WO2007138072A2 - Triazolopyrazine compounds useful for the treatment of degenerative & inflammatory diseases - Google Patents

Triazolopyrazine compounds useful for the treatment of degenerative & inflammatory diseases Download PDF

Info

Publication number
WO2007138072A2
WO2007138072A2 PCT/EP2007/055246 EP2007055246W WO2007138072A2 WO 2007138072 A2 WO2007138072 A2 WO 2007138072A2 EP 2007055246 W EP2007055246 W EP 2007055246W WO 2007138072 A2 WO2007138072 A2 WO 2007138072A2
Authority
WO
WIPO (PCT)
Prior art keywords
triazolo
pyrazin
substituted
pyrazol
piperazin
Prior art date
Application number
PCT/EP2007/055246
Other languages
French (fr)
Other versions
WO2007138072A3 (en
Inventor
Martin James Inglis Andrews
Paul Edwards
Mark Stuart Chambers
Wolfgang Schmidt
Juha Andrew Clase
Gregory Bar
Kim Louise Hirst
Angus Macleod
Original Assignee
Galapagos N.V.
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 EA200870592A priority Critical patent/EA200870592A1/en
Priority to AU2007267121A priority patent/AU2007267121A1/en
Application filed by Galapagos N.V. filed Critical Galapagos N.V.
Priority to NZ573015A priority patent/NZ573015A/en
Priority to BRPI0712531-3A priority patent/BRPI0712531A2/en
Priority to MX2008015057A priority patent/MX2008015057A/en
Priority to EP07729660A priority patent/EP2029602B1/en
Priority to PL07729660T priority patent/PL2029602T3/en
Priority to DE602007006010T priority patent/DE602007006010D1/en
Priority to AT07729660T priority patent/ATE465164T1/en
Priority to JP2009512585A priority patent/JP2009538877A/en
Priority to SI200730288T priority patent/SI2029602T1/en
Priority to DK07729660.6T priority patent/DK2029602T3/en
Priority to CA002653506A priority patent/CA2653506A1/en
Publication of WO2007138072A2 publication Critical patent/WO2007138072A2/en
Publication of WO2007138072A3 publication Critical patent/WO2007138072A3/en
Priority to IL195033A priority patent/IL195033A0/en
Priority to NO20084703A priority patent/NO20084703L/en
Priority to HR20100378T priority patent/HRP20100378T1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/4985Pyrazines or piperazines ortho- or peri-condensed with heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/02Stomatological preparations, e.g. drugs for caries, aphtae, periodontitis
    • 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
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • 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
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • 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

Definitions

  • the present invention relates to a class of triazolopyrazine compounds capable of binding to the active site of a serine/threonine kinase, the expression of which is involved in the pathway resulting in the degradation of extra-cellular matrix (ECM), joint degeneration and diseases involving such degradation and/or inflammation.
  • ECM extra-cellular matrix
  • Diseases involving the degradation of extra-cellular matrix include, but are not limited to, psoriatic arthritis, juvenile arthritis, early arthritis, reactive arthritis, osteoarthritis, ankylosing spondylitis, osteoporosis, muskulo skeletal diseases like tendonitis and periodontal disease, cancer metastasis, airway diseases (COPD, asthma), renal and liver fibrosis, cardio-vascular diseases like atherosclerosis and heart failure, and neurological diseases like neuroinflammation and multiple sclerosis.
  • Diseases involving primarily joint degeneration include, but are not limited to, psoriatic arthritis, juvenile arthritis, early arthritis, reactive arthritis, osteoarthritis, and ankylosing spondylitis.
  • Rheumatoid arthritis is a chronic joint degenerative disease, characterized by inflammation and destruction of the joint structures. When the disease is unchecked, it leads to substantial disability and pain due to loss of joint functionality and even premature death. The aim of an RA therapy, therefore, is not to slow down the disease but to attain remission in order to stop the joint destruction. Besides the severity of the disease outcome, the high prevalence of RA ( ⁇ 0.8% of adults are affected worldwide) means a high socio-economic impact. (For reviews on RA, we refer to Smolen and Steiner (2003); Lee and Weinblatt (2001); Choy and Panayi (2001); O'Dell (2004) and Firestein (2003)).
  • RA auto-immune disease
  • B-cells, T-cells, macrophages, fibroblasts, endothelial cells, dendritic cells and others B-cells, T-cells, macrophages, fibroblasts, endothelial cells, dendritic cells and others.
  • cytokines an increased production of various cytokines is observed in the joints and tissues surrounding the joint (e.g. TNF- ⁇ , IL-6, IL-I, IL-15, IL-18 and others).
  • the pannus mediates the degradation of the adjacent cartilage, leading to the narrowing of the joint space, and has the potential to invade adjacent bone and cartilage.
  • bone and cartilage tissues are composed mainly of collagen type I or II, respectively, the pannus destructive and invasive properties are mediated by the secretion of collagenolytic proteases, principally the matrix metallo proteinases (MMPs).
  • MMPs matrix metallo proteinases
  • the erosion of the bone under and adjacent to the cartilage is also part of the RA process, and results principally from the presence of osteoclasts at the interface of bone and pannus.
  • Osteoclasts are multinucleated cells that, upon adhesion to the bone tissue, form a closed compartment, within which the osteoclasts secrete proteases (Cathepsin K, MMP9) that degrade the bone tissue.
  • the osteoclast population in the joint is abnormally increased by osteoblast formation from precursor cells induced by the secretion of the receptor activator of NFKB ligand (RANKL) by activated SFs and T-cells.
  • RNKL NFKB ligand
  • Collagenstype I and collagen type II are the main components of bone and cartilage, respectively.
  • Collagen proteins typically organise into multimeric structures referred to as collagen fibrils. Native collagen fibrils are very resistant to proteolytic cleavage.
  • MMPs and Cathepsins are the most sensitive collagen in the body.
  • Cathepsins cathepsin K, which is active mainly in osteoclasts, is the best characterised.
  • MMPs MMPl, MMP2, MMP8 MMP13 and MMP14 are known to have collagenolytic properties.
  • MMPl represents a highly relevant collagen degrading protein.
  • cytokines relevant in the RA pathology e.g. TNF- ⁇ and ILlB
  • MMPl expression by SFs therefore is a relevant readout in the field of RA as it is indicative for the activation of SFs towards an erosive phenotype that, in vivo, is responsible for cartilage degradation. Inhibition of the MMPl expression by SFs represents a valuable therapeutic approach towards the treatment of RA.
  • the activity of the ECM-degrading proteins can also be causative or correlate with the progression of various diseases different from RA, e.g. other diseases that involve the degradation of the joints. These diseases include, but are not limited to, psoriatic arthritis, juvenile arthritis, early arthritis, reactive arthritis, osteoarthritis, and ankylosing spondylitis.
  • osteoporosis Other diseases that may be treatable with compounds identified according to the present invention and using the targets involved in the expression of MMPs as described herein are osteoporosis, muscular skeletal diseases like tendonitis and periodontal disease (Gapski et al., 2004), cancer metastasis (Coussens et al., 2002), airway diseases (COPD, asthma) (Suzuki et al., 2004), lung, renal fibrosis (Schanstra et al., 2002), liver fibrosis associated with chronic hepatitis C (Reiff et al., 2005), cardio-vascular diseases like atherosclerosis and heart failure (Creemers et al., 2001), and neurological diseases like neuroinflammation and multiple sclerosis (Rosenberg, 2002). Patients suffering from such diseases may benefit from stabilizing the ECM (by protecting it from degradation).
  • MAPKAPK5 Kinase- Activated Protein Kinase 5
  • PRAK Nuclear Localization signal
  • NES nuclear export signal
  • Endogenous MAPKAPK5 is predominantly present in the cytoplasm, but stress or cytokine activation of the cells mediates its translocation into the nucleus (New et al., 2003). This event is dependent on phosphorylation of MAPKAPK5.
  • Thrl82 is the regulatory phosphorylation site of MAPKAPK5.
  • MAPKAPK5 knock-out mice have been generated that are viable and fertile. The phenotype of these mice is quite different from that of mice deficient for MAPKAPK2, a MAPKAPK5 related kinase that is regulated by p38 ⁇ (Shi et al., 2003). This indicates that the function of each protein is distinct and that neither kinase can compensate for the other's activity. Taken together, MAPKAPK5 and MAPKAPK2 represent distinct targets with a non-redundant role.
  • MAPK6 also referred to as ERK3
  • NSAIDS Non-steroidal anti-inflammatory drugs
  • RA RA-associated joint destruction
  • Corticosteroids were found to decrease the progression of RA as detected radiographically and are used at low doses to treat part of the RA patients (30 to 60%). Serious side effects, however, are associated with long corticosteroid use (skin thinning, osteoporosis, cataracts, hypertension, hyperlipidemia).
  • Synthetic DMARDs Disease-Modifying Anti-Rheumatic Drugs
  • DMARDs Disease-Modifying Anti-Rheumatic Drugs
  • these drugs only have a limited efficacy (joint destruction is only slowed down but not blocked by DMARDs such that disease progression in the long term continues).
  • the lack of efficacy is indicated by the fact that, on average, only 30% of the patients achieve an ACR40 score after 24 months treatment with methotrexate. This means that, according to the American College of Rheumatology, only 30% of the patients achieve a 50% improvement of their symptoms (O'Dell et al., 1996).
  • Biological DMARDs are therapeutic proteins that inactivate cytokines (e.g. TNF- ⁇ ) or cells (e.g. T-cells or B-cells) that have an important role in the RA pathophysiology (Kremer et al., 2003; Edwards et al., 2004).
  • TNF- ⁇ -blockers Infliximab, Etanercept, Adalimumab
  • methotrexate combination therapy is the most effective RA treatment currently available, it is striking that even this therapy only achieves a 50% improvement (ACR40) in disease symptoms in 50-60% of patients after 12 months therapy (St Clair et al., 2004).
  • the TNF- ⁇ blockers also share the general disadvantages of the biological class of therapeutics, which are the unpleasant way of administration (frequent injections accompanied by infusion site reactions) and the high production cost.
  • Newer agents in late development phase target T-cell co-stimulatory molecules and B-cells.
  • the efficacy of these agents is expected to be similar to that of the TNF- ⁇ blockers.
  • the present invention is based on the discovery of that MAPKAPK5 functions in the pathway that results in the expression of MMPl, and that inhibitors of MAPKAPK5 activity, such as the compounds of the present invention, are useful for the treatment of diseases involving the abnormally high expression of MMP activity.
  • the present invention relates to compounds having matrix metallo proteinase inhibiting properties in a mammalian cell, according to formula (I): wherein:
  • E is NH or CR"R6, when k is zero, and is NH or CR"R6a, when k is one;
  • F is sulphur, oxygen or NH
  • T is oxygen or NR
  • U, V, W and X are independently CR"R7 or NR"; Y is CR" or N;
  • Z is hydrogen, amino, hydroxyl, lower alkoxy, carbamoyl, carboxyl, SO 2 RZ, SO 2 NRRZ, -
  • R is independently hydrogen or lower alkyl
  • R" is H or forms a double bond with an adjacent atom
  • Rl is H; R2; or lower alkyl, lower cycloalkyl and lower alkyl-lower cycloalkyl, optionally substituted with one or more R2;
  • R3 is H or forms a double bond with an adjacent R";
  • R2 is H, F, Cl; CN; COOR4; 0R4; C(O)N(R4R5); S(O) 2 N(R4R5); lower alkyl; O- lower alkyl;
  • R6 is hydrogen, amino, hydroxyl, carbamoyl, carboxyl, SO 2 R, NRR', -(CO)-OR, or -(CO)-NRR';
  • R6a is R6, Cl, F, lower alkoxy, cyano, trifluoromethoxy; or together with the adjacent be -
  • R7 is independently hydrogen, halogen, lower alkyl or lower alkoxy;
  • Rz is hydrogen, lower alkyl, lower alkanoyl, phenyl, 1-loweralkyl pyrrolidin-3-yl, pyrazol-4-yl, pyrazol-2-yl, or lower alkyl, lower alkanoyl, phenyl, 1-loweralkyl pyrrolidin-3-yl, pyrazol-4- yl, pyrazol-2-yl or pyrid-3-yl substituted by one or more of hydroxyl, amino, mono- or di- loweralkylamino, acetamidyl, lower alkanoyl, lower alkyl, 4-hydroxy-phenyl, 3- aminomethylphenyl, lower alkyl sulfonyl, 4-diloweralkylaminophenyl, pyrid-3-yl, lH-indol-
  • R and Rz together may be -(CHR) q -T-(CHR) r - and form a five or six member heterocyclic ring with the nitrogen to which they are bonded;
  • Rz and R7 together may be -(CHR") n -NR-(CHR") p -, and form a five or six member heterocyclic ring fused to the ring to which they are bonded;
  • b and d are independently O or 1; provided at least one of b or d is 1; k is O or 1 ; m is O or 1 ; n and p are independently O, 1 or 2; q and r are 1 or 2; x is o or 1 ; with the provisos that at least one of R7 or Rz is other than hydrogen; or a pharmaceutically acceptable salt, hydrate, solvate or prodrug thereof.
  • Another aspect of the present invention is compounds according to formula III:
  • R 1 is H, or substituted or unsubstituted alkyl; and each of R 8 and R 9 is independently selected from substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl; or a pharmaceutically acceptable salt, solvate or prodrug thereof; and stereoisomers, isotopic variants and tautomers thereof.
  • R 8 is selected from substituted or unsubstituted cyclopentyl, cyclohexyl, substituted or unsubstituted phenyl, substituted or unsubstituted pyridyl, substituted or unsubstituted pyrimidine, and substituted or unsubstituted pyrazine, substituted or unsubstituted pyrrole, substituted or unsubstituted pyrazole and substituted or unsubstituted imidazole.
  • R 1 is H, Me, Et, i-Pr or
  • R 1 is H.
  • Another aspect of the present invention relates to compounds according to formula
  • IVa, IVb, IVc, or IVd
  • L is a bond, -CO-, -O(CH 2 ) ml -, -CON(H)(CH 2 ) ml -, or - NHCO-; the subscript ml is selected from 1-4; the ring P is substituted or unsubstituted heterocycloalkyl; the subscript n is selected from 1-4; each R 8a is independently selected from hydrogen, substituted or unsubstituted alkyl, alkoxy, cyano, carbamoyl, CHO, and halo; and R 9 is independently selected from substituted or unsubstituted aryl and heteroaryl; or a pharmaceutically acceptable salt, solvate or prodrug thereof; and stereoisomers, isotopic variants and tautomers thereof.
  • R 9 is substituted or unsubstituted aryl. In another embodiment, R 9 is substituted or unsubstituted phenyl. [0024] In a one embodiment, with respect to compounds of formulae IH-IVd, R 9 is substituted or unsubstituted heteroaryl. In another embodiment, R 9 is substituted or unsubstituted pyridyl.
  • R 9 is selected from substituted or unsubstituted phenyl, indolyl, isoinolyl, pyrrolyl, furanyl, thienyl, pyrazolyl, oxazolyl, and thiazolyl.
  • the present invention provides pharmaceutical compositions comprising a triazolopyrazine compound of the invention, and a pharmaceutical carrier, excipient or diluent.
  • the pharmaceutical composition can comprise one or more of the compounds described herein.
  • the compounds of the present invention useful in the pharmaceutical compositions and treatment methods disclosed herein are all pharmaceutically acceptable as prepared and used.
  • Another aspect of this invention relates to the use of the present compound in a therapeutic method, a pharmaceutical composition, and the manufacture of such composition, useful for the treatment of diseases involving inflammation, collagen degradation, and in particular, diseases characteristic of abnormal matrix metallo protease (MMPl) and/or Mitogen- Activated Protein-Kinase
  • MMPl abnormal matrix metallo protease
  • MAPKAPK5 Activated Protein Kinase 5
  • RA rheumatoid arthritis
  • This invention also relates to processes for the preparation of the present compounds.
  • Figure 1 This diagram shows the striking histological differences between a healthy joint and that of a RA patient.
  • FIG. 1 This chart shows the increased expression of MMPl in synovial fibroblasts triggered with cytokines involved in rheumatoid arthritis pathology.
  • FIG. 3 shows the dose-dependent inhibition of the "TNF- ⁇ -based trigger"-induced expression of MMPl by SFs by a known anti- inflammatory compound.
  • Figure 4. This gel shows the reduction, at the protein level, of the expression of
  • MAPKAPK5 in SFs by infection of the cells with Ad-siRNA virus targeting MAPKAPK5.
  • halo such as fluoro, chloro, bromo
  • -CN -CF 3
  • -OH -OCF 3
  • C 2 -C 6 alkenyl C 3 -C 6 alkynyl
  • CpC 6 alkoxy aryl and di-C r C 6 alkylamino.
  • Alkoxy means alkyl-O-. Exemplary alkoxy includes methoxy, ethoxy, n-propoxy, i-propoxy, n-butoxy, and heptoxy. Preferred alkoxy groups are lower alkoxy.
  • Alkyl means straight or branched aliphatic hydrocarbon having 1 to about 20 carbon atoms. Preferred alkyl has 1 to about 12 carbon atoms. More preferred is lower alkyl. Branched means that one or more lower alkyl groups such as methyl, ethyl or propyl is attached to a linear alkyl chain.
  • alkyl amino means alkyl-NH-.
  • Preferred alkyl amino is (Ci-Ce)-alkyl amino.
  • Exemplary alkyl amino includes methylamino and ethylamino.
  • amino lower alkanoyl means NH 2 -R-CO-, where R is lower alkylene.
  • Preferred groups include aminoethanoyl and aminoacetyl.
  • Carbamoyl lower alkyl means the radical NH 2 CO-lower alkyl-. Preferred groups include carbamoylethyl and carbamoylmethyl.
  • Carboxy lower alkyl ester means a lower alkyl ester of a carboxy radical, -COO- group.
  • Halo or "halogen” means fluoro, chloro, bromo, or iodo.
  • Haldrogen means in the context of a substituent that -H is present at the compound position and also includes its isotope, deuterium.
  • “Lower alkanoyl amino” means an amino group with an organic functional group R-
  • “Lower alkyl” means 1 to about 6 carbon atoms in a linear alkyl chain that may be straight or branched.
  • “Lower alkoxy” means 1 to about 6 carbon atoms in a linear alkyl chain that may be straight or branched, and that is bonded by an oxygen atom.
  • “Lower alkyl sulphonamide” refers to a lower alkyl amide of sulphonamide of the formula -SO 2 NR 515 R*, where R* is hydrogen or lower alkyl, and at least one R* is lower alkyl.
  • “Prophylaxis” means a measure taken for the prevention of a disease.
  • Solvate means a physical association of a compound useful in this invention with one or more solvent molecules. This physical association includes hydrogen bonding. In certain instances the solvate will be capable of isolation, for example when one or more solvent molecules are incorporated in the crystal lattice of the crystalline solid. "Solvate” encompasses both solution-phase and isolable solvates.
  • the compounds of the invention may be prepared e.g. in crystalline form and may be solvated or hydrated. Suitable solvates include pharmaceutically acceptable solvates, such as hydrates, and further include both stoichiometric solvates and non-stoichiometric solvates. Conventional solvents include water, ethanol, acetic acid and the like, therefore, representative solvates include hydrates, ethanolates and methanolates.
  • Substituted means that one atom or group of atoms in a molecule is replaced by another atom or group.
  • Sulphonamide refers to a group of compounds containing the chemical group -
  • “Therapeutically effective amount” means that amount of a drug or pharmaceutical agent that will elicit the biological or medical response of a subject that is being sought by a medical doctor or other clinician.
  • the “therapeutically effective amount” can vary depending on the compound, the disease and its severity, and the age, weight, etc., of the subject to be treated.
  • the term "effective matrix metallo-protease inhibiting amount” is intended to mean that effective amount of an compound of the present invention that will bring about a biologically meaningful decrease in the production of MMP-I in the subject's disease affected tissues such that extracellular matrix degradation is meaningfully reduced.
  • a compound having matrix metallo-protease inhibiting properties or a "matrix metallo-protease inhibiting compound” means a compound of the present invention that provided to a cell in effective amounts is able to cause a biologically meaningful decrease in the production of MMP-I in such cells.
  • Aryl refers to a monovalent aromatic hydrocarbon group derived by the removal of one hydrogen atom from a single carbon atom of a parent aromatic ring system.
  • Typical aryl groups include, but are not limited to, groups derived from aceanthrylene, acenaphthylene, acephenanthrylene, anthracene, azulene, benzene, chrysene, coronene, fluoranthene, fluorene, hexacene, hexaphene, hexalene, ⁇ s-indacene, s-indacene, indane, indene, naphthalene, octacene, octaphene, octalene, ovalene, penta-2,4-diene, pentacene, pentalene, pentaphene, perylene, phenalene, phenanthrene, picene, ple
  • Substituted Aryl includes those groups recited in the definition of "substituted” herein, and particularly refers to an aryl group that may optionally be substituted with 1 or more substituents, for instance from 1 to 5 substituents, particularly 1 to 3 substituents, selected from the group consisting of acyl, acylamino, acyloxy, alkenyl, substituted alkenyl, alkoxy, substituted alkoxy, alkoxycarbonyl, alkyl, substituted alkyl, alkynyl, substituted alkynyl, amino, substituted amino, aminocarbonyl, aminocarbonylamino, aminocarbonyloxy, aryl, aryloxy, azido, carboxyl, cyano, cycloalkyl, substituted cycloalkyl, halogen, hydroxyl, nitro, thioalkoxy, substituted thioalkoxy, thioaryloxy, thiol, alkyl-
  • Bicycloaryl refers to a monovalent aromatic hydrocarbon group derived by the removal of one hydrogen atom from a single carbon atom of a parent bicycloaromatic ring system.
  • Typical bicycloaryl groups include, but are not limited to, groups derived from indane, indene, naphthalene, tetrahydronaphthalene, and the like. Particularly, an aryl group comprises from 8 to 11 carbon atoms.
  • Bicycloheteroaryl refers to a monovalent bicycloheteroaromatic group derived by the removal of one hydrogen atom from a single atom of a parent bicycloheteroaromatic ring system.
  • Typical bicycloheteroaryl groups include, but are not limited to, groups derived from benzofuran, benzimidazole, benzindazole, benzdioxane, chromene, chromane, cinnoline, phthalazine, indole, indoline, indolizine, isobenzofuran, isochromene, isoindole, isoindoline, isoquinoline, benzothiazole, benzoxazole, naphthyridine, benzoxadiazole, pteridine, purine, benzopyran, benzpyrazine, pyridopyrimidine, quinazoline, quinoline, quinolizine, quinoxaline, benzomorphan, tetrahydroisoquinoline, tetrahydroquinoline, and the like.
  • the bicycloheteroaryl group is between 9-11 membered bicycloheteroaryl, with 5-10 membered heteroaryl being particularly preferred.
  • Particular bicycloheteroaryl groups are those derived from benzothiophene, benzofuran, benzothiazole, indole, quinoline, isoquinoline, benzimidazole, benzoxazole and benzdioxane.
  • Carbamoyl refers to the radical -C(O)N(R 42 ) 2 where each R 42 group is independently hydrogen, alkyl, cycloalkyl or aryl, as defined herein, which may be optionally substituted as defined herein.
  • Carbamoyl refers to -C(O)-NH 2 .
  • Cycloalkyl refers to cyclic hydrocarbyl groups having from 3 to about 10 carbon atoms and having a single cyclic ring or multiple condensed rings, including fused and bridged ring systems, which optionally can be substituted with from 1 to 3 alkyl groups.
  • Such cycloalkyl groups include, by way of example, single ring structures such as cyclopropyl, cyclobutyl, cyclopentyl, cyclooctyl, 1 -methylcyclopropyl, 2-methylcyclopentyl, 2-methylcyclooctyl, and the like, and multiple ring structures such as adamantanyl, and the like.
  • Substituted cycloalkyl includes those groups recited in the definition of
  • substituted herein, and particularly refers to a cycloalkyl group having 1 or more substituents, for instance from 1 to 5 substituents, and particularly from 1 to 3 substituents, selected from the group consisting of acyl, acylamino, acyloxy, alkoxy, substituted alkoxy, alkoxycarbonyl, alkoxycarbonylamino, amino, substituted amino, aminocarbonyl, aminocarbonylamino, aminocarbonyloxy, aryl, aryloxy, azido, carboxyl, cyano, cycloalkyl, substituted cycloalkyl, halogen, hydroxyl, keto, nitro, thioalkoxy, substituted thioalkoxy, thioaryloxy, thioketo, thiol, alkyl-S(O)-, aryl-S(O)-, alkyl-S(O) 2 - and aryl-S(O) 2 -.
  • substituents for instance
  • Substituted refers to a group in which one or more hydrogen atoms are each independently replaced with the same or different substituent(s).
  • R 52 and R 53 may be hydrogen and at least one of R 52 and R 53 is each independently selected from alkyl, alkenyl, alkynyl, cycloheteroalkyl, alkanoyl, alkoxy, aryloxy, heteroaryloxy, alkylamino, arylamino, heteroarylamino, NR 54 COR 55 , NR 54 SOR 55 ; NR 54 SO 2 R 57 , COOalkyl, COOaryl, CONR 54 R 55 , CONR 54 OR 55 , NR 54 R 55 , SO 2 NR 54 R 55 , S-alkyl, S-alkyl, SOalkyl, S0 2 alkyl, Saryl, SOaryl, S0 2 aryl; or R 52 and R 53 may be joined to form a cyclic ring (saturated or unsaturated) from 5 to 8 atoms, optionally containing one or more heteroatoms selected from the group N, O or S
  • R 54 , R 55 , and R 56 are independently hydrogen, alkyl, alkenyl, alkynyl, perfluoroalkyl, cycloalkyl, cycloheteroalkyl, aryl, substituted aryl, heteroaryl, substituted or hetero alkyl or the like.
  • Hetero when used to describe a compound or a group present on a compound means that one or more carbon atoms in the compound or group have been replaced by a nitrogen, oxygen, or sulfur heteroatom. Hetero may be applied to any of the hydrocarbyl groups described above such as alkyl, e.g. heteroalkyl, cycloalkyl, e.g. cycloheteroalkyl, aryl, e.g. heteroaryl, cycloalkenyl, cycloheteroalkenyl, and the like having from 1 to 5, and especially from 1 to 3 heteroatoms.
  • Heteroaryl refers to a monovalent heteroaromatic group derived by the removal of one hydrogen atom from a single atom of a parent heteroaromatic ring system.
  • Typical heteroaryl groups include, but are not limited to, groups derived from acridine, arsindole, carbazole, ⁇ -carboline, chromane, chromene, cinnoline, furan, imidazole, indazole, indole, indoline, indolizine, isobenzofuran, isochromene, isoindole, isoindoline, isoquinoline, isothiazole, isoxazole, naphthyridine, oxadiazole, oxazole, perimidine, phenanthridine, phenanthroline, phenazine, phthalazine, pteridine, purine, pyran, pyrazine, pyrazole,
  • the heteroaryl group is between 5-15 membered heteroaryl, with 5-10 membered heteroaryl being particularly preferred.
  • Particular heteroaryl groups are those derived from thiophene, pyrrole, benzothiophene, benzofuran, indole, pyridine, quinoline, imidazole, oxazole and pyrazine.
  • Examples of representative heteroaryls include the following:
  • each Y is selected from carbonyl, N, NR 58 , O, and S; and R 58 is independently hydrogen, alkyl, cycloalkyl, cycloheteroalkyl, aryl, heteroaryl, heteroalkyl or the like.
  • cycloheteroalkyl refers to a stable heterocyclic non- aromatic ring and fused rings containing one or more heteroatoms independently selected from N, O and S.
  • a fused heterocyclic ring system may include carbocyclic rings and need only include one heterocyclic ring.
  • heterocyclic rings include, but are not limited to, piperazinyl, homopiperazinyl, piperidinyl and morpholinyl, and are shown in the following illustrative examples:
  • each X is selected from CR 2 , NR , O and S; and each Y is selected from NR , O and S; and R 58 is independently hydrogen, alkyl, cycloalkyl, cycloheteroalkyl, aryl, heteroaryl, heteroalkyl or the like.
  • cycloheteroalkyl rings may be optionally substituted with one or more groups selected from the group consisting of acyl, acylamino, acyloxy, alkoxy, substituted alkoxy, alkoxycarbonyl, alkoxycarbonylamino, amino, substituted amino, aminocarbonyl, aminocarbonylamino, aminocarbonyloxy, aryl, aryloxy, azido, carboxyl, cyano, cycloalkyl, substituted cycloalkyl, halogen, hydroxyl, keto, nitro, thioalkoxy, substituted thioalkoxy, thioaryloxy, thioketo, thiol, alkyl-S(O)-, aryl-S(O)-, alkyl-S(O) 2 - and aryl-S(O) 2 -.
  • Substituting groups include carbonyl or thiocarbonyl which provide, for example, lactam and urea derivative
  • each X is selected from CR 58 2 , NR 58 , O and S; and each Y is selected from carbonyl, N, NR 58 , O and S; and R 58 is independently hydrogen, alkyl, cycloalkyl, cycloheteroalkyl, aryl, heteroaryl, heteroalkyl or the like.
  • Examples of representative aryl having hetero atoms containing substitution include the following:
  • each X is selected from C-R 58 2 , NR 58 , O and S; and each Y is selected from carbonyl, NR 58 , O and S; and R 58 is independently hydrogen, alkyl, cycloalkyl, cycloheteroalkyl, aryl, heteroaryl, heteroalkyl or the like.
  • heterocyclic ring may have one to four heteroatoms so long as the heteroaromatic ring is chemically feasible and stable.
  • “Pharmaceutically acceptable” means approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopoeia or other generally recognized pharmacopoeia for use in animals, and more particularly in humans.
  • “Pharmaceutically acceptable salt” refers to the non-toxic, inorganic and organic acid addition salts, and base addition salts, of compounds of the present invention, in particular they are pharmaceutically acceptable and possess the desired pharmacological activity of the parent compound. These salts can be prepared in situ during the final isolation and purification of compounds useful in the present invention.
  • Such salts include: (1) acid addition salts, formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like; or formed with organic acids such as acetic acid, propionic acid, hexanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, 3-(4-hydroxybenzoyl) benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, 1,2-ethane-disulfonic acid, 2- hydroxyethanesulfonic acid, benzenesulfonic acid, 4-chlorobenzenesulfonic acid, 2- naphthalenesulfonic acid, 4-toluenesulfonic acid
  • Salts further include, by way of example only, sodium, potassium, calcium, magnesium, ammonium, tetraalkylammonium, and the like; and when the compound contains a basic functionality, salts of non toxic organic or inorganic acids, such as hydrochloride, hydrobromide, tartrate, mesylate, acetate, maleate, oxalate and the like.
  • pharmaceutically acceptable cation refers to a non toxic, acceptable cationic counter-ion of an acidic functional group. Such cations are exemplified by sodium, potassium, calcium, magnesium, ammonium, tetraalkylammonium cations, and the like.
  • “Pharmaceutically acceptable vehicle” refers to a diluent, adjuvant, excipient or carrier with which a compound of the invention is administered.
  • Preventing refers to a reduction in risk of acquiring a disease or disorder (i.e., causing at least one of the clinical symptoms of the disease not to develop in a subject that may be exposed to or predisposed to the disease but does not yet experience or display symptoms of the disease).
  • Prodrugs refers to compounds, including derivatives of the compounds of the invention, which have cleavable groups and become by solvolysis or under physiological conditions the compounds of the invention which are pharmaceutically active in vivo. Such examples include, but are not limited to, choline ester derivatives and the like, N-alkylmorpholine esters and the like.
  • Subject includes humans. The terms “human,” “patient” and “subject” are used interchangeably herein..
  • Treating” or “treatment” of any disease or disorder refers, in one embodiment, to ameliorating the disease or disorder (i.e., arresting or reducing the development of the disease or at least one of the clinical symptoms thereof). In another embodiment “treating” or “treatment” refers to ameliorating at least one physical parameter, which may not be discernible by the subject. In yet another embodiment, “treating” or “treatment” refers to modulating the disease or disorder, either physically, ⁇ e.g., stabilization of a discernible symptom), physiologically, ⁇ e.g., stabilization of a physical parameter), or both. In yet another embodiment, “treating” or “treatment” refers to delaying the onset of the disease or disorder.
  • Prodrugs include acid derivatives well know to practitioners of the art, such as, for example, esters prepared by reaction of the parent acid with a suitable alcohol, or amides prepared by reaction of the parent acid compound with a substituted or unsubstituted amine, or acid anhydrides, or mixed anhydrides.
  • Simple aliphatic or aromatic esters, amides and anhydrides derived from acidic groups pendant on the compounds of this invention are preferred prodrugs.
  • double ester type prodrugs such as (acyloxy)alkyl esters or ((alkoxycarbonyl)oxy)alkylesters.
  • Preferred are the Ci to Cg alkyl, C 2 -C8 alkenyl, aryl, C 7 -C 12 substituted aryl, and C 7 -Ci 2 arylalkyl esters of the compounds of the invention.
  • the term "isotopic variant” refers to a compound that contains unnatural proportions of isotopes at one or more of the atoms that constitute such compound.
  • an "isotopic variant" of a compound can contain one or more non-radioactive isotopes, such as for example, deuterium ( 2 H or D), carbon-13 ( 13 C), nitrogen-15 ( 15 N), or the like.
  • non-radioactive isotopes such as for example, deuterium ( 2 H or D), carbon-13 ( 13 C), nitrogen-15 ( 15 N), or the like.
  • the following atoms, where present may vary, so that for example, any hydrogen may be 2 HZO, any carbon may be 13 C, or any nitrogen may be 15 N, and that the presence and placement of such atoms may be determined within the skill of the art.
  • the invention may include the preparation of isotopic variants with radioisotopes, in the instance for example, where the resulting compounds may be used for drug and/or substrate tissue distribution studies.
  • radioactive isotopes tritium, i.e. 3 H, and carbon-14, i.e. 14 C are particularly useful for this purpose in view of their ease of incorporation and ready means of detection.
  • compounds may be prepared that are substituted with positron emitting isotopes, such as 11 C, 18 F, 15 O and 13 N, and would be useful in Positron Emission Topography (PET) studies for examining substrate receptor occupancy.
  • PET Positron Emission Topography
  • stereoisomers that are not mirror images of one another are termed “diastereomers” and those that are non-superimposable mirror images of each other are termed “enantiomers”.
  • enantiomers When a compound has an asymmetric center, for example, it is bonded to four different groups, a pair of enantiomers is possible.
  • An enantiomer can be characterized by the absolute configuration of its asymmetric center and is described by the R- and S-sequencing rules of Cahn and Prelog, or by the manner in which the molecule rotates the plane of polarized light and designated as dextrorotatory or levorotatory (i.e., as (+) or (-)-isomers respectively).
  • a chiral compound can exist as either individual enantiomer or as a mixture thereof. A mixture containing equal proportions of the enantiomers is called a "racemic mixture".
  • Tautomers refer to compounds that are interchangeable forms of a particular compound structure, and that vary in the displacement of hydrogen atoms and electrons. Thus, two structures may be in equilibrium through the movement of ⁇ electrons and an atom (usually H). For example, enols and ketones are tautomers because they are rapidly interconverted by treatment with either acid or base. Another example of tautomerism is the aci- and nitro- forms of phenylnitromethane, that are likewise formed by treatment with acid or base.
  • Tautomeric forms may be relevant to the attainment of the optimal chemical reactivity and biological activity of a compound of interest.
  • the compounds of this invention may possess one or more asymmetric centers; such compounds can therefore be produced as individual (R)- or (S)- stereoisomers or as mixtures thereof.
  • the present invention is based on the discovery of that MAPKAPK5 functions in the pathway that results in the expression of MMPl, and that inhibitors of MAPKAPK5 activity, such as the compounds of the present invention, are useful for the treatment of diseases involving the abnormally high expression of MMP activity.
  • the present invention relates to compounds having matrix metallo proteinase inhibiting properties in a mammalian cell, according to formula (I):
  • a and B are independently CR2R", NR", oxygen or sulphur;
  • AA is CR2 or N
  • E is NH or CR"R6, when k is zero, and is NH or CR"R6a, when k is one;
  • F is sulphur, oxygen or NH
  • T is oxygen or NR
  • U, V, W and X are independently CR"R7 or NR"; Y is CR" or N;
  • Z is hydrogen, amino, hydroxyl, lower alkoxy, carbamoyl, carboxyl, SO 2 RZ, SO 2 NRRZ, -
  • R is independently hydrogen or lower alkyl
  • R" is H or forms a double bond with an adjacent atom
  • Rl is H; R2; or lower alkyl, lower cycloalkyl and lower alkyl-lower cycloalkyl, optionally substituted with one or more R2;
  • R3 is H or forms a double bond with an adjacent R";
  • R2 is H, F, Cl; CN; COOR4; 0R4; C(O)N(R4R5); S(O) 2 N(R4R5); lower alkyl; O- lower alkyl;
  • R6 is hydrogen, amino, hydroxyl, carbamoyl, carboxyl, SO 2 R, NRR', -(CO)-OR, or -(CO)-NRR';
  • R6a is R6, Cl, F, lower alkoxy, cyano, trifluoromethoxy; or together with the adjacent be -
  • R7 is independently hydrogen, halogen, lower alkyl or lower alkoxy;
  • Rz is hydrogen, lower alkyl, lower alkanoyl, phenyl, 1-loweralkyl pyrrolidin-3-yl, pyrazol-4-yl, pyrazol-2-yl, or lower alkyl, lower alkanoyl, phenyl, 1-loweralkyl pyrrolidin-3-yl, pyrazol-4- yl, pyrazol-2-yl or pyrid-3-yl substituted by one or more of hydroxyl, amino, mono- or di- loweralkylamino, acetamidyl, lower alkanoyl, lower alkyl, 4-hydroxy-phenyl, 3- aminomethylphenyl, lower alkyl sulfonyl, 4-diloweralkylaminophenyl, pyrid-3-yl, lH-indol-
  • R and Rz together may be -(CHR) q -T-(CHR) r - and form a five or six member heterocyclic ring with the nitrogen to which they are bonded;
  • Rz and R7 together may be -(CHR") n -NR-(CHR") p -, and form a five or six member heterocyclic ring fused to the ring to which they are bonded;
  • b and d are independently 0 or 1; provided at least one of b or d is 1;
  • k is 0 or 1 ;
  • m is 0 or 1 ;
  • n and p are independently 0, 1 or 2;
  • q and r are 1 or 2;
  • x is o or 1 ; with the provisos that at least one of R7 or Rz is other than hydrogen; or a pharmaceutically acceptable salt, hydrate, solvate or prodrug thereof.
  • a preferred aspect of the present invention is a subclass of compounds according to formula II,
  • a and B are independently CR2R", NR", oxygen or sulphur;
  • AA is CR2 or N
  • E is NH or CR"R6, when k is zero, and is NH or CR"R6a, when k is one;
  • F is sulphur, oxygen or NH
  • T is oxygen or NR
  • R" is H or forms a double bond with an adjacent atom
  • R is independently hydrogen or lower alkyl
  • Rl is H; R2; or lower alkyl, lower cycloalkyl and lower alkyl-lower cycloalkyl, optionally substituted with one or more R2;
  • R2 is H, F, Cl; CN; COOR4; OR4; C(O)N(R4R5); S(O) 2 N(R4R5); lower alkyl; O- lower alkyl;
  • R4 and R5 are independently H; F, Cl; or lower alkyl, lower cycloalkyl, or lower alkyl-lower cycloalkyl optionally substituted with one or more of F and Cl;
  • R6 is hydrogen, amino, hydroxyl, carbamoyl, carboxyl, SO 2 R, NRR', -(CO)-OR, or -(CO)-NRR';
  • R6a is R6, Cl, F, lower alkoxy, cyano, trifluoromethoxy; or together with the adjacent be - (CHR") n -NR-(CHR") p -, and form a five or six member heterocyclic ring fused to the ring to which they are bonded;
  • R8 is phenyl independently substituted by R a in the ortho-position, by R b in the meta-position, and by R c in the para-position; pyrid-3-yl; pyrid-3-yl substituted by R c in the 5 -position; or cyclohexyl independently substituted by R a in the 2-position, and by Rd in the 4-position;
  • R a is hydrogen, halogen, lower alkyl, trifluoromethyl or lower alkoxy;
  • Rb is hydrogen, trifluoromethyl, lower alkoxy, lower alkyl sulfonamide, carboxyl, -
  • R c is hydrogen, amino, hydroxyl, lower alkoxy, carbamoyl, carboxyl, SO 2 R, SO 2 NR e R f ,
  • Rb and R c can together form a benzdiazole, or indole substituted in the 3 -position by R';
  • Rd is hydroxyl, halogen, amino, lower alkoxy, or NR e R f ;
  • R e and R f are independently hydrogen, 1-loweralkyl pyrrolidin-3-yl, 1 -R-pyrazol-4-yl, lower alkanoyl, phenyl, or lower alkyl optionally substituted by one or more of 4-hydroxy- phenyl, 3-aminomethylphenyl, lower alkyl sulfonyl, 4-diloweralkylaminophenyl, pyrid-3-yl, lH-indol-3-yl, morpholin-4-yl, hydroxyl, amino, mono- or di- loweralkylamino, or by lower alkanoyl; or R' and R" together are -(CHR) n -T- (CHR) n - and form a five or six member heterocyclic ring with the nitrogen to which they are bonded; m is O, 1, or 2; n is 1 or 2; with the proviso that at least one of R a , Rb and R c
  • a particularly preferred embodiment of the present invention relates to compounds according to formula III:
  • R 1 is H, or substituted or unsubstituted alkyl; and each of R 8 and R 9 is independently selected from substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl; or a pharmaceutically acceptable salt, solvate or prodrug thereof; and stereoisomers, isotopic variants and tautomers thereof.
  • R 8 is selected from substituted or unsubstituted cyclopentyl, cyclohexyl, substituted or unsubstituted phenyl, substituted or unsubstituted pyridyl, substituted or unsubstituted pyrimidine, and substituted or unsubstituted pyrazine, substituted or unsubstituted pyrrole, substituted or unsubstituted pyrazole and substituted or unsubstituted imidazole.
  • R 1 is H, Me, Et, i-Pr or
  • R 1 is H.
  • R 8 is selected from substituted or unsubstituted cycloalkyl.
  • R 8 is selected from substituted or unsubstituted cyclohexyl or cyclopentyl.
  • R 8 is selected from substituted or unsubstituted heterocycloalkyl.
  • R 8 is selected from substituted or unsubstituted piperidinyl, morpholinyl, or pyrrolidinyl.
  • R 8 is selected from substituted or unsubstituted phenyl, pyridyl or pyrimidine.
  • R 8 is selected from substituted phenyl, substituted pyridyl, and substituted pyrimidine; and the substitution is -L-R 8d ; and wherein L is selected from a bond, alkylene, heteroalkylene, -O-, -N(R 8e )-, -CO-, -CO 2 -, -SO-,
  • R 8d is selected from substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted heteroaryl, substituted or unsubstituted amino, substituted or unsubstituted aralkyl, substituted or unsubstituted heteroarylalkyl and substituted or unsubstituted aminoalkyl; and
  • R 8e is selected from H, substituted or unsubstituted alkyl and substituted or unsubstituted cycloalkyl. [00100] In one embodiment, with respect to compounds of formula III, R 8 is (R 8a )n
  • L, and R are as described in the preceding paragraph; the subscript n is selected from 1-4; and each R 8a is independently selected from hydrogen, substituted or unsubstituted alkyl, alkoxy, cyano, and halo.
  • R 8 is as described above and the subscript n is 1 and R 8a is Me, Et, Pr, iso-Pr, Cl, F, CN, OMe, or CF 3 .
  • R 8a is at 2-(ortho to -L) position.
  • R 8a is 2-Cl, 2-F, 2-Me or 2-
  • R 8 is as described above and
  • L is a bond, -O-, -CO-, -CON(R 8e )-, or -N(R 8e )CO-;
  • R 8d is selected from substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted heteroaryl, substituted or unsubstituted aralkyl, substituted or unsubstituted heteroarylalkyl and substituted or unsubstituted aminoalkyl; and
  • R 8e is selected from H, substituted or unsubstituted alkyl.
  • R 8 is as described above and
  • L is a bond, -O-, -CO-, -CON(R 8e )-, or -N(R 8e )CO-;
  • R 8d is selected from H, alkylaminoethyl, dialkylaminoethyl, cycloalkyl, heterocycloalkyl, arylalkyl, and heteroarylalkyl.
  • R 8 is as described above and
  • L is a bond, -O-, -CO-, -CON(R 8e )-, or -N(R 8e )CO-;
  • R 8d is selected from methylaminoethyl, ethylaminoethyl, dimethylaminoethyl, diethylaminoethyl, substituted or unsubstituted pyrrolidinyl, benzyl and pyridylmethyl.
  • R 8 is as described above and
  • L is a bond, -CO-, SO 2 , -(CH 2 ) ml -, -O(CH 2 ) ml -, -NH(CH 2 ) ml -, -CON(H)(CH 2 ) ml -, or - SO 2 NH(CH 2 ) mr ; the subscript ml is selected from 1-4; and R 8d is
  • L is a bond, -CO-, -O(CH 2 ) ml -, -CON(H)(CH 2 ) ml -, or - NHCO-;
  • R 8 is as described above; L is a bond; and the ring P is substituted or unsubstituted heterocycloalkyl.
  • R 8 is as described above; L is a bond; and the ring P is substituted or unsubstituted piperidine, substituted or unsubstituted piperazine, and substituted or unsubstituted piperidine, morpholine.
  • R 8 is as described above; L is CO; and the ring P is substituted or unsubstituted heterocycloalkyl.
  • R 8 is as described above; L is CO; and the ring P is substituted or unsubstituted piperidine, substituted or unsubstituted piperazine, and substituted or unsubstituted piperidine, morpholine.
  • R 8 is as described above; L is -(CH 2 ) m r, -O(CH 2 ) m r, or -NH(CH 2 ) m r; the subscript ml is selected from 1-4; and the ring P is substituted or unsubstituted heterocycloalkyl.
  • R 8 is as described above; L is -(CH 2 ) m r, -O(CH 2 ) m r, or -NH(CH 2 ) m r; the subscript ml is 2 or 3; and the ring P is substituted or unsubstituted piperidine, substituted or unsubstituted piperazine, and substituted or unsubstituted piperidine, morpholine.
  • R 8 is as described above; L is -CON(H)(CH 2 ) mr , or -NHCO(CH 2 ) mr ; the subscript ml is selected from 1-4; and the ring P is substituted or unsubstituted heterocycloalkyl.
  • R 8 is as described above; L is -CON(H)(CH 2 ) m r; the subscript ml is 2 or 3; and the ring P is substituted or unsubstituted piperidine, substituted or unsubstituted piperazine, and substituted or unsubstituted piperidine, morpholine.
  • the compound is according to formula IVa, IVb, IVc, or IVd:
  • L and the ring P are as described above; the subscript n, is selected from 1-4; each R > 8a a i ⁇ s independently selected from hydrogen, substituted or unsubstituted alkyl, alkoxy, cyano, and halo; and R 9 is independently selected from substituted or unsubstituted aryl and heteroaryl; or a pharmaceutically acceptable salt, solvate or prodrug thereof; and stereoisomers, isotopic variants and tautomers thereof.
  • L is a bond
  • L is methylene, ethylene, propylene, and butylene.
  • L is -CO-.
  • L is -SO 2 -.
  • L is -CON(H)-
  • L is -OCH 2 -
  • L is a bond
  • the ring P is substituted or unsubstituted piperidine, morpholine or piperazine.
  • L and the ring P are as described above; the subscript n is 1 and R 8a is Me, Et, Pr, iso-Pr, Cl, F, CN, OMe, or CF 3 .
  • R 8a is at 2-(ortho to -L) position.
  • R 8a is 2-Cl, 2-F, 2-
  • R 8 is
  • ring P is substituted or unsubstituted heterocycloalkyl; the subscript n is selected from
  • each R 8a is independently selected from hydrogen, substituted or unsubstituted alkyl, alkoxy, cyano, and halo.
  • R 8 is as described above and the ring P is substituted or unsubstituted piperidine, morpholine or piperazine.
  • R 8 is as described above and the subscript n is 4 and each R 8a is H.
  • R 8 is as described above and the subscript n is 1 and R 8a is Me, Et, Pr, iso-Pr, Cl, F, CN, OMe, or CF 3 .
  • R 8a is at the 2-(ortho to N-ring P) position.
  • R 8a is 2-Cl, 2-F, 2-Me or 2-CF 3 .
  • each R 8a is independently selected from hydrogen, substituted or unsubstituted alkyl, alkoxy, cyano, and halo;
  • R 8b is hydrogen, substituted or unsubstituted alkyl or substituted or unsubstituted cycloalkyl;
  • R 8c is hydrogen, substituted or unsubstituted alkyl and the subscript x is selected from 1-8.
  • each R 8a is independently selected from hydrogen, substituted or unsubstituted alkyl, alkoxy, cyano, and halo;
  • R 8b is hydrogen, substituted or unsubstituted alkyl or substituted or unsubstituted cycloalkyl;
  • R c is hydrogen or
  • each R 8a is independently selected from hydrogen, substituted or unsubstituted alkyl, alkoxy, cyano, and halo.
  • each R 8a is independently selected from hydrogen, substituted or unsubstituted alkyl, alkoxy, cyano, and halo; and R 8b is hydrogen, substituted or unsubstituted alkyl or substituted or unsubstituted cycloalkyl.
  • R 8 is as described above and the subscript n is 4 and each R 8a is H.
  • R 8 is as described above and the subscript n is 1 and R 8a is Me, Et, Pr, iso-Pr, Cl, F, CN, OMe, or CF 3 .
  • R 8a is at 2-(ortho to N-ringP) position.
  • R 8a is 2-Cl, 2-F, 2-Me or 2-CF 3 ..
  • R 8 is as described above and R 8b is H.
  • R 8 is as described above and R 8b is substituted or unsubstiuted alkyl.
  • R 8 is as described above and R 8b is substituted or unsubstituted cycloalkyl.
  • R 8 is as described above and R 8b is Me, Et, Pr, i-Pr, t-Bu, i-Bu, CH 2 CF 3 , CF 3 , CH 2 CONH 2 , cyclopropyl or cyclopropylmethyl.
  • R 8 is as described above and R 8b is i-Pr.
  • R 8a is independently selected from hydrogen, substituted or unsubstituted alkyl, alkoxy, cyano, carbamoyl,
  • R 8a is H, Me, F, or Cl. In a preferred embodiment R 8a is H.
  • the compound is according to formula Va, Vb, Vc, Vd, Ve, or Vf: Va Vb Vc
  • R 9 is as described for formula III and R 8b is hydrogen, substituted or unsubstituted alkyl or substituted or unsubstituted cycloalkyl.
  • R 8b is H.
  • R 8b is substituted or unsubstiuted alkyl.
  • R 8b is substituted or unsubstituted cycloalkyl.
  • R 8b is Me, Et, Pr, i-Pr, t-Bu, i-Bu, CH 2 CF 3 , CF 3 , CH 2 CONH 2 , cyclopropyl or cyclopropylmethyl.
  • R 8b is i-
  • R 9 is substituted or unsubstituted aryl. In another embodiment, R 9 is substituted or unsubstituted phenyl. [00152] In a one embodiment, with respect to compounds of formulae III- Vf, R 9 is substituted or unsubstituted heteroaryl. In another embodiment, R 9 is substituted or unsubstituted pyridyl.
  • R 9 is selected from substituted or unsubstituted phenyl, indolyl, isoinolyl, pyrrolyl, furanyl, thienyl, pyrazolyl, oxazolyl, and thiazolyl. [00154] In one embodiment, with respect to compounds of formulae III- Vf, R 9 is
  • each of A 1 , A 2 and A 3 is independently selected from S, O, N, NR 9a , and CR 9a ; each of R 9a is independently H or substituted or unsubstituted alkyl; and R 9b is CONH 2 , CONHMe, or CN. [00155] In further embodiment, with respect to compounds of formulae III- Vf, R 9 is
  • each R 9d is independently H, substituted or unsubstituted alkyl or halo.
  • R 9 is and wherein the subscript m is selected from 1-3 and each R 9d is independently H, substituted or unsubstituted alkyl or halo.
  • R 9 is as described above; and each R 9d is H.
  • R 9 is as described above; m is 1 or 2 and each R 9d is independently Me, Cl or F.
  • the compound is according to formula Via, VIb, Vic, VId, VIe or VIf:
  • R > 8b i ⁇ s hydrogen, substituted or unsubstituted alkyl or substituted or unsubstituted cycloalkyl.
  • R 8b is H.
  • R 8b is cycloalkyl
  • R 8b is cyclopropyl.
  • R 8b is substituted or unsubstituted alkyl.
  • the compound is according to formula Vila, VIIb, VIIc, VIIc, VIId, VIIe or VIIf:
  • R 8b is hydrogen, substituted or unsubstituted alkyl or substituted or unsubstituted cycloalkyl.
  • R 8b is H.
  • R 8b is cycloalkyl
  • R 8b is cyclopropyl
  • R 8b is substituted or unsubstituted alkyl.
  • the compound is according to formula Villa, VIIIb, VIIIc, VIIId, VIIIe or VIIIf: Villa VIIIc
  • R 8b is H.
  • R 8b is cycloalkyl
  • R 8b is cyclopropyl
  • R 8b is substituted or unsubstituted alkyl.
  • the compound is according to formula IXa, IXb, IXc, IXd, IXe, or IXf: IXa
  • R > 8b i ⁇ s hydrogen, substituted or unsubstituted alkyl or substituted or unsubstituted cycloalkyl.
  • R 8b is H.
  • R 8b is cycloalkyl
  • R 8b is cyclopropyl
  • R 8b is substituted or unsubstituted alkyl.
  • the compound is according to formula Xa, Xb, Xc, Xd, Xe, or Xf: Xa
  • R 8b is hydrogen, substituted or unsubstituted alkyl or substituted or unsubstituted cycloalkyl.
  • R 8b is H.
  • R 8b is cycloalkyl
  • R 8b is cyclopropyl
  • R 8b is substituted or unsubstituted alkyl.
  • the compound is according to formula XIa, XIb, XIc, XId, XIe or XIf:
  • XId XIe XIf and R , 8b is hydrogen, substituted or unsubstituted alkyl or substituted or unsubstituted cycloalkyl;
  • R 9e is hydrogen, Me, or CN.
  • R 9e is H.
  • R 9e is Me.
  • R 9e is CN.
  • R 8b is H.
  • R 8b is cycloalkyl.
  • R 8b is cyclopropyl.
  • R 8b is substituted or unsubstituted alkyl.
  • the compound is according to formula XIIa, XIIb, XIIc or XIId:
  • the compound is according to formula XIIIa, XIIIb, XIIIc or XIIId:
  • the compound is according to formula XIVa, XIVb, XIVc or XIVd:
  • the compound is according to formula XVa, XVb, or XIVc:
  • R 8b is H, Me, i-Pr, t-Bu, CH 2 CONH 2 , cyclopropylmethyl, or CH 2 CF 3 .
  • L is a bond. In another particular embodiment, L is -0-CH 2 -CH 2 - .
  • the present invention provides prodrugs and de ⁇ vatives of the compounds according to the formulae above
  • Prodrugs are derivatives of the compounds of the invention, which have metabolically cleavable groups and become by solvolysis or under physiological conditions the compounds of the invention, which are pharmaceutically active, in vivo
  • a prodrug may be inactive when administered to a subject but is converted in vivo to an active compound of the invention.
  • “Pharmaceutically acceptable prodrugs” as used herein refers to those prodrugs of the compounds useful in the present invention, which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of patients with undue toxicity, irritation, allergic response commensurate with a reasonable benefit/risk ratio, and effective for their intended use of the compounds of the invention.
  • the term “prodrug” means a compound that is transformed in vivo to yield an effective compound useful in the present invention or a pharmaceutically acceptable salt, hydrate or solvate thereof. The transformation may occur by various mechanisms, such as through hydrolysis in blood.
  • the compounds bearing metabolically cleavable groups have the advantage that they may exhibit improved bioavailability as a result of enhanced solubility and/or rate of absorption conferred upon the parent compound by virtue of the presence of the metabolically cleavable group, thus, such compounds act as pro-drugs.
  • a thorough discussion is provided in Design of Prodrugs, H. Bundgaard, ed., Elsevier (1985); Methods in Enzymology; K. Widder et al, Ed., Academic Press, 42, 309-396 (1985); A Textbook of Drug Design and Development, Krogsgaard- Larsen and H. Bandaged, ed., Chapter 5; "Design and Applications of Prodrugs” 113-191 (1991); Advanced Drug Delivery Reviews, H.
  • Such examples include, but are not limited to, choline ester derivatives and the like, N-alkylmorpholine esters and the like.
  • Prodrugs include acid derivatives well know to practitioners of the art, such as, for example, esters prepared by reaction of the parent acid with a suitable alcohol, or amides prepared by reaction of the parent acid compound with a substituted or unsubstituted amine, or acid anhydrides, or mixed anhydrides.
  • Simple aliphatic or aromatic esters, amides and anhydrides derived from acidic groups pendant on the compounds of this invention are preferred prodrugs.
  • double ester type prodrugs such as (acyloxy)alkyl esters or ((alkoxycarbonyl)oxy)alkylesters.
  • Preferred are the Ci to Cg alkyl, C 2 -Cg alkenyl, aryl, C7-C 12 substituted aryl, and C7-C 12 arylalkyl esters of the compounds of the invention.
  • the compounds of this invention are typically administered in the form of a pharmaceutical composition.
  • Such compositions can be prepared in a manner well known in the pharmaceutical art and comprise at least one active compound.
  • the compounds of this invention are administered in a pharmaceutically effective amount. The amount of the compound actually administered will typically be determined by a physician, in the light of the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual compound -administered, the age, weight, and response of the individual patient, the severity of the patient's symptoms, and the like.
  • compositions of this invention can be administered by a variety of routes including oral, rectal, transdermal, subcutaneous, intravenous, intramuscular, and intranasal.
  • routes including oral, rectal, transdermal, subcutaneous, intravenous, intramuscular, and intranasal.
  • the compounds of this invention are preferably formulated as either injectable or oral compositions or as salves, as lotions or as patches all for transdermal administration.
  • compositions for oral administration can take the form of bulk liquid solutions or suspensions, or bulk powders. More commonly, however, the compositions are presented in unit dosage forms to facilitate accurate dosing.
  • unit dosage forms refers to physically discrete units suitable as unitary dosages for human subjects and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient.
  • Typical unit dosage forms include prefilled, premeasured ampules or syringes of the liquid compositions or pills, tablets, capsules or the like in the case of solid compositions.
  • the furansulfonic acid compound is usually a minor component (from about 0.1 to about 50% by weight or preferably from about 1 to about 40% by weight) with the remainder being various vehicles or carriers and processing aids helpful for forming the desired dosing form.
  • Liquid forms suitable for oral administration may include a suitable aqueous or nonaqueous vehicle with buffers, suspending and dispensing agents, colorants, flavors and the like.
  • Solid forms may include, for example, any of the following ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or corn starch; a lubricant such as magnesium stearate; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring.
  • a binder such as microcrystalline cellulose, gum tragacanth or gelatin
  • an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or corn starch
  • Injectable compositions are typically based upon injectable sterile saline or phosphate- buffered saline or other injectable carriers known in the art.
  • the active compound in such compositions is typically a minor component, often being from about 0.05 to 10% by weight with the remainder being the injectable carrier and the like.
  • Transdermal compositions are typically formulated as a topical ointment or cream containing the active ingredient(s), generally in an amount ranging from about 0.01 to about 20% by weight, preferably from about 0.1 to about 20% by weight, preferably from about 0.1 to about 10% by weight, and more preferably from about 0.5 to about 15% by weight.
  • the active ingredients When formulated as a ointment, the active ingredients will typically be combined with either a paraffmic or a water-miscible ointment base. Alternatively, the active ingredients may be formulated in a cream with, for example an oil-in- water cream base.
  • Such transdermal formulations are well-known in the art and generally include additional ingredients to enhance the dermal penetration of stability of the active ingredients or the formulation. All such known transdermal formulations and ingredients are included within the scope of this invention.
  • the compounds of this invention can also be administered by a transdermal device.
  • transdermal administration can be accomplished using a patch either of the reservoir or porous membrane type, or of a solid matrix variety.
  • the compounds of this invention can also be administered in sustained release forms or from sustained release drug delivery systems.
  • sustained release materials can be found in Remington's Pharmaceutical Sciences.
  • a compound of the invention is admixed as a dry powder with a dry gelatin binder in an approximate 1:2 weight ratio.
  • a minor amount of magnesium stearate is added as a lubricant.
  • the mixture is formed into 240-270 mg tablets (80-90 mg of active amide compound per tablet) in a tablet press.
  • a compound of the invention is admixed as a dry powder with a starch diluent in an approximate 1 :1 weight ratio. The mixture is filled into 250 mg capsules (125 mg of active amide compound per capsule).
  • a compound of the invention (125 mg), sucrose (1.75 g) and xanthan gum (4 mg) are blended, passed through a No. 10 mesh U.S. sieve, and then mixed with a previously made solution of microcrystalline cellulose and sodium carboxymethyl cellulose (11 :89, 50 mg) in water.
  • Sodium benzoate (10 mg) flavor, and color are diluted with water and added with stirring. Sufficient water is then added to produce a total volume of 5 mL.
  • a compound of the invention is admixed as a dry powder with a dry gelatin binder in an approximate 1 :2 weight ratio.
  • a minor amount of magnesium stearate is added as a lubricant.
  • the mixture is formed into 450-900 mg tablets (150-300 mg of active amide compound) in a tablet press.
  • a compound of the invention is dissolved or suspended in a buffered sterile saline injectable aqueous medium to a concentration of approximately 5 mg/ml.
  • Stearyl alcohol (250 g) and a white petrolatum (250 g) are melted at about 75°C and then a mixture of a compound of the invention (50 g) methylparaben (0.25 g), propylparaben (0.15 g), sodium lauryl sulfate (10 g), and propylene glycol (120 g) dissolved in water (about 370 g) is added and the resulting mixture is stirred until it congeals.
  • the present compounds are used as therapeutic agents for the treatment of conditions in mammals that are causally related or attributable to aberrant activity of MMPl and / or MAPKAPK5. Accordingly, the compounds and pharmaceutical compositions of this invention find use as therapeutics for preventing and/or treating inflammatory diseases in mammals including humans.
  • this invention provides a method of treating a mammal susceptible to or afflicted with a condition associated with extra-cellular matrix (ECM) degradation, in particular arthritis, and more particularly, rheumatoid arthritis, which method comprises administering an effective amount of one or more of the compounds of the invention or a pharmaceutical composition just described.
  • ECM extra-cellular matrix
  • the invention provides a method of treating a mammal sucepible to or afflicted with a condition associated with an abnormal cellular expression of MMPl, which comprises administering a therapeutically effective amount of a compound of the invention, or a pharmaceutical composition thereof.
  • the present invention provides a method of treatment or prophylaxis of a condition characterized by abnormal matrix metallo proteinase activity, which comprises administering a therapeutically effective matrix metallo proteinase inhibiting amount of one or more of the compounds of the invention, or pharmaceutical composition thereof.
  • this invention provides methods of treating a mammal susceptible to or afflicted with diseases and disorders which are mediated by or result in inflammation such as, for example rheumatoid arthritis and osteoarthritis, myocardial infarction, various autoimmune diseases and disorders, uveitis and atherosclerosis; itch / pruritus such as, for example psoriasis; and renal disorders method comprises administering an effective condition-treating or condition-preventing amount of one or more of the pharmaceutical compositions just described.
  • This invention also relates to the use of the present compounds in the manufacture of a medicament for treatment or prophylaxis of a condition prevented, ameliorated or eliminated by administration of an inhibitor of Mitogen- Activated Protein Kinase- Activated Protein Kinase 5, or a condition characterised by abnormal collagenase activity, or a condition associated with ECM degradation or a condition selected from diseases involving inflammation, most preferably in for the treatment of rheumatoid arthritis.
  • a preferred regimen of the present method comprises the administration to a subject in suffering from a disease condition characterized by inflammatory, with an effective matrix metallo- protease inhibiting amount of a compound of the present invention for a period of time sufficient to reduce the abnormal levels of extracellular matrix degradation in the patient, and preferably terminate, the self-perpetuating processes responsible for said degradation.
  • a special embodiment of the method comprises administering of an effective matrix metallo-protease inhibiting amount of a compound of the present invention to a subject patient suffering from or susceptible to the development of rheumatoid arthritis, for a period of time sufficient to reduce or prevent, respectively, collagen and bone degradation in the joints of said patient, and preferably terminate, the self-perpetuating processes responsible for said degradation.
  • Injection dose levels range from about 0.1 mg/kg/hour to at least 10 mg/kg/hour, all for from about 1 to about 120 hours and especially 24 to 96 hours.
  • a preloading bolus of from about 0.1 mg/kg to about 10 mg/kg or more may also be administered to achieve adequate steady state levels.
  • the maximum total dose is not expected to exceed about 2 g/day for a 40 to 80 kg human patient.
  • each dose provides from about 0.01 to about 20 mg/kg of the compound of the invention, with preferred doses each providing from about 0.1 to about 10 mg/kg and especially about 1 to about 5 mg/kg.
  • Transdermal doses are generally selected to provide similar or lower blood levels than are achieved using injection doses.
  • the compounds of this invention When used to prevent the onset of an inflammatory condition, the compounds of this invention will be administered to a patient at risk for developing the condition, typically on the advice and under the supervision of a physician, at the dosage levels described above.
  • Patients at risk for developing a particular condition generally include those that have a family history of the condition, or those who have been identified by genetic testing or screening to be particularly susceptible to developing the condition.
  • the compounds of this invention can be administered as the sole active agent or they can be administered in combination with other agents, including other compounds that demonstrate the same or a similar therapeutic activity, and that are determined to safe and efficacious for such combined administration.
  • the triazolo[l,5-a]pyridyl compounds of this invention can be prepared from readily available starting materials using the following general methods and procedures. It will be appreciated that where typical or preferred process conditions (i.e., reaction temperatures, times, mole ratios of reactants, solvents, pressures, etc.) are given; other process conditions can also be used unless otherwise stated. Optimum reaction conditions may vary with the particular reactants or solvent used, but such conditions can be determined by one skilled in the art by routine optimization procedures. [00243] Additionally, as will be apparent to those skilled in the art, conventional protecting groups may be necessary to prevent certain functional groups from undergoing undesired reactions.
  • Preparative HPLC Waters XBridge Prep C18 5 ⁇ m ODB 19mm ID x 100mm L (Part No.186002978). All the methods are using MeCN/H2O gradients. H2O contains either 0.1% TFA or 0.1% NH3.
  • Step 3 5,8-Dibromo-[l ,2,4Jtriazolo[l ,5-aJpyrazine
  • N-(3,6-dibromo-pyrazin-2-yl)-jV-hydroxyformamidine (17.4 mg, 58.80 mmol) is treated with polyphosphoric acid (150 g) for one hour at 5O 0 C and then for 1.75 hours at 7O 0 C. After cooling to room temperature, water is added to the reaction mixture. The resultant suspension is brought to pH 8 by careful addition of solid NaHC ⁇ 3 in small portions. The precipitate formed is collected by filtration, washed once with IN NaOH, three times with water and dried in vacuo. The residue is partitioned between ethyl acetate and IN NaOH and the organic phase is washed one more time with IN NaOH and once with brine.
  • Step 4 (5-Bromo-[l,2,4Jtriazolo[l,5-aJpyrazin-8-yl)-(4-morpholin-4-ylphenyl)amine
  • Step 5 (4-Morpholin-4-ylphenyl)-[5-(lH-pyrazol-4-yl)-[l,2,4Jtriazolo[l,5-aJpyrazin-8-ylJamine
  • Step 1 [5-(2-Methoxy-pyridin-4-yl)-[l,2,4Jtriazolo[l,5-aJpyrazin-8-ylJ-[4-(4-methyl-piperazin-l-yl)- phenyl] -amine
  • This compound may be prepared using methods as described for Compound 6, step 4 using (5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-[4-(4-methyl-piperazin-l-yl)-phenyl]-amine
  • Step 2 4-(8-(4-(4-Methylpiperazin-l-yl)phenylamino)-[l,2, 4]triazolo[l, 5a]pyrazin-5-yl)pyridin- 2(lH)-one
  • Step 1 (5-Bromo-[l,2,4Jtriazolo[l,5-aJpyrazin-8-yl)-[4-(4-methyl-piperazin-l-yl)-phenylJ-amine
  • Step 3 4-(4,4,5,5-Tetramethyl-[l,3,2]dioxaborolan-2-yl)-thiophene-2-carboxylic acid amide
  • Step 4 4- ⁇ 8- [4-(4-Methyl-piperazin- 1 -yl)-phenylamino] -[ 1,2,4] triazolo[l,5-a]pyrazin-5-yl ⁇ - thiophene-2-carboxylic acid amide
  • the tube is sealed and the reaction mixture is heated at 9O 0 C overnight. After cooling to room temperature the reaction mixture is partitioned between ethyl acetate and water. The precipitate is collected by filtration and washed with water (Ix) and ether (2x) and dried in vacuo. The crude product is purified by column chromatography (silica gel, DCM/MeOH/NH 3 96:4) yielding the title compound (43 mg) as a yellow solid.
  • Step 2 5-(4,4,5,5-Tetramethyl-[l,3,2Jdioxaborolan-2-yl)-thiophene-2-carboxylic acid amide
  • Step 3 5- ⁇ 8-[4-(4-Methyl-piperazin-l-yl)-phenylamino] -[ 1,2,4] triazolo[l,5-a]pyrazin-5-yl ⁇ - thiophene-2-carboxylic acid amide
  • This compound may be prepared using methods as described for Compound 6, step 4 using 5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-[4-(4-methyl-piperazin-l-yl)-phenyl]-amine
  • This compound may be prepared using methods as described for Compound 6, step 4 using (5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-(4-morpholin-4-yl-phenyl)-amine (0.2g,
  • This compound may be prepared using methods as described for Compound 6, step 4 using (5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-(4-morpholin-4-yl-phenyl)-amine (0.2g,
  • Step 1 5-Methyl-4-(4, 4, 5, 5-tetramethyl-[l , 3, 2]dioxaborolan-2-yl)-lH-pyrazole
  • This compound may be prepared using methods as described for Compound 86, step 2 using 4-bromo-5-methyl-lH-pyrazole (3g, 18.6mmol), bis(pinacolato)diboron (8.52g, 33.5mmol), PdCl 2 dppf (913mg, l.l l ⁇ mmol) and KOAc (5.49mg, 55.9mmol) in dimethylsulfoxide (3OmL).
  • reaction mixture is purified by silica gel column chromatography eluting with 7:3 followed by 1 :1 petroleum ether: ethyl acetate to afford 5-methyl-4-(4,4,5,5-tetramethyl-[l,3,2]dioxaborolan-2-yl)-lH- pyrazole (3.87g, 100%).
  • Step 2 5-(5-Methyl-lH-pyrazol-4-yl)-N-(4-(4-methylpiperazin-l-yl)phenyl)-[l,2,4]triazolo[l,5- aJpyrazin-8-amine
  • This compound may be prepared using methods as described for Compound 6, step 4 using (5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-[4-(4-methyl-piperazin-l-yl)-phenyl]-amine
  • Step 1 (5-Bromo-[l,2,4Jtriazolo[l,5-aJpyrazin-8-yl)-(3- ⁇ uoro-4-morpholin-4-yl-phenyl)-amine
  • This compound may be prepared using methods as described for Compound 6, step 1 using 5,8-dibromo-[l,2,4]triazolo[l,5-a]pyrazine (0.5g, 1.799mmol), 3-fluoro-4-morpholin-4-yl- phenylamine (0.53g, 2.70mmol), and DIPEA (0.47OmL, 2.70mmol) in 2-propanol (6mL).
  • the reaction mixture is partitioned between 10% citric acid aqueous solution and DCM.
  • the organic phase is separated and washed with water and brine, dried over MgSO 4 , filtered and concentrated under vacuum to afford the title compound (697mg, 98%) which is used in the next step without further purification.
  • Step 2 N-(3-Fluoro-4-morpholinophenyl)-5-(lH-pyrazol-4-yl)-[ 1 ,2,4] triazolo [ 1 ,5-a] pyrazin-8-amine
  • This compound may be prepared using methods as described for Compound 6, step 4, using (5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-(3-fluoro-4-morpholin-4-yl-phenyl)-amine (lOOmg, 0.254mmol), 4-(4,4,5,5-tetramethyl-[l,3,2]dioxaborolan-2-yl)-lH-pyrazole (99mg, 0.51mmol) and Pd(PPh 3 ) 4 (73mg, 0.063mmol) in 1.5M Na 2 CO 3 (1.36mL, 2.03mmol) and dioxane (4mL).
  • Step 1 5-(5-Bromo-[l, 2, 4] triazolo [1, 5-a]pyrazin-8-ylamino)-2-morpholin-4-yl-benzamide
  • This compound may be prepared using methods as described for Compound 6, step 1 using 5,8-dibromo-[l,2,4]triazolo[l,5-a]pyrazine (250mg, 0.90mmol) 5-amino-2-morpholin-4-yl benzamide (299mg, 1.35mmol) and N,N-diisopropylethylamine (0.24mL, 1.35mmol) in 2-propanol (7mL). Trituration with 'PrOH and Et 2 O affords the title compound (273mg, 73%).
  • This compound may be prepared using methods as described for Compound 6, step 4 using 5-(5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-ylamino)-2-morpholin-4-yl-benzamide (140mg, 0.33mmol), 4-(4,4,5,5-tetramethyl-[l,3,2]dioxaborolan-2-yl)-lH-pyrazole (130mg, 0.67mmol) and Pd(PPh 3 ) 4 (96mg, 0.083mmol) in 1.5M K 2 CO 3 (aq) (1.93mL) and dioxane (3.44mL).
  • This compound may be prepared using methods as described for Compound 6, step 4, using (5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-[4-(2-morpholin-4-yl-ethoxy)-phenyl]-amine
  • This compound may be prepared using methods as described for Compound 6, step 4, using (5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-[4-(2-morpholin-4-yl-ethoxy)-phenyl]-amine
  • Step 1 l-(Tetrahydro-pyran-2-yl)-lH-pyrazole
  • Step 3 (4-Morpholin-4-yl-phenyl)-[5-(2H-pyrazol-3-yl)-[l,2,4]triazolo[l,5-a]pyrazin-8-yl]-amine
  • This compound may be prepared using methods as described for Compound 6, step 4, using 5-bromo- [l,2,4]triazolo[l,5-a]pyrazin-8-yl)-[4-(morpholin-4-yl)-phenyl]-amine (lOOmg, 0.267mmol), IH- pyrazole-2-boronic acid (60mg, 0.535mmol), Pd(PPh 3 ) 4 (93mg, 0.08mmol) and Na 2 CO 3 (88mg, 0.80mmol) in DMF (2mL). The reaction mixture is placed in a stem-tube and stirred at 100 0 C for 18 hours. After cooling, the mixture is diluted with NaHCO 3 solution and extracted with EtOAc (4x).
  • This compound may be prepared using methods as described for Compound 6, step 4, using 5-bromo-N-(6-(4-isopropylpiperazin-l-yl)pyridin-3-yl)-[l,2,4]triazolo[l,5-a]pyrazin-8-amine (lOOmg, 0.24mmol), 4-(4,4,5,5-tetramethyl-[l,3,2]dioxaborolan-2-yl)-thiophene-2-carboxamide (121mg, 0.48mmol), and Pd(PPh 3 ) 4 (69mg, 0.059mmol) in 1.5M Na 2 CO 3 (1.28mL, 1.92mmol) and dioxane (3.84mL).
  • Step 1 [5-(4-Carbamoyl-furan-2-yl)-[l,2,4]triazolo[l,5-a]pyrazin-8-yl]-(4-morpholin-4-yl-phenyl)- carbamic acid tert-butyl ester
  • This compound may be prepared using methods as described for Compound 35, step 3 using (4-morpholin-4-yl-phenyl)-(5-tributylstannanyl-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-carbamic acid tert-butyl ester (137mg, 0.199mmol), 5-bromo-furan-3-carboxamide (76mg, 0.4mmol) and Pd(PPh 3 ) 4 (23mg, 0.020mmol) in DMF (ImL).
  • This compound may be prepared using methods as described for Compound 6, step 4 using (5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-(4-morpholin-4-yl-phenyl)-amine (1-Og,
  • This compound may be prepared using methods as described for Compound 6, step 4, using (5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-(4-morpholin-4-yl-phenyl)-amine (78mg, O.l ⁇ mmol), 4-(4,4,5,5-tetramethyl-[l,3,2]dioxaborolan-2-yl)-furan-2-carboxylic acid amide (lOOmg, 0.34mmol), and Pd(PPh 3 ) 4 (60mg, 0.052mmol) in 1.5M Na 2 CO 3 (aq) (1.ImL, 1.68mmol), and dioxane (3mL).
  • Step 1 (5-Bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-(4-morpholin-4-yl-phenyl)-carbamic acid tert- butyl ester
  • Step 3 (4-Morpholin-4-yl-phenyl)-[5-(l-oxo-l, 2, 3, 4-tetrahydro-isoquinolin-6-yl)-[ 1 , 2, 4]triazolo[l, 5- aJpyrazin-8-ylJ -carbamic acid tert-butyl ester
  • Step 4 6-[8-(4-Morpholin-4-yl-phenylamino)-[ 1 ,2,4] triazolo [ 1 ,5-a] pyrazin-5-yl] -3 ,4-dihydro-2H- isoquinolin-1-one
  • This compound may be prepared using methods as described for Compound 6, step 4 using 5-bromo-N-(6-(4-isopropylpiperazin-l-yl)pyridin-3-yl)-[l,2,4]triazolo[l,5-a]pyrazin-8-amine (0.6g, 1.44mmol), 5-(4,4,5,5-tetramethyl-[l,3,2]dioxaborolan-2-yl)-2,3-dihydro-isoindol-l-one (0.56g, 2.16mmol) and Pd(PPh 3 ) 4 (0.416g, 0.36mmol) in 1.5M Na 2 CO 3 (7.7mL) and dioxane (23mL).
  • Step 1 (2-Morpholin-4-yl-5-nitro-phenyl)-methanol r .OH r / - ⁇ °
  • Step 4 (5-Bromo-[ 1,2,4] triazolo[l,5-a] pyrazin-8-yl)-[3-(tert-butyl-dimethyl-silanyloxymethyl)-4- morpholin-4-yl-phenyl] -amine
  • Step 5 [5-(5-Bromo-[ 1,2, 4] triazolo[l,5-a]pyrazin-8-ylamino)-2-morpholin-4-yl-phenyl] -methanol
  • Step 6 ⁇ 5-[5-(lH-Pyrazol-4-yl)-[ 1 ,2 ,4] triazolo [ 1 ,5-a] pyrazin-8-ylamino] -2- morpholinophenyljmethanol
  • This compound may be prepared using methods as described for Compound 6, step 4 using [5-(5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-ylamino)-2-morpholin-4-yl-phenyl]-methanol (85mg, 0.21mmol), 4-(4,4,5,5-tetramethyl-[l,3,2]dioxa-borolan-2-yl)-lH-pyrazole (81mg, 0.42mmol) and Pd(PPh 3 ) 4 (60mg, 0.052mmol) in 1.5M Na 2 CO 3 (1.12mL) and dioxane (2.OmL).
  • reaction mixture is purified by silica gel column chromatography, eluting with 95:3 DCM:NH 3 (7M in MeOH) followed by reverse phase preparative HPLC purification.
  • the title compound is obtained as a pale brown solid (lOmg, 12%).
  • This compound may be prepared using the methods as described for Compound 120, using (5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-(6-morpholin-4-yl-pyridin-3-yl)amine in Step 4.
  • LCMS: Rt 0.80 min (95 %), m/z (ESI) 364 (M+H) + .
  • This compound may be prepared using the methods described for Compound 120, using (5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-[6-(4-[cyclopropylmethyl]piperazin-l-yl)pyridin-3- yl]amine in step 4.
  • LCMS: Rt 0.80 min (95 %), m/z (ESI) 417 (M+H) + .
  • Compound 43 [6-(4-Isopropylpiperazin-l-yl)pyridin-3-yl]-[5-(lH-pyrazol-4-yl)-
  • This compound may be prepared using methods as described for Compound 120, using (5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-[6-(4-isopropylpiperazin-l-yl)pyridin-3-yl]amine in Step 4.
  • LCMS: Rt 0.77 min (95 %), m/z (ESI) 405 (M+H) + .
  • This compound may be prepared using methods as described for Compound 120, using (5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)- ⁇ 6-[4-(2,2,2-trifluoroethyl)piperazin-l-yl]-pyridin- 3-yl ⁇ amine in step 4.
  • LCMS: Rt 0.95 min (95 %), m/z (ESI) 445 (M+H) + .
  • This compound may be prepared using methods as described for Compound 120, Step
  • This compound may be prepared using methods as described for Compound 167, using (5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-(6-morpholin-4-yl-pyridin-3-yl)amine in Step 4.
  • LCMS: Rt 0.85 min (95 %), m/z (ESI) 423 (M+H) + .
  • This compound may be prepared using methods as described for Compound 120, using (5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-(4-morpholin-4-ylphenyl)amine and 4,4,5,5- tetramethyl-2-benzo[b]thiophen-3-yl-[l,3,2]dioxaborolane in step 4.
  • LCMS Rt 0.84 min (95 %), m/z (ESI) 402 (M+H) + .
  • This compound may be prepared using methods as described for Compound 120, using (5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-(4-morpholin-4-ylphenyl)amine and 4,4,5,5- tetramethyl-2-thiophen-3-yl-[l,3,2]dioxaborolane in step 4.
  • LCMS Rt 1.19 min (95 %), m/z (ESI) 379 (M+H) + .
  • This compound may be prepared using the methods as described for Compound 50, using (5-bromo[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-[4-(4-isopropylpiperazin-l-yl)-phenyl]amine in the final step.
  • LCMS: rt 1.11 min (95 %), m/z (ESI) 420 (M+H) + .
  • Step 1 3 -Ethyl- lH-pyr azote N-N H
  • Step 3 4-Bromo-5-ethyl-l-(tetrahydro-pyran-2-yl)-lH-pyrazole and 4-Bromo-3 -ethyl- l-(tetrahydro- pyran-2-yl) - lH-pyr azole
  • Step 4 5-Ethyl-l-(tetrahydro-pyran-2-yl)-4-(4, 4, 5, 5-tetramethyl-[l, 3, 2] dioxaborolan-2-yl)-lH- pyrazote and 3-Ethyl-l-(tetrahydro-pyran-2-yl)-4-(4, 4, 5, 5-tetramethyl-fl, 3, 2] dioxaborolan-2-yl)-lH- pyr azote
  • This compound may be prepared using methods as described for Compound 86, step 2 using 4-bromo-5-ethyl-l-(tetrahydro-pyran-2-yl)-lH-pyrazole and 4-bromo-3-ethyl-l-(tetrahydro- pyran-2-yl)-lH-pyrazole (168mg, 0.65mmol), bis(pinacolato)diboron (331mg, 1.3mmol), PdC ⁇ dppf (53mg, 65 ⁇ mol) and KOAc (190mg, 1.95mmol) in dimethylsulfoxide (2mL).
  • the reaction mixture is purified by silica gel column chromatography eluting with 90:10 petroleum ether: ethyl acetate to afford the title compounds ( ⁇ l.lmg, 31%).
  • Step 5 ⁇ 5-[5-Ethyl-l-(tetrahydro-pyran-2-yl)-lH-pyrazol-4-yl]-[l,2,4]triazolo[l,5-a]pyrazin-8-yl ⁇ - (4-morpholin-4-yl-phenyl) -amine and ⁇ 5-[3-Ethyl-l-(tetrahydro-pyran-2-yl)-lH-pyrazol-4-yl]- [l,2,4]triazolo[l,5-a]pyrazin-8-yl ⁇ -(4-morpholin-4-yl-phenyl)-amine
  • This compound may be prepared using methods as described for Compound 6, step 4 using (5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-(4-morpholin-4-yl-phenyl)-amine (0.032g, 0.085mmol), 5-ethyl-l-(tetrahydro-pyran-2-yl)-4-(4,4,5,5-tetramethyl-[l,3,2]dioxaborolan-2-yl)-lH- pyrazole and 3-ethyl-l-(tetrahydro-pyran-2-yl)-4-(4,4,5,5-tetramethyl-[l,3,2]dioxaborolan-2-yl)-lH- pyrazole (0.52mg, 0.17mmol), and Pd(PPh 3 ) 4 (0.025mg, 0.021mmol) in 1.5M Na 2 CO 3 (aq) (0.45mL, O. ⁇
  • Step 6 5-(5-Ethyl-lH-pyrazol-4-yl)-N-(4-morpholinophenyl)-[l,2,4]triazolo[l,5-a]pyrazin-8-amine
  • Step 1 l,l-Dioxo-6-(4,4,5,5-tetramethyl-[l,3,2]dioxaborolan-2-yl)-l,2-dihydro-l ⁇ - benzofdJisothiazol-3-one
  • This compound may be prepared using methods as described for Compound 86, step 2 using 6-bromo-l,l-dioxo-l,2-dihydro-l ⁇ 6 -benzo[d]isothiazol-3-one (0.5, 1.9mmol), bis(pinacolato)diboron (0.53g, 2.1mmol), PdCl 2 dppf (0.047g, 0.058mmol) and KOAc (0.56g, 5.7mmol) in dioxane (1OmL). The solvent is removed in vacuo and the residue is partitioned between DCM and water. The organic layer is washed with sat NaHC ⁇ 3 and 2M HCl, dried over MgSOzi, filtered and evaporated to afford the title compound (990mg, 169%) used in the next step without further purification.
  • Step 2 (4-Morpholin-4-yl-phenyl)-[5-(l, 1, 3-trioxo-2, 3-dihydro-lH-l ⁇ 6 -benzo[d]isothiazol-6-yl)- [l,2,4]triazolo[l,5-a]pyrazin-8-yl]-carbamic acid tert-butyl ester
  • This compound may be prepared using methods as described for Compound 6, step 4 using (5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-(4-morpholin-4-yl-phenyl)-carbamic acid tert-butyl ester (170mg, 0.36mmol), l,l-dioxo-6-(4,4,5,5-tetramethyl-[l,3,2]dioxaborolan-2-yl)-l,2-dihydro-l ⁇ 6 -benzo[d]isothiazol-3-one (374mg, 0.72mmol) and Pd(PPh 3 ) 4 (lOOmg, 0.082mmol) in 1.5M Na 2 CO 3 (2mL, 3mmol) and dioxane (6mL).
  • Step 3 6-[8-(4-Morpholin-4-yl-phenylamino)-[l ,2,4Jtriazolo[l ,5-aJpyrazin-5-ylJ-l ,1-dioxo-l ,2- dihydro-1 ⁇ 6 -benzo[d]isothiazol-3-one
  • This compound may be prepared using the methods described for Compound 120, using (5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-[6-(4-[cyclopropylmethyl]piperazin-l-yl)pyridin-3- yl]amine in step 4.
  • LCMS: Rt 0.85 min (95 %), m/z (ESI) 476 (M+H) + .
  • This compound may be prepared using the methods as described for Compound 167, using (5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-[6-(4-isopropylpiperazin-l-yl)pyridin-3-yl]amine in Step 4.
  • LCMS: Rt 0.82 min (95 %), m/z (ESI) 464 (M+H) + .
  • This compound may be prepared using the methods as described for Compound 167, using (5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)- ⁇ 6-[4-(2,2,2-trifluoroethyl)piperazin-l-yl]-pyridin- 3-yl ⁇ amine in step 4.
  • LCMS: Rt 0.99 min (95 %), m/z (ESI) 504 (M+H) + .
  • This compound may be prepared using methods as described for Compound 58, using
  • This compound may be prepared using methods as described for Compound 46, using
  • This compound may be prepared using methods as described for Compound 46, using
  • This compound may be prepared using the methods described for Compound 120, using (5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-[6-(4-cyclopropylpiperazin-l-yl)pyridin-3- yl]amine in step 4.
  • LCMS: Rt 0.76 min (95 %), m/z (ESI) 403 (M+H) + .
  • This compound may be prepared using methods as described for Compound 73, using ammonia (7 M in MeOH) is used in step 3.
  • LCMS: Rt 1.00 min (95 %), m/z (ESI) 423 (M+H) + .
  • This compound may be prepared using methods as described for Compound 61, using
  • Step 1 l-Trifluoroacetyl-4-(4-nitrophenyl)piperidine
  • Triethlyamine (1.0 mL, 7.3 mmol) and 4-(4-nitrophenyl)piperidine (1.0 g, 4.8 mmol) are stirred in DCM (25 mL) at 0°C under N 2 and trifluoroacetic anhydride (0.81 mL, 5.8 mmol) is added. The mixture is stirred for three days, allowing the temperature to warm to rt. The solution is then diluted with DCM (50 mL) and washed with water (2 x 15 mL), NaHCO 3 (50% sat. aq., 2 x 15 mL) and brine (15 mL). The solvent is dried over MgS ⁇ 4 and evaporated to afford the desired compound (1.46 g, 4.66 mmol).
  • Step 2 l-(2,2,2-Trifluoroethyl)-4-(4-nitrophenyl)piperidine
  • Step 3 4-fl - (2, 2, 2- Trifluoroethyl)piperidin-4-yl]phenylam ine
  • Step 1 (5-Bromo-[ 1, 2,4] triazolo[l,5-a]pyrazin-8-yl)-[4-(2-morpholin-4-yl-ethoxy)-phenyl] -amine
  • This compound may be prepared using the methods as described for Compound 6, step 4, using (5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-[4-(2-morpholin-4-yl-ethoxy)-phenyl]- amine (80mg, 0.19mmol), 5-(4,4,5,5-tetramethyl-[l,3,2]dioxaborolan-2-yl)-2,3-dihydro-isoindol-l- one (74mg, 0.29mmol), and Pd(PPh 3 ) 4 (55mg, 0.047mmol) in 1.5M Na 2 CO 3 (1.02mL, 1.53mmol) and dioxane (3mL).
  • reaction mixture is purified by silica gel column chromatography eluting with DCM and a 97:3 mixture DCM:NH 3 (7M in MeOH). After trituration using diethyl ether, the title compound is isolated as a solid (61.9mg, 69%). Conversion of the material into the mesylate salt, using IM methanesulfonic acid (0.134mL) in MeOH, affords the title compound (57.1mg).
  • Step 1 (5-Bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-(2-fluoro-4-morpholin-4-yl-phenyl)-amine
  • This compound may be prepared using methods as described for Compound 6, step 1, using 5,8-dibromo-[l,2,4]triazolo[l,5-a]pyrazine (0.105g, 0.89mmol), 2-fluoro-4-morpholin-4-yl- phenylamine (93mg, 0.474mmol), DIPEA (0.123mL, 0.706mmol) and l,4-diazabicyclo[2.2.2]octane (53mg, 0.472mmol) in 2-propanol (2mL).
  • Step 2 N-(2-Fluoro-4-morpholinophenyl)-5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5-a]pyrazin-8-amine
  • This compound may be prepared using methods as described for Compound 6, step 4, using (5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-(2-fluoro-4-morpholin-4-yl-phenyl)-amine (80mg, 0.203mmol), 4-(4,4,5,5-tetramethyl-[l,3,2]dioxaborolan-2-yl)-lH-pyrazole (79mg, 0.406mmol), and Pd(PPh 3 ) 4 (59mg, 0.051mmol) in 1.5N Na 2 CO 3 (1.09mL, 1.62mmol) and dioxane (3.25mL).
  • reaction mixture is purified by silica gel column chromatography eluting with 97:3 DCM:NH 3 (7M in MeOH). The fractions containing the product are combined and evaporated in vacuo to afford a solid which is triturated with diethyl ether and petroleum ether to yield the title compound (29mg, 38%).
  • Compound 70 (4-(8-(4-Morpholinophenylamino)-[l,2,4]triazolo[l,5-a]pyrazin-5-yl)-lH-pyrazol- 5-yl)methanol
  • Step 1 4-Bromo-5-(tert-butyl-dimethyl-silanyloxymethyl)-lH-pyrazole
  • Step 2 4-Bromo-3- (tert-butyl-dimethyl-silanyloxymethyl) -I- (tetrahydro-pyran-2-yl) - lH-pyrazole and 4-Bromo-5- (tert-butyl-dimethyl-silanyloxymethyl) -I- (tetrahydro-pyran-2-yl) - lH-pyr azole
  • Step 3 ⁇ 5- [5- (tert-Butyl-dimethyl-silanyloxymethyl) - 1 -(tetrahydro-pyran-2-yl) - lH-pyrazol-4-yl] - fl,2,4Jtriazolo[l,5-aJpyrazin-8-yl ⁇ -(4-morpholin-4-yl-phenyl)- carbamic acid tert-butyl ester and ⁇ 5- [3- (tert-Butyl-dimethyl-silanyloxymethyl) -I- (tetrahydro-pyran-2-yl) - lH-pyrazol-4-yl] - [l,2,4Jtriazolo[l,5-aJpyrazin-8-yl ⁇ -(4-morpholin-4-yl-phenyl)-carbamic acid tert-butyl ester
  • This compound may be prepared using the methods as described for Compound 35, step 3 using (4-morpholin-4-yl-phenyl)-(5-tributylstannanyl-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)- carbamic acid tert-butyl ester (210mg, 0.30mmol) 4-bromo-3-(tert-butyl-dimethyl-silanyloxymethyl)- 1 -(tetrahydro-pyran-2-yl)- 1 H-pyrazole and 4-bromo-5-(tert-butyl-dimethyl-silanyloxymethyl)- 1 - (tetrahydro-pyran-2-yl)- 1 H-pyrazole (170mg, 0.45mmol) and tetrakis(triphenylphosphine)palladium(0) (35mg, 0.030mmol) in DMF (4mL).
  • Step 4 4-[8-(4-Morpholin-4-yl-phenylamino)-[ 1,2,4] 'triazolofl,5-aj ' pyrazin-5-yl] -thiazole-2- carboxylic acid methylamide
  • Step 1 4-Bromo-2-bromomethyl-benzoic acid methyl ester
  • Step 3 5-(4, 4, 5, 5-Tetramethyl-fl, 3, 2] dioxaborolan-2-yl)-2, 3-dihydro-isoindol-l-one
  • Step 4 l-Isopropyl-4-(5-nitro-pyridin-2-yl)-piperazine
  • Step 6 5-Bromo-8-(6-(4-isopropylpiperazin-l-yl)pyridin-3-yl)-[l,2,4]triazolo[l,5-a]pyrazin-8-amine
  • This compound may be prepared using methods as described for Compound 6, step 1, using 5,8-dibromo-[l,2,4]triazolo[l,5-a]pyrazine (0.188g, 0.68mmol), 6-(4-isopropyl-piperazin-l-yl)- pyridin-3-ylamine (0.18Og, 0.816mmol) and N-ethyldiisopropyl-amine (0.2OmL, 1.02mmol) in 2- propanol (2mL). Purification of the crude material by silica gel column chromatography, using DCM followed by 95:5 DCM:MeOH, affords the title compound as a pale brown solid (260mg, 92%).
  • Step 5 5-(8-(6-(4-Isopropylpiperazin-l-yl)pyridin-3-ylamino)-[ 1,2,4] triazolo[l,5-a] pyrazin-5- yl)isoindolin- 1 -one
  • This compound may be prepared using methods as described for Compound 6, step 4, using 5-bromo-N-(6-(4-isopropylpiperazin-l-yl)pyridin-3-yl)-[l,2,4]triazolo[l,5-a]pyrazin-8-amine (50mg, 0.12mmol), 5-(4,4,5,5-tetramethyl-[l,3,2]dioxaborolan-2-yl)-2,3-dihydro-isoindol-l-one (56mg, 0.216mmol) and Pd(PPh 3 ) 4 (35mg, 0.03mmol) in 1.5M Na 2 CO 3 (0.64mL) and dioxane (2mL).
  • the crude product is purified by silica gel column chromatography eluting with 95:5 DCM:MeOH followed by 90:10 DCM:MeOH.
  • the title compound is obtained after trituration with a 10:1 mixture «-hexane:DCM (19mg, 34%).
  • Step 1 l-Bromo-2-(2-ethoxyvinylsulfanyl)benzene
  • Step 3 BenzofbJthiophene-7-carboxylic acid ethyl ester
  • Step 5 Benzo [b] thiophene-7 -carboxylic acid amide
  • the aqueous layer is then extracted with dichloromethane and the resulting organic layers are combined, washed with brine, dried over Na 2 SC ⁇ and evaporated.
  • the crude material is purified by LC using DCM as the eluent to afford the title compound as a white solid (300 mg, 41%).
  • Step 7 3-(4,4,5,5-Tetramethyl[l,3,2Jdioxaborolan-2-yl)benzo[bJthiophene-7-carboxylic acid amide
  • Step 8 3-[8-(4-Morpholin-4-yl-phenylamino)-[ 1 ,2,4] triazolo [ 1 ,5-a] pyrazin-5-yl] -benzo [b] thiophene- 7-carboxylic acid amide
  • This compound may be prepared using the methods as described for Compound 120,
  • This compound may be prepared using methods as described for Compound 80, using
  • Step 3 Acetic acid 4-(4,4,5,5-tetramethyl-[l,3,2Jdioxaborolan-2-yl)pyridin-2-ylmethyl ester
  • Step 4 Acetic acid 4- ⁇ 8-[4-(4-isopropyl-piperazin-l-yl)-phenylamino]-[l,2,4]triazolo[l,5-a]pyrazin- 5-yl ⁇ -pyridin-2-ylmethyl ester
  • Step 5 (4- ⁇ 8-[4-(4-Isopropylpiperazin-l-yl)phenylamino]-[l,2,4]triazolo[l,5-a]pyrazin-5-yl ⁇ pyridin- 2-yl)methanol
  • Acetic acid 4- ⁇ 8-[4-(4-isopropylpiperazin-l-yl)phenylamino]-[l,2,4]triazolo[l,5- a]pyrazin-5-yl ⁇ pyridin-2-ylmethyl ester is stirred at room temperature overnight in 10 mL of a 1.5 M solution of potassium carbonate in methanol. The pH is then brought to neutral by addition of 10% aqueous citric acid and the mixture is extracted with DCM. Purification by LC using 98/2 DCM/2M NH 3 in MeOH as the eluent provides the title compound as a yellow powder (40.3 mg, 12.5% over 2 steps).
  • This compound may be prepared using the methods as described for Compound 92, using pyrazole-4-boronic acid in step 4.
  • Step 2 4-(4,4,5 ,5-Tetramethyl-[ 1 ,3 ,2] dioxaborolan-2-yl)-furan-2-carboxylic acid amide
  • Step 3 4-(8-(6-(4-Isopropylpiperazin-l-yl)pyridin-3-ylamino)-[l,2,4]triazolo[l,5-a]pyrazin-5- yl)furan-2-carboxamide
  • This compound may be prepared using methods as described for Compound 6, step 4 using 5-bromo-8-(6-(4-isopropylpiperazin-l-yl)pyridin-3-yl)-[l,2,4]triazolo[l,5-a]pyrazin-8-amine (60mg, 0.144mmol), 4-(4,4,5,5-tetramethyl-[l,3,2]dioxaborolan-2-yl)-furan-2-carboxylic acid amide (59mg, 0.26mmol), and Pd(PPh 3 ) 4 (42mg, 0.036mmol) in 1.5M Na 2 CO 3 (0.8mL, 1.15mmol) and dioxane (2mL).
  • Step 1 l-Isopropyl-4-(4-nitro-phenyl)-piperazine
  • Step 2 4-(4-Isopropyl-piperazin-l-yl)-phenylamine NH,
  • Step 3 (5-Bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-[4-(4-isopropyl-piperazin-l-yl)-phenyl]-amine
  • This compound may be prepared using methods as described for Compound 6, step 1, using 5,8-dibromo-[l,2,4]triazolo[l,5-a]pyrazine (2g, 7.20mmol), 4-(4-isopropyl-piperazin-l-yl)- phenylamine (1.89g, 8.62mmol) and N,N-diisopropylethylamine (1.88mL, 10.8mmol) in 2-propanol (3OmL) are stirred at 95 0 C overnight. The title compound is isolated after trituration with diethyl ether and petroleum ether as a grey solid (2.59g,
  • Step 4 4-(8-(4-(4-Isopropylpiperazin-l-yl)phenylamino)-[l,2,4]triazolo[l,5-a]pyrazin-5-yl)furan-2- carboxamide
  • This compound may be prepared using methods as described for Compound 6, step 4, using 5-bromo-N-(6-(4-isopropylpiperazin-l-yl)pyridin-3-yl)-[l,2,4]triazolo[l,5-a]pyrazin-8-amine (80mg, 0.21mmol), 4-(4,4,5,5-tetramethyl-[l,3,2]dioxaborolan-2-yl)-furan-2-carboxylic acid amide (lOlmg, 0.42mmol), and Pd(PPh 3 ) 4 (62mg, 0.053mmol) in 1.5M Na 2 CO 3 (aq) (1.143mL, 1.71mmol), and dioxane (4mL).
  • This compound may be prepared using the methods as described for Compound 92, using 2-(aminocarbonyl)thiophene-4-boronic acid in step 4.
  • This compound may be prepared using the methods as described for Compound 83, using (5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-[6-(4-isopropylpiperazin-l-yl)pyridin-3-yl]amine in Step 4.
  • LCMS: Rt 0.71 min (95 %), m/z (ESI) 446 (M+H) + .
  • Step 1 l-Isopropyl-4-(4-nitrophenyl)piperidine
  • 2-iodopropane (240 ⁇ L, 2.4 mmol) are stirred in acetonitrile (3 mL) in a sealed tube at 120°C for 45 min. The mixture is cooled and the solvent removed under reduced pressure. The residue is partitioned between DCM (20 mL) and water (5 mL), the layers are separated and the DCM is washed with water (5 mL) and brine (5 mL) and dried over MgSO 4 . Evaporation of the solvent affords the title compound (300 mg) which is used without further purification.
  • Step 3 (5-Bromo-[ 1 ,2,4] triazolo [ 1 ,5-a] pyrazin-8-yl)-[4-(l-isopropylpiperidin-4-yl)phenyl] amine
  • N,N-Diisopropylethylamine 200 ⁇ L, 1.2 mmol is added to a mixture of 4-(l- isopropylpiperidin-4-yl)phenylamine (220 mg, 1.0 mmol) and 5,8-dibromo-[l,2,4]triazolo[l,5- a]pyrazine (280 mg, 1.0 mmol) in 1 PrOH (5 mL) and heated at reflux for 48 h. The mixture is cooled and the solvent evaporated under reduced pressure to afford an orange-brown solid. This is partitioned between DCM (50 mL) and water (20 mL) and the layers are separated.
  • the organic phase is washed with citric acid (10% aq., 3 x 25 mL).
  • the combined washings are extracted with DCM (25 mL) and then made basic by addition of NaHC ⁇ 3 (s).
  • the mixture is extracted with DCM (3 x 25 mL) and the combined extracts dried over MgS ⁇ 4 and evaporated.
  • the crude product was purified by chromatography on silica gel, eluting with 5% - 10% MeOH in DCM to afford the desired compound, contaminated with the starting aniline. This was recrystallised from MeOH to afford the pure title compound (110 mg).
  • This material may be used to prepare 5- ⁇ 8-[4-(l-isopropylpiperidin-4- yl)phenylamino]-[l,2,4]triazolo[l,5-a]pyrazin-5-yl ⁇ -2,3-dihydroisoindol-l-one in a fashion analogous to step 4 as described for Compound 79.
  • Step 1 4-(4-Amino-phenyl)-3-oxo-piperazine-l-carboxylic acid tert-butyl ester
  • Step 2 l-[4-(5-Bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-ylamino)-phenyl]-piperazin-2-one
  • This compound may be prepared using methods as described for Compound 6, step 1, using 5,8-dibromo-[l,2,4]triazolo[l,5-a]pyrazine (0.283g, l.OOmmol), 4-(4-amino-phenyl)-3-oxo- piperazine-1-carboxylic acid tert-butyl ester (0.30Og, l.OOmmol) and N-ethyldiisopropyl-amine (0.2OmL, 1.02mmol) in 2-propanol (ImL).
  • Step 3 l-(4-(5-(lH-Pyrazol-4-yl)-[l,2,4]triazolo[l,5-a]pyrazin-8-ylamino)phenyl)piperazin-2-one
  • This compound may be prepared using methods as described for Compound 1, step 5 using 1 -[4-(5-bromo-[l ,2,4]triazolo[l ,5-a]pyrazin-8-ylamino)-phenyl]-piperazin-2-one (70mg, 0.18mmol) 4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-lH-pyrazole (64mg, 0.33mmol), Pd(PPh 3 ) 4 (21mg, l ⁇ mol) and NaO'Bu (70mg, 0.72mmol) in 2mL of 3:1 DMF/water.
  • Step 1 l-tert-Butyl-4-(4-nitro-phenyl)-piperazine
  • Step 3 (5-Bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-[4-(4-tert-butyl-piperazin-l-yl)-phenyl]-amine
  • This compound may be prepared using methods as described for Compound 6, step 1, using 5,8-dibromo-[l,2,4]triazolo[l,5-a]pyrazine (0.308g, 1.107mmol), 4-(4-tert-butyl-piperazin-l- yl)-phenylamine (0.31Og, 1.33mmol) and N-ethyldiisopropyl-amine (0.289mL, 1.66mmol) in 2- propanol (5mL). The reaction mixture is partitioned between DCM and IN NaOH, the organic layer is separated and washed with water and brine, dried over MgSOzi, filtered and concentrated under vacuum. Trituration of the residue with diethyl ether and petroleum ether affords the title compound (440mg, 92%) as a cream solid. LCMS: Rt 2.25min (96%).
  • Step 4 5- ⁇ 8-[4-(4-tert-Butyl-piperazin-l-yl)-phenylamino]-[ 1,2,4] triazolo[l,5-a]pyrazin-5-yl ⁇ -2,3- dihydro-isoindol-1-one
  • This compound may be prepared using methods as described for Compound 6, step 4,
  • reaction mixture is partitioned between ethyl acetate and brine, the organic layer is separated, dried over MgSO 4 filtered and evaporated in vacuo.
  • the residue is purified by silica gel column chromatography eluting with 98:2 DCM:NH 3 (7M in MeOH) followed by 95:5 DCM:NH 3 (7M in MeOH) to give a solid which is triturated with diethyl ether and petroleum ether to afford the title compound (71mg, 63%).
  • Step 1 3-Oxo-4- ⁇ 4-[5-(l-oxo-2,3-dihydro-lH-isoindol-5-yl)-[l,2,4]triazolo[l,5-a]pyrazin-8- ylamino] -phenyl ⁇ -piperazine-1 -carboxy Hc acid tert-butyl ester
  • This compound may be prepared using methods as described for Compound 6, step 4, using 4-[4-(5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-ylamino)-phenyl]-3-oxo-piperazine-l-carboxylic acid tert-butyl ester (115mg, 0.24mmol), 5-(4,4,5,5-tetramethyl-[l,3,2]dioxaborolan-2-yl)-2,3- dihydro-isoindol-1-one (92mg, 0.35mmol), and Pd(PPh 3 ) 4 (83mg, 0.072mmol) in 1.5M Na 2 CO 3 (aq) (1.3mL, 1.92mmol), and dioxane (2.5mL).
  • reaction mixture is diluted with brine then toluene is added and a precipitate is formed and collected by filtration.
  • the resultant solid is dissolved in DCM and filtered through a silica cartridge to afford the title compound as a yellow solid (60mg, 47%).
  • Step 2 5- ⁇ 8-[4-(2-Oxo-piperazin-l-yl)-phenylaminoJ-[l,2,4Jtriazolo[l,5-aJpyrazin-5-yl ⁇ -2,3-dihydro- isoindol-1-one
  • Nitromethane (10 mL, 169 mmol, 8 equiv.) is added cautiously to a suspension of sodium hydride (4.05 g, 169 mmol, 8 equiv.) and MgSO 4 (40 g) in DMSO (100 mL) at rt and the resulting slurry is stirred for 0.25 h.
  • To the resulting yellow slurry is added 4-bromo-2,6- difluorobenzoic acid methyl ester (5.3 g, 21 mmol) and the mixture is stirred at rt for 3 days at which point all the starting material has been consumed.
  • Step 4 7-Fluoro-5-(4, 4, 5, 5-tetramethyl-fl, 3, 2] dioxaborolan-2-yl)-2, 3-dihydroisoindol-l-one
  • Step 5 7-Fluoro-5- ⁇ 8-[4-(4-isopropylpiperazin-l-yl)-phenylamino]-[ 1,2,4] triazolo[l,5-a] pyrazin-5- yl ⁇ -2, 3-dihydroisoindol-l-one
  • This compound may be prepared using methods as described for Compound 91 using the above boronate.
  • LCMS: Rt 0.90 min (95 %), m/z 487 (M+H) + .
  • Step 1 l-[4-(5-Bromo-[ 1 ,2 ,4j 'triazolo [ 1 ,5-aJ pyrazin-8-ylamino) -phenyl] ' -4-isopropyl-piperazin-2-one
  • Step 2 5-(8-(4-(4-Isopropyl-2-oxopiperazin-l-yl)phenylamino)-[ 1,2,4] 'triazolo [1,5-aJ pyrazin-5- yl)isoindolin- 1 -one
  • This compound may be prepared using methods as described for Compound 6, step 4, using l-[4-(5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-ylamino)-phenyl]-4-isopropyl-piperazin-2-one (40mg, 0.09mmol), 5-(4,4,5,5-tetramethyl-[l,3,2]dioxaborolan-2-yl)-2,3-dihydro-isoindol-l-one (37mg, 0.14mmol), and Pd(PPh 3 ) 4 (31mg, 0.027mmol) in 1.5M Na 2 CO 3 (aq) (0.5mL, 0.75mmol), and dioxane (1.2mL).
  • reaction mixture is diluted with brine then toluene is added and a precipitate is formed and collected by filtration.
  • the filtrate is washed with diethyl ether, petroleum ether and MeOH and then purified by silica gel column chromatography. Elution with 99:1 DCM:NH 3 (7M in MeOH) followed by 98:2 and 95:5 DCM:NH 3 (7M in MeOH) affords the title compound as a yellow solid (24mg, 55%).
  • Step 1 5-Bromo-2-(4-methoxybenzyl)-3 , 3 -dimethyl- 2, 3-dihydroisoindol-l-one
  • a suspension of sodium hydride (130 mg, 60% dispersion in mineral oil, 3.2 mmol) and tetra- n butylammonium iodide (243 mg, 0.68 mmol) in THF (20 mL) is stirred at rt and a solution of 5-bromo-2,3-dihydroisoindol-l-one (675 mg, 3.2 mmol) in THF (20 mL) and DMF (4 mL) is added. After 75 min 4-methoxybenzyl bromide (460 ⁇ L, 3.2 mmol) is added and stirring is continued for 4 h.
  • Step 3 5- ⁇ 8-[4-(4-Isopropylpiperazin-l-yl)-phenylamino] -[ 1 ,2 ,4] triazolo [ 1 ,5-a] pyrazin-5-yl ⁇ -3 ,3- dimethyl-2,3-dihydroisoindol-l-one
  • Step 1 4-(5-Bromo-[ 1 ,2,4] ' triazolo [ 1 ,5-a] pyrazin-8-ylamino) -benzoic acid methyl ester
  • This compound may be prepared using methods as described for Compound 6, step 1 using 5,8-dibromo-[l,2,4]triazolo[l,5-a]pyrazine (250mg, 0.90mmol), 4-aminobenzoic methyl ester (163mg, 1.08mmol) and N-ethyldiisopropyl-amine (0.19mL, 1.08mmol) in 2-propanol (2.5mL). The title compound is obtained after trituration with 2-propanol as a brown solid (148mg, 47%).
  • Step 2 4-(5-Bromo-[ 1 ,2 ,4j 'triazolo [ 1 ,5-aJ 'pyrazin-8-ylamino) -benzoic acid
  • Step 3 4-(5-Bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-ylamino)-N-pyridin-3-ylmethyl-benzamide
  • Step 4 4-(5-(lH-Pyrazol-4-yl)-[ 1 ,2 ,4j 'triazolo [ 1 ,5-aJ pyrazin-8-ylamino) -N-(pyridin-3- ylmethyl)benzamide
  • This compound may be prepared using methods as described for Compound 6, step 4 using 4-(5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-ylamino)-N-pyridin-3-ylmethyl-benzamide (lOOmg, 0.24mmol), 4-(4,4,5,5-tetramethyl-[l,3,2]dioxaborolan-2-yl)-lH-pyrazole (93mg, 0.48mmol) and Pd(PPh 3 ) 4 (70mg, O.O ⁇ mmol) in 1.5M K 2 CO 3 (aq) (1.4mL) and dioxane (2.5mL).
  • Step 1 4-[5-(2-Methoxy-pyridin-4-yl)-[l,2,4Jtriazolo[l,5-aJpyrazin-8-ylaminoJ-N-pyridin-3-ylmethyl- benzamide
  • This compound may be prepared using methods as described for Compound 6, step 4 using 4-(5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-ylamino)-N-pyridin-3-ylmethyl-benzamide (120mg, 0.28mmol), 2-methoxypyridine-4-boronic acid (87mg, 0.57mmol) and Pd(PPh 3 ) 4 (81mg, 0.07mmol) in 1.5M K 2 CO 3 (aq) (1.6mL) and dioxane (2.9mL).
  • Step 2 4-(5-(2-Oxo-l ,2-dihydropyridin-4-yl)-[l ,2,4Jtriazolo[l ,5-aJpyrazin-8-ylamino)-N-(pyridin-3- ylmethyl)benzamide
  • N-pyridin-3-ylmethyl-benzamide (71.5mg, O.l ⁇ mmol) and pyridinium hydrochloride (91mg, 0.79mmol) in water (0.5mL) in a sealed tube is heated at 15O 0 C for 25 minutes. After this time the solvent is removed in vacuo. The residue is chromatographed on silica gel, eluting with DCM followed by 98:2 and 90:10 DCM:NH 3 (7M in MeOH), and the fractions containing the desired product are combined and evaporated. The title compound is isolated as a yellow solid (34.5mg, 49%).
  • Step 3 4-(5-Bromo-fl,2,4Jtriazolofl,5-aJpyrazin-8-ylamino)-2-hydroxy-N-f(6-methyl-pyridin-3- yl)methyl]benzamide
  • Step 4 2-Methoxy-N-(6-methylpyridin-3-yl)methyl-4-[5-(lH-pyrazol-4-yl)-[l,2,4Jtriazolo[l,5- aJpyrazin-8-ylamino] ' -benzamide
  • the tube is sealed, placed in an ultrasonic bath under a flow of nitrogen gas for 30 seconds and then placed into an oil bath at 85°C.
  • the reaction is stirred for 28 hours, adding additional portions of boronic acid (10 mg) and catalyst (2 mg) after 2 h and 18 h.
  • the crude mixture is absorbed onto SiO 2 and purified by column chromatography, eluting with 1 % - 10% MeOH/DCM.
  • the product obtained is redissolved in MeOH/DCM (4:1, 10 mL) and 0.1 MsOH/MeOH (2 eq) is added, the solvents evaporated and the residue taken up in water and lyophillised to afford the title compound as its bis-mesylate salt (38 mg).
  • LCMS Rt 0.92 min (93.9 %) m/z (ESI) 455 (M+H) + .
  • This compound may be prepared using the methods described for Compound 120, using benzylamine in step 1.
  • This compound may be prepared using methods as described for Compound 129 using
  • This compound may be prepared using methods as described for Compound 129, using 4-isopropylpiperazine in step 5.
  • LCMS: Rt 0.84 min (100 %), m/z (ESI) 432 (M+H) + .
  • Step 1 3-(5-Bromo-[ 1 ,2 ,4] triazolo [ 1 ,5-a] pyrazin-8-ylamino)benzoic acid ethyl ester
  • Step 2 3-[(5-Bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)- ⁇ .& ⁇ -butoxycarbonyl-amino]benzoic acid ethyl ester
  • Step 3 3- ⁇ tert-Butoxycarbonyl-[5-(lH-pyrazol-4-yl)-[ 1,2,4] 'triazolofl,5-aj pyrazin-8-yl] - aminojbenzoic acid ethyl ester

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Rheumatology (AREA)
  • Urology & Nephrology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Pulmonology (AREA)
  • Neurology (AREA)
  • Cardiology (AREA)
  • Vascular Medicine (AREA)
  • Pain & Pain Management (AREA)
  • Neurosurgery (AREA)
  • Oncology (AREA)
  • Dermatology (AREA)
  • Biomedical Technology (AREA)
  • Immunology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Epidemiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)

Abstract

Novel [1.2.4]triazolo[1,5-a]pyrazine compounds are disclosed that have a formula represented by the following (formula I). The compounds may be prepared as pharmaceutical compositions, and may be used for the prevention and treatment of a variety of conditions in mammals including humans, including by way of non-limiting example, arthritis, inflammation, and others.

Description

TRIAZOLOPYRAZINE COMPOUNDS USEFUL FOR THE TREATMENT OF
DEGENERATIVE & INFLAMMATORY DISEASES
FIELD OF THE INVENTION
[0001] The present invention relates to a class of triazolopyrazine compounds capable of binding to the active site of a serine/threonine kinase, the expression of which is involved in the pathway resulting in the degradation of extra-cellular matrix (ECM), joint degeneration and diseases involving such degradation and/or inflammation.
[0002] Diseases involving the degradation of extra-cellular matrix include, but are not limited to, psoriatic arthritis, juvenile arthritis, early arthritis, reactive arthritis, osteoarthritis, ankylosing spondylitis, osteoporosis, muskulo skeletal diseases like tendonitis and periodontal disease, cancer metastasis, airway diseases (COPD, asthma), renal and liver fibrosis, cardio-vascular diseases like atherosclerosis and heart failure, and neurological diseases like neuroinflammation and multiple sclerosis. Diseases involving primarily joint degeneration include, but are not limited to, psoriatic arthritis, juvenile arthritis, early arthritis, reactive arthritis, osteoarthritis, and ankylosing spondylitis. [0003] Rheumatoid arthritis (RA) is a chronic joint degenerative disease, characterized by inflammation and destruction of the joint structures. When the disease is unchecked, it leads to substantial disability and pain due to loss of joint functionality and even premature death. The aim of an RA therapy, therefore, is not to slow down the disease but to attain remission in order to stop the joint destruction. Besides the severity of the disease outcome, the high prevalence of RA (~ 0.8% of adults are affected worldwide) means a high socio-economic impact. (For reviews on RA, we refer to Smolen and Steiner (2003); Lee and Weinblatt (2001); Choy and Panayi (2001); O'Dell (2004) and Firestein (2003)).
[0004] Although it is widely accepted that RA is an auto-immune disease, there is no consensus concerning the precise mechanisms driving the 'initiation stage' of the disease. What is known is that the initial trigger(s) does mediate, in a predisposed host, a cascade of events that leads to the activation of various cell types (B-cells, T-cells, macrophages, fibroblasts, endothelial cells, dendritic cells and others). Concomitantly, an increased production of various cytokines is observed in the joints and tissues surrounding the joint (e.g. TNF-α, IL-6, IL-I, IL-15, IL-18 and others). When the disease progresses, the cellular activation and cytokine production cascade becomes self- perpetuating. At this early stage, the destruction of joint structures is already very clear. Thirty percent of the patients have radiographic evidence of bone erosion at the time of diagnosis and this proportion increases to 60 percent after two years.
[0005] Histological analysis of the joints of RA patients clearly evidences the mechanisms involved in the RA-associated degradative processes. This analysis shows that the main effector responsible for RA-associated joint degradation is the pannus, where the synovial fibroblast, by producing diverse proteolytic enzymes, is the prime driver of cartilage and bone erosion. A joint classically contains two adjacent bones that articulate on a cartilage layer surrounded by the synovial membrane and joint capsule. In the advanced RA patient, the synovium of joint increases in size to form the pannus, due to the proliferation of the synovial fibroblasts and the infiltration of mononuclear cells such as T-cells, B-cells, monocytes, macrophages and neutrophils. The pannus mediates the degradation of the adjacent cartilage, leading to the narrowing of the joint space, and has the potential to invade adjacent bone and cartilage. As bone and cartilage tissues are composed mainly of collagen type I or II, respectively, the pannus destructive and invasive properties are mediated by the secretion of collagenolytic proteases, principally the matrix metallo proteinases (MMPs). The erosion of the bone under and adjacent to the cartilage is also part of the RA process, and results principally from the presence of osteoclasts at the interface of bone and pannus. Osteoclasts are multinucleated cells that, upon adhesion to the bone tissue, form a closed compartment, within which the osteoclasts secrete proteases (Cathepsin K, MMP9) that degrade the bone tissue. The osteoclast population in the joint is abnormally increased by osteoblast formation from precursor cells induced by the secretion of the receptor activator of NFKB ligand (RANKL) by activated SFs and T-cells.
[0006] Various collagen types have a key role in defining the stability of the extracellular matrix (ECM). Collagenstype I and collagen type II, for example, are the main components of bone and cartilage, respectively. Collagen proteins typically organise into multimeric structures referred to as collagen fibrils. Native collagen fibrils are very resistant to proteolytic cleavage. Only a few types of ECM-degrading proteins have been reported to have the capacity to degrade native collagen: MMPs and Cathepsins. Among the Cathepsins, cathepsin K, which is active mainly in osteoclasts, is the best characterised. Among the MMPs, MMPl, MMP2, MMP8 MMP13 and MMP14 are known to have collagenolytic properties. The correlation between an increased expression of MMPl by synovial fibroblasts (SFs) and the progression of the arthritic disease is well-established and is predictive for joint erosive processes (Cunnane et al., 2001). In the context of RA, therefore, MMPl represents a highly relevant collagen degrading protein. In vitro, the treatment of cultured SFs with cytokines relevant in the RA pathology (e.g. TNF-α and ILlB) will increase the expression of MMPl by these cells (Andreakos et al., 2003). Monitoring the levels of MMPl expressed by SFs therefore is a relevant readout in the field of RA as it is indicative for the activation of SFs towards an erosive phenotype that, in vivo, is responsible for cartilage degradation. Inhibition of the MMPl expression by SFs represents a valuable therapeutic approach towards the treatment of RA.
[0007] The activity of the ECM-degrading proteins can also be causative or correlate with the progression of various diseases different from RA, e.g. other diseases that involve the degradation of the joints. These diseases include, but are not limited to, psoriatic arthritis, juvenile arthritis, early arthritis, reactive arthritis, osteoarthritis, and ankylosing spondylitis. Other diseases that may be treatable with compounds identified according to the present invention and using the targets involved in the expression of MMPs as described herein are osteoporosis, muscular skeletal diseases like tendonitis and periodontal disease (Gapski et al., 2004), cancer metastasis (Coussens et al., 2002), airway diseases (COPD, asthma) (Suzuki et al., 2004), lung, renal fibrosis (Schanstra et al., 2002), liver fibrosis associated with chronic hepatitis C (Reiff et al., 2005), cardio-vascular diseases like atherosclerosis and heart failure (Creemers et al., 2001), and neurological diseases like neuroinflammation and multiple sclerosis (Rosenberg, 2002). Patients suffering from such diseases may benefit from stabilizing the ECM (by protecting it from degradation).
[0008] The 471 -amino acid serine/threonine kinase identified as Mitogen- Activated Protein
Kinase- Activated Protein Kinase 5 (MAPKAPK5 or PRAK) is expressed in a wide panel of tissues. The protein contains its catalytic domain at the N-terminal end and both a nuclear localization signal (NLS) and nuclear export signal (NES) at its C-terminal end. Endogenous MAPKAPK5 is predominantly present in the cytoplasm, but stress or cytokine activation of the cells mediates its translocation into the nucleus (New et al., 2003). This event is dependent on phosphorylation of MAPKAPK5. Thrl82 is the regulatory phosphorylation site of MAPKAPK5. Although the p38α kinase is able to phosphorylate MAPKAPK5 in an overexpression setting, experiments with endogenous MAPKAPK5 do not support this hypothesis (Shi et al., 2003). MAPKAPK5 knock-out mice have been generated that are viable and fertile. The phenotype of these mice is quite different from that of mice deficient for MAPKAPK2, a MAPKAPK5 related kinase that is regulated by p38α (Shi et al., 2003). This indicates that the function of each protein is distinct and that neither kinase can compensate for the other's activity. Taken together, MAPKAPK5 and MAPKAPK2 represent distinct targets with a non-redundant role. MAPK6 (also referred to as ERK3) has recently been identified as a physiologically relevant substrate for MAPKAPK5, defining a novel signal transduction pathway (Seternes et al., 2004).
BACKGROUND OF THE INVENTION
[0009] NSAIDS (Non-steroidal anti-inflammatory drugs) are used to reduce the pain associated with RA and improve life quality of the patients. These drugs will not, however, put a brake on the RA-associated joint destruction.
[0010] Corticosteroids were found to decrease the progression of RA as detected radiographically and are used at low doses to treat part of the RA patients (30 to 60%). Serious side effects, however, are associated with long corticosteroid use (skin thinning, osteoporosis, cataracts, hypertension, hyperlipidemia).
[0011] Synthetic DMARDs (Disease-Modifying Anti-Rheumatic Drugs) (e.g. methotrexate, leflunomide, sulfasalazine) mainly tackle the immuno-inflammatory component of RA. As a main disadvantage, these drugs only have a limited efficacy (joint destruction is only slowed down but not blocked by DMARDs such that disease progression in the long term continues). The lack of efficacy is indicated by the fact that, on average, only 30% of the patients achieve an ACR40 score after 24 months treatment with methotrexate. This means that, according to the American College of Rheumatology, only 30% of the patients achieve a 50% improvement of their symptoms (O'Dell et al., 1996). In addition, the precise mechanism of action of DMARDs is often unclear. [0012] Biological DMARDs (Infliximab, Etanercept, Adalimumab, Rituximab, CTLA4-Ig) are therapeutic proteins that inactivate cytokines (e.g. TNF-α) or cells (e.g. T-cells or B-cells) that have an important role in the RA pathophysiology (Kremer et al., 2003; Edwards et al., 2004). Although the TNF-α-blockers (Infliximab, Etanercept, Adalimumab) and methotrexate combination therapy is the most effective RA treatment currently available, it is striking that even this therapy only achieves a 50% improvement (ACR40) in disease symptoms in 50-60% of patients after 12 months therapy (St Clair et al., 2004). Some adverse events warnings for anti-TNF-α drugs exist, shedding a light on the side effects associated to this type of drugs. Increased risk of infections (tuberculosis), hematologic events and demyelinating disorders have been described for the TNF-α blockers (see also Gomez-Reino et al., 2003). Besides the serious side effects, the TNF-α blockers also share the general disadvantages of the biological class of therapeutics, which are the unpleasant way of administration (frequent injections accompanied by infusion site reactions) and the high production cost. Newer agents in late development phase target T-cell co-stimulatory molecules and B-cells. The efficacy of these agents is expected to be similar to that of the TNF-α blockers. The fact that a variety of targeted therapies have similar but limited efficacies, suggests that there is a multiplicity of pathogenic factors for RA. This is also indicative for the deficiencies in our understanding of pathogenic events relevant to RA.
[0013] The current therapies for RA are not satisfactory due to a limited efficacy (No adequate therapy exists for 30% of the patients). This calls for additional strategies to achieve remission. Remission is required since residual disease bears the risk of progressive joint damage and thus progressive disability. Inhibiting the immuno-inflammatory component of the RA disease, which represents the main target of drugs currently used for RA treatment, does not result in a blockade of joint degradation, the major hallmark of the disease.
[0014] US 2005/0009832 describes substituted imidazolo[l,2-a]pyrazine-8-yl-amines as modulators of protein kinases, including MAPKAPK5. WO02/056888 describes inhibitors of MAPKAPK5 as TNF modulators able to regulate the expression of certain cytokines. Neither of these prior art references discloses any compound within the scope of the class of compounds described herein below.
SUMMARY OF THE INVENTION
[0015] The present invention is based on the discovery of that MAPKAPK5 functions in the pathway that results in the expression of MMPl, and that inhibitors of MAPKAPK5 activity, such as the compounds of the present invention, are useful for the treatment of diseases involving the abnormally high expression of MMP activity.
[0016] The compounds of the present invention may be described generally as
[l,2,4]triazolo[l,5-a]pyrazine-8-yl-amines substituted in the 5-position by an aryl and heteroaryl group, and an in the 8-position by an arylamino or a heteroarylamino group.
[0017] More particularly, the present invention relates to compounds having matrix metallo proteinase inhibiting properties in a mammalian cell, according to formula (I):
Figure imgf000007_0001
wherein:
A and B are independently CR2R", NR", oxygen or sulphur; AA is CR2 or N; D is C=O, CR2R" or NR";
E is NH or CR"R6, when k is zero, and is NH or CR"R6a, when k is one;
F is sulphur, oxygen or NH;
T is oxygen or NR;
U, V, W and X are independently CR"R7 or NR"; Y is CR" or N;
Z is hydrogen, amino, hydroxyl, lower alkoxy, carbamoyl, carboxyl, SO2RZ, SO2NRRZ, -
NR(CO)(CH2)d-Rz, -NRRz, -(CO)-ORz, -(CO)-NR(CH2)d-Rz, or
Figure imgf000007_0002
R is independently hydrogen or lower alkyl;
R" is H or forms a double bond with an adjacent atom;
Rl is H; R2; or lower alkyl, lower cycloalkyl and lower alkyl-lower cycloalkyl, optionally substituted with one or more R2;
R3 is H or forms a double bond with an adjacent R"; R2 is H, F, Cl; CN; COOR4; 0R4; C(O)N(R4R5); S(O)2N(R4R5); lower alkyl; O- lower alkyl;
NH-lower alkyl; S-lower alkyl; COO- lower alkyl; OC(O)- lower alkyl; C(0)N(R4)- lower alkyl; S(O)2N(R4)-lower alkyl; S(O)N(R4)-lower alkyl; S(O)2-lower alkyl; S(O)-lower alkyl;
N(R4)S(O)2-lower alkyl; and N(R4)S(O)-lower alkyl; wherein each lower alkyl is optionally substituted with one or more of F and Cl; R4 and R5 are independently
H; F, Cl; or lower alkyl, lower cycloalkyl, or lower alkyl-lower cycloalkyl optionally substituted with one or more of F and Cl;
R6 is hydrogen, amino, hydroxyl, carbamoyl, carboxyl, SO2R, NRR', -(CO)-OR, or -(CO)-NRR'; R6a is R6, Cl, F, lower alkoxy, cyano, trifluoromethoxy; or together with the adjacent be -
(CHR")n-NR-(CHR")p-, and form a five or six member heterocyclic ring fused to the ring to which they are bonded;
R7 is independently hydrogen, halogen, lower alkyl or lower alkoxy; Rz is hydrogen, lower alkyl, lower alkanoyl, phenyl, 1-loweralkyl pyrrolidin-3-yl, pyrazol-4-yl, pyrazol-2-yl, or lower alkyl, lower alkanoyl, phenyl, 1-loweralkyl pyrrolidin-3-yl, pyrazol-4- yl, pyrazol-2-yl or pyrid-3-yl substituted by one or more of hydroxyl, amino, mono- or di- loweralkylamino, acetamidyl, lower alkanoyl, lower alkyl, 4-hydroxy-phenyl, 3- aminomethylphenyl, lower alkyl sulfonyl, 4-diloweralkylaminophenyl, pyrid-3-yl, lH-indol-
3-yl, morpholin-4-yl,; R and Rz together may be -(CHR)q-T-(CHR)r- and form a five or six member heterocyclic ring with the nitrogen to which they are bonded; Rz and R7 together may be -(CHR")n-NR-(CHR")p-, and form a five or six member heterocyclic ring fused to the ring to which they are bonded; b and d are independently O or 1; provided at least one of b or d is 1; k is O or 1 ; m is O or 1 ; n and p are independently O, 1 or 2; q and r are 1 or 2; x is o or 1 ; with the provisos that at least one of R7 or Rz is other than hydrogen; or a pharmaceutically acceptable salt, hydrate, solvate or prodrug thereof. [0018] Another aspect of the present invention is compounds according to formula III:
Figure imgf000008_0001
(III) wherein
R1 is H, or substituted or unsubstituted alkyl; and each of R8 and R9 is independently selected from substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl; or a pharmaceutically acceptable salt, solvate or prodrug thereof; and stereoisomers, isotopic variants and tautomers thereof.
[0019] In one embodiment, with respect to compounds of formula III, R8 is selected from substituted or unsubstituted cyclopentyl, cyclohexyl, substituted or unsubstituted phenyl, substituted or unsubstituted pyridyl, substituted or unsubstituted pyrimidine, and substituted or unsubstituted pyrazine, substituted or unsubstituted pyrrole, substituted or unsubstituted pyrazole and substituted or unsubstituted imidazole.
[0020] In one embodiment, with respect to compounds of formula III, R1 is H, Me, Et, i-Pr or
CF3.
[0021] In one embodiment, with respect to compounds of formula III, R1 is H.
[0022] Another aspect of the present invention relates to compounds according to formula
IVa, IVb, IVc, or IVd:
Figure imgf000009_0001
IVa IVb IVc or IVd
and wherein L is a bond, -CO-, -O(CH2)ml-, -CON(H)(CH2)ml-, or - NHCO-; the subscript ml is selected from 1-4; the ring P is substituted or unsubstituted heterocycloalkyl; the subscript n is selected from 1-4; each R8a is independently selected from hydrogen, substituted or unsubstituted alkyl, alkoxy, cyano, carbamoyl, CHO, and halo; and R9 is independently selected from substituted or unsubstituted aryl and heteroaryl; or a pharmaceutically acceptable salt, solvate or prodrug thereof; and stereoisomers, isotopic variants and tautomers thereof.
[0023] In a one embodiment, with respect to compounds of formulae IH-IVd, R9 is substituted or unsubstituted aryl. In another embodiment, R9 is substituted or unsubstituted phenyl. [0024] In a one embodiment, with respect to compounds of formulae IH-IVd, R9 is substituted or unsubstituted heteroaryl. In another embodiment, R9 is substituted or unsubstituted pyridyl.
[0025] In a one embodiment, with respect to compounds of formulae IH-IVd, R9 is selected from substituted or unsubstituted phenyl, indolyl, isoinolyl, pyrrolyl, furanyl, thienyl, pyrazolyl, oxazolyl, and thiazolyl.
[0026] In a further aspect, the present invention provides pharmaceutical compositions comprising a triazolopyrazine compound of the invention, and a pharmaceutical carrier, excipient or diluent. In this aspect of the invention, the pharmaceutical composition can comprise one or more of the compounds described herein. Moreover, the compounds of the present invention useful in the pharmaceutical compositions and treatment methods disclosed herein, are all pharmaceutically acceptable as prepared and used.
[0027] Another aspect of this invention relates to the use of the present compound in a therapeutic method, a pharmaceutical composition, and the manufacture of such composition, useful for the treatment of diseases involving inflammation, collagen degradation, and in particular, diseases characteristic of abnormal matrix metallo protease (MMPl) and/or Mitogen- Activated Protein-Kinase
Activated Protein Kinase 5 (MAPKAPK5) activity, of which rheumatoid arthritis (RA) is a particular example of such diseases. This invention also relates to processes for the preparation of the present compounds.
[0028] Other objects and advantages will become apparent to those skilled in the art from a consideration of the ensuing detailed description, which proceeds with reference to the following illustrative drawings.
BRIEF DESCRIPTION OF THE DRAWINGS
[0029] Figure 1. This diagram shows the striking histological differences between a healthy joint and that of a RA patient.
[0030] Figure 2. This chart shows the increased expression of MMPl in synovial fibroblasts triggered with cytokines involved in rheumatoid arthritis pathology.
[0031] Figure 3. This graph shows the dose-dependent inhibition of the "TNF-α-based trigger"-induced expression of MMPl by SFs by a known anti- inflammatory compound.
[0032] Figure 4. This gel shows the reduction, at the protein level, of the expression of
MAPKAPK5 in SFs by infection of the cells with Ad-siRNA virus targeting MAPKAPK5.
[0033] Figure 5. This chart shows the reduction of 'complex trigger' induced levels of
MMPl expression by SFs by an Ad-siRNA virus targeting MAPKAPK5.
DETAILED DESCRIPTION OF THE INVENTION Definitions
[0034] When describing the compounds, pharmaceutical compositions containing such compounds and methods of using such compounds and compositions, the following terms have the following meanings unless otherwise indicated. It should also be understood that any of the moieties defined forth below may be substituted with a variety of substituents, and that the respective definitions are intended to include such substituted moieties within their scope. By way of non- limiting example, such substituents may include e.g. halo (such as fluoro, chloro, bromo), -CN, -CF3, -OH, -OCF3, C2-C6 alkenyl, C3-C6 alkynyl, CpC6 alkoxy, aryl and di-CrC6 alkylamino. It should be further understood that the terms "groups" and "radicals" can be considered interchangeable when used herein.
[0035] "Alkoxy" means alkyl-O-. Exemplary alkoxy includes methoxy, ethoxy, n-propoxy, i-propoxy, n-butoxy, and heptoxy. Preferred alkoxy groups are lower alkoxy. [0036] "Alkyl" means straight or branched aliphatic hydrocarbon having 1 to about 20 carbon atoms. Preferred alkyl has 1 to about 12 carbon atoms. More preferred is lower alkyl. Branched means that one or more lower alkyl groups such as methyl, ethyl or propyl is attached to a linear alkyl chain.
[0037] "Alkyl amino" means alkyl-NH-. Preferred alkyl amino is (Ci-Ce)-alkyl amino.
Exemplary alkyl amino includes methylamino and ethylamino.
[0038] "Amino lower alkanoyl" means NH2-R-CO-, where R is lower alkylene. Preferred groups include aminoethanoyl and aminoacetyl.
[0039] "Carbamoyl lower alkyl" means the radical NH2CO-lower alkyl-. Preferred groups include carbamoylethyl and carbamoylmethyl.
[0040] "Carboxy lower alkyl ester" means a lower alkyl ester of a carboxy radical, -COO- group.
[0041] "Compounds of the present invention", and equivalent expressions, are meant to embrace compounds of Formula (I, II or III) as hereinbefore described, which expression includes the prodrugs, the pharmaceutically acceptable salts, and the solvates, e.g., hydrates, where the context so permits. Similarly, reference to intermediates, whether or not they themselves are claimed, is meant to embrace their salts, and solvates, where the context so permits. [0042] "Expression" means endogenous expression.
[0043] "Halo" or "halogen" means fluoro, chloro, bromo, or iodo.
[0044] "Hydrogen" means in the context of a substituent that -H is present at the compound position and also includes its isotope, deuterium.
[0045] "Lower alkanoyl amino" means an amino group with an organic functional group R-
CO-, where R represents a lower alkyl group.
[0046] "Lower alkyl" means 1 to about 6 carbon atoms in a linear alkyl chain that may be straight or branched.
[0047] "Lower alkoxy" means 1 to about 6 carbon atoms in a linear alkyl chain that may be straight or branched, and that is bonded by an oxygen atom.
[0048] "Lower alkyl sulphonamide" refers to a lower alkyl amide of sulphonamide of the formula -SO2NR515R*, where R* is hydrogen or lower alkyl, and at least one R* is lower alkyl. [0049] "Prophylaxis" means a measure taken for the prevention of a disease.
[0050] "Solvate" means a physical association of a compound useful in this invention with one or more solvent molecules. This physical association includes hydrogen bonding. In certain instances the solvate will be capable of isolation, for example when one or more solvent molecules are incorporated in the crystal lattice of the crystalline solid. "Solvate" encompasses both solution-phase and isolable solvates. The compounds of the invention may be prepared e.g. in crystalline form and may be solvated or hydrated. Suitable solvates include pharmaceutically acceptable solvates, such as hydrates, and further include both stoichiometric solvates and non-stoichiometric solvates. Conventional solvents include water, ethanol, acetic acid and the like, therefore, representative solvates include hydrates, ethanolates and methanolates.
[0051] "Substituted" means that one atom or group of atoms in a molecule is replaced by another atom or group.
[0052] "Sulphonamide" refers to a group of compounds containing the chemical group -
SO2NH2.
[0053] "Therapeutically effective amount" means that amount of a drug or pharmaceutical agent that will elicit the biological or medical response of a subject that is being sought by a medical doctor or other clinician. The "therapeutically effective amount" can vary depending on the compound, the disease and its severity, and the age, weight, etc., of the subject to be treated. In particular, with regard to treating an disease condition characterized by the degradation of extracellular matrix, the term "effective matrix metallo-protease inhibiting amount" is intended to mean that effective amount of an compound of the present invention that will bring about a biologically meaningful decrease in the production of MMP-I in the subject's disease affected tissues such that extracellular matrix degradation is meaningfully reduced. A compound having matrix metallo-protease inhibiting properties or a "matrix metallo-protease inhibiting compound" means a compound of the present invention that provided to a cell in effective amounts is able to cause a biologically meaningful decrease in the production of MMP-I in such cells.
[0054] "Aryl" refers to a monovalent aromatic hydrocarbon group derived by the removal of one hydrogen atom from a single carbon atom of a parent aromatic ring system. Typical aryl groups include, but are not limited to, groups derived from aceanthrylene, acenaphthylene, acephenanthrylene, anthracene, azulene, benzene, chrysene, coronene, fluoranthene, fluorene, hexacene, hexaphene, hexalene, αs-indacene, s-indacene, indane, indene, naphthalene, octacene, octaphene, octalene, ovalene, penta-2,4-diene, pentacene, pentalene, pentaphene, perylene, phenalene, phenanthrene, picene, pleiadene, pyrene, pyranthrene, rubicene, triphenylene, trinaphthalene and the like. Particularly, an aryl group comprises from 6 to 14 carbon atoms.
[0055] "Substituted Aryl" includes those groups recited in the definition of "substituted" herein, and particularly refers to an aryl group that may optionally be substituted with 1 or more substituents, for instance from 1 to 5 substituents, particularly 1 to 3 substituents, selected from the group consisting of acyl, acylamino, acyloxy, alkenyl, substituted alkenyl, alkoxy, substituted alkoxy, alkoxycarbonyl, alkyl, substituted alkyl, alkynyl, substituted alkynyl, amino, substituted amino, aminocarbonyl, aminocarbonylamino, aminocarbonyloxy, aryl, aryloxy, azido, carboxyl, cyano, cycloalkyl, substituted cycloalkyl, halogen, hydroxyl, nitro, thioalkoxy, substituted thioalkoxy, thioaryloxy, thiol, alkyl-S(O)-, aryl-S(O)-, alkyl-S(O)2- and aryl-S(O)2-.
[0056] "Bicycloaryl" refers to a monovalent aromatic hydrocarbon group derived by the removal of one hydrogen atom from a single carbon atom of a parent bicycloaromatic ring system. Typical bicycloaryl groups include, but are not limited to, groups derived from indane, indene, naphthalene, tetrahydronaphthalene, and the like. Particularly, an aryl group comprises from 8 to 11 carbon atoms.
[0057] "Bicycloheteroaryl" refers to a monovalent bicycloheteroaromatic group derived by the removal of one hydrogen atom from a single atom of a parent bicycloheteroaromatic ring system. Typical bicycloheteroaryl groups include, but are not limited to, groups derived from benzofuran, benzimidazole, benzindazole, benzdioxane, chromene, chromane, cinnoline, phthalazine, indole, indoline, indolizine, isobenzofuran, isochromene, isoindole, isoindoline, isoquinoline, benzothiazole, benzoxazole, naphthyridine, benzoxadiazole, pteridine, purine, benzopyran, benzpyrazine, pyridopyrimidine, quinazoline, quinoline, quinolizine, quinoxaline, benzomorphan, tetrahydroisoquinoline, tetrahydroquinoline, and the like. Preferably, the bicycloheteroaryl group is between 9-11 membered bicycloheteroaryl, with 5-10 membered heteroaryl being particularly preferred. Particular bicycloheteroaryl groups are those derived from benzothiophene, benzofuran, benzothiazole, indole, quinoline, isoquinoline, benzimidazole, benzoxazole and benzdioxane. [0058] "Carbamoyl" refers to the radical -C(O)N(R42)2 where each R42 group is independently hydrogen, alkyl, cycloalkyl or aryl, as defined herein, which may be optionally substituted as defined herein. In a specific embodiment, the term "carbamoyl" refers to -C(O)-NH2. [0059] "Cycloalkyl" refers to cyclic hydrocarbyl groups having from 3 to about 10 carbon atoms and having a single cyclic ring or multiple condensed rings, including fused and bridged ring systems, which optionally can be substituted with from 1 to 3 alkyl groups. Such cycloalkyl groups include, by way of example, single ring structures such as cyclopropyl, cyclobutyl, cyclopentyl, cyclooctyl, 1 -methylcyclopropyl, 2-methylcyclopentyl, 2-methylcyclooctyl, and the like, and multiple ring structures such as adamantanyl, and the like.
[0060] "Substituted cycloalkyl" includes those groups recited in the definition of
"substituted" herein, and particularly refers to a cycloalkyl group having 1 or more substituents, for instance from 1 to 5 substituents, and particularly from 1 to 3 substituents, selected from the group consisting of acyl, acylamino, acyloxy, alkoxy, substituted alkoxy, alkoxycarbonyl, alkoxycarbonylamino, amino, substituted amino, aminocarbonyl, aminocarbonylamino, aminocarbonyloxy, aryl, aryloxy, azido, carboxyl, cyano, cycloalkyl, substituted cycloalkyl, halogen, hydroxyl, keto, nitro, thioalkoxy, substituted thioalkoxy, thioaryloxy, thioketo, thiol, alkyl-S(O)-, aryl-S(O)-, alkyl-S(O)2- and aryl-S(O)2-.
[0061] "Substituted" refers to a group in which one or more hydrogen atoms are each independently replaced with the same or different substituent(s). Typical substituents include, but are not limited to, -X, -R46, -O", =0, -OR46, -SR46, -S", =S, -NR46R47, =NR46, -CX3, -CF3, -CN, -OCN, - SCN, -NO, -NO2, =N2, -N3, -S(O)2O", -S(O)2OH, -S(O)2R46, -OS(O2)O", -OS(O)2R46, -P(O)(O")2, - P(O)(OR46XO"), -OP(O)(OR46)(OR47), -C(O)R46, -C(S)R46, -C(O)OR46, -C(O)NR46R47, -C(O)O", -C(S)OR46, -NR48C(O)NR46R47, -NR48C(S)NR46R47, -NR49C(NR48)NR46R47 and -C(NR48)NR46R47, where each X is independently a halogen; each R46, R47, R48 and R49 are independently hydrogen, alkyl, substituted alkyl, aryl, substituted alkyl, arylalkyl, substituted alkyl, cycloalkyl, substituted alkyl, cycloheteroalkyl, substituted cycloheteroalkyl, heteroalkyl, substituted heteroalkyl, heteroaryl, substituted heteroaryl, heteroarylalkyl, substituted heteroarylalkyl, -NR50R51, -C(O)R50 or -S(O)2R50 or optionally R50 and R51 together with the atom to which they are both attached form a cycloheteroalkyl or substituted cycloheteroalkyl ring; and R50 and R51 are independently hydrogen, alkyl, substituted alkyl, aryl, substituted alkyl, arylalkyl, substituted alkyl, cycloalkyl, substituted alkyl, cycloheteroalkyl, substituted cycloheteroalkyl, heteroalkyl, substituted heteroalkyl, heteroaryl, substituted heteroaryl, heteroarylalkyl or substituted heteroarylalkyl. [0062] Examples of representative substituted aryls include the following
Figure imgf000014_0001
[0063] In these formulae one of R52 and R53 may be hydrogen and at least one of R52 and R53 is each independently selected from alkyl, alkenyl, alkynyl, cycloheteroalkyl, alkanoyl, alkoxy, aryloxy, heteroaryloxy, alkylamino, arylamino, heteroarylamino, NR54COR55, NR54SOR55 ; NR54SO2R57, COOalkyl, COOaryl, CONR54R55, CONR54OR55, NR54R55, SO2NR54R55, S-alkyl, S-alkyl, SOalkyl, S02alkyl, Saryl, SOaryl, S02aryl; or R52 and R53 may be joined to form a cyclic ring (saturated or unsaturated) from 5 to 8 atoms, optionally containing one or more heteroatoms selected from the group N, O or S. R54, R55, and R56 are independently hydrogen, alkyl, alkenyl, alkynyl, perfluoroalkyl, cycloalkyl, cycloheteroalkyl, aryl, substituted aryl, heteroaryl, substituted or hetero alkyl or the like.
[0064] "Hetero" when used to describe a compound or a group present on a compound means that one or more carbon atoms in the compound or group have been replaced by a nitrogen, oxygen, or sulfur heteroatom. Hetero may be applied to any of the hydrocarbyl groups described above such as alkyl, e.g. heteroalkyl, cycloalkyl, e.g. cycloheteroalkyl, aryl, e.g. heteroaryl, cycloalkenyl, cycloheteroalkenyl, and the like having from 1 to 5, and especially from 1 to 3 heteroatoms. [0065] "Heteroaryl" refers to a monovalent heteroaromatic group derived by the removal of one hydrogen atom from a single atom of a parent heteroaromatic ring system. Typical heteroaryl groups include, but are not limited to, groups derived from acridine, arsindole, carbazole, β-carboline, chromane, chromene, cinnoline, furan, imidazole, indazole, indole, indoline, indolizine, isobenzofuran, isochromene, isoindole, isoindoline, isoquinoline, isothiazole, isoxazole, naphthyridine, oxadiazole, oxazole, perimidine, phenanthridine, phenanthroline, phenazine, phthalazine, pteridine, purine, pyran, pyrazine, pyrazole, pyridazine, pyridine, pyrimidine, pyrrole, pyrrolizine, quinazoline, quinoline, quinolizine, quinoxaline, tetrazole, thiadiazole, thiazole, thiophene, triazole, xanthene, and the like. Preferably, the heteroaryl group is between 5-15 membered heteroaryl, with 5-10 membered heteroaryl being particularly preferred. Particular heteroaryl groups are those derived from thiophene, pyrrole, benzothiophene, benzofuran, indole, pyridine, quinoline, imidazole, oxazole and pyrazine. [0066] Examples of representative heteroaryls include the following:
Figure imgf000015_0001
Figure imgf000015_0002
Figure imgf000015_0003
wherein each Y is selected from carbonyl, N, NR58, O, and S; and R58 is independently hydrogen, alkyl, cycloalkyl, cycloheteroalkyl, aryl, heteroaryl, heteroalkyl or the like.
[0067] As used herein, the term "cycloheteroalkyl" refers to a stable heterocyclic non- aromatic ring and fused rings containing one or more heteroatoms independently selected from N, O and S. A fused heterocyclic ring system may include carbocyclic rings and need only include one heterocyclic ring. Examples of heterocyclic rings include, but are not limited to, piperazinyl, homopiperazinyl, piperidinyl and morpholinyl, and are shown in the following illustrative examples:
Figure imgf000015_0004
wherein each X is selected from CR 2, NR , O and S; and each Y is selected from NR , O and S; and R58 is independently hydrogen, alkyl, cycloalkyl, cycloheteroalkyl, aryl, heteroaryl, heteroalkyl or the like. These cycloheteroalkyl rings may be optionally substituted with one or more groups selected from the group consisting of acyl, acylamino, acyloxy, alkoxy, substituted alkoxy, alkoxycarbonyl, alkoxycarbonylamino, amino, substituted amino, aminocarbonyl, aminocarbonylamino, aminocarbonyloxy, aryl, aryloxy, azido, carboxyl, cyano, cycloalkyl, substituted cycloalkyl, halogen, hydroxyl, keto, nitro, thioalkoxy, substituted thioalkoxy, thioaryloxy, thioketo, thiol, alkyl-S(O)-, aryl-S(O)-, alkyl-S(O)2- and aryl-S(O)2-. Substituting groups include carbonyl or thiocarbonyl which provide, for example, lactam and urea derivatives. [0068] Examples of representative cycloheteroalkenyls include the following:
Figure imgf000016_0001
Figure imgf000016_0002
wherein each X is selected from CR58 2, NR58, O and S; and each Y is selected from carbonyl, N, NR58, O and S; and R58 is independently hydrogen, alkyl, cycloalkyl, cycloheteroalkyl, aryl, heteroaryl, heteroalkyl or the like.
[0069] Examples of representative aryl having hetero atoms containing substitution include the following:
Figure imgf000016_0003
wherein each X is selected from C-R58 2, NR58, O and S; and each Y is selected from carbonyl, NR58, O and S; and R58 is independently hydrogen, alkyl, cycloalkyl, cycloheteroalkyl, aryl, heteroaryl, heteroalkyl or the like.
[0070] One having ordinary skill in the art of organic synthesis will recognize that the maximum number of heteroatoms in a stable, chemically feasible heterocyclic ring, whether it is aromatic or non aromatic, is determined by the size of the ring, the degree of unsaturation and the valence of the heteroatoms. In general, a heterocyclic ring may have one to four heteroatoms so long as the heteroaromatic ring is chemically feasible and stable.
[0071] "Pharmaceutically acceptable" means approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopoeia or other generally recognized pharmacopoeia for use in animals, and more particularly in humans.
[0072] "Pharmaceutically acceptable salt" refers to the non-toxic, inorganic and organic acid addition salts, and base addition salts, of compounds of the present invention, in particular they are pharmaceutically acceptable and possess the desired pharmacological activity of the parent compound. These salts can be prepared in situ during the final isolation and purification of compounds useful in the present invention. Such salts include: (1) acid addition salts, formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like; or formed with organic acids such as acetic acid, propionic acid, hexanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, 3-(4-hydroxybenzoyl) benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, 1,2-ethane-disulfonic acid, 2- hydroxyethanesulfonic acid, benzenesulfonic acid, 4-chlorobenzenesulfonic acid, 2- naphthalenesulfonic acid, 4-toluenesulfonic acid, camphorsulfonic acid, 4-methylbicyclo[2.2.2]-oct-2- ene-1-carboxylic acid, glucoheptonic acid, 3-phenylpropionic acid, trimethylacetic acid, tertiary butylacetic acid, lauryl sulfuric acid, gluconic acid, glutamic acid, hydroxynaphthoic acid, salicylic acid, stearic acid, muconic acid, and the like; or (2) salts formed when an acidic proton present in the parent compound either is replaced by a metal ion, e.g., an alkali metal ion, an alkaline earth ion, or an aluminum ion; or coordinates with an organic base such as ethanolamine, diethanolamine, triethanolamine, N-methylglucamine and the like. Salts further include, by way of example only, sodium, potassium, calcium, magnesium, ammonium, tetraalkylammonium, and the like; and when the compound contains a basic functionality, salts of non toxic organic or inorganic acids, such as hydrochloride, hydrobromide, tartrate, mesylate, acetate, maleate, oxalate and the like. The term "pharmaceutically acceptable cation" refers to a non toxic, acceptable cationic counter-ion of an acidic functional group. Such cations are exemplified by sodium, potassium, calcium, magnesium, ammonium, tetraalkylammonium cations, and the like.
[0073] "Pharmaceutically acceptable vehicle" refers to a diluent, adjuvant, excipient or carrier with which a compound of the invention is administered.
[0074] "Preventing" or "prevention" refers to a reduction in risk of acquiring a disease or disorder (i.e., causing at least one of the clinical symptoms of the disease not to develop in a subject that may be exposed to or predisposed to the disease but does not yet experience or display symptoms of the disease).
[0075] "Prodrugs" refers to compounds, including derivatives of the compounds of the invention, which have cleavable groups and become by solvolysis or under physiological conditions the compounds of the invention which are pharmaceutically active in vivo. Such examples include, but are not limited to, choline ester derivatives and the like, N-alkylmorpholine esters and the like. [0076] "Subject" includes humans. The terms "human," "patient" and "subject" are used interchangeably herein..
[0077] "Treating" or "treatment" of any disease or disorder refers, in one embodiment, to ameliorating the disease or disorder (i.e., arresting or reducing the development of the disease or at least one of the clinical symptoms thereof). In another embodiment "treating" or "treatment" refers to ameliorating at least one physical parameter, which may not be discernible by the subject. In yet another embodiment, "treating" or "treatment" refers to modulating the disease or disorder, either physically, {e.g., stabilization of a discernible symptom), physiologically, {e.g., stabilization of a physical parameter), or both. In yet another embodiment, "treating" or "treatment" refers to delaying the onset of the disease or disorder.
[0078] Other derivatives of the compounds of this invention have activity in both their acid and acid derivative forms, but in the acid sensitive form often offers advantages of solubility, tissue compatibility, or delayed release in the mammalian organism (see, Bundgard, H., Design of Prodrugs, pp. 7-9, 21-24, Elsevier, Amsterdam 1985). Prodrugs include acid derivatives well know to practitioners of the art, such as, for example, esters prepared by reaction of the parent acid with a suitable alcohol, or amides prepared by reaction of the parent acid compound with a substituted or unsubstituted amine, or acid anhydrides, or mixed anhydrides. Simple aliphatic or aromatic esters, amides and anhydrides derived from acidic groups pendant on the compounds of this invention are preferred prodrugs. In some cases it is desirable to prepare double ester type prodrugs such as (acyloxy)alkyl esters or ((alkoxycarbonyl)oxy)alkylesters. Preferred are the Ci to Cg alkyl, C2-C8 alkenyl, aryl, C7-C 12 substituted aryl, and C7-Ci2 arylalkyl esters of the compounds of the invention. [0079] As used herein, the term "isotopic variant" refers to a compound that contains unnatural proportions of isotopes at one or more of the atoms that constitute such compound. For example, an "isotopic variant" of a compound can contain one or more non-radioactive isotopes, such as for example, deuterium (2H or D), carbon-13 (13C), nitrogen-15 (15N), or the like. It will be understood that, in a compound where such isotopic substitution is made, the following atoms, where present, may vary, so that for example, any hydrogen may be 2HZO, any carbon may be 13C, or any nitrogen may be 15N, and that the presence and placement of such atoms may be determined within the skill of the art. Likewise, the invention may include the preparation of isotopic variants with radioisotopes, in the instance for example, where the resulting compounds may be used for drug and/or substrate tissue distribution studies. The radioactive isotopes tritium, i.e. 3H, and carbon-14, i.e. 14C, are particularly useful for this purpose in view of their ease of incorporation and ready means of detection. Further, compounds may be prepared that are substituted with positron emitting isotopes, such as 11C, 18F, 15O and 13N, and would be useful in Positron Emission Topography (PET) studies for examining substrate receptor occupancy.
[0080] All isotopic variants of the compounds provided herein, radioactive or not, are intended to be encompassed within the scope of the invention.
[0081] It is also to be understood that compounds that have the same molecular formula but differ in the nature or sequence of bonding of their atoms or the arrangement of their atoms in space are termed "isomers". Isomers that differ in the arrangement of their atoms in space are termed "stereoisomers".
[0082] Stereoisomers that are not mirror images of one another are termed "diastereomers" and those that are non-superimposable mirror images of each other are termed "enantiomers". When a compound has an asymmetric center, for example, it is bonded to four different groups, a pair of enantiomers is possible. An enantiomer can be characterized by the absolute configuration of its asymmetric center and is described by the R- and S-sequencing rules of Cahn and Prelog, or by the manner in which the molecule rotates the plane of polarized light and designated as dextrorotatory or levorotatory (i.e., as (+) or (-)-isomers respectively). A chiral compound can exist as either individual enantiomer or as a mixture thereof. A mixture containing equal proportions of the enantiomers is called a "racemic mixture".
[0083] "Tautomers" refer to compounds that are interchangeable forms of a particular compound structure, and that vary in the displacement of hydrogen atoms and electrons. Thus, two structures may be in equilibrium through the movement of π electrons and an atom (usually H). For example, enols and ketones are tautomers because they are rapidly interconverted by treatment with either acid or base. Another example of tautomerism is the aci- and nitro- forms of phenylnitromethane, that are likewise formed by treatment with acid or base.
[0084] Tautomeric forms may be relevant to the attainment of the optimal chemical reactivity and biological activity of a compound of interest.
[0085] The compounds of this invention may possess one or more asymmetric centers; such compounds can therefore be produced as individual (R)- or (S)- stereoisomers or as mixtures thereof.
Unless indicated otherwise, the description or naming of a particular compound in the specification and claims is intended to include both individual enantiomers and mixtures, racemic or otherwise, thereof. The methods for the determination of stereochemistry and the separation of stereoisomers are well-known in the art.
THE COMPOUNDS
[0086] The present invention is based on the discovery of that MAPKAPK5 functions in the pathway that results in the expression of MMPl, and that inhibitors of MAPKAPK5 activity, such as the compounds of the present invention, are useful for the treatment of diseases involving the abnormally high expression of MMP activity.
[0087] The compounds of the present invention may be described generally as
[l,2,4]triazolo[l,5-a]pyrazine-8-yl-amines substituted in the 5-position by an aryl and heteroaryl group, and an in the 8-position by an arylamino or a heteroarylamino group.
[0088] More particularly, the present invention relates to compounds having matrix metallo proteinase inhibiting properties in a mammalian cell, according to formula (I):
Figure imgf000019_0001
wherein:
A and B are independently CR2R", NR", oxygen or sulphur;
AA is CR2 or N;
D is C=O, CR2R" or NR"; E is NH or CR"R6, when k is zero, and is NH or CR"R6a, when k is one;
F is sulphur, oxygen or NH;
T is oxygen or NR;
U, V, W and X are independently CR"R7 or NR"; Y is CR" or N;
Z is hydrogen, amino, hydroxyl, lower alkoxy, carbamoyl, carboxyl, SO2RZ, SO2NRRZ, -
NR(CO)(CH2)d-Rz, -NRRz, -(CO)-ORz, -(CO)-NR(CH2)d-Rz, or
Figure imgf000020_0001
R is independently hydrogen or lower alkyl;
R" is H or forms a double bond with an adjacent atom;
Rl is H; R2; or lower alkyl, lower cycloalkyl and lower alkyl-lower cycloalkyl, optionally substituted with one or more R2;
R3 is H or forms a double bond with an adjacent R"; R2 is H, F, Cl; CN; COOR4; 0R4; C(O)N(R4R5); S(O)2N(R4R5); lower alkyl; O- lower alkyl;
NH-lower alkyl; S-lower alkyl; COO- lower alkyl; OC(O)- lower alkyl; C(O)N(R4)- lower alkyl; S(O)2N(R4)-lower alkyl; S(O)N(R4)-lower alkyl; S(O)2-lower alkyl; S(O)-lower alkyl;
N(R4)S(O)2-lower alkyl; and N(R4)S(O)-lower alkyl; wherein each lower alkyl is optionally substituted with one or more of F and Cl; R4 and R5 are independently
H; F, Cl; or lower alkyl, lower cycloalkyl, or lower alkyl-lower cycloalkyl optionally substituted with one or more of F and Cl;
R6 is hydrogen, amino, hydroxyl, carbamoyl, carboxyl, SO2R, NRR', -(CO)-OR, or -(CO)-NRR'; R6a is R6, Cl, F, lower alkoxy, cyano, trifluoromethoxy; or together with the adjacent be -
(CHR")n-NR-(CHR")p-, and form a five or six member heterocyclic ring fused to the ring to which they are bonded;
R7 is independently hydrogen, halogen, lower alkyl or lower alkoxy; Rz is hydrogen, lower alkyl, lower alkanoyl, phenyl, 1-loweralkyl pyrrolidin-3-yl, pyrazol-4-yl, pyrazol-2-yl, or lower alkyl, lower alkanoyl, phenyl, 1-loweralkyl pyrrolidin-3-yl, pyrazol-4- yl, pyrazol-2-yl or pyrid-3-yl substituted by one or more of hydroxyl, amino, mono- or di- loweralkylamino, acetamidyl, lower alkanoyl, lower alkyl, 4-hydroxy-phenyl, 3- aminomethylphenyl, lower alkyl sulfonyl, 4-diloweralkylaminophenyl, pyrid-3-yl, lH-indol-
3-yl, morpholin-4-yl,; R and Rz together may be -(CHR)q-T-(CHR)r- and form a five or six member heterocyclic ring with the nitrogen to which they are bonded; Rz and R7 together may be -(CHR")n-NR-(CHR")p-, and form a five or six member heterocyclic ring fused to the ring to which they are bonded; b and d are independently 0 or 1; provided at least one of b or d is 1; k is 0 or 1 ; m is 0 or 1 ; n and p are independently 0, 1 or 2; q and r are 1 or 2; x is o or 1 ; with the provisos that at least one of R7 or Rz is other than hydrogen; or a pharmaceutically acceptable salt, hydrate, solvate or prodrug thereof.
[0089] A preferred aspect of the present invention is a subclass of compounds according to formula II,
Figure imgf000021_0001
wherein
A and B are independently CR2R", NR", oxygen or sulphur;
AA is CR2 or N;
D is C=O, CR2R" or NR";
E is NH or CR"R6, when k is zero, and is NH or CR"R6a, when k is one;
F is sulphur, oxygen or NH;
T is oxygen or NR;
R" is H or forms a double bond with an adjacent atom;
R is independently hydrogen or lower alkyl;
Rl is H; R2; or lower alkyl, lower cycloalkyl and lower alkyl-lower cycloalkyl, optionally substituted with one or more R2; R2 is H, F, Cl; CN; COOR4; OR4; C(O)N(R4R5); S(O)2N(R4R5); lower alkyl; O- lower alkyl;
NH-lower alkyl; S-lower alkyl; COO- lower alkyl; OC(O)- lower alkyl; C(O)N(R4)- lower alkyl; S(O)2N(R4)-lower alkyl; S(O)N(R4)-lower alkyl; S(O)2-lower alkyl; S(O)-lower alkyl;
N(R4) S (O)2- lower alkyl; and N(R4)S(O)-lower alkyl; wherein each lower alkyl is optionally substituted with one or more of F and Cl; R4 and R5 are independently H; F, Cl; or lower alkyl, lower cycloalkyl, or lower alkyl-lower cycloalkyl optionally substituted with one or more of F and Cl;
R6 is hydrogen, amino, hydroxyl, carbamoyl, carboxyl, SO2R, NRR', -(CO)-OR, or -(CO)-NRR'; R6a is R6, Cl, F, lower alkoxy, cyano, trifluoromethoxy; or together with the adjacent be - (CHR")n-NR-(CHR")p-, and form a five or six member heterocyclic ring fused to the ring to which they are bonded;
R8 is phenyl independently substituted by Ra in the ortho-position, by Rb in the meta-position, and by Rc in the para-position; pyrid-3-yl; pyrid-3-yl substituted by Rc in the 5 -position; or cyclohexyl independently substituted by Ra in the 2-position, and by Rd in the 4-position; Ra is hydrogen, halogen, lower alkyl, trifluoromethyl or lower alkoxy; Rb is hydrogen, trifluoromethyl, lower alkoxy, lower alkyl sulfonamide, carboxyl, -
NReRf, -(CO)-OR or -(C0)-NReRf; Rc is hydrogen, amino, hydroxyl, lower alkoxy, carbamoyl, carboxyl, SO2R, SO2NReRf,
NReRf, -(CO)-OR, or -(C0)-NReRf; or
Rb and Rc can together form a benzdiazole, or indole substituted in the 3 -position by R'; Rd is hydroxyl, halogen, amino, lower alkoxy, or NReRf;
Re and Rf are independently hydrogen, 1-loweralkyl pyrrolidin-3-yl, 1 -R-pyrazol-4-yl, lower alkanoyl, phenyl, or lower alkyl optionally substituted by one or more of 4-hydroxy- phenyl, 3-aminomethylphenyl, lower alkyl sulfonyl, 4-diloweralkylaminophenyl, pyrid-3-yl, lH-indol-3-yl, morpholin-4-yl, hydroxyl, amino, mono- or di- loweralkylamino, or by lower alkanoyl; or R' and R" together are -(CHR)n-T- (CHR)n- and form a five or six member heterocyclic ring with the nitrogen to which they are bonded; m is O, 1, or 2; n is 1 or 2; with the proviso that at least one of Ra, Rb and Rc is other than hydrogen; or a pharmaceutically acceptable salt, hydrate, solvate or prodrug thereof.
[0090] A particularly preferred embodiment of the present invention relates to compounds according to formula III:
Figure imgf000022_0001
(III) wherein R1 is H, or substituted or unsubstituted alkyl; and each of R8 and R9 is independently selected from substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl; or a pharmaceutically acceptable salt, solvate or prodrug thereof; and stereoisomers, isotopic variants and tautomers thereof. [0091] In one embodiment, with respect to compounds of formula III, R8 is selected from substituted or unsubstituted cyclopentyl, cyclohexyl, substituted or unsubstituted phenyl, substituted or unsubstituted pyridyl, substituted or unsubstituted pyrimidine, and substituted or unsubstituted pyrazine, substituted or unsubstituted pyrrole, substituted or unsubstituted pyrazole and substituted or unsubstituted imidazole.
[0092] In one embodiment, with respect to compounds of formula III, R1 is H, Me, Et, i-Pr or
CF3.
[0093] In one embodiment, with respect to compounds of formula III, R1 is H.
[0094] In one embodiment, with respect to compounds of formula III, R8 is selected from substituted or unsubstituted cycloalkyl.
[0095] In another embodiment, with respect to compounds of formula III, R8 is selected from substituted or unsubstituted cyclohexyl or cyclopentyl.
[0096] In one embodiment, with respect to compounds of formula III, R8 is selected from substituted or unsubstituted heterocycloalkyl.
[0097] In another embodiment, with respect to compounds of formula III, R8 is selected from substituted or unsubstituted piperidinyl, morpholinyl, or pyrrolidinyl.
[0098] In one embodiment, with respect to compounds of formula III, R8 is selected from substituted or unsubstituted phenyl, pyridyl or pyrimidine.
[0099] In one embodiment, with respect to compounds of formula III, R8 is selected from substituted phenyl, substituted pyridyl, and substituted pyrimidine; and the substitution is -L-R8d; and wherein L is selected from a bond, alkylene, heteroalkylene, -O-, -N(R8e)-, -CO-, -CO2-, -SO-,
-SO2-, -CON(R8e)-, -SO2N(R8e)-, -N(R8e)CO-, -N(R8e)SO2-, -N(R8e)CO N(R8e)-, -
N(R8e)SO2 N(R8e)-; and
R8d is selected from substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted heteroaryl, substituted or unsubstituted amino, substituted or unsubstituted aralkyl, substituted or unsubstituted heteroarylalkyl and substituted or unsubstituted aminoalkyl; and
R8e is selected from H, substituted or unsubstituted alkyl and substituted or unsubstituted cycloalkyl. [00100] In one embodiment, with respect to compounds of formula III, R8 is (R8a)n
Figure imgf000024_0001
wherein L, and R are as described in the preceding paragraph; the subscript n is selected from 1-4; and each R8a is independently selected from hydrogen, substituted or unsubstituted alkyl, alkoxy, cyano, and halo.
[00101] In one embodiment, with respect to compounds of formula III, R8 is as described above and the subscript n is 1 and R8a is Me, Et, Pr, iso-Pr, Cl, F, CN, OMe, or CF3. In another embodiment, R8a is at 2-(ortho to -L) position. In yet another embodiment, R8a is 2-Cl, 2-F, 2-Me or 2-
CF3.
[00102] In one embodiment, with respect to compounds of formula III, R8 is as described above and
L is a bond, -O-, -CO-, -CON(R8e)-, or -N(R8e)CO-;
R8d is selected from substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted heteroaryl, substituted or unsubstituted aralkyl, substituted or unsubstituted heteroarylalkyl and substituted or unsubstituted aminoalkyl; and
R8e is selected from H, substituted or unsubstituted alkyl.
[00103] In one embodiment, with respect to compounds of formula III, R8 is as described above and
L is a bond, -O-, -CO-, -CON(R8e)-, or -N(R8e)CO-; and
R8d is selected from H, alkylaminoethyl, dialkylaminoethyl, cycloalkyl, heterocycloalkyl, arylalkyl, and heteroarylalkyl.
[00104] In one embodiment, with respect to compounds of formula III, R8 is as described above and
L is a bond, -O-, -CO-, -CON(R8e)-, or -N(R8e)CO-; and
R8d is selected from methylaminoethyl, ethylaminoethyl, dimethylaminoethyl, diethylaminoethyl, substituted or unsubstituted pyrrolidinyl, benzyl and pyridylmethyl.
[00105] In one embodiment, with respect to compounds of formula III, R8 is as described above and
L is a bond, -CO-, SO2, -(CH2)ml-, -O(CH2)ml-, -NH(CH2)ml-, -CON(H)(CH2)ml-, or - SO2NH(CH2)mr; the subscript ml is selected from 1-4; and R8d is
Figure imgf000024_0002
and wherein the ring P is substituted or unsubstituted heterocycloalkyl. In another embodiment, L is a bond, -CO-, -O(CH2)ml-, -CON(H)(CH2)ml-, or - NHCO-; [00106] In one embodiment, with respect to compounds of formula III, R8 is as described above; L is a bond; and the ring P is substituted or unsubstituted heterocycloalkyl. [00107] In one embodiment, with respect to compounds of formula III, R8 is as described above; L is a bond; and the ring P is substituted or unsubstituted piperidine, substituted or unsubstituted piperazine, and substituted or unsubstituted piperidine, morpholine. [00108] In one embodiment, with respect to compounds of formula III, R8 is as described above; L is CO; and the ring P is substituted or unsubstituted heterocycloalkyl.
[00109] In one embodiment, with respect to compounds of formula III, R8 is as described above; L is CO; and the ring P is substituted or unsubstituted piperidine, substituted or unsubstituted piperazine, and substituted or unsubstituted piperidine, morpholine.
[00110] In one embodiment, with respect to compounds of formula III, R8 is as described above; L is -(CH2)mr, -O(CH2)mr, or -NH(CH2)mr; the subscript ml is selected from 1-4; and the ring P is substituted or unsubstituted heterocycloalkyl.
[00111] In one embodiment, with respect to compounds of formula III, R8 is as described above; L is -(CH2)mr, -O(CH2)mr, or -NH(CH2)mr; the subscript ml is 2 or 3; and the ring P is substituted or unsubstituted piperidine, substituted or unsubstituted piperazine, and substituted or unsubstituted piperidine, morpholine.
[00112] In one embodiment, with respect to compounds of formula III, R8 is as described above; L is -CON(H)(CH2)mr, or -NHCO(CH2)mr; the subscript ml is selected from 1-4; and the ring P is substituted or unsubstituted heterocycloalkyl.
[00113] In one embodiment, with respect to compounds of formula III, R8 is as described above; L is -CON(H)(CH2)mr; the subscript ml is 2 or 3; and the ring P is substituted or unsubstituted piperidine, substituted or unsubstituted piperazine, and substituted or unsubstituted piperidine, morpholine.
[00114] In one embodiment, with respect to compounds of formula III, the compound is according to formula IVa, IVb, IVc, or IVd:
Figure imgf000025_0001
IVa IVb IVc or IVd
and wherein L and the ring P are as described above; the subscript n, is selected from 1-4; each R > 8aa i s independently selected from hydrogen, substituted or unsubstituted alkyl, alkoxy, cyano, and halo; and R9 is independently selected from substituted or unsubstituted aryl and heteroaryl; or a pharmaceutically acceptable salt, solvate or prodrug thereof; and stereoisomers, isotopic variants and tautomers thereof.
[00115] In one embodiment, with respect to compounds of formulae IVa-IVd, L is a bond.
[00116] In one embodiment, with respect to compounds of formulae IVa-IVd, L is methylene, ethylene, propylene, and butylene.
[00117] In one embodiment, with respect to compounds of formulae IVa-IVd, L is -CO-.
[00118] A compound according to Claim 9, wherein L is -CO-.
[00119] A compound according to Claim 9, wherein L is -NHCO- or -CONH-.
[00120] A compound according to Claim 9, wherein L is -CON(H)-CH2-CH2-, or -N(H)-CO-
CH2-CH2-.
[00121] A compound according to Claim 9, wherein L is -OCH2-CH2- or -NHCH2-CH2-.
[00122] In one embodiment, with respect to compounds of formulae IVa-IVd, L is -SO2-.
[00123] In one embodiment, with respect to compounds of formulae IVa-IVd, L is -CON(H)-
CH2-CH2-, or - SO2NH-CH2-CH2-.
[00124] In one embodiment, with respect to compounds of formulae IVa-IVd, L is -OCH2-
CH2- or -NHCH2-CH2-.
[00125] In a preferred embodiment L is a bond.
[00126] In one embodiment, with respect to compounds of formulae IVa-IVd, the ring P is substituted or unsubstituted piperidine, morpholine or piperazine.
[00127] In one embodiment, with respect to compounds of formulae IVa-IVd, L and the ring P are as described above; the subscript n is 4 and each R8a is H.
[00128] In one embodiment, with respect to compounds of formulae IVa-IVd, L and the ring P are as described above; the subscript n is 1 and R8a is Me, Et, Pr, iso-Pr, Cl, F, CN, OMe, or CF3. In another embodiment, R8a is at 2-(ortho to -L) position. In yet another embodiment, R8a is 2-Cl, 2-F, 2-
Me or 2-CF3.
[00129] In a further embodiment, with respect to compounds of formula III, R8 is
Figure imgf000026_0001
and wherein the ring P is substituted or unsubstituted heterocycloalkyl; the subscript n is selected from
1 -4 and each R8a is independently selected from hydrogen, substituted or unsubstituted alkyl, alkoxy, cyano, and halo.
[00130] In one embodiment, with respect to compounds of formula III, R8 is as described above and the ring P is substituted or unsubstituted piperidine, morpholine or piperazine.
[00131] In one embodiment, with respect to compounds of formula III, R8 is as described above and the subscript n is 4 and each R8a is H. [00132] In one embodiment, with respect to compounds of formula III, R8 is as described above and the subscript n is 1 and R8a is Me, Et, Pr, iso-Pr, Cl, F, CN, OMe, or CF3. In another embodiment, R8a is at the 2-(ortho to N-ring P) position. In yet another embodiment, R8a is 2-Cl, 2-F, 2-Me or 2-CF3.
[00133] In a further embodiment, with respect to compounds of formula III, R 8 i-s
Figure imgf000027_0001
and wherein the subscript n is selected from 1-4; each R8a is independently selected from hydrogen, substituted or unsubstituted alkyl, alkoxy, cyano, and halo; R8b is hydrogen, substituted or unsubstituted alkyl or substituted or unsubstituted cycloalkyl; R8c is hydrogen, substituted or unsubstituted alkyl and the subscript x is selected from 1-8.
[00134] In a further embodiment, with respect to compounds of formula III, R 8 i-s
Figure imgf000027_0002
and wherein the subscript n is selected from 1-4; each R8a is independently selected from hydrogen, substituted or unsubstituted alkyl, alkoxy, cyano, and halo; R 8b is hydrogen, substituted or unsubstituted alkyl or substituted or unsubstituted cycloalkyl; R c is hydrogen or
Me.
[00135] In a further embodiment, with respect to compounds of formula III, R 8 i-s
Figure imgf000027_0003
and wherein the subscript n is selected from 1-4; each R8a is independently selected from hydrogen, substituted or unsubstituted alkyl, alkoxy, cyano, and halo.
[00136] In a further embodiment, with respect to compounds of formula III, R 8 i-s
Figure imgf000028_0001
and wherein the subscript n is selected from 1-4; each R8a is independently selected from hydrogen, substituted or unsubstituted alkyl, alkoxy, cyano, and halo; and R8b is hydrogen, substituted or unsubstituted alkyl or substituted or unsubstituted cycloalkyl.
[00137] In one embodiment, with respect to compounds of formula III, R8 is as described above and the subscript n is 4 and each R8a is H.
[00138] In one embodiment, with respect to compounds of formula III, R8 is as described above and the subscript n is 1 and R8a is Me, Et, Pr, iso-Pr, Cl, F, CN, OMe, or CF3. In another embodiment, R8a is at 2-(ortho to N-ringP) position. In yet another embodiment, R8a is 2-Cl, 2-F, 2-Me or 2-CF3..
[00139] In one embodiment, with respect to compounds of formula III, R8 is as described above and R8b is H.
[00140] In one embodiment, with respect to compounds of formula III, R8 is as described above and R8b is substituted or unsubstiuted alkyl.
[00141] In one embodiment, with respect to compounds of formula III, R8 is as described above and R8b is substituted or unsubstituted cycloalkyl.
[00142] In one embodiment, with respect to compounds of formula III, R8 is as described above and R8b is Me, Et, Pr, i-Pr, t-Bu, i-Bu, CH2CF3, CF3, CH2CONH2, cyclopropyl or cyclopropylmethyl.
[00143] In one particular embodiment, with respect to compounds of formula III, R8 is as described above and R8b is i-Pr.
[00144] In a further embodiment, with respect to compounds of formulae IVa-IVd, R8a is independently selected from hydrogen, substituted or unsubstituted alkyl, alkoxy, cyano, carbamoyl,
CHO, and halo. In one embodiment, R8a is H, Me, F, or Cl. In a preferred embodiment R8a is H.
[00145] In one embodiment, with respect to compounds of formulae IVa-IVd, the compound is according to formula Va, Vb, Vc, Vd, Ve, or Vf:
Figure imgf000029_0001
Va Vb Vc
Figure imgf000029_0002
Vd Ve Vf and wherein R9 is as described for formula III and R8b is hydrogen, substituted or unsubstituted alkyl or substituted or unsubstituted cycloalkyl.
[00146] In one embodiment, with respect to compounds of formulae Va-Vf, R8b is H.
[00147] In one embodiment, with respect to compounds of formulae Va-Vf, R8b is substituted or unsubstiuted alkyl.
[00148] In one embodiment, with respect to compounds of formulae Va-Vf, R8b is substituted or unsubstituted cycloalkyl.
[00149] In one embodiment, with respect to compounds of formulae Va-Vf, R8b is Me, Et, Pr, i-Pr, t-Bu, i-Bu, CH2CF3, CF3, CH2CONH2, cyclopropyl or cyclopropylmethyl.
[00150] In one particular embodiment, with respect to compounds of formulae Va-Vf, R8b is i-
Pr.
[00151] In a one embodiment, with respect to compounds of formulae III- Vf, R9 is substituted or unsubstituted aryl. In another embodiment, R9 is substituted or unsubstituted phenyl. [00152] In a one embodiment, with respect to compounds of formulae III- Vf, R9 is substituted or unsubstituted heteroaryl. In another embodiment, R9 is substituted or unsubstituted pyridyl. [00153] In a one embodiment, with respect to compounds of formulae III- Vf, R9 is selected from substituted or unsubstituted phenyl, indolyl, isoinolyl, pyrrolyl, furanyl, thienyl, pyrazolyl, oxazolyl, and thiazolyl. [00154] In one embodiment, with respect to compounds of formulae III- Vf, R9 is
Figure imgf000029_0003
and each of A1, A2 and A3 is independently selected from S, O, N, NR9a, and CR9a; each of R9a is independently H or substituted or unsubstituted alkyl; and R9b is CONH2, CONHMe, or CN. [00155] In further embodiment, with respect to compounds of formulae III- Vf, R9 is
Figure imgf000030_0001
[00156] In further embodiment, with respect to compounds of formulae III- Vf, R9 is
Figure imgf000030_0002
Figure imgf000030_0003
[00157] In further embodiment, with respect to compounds of formulae III- Vf, R is
Figure imgf000030_0004
and wherein the subscript m is selected from 1-4 and each R9d is independently H, substituted or unsubstituted alkyl or halo.
[00158] In further embodiment, with respect to compounds of formulae III- Vf, R9 is
Figure imgf000030_0005
and wherein the subscript m is selected from 1 -4 and each R9d is independently H, substituted or unsubstituted alkyl or halo. [00159] In a further embodiment, with respect to compounds of formulae III- Vf, R9 is
Figure imgf000031_0001
and wherein the subscript m is selected from 1-3 and each R9d is independently H, substituted or unsubstituted alkyl or halo.
[00160] In a further embodiment, with respect to compounds of formulae III- Vf, R9 is as described above; and each R9d is H.
[00161] In a further embodiment, with respect to compounds of formulae III- Vf, R9 is as described above; m is 1 or 2 and each R9d is independently Me, Cl or F.
[00162] In one embodiment, with respect to compounds of formula III, the compound is according to formula Via, VIb, Vic, VId, VIe or VIf:
Figure imgf000031_0002
Via VIb VIc
Figure imgf000031_0003
and R > 8b i s hydrogen, substituted or unsubstituted alkyl or substituted or unsubstituted cycloalkyl.
[00163] In a further embodiment, with respect to compounds of formulae VIa-VIf, R8b is H.
[00164] In a further embodiment, with respect to compounds of formulae VIa-VIf, R8b is cycloalkyl.
[00165] In a further embodiment, with respect to compounds of formulae VIa-VIf, R8b is cyclopropyl. [00166] In a further embodiment, with respect to compounds of formulae VIa-VIf, R8b is substituted or unsubstituted alkyl.
[00167] In a further embodiment, with respect to compounds of formulae VIa-VIf, R8b is Me,
Et, Pr, i-Pr, t-Bu, i-Bu, CF3, CH2CF3, CH2CONH2, or cyclopropylmethyl. [00168] In one embodiment, with respect to compounds of formula III, the compound is according to formula Vila, VIIb, VIIc, VIIc, VIId, VIIe or VIIf:
Figure imgf000032_0001
and R8b is hydrogen, substituted or unsubstituted alkyl or substituted or unsubstituted cycloalkyl.
[00169] In a further embodiment, with respect to compounds of formulae VIIa-VIId, R8b is H.
[00170] In a further embodiment, with respect to compounds of formulae VIIa-VIIf, R8b is cycloalkyl.
[00171] In a further embodiment, with respect to compounds of formulae VIIa-VIIf, R8b is cyclopropyl.
[00172] In a further embodiment, with respect to compounds of formulae VIIa-VIIf, R8b is substituted or unsubstituted alkyl.
[00173] In a further embodiment, with respect to compounds of formulae VIIa-VIIf, R8b is Me,
Et, Pr, i-Pr, t-Bu, i-Bu, CF3, CH2CF3, CH2CONH2, or cyclopropylmethyl.
[00174] In one embodiment, with respect to compounds of formula III, the compound is according to formula Villa, VIIIb, VIIIc, VIIId, VIIIe or VIIIf:
Figure imgf000033_0001
Villa VIIIc
VIIIb
Figure imgf000033_0002
VIIIf VIIIe and R > 8b i s hydrogen, substituted or unsubstituted alkyl or substituted or unsubstituted cycloalkyl.
[00175] In a further embodiment, with respect to compounds of formulae VIIIa-VIIIf, R8b is H.
[00176] In a further embodiment, with respect to compounds of formulae VIIIa-VIIIf, R8b is cycloalkyl.
[00177] In a further embodiment, with respect to compounds of formulae VIIIa-VIIIf, R8b is cyclopropyl.
[00178] In a further embodiment, with respect to compounds of formulae VIIIa-VIIIf, R8b is substituted or unsubstituted alkyl.
[00179] In a further embodiment, with respect to compounds of formulae VIIIa-VIIIf, R8b is
Me, Et, Pr, i-Pr, t-Bu, i-Bu, CF3, CH2CF3, CH2CONH2, or cyclopropylmethyl.
[00180] In one embodiment, with respect to compounds of formula III, the compound is according to formula IXa, IXb, IXc, IXd, IXe, or IXf:
Figure imgf000034_0001
IXa
IXb IXc
Figure imgf000034_0002
and R > 8b i s hydrogen, substituted or unsubstituted alkyl or substituted or unsubstituted cycloalkyl.
[00181] In a further embodiment, with respect to compounds of formulae IXa-IXf, R8b is H.
[00182] In a further embodiment, with respect to compounds of formulae IXa-IXf, R8b is cycloalkyl.
[00183] In a further embodiment, with respect to compounds of formulae IXa-IXf, R8b is cyclopropyl.
[00184] In a further embodiment, with respect to compounds of formulae IXa-IXf, R8b is substituted or unsubstituted alkyl.
[00185] In a further embodiment, with respect to compounds of formulae IXa-IXf, R8b is Me,
Et, Pr, i-Pr, t-Bu, i-Bu, CF3, CH2CF3, CH2CONH2, or cyclopropylmethyl.
[00186] In one embodiment, with respect to compounds of formula III, the compound is according to formula Xa, Xb, Xc, Xd, Xe, or Xf:
Figure imgf000035_0001
Xa
Xb Xc
Figure imgf000035_0002
and R8b is hydrogen, substituted or unsubstituted alkyl or substituted or unsubstituted cycloalkyl.
[00187] In a further embodiment, with respect to compounds of formulae Xa-Xf, R8b is H.
[00188] In a further embodiment, with respect to compounds of formulae Xa-Xf, R8b is cycloalkyl.
[00189] In a further embodiment, with respect to compounds of formulae Xa-Xf, R8b is cyclopropyl.
[00190] In a further embodiment, with respect to compounds of formulae Xa-Xf, R8b is substituted or unsubstituted alkyl.
[00191] In a further embodiment, with respect to compounds of formulae Xa-Xf, R8b is Me, Et,
Pr, i-Pr, t-Bu, i-Bu, CF3, CH2CF3, CH2CONH2, or cyclopropylmethyl.
[00192] In one embodiment, with respect to compounds of formula III, the compound is according to formula XIa, XIb, XIc, XId, XIe or XIf:
Figure imgf000036_0001
XIa
XIb XIc
Figure imgf000036_0002
XId XIe XIf and R , 8b is hydrogen, substituted or unsubstituted alkyl or substituted or unsubstituted cycloalkyl; and
R9e is hydrogen, Me, or CN.
[00193] In one embodiment, with respect to compounds of formulae XIa-XIf, R9e is H.
[00194] In one embodiment, with respect to compounds of formulae XIa-XIf, R9e is Me. [00195] In one embodiment, with respect to compounds of formulae XIa-XIf, R9e is CN. [00196] In a further embodiment, with respect to compounds of formulae XIa-XIf, R8b is H. [00197] In a further embodiment, with respect to compounds of formulae XIa-XIf, R8b is cycloalkyl. [00198] In a further embodiment, with respect to compounds of formulae XIa-XIf, R8b is cyclopropyl.
[00199] In a further embodiment, with respect to compounds of formulae XIa-XIf, R8b is substituted or unsubstituted alkyl.
[00200] In a further embodiment, with respect to compounds of formulae XIa-XIf, R8b is Me,
Et, Pr, i-Pr, t-Bu, i-Bu, CF3, CH2CF3, CH2CONH2, or cyclopropylmethyl.
[00201] In one embodiment, with respect to compounds of formula III, the compound is according to formula XIIa, XIIb, XIIc or XIId:
Figure imgf000037_0001
XIIa
XIIb XlIc
[00202] In one embodiment, with respect to compounds of formula III, the compound is according to formula XIIIa, XIIIb, XIIIc or XIIId:
Figure imgf000037_0002
XIIIa
XIlIb X ΛIlIlIlcc o Uri XIIId
[00203] In one embodiment, with respect to compounds of formula III, the compound is according to formula XIVa, XIVb, XIVc or XIVd:
Figure imgf000037_0003
XIVa
XIVb XIVc XIVd
[00204] In one embodiment, with respect to compounds of formula III, the compound is according to formula XVa, XVb, or XIVc:
Figure imgf000038_0001
XVa XVb or XVc and L is a bond or -0-CH2-CH2- , the πng P is
Figure imgf000038_0002
and R8b is H, Me, i-Pr, t-Bu, CH2CONH2, cyclopropylmethyl, or CH2CF3.
[00205] In one particular embodiment, with respect to compounds of formulae XVa-XVc, L is a bond. In another particular embodiment, L is -0-CH2-CH2- .
[00206] In one particular embodiment, with respect to compounds of formulae XVa-XVc, the πng P is
Figure imgf000038_0003
[00207] In more particular embodiment, with respect to compounds of formulae XVa-XVc, the πng P is
Figure imgf000038_0004
[00208] In another embodiment, with respect to compounds of formula III, the compound is selected from Table 1
[00209] In another embodiment, with respect to compounds of formula III, the compound is selected from Table 2
[00210] In certain aspects, the present invention provides prodrugs and deπvatives of the compounds according to the formulae above Prodrugs are derivatives of the compounds of the invention, which have metabolically cleavable groups and become by solvolysis or under physiological conditions the compounds of the invention, which are pharmaceutically active, in vivo
A prodrug may be inactive when administered to a subject but is converted in vivo to an active compound of the invention. "Pharmaceutically acceptable prodrugs" as used herein refers to those prodrugs of the compounds useful in the present invention, which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of patients with undue toxicity, irritation, allergic response commensurate with a reasonable benefit/risk ratio, and effective for their intended use of the compounds of the invention. The term "prodrug" means a compound that is transformed in vivo to yield an effective compound useful in the present invention or a pharmaceutically acceptable salt, hydrate or solvate thereof. The transformation may occur by various mechanisms, such as through hydrolysis in blood. The compounds bearing metabolically cleavable groups have the advantage that they may exhibit improved bioavailability as a result of enhanced solubility and/or rate of absorption conferred upon the parent compound by virtue of the presence of the metabolically cleavable group, thus, such compounds act as pro-drugs. A thorough discussion is provided in Design of Prodrugs, H. Bundgaard, ed., Elsevier (1985); Methods in Enzymology; K. Widder et al, Ed., Academic Press, 42, 309-396 (1985); A Textbook of Drug Design and Development, Krogsgaard- Larsen and H. Bandaged, ed., Chapter 5; "Design and Applications of Prodrugs" 113-191 (1991); Advanced Drug Delivery Reviews, H. Bundgard, 8 , 1-38, (1992); J. Pharm. Sci., 77,285 (1988); Chem. Pharm. Bull., N. Nakeya et al, 32, 692 (1984); Pro-drugs as Novel Delivery Systems, T. Higuchi and V. Stella, 14 A.C.S. Symposium Series, and Bioreversible Carriers in Drug Design, E.B. Roche, ed., American Pharmaceutical Association and Pergamon Press, 1987, which are incorporated herein by reference. Such examples include, but are not limited to, choline ester derivatives and the like, N-alkylmorpholine esters and the like.
[00211] Other derivatives of the compounds of this invention have activity in both their acid and acid derivative forms, but the acid sensitive form often offers advantages of solubility, tissue compatibility, or delayed release in the mammalian organism (see, Bundgard, H., Design of Prodrugs, pp. 7-9, 21-24, Elsevier, Amsterdam 1985). Prodrugs include acid derivatives well know to practitioners of the art, such as, for example, esters prepared by reaction of the parent acid with a suitable alcohol, or amides prepared by reaction of the parent acid compound with a substituted or unsubstituted amine, or acid anhydrides, or mixed anhydrides. Simple aliphatic or aromatic esters, amides and anhydrides derived from acidic groups pendant on the compounds of this invention are preferred prodrugs. In some cases it is desirable to prepare double ester type prodrugs such as (acyloxy)alkyl esters or ((alkoxycarbonyl)oxy)alkylesters. Preferred are the Ci to Cg alkyl, C2-Cg alkenyl, aryl, C7-C12 substituted aryl, and C7-C12 arylalkyl esters of the compounds of the invention.
PHARMACEUTICAL COMPOSITIONS
[00212] When employed as pharmaceuticals, the compounds of this invention are typically administered in the form of a pharmaceutical composition. Such compositions can be prepared in a manner well known in the pharmaceutical art and comprise at least one active compound. [00213] Generally, the compounds of this invention are administered in a pharmaceutically effective amount. The amount of the compound actually administered will typically be determined by a physician, in the light of the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual compound -administered, the age, weight, and response of the individual patient, the severity of the patient's symptoms, and the like.
[00214] The pharmaceutical compositions of this invention can be administered by a variety of routes including oral, rectal, transdermal, subcutaneous, intravenous, intramuscular, and intranasal. Depending on the intended route of delivery, the compounds of this invention are preferably formulated as either injectable or oral compositions or as salves, as lotions or as patches all for transdermal administration.
[00215] The compositions for oral administration can take the form of bulk liquid solutions or suspensions, or bulk powders. More commonly, however, the compositions are presented in unit dosage forms to facilitate accurate dosing. The term "unit dosage forms" refers to physically discrete units suitable as unitary dosages for human subjects and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient. Typical unit dosage forms include prefilled, premeasured ampules or syringes of the liquid compositions or pills, tablets, capsules or the like in the case of solid compositions. In such compositions, the furansulfonic acid compound is usually a minor component (from about 0.1 to about 50% by weight or preferably from about 1 to about 40% by weight) with the remainder being various vehicles or carriers and processing aids helpful for forming the desired dosing form.
[00216] Liquid forms suitable for oral administration may include a suitable aqueous or nonaqueous vehicle with buffers, suspending and dispensing agents, colorants, flavors and the like. Solid forms may include, for example, any of the following ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or corn starch; a lubricant such as magnesium stearate; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring. [00217] Injectable compositions are typically based upon injectable sterile saline or phosphate- buffered saline or other injectable carriers known in the art. As before, the active compound in such compositions is typically a minor component, often being from about 0.05 to 10% by weight with the remainder being the injectable carrier and the like.
[00218] Transdermal compositions are typically formulated as a topical ointment or cream containing the active ingredient(s), generally in an amount ranging from about 0.01 to about 20% by weight, preferably from about 0.1 to about 20% by weight, preferably from about 0.1 to about 10% by weight, and more preferably from about 0.5 to about 15% by weight. When formulated as a ointment, the active ingredients will typically be combined with either a paraffmic or a water-miscible ointment base. Alternatively, the active ingredients may be formulated in a cream with, for example an oil-in- water cream base. Such transdermal formulations are well-known in the art and generally include additional ingredients to enhance the dermal penetration of stability of the active ingredients or the formulation. All such known transdermal formulations and ingredients are included within the scope of this invention.
[00219] The compounds of this invention can also be administered by a transdermal device.
Accordingly, transdermal administration can be accomplished using a patch either of the reservoir or porous membrane type, or of a solid matrix variety.
[00220] The above-described components for orally administrable, injectable or topically administrable compositions are merely representative. Other materials as well as processing techniques and the like are set forth in Part 8 of Remington's Pharmaceutical Sciences, 17th edition,
1985, Mack Publishing Company, Easton, Pennsylvania, which is incorporated herein by reference.
[00221] The compounds of this invention can also be administered in sustained release forms or from sustained release drug delivery systems. A description of representative sustained release materials can be found in Remington's Pharmaceutical Sciences.
[00222] The following formulation examples illustrate representative pharmaceutical compositions of this invention. The present invention, however, is not limited to the following pharmaceutical compositions.
Formulation 1 - Tablets
[00223] A compound of the invention is admixed as a dry powder with a dry gelatin binder in an approximate 1:2 weight ratio. A minor amount of magnesium stearate is added as a lubricant. The mixture is formed into 240-270 mg tablets (80-90 mg of active amide compound per tablet) in a tablet press.
Formulation 2 - Capsules
[00224] A compound of the invention is admixed as a dry powder with a starch diluent in an approximate 1 :1 weight ratio. The mixture is filled into 250 mg capsules (125 mg of active amide compound per capsule).
Formulation 3 - Liquid
[00225] A compound of the invention (125 mg), sucrose (1.75 g) and xanthan gum (4 mg) are blended, passed through a No. 10 mesh U.S. sieve, and then mixed with a previously made solution of microcrystalline cellulose and sodium carboxymethyl cellulose (11 :89, 50 mg) in water. Sodium benzoate (10 mg), flavor, and color are diluted with water and added with stirring. Sufficient water is then added to produce a total volume of 5 mL.
Formulation 4 - Tablets
[00226] A compound of the invention is admixed as a dry powder with a dry gelatin binder in an approximate 1 :2 weight ratio. A minor amount of magnesium stearate is added as a lubricant. The mixture is formed into 450-900 mg tablets (150-300 mg of active amide compound) in a tablet press.
Formulation 5 - Injection
[00227] A compound of the invention is dissolved or suspended in a buffered sterile saline injectable aqueous medium to a concentration of approximately 5 mg/ml. Formulation 6 - Topical
[00228] Stearyl alcohol (250 g) and a white petrolatum (250 g) are melted at about 75°C and then a mixture of a compound of the invention (50 g) methylparaben (0.25 g), propylparaben (0.15 g), sodium lauryl sulfate (10 g), and propylene glycol (120 g) dissolved in water (about 370 g) is added and the resulting mixture is stirred until it congeals.
METHODS OF TREATMENT
[00229] The present compounds are used as therapeutic agents for the treatment of conditions in mammals that are causally related or attributable to aberrant activity of MMPl and / or MAPKAPK5. Accordingly, the compounds and pharmaceutical compositions of this invention find use as therapeutics for preventing and/or treating inflammatory diseases in mammals including humans.
[00230] In a method of treatment aspect, this invention provides a method of treating a mammal susceptible to or afflicted with a condition associated with extra-cellular matrix (ECM) degradation, in particular arthritis, and more particularly, rheumatoid arthritis, which method comprises administering an effective amount of one or more of the compounds of the invention or a pharmaceutical composition just described.
[00231] In another method of treatment aspect, the invention provides a method of treating a mammal sucepible to or afflicted with a condition associated with an abnormal cellular expression of MMPl, which comprises administering a therapeutically effective amount of a compound of the invention, or a pharmaceutical composition thereof.
[00232] In another method of treatment aspect, the present invention provides a method of treatment or prophylaxis of a condition characterized by abnormal matrix metallo proteinase activity, which comprises administering a therapeutically effective matrix metallo proteinase inhibiting amount of one or more of the compounds of the invention, or pharmaceutical composition thereof. [00233] In yet another method of treatment aspect, this invention provides methods of treating a mammal susceptible to or afflicted with diseases and disorders which are mediated by or result in inflammation such as, for example rheumatoid arthritis and osteoarthritis, myocardial infarction, various autoimmune diseases and disorders, uveitis and atherosclerosis; itch / pruritus such as, for example psoriasis; and renal disorders method comprises administering an effective condition-treating or condition-preventing amount of one or more of the pharmaceutical compositions just described. [00234] This invention also relates to the use of the present compounds in the manufacture of a medicament for treatment or prophylaxis of a condition prevented, ameliorated or eliminated by administration of an inhibitor of Mitogen- Activated Protein Kinase- Activated Protein Kinase 5, or a condition characterised by abnormal collagenase activity, or a condition associated with ECM degradation or a condition selected from diseases involving inflammation, most preferably in for the treatment of rheumatoid arthritis.
[00235] As a further aspect of the invention there is provided the present compounds for use as a pharmaceutical especially in the treatment or prevention of the aforementioned conditions and diseases. Also provided herein is the use of the present compounds in the manufacture of a medicament for the treatment or prevention of one of the aforementioned conditions and diseases. [00236] A preferred regimen of the present method comprises the administration to a subject in suffering from a disease condition characterized by inflammatory, with an effective matrix metallo- protease inhibiting amount of a compound of the present invention for a period of time sufficient to reduce the abnormal levels of extracellular matrix degradation in the patient, and preferably terminate, the self-perpetuating processes responsible for said degradation. A special embodiment of the method comprises administering of an effective matrix metallo-protease inhibiting amount of a compound of the present invention to a subject patient suffering from or susceptible to the development of rheumatoid arthritis, for a period of time sufficient to reduce or prevent, respectively, collagen and bone degradation in the joints of said patient, and preferably terminate, the self-perpetuating processes responsible for said degradation.
[00237] Injection dose levels range from about 0.1 mg/kg/hour to at least 10 mg/kg/hour, all for from about 1 to about 120 hours and especially 24 to 96 hours. A preloading bolus of from about 0.1 mg/kg to about 10 mg/kg or more may also be administered to achieve adequate steady state levels. The maximum total dose is not expected to exceed about 2 g/day for a 40 to 80 kg human patient.
[00238] For the prevention and/or treatment of long-term conditions, such as inflammatory and autoimmune conditions, the regimen for treatment usually extends over many months or years, and accordingly oral dosing is preferred for patient convenience and tolerance. With oral dosing, one to five and especially two to four and typically three oral doses per day are representative regimens. Using these dosing patterns, each dose provides from about 0.01 to about 20 mg/kg of the compound of the invention, with preferred doses each providing from about 0.1 to about 10 mg/kg and especially about 1 to about 5 mg/kg.
[00239] Transdermal doses are generally selected to provide similar or lower blood levels than are achieved using injection doses.
[00240] When used to prevent the onset of an inflammatory condition, the compounds of this invention will be administered to a patient at risk for developing the condition, typically on the advice and under the supervision of a physician, at the dosage levels described above. Patients at risk for developing a particular condition generally include those that have a family history of the condition, or those who have been identified by genetic testing or screening to be particularly susceptible to developing the condition.
[00241] The compounds of this invention can be administered as the sole active agent or they can be administered in combination with other agents, including other compounds that demonstrate the same or a similar therapeutic activity, and that are determined to safe and efficacious for such combined administration.
GENERAL SYNTHETIC PROCEDURES [00242] The triazolo[l,5-a]pyridyl compounds of this invention can be prepared from readily available starting materials using the following general methods and procedures. It will be appreciated that where typical or preferred process conditions (i.e., reaction temperatures, times, mole ratios of reactants, solvents, pressures, etc.) are given; other process conditions can also be used unless otherwise stated. Optimum reaction conditions may vary with the particular reactants or solvent used, but such conditions can be determined by one skilled in the art by routine optimization procedures. [00243] Additionally, as will be apparent to those skilled in the art, conventional protecting groups may be necessary to prevent certain functional groups from undergoing undesired reactions. The choice of a suitable protecting group for a particular functional group as well as suitable conditions for protection and deprotection are well known in the art. For example, numerous protecting groups, and their introduction and removal, are described in T. W. Greene and P. G. M. Wuts, Protecting Groups in Organic Synthesis, Second Edition, Wiley, New York, 1991, and references cited therein.
[00244] The following methods are presented with details as to the preparation of representative bicycloheteroaryls that have been listed hereinabove. The compounds of the invention may be prepared from known or commercially available starting materials and reagents by one skilled in the art of organic synthesis. General synthetic route
Figure imgf000044_0001
General
[00245] All reagents were of commercial grade and were used as received without further purification, unless otherwise stated. Commercially available anhydrous solvents were used for reactions conducted under inert atmosphere. Reagent grade solvents were used in all other cases, unless otherwise specified. Column chromatography was performed on silica gel 60 (35-70 μm). Thin layer chromatography was carried out using pre-coated silica gel F-254 plates (thickness 0.25 mm). 1H NMR spectra were recorded on a Bruker DPX 400 NMR spectrometer (400 MHz). Chemical shifts
(δ) for 1H NMR spectra are reported in parts per million (ppm) relative to tetramethylsilane (δ 0.00) or the appropriate residual solvent peak, i.e. CHCI3 (δ 7.27), as internal reference. Multiplicities are given as singlet (s), doublet (d), triplet (t), quartet (q), multiplet (m) and broad (br). Coupling constants (J) are given in Hz. Electrospray MS spectra were obtained on a Micromass platform LC/MS spectrometer. Column Used for all LCMS analysis: Waters Acquity UPLC BEH Cl 8 1.7μm, 2.1mm ID x 50mm L (Part No.186002350)). Preparative HPLC: Waters XBridge Prep C18 5μm ODB 19mm ID x 100mm L (Part No.186002978). All the methods are using MeCN/H2O gradients. H2O contains either 0.1% TFA or 0.1% NH3.
Representative Synthesis of Compounds of Invention
Compound 1: (4-Morpholin-4-ylphenyl)-[5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5-a]pyrazin-8- yl] amine
Step 1: N'-(3, 6-Dibromo-pyrazin-2-yl)-N,N-dimethylformamidine
Figure imgf000045_0001
[00246] A mixture of 3,6-dibromo-pyrazin-2-ylamine (15.37 g, 60.80 mmol) and N,N- dimethylformamide dimethyl acetal (10. ImL, 76.00 mmol), suspended in ethanol (15O mL), is refluxed for 2 hours. The reaction mixture is evaporated in vacuo affording the title compound (18.6 g). 1H-NMR (400MHz, CDCl3) δ(ppm) 3.20 (s, 3H), 3.21 (s, 3H), 7.93 (s, IH), 8.48 (s, IH). LCMS: Rt 3.81 min (99.1%), m/z (APCI) 307 (M+H)+.
Step 2: N-(3,6-Dibromo-pyrazin-2-yl)-N'-hydroxyformamidine
Figure imgf000045_0002
[00247] To a solution of jV-(3,6-dibromo-pyrazin-2-yl)-N,N-dimethylformamidine (18.6 g,
60.80 mmol) in methanol (20O mL) is added hydroxylamine hydrochloride (5.91 g, 85.12 mmol) in one portion. The reaction is stirred at room temperature for 16 hours. The solvent is evaporated and the solid residue is treated with cold (ice cooling) water and collected by filtration. The precipitate is washed twice with water and petroleum ether and dried in vacuo yielding the title compound (17.45 g) as a white solid. 1H-NMR (400MHz, Cl6-DMSO) δ(ppm) 7.82 (IH, br s), 8.21 (IH, s), 8.34 (IH, m), 11.17 (IH, br s). LCMS: Rt 3.17 min (98.7 %), m/z (APCI) 295 (M+H)+.
Step 3: 5,8-Dibromo-[l ,2,4Jtriazolo[l ,5-aJpyrazine
Figure imgf000046_0001
[00248] N-(3,6-dibromo-pyrazin-2-yl)-jV-hydroxyformamidine (17.4 mg, 58.80 mmol) is treated with polyphosphoric acid (150 g) for one hour at 5O0C and then for 1.75 hours at 7O0C. After cooling to room temperature, water is added to the reaction mixture. The resultant suspension is brought to pH 8 by careful addition of solid NaHCθ3 in small portions. The precipitate formed is collected by filtration, washed once with IN NaOH, three times with water and dried in vacuo. The residue is partitioned between ethyl acetate and IN NaOH and the organic phase is washed one more time with IN NaOH and once with brine. The organic phase is dried over MgSOzi, filtered and evaporated to give the title compound (10.15 g) as a white solid. 1H-NMR (400MHz, Cl6-DMSO) δ(ppm) 8.43 (s, IH), 8.92 (s, IH). LCMS: Rt 2.73 min (94.2 %), m/z (APCI) 277 (M+H)+.
Step 4: (5-Bromo-[l,2,4Jtriazolo[l,5-aJpyrazin-8-yl)-(4-morpholin-4-ylphenyl)amine
Figure imgf000046_0002
[00249] A mixture of 5,8-dibromo-[l,2,4]triazolo[l,5-a]pyrazine (123 mg, 443 μmol), 4-(4- morpholino)aniline (118 mg. 0.664mmol) and N-ethyldiisopropylamine (116 mL, 0.664mmol) is heated at reflux in 2-propanol (3 mL) for 4.5 hours. The reaction mixture is evaporated to dryness and the residue partitioned between dichloromethane and citric acid (10 %). The organic phase is washed once with water and brine, dried over MgSOzi, filtered and evaporated to furnish the title compound (156 mg, 94 %) as a yellow solid. 1H-NMR (400MHz, Cl6-DMSO) δ(ppm) 3.11 (m, 4H), 3.78 (m, 4H), 6.97 (d, 2H), 7.82 (d, 2H), 7.87 (s, IH), 8.71 (s, IH), 9.93 (br s, IH). LCMS, Rt 3.32 min (96.8 %) m/z (APCI) 375 (M+H)+.
Step 5: (4-Morpholin-4-ylphenyl)-[5-(lH-pyrazol-4-yl)-[l,2,4Jtriazolo[l,5-aJpyrazin-8-ylJamine
Figure imgf000047_0001
[00250] A suspension of (5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-(4-morpholin-4- ylphenyl)amine (220 mg, 0.586mmol), 4-(4,4,5,5-tetratmethyl-l,3,2-dioxaborolan-2-yl)-lH-pyrazole (171 mg, 0.879mmol), Pd(PPh3)4 (68mg, 59μmol) and NaO'Bu (225mg, 2.34mmol) in 4mL of DMF/water (3:1) is degassed for 5 min in a sealed tube. The reaction mixture is heated in the sealed tube at 9O0C overnight. After evaporation of the solvents, the residue is collected by filtration, washed with water (3x) and ether (2x) and dried in vacuo. The crude product is purified by column chromatography (silica gel, 96:4 DCM/MeOH) yielding the title compound (76 mg, 36 %) as a yellow solid. 1H-NMR (400MHz, ds-DMSO) δ(ppm) 3.11 (4H, m), 3.79 (4H, m), 6.98 (2H, d), 7.90 (2H, d), 8.17 (IH, s), 8.35 (IH, br s), 8.64 (IH, br s), 8.75 (IH, s), 9.73 (IH, br s). LCMS: Rt 2.68 min (97.7 %) m/z (APCI) 363 (M+H)+.
Compound 4: 4-(8-(4-(4-methylpiperazin-l-yl)phenylamino)-[l,2,4]triazolo[l,5a]pyrazin-5- yl)pyridin-2(lH)-one
Step 1: [5-(2-Methoxy-pyridin-4-yl)-[l,2,4Jtriazolo[l,5-aJpyrazin-8-ylJ-[4-(4-methyl-piperazin-l-yl)- phenyl] -amine
Figure imgf000047_0002
[00251] This compound may be prepared using methods as described for Compound 6, step 4 using (5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-[4-(4-methyl-piperazin-l-yl)-phenyl]-amine
(80mg, 0.206mmol), 2-methoxypyridine-4-boronic acid (63mg, 0.412mmol), Pd(PPh3)4 (0.052mmol) and 1.5N Na2CO3 (1.ImL, 1.65mmol) in 2:1 DMF/dioxane (2.2mL). The crude product is purified by silica gel column chromatography using 96:4 DCM:NH3 (7M in MeOH) and the fractions containing the desired product are combined and evaporated to afford the title compound (40mg, 47%). HPLC (254nm): Rt 2.26min (65%). Step 2: 4-(8-(4-(4-Methylpiperazin-l-yl)phenylamino)-[l,2, 4]triazolo[l, 5a]pyrazin-5-yl)pyridin- 2(lH)-one
Figure imgf000048_0001
[00252] A solution of [5-(2-methoxy-pyridin-4-yl)-[l,2,4]triazolo[l,5-a]pyrazm-8-yl]-[4-(4- methyl-piperazin-l-yl)-phenyl]-amine (40mg, 0.096mmol) and pyridine hydrochloride (55mg, 0.48mmol) in water (ImL) is stirred at room temperature for 1 hour. The solvent is removed in vacuo and the residue is purified by silica gel column chromatography eluting with 96:4 DCM:NH3 (7M in MeOH). The title compound is isolated (23mg, 60%).
[00253] 1 lmg (0.0273mmol) of the free base compound are dissolved in the minimum amount of MeOH/DCM, (refluxed to dissolve) and 0.1M methanesulfonic acid (0.273mL) in MeOH is added. After evaporation of the solvent the residue is triturated several times with a mixture of 1 : 1 ethyl acetate-diethyl ether and DCM-diethyl ether, filtered and dried in vacuo to afford the target compound as a mesylate salt (13mg, 99%). 1H-NMR (400MHz, ds-DMSO) δ(ppm) 2.21 (3H, s), 2.91 (3H, s, MsOH), 3.00 (2H, t), 3.18-3.26 (2H, m), 3.58 (2H, d), 3.90 (2H, d), 6.83 (IH, s), 7.07 (2H, d), 7.25 (IH, s), 7.53 (IH, d), 7.93 (2H, d), 8.14 (IH, s), 8.76 (IH, s), 9.69 (IH, br s), 10.21 (IH, s), 13.3 (IH, br s). LCMS: Rt 1.71min (97.5%), m/z (APCI) 403 (M+H)+.
Compound 6: 4-{8-[4-(4-Methyl-piperazin-l-yl)-phenylamino]-[l,2,4]triazolo[l,5-a]pyrazin-5- yl}-thiophene-2-carboxylic acid amide
Step 1: (5-Bromo-[l,2,4Jtriazolo[l,5-aJpyrazin-8-yl)-[4-(4-methyl-piperazin-l-yl)-phenylJ-amine
Figure imgf000048_0002
[00254] A mixture of 5,8-dibromo-[l,2,4]triazolo[l,5-a]pyrazine (2 g, 7.20 mmol), 4-(4- methyl-piperazin-l-yl)-phenylamine (1.65 g, 8.64 mmol) and N-ethyldiisopropyl-amine (1.5 mL, 8.64 mmol) is heated at 8O0C in 2-propanol (50 mL) for 8 hours. The reaction mixture is evaporated to dryness and the residue partitioned between dichloromethane and water. The aqueous phase is extracted twice with dichloromethane. The organic layers are washed with brine, dried over MgSOzi, filtered and evaporated to furnish the title compound (1.41 g) as a grey solid. 1H-NMR (400MHz, c^- DMSO) δ(ppm) 2.27 (3H, s), 2.54 (4H, m), 3.14 (4H, m), 6.97 (2H, d), 7.80 (2H, d), 7.87 (IH, s), 8.72 (IH, s), 9.92 (IH, br s). LCMS: Rt 2.07 min (77.4 %), m/z (APCI) 388 (M+H)+.
Step 2: 4-Bromo-thiophene-2-carboxylic acid amide
Figure imgf000049_0001
[00255] A solution of 4 bromo-thiophene-2-carbo xylic acid (2.Og, 9.66mmol), l-ethyl-3-(3'- dimethylaminopropyl)carbodiimide hydrochloride (2.04g, 10.63mmol) and 1 -hydroxybenzotriazole hydrate (1.44g, 10.63mmol) in DMF (2OmL) is stirred at room temperature for 2 hours. The reaction mixture is then cooled to O0C and aq. NH3 (ImL, 17.3mmol) is added. The mixture is stirred at room temperature for an additional 5 hours, then water is added to the reaction mixture and the resultant precipitate is collected by filtration and washed with IM NaOH, H2O and petroleum ether. The title compound is isolated as a white solid (1.56g, 78%).
Step 3: 4-(4,4,5,5-Tetramethyl-[l,3,2]dioxaborolan-2-yl)-thiophene-2-carboxylic acid amide
Figure imgf000049_0002
[00256] 4-Bromo-thiophene-2-carboxylic acid amide (1.3g, 6.34mmol), bis(pinacolato)diboron (3.22g, 12.7mmol), PdCl2dppf (0.26g, 0.318mol) and KOAc (1.87g, 19.10mmol) are suspended in dioxane (2OmL), purged with nitrogen for 5 minutes and then heated at 9O0C overnight. The solvent is removed in vacuo and the residue partitioned between ethyl acetate and water. The aqueous layer is extracted three times with ethyl acetate and the combined organic phases are washed with brine, filtered through MgSθ4 and evaporated. The title product is crystallised from EtO Ac-petroleum ether (2.135g, 77% pure by LCMS).
Step 4: 4-{8- [4-(4-Methyl-piperazin- 1 -yl)-phenylamino] -[ 1,2,4] triazolo[l,5-a]pyrazin-5-yl}- thiophene-2-carboxylic acid amide
Figure imgf000050_0001
[00257] A suspension of 5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-[4-(4-methyl-piperazin- l-yl)-phenyl] -amine (100 mg, 258 mmol), 4-(4,4,5,5-tetramethyl-[l,3,2]dioxa-borolan-2-yl)- thiophene-2-carboxylic acid amide (130 mg, 516 mmol) and Pd(PPh3)4 (74 mg, 64.5 mmol) in aqueous Na2CO3 (1.37 mL, 1.5 M, 2.06mmol) and DMF/dioxane 2/1 (2.75 mL) is degassed for 5 min in a reaction tube. The tube is sealed and the reaction mixture is heated at 9O0C overnight. After cooling to room temperature the reaction mixture is partitioned between ethyl acetate and water. The precipitate is collected by filtration and washed with water (Ix) and ether (2x) and dried in vacuo. The crude product is purified by column chromatography (silica gel, DCM/MeOH/NH3 96:4) yielding the title compound (43 mg) as a yellow solid. 1H-NMR (400MHz, ds-DMSO) δ(ppm) 2.26 (3H, s), 2.50 (4H, m), 3.15 (4H, m,), 6.99 (2H, d), 7.60 (IH, br s), 7.88 (2H, d), 8.10 (2H, m), 8.48 (IH, s), 8.66 (IH, s), 8.79 (IH, s), 9.97 (IH, br s). LCMS: Rt 1.99 min (97.6 %), m/z (APCI) 435 (M+H)+.
Compound 9: 5-{8-[4-(4-Methyl-piperazin-l-yl)-phenylamino]-[l,2,4]triazolo[l,5-a]pyrazin-5- yl}-thiophene-2-carboxylic acid amide
Step 1: 5-Bromo-thiophene-2-carboxylic acid amide
Ox I/ \\
H2N
[00258] A solution of 5-bromo-thiophene-2-carboxylic acid (4.51g, 21.78mmol), 3- hydroxybenzotriazole hydrate (3.24g, 23.96mmol), l-ethyl-3-(3'-dimethylaminopropyl)carbodiimide (4.6g, 23.96mmol) in DMF (7OmL) is stirred at room temperature for 2 hours. The reaction mixture is then cooled to O0C and aq. 35% NH3 (2.2mL) is added. The mixture is stirred at room temperature overnight. The solvent is removed in vacuo and the residue dissolved in EtOAc, washed with IN NaHCO3, and brine. The organic layers are combined, dried over MgSO4, filtered and concentrated to afford the title compound (3.78g, 84%). HPLC (254nm): Rt 2.46min (96.5%).
Step 2: 5-(4,4,5,5-Tetramethyl-[l,3,2Jdioxaborolan-2-yl)-thiophene-2-carboxylic acid amide
Figure imgf000050_0002
[00259] 5-Bromo-thiophene-2-carboxylic acid amide (0.5g, 2.426mmol), bis(pinacolato)diboron (678mg, 2.669mmol), PdCl2dppf (59mg, 0.072mmol) and KOAc (0.714g, 7.28mmol) are suspended in dioxane (5mL), purged with nitrogen for 5 minutes and then heated at 850C overnight. The solvent is removed in vacuo and the residue partitioned between ethyl acetate and water. The aqueous layer is extracted again with ethyl acetate and the combined organic phases are washed with brine, filtered through MgSθ4 and evaporated in vacuo to afford the title compound (417mg, 68%).
Step 3: 5-{8-[4-(4-Methyl-piperazin-l-yl)-phenylamino] -[ 1,2,4] triazolo[l,5-a]pyrazin-5-yl}- thiophene-2-carboxylic acid amide
Figure imgf000051_0001
[00260] This compound may be prepared using methods as described for Compound 6, step 4 using 5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-[4-(4-methyl-piperazin-l-yl)-phenyl]-amine
(lOOmg, 0.258mmol), 5-(4,4,5,5-tetramethyl-[l,3,2]dioxa-borolan-2-yl)-thiophene-2-carboxylic acid amide (130mg, 0.516mmol) and Pd(PPh3)4 (74mg, 0.0645mmol) in aqueous 1.5M Na2CO3 (1.37mL, 2.06mmol) and dioxane (2.75mL). After evaporation of the solvent, the residue is purified by silica gel column chromatography eluting with 96:6 DCM:NH3 (7M in MeOH) to afford the title compound (23mg, 21%). LCMS: Rt 1.94min (97.9%). 1H-NMR (400MHz, ds-DMSO) δ(ppm) 2.26 (3H, s), 2.49 (4H, m), 3.14 (4H, m), 6.98 (2H, d), 7.51 (IH, br s), 7.85-7.89 (2H, m), 8.03 (2H, d), 8.10 (IH, br s), 8.38 (IH, s), 8.82 (IH, s), 10.10 (IH, s). LCMS: Rt 1.94mm, (97.9%), m/z (APCI) 435 (M+H)+.
Compound 12: 5-(5-Methyl-lH-pyrazol-4-yl)-N-(4-morpholinophenyl)-[l,2,4]triazolo[l,5- a]pyrazin-8-amine
Figure imgf000051_0002
[00261] This compound may be prepared using methods as described for Compound 6, step 4 using (5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-(4-morpholin-4-yl-phenyl)-amine (0.2g,
0.53mmol), 5-methyl-4-(4,4,5,5-tetramethyl-[l,3,2]dioxaborolan-2-yl)-lH-pyrazole (222mg, l.Oόmmol), and Pd(PPh3)4 (0.154mg, 0.134mmol) in 1.5M Na2CO3 (aq) (2.84mL, 4.26mmol) and dioxane (8.5mL). The reaction mixture is purified by silica gel column chromatography using 98:2 and 97:3 DCM:NH3 (7M in MeOH) to give the title compound (15mg, 7.5%). Conversion into the mesylate salt using O. IM methanesulfonic acid in MeOH (0.398mL) affords the title compound (15mg) as a pale green solid. 1H-NMR (400MHz, ds-DMSO) δ(ppm) 2.41 (3H, s, MsOH), 2.46 (3H, s), 3.20 (4H, m), 3.71 (4H, m), 7.11 (2H, d), 7.74 (IH, s), 7.97 (2H, d), 8.15 (IH, s), 8.69 (IH, s), 9.88 (IH, s). LCMS: Rt 2.41min (98.3%), m/z (APCI) 377 (M+H)+.
Compound 15: 4-[8-(4-Morpholin-4-yl-phenylamino)-[l,2,4]triazolo[l,5-a]pyrazin-5-yl]- thiophene-2-carboxylic acid amide
Figure imgf000052_0001
[00262] This compound may be prepared using methods as described for Compound 6, step 4 using (5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-(4-morpholin-4-yl-phenyl)-amine (0.2g,
0.53mmol), 4-(4,4,5,5-tetramethyl-[l,3,2]dioxaborolan-2-yl)-thiophene-2-carboxylic acid amide (0.27mg, 1.06mmol), and Pd(PPh3)4 (0.15mg, 0.133mmol) in 1.5M Na2CO3 (aq) (2.84mL, 4.26mmol) and dioxane (1OmL). The reaction mixture is partitioned between water and ethyl acetate. A precipitate is formed, collected by filtration and purified by silica gel column chromatography using 95:5 DCM:MeOH to give the title compound (84.1mg, 37.4%). 1H-NMR (400MHz, ds-DMSO) δ(ppm) 3.13 (4H, m), 3.79 (4H, m), 7.01 (2H, d), 7.60 (IH, br s), 7.91 (2H, d), 8.10 (2H, s), 8.48 (IH, s), 8.66 (IH, s), 8.79 (IH, s), 9.96 (IH, s). LCMS: Rt 2.61min (97.1%), m/z (APCI) 422 (M+H)+.
Compound 16: 5-(5-Methyl-lH-pyrazol-4-yl)-N-(4-(4-methylpiperazin-l-yl)phenyl)-
[l,2,4]triazolo [l,5-a]pyrazin-8-amine
Step 1 : 5-Methyl-4-(4, 4, 5, 5-tetramethyl-[l , 3, 2]dioxaborolan-2-yl)-lH-pyrazole
Figure imgf000052_0002
[00263] This compound may be prepared using methods as described for Compound 86, step 2 using 4-bromo-5-methyl-lH-pyrazole (3g, 18.6mmol), bis(pinacolato)diboron (8.52g, 33.5mmol), PdCl2dppf (913mg, l.l lδmmol) and KOAc (5.49mg, 55.9mmol) in dimethylsulfoxide (3OmL). The reaction mixture is purified by silica gel column chromatography eluting with 7:3 followed by 1 :1 petroleum ether: ethyl acetate to afford 5-methyl-4-(4,4,5,5-tetramethyl-[l,3,2]dioxaborolan-2-yl)-lH- pyrazole (3.87g, 100%).
Step 2: 5-(5-Methyl-lH-pyrazol-4-yl)-N-(4-(4-methylpiperazin-l-yl)phenyl)-[l,2,4]triazolo[l,5- aJpyrazin-8-amine
Figure imgf000053_0001
[00264] This compound may be prepared using methods as described for Compound 6, step 4 using (5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-[4-(4-methyl-piperazin-l-yl)-phenyl]-amine
(132mg, 0.340mmol), 5-methyl-4-(4,4,5,5-tetramethyl-[l,3,2]dioxaborolan-2-yl)-lH-pyrazole (141mg, 0.68mmol), Pd(PPh3)4 (98mg, 0.085mmol) and 1.5N Na2CO3 (1.8ImL, 2.72mmol) in dioxane (5.4mL). The crude product is purified by silica gel column chromatography eluting with 95:5 DCM:NH3 (7M in MeOH) followed by trituration with diethyl ether and petroleum ether to afford the title compound (9mg, 7%) as a pale green solid. 1H-NMR (400MHz, de-DMSO) δ(ppm) 2.27 (3H, s), 2.38-2.49 (7H, m), 3.14 (4H, m), 6.97 (2H, d), 7.70 (IH, s), 7.88 (2H, d), 7.99 and 8.43 (IH, br s), 8.68 (IH, s), 9.71 (IH, s), 12.92 and 12.98 (IH, br s).
[00265] Conversion into the mesylate salt using 0.1M methane sulfonic acid (0.23ImL) yields the target compound (1 lmg). LCMS: Rt 1.78min (86%), m/z (APCI) 390 (M+H)+.
Compound 17: -(3-Fluoro-4-morpholinophenyl)-5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5- a]pyrazin-8-amine
Step 1: (5-Bromo-[l,2,4Jtriazolo[l,5-aJpyrazin-8-yl)-(3-βuoro-4-morpholin-4-yl-phenyl)-amine
Figure imgf000053_0002
[00266] This compound may be prepared using methods as described for Compound 6, step 1 using 5,8-dibromo-[l,2,4]triazolo[l,5-a]pyrazine (0.5g, 1.799mmol), 3-fluoro-4-morpholin-4-yl- phenylamine (0.53g, 2.70mmol), and DIPEA (0.47OmL, 2.70mmol) in 2-propanol (6mL). The reaction mixture is partitioned between 10% citric acid aqueous solution and DCM. The organic phase is separated and washed with water and brine, dried over MgSO4, filtered and concentrated under vacuum to afford the title compound (697mg, 98%) which is used in the next step without further purification. LCMS: Rt 3.52min (98.2%).
Step 2: N-(3-Fluoro-4-morpholinophenyl)-5-(lH-pyrazol-4-yl)-[ 1 ,2,4] triazolo [ 1 ,5-a] pyrazin-8-amine
Figure imgf000054_0001
[00267] This compound may be prepared using methods as described for Compound 6, step 4, using (5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-(3-fluoro-4-morpholin-4-yl-phenyl)-amine (lOOmg, 0.254mmol), 4-(4,4,5,5-tetramethyl-[l,3,2]dioxaborolan-2-yl)-lH-pyrazole (99mg, 0.51mmol) and Pd(PPh3)4 (73mg, 0.063mmol) in 1.5M Na2CO3 (1.36mL, 2.03mmol) and dioxane (4mL). The crude material is purified by silica gel column chromatography eluting with 98:2 DCM:NH3 (7M in MeOH) and by trituration with diethyl ether and petroleum ether to yield the target compound (43mg, 44%). 1H-NMR (400MHz, ds-DMSO) δ(ppm) 3.01 (4H, m), 3.79 (4H, m), 7.08 (IH, t), 7.79 (IH, d), 8.06 (IH, d), 8.26 (IH, s), 8.38 (IH, s), 8.67 (IH, s), 8.79 (IH, s), 10.03 (IH, s), 13.3 (IH, br s). LCMS: Rt 2.83min (99%), m/z (APCI) 381 (M+H)+.
Compound 19: 5-(5-(lH-Pyrazol-4-yl)-[l,2,4]triazolo[l,5-a]pyrazin-8-ylamino)-2- morpholinobenzamide
Step 1 : 5-(5-Bromo-[l, 2, 4] triazolo [1, 5-a]pyrazin-8-ylamino)-2-morpholin-4-yl-benzamide
Figure imgf000054_0002
[00268] This compound may be prepared using methods as described for Compound 6, step 1 using 5,8-dibromo-[l,2,4]triazolo[l,5-a]pyrazine (250mg, 0.90mmol) 5-amino-2-morpholin-4-yl benzamide (299mg, 1.35mmol) and N,N-diisopropylethylamine (0.24mL, 1.35mmol) in 2-propanol (7mL). Trituration with 'PrOH and Et2O affords the title compound (273mg, 73%).
Step 2: 5-(5-(lH-Pyrazol-4-yl)-[l,2,4]triazolo[l,5-a]pyrazin-8-ylamino)-2-morpholinobenzamide
Figure imgf000055_0001
[00269] This compound may be prepared using methods as described for Compound 6, step 4 using 5-(5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-ylamino)-2-morpholin-4-yl-benzamide (140mg, 0.33mmol), 4-(4,4,5,5-tetramethyl-[l,3,2]dioxaborolan-2-yl)-lH-pyrazole (130mg, 0.67mmol) and Pd(PPh3)4 (96mg, 0.083mmol) in 1.5M K2CO3 (aq) (1.93mL) and dioxane (3.44mL). The crude material is purified by silica gel column chromatography eluting with DCM followed by 99:1 then 97:3 then 95:5 DCM:NH3 (7M in MeOH) to afford the title compound as a white solid (40.5mg, 30%). 1H-NMR (400MHz, d6-DMSO) δ(ppm) 2.96 (4H, m), 3.79 (4H, m), 7.28 (2H, d), 7.45 (IH, m), 7.54 (IH, br s), 8.01-8.09 (IH, dd), 8.25 (IH, s), 8.50 (IH, d), 8.69 (IH, br s), 8.78 (IH, s), 9.98 (IH, s), 13.3 (IH, br s). LCMS: Rt 2.23min (96.9%), m/z (APCI) 406 (M+H)+.
Compound 21: 4-(8-(4-(2-Morpholinoethoxy)phenylamino)-[l,2,4]triazolo[l,5-a]pyrazin-5- yl)thiophene-2-carboxamide
Figure imgf000055_0002
[00270] This compound may be prepared using methods as described for Compound 6, step 4, using (5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-[4-(2-morpholin-4-yl-ethoxy)-phenyl]-amine
(113mg, 0.27mmol), 4-(4,4,5,5-tetramethyl-[l,3,2]dioxaborolan-2-yl)-thiophene-2-carboxamide (136mg, 0.537mmol), and Pd(PPh3)4 (78mg, 0.067mmol) in 1.5M Na2CO3 (1.44mL, 2.16mmol) and dioxane (4.3mL). The reaction mixture is purified by silica gel column chromatography eluting with 97:3 DCM:NH3 (7M in MeOH) to yield the title compound (65mg, 52%). LCMS: Rt 1.98min (98.9%). Conversion into the mesylate salt using 0.1M methanesulfonic acid in MeOH (1.1385mL) affords the title compound (50mg, 95%). 1H-NMR (400MHz, ds-DMSO) δ(ppm) 2.37 (3H, s, MsOH), 3.21-3.64 (6H, m), 3.76 (2H, t), 4.05 (2H, d), 4.40 (2H, m), 7.09 (2H, d), 7.61 (IH, br s), 8.02 (2H, d), 8.11 (2H, s), 8.48 (IH, s), 8.67 (IH, s), 8.82 (IH, s), 9.91 (IH, br s), 10.09 (IH, s). LCMS: Rt 1.98min (97.7%), m/z (APCI) 466 (M+H)+. Compound 22: 5-(8-(4-(2-Morpholinoethoxy)phenylamino)-[l,2,4]triazolo[l,5-a]pyrazin-5- yl)thiophene-2-carboxamide
Figure imgf000056_0001
[00271] This compound may be prepared using methods as described for Compound 6, step 4, using (5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-[4-(2-morpholin-4-yl-ethoxy)-phenyl]-amine
(118mg, 0.28mmol), 5-(4,4,5,5-tetramethyl-[l,3,2]dioxaborolan-2-yl)-thiophene-2-carboxamide (142mg, 0.56mmol), and Pd(PPh3)4 (81mg, 0.07mmol) in 1.5M Na2CO3 (1.5mL, 2.24mmol) and dioxane (4.5mL). The reaction mixture is purified by silica gel column chromatography eluting with 97:3 DCM:NH3 (7M in MeOH) to yield the title compound (33mg, 25%). LCMS: Rt 2.66min (99%). Conversion into the mesylate salt using O. IM methanesulfonic acid in MeOH (0.569mL) affords the title compound (24mg, 86%). 1H-NMR (400MHz, ds-DMSO) δ(ppm) 2.37 (3H, s, MsOH), 3.25 (2H, m), 3.50-3.64 (4H, m), 3.76 (2H, t), 4.05 (2H, d), 4.40 (2H, m), 7.09 (2H, d), 7.51 (IH, br s), 7.87 (IH, d), 7.99-8.05 (3H, m), 8.11 (IH, br s), 8.37 (IH, s), 8.84 (IH, s), 9.92 (IH, br s), 10.21 (IH, s). LCMS: Rt 1.95min (98.8%), m/z (APCI) 466 (M+H)+.
Compound 23: (4-Morpholin-4-yl-phenyl)-[5-(2H-pyrazol-3-yl)-[l,2,4]triazolo[l,5-a]pyrazin-8- yl]-amine
Step 1: l-(Tetrahydro-pyran-2-yl)-lH-pyrazole
Figure imgf000056_0002
[00272] 7H-Pyrazole (14.3g, 0.21mol) is dissolved in 3,4-dihydro-2Η-pyran (26.74g, 0.32mol) in the presence of a catalytic amount of TFA (O.lmL, 1.3mmol). The reaction mixture is stirred at 950C for 5 hours, cooled and then quenched using NaH (0.2g, 5mmol). The solvent is removed to give the title compound as a brown oil (33.3g, 99%), which is used in the next step without further purification. Step 2: lH-Pyrazole-2-boronic acid
Figure imgf000057_0001
[00273] To a cooled solution (-780C) of l-(tetrahydro-pyran-2-yl)-lH-pyrazole (7.6g,
52mmol) in THF (5OmL), «BuLi (33mL, 2.5M in hexane, 82.5mmol) and triisopropyl borane (12.7mL, 55mmol) are added dropwise maintaining the temperature at -7O0C. The reaction mixture is stirred at -7O0C for one hour and then allowed to reach room temperature over 4 hours. After quenching the reaction with 2M HCl, the solvent is removed in vacuo and the pH is adjusted to pH 6 using IM NaOH. A precipitate is formed, collected by filtration and washed with toluene and petroleum ether. Trituration with ethyl acetate affords the target compound as a white solid (2.7g, 48%), which is used in the next step without further purification.
Step 3: (4-Morpholin-4-yl-phenyl)-[5-(2H-pyrazol-3-yl)-[l,2,4]triazolo[l,5-a]pyrazin-8-yl]-amine
Figure imgf000057_0002
This compound may be prepared using methods as described for Compound 6, step 4, using 5-bromo- [l,2,4]triazolo[l,5-a]pyrazin-8-yl)-[4-(morpholin-4-yl)-phenyl]-amine (lOOmg, 0.267mmol), IH- pyrazole-2-boronic acid (60mg, 0.535mmol), Pd(PPh3)4 (93mg, 0.08mmol) and Na2CO3 (88mg, 0.80mmol) in DMF (2mL). The reaction mixture is placed in a stem-tube and stirred at 1000C for 18 hours. After cooling, the mixture is diluted with NaHCO3 solution and extracted with EtOAc (4x). The organic layer is washed with water, dried over MgSO4, filtered and evaporated to afford a crude product that is purified by silica gel column chromatography eluting with DCM followed by 96:4 DCM:NH3 (7M in MeOH). The title compound is isolated after trituration with diethyl ether (12.4mg, 13%). Conversion into the mesylate salt using 0.1M methanesulfonic acid (0.342mL, 0.0342mmol) gives the target compound (lOmg, 81%). 1H-NMR (400MHz, de-DMSO) δ(ppm) 2.34 (3H, s, MsOH), 3.14 (4H, m), 3.81 (4H, m), 7.04 (2H, d), 7.27 (IH, m), 7.92 (3H, m), 8.24 (IH, s), 8.76 (IH, s), 9.92 (IH, s). LCMS: Rt 2.45min (97.6%), m/z (APCI) 363 (M+H)+.
Compound 28: 4-(8-(4-(4-Isopropylpiperazin-l-yl)phenylamino)-[l,2,4]triazolo[l,5-a]pyrazin-5- yl)thiophene-2-carboxamide
Figure imgf000058_0001
[00274] This compound may be prepared using methods as described for Compound 6, step 4, using 5-bromo-N-(6-(4-isopropylpiperazin-l-yl)pyridin-3-yl)-[l,2,4]triazolo[l,5-a]pyrazin-8-amine (lOOmg, 0.24mmol), 4-(4,4,5,5-tetramethyl-[l,3,2]dioxaborolan-2-yl)-thiophene-2-carboxamide (121mg, 0.48mmol), and Pd(PPh3)4 (69mg, 0.059mmol) in 1.5M Na2CO3 (1.28mL, 1.92mmol) and dioxane (3.84mL). The crude product is purified by silica gel column chromatography eluting with DCM and 97:3 DCM:NH3 (7M in MeOH) to yield the title compound (68mg, 61%) as a pale green solid. Conversion into the mesylate salt using O. IM methanesulfonic acid in MeOH (1.47mL, 0.147mmol) followed by trituration with DCM and diethyl ether, affords the title compound (67mg, 98.5%). 1H-NMR (400MHz, ds-DMSO) δ(ppm) 1.35 (6H, d), 2.34 (3H, s, MsOH), 3.00 (2H, t), 3.22- 3.25 (2H, m), 3.56-3.61 (3H, m), 3.89 (2H, d), 7.09 (2H, d), 7.60 (IH, br s), 7.97 (2H, d), 8.10 (2H, s), 8.48 (IH, s), 8.67 (IH, s), 8.81 (IH, s), 9.23 (lH, br s), 10.03 (IH, s). LCMS: Rt 2.13min (98.4%), m/z (APCI) 463 (M+H)+.
Compound 29: 5-(8-(4-Morpholinophenylamino)-[l,2,4]triazolo[l,5-a]pyrazin-5-yl)furan-3- carboxamide
Step 1: [5-(4-Carbamoyl-furan-2-yl)-[l,2,4]triazolo[l,5-a]pyrazin-8-yl]-(4-morpholin-4-yl-phenyl)- carbamic acid tert-butyl ester
Figure imgf000058_0002
[00275] This compound may be prepared using methods as described for Compound 35, step 3 using (4-morpholin-4-yl-phenyl)-(5-tributylstannanyl-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-carbamic acid tert-butyl ester (137mg, 0.199mmol), 5-bromo-furan-3-carboxamide (76mg, 0.4mmol) and Pd(PPh3)4 (23mg, 0.020mmol) in DMF (ImL). Purification of the reaction mixture eluting with 98:2 DCM:MeOH and 96:4 DCM:NH3 (7M in MeOH) affords the title compound (33mg, 33%). Step 2: 5-(8-(4-Morpholinophenylamino)-[l,2,4]triazolo[l,5-a]pyrazin-5-yl)furan-3-carboxamide
Figure imgf000059_0001
[00276] A solution of [5-(4-carbamoyl-furan-2-yl)-[l,2,4]triazolo[l,5-a]pyrazin-8-yl]-(4- morpholin-4-yl-phenyl)-carbamic acid tert-butyl ester (33mg, 0.065mmol) in a mixture 1:1 DCM:TFA (2mL) (2 drops of water) is stirred at room temperature for 2 hours. After addition of sat. Na2CO3, a precipitate is formed, collected by filtration and washed with water, diethyl ether and petroleum ether. After drying under vacuum, the title compound is isolated (20mg, 76%). Conversion into the mesylate salt using 0.1 M methanesulfonic acid in MeOH (0.444mL) yields the title compound (19mg, 100%). 1H-NMR (400MHz, d6-DMSO) δ(ppm) 2.36 (3H, s, MsOH), 3.22 (4H, m), 3.82-3.89 (4H, m), 7.13 (2H, d), 7.36 (IH, br s), 7.86 (IH, s), 7.95 (3H, m), 8.17 (IH, s), 8.40 (IH, s), 8.84 (IH, s), 10.17 (IH, s). LCMS: Rt 2.60min (96.9%), m/z (APCI) 406 (M+H)+.
Compound 33: 5-(8-(4-Morpholinophenylamino)-[l,2,4]triazolo[l,5-a]pyrazin-5-yl)isoindolin-l- one
Figure imgf000059_0002
[00277] This compound may be prepared using methods as described for Compound 6, step 4 using (5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-(4-morpholin-4-yl-phenyl)-amine (1-Og,
2.67mmol), 5-(4,4,5,5-tetramethyl-[l,3,2]dioxaborolan-2-yl)-2,3-dihydro-isoindol-l-one (see description for Compound 79) (1.03g, 4.01mmol) and Pd(PPh3)4 (0.77g, 0.67mmol) in 1.5M Na2CO3 (14.3mL) and dioxane (4OmL). Purification by silica gel column chromatography eluting with DCM followed by 96:4 DCM:NH3 (7M in MeOH) affords the title compound (0.895g, 79%). 1H-NMR (400MHz, ds-DMSO) δ(ppm) 3.12 (4H, m), 3.79 (4H, m), 4.51 (2H, s), 7.01 (2H, d), 7.84 (IH, d), 7.92 (2H, d), 8.01 (IH, s), 8.09 (IH, d), 8.23 (IH, s), 8.72 (IH, s), 8.73 (IH, s), 10.02 (IH, s). LCMS: Rt 2.51min (97.8%), m/z (APCI) 428 (M+H)+. Compound 34: 4-(8-(4-Morpholinophenylamino)-[l,2,4]triazolo[l,5-a]pyrazin-5-yl)furan-2- carboxamide
Figure imgf000060_0001
[00278] This compound may be prepared using methods as described for Compound 6, step 4, using (5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-(4-morpholin-4-yl-phenyl)-amine (78mg, O.lόmmol), 4-(4,4,5,5-tetramethyl-[l,3,2]dioxaborolan-2-yl)-furan-2-carboxylic acid amide (lOOmg, 0.34mmol), and Pd(PPh3)4 (60mg, 0.052mmol) in 1.5M Na2CO3 (aq) (1.ImL, 1.68mmol), and dioxane (3mL). The reaction mixture is partitioned between water and ethyl acetate, the title compound precipitates and is collected by filtration. Purification of the solid by silica gel column chromatography, eluting with DCM and 95:5 DCM:NH3 (7M in MeOH), affords the title compound (42mg, 65%). Conversion into the mesylate salt using O. IM methanesulfonic acid (0.82mL) gives a solid which is triturated with diethyl ether to afford the title compound (30.5mg). 1H-NMR (400MHz, ds-DMSO) δ(ppm) 2.19 (3H, s, MsOH), 3.03-3.07 (4H, m), 3.67 (4H, m), 6.99 (2H, br s), 7.43 (IH, d), 7.74-7.82 (4H, m), 8.06 (IH, br s), 8.59 (IH, br s), 8.64 (IH, s), 9.91 (IH, br s). LCMS: Rt 2.64min (98.1%), m/z (APCI) 406 (M+H)+.
Compound 35: 6-(8-(4-Morpholinophenylamino)-[l,2,4]triazolo[l,5-a]pyrazin-5-yl)-3,4- dihydroisoquinolin-1 (2H)-one
Step 1: (5-Bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-(4-morpholin-4-yl-phenyl)-carbamic acid tert- butyl ester
Figure imgf000060_0002
[00279] A solution of 5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-[4-(morpholin-4-yl)- phenyl]-amine (300mg, 0.800mmol), dimethylaminopyridine (lOmg, 0.08mmol) and άi-tert butyl dicarbonate (523mg, 2.4mmol) in dichloromethane (5ml) is stirred at 5O0C overnight. The reaction mixture is partitioned between DCM and water and the organic layer is washed with IN NaOH and brine. The organic layer is dried over MgSO4, filtered and concentrated in vacuo to afford a crude compound purified by silica gel column chromatography. Elution with 98:2 DCM:MeOH yields the title compound (352mg, 93%). LCMS: Rt 3.45min (97.8%). Step 2: (4-Morpholin-4-yl-phenyl)-(5-tributylstannanyl-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-carbamic acid tert-butyl ester
Figure imgf000061_0001
[00280] To a cooled (-780C) solution of (5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-(4- morpholin-4-yl-phenyl)-carbamic acid tert-butyl ester (370mg, 0.78mmol) in THF (12mL), are added 2M isopropylmagnesium chloride in THF (0.78mL, 1.56mmol), and after 5 minutes stirring, tributyltin chloride (0.42, 1.56mmol) is added. The reaction mixture is stirred at -780C for 15 minutes and at room temperature for an additional 15 minutes. After removing the solvent, the residue is purified by silica gel column chromatography eluting with 5:1 petroleum etheπethyl acetate followed by 1 :1 petroleum etheπethyl acetate. The title compound is isolated (105mg, 20%).
Step 3: (4-Morpholin-4-yl-phenyl)-[5-(l-oxo-l, 2, 3, 4-tetrahydro-isoquinolin-6-yl)-[ 1 , 2, 4]triazolo[l, 5- aJpyrazin-8-ylJ -carbamic acid tert-butyl ester
Figure imgf000061_0002
[00281] To a degassed solution of (4-morpholin-4-yl-phenyl)-(5-tributylstannanyl-
[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-carbamic acid tert-butyl ester (lOOmg, 0.15mmol) and 6-bromo- 3,4-dihydro-2H-isoquinolin-l-one in DMF (ImL) is added tetrakis(triphenylphosphine)palladium(0) (17mg, 0.015mmol) and the reaction mixture is stirred at 9O0C overnight. After removing the solvent in vacuo, the residue is purified by silica gel column chromatography. Elution with 1 : 1 petroleum etheπethyl acetate and ethyl acetate affords the target compound as a yellow solid (30mg,
Step 4: 6-[8-(4-Morpholin-4-yl-phenylamino)-[ 1 ,2,4] triazolo [ 1 ,5-a] pyrazin-5-yl] -3 ,4-dihydro-2H- isoquinolin-1-one
Figure imgf000062_0001
[00282] A solution of (4-morpholin-4-yl-phenyl)-[5-(l-oxo-l,2,3,4-tetrahydro-isoquinolin-6- yl)-[l,2,4]triazolo[l,5-a]pyrazin-8-yl]-carbamic acid tert-butyl ester (30mg, 0.055mmol) in a 1:1 mixture of TFA:DCM (1 drop of H2O), is stirred at room temperature for 2 hours. The solvent is removed in vacuo and the residue partitioned between ethyl acetate and sat. NaHCθ3 (aq). The water layer is extracted with ethyl acetate (2x). The organic layers are combined and evaporated to afford a residue purified by silica gel column chromatography eluting with ethyl acetate. The title compound is isolated (18mg, 75%) and converted into the mesylate salt (16.6mg, 97%) using 0.1M methanesulfonic acid (0.317mL) in MeOH.
1H-NMR (400MHz, ds-DMSO) δ(ppm) 2.36 (3H, s MsOH), 3.04 (2H, t), 3.21 (4H, m), 3.47 (2H, t), 3.83 (4H, m), 7.11 (2H, d), 7.95-8.02 (6H, m), 8.05 (IH, s), 8.74 (IH, s), 10.09 (IH, br s). LCMS: Rt 2.81min (97.9%), m/z (APCI) 442 (M+H)+.
Compound 36: 5-(8-(4-(4-Isopropylpiperazin-l-yl)phenylamino)-[l,2,4]triazolo[l,5-a]pyrazin-5- yl)isoindolin-l-one
Figure imgf000062_0002
[00283] This compound may be prepared using methods as described for Compound 6, step 4 using 5-bromo-N-(6-(4-isopropylpiperazin-l-yl)pyridin-3-yl)-[l,2,4]triazolo[l,5-a]pyrazin-8-amine (0.6g, 1.44mmol), 5-(4,4,5,5-tetramethyl-[l,3,2]dioxaborolan-2-yl)-2,3-dihydro-isoindol-l-one (0.56g, 2.16mmol) and Pd(PPh3)4 (0.416g, 0.36mmol) in 1.5M Na2CO3 (7.7mL) and dioxane (23mL). Purification by silica gel column chromatography eluting with DCM followed by 98:2 DCM:NH3 (7M in MeOH) affords the title compound (0.36g, 53%) which is converted into the mesylate salt using IM methanesulfonic acid in MeOH (0.77mL). After trituration with diethyl ether and DCM the title compound is isolated as a solid (0.41Og). 1H-NMR (400MHz, ds-DMSO) δ(ppm) 1.35 (6H, d), 2.35 (3H, s, MsOH), 3.04 (2H, t), 3.20-3.27 (2H, m), 3.56-3.63 (3H, m), 3.89 (2H, d), 4.52 (2H, s), 7.09 (2H, d), 7.85 (IH, d), 7.95-8.01 (3H, m), 8.10 (IH, d), 8.24 (IH, s), 8.72 (IH, s), 8.74 (IH, s), 9.27 (IH, br s), 10.07 (IH, s). LCMS: Rt 2.93min (97.9%), m/z (ES+) 469 (M+H)+. Compound 37: {5-[5-(lH-Pyrazol-4-yl)-[l,2,4]triazolo[l,5-a]pyrazin-8-ylamino]-2- morpholinophenyl}methanol
Step 1: (2-Morpholin-4-yl-5-nitro-phenyl)-methanol r .OH r /-\°
O2N X ^^T
[00284] To a cooled (O0C) solution of 2-morpholin-4-yl-5-nitro-benzaldehyde (0.8g,
3.39mmol) in MeOH (5mL) is added NaBH4 (0.125g, 3.39mmol) and the reaction mixture is stirred at room temperature for 3 hours. After quenching the reaction with water, the solvent is removed in vacuo and the residue dissolved in ethyl acetate and washed with brine. The organic layer is dried over MgSO4, filtered and concentrated to afford the title compound (870mg), which is used in the next step without further purification.
Step 2: (5-Amino-2-morpholin-4-yl-phenyl)-methanol
Figure imgf000063_0001
[00285] To a solution of (2-morpholin-4-yl-5-nitro-phenyl)-methanol (870mg) in ethanol
(4OmL) palladium hydroxide (87mg) is added and the mixture is stirred in a Parr-apparatus under hydrogen pressure (10 bars) for 4 hours. The reaction mixture is filtrated over Celite 521, washed with ethanol and concentrated in vacuo to give the title compound (640mg, 83%).
Step 3: 3-(tert-Butyl-dimethyl-silanyloxymethyl)-4-morpholin-4-yl-phenylamine
\ \ / /
\ r ~ SI —
X XO
H2N ^-T^
[00286] A solution of (5-amino-2-morpholin-4-yl-phenyl)-methanol (640mg, 3.07mmol), tert- butyldimethylsilyl chloride (509mg, 3.38mmol) and imidazole (250mg, 3.68mmol) in dimethylformamide (2OmL) is stirred at room temperature overnight. The solvent is removed in vacuo and the residue partitioned between water and ethyl acetate. The organic layer is washed with brine, dried over MgSO4, filtered and concentrated to afford a crude product. Purification, using silica gel column chromatography, eluting with DCM followed by a 95:5 mixture DCM:MeOH, affords the title compound as a pink solid (390mg, 27%).
Step 4: (5-Bromo-[ 1,2,4] triazolo[l,5-a] pyrazin-8-yl)-[3-(tert-butyl-dimethyl-silanyloxymethyl)-4- morpholin-4-yl-phenyl] -amine
Figure imgf000064_0001
[00287] A mixture of 5,8-dibromo-[l,2,4]triazolo[l,5-a]pyrazine (0.391g, 1.41mmol), 3-(tert- butyl-dimethyl-silanyloxymethyl)-4-morpholin-4-yl-phenylamine (0.5g, 1.55mmol) and N- ethyldiisopropyl-amine (0.27mL, 1.55mmol) is heated at 9O0C in 2-propanol (1OmL) for 8 hours. The reaction mixture is evaporated to dryness and the residue partitioned between dichloromethane and water. The aqueous phase is extracted twice with dichloromethane. The organic layers are combined, washed with brine, dried over MgSOzi, filtrated and concentrated in vacuo to afford the title compound (335mg, 46%).
Step 5: [5-(5-Bromo-[ 1,2, 4] triazolo[l,5-a]pyrazin-8-ylamino)-2-morpholin-4-yl-phenyl] -methanol
Figure imgf000064_0002
[00288] A solution of 5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-[3-(tert-butyl-dimethyl- silanyloxymethyl)-4-morpholin-4-yl-phenyl]-amine (285mg, 0.549mmol) in IM tetrabutylammonium fluoride solution in THF (0.63 mL), is stirred at room temperature for 1 hour. The reaction mixture is partitioned between ethyl acetate and water. The organic layer is washed with 10% citric acid solution and brine, dried over MgSOzi, filtered and concentrated to afford the title compound as a cream solid (lOOmg, 45%).
Step 6: {5-[5-(lH-Pyrazol-4-yl)-[ 1 ,2 ,4] triazolo [ 1 ,5-a] pyrazin-8-ylamino] -2- morpholinophenyljmethanol
Figure imgf000064_0003
[00289] This compound may be prepared using methods as described for Compound 6, step 4 using [5-(5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-ylamino)-2-morpholin-4-yl-phenyl]-methanol (85mg, 0.21mmol), 4-(4,4,5,5-tetramethyl-[l,3,2]dioxa-borolan-2-yl)-lH-pyrazole (81mg, 0.42mmol) and Pd(PPh3)4 (60mg, 0.052mmol) in 1.5M Na2CO3 (1.12mL) and dioxane (2.OmL). The reaction mixture is purified by silica gel column chromatography, eluting with 95:3 DCM:NH3 (7M in MeOH) followed by reverse phase preparative HPLC purification. The title compound is obtained as a pale brown solid (lOmg, 12%). 1H-NMR (400MHz, ds-DMSO) δ(ppm) 2.87 (4H, m), 3.77 (4H, m), 4.63 (2H, d), 5.12 (IH, t), 7.11 (IH, d), 7.84 (IH, d), 8.12 (IH, s), 8.21 (IH, s), 8.39 (IH, s), 8.69 (IH, s), 8.77 (IH, s), 9.78 (IH, s), 13.29 (IH, br s). LCMS: Rt 2.45min (96.3%), m/z (APCI) 393 (M+H)+.
Compound 41: (6-[Morpholin-4-yl]pyridin-3-yl)-[5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5- a] pyrazin-8-yl] amine
Figure imgf000065_0001
[00290] This compound may be prepared using the methods as described for Compound 120, using (5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-(6-morpholin-4-yl-pyridin-3-yl)amine in Step 4. LCMS: Rt = 0.80 min (95 %), m/z (ESI) 364 (M+H)+.
Compound 42: [6-(4-[Cyclopropylmethyl]piperazin-l-yl)pyridin-3-yl]-[5-(lH-pyrazol-4-yl)- [l,2,4]triazolo[l,5-a]pyrazin-8-yl]amine
Figure imgf000065_0002
[00291] This compound may be prepared using the methods described for Compound 120, using (5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-[6-(4-[cyclopropylmethyl]piperazin-l-yl)pyridin-3- yl]amine in step 4. LCMS: Rt = 0.80 min (95 %), m/z (ESI) 417 (M+H)+. Compound 43: [6-(4-Isopropylpiperazin-l-yl)pyridin-3-yl]-[5-(lH-pyrazol-4-yl)-
[l,2,4]triazolo[l,5-a]pyrazin-8-yl]amine
Figure imgf000066_0001
[00292] This compound may be prepared using methods as described for Compound 120, using (5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-[6-(4-isopropylpiperazin-l-yl)pyridin-3-yl]amine in Step 4. LCMS: Rt = 0.77 min (95 %), m/z (ESI) 405 (M+H)+.
Compound 44: [5-(lH-Pyrazol-4-yl)-[l,2,4]triazolo[l,5-a]pyrazin-8-yl]-{6-[4-(2,2,2- trifluoroethyl) piperazin-l-yl]pyridin-3-yl}amine
Figure imgf000066_0002
[00293] This compound may be prepared using methods as described for Compound 120, using (5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-{6-[4-(2,2,2-trifluoroethyl)piperazin-l-yl]-pyridin- 3-yl} amine in step 4. LCMS: Rt = 0.95 min (95 %), m/z (ESI) 445 (M+H)+.
Compound 46: {4-[4-(Cyclopropylmethyl)piperazin-l-yl]phenyl}-[5-(lH-pyrazol-4-yl)-
[l,2,4]triazolo[l,5-a]pyrazin-8-yl]amine
Figure imgf000067_0001
[00294] This compound may be prepared using methods as described for Compound 120, Step
4, using (5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-{4-[4-cyclopropylmethyl)piperazin-l-yl]phenyl} amine. LCMS: Rt = 1.02 (95 %), m/z (ESI) = 444 (M+H)+.
Compound 47: 4-[8-(6-[Morpholin-4-yl]pyridin-3-ylamino)-[l,2,4]triazolo[l,5-a]pyrazin-5- yl]thiophene-2-carboxylic acid amide
Figure imgf000067_0002
[00295] This compound may be prepared using methods as described for Compound 167, using (5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-(6-morpholin-4-yl-pyridin-3-yl)amine in Step 4. LCMS: Rt = 0.85 min (95 %), m/z (ESI) 423 (M+H)+.
Compound 48: (5-Benzo[b]thiophen-3-yl-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-(4-morpholin-4- ylphenyl)amine
Figure imgf000067_0003
[00296] This compound may be prepared using methods as described for Compound 120, using (5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-(4-morpholin-4-ylphenyl)amine and 4,4,5,5- tetramethyl-2-benzo[b]thiophen-3-yl-[l,3,2]dioxaborolane in step 4. LCMS: Rt 0.84 min (95 %), m/z (ESI) 402 (M+H)+.
Compound 50: (4-Morpholin-4-ylphenyl)-(5-thiophen-3-yl-[l,2,4]triazolo[l,5-a]pyrazin-8- yl)amine
Figure imgf000068_0001
[00297] This compound may be prepared using methods as described for Compound 120, using (5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-(4-morpholin-4-ylphenyl)amine and 4,4,5,5- tetramethyl-2-thiophen-3-yl-[l,3,2]dioxaborolane in step 4. LCMS: Rt 1.19 min (95 %), m/z (ESI) 379 (M+H)+.
Compound 51: [4-(4-Isopropylpiperazin-l-yl)phenyl]-(5-thiophen-3-yl-[l,2,4]triazolo[l,5- a]pyrazin-8-yl)amine
Figure imgf000068_0002
[00298] This compound may be prepared using the methods as described for Compound 50, using (5-bromo[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-[4-(4-isopropylpiperazin-l-yl)-phenyl]amine in the final step. LCMS: rt = 1.11 min (95 %), m/z (ESI) 420 (M+H)+.
Compound 52: 5-(5-Ethyl-lH-pyrazol-4-yl)-N-(4-morpholinophenyl)-[l,2,4]triazolo[l,5- a]pyrazin-8-amine
Step 1: 3 -Ethyl- lH-pyr azote N-N H
[00299] To a stirred solution of l-trimethylsilanyl-pent-l-yn-3-one (0.5g, 3.25mmol) and hydrazine sulphate salt (0.56g, 4.33mmol) in ethanol (15mL), is added sat. Na2CC^ (0.52g, 4.87mmol) and the mixture is refluxed at 9O0C for 5 hours. The reaction mixture is diluted with water and brine and extracted using diethyl ether (3x). The organic layers are combined, dried over MgSOzi, filtered and concentrated to afford a crude product, purified by silica gel column chromatography. Elution with 90:10 petroleum etheπethyl acetate afford the title compound (0.1196g, 36%).
Step 2: 4-Bromo-5-ethyl-lH-pyrazole
Br
W λ
[00300] To a stirred solution of 3-ethyl-lH-pyrazole (0.114g, 1.186mmol) in acetic acid
(2mL), bromine (0.06ImL, 1.186mmol) is added and the mixture is stirred at room temperature for 2 hours. The reaction mixture is basified with sat. NaHCθ3, and extracted using ethyl acetate (3x). The organic layers are combined, dried over MgSOzi, filtered and concentrated to give the title compound (0.180g, 87%). The compound is used in the next step without further purification.
Step 3: 4-Bromo-5-ethyl-l-(tetrahydro-pyran-2-yl)-lH-pyrazole and 4-Bromo-3 -ethyl- l-(tetrahydro- pyran-2-yl) - lH-pyr azole
Figure imgf000069_0001
[00301] 4-Bromo-5-ethyl-lH-pyrazole (0.179g, 1.032mmol) is dissolved in 3,4-dihydro-2H- pyran (0.28mL, 3.098mmol) in the presence of a catalytic amount of TFA (0.00ImL, 0.00103mmol). The reaction mixture is stirred at 9O0C for 3 hours, cooled and then quenched using NaH (1.5mg, 0.0061mmol). After removing the solvent, the residue is purified by silica gel column chromatography eluting with a mixture of 90:10 petroleum ether-ethyl acetate. The fractions containing the desired compounds are collected and concentrated in vacuo to afford the title compounds (175mg, 66%).
Step 4: 5-Ethyl-l-(tetrahydro-pyran-2-yl)-4-(4, 4, 5, 5-tetramethyl-[l, 3, 2] dioxaborolan-2-yl)-lH- pyrazote and 3-Ethyl-l-(tetrahydro-pyran-2-yl)-4-(4, 4, 5, 5-tetramethyl-fl, 3, 2] dioxaborolan-2-yl)-lH- pyr azote
Figure imgf000070_0001
[00302] This compound may be prepared using methods as described for Compound 86, step 2 using 4-bromo-5-ethyl-l-(tetrahydro-pyran-2-yl)-lH-pyrazole and 4-bromo-3-ethyl-l-(tetrahydro- pyran-2-yl)-lH-pyrazole (168mg, 0.65mmol), bis(pinacolato)diboron (331mg, 1.3mmol), PdC^dppf (53mg, 65μmol) and KOAc (190mg, 1.95mmol) in dimethylsulfoxide (2mL). The reaction mixture is purified by silica gel column chromatography eluting with 90:10 petroleum ether: ethyl acetate to afford the title compounds (όl.lmg, 31%).
Step 5: {5-[5-Ethyl-l-(tetrahydro-pyran-2-yl)-lH-pyrazol-4-yl]-[l,2,4]triazolo[l,5-a]pyrazin-8-yl}- (4-morpholin-4-yl-phenyl) -amine and {5-[3-Ethyl-l-(tetrahydro-pyran-2-yl)-lH-pyrazol-4-yl]- [l,2,4]triazolo[l,5-a]pyrazin-8-yl}-(4-morpholin-4-yl-phenyl)-amine
Figure imgf000070_0002
[00303] This compound may be prepared using methods as described for Compound 6, step 4 using (5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-(4-morpholin-4-yl-phenyl)-amine (0.032g, 0.085mmol), 5-ethyl-l-(tetrahydro-pyran-2-yl)-4-(4,4,5,5-tetramethyl-[l,3,2]dioxaborolan-2-yl)-lH- pyrazole and 3-ethyl-l-(tetrahydro-pyran-2-yl)-4-(4,4,5,5-tetramethyl-[l,3,2]dioxaborolan-2-yl)-lH- pyrazole (0.52mg, 0.17mmol), and Pd(PPh3)4 (0.025mg, 0.021mmol) in 1.5M Na2CO3 (aq) (0.45mL, O.όmmol) and dioxane (2mL). The reaction mixture is purified by silica gel column chromatography eluting with 1 :1 petroleum etheπethyl acetate followed by 1:4 petroleum etheπethyl acetate to yield the title compound (45mg).
Step 6: 5-(5-Ethyl-lH-pyrazol-4-yl)-N-(4-morpholinophenyl)-[l,2,4]triazolo[l,5-a]pyrazin-8-amine
Figure imgf000070_0003
[00304] A solution of {5-[5-ethyl-l-(tetrahydro-pyran-2-yl)-lH-pyrazol-4-yl]-
[l,2,4]triazolo[l,5-a]pyrazin-8-yl}-(4-morpholin-4-yl-phenyl)-amine and {5-[3-ethyl-l-(tetrahydro- pyran-2-yl)-lH-pyrazol-4-yl]-[l,2,4]triazolo[l,5-a]pyrazin-8-yl}-(4-morpholin-4-yl-phenyl)-amine (40mg, 0.077mmol) and cone. HCl (0.3mL) in MeOH (1OmL) is stirred at room temperature for 5 hours. After removing the solvent, the solid residue is partitioned between ethyl acetate and sat. NaHCθ3. The undissolved solid is collected by filtration, washed with water, diethyl ether and petroleum ether and dried to afford the title compound (5mg, 17%). Conversion into the mesylate salt using 0.1M methanesulfonic acid (0.128mL) affords the title compound (5.6mg, 89%) as a solid. 1H-NMR (400MHz, d6-DMSO) δ(ppm) 1.13 (3H, m), 2.29 (3H, s, MsOH), 2.74 (2H, m), 3.29 (4H, m), 3.77 (4H, m), 7.06 (2H, m), 7.62 (IH, s), 7.89 (2H, d), 8.00 (IH, s), 8.61 (IH, s), 9.83 (IH, s). LCMS: Rt 2.69min (98.4%), m/z (APCI) 391 (M+H)+.
Compound 53: 6-[8-(4-Morpholin-4-yl-phenylamino)-[l,2,4]triazolo[l,5-a]pyrazin-5-yl]-l,l- dioxo-l,2-dihydro-lλ6-benzo[d]isothiazol-3-one
Step 1: l,l-Dioxo-6-(4,4,5,5-tetramethyl-[l,3,2]dioxaborolan-2-yl)-l,2-dihydro-l λ - benzofdJisothiazol-3-one
Figure imgf000071_0001
[00305] This compound may be prepared using methods as described for Compound 86, step 2 using 6-bromo-l,l-dioxo-l,2-dihydro-l λ6-benzo[d]isothiazol-3-one (0.5, 1.9mmol), bis(pinacolato)diboron (0.53g, 2.1mmol), PdCl2dppf (0.047g, 0.058mmol) and KOAc (0.56g, 5.7mmol) in dioxane (1OmL). The solvent is removed in vacuo and the residue is partitioned between DCM and water. The organic layer is washed with sat NaHCθ3 and 2M HCl, dried over MgSOzi, filtered and evaporated to afford the title compound (990mg, 169%) used in the next step without further purification.
Step 2: (4-Morpholin-4-yl-phenyl)-[5-(l, 1, 3-trioxo-2, 3-dihydro-lH-l λ6-benzo[d]isothiazol-6-yl)- [l,2,4]triazolo[l,5-a]pyrazin-8-yl]-carbamic acid tert-butyl ester
Figure imgf000072_0001
[00306] This compound may be prepared using methods as described for Compound 6, step 4 using (5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-(4-morpholin-4-yl-phenyl)-carbamic acid tert-butyl ester (170mg, 0.36mmol), l,l-dioxo-6-(4,4,5,5-tetramethyl-[l,3,2]dioxaborolan-2-yl)-l,2-dihydro-l λ6-benzo[d]isothiazol-3-one (374mg, 0.72mmol) and Pd(PPh3)4 (lOOmg, 0.082mmol) in 1.5M Na2CO3 (2mL, 3mmol) and dioxane (6mL). Purification of the reaction mixture by silica gel column chromatography using 1 :1 petroleum ether: ethyl acetate affords a compound still impure. A second silica gel column chromatography eluting with 10:1 DCM:MeOH affords the title compound (91mg, 44%).
Step 3: 6-[8-(4-Morpholin-4-yl-phenylamino)-[l ,2,4Jtriazolo[l ,5-aJpyrazin-5-ylJ-l ,1-dioxo-l ,2- dihydro-1 λ6-benzo[d]isothiazol-3-one
Figure imgf000072_0002
[00307] A suspension of (4-morpholin-4-yl-phenyl)-[5-(l,l,3-trioxo-2,3-dihydro-lH-l λ6- benzo[d]isothiazol-6-yl)-[l,2,4]triazolo[l,5-a]pyrazin-8-yl]-carbamic acid tert-butyl ester (lOOmg, 0.173mmol) in 4M HCl (2.5mL) in dioxane is stirred at room temperature for 2 hours. The solvent is removed under vacuum and the residue is triturated with DCM, diethyl ether and petroleum ether to afford the title compound (84mg, 100%). 1H-NMR (400MHz, ds-DMSO) δ(ppm) 3.25 (4H, m), 3.85 (4H, m), 7.20 (2H, d), 7.88 (2H, d), 8.16 (IH, d), 8.24 (IH, s), 8.58 (IH, d), 8.82 (2H, s), 8.96 (IH, s), 10.31 (IH, s). LCMS: Rt 2.31min (95.7%), m/z (APCI) 478 (M+H)+.
Compound 54: 4-{8-[6-(4-[Cyclopropylmethyl]piperazin-l-yl)-pyridin-3-ylamino]-[l,2,4]triazolo [l,5-a]pyrazin-5-yl}thiophene-2-carboxylic acid amide
Figure imgf000073_0001
[00308] This compound may be prepared using the methods described for Compound 120, using (5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-[6-(4-[cyclopropylmethyl]piperazin-l-yl)pyridin-3- yl]amine in step 4. LCMS: Rt = 0.85 min (95 %), m/z (ESI) 476 (M+H)+.
Compound 55: 4-{8-[6-(4-Isopropylpiperazin-l-yl)pyridin-3-ylamino]-[l,2,4]triazolo[l,5- a]pyrazin-5-yl}thiophene-2-carboxylic acid amide
Figure imgf000073_0002
[00309] This compound may be prepared using the methods as described for Compound 167, using (5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-[6-(4-isopropylpiperazin-l-yl)pyridin-3-yl]amine in Step 4. LCMS: Rt = 0.82 min (95 %), m/z (ESI) 464 (M+H)+.
Compound 56: 4-(8-{6-[4-(2,2,2-Trifluoroethyl)piperazin-l-yl]-pyridin-3-ylamino}-[l,2,4]triazolo [l,5-a]pyrazin-5-yl)thiophene-2-carboxylic acid amide
Figure imgf000074_0001
[00310] This compound may be prepared using the methods as described for Compound 167, using (5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-{6-[4-(2,2,2-trifluoroethyl)piperazin-l-yl]-pyridin- 3-yl}amine in step 4. LCMS: Rt = 0.99 min (95 %), m/z (ESI) 504 (M+H)+.
Compound 57: 4-(8-{4-[4-(2,2,2-Trifluoroethyl)piperazin-l-yl]-phenylamino}-[l,2,4]triazolo[l,5- a]pyrazin-5-yl)thiophene-2-carboxylic acid amide
Figure imgf000074_0002
[00311] This compound may be prepared using methods as described for Compound 58, using
(5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-{4-[4-(2,2,2-trifluoroethyl)piperazin-l-yl]phenyl} amine. LCMS: Rt = 1.07 min (95 %), m/z (ESI) 503 (M+H)+.
Compound 58: 4-{8-[4-(4-(Cyclopropylmethyl)piperazin-l-yl)-phenylamino]-[l,2,4]triazolo[l,5- a]pyrazin-5-yl}thiophene-2-carboxylic acid amide
Figure imgf000075_0001
[00312] This compound may be prepared using methods as described for Compound 46, using
2-(aminocarbonyl)thiophene-4-boronic acid. LCMS: Rt = 0.93 (95 %), m/z (ESI) 475 (M+H)+.
Compound 59: [4-(4-Cyclopropylpiperazin-l-yl)phenyl]-[5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5- a] pyrazin-8-yl] amine
Figure imgf000075_0002
[00313] This compound may be prepared using methods as described for Compound 46, using
(5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-[4-(4-cyclopropylpiperazin-l-yl)-phenyl]amine. LCMS: Rt = 0.84 (95 %), m/z (ESI) 402 (M+H)+.
Compound 60: [6-(4-Cyclopropylpiperazin-l-yl)pyridin-3-yl]-[5-(lH-pyrazol-4-yl)-[l,2,4]triazolo [l,5-a]pyrazin-8-yl]amine
N
Figure imgf000075_0003
[00314] This compound may be prepared using the methods described for Compound 120, using (5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-[6-(4-cyclopropylpiperazin-l-yl)pyridin-3- yl]amine in step 4. LCMS: Rt = 0.76 min (95 %), m/z (ESI) 403 (M+H)+.
Compound 61: 4-[8-(4-Morpholin-4-ylphenylamino)-[l,2,4]triazolo[l,5-a]pyrazin-5-yl]thiazole- 2-carboxylic acid amide
Figure imgf000076_0001
[00315] This compound may be prepared using methods as described for Compound 73, using ammonia (7 M in MeOH) is used in step 3. LCMS: Rt = 1.00 min (95 %), m/z (ESI) 423 (M+H)+.
Compound 62: 4-{8-[4-(4-Isopropylpiperazin-l-yl)phenylamino]-[l,2,4]triazolo[l,5-a]pyrazin-5- yl}thiazole-2-carboxylic acid amide
Figure imgf000076_0002
[00316] This compound may be prepared using methods as described for Compound 61, using
(5-bromo[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-[4-(4-isopropylpiperazin-l-yl)-phenyl]amine in the final step. LCMS: rt = 0.96 min (95 %), m/z (ESI) 464 (M+H)+.
Compound 63: 4-(8-{4-[l-(2,2,2-Trifluoroethyl)piperidin-4-yl]phenylamino}-[l,2,4]triazolo[l,5- a]pyrazin-5-yl)thiophene-2-carboxylic acid amide
Step 1: l-Trifluoroacetyl-4-(4-nitrophenyl)piperidine
Figure imgf000077_0001
[00317] Triethlyamine (1.0 mL, 7.3 mmol) and 4-(4-nitrophenyl)piperidine (1.0 g, 4.8 mmol) are stirred in DCM (25 mL) at 0°C under N2 and trifluoroacetic anhydride (0.81 mL, 5.8 mmol) is added. The mixture is stirred for three days, allowing the temperature to warm to rt. The solution is then diluted with DCM (50 mL) and washed with water (2 x 15 mL), NaHCO3 (50% sat. aq., 2 x 15 mL) and brine (15 mL). The solvent is dried over MgSθ4 and evaporated to afford the desired compound (1.46 g, 4.66 mmol).
Step 2: l-(2,2,2-Trifluoroethyl)-4-(4-nitrophenyl)piperidine
Figure imgf000077_0002
[00318] A solution of l-trifluoroacetyl-4-(4-nitrophenyl)piperidine (1.42 g, 4.7 mmol) in THF
(15 mL) is stirred in 25 mL 2-necked flask fitted with a condenser and pressure-equalising addition funnel. The system is flushed with N2, NaBH4 (210 mg, 5.6 mmol) is added and the flask is cooled to 0°C. A solution of iodine (600 mg, 2.3 mmol) in THF (5 mL) is then added dropwise over 20 minutes, after which the addition funnel is removed and the mixture heated at reflux overnight. The resulting pale yellow suspension is cooled to rt and MeOH (1.5 mL) is added cautiously, causing vigorous evolution of a gas. Evaporation of the solvents affords the title compound, which is used without further purification.
Step 3 : 4-fl - (2, 2, 2- Trifluoroethyl)piperidin-4-yl]phenylam ine
Figure imgf000077_0003
[00319] Ammonium formate (1.38 g, 22 mmol) and 10% Pd/C (230 mg, 0.2 mmol) are added to a solution of l-(2,2,2-trifluoroethyl)-4-(4-nitrophenyl)piperidine (1.26 g, 4.4 mmol) in EtOH (10 mL) and EtOAc (10 mL). The suspension is heated at reflux for 24 hours, adding further portions of ammonium formate (2 g) after 4 h and 8 h. The mixture is filtered through celite and evaporated to afford an orange solid. This is partitioned between DCM (40 mL) and water (20 mL) and the layers separated. The aqueous phase is extracted with DCM (2 x 20 mL) and the combined organic layers are dried over MgSO4 and evaporated under reduced pressure to afford N- {4-[l -(2,2,2- trifluoroethyl)piperidin-4-yl]phenyl} formamide as a pale orange solid (980 mg). [00320] A solution of the formamide in MeOH (20 mL) is stirred at rt and HCl (cone, 1 mL) is added. The deep purple solution is heated at reflux for 1 h, cooled and the MeOH is evaporated. The residue is stirred with water (20 mL) and ΝaHCθ3 (sat. aq.) is added until bubbling ceases. The mixture is extracted with DCM (20 mL, 2 x 10 mL) and the combined extracts are dried over MgSθ4 and evaporated under reduced pressure to afford the title compound as an orange solid (870 mg). [00321] This material is used to prepare 4-(8-{4-[l-(2,2,2-trifluoroethyl)piperidin-4- yl]phenylamino}-[l,2,4]triazolo[l,5-a]pyrazin-5-yl)thiophene-2-carboxylic acid amide using methods analogous to those used for Compound 89.
Figure imgf000078_0001
[00322] LCMS: Rt = 2.11 min (100%), m/z (ESI) 502 (M+H)+.
Compound 65: 5-(8-(4-(2-Morpholinoethoxy)phenylamino)-[l,2,4]triazolo[l,5-a]pyrazin-5- yl)isoindolin-l-one
Step 1: (5-Bromo-[ 1, 2,4] triazolo[l,5-a]pyrazin-8-yl)-[4-(2-morpholin-4-yl-ethoxy)-phenyl] -amine
Figure imgf000078_0002
[00323] According to the procedure described for Compound 6, step 1, 5,8-dibromo-
[l,2,4]triazolo[l,5-a]pyrazine (0.5g, 1.799mmol), 4-(2-morpholin-4-yl-ethoxy)-phenylamine (0.6g, 4.5mmol) and N,N-diisopropylethylamine (0.47mL, 2.7mmol) in 2-propanol (6mL) are stirred at 950C overnight. After evaporation of the solvent the residue is dissolved in DCM and washed with water (2x) and brine. The organic layer is dried over MgSOzi, filtered and concentrated in vacuo to afford an oil purified by silica gel column chromatography. Elution with a 97: 3 mixture DCM:MeOH gives the title compound (650mg 86%) as a light yellow solid. LCMS: Rt 2.06min (97.7%). Step 2: 5-(8-(4-(2-Morpholinoethoxy)phenylamino)-[l,2,4]triazolo[l,5-a]pyrazin-5-yl)isoindolin-l- one
Figure imgf000079_0001
[00324] This compound may be prepared using the methods as described for Compound 6, step 4, using (5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-[4-(2-morpholin-4-yl-ethoxy)-phenyl]- amine (80mg, 0.19mmol), 5-(4,4,5,5-tetramethyl-[l,3,2]dioxaborolan-2-yl)-2,3-dihydro-isoindol-l- one (74mg, 0.29mmol), and Pd(PPh3)4 (55mg, 0.047mmol) in 1.5M Na2CO3 (1.02mL, 1.53mmol) and dioxane (3mL). The reaction mixture is purified by silica gel column chromatography eluting with DCM and a 97:3 mixture DCM:NH3 (7M in MeOH). After trituration using diethyl ether, the title compound is isolated as a solid (61.9mg, 69%). Conversion of the material into the mesylate salt, using IM methanesulfonic acid (0.134mL) in MeOH, affords the title compound (57.1mg). 1H-NMR (400MHz, ds-DMSO) δ(ppm) 2.40 (3H, s, MsOH), 3.25-3.38 (2H, m), 3.60-3.78 (4H, m), 3.84 (2H, t), 4.09 (2H, d), 4.45 (2H, m), 4.56 (2H, s), 7.14 (2H, d), 7.90 (IH, d), 8.05-8.19 (4H, m), 8.28 (IH, s), 8.76 (IH, br s), 8.79 (IH, s), 10.00 (IH, br s), 10.16 (IH, s). LCMS: Rt 1.98min (99.1%), m/z (APCI) 472 (M+H)+.
Compound 67: N-(2-Fluoro-4-morpholinophenyl)-5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5- a]pyrazin-8-amine
Step 1: (5-Bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-(2-fluoro-4-morpholin-4-yl-phenyl)-amine
Figure imgf000079_0002
[00325] This compound may be prepared using methods as described for Compound 6, step 1, using 5,8-dibromo-[l,2,4]triazolo[l,5-a]pyrazine (0.105g, 0.89mmol), 2-fluoro-4-morpholin-4-yl- phenylamine (93mg, 0.474mmol), DIPEA (0.123mL, 0.706mmol) and l,4-diazabicyclo[2.2.2]octane (53mg, 0.472mmol) in 2-propanol (2mL). The reaction mixture is partitioned between DCM and 10% citric acid (aq) solution, the organic layer is separated and washed with 10% citric acid solution, water and brine, dried over MgSOzi, filtered and concentrated in vacuo. The title compound is isolated as a pale red solid (77mg, 42%) and used in the next step without further purification. LCMS: Rt 3.48min (89%).
Step 2: N-(2-Fluoro-4-morpholinophenyl)-5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5-a]pyrazin-8-amine
Figure imgf000080_0001
[00326] This compound may be prepared using methods as described for Compound 6, step 4, using (5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-(2-fluoro-4-morpholin-4-yl-phenyl)-amine (80mg, 0.203mmol), 4-(4,4,5,5-tetramethyl-[l,3,2]dioxaborolan-2-yl)-lH-pyrazole (79mg, 0.406mmol), and Pd(PPh3)4 (59mg, 0.051mmol) in 1.5N Na2CO3 (1.09mL, 1.62mmol) and dioxane (3.25mL). The reaction mixture is purified by silica gel column chromatography eluting with 97:3 DCM:NH3 (7M in MeOH). The fractions containing the product are combined and evaporated in vacuo to afford a solid which is triturated with diethyl ether and petroleum ether to yield the title compound (29mg, 38%). LCMS: Rt 2.90min (96%).
[00327] Conversion into the mesylate salt using 0.1M methanesulfonic acid solution in MeOH
(0.762mL, 0.076mmol) affords the title compound (35mg). 1H-NMR (400MHz, de-DMSO) δ(ppm) 2.36 (3H, s, MsOH), 3.19 (4H, m), 3.79 (4H, m), 6.84-6.94 (2H, m), 7.54 (IH, t), 8.08 (IH, s), 8.48 (2H, s), 8.76 (IH, s), 9.31 (IH, s). LCMS: Rt 2.91min (95.4%), m/z (APCI) 381 (M+H)+.
Compound 70: (4-(8-(4-Morpholinophenylamino)-[l,2,4]triazolo[l,5-a]pyrazin-5-yl)-lH-pyrazol- 5-yl)methanol
Step 1 : 4-Bromo-5-(tert-butyl-dimethyl-silanyloxymethyl)-lH-pyrazole
Figure imgf000080_0002
[00328] A solution of (4-bromo-2H-pyrazol-3-yl)-methanol (0.64mg, 3.63mmol), tert- butyldimethylsilyl chloride (0.82g, 5.45mmol) and imidazole (0.42g, 6.18mmol) in NN- dimethylformamide (2OmL) is stirred at room temperature overnight. The reaction mixture is diluted with a 50:50 mixture diethyl etheπethyl acetate and washed with water (3x). The organic layers are dried over MgSO4, filtered and concentrated. The residue is purified by silica gel column chromatography eluting with 80:20 petroleum etheπethyl acetate to afford the title compound (1.035g, 98%). Step 2: 4-Bromo-3- (tert-butyl-dimethyl-silanyloxymethyl) -I- (tetrahydro-pyran-2-yl) - lH-pyrazole and 4-Bromo-5- (tert-butyl-dimethyl-silanyloxymethyl) -I- (tetrahydro-pyran-2-yl) - lH-pyr azole
Figure imgf000081_0001
[00329] 4-Bromo-5-(tert-butyl-dimethyl-silanyloxymethyl)-l H-pyrazole (Ig, 3.45mmol) is dissolved in 3,4-dihydro-2H-pyran (0.944mL, 10.34mmol) in the presence of a catalytic amount of TFA (0.0026mL, 0.035mmol). The reaction mixture is stirred at 9O0C for 18 hours, cooled and then quenched using NaH (4.69mg, 0.206mmol). After removing the solvent, the residue is purified by silica gel column chromatography eluting with a mixture of 95:5 petroleum ether-ethyl acetate, followed by 90:10 petroleum ether-ethyl acetate. The fractions containing the desired compounds are collected and concentrated in vacuo to afford 4-bromo-3-(tert-butyl-dimethyl-silanyloxymethyl)-l- (tetrahydro-pyran-2-yl)- 1 H-pyrazole and 4-bromo-5-(tert-butyl-dimethyl-silanyloxymethyl)- 1 - (tetrahydro-pyran-2-yl)- lH-pyrazole (724mg, 56%).
Step 3 : {5- [5- (tert-Butyl-dimethyl-silanyloxymethyl) - 1 -(tetrahydro-pyran-2-yl) - lH-pyrazol-4-yl] - fl,2,4Jtriazolo[l,5-aJpyrazin-8-yl}-(4-morpholin-4-yl-phenyl)- carbamic acid tert-butyl ester and {5- [3- (tert-Butyl-dimethyl-silanyloxymethyl) -I- (tetrahydro-pyran-2-yl) - lH-pyrazol-4-yl] - [l,2,4Jtriazolo[l,5-aJpyrazin-8-yl}-(4-morpholin-4-yl-phenyl)-carbamic acid tert-butyl ester
Figure imgf000081_0002
[00330] This compound may be prepared using the methods as described for Compound 35, step 3 using (4-morpholin-4-yl-phenyl)-(5-tributylstannanyl-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)- carbamic acid tert-butyl ester (210mg, 0.30mmol) 4-bromo-3-(tert-butyl-dimethyl-silanyloxymethyl)- 1 -(tetrahydro-pyran-2-yl)- 1 H-pyrazole and 4-bromo-5-(tert-butyl-dimethyl-silanyloxymethyl)- 1 - (tetrahydro-pyran-2-yl)- 1 H-pyrazole (170mg, 0.45mmol) and tetrakis(triphenylphosphine)palladium(0) (35mg, 0.030mmol) in DMF (4mL). The crude compound is purified by silica gel column chromatography eluting with 7:3 then 3:7 petroleum etheπethyl acetate to afford the title compounds (62.5mg). Step 4: (4-(8-(4-Morpholinophenylamino)-[l,2,4]triazolo[l,5-a]pyrazin-5-yl)-lH-pyrazol-5- yl)methanol
Figure imgf000082_0001
[00331] A solution of {5-[3-(tert-butyl-dimethyl-silanyloxymethyl)-l-(tetrahydro-pyran-3-yl)- lH-pyrazol-4-yl]-[l,2,4]triazolo[l,5-a]pyrazin-8-yl}-(4-morpholin-4-yl-phenyl)-amine and {5-[5- (tert-butyl-dimethyl-silanyloxymethyl)-l-(tetrahydro-pyran-3-yl)-lH-pyrazol-4-yl]- [l,2,4]triazolo[l,5-a]pyrazin-8-yl}-(4-morpholin-4-yl-phenyl)-amine (53.6mg, 0.077mmol) and cone. HCl (0.27mL) in MeOH (5mL) is stirred at room temperature for 2 hours. After removing the solvent, the residue is partitioned between ethyl acetate and water. The aqueous layer is washed with ethyl acetate (3x) and the combined organic layers are dried over MgSOzi, filtered and concentrated. The residue is dissolved in 4M HCl in dioxane, concentrated in vacuo and then partitioned between ethyl acetate and sat. NaHCθ3. The organic layer is dried over MgSOzi, filtered and concentrated in vacuo to afford a crude compound purified by silica gel column chromatography. The title compound is isolated eluting with EtOAc and a 95:5 mixture DCMMeOH (5.4mg, 18%).
1H-NMR (400MHz, d6-DMSO) δ(ppm) 3.11 (4H, m), 3.79 (4H, m), 4.65 (2H, d), 5.25 and 5.53 (IH, br s), 6.99 (2H, m), 7.84 and 8.51 (IH, br s), 7.92 (2H, d), 8.12 (IH, d), 8.70 (IH, s), 9.74 (IH, s), 13.12 and 13.23 (IH, br s). LCMS: Rt 2.26min (96.2%), m/z (APCI) 393 (M+H)+.
Compound 73: 4-[8-(4-Morpholin-4-yl-phenylamino)-[l,2,4]triazolo[l,5-a]pyrazin-5-yl]-thiazole- 2-carboxylic acid methylamide
Step 1: 2,4-Dibromothiazole
Figure imgf000082_0002
[00332] Thiazolidinone (3.43 g, 29.32 mmol) and POBr3 (25 g, 87.96 mmol, 3 equiv.) are mixed under nitrogen as solids. The reaction mixture is then heated to HO0C under stirring for 3 h causing the formation of a black syrup. The reaction mixture is then allowed to cool down to room temperature and a mixture of water/ice (200 mL) is added very cautiously. The resulting grey suspension is extracted with diethyl ether (3 x 50 mL), the organic layers are combined, filtered through a silica plug and evaporated to afford the title compound as an orange oil (4 g, 57%) which is used in the next step without further purification. Step 2: 4-Bromothiazole-2-carboxylic acid ethyl ester
Figure imgf000083_0001
[00333] To a solution of dibromothiazole (1 g, 4.15 mmol) in THF (15 mL) at O0C, is added dropwise a solution of iPrMgCl in THF (2 M, 2.3 mL, 4.57 mmol, 1.10 equiv.). The reaction is stirred at 0°C for 0.25 h. To the resulting orange solution is added via a cannula, diethyl carbonate (3 mL) in THF (5 ml). The resulting green solution is stirred further at room temperature for 0.5 h, at which point the reaction is quenched by adding saturated NH4Cl. The title compound is purified by LC using 6/4 cyclohexane/DCM as the eluent to afford 514.2 mg (52%) of the title compound as a pale yellow powdery solid.
Step 3: 4-Bromothiazole-2-carboxylic acid methylamide
Figure imgf000083_0002
[00334] 4-Bromothiazole-2-carboxylic acid ethyl ester (500 mg, 2.13 mmol) is dissolved in methanol (1 mL) and methyl amine in methanol (10 mL) is added. The mixture is stirred overnight at room temperature. Evaporation of the solvent under reduced pressure affords the title compound as yellow solid. LCMS: Rt 0.92 min (100%) m/z (ESI) 219/221 (M+H)+.
Step 4: 4-[8-(4-Morpholin-4-yl-phenylamino)-[ 1,2,4] 'triazolofl,5-aj ' pyrazin-5-yl] -thiazole-2- carboxylic acid methylamide
Figure imgf000083_0003
[00335] 4-Bromothiazole-2-carboxylic acid methylamide is converted to the boronate in a fashion analogous to that described for Compound 6, Step 3. The title compound is then prepared using methods as described for Compound 120,, step 4. LCMS: Rt = 1.06 min (95 %), m/z (ESI) 437 (M+H)+.
Compound 79: 5-(8-(6-(4-Isopropylpiperazin-l-yl)pyridin-3-ylamino)-[l,2,4]triazolo[l,5- a]pyrazin-5-yl)isoindolin-l-one
Step 1 : 4-Bromo-2-bromomethyl-benzoic acid methyl ester
Figure imgf000084_0001
[00336] 4-Bromo-2-methyl-benzoic acid (4.6 g, 21.39 mmol) is dissolved in 2M HCl in
MeOH and refluxed for 3 hours. The solvent is evaporated to give the 4-bromo-2-methyl-benzoic acid methyl ester (4.24 g, 86 %). This intermediate (18.51 mmol) is dissolved in carbon tetrachloride (100 mL) and N-bromosuccinimide (NBS) (5.57 g, 24.06 mmol) is added. AIBN (122 mg, 740 μmol) is then added and the mixture purged with nitrogen for 5 min. The reaction mixture is then refluxed for 4 hours. After cooling to room temperature the reaction mixture is filtered and the filtrate is evaporated. The residue is purified by flash chromatography (silica gel, 2:1 petroleum ether/ethyl acetate) to give the title compound (3.42g, 60 %).
Step 2: 5-Bromo-2,3-dihydro-isoindol-l-one
Figure imgf000084_0002
[00337] 4-Bromo-2-bromomethyl-benzoic acid methyl ester (0.5g, 16.2mmol) is treated with methanolic ammonia (1OmL, 7 N NH3 in MeOH) for 5 minutes at 9O0C. After cooling to room temperature a precipitate is formed, collected by filtration and washed with a small amount of methanol to afford the title compound as a colourless solid (224mg, 65%). 1H-NMR (400MHz, dζ- DMSO) δ(ppm) 4.41 (2H, s), 7.64 (IH, d), 7.70 (IH, d), 7.87 (IH, s), 8.67 (IH, br s). LCMS: Rt 2.49min, (99.6%), m/z (APCI) 212 (M+H)+.
Step 3: 5-(4, 4, 5, 5-Tetramethyl-fl, 3, 2] dioxaborolan-2-yl)-2, 3-dihydro-isoindol-l-one
Figure imgf000085_0001
[00338] 5-Bromo-2,3-dihydro-isoindol-l-one (230mg, 1.08mmol), bis(pinacolato)diboron
(300mg, l.lδmmol), PdCl2dppf (25mg, 31μmol) and KOAc (320mg, 3.26mmol) are suspended in dioxane (4mL), purged with nitrogen for 5 minutes and then heated at 850C overnight. The solvent is removed in vacuo and the residue partitioned between ethyl acetate and water. The aqueous layer is extracted with ethyl acetate (3x) and the combined organic phases are washed once with brine, filtered through MgSθ4 and evaporated. The solid residue is triturated with hexane and dried in vacuo to yield the title compound (185mg, 66 %) as a grey solid. 1H-NMR (400MHz, CDCl3) δ(ppm) 1.37 (12H, s), 4.45 (2H, s), 6.38 (IH, br s), 7.87 (IH, d), 7.93 (2H, m).
Step 4: l-Isopropyl-4-(5-nitro-pyridin-2-yl)-piperazine
Figure imgf000085_0002
[00339] To a solution of 2-chloro-5-nitropyridine (2.5g, 15.7mmol) in THF (25mL), are added
1 -isopropylpiperazine (2.01g, 15.7mmol) and K2CO3 (3.25g, 23.6mmol). The reaction mixture is stirred at 5O0C for 4 hours and then at 7O0C overnight. The solvent is removed in vacuo and the resultant orange solid is triturated using 10:1 petroleum ether-diethyl ether. The isolated compound (3.7g, 94%) is used in the next step without further purification.
Step 5: 6-(4-Isopropyl-piperazin-l-yl)-pyridin-3-yl-amine
Figure imgf000085_0003
[00340] l-Isopropyl-4-(5-nitro-pyridin-2-yl)-piperazine (0.9g, 3.6mmol) is dissolved in MeOH
(20 mL) and tin (II) dichloride dihydrate (4g, 18mmol) is added. The mixture is cooled using a water bath and cone. HCl is added (4mL). The reaction is stirred at room temperature overnight. After removing the methanol, the resultant light yellow solution is basified using cone. NaOH (pH 11) and a white precipitate is formed. The solid is collected by filtration and the water is extracted with diethyl ether (5x). The organic layers are combined, dried over MgSOzi, filtered, concentrated under vacuum to afford an orange oil which crystallizes on standing to afford an orange solid (0.68g, 86%).
Step 6: 5-Bromo-8-(6-(4-isopropylpiperazin-l-yl)pyridin-3-yl)-[l,2,4]triazolo[l,5-a]pyrazin-8-amine
Figure imgf000086_0001
[00341] This compound may be prepared using methods as described for Compound 6, step 1, using 5,8-dibromo-[l,2,4]triazolo[l,5-a]pyrazine (0.188g, 0.68mmol), 6-(4-isopropyl-piperazin-l-yl)- pyridin-3-ylamine (0.18Og, 0.816mmol) and N-ethyldiisopropyl-amine (0.2OmL, 1.02mmol) in 2- propanol (2mL). Purification of the crude material by silica gel column chromatography, using DCM followed by 95:5 DCM:MeOH, affords the title compound as a pale brown solid (260mg, 92%).
Step 5: 5-(8-(6-(4-Isopropylpiperazin-l-yl)pyridin-3-ylamino)-[ 1,2,4] triazolo[l,5-a] pyrazin-5- yl)isoindolin- 1 -one
Figure imgf000086_0002
[00342] This compound may be prepared using methods as described for Compound 6, step 4, using 5-bromo-N-(6-(4-isopropylpiperazin-l-yl)pyridin-3-yl)-[l,2,4]triazolo[l,5-a]pyrazin-8-amine (50mg, 0.12mmol), 5-(4,4,5,5-tetramethyl-[l,3,2]dioxaborolan-2-yl)-2,3-dihydro-isoindol-l-one (56mg, 0.216mmol) and Pd(PPh3)4 (35mg, 0.03mmol) in 1.5M Na2CO3 (0.64mL) and dioxane (2mL). The crude product is purified by silica gel column chromatography eluting with 95:5 DCM:MeOH followed by 90:10 DCM:MeOH. The title compound is obtained after trituration with a 10:1 mixture «-hexane:DCM (19mg, 34%). Conversion into the mesylate salt, using 0.1M methanesulfonic acid (0.35mL), gives the title compound as a yellow solid (20mg, 69%).
1H-NMR (400MHz, ds-DMSO) δ(ppm) 1.34 (6H, d), 2.36 (6H, s, 2xMsOH), 3.16-3.21 (5H, m), 3.57 (2H, m), 4.45 (2H, d), 4.52 (2H, s), 7.11 (IH, d), 7.86 (IH, d), 8.01 (IH, s), 8.10 (IH, d), 8.24-8.29 (2H, m), 8.72 (IH, s), 8.76 (IH, s), 8.82 (IH, d), 9.39 (IH, br s), 10.21 (IH, s). LCMS: Rt 1.92min (98.5%), m/z (APCI) 470 (M+H)+.
Compound 80: 3-[8-(4-Morpholin-4-ylphenylamino)-[l,2,4]triazolo[l,5-a]pyrazin-5-yl]-benzo[b] thiophene-7-carboxylic acid amide
Step 1: l-Bromo-2-(2-ethoxyvinylsulfanyl)benzene
Figure imgf000087_0001
[00343] To a solution of 2-bromothiophenol (10.50 g, 55.87 mmol) in dry DMF (50 mL) under nitrogen is added cautiously potassium carbonate (9.2 mL, 61.46 mmol, 1.10 equiv.). Once bubbling has subsided, 2-bromo-l,l-diethoxy ethane (8.46 g, 61.46 mmol, 1.10 equiv.) is added to the mixture and stirred for 2h at room temperature. The resulting suspension is poured onto 20OmL of iced water and extracted with diethyl ether (3 x 50 mL). The organic layers are washed with brine, dried over MgSθ4 to afford after solvent removal the title compound as a viscous orange oil, used without further purification.
Step 2: 7-bromobenzofbJthiophene
Figure imgf000087_0002
[00344] l-Bromo-2-(2-ethoxyvinylsulfanyl)benzene is dissolved in chlorobenzene (50 mL) and the solution is heated to 7O0C. PPA (10.5 mL) is then added carefully and the biphasic mixture is heated at 15O0C overnight. The resulting dark syrup is allowed to cool down and the supernatant solvent removed by pipette. Chlorobenzene (15mL) is added to the residue and heated to 15O0C for 30 min. The solvent is removed again and the residue washed with small amounts of dichloromethane until the washings are clear. The organic fractions are combined and filtered through celite to afford a clear yellow solution. Concentration under vacuum followed by LC using cyclohexane as the eluent provided the title compound as a viscous colourless oil (1.21 g, 35%).
Step 3: BenzofbJthiophene-7-carboxylic acid ethyl ester
Figure imgf000087_0003
[00345] 7-bromobenzo[b]thiophene (500 mg, 2.36 mmol) is dissolved in dry THF (5.0 mL) under nitrogen. Magnesium turnings (530 mg, 2.83 mmol, 1.20 equiv.) are added and the resulting mixture is heated at reflux until dissolution of the magnesium has occurred. The resulting cloudy yellow solution is allowed to cool to rt and diethyl carbonate (2 mL, excess) is added and stirring continued for one hour, when ammonium chloride (10% aq) is added. The resulting mixture is partitioned between DCM and water and the aqueous layer is extracted with DCM. The organic fractions are combined, washed with brine, dried over Na2SC^ and adsorbed onto silica. Purification by LC using 8/2 cyclohexane/dichloromethane as the eluent afforded the title compound as a pale orange oil (346.2 mg, 71%).
Step4: BenzofbJthiophene-7-carboxylic acid
Figure imgf000088_0001
[00346] To a solution of benzo[b]thiophene-7-carboxylic acid ethyl ester (1.0 g, 48 mmol) in methanol (10 mL) and water (10 mL) is added sodium hydroxide (5 g, excess). The solution is stirred at room temperature for 30 min, at which point all the ester has been consumed. The reaction mixture is adjusted to pH 1 by adding 6M HCl solution, and extracted with DCM. The combined organic layers are filtered through a silica plug to afford the title compound as a yellow solid (505.2 mg, 59%).
Step 5: Benzo [b] thiophene-7 -carboxylic acid amide
Figure imgf000088_0002
[00347] A solution of benzo [b]thiophene-7-carboxylic acid (505 mg, 2.84 mmol) in dichloromethane (10 mL) is stirred at rt and thionyl chloride (669 mg, 5.68 mmol, 2.0 equiv.) is added, followed by DMF (0.06 mL), causing gas evolution. The resulting solution is stirred at rt for 1 hour. Aqueous ammonia (10 mL) is then added cautiously to the mixture causing vigorous gas evolution. The resulting mixture is then diluted with water (50 mL), and pH is brought to neutral by adding saturated NaHCθ3 (aq.). The aqueous layer is extracted with DCM, and the organic layers are combined and dried over Na2SOzI. Evaporation of the solvent under reduced pressure affords the title compound (81% pure) as a yellow solid (500 mg, 98%) which is used without further purification.
Step 6: 3 -Bromobenzo[b] thiophene-7 -carboxylic acid amide
Figure imgf000088_0003
[00348] A solution of benzo [b]thiophene-7-carboxylic acid amide (500 mg, 2.82 mmol) in dichloromethane (5 mL) is stirred at rt and acetic acid (5 mL) is added followed by NBS (750 mg, 4.23 mmol, 1.5 equiv.). The reaction is stirred at room temperature for 1 hour at which point LC-MS analysis shows full conversion of the starting material to the desired compound (79%) and dibrominated material (21%). The reaction is diluted with water (50 mL), neutralized with aqueous potassium disulfite, followed by sodium bicarbonate. The aqueous layer is then extracted with dichloromethane and the resulting organic layers are combined, washed with brine, dried over Na2SC^ and evaporated. The crude material is purified by LC using DCM as the eluent to afford the title compound as a white solid (300 mg, 41%).
Step 7: 3-(4,4,5,5-Tetramethyl[l,3,2Jdioxaborolan-2-yl)benzo[bJthiophene-7-carboxylic acid amide
Figure imgf000089_0001
[00349] A solution of 3-bromobenzo[b]thiophene-7-carboxylic acid amide (300 mg, 1.18 mmol) in dioxane (5 mL) is stirred under nitrogen. Pd(dppf)Cl2 (29 mg, 3 mol%), potassium acetate (230 mg, 2.35 mmol, 2.0 equiv.) and bispinacolatodiboron (450 mg, 1.77 mmol, 1.5 equiv.) are added, and the reaction is heated to 8O0C and stirred overnight. The resulting orange suspension is diluted with DCM, filtered through celite and concentrated under vacuum to afford an oil that is used in the next step without further purification.
Step 8: 3-[8-(4-Morpholin-4-yl-phenylamino)-[ 1 ,2,4] triazolo [ 1 ,5-a] pyrazin-5-yl] -benzo [b] thiophene- 7-carboxylic acid amide
Figure imgf000089_0002
[00350] This compound may be prepared using the methods as described for Compound 120,
Step 4. 1H-NMR (400MHz, ds-DMSO) δ(ppm) 3.19 (4H, br s), 3.76 (4H, br s), 5.70 (IH, d), 7.12 (2H, m), 7.43 (IH, t), 7.63 (IH, br s), 7.76-7.80 (2H, m), 7.92 (IH, d), 8.03 (IH, d), 8.21 (IH, s), 8.28 (IH, s), 8.6 (IH, s), 10.09 (IH, s). LCMS: Rt = 1.01 min (95%), m/z (ESI) 472 (M+H)+. Compound 81: 3-{8-[4-(4-Isopropylpiperazin-l-yl)phenylamino]-[l,2,4]triazolo[l,5-a]pyrazin-5- yl}benzo[b]thiophene-7-carboxylic acid amide
Figure imgf000090_0001
[00351] This compound may be prepared using methods as described for Compound 80, using
(5-bromo[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-[4-(4-isopropylpiperazin-l-yl)-phenyl]amine in the final step. LCMS: rt = 0.94 min (95 %), m/z (ESI) 513 (M+H)+.
Compound 83: (4-{8-[4-(4-Isopropylpiperazin-l-yl)-phenylamino]-[l,2,4]triazolo[l,5-a]pyrazin- 5-yl}pyridin-2-yl)methanol
Step 1: 4-Bromo-2-methylpyridine-l -oxide
Figure imgf000090_0002
[00352] A solution of 4-bromo-2-methylpyridine (5 g, 29 mmol) in DCM (20 mL) is cooled to
O0C and m-CPBA (7.55 g, 43.87 mmol, 1.5 equiv.) is added portionwise over 30 min. The ice bath is then removed and the mixture is allowed to stir at room temperature for 3 hours. The resulting solution is diluted with sodium bicarbonate (sat. aq.) and extracted with DCM. The organic layers are combined, washed with sodium bicarbonate (sat. aq.), dried over Na2SOzI, and concentrated under vacuum to afford the title compound as a pale orange oil which is without further purification.
Step 2: Acetic acid 4-bromopyridin-2-ylmethyl ester
Figure imgf000090_0003
[00353] To a solution of 4-bromo-2-methylpyridine-l -oxide (4.00 g, 21.40 mmol) in DCM (20 mL) is added acetic anhydride (6 mL). The mixture is stirred at room temperature for 1 hour and then heated at reflux overnight. The solvent is removed under vacuum and the crude product is filtered through a silica plug, eluting with DCM to afford the title compound as an orange oil (815 mg).
Step 3: Acetic acid 4-(4,4,5,5-tetramethyl-[l,3,2Jdioxaborolan-2-yl)pyridin-2-ylmethyl ester
Figure imgf000091_0001
[00354] A solution of acetic acid 4-bromopyridin-2-ylmethyl ester (800 mg, 3.49 mmol) in dioxane (5 mL) is stirred under nitrogen. Pd(dppf)Cl2 (85 mg, 3 mol%), potassium acetate (1.03 g, 10.5 mmol, 3.0 equiv.) and bispinacolatodiboron (1.33 g, 5.24 mmol, 1.5 equiv.) are added, and the reaction is heated at 8O0C overnight. The resulting orange suspension is diluted with DCM, filtered through celite and concentrated under vacuum to afford an oil that is used in the next step without further purification.
Step 4: Acetic acid 4-{8-[4-(4-isopropyl-piperazin-l-yl)-phenylamino]-[l,2,4]triazolo[l,5-a]pyrazin- 5-yl}-pyridin-2-ylmethyl ester
Figure imgf000091_0002
[00355] A suspension of (5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-[4-(4-isopropyl- piperazin-l-yl)phenyl]amine (300 mg, 0.723 mmol) and Pd(dppf)Cl2 (59 mg, 10 mol%) in 4/1 dioxane/water (5 mL) is stirred at rt and potassium carbonate (200 mg, 1.45 mmol, 2.0 equiv.) and acetic acid 4-(4,4,5,5-tetramethyl-[l,3,2]dioxaborolan-2-yl)pyridin-2-ylmethyl ester (300 mg, 1.08 mmol, 1.5 equiv.) are added. The resulting mixture is heated at 850C overnight. The resulting solution is partitioned between dichloromethane and water, and the organic layer is concentrated onto silica and purified by column chromatography (98/2 DCM/MeOH) to afford the title compound as a yellow solid. LCMS: Rt = 0.78min (100%), m/z 445 (M+H)+.
Step 5: (4-{8-[4-(4-Isopropylpiperazin-l-yl)phenylamino]-[l,2,4]triazolo[l,5-a]pyrazin-5-yl}pyridin- 2-yl)methanol
Figure imgf000092_0001
[00356] Acetic acid 4-{8-[4-(4-isopropylpiperazin-l-yl)phenylamino]-[l,2,4]triazolo[l,5- a]pyrazin-5-yl}pyridin-2-ylmethyl ester is stirred at room temperature overnight in 10 mL of a 1.5 M solution of potassium carbonate in methanol. The pH is then brought to neutral by addition of 10% aqueous citric acid and the mixture is extracted with DCM. Purification by LC using 98/2 DCM/2M NH3 in MeOH as the eluent provides the title compound as a yellow powder (40.3 mg, 12.5% over 2 steps). 1H-NMR (400MHz, d6-DMSO) δ(ppm) 1.02 (6H, m), 2.33 (IH, m), 2.60 (4H, br m), 3.10 (4H, br m), 4.65 (2H, d), 5.51 (IH, t), 6.96 (2H, d), 7.84-7.89 (3H, m), 8.11 (IH, s), 8.17 (IH, m), 8.61 (IH, d), 8.73 (IH, m), 10.08 (IH, s). LCMS: Rt = 0.78 min (100%), m/z (ESI) 445 (M+H)+.
Compound 84: [4-(l-Isopropylpiperidin-4-yl)phenyl]-[5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5- a] pyrazin-8-yl] amine
Figure imgf000092_0002
[00357] This compound may be prepared using the methods as described for Compound 92, using pyrazole-4-boronic acid in step 4. LCMS: Rt 0.89 min (100 %) m/z (ESI) 403 (M+H)+.
Compound 86: 4-(8-(6-(4-Isopropylpiperazin-l-yl)pyridin-3-ylamino)-[l,2,4]triazolo[l,5- a]pyrazin-5-yl)furan-2-carboxamide
Step 1: 4-Bromo-furan-2-carboxylic acid amide
Figure imgf000093_0001
[00358] To a solution of 4,5-dibromo-furan-2-carboxylic acid {1.19%, 28.85mmol) in NH4OH
(10OmL) is added zinc dust (2.29g, 34.62mmol) in small portions. The reaction mixture is stirred at room temperature for 7 minutes then filtered over celite and washed with water and 2M HCl. The filtrate is acidified to pH 1 using cone. HCl and extracted with ethyl acetate (3x). The organic phase is washed with brine, dried over MgSO4, filtrated and concentrated in vacuo to give an oil (4.96g) which solidifies on standing to give a white solid, which is used without further purification. [00359] The solid (4.93g, 25.81mmol) is dissolved in thionyl chloride (44.2mL) and refluxed for 1 hour. After removing the solvent in vacuo the residue is dissolved in dichloromethane (75mL) and a solution of 0.5 M NH3 in dioxane (52mL) is added. The reaction mixture is stirred at room temperature for 1 hour, then 33% aq. NH3 (5mL) is added and the reaction stirred for additional 2 hours. The solvent is removed in vacuo and the residue taken-up with a solution of sat. NaHCO3. The basic solution is extracted using ethyl acetate (3x), the combined organic layers are dried over MgSO4 and concentrated in vacuo. Purification by silica gel column chromatography eluting with a mixture of (50:49:1) ethyl acetate:petroleum etheπacetic acid, affords the title compound (1.2g, 22%).
Step 2: 4-(4,4,5 ,5-Tetramethyl-[ 1 ,3 ,2] dioxaborolan-2-yl)-furan-2-carboxylic acid amide
Figure imgf000093_0002
[00360] 4-Bromo-furan-2-carboxylic acid amide (1.2g, 6.32mmol), bis(pinacolato)diboron
(1.76g, 6.94mmol), PdCl2dppf (0.154g, 189mol) and KOAc (1.85g, 18.94mmol) are suspended in dioxane (2OmL), purged with nitrogen for 5 minutes and then heated at 9O0C overnight. The solvent is removed in vacuo and the residue partitioned between ethyl acetate and water. The aqueous layer is extracted three times with ethyl acetate and the combined organic phases are washed with brine, filtered through MgSO4 and evaporated. The solid residue is triturated with hexane and dried in vacuo to afford the title compound as a solid (0.984g, 66%). Step 3: 4-(8-(6-(4-Isopropylpiperazin-l-yl)pyridin-3-ylamino)-[l,2,4]triazolo[l,5-a]pyrazin-5- yl)furan-2-carboxamide
Figure imgf000094_0001
[00361] This compound may be prepared using methods as described for Compound 6, step 4 using 5-bromo-8-(6-(4-isopropylpiperazin-l-yl)pyridin-3-yl)-[l,2,4]triazolo[l,5-a]pyrazin-8-amine (60mg, 0.144mmol), 4-(4,4,5,5-tetramethyl-[l,3,2]dioxaborolan-2-yl)-furan-2-carboxylic acid amide (59mg, 0.26mmol), and Pd(PPh3)4 (42mg, 0.036mmol) in 1.5M Na2CO3 (0.8mL, 1.15mmol) and dioxane (2mL). After evaporation of the solvent the crude material is purified by silica gel column chromatography using DCM followed by 97.5:2.5 and 95:5 DCM:NH3 (7M in MeOH). The title compound is obtained as a yellow solid (30mg, 47%). 1H-NMR (400MHz, de-DMSO) δ(ppm) 1.05 (6H, d), 2.54-2.59 (4H, m), 2.70-2.74 (IH, m), 3.47 (4H, m), 6.90 (IH, d), 7.60 (IH, br s), 7.91 (IH, s), 7.99 (IH, br s), 8.12 (IH, dd), 8.19 (IH, s), 8.68 (IH, s), 8.75 (IH, s), 8.81 (IH, s), 10.03 (IH, s). LCMS: Rt 2.64min (98.1%), m/z (APCI) 406 (M+H)+. LCMS: Rt 2.74min (93%), m/z (ES+) 448 (M+H)+.
Compound 88: 4-(8-(4-(4-Isopropylpiperazin-l-yl)phenylamino)-[l,2,4]triazolo[l,5-a]pyrazin-5- yl)furan-2-carboxamide
Step 1: l-Isopropyl-4-(4-nitro-phenyl)-piperazine
Figure imgf000094_0002
[00362] To a solution of 4-fluoronitrobenzene (5g, 35.4mmol) in THF (5OmL), 1- isopropylpiperazine (4.54g, 35.4mmol) and K2CO3 (7.35g, 53.2mmol) are added. The reaction mixture is stirred at room temperature overnight. The solvent is removed in vacuo and the residue is partitioned between EtOAc and water. The organic layer is washed with brine, dried over MgSO4, filtered and concentrated. The crude compound is purified by silica gel column chromatography using 99:1 and 98:2 DCM:NH3 (7M in MeOH) to give the title compound (8.2g, 94%).
Step 2: 4-(4-Isopropyl-piperazin-l-yl)-phenylamine NH,
[00363] l-Isopropyl-4-(4-nitro-phenyl)-piperazine (8.3g, 33.2mmol) is dissolved in MeOH
(120 mL) and tin (II) dichloride dihydrate (37.4g, 0.165mol) is added. The mixture is cooled using a water bath and cone. HCl is added (36mL). The reaction is stirred at room temperature overnight. After removing the methanol, the resultant solution is basified using cone. NaOH (pH 11). The water phase is extracted with diethyl ether (3x) and the organic layers combined, dried over MgSOzi, filtered, concentrated in vacuo to afford the title compound (6.4g, 88%).
Step 3 (5-Bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-[4-(4-isopropyl-piperazin-l-yl)-phenyl]-amine
Figure imgf000095_0001
[00364] This compound may be prepared using methods as described for Compound 6, step 1, using 5,8-dibromo-[l,2,4]triazolo[l,5-a]pyrazine (2g, 7.20mmol), 4-(4-isopropyl-piperazin-l-yl)- phenylamine (1.89g, 8.62mmol) and N,N-diisopropylethylamine (1.88mL, 10.8mmol) in 2-propanol (3OmL) are stirred at 950C overnight. The title compound is isolated after trituration with diethyl ether and petroleum ether as a grey solid (2.59g,
Step 4: 4-(8-(4-(4-Isopropylpiperazin-l-yl)phenylamino)-[l,2,4]triazolo[l,5-a]pyrazin-5-yl)furan-2- carboxamide
Figure imgf000095_0002
[00365] This compound may be prepared using methods as described for Compound 6, step 4, using 5-bromo-N-(6-(4-isopropylpiperazin-l-yl)pyridin-3-yl)-[l,2,4]triazolo[l,5-a]pyrazin-8-amine (80mg, 0.21mmol), 4-(4,4,5,5-tetramethyl-[l,3,2]dioxaborolan-2-yl)-furan-2-carboxylic acid amide (lOlmg, 0.42mmol), and Pd(PPh3)4 (62mg, 0.053mmol) in 1.5M Na2CO3 (aq) (1.143mL, 1.71mmol), and dioxane (4mL). The crude product is purified by silica gel column chromatography eluting with DCM and 97:3 DCM:NH3 (7M in MeOH). The title compound is isolated after trituration with diethyl ether (35.4mg, 42%). Conversion into the mesylate salt using IM methanesulfonic acid in MeOH (0.0793mL) affords the title compound as a solid (35mg).
H-NMR (400MHz, ds-DMSO) δ(ppm) 1.35 (6H, d), 2.35 (3H, s, MsOH), 3.01 (2H, t), 3.22-3.35 (2H, m), 3.58 (3H, m), 3.88 (2H, d), 7.09 (2H, d), 7.61 (IH, br s), 7.94 (2H, m), 8.00 (IH, br s), 8.22 (IH, s), 8.76 (IH, s), 8.82 (IH, s), 9.27 (IH, br s), 10.02 (IH, s). LCMS: Rt 2.02min (98.9%), m/z (APCI) 447 (M+H)+.
Compound 89: 4-{8-[4-(l-Isopropylpiperidin-4-yl)phenylamino]-[l,2,4]triazolo[l,5-a]pyrazin-5- yl}-thiophene-2-carboxylic acid amide
Figure imgf000096_0001
[00366] This compound may be prepared using the methods as described for Compound 92, using 2-(aminocarbonyl)thiophene-4-boronic acid in step 4. LCMS: Rt 0.94 min (100 %) m/z (ESI) 462 (M+H)+.
Compound 90: (4-{8-[6-(4-Isopropylpiperazin-l-yl)pyridin-3-ylamino]-[l,2,4]triazolo[l,5- a]pyrazin-5-yl}pyridin-2-yl)methanol
Figure imgf000097_0001
[00367] This compound may be prepared using the methods as described for Compound 83, using (5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-[6-(4-isopropylpiperazin-l-yl)pyridin-3-yl]amine in Step 4. LCMS: Rt = 0.71 min (95 %), m/z (ESI) 446 (M+H)+.
Compound 92: 5-{8-[4-(l-Isopropylpiperidin-4-yl)phenylamino]-[l,2,4]triazolo[l,5-a]pyrazin-5- yl}-2,3-dihydroisoindol-l-one
Step 1: l-Isopropyl-4-(4-nitrophenyl)piperidine
Figure imgf000097_0002
[00368] 4-(4-Nitrophenyl)piperidine (250 mg, 1.21 mmol), K2CO3 (170 mg, 1.21 mmol) and
2-iodopropane (240 μL, 2.4 mmol) are stirred in acetonitrile (3 mL) in a sealed tube at 120°C for 45 min. The mixture is cooled and the solvent removed under reduced pressure. The residue is partitioned between DCM (20 mL) and water (5 mL), the layers are separated and the DCM is washed with water (5 mL) and brine (5 mL) and dried over MgSO4. Evaporation of the solvent affords the title compound (300 mg) which is used without further purification.
Step 2: 4-(l-Isopropylpiperidin-4-yl)phenylamine
Figure imgf000097_0003
[00369] Hydrazine (35% by weight in water, 0.67 mL, 7.2 mmol) and 10% Pd/C (38 mg, 0.03 mmol) are added to a solution of l-isopropyl-4-(4-nitrophenyl)piperidine (180 mg, 0.72 mmol) in EtOH (10 mL) and the mixture is heated at reflux for 3 h. After cooling, the mixture is filtered through celite and the solvent evaporated. The residue is redissolved in DCM (25 mL), dried over MgSθ4 and the solvent evaporated to afford the desired compound as a pale yellow solid (113 mg, 0.52 mmol) which was used without further purification.
Step 3: (5-Bromo-[ 1 ,2,4] triazolo [ 1 ,5-a] pyrazin-8-yl)-[4-(l-isopropylpiperidin-4-yl)phenyl] amine
Figure imgf000098_0001
[00370] N,N-Diisopropylethylamine (200 μL, 1.2 mmol) is added to a mixture of 4-(l- isopropylpiperidin-4-yl)phenylamine (220 mg, 1.0 mmol) and 5,8-dibromo-[l,2,4]triazolo[l,5- a]pyrazine (280 mg, 1.0 mmol) in 1PrOH (5 mL) and heated at reflux for 48 h. The mixture is cooled and the solvent evaporated under reduced pressure to afford an orange-brown solid. This is partitioned between DCM (50 mL) and water (20 mL) and the layers are separated. The organic phase is washed with citric acid (10% aq., 3 x 25 mL). The combined washings are extracted with DCM (25 mL) and then made basic by addition of NaHCθ3 (s). The mixture is extracted with DCM (3 x 25 mL) and the combined extracts dried over MgSθ4 and evaporated. The crude product was purified by chromatography on silica gel, eluting with 5% - 10% MeOH in DCM to afford the desired compound, contaminated with the starting aniline. This was recrystallised from MeOH to afford the pure title compound (110 mg).
[00371] This material may be used to prepare 5-{8-[4-(l-isopropylpiperidin-4- yl)phenylamino]-[l,2,4]triazolo[l,5-a]pyrazin-5-yl}-2,3-dihydroisoindol-l-one in a fashion analogous to step 4 as described for Compound 79.
Figure imgf000098_0002
LCMS: Rt 0.91 min (100 %) m/z (ESI) 468 (M+H)+ Compound 100: l-(4-(5-(lH-Pyrazol-4-yl)-[l,2,4]triazolo[l,5-a]pyrazin-8- ylamino)phenyl)piperazin-2-one
Step 1: 4-(4-Amino-phenyl)-3-oxo-piperazine-l-carboxylic acid tert-butyl ester
Figure imgf000099_0001
[00372] A suspension of />-iodo-aniline (918mg, 4.8mmol), 3-oxo-piperazine-l-carboxylic acid tert-butyl ester (960mg, 4.2mmol) (7R,2R)-cyclohexane-l,2-diamine (0.05mL, 0.42mmol), copper (I) iodide (14.9mg,0.0042mmol) and K2CO3 (1.19g, 2.04mmol) in dioxane (4mL), is purged with nitrogen for 5 min in a reaction tube. The tube is sealed and the reaction mixture is heated at 1190C for 15 hours. After cooling to room temperature, the reaction mixture is filtered through a silica cartridge washing with ethyl acetate (4OmL). The filtrate is concentrated in vacuo to afford the title compound as a brown liquid (1.06g, 87%). LCMS: Rt 0.88min (91%).
Step 2: l-[4-(5-Bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-ylamino)-phenyl]-piperazin-2-one
Figure imgf000099_0002
[00373] This compound may be prepared using methods as described for Compound 6, step 1, using 5,8-dibromo-[l,2,4]triazolo[l,5-a]pyrazine (0.283g, l.OOmmol), 4-(4-amino-phenyl)-3-oxo- piperazine-1-carboxylic acid tert-butyl ester (0.30Og, l.OOmmol) and N-ethyldiisopropyl-amine (0.2OmL, 1.02mmol) in 2-propanol (ImL). Purification of the crude material by silica gel column chromatography eluting with DCM followed by 98:2 DCM:MeOH affords 4-[4-(5-bromo- [l,2,4]triazolo[l,5-a]pyrazin-8-ylamino)-phenyl]-3-oxo-piperazine-l-carboxylic acid tert-butyl ester as a white solid (0.252g, 51%).
[00374] A solution of 4-[4-(5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-ylamino)-phenyl]-3-oxo- piperazine-1-carboxylic acid tert-butyl ester (0.252mg, O.όmmol) in 2:1 DCM:TFA (4.8mL) is stirred for 1 hour at room temperature. The mixture is then diluted with DCM and basified with sat. NaHCC"3. The aqueous layer is extracted with DCM (3x) and the organic layers are combined, dried over MgSOzi, filtered and concentrated to afford the title compound as a solid (180mg, 78%).
Step 3: l-(4-(5-(lH-Pyrazol-4-yl)-[l,2,4]triazolo[l,5-a]pyrazin-8-ylamino)phenyl)piperazin-2-one
Figure imgf000100_0001
[00375] This compound may be prepared using methods as described for Compound 1, step 5 using 1 -[4-(5-bromo-[l ,2,4]triazolo[l ,5-a]pyrazin-8-ylamino)-phenyl]-piperazin-2-one (70mg, 0.18mmol) 4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-lH-pyrazole (64mg, 0.33mmol), Pd(PPh3)4 (21mg, lδμmol) and NaO'Bu (70mg, 0.72mmol) in 2mL of 3:1 DMF/water. The reaction mixture is concentrated under vacuum and the residue is purified by silica gel column chromatography eluting with a gradient 99:1 to 90:10 DCM:NH3 (7M in MeOH). The title compound is isolated as a pale green solid (13mg, 19%). 1H-NMR (400MHz, ds-DMSO) δ(ppm) 3.05 (2H, m), 3.42 (2H, s), 3.62 (2H, m), 7.32 (2H, d), 8.07 (2H, d), 8.26 (IH, s), 8.39 (IH, br s), 8.67 (IH, br s), 8.81 (IH, s), 10.05 (IH, s), 13.32 (IH, br s). LCMS: Rt 6.98min (92.4%), m/z (APCI) 376 (M+H)+.
Compound 102: 5-{8-[4-(4-tert-Butyl-piperazin-l-yl)-phenylamino]-[l,2,4]triazolo[l,5- a]pyrazin-5-yl}-2,3-dihydro-isoindol-l-one
Step 1: l-tert-Butyl-4-(4-nitro-phenyl)-piperazine
Figure imgf000100_0002
[00376] To a solution of l-fluoro-4-nitro-benzene (314mg, 2.23mmol) and 4-tert-butyl- piperazine (Ig, 3.34mmol) in dioxane (15mL) is added K2CO3 (1.65mg, 11.9mmol) and the reaction is stirred at 13O0C overnight. The solvent is evaporated under vacuum and the residue is partitioned between ethyl acetate and water. The organic layer is washed with brine, dried over MgSO4, filtered and concentrated to give a crude compound. Purification by silica gel column chromatography eluting with DCM followed by 99:1 DCM:NH3 (7M in MeOH) affords the title compound (348mg, 55.4%). LCMS: Rt 3.87min (99%).
Step 2: 4-(4-tert-Butyl-piperazin-l-yl)-phenylamine
Figure imgf000100_0003
[00377] l-tert-Butyl-4-(4-mtro-phenyl)-piperazine (348mg, 1.32mmol) is dissolved in MeOH
(10 mL) and tin (II) dichloride dihydrate (1.08g, 4.79mmol) is added. The mixture is cooled using a water bath and cone. HCl is added (3mL). The reaction is stirred at 4O0C overnight. After removing the methanol, the resultant solution is basified using cone. NaOH (pH 11). The water phase is extracted with diethyl ether (3x) and the organic layers combined, dried over MgSOzi, filtered, concentrated in vacuo to afford the title compound (308mg, 99%) which is used in the next step without further purification. LCMS: Rt 2.16min (87%)
Step 3: (5-Bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-[4-(4-tert-butyl-piperazin-l-yl)-phenyl]-amine
Figure imgf000101_0001
[00378] This compound may be prepared using methods as described for Compound 6, step 1, using 5,8-dibromo-[l,2,4]triazolo[l,5-a]pyrazine (0.308g, 1.107mmol), 4-(4-tert-butyl-piperazin-l- yl)-phenylamine (0.31Og, 1.33mmol) and N-ethyldiisopropyl-amine (0.289mL, 1.66mmol) in 2- propanol (5mL). The reaction mixture is partitioned between DCM and IN NaOH, the organic layer is separated and washed with water and brine, dried over MgSOzi, filtered and concentrated under vacuum. Trituration of the residue with diethyl ether and petroleum ether affords the title compound (440mg, 92%) as a cream solid. LCMS: Rt 2.25min (96%).
Step 4: 5-{8-[4-(4-tert-Butyl-piperazin-l-yl)-phenylamino]-[ 1,2,4] triazolo[l,5-a]pyrazin-5-yl}-2,3- dihydro-isoindol-1-one
Figure imgf000101_0002
[00379] This compound may be prepared using methods as described for Compound 6, step 4,
(5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-[4-(4-tert-butyl-piperazin-l-yl)-phenyl]-amine (lOOmg, 0.232mmol), 5-(4,4,5,5-tetramethyl-[l,3,2]dioxaborolan-2-yl)-2,3-dihydro-isoindol-l-one (90mg, 0.32mmol), and Pd(PPh3)4 (67mg, 0.058mmol) in 1.5M Na2CO3 (aq) (1.24mL), and dioxane (3.7mL). The reaction mixture is partitioned between ethyl acetate and brine, the organic layer is separated, dried over MgSO4 filtered and evaporated in vacuo. The residue is purified by silica gel column chromatography eluting with 98:2 DCM:NH3 (7M in MeOH) followed by 95:5 DCM:NH3 (7M in MeOH) to give a solid which is triturated with diethyl ether and petroleum ether to afford the title compound (71mg, 63%). 1H-NMR (400MHz, dg-DMSO) δ(ppm) 1.43 (9H, s), 2.36 (3H, s, MsOH), 3.02 (2H, m), 3.19-3.26 (2H, m), 3.67 (2H, d), 3.90 (2H, d), 4.52 (2H, s), 7.10 (2H, d), 7.86 (IH, d), 7.96-8.01 (3H, m), 8.10 (IH, d), 8.24 (IH, s), 8.72 (IH, s), 8.75 (IH, s), 9.09 (IH, br s), 10.09 (IH, s). LCMS: Rt 2.07min (99%), m/z (APCI) 483 (M+H)+.
Compound 105: 5-{8-[4-(2-Oxo-piperazin-l-yl)-phenylamino]-[l,2,4]triazolo[l,5-a]pyrazin-5- yl}-2,3-dihydro-isoindol-l-one
Step 1: 3-Oxo-4-{4-[5-(l-oxo-2,3-dihydro-lH-isoindol-5-yl)-[l,2,4]triazolo[l,5-a]pyrazin-8- ylamino] -phenyl} -piperazine-1 -carboxy Hc acid tert-butyl ester
Figure imgf000102_0001
[00380] This compound may be prepared using methods as described for Compound 6, step 4, using 4-[4-(5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-ylamino)-phenyl]-3-oxo-piperazine-l-carboxylic acid tert-butyl ester (115mg, 0.24mmol), 5-(4,4,5,5-tetramethyl-[l,3,2]dioxaborolan-2-yl)-2,3- dihydro-isoindol-1-one (92mg, 0.35mmol), and Pd(PPh3)4 (83mg, 0.072mmol) in 1.5M Na2CO3 (aq) (1.3mL, 1.92mmol), and dioxane (2.5mL). The reaction mixture is diluted with brine then toluene is added and a precipitate is formed and collected by filtration. The resultant solid is dissolved in DCM and filtered through a silica cartridge to afford the title compound as a yellow solid (60mg, 47%).
Step 2: 5-{8-[4-(2-Oxo-piperazin-l-yl)-phenylaminoJ-[l,2,4Jtriazolo[l,5-aJpyrazin-5-yl}-2,3-dihydro- isoindol-1-one
Figure imgf000102_0002
[00381] A solution of 3-oxo-4-{4-[5-(l-oxo-2,3-dihydro-lH-isoindol-5-yl)-[l,2,4]triazolo[l,5- a]pyrazin-8-ylamino]-phenyl}-piperazine-l-carboxylic acid tert-butyl ester (59mg, O.lmmol) in 3:1 DCM:TFA (1.2mL) is stirred at room temperature for 1 hour. The reaction mixture is diluted with DCM and basified with sat. NaHCθ3. The aqueous layer is extracted with DCM (3x) and the organic layers are combined, dried over MgSOzi, filtered and concentrated under vacuum to afford the title compound as a yellow solid (38mg, 86%). 1H-NMR (400MHz, de-DMSO) δ(ppm) 3.07 (2H, m), 3.44 (2H, s), 3.64 (2H, m), 4.50 (2H, s), 7.32 (2H, d), 7.83 (IH, d), 8.04-8.10 (4H, m), 8.22 (IH, s), 8.70 (IH, br s), 8.74 (IH, s), 10.25 (IH, s). LCMS: Rt 7.17min (93.7%), m/z (APCI) 441 (M+H)+.
Compound 108: 7-Fluoro-5-{8-[4-(4-isopropylpiperazin-l-yl)-phenylamino]-[l,2,4]triazolo[l,5- a]pyrazin-5-yl}-2,3-dihydroisoindol-l-one
Step 1: 4-Bromo-2,6-difluorobenzoic acid methyl ester
Figure imgf000103_0001
[00382] To a suspension of 4-Bromo-2,6-difluoro-benzoic acid (5 g, 21 mmol) in DCM
(1OmL) is added thionyl chloride (15 mL) and DMF (0.5mL). The mixture is stirred at room temperature for 2.5 h. It is then cooled to O0C and MeOH (2OmL) is added carefully causing vigorous HCl evolution. After stirring for an additional 0.5 h, the clear solution is partitioned between DCM (50 mL) and water (50 mL). The organic layer is washed with saturated NaHCθ3, brine, dried over MgSO4 and the solvent is removed under vacuum to afford the title compound as a pale yellow oil which is used without further purification.
Step 2: 4-Bromo-2-fluoro-6-(nitromethyl)benzoic acid methyl ester
Figure imgf000103_0002
[00383] Nitromethane (10 mL, 169 mmol, 8 equiv.) is added cautiously to a suspension of sodium hydride (4.05 g, 169 mmol, 8 equiv.) and MgSO4 (40 g) in DMSO (100 mL) at rt and the resulting slurry is stirred for 0.25 h. To the resulting yellow slurry is added 4-bromo-2,6- difluorobenzoic acid methyl ester (5.3 g, 21 mmol) and the mixture is stirred at rt for 3 days at which point all the starting material has been consumed. Water (200 mL) and 6M HCl (50 mL) is added followed by DCM (200 mL). More water (500 mL) is added to yield a clear biphasic system. The aqueous layer is extracted with DCM (3 x 100 mL), the DCM layers are then combined, washed with saturated NaHCθ3 and brine, and dried over MgSO4. Evaporation of the solvent affords an orange solid containing 64% of the desired material, used without further purification. LCMS; Rt = 1.27 min
(64%). Step 3: 5-Bromo-7-fluoro-2,3-dihydro-isoindol-l-one
Figure imgf000104_0001
[00384] Crude 4-bromo-2-fluoro-6-(nitromethyl)benzoic acid methyl ester from the previous step is dissolved in MeOH (100 mL). To this clear orange solution is added Zinc dust (3.35 g, 51.3 mmol, 3 equiv.) followed by ammonium formate (3.23 g, 51.3 mmol, 3 equiv.) which results in an exothermic reaction. After 0.3 h, 7M NH3 in MeOH (50 mL) is added and the mixture is stirred overnight at room temperature. The resulting mixture is filtered through celite, and the yellow filtrate is adsorbed on silica and roughly cleaned by LC using 94/6 DCM/ MeOH 7M NH3. Removal of the solvent affords a tan solid that is redissolved in DCM, washed with 10% NaOH, and concentrated under reduced pressure to leave a white solid, which is triturated further with small amounts of DCM to afford the title compound. LCMS: Rt 0.96 min (100%) m/z 230/232 (M+H)+.
Step 4: 7-Fluoro-5-(4, 4, 5, 5-tetramethyl-fl, 3, 2] dioxaborolan-2-yl)-2, 3-dihydroisoindol-l-one
Figure imgf000104_0002
[00385] A solution of 5-bromo-7-fluoro-2,3-dihydroisoindol-l-one (531 mg, 2.31 mmol) in dioxane (5 mL) is stirred under nitrogen. Pd(dppf)Cl2 (94 mg, 5 mol%), potassium acetate (453 mg, 4.62 mmol, 2.0 equiv.) and bispinacolatodiboron (1.17 g, 4.62 mmol, 2 equiv.) are added, and the reaction is heated at 8O0C for 3 h. The resulting orange suspension is diluted with DCM, filtered through celite and concentrated under vacuum to afford an oil that is redissolved in a minimum of DCM. Diethyl ether is slowly added to afford the title compound as a tan solid. LCMS: Rt 1.18 min (100%) m/z 277/279 (M+H)+.
Step 5: 7-Fluoro-5-{8-[4-(4-isopropylpiperazin-l-yl)-phenylamino]-[ 1,2,4] triazolo[l,5-a] pyrazin-5- yl} -2, 3-dihydroisoindol-l-one
Figure imgf000105_0001
[00386] This compound may be prepared using methods as described for Compound 91 using the above boronate. LCMS: Rt = 0.90 min (95 %), m/z 487 (M+H)+.
Compound 112: 5-(8-(4-(4-Isopropyl-2-oxopiperazin-l-yl)phenylamino)-[l,2,4]triazolo[l,5- a]pyrazin-5-yl)isoindolin-l-one
Step 1: l-[4-(5-Bromo-[ 1 ,2 ,4j 'triazolo [ 1 ,5-aJ pyrazin-8-ylamino) -phenyl] ' -4-isopropyl-piperazin-2-one
Figure imgf000105_0002
[00387] To a solution of l-[4-(5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-ylamino)-phenyl]- piperazin-2-one (example 31, step 2) (180mg, 0.47mmol) in MeOH (4mL) are added acetic acid (0.03mL, 0.47mmol), NaOAc (38mg, 0.47mmol) and acetone (0.2mL, l.lδmmol). The reaction mixture is stirred at room temperature for 1 hour then NaCNBH3 (60mg, 0.94mmol) is added and the mixture is stirred at 4O0C overnight. After cooling to room temperature, the reaction mixture is acidified with cone. HCl (pH 1) and concentrated in vacuo. The residue is partitioned between 6N NaOH and DCM. The aqueous layer is extracted with DCM (3x) and the combined organic layers are dried over MgSθ4 and concentrated in vacuo to afford a crude compound which is purified by silica gel column chromatography. Elution with DCM and 98:2 DCM:MeOH affords the title compound as a yellow solid (45mg, 25%).
Step 2: 5-(8-(4-(4-Isopropyl-2-oxopiperazin-l-yl)phenylamino)-[ 1,2,4] 'triazolo [1,5-aJ pyrazin-5- yl)isoindolin- 1 -one
Figure imgf000106_0001
[00388] This compound may be prepared using methods as described for Compound 6, step 4, using l-[4-(5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-ylamino)-phenyl]-4-isopropyl-piperazin-2-one (40mg, 0.09mmol), 5-(4,4,5,5-tetramethyl-[l,3,2]dioxaborolan-2-yl)-2,3-dihydro-isoindol-l-one (37mg, 0.14mmol), and Pd(PPh3)4 (31mg, 0.027mmol) in 1.5M Na2CO3 (aq) (0.5mL, 0.75mmol), and dioxane (1.2mL). The reaction mixture is diluted with brine then toluene is added and a precipitate is formed and collected by filtration. The filtrate is washed with diethyl ether, petroleum ether and MeOH and then purified by silica gel column chromatography. Elution with 99:1 DCM:NH3 (7M in MeOH) followed by 98:2 and 95:5 DCM:NH3 (7M in MeOH) affords the title compound as a yellow solid (24mg, 55%).
1H-NMR (400MHz, d6-DMSO) δ(ppm) 1.08 (6H, d), 2.73-2.87 (3H, m), 3.27 (2H, s), 3.62-3.68 (2H, m), 4.52 (2H, s), 7.36 (2H, d), 7.84 (IH, d), 8.07-8.10 (4H, m), 8.25 (IH, s), 8.70 (IH, br s), 8.77 (IH, s), 10.28 (IH, s). LCMS: Rt 1.94min (95.2%), m/z (APCI) 483 (M+H)+.
Compound 114: 5-{8-[4-(4-Isopropylpiperazin-l-yl)-phenylamino]-[l,2,4]triazolo[l,5-a]pyrazin- 5-yl}-3,3-dimethyl-2,3-dihydroisoindol-l-one
Step 1 : 5-Bromo-2-(4-methoxybenzyl)-3 , 3 -dimethyl- 2, 3-dihydroisoindol-l-one
Figure imgf000106_0002
[00389] A suspension of sodium hydride (130 mg, 60% dispersion in mineral oil, 3.2 mmol) and tetra-nbutylammonium iodide (243 mg, 0.68 mmol) in THF (20 mL) is stirred at rt and a solution of 5-bromo-2,3-dihydroisoindol-l-one (675 mg, 3.2 mmol) in THF (20 mL) and DMF (4 mL) is added. After 75 min 4-methoxybenzyl bromide (460 μL, 3.2 mmol) is added and stirring is continued for 4 h. Sodium hydride (635 mg, 60% dispersion in mineral oil, 15.9 mmol) is then added and stirring is continued for 30 min before iodomethane (1.19 mL, 19 mmol) is added and the mixture heated to 70°C for 30 min. After cooling, NH4Cl (sat. aq.) is added and the mixture is diluted with ethyl acetate (120 mL). The layers are separated, the organic phase is dried over MgSO4 and the solvents removed under reduced pressure. The residue is purified by silica chromatography, eluting with 5% to 10% ethyl acetate in petroleum ether to afford the title compound as a yellow oil (640 mg, 1.78 mmol).
Step 2: 5-Bromo-3,3-dimethyl-2,3-dihydroisoindol-l-one
Figure imgf000107_0001
[00390] A solution of 5-bromo-2-(4-methoxybenzyl)-3,3-dimethyl-2,3-dihydroisoindol-l-one
(640 mg, 1.78 mmol) and eerie ammonium nitrate (2.91 g, 5.33 mmol) in acetonitrile (11 mL) and water (5 mL) is stirred at 0°C for 45 min. The solution is diluted with ethyl acetate (100 mL) and washed with brine (40 mL). The organic solvents are dried over MgSθ4 and evaporated under reduced pressure. The residue is purified by silica chromatography, eluting with 10% to 50% ethyl acetate in petroleum ether to afford the title compound (367 mg, 1.53 mmol).
Step 3: 5-{8-[4-(4-Isopropylpiperazin-l-yl)-phenylamino] -[ 1 ,2 ,4] triazolo [ 1 ,5-a] pyrazin-5-yl}-3 ,3- dimethyl-2,3-dihydroisoindol-l-one
Figure imgf000107_0002
[00391] 5-Bromo-3,3-dimethyl-2,3-dihydroisoindol-l-one is converted to the corresponding boronate in a fashion analogous to Compound 6, Step 3. This is then used to prepare the title compound from (5-bromo[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-[4-(4-isopropylpiperazin-l-yl)- phenyl]amine using methods as described for Compound 120, Step 4. LCMS: Rt = 0.92 (100%), m/z = 497 (M+H)+.
Compound 118: 4-(5-(lH-Pyrazol-4-yl)-[l,2,4]triazolo[l,5-a]pyrazin-8-ylamino)-N-(pyridin-3- ylmethyl)benzamide
Step 1: 4-(5-Bromo-[ 1 ,2,4] ' triazolo [ 1 ,5-a] pyrazin-8-ylamino) -benzoic acid methyl ester
Figure imgf000108_0001
[00392] This compound may be prepared using methods as described for Compound 6, step 1 using 5,8-dibromo-[l,2,4]triazolo[l,5-a]pyrazine (250mg, 0.90mmol), 4-aminobenzoic methyl ester (163mg, 1.08mmol) and N-ethyldiisopropyl-amine (0.19mL, 1.08mmol) in 2-propanol (2.5mL). The title compound is obtained after trituration with 2-propanol as a brown solid (148mg, 47%).
Step 2: 4-(5-Bromo-[ 1 ,2 ,4j 'triazolo [ 1 ,5-aJ 'pyrazin-8-ylamino) -benzoic acid
Figure imgf000108_0002
[00393] 4-(5-Bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-ylamino)-benzoic acid methyl ester
(460mg, 1.32mmol) is suspended in THF (HmL) and a solution of lithium hydroxide monohydrate (554mg, 13.2mmol) in water (1 ImL) is added. The reaction mixture is stirred at room temperature for 4 hours then methanol (1 ImL) is added and the mixture is stirred at 5O0C for 24 hours. The mixture is partitioned between water and DCM, the aqueous phase is acidified with 2M HCl (pH 2) and a yellow precipitate is formed. The precipitate is collected by filtration, washed with water and diethyl ether and dried to afford the title compound (215mg, 49%).
Step 3: 4-(5-Bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-ylamino)-N-pyridin-3-ylmethyl-benzamide
Figure imgf000108_0003
[00394] A solution of 4-(5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-ylamino)-benzoic acid
(0.23Og, 0.69mmol), 3-hydroxybenzotriazole (0.103g, 0.76mmol), l-ethyl-3-(3'- dimethylaminopropyl)carbodiimide hydrate (0.146g, 0.76mmol) in DMF (5mL) and 3-picolylamine (0.077mL, 0.76mmol) is stirred at room temperature for 21 hours. The solvent is removed in vacuo and the residue is triturated with diethyl ether, ethyl acetate and dichloromethane. A pale yellow solid is obtained, washed with water and dried to afford the title compound (241mg, 82%).
Step 4: 4-(5-(lH-Pyrazol-4-yl)-[ 1 ,2 ,4j 'triazolo [ 1 ,5-aJ pyrazin-8-ylamino) -N-(pyridin-3- ylmethyl)benzamide
Figure imgf000109_0001
[00395] This compound may be prepared using methods as described for Compound 6, step 4 using 4-(5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-ylamino)-N-pyridin-3-ylmethyl-benzamide (lOOmg, 0.24mmol), 4-(4,4,5,5-tetramethyl-[l,3,2]dioxaborolan-2-yl)-lH-pyrazole (93mg, 0.48mmol) and Pd(PPh3)4 (70mg, O.Oόmmol) in 1.5M K2CO3 (aq) (1.4mL) and dioxane (2.5mL). The crude material is purified by silica gel column chromatography eluting with DCM followed by 98:2 then 96:4 then 90:10 DCM:NH3 (7M in MeOH) to afford the title compound as a white solid (19.6mg, 20%). 1H-NMR (400MHz, ds-DMSO) δ(ppm) 4.52 (2H, d), 7.40 (IH, m), 7.78 (IH, d), 7.93 (2H, d), 8.18 (2H, d), 8.32 (IH, s), 8.48-8.62 (4H, m), 8.82 (IH, s), 9.01 (IH, t), 10.21 (IH, br s), 13.3 (IH, br s). LCMS: Rt 1.86min (93%), m/z (ES+) 412 (M+H)+.
Compound 119: 4-(5-(2-Oxo-l,2-dihydropyridin-4-yl)-[l,2,4]triazolo[l,5-a]pyrazin-8-ylamino)- N-(pyridin-3-ylmethyl)benzamide
Step 1: 4-[5-(2-Methoxy-pyridin-4-yl)-[l,2,4Jtriazolo[l,5-aJpyrazin-8-ylaminoJ-N-pyridin-3-ylmethyl- benzamide
Figure imgf000109_0002
[00396] This compound may be prepared using methods as described for Compound 6, step 4 using 4-(5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-ylamino)-N-pyridin-3-ylmethyl-benzamide (120mg, 0.28mmol), 2-methoxypyridine-4-boronic acid (87mg, 0.57mmol) and Pd(PPh3)4 (81mg, 0.07mmol) in 1.5M K2CO3 (aq) (1.6mL) and dioxane (2.9mL). The crude material is purified by silica gel column chromatography eluting with DCM followed by 99:1 then 97:3 then 95:5 DCM:NH3 (7M in MeOH) to afford the title compound as a white solid (92.6mg, 73%).
Step 2: 4-(5-(2-Oxo-l ,2-dihydropyridin-4-yl)-[l ,2,4Jtriazolo[l ,5-aJpyrazin-8-ylamino)-N-(pyridin-3- ylmethyl)benzamide
Figure imgf000110_0001
[00397] A mixture of 4-[5-(2-methoxy-pyridin-4-yl)-[l,2,4]triazolo[l,5-a]pyrazin-8-ylamino]-
N-pyridin-3-ylmethyl-benzamide (71.5mg, O.lόmmol) and pyridinium hydrochloride (91mg, 0.79mmol) in water (0.5mL) in a sealed tube is heated at 15O0C for 25 minutes. After this time the solvent is removed in vacuo. The residue is chromatographed on silica gel, eluting with DCM followed by 98:2 and 90:10 DCM:NH3 (7M in MeOH), and the fractions containing the desired product are combined and evaporated. The title compound is isolated as a yellow solid (34.5mg, 49%).
[00398] 1H-NMR (400MHz, d6-DMSO) δ(ppm) 4.52 (2H, d), 6.82 (IH, m), 7.22 (IH, s), 7.38
(IH, m), 7.51 (IH, d), 7.78 (2H, d), 8.18-8.23 (3H, m), 8.51 (IH, d), 8.62 (IH, s), 8.82 (IH, s), 9.01 (IH, t), 10.58 (IH, br s), 11.8 (IH, br s). LCMS: Rt 1.72min (97%), m/z (ES+) 439 (M+H)+.
Compound 120: 2-Methoxy-N-(6-methylpyridin-3-yl)methyl-4-[5-(lH-pyrazol-4-yl)-
[l,2,4]triazolo [l,5-a]pyrazin-8-ylamino]-benzamide
Step 1 : 2-Methoxy-N-(6-methylpyridin-3-yl)methyl-4-nitrobenzamide
Figure imgf000110_0002
[00399] 2-Methoxy-4-nitrobenzoic acid (293 mg, 1.49 mmol) is dissolved in DMF (2 mL) and
4-methylmorpholine (220 μL, 3.0 mmol) and TBTU (1.79 g, 1.7 mmol) are added. The mixture is stirred at rt for 30 min and C-(6-methylpyridin-3-yl)methylamine (400 mg, 3.27 mmol) is added. Stirring is continued at rt for 12 h. DCM (10 mL) is added and the organic phase is washed with Na2CO3 (5% aq.), HCl (3% aq.) and water, and then dried over Na2SOzI. After evaporation of the solvents, the residue is triturated with ether-hexane to afford the title compound as a white solid.
Step 2: 4-Amino-2-methoxy-N-[(6-methylpyridin-3-yl)methylJbenzamide
Figure imgf000110_0003
[00400] A solution of 2-methoxy-N-(6-methylpyridin-3-yl)methyl-4-nitrobenzamide (448 mg,
1.49 mmol) in EtOH and EtOAc (8 mL each) is stirred and ammonium formate (375 mg, 6 mmol) and 10% Pd/C (100 mg) are added. The mixture is heated at reflux for 20 min, cooled, filtered through celite, the solid is washed with EtOH and the combined solvents are evaporated to afford the title compound.
Step 3: 4-(5-Bromo-fl,2,4Jtriazolofl,5-aJpyrazin-8-ylamino)-2-hydroxy-N-f(6-methyl-pyridin-3- yl)methyl]benzamide
Figure imgf000111_0001
[00401] 5,8-Dibromo-[l,2,4]triazolo[l,5-a]pyrazine (285 mg, 1.03 mmol) and 4-amino-2- methoxy-N-[(6-methylpyridin-3-yl)methyl]benzamide (280 mg, 1.03 mmol) are stirred in 1PrOH (5 mL) and HBr (48% aq., 380 μL) is added. The mixture is heated at reflux for 24 hours. The cooled suspension is poured into NaHCθ3 (sat. aq., 25 mL) and water (25 mL) and extracted with CHCI3 (4 x 30 mL). The extracts are dried over MgSθ4 and evaporated. The residue is purified by column chromatography, eluting with 10% CH2CVMeOH. The product-containing fractions are evaporated and the residue is triturated with MeOH to afford the title compound as a brown solid.
Step 4: 2-Methoxy-N-(6-methylpyridin-3-yl)methyl-4-[5-(lH-pyrazol-4-yl)-[l,2,4Jtriazolo[l,5- aJpyrazin-8-ylamino] ' -benzamide
Figure imgf000111_0002
[00402] Pyrazole-4-boronic acid (19 mg, 0.17 mmol), 4-(5-bromo-[l,2,4]triazolo[l,5- a]pyrazin-8-ylamino)-2-hydroxy-N-[(6-methyl-pyridin-3-yl)methyl]benzamide (40 mg, 0.085 mmol), K2CO3 (24 mg, 0.17 mmol) and Pd(dppf)Cl2 CH2Cl2 (4 mg, 0.005 mmol) are weighed into a sealable tube. The tube is flushed with nitrogen and dioxane-water (4:1, 4 mL) is added. The tube is sealed, placed in an ultrasonic bath under a flow of nitrogen gas for 30 seconds and then placed into an oil bath at 85°C. The reaction is stirred for 28 hours, adding additional portions of boronic acid (10 mg) and catalyst (2 mg) after 2 h and 18 h. The crude mixture is absorbed onto SiO2 and purified by column chromatography, eluting with 1 % - 10% MeOH/DCM. The product obtained is redissolved in MeOH/DCM (4:1, 10 mL) and 0.1 M MsOH/MeOH (2 eq) is added, the solvents evaporated and the residue taken up in water and lyophillised to afford the title compound as its bis-mesylate salt (38 mg). LCMS: Rt 0.92 min (93.9 %) m/z (ESI) 455 (M+H)+.
Compound 122: N-Benzyl-2-methoxy-4-[5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5-a]pyrazin-8- ylamino]-benzamide
Figure imgf000112_0001
[00403] This compound may be prepared using the methods described for Compound 120, using benzylamine in step 1. LCMS: Rt 1.62 min (100 %) m/z (ESI) 440 (M+H)+.
Compound 124: jty-Pyridin-2-ylmethyl 3-[5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5-a]pyrazin-8- ylaminojbenzamide
Figure imgf000112_0002
[00404] This compound may be prepared using methods as described for Compound 129 using
2-pyridylmethylamine in step 5. LCMS: Rt = 0.84 min (100 %), m/z (ESI) 412 (M+H)+.
Compound 127: (4-Isopropylpiperazin-l-yl)-{3-[5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5- a]pyrazin-8-ylamino]phenyl}methanone
Figure imgf000113_0001
[00405] This compound may be prepared using methods as described for Compound 129, using 4-isopropylpiperazine in step 5. LCMS: Rt = 0.84 min (100 %), m/z (ESI) 432 (M+H)+.
Compound 129: TV-Ethyl 3-[5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5-a]pyrazin-8- ylamino] benzamide
Step 1: 3-(5-Bromo-[ 1 ,2 ,4] triazolo [ 1 ,5-a] pyrazin-8-ylamino)benzoic acid ethyl ester
Figure imgf000113_0002
[00406] 5,8-Dibromo-[l,2,4]triazolo[l,5-a]pyrazine (1.39 g, 5.00 mmol) and ethyl 3- aminobenzoate (0.93 g, 5.60 mmol) are stirred in 1PrOH (10 mL) and HBr (48% aq., 1.14 mL, 10 mmol) is added. The mixture is heated at 85°C for 4 h and then cooled to rt and quenched with NaHCC"3 (sat. aq., 25 mL). The resulting suspension is cooled to 0°C and the white solid is collected by suction filtration and washed with water (10 mL). The crude product is taken up in EtOH and the solvent evaporated to afford the title compound as an off- white solid (1.79 g, 4.9 mmol) which is used without further purification.
Step 2: 3-[(5-Bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-\.&Λ-butoxycarbonyl-amino]benzoic acid ethyl ester
Figure imgf000113_0003
[00407] A solution of 3-(5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-ylamino)benzoic acid ethyl ester (1.79 g, 4.93 mmol) in DCM (20 mL) is stirred under N2 at rt and BOC2O (1.34 g, 6.16 mmol) and DMAP (0.30 g, 2.46 mmol) are added. Stirring is continued for 2 h, when LCMS indicates complete conversion of the starting material. The mixture is filtered and the filtrate is washed with dilute citric acid (pH 6, 5 mL) and brine (5 mL) and dried over Na2SOzI. Evaporation of the solvent affords the title compound as a yellow oil (1.87 g, 4.0 mmol) which is used without further purification.
Step 3: 3-{tert-Butoxycarbonyl-[5-(lH-pyrazol-4-yl)-[ 1,2,4] 'triazolofl,5-aj pyrazin-8-yl] - aminojbenzoic acid ethyl ester
Figure imgf000114_0001
[00408] Nitrogen is bubbled through a mixture of 3-[(5-bromo-[l,2,4]triazolo[l,5-a]pyrazm-8- yl)-tert-butoxycarbonyl-amino]benzoic acid ethyl ester (0.58 g, 1.25 mmol), 4-(4,4,5,5-tetramethyl- [l,3,2]dioxaborolan-2-yl)-lH-pyrazole (0.49 g, 2.50 mmol), K2CO3 (0.35 g, 2.50 mmol) and Pd(dppf)Cl2 DCM (102 mg, 0.125 mmol) in dioxane (37.5 mL) and water (9.6 mL). A condenser is fitted to the flask, the system is evacuated and purged with N2 (g) and then heated at 115°C for 25 min. The heating bath is removed and CO2 (s) is added to cool and buffer the system. The solvent is removed under reduced pressure and the residue is purified by silica chromatography, eluting with 10% to 50% ethyl acetate in DCM, to afford the title compound as an orange oil (0.61 g, > 100%) which is used without further purification.
Step 4: 3-{tert-Butoxycarbonyl-[5-(lH-pyrazol-4-yl)-[ '1,2,4] 'triazolofl ,5-a]pyrazin-8-yl]- aminojbenzoic acid
Figure imgf000114_0002
[00409] Impure 3-{tert-butoxycarbonyl-[5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5-a]pyrazin-8- yl]-amino}benzoic acid ethyl ester (0.61 g, 1.25 mmol) is dissolved in THF (2.5 mL) and KOH (2M aq., 2.5 mL, 5 mmol) is added and the mixture is heated at 60°C overnight. The solution is cooled and filtered through celite, rinsing with water. The filtrate is poured into citric acid (2M aq., 2.5 mL) and stirred at 0°C. The resulting solid is collected by suction filtration, washing with water and dried by suspending in EtOH and evaporation under reduced pressure, to afford a brown gum (280 mg). [00410] Further product is obtained by extraction of the filtrate with DCM, to afford a combined total of 352 mg of the title compound. This is used without further purification.
Step 5: N-Ethyl 3-[5-(lH-pyrazol-4-yl)-[l,2,4Jtriazolo[l,5-aJpyrazin-8-ylaminoJbenzamide
Figure imgf000115_0001
[00411] 3-{tert-Butoxycarbonyl-[5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5-a]pyrazin-8-yl]- amino}benzoic acid (42 mg, 0.10 mmol), ethylamine (12 M aq., 42 μL, 0.50 mmol), and triethylamine (28 μL, 0.20 mmol) are stirred in DMF (0.30 mL) and PyBOP (57 mg, 0.11 mmol) is added. Stirring is continued overnight. The reaction mixture is partitioned between EtOAc (7 mL) and NaHCθ3 (sat. aq., 7 mL) and the layers are separated. The combined organic layers are washed with brine (2 x 5 mL) and dried over Na2SOzI. Evaporation of the solvent affords an oil which is dissolved in MeOH (0.3 mL) containing HCl (12 M aq., 0.15 mL) and stirred overnight. Triethylamine (0.25 mL) and EtOH (2 mL) are added and the solvents removed under reduced pressure. The residue is purified by preparative HPLC to afford the title compound as a white solid (7 mg). 1H NMR (DMSO-d6, 400 MHz): δ = 13.29 ppm (s, 0.8 H); 10.01 (s, 0.9 H); 8.77 (s, 1.0 H); 8.65 (s, 1.0 H); 8.52 (s, 1.1 H); 8.41- 8.37 (m, 2.1 H); 8.25 (s, 1.0 H); 8.08 (d, 1.0 H); 7.47 (d, 1.0 H); 7.41 (t, 1.1 H); 7.04-7.03 (m, 0.5 H); 3.32 (s, 29.2 H (water)); 1.14 (t, 3.5 H). LCMS: Rt = 0.95 min (100 %), m/z (ESI) 349 (M+H)+.
Compound 131: Λ7-(4-Hydroxybenzyl) 3-[5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5-a]pyrazin-8- ylamino] benzamide
Figure imgf000115_0002
[00412] This compound may be prepared using methods as described for Compound 129, using 4-hydroxybenzylamine in step 5. LCMS: Rt = 0.98 min (100 %), m/z (ESI) 427 (M+H)+. Compound 134: Λf-BenzyWV-methyl 3-[5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5-a]pyrazin-8- ylamino] benzamide
[00413] This compound may be prepared using methods as described for Compound 129, using N-methylbenzylamine in step 5. LCMS: Rt = 1.15 min (100 %), m/z (ESI) 425 (M+H)+.
Compound 167: 4-[8-(6-Phenylacetylamino-pyridin-3-ylamino)-[l,2,4]triazolo[l,5-a]pyrazin-5- yl]-thiophene-2-carboxylic acid amide
Step 1: Η-(5-Nitropyridin-2-yl)-2-phenylacetamide
Figure imgf000116_0002
[00414] A solution of 2-amino-5-nitropyridine (4.17 g, 30 mmol) in pyridine (30 mL) is stirred at rt and a solution of phenylacetyl chloride (4.64 g, 30 mmol) in THF (30 mL) is added dropwise. The mixture is stirred for 24 h and then poured into ice-water (250 mL) to afford a brown solid, which is used without further purification.
[00415] This material is used to prepare N-[5-(5-bromo-[l,2,4]triazolo[l,5-a ]pyrazin-8- ylamino)-pyridin-2-yl]-2-phenylacetamide in a fashion analogous to steps 2 and 3 of the methods as described for Compound 120.
Step 4: 4-[8-(6-Phenylacetylamino-pyridin-3-ylamino)-[l,2,4]triazolo[l,5-a]pyrazin-5-yl]-thiophene- 2-carboxylic acid amide
Figure imgf000116_0003
[00416] This compound may be prepared using methods as decribed for Compound 120, step
4, using N-[5-(5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-ylamino)-pyridin-2-yl]-2-phenylacetamide and 2-(aminocarbonyl)thiophene-4-boronic acid. LCMS: Rt 1.06 min (100 %) m/z (ESI) 471 (M+H)+.
Compound 169: 2-(4-{4-[5-(lH-Pyrazol-4-yl)-[l,2,4]triazolo[l,5-a]pyrazin-8-ylamino]phenyl}- piperidin-l-yl)acetamide
Figure imgf000117_0001
[00417] This compound may be prepared using methods as described for Compound 84, using
2-bromoacetamide in step 1. LCMS: Rt = 0.81 min (100%), m/z (ESI) 418 (M+H)+.
Compound 170: 5-{8-[4-(4-Isopropylpiperazin-l-yl)-3-trifluoromethylphenylamino]-
[l,2,4]triazolo[l,5-a]pyrazin-5-yl}-2,3-dihydroisoindol-l-one
Figure imgf000117_0002
[00418] This compound may be prepared using the methods as described for Compound
GB15, using 5-(4,4,5,5-tetramethyl-[l,3,2]dioxaborolan-2-yl)-2,3-dihydroisoindol-l-one. LCMS: Rt = 0.99 min (95 %), m/z (ESI) 537 (M+H)+.
Compound 171 : [4-(4-Isopropylpiperazin-l-yl)-3-trifluoromethylphenyl]-[5-(lH-pyrazol-4-yl)- [l,2,4]triazolo[l,5-a]pyrazin-8-yl]amine
Figure imgf000118_0001
[00419] This compound may be prepared using methods as described for Compound 46, using
(5-bromo-[l,2,4]triazolo[l,5-a]pyrazin-8-yl)-[4-(4-isopropylpiperazin-l-yl)-3- trifluoromethylphenyl]amine. LCMS: Rt = 0.99 (95 %), m/z (ESI) 472 (M+H)+.
[00420] Other examples of compounds of the invention, prepared by the above procedures are described hereinabove.
Purification Conditions and Characterization
[00421] Routinely, post-synthesis all compounds may be purified using reverse phase HPLC using a Gilson preparative HPLC system (322 pump, 155 UV/VIS detector, 215 liquid handler). The Gilson 215 acts as both auto-sampler and fraction collector. Compounds can also be purified by flash chromatography on silica gel.
[00422] Compounds are characterised by mass spectrometry using single quadrupole instrumentation with an electrospray source.
Biological Assays Demonstrating the Usefulness of the Compounds
Example 1: MAPKAP-K5 Assay
[00423] MAPKAP-K5 reactions are performed in FlashPlate format using 0.1 or 0.2 μCi 33P-
ATP; 0.6μM ATP; ImU MAPKAP-K5; 3μM MAPKAP-K5 peptide substrate, incubated at room temperature for 30 minutes.
Flashplate assay:
[00424] The MAPKAP-K5 kinase reaction is performed in a 384 well polypropylene plate
(Matrix Technologies) and then transferred to a streptavidin-coated 384 well flashplate (Perkin-
Elmer).
[00425] To wells containing 2μL test compound or standard inhibitor, 13μL Enzyme mix or diluent are added using a Hydra (Robbins Scientific).
[00426] Reactions are started by addition of lOμL of [2.5x] substrate cocktail using a
Multidrop (Thermo-Labsystems), to give final concentrations in the assay of: ImU MAPKAP-K5 3μM MAPKAP-K5 peptide substrate 0.6μM ATP
0.004μCi [33P]-γ-ATP/μL Ix reaction buffer
[00427] Plates are incubated at room temperature for 30 minutes.
[00428] Reactions are terminated by the addition of 25 μL EDTA (5OmM) to each well using a
Micro-fill (Biotek).
[00429] Reactions are transferred to a streptavidin-coated flashplate using a Zymark robotic system. Plates are incubated for 60 minutes at room temperature.
[00430] All wells are washed 3 times with lOOμl phosphate buffered saline using a Tecan plate washer.
[00431] Radioactivity is determined by scintillation counting of the flashplate (empty wells) on a Packard TopCount.
Enzyme Mix:
Enzyme
5OmM Tris HcI (pH 7.5) 0. ImM EGTA 2mM DTT lmg/mL BSA Reaction Buffer:
5OmM Tris HcI (pH 7.5)
0. ImM EGTA
1 OmM Magnesium acetate
2mM DTT
[00432] The following compounds have been or can be prepared according to the synthetic methods described above. For the purpose of Table 1 and Table 2 below, activity of each compound, which can be determined using the MAPKAPK5 assay method described in Example 1 , is expressed as follows:
++++ compound exhibited MAPKAPK5 IC50 1 - 100 nM
+++ compound exhibited MAPKAPK5 IC50 101 -500 nM
++ compound exhibited MAPKAPK5 IC50 501 - 1000 nM
+ compound exhibited MAPKAPK5 IC50 > 1000 nM
TABLE 1 : Structure and activity of of Exemplary Compounds
Figure imgf000120_0001
Figure imgf000121_0001
Figure imgf000122_0001
Figure imgf000123_0001
Figure imgf000124_0001
Figure imgf000125_0001
Figure imgf000126_0001
Figure imgf000127_0001
Figure imgf000128_0001
Figure imgf000129_0001
Figure imgf000130_0001
Figure imgf000131_0001
Figure imgf000132_0001
Figure imgf000133_0001
Figure imgf000134_0001
Figure imgf000135_0001
Figure imgf000136_0001
TABLE 2: Structure and activity of Exemplary Benzamide Compounds
Figure imgf000137_0001
Figure imgf000138_0001
Figure imgf000139_0001
Figure imgf000140_0001
Figure imgf000141_0001
Figure imgf000142_0001
Figure imgf000143_0001
Figure imgf000144_0001
Example 2. Development of an assay for the identification of regulators of the expression of MMPl by activated primary synovial fibroblasts.
[00433] To identify compounds that decrease the ECM- degrading activity of cells, the ECM- degrading activity of cells may be induced to allow proper detection of this activity, and to achieve a clearer read-out. In the context of RA, the cells of choice are mammalian synovial fibroblasts and the triggers that may be used to induce the ECM-degrading activity are cytokines relevant in the field of arthritis: for instance TNF-α, ILlB, IL6, OSM, IL17, and MIFl-α. This list is not comprehensive due to the plethora of cytokines potentially involved in the RA pathogenesis (Smolen and Steiner, 2003). To set up an in vitro assay that is as close as possible to the complexity of the pathology, the trigger applied should be a mixture of factors generated by contacting cytokine-producing cells relevant in the field of arthritis, such as monocytes, macrophages, T-cells, and B-cells, with a trigger. The cytokine- producing cells will respond to the contact by producing a complex and unbiased mixture of factors. If the cytokine-producing cell used is also found in a pannus, and the cytokine applied to produce this trigger is found in the synovial fluid of rheumatoid arthritis patients, the mixture of factors ultimately produced will contain part of the factors that are present in the joints of arthritis patients. Principle of the "MMP assay'
[00434] Matrix Metallo Proteases (MMPs) possess various physiological roles, as e.g. the maturation of other proteases, growth factors, and the degradation of extra-cellular matrix components. MMPl is one of the members of the MMP family that is able to degrade native collagen, the main component of bone and cartilage. An increased expression of MMPl by synovial fibroblasts (SFs) is diagnostic for the progression of the arthritic disease and is predictive for erosive processes in the joint (Cunnane et al., 2001). The expression of MMPl by SFs can be increased by the activation of SFs with triggers relevant for rheumatoid arthritis, as cytokines like TNF-α or IL IB (Andreakos et al., 2003). Taken together, measurement of the levels of MMPl produced by activated SFs is a readout that is highly relevant in the context of RA as this event reflects the level of activation of SFs towards an erosive phenotype as it is seen in the pannus. If a reduced expression of a candidate drug target in activated SFs leads to the reduction of MMPl expression by these cells, the drug target is then proven to be involved in the regulation of MMPl expression and thus considered relevant for the development of therapeutic strategies for the treatment of RA.
[00435] In the following examples, the development of an assay, further referred to as 'MMP assay', monitors the MMPl production by synovial fibroblasts (SFs) in response to diverse activating triggers (Example 2.1). The use of this assay is then described for the validation of gene products that are considered drug targets for the development of RA therapies (Example 2.2). The validation of drug targets is performed using recombinant adenoviruses, further referred to as knock-down viruses or Ad-siRNAs, that mediate the expression in cells of shRNA's which reduce the expression levels of targeted genes by a RNAi (RNA interference)-based mechanism (see WO 03/020931). The identification of compounds modulating the activity of the validated drug targets is then described in Table 3. The use of the 'MMP assay' for the testing of compounds that modulate the activity of the drug targets identified is described further below.
Assay Examples Control viruses used:
[00436] The control viruses used in these studies are listed below. dEl/dE2A adenoviruses are generated from these adapter plasmids by co-transfection of the helper plasmid pWEAd5AflII- rlTR. dE2A in PER.E2A packaging cells, as described in WO99/64582. Negative control viruses:
Ad5-eGFP_KD: Target sequence: GCTGACCCTGAAGTTCATC (SEQ ID NO: 1). Cloned using Sap 1 -sites into vector and virus generated as described in WO03/020931. Ad5-Luc_vl3_KD: Target sequence GGTTACCTAAGGGTGTGGC (SEQ ID NO: 2). Cloned using Sap 1 -sites into vector and virus generated as described in WO03/020931.
Ad5-M6PR_vl_KD: Target sequence CTCTGAGTGCAGTGAAATC (SEQ ID NO: 3). Cloned using Sap 1 -sites into vector and virus generated as described in WO03/020931.
Positive control viruses:
Ad5-MMPl_vlO_KD: Target sequence ACAAGAGCAAGATGTGGAC (SEQ ID NO: 4). Cloned using Sap 1 -sites into vector and virus generated as described in WO03/020931.
Viruses used for target validation:
Ad5-MAPKAPK5_vl3_KD: Target sequence CGGCACTTTACAGAGAAGC (SEQ ID NO: 5). Cloned using Sap 1 -sites into vector and virus generated as described in WO03/020931.
Ad5-MAPKAPK5_vl2_KD: Target sequence ATGATGTGTGCCACACACC (SEQ ID NO: 6). Cloned using Sap 1 -sites into vector and virus generated as described in WO03/020931.
Example 2.1: Development of the MMP assay
[00437] A 384-well format ELISA for measurement of MMPl is developed. Various primary antibodies are tested, as well as various ELISA protocols. The following protocol is developed and validated to measure MMPl levels in SF supernatant in 384 well plates: white Lumitrac 600 384 well plates (Greiner) are coated with 2 μg/mL anti-MMPl antibody MAB1346 (Chemicon). The antibody is diluted in buffer 40 (1.21 g Tris base (Sigma), 0.58 g NaCl (Calbiochem) and 5 ml 10% NaN3 (Sigma) in 1 L milliQ water and adjusted to pH 8.5). After overnight incubation at 4°C, plates are washed with PBS (80 g NaCl, 2g KCl (Sigma), 11.5 g Na2HPO4JH2O and 2 g KH2PO4 in 10 L milliQ; pH 7.4) and blocked with 100 μL/well Casein buffer (2% Casein (VWR International) in PBS). Next day, casein buffer is removed from ELISA plates and replaced by 50 μL/well EC buffer (4 g casein, 2.13 g Na2HPO4 (Sigma), 2 g bovine albumin (Sigma), 0.69 g NaH2PO4-H2O (Sigma), 0.5 g CHAPS (Roche), 23.3 g NaCl, 4 ml 0,5 M EDTA pH 8 (Invitrogen), 5 ml 10% NaN3 in 1 L milliQ and adjusted to pH 7.0). 0.25 mM DTT (Sigma) is added to the thawed samples plates. After removal of the EC buffer, 20 μL of sample is transferred to the ELISA plates. After overnight incubation at 4°C plates are washed twice with PBS and once with PBST (PBS with 0.05% Tween-20 (Sigma)) and incubated with 35 μL/well biotinylated anti-MMPl antibody solution (R&D). This secondary antibody is diluted in buffer C (0.82 g NaH2PO4-H2O, 4.82 g Na2HPO4, 46.6 g NaCl, 20 g bovine albumin and 4 mL 0,5M EDTA pH 8 in 2 L milliQ and adjusted to pH 7.0) at a concentration of 5 μg/mL. After 2 h of incubation at RT, plates are washed as described above and incubated with 50 μL/well streptavidin- HRP conjugate (Biosource). Streptavidin-HRP conjugate is diluted in buffer C at a concentration of 0.25 μg/mL. After 45 min, plates are washed as described above and incubated for 5 min with 50 μL/well BM Chem ELISA Substrate (Roche). Readout is performed on the Luminoscan Ascent Luminometer (Labsystems) with an integration time of 200 msec or with an Envision reader (Perkin
Elmer). [00438] The increase of MMPl expression by SFs upon treatment with cytokines relevant in the field of RA (TNF-α, ILlB and OSM) or a combination thereof is shown in Figure 2 as white bars. For this experiment, SFs are seeded in 96 well plates, 3,000 cells/well. 24 h later, the medium is changed to Ml 99 medium supplemented with 1% FBS. One day after the medium change, cytokines or combinations thereof are added to the cultures, each cytokine being added to a final concentration of 25 ng/mL. 72 h after cytokine addition, the supernatant is collected and processed in the MMPl ELISA as described in the protocol given above. In parallel with this experiment, SFs are triggered, using the same protocol, with the supernatant of THPl cells (2-fold diluted in Ml 99 + 1% FBS) treated with the same cytokines or combinations of cytokines for 48 h in Ml 99 medium + 1% FBS. MMPl levels for these samples are shown in Figure 2 as grey bars. The induction of the MMPl expression by SFs triggered with the supernatants of TNF-α-treated THPl cells is stronger (>4.5 fold induction) as compared to the SFs triggered with recombinant TNF-α alone (3 -fold induction) and almost equals the 5-fold induction obtained by a mixture of 3 purified cytokines (TNF-α, ILlBb, OSM). This result indicates that the supernatant of TNF-α-induced THPl cells contains, besides TNF- α, additional pro-inflammatory factors that activate SFs towards MMPl expression. As the role of TNF-α in the RA pathogenesis is validated (TNF-α-blockers such as Infliximab and Etanercept show some efficacy in the treatment of RA patients) and the THP-I cells are representative for monocytes / macrophages present in the joint of RA patients, the TNF-α-based trigger mixture prepared by contacting THP-I cells with TNF-α will contain factors present in the joints of RA patients and subsequently is relevant to RA. This TNF-α-based complex trigger, further referred to as the 'complex trigger', will further be used as basis for the 'MMP assay'.
[00439] Inhibition of the activation of SF by the 'complex trigger' is shown using dexamethasone, a potent anti-inflammatory agent that also strongly reduces collagen-induced arthritis in rodents (Yang et al., 2004) (Figure 3). Dexamethasone is shown to dose-dependently reduce amounts of MMPl produced by complex trigger activated SFs. SFs are seeded at a density of 3000 cells/well in 96 well plates. 24hrs after seeding, increasing concentrations of dexamethasone are added to the cells. After overnight incubation, medium of every well is refreshed to supernatant of THP-I cells treated with TNF-α (50% diluted in Ml 99 + 0.5%FBS), and the same concentration of dexamethasone as added the day before. 48hrs after treatment, the supernatant is collected and subjected to the MMPl ELISA described above. The addition of dexamethasone clearly reduced the MMPl expression by SFs, with an IC5O value of about InM (see Figure 3). These data show that the MMPl expression by activated SFs can be reduced by the addition of a physiologically relevant inhibitor and represent a proof of principle for the 'MMP assay'.
Example 2.2: MAPKAPK5 Modulates SF 'Complex Trigger '-induced MMPl Expression (A) Ad-siRNA Virus Functions to Knock Down MAPKAPK5 Expression. [00440] Recombinant adenoviruses mediating the expression of siRNA's targeting
MAPKAPK5 and eGFP are generated according to the procedure described in WO03/020931. The target sequence used in the recombinant adenovirus is: CGGCACTTTACAGAGAAGC (SEQ ID NO: 5) as well as ATGATGTGTGCCACACACC (SEQ ID NO: 6). The target sequence within the eGFP mRNA used in the recombinant adenovirus is: GCTGACCCTGAAGTTCATC (SEQ ID NO: 1). These sequences are cloned into the adapter plasmid using Sapl sites. dEl/dE2A adenoviruses are generated from these adapter plasmids by co-transfection of the helper plasmid pWEAd5AflII- rITR.dE2A in PER.E2A packaging cells, as described in WO99/64582.
[00441] The functionality of an adenovirus targeting MAPKAPK5 is tested as follows. These adenoviruses are used to infect primary human SFs cultured in petri dishes as follows. On day 1, 500.000 SFs are seeded per petri dish. One day later, the cells are infected with Ad5-MAPKAPK5- vl3_KD (1.6E9 VP/mL) or Ad5-eGFP-v5_KD (1.3E10 VP/mL) at an MOI of 4000 (based on the titers (number of virus particles per mL) defined for the viruses by Q-rt-PCR). On day 7, cells are detached from the petri dish according to standard procedure using a trypsin EDTA solution. The trypsin is then neutralized by addition of DMEM growth medium supplemented with 10%FBS. The cells are then collected by a centrifugation step (1000 rpm, 5 min). The pellet is lysed in lOOμL of fresh RIPA buffer (5OmM Tris pH7.5, 15OmM NaCl, 1% deoxycholate, 1% Triton XlOO, 0.1% SDS). The samples are then sonicated for lOsec. The protein concentration of the samples is then determined using the BCA kit (Pierce, Cat N° 23227) as described by the provider, using BSA as a standard. To 30μg of cell lysate diluted to 19.5μl in RIPA buffer, 3.5μL of reducing agent (NuPage reducing agent N°10, Invitrogen NP0004) and 7.5μL of sample buffer (NuPage LDS sample buffer, Invitrogen NP0007) are added. The 30μL sample is then boiled for 5min and loaded on a 10% polyacrylamide gel (Invitrogen NP0301). To allow the estimation of the level of protein knock-down, 15μg, 7.5μg and 3.75μg of the lysate of the Ad5-eGFP-v5_KD infected cells are also loaded onto the gel. The gel is then run for 2 hours at 100V in Ix MOPS/SDS NuPage running buffer (Invitrogen NPOOl). lOμl of Seablue Plus Prestained standard (Invitrogen LC5925) is used to estimate protein size on the gel. The proteins on the gel are then transferred onto a PVDF membrane (Invitrogen LC2002) by a wet blotting procedure using a transfer buffer prepared by mixing 100ml Nupage Transfer buffer 20* (NP0006-1), 40OmL methanol and 150OmL Milli Q water. Before the transfer, the membrane is first soaked in methanol and in transfer buffer. The transfer is performed at 100V for 90 minutes. The membrane is then blocked by 30 min soaking in blocking buffer (2% blocking blocking powder (Amersham, RPN 2109) prepared in PBST (PBS supplemented with 0,1% Tween 20 (Sigma, P1379)). After blocking, the immunodetection is performed using a mouse monoclonal antibody against MAPKAPK5 (BD Biosciences, Cat N°612080) diluted 250 fold in blocking buffer. After overnight incubation with this primary antibody, the membrane is washed 3 times with PBST and incubated 1 hr with the secondary antibody ((Polyclonal goat anti-mouse Ig, HRP conjugated (DAKO P0447) diluted 50000 fold in blocking buffer. The blot is then washed 3 times in PBST and the detection is performed with ECL advance (RPN2109, Amersham) on a Kodakimager according to the manufacturers instructions. The Western Blotting revealed a lower expression level of MAPKAPK5 in the Ad5-MAPKAPK5-vl3_KD infected cells compared to the cells infected with the Ad5-eGFP-v5_KD negative control virus. Comparison with the diluted Ad5-eGFP-v5_KD infected samples allowed to estimate the reduction in expression to be 2-fold. Equal loading of the 30μg samples is demonstrated by immunodetection of β- actin after removal of the MAPKAPK5 antibody by a 'stripping procedure' (5 minutes boiling of the membrane in PBST). Immunodetection of β-actin is performed according to the method described for MAPKAPK5 detection, but using a goat polyclonal antibody against β-actin (Santa Cruz, Cat N° SC- 1615) at a 1000 fold dilution as primary antibody and a rabbit anti goat antibody at a 50000 fold dilution as a secondary antibody. Results of this experiment are given in Figure 4. Taken together, this experiment demonstrated the functionality of the Ad-siRNA virus produced to reduce the MAPKAPK5 expression levels in primary human SFs.
(B) MAPKAPK5 knock-down Ad-siRNA Reduces SF-induced MMPl Expression
[00442] The efficacy of Ad5-MAPKAPK5-vl3_KD virus in the 'MMP assay' is tested as follows. Day 1, SFs (passage 9 to 10) are seeded in 96 well plates at a density of 3000 cells per well in complete synovial growth medium (Cell Applications). One day later, the cells are infected with increasing amounts (3, 6; 9, 12 or 15 μl) of following viruses: Ad5-eGFP-v5_KD, Ad5-MAPKAPK5- vl2_KD, Ad5-MAPKAPK5-vl3_KD, Ad5-MMPl-vlO_KD. The virus load is corrected by addition of the neutral virus Ad5-Luc-vl3_KD to bring the final virus volume on the cells to 15μL in every well. This correction guarantees that the effects observed do not result from the virus load applied to the cells. The cells are then incubated for 5 days before the activation step. This step involves the replacement, in every well, of the growth medium by 75μL of M199 medium supplemented with 25μL of 'complex trigger'. 48 hrs after the activation step, the supernatant is collected and subjected to the MMPl ELISA as described in Example 1. The results of the experiment are shown in Figure 5. The quality of the experiment is demonstrated by the efficacy of the Ad-siRNA virus targeting MMPl itself. This positive control virus strongly reduces the MMPl expression by SFs, whereas the negative control virus, designed to target the expression of luciferase, does not influence the levels of MMPl expression. Two viruses used to validate the MAPKAPK5 target (Ad5-MAPKAPK5-vl2_KD and Ad5-MAPKAPK5-vl3) do also lead to a clear reduction of the complex trigger induced MMPl expression by primary human SFs. It can be concluded, from this experiment, that MAPKAPK5 represents a valuable drug target that is shown to modulate MMPl expression in SFs. Similarly, the inhibition of MAPKAPK5 enzymatic activity by a small molecule compound is expected to reduce the 'complex cytokine' induced MMPl expression in the 'MMP assay'. The inhibition of MAPKAPK5 enzymatic activity by a small molecule compound is also predicted to reduce the degradation of the joint associated with RA. (C) In vitro 'MMP assay' Testing of Compounds Inhibiting MAPKAPK5
[00443] Compounds inhibiting the MAPKAPK5 activity in a biochemical assay (i.e. cell free, using purified enzyme), are tested in the 'MMP assay' according to following protocol. [00444] The compound master stocks (all at 1OmM concentration in 100% DMSO) are diluted
10-fold in water (Distilled water, GIBCO, DNAse and RNAse free) to obtain a ImM intermediate work stock in 10% DMSO. This intermediate work stock is further diluted either 3-fold (or 10-fold) in 10%DMSO to obtain an intermediate work stock of 333μM (or lOOμM) concentration, respectively, in 10% DMSO. The ImM as well as 333μM (or lOOμM) intermediate work stocks are then further diluted 10-fold in 1.1% DMSO to obtain the 1Ox workstocks at lOOμM and 33.3μM (or lOμM) concentration in 2% DMSO. This 1Ox work stock is then diluted 10-fold in Ml 99 medium supplemented with 1%FBS to obtain the final 'Ix compound preparation' containing the compounds at lOμM and 3.33μM (or lμM) as well as 0.2% DMSO. These are the final conditions at which the compounds are tested on the cells. In parallel, the 1Ox work stock is diluted 10-fold in 'complex trigger' (i.e. the supernatant of TNF-α treated THPl cells produced as described in Example 1) that is diluted 2-fold in M199 supplemented with 1% FBS to produce the 'Ix compound in 50% complex trigger preparation' .
[00445] At day 1, RASFs are seeded in 96 well plates (Flat bottom, tissue culture treated,
Greiner) at a density of 3000 cells/ well in complete synovial growth medium (Cell Applications). Day 5, the compounds are added to the cultured cells as follows. Medium is completely removed from the cells and replaced by 75 μL of the 'Ix compound preparations' containing the compounds at either lOμM or 3.33μM (or lμM) in M199 medium supplemented with 1%FBS and 0.2% DMSO. After an incubation period of 2 hours, which allows the compounds to equilibrate and enter the cells, 25 μL of the 'Ix compound in 50% complex trigger preparations' are added to the wells on top of the 'Ix compound preparation', in the wells containing the corresponding compounds at corresponding concentration. In this way, an 8-fold diluted complex trigger is ultimately applied to the cells. An incubation of 48 hrs is then performed and 20μl of the cell supernatant is then processed in the MMPl ELISA as described above, delivering raw data (RLU: relative luminescence units). Following controls are included in the experiments. A maximal signal control, in which the cells are activated by the complex trigger but only the 0.2% DMSO vehicle (and thus no compound) is added. This control indicates the maximal level of MMPl that can be achieved in the test. A minimal signal control is also included in these experiments. Here, cells are not triggered. The medium of the cells is then changed to lOOμl Ml 99 medium supplemented with 1% FBS at day 5. This control returns the basal MMPl levels produced by the RASFs. The percent inhibition of the MMPl expression achieved by the compounds is then calculated based on the RLU data returned by the ELISA with following formula: [[(maximal MMPl levels - minimal MMPl levels) - (MMPl level compound X at concentration Y- minimal MMPl levels)]/(maximal MMPl levels - minimal MMPl levels)]xlOO.. [00446] Toxicity of the compounds is assessed as follows. Day 1, SFs are seeded in white, tissue culture treated 96 well plates at a density of 3000 cells per well in lOOμL complete synovial growth medium. The compound handling, compound addition to the cells as well as activation of the cells is further performed as described above in this example for the determination of the MMPl levels. After the 48hrs incubation period, the medium is removed from the wells, replaced by 50μL fresh Ml 99 medium supplemented with 1% FBS. 50μL of substrate (Promega Celltiter Glow cell viability kit) is then added to the wells. After an incubation period of 10 min, luminescence signal is measured. A reduction of the luminescence signal by more than 50% as compared to the maximal control wells is considered to reflect significant toxicity. No toxicity is observed for the compounds tested in the 'MMP assay'.
[00447] It should be understood that factors such as the differential cell penetration capacity of the various compounds can contribute to discrepancies between the activity of the compounds in the in vitro biochemical and cellular MMP assays.
[00448] For the purpose of Table 3 and Table 4 below, MMPl EC5O of each compound, which can be determined using the assay method described herein, is expressed as follows:
* * * * compound exhibited MMP 1 EC50 1 - 100 nM
*** compound exhibited MMPl EC50 101-500 nM
** compound exhibited MMPl EC50 501-1000 nM compound exhibited MMPl EC50 >1000 nM TABLE 3
Figure imgf000151_0001
Figure imgf000151_0002
Figure imgf000152_0001
Figure imgf000152_0002
Figure imgf000153_0001
Figure imgf000153_0002
Figure imgf000154_0003
Figure imgf000154_0001
Figure imgf000154_0002
TABLE 4
Figure imgf000154_0004
Figure imgf000154_0005
Figure imgf000155_0002
Figure imgf000155_0001
Example 3: Assay to assess effect of compounds on cytokine release by human PBMCs
[00450] Human peripheral blood mononuclear cells (PBMCs) are isolated from "buffy coats" prepared from the blood of healthy volunteers, isolated essentially according to method of Bøyum (1984). In brief, buffy coat is diluted 1 :1 with Ix PBS (Gibco) and 30 mL is carefully put on top of 20 mL Lymphoprep™ (Lucron Bioproducts) in 50 mL Falcon tubes. After centrifugation (35 min, 400 g, 180C) the mononuclear cells are collected from the white interphase and washed 3 times with Ix PBS by resuspending and centrifugation (10 min, 200 g). Isolated PBMCs are finally resuspended in RPMI 1640 (Cat.No. 21875, Gibco) that is supplemented with 10% heat-inactivated FBS (Hyclone). [00451] For the assay PBMCs are seeded at 2.5E6 cells/mL in 160 μL in 96-well plates
(Nunc). Serial dilution of the test compounds are made first in DMSO (Sigma) and then diluted 50- fold in M199 medium (Gibco) containing 1% heat-inactivated FBS. Compounds are further 1/10 diluted in the assay plates to obtain final DMSO concentration of 0.2%. Cells are preincubated with the compounds for 1 hr at 370C, 5% CO2. Then, cells are stimulated with LPS (Escherichia coli serotype 026:B6, Cat.No. L2654, Sigma) that is added in a volume of 20 μL to a final concentration of 1 μg/mL and cells are further cultured for 24 hr. The plates are centrifuged and the supernatant is collected and stored at -8O0C until analysis of appropriate dilutions in ELISAs. [00452] The following 384-well chemiluminescent ELISA protocol was developed to measure
TNFα levels in the supernatant : White Lumitrac 600 384-well plates (Greiner) are coated with (40 μL/well) anti-TNFα capture antibody (Cat.No. 551220, BD Pharmingen) that is diluted to 1 μg/mL in Ix PBS (Gibco). After overnight incubation at 4°C, plates are washed with Ix PBS (80 g NaCl, 2g KCl (Sigma), 11.5 g Na2HPO4.7H2O and 2 g KH2PO4 in 10 L milliQ; pH 7.4) and blocked with 100 μL/well buffer B (Ix PBS containing 1% BSA (Sigma), 5% sucrose (Sigma) and 0.05% NaN3 (Sigma)). After 4 hr incubation at RT, blocking buffer is removed and plates are washed once with PBST (Ix PBS with 0.05% Tween-20 (Sigma)). Then, 40 μL of sample is transferred to the ELISA plates and plates are incubated at 40C. Next day, plates are washed 3 times (twice with PBST and once with PBS) and 35 μL/well biotinylated anti-TNFα antibody (Cat.No. 554511, BD Pharmingen) diluted first to a concentration of 250 ng/ml in buffer D (Ix PBS with 1% BSA) is added. After 2 h of incubation at RT, plates are washed as described above and 35 μL/well of a 1/2000 dilution of streptavidin-HRP conjugate (Cat.No. SNN2004, Biosource) in buffer D is added. After 45 min, plates are washed as described above and incubated for 5 min with 50 μL/well BM Chemiluminescence ELISA Substrate POD (Roche). Readout is performed on the Luminoscan Ascent Luminometer (Labsystems) with an integration time of 100 msec delivering raw data (RLU: relative luminescence units). The following controls are included in the experiments, a maximal signal control, in which the cells are activated by LPS but only the 0.2% DMSO vehicle (and thus no compound) is added. This control indicates the maximal level of TNFα that can be achieved in the test. A minimal signal control is also included in these experiments. Here, cells are not triggered. This control returns the basal TNFα levels produced by the PBMCs. The percent inhibition (PIN) of the TNFα release, achieved by the compounds is then calculated based on the RLU data returned by the ELISA with following formula: 100 - [((TNFα level compound X at concentration Y- minimal TNFα levels)/(maximal TNFα levels - minimal TNFα levels))xlOO]. Where compounds are tested at 8 concentrations (1/3 serial dilution), EC50-values can be calculated by curve fitting of the means of the PIN data achieved for a compound at each test concentration.
[00453] To assay the effect of c ompounds on the release of ILl and IL6 by LPS stimulated
PBMC cultures, appropriate dilutions of the supernatant can be measured using the same ELISA protocol as described above. Matched pair antibodies for ILl and IL6 ELISA (all from R&D Systems) may be used as follows : anti-ILl capture antibody (Cat.No. MAB601) used at 0.5 μg/mL , biotinylated anti-ILl detection antibody (Cat.No. BAF201) used at 50 ng/mL ; anti-IL6 capture antibody (Cat.No. MAB206) used at 1 μg/mL; biotinylated anti-IL6 detection antibody (Cat.No. BAF206) used at 50 ng/mL.
[00454] For the purpose of Table 5 below, PBMC EC5O of each compound, which can be determined using the assay method described herein, is expressed as follows:
#### compound exhibited PBMC EC50 1 - 100 nM
### compound exhibited PBMC EC50 101 -500 nM
## compound exhibited PBMC EC50 501 - 1000 nM
# compound exhibited PBMC EC50 >1000 nM
[00455] TABLE 5
Figure imgf000156_0001
Figure imgf000156_0002
Figure imgf000157_0001
Figure imgf000157_0002
Figure imgf000158_0002
Figure imgf000158_0001
[00456] The present invention relates also to a method of treatment or prevention of inflammatory diseases, which comprises administering to a subject in need thereof, a therapeutically effective inhibitor of Mitogen- Activated Protein Kinase- Activated Protein Kinase 5 inhibiting amount of a compound according to Formula 1.
[00457] Another aspect of the present method invention relates to a method of treatment or prophylaxis of a condition characterised by abnormal matrix metallo proteinase activity, which comprises administering a therapeutically effective amount of a matrix metallo proteinase inhibiting compound according to Formula 1.
[00458] A further aspect of the present method invention is a method of treatment or prophylaxis of a condition selected from diseases involving degradation of extra-cellular matrix, which comprises administering a therapeutically effective matrix metallo proteinase inhibiting amount of a compound according to Formula 1.
[00459] A yet further aspect of the present method invention is a method of treatment or prophylaxis of a condition selected from diseases involving abnormal cellular expression of MMPl, which comprises administering a therapeutically effective matrix metallo proteinase inhibiting amount of a compound according to Formula 1.
[00460] A special embodiment of the present method invention is a method of treatment or prevention of rheumatoid arthritis, which comprises administering to a subject in need thereof, a therapeutically effective amount of a compound according to Formula 1.
[00461] This invention also relates to the use of the present compounds in the manufacture of a medicament for treatment or prophylaxis of a condition prevented, ameliorated or eliminated by administration of an inhibitor of Mitogen- Activated Protein Kinase- Activated Protein Kinase 5, or a condition characterised by abnormal collagenase activity, or a condition selected from diseases involving inflammation, most preferably in for the treatment of rheumatoid arthritis. [00462] Administering of the compound of the present invention to the subject patient includes both self-administration and administration by another person. The patient may be in need of treatment for an existing disease or medical condition, or may desire prophylactic treatment to prevent or reduce the risk for diseases and medical conditions affected by a disturbance in bone metabolism. The compound of the present invention may be delivered to the subject patient orally, transdermally, via inhalation, injection, nasally, rectally or via a sustained release formulation. [00463] A preferred regimen of the present method comprises the administration to a subject in suffering from a disease condition characterized by inflammatory, with an effective matrix metallo- protease inhibiting amount of a compound of the present invention for a period of time sufficient to reduce the abnormal levels of extracellular matrix degradation in the patient, and preferably terminate, the self-perpetuating processes responsible for said degradation. A special embodiment of the method comprises administering of an effective matrix metallo-protease inhibiting amount of a compound of the present invention to a subject patient suffering from or susceptible to the development of rheumatoid arthritis, for a period of time sufficient to reduce or prevent, respectively, collagen and bone degradation in the joints of said patient, and preferably terminate, the self-perpetuating processes responsible for said degradation.
[00464] Toxicity and therapeutic efficacy of such compounds can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD50/ED50. Compounds that exhibit large therapeutic indices are preferred. While compounds that exhibit toxic side effects may be used, care should be taken to design a delivery system that targets such compounds to the site of affected tissue in order to minimize potential damage to uninfected cells and, thereby, reduce side effects.
[00465] The data obtained from the cell culture assays and animal studies can be used in formulating a range of dosage for use in humans. The dosage of such compounds lies preferably within a range of circulating concentrations that include the ED50 with little or no toxicity. The dosage may vary within this range depending upon the dosage form employed and the route of administration utilized. For any compound used in the method of the invention, the therapeutically effective dose can be estimated initially from cell culture assays. A dose may be formulated in animal models to achieve a circulating plasma concentration range that includes the IC50 (i.e., the concentration of the test compound which achieves a half-maximal inhibition of symptoms) as determined in cell culture. Such information can be used to more accurately determine useful doses in humans. Levels in plasma may be measured, for example, by high performance liquid chromatography.
[00466] A preferred therapeutically effective amount of the compound of the present invention to administer to a subject patient is about 0.1 mg/kg to about 10 mg/kg administered from once to three times a day. For example, an effective regimen of the present method may administer about 5 mg to about 1000 mg of said compound of the present invention from once to three times a day. It will be understood, however, that the specific dose level for any particular subject patient will depend upon a variety of factors including the age, body weight, general health, sex, diet, time of administration, route of administration, rate of excretion, drug combination and the severity of the particular inflammatory condition. A consideration of these factors is well within the purview of the ordinarily skilled clinician for the purpose of determining the therapeutically effective or prophylactically effective dosage amount needed to prevent, counter, or arrest the progress of the condition.
[00467] Compounds of the invention can be incorporated into pharmaceutical compositions suitable for administration. Such compositions typically comprise at least one compound of the invention and at least one pharmaceutically acceptable carrier. As used herein the language "pharmaceutically acceptable carrier" is intended to include solid carriers such as lactose, magnesium stearate, terra alba, sucrose, talc, stearic acid, gelatin, agar, pectin, acacia or the like; and liquids such as vegetable oils, arachis oil and sterile water, or the like, any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like, compatible with pharmaceutical administration. This listing of pharmaceutically acceptable carriers is not to be construed as limiting. The use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active compound, use thereof in the compositions is contemplated. Supplementary active compounds can also be incorporated into the compositions. [00468] A pharmaceutical composition of the invention is formulated to be compatible with its intended route of administration. Examples of routes of administration include parenteral, e.g., intravenous, intradermal, subcutaneous, oral (e.g., inhalation), transdermal (topical), transmucosal, and rectal administration. Solutions or suspensions used for parenteral, intradermal, or subcutaneous application can include the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose. The pH can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide. The parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic.
[00469] Pharmaceutical compositions suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion. For intravenous administration, suitable carriers include physiological saline, bacteriostatic water, Cremophor ELTM (BASF, Parsippany, NJ) or phosphate buffered saline (PBS). In all cases, the composition must be sterile and should be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyetheylene glycol, and the like), and suitable mixtures thereof. The proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. Prevention of the action of microorganisms can be achieved by various antibacterial and antifungal agents, for example, parabens, 'chlorobutanol, phenol, ascorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars, polyalcohols such as manitol, sorbitol, sodium chloride in the composition. Prolonged absorption of the injectable compositions can be brought about by including in the composition an agent which delays absorption, for example, aluminum mono stearate and gelatin.
[00470] Sterile injectable solutions can be prepared by incorporating the active compound
(e.g., a compound according to an embodiment of the invention) in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the active compound into a sterile vehicle which contains a basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum drying and freeze-drying which yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
[00471] Oral compositions generally include an inert diluent or an edible carrier. They can be enclosed in gelatin capsules or compressed into tablets. For the purpose of oral therapeutic administration, the active compound can be incorporated with excipients and used in the form of tablets, troches, or capsules. Oral compositions can also be prepared using a fluid carrier for use as a mouthwash, wherein the compound in the fluid carrier is applied orally and swished and expectorated or swallowed.
[00472] Pharmaceutically compatible binding agents, and/or adjuvant materials can be included as part of the composition. The tablets, pills, capsules, troches and the like can contain any of the following ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or corn starch; a lubricant such as magnesium stearate or Sterotes; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring.
[00473] For administration by inhalation, the compounds are delivered in the form of an aerosol spray from pressured container or dispenser which contains a suitable propellant, e.g., a gas such as carbon dioxide, or a nebulizer.
[00474] Systemic administration can also be by transmucosal or transdermal means. For transmucosal or transdermal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art, and include, for example, for transmucosal administration, detergents, bile salts, and fusidic acid derivatives. Transmucosal administration can be accomplished through the use of nasal sprays or suppositories. For transdermal administration, the active compounds are formulated into ointments, salves, gels, or creams as generally known in the art.
[00475] The compounds can also be prepared in the form of suppositories (e.g., with conventional suppository bases such as cocoa butter and other glycerides) or retention enemas for rectal delivery.
[00476] In one embodiment, the active compounds are prepared with carriers that will protect the compound against rapid elimination from the body, such as a controlled release formulation, including implants and microencapsulated delivery systems. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Methods for preparation of such formulations will be apparent to those skilled in the art. The materials can also be obtained commercially from Alza Corporation and Nova Pharmaceuticals, Inc. Liposomal suspensions (including liposomes targeted to infected cells with monoclonal antibodies to viral antigens) can also be used as pharmaceutically acceptable carriers. These can be prepared according to methods known to those skilled in the art.
[00477] It is especially advantageous to formulate oral or parenteral compositions in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the subject to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. The specification for the dosage unit forms of the invention are dictated by and directly dependent on the unique characteristics of the active compound and the particular therapeutic effect to be achieved, and the limitations inherent in the art of compounding such an active compound for the treatment of individuals.
[00478] The pharmaceutical compositions can be included in a container, pack, or dispenser together with instructions for administration. [00479] A compound according to an embodiment of the invention may be provided as a salt, preferably as a pharmaceutically acceptable salt of compounds of formula I. Examples of pharmaceutically acceptable salts of these compounds include those derived from organic acids such as acetic acid, malic acid, tartaric acid, citric acid, lactic acid, oxalic acid, succinic acid, fumaric acid, maleic acid, benzoic acid, salicylic acid, phenylacetic acid, mandelic acid, methanesulphonic acid, benzenesulphonic acid and />-toluenesulphonic acid, mineral acids such as hydrochloric and sulphuric acid and the like, giving methanesulphonate, benzenesulphonate, />-toluenesulphonate, hydrochloride and sulphate, and the like, respectively or those derived from bases such as organic and inorganic bases. Examples of suitable inorganic bases for the formation of salts of compounds for this invention include the hydroxides, carbonates, and bicarbonates of ammonia, lithium, sodium, calcium, potassium, aluminium, iron, magnesium, zinc and the like. Salts can also be formed with suitable organic bases. Such bases suitable for the formation of pharmaceutically acceptable base addition salts with compounds of the present invention include organic bases which are nontoxic and strong enough to form salts. Such organic bases are already well known in the art and may include amino acids such as arginine and lysine, mono-, di-, or trihydroxyalkylamines such as mono-, di-, and triethanolamine, choline, mono-, di-, and trialkylamines, such as methylamine, dimethylamine, and trimethylamine, guanidine; N-methylglucosamine; N-methylpiperazine; morpholine; ethylenediamine; N- benzylphenethylamine; tris(hydroxymethyl) aminomethane; and the like.
[00480] Salts of compounds according to an embodiment of the invention may be prepared in a conventional manner using methods well known in the art. Acid addition salts of said basic compounds may be prepared by dissolving the free base compounds according to the first or second aspects of the invention in aqueous or aqueous alcohol solution or other suitable solvents containing the required acid. Where a compound of the invention contains an acidic function, a base salt of said compound may be prepared by reacting said compound with a suitable base. The acid or base salt may separate directly or can be obtained by concentrating the solution e.g. by evaporation. The compounds of this invention may also exist in solvated or hydrated forms.
[00481] It will be appreciated by those skilled in the art that the foregoing description is exemplary and explanatory in nature, and is intended to illustrate the invention and its preferred embodiments. Through routine experimentation, an artisan will recognise apparent modifications and variations that may be made without departing from the spirit of the invention. Thus, the invention is intended to be defined not by the above description, but by the following claims and their equivalents. [00482] REFERENCES
Choy EH, Panayi GS. (2001). N Engl J Med. 344: 907-16.
Firestein GS. (2003). Nature. 423:356-61.
Smolen JS, Steiner G. (2003). Nat Rev Drug Discov. 2: 473-88.
Lee DM, Weinblatt ME (2001). Lancet. 358: 903-11. Kremer J. M., Westhovens R., Leon M., Di Giorgio E., Alten R., Steinfeld S., Russell A.,
Dougados M., Emery P., Nuamah I. F., Williams G. R., Becker J.-C, Hagerty D. T., Moreland
L. W. (2003) N Engl J Med. 349:1907-1915.
Edwards J. C.W., Szczepanski L., Szechinski J., Filipowicz-Sosnowska A., Emery P., Close
D. R., Stevens R. M., Shaw T. (2004) N Engl J Med. 350:2572-2581.
O1DeU JR, Leff R, Paulsen G, Haire C, Mallek J, Eckhoff PJ, Fernandez A, Blakely K, Wees
S, Stoner J, Hadley S, Felt J, Palmer W, Waytz P, Churchill M, Klassen L, Moore G. (2002)
Arthritis Rheum. 46:1164-70.
St Clair EW, van der Heijde DM, Smolen JS, Maini RN, Bathon JM, Emery P, Keystone E,
Schiff M, Kalden JR, Wang B, Dewoody K, Weiss R, Baker D; (2004) Combination of infliximab and methotrexate therapy for early rheumatoid arthritis: a randomized, controlled trial. Arthritis Rheum. 50 :3432-43.
Gomez-Reino JJ, et al. (2003). Arthritis Rheum. 48: 2122-7.
O'Dell JR. (2004) Therapeutic strategies for rheumatoid arthritis. N Engl J Med.
350(25):2591-602.
New L, Jiang Y, Han J. (2003) Regulation of PRAK subcellular location by p38 MAP kinases.
MoI Biol Cell. 14(6):2603-16.
Shi Y, Kotlyarov A, Laabeta K, Gruber AD, Butt E, Marcus K, Meyer HE, Friedrich A, VoIk
HD, Gaestel M. (2003) Elimination of protein kinase MK5/PRAK activity by targeted homologous recombination. MoI Cell Biol. 23:7732-41.
Seternes OM, Mikalsen T, Johansen B, Michaelsen E, Armstrong CG, Morrice NA, Turgeon
B, Meloche S, Moens U, Keyse SM. (2004) Activation of MK5/PRAK by the atypical MAP kinase ERK3 defines a novel signal transduction pathway.
EMBO J. 23:4780-91.
Andreakos E, et al. (2003). Arthritis Rheum. 48: 1901-12.
Cunnane G, et al. (2001). Arthritis Rheum 44: 2263-74.
Coussens LM, et al. (2002). Science 295: 2387-92.
Creemers EE, et al. (2001). Circ Res. 2001 89:201-10
Gapski R, et al. (2004). J Periodontal. 75:441-52.
Reif S, Somech R, Brazovski E, Reich R, Belson A, Konikoff FM, Kessler A. (2005)
Digestion. 71 :124-130.
Rosenberg GA. (2002). Glia. 39:279-91.
Schanstra JP, et al. (2002). J Clin Invest. 110:371-9.
Suzuki R, et al. (2004). Treat Respir Med. 3:17-27.
[00483] From the foregoing description, various modifications and changes in the compositions and methods of this invention will occur to those skilled in the art. All such modifications coming within the scope of the appended claims are intended to be included therein. [00484] It should be understood that factors such as the differential cell penetration capacity of the various compounds can contribute to discrepancies between the activity of the compounds in the in vitro biochemical and cellular assays.
[00485] All publications, including but not limited to patents and patent applications, cited in this specification are herein incorporated by reference as if each individual publication were specifically and individually indicated to be incorporated by reference herein as though fully set forth. [00486] The chemical names of compounds of the invention given in this application are generated using MDL's ISIS Draw Autonom Software tool and are not verified. Preferably, in the event of inconsistency, the depicted structure governs.

Claims

WHAT IS CLAIMED IS:
1. A compound according to formula III:
Figure imgf000166_0001
(III) wherein
R1 is H, or substituted or unsubstituted alkyl; and each of R8 and R9 is independently selected from substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl; or a pharmaceutically acceptable salt, solvate or prodrug thereof; and stereoisomers, isotopic variants and tautomers thereof.
2. A compound according to Claim 1, wherein R8 is selected from substituted or unsubstituted cyclopentyl, cyclohexyl, substituted or unsubstituted phenyl, substituted or unsubstituted pyridyl, substituted or unsubstituted pyrimidine, and substituted or unsubstituted pyrazine, substituted or unsubstituted pyrrole, substituted or unsubstituted pyrazole and substituted or unsubstituted imidazole.
3. A compound according to Claim 1 , wherein R8 is selected from substituted phenyl, substituted pyridyl, and substituted pyrimidine; and the substitution is
-L-R8d; and wherein
L is selected from bond, alkylene, heteroalkylene, -O-, -N(R8e)-, -CO-, -CO2-, -SO-, -SO2-, -C0N(R8e>, -SO2N(R86)-, -N(R8e)C0-, -N(R8e)SO2-, -N(R8e)C0 N(R8e>, -N(R8e)SO2 N(R8e)-; and
R8d is selected from substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted heteroaryl, substituted or unsubstituted amino, substituted or unsubstituted aralkyl, substituted or unsubstituted heteroarylalkyl and substituted or unsubstituted aminoalkyl; and
R8e is selected from H, substituted or unsubstituted alkyl and substituted or unsubstituted cycloalkyl.
4. A compound according to Claim 1 , wherein R8 is
Figure imgf000167_0001
wherein L, and R8d are as in claim 3; the subscript n is selected from 1-4; and each R8a is independently selected from hydrogen, substituted or unsubstituted alkyl, alkoxy, cyano, and halo.
5. A compound according to Claim 4, wherein L is a bond, -O-, -CO-, -CON(R8e)-, or -N(R8e)CO-
R8 is selected from substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted heteroaryl, substituted or unsubstituted aralkyl, substituted or unsubstituted heteroarylalkyl and substituted or unsubstituted aminoalkyl; and R8e is selected from H, substituted or unsubstituted alkyl.
6. A compound according to Claim 4, wherein L is a bond, -0-, -CO-, -CON(R8e)-, or -N(R8e)CO- ; and
R8d is selected from H, alkylaminoethyl, dialkylaminoethyl, cycloalkyl, heterocycloalkyl, arylalkyl, and heteroarylalkyl.
7. A compound according to Claim 4, wherein L is a bond, -0-, -CO-, -CON(R8e)-, or -N(R8e)CO- ; and
R8d is selected from H, methylaminoethyl, ethylaminoethyl, dimethylaminoethyl, diethylaminoethyl, substituted or unsubstituted pyrrolidinyl, benzyl and pyridylmethyl.
8. A compound according to Claim 4, wherein L is bond, -CO-, -O(CH2)mi-, -CON(H)(CH2)mi-, or - NHCO-; the subscript ml is selected from 1 -4; and R8d is
Figure imgf000167_0002
and wherein the ring P is substituted or unsubstituted heterocycloalkyl.
9. A compound according to formula IVa, IVTJ, IVC, or IVd:
Figure imgf000167_0003
IVa IVb IVc or IVd and wherein L and the ring P are as in claim 8; the subscript n, is selected from 1-4; each R a is independently selected from hydrogen, substituted or unsubstituted alkyl, alkoxy, cyano, and halo; and R9 is independently selected from substituted or unsubstituted aryl and heteroaryl; or a pharmaceutically acceptable salt, solvate or prodrug thereof; and stereoisomers, isotopic variants and tautomers thereof.
10. A compound according to Claim 9, wherein L is a bond.
11. A compound according to Claim 9, wherein L is -CO-.
12. A compound according to Claim 9, wherein L is -NHCO- or -CONH-.
13. A compound according to Claim 9, wherein L is -CON(H)-CH2-CH2-, or - -N(H)-CO-CH2-
CH2-.
14. A compound according to Claim 9, wherein L is -OCH2-CH2- or -NHCH2-CH2-.
15. A compound according to any one of claims 9-14, wherein the ring P is substituted or unsubstituted piperidine, morpholine or piperazine.
16. A compound according to Claim 9, wherein each R8a is H.
17. A compound according to Claim 9, wherein subscript n is 1 and R8a is Me, Et, Pr, iso-Pr, Cl, F,
CN, OMe, or CF3.
18. A compound according to Claim 1, wherein the compound is according to formula Va, Vb, VC,
Vd, Ve, or Vf:
Figure imgf000168_0001
Va Vb Vc
Figure imgf000168_0002
Vd Ve or Vf and wherein R9 is as in Claim 1 and R8b is hydrogen, substituted or unsubstituted alkyl or substituted or unsubstituted cycloalkyl.
19. A compound according to claim 18, wherein R8b is H.
20. A compound according to claim 18, wherein R8 is substituted or unsubstiuted alkyl or substituted or unsubstituted cycloalkyl.
21. A compound according to claim 18, wherein R8b is Me, Et, Pr, i-Pr, t-Bu, i-Bu, CH2CONH2, cyclopropyl or cyclopropylmethyl.
22. A compound according to Claim 1, wherein R9 is selected from substituted or unsubstituted aryl.
23. A compound according to Claim 1, wherein R9 is selected from substituted or unsubstituted phenyl.
24. A compound according to Claim 1, wherein R9 is selected from substituted or unsubstituted heteroaryl.
25. A compound according to Claim 1, wherein R9 is selected from substituted or unsubstituted phenyl, pyridyl, indolyl, isoindolyl, pyrrolyl, furanyl, thienyl, pyrazolyl, oxazolyl, and thiazolyl.
26. A compound according to any one of claims 1 -25, wherein R9 is
Figure imgf000169_0001
and each of A1, A2 and A3 is independently selected from S, O, N, NR9a, and CR9a; each of R9a is independently H or substituted or unsubstituted alkyl; and R9b is CONH2, CONHMe, or CN.
27. A compound according to any one of claims 1 -25, wherein R9 is
Figure imgf000169_0002
28. A compound according to any one of claims 1 -25, wherein R9 is
Figure imgf000170_0001
Figure imgf000170_0002
29. A compound according to any one of claims 1 -25, wherein R9 is
Figure imgf000170_0003
and wherein the subscript m is selected from 1 -4 and each R9d is independently H, substituted or unsubstituted alkyl or halo.
30. A compound according to any one of claims 1 -25, wherein R9 is
Figure imgf000170_0004
and wherein the subscript m is selected from 1 -4 and each R9d is independently H, substituted or unsubstituted alkyl or halo.
31. A compound according to any one of claims 1 -25, wherein R9 is
Figure imgf000170_0005
and wherein the subscript m is selected from 1 -3 and each R9d is independently H, substituted or unsubstituted alkyl or halo.
32. A compound according to any one of claims 29-31, wherein each R9d is H.
33. A compound according to any one of claims 29-31, wherein m is 1 or 2 and each R9d is Me, Cl or F.
34. A compound according to Claim 1, wherein the compound is according to formula Via, VIb,
VIc, VId, VIe or VIf:
Figure imgf000171_0001
Via
VIb VIc
Figure imgf000171_0002
and R8b is hydrogen, substituted or unsubstituted alkyl or substituted or unsubstituted cycloalkyl.
35. A compound according to Claim 1, wherein the compound is according to formula Vila, VIIb, VIIc, VIIc, VIId, VIIe or VIIf:
Figure imgf000172_0001
and R8b is hydrogen, substituted or unsubstituted alkyl or substituted or unsubstituted cycloalkyl.
36. A compound according to Claim 1, wherein the compound is according to formula Villa, VIIIb, VIIIc, VIIId, VIIIe or VIIIf:
Figure imgf000173_0001
VIIIc
Figure imgf000173_0002
Figure imgf000173_0003
and R8b is hydrogen, substituted or unsubstituted alkyl or substituted or unsubstituted cycloalkyl.
37. A compound according to Claim 1, wherein the compound is according to formula IXa, IXb, IXc, IXd, IXe, or IXf:
Figure imgf000174_0001
IXa
IXb IXc
Figure imgf000174_0002
and R8b is hydrogen, substituted or unsubstituted alkyl or substituted or unsubstituted cycloalkyl
38. A compound according to Claim 1 , wherein the compound is according to formula Xa, Xb, Xc, Xd, Xe, or Xf:
Figure imgf000175_0001
Xa
Xb
Figure imgf000175_0002
and R8b is hydrogen, substituted or unsubstituted alkyl or substituted or unsubstituted cycloalkyl.
39. A compound according to Claim 1, wherein the compound is according to formula XIa, XIb, XIc, XId, XIe or XIf:
Figure imgf000176_0001
XIa
XIb XIc
Figure imgf000176_0002
XId XIe XIf and R8b is hydrogen, substituted or unsubstituted alkyl or substituted or unsubstituted cycloalkyl; and R9e is hydrogen, Me, or CN.
40. A compound according to any one of claims 34-39, wherein R8b is H.
41. A compound according to any one of claims 34-39, wherein R8b is cycloalkyl.
42. A compound according to any one of claims 34-39, wherein R8b is cyclopropyl.
43. A compound according to any one of claims 34-39, wherein R8b is substituted or unsubstituted alkyl.
44. A compound according to any one of claims 34-39, wherein R8b is Me, Et, Pr, i-Pr, t-Bu, i-Bu, CF3, CH2CF3, CH2CONH2, or cyclopropylmethyl.
45. A compound according to Claim 1, wherein the compound is according to formula XIIa, XIIb, XIIc or XIId:
Figure imgf000177_0001
XIIa
XIIb XIIc XIId
46. A compound according to Claim 1, wherein the compound is according to formula XIIIa, XIIIb, XIIIc or XIIId:
Figure imgf000177_0002
XIIIa
XIIIb XIIIc XIIId
47. A compound according to Claim 1, wherein the compound is according to formula XIVa, XIVb, XIVc or XIVd:
Figure imgf000177_0003
XIVa
XIVb XIVc XIVd
48. A compound according to Claim 1, wherein the compound is according to formula XVa, XVb, or XIVc:
Figure imgf000178_0001
XVc
XVa XVb
and L is a bond, -CO-, or -0-CH2-CH2- ; the ring P is
Figure imgf000178_0002
and R8b is H, Me, i-Pr, t-Bu, CH2CONH2, cyclopropylmethyl, or CH2CF3.
49. A compound according to Claim 48, wherein L is a bond and the ring P is
Figure imgf000178_0003
50. A compound according to claim 1 wherein the compound is selected from
(4-Moφholin-4-yl-phenyl)-[5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5-α]pyrazin-8-yl]-amine;
[4-(4-Methyl-piperazin-l-yl)-phenyl]-[5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5-α]pyrazin-8-yl]- amine;
4-{8-[4-(4-Methyl-piperazin-l-yl)-phenylamino]-[l,2,4]triazolo[l,5-α]pyrazin-5-yl}- benzamide;
4-{ 8-[4-(4-Methyl-piperazin- 1 -yl)-phenylamino] -[ 1 ,2,4]triazolo[ 1 ,5-α]pyrazin-5-yl} -IH- pyridin-2-one;
4-[8-(4-Moφholin-4-yl-phenylamino)-[l,2,4]triazolo[l,5-α]pyrazin-5-yl]-benzamide;
4-{8-[4-(4-Methyl-piperazin-l-yl)-phenylamino]-[l,2,4]triazolo[l,5-α]pyrazin-5-yl}- thiophene-2-carboxylic acid amide;
2-Fluoro-4-{8-[4-(4-methyl-piperazin-l-yl)-phenylamino]-[l,2,4]triazolo[l,5-α]pyrazin-5-yl}- benzamide;
3-Fluoro-4-{8-[4-(4-methyl-piperazin-l-yl)-phenylamino]-[l,2,4]triazolo[l,5-α]pyrazin-5-yl}- benzamide; 5 - { 8- [4-(4-Methyl-piperazin- 1 -yl)-phenylamino] -[ 1 ,2,4]triazolo[ 1 ,5 -α]pyrazin-5 -yl} - thiophene-2-carboxylic acid amide;
3 -Fluoro-4- [8 -(4-morpholin-4-yl-phenylamino)- [ 1 ,2,4]triazolo [1,5 -α]pyrazin-5 -yl] -benzamide;
2-Fluoro-4- [8 -(4-morpholin-4-yl-phenylamino)- [ 1 ,2,4]triazolo [ 1 ,5 -α]pyrazin-5 -yl] -benzamide;
[5-(5-Methyl-lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5-α]pyrazin-8-yl]-(4-moφholin-4-yl-phenyl)- amine;
[5-(5-Methyl-lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5-α]pyrazin-8-yl]-(4-moφholin-4-yl-phenyl)- amine;
5-[8-(4-Moφholin-4-yl-phenylamino)-[l,2,4]triazolo[l,5-α]pyrazin-5-yl]-thiophene-2- carboxylic acid amide;
2,6-Difluoro-4-[8-(4-moφholin-4-yl-phenylamino)-[l,2,4]triazolo[l,5-α]pyrazin-5-yl]- benzamide;
4-[8-(4-Moφholin-4-yl-phenylamino)-[l,2,4]triazolo[l,5-α]pyrazin-5-yl]-thiophene-2- carboxylic acid amide;
[4-(4-Methyl-piperazin-l-yl)-phenyl]-[5-(5-methyl-lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5- α]pyrazin-8 -yl] -amine;
(3-Fluoro-4-morpholin-4-yl-phenyl)-[5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5-α]pyrazin-8-yl]- amine;
(3-Chloro-4-moφholin-4-yl-phenyl)-[5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5-α]pyrazin-8-yl]- amine;
[4-(2-Moφholin-4-yl-ethoxy)-phenyl]-[5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5-α]pyrazin-8- yl]-amine;
4-{8-[4-(2-Moφholin-4-yl-ethoxy)-phenylamino]-[l,2,4]triazolo[l,5-α]pyrazin-5-yl}- thiophene-2-carboxylic acid amide;
5 -{ 8-[4-(2-Moφholin-4-yl-ethoxy)-phenylamino] -[1 ,2,4]triazolo[ 1 ,5 -α]pyrazin-5 -yl} - thiophene-2-carboxylic acid amide;
(4-Morpholin-4-yl-phenyl)-[5-(2H-pyrazol-3-yl)-[l,2,4]triazolo[l,5-α]pyrazin-8-yl]-amine;
4-{8-[4-((2R,6S)-2,6-Dimethyl-moφholin-4-yl)-phenylamino]-[l,2,4]triazolo[l,5-α]pyrazin-5- yl} -thiophene-2-carboxylic acid amide;
(4-Morpholin-4-yl-3-trifluoromethyl-phenyl)-[5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5- α]pyrazin-8 -yl] -amine;
2,6-Difluoro-4-{8-[4-(4-isopropyl-piperazin-l-yl)-phenylamino]-[l,2,4]triazolo[l,5-α]pyrazin-
5 -yl} -benzamide;
4-[8-(4-Piperazin-l-yl-phenylamino)-[l,2,4]triazolo[l,5-α]pyrazin-5-yl]-thiophene-2- carboxylic acid amide; 4- { 8- [4-(4-Isopropyl-piperazin- 1 -yl)-phenylamino]- [ 1 ,2,4]triazolo [ 1 ,5 -α]pyrazin-5 -yl} - thiophene-2-carboxylic acid amide;
5-[8-(4-Moφholin-4-yl-phenylamino)-[l,2,4]triazolo[l,5-α]pyrazin-5-yl]-furan-3-carboxylic acid amide;
5 - { 8- [4-(4-Isopropyl-piperazin- 1 -yl)-phenylamino]- [ 1 ,2,4]triazolo[ 1 ,5 -α]pyrazin-5 -yl} -furan-
3-carboxylic acid amide;
[5-(lH-Pyrazol-4-yl)-[l,2,4]1riazolo[l,5-α]pyrazin-8-yl]-[4-((2S,5R)-2,4,5-trimetliyl-piperazin-
1 -yl)-ρhenyl] -amine;
4-{8-[4-((2S,5R)-2,4,5-Trimethyl-piperazin-l-yl)-phenylamino]-[l,2,4]triazolo[l,5-α]pyrazin-
5 -yl} -thiophene-2-carboxylic acid amide;
5-[8-(4-Moφholin-4-yl-phenylamino)-[l,2,4]triazolo[l,5-α]pyrazin-5-yl]-2,3-dihydro-isoindol-
1 -one;
4-[8-(4-Moφholin-4-yl-phenylamino)-[l,2,4]triazolo[l,5-α]pyrazin-5-yl]-furan-2-carboxylic acid amide;
6-[8-(4-Moφholin-4-yl-phenylamino)-[l,2,4]triazolo[l,5-α]pyrazin-5-yl]-3,4-dihydro-2H- isoquinolin- 1 -one;
5-{8-[4-(4-Isopropyl-piperazin-l-yl)-phenylamino]-[l,2,4]triazolo[l,5-α]pyrazin-5-yl}-2,3- dihydro-isoindol- 1 -one;
{2-Moφholin-4-yl-5-[5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5-α]pyrazin-8-ylamino]-phenyl}- methanol;
[4-(4-Isopropyl-piperazin-l-yl)-phenyl]-[5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5-α]pyrazin-8- yl]-amine;
6-[8-(4-Moφholin-4-yl-phenylamino)-[l,2,4]triazolo[l,5-α]pyrazin-5-yl]-3,4-dihydro-lH- quinolin-2-one;
(4-Piperazin-l-yl-phenyl)-[5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5-α]pyrazin-8-yl]-amine;
(6-Moφholin-4-yl-pyridin-3-yl)-[5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5-α]pyrazin-8-yl]- amine;
[6-(4-Cyclopropylmethyl-piperazin-l-yl)-pyridin-3-yl]-[5-(lH-pyrazol-4-yl)-
[ 1 ,2,4]triazolo[ 1 ,5-α]pyrazin-8-yl]-amine;
[6-(4-Isopropyl-piperazin-l-yl)-pyridin-3-yl]-[5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5- α]pyrazin-8 -yl] -amine;
[5-(lH-Pyrazol-4-yl)-[l,2,4]triazolo[l,5-α]pyrazin-8-yl]-{6-[4-(2,2,2-trifluoro-ethyl)-piperazin-
1 -yl] -pyridin-3 -yl} -amine;
[5-(lH-Pyrazol-4-yl)-[l,2,4]triazolo[l,5-α]pyrazin-8-yl]-{4-[4-(2,2,2-trifluoro-ethyl)-piperazin-
1 -yl] -phenyl} -amine; [4-(4-Cyclopropylmethyl-piperazin- 1 -yl)-phenyl]- [5 -( lH-pyrazol-4-yl)- [ 1 ,2,4]triazolo[ 1 ,5 - α]pyrazin-8 -yl] -amine;
4-[8-(6-Moφholin-4-yl-pyridin-3-ylamino)-[l,2,4]triazolo[l,5-α]pyrazin-5-yl]-thiophene-2- carboxylic acid amide;
(5-Benzo[#b!]thiophen-3-yl-[l,2,4]triazolo[l,5-α]pyrazin-8-yl)-(4-moφholin-4-yl-phenyl)- amine;
(5-Benzo[#b!]thiophen-3-yl-[l,2,4]triazolo[l,5-α]pyrazin-8-yl)-[4-(4-isopropyl-piperazin-l-yl)- phenyl] -amine;
(4-Moφholin-4-yl-phenyl)-(5-thiophen-3-yl-[l,2,4]triazolo[l,5-α]pyrazin-8-yl)-amine;
[4-(4-Isopropyl-piperazin-l-yl)-phenyl]-(5-thiophen-3-yl-[l,2,4]triazolo[l,5-α]pyrazin-8-yl)- amine;
[5 -(5 -Ethyl- lH-pyrazol-4-yl)- [ 1 ,2,4]triazolo [ 1 ,5 -α]pyrazin-8 -yl] -(4-moφholin-4-yl-phenyl)- amine;
6-[8-(4-Moφholin-4-yl-phenylamino)-[l,2,4]triazolo[l,5-α]pyrazin-5-yl]-l,l-dioxo-l,2- dihydro-l$l%6&-benzo[#d!]isothiazol-3-one;
4-{8-[6-(4-Cyclopropylmethyl-piperazin-l-yl)-pyridin-3-ylamino]-[l,2,4]triazolo[l,5- α]pyrazin-5-yl}-thiophene-2-carboxylic acid amide;
4-{8-[6-(4-Isopropyl-piperazin-l-yl)-pyridin-3-ylamino]-[l,2,4]triazolo[l,5-α]pyrazin-5-yl}- thiophene-2-carboxylic acid amide;
4-(8-{6-[4-(2,2,2-Trifluoro-ethyl)-piperazin-l-yl]-pyridin-3-ylamino}-[l,2,4]triazolo[l,5- α]pyrazin-5-yl)-thiophene-2-carboxylic acid amide;
4-(8- {4-[4-(2,2,2-Trifluoro-ethyl)-piperazin-l -yl]-phenylamino} -[ 1 ,2,4]triazolo[ 1 ,5 -α]pyrazin-
5-yl)-thiophene-2-carboxylic acid amide;
4-{8-[4-(4-Cyclopropylmethyl-piperazin-l-yl)-phenylamino]-[l,2,4]triazolo[l,5-α]pyrazin-5- yl}-thiophene-2-carboxylic acid amide;
[4-(4-Cyclopropyl-piperazin-l-yl)-phenyl]-[5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5-α]pyrazin-
8 -yl] -amine;
[6-(4-Cyclopropyl-piperazin- 1 -yl)-pyridin-3 -yl] - [5 -( lH-pyrazol-4-yl)- [ 1 ,2,4]triazolo[ 1 ,5 - α]pyrazin-8 -yl] -amine;
4-[8-(4-Moφholin-4-yl-phenylamino)-[l,2,4]triazolo[l,5-α]pyrazin-5-yl]-thiazole-2-carboxylic acid amide;
4-{8-[4-(4-Isopropyl-piperazin-l-yl)-phenylamino]-[l,2,4]triazolo[l,5-α]pyrazin-5-yl}- thiazole-2-carboxylic acid amide;
4-(8-{4-[l-(2,2,2-Trifluoro-ethyl)-piperidin-4-yl]-phenylamino}-[l,2,4]triazolo[l,5-α]pyrazin-
5-yl)-thiophene-2-carboxylic acid amide; [5-(lH-Pyrazol-4-yl)-[l,2,4]triazolo[l,5-α]pyrazin-8-yl]-{4-[l-(2,2,2-trifluoro-ethyl)-piperidin-
4-yl]-phenyl} -amine;
5 -{ 8-[4-(2-Moφholin-4-yl-ethoxy)-phenylamino] -[1 ,2,4]triazolo[ 1 ,5 -α]pyrazin-5 -yl} -2,3 - dihydro-isoindol- 1 -one;
(5 -Benzothiazol-6-yl- [ 1 ,2,4]triazolo [ 1 ,5 -α]pyrazin-8 -yl)-(4-moφholin-4-yl-phenyl)-amine;
(2-Fluoro-4-morpholin-4-yl-phenyl)-[5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5-α]pyrazin-8-yl]- amine;
(2-Chloro-4-morpholin-4-yl-phenyl)-[5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5-α]pyrazin-8-yl]- amine;
1 - { 5 - [8 -(4-Moφholin-4-yl-phenylamino)- [ 1 ,2,4]triazolo [ 1 ,5 -α]pyrazin-5 -yl] -thiophen-2-yl} - ethanone;
{4-[8-(4-Moφholin-4-yl-phenylamino)-[l,2,4]triazolo[l,5-α]pyrazin-5-yl]-2H-pyrazol-3-yl}- methanol;
6-{4-[8-(4-Moφholin-4-yl-phenylamino)-[l,2,4]triazolo[l,5-α]pyrazin-5-yl]-phenyl}-4,5- dihydro-2H-pyridazin-3 -one;
(5-Benzo[l,2,5]oxadiazol-5-yl-[l,2,4]triazolo[l,5-α]pyrazin-8-yl)-(4-moφholin-4-yl-phenyl)- amine;
4-[8-(4-Moφholin-4-yl-phenylamino)-[l,2,4]triazolo[l,5-α]pyrazin-5-yl]-thiazole-2-carboxylic acid methylamide;
4-{8-[4-(4-Isopropyl-piperazin-l-yl)-phenylamino]-[l,2,4]triazolo[l,5-α]pyrazin-5-yl}- thiazole-2-carboxylic acid methylamide;
5-[8-(2-Fluoro-4-moφholin-4-yl-phenylamino)-[l,2,4]triazolo[l,5-α]pyrazin-5-yl]-2,3- dihydro-isoindol- 1 -one;
5-[8-(2-Chloro-4-moφholin-4-yl-phenylamino)-[l,2,4]triazolo[l,5-α]pyrazin-5-yl]-2,3- dihydro-isoindol- 1 -one;
5-{8-[2-Chloro-4-(4-isopropyl-piperazin-l-yl)-phenylamino]-[l,2,4]triazolo[l,5-α]pyrazin-5- yl} -2,3 -dihydro-isoindol- 1 -one;
[5 -(2-Amino-pyrimidin-5 -yl)- [ 1 ,2,4]triazolo [ 1 ,5 -α]pyrazin-8 -yl] -(4-moφholin-4-yl-phenyl)- amine;
5-{8-[6-(4-Isopropyl-piperazin-l-yl)-pyridin-3-ylamino]-[l,2,4]triazolo[l,5-α]pyrazin-5-yl}-
2,3-dihydro-isoindol-l-one;
3-[8-(4-Moφholin-4-yl-phenylamino)-[l,2,4]triazolo[l,5-α]pyrazin-5-yl]-benzo[#b!]thiophene-
7-carboxylic acid amide;
3-{8-[4-(4-Isopropyl-piperazin-l-yl)-phenylamino]-[l,2,4]triazolo[l,5-α]pyrazin-5-yl}- benzo[#b!]thiophene-7-carboxylic acid amide; {4-[8-(4-Moφholin-4-yl-phenylamino)-[l,2,4]triazolo[l,5-α]pyrazin-5-yl]-pyridin-2-yl}- methanol;
(4- { 8 -[4-(4-Isopropyl-piperazin- 1 -yl)-phenylamino] - [ 1 ,2,4]triazolo [ 1 ,5 -α]pyrazin-5 -yl} - pyridin-2-yl)-methanol;
[4-(l-Isopropyl-piperidin-4-yl)-phenyl]-[5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5-α]pyrazin-8- yl]-amine;
{5-[4-(2-Amino-thiazol-4-yl)-phenyl]-[l,2,4]triazolo[l,5-α]pyrazin-8-yl}-(4-moφholin-4-yl- phenyl)-amine;
4-{8-[6-(4-Isopropyl-piperazin-l-yl)-pyridin-3-ylamino]-[l,2,4]triazolo[l,5-α]pyrazin-5-yl}- furan-2-carboxylic acid amide;
5 -[8-(6-Morpholin-4-yl-pyridin-3 -ylamino)- [ 1 ,2,4]triazolo [ 1 ,5 -α]pyrazin-5 -yl] -2,3 -dihydro- isoindol-1-one;
4-{8-[4-(4-Isopropyl-piperazin-l-yl)-phenylamino]-[l,2,4]triazolo[l,5-α]pyrazin-5-yl}-furan-
2-carboxylic acid amide;
4-{8-[4-(l-Isopropyl-piperidin-4-yl)-phenylamino]-[l,2,4]triazolo[l,5-α]pyrazin-5-yl}- thiophene-2-carboxylic acid amide;
(4-{8-[6-(4-Isopropyl-piperazin-l-yl)-pyridin-3-ylamino]-[l,2,4]triazolo[l,5-α]pyrazin-5-yl}- pyridin-2-yl)-methanol;
[5-(2-Fluoromethyl-pyridin-4-yl)-[l,2,4]triazolo[l,5-α]pyrazin-8-yl]-[4-(4-isopropyl-piperazin-
1 -yl)-ρhenyl] -amine;
5-{8-[4-(l-Isopropyl-piperidin-4-yl)-phenylamino]-[l,2,4]triazolo[l,5-α]pyrazin-5-yl}-2,3- dihydro-isoindol- 1 -one;
[5-(lH-Indazol-6-yl)-[l,2,4]triazolo[l,5-α]pyrazin-8-yl]-(4-moφholin-4-yl-phenyl)-amine;
4-[8-(6-Moφholin-4-yl-pyridin-3-ylamino)-[l,2,4]triazolo[l,5-α]pyrazin-5-yl]-furan-2- carboxylic acid amide;
4-{8-[4-(4-Isopropyl-piperazin-l-yl)-phenylamino]-[l,2,4]triazolo[l,5-α]pyrazin-5-yl}-5- methyl-thiophene-2-carboxylic acid amide;
4-{8-[6-(4-Isopropyl-piperazin-l-yl)-pyridin-3-ylamino]-[l,2,4]triazolo[l,5-α]pyrazin-5-yl}-5- methyl-thiophene-2-carboxylic acid amide;
4-{8-[4-(4-Isopropyl-piperazin-l-yl)-phenylamino]-[l,2,4]triazolo[l,5-α]pyrazin-5-yl}-2,3- dihydro-isoindol- 1 -one;
4-{8-[6-(4-Isopropyl-piperazin-l-yl)-pyridin-3-ylamino]-[l,2,4]triazolo[l,5-α]pyrazin-5-yl}-
2,3-dihydro-isoindol-l-one;
5 -[8-(2-Moφholin-4-yl-pyrimidin-5 -ylamino)- [ 1 ,2,4]triazolo[ 1 ,5 -α]pyrazin-5-yl]-2,3 -dihydro- isoindol-1-one; l-{4-[5-(lH-Pyrazol-4-yl)-[l,2,4]triazolo[l,5-α]pyrazin-8-ylamino]-phenyl}-piperazin-2-one; 5 - { 8- [2-(4-Isopropyl-piperazin- 1 -yl)-pyrimidin-5 -ylamino] -[ 1 ,2,4]triazolo[ 1 ,5 -α]pyrazin-5 -yl} - 2,3-dihydro-isoindol-l-one;
5 - { 8- [4-(4-#tert !-Butyl-piperazin- 1 -yl)-phenylamino] - [ 1 ,2,4]triazolo [ 1 ,5 -α]pyrazin-5 -yl} -2,3 - dihydro-isoindol- 1 -one;
[4-(4-#tert ! -Butyl-piperazin- 1 -yl)-phenyl] - [5 -( lH-pyrazol-4-yl)- [ 1 ,2,4]triazolo[ 1 ,5 -α]pyrazin- 8 -yl] -amine;
[5 -( lH-Indazol-5 -yl)- [ 1 ,2,4]triazolo[ 1 ,5 -α]pyrazin-8 -yl] -(4-moφholin-4-yl-phenyl)-amine; 5 -{ 8-[4-(2-Oxo-piperazin- 1 -yl)-phenylamino]- [ 1 ,2,4]triazolo[ 1 ,5 -α]pyrazin-5-yl} -2,3 -dihydro- isoindol- 1 -one;
[5-(lH-Indazol-6-yl)-[l,2,4]triazolo[l,5-α]pyrazin-8-yl]-[4-(4-isopropyl-piperazin-l-yl)- phenyl] -amine;
[6-(4-Isopropyl-piperazin-l-yl)-pyridin-3-yl]-(5-{(E)-l-[4-methylene-2,4-dihydro-pyrazol- (3E)-ylidenemethyl]-propenyl}-[l,2,4]triazolo[l,5-α]pyrazin-8-yl)-amine; 7-Fluoro-5-{8-[4-(4-isopropyl-piperazin-l-yl)-phenylamino]-[l,2,4]triazolo[l,5-α]pyrazin-5- yl} -2,3 -dihydro-isoindol- 1 -one;
6-{8-[4-(4-Isopropyl-piperazin-l-yl)-phenylamino]-[l,2,4]triazolo[l,5-α]pyrazin-5-yl}-2,3- dihydro-isoindol- 1 -one;
(3 -Methylaminomethyl-4-morpholin-4-yl-phenyl)- [5 -( lH-pyrazol-4-yl)- [ 1 ,2,4]triazolo[ 1 ,5 - α]pyrazin-8 -yl] -amine;
2-[8-(4-Moφholin-4-yl-phenylamino)-[l,2,4]triazolo[l,5-α]pyrazin-5-yl]-5,6-dihydro-furo[2,3- #c!]pyrrol-4-one;
5-{8-[4-(4-Isopropyl-2-oxo-piperazin-l-yl)-phenylamino]-[l,2,4]triazolo[l,5-α]pyrazin-5-yl}- 2,3-dihydro-isoindol-l-one;
2-[8-(4-Moφholin-4-yl-phenylamino)-[l,2,4]triazolo[l,5-α]pyrazin-5-yl]-4,5-dihydro- thieno[2,3-#c!]pyrrol-6-one;
5-{8-[4-(4-Isopropyl-piperazin-l-yl)-phenylamino]-[l,2,4]triazolo[l,5-α]pyrazin-5-yl}-3,3- dimethyl-2,3 -dihydro-isoindol- 1 -one;
5-(8-Cyclohexylamino-[l,2,4]triazolo[l,5-α]pyrazin-5-yl)-2,3-dihydro-isoindol-l-one; 5-[8-(Tetrahydro-pyran-4-ylamino)-[l,2,4]triazolo[l,5-α]pyrazin-5-yl]-2,3-dihydro-isoindol-l- one; and
5-[8-(4-Fluoro-phenylamino)-[l,2,4]triazolo[l,5-α]pyrazin-5-yl]-2,3-dihydro-isoindol-l-one.
51. A compound according to claim 1 wherein the compound is selected from:
4-[5-(lH-Pyrazol-4-yl)-[l,2,4]triazolo[l,5-α]pyrazin-8-ylamino]-N-pyridin-3-ylmethyl- benzamide;
4- [5 -(2-Oxo- 1 ,2-dihydro-pyridin-4-yl)- [ 1 ,2,4]triazolo [ 1 ,5 -α]pyrazin-8 -ylamino] -N-pyridin-3 - ylmethyl-benzamide; 2-Methoxy-N-(6-methyl-pyridin-3 -ylmethyl)-4- [5 -( lH-pyrazol-4-yl)- [ 1 ,2,4]triazolo[ 1 ,5 - α]pyrazin-8-ylamino] -benzamide;
4-(8-{3-Methoxy-4-[(6-methyl-pyridin-3-ylmethyl)-carbamoyl]-phenylamino}-
[l,2,4]triazolo[l,5-α]pyrazin-5-yl)-thiophene-2-carboxylic acid amide;
N-Benzyl-2-methoxy-4-[5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5-α]pyrazin-8-ylamino]- benzamide;
N-Benzyl-3-[5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5-α]pyrazin-8-ylamino]-benzamide;
3-[5-(lH-Pyrazol-4-yl)-[l,2,4]triazolo[l,5-α]pyrazin-8-ylamino]-N-pyridin-2-ylmethyl- benzamide;
N,N-Diethyl-3-[5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5-α]pyrazin-8-ylamino]-benzamide;
{3-[5-(lH-Pyrazol-4-yl)-[l,2,4]triazolo[l,5-α]pyrazin-8-ylamino]-phenyl}-pyrrolidin-l-yl- methanone;
(4-Isopropyl-piperazin-l-yl)-{3-[5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5-α]pyrazin-8- ylamino] -phenyl} -methanone;
3-[5-(lH-Pyrazol-4-yl)-[l,2,4]triazolo[l,5-α]pyrazin-8-ylamino]-benzamide;
N-Ethyl-3-[5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5-α]pyrazin-8-ylamino]-benzamide;
N-Cyclohexylmethyl-3 -[5 -(lH-pyrazol-4-yl)- [ 1 ,2,4]triazolo [ 1 ,5 -α]pyrazin-8 -ylamino] - benzamide;
N-(4-Ηydroxy-benzyl)-3-[5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5-α]pyrazin-8-ylamino]- benzamide;
4-[8-(4-Benzylcarbamoyl-3-methoxy-phenylamino)-[l,2,4]triazolo[l,5-α]pyrazin-5-yl]- thiophene-2-carboxylic acid amide;
4-[8-(6-Benzoylamino-pyridin-3-ylamino)-[l,2,4]triazolo[l,5-α]pyrazin-5-yl]-thiophene-2- carboxylic acid amide;
N-Benzyl-N-methyl-3 - [5 -( lH-pyrazol-4-yl)- [ 1 ,2,4]triazolo [ 1 ,5 -α]pyrazin-8-ylamino] - benzamide;
N- {5-[5-(lH-Pyrazol-4-yl)-[l,2,4]triazolo[l, 5 -α]pyrazin-8-ylamino]-pyridin-2-yl} -benzamide; l-{4-[5-(lH-Pyrazol-4-yl)-[l,2,4]triazolo[l,5-α]pyrazin-8-ylamino]-benzoyl}-piperidine-4- carboxylic acid methylamide;
4-[5-(lH-Pyrazol-4-yl)-[l,2,4]triazolo[l,5-α]pyrazin-8-ylamino]-N-(2-pyridin-4-yl-ethyl)- benzamide;
(4-Ethyl-piperazin-l-yl)-{4-[5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5-α]pyrazin-8-ylamino]- phenyl} -methanone;
N-(l-Ethyl-pyrrolidin-2-ylmethyl)-4-[5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5-α]pyrazin-8- ylamino] -benzamide; (2,5-Dihydro-pyrrol-l-yl)-{4-[5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5-α]pyrazin-8-ylamino]- phenyl} -methanone;
[4-(2-Ethoxy-ethyl)-piperazin-l-yl]-{4-[5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5-α]pyrazin-8- ylamino] -phenyl} -methanone;
N-Cyclopropylmethyl-4-[5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5-α]pyrazin-8-ylamino]- benzamide;
N-Methyl-N-(l-{4-[5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5-α]pyrazin-8-ylamino]-benzoyl}- pyrrolidin-3 -yl)-acetamide;
[4-(4-Fluoro-benzyl)-piperazin-l-yl]-{4-[5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5-α]pyrazin-8- ylamino] -phenyl} -methanone;
(4-Phenyl-piperazin-l-yl)-{4-[5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5-α]pyrazin-8-ylamino]- phenyl} -methanone;
((S)-2-Methoxymethyl-pyrrolidin-l-yl)-{4-[5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5-α]pyrazin-
8 -ylamino] -phenyl} -methanone;
N-((R)-l-Benzyl-pyrrolidin-3-yl)-4-[5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5-α]pyrazin-8- ylamino] -benzamide;
{4-[5-(lH-Pyrazol-4-yl)-[l ,2,4]triazolo[l ,5-α]pyrazin-8-ylamino]-phenyl} -(4-pyridin-2-yl- piperazin- 1 -yl)-methanone;
N-(l-Methoxymethyl-propyl)-4-[5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5-α]pyrazin-8- ylamino] -benzamide;
[4-(2-Methoxy-ethyl)-piperazin-l-yl]-{4-[5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5-α]pyrazin-8- ylamino] -phenyl} -methanone;
N-[2-(4-Ηydroxy-phenyl)-ethyl]-4-[5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5-α]pyrazin-8- ylamino] -benzamide;
(4-#sec!-Butyl-piperazin-l-yl)-{4-[5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5-α]pyrazin-8- ylamino] -phenyl} -methanone;
N-[4-(4-Methyl-piperazin-l-yl)-phenyl]-4-[5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5-α]pyrazin-
8 -ylamino] -b enzamide;
(Benzyl-{4-[5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5-α]pyrazin-8-ylamino]-benzoyl}-amino)- acetic acid ethyl ester;
N-Isopropyl-2-(4-{4-[5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5-α]pyrazin-8-ylamino]-benzoyl}- piperazin- 1 -yl)-acetamide;
N-(2-Methoxy-ethyl)-N-methyl-4-[5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5-α]pyrazin-8- ylamino] -benzamide;
N-Ethyl-N-(2-methoxy-ethyl)-4-[5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5-α]pyrazin-8- ylamino] -benzamide; 4-[5-(lH-Pyrazol-4-yl)-[l,2,4]triazolo[l,5-α]pyrazin-8-ylamino]-N-(tetrahydro-furan-2- ylmethyl)-benzamide;
(3,6-Dihydro-2H-pyridin-l-yl)-{4-[5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5-α]pyrazin-8- ylamino] -phenyl} -methanone;
N-(2-Methylsulfanyl-ethyl)-4-[5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5-α]pyrazin-8-ylamino]- benzamide;
N-(2,2-Dimethyl-[l,3]dioxolan-4-ylmethyl)-4-[5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5- α]pyrazin-8-ylamino] -benzamide;
N-(2-Diisopropylamino-ethyl)-4-[5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5-α]pyrazin-8- ylamino] -benzamide;
N-((S)-l-Ethyl-pyrrolidin-2-ylmethyl)-4-[5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5-α]pyrazin-8- ylamino] -benzamide;
N-Isobutyl-4-[5 -( lH-pyrazol-4-yl)- [ 1 ,2,4]triazolo [ 1 ,5 -α]pyrazin-8 -ylamino] -benzamide; [l,4']Bipiperidinyl-l'-yl-{4-[5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5-α]pyrazin-8-ylamino]- phenyl} -methanone;
N-(2-Ηydroxy-ethyl)-4-[5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5-α]pyrazin-8-ylamino]- benzamide; and
4-[8-(6-Phenylacetylamino-pyridin-3-ylamino)-[l,2,4]triazolo[l,5-α]pyrazin-5-yl]-thiophene- 2-carboxylic acid amide.
52. A compound according to claim 1 wherein the compound is selected from
4- { 8 -[4-(4-Methyl-piperazin- 1 -yl)-phenylamino] - [ 1 ,2,4]triazolo[ 1 ,5 -α]pyrazin-5 - yl}-thiophene-2-carboxylic acid amide;
5 - { 8 -[4-(4-Methyl-piperazin- 1 -yl)-phenylamino] - [ 1 ,2,4]triazolo[ 1 ,5 -α]pyrazin-5 - yl}-thiophene-2-carboxylic acid amide;
[5-(5-Methyl-lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5-α]pyrazin-8-yl]-(4-morpholin-4- yl-phenyl)-amine;
4-[8-(4-Moφholin-4-yl-phenylamino)-[l,2,4]triazolo[l,5-α]pyrazin-5-yl]- thiophene-2-carboxylic acid amide;
4-{8-[4-(2-Morpholin-4-yl-ethoxy)-phenylamino]-[l,2,4]triazolo[l,5-α]pyrazin-5- yl}-thiophene-2-carboxylic acid amide;
4-{8-[4-((2R,6S)-2,6-Dimethyl-moφholin-4-yl)-phenylamino]-[l,2,4]triazolo[l,5- α]pyrazin-5-yl}-thiophene-2-carboxylic acid amide;
4-[8-(4-Piperazin-l-yl-phenylamino)-[l,2,4]triazolo[l,5-α]pyrazin-5-yl]- thiophene-2-carboxylic acid amide;
4-{8-[4-(4-Isopropyl-piperazin-l-yl)-phenylamino]-[l,2,4]triazolo[l,5-α]pyrazin- 5 -yl} -thiophene-2-carboxylic acid amide;
4-{8-[4-((2S,5R)-2,4,5-Trimethyl-piperazin-l-yl)-phenylamino]-
[1 ,2,4]triazolo[l,5-α]pyrazin-5-yl} -thiophene-2-carboxylic acid amide;
5 - [8 -(4-Morpholin-4-yl-phenylamino)- [ 1 ,2,4]triazolo[ 1 ,5 -α]pyrazin-5 -yl] -2,3 - dihydro-isoindol-1 -one;
4-[8-(4-Morpholin-4-yl-phenylamino)-[l,2,4]triazolo[l,5-α]pyrazin-5-yl]-furan-2- carboxylic acid amide;
5-{8-[4-(4-Isopropyl-piperazin-l-yl)-phenylamino]-[l,2,4]triazolo[l,5-α]pyrazin-
5-yl} -2,3 -dihydro-isoindol-1 -one;
4-[8-(6-Moφholin-4-yl-pyridin-3-ylamino)-[l,2,4]triazolo[l,5-α]pyrazin-5-yl]- thiophene-2-carboxylic acid amide;
4-{8-[6-(4-Cyclopropylmethyl-piperazin-l-yl)-pyridin-3-ylamino]-
[1 ,2,4]triazolo[l,5-α]pyrazin-5-yl} -thiophene-2-carboxylic acid amide;
4-{8-[6-(4-Isopropyl-piperazin-l-yl)-pyridin-3-ylamino]-[l,2,4]triazolo[l,5- α]pyrazin-5-yl} -thiophene-2-carboxylic acid amide;
4-(8-{6-[4-(2,2,2-Trifluoro-ethyl)-piperazin-l-yl]-pyridin-3-ylamino}-
[1 ^^jtriazolofl^-αlpyrazin-S-yO-thiophene^-carboxylic acid amide;
4-(8-{4-[4-(2,2,2-Trifluoro-ethyl)-piperazin-l-yl]-phenylamino}-
[1 ^^jtriazolofl^-αlpyrazin-5-yl-thiophene^-carboxylic acid amide;
4- { 8 -[4-(4-Cyclopropylmethyl-piperazin- 1 -yl)-phenylamino] - [ 1 ,2,4]triazolo [1,5- α]pyrazin-5-yl} -thiophene-2-carboxylic acid amide;
5-{8-[4-(2-Morpholin-4-yl-ethoxy)-phenylamino]-[l,2,4]triazolo[l,5-α]pyrazin-5- yl} -2,3 -dihydro-isoindol-1 -one;
5-{8-[6-(4-Isopropyl-piperazin-l-yl)-pyridin-3-ylamino]-[l,2,4]triazolo[l,5- α]pyrazin-5-yl} -2,3 -dihydro-isoindol-1 -one;
5-[8-(6-Moφholin-4-yl-pyridin-3-ylamino)-[l,2,4]triazolo[l,5-α]pyrazin-5-yl]-
2,3 -dihydro-isoindol-1 -one;
4-{8-[4-(4-Isopropyl-piperazin-l-yl)-phenylamino]-[l,2,4]triazolo[l,5-α]pyrazin-
5-yl}-furan-2-carboxylic acid amide;
4-{8-[4-(l-Isopropyl-piperidin-4-yl)-phenylamino]-[l,2,4]triazolo[l,5-α]pyrazin-
5 -yl} -thiophene-2-carboxylic acid amide;
5-{8-[4-(l-Isopropyl-piperidin-4-yl)-phenylamino]-[l,2,4]triazolo[l,5-α]pyrazin-
5-yl} -2,3 -dihydro-isoindol-1 -one;
5 - [8 -(2-Moφholin-4-yl-pyrimidin-5 -ylamino)- [ 1 ,2,4]triazolo[ 1 ,5 -α]pyrazin-5 -yl]-
2,3 -dihydro-isoindol-1 -one; [4-(4-rerr-Butyl-piperazin-l-yl)-phenyl]-[5-(lH-pyrazol-4-yl)-[l,2,4]triazolo[l,5- α]pyrazin-8-yl]-amine; and
2-[8-(4-Moφholin-4-yl-phenylamino)-[l,2,4]triazolo[l,5-α]pyrazin-5-yl]-4,5- dihydro-thieno[2,3-c]pyrrol-6-one.
53. A pharmaceutical composition comprising a pharmaceutically acceptable carrier and a pharmaceutically effective amount of a compound according to any one of claims 1 to 52.
54. The pharmaceutical composition of claim 53, wherein the carrier is a parenteral carrier.
55. The pharmaceutical composition of claim 53, wherein the carrier is an oral carrier.
56. The pharmaceutical composition of claim 53, wherein the carrier is a topical carrier.
57. The use of a compound according to any one of claims 1 to 52 in the manufacture of a medicament for treatment or prophylaxis of a condition characterized by ECM degradation.
58. The use of a compound according to any one of claims 1 to 52 in the manufacture of a medicament for treatment or prophylaxis of a condition selected from diseases involving inflammation.
59. The use of a compound according to claim 58, wherein said disease is rheumatoid arthritis.
60. The use of a compound according to any one of claims 1 to 52 in the manufacture of a medicament for treatment or prophylaxis of a condition prevented, ameliorated or eliminated by administration of an inhibitor of Mitogen-Activated Protein Kinase- Activated Protein Kinase 5.
61. A method of treatment of a condition characterised by abnormal matrix metallo proteinase activity, which comprises administering a therapeutically effective amount of a matrix metallo proteinase inhibiting compound according to any one of claims 1 to 52.
62. A method of treatment of a condition selected from diseases involving degradation of extracellular matrix, which comprises administering a therapeutically effective matrix metallo proteinase inhibiting amount of a compound according to any one of claims 1 to 52.
63. A method of treatment of a condition selected from diseases involving abnormal cellular expression of MMPl, which comprises administering a therapeutically effective matrix metallo proteinase inhibiting amount of a compound according to any one of claims 1 to 52.
64. A method of treatment or prevention of inflammatory diseases, which comprises administering to a subject in need thereof, a therapeutically effective amount of a compound according to any one of claims 1 to 52.
65. A method of treatment or prevention of rheumatoid arthritis, which comprises administering to a subject in need thereof, a therapeutically effective amount of a compound according to any one of claims 1 to 52.
PCT/EP2007/055246 2006-05-31 2007-05-30 Triazolopyrazine compounds useful for the treatment of degenerative & inflammatory diseases WO2007138072A2 (en)

Priority Applications (16)

Application Number Priority Date Filing Date Title
CA002653506A CA2653506A1 (en) 2006-05-31 2007-05-30 Triazolopyrazine compounds useful for the treatment of degenerative & inflammatory diseases
DE602007006010T DE602007006010D1 (en) 2006-05-31 2007-05-30 TRIAZOLOPYRAZIN COMPOUNDS FOR THE TREATMENT OF DEGENERATIVE AND INFLAMMATORY DISEASES
NZ573015A NZ573015A (en) 2006-05-31 2007-05-30 Triazolopyrazine compounds useful for the treatment of degenerative & inflammatory diseases
BRPI0712531-3A BRPI0712531A2 (en) 2006-05-31 2007-05-30 triazolopyrazine compound useful for the treatment of degenerative and inflammatory diseases
MX2008015057A MX2008015057A (en) 2006-05-31 2007-05-30 Triazolopyrazine compounds useful for the treatment of degenerative & inflammatory diseases.
EP07729660A EP2029602B1 (en) 2006-05-31 2007-05-30 Triazolopyrazine compounds useful for the treatment of degenerative & inflammatory diseases
PL07729660T PL2029602T3 (en) 2006-05-31 2007-05-30 Triazolopyrazine compounds useful for the treatment of degenerative & inflammatory diseases
EA200870592A EA200870592A1 (en) 2006-05-31 2007-05-30 TRIAZOLOPIRASIN CONNECTIONS SUITABLE FOR THE TREATMENT OF DEGENERATIVE AND INFLAMMATORY DISEASES
AT07729660T ATE465164T1 (en) 2006-05-31 2007-05-30 TRIAZOLOPYRAZINE COMPOUNDS FOR THE TREATMENT OF DEGENERATIONAL AND INFLAMMATORY DISEASES
SI200730288T SI2029602T1 (en) 2006-05-31 2007-05-30 Triazolopyrazine compounds useful for the treatment of degenerative & inflammatory diseases
JP2009512585A JP2009538877A (en) 2006-05-31 2007-05-30 Triazolopyrazine compounds useful for the treatment of degenerative and inflammatory diseases
DK07729660.6T DK2029602T3 (en) 2006-05-31 2007-05-30 Triazolopyrazine compounds that can be used to treat degenerative and inflammatory diseases
AU2007267121A AU2007267121A1 (en) 2006-05-31 2007-05-30 Triazolopyrazine compounds useful for the treatment of degenerative & inflammatory diseases
IL195033A IL195033A0 (en) 2006-05-31 2008-10-30 Triazolopyrazine compounds useful for the treatment of degenerative & inflammatory diseases
NO20084703A NO20084703L (en) 2006-05-31 2008-11-10 Triazolopyrazine compounds useful for the treatment of degenerative and inflammatory diseases
HR20100378T HRP20100378T1 (en) 2006-05-31 2010-07-06 Triazolopyrazine compounds useful for the treatment of degenerative & inflammatory diseases

Applications Claiming Priority (10)

Application Number Priority Date Filing Date Title
US80355206P 2006-05-31 2006-05-31
US60/803,552 2006-05-31
US93176407P 2007-05-25 2007-05-25
US93176307P 2007-05-25 2007-05-25
US93184407P 2007-05-25 2007-05-25
US60/931,844 2007-05-25
US60/931,763 2007-05-25
US60/931,764 2007-05-25
US93210007P 2007-05-29 2007-05-29
US60/932,100 2007-05-29

Publications (2)

Publication Number Publication Date
WO2007138072A2 true WO2007138072A2 (en) 2007-12-06
WO2007138072A3 WO2007138072A3 (en) 2008-02-14

Family

ID=38618932

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2007/055246 WO2007138072A2 (en) 2006-05-31 2007-05-30 Triazolopyrazine compounds useful for the treatment of degenerative & inflammatory diseases

Country Status (20)

Country Link
US (3) US7501411B2 (en)
JP (1) JP2009538877A (en)
KR (1) KR20090026288A (en)
AT (1) ATE465164T1 (en)
AU (1) AU2007267121A1 (en)
BR (1) BRPI0712531A2 (en)
CA (1) CA2653506A1 (en)
CR (1) CR10458A (en)
DE (1) DE602007006010D1 (en)
DK (1) DK2029602T3 (en)
EA (1) EA200870592A1 (en)
EC (1) ECSP088923A (en)
HR (1) HRP20100378T1 (en)
IL (1) IL195033A0 (en)
MX (1) MX2008015057A (en)
NI (1) NI200800299A (en)
NO (1) NO20084703L (en)
NZ (1) NZ573015A (en)
PT (1) PT2029602E (en)
WO (1) WO2007138072A2 (en)

Cited By (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008138842A1 (en) * 2007-05-10 2008-11-20 Galapagos N.V. Imidazopyrazines and triazolopyrazine for the treatment of joint degenerative and inflammatory diseases
WO2008138843A1 (en) * 2007-05-10 2008-11-20 Galapagos N.V. Imidazopyridines and triazolopyrimidines useful for the treatment of joint degenerative & inflammatory diseases
DE102007060172A1 (en) 2007-12-13 2009-06-25 Bayer Healthcare Ag New nitrogen substituted heterocyclic compounds are glycogen synthase kinase 3-beta inhibitors useful e.g. to treat and/or prevent hematological disease, preferably leukopenia, and for ex-vivo increase of adult hematopoietic stem cells
WO2009135885A1 (en) * 2008-05-07 2009-11-12 Galapagos Nv Fused pyrazine compounds useful for the treatment of degenerative and inflammatory diseases
WO2009138176A1 (en) 2008-05-15 2009-11-19 Bayer Schering Pharma Aktiengesellschaft Substituted imidazo- and triazolopyrimidines, imidazo- and pyrazolopyrazines and imidazotriazines as gsk3beta-inhibitors
DE102008035209A1 (en) 2008-07-29 2010-02-04 Bayer Schering Pharma Aktiengesellschaft New nitrogen substituted heterocyclic compounds are glycogen synthase kinase 3-beta inhibitors useful e.g. to treat and/or prevent hematological disease, preferably leukopenia, and for ex-vivo increase of adult hematopoietic stem cells
WO2011054922A1 (en) * 2009-11-05 2011-05-12 Galapagos Nv Salts of fused pyrazine compounds, useful for the treatment of degenerative and inflammatory diseases.
US7977352B2 (en) 2006-12-01 2011-07-12 Galapagos Nv Triazolopyridine compounds useful for the treatment of degenerative and inflammatory diseases
US8299056B2 (en) 2008-09-08 2012-10-30 Signal Pharmaceuticals, Llc Aminotriazolopyridines, compositions thereof, and methods of treatment therewith
WO2015101622A1 (en) 2014-01-03 2015-07-09 Bayer Cropscience Ag Novel pyrazolyl-heteroarylamides as pesticides
WO2015166370A1 (en) 2014-04-28 2015-11-05 Pfizer Inc. Heteroaromatic compounds and their use as dopamine d1 ligands
AU2011224316B2 (en) * 2010-03-10 2016-09-15 Kalypsys, Inc. Heterocyclic inhibitors of histamine receptors for the treatment of disease
WO2017146128A1 (en) * 2016-02-26 2017-08-31 大日本住友製薬株式会社 Imidazolylamide derivative
US9951069B1 (en) 2017-01-11 2018-04-24 Rodin Therapeutics, Inc. Bicyclic inhibitors of histone deacetylase
CN109206357A (en) * 2017-07-04 2019-01-15 浙江九洲药业股份有限公司 A kind of general formula compound and its preparation method and application of gliclazide intermediate
US10294237B2 (en) 2015-06-22 2019-05-21 Sumitomo Dainippon Pharma Co., Ltd. Bicyclic heterocyclic amide derivative
US10421756B2 (en) 2015-07-06 2019-09-24 Rodin Therapeutics, Inc. Heterobicyclic N-aminophenyl-amides as inhibitors of histone deacetylase
US10919902B2 (en) 2015-07-06 2021-02-16 Alkermes, Inc. Hetero-halo inhibitors of histone deacetylase
US11225475B2 (en) 2017-08-07 2022-01-18 Alkermes, Inc. Substituted pyridines as inhibitors of histone deacetylase
AU2018210770B2 (en) * 2017-01-23 2022-03-24 Revolution Medicines, Inc. Bicyclic compounds as allosteric SHP2 inhibitors
US11661401B2 (en) 2016-07-12 2023-05-30 Revolution Medicines, Inc. 2,5-disubstituted 3-methyl pyrazines and 2,5,6-trisubstituted 3-methyl pyrazines as allosteric SHP2 inhibitors
US11673896B2 (en) 2017-01-23 2023-06-13 Revolution Medicines, Inc. Pyridine compounds as allosteric SHP2 inhibitors
US11673901B2 (en) 2017-12-15 2023-06-13 Revolution Medicines, Inc. Polycyclic compounds as allosteric SHP2 inhibitors
US11702411B2 (en) 2017-10-12 2023-07-18 Revolution Medicines, Inc. Pyridine, pyrazine, and triazine compounds as allosteric SHP2 inhibitors

Families Citing this family (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP4832426B2 (en) 2004-04-02 2011-12-07 オーエスアイ・ファーマスーティカルズ・インコーポレーテッド 6,6-bicyclic substituted heterobicyclic protein kinase inhibitors
US7893058B2 (en) * 2006-05-15 2011-02-22 Janssen Pharmaceutica Nv Imidazolopyrazine compounds useful for the treatment of degenerative and inflammatory diseases
ATE465164T1 (en) * 2006-05-31 2010-05-15 Galapagos Nv TRIAZOLOPYRAZINE COMPOUNDS FOR THE TREATMENT OF DEGENERATIONAL AND INFLAMMATORY DISEASES
CN102405214A (en) 2009-04-20 2012-04-04 Osi药物有限责任公司 Preparation of c-pyrazine-methylamines
JP2012526138A (en) * 2009-05-07 2012-10-25 オーエスアイ・ファーマシューティカルズ,エルエルシー Use of OSI-906 to treat adrenocortical cancer
EP2343297A1 (en) 2009-11-30 2011-07-13 Bayer Schering Pharma AG Triazolopyridines
US9512126B2 (en) 2012-03-14 2016-12-06 Bayer Intellectual Property Gmbh Substituted imidazopyridazines
HUE033131T2 (en) 2013-06-11 2017-11-28 Bayer Pharma AG Prodrug derivatives of substituted triazolopyridines
CN105339370B (en) * 2013-06-19 2017-09-15 加拉帕戈斯股份有限公司 Compound and and its pharmaceutical composition for treating diseases associated with inflammation
CN112384515A (en) 2018-02-27 2021-02-19 因赛特公司 Imidazopyrimidines and triazolopyrimidines as A2A/A2B inhibitors
CN108379583B (en) * 2018-03-27 2021-08-17 北京大学 Target for drug therapy of tumor metastasis and application thereof
MA52940A (en) 2018-05-18 2021-04-28 Incyte Corp FUSION PYRIMIDINE DERIVATIVES USED AS A2A / A2B INHIBITORS
EP3818063A1 (en) 2018-07-05 2021-05-12 Incyte Corporation Fused pyrazine derivatives as a2a / a2b inhibitors
TWI829857B (en) 2019-01-29 2024-01-21 美商英塞特公司 Pyrazolopyridines and triazolopyridines as a2a / a2b inhibitors

Family Cites Families (31)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6316444B1 (en) * 1999-06-30 2001-11-13 Merck & Co., Inc. SRC kinase inhibitor compounds
KR20020038804A (en) * 1999-10-08 2002-05-23 파퀘스, 파우쓰-베르구스 Bicyclic imidazo-3-yl-amine derivatives which are substituted on the sixth ring
CN1257169C (en) * 1999-10-08 2006-05-24 格吕伦塔尔有限公司 Bicyclic imidazo-3-yl-amine derivatives
DK1218383T3 (en) * 1999-10-08 2009-01-19 Gruenenthal Gmbh Biocyclic imidazo-5-yl amino derivatives
DE10019714A1 (en) * 2000-04-20 2002-01-10 Gruenenthal Gmbh Salts of bicyclic, N-acylated imidazo-3-amines and imidazo-5-amines
NO311460B1 (en) 2000-07-14 2001-11-26 Zopa As Method and apparatus for cooling products in a transport system
DE10050663A1 (en) * 2000-10-13 2002-04-18 Gruenenthal Gmbh Use of amino-substituted imidazo(1,2-a)pyridine, imidazo(1,2-a)pyrimidine and imidazo(1,2-a)pyrazine derivatives as NO synthase inhibitors, e.g. in treatment of migraine and neurodegenerative diseases
US20020132823A1 (en) 2001-01-17 2002-09-19 Jiahuai Han Assay method
CA2436487A1 (en) * 2001-01-30 2002-08-08 Cytopia Pty Ltd. Methods of inhibiting kinases
IL164703A0 (en) * 2002-04-19 2005-12-18 Cellular Genomics Inc ImidazoÄ1,2-AÜpyrazin-8-ylamines method of making and method of use thereof
EP1543008B1 (en) * 2002-09-23 2007-11-07 Schering Corporation Imidazopyrazines as cyclin dependent kinase inhibitors
AU2003286711A1 (en) 2002-10-25 2004-05-13 Vertex Pharmaceuticals Incorporated Indazolinone compositions useful as kinase inhibitors
US7160885B2 (en) 2003-02-10 2007-01-09 Cgi Pharmaceuticals, Inc. Certain 6, 8-(heteroaryl or aryl) disubstituted imidazo[1,2-a]pyrazines as modulators of Hsp90 complex activity
US7186832B2 (en) * 2003-02-20 2007-03-06 Sugen Inc. Use of 8-amino-aryl-substituted imidazopyrazines as kinase inhibitors
US7157460B2 (en) * 2003-02-20 2007-01-02 Sugen Inc. Use of 8-amino-aryl-substituted imidazopyrazines as kinase inhibitors
WO2004092177A1 (en) * 2003-04-09 2004-10-28 Biogen Idec Ma Inc. Triazolopyrazines and methods of making and using the same
AU2004282219C1 (en) * 2003-10-15 2009-12-17 Osi Pharmaceuticals, Inc. Imidazo [1, 5 - a] pyrazine tyrosine kinase inhibitors
WO2005085252A1 (en) 2004-03-04 2005-09-15 Biofocus Discovery Limited Imidazo ‘1,2-a’ pyrazine compounds which interact with protein kinases
EP1765351B9 (en) 2004-06-09 2010-03-24 Oncalis AG Protein kinase inhibitors
US8030327B2 (en) * 2004-11-08 2011-10-04 Mds K.K. Fused imidazole derivative
WO2006053121A2 (en) * 2004-11-10 2006-05-18 Cgi Pharmaceuticals, Inc. Imidazo[1 , 2-a] pyrazin-8-ylamines useful as modulators of kinase activity
US20060149535A1 (en) * 2004-12-30 2006-07-06 Lg Electronics Inc. Method for controlling speed of audio signals
WO2006086545A2 (en) 2005-02-08 2006-08-17 Robert W Desimone Method to identify novel therapeutics
ES2543607T3 (en) * 2005-03-10 2015-08-20 Gilead Connecticut, Inc. Certain substituted amides, method of obtaining, and method of use
EP1928237A4 (en) * 2005-09-02 2011-03-09 Abbott Lab Novel imidazo based heterocycles
RU2008122967A (en) * 2005-11-10 2009-12-20 Шеринг Корпорейшн (US) IMIDAZOPYRAZINES AS PROTEINKINASE INHIBITORS
CA2628534A1 (en) * 2005-11-10 2007-05-18 Schering Corporation Methods for inhibiting protein kinases
EA014906B1 (en) * 2005-12-01 2011-02-28 Элан Фармасьютикалз, Инк. 5-(arylsulfonyl)-pyrazolopiperidines
US7893058B2 (en) * 2006-05-15 2011-02-22 Janssen Pharmaceutica Nv Imidazolopyrazine compounds useful for the treatment of degenerative and inflammatory diseases
ATE465164T1 (en) * 2006-05-31 2010-05-15 Galapagos Nv TRIAZOLOPYRAZINE COMPOUNDS FOR THE TREATMENT OF DEGENERATIONAL AND INFLAMMATORY DISEASES
JP2010502751A (en) 2006-09-11 2010-01-28 シージーアイ ファーマシューティカルズ,インコーポレイティド Kinase inhibitors and methods of using and identifying kinase inhibitors

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
None

Cited By (43)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7977352B2 (en) 2006-12-01 2011-07-12 Galapagos Nv Triazolopyridine compounds useful for the treatment of degenerative and inflammatory diseases
US8133895B2 (en) * 2007-05-10 2012-03-13 Janssen Pharmaceutica N.V. Fused pyrazine compounds useful for the treatment of degenerative and inflammatory diseases
WO2008138843A1 (en) * 2007-05-10 2008-11-20 Galapagos N.V. Imidazopyridines and triazolopyrimidines useful for the treatment of joint degenerative & inflammatory diseases
WO2008138842A1 (en) * 2007-05-10 2008-11-20 Galapagos N.V. Imidazopyrazines and triazolopyrazine for the treatment of joint degenerative and inflammatory diseases
US9006237B2 (en) 2007-05-10 2015-04-14 Janssen Pharmaceutica Nv Fused pyrazine compounds useful for the treatment of degenerative and inflammatory diseases
US8148369B2 (en) 2007-05-10 2012-04-03 Janssen Pharmaceutica Nv Fused pyrazine compounds useful for the treatment of degenerative and inflammatory diseases
US8138193B2 (en) 2007-05-10 2012-03-20 Janssen Pharmaceutica N.V. Fused pyrazine compounds useful for the treatment of degenerative and inflammatory diseases
DE102007060172A1 (en) 2007-12-13 2009-06-25 Bayer Healthcare Ag New nitrogen substituted heterocyclic compounds are glycogen synthase kinase 3-beta inhibitors useful e.g. to treat and/or prevent hematological disease, preferably leukopenia, and for ex-vivo increase of adult hematopoietic stem cells
WO2009135885A1 (en) * 2008-05-07 2009-11-12 Galapagos Nv Fused pyrazine compounds useful for the treatment of degenerative and inflammatory diseases
CN102036997A (en) * 2008-05-07 2011-04-27 加拉帕戈斯股份有限公司 Fused pyrazine compounds useful for the treatment of degenerative and inflammatory diseases
US8012983B2 (en) 2008-05-07 2011-09-06 Galapagos Nv Substituted triazolopyrazines useful for the treatment of degenerative and inflammatory diseases
WO2009138176A1 (en) 2008-05-15 2009-11-19 Bayer Schering Pharma Aktiengesellschaft Substituted imidazo- and triazolopyrimidines, imidazo- and pyrazolopyrazines and imidazotriazines as gsk3beta-inhibitors
DE102008023801A1 (en) 2008-05-15 2009-11-19 Bayer Schering Pharma Aktiengesellschaft Substituted imidazo and triazolopyrimidines, imidazo and pyrazolopyrazines and imidazotriazines
DE102008035209A1 (en) 2008-07-29 2010-02-04 Bayer Schering Pharma Aktiengesellschaft New nitrogen substituted heterocyclic compounds are glycogen synthase kinase 3-beta inhibitors useful e.g. to treat and/or prevent hematological disease, preferably leukopenia, and for ex-vivo increase of adult hematopoietic stem cells
US8299056B2 (en) 2008-09-08 2012-10-30 Signal Pharmaceuticals, Llc Aminotriazolopyridines, compositions thereof, and methods of treatment therewith
WO2011054922A1 (en) * 2009-11-05 2011-05-12 Galapagos Nv Salts of fused pyrazine compounds, useful for the treatment of degenerative and inflammatory diseases.
AU2011224316B2 (en) * 2010-03-10 2016-09-15 Kalypsys, Inc. Heterocyclic inhibitors of histamine receptors for the treatment of disease
WO2015101622A1 (en) 2014-01-03 2015-07-09 Bayer Cropscience Ag Novel pyrazolyl-heteroarylamides as pesticides
US9856263B2 (en) 2014-04-28 2018-01-02 Pfizer Inc. Heteroaromatic compounds and their use as dopamine D1 ligands
WO2015166370A1 (en) 2014-04-28 2015-11-05 Pfizer Inc. Heteroaromatic compounds and their use as dopamine d1 ligands
US10294237B2 (en) 2015-06-22 2019-05-21 Sumitomo Dainippon Pharma Co., Ltd. Bicyclic heterocyclic amide derivative
US11858939B2 (en) 2015-07-06 2024-01-02 Alkermes, Inc. Hetero-halo inhibitors of histone deacetylase
US10421756B2 (en) 2015-07-06 2019-09-24 Rodin Therapeutics, Inc. Heterobicyclic N-aminophenyl-amides as inhibitors of histone deacetylase
US10919902B2 (en) 2015-07-06 2021-02-16 Alkermes, Inc. Hetero-halo inhibitors of histone deacetylase
US10898469B2 (en) 2016-02-26 2021-01-26 Sumitomo Dainippon Pharma Co., Ltd. Imidazolylamide derivative
JPWO2017146128A1 (en) * 2016-02-26 2018-12-20 大日本住友製薬株式会社 Imidazolylamide derivatives
WO2017146128A1 (en) * 2016-02-26 2017-08-31 大日本住友製薬株式会社 Imidazolylamide derivative
US11661401B2 (en) 2016-07-12 2023-05-30 Revolution Medicines, Inc. 2,5-disubstituted 3-methyl pyrazines and 2,5,6-trisubstituted 3-methyl pyrazines as allosteric SHP2 inhibitors
US11225479B2 (en) 2017-01-11 2022-01-18 Alkermes, Inc. Bicyclic inhibitors of histone deacetylase
US10696673B2 (en) 2017-01-11 2020-06-30 Rodin Therapeutics, Inc. Bicyclic inhibitors of histone deacetylase
US10519149B2 (en) 2017-01-11 2019-12-31 Rodin Therapeutics, Inc. Bicyclic inhibitors of histone deacetylase
US10793567B2 (en) 2017-01-11 2020-10-06 Rodin Therapeutics, Inc. Bicyclic inhibitors of histone deacetylase
US11286256B2 (en) 2017-01-11 2022-03-29 Alkermes, Inc. Bicyclic inhibitors of histone deacetylase
US11987580B2 (en) 2017-01-11 2024-05-21 Alkermes, Inc. Bicyclic inhibitors of histone deacetylase
US9951069B1 (en) 2017-01-11 2018-04-24 Rodin Therapeutics, Inc. Bicyclic inhibitors of histone deacetylase
US11739093B2 (en) 2017-01-23 2023-08-29 Revolution Medicines, Inc. Substituted pyrazolopyrazines, imidazopyrazines and [1,2,4]triazolopyrazines as allosteric SHP2 inhibitors
AU2018210770B2 (en) * 2017-01-23 2022-03-24 Revolution Medicines, Inc. Bicyclic compounds as allosteric SHP2 inhibitors
US11673896B2 (en) 2017-01-23 2023-06-13 Revolution Medicines, Inc. Pyridine compounds as allosteric SHP2 inhibitors
CN109206357A (en) * 2017-07-04 2019-01-15 浙江九洲药业股份有限公司 A kind of general formula compound and its preparation method and application of gliclazide intermediate
US11225475B2 (en) 2017-08-07 2022-01-18 Alkermes, Inc. Substituted pyridines as inhibitors of histone deacetylase
US11912702B2 (en) 2017-08-07 2024-02-27 Alkermes, Inc. Substituted pyridines as inhibitors of histone deacetylase
US11702411B2 (en) 2017-10-12 2023-07-18 Revolution Medicines, Inc. Pyridine, pyrazine, and triazine compounds as allosteric SHP2 inhibitors
US11673901B2 (en) 2017-12-15 2023-06-13 Revolution Medicines, Inc. Polycyclic compounds as allosteric SHP2 inhibitors

Also Published As

Publication number Publication date
DK2029602T3 (en) 2010-08-09
BRPI0712531A2 (en) 2012-12-25
CR10458A (en) 2009-02-26
US7501411B2 (en) 2009-03-10
NZ573015A (en) 2010-11-26
PT2029602E (en) 2010-07-08
DE602007006010D1 (en) 2010-06-02
US7915256B2 (en) 2011-03-29
HRP20100378T1 (en) 2010-08-31
EA200870592A1 (en) 2009-08-28
IL195033A0 (en) 2009-08-03
NI200800299A (en) 2010-05-25
US20080090818A1 (en) 2008-04-17
AU2007267121A1 (en) 2007-12-06
CA2653506A1 (en) 2007-12-06
US20110178067A1 (en) 2011-07-21
WO2007138072A3 (en) 2008-02-14
US20100298300A1 (en) 2010-11-25
MX2008015057A (en) 2008-12-10
KR20090026288A (en) 2009-03-12
ECSP088923A (en) 2009-01-30
JP2009538877A (en) 2009-11-12
ATE465164T1 (en) 2010-05-15
NO20084703L (en) 2009-02-19

Similar Documents

Publication Publication Date Title
WO2007138072A2 (en) Triazolopyrazine compounds useful for the treatment of degenerative & inflammatory diseases
EP2027124B1 (en) Imidazolopyrazine compounds useful for the treatment of degenerative and inflammatory diseases
AU2020202707B2 (en) Heteroaryl pyridone and aza-pyridone compounds as inhibitors of Btk activity
US9505785B2 (en) Pyridone and aza-pyridone compounds and methods of use
JP5841602B2 (en) Pyridinone / pyrazinone, methods for making and using the same
RU2622391C2 (en) Compound 8-fluorophthalazine-1(2h)-one as inhibitors of bruton tyrosine kinase
TWI494314B (en) 5,7-substituted-imidazo(1,2-c)pyrimidines
IL274938B1 (en) Novel compounds and pharmaceutical compositions thereof for the treatment of diseases
WO2008138842A1 (en) Imidazopyrazines and triazolopyrazine for the treatment of joint degenerative and inflammatory diseases
CN118005640A (en) GCN2 inhibitors and uses thereof
MXPA06013250A (en) Kinase inhibitors as therapeutic agents.
WO2008138843A1 (en) Imidazopyridines and triazolopyrimidines useful for the treatment of joint degenerative & inflammatory diseases
JP2013522334A (en) Pyrrolopyrazine kinase inhibitors
CA2821712A1 (en) Substituted n-(1h-indazol-4-yl)imidazo[1,2-a]pyridine-3-carboxamide compounds as type iii receptor tyrosine kinase inhibitors
WO2009135885A1 (en) Fused pyrazine compounds useful for the treatment of degenerative and inflammatory diseases
EP2029602B1 (en) Triazolopyrazine compounds useful for the treatment of degenerative & inflammatory diseases
CN101454326A (en) Triazolopyrazine compounds useful for the treatment of degenerative & inflammatory diseases
ES2345066T3 (en) USEFUL TRIAZOLOPIRAZINE COMPOUNDS FOR THE TREATMENT OF DEGENERATIVE AND INFLAMMATORY DISEASES.

Legal Events

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

Ref document number: 200780019156.2

Country of ref document: CN

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

Ref document number: 07729660

Country of ref document: EP

Kind code of ref document: A2

WWE Wipo information: entry into national phase

Ref document number: 2007729660

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 195033

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 2008111842

Country of ref document: EG

WWE Wipo information: entry into national phase

Ref document number: 2007267121

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 573015

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 2653506

Country of ref document: CA

Ref document number: CR2008-010458

Country of ref document: CR

WWE Wipo information: entry into national phase

Ref document number: MX/a/2008/015057

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 12008502621

Country of ref document: PH

Ref document number: 08126436

Country of ref document: CO

WWE Wipo information: entry into national phase

Ref document number: 2009512585

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 10012/DELNP/2008

Country of ref document: IN

ENP Entry into the national phase

Ref document number: 2007267121

Country of ref document: AU

Date of ref document: 20070530

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 1020087030751

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 200870592

Country of ref document: EA

WWE Wipo information: entry into national phase

Ref document number: A200815160

Country of ref document: UA

WWE Wipo information: entry into national phase

Ref document number: P-2010/0332

Country of ref document: RS

ENP Entry into the national phase

Ref document number: PI0712531

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20081128