WO2005103028A1 - 2-pyridyl substituted imidazoles as alk5 and/or alk4 inhibitors - Google Patents

2-pyridyl substituted imidazoles as alk5 and/or alk4 inhibitors Download PDF

Info

Publication number
WO2005103028A1
WO2005103028A1 PCT/KR2004/002891 KR2004002891W WO2005103028A1 WO 2005103028 A1 WO2005103028 A1 WO 2005103028A1 KR 2004002891 W KR2004002891 W KR 2004002891W WO 2005103028 A1 WO2005103028 A1 WO 2005103028A1
Authority
WO
WIPO (PCT)
Prior art keywords
imidazol
benzamide
quinoxalin
alkyl
methyl
Prior art date
Application number
PCT/KR2004/002891
Other languages
French (fr)
Inventor
Dae-Kee Kim
Yung-Jue Bang
Hun-Taek Kim
Il-Sang Cho
Myoung-Soon Park
Young Jae An
Joon Hun Choi
Original Assignee
In2Gen Co., Ltd
Sk Chemicals Co., Ltd.
Ewha University Collaboration Foundation
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
Priority to US10/983,227 priority Critical patent/US7407958B2/en
Application filed by In2Gen Co., Ltd, Sk Chemicals Co., Ltd., Ewha University Collaboration Foundation filed Critical In2Gen Co., Ltd
Priority to JP2007509388A priority patent/JP4678540B2/en
Priority to MXPA06012096A priority patent/MXPA06012096A/en
Priority to CN2004800433657A priority patent/CN1972921B/en
Priority to BRPI0418765A priority patent/BRPI0418765B8/en
Priority to EP04800071A priority patent/EP1620426A1/en
Priority to CA2564442A priority patent/CA2564442C/en
Priority to AU2004318777A priority patent/AU2004318777B2/en
Publication of WO2005103028A1 publication Critical patent/WO2005103028A1/en
Priority to NO20064771A priority patent/NO20064771L/en
Priority to IL178757A priority patent/IL178757A/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C04CEMENTS; CONCRETE; ARTIFICIAL STONE; CERAMICS; REFRACTORIES
    • C04BLIME, MAGNESIA; SLAG; CEMENTS; COMPOSITIONS THEREOF, e.g. MORTARS, CONCRETE OR LIKE BUILDING MATERIALS; ARTIFICIAL STONE; CERAMICS; REFRACTORIES; TREATMENT OF NATURAL STONE
    • C04B35/00Shaped ceramic products characterised by their composition; Ceramics compositions; Processing powders of inorganic compounds preparatory to the manufacturing of ceramic products
    • C04B35/622Forming processes; Processing powders of inorganic compounds preparatory to the manufacturing of ceramic products
    • C04B35/626Preparing or treating the powders individually or as batches ; preparing or treating macroscopic reinforcing agents for ceramic products, e.g. fibres; mechanical aspects section B
    • C04B35/63Preparing or treating the powders individually or as batches ; preparing or treating macroscopic reinforcing agents for ceramic products, e.g. fibres; mechanical aspects section B using additives specially adapted for forming the products, e.g.. binder binders
    • C04B35/632Organic additives
    • 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
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • 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
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/10Drugs for genital or sexual disorders; Contraceptives for impotence
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • 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
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • 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
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings

Definitions

  • This invention relates to 2-pyridyl substituted imidazoles which are inhibitors of the transforming growth factor- ⁇ (TGF- ⁇ ) type I receptor (ALK5) and/or the activin type I receptor (ALK4), methods for their preparation, and their use in medicine, specifically in the treatment and prevention of a disease state mediated by these receptors.
  • TGF- ⁇ transforming growth factor- ⁇
  • ALK4 activin type I receptor
  • TGF- ⁇ denotes a family of proteins, TGF- ⁇ 1, TGF- ⁇ 2 and TGF- ⁇ 3, which are pleiotropic modulators of cell proliferation and differentiation, wound healing, extracellular matrix production and immunosuppression.
  • Other members of this superfamily include activins, inhibins, bone morphogenetic proteins, growth and differentiation factors and M ⁇ llerian inhibiting substance.
  • TGF- ⁇ 1 transduces signals through two highly conserved single transmembrane serine/threonine kinases, the type I (ALK5) and type II TGF- ⁇ receptors.
  • the type II receptor hyperphosphorylates serine/threonine residues in the GS region of the ALK5, which leads to activation of the ALK5 by creating a binding site for Smad proteins.
  • the activated ALK5 in turn phosphorylates Smad2 and Smad3 proteins at the C- terminal SSXS-motif thereby causing their dissociation from the receptor and heteromeric complex formation with Smad4.
  • Smad complexes translocate to the nucleus, assemble with specific DNA-binding co-factors and co-modulators to finally activate transcription of extracellular matrix components and inhibitors of matrix-degrading proteases.
  • Activins transduce signals in a manner similar to TGF- ⁇ .
  • Activins bind to serine/thereonine kinase, the activin type II receptor (ActRIIB), and the activated type II receptor hyperphosphorylates serine/threonine residues in the GS region of the ALK4.
  • the activated ALK4 in turn phosphorylates Smad2 and Smad3.
  • the consequent formation of a hetero-Smad complex with Smad4 results in the activin-induced regulation of gene transcription.
  • TGF- ⁇ glomerular expression of TGF- ⁇ and fibrosis
  • Thy-1 rat model of proliferative glomerulonephritis, anti-GBM glomerulonephritis in rabbits and the 5/6 nephrectomy rat model of focal segmental glomerulosclerosis, as has been reviewed recently (e.g., Bitzer, M. et al., Kidney Blood Press. Res. 21:1 — 12 (1998)).
  • Neutralizing antibody to TGF- ⁇ improves glomerular histology in the Thy-1 nephritis model (e.g., Border, W. A. et al., Nature 346: 371 — 374 (1990)).
  • Hyperglycemic conditions increase TGF- ⁇ mRNA and protein synthesis in both murine proximal tubule cells and human mesangial cells (e.g., Wahab, N. A. et al., Biochem. J. 316:985—992 (1996); Rocco, M. V. et al., Kidney Int. 41: 107—114 (1992)).
  • Diabetic patients with early kidney disease show increased accumulation of TGF- ⁇ mRNA and protein within the glomerulus (e.g., Yoshioka, K. et al., Lab. Invest. 68: 15 — 163 (1993)).
  • kidneys with chronic renal interstitial fibrosis the hallmarks are thickened tubular basement membranes and an expanded interstitial compartment, with interstitial fibrosis characterized by an increase in collagens I, III, V, Nil, and fibronectin (e.g., Eddy, A. A., J. Am. Soc. Nephrol 7: 2495—2508 (1996)).
  • TGF- ⁇ gene expression and protein production are increased in a variety of animal models of pulmonary fibrosis including bleomycin, silica, asbestos, and radiation (e.g., Phan, S.
  • COPD is a slowly progressive and irreversible disorder characterized by the functional abnormality of airway obstruction.
  • TGF-D has been hypothesized to be involved in airway remodeling found in chronic airway inflammatory disorders such as COPD (e.g., Takizawa, H. Int. J. Mol. Med. 1: 367—378 (1998); Ning, W et al., Rroc. Natl. Acad. Sci. USA 101:14895—14900 (2004)).
  • Hepatic stellate cells are the major source of extracellular matrix proteins in hepatic fibrosis. Extracellular matrix production by activated hepatic stellate cells is markedly increased through the action of TGF- ⁇ l (e.g., Friedman, S. L., Prog. Liver Dis. 14: 101 — 130 (1996); Pietrangelo, A., Semin. Liver Dis. 16:13 — 30 (1996)).
  • TGF- ⁇ l e.g., Friedman, S. L., Prog. Liver Dis. 14: 101 — 130 (1996); Pietrangelo, A., Semin. Liver Dis. 16:13 — 30 (1996).
  • Transgenic mice that overexpress TGF- ⁇ l in the liver develop hepatic fibrosis as well as extrahepatic pathologies such as renal fibrosis (e.g., Sanderson, N. et al., Proc. Natl. Acad. Sci. USA 92:2572—2576 (1995
  • TGF- ⁇ l and its receptors are overexpressed in injured blood vessels and in fibroproliferative vascular lesions leading to overproduction of extracellular matrix (e.g., Saltis, J. et al., Clin. Exp. Pharmacol. Physiol. 23: 193—200 (1996); McCaffrey, T. A. et al, J. Clin. Invest. 96: 2667—2675 (1995)).
  • Anti-TGF- ⁇ antibodies reduce scar formation and improve the cytoarchitecture of the neodermis in rats (e.g., Shah, M., J. Cell. Sci. 108: 985—1002 (1995)), improve healing of corneal wounds in rabbits (e.g., Moller-Pedersen, T., Curr. Eye Res. 17:736 — 747 (1998)), and accelerate wound healing of gastric ulcers in rats (e.g., Ernst, H., Gut 39: 172 — 175 (1996)).
  • TGF- ⁇ l plays a central role in the initiation, development, and persistence of radiation fibrosis, as has been reviewed recently (e.g., Martin, M. et al., Int. J. Radiat. Oncol. Biol. Phys. 47:277—290 (2000)).
  • Organ transplantation is complicated in many instances by chronic rejection and for some organs such as the kidney, it is the major forms of graft loss.
  • chronic rejection of lung and kidney transplants is associated with increased expression of TGF- ⁇ within the tissue (e.g., El-Gamel, A. et al., Eur. J. Cardiothorac. Surg. 13: 424—430 (1998); Shihab, F. S. et al., J. Am. Soc. Nephrol. 6:286—294 (1995)).
  • TGF- ⁇ is implicated in peritoneal adhesions (e.g., Saed, G. M. et al., Wound Repair Regeneration 7: 504 — 510 (1999)).
  • the peritoneal and sub-dermal fibrotic adhesions could be prevented by inhibitors of ALK5 and/or ALK4.
  • the tumor cells and the stromal cells within the tumors in late stages of various cancers generally overexpress TGF- ⁇ . This leads to stimulation of angiogenesis and cell motility, suppression of the immune system, and increased interaction of tumor cells with the extracellular matrix (e.g., Hojo, M. et al., Nature 397: 530 — 534 (1999)). Consequently, the tumor cells become more invasive and metastasize to distant organs (e.g., Maehara, Y. et al., J. Clin. Oncol. 17: 607 — 614 (1999); Picon, A. et al., Cancer Epidemiol. Biomarkers Prev. 7:497—504 (1998)).
  • Plasminogen activator inhibitor- 1 is the major physiological inhibitor of both tissue-type plasminogen activator and urokinase-type plasminogen activator. Elevated levels of PAI-1 are associated with thrombosis and vascular disease, suggesting that high plasma PAI-1 may promote a hypercoagulable state by disrupting the natural balance between fibrinolysis and coagulation (e.g., Naughan, D. E., J. Invest. Med. 46: 370—376 (1998)). It is known that TGF-D stimulates the expression of PAI-1 (e.g., Dennler, S. et al., EMBO J 17: 3091—3100 (1998)). Accordingly, inhibition of the production of PAI-1 with an inhibitor of the TGF-D signaling pathway could produce a novel fibrinolytic therapy.
  • PAI-1 Plasminogen activator inhibitor- 1
  • Activin signaling and overexpression of activin is linked to pathological disorders that involve extracellular matrix accumulation and fibrosis (e.g., Matsuse, T. et al., Am. J. Respir. Cell Mol Biol. 13:17—24 (1995); Inoue, S. et al., Biochem. Biophys. Res. Comm. 205:441— 448 (1994); Matsuse, T. et al., Am. J. Pathol. 148:707—713 (1996); De Bleser et al,
  • Cipriano S. C. et al., Endocrinology 141:2319 — 2327 (2000)
  • diseases or pathological responses in the central nervous system e.g., Logan, A. et al., Eur. J. Neurosci. 11:2367 — 2374
  • TGF- ⁇ and activin can act synergistically to induce extracellular matrix production
  • WO 00/61576 and US 2003/0149277 Al disclose triarylimidazole derivatives and their use as ALK5 inhibitors.
  • WO 01/62756 Al discloses pyridinylimidazole derivatives and their use as ALK5 inhibitors.
  • WO 02/055077 Al discloses use of imidazolyl cyclic acetal derivatives as
  • WO 03/087304 A2 discloses tri-substituted heteroaryls and their use as ALK5 and/or ALK4 inhibitors.
  • ALK5 and/or ALK4 function as potent and selective inhibitors of ALK5 and/or ALK4 and therefore, have utility in the treatment and prevention of various disease states mediated by ALK5 and/or ALK4, such as glomerulonephritis, diabetic nephropathy, lupus nephritis, hypertension-induced nephropathy, renal interstitial fibrosis, renal fibrosis resulting from complications of drug exposure, HIN-associated nephropathy, transplant necropathy, liver fibrosis due to all etiologies, hepatic dysfunction attributable to infections, alcohol-induced hepatitis, disorders of the biliary tree, pulmonary fibrosis, acute lung injury, adult respiratory distress syndrome, idiopathic pulmonary fibrosis, chronic obstructive pulmonary disease, pulmonary disease due to infectious or toxic agents, post-infarction cardiac fibrosis, conges
  • Ri is naphthyl, anthracenyl, or phenyl optionally substituted with substituents selected from halo, OH, -O-C ⁇ ---- 6 alkyl, -S-C ⁇ _ 6 alkyl, C ⁇ _ 6 alkyl, C ⁇ -_ 6 haloalkyl, -O-(CH 2 ) n -Ph, - S-(CH 2 ) n -Ph, cyano, phenyl, and CO 2 R, wherein R is H or Ci— ⁇ alkyl, and n is 0, 1, 2, or 3; or Ri is phenyl or pyridyl fused with an aromatic or non-aromatic cyclic ring of 5 — 7 members wherein said cyclic ring optionally contains up to three heteroatoms, independently selected from ⁇ , O and S, and the fused phenyl or pyridyl may be further optionally substituted by halo, OH, -O-C ⁇ _ 6 al
  • R 4 and R 5 are independently H or C ⁇ _ 6 alkyl; R 6 is C ⁇ _ 6 alkyl; p is 0, 1, 2, 3, or 4; q is 1, 2, 3, or 4; X is d—ioalkylene, C 2 — l oalkenylene, or C 2 — ⁇ 0 alkynylene; One of Ai and A2 is N and the other is NR 7 ; and R 7 is H, OH, C ⁇ _ 6 alkyl, or C 3 -_ 7 cycloalkyl.
  • the double bond indicated by the dotted lines of formula (I), represent the possible tautomeric ring forms of the compounds falling within the scope of this invention, the double bond being to the unsubstituted nitrogen.
  • Ri is optionally substituted naphthyl or phenyl.
  • Ri is phenyl optionally substituted with substituents selected from halo, OH, -O-C-_- 6 alkyl, -S-C ⁇ _ 6 alkyl, and phenyl; or Ri is phenyl fused with an aromatic or non-aromatic cyclic ring of 5 — 7members wherein said cyclic ring optionally contains up to two heteroatoms, independently selected from N, O and S, and the fused phenyl or pyridyl may be further optionally substituted by halo, OH, -O-C ⁇ _ 6 alkyl, -S-C-_ 6 alkyl, C ⁇ _ 6 alkyl, C ⁇ _ 6 haloalkyl or O.
  • Ri represents benzo[l,3]dioxolyl, 2,3-dihydrobenzo[l,4]dioxinyl, benzoxazolyl, benzothiazolyl, benzo[l,2,5]oxadiazolyl, benzo[l,2,5]thiadiazolyl, quinoxalinyl, dihydrobenzofuranyl, benzimidazolyl, C--_ 6 benzimidazolyl, [l,2,4]triazolo[l,5-c-] ⁇ yridyl, benzo[l,4]oxazinyl-3-one, benzoxazolyl-2-one or benzo[l,4]oxazinyl.
  • R2 is other than H.
  • R 2 is other than H, it is preferably positioned ortho to the nitrogen of the pyridyl ring.
  • R 2 is preferably C alkyl.
  • R 3 is - CH 2 ) p -CONHOH, -(CH 2 ) P -CN, -(CH 2 ) p -CO 2 H, -(CH 2 ) p -CONR 4 R 5 , or -(CH 2 ) p -tetrazole.
  • R4 and R 5 are independently H or C ⁇ _ 3 alkyl.
  • p is 0, 1, or 2.
  • X is C ⁇ _ 6 alkylene.
  • one of Ai and A 2 is N and the other is NR 7 , wherein R 7 is H.
  • the compounds of the present invention typically are small organic molecules (non- peptide small molecules), generally less than about 1,000 daltons in size.
  • Preferred non-peptide small molecules have molecular weights of less than about 750 daltons, more preferably less than about 500 daltons, and even more preferably less than about 300 daltons.
  • Compounds of formula (I) may also be supplied in the form of a "prodrug" which is designed to release compound of formula (I) when administered to a subject.
  • Prodrug formed designs are well known in the art, and depend on the substituents contained in compound of formula (I).
  • a substituent containing hydroxyl could be. coupled to a carrier which renders the compound biologically inactive until it is removed by endogenous enzymes or, for example, by enzymes targeted to a particular receptor or location in the subject.
  • a compound of formula (I) that is acidic in nature e.g., having a carboxyl or phenolic hydroxyl group
  • can form a pharmaceutically acceptable salt such as a sodium, potassium, calcium, or gold salt.
  • salts formed with pharmaceutically acceptable amines such as ammonia, alkyl amines, hydroxyalkylamines, and N-methylglycamine.
  • a compound of formula (I) can be treated with an acid to form acid addition salts.
  • acids examples include hydrochloric acid, hydrobromic acid, hydroiodic acid, sulfuric acid, methanesulfonic acid, phosphoric acid, -bromophenylsulfonic acid, carbonic acid, succinic acid, citric acid, benzoic acid, oxalic acid, malonic acid, salicylic acid, malic acid, fumaric acid, ascorbic acid, maleic acid, acetic acid, and other mineral and organic acids well known to those skilled in the art.
  • the acid addition salts can be prepared by treating a compound of formula (I) in its free base form with a sufficient amount of an acid (e.g., hydrochloric acid) to produce an acid addition salt (e.g., a hydrochloride salt).
  • the acid addition salt can be converted back to its free base form by treating the salt with a suitable dilute aqueous basic solution (e.g., sodium hydroxide, sodium bicarbonate, potassium carbonate, or ammonia).
  • Some of the compounds of this invention may be crystallized or recrystallized from solvents such as aqueous and organic solvents, h such cases solvates may be formed.
  • This invention includes within its scope stoichiometric solvates including hydrates as well as compounds containing variable amounts of water that may be produced by processes such as lyophilization.
  • Compounds of formula (I) may contain one or more asymmetric centers and thus can exist as enantiomers or diastereomers. It is to be understood that the invention includes both mixtures and separate individual isomers of compounds of the formula (I).
  • certain compounds of the formula (I) which contain alkenyl groups may exist as cis- or trans-isomers. In each instance, the invention includes both mixtures and separate individual isomers.
  • alkyl refers to a saturated aliphatic hydrocarbon group containing 1 — 10 (e.g., 1 — 6 or 1 — 4) carbon atoms.
  • An alkyl group can be straight or branched. Examples of an alkyl group include, but are not limited to, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, «-heptyl, and 2-ethylhexyl.
  • An alkyl group can be optionally substituted with one or more substituents such as alkoxy, cycloalkoxy, amino, nitro, carboxy, cyano, halo, hydroxy, sulfo, or mercapto.
  • alkylene refers to a saturated aliphatic hydrocarbon group containing 1 — 10 (e.g., 1 — 6 or 1 — 4) carbon atoms.
  • An alkylene group can be straight or branched. Examples of an alkylene group include, but are not limited to, methylene, ethylene, propylene, isopropylene, butylene, isobutylene, sec-butylene, tert-butylene, R-pentylene, n- heptylene, and 2-ethylhexylene.
  • An alkylene group can be optionally substituted with one or more substituents such as alkoxy, cycloalkoxy, amino, nitro, carboxy, cyano, halo, hydroxy, sulfo, or mercapto.
  • alkenylene refers to an aliphatic carbon group that contains 2 — 10 (e.g., 2 — 6 or 2 — 4) carbon atoms and at least one double bond. Like an alkylene group, an alkenylene group can be straight or branched. Examples of an alkenylene group include, but are not limited to, allylene, isoprenylene, 2-butenylene, and 2-hexenylene.
  • An alkenylene group can be optionally substituted with one or more substituents such as alkoxy, cycloalkyloxy, heterocycloalkyloxy, aryloxy, heteroaryloxy, aralkyloxy, heteroarylalkoxy, amino, nitro, carboxy, cyano, halo, hydroxy, sulfo, mercapto, alkylsulfanyl, alkylsulfinyl, alkylsulfonyl, aminocarbonyl, alkylcarbonylamino, cycloalkylcarbonylamino, cycloalkylalkylcarbonylamino, arylcarbonylamino, aralkylcarbonylamino, heterocycloalkylcarbonylamino, heterocycloalkylalkylcarbonylamino, heteroarylcarbonylamino, heteroaralkylcarbonylamino, urea, thiourea, sulfamoyl, sul
  • alkynylene refers to an aliphatic carbon group that contains 2 — 10 (e.g., 2 — 6 or 2 — 4) carbon atoms and has at least one triple bond.
  • An alkynylene group can be straight or branched. Examples of an alkynylene group include, but are not limited to, propargylene and butynylene.
  • An alkynylene group can be optionally substituted with one or more substituents such as alkoxy, cycloalkyloxy, heterocycloalkyloxy, aryloxy, heteroaryloxy, aralkyloxy, heteroarylalkoxy, amino, nitro, carboxy, cyano, halo, hydroxy, sulfo, mercapto, alkylsulfanyl, alkylsulfmyl, alkylsulfonyl, aminocarbonyl, alkylcarbonylamino, cycloalkylcarbonylamino, cycloalkylalkylcarbonylamino, arylcarbonylamino, aralkylcarbonylamino, heterocycloalkylcarbonylamino, heterocycloalkylalkylcarbonylamino, heteroarylcarbonylamino, heteroaralkylcarbonylamino, urea, thiourea, sulfamoyl, sul
  • cycloalkyl refers to an aliphatic carbocyclic ring of
  • cycloalkyl groups include cyclopropyl, cyclopentyl, cyclohexyl, cycloheptyl, adamantly, norbornyl, cubyl, octahydroindenyl, decahydronaphthyl; bicyclo[3.2.1]octyl, bicyclo[2.2.2]octyl, bicyclo[3.3.1]nonyl, and bicyclo[3.2.3]nonyl.
  • alkoxy group refers to an alkyl-O-group where "alkyl” has been defined previously.
  • haloalkyl group refers to an alkyl group containing one or more halogen atoms.
  • haloalkyl groups include fluoromethyl, chloromethyl, bromomethyl, and trifluoromethyl.
  • halogen or halo group refers to fluorine, chlorine, bromine or iodine.
  • ALK5 and/or ALK4 inhibitor refers to a compound, other than inhibitory Smads, e.g. Smad6 and Smad7, which selectively inhibits the ALK5 and/or ALK4 receptors preferentially over p38 or type II receptors.
  • ALK5- and/or ALK4-mediated disease state refers to any disease state which is mediated (or modulated) by ALK5 and/or ALK4, for example, a disease which is modulated by the inhibition of the phosphorylation of Smad2 and Smad3 in the TGF- ⁇ and/or activin signaling pathways.
  • the term “ulcers” is used to include, but not to be limited to, diabetic ulcers, chronic ulcers, gastric ulcers, and duodenal ulcers.
  • Compounds of formula (I) may be prepared by a number of known methods from commercially available or known starting materials. If the starting materials are unavailable from a commercial source, they can be prepared by procedures known in the art.
  • compounds of formula (I) wherein Ai is N and A 2 is NH, or A] is NH and A 2 is N are prepared according to Scheme 1.
  • optionally substituted 2- methylpyridine (II) is deprotonated by a base such as LDA or LiHMDS before reacting with
  • the methoxy-methyl-amide (V) can be prepared by reacting a corresponding acid chloride (IN) with N,O-dimethylhydroxylamine hydrochloride.
  • the ketone (NI) may be oxidized to a diketone (Nil) with HBr in DMSO.
  • This diketone (Nil) can then be condensed with an appropriately substituted aldehyde (NIII) or protected aldehyde derivative in the presence of ammonium acetate to yield a compound of formula (I).
  • R_, R 2 , R 3 , and X have been defined as above.
  • the aldehyde (NIII) can be prepared according to the methods outlined in WO 02/096875 Al and Liquid Crystals 10:273—287 (1991).
  • the ketone (NI) can be treated with sodium nitrite in HC1 or acetic acid to afford an ⁇ -keto-oxime (IX), which can be then condensed with an appropriately substituted aldehyde (NIII) or protected aldehyde derivative in the presence of ammonium acetate to give the N-hydroxyimidazoles. Treatment of this with triethylphophite affords a compound of formula (I).
  • Ri, R 2 , X and P have been defined as above and R4 and R 5 are independently H or C ⁇ _ 6 alkyl.
  • the resulting compounds of this invention represented by the formula (I) — (IX) can be separated and purified by appropriate conventional methods such as column chromatography and recrystallization.
  • Compounds of the invention may be administered by any suitable route, for example by oral, buccal, sub-lingual, rectal, vaginal, nasal, topical or parenteral (including intravenous, intramuscular, subcutaneous and intracoronary) administration.
  • topical formulations of the present invention may be presented as, for instance, ointments, creams or lotions, eye ointments and eye or ear drops, impregnated dressings and aerosols, and may contain appropriate conventional additives such as preservatives, solvents to assist drug penetration and emollients in ointments and creams.
  • the formulations may also contain compatible conventional carriers, such as cream or ointment bases and ethanol or oleyl alcohol for lotions.
  • suitable conventional carriers such as cream or ointment bases and ethanol or oleyl alcohol for lotions.
  • Such carriers may be present as from about 1% up to about 98% of the formulation. More usually, they will form up to about 80% of the formulation.
  • oral, buccal or sub-lingual dosages of a compound of formula (I) will generally be in the range of from 50 — 5000 mg daily for an average adult patient (70 kg).
  • individual tablets or capsules contain from 25 — 500 mg of active compound, in a suitable pharmaceutically acceptable vehicle or carrier, for administration in single or multiple doses, once or several times per day.
  • Dosages for parenteral administration will typically be within the range of from 25 — 250 mg per single dose as required.
  • the physician will determine the actual dosing regimen which will be most suitable for an individual patient and it will vary with the age, weight and response of the particular patient.
  • the above dosages are exemplary of the average case but there can be individual instances in which higher or lower dosage ranges may be merited, and such are within the scope of this invention.
  • a compound of formula (I) can be administered alone, but will generally be administered in admixture with a pharmaceutical carrier selected with regard to the intended route of administration and standard pharmaceutical practice.
  • the compound may be administered orally, buccally or sublingually, in the form of tablets containing excipients such as starch or lactose, or in capsules or ovules either alone or in admixture with excipients, or in the form of elixirs or suspensions containing flavoring or coloring agents.
  • Such liquid preparations may be prepared with pharmaceutically acceptable additives such as suspending agent (e.g.
  • a compound may also be injected parenterally, for example intravenously, intramuscularly, subcutaneously or intracoronarily.
  • the compound is best used in the form of a sterile aqueous solution which may contain other substances, for example, salts, or monosaccharides such as mannitol or glucose, to make the solution isotonic with blood.
  • the invention provides in a further aspect a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of formula (I), or a pharmaceutically acceptable salt or solvate thereof, together with a pharmaceutically acceptable diluent or carrier therefor.
  • the invention also provides a compound of formula (I), or a pharmaceutically acceptable salt or solvate thereof, or a pharmaceutical composition containing either entity, for use in therapy.
  • the invention further provides the use of a compound of formula (I), or a pharmaceutically acceptable salt or solvate thereof, or a pharmaceutical composition containing either entity, for the manufacture of a medicament for the treatment of a disease, mediated by the ALK5 and/or ALK4 receptors in mammals.
  • ALK5- and/or ALK4- mediated disease states include, but are not limited to, glomerulonephritis, diabetic nephropathy, lupus nephritis, hypertension-induced nephropathy, renal interstitial fibrosis, renal fibrosis resulting from complications of drug exposure, HIN- associated nephropathy, transplant necropathy, liver fibrosis due to all etiologies, hepatic dysfunction attributable to infections, alcohol-induced hepatitis, disorders of the biliary tree, pulmonary fibrosis, acute lung injury, adult respiratory distress syndrome, idiopathic pulmonary fibrosis, chronic obstructive pulmonary disease, pulmonary fibrosis due to infectious or toxic agents, post-infarction cardiac fibrosis, congestive heart failure, dilated cardiomyopathy, myocarditis, vascular stenosis, restenosis, atherosclerosis, ocular scarring, corneal scarring, pro
  • the invention further provides a method of inhibiting the TGF- ⁇ and/or activin signaling pathways in mammals, for example, inhibiting the phosphorylation of Smad2 or Smad3 byALK5 and or ALK4.
  • the invention further provides a method of reducing the accumulation of excess extracellular matrix in mammals by inhibiting the TGF- ⁇ and/or activin signaling pathways, for example, inhibiting the phosphorylation of Smad2 or Smad3 by ALK5 and/or ALK4.
  • the invention further provides a method of inhibiting metastasis of tumor cells in mammals by inhibiting the TGF- ⁇ signaling pathway.
  • the invention further provides a method of treating carcinomas mediated by an overexpression of TGF- ⁇ in mammals by inhibiting the TGF- ⁇ signaling pathway.
  • Figure 1 shows effect of Examples 6, 9, 10, 13, and 31 on TGF- ⁇ l -induced SBE-Luc reporter activity in HepG2 cells
  • Figure 2 shows effect of Example 41 on TGF- ⁇ l -induced SBE-Luc reporter activity in HepG2 cells
  • Figure 3 shows effect of Example 41 on TGF- ⁇ l -induced 3TP-Lux reporter activity in HepG2 cells
  • Figure 4 shows effect of Example 41 on TGF- ⁇ l -induced PAI-1 promoter-Luc reporter activity in HepG2 cells
  • Figure 5 shows effect of Example 41 on TGF- ⁇ l -induced PAI-1 expression in HaCaT human keratinocytes
  • Figure 6 shows effect of Example 41 on TGF- ⁇ l -induced Smad2 and Smad3 phosphorylation in human dermal fibroblasts
  • Figures 7 A and 7B show effect of Example 41 on liver fibrosis in rats with bile duct ligation
  • Table 1 shows structures and 1H NMR and MS spectral data of Examples 1 61
  • Figure 8 shows effect of Example 41 on the activities of protein kinases in vitro.
  • the pH of the cooled reaction mixture was adjusted to pH 7 — 8 at 0 ° C with 28 % NH 4 OH, and the reaction mixture was extracted with CH2CI2 (10 mL).
  • the CH2CI 2 solution was washed with water (5 mL) and brine (5 mL), dried (anhydrous Na 2 SO 4 ), filtered, and evaporated to dryness under reduced pressure.
  • the pH of the reaction mixture was adjusted to pH 7 — 8 at 0 ° C with saturated NaHCO 3 solution.
  • the reaction mixture was partitioned between CH 2 CI 2 (40 mL) and water (40 mL).
  • the aqueous layer was repeatedly extracted with CH 2 C1 2 (3 x 15 mL).
  • the combined CH 2 CI 2 solution was dried (anhydrous Na 2 SO 4 ), filtered, and evaporated to dryness under reduced pressure.
  • the ethanol solvent was evaporated off under reduced pressure, and the residue was partitioned between CH 2 CI 2 (30 mL) and H 2 O (50 mL). The aqueous layer was saturated with NaCI and extracted with CH 2 CI 2 (3 x 30 mL). The combined CH2CI2 solution was washed with brine (30 mL), dried (anhydrous Na 2 SO 4 ), filtered, and evaporated to dryness under reduced pressure. The residue was dissolved in anhydrous DMF (20 mL) and treated with triethyl phosphite (2.39 mmol). The mixture was heated at 110 ° C for 3 days, cooled to room temperature, and evaporated to dryness under reduced pressure.
  • the reaction mixture was partitioned between CH 2 C1 2 (30 mL) and water (50 mL), and the aqueous layer was extracted with CH 2 C1 2 (2 x 30 mL), The combined CH 2 C1 2 solution was washed with saturated NaHCO 3 solution (40 mL) and brine (50 mL), dried (anhydrous Na 2 SO 4 ), filtered, and evaporated to dryness under reduced pressure.
  • the pH of the cooled reaction mixture was adjusted to pH 7 — 8 at 0 ° C with 28% NH 4 OH, and the reaction mixture was extracted with CH2CI 2 (3 x 30 mL). The combined CH 2 Cl 2 solution was dried (anhydrous Na 2 SO 4 ), filtered, and evaporated to dryness under reduced pressure.
  • the biological activity of the compounds of the invention may be assessed using the following assays: Cell-Free Assay for Evalutating Inhibition of ALK5 Kinase Phosphorylation of Smad3
  • the His-tagged, constitutively active ALK5 (T204D) and Smad3 full protein were expressed in insect cells using the Invitrogen BacNBlue baculovirus expression system. Expressed proteins were purified with Qiagen Ni-NTA resin column. The purified smad3 protem 200 ng was mixed with 100 ⁇ L of 0.1 M sodium bicarbonate coating buffer and coated into Flash-Plates by pipetting. Plates were covered and incubated at 4 ° C for 16 hours.
  • the purified ALK5 protein 100 ng was mixed with 100 ⁇ L of reaction buffer containing 20 mM Tris-HCl (pH 7.4), 5 mM MgCl 2 , 1 mM CaCl 2 , 1 M DTT, 1 ⁇ M ATP and 2 ⁇ Ci ⁇ - 32 p-ATP, and 1 ⁇ L of each test compound of formula ( I ) prepared in 100% DMSO solution at different concentrations.
  • the assay was then initiated with the addition of ALK5 reaction mixture into Smad3 -coated Flash-Plates, followed by incubation at 30 ° C for 3 hours.
  • Inhibition of the ALK4 kinase phosphorylation of Smad3 by test compounds of formula ( I ) can be determined in a similar manner to that described above for ALK5 inhibition except that a similarly His-tagged ALK4 is used in place of the His-tagged, constitutively active ALK5.
  • Biological activity of the compounds of formula ( I ) was determined by measuring their ability to inhibit TGF-/31 -induced Smad binding element-luciferase (SBE-Luc) reporter activity and PAI-1 -luciferase (p3TP-Lux) reporter activity in HepG2 cells.
  • HepG2 cells were transiently transfected with either SBE-Luc reporter construct or p3TP-Lux reporter construct grown in DMEM medium containing 10% FBS, penicillin 100 U/mL, streptomycin 100 ⁇ g/mL, L-glutamine 2 mM, sodium pyruvate 1 mM, and non-essential amino acids.
  • the transfected cells were then plated at a concentration of 2.5 x 10 cells/well in 96 well plates and starved for 3—6 hours in media with 0.5% FBS at 37 ° C in a 5% CO 2 incubator.
  • the cells were then stimulated with 5 ng/mL TGF-/31 ligand in the starvation media containing 1% DMSO either in the presence or absence of a test compound of formula ( I ) and incubated at 37 °C in a 5% CO 2 incubator for 24 hours.
  • the media was washed out, and the luciferase activity in cell lysates was determined by using a luciferase assay system (Promega).
  • Compounds of formula ( I ) typically exhibited IC 50 values of less than 10 ⁇ M; some exhibited IC50 values of less than 1 ⁇ M; and some even exhibited IC 50 values of less than 50 nM.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Pharmacology & Pharmacy (AREA)
  • General Health & Medical Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Ceramic Engineering (AREA)
  • Manufacturing & Machinery (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Neurology (AREA)
  • Biomedical Technology (AREA)
  • Rheumatology (AREA)
  • Neurosurgery (AREA)
  • Structural Engineering (AREA)
  • Materials Engineering (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Urology & Nephrology (AREA)
  • Inorganic Chemistry (AREA)
  • Vascular Medicine (AREA)
  • Psychiatry (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Pulmonology (AREA)
  • Dermatology (AREA)
  • Hospice & Palliative Care (AREA)
  • Ophthalmology & Optometry (AREA)
  • Immunology (AREA)
  • Oncology (AREA)
  • Gynecology & Obstetrics (AREA)
  • Endocrinology (AREA)

Abstract

This invention relates to 2-pyridyl substituted imidazoles which are inhibitors of the transforming growth factor-β (TGF-β) type I receptor (ALK5) and/or the activin type I receptor (ALK4), methods for their preparation, and their use in medicine, specifically in the treatment and prevention of a disease state mediated by these receptors.

Description

2-PYRIDYL SUBSTITUTED IMIDAZOLES AS ALK5 and/or ALK4 INHIBITORS
Technical Field of the Invention This invention relates to 2-pyridyl substituted imidazoles which are inhibitors of the transforming growth factor-β (TGF-β) type I receptor (ALK5) and/or the activin type I receptor (ALK4), methods for their preparation, and their use in medicine, specifically in the treatment and prevention of a disease state mediated by these receptors.
Background of the Invention TGF-β denotes a family of proteins, TGF-β 1, TGF-β2 and TGF-β3, which are pleiotropic modulators of cell proliferation and differentiation, wound healing, extracellular matrix production and immunosuppression. Other members of this superfamily include activins, inhibins, bone morphogenetic proteins, growth and differentiation factors and Mϋllerian inhibiting substance. TGF-β 1 transduces signals through two highly conserved single transmembrane serine/threonine kinases, the type I (ALK5) and type II TGF-β receptors. Upon ligand induced oligomerization, the type II receptor hyperphosphorylates serine/threonine residues in the GS region of the ALK5, which leads to activation of the ALK5 by creating a binding site for Smad proteins. The activated ALK5 in turn phosphorylates Smad2 and Smad3 proteins at the C- terminal SSXS-motif thereby causing their dissociation from the receptor and heteromeric complex formation with Smad4. Smad complexes translocate to the nucleus, assemble with specific DNA-binding co-factors and co-modulators to finally activate transcription of extracellular matrix components and inhibitors of matrix-degrading proteases. Activins transduce signals in a manner similar to TGF-β. Activins bind to serine/thereonine kinase, the activin type II receptor (ActRIIB), and the activated type II receptor hyperphosphorylates serine/threonine residues in the GS region of the ALK4. The activated ALK4 in turn phosphorylates Smad2 and Smad3. The consequent formation of a hetero-Smad complex with Smad4 results in the activin-induced regulation of gene transcription. Numerous experimental animal studies demonstrate an association between glomerular expression of TGF-β and fibrosis, including the Thy-1 rat model of proliferative glomerulonephritis, anti-GBM glomerulonephritis in rabbits, and the 5/6 nephrectomy rat model of focal segmental glomerulosclerosis, as has been reviewed recently (e.g., Bitzer, M. et al., Kidney Blood Press. Res. 21:1 — 12 (1998)). Neutralizing antibody to TGF-β improves glomerular histology in the Thy-1 nephritis model (e.g., Border, W. A. et al., Nature 346: 371 — 374 (1990)).
Hyperglycemic conditions increase TGF-β mRNA and protein synthesis in both murine proximal tubule cells and human mesangial cells (e.g., Wahab, N. A. et al., Biochem. J. 316:985—992 (1996); Rocco, M. V. et al., Kidney Int. 41: 107—114 (1992)). Diabetic patients with early kidney disease show increased accumulation of TGF-β mRNA and protein within the glomerulus (e.g., Yoshioka, K. et al., Lab. Invest. 68: 15 — 163 (1993)). In kidneys with chronic renal interstitial fibrosis, the hallmarks are thickened tubular basement membranes and an expanded interstitial compartment, with interstitial fibrosis characterized by an increase in collagens I, III, V, Nil, and fibronectin (e.g., Eddy, A. A., J. Am. Soc. Nephrol 7: 2495—2508 (1996)).
TGF-β gene expression and protein production are increased in a variety of animal models of pulmonary fibrosis including bleomycin, silica, asbestos, and radiation (e.g., Phan, S.
H. and Kunkel, S. L., Exp. Lung Res. 18: 29—43 (1992); Williams, A. O. et al, Am. J. Pathol.
142: 1831—1840 (1993); Rube, C. E. et al, Int. J. Radial Oncol. Biol. Phys. 47: 1033—1042
(2000)). Coincident increase in TGF-β 1 protein and collagen gene expression in adjacent tissue slices from idiopathic pulmonary fibrosis is observed in human pulmonary fibrotic disease (e.g., Broekelmann, T. J. et al., Proc. Natl. Acad. Sci. USA 88:6642—6646 (1991)). Increased TGF-β production has been documented in patients with sarcoidosis, pneumoconiosis, asbestosis, and radiation-induced fibrosis (e.g., Khalil, Ν. et al., Am. J. Respir. Cell. Mol. Biol. 14: 131 — 138
(1996); Jagirdar, . et al., Environ. Health Perspect. 105: 1197—1203 (1997)). Anti-TGF-β antibodies and TGF-β-soluble receptors could partially inhibit fibrosis in bleomycin-induced lung fibrosis rodent models (e.g., Giri, S. Ν. et al., Thorax 48: 959—966 (1993); Wang, Q. et al, Thorax 54: 805 — 812 (1999)). Tobacco smoke has been implicated as one of the most important factors that can cause small airway disease followed by chronic obstructive pulmonary disease (COPD) (e.g., Wright, J. M. et al., Am. Rev. Respir. Dis. 146: 240—262 (1992)). COPD is a slowly progressive and irreversible disorder characterized by the functional abnormality of airway obstruction. TGF-D has been hypothesized to be involved in airway remodeling found in chronic airway inflammatory disorders such as COPD (e.g., Takizawa, H. Int. J. Mol. Med. 1: 367—378 (1998); Ning, W et al., Rroc. Natl. Acad. Sci. USA 101:14895—14900 (2004)).
Hepatic stellate cells (HSC) are the major source of extracellular matrix proteins in hepatic fibrosis. Extracellular matrix production by activated hepatic stellate cells is markedly increased through the action of TGF-βl (e.g., Friedman, S. L., Prog. Liver Dis. 14: 101 — 130 (1996); Pietrangelo, A., Semin. Liver Dis. 16:13 — 30 (1996)). Transgenic mice that overexpress TGF-βl in the liver develop hepatic fibrosis as well as extrahepatic pathologies such as renal fibrosis (e.g., Sanderson, N. et al., Proc. Natl. Acad. Sci. USA 92:2572—2576 (1995)).
TGF-βl and its receptors are overexpressed in injured blood vessels and in fibroproliferative vascular lesions leading to overproduction of extracellular matrix (e.g., Saltis, J. et al., Clin. Exp. Pharmacol. Physiol. 23: 193—200 (1996); McCaffrey, T. A. et al, J. Clin. Invest. 96: 2667—2675 (1995)).
Anti-TGF-β antibodies reduce scar formation and improve the cytoarchitecture of the neodermis in rats (e.g., Shah, M., J. Cell. Sci. 108: 985—1002 (1995)), improve healing of corneal wounds in rabbits (e.g., Moller-Pedersen, T., Curr. Eye Res. 17:736 — 747 (1998)), and accelerate wound healing of gastric ulcers in rats (e.g., Ernst, H., Gut 39: 172 — 175 (1996)).
Radiation fibrosis is a frequent sequel of therapeutic or accidental radiation overexposure in normal human tissues. TGF-βl plays a central role in the initiation, development, and persistence of radiation fibrosis, as has been reviewed recently (e.g., Martin, M. et al., Int. J. Radiat. Oncol. Biol. Phys. 47:277—290 (2000)). Organ transplantation is complicated in many instances by chronic rejection and for some organs such as the kidney, it is the major forms of graft loss. In human patients, chronic rejection of lung and kidney transplants is associated with increased expression of TGF-β within the tissue (e.g., El-Gamel, A. et al., Eur. J. Cardiothorac. Surg. 13: 424—430 (1998); Shihab, F. S. et al., J. Am. Soc. Nephrol. 6:286—294 (1995)).
TGF-β is implicated in peritoneal adhesions (e.g., Saed, G. M. et al., Wound Repair Regeneration 7: 504 — 510 (1999)). The peritoneal and sub-dermal fibrotic adhesions could be prevented by inhibitors of ALK5 and/or ALK4.
The tumor cells and the stromal cells within the tumors in late stages of various cancers generally overexpress TGF-β. This leads to stimulation of angiogenesis and cell motility, suppression of the immune system, and increased interaction of tumor cells with the extracellular matrix (e.g., Hojo, M. et al., Nature 397: 530 — 534 (1999)). Consequently, the tumor cells become more invasive and metastasize to distant organs (e.g., Maehara, Y. et al., J. Clin. Oncol. 17: 607 — 614 (1999); Picon, A. et al., Cancer Epidemiol. Biomarkers Prev. 7:497—504 (1998)).
Plasminogen activator inhibitor- 1 (PAI-1) is the major physiological inhibitor of both tissue-type plasminogen activator and urokinase-type plasminogen activator. Elevated levels of PAI-1 are associated with thrombosis and vascular disease, suggesting that high plasma PAI-1 may promote a hypercoagulable state by disrupting the natural balance between fibrinolysis and coagulation (e.g., Naughan, D. E., J. Invest. Med. 46: 370—376 (1998)). It is known that TGF-D stimulates the expression of PAI-1 (e.g., Dennler, S. et al., EMBO J 17: 3091—3100 (1998)). Accordingly, inhibition of the production of PAI-1 with an inhibitor of the TGF-D signaling pathway could produce a novel fibrinolytic therapy.
Activin signaling and overexpression of activin is linked to pathological disorders that involve extracellular matrix accumulation and fibrosis (e.g., Matsuse, T. et al., Am. J. Respir. Cell Mol Biol. 13:17—24 (1995); Inoue, S. et al., Biochem. Biophys. Res. Comm. 205:441— 448 (1994); Matsuse, T. et al., Am. J. Pathol. 148:707—713 (1996); De Bleser et al,
Hepatology 26:905—912 (1997); Pawlowski, J. E., et al., J. Clin. Invest. 100:639—648 (1997);
Sugiyama, M. et al, Gastroenterology 114:550—558 (1998); Munz, B. et al., EMBO J.
18:5205 — 5215 (1999)), inflammatory responses (e.g., Rosendahl, A. et al., Am. J. Respir. Cell Mol. Biol. 25:60—68 (2001), cachexia or wasting (Matzuk, M. M. et al., Proc. Natl. Acd. Sci.
USA 91:8817—8821 (1994); Coerver, K. A. et al., Mol. Endocrinol. 10:534 — 543 (1996);
Cipriano, S. C. et al., Endocrinology 141:2319 — 2327 (2000)), diseases or pathological responses in the central nervous system (e.g., Logan, A. et al., Eur. J. Neurosci. 11:2367 — 2374
(1999); Logan, A. et al., Exp. Neurol. 159:504—510 (1999); Masliah, E. et al., Neurochem. Int. 39:393 — 400 (2001); De Groot, C. J. A. et al., J. Neuropathol. Exp. Neurol. 58:174—187
(1999); John, G R. et al., Nat. Med. 8:1115—1121 (2002)) and hypertension (e.g., Dahly, A. J. et al., Am. J. Physiol Regul Integr. Comp. Physiol. 283: R757— 767 (2002)). Studies have shown that TGF-β and activin can act synergistically to induce extracellular matrix production
(e.g., Sugiyama, M. et al., Gastroenterology 114;550 — 558 (1998)). Therefore, it becomes evident that inhibition of ALK5 and/or ALK4 phosphorylation of Smad2 and Smad3 by the preferred compounds of this invention could treat and prevent disorders involving these signaling pathways.
WO 00/61576 and US 2003/0149277 Al disclose triarylimidazole derivatives and their use as ALK5 inhibitors. WO 01/62756 Al discloses pyridinylimidazole derivatives and their use as ALK5 inhibitors. WO 02/055077 Al discloses use of imidazolyl cyclic acetal derivatives as
ALK5 inhibitors. And, also, WO 03/087304 A2 discloses tri-substituted heteroaryls and their use as ALK5 and/or ALK4 inhibitors.
Detailed Description of Preferred Embodiments
Surprisingly, it has now been discovered that a class of 2-pyridyl substituted imidazoles function as potent and selective inhibitors of ALK5 and/or ALK4 and therefore, have utility in the treatment and prevention of various disease states mediated by ALK5 and/or ALK4, such as glomerulonephritis, diabetic nephropathy, lupus nephritis, hypertension-induced nephropathy, renal interstitial fibrosis, renal fibrosis resulting from complications of drug exposure, HIN-associated nephropathy, transplant necropathy, liver fibrosis due to all etiologies, hepatic dysfunction attributable to infections, alcohol-induced hepatitis, disorders of the biliary tree, pulmonary fibrosis, acute lung injury, adult respiratory distress syndrome, idiopathic pulmonary fibrosis, chronic obstructive pulmonary disease, pulmonary disease due to infectious or toxic agents, post-infarction cardiac fibrosis, congestive heart failure, dilated cardiomyopathy, myocarditis, vascular stenosis, restenosis, atherosclerosis, ocular scarring, corneal scarring, proliferative vitreoretinopathy, excessive or hypertrophic scar or keloid formation in the dermis occurring during wound healing resulting from trauma or surgical wounds, peritoneal and sub- dermal adhesion, scleroderma, fibrosclerosis, progressive systemic sclerosis, dermatomyositis, polymyositis, arthritis, osteoporosis, ulcers, impaired neurological function, male erectile dysfunction, Alzheimer's disease, Raynaud's syndrome, fibrotic cancers, tumor metastasis growth, radiation-induced fibrosis, and thrombosis. In an embodiment of the present invention, there is provided a compound of formula
(I) or a pharmaceutically acceptable salt thereof:
Figure imgf000008_0001
wherein Ri is naphthyl, anthracenyl, or phenyl optionally substituted with substituents selected from halo, OH, -O-Cι----6alkyl, -S-Cι_6alkyl, Cι_6alkyl, Cι-_6haloalkyl, -O-(CH2)n-Ph, - S-(CH2)n-Ph, cyano, phenyl, and CO2R, wherein R is H or Ci—βalkyl, and n is 0, 1, 2, or 3; or Ri is phenyl or pyridyl fused with an aromatic or non-aromatic cyclic ring of 5 — 7 members wherein said cyclic ring optionally contains up to three heteroatoms, independently selected from Ν, O and S, and the fused phenyl or pyridyl may be further optionally substituted by halo, OH, -O-Cι_6alkyl, -S-Cι_6alkyl, Cι_6alkyl, Cι_6haloalkyl, cyano, phenyl or =O; R2 is H, OH, -O-Cι_6alkyl, -S-Cι_6alkyl, Cι_6alkyl, phenyl, Cι_6haloalkyl, ΝH2, NH(CH2)r--Ph. NH-Cι_6alkyl, halo, CN, NO2, CONHR or SO2NHR, wherein R is H or Cι_ 6alkyl, and n is 0, 1, 2, or 3; R3 is H, Ci-δalkyl, C3_7cycloalkyl, -(CH2)p-NO2, -(CH2)P-NR4R5, -(CH2)p-CHO, -
(CH2)p-CONHOH, -(CH2)P-CN, -(CH2)P-CO2H, -(CH2)p-CO2R4, -(CH^-CONR-tRs, -(CH2)P- tetrazole, -(CH2)p-COR4, -(CH2)q-(OR<.)2, -(CH2)p-OR4, -(CH2)P-CH=CH-CN, -(CH2)P-CH=CH-
CO2H, -(CH2)p-CH=CH-CO2R45 -(CH2)p-CH=CH-CONR4R5, -(CH2)p-NHCOR4, -(CH2)P-
NHCOsR,, -(CH2)p-CONHSO2R4,-(CH2)p-NHSO2R4 or -(CH2)p-CH=CH-tetrazole; R4 and R5 are independently H or Cι_6alkyl; R6 is Cι_6alkyl; p is 0, 1, 2, 3, or 4; q is 1, 2, 3, or 4; X is d—ioalkylene, C2loalkenylene, or C2— ι0alkynylene; One of Ai and A2 is N and the other is NR7; and R7 is H, OH, Cι_6alkyl, or C3-_7cycloalkyl.
As used herein, the double bond indicated by the dotted lines of formula (I), represent the possible tautomeric ring forms of the compounds falling within the scope of this invention, the double bond being to the unsubstituted nitrogen. Preferably, Ri is optionally substituted naphthyl or phenyl. More preferably, Ri is phenyl optionally substituted with substituents selected from halo, OH, -O-C-_-6alkyl, -S-Cι_ 6alkyl, and phenyl; or Ri is phenyl fused with an aromatic or non-aromatic cyclic ring of 5 — 7members wherein said cyclic ring optionally contains up to two heteroatoms, independently selected from N, O and S, and the fused phenyl or pyridyl may be further optionally substituted by halo, OH, -O-Cι_6alkyl, -S-C-_6alkyl, Cι_6alkyl, Cι_6haloalkyl or O. For example, Ri represents benzo[l,3]dioxolyl, 2,3-dihydrobenzo[l,4]dioxinyl, benzoxazolyl, benzothiazolyl, benzo[l,2,5]oxadiazolyl, benzo[l,2,5]thiadiazolyl, quinoxalinyl, dihydrobenzofuranyl, benzimidazolyl, C--_6benzimidazolyl, [l,2,4]triazolo[l,5-c-]ρyridyl, benzo[l,4]oxazinyl-3-one, benzoxazolyl-2-one or benzo[l,4]oxazinyl.
Preferably, R2 is other than H. When R2 is other than H, it is preferably positioned ortho to the nitrogen of the pyridyl ring. R2 is preferably C alkyl. Preferably, R3 is - CH2)p-CONHOH, -(CH2)P-CN, -(CH2)p-CO2H, -(CH2)p-CONR4R5, or -(CH2)p-tetrazole. Preferably, R4 and R5 are independently H or Cι_3alkyl. Preferably, p is 0, 1, or 2. Preferably, X is Cι_6alkylene. Preferably, one of Ai and A2 is N and the other is NR7, wherein R7 is H.
Specific compounds of the invention which may be mentioned include the following and pharmaceutically acceptable salts thereof:
4-((4-(benzo[ 1 ,3]dioxol-5-yl)-5-(6-methylρyridin-2-yl)- 1 (3)H-imidazol-2- yl)methyl)benzonitrile; 4-((4-(benzo[ 1 ,3]dioxol-5-yl)-5-(6-methylρyridin-2-yl)- 1 (3)H-imidazol-2- yl)methyl)benzamide; 3-((4-(benzo[l,3]dioxol-5-yl)-5-(6-methylpyridin-2-yl)-l(3)H-imidazol-2- yl)methyl)benzonitrile; 3-((4-(benzo[l,3]dioxol-5-yl)-5-(6-methylpyridin-2-yl)-l(3)H-imidazol-2- yl)methyl)benzamide; 4-(2-(4-(benzo[l,3]dioxol-5-yl)-5-(6-methylpyridin-2-yl)-l(3)H-imidazol-2- yl)ethyl)benzonitrile; 4-(2-(4-(benzo[l,3]dioxol-5-yl)-5-(6-methylpyridin-2-yl)-l(3)H-imidazol-2- yl)ethyl)benzamide; 4-(3-(4-(benzo[l,3]dioxol-5-yl)-5-(6-methylpyridin-2-yl)-l(3)H-imidazol-2- yl)propyl)benzonitrile; 4-(3-(4-(benzo[l,3]dioxol-5-yl)-5-(6-methylpyridin-2-yl)-l(3)H-imidazol-2- yl)propyl)benzamide; 4-((5-(6-methylpyridin-2-yl)-4-(quinoxalin-6-yl)-l(3)H-imidazol-2-yl)methyl)benzamide; 4-((5-(6-ethylpyridin-2-yl)-4-(quinoxalin-6-yl)-l(3)H-imidazol-2-yl)methyl)benzamide; 4-((5-(6-«-propylpyridin-2-yl)-4-(quinoxalin-6-yl)-l(3)H-imidazol-2-yl)methyl)benzamide; 4-((5-(6-isopropylpyridin-2-yl)-4-(quinoxalin-6-yl)- 1 (3)H-imidazol-2-yl)methyl)benzamide; 4-((5-(6-n-butylpyridin-2-yl)-4-(quinoxalin-6-yl)-l(3)H-imidazol-2-yl)methyl)benzamide; 3-((5-(6-methylpyridin-2-yl)-4-(quinoxalin-6-yl)-l(3)H-imidazol-2-yl)methyl)benzonitrile; 3-((5-(6-methylpyridin-2-yl)-4-(quinoxalin-6-yl)-l(3)H-imidazol-2-yl)methyl)benzamide; 3-((5-(6-ethylpyridin-2-yl)-4-(quinoxalin-6-yl)-l(3)H-imidazol-2-yl)methyl)benzamide; 3 -((5 -(6-«-propylpyridin-2-yl)-4-(quinoxalin-6-yl)- 1 (3)H-imidazol-2-yl)methyl)benzamide; 3-((5-(6-isopropylpyridin-2-yl)-4-(quinoxalin-6-yl)- 1 (3)H-imidazol-2-yl)methyl)benzamide; 3-((5-(6-7z-butylpyridin-2-yl)-4-(quinoxalin-6-yl)-l(3)H-imidazol-2-yl)methyl)benzamide; 4-(2-(5-(6-methylpyridin-2-yl)-4-(quinoxalin-6-yl)-l(3)H-imidazol-2-yl)ethyl)benzamide; 4-(2-(5-(6-ethylpyridin-2-yl)-4-(quinoxalin-6-yl)-l(3)H-imidazol-2-yl)ethyl)benzamide; 4-(2-(5-(6-Η-propylpyridin-2-yl)-4-(quinoxalin-6-yl)-l(3)H-imidazol-2-yl)ethyl)benzamide; 4-(2-(5-(6-isopropylpyridin-2-yl)-4-(quinoxalin-6-yl)- 1 (3)H-imidazol-2-yl)ethyl)benzamide; 4-(2-(5-(6-«-butylpyridin-2-yl)-4-(quinoxalin-6-yl)-l(3)H-imidazol-2-yl)ethyl)benzamide; 3-(2-(5-(6-methylpyridin-2-yl)-4-(quinoxalin-6-yl)-l(3)H-imidazol-2-yl)ethyl)benzamide; 3-(2-(5-(6-ethylpyridin-2-yl)-4-(quinoxalin-6-yl)-l(3)H-imidazol-2-yl)ethyl)benzamide; 3-(2-(5-(6-«-propylpyridin-2-yl)-4-(quinoxalin-6-yl)-l(3)H-imidazol-2-yl)ethyl)benzamide; 3-(2-(5-(6-isopropylpyridin-2-yl)-4-(quinoxalin-6-yl)-l(3)H-imidazol-2-yl)ethyl)benzamide; 3-(2-(5-(6-«-butylpyridin-2-yl)-4-(quinoxalin-6-yl)-l(3)H-imidazol-2-yl)ethyl)benzamide.
The compounds of the present invention typically are small organic molecules (non- peptide small molecules), generally less than about 1,000 daltons in size. Preferred non-peptide small molecules have molecular weights of less than about 750 daltons, more preferably less than about 500 daltons, and even more preferably less than about 300 daltons.
Compounds of formula (I) may also be supplied in the form of a "prodrug" which is designed to release compound of formula (I) when administered to a subject. Prodrug formed designs are well known in the art, and depend on the substituents contained in compound of formula (I). For example, a substituent containing hydroxyl could be. coupled to a carrier which renders the compound biologically inactive until it is removed by endogenous enzymes or, for example, by enzymes targeted to a particular receptor or location in the subject. A compound of formula (I) that is acidic in nature (e.g., having a carboxyl or phenolic hydroxyl group) can form a pharmaceutically acceptable salt such as a sodium, potassium, calcium, or gold salt. Also within the scope of the invention are salts formed with pharmaceutically acceptable amines such as ammonia, alkyl amines, hydroxyalkylamines, and N-methylglycamine. A compound of formula (I) can be treated with an acid to form acid addition salts. Examples of such acids include hydrochloric acid, hydrobromic acid, hydroiodic acid, sulfuric acid, methanesulfonic acid, phosphoric acid, -bromophenylsulfonic acid, carbonic acid, succinic acid, citric acid, benzoic acid, oxalic acid, malonic acid, salicylic acid, malic acid, fumaric acid, ascorbic acid, maleic acid, acetic acid, and other mineral and organic acids well known to those skilled in the art. The acid addition salts can be prepared by treating a compound of formula (I) in its free base form with a sufficient amount of an acid (e.g., hydrochloric acid) to produce an acid addition salt (e.g., a hydrochloride salt). The acid addition salt can be converted back to its free base form by treating the salt with a suitable dilute aqueous basic solution (e.g., sodium hydroxide, sodium bicarbonate, potassium carbonate, or ammonia).
Some of the compounds of this invention may be crystallized or recrystallized from solvents such as aqueous and organic solvents, h such cases solvates may be formed. This invention includes within its scope stoichiometric solvates including hydrates as well as compounds containing variable amounts of water that may be produced by processes such as lyophilization. Compounds of formula (I) may contain one or more asymmetric centers and thus can exist as enantiomers or diastereomers. It is to be understood that the invention includes both mixtures and separate individual isomers of compounds of the formula (I). Furthermore, certain compounds of the formula (I) which contain alkenyl groups may exist as cis- or trans-isomers. In each instance, the invention includes both mixtures and separate individual isomers.
Compounds of formula (I) may also exist in tautomeric forms and the invention includes both mixtures and separate individual tautomers thereof.
Also included in the invention are radiolabelled derivatives of compounds of formula (I) which are suitable for biological studies. As used herein, the term "alkyl" group refers to a saturated aliphatic hydrocarbon group containing 1 — 10 (e.g., 1 — 6 or 1 — 4) carbon atoms. An alkyl group can be straight or branched. Examples of an alkyl group include, but are not limited to, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, «-heptyl, and 2-ethylhexyl. An alkyl group can be optionally substituted with one or more substituents such as alkoxy, cycloalkoxy, amino, nitro, carboxy, cyano, halo, hydroxy, sulfo, or mercapto.
As used herein, the term "alkylene" group refers to a saturated aliphatic hydrocarbon group containing 1 — 10 (e.g., 1 — 6 or 1 — 4) carbon atoms. An alkylene group can be straight or branched. Examples of an alkylene group include, but are not limited to, methylene, ethylene, propylene, isopropylene, butylene, isobutylene, sec-butylene, tert-butylene, R-pentylene, n- heptylene, and 2-ethylhexylene. An alkylene group can be optionally substituted with one or more substituents such as alkoxy, cycloalkoxy, amino, nitro, carboxy, cyano, halo, hydroxy, sulfo, or mercapto.
As used herein, the term "alkenylene" group refers to an aliphatic carbon group that contains 2 — 10 (e.g., 2 — 6 or 2 — 4) carbon atoms and at least one double bond. Like an alkylene group, an alkenylene group can be straight or branched. Examples of an alkenylene group include, but are not limited to, allylene, isoprenylene, 2-butenylene, and 2-hexenylene. An alkenylene group can be optionally substituted with one or more substituents such as alkoxy, cycloalkyloxy, heterocycloalkyloxy, aryloxy, heteroaryloxy, aralkyloxy, heteroarylalkoxy, amino, nitro, carboxy, cyano, halo, hydroxy, sulfo, mercapto, alkylsulfanyl, alkylsulfinyl, alkylsulfonyl, aminocarbonyl, alkylcarbonylamino, cycloalkylcarbonylamino, cycloalkylalkylcarbonylamino, arylcarbonylamino, aralkylcarbonylamino, heterocycloalkylcarbonylamino, heterocycloalkylalkylcarbonylamino, heteroarylcarbonylamino, heteroaralkylcarbonylamino, urea, thiourea, sulfamoyl, sulfamide, alkoxycarbonyl, or alkylcarbonyloxy. As used herein, the term "alkynylene" group refers to an aliphatic carbon group that contains 2 — 10 (e.g., 2 — 6 or 2 — 4) carbon atoms and has at least one triple bond. An alkynylene group can be straight or branched. Examples of an alkynylene group include, but are not limited to, propargylene and butynylene. An alkynylene group can be optionally substituted with one or more substituents such as alkoxy, cycloalkyloxy, heterocycloalkyloxy, aryloxy, heteroaryloxy, aralkyloxy, heteroarylalkoxy, amino, nitro, carboxy, cyano, halo, hydroxy, sulfo, mercapto, alkylsulfanyl, alkylsulfmyl, alkylsulfonyl, aminocarbonyl, alkylcarbonylamino, cycloalkylcarbonylamino, cycloalkylalkylcarbonylamino, arylcarbonylamino, aralkylcarbonylamino, heterocycloalkylcarbonylamino, heterocycloalkylalkylcarbonylamino, heteroarylcarbonylamino, heteroaralkylcarbonylamino, urea, thiourea, sulfamoyl, sulfamide, alkoxycarbonyl, or alkylcarbonyloxy.
As used herein, the term "cycloalkyl" group refers to an aliphatic carbocyclic ring of
3—10 (e.g., 4 — 8) carbon atoms. Examples of cycloalkyl groups include cyclopropyl, cyclopentyl, cyclohexyl, cycloheptyl, adamantly, norbornyl, cubyl, octahydroindenyl, decahydronaphthyl; bicyclo[3.2.1]octyl, bicyclo[2.2.2]octyl, bicyclo[3.3.1]nonyl, and bicyclo[3.2.3]nonyl.
As used herein, the term "alkoxy" group refers to an alkyl-O-group where "alkyl" has been defined previously.
As used herein, the term "haloalkyl" group refers to an alkyl group containing one or more halogen atoms. Examples of haloalkyl groups include fluoromethyl, chloromethyl, bromomethyl, and trifluoromethyl.
As used herein, the term "halogen" or "halo" group refers to fluorine, chlorine, bromine or iodine.
As used herein, the term "ALK5 and/or ALK4 inhibitor" refers to a compound, other than inhibitory Smads, e.g. Smad6 and Smad7, which selectively inhibits the ALK5 and/or ALK4 receptors preferentially over p38 or type II receptors.
As used herein, the term "ALK5- and/or ALK4-mediated disease state" refers to any disease state which is mediated (or modulated) by ALK5 and/or ALK4, for example, a disease which is modulated by the inhibition of the phosphorylation of Smad2 and Smad3 in the TGF-β and/or activin signaling pathways.
As used herein, the term "ulcers" is used to include, but not to be limited to, diabetic ulcers, chronic ulcers, gastric ulcers, and duodenal ulcers.
Compounds of formula (I) may be prepared by a number of known methods from commercially available or known starting materials. If the starting materials are unavailable from a commercial source, they can be prepared by procedures known in the art.
Scheme 1
Figure imgf000015_0001
In one method, compounds of formula (I) wherein Ai is N and A2 is NH, or A] is NH and A2 is N are prepared according to Scheme 1. Specifically, optionally substituted 2- methylpyridine (II) is deprotonated by a base such as LDA or LiHMDS before reacting with
RiCOORs (III) wherein R8 is d-^alkyl, R-COC1 (IN), or Rj -substituted carboxylic acid methoxy-methyl-amide (V) to form a ketone (VI). The methoxy-methyl-amide (V) can be prepared by reacting a corresponding acid chloride (IN) with N,O-dimethylhydroxylamine hydrochloride. The ketone (NI) may be oxidized to a diketone (Nil) with HBr in DMSO. This diketone (Nil) can then be condensed with an appropriately substituted aldehyde (NIII) or protected aldehyde derivative in the presence of ammonium acetate to yield a compound of formula (I). R_, R2, R3, and X have been defined as above. The aldehyde (NIII) can be prepared according to the methods outlined in WO 02/096875 Al and Liquid Crystals 10:273—287 (1991). Alternatively, the ketone (NI) can be treated with sodium nitrite in HC1 or acetic acid to afford an α-keto-oxime (IX), which can be then condensed with an appropriately substituted aldehyde (NIII) or protected aldehyde derivative in the presence of ammonium acetate to give the N-hydroxyimidazoles. Treatment of this with triethylphophite affords a compound of formula (I).
In another method, when R3 in compounds of formula ( I ) is -(CH2)p-CΝ or -(CH2)P- CH=CH-CN, it can be further functionahzed to form a compound of formula ( I ) as depicted in Scheme 2. Ri, R2, X and P have been defined as above and R4 and R5 are independently H or Cι_6alkyl.
Scheme 2
Figure imgf000017_0001
The resulting compounds of this invention represented by the formula (I) — (IX) can be separated and purified by appropriate conventional methods such as column chromatography and recrystallization. Compounds of the invention may be administered by any suitable route, for example by oral, buccal, sub-lingual, rectal, vaginal, nasal, topical or parenteral (including intravenous, intramuscular, subcutaneous and intracoronary) administration.
The topical formulations of the present invention may be presented as, for instance, ointments, creams or lotions, eye ointments and eye or ear drops, impregnated dressings and aerosols, and may contain appropriate conventional additives such as preservatives, solvents to assist drug penetration and emollients in ointments and creams.
The formulations may also contain compatible conventional carriers, such as cream or ointment bases and ethanol or oleyl alcohol for lotions. Such carriers may be present as from about 1% up to about 98% of the formulation. More usually, they will form up to about 80% of the formulation.
For administration to man in the curative or prophylactic treatment of the disorders identified above, oral, buccal or sub-lingual dosages of a compound of formula (I) will generally be in the range of from 50 — 5000 mg daily for an average adult patient (70 kg). Thus for a typical adult patient, individual tablets or capsules contain from 25 — 500 mg of active compound, in a suitable pharmaceutically acceptable vehicle or carrier, for administration in single or multiple doses, once or several times per day. Dosages for parenteral administration will typically be within the range of from 25 — 250 mg per single dose as required. In practice the physician will determine the actual dosing regimen which will be most suitable for an individual patient and it will vary with the age, weight and response of the particular patient. The above dosages are exemplary of the average case but there can be individual instances in which higher or lower dosage ranges may be merited, and such are within the scope of this invention.
For human use, a compound of formula (I) can be administered alone, but will generally be administered in admixture with a pharmaceutical carrier selected with regard to the intended route of administration and standard pharmaceutical practice. For example, the compound may be administered orally, buccally or sublingually, in the form of tablets containing excipients such as starch or lactose, or in capsules or ovules either alone or in admixture with excipients, or in the form of elixirs or suspensions containing flavoring or coloring agents. Such liquid preparations may be prepared with pharmaceutically acceptable additives such as suspending agent (e.g. methylcellulose, a semi-synthetic glyceride such as witepsol or mixtures of glycerides such as a mixture of apricot kernel oil and PEG-6 esters or mixtures of PEG-8 and caprylic/capric glycerides). A compound may also be injected parenterally, for example intravenously, intramuscularly, subcutaneously or intracoronarily. For parenteral administration, the compound is best used in the form of a sterile aqueous solution which may contain other substances, for example, salts, or monosaccharides such as mannitol or glucose, to make the solution isotonic with blood.
Thus, the invention provides in a further aspect a pharmaceutical composition comprising a compound of formula (I), or a pharmaceutically acceptable salt or solvate thereof, together with a pharmaceutically acceptable diluent or carrier therefor.
The invention also provides a compound of formula (I), or a pharmaceutically acceptable salt or solvate thereof, or a pharmaceutical composition containing either entity, for use in therapy.
The invention further provides the use of a compound of formula (I), or a pharmaceutically acceptable salt or solvate thereof, or a pharmaceutical composition containing either entity, for the manufacture of a medicament for the treatment of a disease, mediated by the ALK5 and/or ALK4 receptors in mammals.
ALK5- and/or ALK4- mediated disease states include, but are not limited to, glomerulonephritis, diabetic nephropathy, lupus nephritis, hypertension-induced nephropathy, renal interstitial fibrosis, renal fibrosis resulting from complications of drug exposure, HIN- associated nephropathy, transplant necropathy, liver fibrosis due to all etiologies, hepatic dysfunction attributable to infections, alcohol-induced hepatitis, disorders of the biliary tree, pulmonary fibrosis, acute lung injury, adult respiratory distress syndrome, idiopathic pulmonary fibrosis, chronic obstructive pulmonary disease, pulmonary fibrosis due to infectious or toxic agents, post-infarction cardiac fibrosis, congestive heart failure, dilated cardiomyopathy, myocarditis, vascular stenosis, restenosis, atherosclerosis, ocular scarring, corneal scarring, proliferative vitreoretinopathy, excessive or hypertrophic scar or keloid formation in the dermis occurring during wound healing resulting from trauma or surgical wounds, peritoneal and sub- dermal adhesion, scleroderma, fibrosclerosis, progressive systemic sclerosis, dermatomyositis, polymyositis, arthritis, osteoporosis, ulcers, impaired neurological function, male erectile dysfunction, Alzheimer's disease, Raynaud's syndrome, fϊbrotic cancers, tumor metastasis growth, radiation-induced fibrosis, and thrombosis.
The invention further provides a method of inhibiting the TGF-β and/or activin signaling pathways in mammals, for example, inhibiting the phosphorylation of Smad2 or Smad3 byALK5 and or ALK4.
The invention further provides a method of reducing the accumulation of excess extracellular matrix in mammals by inhibiting the TGF-β and/or activin signaling pathways, for example, inhibiting the phosphorylation of Smad2 or Smad3 by ALK5 and/or ALK4.
The invention further provides a method of inhibiting metastasis of tumor cells in mammals by inhibiting the TGF-β signaling pathway. The invention further provides a method of treating carcinomas mediated by an overexpression of TGF-β in mammals by inhibiting the TGF-β signaling pathway.
Brief Description of the Drawings
The aforementioned aspects and other features of the present invention will be explained in the following description, taken in conjunction with the accompanying drawings, wherein:
Figure 1 shows effect of Examples 6, 9, 10, 13, and 31 on TGF-βl -induced SBE-Luc reporter activity in HepG2 cells,
Figure 2 shows effect of Example 41 on TGF-βl -induced SBE-Luc reporter activity in HepG2 cells,
Figure 3 shows effect of Example 41 on TGF-βl -induced 3TP-Lux reporter activity in HepG2 cells,
Figure 4 shows effect of Example 41 on TGF-βl -induced PAI-1 promoter-Luc reporter activity in HepG2 cells, Figure 5 shows effect of Example 41 on TGF-βl -induced PAI-1 expression in HaCaT human keratinocytes,
Figure 6 shows effect of Example 41 on TGF-βl -induced Smad2 and Smad3 phosphorylation in human dermal fibroblasts,
Figures 7 A and 7B show effect of Example 41 on liver fibrosis in rats with bile duct ligation, Table 1 shows structures and 1H NMR and MS spectral data of Examples 1 61, and Figure 8 shows effect of Example 41 on the activities of protein kinases in vitro.
Examples The present invention is further illustrated in the following Examples, which should not be taken to limit the scope of the invention described in the claims. In the Examples, electrospray ionization mass spectra (ESI-MS) were obtained on a Q-Tof2 mass spectrometer (Micromass, Manchester, UK). Practice Example 1: Preparation of 3-((4-(benzo[l,3]dioxol-5-yl)-5-(6-methylpyridin-
2-yl)-l(3)H-imidazol-2-yl)methyl)benzonitrile (Example 5)
Figure imgf000021_0001
To a stirred solution of l-(benzo[l,3]dioxol-5-yl)-2-(6-methylpyridin-2-yl)ethane-l,2- dione (50 mg, 0.19 mmol) (prepared according to the method described in WO 01/62756 Al) in AcOΗ (3 mL) were added 3-(formylmethyl)benzonitrile (28 mg, 0.19 mmol) (prepared according to the method described in WO 02/096875 Al) and NΗ4OAc (86 mg, 1.11 mmol), and the mixture was heated at 120 °C for 3 h. The pH of the cooled reaction mixture was adjusted to pH 7 — 8 at 0°C with 28 % NH4OH, and the reaction mixture was extracted with CH2CI2 (10 mL). The CH2CI2 solution was washed with water (5 mL) and brine (5 mL), dried (anhydrous Na2SO4), filtered, and evaporated to dryness under reduced pressure. The residue was purified by MPLC on silica gel using a mixture of MeOH and CH2C12 (1:19 (v/v)) as eluent to give 26 mg (36 %) of 3-((4-benzo[l,3]dioxol-5-yl)-5-(6-methylpyridin-2-yl)-l(3)H-imidazol- 2-yl)methyl)benzonitrile as a solid. MS (ESI) mfz 395.13 (MR*). 1H NMR (400 MHz, CDC13) h 7.52 (s, 1 H), 7.46 (m, 1 H), 7.40 (t, 1 H), 7.33 (d, 1 H), 7.32 - 7.26 (m, 2 H), 7.09—7.04 (m, 2 H), 6.90 (d, 1 H), 6.82 (d, 1 H), 5.96 (s, 2 H), 4.10 (s, 2 H), 2.38 (s, 3 H). Practice Example 2: Preparation of 3-((4-(benzo[l,3]dioxol-5-yl)-5-(6-methylpyridin- 2-yl)-l(3)H-imidazol-2-yl)methyl)benzamide (Example 6)
Figure imgf000022_0001
To a stirred solution of 3-((4-(benzo[l,3]dioxol-5-yl)-5-(6-methylpyridin-2-yl)-l(3)H- imidazol-2-yl)methyl)benzonitrile (70 mg, 0.17 mmol) in EtOΗ (4 mL) at room temperature were added 30 % Η2O2 (0.59 mmol) and 6 N NaOH (0.04 mmol) solution. The mixture was warmed to 50 — 60 °C and stirred for 3 h, and to it, 1 N HC1 solution was added to adjust to pH 7 — 8 at 0°C . The ethanol solvent was evaporated off under reduced pressure, and the residue was dissolved in CH2C12 (30 mL). The CH2C12 solution was washed with water (15 mL) and brine (15 mL), dried (anhydrous Na2SO4), filtered, and evaporated to dryness under reduced pressure. The residue was purified by MPLC on silica gel using a mixture of MeOH and CH2C12 (1:9 (v/v)) as eluent to give 23 mg (33 %) of 3-((4-benzo[l,3]dioxol-5-yl)-5-(6-methylpyridin-2- yl)-l(3)H-imidazol-2-yl)methyl)benzamide as a solid. MS (ESI) m/z 413.11 (MΗ*). 1H NMR (400 MHz, CDC13) δ 7.59 (s, 1 H), 7.52 (d, 1 H), 7.37 (dd, 1 H), 7.24 (m, 2 H), 7.15 (t, 1 H), 7.01 (overlapped, 1 H), 7.00 (s, 1 H), 6.88 (d, 1 H), 6.75 (d, 1 H), 6.70 (br s, 1 H), 6.02 (br s, 1 H), 5.92 (s, 2 H), 4.00 (s, 2 H), 2.34 (s, 3 H).
Practice Example 3: Preparation of 4-((l(3)-hydroxy-5-(6-methylpyridin-2-yl)-4- (quinoxalin-6-yl)- 1 (3)H-imidazol-2-yl)methyl)benzonitrile (Example 30)
Figure imgf000023_0001
To a stirred solution of l-(6-methylpyridin-2-yl)-2-(quinoxalin-6-yl)ethane-l,2-dione 1-oxime (67 mg, 0.23 mmol) (prepared according to the method described in WO 01/62756 Al) in tert-butyl methyl ether (2.5 mL) were added 4-(formylmethyl)benzonitrile (101 mg, 0.69 mmol) (prepared according to the method described in WO 02/096875 Al) and NH4OAc (89 mg, 1.15 mmol) dissolved in MeOH (1.2 mL), and the mixture was stirred at room temperature overnight. The pH of the reaction mixture was adjusted to pH 7 — 8 at 0°C with saturated NaHCO3 solution. The reaction mixture was partitioned between CH2CI2 (40 mL) and water (40 mL). The aqueous layer was repeatedly extracted with CH2C12 (3 x 15 mL). The combined CH2CI2 solution was dried (anhydrous Na2SO4), filtered, and evaporated to dryness under reduced pressure. The residue was purified by MPLC on silica gel using a mixture of MeOH and CH2C12 (1:30, then 1:19 (v/v)) as eluent to give 38 mg (40 %) of 4-((l(3)-hydroxy-5-(6- methylpyridin-2-yl)-4-(quinoxalin-6-yl)-l(3)H-imidazol-2-yl)methyl)benzonitrile as a solid. MS (ESI) m/z 419.23 (MΗ^. 1H NMR (400 MHz, CDC13) δ 8.86 (m, 2 H), 8.35 (d, 1 H), 8.17 (d, 1 H), 8.06 (dd, 1 H), 7.62 (m, 2 H), 7.56 ( , 2 H), 7.52 (t, 1 H), 7.36 (d, 1 H), 7.06 (d, 1 H), 4.31 (s, 2 H), 2.61 (s, 3 H).
Practice Example 4: Preparation of 4-((5-(6-methylpyridin-2-yl)-4-(quinoxalin-6-yl)- l(3)H-imidazol-2-yl)methyl)benzamide (Example 31)
Figure imgf000024_0001
To a stirred solution of 4-((l(3)-hydroxy-5-(6-methylpyridin-2-yl)-4-(quinoxalin-6-yl)- l(3)H-imidazol-2-yl)methyl)benzonitrile (264 mg, 0.631 mmol) in a mixture of EtOΗ (16 mL) and DMSO (4 mL) at room temperature were added 30% Η2O2 (6.62 mmol) and 6 N NaOH (0.47 mmol) solution. The mixture was warmed to 50 — 60 °C and stirred overnight, and to it, 1 N HC1 solution was added to adjust to pH 7 — 8 at 0°C. The ethanol solvent was evaporated off under reduced pressure, and the residue was partitioned between CH2CI2 (30 mL) and H2O (50 mL). The aqueous layer was saturated with NaCI and extracted with CH2CI2 (3 x 30 mL). The combined CH2CI2 solution was washed with brine (30 mL), dried (anhydrous Na2SO4), filtered, and evaporated to dryness under reduced pressure. The residue was dissolved in anhydrous DMF (20 mL) and treated with triethyl phosphite (2.39 mmol). The mixture was heated at 110°C for 3 days, cooled to room temperature, and evaporated to dryness under reduced pressure. The reaction mixture was partitioned between CH2C12 (30 mL) and water (50 mL), and the aqueous layer was extracted with CH2C12 (2 x 30 mL), The combined CH2C12 solution was washed with saturated NaHCO3 solution (40 mL) and brine (50 mL), dried (anhydrous Na2SO4), filtered, and evaporated to dryness under reduced pressure. The residue was purified by MPLC on silica gel using a mixture of MeOH and CH2C12 (1:9, then 1:5 (v/v)) as eluent to give 96 mg (36 %) of 4-((5-(6-methylρyridin-2-yl)-4-(quinoxalin-6-yl)-l(3)H-imidazol-2- yl)methyl)benzamide as a solid. MS (ESI) m/z 421.14 (MΗ+). 1H NMR (400 MHz, CDC13) δ 12.01 (br s, 1 H), 8.83 (m, 2 H), 8.38 (s, 1 H), 8.15 (dd, 2 H), 7.55 (d, 2 H), 7.42 (dd, 1 H), 7.33 (d, 1 H), 7.21 (d, 2 H), 6.95 (d, 1 H), 6.62 (br s, 1 H), 5.83 (br s, 1 H), 4.13 (s, 2 H), 2.29 (s, 3 H).
Practice Example 5: Preparation of 4-(2-(5-(6-methylpyridin-2-yl)-4-(quinoxalin-6-yl)- l(3)H-imidazol-2-yl)ethyl)benzamide (Example 48)
Figure imgf000025_0001
To a stirred solution of 4-(2-(l,3-dioxolan-2-yl)ethyl)benzonitrile (1.50 g, 7.34 mmol)
(prepared according to the method described in Kelly, S. M., Liquid Crystals 10: 273 — 287 (1991)) in MeOH (50 mL) at room temperature were added 30% H2O2 (25.70 mmol) and 6 N
NaOH (7.34 mmol) solution. The mixture was warmed to 50 — 60 °C and stirred for 2 h, and to it,
1 N HC1 solution was added to adjust to pH 7 — 8 at 0°C . The MeOH solvent was evaporated off under reduced pressure, and the residue was extracted with CH2C12 (3 x 30 mL). The CH2C12 solution was washed with brine (30 mL), dried (anhydrous Na2SO4), filtered, and evaporated to dryness under reduced pressure. The residue was purified by MPLC on silica gel using a mixture of MeOH and CH2C12 (1:19, then 1:9 (v/v)) as eluent to give 1.58 g (97%) of 4-(2-(l,3- dioxolan-2-yl)ethyl)benzamide as a solid. 1H NMR (400 MHz, CDC13) δ 7.74 (dd, 2 H), 7.29 (d,
2 H), 6.01 (br s, 1 H), 5.71 (br s, 1 H), 4.89 (t, 1 H), 3.99 (m, 2 H), 3.88 (m, 2 H), 2.80 (m, 2 H), 1.99 (m, 2 H). To a stirred solution of 4-(2-(l,3-dioxolan-2-yl)ethyl)benzamide (0.50 g, 2.26 mmol) in
THF (22 mL) was added 1 N HC1 solution (20 mL) at room temperature. The mixture was heated at 80 — 90 °C for 1 h and cooled to room temperature. After saturation with NaCI, the reaction mixture was extracted repeatedly with CHC13 ( 5 x 20 mL). The combined CHC13 solution was dried (anhydrous Na2SO4), filtered, and evaporated under reduced pressure to give 0.40 g (98 %) of 4-(2-formylethyl)benzamide as a solid which was used to the next step without further purification. Η NMR (400 MHz, CDC13) δ 9.82 (t, 1 H), 7.74 (m, 2 H), 7.27 (m, 2 H), 6.14 (br s, 1 H), 6.03 (br s, 1 H), 3.01 (m, 2 H), 2. 81 (m, 2 H).
To a stirred solution of l-(6-methylpyridin-2-yl)-2-(quinoxalin-6-yl)ethane-l,2-dione (1.05 g, 3.79 mmol) (prepared according to the method described in WO 02/055077 Al) in a mixture of tert-butyl methyl ether (35 mL) and MeOH (25 L) were added 4-(2- formylethyl)benzamide (1.00 g, 5.69 mmol) and NH4OAc (1.46 g, 18.95 mmol), and the mixture was stirred at room temperature overnight. The pH of the reaction mixture was adjusted to pH 7 — 8 at 0°C with saturated NaHCO3 solution. After removal of solvent, the reaction mixture was extracted with CH2C12 (3 x 25 mL), and the CH2CI2 solution was dried (anhydrous Na2SO4), filtered and evaporated to dryness under reduced pressure. The residue was purified by MPLC on silica gel using a mixture of MeOH and CH2CI2 (1:19, then 1:9 (v/v)) as eluent to give 1.08 g (66 %) of 4-(2-(5-(6-methylpyridin-2-yl)-4-(quinoxalin-6-yl)-l(3)H-imidazol-2- yl)ethylbenzamide as a solid. MS (ESI) m/z 435.19. 1H NMR (400 MHz, CDC13) δ 8.82 (m, 2 H), 8.36 (s, 1 H), 8.09 (d, 2 H), 7.64 (d, 2 H), 7.43 (t, 1 H), 7.32 (d, 1 H), 7.17 (d, 2 H), 6.99 (d, 1 H), 6.58 (br s, 1 H), 6.09 (br s, 1 H), 3.06 (s, 4 H), 2.45 (s, 3 H).
Practice Example 6: Preparation of 4-(3-(4-(benzo[l,3]dioxol-5-yl)-5-(6- methylpyridin-2-yl)- 1 (3)H-imidazol-2-yl)propyl)benzonitrile (Example 13)
Figure imgf000026_0001
To a stirred solution of l-(benzo[l,3]dioxol-5-yl)-2-(6-methylpyridin-2-yl)ethane-l,2- dione (230 mg, 0.86 mmol) in AcOΗ (8 mL) were added 4-(3-formylpropyl)benzonitrile (156 mg, 0.90 mmol) (prepared according to the method described in Kelly, S. M., Liquid Crystals 10: 273—287 (1991)) and Η OAc (396 mg, 5.14 mmol), and the mixture was heated at 120 °C for 3 h. The pH of the cooled reaction mixture was adjusted to pH 7 — 8 at 0°C with 28% NH4OH, and the reaction mixture was extracted with CH2CI2 (3 x 30 mL). The combined CH2Cl2solution was dried (anhydrous Na2SO4), filtered, and evaporated to dryness under reduced pressure. The residue was purified by MPLC on silica gel using a mixture of MeOH and CH2CI2 (1:30, then 1:19 (v/v)) as eluent to give 130 mg (36 %) of 4-(3-(4- (benzo[l,3]dioxol-5-yl)-5-(6-methylpyridin-2-yl)-l(3)H-imidazol-2-yl)propyl)benzonitrile as a solid. MS (ESI) m/z 423.14. 1H NMR (400 MHz, CDC13) δ 10.63 (br s, 1 H), 7. 1 (d, 2 H), 7.42 (dd, 1 H), 7.30 (d, 1 H), 7.24 (d, 2 H), 7.10—7.04 (m, 2 H), 6.93 (d, 1 H), 6.82 (d, 1 H), 5.98 (s, 2 H), 2.74 (m. 4 H), 2.47 (s, 3 H), 2.07 (m, 2 H).
Practice Example 7: Preparation of 3-((5-(6-methylpyridin-2-yl)-4-(quinoxalin-6-yl)-l(3)H- imidazol-2-yl)methyl)benzonitrile (Example 40)
Figure imgf000027_0001
To a solution of l-(6-methylpyridin-2-yl)-2-(quinoxalin-6-yl)ethane-l,2-dione (40.0 g, 144.26 mmol) (prepared according to the method described in WO 02/055077 Al) in a mixture oϊtert- butyl methyl ether (350 mL) and MeOΗ (350 mL) were added 3-(formylmethyl)b'enzonitrile
(23.03 g, 158.69 mmol) (prepared according to the method described in WO 02/096875 Al ) in tert-butyl methyl ether (350 mL) andNΗ4OAc (111.00 g, 1.44 mol) in MeOH (350 mL), and the mixture was stirred at 45 °C for overnight. The pH of the reaction mixture was adjusted to pH 7-8 at 0 °C with saturated NaHCO3 solution. After removal of solvent, the reaction mixture was extracted with CH2CI2 (3 x 800 mL), and the CH2CI2 solution was dried (anhydrous MgSO4), filtered and evaporated to dryness under reduced pressure. The residue was purified by MPLC on silica gel using a mixture of MeOH and CH2CI2 (1:19, then 1:9 (v/v)) as eluent to give 28.98 g
(50%, crude compound) of 3-((5-(6-methylpyridin-2-yl)-4-(quinoxalin-6-yl)-l(3)H-imidazol-2- yl)methyl)benzonitrile as a solid. MS (ESI) m/z 403.14 (MΗ+). 1H NMR (400 MHz, CDC13) δ
8.84 (m, 2 H), 8.39 (s, 1 H), 8.15 (s, 2 H), 7.56 (s, 1 H), 7.46 (m, 3 H), 7.37 (d, 1 H), 7.32 (t, 1
H), 6.99 (d, 1 H), 4.17 (s, 2 H), 2.37 (s, 3 H).
Practice Example 8: Preparation of 3-((5-(6-methylpyridin-2-yl)-4-(quinoxalin-6-yl)-l(3)H- imidazol-2-yl)methyl)benzamide (Example 41)
Figure imgf000028_0001
To a stirred solution of 3-((5-(6-methylpyridin-2-yl)-4-(quinoxalin-6-yl)-l(3)H-imidazol-2- yl)methyl)benzonitrile (28.98 g, 72.58 mmol) in EtOΗ (250 mL) were added 30% Η2O2 (4.85 mL, 39.92 mmol) and 1 N NaOH (250 mL). The mixture was warmed to 40 °C for 30 min and then, to it, was added 30% H2O2 (4.85 mL, 39.92 mmol). After 30 min, to the reaction mixture was added 1 N HC1 at 0 °C to adjust pH 7-8. After removal of solvent, the residue was extracted repeatedly with CH2C12 (3 x 450 mL). The combined CH2C12 solution was dried (anhydrous MgSO4), filtered, and evaporated to dryness under reduced pressure. The residue was purified by MPLC on silica gel using a mixture of MeOH and CH2Cl2 (l:9, then 1:5 (v/v)) as eluent to give 18.49 g (61%) of 3-((5-(6-methylpyridin-2-yl)-4-(quinoxalin-6-yl)-l(3)H-imidazol-2- yl)methyl)benzamide as a solid. MS (ESI) m/z 421.23 (MΗ ). !Η NMR (400 MHz, CDC13) δ 8.79 (s, 2 H), 8.34 (s, 1 H), 8.06 (s, 2 H), 7.71 (s, 1 H), 7.55 (d, 1 H), 7.41 (t, 1 H), 7.34 (d, 1 H), 7.29 (d, 1 H), 7.19 (t, 1 H), 6.96 (d, 1 H), 6.83 (br s, 1 H), 6.30 (br s, 1 H), 4.13 (s, 2 H), 2.36 (s, 3 H).
Practice Example 9: Preparation of 3-((5-(6-methylpyridin-2-yl)-4-(quinoxalin-6-yl)-l(3)H- imidazol-2-yl)methyl)benzamide hydrochloride (Example 58)
Figure imgf000028_0002
3-((5-(6-Methylpyridin-2-yl)-4-(quinoxalin-6-yl)-l(3)H-imidazol-2-yl)methyl)benzamide (5.0 g, 11.89 mmol) was dissolved in dry CΗ2CI2 (40 mL), filtered through a glass filter, and washed with dry CH2C12 (20 mL). The CH2C12 solution was cooled to 0 °C and, to it, 1.0 M HCl in Et2O (18 mL, 18 mmol) was added. The reaction mixture was stirred at room temperature for 15 min, evaporated to dryness under reduced pressure, and dried overnight over P2O5 to obtain 5.25 g (97%) of 3-((5-(6-methylpyridin-2-yl)-4-(quinoxalin-6-yl)- 1 (3)H-imidazol-2- yl)methyl)benzamide hydrochloride as a pale yellow powder. 1H NMR (400 MHz, OMSO-dβ) δ 9.03 (s, 2 H), 8.48 (d, 1 H), 8.19 (d, 1 H), 8.11 (m, 1 H), 8.06 (d, 1 H), 8.04 (d, 1 H), 7.83 (m, 1 H), 7.80 (t, 1 H), 7.74 (m, 1 H), 7.48 (m, 2 H), 7.45 (br s, 1 H), 7.38 (d, 1 H), 4.54 (s, 2 H), 2.54 (s, 3 H).
Practice Example 10: Preparation of 3-((5-(6-methylpyridin-2-yl)-4-(quinoxalin-6-yl)-l(3)H- imidazol-2-yl)methyl)benzamide sulfate (Example 59)
Figure imgf000029_0001
To a stirred solution of 3-((5-(6-methylpyridin-2-yl)-4-(quinoxalin-6-yl)-l(3)H-imidazol-2- yl)methyl)benzamide (50 mg, 0.12 mmol) in dry CΗ2C12 (2 mL) was added 10 % H2SO4 in EtOH ( 80 μL, 0.14 mmol), and the mixture was stirred at room temperature for 15 min. The reaction mixture was added to MeOH (1 mL) and then poured into anhydrous Et2O (15 mL). The precipitated solid was colleted by filtration and dried overnight over P2O5 to obtain 54 mg (88%) of 3-((5-(6-methylpyridin-2-yl)-4-(quinoxalin-6-yl)-l(3)H-imidazol-2-yl)methyl)benzamide sulfate as a pale yellow powder.
1H NMR (400 MHz, DMSO- 6) δ 9.03 (s, 2 H), 8.41 (d, 1 H), 8.22 (d, 1 H), 8.01 - 7.88 (m, 3 H), 7.83 (m, 1 H), 7.79 (t, 1 H), 7.62 (d, 1 H), 7.49 (t, 1 H), 7.44 (br s, 1 H), 7.39 (d, 1 H), 7.35 (d, 1 H), 4.45 (s, 2 H), 2.58 (s, 3 H). The compounds listed in the following Table 1 were prepared in an analogous manner to those described in the Practice Examples 1 — 10 above. The mass spectroscopy data of these compounds are included in the Table 1.
Table 1
Figure imgf000030_0001
Figure imgf000031_0001
Figure imgf000032_0001
Figure imgf000033_0001
Figure imgf000034_0001
Figure imgf000035_0001
Figure imgf000036_0001
Figure imgf000037_0001
Figure imgf000038_0001
Figure imgf000039_0001
Figure imgf000040_0001
Figure imgf000041_0001
Figure imgf000042_0001
Biological Data
The biological activity of the compounds of the invention may be assessed using the following assays: Cell-Free Assay for Evalutating Inhibition of ALK5 Kinase Phosphorylation of Smad3 The His-tagged, constitutively active ALK5 (T204D) and Smad3 full protein were expressed in insect cells using the Invitrogen BacNBlue baculovirus expression system. Expressed proteins were purified with Qiagen Ni-NTA resin column. The purified smad3 protem 200 ng was mixed with 100 μL of 0.1 M sodium bicarbonate coating buffer and coated into Flash-Plates by pipetting. Plates were covered and incubated at 4°C for 16 hours. Then the plates were washed 3 times with 200 μL of coating buffer and allowed to block in 1% BSA in PBS at room temperature for 1 hour. The purified ALK5 protein 100 ng was mixed with 100 μL of reaction buffer containing 20 mM Tris-HCl (pH 7.4), 5 mM MgCl2, 1 mM CaCl2, 1 M DTT, 1 μM ATP and 2 μCi γ-32p-ATP, and 1 μL of each test compound of formula ( I ) prepared in 100% DMSO solution at different concentrations. The assay was then initiated with the addition of ALK5 reaction mixture into Smad3 -coated Flash-Plates, followed by incubation at 30°C for 3 hours. After incubation, the assay buffer was removed and washed 3 times with 200 μL of 10 mM sodium pyrophosphate solution. Then, the Flash-Plates were air-dried and counted on a Packard TopCount. Compounds of formula ( I ) typically exhibited IC50 values of less than 10 μM; some exhibited IC50 values of less than 1 μM; and some even exhibited IC50 values less than 50 nM.
Cell-Free Assay for Evaluating Inhibition of ALK4 Kinase Phosphorylation of
Smad3
Inhibition of the ALK4 kinase phosphorylation of Smad3 by test compounds of formula ( I ) can be determined in a similar manner to that described above for ALK5 inhibition except that a similarly His-tagged ALK4 is used in place of the His-tagged, constitutively active ALK5.
Compounds of formula ( I ) typically exhibited IC50 values of less than 10 μM; some exhibited IC50 values of less than 1 μM. Assay for Evaluating Cellular Inhibition of TGF-/3 Signaling
Biological activity of the compounds of formula ( I ) was determined by measuring their ability to inhibit TGF-/31 -induced Smad binding element-luciferase (SBE-Luc) reporter activity and PAI-1 -luciferase (p3TP-Lux) reporter activity in HepG2 cells. HepG2 cells were transiently transfected with either SBE-Luc reporter construct or p3TP-Lux reporter construct grown in DMEM medium containing 10% FBS, penicillin 100 U/mL, streptomycin 100 μg/mL, L-glutamine 2 mM, sodium pyruvate 1 mM, and non-essential amino acids. The transfected cells were then plated at a concentration of 2.5 x 10 cells/well in 96 well plates and starved for 3—6 hours in media with 0.5% FBS at 37 °C in a 5% CO2 incubator. The cells were then stimulated with 5 ng/mL TGF-/31 ligand in the starvation media containing 1% DMSO either in the presence or absence of a test compound of formula ( I ) and incubated at 37 °C in a 5% CO2 incubator for 24 hours. The media was washed out, and the luciferase activity in cell lysates was determined by using a luciferase assay system (Promega). Compounds of formula ( I ) typically exhibited IC50 values of less than 10 μM; some exhibited IC50 values of less than 1 μM; and some even exhibited IC50 values of less than 50 nM.

Claims

Claims
What is claimed is: A compound of the formula (I) or a pharmaceutically acceptable salt thereof:
Figure imgf000045_0001
wherein Ri is naphthyl, anthracenyl, or phenyl optionally substituted with substituents selected from the group consisting of halo, OH, -O-Cι-_6alkyl, -S-Cι-_ 6alkyl, Ci—βalkyl, Cι-_ ehaloalkyl, -O-(CH2)„-Ph, -S-(CH2)n-Ph, cyano, phenyl, and CO2R, wherein R is H or Ci-βal yl, and n is 0, 1, 2, or 3; or Ri is phenyl or pyridyl fused with an aromatic or non-aromatic cyclic ring of 5 — 7 members wherein said cyclic ring optionally contains up to three heteroatoms, independently selected from the group consisting of N, O and S, and the fused phenyl or pyridyl are further optionally substituted by halo, OH, -O-Cι_6alkyl, -S-C_-_6alkyl, Cι_6alkyl, C__ 6haloalkyl, cyano, phenyl or =O; R2 is H, OH, -O-Cι_6alkyl, -S-Cι_6alkyl, C-_6alkyl, phenyl, Cι-_6haloalkyl, NH2, NH(CH2)n-Ph, NH-Cι_6alkyl, halo, CN, NO2, CONHR or SO2NHR, wherein R is H or Cι_ 6alkyl, and n is 0, 1, 2, or 3; R3 is H, d-ealkyl, C3_7cycloalkyl, -(CH2)p-NO2, -(CH2)P-NR4R5, -(CH2)p-CHO, - (CH2)p-CONHOH, -(CH2)P-CN, -(CH2)P-CO2H, -(CH2)p-CO2R4, -(CH2)p-CONR4R5, -(CH2)p- tetrazole, -(CH2)p-COR4, -(CH2)q-(OR6)2, -(CH2)p-OR4, -(CH2)P-CH=CH-CN, -(CH2)P-CH=CH- CO2H, -(CH2)p-CH=CH-CO2R , -(CH2)P-CH=CH-CONR4R5, -(CH2)p-NHCOR4, -(CH2)P- NHCOzI^, -(CH2)p-CONHSO2R4, -(CH2)P-NHSO2R4 or -(CH2)p-CH=CH-tetrazole; R4 and R5 are independently H or Cι-_6alkyl; R6 is C--_6alkyl; p is 0, 1, 2, 3, or 4; q is 1, 2, 3, or 4; X is Ci— -oalkylene, C2loalkenylene or C2-_ιoalkynylene; one of Ai and A2 is N and the other is NR7; and R7 is H, OH, Ci-βalkyl, or C3_7cycloalkyl.
2. The compound according to claim 1, wherein R. is phenyl optionally substituted with substituents selected from the group consisting of halo, OH, -O-C-_6alkyl, -S-Cι_6alkyl, and phenyl; or R. is phenyl fused with an aromatic or non-aromatic cyclic ring of 5 — 7members wherein said cyclic ring optionally contains up to two heteroatoms, independently selected from the group consisting of N, O and S, and the fused phenyl or pyridyl are further optionally substituted by halo, OH, -O-C--_6alkyl, -S-Cι---6alkyl, Cι-_δalkyl, Cι----6- aloalkyl or =O.
R2 is other than H and is positioned ortho to the nitrogen of the pyridyl ring; R3 is -(CH2)P-CONHOH, -(CH2)P-CN, -(CH2)p-CO2H,
Figure imgf000046_0001
or -(CH2)p-tetrazole; I . and R5 are independently H or Cι_3alkyl; p is 0, 1, or 2; X is Cι_6alkylene; and one of Ai and A2 is N and the other is NR , wherein R7 is H.
3. The compound according to claim l^ selected from the group consisting of: 4-((4-(benzo[l,3]dioxol-5-yl)-5-(6-methylpyridin-2-yl)-l(3)H-imidazol-2- yl)methyl)benzonitrile; 4-((4-(benzo[ 1 ,3]dioxol-5-yl)-5-(6-methylpyridin-2-yl)- 1 (3)H-imidazol-2- yl)methyl)benzamide; 3-((4-(benzo[l,3]dioxol-5-yl)-5-(6-methylpyridin-2-yl)-l(3)H-imidazol-2- yl)methyl)benzonitrile; 3-((4-(benzo[l,3]dioxol-5-yl)-5-(6-methylρyridin-2-yl)-l(3)H-imidazol-2- yl)methyl)benzamide; 4-(2-(4-(benzo[l,3]dioxol-5-yl)-5-(6-methylpyridin-2-yl)-l(3)H-imidazol-2- yl)ethyl)benzonitrile; 4-(2-(4-(benzo[ 1 ,3]dioxol-5-yl)-5-(6-methylpyridin-2-yl)- 1 (3)H-imidazol-2- yl)ethyl)benzamide; 4-(3-(4-(benzo[l,3]dioxol-5-yl)-5-(6-methylpyridin-2-yl)-l(3)H-imidazol-2- yl)propyl)benzonitrile; 4-(3-(4-(benzo[ 1 ,3]dioxol-5-yl)-5-(6-methylpyridin-2-yl)- 1 (3)H-imidazol-2- yl)propyl)benzamide; 4-((5-(6-methylpyridin-2-yl)-4-(quinoxalin-6-yl)-l(3)H-imidazol-2-yl)methyl)benzamide; 4-((5-(6-ethylpyridin-2-yl)-4-(quinoxalin-6-yl)- 1 (3)H-imidazol-2-yl)methyl)benzamide; 4-((5-(6-n-propylpyridin-2-yl)-4-(quinoxalin-6-yl)-l(3)H-imidazol-2-yl)methyl)benzamide; 4-((5-(6-isopropylpyridin-2-yl)-4-(quinoxalin-6-yl)-l(3)H-imidazol-2-yl)methyl)benzamide; 4-((5-(6-π-butylpyridin-2-yl)-4-(quinoxalin-6-yl)-l(3)H-imidazol-2-yl)methyl)benzamide; 3-((5-(6-methylpyridin-2-yl)-4-(quinoxalin-6-yl)-l(3)H-imidazol-2-yl)methyl)benzonitrile; 3-((5-(6-methylpyridin-2-yl)-4-(quinoxalin-6-yl)- 1 (3)H-imidazol-2-yl)methyl)benzamide; 3-((5-(6-ethylpyridin-2-yl)-4-(quinoxalin-6-yl)-l(3)H-imidazol-2-yl)methyl)benzamide; 3-((5-(6-n-propylpyridin-2-yl)-4-(quinoxalin-6-yl)-l(3)H-imidazol-2-yl)methyl)benzamide; 3-((5-(6-isopropylpyridin-2-yl)-4-(quinoxalin-6-yl)-l(3)H-imidazol-2-yl)methyl)benzamide; 3-((5-(6-«-butylpyridin-2-yl)-4-(quinoxalin-6-yl)-l(3)H-imidazol-2-yl)methyl)benzamide; 4-(2-(5-(6-methylpyridin-2-yl)-4-(quinoxalin-6-yl)-l(3)H-imidazol-2-yl)ethyl)benzamide; 4-(2-(5-(6-ethylpyridin-2-yl)-4-(quinoxalin-6-yl)-l(3)H-imidazol-2-yl)ethyl)benzamide; 4-(2-(5-(6-n-propylpyridin-2-yl)-4-(quinoxalin-6-yl)-l(3)H-imidazol-2-yl)ethyl)benzamide; 4-(2-(5-(6-isopropylpyridin-2-yl)-4-(quinoxalin-6-yl)-l(3)H-imidazol-2-yl)ethyl)benzamide; 4-(2-(5-(6-«-butylpyridin-2-yl)-4-(quinoxalin-6-yl)-l(3)H-imidazol-2-yl)ethyl)benzamide; 3-(2-(5-(6-methylpyridin-2-yl)-4-(quinoxalin-6-yl)-l(3)H-imidazol-2-yl)ethyl)benzamide; 3 -(2-(5-(6-ethylpyridin-2-yl)-4-(quinoxalin-6-yl)- 1 (3)H-imidazol-2-yl)ethyl)benzamide; 3-(2-(5-(6-n-propylpyridin-2-yl)-4-(quinoxalin-6-yl)-l(3)H-imidazol-2-yl)ethyl)benzamide; 3-(2-(5-(6-isopropylpyridin-2-yl)-4-(quinoxalin-6-yl)-l(3)H-imidazol-2-yl)ethyl)benzamide; 3-(2-(5-(6-n-butylpyridin-2-yl)-4-(quinoxalin-6-yl)-l(3)H-imidazol-2-yl)ethyl)benzamide, and pharmaceutically acceptable salts thereof.
4. A pharmaceutical composition comprising a compound of the formula (I) as claimed in claim 1, or a pharmaceutically acceptable salt or solvate thereof, and a pharmaceutically acceptable diluent or carrier.
5. A method of inhibiting the TGF-β or activin signaling pathways or both in mammals, comprising administering to a mammal in need of such treatment, a therapeuticaUy effective amount of a compound according to claim 1, or a pharmaceutically acceptable salt or solvate thereof.
6. A method of reducing the accumulation of excess extracellular matrix in mammals, comprising administering to a mammal in need of such treatment, a therapeuticaUy effective amount of a compound according to claim 1, or a pharmaceutically acceptable salt or solvate thereof.
7. A method of inhibiting metastasis of tumor cells in mammals, comprising administering to a mammal in need of such treatment, a therapeuticaUy effective amount of a compound according to claim 1, or a pharmaceutically acceptable salt or solvate thereof.
8. A method of treating carcinomas mediated by an overexpression of TGF-β by inhibiting a TGF- β signaling pathway in mammals, comprising administering to a mammal in need of such treatment, a therapeuticaUy effective amount of a compound according to claim 1, or a pharmaceutically acceptable salt or solvate thereof.
9. A method for treating a disease selected from the group consisting of_glomerulonephritis, diabetic nephropathy, lupus nephritis, hypertension-induced nephropathy, renal interstitial fibrosis, renal fibrosis resulting from complications of drug exposure, HIN-associated nephropathy, transplant necropathy, liver fibrosis due to all etiologies, hepatic dysfunction attributable to infections, alcohol-induced hepatitis, disorders of the biliary tree, pulmonary fibrosis, acute lung injury, adult respiratory distress syndrome, idiopathic pulmonary fibrosis, chronic obstructive pulmonary disease, pulmonary fibrosis due to infectious or toxic agents, post-infarction cardiac fibrosis, congestive heart failure, dilated cardiomyopathy, myocarditis, vascular stenosis, restenosis, atherosclerosis, ocular scarring, corneal scarring, proliferative vitreoretinopathy, excessive or hypertrophic scar or keloid formation in the dermis occurring during wound healing resulting from trauma or surgical wounds, peritoneal and sub-dermal adhesion, scleroderma, fibrosclerosis, progressive systemic sclerosis, dermatomyositis, polymyositis, arthritis, osteoporosis, ulcers, impaired neurological function, male erectile dysfunction, Alzheimer's disease, Raynaud's syndrome, fibrotic cancers, tumor metastasis growth, radiation-induced fibrosis, any disease wherein fibrosis is a major component, and thrombosis, comprising administering to a mammal in need of such treatment, a therapeuticaUy 5 effective amount of a compound according to claim 1, or a pharmaceutically acceptable salt or solvate thereof.
10. The method of claim 8, wherein said mammal is human.
10 11. The method of claim 9, wherein said mammal is human.
12. The method of claim 10, wherein said mammal is human.
13. The method of claim 11, wherein said mammal is human. 15
14. The compound of claim 2, wherein R2 is CM alkyl.
15. A prodrug of the compound of claim 1, which releases the compound of claim 1 when administered to a mammal.
20 16. The prodrug of claim 15, which is the compound of claim 1 containing a hydroxyl group to which is bonded a carrier which renders the compound of claim 1 inactive until removed endogenously.
25 17. The compound of claim 1, which contains one or more chiral centers.
18. The compound of claim 17, which is an enantiomer.
19. The compound of claim 17, which is a diastereomer.
30 20. A method of inhibiting thrombosis in mammals, comprising administering to a mammal in need of such treatment, a therapeuticaUy effective amount of a compound according to claim 1, or a pharmaceutically acceptable salt or solvate thereof.
21. The method of claim 20, wherein said mammal is human.
PCT/KR2004/002891 2004-04-21 2004-11-09 2-pyridyl substituted imidazoles as alk5 and/or alk4 inhibitors WO2005103028A1 (en)

Priority Applications (10)

Application Number Priority Date Filing Date Title
US10/983,227 US7407958B2 (en) 2004-04-21 2004-11-08 2-pyridyl substituted imidazoles as ALK5 and/or ALK4 inhibitors
JP2007509388A JP4678540B2 (en) 2004-04-21 2004-11-09 2-Pyridyl-substituted imidazoles as inhibitors of ALK5 and / or ALK4
MXPA06012096A MXPA06012096A (en) 2004-04-21 2004-11-09 2-pyridyl substituted imidazoles as alk5 and/or alk4 inhibitors.
CN2004800433657A CN1972921B (en) 2004-04-21 2004-11-09 2-pyridyl substituted imidazoles as alk5 and/or alk4 inhibitors
BRPI0418765A BRPI0418765B8 (en) 2004-04-21 2004-11-09 compound of formula (I) and pharmaceutical composition
EP04800071A EP1620426A1 (en) 2004-04-21 2004-11-09 2-pyridyl substituted imidazoles as alk5 and/or alk4 inhibitors
CA2564442A CA2564442C (en) 2004-04-21 2004-11-09 2-pyridyl substituted imidazoles as alk5 and/or alk4 inhibitors
AU2004318777A AU2004318777B2 (en) 2004-04-21 2004-11-09 2-pyridyl substituted imidazoles as ALK5 and/or ALK4 inhibitors
NO20064771A NO20064771L (en) 2004-04-21 2006-10-19 2-Pyridyl substituted imidazoles as ALK5 and / or ALK4 inhibitors
IL178757A IL178757A (en) 2004-04-21 2006-10-19 2-pyridyl substituted imidazoles as alk5 and/or alk4 inhibitors

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR1020040027591A KR100749566B1 (en) 2004-04-21 2004-04-21 2-PYRIDYL SUBSTITUTED IMIDAZOLES AS ALK5 and/or ALK4 INHIBITORS
KR10-2004-0027591 2004-04-21

Publications (1)

Publication Number Publication Date
WO2005103028A1 true WO2005103028A1 (en) 2005-11-03

Family

ID=35196912

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2004/002891 WO2005103028A1 (en) 2004-04-21 2004-11-09 2-pyridyl substituted imidazoles as alk5 and/or alk4 inhibitors

Country Status (14)

Country Link
US (1) US7407958B2 (en)
EP (1) EP1620426A1 (en)
JP (1) JP4678540B2 (en)
KR (1) KR100749566B1 (en)
CN (1) CN1972921B (en)
AU (1) AU2004318777B2 (en)
BR (1) BRPI0418765B8 (en)
CA (1) CA2564442C (en)
IL (1) IL178757A (en)
MX (1) MXPA06012096A (en)
NO (1) NO20064771L (en)
RU (1) RU2348626C2 (en)
WO (1) WO2005103028A1 (en)
ZA (1) ZA200609626B (en)

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009115572A2 (en) * 2008-03-21 2009-09-24 Novartis Ag Novel heterocyclic compounds and uses therof
KR20110022662A (en) * 2008-06-12 2011-03-07 에스케이케미칼주식회사 2-pyridyl substituted imidazoles as alk5 and/or alk4 inhibitors
US8080568B1 (en) 2010-06-29 2011-12-20 Ewha University - Industry Collaboration Foundation 2-pyridyl substituted imidazoles as therapeutic ALK5 and/or ALK4 inhibitors
US8410146B2 (en) * 2004-04-21 2013-04-02 Sk Chemicals Co., Ltd. 2-pyridyl substituted imidazoles as ALK5 and/or ALK4 inhibitors
US8420685B2 (en) * 2004-04-21 2013-04-16 Sk Chemicals Co., Ltd. 2-pyridyl substituted imidazoles as ALK5 and/or ALK4 inhibitors
US8513222B2 (en) 2010-06-29 2013-08-20 EWHA University—Industry Collaboration Foundation Methods of treating fibrosis, cancer and vascular injuries
US8865732B2 (en) 2008-03-21 2014-10-21 Novartis Ag Heterocyclic compounds and uses thereof
US9242969B2 (en) 2013-03-14 2016-01-26 Novartis Ag Biaryl amide compounds as kinase inhibitors
US9573969B2 (en) 2014-09-12 2017-02-21 Novartis Ag Compounds and compositions as kinase inhibitors
USRE47141E1 (en) 2010-06-29 2018-11-27 EWHA University—Industry Collaboration Foundation Methods of treating fibrosis, cancer and vascular injuries
WO2021142086A1 (en) 2020-01-08 2021-07-15 Synthis Therapeutics, Inc. Alk5 inhibitor conjugates and uses thereof

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
PL3520815T3 (en) * 2005-02-08 2022-04-11 Genzyme Corporation Antibodies to tgfbeta
WO2009146301A1 (en) * 2008-05-27 2009-12-03 Yale University TARGETING TGF-β AS A THERAPY FOR ALZHEIMER'S DISEASE
EP2285380A4 (en) * 2008-05-30 2012-03-14 Summa Health Systems Llc Methods for using tgf-b receptor inhibitors or activin-like kinase (alk) 5 inhibitors a-83-01 and sb-431542 to treat eye disease and wound healing conditions
US9782452B2 (en) 2011-11-22 2017-10-10 Cornell University Methods for stimulating hematopoietic recovery by inhibiting TGFβ signaling
RU2475243C1 (en) * 2012-02-16 2013-02-20 Федеральное государственное бюджетное учреждение науки Новосибирский институт органической химии им. Н.Н. Ворожцова Сибирского отделения Российской академии наук (НИОХ СО РАН) Agent possessing antihypertensive activity
KR102434226B1 (en) * 2016-06-30 2022-08-19 한미약품 주식회사 Novel substituted pyrazole derivatives as a alk5 inhibitors and use thereof
CN110066277B (en) * 2018-01-24 2021-07-23 上海璎黎药业有限公司 Aromatic heterocyclic substituted olefin compound, preparation method, pharmaceutical composition and application thereof
CN112266378B (en) * 2020-11-09 2021-10-12 延边大学 Imidazole derivative containing indazole structure and preparation method and application thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5656644A (en) * 1994-07-20 1997-08-12 Smithkline Beecham Corporation Pyridyl imidazoles
WO2000061576A1 (en) * 1999-04-09 2000-10-19 Smithkline Beecham Corporation Triarylimidazoles
US20030166633A1 (en) * 2000-02-21 2003-09-04 Gaster Laramie Mary Pyridinylimidazoles

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB0007405D0 (en) * 2000-03-27 2000-05-17 Smithkline Beecham Corp Compounds
WO2002096875A1 (en) 2001-05-25 2002-12-05 Mochida Pharmaceutical Co., Ltd. 4-hydroxpiperidine derivative with analgetic activity
JP2003040888A (en) * 2001-07-30 2003-02-13 Sankyo Co Ltd Imidazole derivative
GB0218079D0 (en) * 2002-08-03 2002-09-11 Boatman Peter J A pipe and pipe assembly
EA200500378A1 (en) * 2002-09-18 2005-08-25 Пфайзер Продактс Инк. NEW IMIDAZOL COMPOUNDS AS A TRANSFORMING GROWTH FACTOR INHIBITORS (TGF) INHIBITORS
US7250434B2 (en) * 2003-12-22 2007-07-31 Janssen Pharmaceutica N.V. CCK-1 receptor modulators

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5656644A (en) * 1994-07-20 1997-08-12 Smithkline Beecham Corporation Pyridyl imidazoles
WO2000061576A1 (en) * 1999-04-09 2000-10-19 Smithkline Beecham Corporation Triarylimidazoles
US20030166633A1 (en) * 2000-02-21 2003-09-04 Gaster Laramie Mary Pyridinylimidazoles

Cited By (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8410146B2 (en) * 2004-04-21 2013-04-02 Sk Chemicals Co., Ltd. 2-pyridyl substituted imidazoles as ALK5 and/or ALK4 inhibitors
US8420685B2 (en) * 2004-04-21 2013-04-16 Sk Chemicals Co., Ltd. 2-pyridyl substituted imidazoles as ALK5 and/or ALK4 inhibitors
US8865732B2 (en) 2008-03-21 2014-10-21 Novartis Ag Heterocyclic compounds and uses thereof
WO2009115572A3 (en) * 2008-03-21 2009-12-03 Novartis Ag Novel heterocyclic compounds and uses therof
WO2009115572A2 (en) * 2008-03-21 2009-09-24 Novartis Ag Novel heterocyclic compounds and uses therof
US8129394B2 (en) 2008-03-21 2012-03-06 Novartis Ag Heteroaryl-substituted imidazole compounds and uses thereof
AU2009227013B2 (en) * 2008-03-21 2013-01-10 Novartis Ag Novel heterocyclic compounds and uses therof
KR20110022662A (en) * 2008-06-12 2011-03-07 에스케이케미칼주식회사 2-pyridyl substituted imidazoles as alk5 and/or alk4 inhibitors
EP2303860A2 (en) * 2008-06-12 2011-04-06 Sk Chemicals Co., Ltd. 2-pyridyl substituted imidazoles as alk4 and/or alk4 inhibitors
EP2303860A4 (en) * 2008-06-12 2011-07-06 Sk Chemicals Co Ltd 2-pyridyl substituted imidazoles as alk4 and/or alk4 inhibitors
JP2011522877A (en) * 2008-06-12 2011-08-04 エスケイ ケミカルズ カンパニー リミテッド Imidazoles substituted with 2-pyridyl as ALK5 and / or ALK4 inhibitors
KR101654859B1 (en) * 2008-06-12 2016-09-07 에스케이케미칼주식회사 2-PYRIDYL SUBSTITUTED IMIDAZOLES AS ALK5 and/or ALK4 INHIBITORS
US8080568B1 (en) 2010-06-29 2011-12-20 Ewha University - Industry Collaboration Foundation 2-pyridyl substituted imidazoles as therapeutic ALK5 and/or ALK4 inhibitors
USRE47122E1 (en) 2010-06-29 2018-11-13 EWHA University—Industry Collaboration Foundation 2-pyridyl substituted imidazoles as therapeutic ALK5 and/or ALK4 inhibitors
AU2011272149B2 (en) * 2010-06-29 2014-05-22 Ewha University-Industry Collaboration Foundation 2-pyridyl substituted imidazoles as therapeutic ALK5 and/or ALK4 inhibitors
US8513222B2 (en) 2010-06-29 2013-08-20 EWHA University—Industry Collaboration Foundation Methods of treating fibrosis, cancer and vascular injuries
EP2947081A1 (en) 2010-06-29 2015-11-25 Ewha University-Industry Collaboration Foundation 2-Pyridyl substituted imidazoles as therapeutic Alk5 and/or Alk4 inhibitors
USRE47141E1 (en) 2010-06-29 2018-11-27 EWHA University—Industry Collaboration Foundation Methods of treating fibrosis, cancer and vascular injuries
EP2588479A2 (en) * 2010-06-29 2013-05-08 Ewha University-Industry Collaboration Foundation 2-pyridyl substituted imidazoles as therapeutic alk5 and/or alk4 inhibitors
EP2588479A4 (en) * 2010-06-29 2013-11-20 Univ Ewha Ind Collaboration 2-pyridyl substituted imidazoles as therapeutic alk5 and/or alk4 inhibitors
US9694016B2 (en) 2013-03-14 2017-07-04 Novartis Ag Biaryl amide compounds as kinase inhibitors
US9242969B2 (en) 2013-03-14 2016-01-26 Novartis Ag Biaryl amide compounds as kinase inhibitors
US10245267B2 (en) 2013-03-14 2019-04-02 Novartis Ag Biaryl amide compounds as kinase inhibitors
US10709712B2 (en) 2013-03-14 2020-07-14 Novartis Ag Biaryl amide compounds as kinase inhibitors
US9809610B2 (en) 2014-09-12 2017-11-07 Novartis Ag Compounds and compositions as kinase inhibitors
US9573969B2 (en) 2014-09-12 2017-02-21 Novartis Ag Compounds and compositions as kinase inhibitors
WO2021142086A1 (en) 2020-01-08 2021-07-15 Synthis Therapeutics, Inc. Alk5 inhibitor conjugates and uses thereof

Also Published As

Publication number Publication date
JP4678540B2 (en) 2011-04-27
AU2004318777B2 (en) 2008-10-16
KR100749566B1 (en) 2007-08-16
RU2006140989A (en) 2008-05-27
US20050261299A1 (en) 2005-11-24
ZA200609626B (en) 2008-08-27
AU2004318777A1 (en) 2005-11-03
CA2564442A1 (en) 2005-11-03
BRPI0418765B1 (en) 2019-06-04
CN1972921A (en) 2007-05-30
NO20064771L (en) 2007-01-19
KR20050102752A (en) 2005-10-27
CA2564442C (en) 2011-06-14
IL178757A0 (en) 2007-02-11
CN1972921B (en) 2012-01-04
EP1620426A1 (en) 2006-02-01
RU2348626C2 (en) 2009-03-10
BRPI0418765B8 (en) 2021-05-25
IL178757A (en) 2011-08-31
US7407958B2 (en) 2008-08-05
MXPA06012096A (en) 2007-11-20
BRPI0418765A (en) 2007-10-09
JP2007533734A (en) 2007-11-22

Similar Documents

Publication Publication Date Title
IL178757A (en) 2-pyridyl substituted imidazoles as alk5 and/or alk4 inhibitors
EP2731949B1 (en) 2-pyridyl substituted imidazoles as alk5 and/or alk4 inhibitors
US8410146B2 (en) 2-pyridyl substituted imidazoles as ALK5 and/or ALK4 inhibitors
US20130245066A1 (en) 2-Pyridyl substituted imidazoles as ALK5 and/or ALK4 inhibitors
US8420685B2 (en) 2-pyridyl substituted imidazoles as ALK5 and/or ALK4 inhibitors

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 2004800071

Country of ref document: EP

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 KP KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL 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 LU 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
WWP Wipo information: published in national office

Ref document number: 2004800071

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2004318777

Country of ref document: AU

Ref document number: 178757

Country of ref document: IL

Ref document number: PA/a/2006/012096

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 2007509388

Country of ref document: JP

Ref document number: 2564442

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

WWW Wipo information: withdrawn in national office

Ref document number: DE

ENP Entry into the national phase

Ref document number: 2004318777

Country of ref document: AU

Date of ref document: 20041109

Kind code of ref document: A

WWP Wipo information: published in national office

Ref document number: 2004318777

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 6802/DELNP/2006

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 200609626

Country of ref document: ZA

WWE Wipo information: entry into national phase

Ref document number: 2006140989

Country of ref document: RU

WWE Wipo information: entry into national phase

Ref document number: 200480043365.7

Country of ref document: CN

ENP Entry into the national phase

Ref document number: PI0418765

Country of ref document: BR