US20220281846A1 - Compounds and compositions for the treatment of respiratory diseases - Google Patents

Compounds and compositions for the treatment of respiratory diseases Download PDF

Info

Publication number
US20220281846A1
US20220281846A1 US17/291,694 US201917291694A US2022281846A1 US 20220281846 A1 US20220281846 A1 US 20220281846A1 US 201917291694 A US201917291694 A US 201917291694A US 2022281846 A1 US2022281846 A1 US 2022281846A1
Authority
US
United States
Prior art keywords
alkyl
alkylc
cycloalkyl
compound
prodrug
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
US17/291,694
Other languages
English (en)
Inventor
Alastair Stewart
Spencer Williams
Zalihe Hakki
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Tianli Biotech Pty Ltd
Original Assignee
University of Melbourne
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 claimed from AU2018904242A external-priority patent/AU2018904242A0/en
Application filed by University of Melbourne filed Critical University of Melbourne
Assigned to THE UNIVERSITY OF MELBOURNE reassignment THE UNIVERSITY OF MELBOURNE ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: STEWART, ALASTAIR, HAKKI, Zalihe, WILLIAMS, SPENCER
Publication of US20220281846A1 publication Critical patent/US20220281846A1/en
Assigned to TIANLI BIOTECH PTY LTD reassignment TIANLI BIOTECH PTY LTD ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: THE UNIVERSITY OF MELBOURNE
Pending legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/14Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/05Isotopically modified compounds, e.g. labelled

Definitions

  • the present invention provides imidazole-based compounds that show potential in the treatment of respiratory diseases, such as asthma and related conditions, acute and chronic inflammatory conditions, and fibrotic diseases or conditions in which fibrosis contributes to the pathology of the condition.
  • respiratory diseases such as asthma and related conditions, acute and chronic inflammatory conditions, and fibrotic diseases or conditions in which fibrosis contributes to the pathology of the condition.
  • Asthma is a syndrome that encompasses various different types of diseases, which vary in their severity and in their causes and triggers.
  • the common features of the asthma syndrome are reversible airway obstruction, airway hyper-responsiveness and airway inflammation, with infiltration of the airway wall by eosinophils and T lymphocytes the most prominent features in addition to activation of mast cells.
  • SABA and LABA short- and long-acting ⁇ 2 -adrenoceptor selective agonists
  • ICS inhaled corticosteroids
  • SAMA and LAMA Short and long-acting muscarinic receptor antagonists
  • SAMA and LAMA are used in some patients, usually in combination with other bronchodilators and anti-inflammatory drugs, especially ICS.
  • Leukotriene receptor antagonists (LTRA) may also be added to different therapeutic regimens. More recently, the monoclonal antibody mepolizumab, which neutralises a chemoattractant for eosinophils, interleukin-5, has been shown to have benefit additional to the ICS and LABA combinations in selected patients.
  • exacerbations are still symptomatic and have periodic worsening of disease, referred to as exacerbations.
  • exacerbations There is a considerable unmet need in the drug treatment of severe asthma.
  • these asthma exacerbations are considered to be caused by respiratory viral infection of the lower respiratory tract.
  • the viruses that cause these exacerbations include respiratory syncytial virus, influenza virus and rhinoviruses, which infect the respiratory epithelium.
  • the epithelium of asthmatic individuals is considered to be especially susceptible to such infections and is implicated in the worsening of the inflammatory response.
  • TGF- ⁇ is able to compromise the effectiveness of ICS. Furthermore, the inventors have demonstrated that viral infection of the airway epithelium compromises ICS activity through induction of TGF- ⁇ activity. Drug targeting of TGF- ⁇ carries risk of autoimmune and mitral valve defects. The inventors surprisingly identified casein kinase 1 ⁇ / ⁇ as a mediator of TGF- ⁇ induced ICS insensitivity, using the compound PF670462 (WO2016/149756), and demonstrated the utility of this agent to reverse steroid insensitivity.
  • Described herein is a compound of formula (I) or a salt, solvate, N-oxide, tautomer, stereoisomer, polymorph and/or prodrug thereof:
  • R 1 and R 2 are each independently selected from the group consisting of H, C 1-6 alkyl, C 1 alkylC 6 aryl, C 3-6 cycloalkyl and C 3-6 heterocyclyl;
  • R 3 is selected from the group consisting of F, Cl and CH 3 ;
  • R 4 is selected from the group consisting of C 0-3 alkylC 3-12 cycloalkyl and C 1-12 alkyl; wherein each of R 1 , R 2 , R 3 and R 4 is optionally substituted.
  • the invention provides a compound of formula (I), or salt, solvate, N-oxide, tautomer, stereoisomer, polymorph and/or prodrug thereof,
  • R 1 is selected from the group consisting of H, C 2-6 alkyl, hydroxyC 1-6 alkyl, C 1 alkylC 6 aryl, C 1 alkylC 12 aryl, C 1 alkylC 6 arylhalo, C 1 alkyl(C 1 alkyl)C 6 aryl, C 3-6 cycloalkyl, haloC 3-6 cycloalkyl, hydroxyC 3-6 cycloalkyl, and C 3-5 heterocyclyl;
  • R 2 is H
  • R 3 is selected from the group consisting of F, Cl and CH 3 ;
  • R 4 is selected from the group consisting of C 0-3 alkylC 3-12 cycloalkyl, C 0-3 alkylC 3-12 cycloalkylhalo, C 1-12 alkyl and haloC 1-12 alkyl; wherein each of R 1 , R 2 , R 3 and R 4 is optionally substituted; and wherein when R 1 is H, R 4 is selected from the group consisting of C 0-3 alkylC 4-12 cycloalkyl and C 1-12 alkyl, wherein when R 4 is C 6 cycloalkyl, R 4 is substituted by one or more groups selected from C 1-6 alkyl, C 1-6 haloalkyl, aryl, heteroaryl, and halo, and when R 4 is C 1-3 alkyl, R 4 is substituted by one or more halo groups, and when R 1 is C 3 cycloalkyl, C 1 alkylC 6
  • a method of treating or preventing a respiratory disease in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a compound of formula (I) or a salt, solvate, N-oxide, tautomer, stereoisomer, polymorph and/or prodrug thereof, thereby treating or preventing the respiratory disease in the subject.
  • the respiratory disease may be selected from asthma, chronic obstructive pulmonary disease, interstitial lung diseases (such as idiopathic pulmonary fibrosis) and other conditions relating to tissue remodelling, primary or secondary lung tumour, hayfever, chronic and acute sinusitis, and chronic and acute viral, fungal and bacterial infections of the respiratory tract.
  • interstitial lung diseases such as idiopathic pulmonary fibrosis
  • a method of improving respiratory function in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a compound of formula (I) or a salt, solvate, N-oxide, tautomer, stereoisomer, polymorph and/or prodrug thereof, thereby improving respiratory function of the subject.
  • the improvement in respiratory function may be selected from a decrease in the level of constriction of the lungs, a decrease in the elastic stiffness of the respiratory system, and/or an increase in the ease with which the respiratory system can be extended.
  • the improvement is selected from a decrease in the level of constriction of the lungs, and a decrease in the elastic stiffness of the respiratory system.
  • a composition comprising a compound according to formula (I) or a salt, solvate, N-oxide, tautomer, stereoisomer, polymorph and/or prodrug thereof, and a pharmaceutically acceptable excipient.
  • composition may be formulated for oral administration or administration by inhalation or injection.
  • FIG. 1 Defines a number of known compounds.
  • FIG. 2 P IC 50 values (the negative log of the concentration suppressing IL-11 level by 50%) for inhibition of TGF- ⁇ -induced IL-11 (interpolated from the linear regression of log concentration small molecule versus IL-11 level).
  • FIG. 3 Defines compounds disclosed in WO2018/201192.
  • FIG. 4 Plasma concentrations over time following IV or oral administration of PF670462 (A) and ZH3-138 (B) to male C57BL/6 mice.
  • FIG. 5 Comparison of PF670462 and ZH3-138 for inhibition of TGF- ⁇ -induced IL-11 levels (A—a chart of concentration of IL-11 in supernatant taken from MRC5 cells following exposure to vehicle, PF670462 and ZH3-138 at 0.1 ⁇ M, 1 ⁇ M and 10 ⁇ M; B—a chart of IL-11 concentration in supernatant of A549 cells treated with various concentrations of PF670462 or ZH3-138 30 minutes prior to TGF- ⁇ for 24 h).
  • A a chart of concentration of IL-11 in supernatant taken from MRC5 cells following exposure to vehicle, PF670462 and ZH3-138 at 0.1 ⁇ M, 1 ⁇ M and 10 ⁇ M
  • B a chart of IL-11 concentration in supernatant of A549 cells treated with various concentrations of PF670462 or ZH3-138 30 minutes prior to TGF- ⁇ for 24 h).
  • FIG. 6 Effects of ZH3-126 on TGF- ⁇ -induced IL-11 concentration in supernatant taken from MRC5 cells at 0.1 ⁇ M, 1 ⁇ M and 10 ⁇ M compared to vehicle.
  • FIG. 7 Comparison of effects of PF670462 and compounds of the invention (ZH3-126 and ZH3-138) on supernatant concentrations of various inflammatory biomarkers (A: IL-6; B: IL-8; C: GM-CSF) across a range of concentrations.
  • the present invention provides a compound of formula (I) or a salt, solvate, N-oxide, tautomer, stereoisomer, polymorph and/or prodrug thereof:
  • R 1 and R 2 are each independently selected from the group consisting of H, C 1-6 alkyl, C 1 alkylC 6 aryl, C 3-6 cycloalkyl and C 3-6 heterocyclyl;
  • R 3 is selected from the group consisting of F, Cl and CH 3 ;
  • R 4 is selected from the group consisting of C 0-3 alkylC 3-12 cycloalkyl and C 1-12 alkyl; wherein each of R 1 , R 2 , R 3 and R 4 is optionally substituted.
  • the invention provides a compound of formula (I),
  • R 1 is selected from the group consisting of H, C 2-6 alkyl, hydroxyC 1-6 alkyl, C 1 alkylC 6 aryl, C 1 alkylC 12 aryl, C 1 alkylC 6 arylhalo, C 1 alkyl(C 1 alkyl)C 6 aryl, C 3-6 cycloalkyl, haloC 3 -cycloalkyl, hydroxyC 3-6 cycloalkyl, and C 3-5 heterocyclyl;
  • R 2 is H
  • R 3 is selected from the group consisting of F, Cl and CH 3 ;
  • R 4 is selected from the group consisting of C 0-3 alkylC 3-12 cycloalkyl, C 0-3 alkylC 3-12 cycloalkylhalo, C 1-12 alkyl and haloC 1-12 alkyl; wherein each of R 1 , R 2 , R 3 and R 4 is optionally substituted wherein when R 1 is H, R 4 is selected from the group consisting of C 0-3 alkylC 4-12 cycloalkyl and C 1-12 alkyl, wherein when R 4 is C 6 cycloalkyl, R 4 is substituted by one or more groups selected from C 1-6 alkyl, C 1-6 haloalkyl, aryl, heteroaryl, and halo, and when R 4 is C 1-3 alkyl, R 4 is substituted by one or more halo groups, and when R 1 is C 3 cycloalkyl, C 1 alkylC 6 aryl
  • the compounds of Formula (I) are inhibitors of CK1 ⁇ .
  • at least preferred embodiments of these compounds are selective inhibitors of CK1 ⁇ compared with other kinases.
  • Selective CK1 ⁇ inhibitors may avoid undesired effects associated with the activity of the other kinases (ie the kinases for which the CK1 ⁇ inhibitors select against).
  • at least preferred embodiments provided lower clearance rates following administration (eg IV or oral administration) compared to known CK1 ⁇ inhibitors, such as PF670462.
  • the present invention provides a compound of formula (I) provided that the compound is not selected from the list of compounds in FIGS. 1 and/or 3 .
  • Various compounds are also described in Keenan, et al. Frontiers in Pharmacology, 2018, vol 9, article 738; WO2016/149756A1; WO1996/021654A1; WO1999/032121A1; WO1997/035856A1; Kim, D-K., et al. Bioorganic & Medicinal Chemistry Letters, 2008, 18, 4006-4010; WO1999/001136A1; U.S. Pat. No. 6,369,068B1; and WO2018/201192A1.
  • any specific compound described in one or more of these documents may be excluded by way of proviso.
  • R 1 is C 2-6 alkyl, preferably C 2-3 alkyl.
  • R 2 may be H.
  • R 1 may be C 1-3 alkyl substituted with one or more hydroxyl groups.
  • R 1 may be hydroxyC 1-6 alkyl. In some embodiments, the hydroxyC 1-6 alkyl comprises one hydroxyl substituent.
  • the present invention provides a compound of formula (I) wherein R 2 is H.
  • R 1 is C 3-6 cycloalkyl.
  • R 1 may be selected from cyclobutyl, cyclopentyl and cyclohexyl.
  • R 1 may be substituted or unsubstituted.
  • the substituent may be selected from one or more OH groups and/or one or more halo groups.
  • R 1 is C 3-6 cycloalkyl, R 2 may be H.
  • R 1 is haloC 3-6 cycloalkyl.
  • R 1 may comprise 1 or 2 halo groups, which may be the same or different.
  • R 1 comprises 2 halo groups, which may be attached to the same carbon atom.
  • the haloC 3-6 cycloalkyl is a fluoroC 3-6 cycloalkyl.
  • the halo group is in a para position relative to the point of attachment of R 1 to the pyrimidyl amine of formula (I).
  • R 1 is hydroxyC 3-6 cycloalkyl.
  • R 1 may comprise 1 hydroxy group.
  • the hydroxy group is in a para position relative to the point of attachment to the pyrimidyl amine of formula (I).
  • R 1 is C 3-6 heterocyclyl.
  • R 1 may be selected from an oxygen-containing heterocyclyl group, a nitrogen-containing heterocyclyl group, or a sulphur-containing heterocyclyl group, or a heterocyclyl group containing a combination of two or more oxygen, nitrogen and sulphur atoms.
  • R 1 is an optionally substituted 4-7 membered heterocyclyl or a 4-6 membered heterocyclyl.
  • R 1 is selected from oxetanyl, thietanyl, pyrrolidinyl, pyrazolidinyl, imidazolidinyl, terahydrofuranyl, 1,3-dioxolanyl, tetrahydropyranyl, tetrahydrothipheneyl, and 1,2- and 1,3-oxathiolanyl groups.
  • R 1 when R 1 is C 3-5 heterocyclyl, R 2 is H.
  • the present invention provides a compound of formula (I) wherein R 1 and R 2 are both the same.
  • R 1 and R 2 may both be H, or R 1 and R 2 may both be C 1-6 alkyl (e.g. methyl, ethyl, propyl or butyl).
  • R 1 is C 1 alkylC 6 aryl.
  • the C 1 alkylC 6 aryl group may be substituted.
  • the substituents may be selected from one or more alkyl groups, one or more hydroxyl groups and one or more halo groups.
  • the aryl group may be substituted.
  • the alkyl group may be substituted.
  • R 1 is C 1 alkylC 12 aryl.
  • the C 12 aryl is biphenyl.
  • the covalent bond between the phenyl rings is para relative to the C 1 alkyl moiety.
  • the C 1 alkylC 12 aryl group is connected to the pyrimidyl amine through the C 1 alkyl moiety.
  • R 1 is C 1 alkylC 6 arylhalo.
  • the halo moiety is a substituent of the C 6 aryl ring.
  • the halo group may be at any one or more of ortho, meta or para position(s) relative to the C 1 alkyl moiety.
  • the C 1 alkylC 6 arylhalo is a C 1 alkylC 6 arylfluoro.
  • the C 1 alkylC 6 arylhalo group is connected to the pyrimidyl amine through the C 1 alkyl moiety.
  • R 1 is C 1 alkyl(C 1 alkyl)C 6 aryl.
  • the C 1 alkyl(C 1 alkyl)C 6 aryl group is connected to the pyrimidyl amine through the C 1 alkyl moiety, which may be denoted as —C 1 alkyl(C 1 alkyl)C 6 aryl.
  • R 1 is selected from H, ethyl, hydroxyethyl, halobenzyl, hydroxypropyl, cyclopropyl, cyclobutyl, oxetanyl, halocyclobutyl (eg difluorocyclobutyl), hydroxycyclobutyl, cyclopentyl, cyclohexyl, halocyclohexyl (eg difluorocyclohexyl), pyranyl and S,S-dioxythianyl.
  • R 3 is CH 3 . In any one of the embodiments, R 3 is F or Cl. Preferably, R 3 is F.
  • R 4 is C 0-3 alkylC 3-12 cycloalkyl.
  • R 4 may be C 0-3 alkylC 3-12 cycloalkyl, wherein the C 3-12 cycloalkyl group is selected from cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and cycloheptyl.
  • R 4 is C 3-12 cycloalkyl. More preferably, R 4 is selected from cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and cycloheptyl.
  • R 4 is C 1-2 alkylC 3-12 cycloalkyl.
  • R 4 may be C 1 alkylC 3-12 cycloalkyl.
  • the C 3-12 cycloalkyl group may be selected from cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and cycloheptyl.
  • R 4 may be substituted.
  • the substituent may be selected from one or more OH groups, one or more C 1-6 alkyl groups, and one or more halo groups.
  • R 4 is selected from C 0-3 alkylC 4-12 cycloalkyl and C 3 cycloalkyl and C 2-3 alkylC 3 cycloalkyl, each of which may be optionally substituted.
  • R 4 is C 0-3 alkylC 3-12 cycloalkylhalo.
  • the halo is a substituent of the cycloalkyl moiety.
  • the group may comprise 1 or 2 halo groups. In embodiments comprising 2 halo groups, these may be attached to the same carbon atom.
  • the halo substituent is attached to a position of the cycloalkyl group para to the C 0-3 alkyl moiety or the point of attachment to the imidazole of formula (I).
  • the C 3-12 cycloalkyl and/or C 0-3 alkyl moiety of the C 0-3 alkylC 3-12 cycloalkylhalo group may be any of the preferred C 0-3 alkyl and/or C 3-12 cycloalkyl groups described for any embodiment of R 4 .
  • Preferred groups include dihalocyclobutyl (eg 3,3-difluorocyclobut-1-yl), dihalocyclobutylmethyl (eg 3,3-difluorocyclobutyl-1-methyl), dihalocyclohexyl (eg 4,4-difluorocyclohex-1-yl) and dihalocyclohexylmethyl (eg 4,4-difluorocyclohexyl-1-methyl).
  • the halo is fluoro.
  • the C 0-3 alkylC 3-12 cycloalkylhalo is connected to the imidazole nitrogen through the C 0-3 alkyl moiety (when present) which may be denoted as —C 0-3 alkylC 3-12 cycloalkyl.
  • R 4 is C 1-12 alkyl.
  • R 4 may be a methyl, ethyl, propyl or butyl group.
  • R 4 may be a branched C 1-12 alkyl group, such as a branched C 3 , C 4 or C 5 alkyl group.
  • R 4 may be substituted by one or more groups selected from halo and OH.
  • R 4 may be substituted by one, two, or more halo groups.
  • R 4 is haloC 1-12 alkyl.
  • R 4 is a haloC 1-6 alkyl or a haloC 1-4 alkyl.
  • the group may comprise 1, 2 or 3 halo groups. In embodiments comprising 2 or more halo groups, these may be attached to the same carbon atom.
  • the halo group is attached the carbon atom distal from the point of attachment to the imidazole moiety of formula (I).
  • the halo is fluoro.
  • R 4 is selected from trihalomethyl, trihaloethyl (eg 2,2,2-trifluoroeth-1-yl), dihalomethyl (eg difluoromethyl), halomethyl (eg fluoromethyl), dihaloethyl (eg 2,2-difluoroeth-1-yl), hexahalopropyl (eg 1,1,1,3,3,3-hexafluoroprop-2-yl), trihalopropyl (eg 3,3,3-trifluoroprop-1-yl and 1,1,1-trifluoroprop-2-yl), halopropyl (eg 2-fluoroprop-2-yl), dihalopropyl (eg 3,3-difluoroprop-1-yl), haloethyl (eg 2-fluoroeth-1-yl).
  • trihalomethyl eg 2,2,2-trifluoroeth-1-yl
  • dihalomethyl eg difluoromethyl
  • R 4 is selected from ethyl, pentyl, cyclopentyl, cyclohexyl, halocyclohexylmethyl, halocyclohexyl and haloethyl. In some embodiments, R 4 is selected from ethyl, pentyl, cyclopentyl, cyclohexyl, difluorocyclohexyl (eg 4,4-diflurocyclohex-1-yl), difluorocyclohexylmethyl (eg 4,4-diflurocyclohexyl-1-methyl) and trifluoroethyl (eg 2,2,2-trifluoroeth-1-yl).
  • difluorocyclohexyl eg 4,4-diflurocyclohex-1-yl
  • difluorocyclohexylmethyl eg 4,4-diflurocyclohexyl-1-methyl
  • R 4 when R 1 is H, R 4 is selected from the group consisting of C 0-3 alkylC 4-12 cycloalkyl and C 1-12 alkyl. In these embodiments, when R 4 is C 6 cycloalkyl, R 4 is substituted by one or more groups selected from C 1-6 alkyl, C 1-6 haloalkyl, aryl, heteroaryl, and halo, preferably halo (eg fluoro); and when R 4 is C 1-3 alkyl, R 4 is substituted by one or more halo groups.
  • R 4 is selected from haloC 1-12 alkyl and C 0-3 alkylC 3-12 cycloalkyl.
  • R 1 , R 2 , R 3 and R 4 are optionally substituted by one or more groups selected from OH, C 1-6 alkoxy, halo, amino, mercapto and C 1-6 alkyl. In some embodiments, R 1 , R 2 , R 3 and R 4 are unsubstituted.
  • C 1-12 alkyl refers to a straight or branched chain hydrocarbon radical having from one to twelve carbon atoms, or any range between, i.e. it contains 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12 carbon atoms.
  • the alkyl group is optionally substituted with substituents, multiple degrees of substitution being allowed.
  • Examples of “C 1-12 alkyl” as used herein include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, t-butyl, n-pentyl, isopentyl, neopentyl and the like.
  • C 1-3 alkyl “C 1-4 alkyl” and “C 1-6 alkyl” are preferred. These groups refer to an alkyl group containing 1-3, 1-4 or 1-6 carbon atoms, respectively, or any range in between (e.g. alkyl groups containing 2-5 carbon atoms, i.e. 2, 3, 4 or 5 carbon atoms, are also within the range of C 1-6 ). Where the term “C 0-2 alkyl”, or the like, is used, there may be no alkyl group, or an alkyl group containing 1 or 2 carbon atoms.
  • C 2-6 alkenyl refers to optionally substituted straight chain or branched chain hydrocarbon groups having at least one double bond of either E or Z stereochemistry where applicable and 2 to 6 carbon atoms. Examples include vinyl, 1-propenyl, 1- and 2-butenyl and 2-methyl-2-propenyl. Unless the context requires otherwise, the term “C 2-6 alkenyl” also encompasses alkenyl groups containing one less hydrogen atom such that the group is attached via two positions i.e. divalent. “C 2-4 alkenyl” and “C 2-3 alkenyl” including ethenyl, propenyl and butenyl are preferred with ethenyl being particularly preferred.
  • C 2-6 alkynyl refers to optionally substituted straight chain or branched chain hydrocarbon groups having at least one triple bond and 2 to 6 carbon atoms. Examples include ethynyl, 1-propynyl, 1- and 2-butynyl, 2-methyl-2-propynyl, 2-pentynyl, 3-pentynyl, 4-pentynyl, 2-hexynyl, 3-hexynyl, 4-hexynyl and 5-hexynyl and the like. Unless the context indicates otherwise, the term “C 2-6 alkynyl” also encompasses alkynyl groups containing one less hydrogen atom such that the group is attached via two positions i.e. divalent. C 2-3 alkynyl is preferred.
  • halogen refers to fluorine (F), chlorine (Cl), bromine (Br), or iodine (I) and the term “halo” refers to the halogen radicals fluoro (—F), chloro (—Cl), bromo (—Br), and iodo (—I).
  • halo is fluoro.
  • cycloalkyl refers to a non-aromatic cyclic hydrocarbon ring.
  • C 3-7 cycloalkyl refers to a non-aromatic cyclic hydrocarbon ring having from three to seven carbon atoms, or any range of integers in between.
  • the C 3-7 cycloalkyl group would also include cycloalkyl groups containing 4 to 6 (i.e. 4, 5 or 6) carbon atoms.
  • the alkyl group is as defined above, and may be substituted.
  • C 3-7 cycloalkyl groups useful in the present invention include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and cycloheptyl.
  • Cycloalkyl groups may optionally be fused to one or more heterocyclic or cycloalkyl rings. Cycloalkyl rings may be substituted at any of the carbon atoms on the ring with another cycloalkyl or heterocyclic moiety to form a spirocycloalkyl or spiroheteroalkyl compound.
  • Two non-adjacent atoms on the cycloalkyl group may be bridged by an alkyl or heteroalkyl group to form a bridged system.
  • the bridging group is 1-3 atoms in length.
  • heterocyclic or “heterocyclyl” refer to a non-aromatic heterocyclic ring, being saturated or having one or more degrees of unsaturation, containing one or more heteroatom substitution selected from S, S(O), S(O) 2 , O, or N.
  • C 3-7 heterocyclyl refers to a non-aromatic cyclic hydrocarbon ring having from three to seven carbon atoms (i.e. 3, 4, 5, 6 or 7 carbon atoms) containing one or more heteroatom substitutions as referred to herein.
  • the heterocyclic moiety may be substituted, multiple degrees of substitution being allowed.
  • C 3-7 heterocyclyl also includes heterocyclyl groups containing C 4-5 , C 5-7 , C 6-7 , C 4-7 , C 4-6 and C 5-6 carbon atoms.
  • the heterocyclic ring contains four to six carbon atoms and one or two heteroatoms. More preferably, the heterocyclic ring contains five carbon atoms and one heteroatom, or four carbon atoms and two heteroatom substitutions, or five carbon atoms and one heteroatom.
  • the heterocyclyl groups may be 3 to 10-membered ring systems, which denotes the total number of atoms (carbon atoms and heteroatoms) contained within the ring system.
  • the prefixs 3-, 4-, 5-, 6-, 7-, 8-, 9- and 10-membered denote the number of ring atoms, or range of ring atoms, whether carbon atoms or heteroatoms.
  • the term “3-10 membered heterocylyl”, as used herein, pertains to a heterocyclyl group having 3, 4, 5, 6, 7, 8, 9 or 10 ring atoms.
  • heterocylyl groups include 5-6-membered monocyclic heterocyclyls and 9-10 membered fused bicyclic heterocyclyls.
  • heterocyclyl heterocyclyl rings may be optionally fused to one or more other “heterocyclic” ring(s), cycloalkyl ring(s), aryl ring(s) or heteroaryl ring(s).
  • heterocyclic moieties include, but are not limited to, tetrahydrofuran, pyran, oxetane, 1,4-dioxane, 1,3-dioxane, piperidine, piperazine, N-methylpiperazinyl, 2,4-piperazinedione, pyrrolidine, imidazolidine, pyrazolidine, morpholine, thiomorpholine, tetrahydrothiopyran, tetrahydrothiophene, and the like.
  • Heterocyclic groups may be substituted at any of the carbons on the ring with another heterocyclic or cycloalkyl moiety to form a spirocycloalkyl or spiroheterocyclyl compound.
  • Two non-adjacent atoms on the heterocyclic group may further be bridged by an alkyl or heteroalkyl group to form a bridged system.
  • the bridging group is 1-3 atoms in length.
  • C 0-2 alkylC 3-7 heterocyclyl includes heterocyclyl groups containing either no alkyl group as a linker between the compound and the heterocycle, or an alkyl group containing 1 or 2 carbon atoms as a linker between the compound and the heterocycle (i.e. heterocycle, —CH 2 -heterocycle or —CH 2 CH 2 -heterocycle). These heterocycles may be further substituted.
  • Substituted cycloalkyl and heterocyclyl groups may be substituted with any suitable substituent as described below.
  • aryl refers to an optionally substituted benzene ring or to an optionally substituted benzene ring system fused to one or more optionally substituted benzene rings to form, for example, anthracene, phenanthrene, or naphthalene ring systems.
  • aryl groups include, but are not limited to, phenyl, 2-naphthyl, 1-naphthyl, biphenyl, as well as substituted derivatives thereof.
  • Preferred substituted aryl groups include arylamino, arylalkyl, arylalkylhalo, arylhalo, and aralkoxy groups.
  • heteroaryl refers to a monocyclic five, six or seven membered aromatic ring, or to a fused bicyclic or tricyclic aromatic ring system comprising at least one monocyclic five, six or seven membered aromatic ring.
  • These heteroaryl rings contain one or more nitrogen, sulfur, and/or oxygen heteroatoms, and may be optionally substituted with up to three members.
  • N-containing heteroaryls may be in the form of an N-oxide and S-containing heteroaryls may be in the form of sulfur oxides and dioxides.
  • heteroaryl groups used herein include furanyl, thiophenyl, pyrrolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, thiazolyl, oxazolyl, isoxazolyl, oxadiazolyl, oxo-pyridyl, thiadiazolyl, isothiazolyl, pyridyl, pyridazyl, pyrazinyl, pyrimidyl, quinolinyl, isoquinolinyl, benzofuranyl, benzothiophenyl, indolyl, indazolyl, benzimidazolyl, and substituted versions thereof.
  • hydroxy and “hydroxyl” refer to the group —OH.
  • oxo refers to the group ⁇ O.
  • C 1-6 alkoxy refers to an alkyl group as defined above covalently bound via an O linkage containing 1 to 6 carbon atoms, such as methoxy, ethoxy, propoxy, isoproxy, butoxy, tert-butoxy and pentoxy.
  • C 1-4 alkoxy and “C 1-3 alkoxy” including methoxy, ethoxy, propoxy and butoxy are preferred with methoxy being particularly preferred.
  • haloC 1-6 alkyl and “C 1-6 alkylhalo” refer to a C 1-6 alkyl which is substituted with one or more halogens.
  • HaloC 1-3 alkyl groups are preferred, such as for example, —CH 2 CF 3 , and —CF 3 .
  • haloC 1-6 alkoxy and “C 1-6 alkoxyhalo” refer to a C 1-6 alkoxy which is substituted with one or more halogens.
  • C 1-3 alkoxyhalo groups are preferred, such as for example, —OCF 3 .
  • carboxylate or “carboxyl” refers to the group —COO— or —COOH.
  • esters refers to a carboxyl group having the hydrogen replaced with, for example a C 1-6 alkyl group (“carboxylC 1-6 alkyl” or “alkylester”), an aryl or aralkyl group (“arylester” or “aralkylester”) and so on.
  • CO 2 C 1-3 alkyl groups are preferred, such as for example, methylester (CO 2 Me), ethylester (CO 2 Et) and propylester (CO 2 Pr) and includes reverse esters thereof (e.g. —OC(O)Me, —OC(O)Et and —OC(O)Pr).
  • cyano and “nitrile” refer to the group —CN.
  • nitro refers to the group —NO 2 .
  • amino refers to the group —NH 2 .
  • substituted amino or “secondary amino” refers to an amino group having a hydrogen replaced with, for example a C 1-6 alkyl group (“C 1-6 alkylamino”), an aryl or aralkyl group (“arylamino”, “aralkylamino”) and so on.
  • C 1-3 alkylamino groups are preferred, such as for example, methylamino (NHMe), ethylamino (NHEt) and propylamino (NHPr).
  • disubstituted amino or “tertiary amino” refers to an amino group having the two hydrogens replaced with, for example a C 1-6 alkyl group, which may be the same or different (“dialkylamino”), an aryl and alkyl group (“aryl(alkyl)amino”) and so on.
  • Di(C 1-3 alkyl)amino groups are preferred, such as for example, dimethylamino (NMe 2 ), diethylamino (NEt 2 ), dipropylamino (NPr 2 ) and variations thereof (e.g. N(Me)(Et) and so on).
  • aldehyde refers to the group —C( ⁇ O)H.
  • acyl refers to the group —C(O)CH 3 .
  • ketone refers to a carbonyl group which may be represented by —C(O)—.
  • substituted ketone refers to a ketone group covalently linked to at least one further group, for example, a C 1-6 alkyl group (“C 1-6 alkylacyl” or “alkylketone” or “ketoalkyl”), an aryl group (“arylketone”), an aralkyl group (“aralkylketone) and so on.
  • C 1-3 alkylacyl groups are preferred.
  • amido or “amide” refers to the group —C(O)NH 2 .
  • substituted amido or “substituted amide” refers to an amido group having a hydrogen replaced with, for example a C 1-6 alkyl group (“C 1-6 alkylamido” or “C 1-6 alkylamide”), an aryl (“arylamido”), aralkyl group (“aralkylamido”) and so on.
  • C 1-3 alkylamide groups are preferred, such as for example, methylamide (—C(O)NHMe), ethylamide (—C(O)NHEt) and propylamide (—C(O)NHPr) and includes reverse amides thereof (e.g. —NHMeC(O)—, —NHEtC(O)— and —NHPrC(O)—).
  • disubstituted amido or “disubstituted amide” refers to an amido group having the two hydrogens replaced with, for example a C 1-6 alkyl group (“di(C 1-6 alkyl)amido” or “di(C 1-6 alkyl)amide”), an aralkyl and alkyl group (“alkyl(aralkyl)amido”) and so on.
  • Di(C 1-3 alkyl)amide groups are preferred, such as for example, dimethylamide (—C(O)NMe 2 ), diethylamide (—C(O)NEt 2 ) and dipropylamide ((—C(O)NPr 2 ) and variations thereof (e.g. —C(O)N(Me)Et and so on) and includes reverse amides thereof.
  • thiol refers to the group —SH.
  • C 1-6 alkylthio refers to a thiol group having the hydrogen replaced with a C 1-6 alkyl group.
  • C 1-3 alkylthio groups are preferred, such as for example, thiolmethyl, thiolethyl and thiolpropyl.
  • thioxo refer to the group ⁇ S.
  • sulfinyl refers to the group —S( ⁇ O)H.
  • substituted sulfinyl or “sulfoxide” refers to a sulfinyl group having the hydrogen replaced with, for example a C 1-6 alkyl group (“C 1-6 alkylsulfinyl” or “C 1-6 alkylsulfoxide”), an aryl (“arylsulfinyl”), an aralkyl (“aralkyl sulfinyl”) and so on.
  • C1-3alkylsulfinyl groups are preferred, such as for example, —SOmethyl, —SOethyl and —SOpropyl.
  • sulfonyl refers to the group —SO 2 H.
  • substituted sulfonyl refers to a sulfonyl group having the hydrogen replaced with, for example a C 1-6 alkyl group (“sulfonylC 1-6 alkyl”), an aryl (“arylsulfonyl”), an aralkyl (“aralkylsulfonyl”) and so on.
  • SulfonylC 1-3 alkyl groups are preferred, such as for example, —SO 2 Me, —SO 2 Et and —SO 2 Pr.
  • sulfonylamido or “sulfonamide” refers to the group —SO 2 NH 2 .
  • substituted sulfonamido or “substituted sulphonamide” refers to an sulfonylamido group having a hydrogen replaced with, for example a C 1-6 alkyl group (“sulfonylamidoC 1-6 alkyl”), an aryl (“arylsulfonamide”), aralkyl (“aralkylsulfonamide”) and so on.
  • SulfonylamidoC 1-3 alkyl groups are preferred, such as for example, —SO 2 NHMe, —SO 2 NHEt and —SO 2 NHPr and includes reverse sulfonamides thereof (e.g. —NHSO 2 Me, —NHSO 2 Et and —NHSO 2 Pr).
  • disubstituted sufonamido or “disubstituted sulphonamide” refers to an sulfonylamido group having the two hydrogens replaced with, for example a C 1-6 alkyl group, which may be the same or different (“sulfonylamidodi(C 1-6 alkyl)”), an aralkyl and alkyl group (“sulfonamido(aralkyl)alkyl”) and so on.
  • Sulfonylamidodi(C 1-3 alkyl) groups are preferred, such as for example, —SO 2 NMe 2 , —SO 2 NEt 2 and —SO 2 NPr 2 and variations thereof (e.g. —SO 2 N(Me)Et and so on) and includes reserve sulfonamides thereof (e.g. —N(Me)SO 2 Me and so on).
  • sulfate refers to the group OS(O) 2 OH and includes groups having the hydrogen replaced with, for example a C 1-6 alkyl group (“alkylsulfates”), an aryl (“arylsulfate”), an aralkyl (“aralkylsulfate”) and so on.
  • alkylsulfates groups having the hydrogen replaced with, for example a C 1-6 alkyl group
  • arylsulfate an aryl
  • aralkyl aralkyl
  • C 1-3 sulfates are preferred, such as for example, OS(O) 2 OMe, OS(O) 2 OEt and OS(O) 2 OPr.
  • sulfonate refers to the group SO 3 H and includes groups having the hydrogen replaced with, for example a C 1-6 alkyl group (“alkylsulfonate”), an aryl (“arylsulfonate”), an aralkyl (“aralkylsulfonate”) and so on.
  • alkylsulfonate a C 1-6 alkyl group
  • arylsulfonate an aryl
  • aralkyl aralkylsulfonate
  • C 1-3 sulfonates are preferred, such as for example, SO 3 Me, SO 3 Et and SO 3 Pr.
  • a “ring substituent” may be a moiety such as a halogen, alkyl group, or other substituent described herein that is covalently bonded to an atom, preferably a carbon or nitrogen atom, that is a ring member.
  • substituted means that any one or more hydrogens on the designated atom is replaced with a selection from the indicated substituents, provided that the designated atom's normal valence is not exceeded, and that the substitution results in a stable compound, i.e., a compound that can be isolated, characterized and tested for biological activity.
  • an optionally substituted moiety may or may not be further substituted with 1, 2, 3, 4 or more groups, preferably 1, 2 or 3, more preferably 1 or 2 groups.
  • substituents include but are not limited to:
  • Optional substituents in the case of heterocycles (heterocyclyl and heteroaryl groups) containing N may also include but are not limited to C 1-6 alkyl i.e. N—C 1-3 alkyl, more preferably methyl particularly N-methyl.
  • cyclic or heterocyclic substituents may form a spirocycloalkyl or spiroheteroalkyl substituent with a carbon in the moiety from which the cyclic or heterocyclic group is substituted.
  • cyclic or heterocyclic substituents may be bridged.
  • optionally substituted “C 1-6 alkyl”, “C 2-6 alkenyl” and “C 2-6 alkynyl”, the optional substituent or substituents are preferably selected from halo, aryl, heterocyclyl, C 3-8 cycloalkyl, C 1-6 alkoxy, hydroxyl, oxo, aryloxy, haloC 1-6 alkyl, haloC 1-6 alkoxyl and carboxyl.
  • the optionally substituted “C 1-6 alkyl”, “C 2-6 alkenyl” and “C 2-6 alkynyl” may be optionally substituted by any subset of optional substituents selected from those described above.
  • any of these groups may be further substituted by any of the above-mentioned groups, where appropriate.
  • salts of the compounds of formula (I) are preferably pharmaceutically acceptable, but it will be appreciated that non-pharmaceutically acceptable salts also fall within the scope of the present disclosure, since these may be useful as intermediates in the preparation of pharmaceutically acceptable salts.
  • pharmaceutically acceptable may be used to describe any pharmaceutically acceptable salt, solvate, tautomer, N-oxide, stereoisomer polymorph and/or prodrug, or any other compound which upon administration to a subject, is capable of providing (directly or indirectly) a compound of formula (I), or an active metabolite or residue thereof and typically that is not deleterious to the subject.
  • Suitable pharmaceutically acceptable salts include, but are not limited to, salts of pharmaceutically acceptable inorganic acids such as hydrochloric, sulphuric, phosphoric, nitric, carbonic, boric, sulfamic, and hydrobromic acids, or salts of pharmaceutically acceptable organic acids such as acetic, propionic, butyric, tartaric, maleic, hydroxymaleic, fumaric, malic, citric, lactic, mucic, gluconic, benzoic, succinic, oxalic, phenylacetic, methanesulphonic, toluenesulphonic, benzenesulphonic, salicylic, sulphanilic, aspartic, glutamic, edetic, stearic, palmitic, oleic, lauric, pantothenic, tannic, ascorbic, valeric and xinafoic acids.
  • pharmaceutically acceptable inorganic acids such as hydrochloric, sulphuri
  • Base salts include, but are not limited to, those formed with pharmaceutically acceptable cations, such as sodium, potassium, lithium, calcium, magnesium, zinc, ammonium, alkylammonium such as salts formed from triethylamine, alkoxyammonium such as those formed with ethanolamine and salts formed from ethylenediamine, choline or amino acids such as arginine, lysine or histidine.
  • pharmaceutically acceptable cations such as sodium, potassium, lithium, calcium, magnesium, zinc, ammonium, alkylammonium such as salts formed from triethylamine, alkoxyammonium such as those formed with ethanolamine and salts formed from ethylenediamine, choline or amino acids such as arginine, lysine or histidine.
  • Basic nitrogen-containing groups may be quarternised with such agents as lower alkyl halide, such as methyl, ethyl, propyl, and butyl chlorides, bromides and iodides; dialkyl sulfates like dimethyl and diethyl sulfate; and others.
  • lower alkyl halide such as methyl, ethyl, propyl, and butyl chlorides, bromides and iodides
  • dialkyl sulfates like dimethyl and diethyl sulfate; and others.
  • a “prodrug” is a compound that may not fully satisfy the structural requirements of the compounds provided herein, but is modified in vivo, following administration to a subject or patient, to produce a compound of formula (I) provided herein.
  • a prodrug may be an acylated derivative of a compound as provided herein.
  • Prodrugs include compounds wherein hydroxy, carboxy, amine or sulfhydryl groups are bonded to any group that, when administered to a mammalian subject, cleaves to form a free hydroxy, carboxy, amino, or sulfhydryl group, respectively.
  • prodrugs include, but are not limited to, acetate, formate, phosphate and benzoate derivatives of alcohol and amine functional groups within the compounds provided herein.
  • Prodrugs of the compounds provided herein may be prepared by modifying functional groups present in the compounds in such a way that the modifications are cleaved in vivo to generate the parent compounds.
  • Prodrugs include compounds wherein an amino acid residue, or a polypeptide chain of two or more (e.g., two, three or four) amino acid residues which are covalently joined to free amino and/or amido groups of compounds of formula (I).
  • the amino acid residues include the 20 naturally occurring amino acids commonly designated by three letter symbols and also include, 4-hydroxyproline, hydroxylysine, demosine, isodemosine, 3-methylhistidine, norvlin, beta-alanine, gamma-aminobutyric acid, citrulline, homocysteine, homoserine, ornithine and methionine sulfone.
  • Prodrugs also include compounds wherein carbonates, carbamates, amides and alkyl esters which are covalently bonded to the above substituents of formula (I) through the carbonyl carbon prodrug sidechain.
  • Prodrugs can include covalent irreversible and reversible inhibitors.
  • inventive compounds, agents and salts may exist in different crystalline or polymorphic forms, all of which are intended to be within the scope of the present invention and specified formulae.
  • the invention includes all crystalline forms of a compound of Formula (I) including anhydrous crystalline forms, hydrates, solvates and mixed solvates. If any of these crystalline forms demonstrates polymorphism, all polymorphs are within the scope of this invention.
  • Formula (I) is intended to cover, where applicable, solvated as well as unsolvated forms of the compounds.
  • Formula (I) includes compounds having the indicated structures, including the hydrated or solvated forms, as well as the non-hydrated and non-solvated forms.
  • the compounds of Formula (I) or salts, tautomers, N-oxides, polymorphs or prodrugs thereof may be provided in the form of solvates.
  • Solvates contain either stoichiometric or non-stoichiometric amounts of a solvent, and may be formed during the process of crystallization with pharmaceutically acceptable solvents such as water, alcohols such as methanol, ethanol or isopropyl alcohol, DMSO, acetonitrile, dimethyl formamide (DMF), acetic acid, and the like with the solvate forming part of the crystal lattice by either non-covalent binding or by occupying a hole in the crystal lattice. Hydrates are formed when the solvent is water, alcoholates are formed when the solvent is alcohol.
  • Solvates of the compounds of the present invention can be conveniently prepared or formed during the processes described herein. In general, the solvated forms are considered equivalent to the unsolvated forms for the purposes of the invention.
  • the compound of Formula (I) or salts, tautomers, N-oxides, solvates and/or prodrugs thereof that form crystalline solids may demonstrate polymorphism. All polymorphic forms of the compounds, salts, tautomers, N-oxides, solvates and/or prodrugs are within the scope of the invention.
  • the compound of Formula (I) may demonstrate tautomerism.
  • Tautomers are two interchangeable forms of a molecule that typically exist within an equilibrium. Any tautomers of the compounds of Formula (I) are to be understood as being within the scope of the invention.
  • the compound of Formula (I) may contain one or more stereocentres. All stereoisomers of the compounds of formula (I) are within the scope of the invention. Stereoisomers include enantiomers, diastereomers, geometric isomers (E and Z olephinic forms and cis and trans substitution patterns) and atropisomers.
  • the compound is a stereoisomerically enriched form of the compound of formula (I) at any stereocentre. The compound may be enriched in one stereoisomer over another by at least about 60, 70, 80, 90, 95, 98 or 99%.
  • the compound of Formula (I) or its salts, tautomers, solvates, N-oxides, and/or stereoisomers may be isotopically enriched with one or more of the isotopes of the atoms present in the compound.
  • the compound may be enriched with one or more of the following minor isotopes: 2 H, 3 H, 13 C, 14 C 15 N and/or 17 O.
  • An isotope may be considered enriched when its abundance is greater than its natural abundance.
  • the compounds of formula (I) or a salt, solvate, N-oxide, tautomer, stereoisomer, polymorph and/or prodrug thereof have a pIC 50 of at least 7 M.
  • the inhibitory activity can be determined using a kinase assay. Such assays are well-known to a person skilled in the art, and an example of a suitable assay is that described in the Examples.
  • composition comprising a compound according to formula (I) or a salt, solvate, N-oxide, tautomer, stereoisomer, polymorph and/or prodrug thereof, and a pharmaceutically acceptable excipient.
  • An appropriate dosage level will generally be about 0.01 to 500 mg per kg patient body weight per day which can be administered in single or multiple doses.
  • the dosage level will be about 0.1 to about 250 mg/kg per day; more preferably about 0.5 to about 100 mg/kg per day.
  • a suitable dosage level may be about 0.01 to 250 mg/kg per day, about 0.05 to 100 mg/kg per day, or about 0.1 to 50 mg/kg per day. Within this range the dosage may be 0.05 to 0.5, 0.5 to 5, or 5 to 50 mg/kg per day.
  • compositions are preferably provided in the form of tablets containing 1.0 to 1000 milligrams of the active ingredient, particularly 1.0, 5.0, 10.0, 15.0, 20.0, 25.0, 50.0, 75.0, 100.0, 150.0, 200.0, 250.0, 300.0, 400.0, 500.0, 600.0, 750.0, 800.0, 900.0, and 1000.0 milligrams of the active ingredient for the symptomatic adjustment of the dosage to the patient to be treated.
  • the compounds may be administered on a regimen of 1 to 4 times per day, preferably once or twice per day.
  • the specific dose level and frequency of dosage for any particular patient may be varied and will depend upon a variety of factors including the activity of the specific compound employed, the metabolic stability and length of action of that compound, the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the particular condition, and the host undergoing therapy.
  • the amount of a compound of the present invention in the composition will also depend upon the particular compound in the composition.
  • the typical inhalation dose is less than with other forms of dosing starting at 1 microgram and rising to 1000 microgram for a single puff.
  • the dose ranges from 25 microgram to 250 microgram per puff.
  • the dosage ranges from 500 to 1000 micrograms per puff.
  • the dosage is selected from the group consisting of 1, 2.5, 10.0, 25.0, 50.0, 75.0, 100.0, 150.0, 200.0, 250.0, 300.0, 400.0, 500.0, 600.0, 750.0, 800.0, 900.0, and 1000.0 micrograms per puff or any range in between and including two of these values.
  • the medication may be one puff per day or increase up to two puffs four times a day.
  • the pharmaceutical composition may further comprise other therapeutically active compounds which are usually applied in the treatment of the disclosed disorders or conditions. Selection of the appropriate agents for use in combination therapy may be made by one of ordinary skill in the art, according to conventional pharmaceutical principles.
  • the combination of therapeutic agents may act synergistically to effect the treatment or prevention of the various disorders or conditions disclosed herein. Using this approach, one may be able to achieve therapeutic efficacy with lower dosages of each agent, thus reducing the potential for adverse side effects.
  • compositions of the invention may be formulated for any appropriate route of administration including, for example, topical (for example, transdermal or ocular), pulmonary, oral, buccal, nasal, vaginal, rectal or parenteral administration.
  • parenteral as used herein includes subcutaneous, intradermal, intravascular (for example, intravenous), intramuscular, spinal, intracranial, intrathecal, intraocular, periocular, intraorbital, intrasynovial and intraperitoneal injection, as well as any similar injection or infusion technique.
  • compositions in a form suitable for oral use or parenteral use are preferred.
  • Suitable oral forms include, for example, tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsions, hard or soft capsules, or syrups or elixirs.
  • compositions provided herein may be formulated as a lyophilizate.
  • the composition is suitable for administration to the respiratory tract. In another form, the composition is suitable for oral administration.
  • the various dosage units are each preferably provided as a discrete dosage tablet, capsules, lozenge, dragee, gum, or other type of solid formulation.
  • Capsules may encapsulate a powder, liquid, or gel.
  • the solid formulation may be swallowed, or may be of a suckable or chewable type (either frangible or gum-like).
  • the present invention contemplates dosage unit retaining devices other than blister packs; for example, packages such as bottles, tubes, canisters, packets.
  • the dosage units may further include conventional excipients well-known in pharmaceutical formulation practice, such as binding agents, gellants, fillers, tableting lubricants, disintegrants, surfactants, and colorants; and for suckable or chewable formulations.
  • compositions intended for oral use may further comprise one or more components such as sweetening agents, flavouring agents, colouring agents and/or preserving agents in order to provide appealing and palatable preparations.
  • Tablets contain the active ingredient in admixture with physiologically acceptable excipients that are suitable for the manufacture of tablets.
  • excipients include, for example, inert diluents such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate, granulating and disintegrating agents such as corn starch or alginic acid, binding agents such as starch, gelatine or acacia, and lubricating agents such as magnesium stearate, stearic acid or talc.
  • the tablets may be uncoated or they may be coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period.
  • a time delay material such as glyceryl monosterate or glyceryl distearate may be employed.
  • Formulations for oral use may also be presented as hard gelatine capsules wherein the active ingredient is mixed with an inert solid diluent such as calcium carbonate, calcium phosphate or kaolin, or as soft gelatine capsules wherein the active ingredient is mixed with water or an oil medium such as peanut oil, liquid paraffin or olive oil.
  • an inert solid diluent such as calcium carbonate, calcium phosphate or kaolin
  • an oil medium such as peanut oil, liquid paraffin or olive oil.
  • Aqueous suspensions contain the active ingredient(s) in admixture with excipients suitable for the manufacture of aqueous suspensions.
  • excipients include suspending agents such as sodium carboxymethylcellulose, methylcellulose, hydroxypropylmethylcellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia, and dispersing or wetting agents such as naturally-occurring phosphatides (for example, lecithin), condensation products of an alkylene oxide with fatty acids such as polyoxyethylene stearate, condensation products of ethylene oxide with long chain aliphatic alcohols such as heptadecaethyleneoxycetanol, condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such as polyoxyethylene sorbitol mono-oleate, or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides such as polyethylene sorbitan monooleate.
  • Aqueous suspensions may also comprise one or more preservatives, for example ethyl or n-propyl p-hydroxybenzoate, one or more colouring agents, one or more flavouring agents, and one or more sweetening agents, such as sucrose or saccharin.
  • preservatives for example ethyl or n-propyl p-hydroxybenzoate
  • colouring agents for example ethyl or n-propyl p-hydroxybenzoate
  • flavouring agents such as sucrose or saccharin.
  • sweetening agents such as sucrose or saccharin.
  • Oily suspensions may be formulated by suspending the active ingredients in a vegetable oil such as arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin.
  • the oily suspensions may contain a thickening agent such as beeswax, hard paraffin or cetyl alcohol. Sweetening agents such as those set forth above, and/or flavouring agents may be added to provide palatable oral preparations.
  • Such suspensions may be preserved by the addition of an antioxidant such as ascorbic acid.
  • Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, suspending agent and one or more preservatives.
  • a dispersing or wetting agent e.g., kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, mannitol, mannitol, mannitol, mannitol, mannitol, mannitol, mannitol, mannitol, mannitol, mannitol,
  • compositions may also be in the form of oil-in-water emulsions.
  • the oily phase may be a vegetable oil such as olive oil or arachis oil, a mineral oil such as liquid paraffin, or a mixture thereof.
  • Suitable emulsifying agents include naturally-occurring gums such as gum acacia or gum tragacanth, naturally-occurring phosphatides such as soy bean lecithin, and esters or partial esters derived from fatty acids and hexitol, anhydrides such as sorbitan monoleate, and condensation products of partial esters derived from fatty acids and hexitol with ethylene oxide such as polyoxyethylene sorbitan monoleate.
  • An emulsion may also comprise one or more sweetening and/or flavouring agents.
  • Syrups and elixirs may be formulated with sweetening agents, such as glycerol, propylene glycol, sorbitol or sucrose. Such formulations may also comprise one or more demulcents, preservatives, flavouring agents and/or colouring agents.
  • sweetening agents such as glycerol, propylene glycol, sorbitol or sucrose.
  • Such formulations may also comprise one or more demulcents, preservatives, flavouring agents and/or colouring agents.
  • compositions of the invention may be formulated for local or topical administration, such as for topical application to the skin.
  • Formulations for topical administration typically comprise a topical vehicle combined with active agent(s), with or without additional optional components.
  • Topical vehicles include organic solvents such as alcohols (for example, ethanol, iso-propyl alcohol or glycerine), glycols such as butylene, isoprene or propylene glycol, aliphatic alcohols such as lanolin, mixtures of water and organic solvents and mixtures of organic solvents such as alcohol and glycerine, lipid-based materials such as fatty acids, acylglycerols including oils such as mineral oil, and fats of natural or synthetic origin, phosphoglycerides, sphingolipids and waxes, protein-based materials such as collagen and gelatine, silicone-based materials (both nonvolatile and volatile), and hydrocarbon-based materials such as microsponges and polymer matrices.
  • organic solvents such as alcohols (for example, ethanol, iso-propyl alcohol or glycerine), glycols such as butylene, isoprene or propylene glycol, aliphatic alcohols such as lanolin, mixtures of
  • a composition may further include one or more components adapted to improve the stability or effectiveness of the applied formulation, such as stabilizing agents, suspending agents, emulsifying agents, viscosity adjusters, gelling agents, preservatives, antioxidants, skin penetration enhancers, moisturizers and sustained release materials.
  • stabilizing agents such as hydroxymethylcellulose or gelatine-microcapsules, liposomes, albumin microspheres, microemulsions, nanoparticles or nanocapsules.
  • a topical formulation may be prepared in a variety of physical forms including, for example, solids, pastes, creams, foams, lotions, gels, powders, aqueous liquids, emulsions, sprays and skin patches.
  • the physical appearance and viscosity of such forms can be governed by the presence and amount of emulsifier(s) and viscosity adjuster(s) present in the formulation.
  • Solids are generally firm and non-pourable and commonly are formulated as bars or sticks, or in particulate form.
  • Solids can be opaque or transparent, and optionally can contain solvents, emulsifiers, moisturizers, emollients, fragrances, dyes/colorants, preservatives and other active ingredients that increase or enhance the efficacy of the final product.
  • Creams and lotions are often similar to one another, differing mainly in their viscosity. Both lotions and creams may be opaque, translucent or clear and often contain emulsifiers, solvents, and viscosity adjusting agents, as well as moisturizers, emollients, fragrances, dyes/colorants, preservatives and other active ingredients that increase or enhance the efficacy of the final product.
  • Gels can be prepared with a range of viscosities, from thick or high viscosity to thin or low viscosity. These formulations, like those of lotions and creams, may also contain solvents, emulsifiers, moisturizers, emollients, fragrances, dyes/colorants, preservatives and other active ingredients that increase or enhance the efficacy of the final product. Liquids are thinner than creams, lotions, or gels, and often do not contain emulsifiers. Liquid topical products often contain solvents, emulsifiers, moisturizers, emollients, fragrances, dyes/colorants, preservatives and other active ingredients that increase or enhance the efficacy of the final product.
  • Emulsifiers for use in topical formulations include, but are not limited to, ionic emulsifiers, cetearyl alcohol, non-ionic emulsifiers like polyoxyethylene oleyl ether, PEG-40 stearate, ceteareth-12, ceteareth-20, ceteareth-30, ceteareth alcohol, PEG-100 stearate and glyceryl stearate.
  • Suitable viscosity adjusting agents include, but are not limited to, protective colloids or nonionic gums such as hydroxyethylcellulose, xanthan gum, magnesium aluminum silicate, silica, microcrystalline wax, beeswax, paraffin, and cetyl palmitate.
  • a gel composition may be formed by the addition of a gelling agent such as chitosan, methyl cellulose, ethyl cellulose, polyvinyl alcohol, polyquaterniums, hydroxyethylcellulose, hydroxypropylcellulose, hydroxypropylmethylcellulose, carbomer or ammoniated glycyrrhizinate.
  • a gelling agent such as chitosan, methyl cellulose, ethyl cellulose, polyvinyl alcohol, polyquaterniums, hydroxyethylcellulose, hydroxypropylcellulose, hydroxypropylmethylcellulose, carbomer or ammoniated glycyrrhizinate.
  • Suitable surfactants include, but are not limited to, nonionic, amphoteric, ionic and anionic surfactants.
  • dimethicone copolyol polysorbate 20, polysorbate 40, polysorbate 60, polysorbate 80, lauramide DEA, cocamide DEA, and cocamide MEA, oleyl betaine, cocamidopropyl phosphatidyl PG-diimonium chloride, and ammonium laureth sulfate may be used within topical formulations.
  • Preservatives include, but are not limited to, antimicrobials such as methylparaben, propylparaben, sorbic acid, benzoic acid, and formaldehyde, as well as physical stabilizers and antioxidants such as vitamin E, sodium ascorbate/ascorbic acid and propyl gallate.
  • Suitable moisturizers include, but are not limited to, lactic acid and other hydroxy acids and their salts, glycerine, propylene glycol, and butylene glycol.
  • Suitable emollients include lanolin alcohol, lanolin, lanolin derivatives, cholesterol, petrolatum, isostearyl neopentanoate and mineral oils.
  • Suitable fragrances and colours include, but are not limited to, FD&C Red No. 40 and FD&C Yellow No. 5.
  • Other suitable additional ingredients that may be included in a topical formulation include, but are not limited to, abrasives, absorbents, anticaking agents, antifoaming agents, antistatic agents, astringents (such as witch hazel), alcohol and herbal extracts such as chamomile extract, binders/excipients, buffering agents, chelating agents, film forming agents, conditioning agents, propellants, opacifying agents, pH adjusters and protectants.
  • Typical modes of delivery for topical compositions include application using the fingers, application using a physical applicator such as a cloth, tissue, swab, stick or brush, spraying including mist, aerosol or foam spraying, dropper application, sprinkling, soaking, and rinsing.
  • Controlled release vehicles can also be used, and compositions may be formulated for transdermal administration (for example, as a transdermal patch).
  • compositions may be formulated as sustained release formulations such as a capsule that creates a slow release of modulator following administration.
  • sustained release formulations such as a capsule that creates a slow release of modulator following administration.
  • Such formulations may generally be prepared using well-known technology and administered by, for example, oral, rectal or subcutaneous implantation, or by implantation at the desired target site.
  • Carriers for use within such formulations are biocompatible, and may also be biodegradable.
  • the formulation provides a relatively constant level of modulator release.
  • the amount of modulator contained within a sustained release formulation depends upon, for example, the site of implantation, the rate and expected duration of release and the nature of the disorder to be treated or prevented.
  • a pharmaceutical composition may be formulated as inhaled formulations, including sprays, mists, or aerosols.
  • inhaled formulations including sprays, mists, or aerosols.
  • the inhaled formulation may be for application to the upper (including the nasal cavity, pharynx and larynx) and lower respiratory tract (including trachea, bronchi and lungs).
  • the composition or combination provided herein may be delivered via any inhalation methods known to a person skilled in the art.
  • Such inhalation methods and devices include, but are not limited to, metered dose inhalers with propellants such as HFA or propellants that are physiologically and environmentally acceptable.
  • Aerosol formulations for use in the subject method typically include propellants, surfactants and co-solvents and may be filled into conventional aerosol containers that are closed by a suitable metering valve.
  • propellants including propellants, surfactants and co-solvents
  • Different devices and excipients can be used depending on whether the application is to the upper (including the nasal cavity, pharynx and larynx) or lower respiratory tract (including trachea, bronchi and lungs) and can be determined by those skilled in the art.
  • processes for micronisation and nanoparticle formation for the preparation of compounds described herein for use in an inhaler such as a dry powder inhaler, are also known by those skilled in the art.
  • Inhalant compositions may comprise liquid or powdered compositions containing the active ingredient that are suitable for nebulization and intrabronchial use, or aerosol compositions administered via an aerosol unit dispensing metered doses.
  • Suitable liquid compositions comprise the active ingredient in an aqueous, pharmaceutically acceptable inhalant solvent such as isotonic saline or bacteriostatic water.
  • the solutions are administered by means of a pump or squeeze-actuated nebulized spray dispenser, or by any other conventional means for causing or enabling the requisite dosage amount of the liquid composition to be inhaled into the patient's lungs.
  • Suitable formulations, wherein the carrier is a liquid, for administration, as for example, a nasal spray or as nasal drops, include aqueous or oily solutions of the active ingredient.
  • inhalation drug delivery devices are described in Bennett et al. Medical Devices: Evidence and Research 2015:8 131-139, are contemplated for use in the present invention.
  • a method of treating or preventing a respiratory disease in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a compound of formula (I) or a salt, solvate, N-oxide, tautomer, stereoisomer, polymorph and/or prodrug thereof, thereby treating or preventing a respiratory disease in a subject.
  • a compound of formula (I) or a salt, solvate, N-oxide, tautomer, stereoisomer, polymorph and/or prodrug thereof for use in the treatment or prevention of a respiratory disease in a subject in need thereof.
  • ‘preventing’ or ‘prevention’ is intended to refer to at least the reduction of likelihood of the risk of (or susceptibility to) acquiring a disease or disorder (i.e., causing at least one of the clinical symptoms of the disease not to develop in a patient that may be exposed to or predisposed to the disease but does not yet experience or display symptoms of the disease).
  • a disease or disorder i.e., causing at least one of the clinical symptoms of the disease not to develop in a patient that may be exposed to or predisposed to the disease but does not yet experience or display symptoms of the disease.
  • Biological and physiological parameters for identifying such patients are provided herein and are also well known by physicians.
  • treatment includes the application or administration of a compound of the invention to a subject (or application or administration of a compound of the invention to a cell or tissue from a subject) with the purpose of delaying, slowing, stabilizing, curing, healing, alleviating, relieving, altering, remedying, lessening, worsening, ameliorating, improving, or affecting the disease or condition, the symptom of the disease or condition, or the risk of (or susceptibility to) the disease or condition.
  • treating refers to any indication of success in the treatment or amelioration of an injury, pathology or condition, including any objective or subjective parameter such as abatement; remission; lessening of the rate of worsening; lessening severity of the disease; stabilization, diminishing of symptoms or making the injury, pathology or condition more tolerable to the subject; slowing in the rate of degeneration or decline; making the final point of degeneration less debilitating; or improving a subject's physical or mental well-being.
  • antiagonizing used herein is intended to mean ‘decreasing’ or ‘reducing’.
  • a sufficient period of time can be during one week, or between 1 week to 1 month, or between 1 to 2 months, or 2 months or more.
  • the compound of the present invention can be advantageously administered for life time period.
  • respiratory refers to the process by which oxygen is taken into the body and carbon dioxide is discharged, through the bodily system including the nose, throat, larynx, trachea, bronchi and lungs.
  • respiratory disease or ‘respiratory condition’ refers to any one of several ailments that may involve inflammation and/or tissue remodelling affecting a component of the respiratory system including the upper (including the nasal cavity, pharynx and larynx) and lower respiratory tract (including trachea, bronchi and lungs).
  • Such ailments include pulmonary fibrosis (interstitial lung diseases), rhino sinusitis, influenza, sarcoidosis, bronchial carcinoma (including but not limited to non-small cell and small cell carcinoma of the lung, and lung metastases from tumours of other organs), silicosis, pneumoconiosis, acute lung injury, ventilation-induced lung injury, congenital emphysema, bronchopulmonary dysplasia, bronchiectasis, atelectasis, nasal polyps, asbestosis, mesothelioma, pulmonary eosinophilia, diffuse pulmonary haemorrhage syndromes, bronchiolitis obliterans, alveolar proteinosis, collagen and vascular disorders affecting the lung, and cough.
  • pulmonary fibrosis internal lung diseases
  • rhino sinusitis influenza
  • sarcoidosis bronchial carcinoma
  • bronchial carcinoma including but not limited to non-small cell and small cell carcinoma of the lung
  • the respiratory disease is an obstructive airway disease
  • such ailments include asthmatic conditions including hay fever, allergen-induced asthma, exercise-induced asthma, pollution-induced asthma, cold-induced asthma, stress-induced asthma and viral-induced-asthma, obesity-related asthma, occupational asthma, thunderstorm-induced asthma, asthma COPD overlap syndrome (ACOS) chronic obstructive pulmonary diseases including chronic bronchitis with normal airflow, chronic bronchitis with airway obstruction (chronic obstructive bronchitis), emphysema, asthmatic bronchitis, and bullous disease, and other pulmonary diseases involving inflammation including cystic fibrosis, pigeon fancier's disease, farmer's lung, acute respiratory distress syndrome, pneumonia of fungal, viral, bacterial, mixed or unknown aetiology, aspiration or inhalation injury, fat embolism in the lung, acidosis inflammation of the lung, acute pulmonary edema, acute mountain sickness, post-cardiac surgery,
  • the inflammation in the upper and lower respiratory tract may be associated with or caused by viral infection or an allergen. It is expected that the anti-inflammatory activity of the compounds either alone or when co-administered with a glucocorticoid would make them particularly suitable for treatment of these disease or conditions.
  • the respiratory disease or condition may be associated with or caused by an allergen, such as house dust mite.
  • the respiratory disease or condition may be the result of an allergen-induced inflammation.
  • the present invention finds particular application to allergic disease of the airway or lung and exacerbations of that disease, such as exacerbations resulting from viral infection (e.g. RSV infection).
  • a symptom of respiratory disease may include cough, excess sputum production, a sense of breathlessness or chest tightness with audible wheeze.
  • Exercise capacity may be quite limited.
  • the FEV1.0 force expiratory volume in one second
  • COPD the FEV1.0 as a ratio of the forced vital capacity (FVC) is typically reduced to less than 0.7.
  • IPF there is a progressive fall in FVC.
  • the impact of each of these conditions may also be measured by days of lost work/school, disturbed sleep, requirement for bronchodilator drugs, requirement for glucocorticoids including oral glucocorticoids.
  • Medical imaging procedures including but not limited to X-ray, high resolution computed tomography, magnetic resonance imaging, positron emission tomography, ultra sound, optical coherence tomography and fluoroscopy may also be used to assess disease and therapeutic response.
  • a parameter measured may be the presence or degree of lung function, signs and symptoms of obstruction; exercise tolerance; night time awakenings; days lost to school or work; bronchodilator usage; ICS dose; oral GC usage; need for other medications; need for medical treatment; hospital admission.
  • the term ‘asthma’ refers to a respiratory disorder characterized by episodic difficulty in breathing brought on by any one or a combination of three primary factors including: 1) bronchospasm (i.e., variable and reversible airway obstruction due to airway muscle contraction), 2) inflammation of the airway lining, and 3) bronchial hyper-responsiveness resulting in excessive mucous in the airways, which may be triggered by exposure to an allergen or combination of allergens (i.e., dust mites and mold), viral or bacterial infection (i.e., common cold virus), environmental pollutants (i.e., chemical fumes or smoke), physical exertion (i.e., during exercise), stress, or inhalation of cold air.
  • allergen or combination of allergens i.e., dust mites and mold
  • viral or bacterial infection i.e., common cold virus
  • environmental pollutants i.e., chemical fumes or smoke
  • physical exertion i.e., during exercise
  • stress
  • asthma condition refers to the characteristic of an individual to suffer from an attack of asthma upon exposure to any one or a number of asthma triggers for that individual.
  • An individual may be characterized as suffering from, for example, allergen-induced asthma, exercise-induced asthma, pollution-induced asthma, viral-induced asthma, or cold-induced asthma.
  • the efficacy of a treatment for asthma may be measured by methods well-known in the art, for example, increase in pulmonary function (spirometry), decrease in asthma exacerbations, increase in morning peak expiratory flow rate, decrease in rescue medication use, decrease in daytime and night-time asthma symptoms, increase in asthma-free days, increase in time to asthma exacerbation, and increase in forced expiratory volume in one second (FEV1.0).
  • increase in pulmonary function spirometry
  • decrease in asthma exacerbations increase in morning peak expiratory flow rate
  • decrease in rescue medication use decrease in daytime and night-time asthma symptoms
  • increase in asthma-free days increase in time to asthma exacerbation
  • increase in forced expiratory volume in one second FEV1.0
  • chronic obstructive pulmonary disease and ‘COPD’ as used interchangeably herein refers to a chronic disorder or combination of disorders characterized by reduced maximal expiratory flow and slow forced emptying of the lungs that does not change markedly over several months and is not, or is only minimally, reversible with traditional bronchodilators.
  • COPD is a combination of chronic bronchitis, i.e. the presence of cough and sputum for more than three months for about two consecutive years, and emphysema, i.e. alveolar damage.
  • COPD can involve chronic bronchitis with normal airflow, chronic bronchitis with airway obstruction (chronic obstructive bronchitis), emphysema, asthmatic bronchitis, and bullous disease, and combinations thereof.
  • Chronic obstructive pulmonary disease is a condition usually but not exclusively resulting from chronic lung damage induced by exposure to tobacco smoke.
  • Other noxious airborne pollutants, such as indoor cooking exhaust and car exhaust may over the long-term cause or increase the risk of COPD, as does ageing.
  • a condition of the airway or lung involving fibrosis or ‘a condition of the airway or lung having a fibrotic component’ includes any disease or condition where there is the formation or development of excess fibrous connective tissue (fibrosis) in the airway or lung thereby resulting in the development of scarred (fibrotic) tissue.
  • IPF idiopathic pulmonary fibrosis
  • IIP interstitial pneumonia
  • DPLD diffuse parenchymal lung disease
  • UIP interstitial pneumonia
  • the existence of, improvement in, treatment of or prevention of a condition of the airway or lung involving fibrosis, particularly pulmonary fibrosis/lung fibrosis or Idiopathic pulmonary fibrosis may be by any clinically or biochemically relevant method of the subject or a biopsy therefrom.
  • the rate of decline in FVC or the appearance of high resolution computed tomographic images of the lung may be useful in diagnosing IPF.
  • a parameter measured may be the presence or degree of fibrosis, the content of collagen, fibronectin, or another extracellular matrix protein, the proliferation rate of the cells or any extracellular matrix components in the cells or transdifferentiation of the cells to myofibroblasts.
  • the respiratory disease is selected from asthma, chronic obstructive pulmonary disease, interstitial lung diseases (such as idiopathic pulmonary fibrosis) and other conditions relating to tissue remodelling, primary or secondary lung tumour, hayfever, chronic and acute sinusitis, and chronic and acute viral, fungal and bacterial infections of the respiratory tract.
  • the improvement in respiratory function may be selected from a decrease in the level of constriction of the lungs, a decrease in the elastic stiffness of the respiratory system, and/or an increase in the ease with which the respiratory system can be extended.
  • the improvement is selected from a decrease in the level of constriction of the lungs, and a decrease in the elastic stiffness of the respiratory system.
  • a composition comprising a compound according to formula (I) or a salt, solvate, N-oxide, tautomer, stereoisomer, polymorph and/or prodrug thereof, and a pharmaceutically acceptable excipient.
  • the therapeutically effective amount of the formulation depends on the severity of the specific respiratory disease indication (e.g. severe chronic asthma), the patient's clinical history and response, and the discretion of the attending physician.
  • the formulation may be administered to the patient at one time or over a series of treatments.
  • An initial candidate dosage may be administered to a patient and the proper dosage and treatment regimen established by monitoring the progress of this patient using conventional techniques well known to those of ordinary skill in the art.
  • the therapeutically effective concentration of the active compound will be in the range 0.1 nM to 100 ⁇ M. More preferably the range will be 0.1-10 ⁇ M.
  • the method of treatment of the present invention further comprises administering a concomitant medication for the target disease indication.
  • concomitant asthma medications for both chronic and acute
  • concomitant asthma medications include but are not limited to: inhaled and oral steroids (e.g. beclomethasone, budesonide, flunisolide, fluticasone, triamcinolone, mometasone); systemic corticosteroids (e.g. methylprednisolone, prednisolone, prednisone, dexamethasone, and deflazacort); inhaled or oral ⁇ 2 -adrenoceptor agonists (e.g.
  • zafirlukast zafirlukast, zileuton, montekulast and pranlukast
  • anticholinergics e.g. ipatropium bromide
  • other therapeutic antibodies of any format e.g. antibodies directed against interleukin 5, such as mepolizumab, or against IgE, such as omalizumab, those antibodies in monoclonal form, Fab, scFV, multivalent compositions, xenoantibodies etc.
  • natural or engineered antibody mimetics e.g.
  • thromboxane A 2 synthetase inhibitors thromboxane prostanoid receptor antagonists
  • other eicosanoid modifiers e.g. alprostadil vs. PGE 1 , dinoprostone vs. PGE 2 , epoprostenol vs. prostacyclin and PGI 2 analogues (e.g. PG1 2 beraprost), seratrodast, phosphodiesterase 4 isoenzyme inhibitors, thromboxane A 2 synthetase inhibitors (e.g.
  • ozmagrel, dazmegrel or ozagrel ditec (low dose disodium cromoglycate and fenoterol); platelet activating factor receptor antagonists; antihistamines or histamine antagonists: promethazine, chlorpheniramine, loratadine, cetirazine, azelastine; thromboxane A 2 receptor antagonists; bradykinin receptor antagonists (e.g. icatibant); agents that inhibit activated eosinophils and T-cell recruitment (e.g. ketotifen), IL-13 blockers (e.g. soluble IL-13 receptor fragments), IL-4 blockers (e.g.
  • soluble IL-4 receptor fragments ligands that bind and block the activity of IL-13 or IL-4, and xanthine derivatives (e.g. pentoxifylline); chemokine receptor antagonists and antagonists of the CRTH2 receptor.
  • the method of treatment of the present invention includes the concomitant provision to the subject of inhibitory RNA molecules (RNA interference molecules), for the purpose of reducing, inhibiting or preventing the expression of genes which encode target proteins.
  • the inhibitory RNA molecules may be used for reducing or inhibiting the expression of one or more of: proteins associated with pathogens (viral, bacterial, fungal) or mammalian cells, including but not limited to casein kinase 1 isoforms and other components of the CLOCK regulatory network (eg ARNT1, period 1-3) and other proteins that contribute to the inflammatory response in the respiratory system such as interleukin-5 and the NALP inflammasome.
  • inhibitory RNA molecules may be any one of: short interfering RNA (siRNA), microRNA mimetic (miRNA), short hairpin RNA (shRNA) or long double stranded RNA (long dsRNA) molecules.
  • siRNA short interfering RNA
  • miRNA microRNA mimetic
  • shRNA short hairpin RNA
  • long dsRNA long double stranded RNA
  • the inhibitory RNA molecule may be administered directly to the subject requiring treatment (for example by inhalation, intratracheal, oral or nasal administration or by parenteral administration), or alternatively, be formed in the subject receiving treatment, following the administration of a polynucleotide (vector) construct which encodes a double stranded RNA (dsRNA) molecule which is capable of forming an inhibitory RNA molecule.
  • dsRNA double stranded RNA
  • the skilled person will also be familiar with various methods known in the art for formulating inhibitory RNA molecules for administration (for example, in liposomes, nanoparticles and the like).
  • the invention also includes the administration of an inhibitor of casein kinase 1 and a medication for the target disease indication as described above where either or both are administered by inhalation or formulated for oral administration.
  • the invention finds application in humans, the invention is also useful for therapeutic veterinary purposes.
  • the invention is useful for domestic or farm animals such as cattle, sheep, horses and poultry; for companion animals such as cats and dogs; and for zoo animals.
  • a ‘subject’ refers to an animal, such as a mammalian or an avian species, including a human, an ape, a horse, a cow, a sheep, a goat, a dog, a cat, a guinea pig, a rat, a mouse, a chicken etc.
  • CK1 ⁇ homologues are ubiquitious in nature, including in protozoa such as malaria, and in funghi and bacteria. Therefore, it is envisioned that the compounds of this invention may be used in any application requiring inhibition of CK1 ⁇ homologues. Such uses may include administration of a compound of the invention to a subject suffering from a disease, condition and/or disorder associated with infection and/or infestation with protozoa, fungii and/or bacteria.
  • the present invention provides a method of inhibiting casein kinase 1 ⁇ (CK1 ⁇ ), comprising contacting a cell with an effective amount of a compound of formula (I) as defined herein, or a salt, solvate, N-oxide, tautomer, stereoisomer, polymorph and/or prodrug thereof.
  • CK1 ⁇ casein kinase 1 ⁇
  • the compounds of the present invention or a salt, solvate, N-oxide, tautomer, stereoisomer, polymorph and/or prodrug thereof may serve as a selective inhibitor of CK1 ⁇ .
  • the compounds of the invention in any of their disclosed forms may selectively inhibit CK1 ⁇ compared to one or more other kinases, such as ERBB4/HER4, MINK/MINK1 and the like.
  • the compounds of the invention may be selective for CK1 ⁇ over at least one kinase by at least about 1, 5, 10 or 100-fold.
  • the present invention provides a kit or article of manufacture including a compound of formula (I) or pharmaceutical compositions including a compound of formula (I) as described herein.
  • kits for use in a therapeutic or prophylactic application mentioned herein including: a container holding a compound of formula (I) or pharmaceutical composition including a compound of formula (I); and a label or package insert with instructions for use.
  • the kit or ‘article of manufacture’ may comprise a container and a label or package insert on or associated with the container.
  • Suitable containers include, for example, bottles, vials, syringes, blister pack(s), etc.
  • the containers may be formed from a variety of materials such as glass or plastic.
  • the container holds a compound of formula (I), or composition which is effective for treating the condition and may have a sterile access port (for example the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle).
  • the label or package insert indicates that the composition is used for treating a disorder.
  • the label or package insert includes instructions for use and indicates that the therapeutic or prophylactic composition can be used to treat a disorder described herein.
  • the kit may comprise (a) a therapeutic or prophylactic composition; and (b) a second container with a second active principle or ingredient contained therein.
  • the kit in this embodiment of the invention may further comprise a package insert indicating the composition and other active principle can be used to treat a disorder or prevent a complication stemming from a disorder described herein.
  • the kit may further comprise a second (or third) container comprising a pharmaceutically-acceptable buffer, such as bacteriostatic water for injection (BWFI), phosphate-buffered saline, Ringer's solution and dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, and syringes.
  • BWFI bacteriostatic water for injection
  • the therapeutic composition may be provided in the form of a device, disposable or reusable, including a receptacle for holding the compound of formula (I) or therapeutic or prophylactic pharmaceutical composition including a compound of formula (I).
  • the device is a syringe.
  • the therapeutic or prophylactic composition may be provided in the device in a state that is ready for use or in a state requiring mixing or addition of further components.
  • the assay used was the HotSpot assay (Reaction Biology Corp).
  • Base Reaction buffer 20 mM Hepes (pH 7.5), 10 mM MgCl 2 , 1 mM EGTA, 0.02% Brij35, 0.02 mg/mL BSA, 0.1 mM Na 3 VO 4 , 2 mM DTT, 1% DMSO.
  • pFbs Primary human parenchymal fibroblast cells
  • DMEM Dulbecco's Modified Eagle's Media
  • FCS heat-inactivated fetal calf serum
  • FCS heat-inactivated fetal calf serum
  • FCS heat-inactivated fetal calf serum
  • FCS heat-inactivated fetal calf serum
  • 0.2% (v/v) sodium bicarbonate
  • 2 mM L-glutamine 1%
  • non-essential amino acids 1% (v/v) sodium pyruvate
  • 2.5 ⁇ g/mL amphotericin 5 IU/mL penicillin and 50 ⁇ g/mL streptomycin.
  • MRC5 sourced from ATTC
  • the MRC5 line was maintained under the same conditions as described for the pFb.
  • pFb Prior to experimentation, pFb were incubated in serum-free DMEM containing 0.25% bovine serum albumin (BSA) and insulin-transferrin-selenium-containing supplement (Monomed A; CSL, Parkville, Melbourne, Australia). The cells were incubated with small molecular CK1 ⁇ inhibitors (0.1-10 ⁇ M) for 30 min prior to 100 ⁇ M TGF- ⁇ 1 (R&D Systems, Minneapolis, Minn.) and the incubation continued for 16-24 hours prior to harvest of supernatant for detection of immunoreactive IL-11. Stock solutions were made as 10 mM in 100% DMSO and diluted to the required concentration in medium containing 0.1% DMSO (final concentration).
  • Supernatants were collected for measurement of IL-11 (R&D DuoSet, DY218) by ELISA following the manufacturers' instructions.
  • capture antibodies were initially diluted to the recommended concentrations using PBS buffer, and then used to coat the wells of 96-well microplates (Greiner, #655061) by adding 50 ⁇ L/well and incubated overnight at room temperature. Next day, solutions were discarded and wells were washed 3 times with wash buffer (PBS containing 0.1% (v/v) Tween-20) prior to the addition of 200 ⁇ L of blocking solution (PBS containing 1% (v/v) BSA) for 1 hour to block non-specific sites.
  • wash buffer PBS containing 0.1% (v/v) Tween-20
  • blocking solution PBS containing 1% (v/v) BSA
  • P IC 50 values (the negative log of the concentration suppressing IL-11 level by 50%) for inhibition of TGF- ⁇ -induced IL-11 were interpolated from the linear regression of log concentration small molecule versus IL-11 level (Table 4 and FIG. 2 ).
  • A549 cells were treated with various concentrations of PF670462 or ZH3-138 30 min prior to TGF-b (40 ⁇ M) for 24 h. Supernatant was collected for the determination of IL-11 or PAI-1 by ELISA. Data are presented in FIG. 5B as mean ⁇ SEM of three replicates. A similar experiment assessing the ability of ZH3-126 to effect IL-11 levels following administration was also carried out. Data from this experiment are presented in FIG. 6 as mean ⁇ SEM of three replicates.
  • compounds of the invention demonstrate the ability to reduce supernatant concentrations of other inflammatory biomarkers (such as interleukin-1 ⁇ (IL1 ⁇ )-mediated cytokine production) in a dose dependant manner.
  • IL1 ⁇ interleukin-1 ⁇
  • the anti-inflammatory potential of selected compounds was examined by establishing the effects on interleukin-1 ⁇ (IL1 ⁇ )-mediated cytokine production by the lung adenocarcinoma cell line A549.
  • Serum starved A549 cells were treated with various concentrations of the relevant CK1 ⁇ inhibitor (PF670462, ZH3-126 and ZH3-138) 30 min prior to IL-1 ⁇ (1 ng/ml) for 24 h.
  • the metabolic stability assay was performed by incubating each test compound in human liver microsomes at 37° C. and a protein concentration of 0.4 mg/mL. The metabolic reaction was initiated by the addition of an NADPH-regenerating system and quenched at various time points over a 60 minute incubation period by the addition of acetonitrile containing diazepam as internal standard. Control samples (containing no NADPH) were included (and quenched at 2, 30 and 60 minutes) to monitor for potential degradation in the absence of cofactor.
  • the human liver microsomes used in this experiment were supplied by XenoTech, lot #1410230. Microsomal incubations were performed at a substrate concentration of 1 ⁇ M. Data analysis: Species scaling factors from Ring et al.
  • Species scaling calculations are based on two assumptions: 1) NADPH-dependent oxidative metabolism predominates over other metabolic routes (i.e. direct conjugative metabolism, reduction, hydrolysis, etc.), and; 2) rates of metabolism and enzyme activities in vitro are truly reflective of those that exist in vivo. If significant non-NADPH-mediated degradation is observed in microsome control samples, then assumption (1) is invalid and predicted clearance parameters are therefore not reported.
  • PF670462 (2HCl.0.4 mol. eq. isopropanol) and ZH3-138 (unionised) were administered to male C57BL/6 mice by IV (3 mg/kg) or orally (10 mg/kg).
  • IV administration was achieved by bolus injection into the tail vein of mice using 1 mL syringe with 25G ⁇ 1′′ needle at a volume of 3 mL/kg.
  • Oral administration was achieved by gavage needle at a volume of 3 mL/kg.
  • the concentration-time profile for ZH3-138 was distinctly different to PF670462 ( FIG. 4 ). Plasma concentrations remained above the analytical lower limit of quantitation (LLQ) for the duration of the 24 h sampling period, and the apparent half-life was approximately 3 h. In addition the concentration-time profile exhibited an apparent secondary peak at 2 h post-dose; this may be indicative of enterohepatic recirculation where compound is excreted in bile (either unchanged, or as a conjugated metabolite) and then reabsorbed into the systemic circulation from the gastrointestinal tract. The apparent blood volume of distribution was moderate and the apparent blood clearance was low for ZH3-138.
  • IV and oral formulations were prepared using the same method. On the day of dosing, each of the solid compound was dissolved in DMSO to which an aqueous solution containing 10% Captisol was added. Hydrochloric acid (1 M) was used to modify pH for the formulation of ZH3-138 with the purpose of solubilising the compound (see Section B for the final pH of each formulation). The formulations of each compound were thoroughly vortexed, producing colourless solutions for all three compounds.
  • the IV formulation was filtered through a 0.22 ⁇ m syringe filter prior to IV dosing.
  • Blood was collected into polypropylene Eppendorf tubes containing heparin as anticoagulant and stabilisation cocktail (containing Complete® (a protease inhibitor cocktail) and potassium fluoride) to minimise the potential for ex vivo compound degradation in blood/plasma samples.
  • heparin as anticoagulant and stabilisation cocktail (containing Complete® (a protease inhibitor cocktail) and potassium fluoride) to minimise the potential for ex vivo compound degradation in blood/plasma samples.
  • Plasma samples were quantified against calibration standards prepared by spiking blank mouse plasma (50 ⁇ L) with solution standards (10 ⁇ L) obtained by diluting a stock solution of test compound (1 mg/mL in DMSO) with 50% acetonitrile in water. Diazepam (10 ⁇ L of 5 ⁇ g/mL in 50% acetonitrile/water) was added to all plasma calibration standards and samples as an internal standard (IS). The extraction of the test compound and IS from plasma was conducted using protein precipitation with acetonitrile. Protein precipitation was carried out by the addition of acetonitrile followed by vortexing and centrifugation (10,000 rpm) for 3 minutes to obtain supernatant for analysis using the LC-MS conditions described in Table 7.
  • B/P partitioning samples were conducted similarly and quantified against calibration standards samples prepared using a 1:1 v/v mixture of blank mouse blood and plasma.
  • the matrix matched B/P partitioning samples and standards were processed using protein precipitation with acetonitrile as described above.
  • Formulation aliquots were analysed against standard samples prepared in 50% acetonitrile/water. Formulation aliquots (50 or 100 ⁇ L) were dissolved in DMSO (1 mL final volume) and diluted using 50% acetonitrile/water to be within the calibration range.
  • the B/P ratio was obtained by dividing the average measured concentration in blood by the average concentration measured in plasma following centrifugation of whole blood.
  • HRMS High resolution mass spectra
  • ESI electrospray ionization
  • CH 2 Cl 2 dry THF and CH 2 Cl 2 were obtained by the method of Pangborn et al. 2 Pet. spirits refers to petroleum ether, boiling range 40-60° C. All other commercially available reagents were used as received.
  • Ethanolamine (46.0 ⁇ L, 0.762 mmol) was added to a mixture of 4 (33.0 mg, 0.095 mmol) in THF (5 mL) and the mixture was stirred at 40° C. for 3 d. The mixture was concentrated under vacuum and the residue purified by flash chromatography (EtOAc 100% to MeOH/EtOAc 20%) to give a pale orange oil (24.9 mg, 78%).
  • 4,4-Difluorocyclohexan-1-amine (67.0 mg, 0.499 mmol) was added to a solution of 14 (20.0 mg, 0.050 mmol) in THF (5 mL) and the mixture was stirred at 40° C. for 3 d. The mixture was concentrated under vacuum and the residue purified by flash chromatography (EtOAc/pet. spirits 70% to 100%) to give a pale orange solid (22.2 mg, 98%).
  • trans-3-Amino-cyclobutanol (29.0 mg, 0.328 mmol) was added to a solution of 14 (26.3 mg, 0.066 mmol) in THF (5 mL) and the mixture was stirred at r.t. for 5 d. The mixture was concentrated under vacuum and the residue purified by flash chromatography (EtOAc/pet. spirits 80% to 10%) to give a pale yellow solid (26.5 mg, 99%).
  • Tetrahydrothiopyran-4-ylamine (57.0 mg, 0.486 mmol) was added to a solution of 14 (29.9 mg, 0.075 mmol) in THF (5 mL) and the mixture was stirred at 40° C. for 3 d. The mixture was concentrated under vacuum and the residue purified by flash chromatography (EtOAc/pet. spirits 50% to 70%) to give a pale yellow solid (27.0 mg, 83%).
  • 2,2,2-Trifluoroethylamine hydrochloride (1.96 g, 14.5 mmol) and K 2 CO 3 (3.68 g, 26.6 mmol) were added to a solution of the crude aldehyde (1.87 g, 12.1 mmol) in CH 2 Cl 2 (20 mL). The reaction mixture was stirred at r.t. overnight. 1 H NMR analysis indicated complete consumption of the aldehyde. The reaction mixture containing the imine was used directly in the next step without isolation.
  • Ethanolamine (8.00 ⁇ L, 0.130 mmol) was added to a mixture of sulfone (13.0 mg, 0.032 mmol) in THF (5 mL) and the mixture was stirred at r.t. for 48 h. The mixture was concentrated under vacuum and the compound purified by flash chromatography (EtOAc/pet. spirits 50% to 100%) to afford an oil (8.10 mg, 66%).
  • Cis-3-amincyclobutanol (17.0 ⁇ L, 0.190 mmol) was added to a mixture of sulfone (19.1 mg, 0.047 mmol) in THF (5 mL) and the mixture was stirred at 40° C. for 72 h. The mixture was concentrated under vacuum and the compound purified by flash chromatography (EtOAc/pet. spirits 50% to 100%) to afford a pale yellow oil (15.8 mg, 83%).
  • 3,3-Difluorocyclobutanamine hydrochloride (30.0 mg, 0.206 mmol) was added to a mixture of sulfone (20.6 mg, 0.051 mmol) and aq. 20% K 2 CO 3 (140 ⁇ L, 0.206 mmol) in THF (5 mL) and the mixture was stirred at r.t. for 48 h. The mixture was concentrated under vacuum and the compound purified by flash chromatography (EtOAc/pet. spirits 20% to 40%) to afford a pale yellow solid (11.2 mg, 51%).
  • R 1 and R 2 are each independently selected from the group consisting of H, C 1-6 alkyl, C 1 alkylC 6 aryl, C 3-6 cycloalkyl and C 3-5 heterocyclyl;
  • R 3 is selected from the group consisting of F, Cl and CH 3 ;
  • R 4 is selected from the group consisting of C 0-3 alkylC 3-12 cycloalkyl and C 1-12 alkyl; wherein each of R 1 , R 2 , R 3 and R 4 is optionally substituted.
  • R 1 is C 1 alkylC 6 aryl.
  • R 1 is C 1 alkylC 6 aryl.
  • a compound according to embodiment 14, wherein the substituent is selected from one or more alkyl groups, one or more hydroxyl groups and/or one or more halo groups. 16.
  • R 2 is H. 17.
  • 28. A compound according to embodiment 27, wherein R 4 is a methyl, ethyl, propyl or butyl group.
  • 29. A compound according to embodiment 27, wherein R 4 is a branched alkyl group.
  • 30. A compound according to any one of embodiments 27 to 29, wherein R 4 is substituted.
  • 31. A compound according to embodiment 30, wherein by the substituent is selected from one or more OH groups and/or one or more halo groups. 32.
  • 33. A method of treating or preventing a respiratory disease in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of a compound of formula (I) according to any one of embodiments 1 to 32, thereby treating or preventing a respiratory disease in a subject.
  • a composition comprising a compound of formula (I) according to any one of embodiments 1 to 32, and a pharmaceutically acceptable excipient. 36. Use of a compound according to any one of embodiments 1 to 32, or a composition according to embodiment 35, in the preparation of a medicament for the treatment or prevention of a respiratory disease in a subject.

Landscapes

  • Organic Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Pulmonology (AREA)
  • Medicinal Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)
US17/291,694 2018-11-07 2019-11-07 Compounds and compositions for the treatment of respiratory diseases Pending US20220281846A1 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
AU2018904242A AU2018904242A0 (en) 2018-11-07 Compounds and compositions for the treatment of respiratory diseases
AU2018904242 2018-11-07
AU2018904241 2018-11-07
AU2018904241A AU2018904241A0 (en) 2018-11-07 Novel compounds for the treatment of respiratory diseases
PCT/AU2019/051224 WO2020093097A1 (en) 2018-11-07 2019-11-07 Compounds and compositions for the treatment of respiratory diseases

Publications (1)

Publication Number Publication Date
US20220281846A1 true US20220281846A1 (en) 2022-09-08

Family

ID=70610668

Family Applications (2)

Application Number Title Priority Date Filing Date
US17/291,694 Pending US20220281846A1 (en) 2018-11-07 2019-11-07 Compounds and compositions for the treatment of respiratory diseases
US17/291,707 Pending US20220002269A1 (en) 2018-11-07 2019-11-07 Novel compounds for the treatment of respiratory diseases

Family Applications After (1)

Application Number Title Priority Date Filing Date
US17/291,707 Pending US20220002269A1 (en) 2018-11-07 2019-11-07 Novel compounds for the treatment of respiratory diseases

Country Status (6)

Country Link
US (2) US20220281846A1 (zh)
EP (2) EP3877383A4 (zh)
JP (2) JP2022507118A (zh)
CN (2) CN113272298A (zh)
AU (2) AU2019376684A1 (zh)
WO (2) WO2020093097A1 (zh)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022266316A1 (en) 2021-06-18 2022-12-22 Hongene Biotech Corporation Functionalized n-acetylgalactosamine nucleosides
EP4396193A1 (en) 2021-08-30 2024-07-10 Hongene Biotech Corporation Functionalized n-acetylgalactosamine analogs
WO2023114746A1 (en) 2021-12-15 2023-06-22 Hongene Biotech Corporation Functionalized n-acetylgalactosamine analogs

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1996021654A1 (en) * 1995-01-12 1996-07-18 Smithkline Beecham Corporation Novel compounds
US6218537B1 (en) * 1995-06-07 2001-04-17 Smithkline Beecham Corporation Process for making pyridyl and pyrimidine substituted imidazole compounds
US6369068B1 (en) * 1995-06-07 2002-04-09 Smithkline Beecham Corporation Amino substituted pyrimidine containing compounds
US8518944B2 (en) * 2009-10-28 2013-08-27 Pfizer Inc. Compounds as casein kinase inhibitors

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5670527A (en) * 1993-07-16 1997-09-23 Smithkline Beecham Corporation Pyridyl imidazole compounds and compositions
US5593992A (en) * 1993-07-16 1997-01-14 Smithkline Beecham Corporation Compounds
UA48219C2 (uk) * 1996-03-25 2002-08-15 Елі Ліллі Енд Компані Композиція для вгамування болю (варіанти) та спосіб вгамування болю
JP2000507558A (ja) * 1996-03-25 2000-06-20 スミスクライン・ビーチャム・コーポレイション Cns損傷についての新規な治療
TW517055B (en) * 1997-07-02 2003-01-11 Smithkline Beecham Corp Novel substituted imidazole compounds
CA2316296A1 (en) * 1997-10-10 1999-04-22 Imperial College Of Science, Technology And Medicine Use of csaidtm compounds for the management of uterine contractions
WO1999032121A1 (en) * 1997-12-19 1999-07-01 Smithkline Beecham Corporation Compounds of heteroaryl substituted imidazole, their pharmaceutical compositions and uses
EP3273955A4 (en) * 2015-03-23 2019-05-01 The University of Melbourne TREATMENT OF RESPIRATORY DISEASES
AU2018262108A1 (en) * 2017-05-03 2019-11-21 The University Of Melbourne Compounds for the treatment of respiratory diseases

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1996021654A1 (en) * 1995-01-12 1996-07-18 Smithkline Beecham Corporation Novel compounds
US6218537B1 (en) * 1995-06-07 2001-04-17 Smithkline Beecham Corporation Process for making pyridyl and pyrimidine substituted imidazole compounds
US6369068B1 (en) * 1995-06-07 2002-04-09 Smithkline Beecham Corporation Amino substituted pyrimidine containing compounds
US8518944B2 (en) * 2009-10-28 2013-08-27 Pfizer Inc. Compounds as casein kinase inhibitors

Also Published As

Publication number Publication date
EP3877382A4 (en) 2022-07-27
EP3877383A4 (en) 2022-09-21
AU2019376684A1 (en) 2021-05-27
JP2022507117A (ja) 2022-01-18
WO2020093098A1 (en) 2020-05-14
AU2019374731A1 (en) 2021-05-27
EP3877382A1 (en) 2021-09-15
US20220002269A1 (en) 2022-01-06
CN113260616A (zh) 2021-08-13
JP2022507118A (ja) 2022-01-18
CN113272298A (zh) 2021-08-17
WO2020093097A1 (en) 2020-05-14
EP3877383A1 (en) 2021-09-15

Similar Documents

Publication Publication Date Title
US11975005B2 (en) Treatment of respiratory diseases
AU2012358805B2 (en) Pyrazolo[1,5-a]pyrimidines as antiviral agents
US20220380343A1 (en) Compounds for the treatment of respiratory diseases
US20220281846A1 (en) Compounds and compositions for the treatment of respiratory diseases
US20240041896A1 (en) Certain (2s)-n-[(1s)-1-cyano-2-phenylethyl]-1,4-oxazepane-2-carboxamides for treating bronchiectasis
TW201014590A (en) Heteroalkyl antagonists of prostaglandin D2 receptors
NZ587709A (en) N,n-disubstituted aminoalkylbiphenyl antagonists of prostaglandin d2 receptors
US8969350B2 (en) Pharmaceutical product comprising a p38 kinase inhibitor and a second active ingredient
CN111620815B (zh) 手性氯喹、羟氯喹和其衍生物及其制备方法与用途
JP2021508341A (ja) 医薬化合物
NZ621440B2 (en) 1-pyrazolyl-3-(4-((2-anilinopyrimidin-4-yl)oxy) napththalen-1-yl) ureas as p38 map kinase inhibitors

Legal Events

Date Code Title Description
AS Assignment

Owner name: THE UNIVERSITY OF MELBOURNE, AUSTRALIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:STEWART, ALASTAIR;WILLIAMS, SPENCER;HAKKI, ZALIHE;SIGNING DATES FROM 20210714 TO 20210910;REEL/FRAME:057535/0299

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

AS Assignment

Owner name: TIANLI BIOTECH PTY LTD, AUSTRALIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:THE UNIVERSITY OF MELBOURNE;REEL/FRAME:065681/0903

Effective date: 20230725

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED