroid dehydrogenase type I inhibitors
US8158648B2 (en) 2005-06-09 2012-04-17 Li James J Heteroaryl 11-beta hydroxysteroid dehydrogenase type I inhibitors
US8217177B2 (en) 2006-07-14 2012-07-10 Amgen Inc. Fused heterocyclic derivatives and methods of use
US8524900B2 (en) 2006-07-14 2013-09-03 Amgen Inc. Fused heterocyclic derivatives and methods of use
EP2081931B1 (en) * 2006-07-14 2016-08-31 Amgen Inc. [1,2,4]Triazolo[4,3-a]pyridine derivatives useful as inhibitors of the hepatocyte growth factor receptor
US8212041B2 (en) 2006-07-14 2012-07-03 Amgen Inc. Fused heterocyclic derivatives and methods of use
US8198448B2 (en) 2006-07-14 2012-06-12 Amgen Inc. Fused heterocyclic derivatives and methods of use
EP3093289A1 (en) * 2006-07-14 2016-11-16 Amgen, Inc [1,2,4]triazolo[4,3-a]pyridine derivatives useful as inhibitors of the hepatocyte growth factor receptor
KR101140474B1 (en) * 2006-07-14 2012-05-08 암젠 인크 Fused heterocyclic derivatives useful as inhibitors of the hepatocyte growth factor receptor
AU2007272783B2 (en) * 2006-07-14 2012-04-26 Amgen Inc. Fused heterocyclic derivatives and methods of use
US9066954B2 (en) 2006-07-14 2015-06-30 Amgen Inc. Fused heterocyclic derivatives and methods of use
US7943659B2 (en) 2006-10-31 2011-05-17 Takeda Pharmaceutical Company Limited MAPK/ERK kinase inhibitors
US8470837B2 (en) 2006-12-20 2013-06-25 Takeda Pharmaceutical Company Limted MAPK/ERK kinase inhibitors
US8030317B2 (en) 2006-12-20 2011-10-04 Takeda Pharmaceutical Company Limited MAPK/ERK kinase inhibitors
JP2010516698A (en) * 2007-01-19 2010-05-20 アルデア バイオサイエンシズ,インコーポレイティド MEK inhibitor
US8119658B2 (en) 2007-10-01 2012-02-21 Bristol-Myers Squibb Company Triazolopyridine 11-beta hydroxysteroid dehydrogenase type I inhibitors
US8541444B2 (en) 2007-10-01 2013-09-24 Bristol-Myers Squibb Company Triazolopyridine 11-beta hydroxysteroid dehydrogenase type I inhibitors
US8022057B2 (en) 2007-11-12 2011-09-20 Takeda Pharmaceutical Company Limited MAPK/ERK kinase inhibitors
JP2011507889A (en) * 2007-12-19 2011-03-10 ジェネンテック, インコーポレイテッド 5-Anilinoimidazopyridine and method of use
AU2008343062B2 (en) * 2007-12-19 2013-03-07 Genentech, Inc. 8-Anilinoimidazopyridines and their use as anti-cancer and/or anti-inflammatory agents
JP2011507888A (en) * 2007-12-19 2011-03-10 ジェネンテック, インコーポレイテッド 8-Anilinoimidazopyridine and its use as an anti-cancer and / or anti-inflammatory agent
US9206174B2 (en) 2007-12-19 2015-12-08 Genentech, Inc. 5-anilinoimidazopyridines and methods of use
CN101903387B (en) * 2007-12-19 2014-05-21 健泰科生物技术公司 8-anilin0imidaz0pyridines and application thereof as anti-cancer and/or anti-inflammatory agents
WO2009085980A1 (en) * 2007-12-19 2009-07-09 Genentech, Inc. 8-anilin0imidaz0pyridines and their use as anti-cancer and/or anti-inflammatory agents
US8557809B2 (en) 2008-08-19 2013-10-15 Array Biopharma Inc. Triazolopyridine compounds as PIM kinase inhibitors
US8575145B2 (en) 2008-08-19 2013-11-05 Array Biopharma Inc. Triazolopyridine compounds as PIM kinase inhibitors
US8895550B2 (en) 2008-08-19 2014-11-25 Array Biopharma Inc. Triazolopyridine compounds as PIM kinase inhibitors
US8987251B2 (en) 2008-08-19 2015-03-24 Array Biopharma Inc. Triazolopyridine compounds as PIM kinase inhibitors
WO2010022081A1 (en) * 2008-08-19 2010-02-25 Array Biopharma Inc. Triazolopyridine compounds as pim kinase inhibitors
WO2010068738A1 (en) 2008-12-10 2010-06-17 Dana-Farber Cancer Institute, Inc. Mek mutations conferring resistance to mek inhibitors
US9084781B2 (en) 2008-12-10 2015-07-21 Novartis Ag MEK mutations conferring resistance to MEK inhibitors
EP3028699A1 (en) 2010-02-25 2016-06-08 Dana-Farber Cancer Institute, Inc. Braf mutations conferring resistance to braf inhibitors
WO2011106298A1 (en) 2010-02-25 2011-09-01 Dana-Farber Cancer Institute, Inc. Braf mutations conferring resistance to braf inhibitors
US9279144B2 (en) 2010-02-25 2016-03-08 Dana-Farber Cancer Institute, Inc. Screening method for BRAF inhibitors
US11078540B2 (en) 2010-03-09 2021-08-03 Dana-Farber Cancer Institute, Inc. Methods of diagnosing and treating cancer in patients having or developing resistance to a first cancer therapy
US8889704B2 (en) 2011-02-25 2014-11-18 Array Biopharma Inc. Triazolopyridine compounds as PIM kinase inhibitors
WO2013169858A1 (en) 2012-05-08 2013-11-14 The Broad Institute, Inc. Diagnostic and treatment methods in patients having or at risk of developing resistance to cancer therapy
US11247992B2 (en) 2014-02-13 2022-02-15 Incyte Corporation Cyclopropylamines as LSD1 inhibitors
US9527835B2 (en) 2014-02-13 2016-12-27 Incyte Corporation Cyclopropylamines as LSD1 inhibitors
US9670210B2 (en) 2014-02-13 2017-06-06 Incyte Corporation Cyclopropylamines as LSD1 inhibitors
US10174030B2 (en) 2014-02-13 2019-01-08 Incyte Corporation Cyclopropylamines as LSD1 inhibitors
US11155532B2 (en) 2014-02-13 2021-10-26 Incyte Corporation Cyclopropylamines as LSD1 inhibitors
US9493442B2 (en) 2014-02-13 2016-11-15 Incyte Corporation Cyclopropylamines as LSD1 inhibitors
US9493450B2 (en) 2014-02-13 2016-11-15 Incyte Corporation Cyclopropylamines as LSD1 inhibitors
US10717737B2 (en) 2014-02-13 2020-07-21 Incyte Corporation Cyclopropylamines as LSD1 inhibitors
US9994546B2 (en) 2014-02-13 2018-06-12 Incyte Corporation Cyclopropylamines as LSD1 inhibitors
US10676457B2 (en) 2014-02-13 2020-06-09 Incyte Corporation Cyclopropylamines as LSD1 inhibitors
US10513493B2 (en) 2014-02-13 2019-12-24 Incyte Corporation Cyclopropylamines as LSD1 inhibitors
US10300051B2 (en) 2014-02-13 2019-05-28 Incyte Corporation Cyclopropylamines as LSD1 inhibitors
US9695180B2 (en) 2014-07-10 2017-07-04 Incyte Corporation Substituted imidazo[1,2-a]pyrazines as LSD1 inhibitors
US10968221B2 (en) 2014-07-10 2021-04-06 Incyte Corporation Substituted [1,2,4]triazolo[1,5-a]pyrazines as LSD1 inhibitors
US10138249B2 (en) 2014-07-10 2018-11-27 Incyte Corporation Triazolopyridines and triazolopyrazines as LSD1 inhibitors
US10125133B2 (en) 2014-07-10 2018-11-13 Incyte Corporation Substituted [1,2,4]triazolo[1,5-a]pyridines and substituted [1,2,4]triazolo[1,5-a]pyrazines as LSD1 inhibitors
US9695167B2 (en) 2014-07-10 2017-07-04 Incyte Corporation Substituted triazolo[1,5-a]pyridines and triazolo[1,5-a]pyrazines as LSD1 inhibitors
US10112950B2 (en) 2014-07-10 2018-10-30 Incyte Corporation Substituted imidazo[1,2-a]pyrazines as LSD1 inhibitors
US10556908B2 (en) 2014-07-10 2020-02-11 Incyte Corporation Substituted imidazo[1,2-a]pyrazines as LSD1 inhibitors
US10640503B2 (en) 2014-07-10 2020-05-05 Incyte Corporation Imidazopyridines and imidazopyrazines as LSD1 inhibitors
US10047086B2 (en) 2014-07-10 2018-08-14 Incyte Corporation Imidazopyridines and imidazopyrazines as LSD1 inhibitors
US9695168B2 (en) 2014-07-10 2017-07-04 Incyte Corporation Substituted imidazo[1,5-α]pyridines and imidazo[1,5-α]pyrazines as LSD1 inhibitors
US9758523B2 (en) 2014-07-10 2017-09-12 Incyte Corporation Triazolopyridines and triazolopyrazines as LSD1 inhibitors
US10800779B2 (en) 2015-04-03 2020-10-13 Incyte Corporation Heterocyclic compounds as LSD1 inhibitors
US11401272B2 (en) 2015-04-03 2022-08-02 Incyte Corporation Heterocyclic compounds as LSD1 inhibitors
US9944647B2 (en) 2015-04-03 2018-04-17 Incyte Corporation Heterocyclic compounds as LSD1 inhibitors
US11498900B2 (en) 2015-08-12 2022-11-15 Incyte Corporation Salts of an LSD1 inhibitor
US10723700B2 (en) 2015-08-12 2020-07-28 Incyte Corporation Salts of an LSD1 inhibitor
US10329255B2 (en) 2015-08-12 2019-06-25 Incyte Corporation Salts of an LSD1 inhibitor
US10166221B2 (en) 2016-04-22 2019-01-01 Incyte Corporation Formulations of an LSD1 inhibitor
US10968200B2 (en) 2018-08-31 2021-04-06 Incyte Corporation Salts of an LSD1 inhibitor and processes for preparing the same
US11512064B2 (en) 2018-08-31 2022-11-29 Incyte Corporation Salts of an LSD1 inhibitor and processes for preparing the same
WO2020188015A1 (en) 2019-03-21 2020-09-24 Onxeo A dbait molecule in combination with kinase inhibitor for the treatment of cancer
WO2021089791A1 (en) 2019-11-08 2021-05-14 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for the treatment of cancers that have acquired resistance to kinase inhibitors
WO2021148581A1 (en) 2020-01-22 2021-07-29 Onxeo Novel dbait molecule and its use

Also Published As

Publication number Publication date
CA2586796A1 (en) 2006-06-08
ATE443063T1 (en) 2009-10-15
EP1828184B1 (en) 2009-09-16
AU2005311451A1 (en) 2006-06-08
EP1828184A1 (en) 2007-09-05
US20070299103A1 (en) 2007-12-27
DE602005016718D1 (en) 2009-10-29
ES2330872T3 (en) 2009-12-16
NO20073393L (en) 2007-09-03
JP2008521858A (en) 2008-06-26
IL183340A0 (en) 2007-09-20

Similar Documents

Publication Publication Date Title
EP1828184B1 (en) [1,2,4]triazolo[4,3-a]pyridine derivatives for the treatment of hyperproliferative diseases
US20070293544A1 (en) Novel 4-Arylamino Pyridone Derivatives as Mek Inhibitors for the Treatment of Hyper-Proliferative Disorders
CA2582247C (en) 3-arylamino pyridine derivatives
JP5710268B2 (en) 8-Anilinoimidazopyridine and its use as an anti-cancer and / or anti-inflammatory agent
CA2732828C (en) Phenylamino isonicotinamide compounds
JP2011507889A (en) 5-Anilinoimidazopyridine and method of use
WO2005051300A2 (en) Bicyclic inhibitors of mek and methods of use thereof
BRPI0714629A2 (en) compound, pharmaceutical composition, method for inhibiting cell growth, method for treating an inflammatory disease and method for treating an autoimmune disease, destructive bone damage, proliferative disorders, infectious diseases, viral diseases, fibrotic diseases or neurodegenerative diseases
Deppe et al. HAMILTON, BROOK, SMITH 4 REYNOLDS

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KM KN KP KR KZ LC LK LR LS LT LU LV LY MA MD MG MK MN MW MX MZ NA NG NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SM SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): BW GH GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LT LU LV MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2586796

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2005311451

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2005826556

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 183340

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 11791799

Country of ref document: US

ENP Entry into the national phase

Ref document number: 2005311451

Country of ref document: AU

Date of ref document: 20051201

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2007543779

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

WWP Wipo information: published in national office

Ref document number: 2005826556

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 11791799

Country of ref document: US