WO2012078687A1 - Substituted pyrimidine urea compounds - Google Patents

Substituted pyrimidine urea compounds Download PDF

Info

Publication number
WO2012078687A1
WO2012078687A1 PCT/US2011/063611 US2011063611W WO2012078687A1 WO 2012078687 A1 WO2012078687 A1 WO 2012078687A1 US 2011063611 W US2011063611 W US 2011063611W WO 2012078687 A1 WO2012078687 A1 WO 2012078687A1
Authority
WO
WIPO (PCT)
Prior art keywords
group
alkyl
substituents
heteroaryl
optionally substituted
Prior art date
Application number
PCT/US2011/063611
Other languages
French (fr)
Inventor
Shaun R. Selness
Joseph B. Monahan
John F. Schindler
Balekudru Devadas
Original Assignee
Confluence Life Sciences, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Confluence Life Sciences, Inc. filed Critical Confluence Life Sciences, Inc.
Publication of WO2012078687A1 publication Critical patent/WO2012078687A1/en

Links

Classifications

    • 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
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/32One oxygen, sulfur or nitrogen atom
    • C07D239/42One nitrogen atom
    • 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
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings

Definitions

  • the present disclosure generally relates to a compound having enzyme inhibitory activity, pharmaceutical compositions comprising the compound, and methods useful for treating diseases. More specifically, the present disclosure relates to a class of pyrimidine urea compounds, pharmaceutical compositions comprising the compound, and methods useful for treating p38 kinase mediated diseases.
  • MAPK Mitogen-activated protein kinases
  • the MAPK are proline-directed serine/threonine-specific protein kinases that regulate cellular activities, such as gene expression, mitosis, differentiation, and cell survival/apoptosis.
  • ERK1 and 2 extracellular signaling kinases
  • JNK1-3 the extracellular signaling kinases
  • JNK1-3 the c-jun N-terminal kinase- 1
  • p38 MAPK ⁇ 38 ⁇ , ⁇ , ⁇ , and ⁇
  • ERK5 the extracellular signaling kinases
  • the MAPK are activated by the dual phosphorylation of Thr and Tyr residues within a TXY activation motif by coordinated dual- specificity MAPKK, where X is Glu, Pro, and Gly in ERK, JNK, and p38 MAPK, respectively.
  • MAPK are 60-70% identical to each other, yet differ in their activation loop sequences and sizes.
  • the activation loop is adjacent to the enzyme-active site, and its phosphorylation allows the enzyme to reposition active-site residues into the optimal orientation for substrate binding and catalysis.
  • MAPK mitogen-activated protein-kinase-activated protein
  • transcription factors the phosphorylation of which, either directly or indirectly, regulates gene expression at several points, including transcription, nuclear export, and mRNA stability and translation.
  • MAPK activation include inflammation, apoptosis, differentiation, and proliferation.
  • Distinct genes encode 4 p38 MAPK in humans: ⁇ 38 ⁇ , ⁇ , ⁇ , and ⁇ . Significant amino acid sequence homology is observed among the 4 isoforms, with 60 -75 overall sequence identity and > 90% identity within the kinase domains. Tissue- selective expression is observed, with ⁇ 38 ⁇ found predominantly in skeletal muscle, ⁇ 38 ⁇ in the testes, pancreas, and small intestine. In contrast, p38a and ⁇ are more ubiquitously expressed.
  • cytokines including TNFa, IL-1, IL-6, and IL-8
  • p38 MAPK is also responsible for the induction of key inflammatory enzymes such as COX2 and iNOS, the major sources of eicosanoids and nitric oxide at sites of inflammation, respectively.
  • COX2 and iNOS key inflammatory enzymes
  • iNOS key inflammatory enzymes
  • the p38 MAPK pathway regulates the expression of matrix metalloproteinases (MMP), including MMP2, MMP9, and MMP13.
  • MMP matrix metalloproteinases
  • p38 MAPK can directly phosphorylate several transcription factors, such as myocyte - specific enhancer binding factor 2C (MEF2C), CHOP, peroxisome proliferator- activated receptor (PPAR) a, PPAR ⁇ co-activator 1 and p53.
  • MEF2C myocyte - specific enhancer binding factor 2C
  • CHOP peroxisome proliferator- activated receptor
  • PPAR peroxisome proliferator- activated receptor
  • PPAR ⁇ co-activator 1 and p53 are involved in cellular functions such as apoptosis, gluconeogenesis, and synthesis of enzymes involved in fatty acid oxidation.
  • p38 MAPK is also involved in the direct or indirect phosphorylation of enzyme substrates, such as cytosolic phospholipase A2, and the Cdc25 phosphatases, which are involved in the activation of cyclin-dependent protein kinase activity and cell-cycle regulation. Therefore in addition to its role in the inflammatory response, p38 MAPK has other functions associated with normal and abnormal cell growth and survival as well as cellular function and homeostasis.
  • the MAPKAP kinases— MK2, MK-3, and PRAK— are selectively phosphorylated by p38 MAPK, while the phosphorylation of MSK1/2, MNK1/2, and RSKb is catalyzed by both p38 MAPK and ERK.
  • MNK is involved in the phosphorylation of eukaryotic initiation factor-4E, which binds to the 'cap' structure of mRNA and enhances protein translation. MNK phosphorylates the mRNA binding protein hnRNP-AO, a protein that regulates mRNA stability of transcripts encoding inflammatory proteins. MSK1/2 is involved in the phosphorylation of the transcription factors CREB and ATF- 1, which regulate AP-1 binding proteins. In addition, MSK1/2 can phosphorylate Histone H3, which is involved in chromatin remodeling. While evidence suggests that MSK and MNK play a role in the mediation of pro-inflammatory cytokines, in vivo data with selective inhibitors and/or knockout mice are lacking.
  • MK-2, MK-3, and PRAK once phosphorylated and activated by p38 MAPK, share similar substrate specificities. All of these kinases can phosphorylate the small heat-shock protein Hsp27.
  • PRAK- and MK3-deficient mice do not display any resistance to endotoxic shock or a decrease in lipopolysaccharide-(LPS)-induced cytokine production.
  • LPS lipopolysaccharide-(LPS)-induced cytokine production.
  • MK-2-deficient mice show a resistance to endotoxic shock and an impaired inflammatory response, as well as a significantly decreased production of cytokines such as TNFa, IFNyD and IL-6.
  • the p38/MK2 axis specifically is necessary and sufficient for mediating pro-inflammatory responses.
  • This inhibitor demonstrated substrate selectivity by preventing the p38a dependent phosphorylation of MK2 (Ki app 300 nM) while sparing the p38a dependent phosphorylation of ATF2 (Ki app > 20 uM).
  • This novel inhibitor is functionally unique compared with traditional p38 ATP competitive inhibitors that block the p38-dependent phosphorylation of all p38 substrates.
  • a second independent study also describes p38 inhibitors with unique mechanistic properties. This work demonstrates a novel mechanism for the selective inhibition of the p38 dependent phosphorylation of MK2. Unlike the previous study of Davidson et al., these mechanistically unique compounds are competitive with ATP and stabilize the p38/MK2 complex.
  • IL- ⁇ mediated diseases could be impacted by a p38 inhibitor based upon the key role for the p38 MAPK pathway in the biosynthesis and activity of this cytokine.
  • diseases include the family of cryopyrin associated periodic disorders (CAPS), chronic gout, diabetes, Still's disease, Familial Mediterranean Fever among others.
  • p38 MAPK has been linked to canine B cell growth and survival.
  • the role of p38 MAPK in B cell growth suggests that inhibition of this enzyme may be therapeutically beneficial for the treatment of canine B cell lymphoma.
  • Canine lymphoma is one of the most common malignancies diagnosed in companion animals representing 10-25% of canine neoplasms and >80% of the hematopoietic tumors.
  • An orally available, selective B cell growth inhibitor would meet a significant unmet medical need.
  • Compounds useful for treating diseases and conditions caused or exacerbated by unregulated p38 MAP Kinase and/or TNF activity are described in WO 2000/017175 published 30 March 2000. The compounds described therein include a class of substituted urea compounds.
  • WO 2008/062905 published 29 May 2008.
  • the compounds described therein include an alkyl-pyrimidinone-phenyl compounds wherein the phenyl fragment is substituted with a cyclopropyl radical, e.g., 6-butyl-3-(3- cyclopropylphenyl)-2methyl-5- ⁇ [2'-(5-oxo-4,5-dihydro-l,2,4-oxadizol-3-yl)biphenyl-4- yl]methyl ⁇ pyrimidin-4(3H)-one.
  • a cyclopropyl radical e.g., 6-butyl-3-(3- cyclopropylphenyl)-2methyl-5- ⁇ [2'-(5-oxo-4,5-dihydro-l,2,4-oxadizol-3-yl)biphenyl-4- yl]methyl ⁇ pyrimidin-4(3H)-one.
  • Various potential inhibitors or modulators of p38 kinase and the p38 kinase pathway are described in WO 2005/018557 published 3 March 2005.
  • the compounds described therein include di-fluorophenyl-methoxy-pyridinone-pyridyl compounds wherein the pyridyl fragment is substituted with various radicals including: alkyl, alkenyl, hydroxyalkyl, halo, cyano, amino, carboxy, carbamoyl, methoxycarbonyl and hydroxyalkenylimino radicals.
  • the compounds described therein include di-fluorophenyl-methoxy-pyrimidinone-phenyl compounds wherein the phenyl fragment is substituted with a cyclopropanyl or a morpholinyl radical through an amidoalkylamido bridge.
  • Pyrimidinone derivatives (as inhibitors of protein kinases and useful in treating disorders related to abnormal protein kinase activities such as inflammatory diseases and certain types of cancer) are described in WO 2008/153942 published 18 December 2008.
  • the compounds described therein include di-fluorophenyl-methoxy-pyrimidinone-phenyl compounds where the phenyl radical is substituted with cyclopentyl or a cyclohexyl radical through an amido bridge.
  • a pharmaceutical composition comprising a compound of Formula (I) or a pharmaceutically acceptable salt thereof, and a pharmaceutically- acceptable carrier.
  • the pharmaceutical composition further comprises one or more additional pharmaceutically active compounds.
  • a method for treating a condition comprising administering to a subject a therapeutically effective amount of a compound of Formula (I), wherein the condition to be treated includes, but is not limited to, autoimmune disorders, chronic inflammatory disorders, acute inflammatory disorders, auto-inflammatory disorders, pain, atherosclerosis, diabetes, fibrotic diseases, metabolic disorders, cancer, neoplasia, leukemia, and lymphoma.
  • the method comprises administering a combination of a compound of Formula (I) and at least one additional pharmaceutically active compound.
  • the invention comprises use of a compound having the structure of Formula I, or pharmaceutically acceptable salts thereof, for the manufacture of a medicament for the treatment of a condition in a subject.
  • the conditions that can be treated in accordance with the present invention include autoimmune disorders, chronic inflammatory disorders, acute inflammatory disorders, auto-inflammatory disorders, pain, atherosclerosis, diabetes, fibrotic diseases, metabolic disorders, cancer, neoplasia, leukemia, lymphoma and the like.
  • the invention comprises methods for making a compound having the structure of Formula I, or pharmaceutically acceptable salts thereof.
  • the invention comprises intermediates useful in making a compound having the structure of Formula I, or pharmaceutically acceptable salts thereof.
  • hydro denotes a single -H atom (H) and may be used interchangeably with the symbol "H” or the term “hydrogen”.
  • substituent may be either (1) not substituted or (2) substituted. If a substitutable position is not substituted, the default substituent is a hydrido radical.
  • alkyl refers to an acyclic alkyl radical containing 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 carbon atoms.
  • alkyl is a C Qo alkyl group or a Ci-C6 alkyl group.
  • alkyl groups include, but are not limited to, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, t- butyl, pentyl, hexyl, heptyl, octyl, nonyl and decyl.
  • alkoxy is RO- where R is alkyl as defined herein.
  • alkoxy groups include methoxy, ethoxy and propoxy.
  • alkyloxy and alkoxy may be used interchangeably.
  • alkoxyalkyl refers to an alkyl moiety substituted with an alkoxy group.
  • alkoxyalkyl groups include methoxymethyl, methoxyethyl, methoxypropyl and ethoxyethyl.
  • aralkoxy embraces an arylalkyl radical attached through an oxygen atom to the parent molecular scaffold.
  • arylalkoxy and “aralkoxy” may be used interchangeable.
  • aryl refers to any monocyclic, bicyclic or tricyclic carbon ring of up to 6 atoms in each ring, wherein at least one ring is aromatic, or an aromatic ring system of 5 to 14 carbons atoms which includes a carbocyclic aromatic group fused with a 5-or 6-membered cycloalkyl group.
  • aryl groups include, but are not limited to, phenyl, naphthyl, tetrahydronaphthyl and indanyl.
  • arylalkyl embraces an aryl-substituted alkyl radical and may be used interchangeably with the term “aralkyl”. Examples include benzyl, diphenylmethyl, triphenylmethyl, phenylethyl and diphenylethyl. The terms benzyl and phenylmethyl are interchangeable.
  • aryloxy is RO-, where R is aryl.
  • Arylthio is RS-, where R is aryl.
  • aryloxyalkyl embraces an aryloxy radical attached to an alkyl group.
  • cyano denotes a carbon radical having 3 of 4 covalent bonds shared by a nitrogen atom.
  • cycloalkyl is a hydrocarbyl group containing at least one saturated or partially unsaturated ring structure, and attached via a ring carbon. In various embodiments, it refers to a saturated or a partially unsaturated C 3 -C 12 cyclic moiety.
  • cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cycloheptyl and cyclooctyl.
  • halo refers to fluoro (-F), chloro (-C1), bromo (-Br), or iodo (-1).
  • haloalkyl refers to an alkyl moiety substituted with one or more halo groups.
  • haloalkyl groups include -CF 3 and -CHF 2 .
  • haloaralkoxy refers to aralkoxy group substituted with one or more halo radicals.
  • haloaralkoxy groups include fluorobenzyloxy, difluorobenzyloxy.
  • haloaralkoxy is 4-fluorobenzyloxy or 2,4- difluorobenzyloxy.
  • heterocyclyl includes the heteroaryls defined below and refers to a saturated or partially unsaturated monocyclic, bicyclic or tricyclic group of 2 to 14 ring-carbon atoms and, in addition to ring-carbon atoms, 1 to 4 heteroatoms selected from P, N, O and S.
  • the heterocyclic group is attached to another moiety through carbon or through a heteroatom, and is optionally substituted on carbon or a heteroatom.
  • heterocyclyl examples include azetidinyl, benzoimidazolyl, benzofuranyl, benzofurazanyl, benzopyrazolyl, benzotriazolyl, benzothiophenyl, benzoxazolyl, carbazolyl, carbolinyl, cinnolinyl, furanyl, imidazolyl, indolinyl, indolyl, indolazinyl, indazolyl, isobenzofuranyl, isoindolyl, isoquinolyl, isothiazolyl, isoxazolyl, naphthpyridinyl, oxadiazolyl, oxazolyl, oxazoline, isoxazoline, oxetanyl, pyranyl, pyrazinyl, pyrazolyl, pyridazinyl, pyridopyridinyl, pyridazinyl, pyridyl,
  • heteroaryl refers to a monocyclic, bicyclic or tricyclic ring having up to 6 atoms in each ring, wherein at least one ring is aromatic and contains from 1 to 4 heteroatoms in the ring selected from the group consisting of N, O and S.
  • heteroaryl examples include pyridyl, thienyl, furanyl, pyrimidyl, imidazolyl, pyranyl, pyrazolyl, thiazolyl, thiadiazolyl, isothiazolyl, oxazolyl, isoxazoyl, pyrrolyl, pyridazinyl, pyrazinyl, quinolinyl, isoquinolinyl, benzofuranyl, dibenzofuranyl, dibenzothiophenyl, benzothienyl, indolyl, benzothiazolyl, benzooxazolyl, benzimidazolyl, isoindolyl, benzotriazolyl, purinyl, thianaphthenyl and pyrazinyl.
  • heteroaryl can occur via an aromatic ring, or, if heteroaryl is bicyclic or tricyclic and one of the rings is not aromatic or contains no heteroatoms, through a non-aromatic ring or a ring containing no heteroatoms.
  • Heteroaryl is also understood to include the N-oxide derivative of any nitrogen containing heteroaryl.
  • heteroarylkoxy embraces a heteroarylalkyl radical attached through an oxygen atom to the molecular scaffold.
  • a class of preferred heteroaralkoxy radicals is "lower heteroaralkoxy” radicals having an alkyl range of 1-3 carbon atoms.
  • a preferred class of C 6 - heteroarylalkoxy radicals is (pyridin-2-yl)methoxy.
  • heteroaryloxy is RO-, where R is heteroaryl as defined herein.
  • examples include thiophen-2-yl-oxy, pyridin-2-yl-oxy, pyridin-3-yl-oxy, and pyridin-4-yl-oxy.
  • heteroaryloxyalkyl is a heteroaryloxy radical further attached to an alkyl radical.
  • hydroxyl refers to -OH radical and may be used interchangeably with “hydroxyl”.
  • hydroxyalkyl refers to a linear or branched monovalent C Qo hydrocarbon group substituted with at least one hydroxy group and examples of hydroxyalkyl groups include, but are not limited to, hydroxymethyl, hydroxyethyl, hydroxypropyl and hydroxybutyl.
  • the number of carbon atoms in a hydrocarbyl substituent can be indicated by the prefix “Cx-Cy” where X is the minimum and Y is the maximum number of carbon atoms in the substituent.
  • pharmaceutically-acceptable means suitable for use in pharmaceutical preparations, generally considered as safe for such use, officially approved by a regulatory agency of a national or state government for such use, or being listed in the U. S. Pharmacopoeia or other generally recognized pharmacopoeia for use in animals, and more particularly in humans.
  • pharmaceutically-acceptable salt refers to a salt which may enhance desired pharmacological activity.
  • pharmaceutically-acceptable salts include acid addition salts formed with inorganic or organic acids, metal salts and amine salts.
  • acid addition salts formed with inorganic acids include salts with hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid and phosphoric acid.
  • Examples of acid addition salts formed with organic acids such as acetic acid, propionic acid, hexanoic acid, heptanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, o-(4-hydroxy- benzoyl)-benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, 1,2-ethanedisulfonic acid, 2-hydroxyethane-sulfonic acid, benzenesulfonic acid, p- chlorobenzenesulfonic acid, 2-naphthalenesulfonic acid, p-toluenesulfonic acid, camphorsulfonic acid, 4-methyl-bicyclo[2.2.2]oct-2-enel-carboxy
  • terapéuticaally-effective amount refers to an amount of a compound that, when administered to a subject for treating a disease, is sufficient to effect treatment for the disease. "Therapeutically effective amount” can vary depending on the compound, the disease and its severity, the age, the weight, etc. of the subject to be treated.
  • Compounds of the present invention can exist in tautomeric, geometric or stereoisomeric forms.
  • the compounds' corresponding esters, metabolites, oximes, prodrugs, oniums and N-oxides are also embraced by the invention.
  • the present invention contemplates all such compounds, including cis- and trans-geometric isomers, E- and Z-geometric isomers, R- and S-enantiomers, diastereomers, d-isomers, 1-isomers, mixtures of isomers and racemates thereof, as falling within the scope of the invention.
  • cis and trans denote a form of geometric isomerism in which two carbon atoms connected by a double bond will each have a radical atom on the same side of the double bond ("cis”) or on opposite sides of the double bond ("trans").
  • Some of the compounds described contain one or more stereocenters and are meant to include R, S and mixtures of R and S forms for each stereocenter present.
  • the compounds of the invention may also exist as atropisomers, i.e., chiral rotational isomers.
  • the invention encompasses the racemic, resolved atropisomers, and mixtures thereof.
  • the present disclosure provides a class of compounds, including pharmaceutically acceptable salts of the compounds wherein the compounds have the structure of Formula I:
  • V and W are independently selected from the group consisting of CH and N;
  • R 1 is selected from the group consisting of cycloalkyl, aryl, heterocyclyl and heteroaryl; wherein the cycloalkyl and aryl substituents may be optionally substituted with one or more substituents independently selected from the group consisting of halo, cyano, alkyl and alkoxy; and wherein the heterocyclyl and heteroaryl substituents may be optionally substituted with one or more substituents independently selected from the group consisting of cyano and alkyl;
  • R 3 J and R 5 J are independently selected from the group consisting of alkyl, halo and hydrogen; and R 4 is selected from the group consisting of cycloalkyl, aryl, heterocyclyl and heteroaryl; wherein the cycloalkyl and aryl substituents may be optionally substituted with one or more substituents independently selected from the group consisting of halo, hydroxy, cyano, alkyl, alkoxy, hydroxyalkyl, alkoxyalkyl and aminoalkyl; and wherein the heterocyclyl and heteroaryl substituents may be optionally substituted with one or more substituents independently selected from the group consisting of cyano, alkyl, hydroxyalkyl, alkoxyalkyl and aminoalkyl.
  • V is CH and W is selected from the group consisting of CH and N;
  • R 1 is selected from the group consisting of heterocyclyl and heteroaryl; wherein the heterocyclyl and heteroaryl substituents may be optionally substituted with one or more substituents independently selected from the group consisting of (Ci-C 3 )-alkyl and cyano;
  • R 3 J and R 5 J are independently selected from the group consisting of hydrogen, halo and (C 1 -C 3 )-alkyl;
  • R 4 is (Cs-C 6 )-heteroaryl; wherein the (Cs-C 6 )-heteroaryl substituent may be optionally substituted with one or more substituents independently selected from the group consisting of alkyl and hydroxyalkyl.
  • V is CH and W is selected from the group consisting of CH and N;
  • R 1 is (Cs-C 6 )-heteroaryl; wherein the (Cs-C 6 )-heteroaryl substituent may be optionally substituted with one or more substituents independently selected from (Ci-C 3 )-alkyl;
  • R 3 is selected from the group consisting of methyl and fluoro;
  • R 4 is selected from the group consisting of pyridine and pyrimidine; wherein the pyridine and pyrimidine substituents may be optionally substituted with one or more substituents independently selected from the group consisting of alkyl and hydroxyalkyl; and
  • R 5 is selected from the group consisting of hydrogen and fluoro.
  • V is CH and W is selected from the group consisting of CH and N;
  • R 1 is Cs-heteroaryl, comprising one or more heteroatoms selected from the group consisting of O, N and S; and wherein the Cs-heteroaryl may be optionally substituted with one or more methyl substituents;
  • R is selected from the group consisting of methyl and fluoro;
  • R 4 is selected from the group consisting of pyridine and pyrimidine; wherein the pyridine and pyrimidine substituents may be optionally substituted with one or more substituents independently selected from the group consisting of alkyl and hydroxyalkyl; and
  • R 5 is selected from the group consisting of hydrogen and fluoro.
  • the present invention is also directed to a subclass of compounds, including pharmaceutically acceptable salts of the compounds, wherein the compounds have the structure of Formula II:
  • W is selected from the group consisting of CH and N;
  • X is selected from the group consisting of O, S and CH;
  • Y is selected from the group consisting of N and C;
  • R is selected from the group consisting of methyl and fluoro
  • R 5 is selected from the group consisting of hydrogen and fluoro
  • R 10 is selected from (C 1 -C3)-alkyl
  • R 40 is selected from the group consisting of methyl and hydroxy.
  • Non-limiting examples of Formula (II) compounds include the following compounds and pharmaceutically acceptable salts thereof:
  • V is CH and W is selected from the group consisting of CH and N;
  • R 1 is pyridine;
  • R 3 is selected from the group consisting of methyl and fluoro;
  • R is selected from the group consisting of pyridine and pyrimidine; wherein the pyridine and pyrimidine substituents may be optionally substituted with one or more substituents independently selected from the group consisting of alkyl and hydroxyalkyl; and
  • R 5 is selected from the group consisting of hydrogen and fluoro.
  • the present invention is also directed to a subclass of compounds, including pharmaceutically acceptable salts of the compounds, wherein the compounds have the structure of Formula III:
  • W is selected from the group consisting of CH and N;
  • R is selected from the group consisting of methyl and fluoro
  • R 5 is selected from the group consisting of hydrogen and fluoro
  • R 40 is selected from the group consisting of methyl and hydroxy.
  • the compound of Formula III is l-(5-(2-(2-hydroxypropan-2- yl)pyrimidin-4-yl)-2-methylphenyl)-l-(2-(pyridin-2-yl)pyrimidin-4-yl)urea.
  • V is CH and W is selected from the group consisting of CH and N;
  • R 1 is selected from the group consisting of cycloalkyl and aryl; wherein the cycloalkyl and aryl substituents may be optionally substituted with one or more substituents independently selected from the group consisting of (Ci-C 3 )-alkyl and halo;
  • R and R 5 are independently selected from the group consisting of hydrogen, halo and (C 1 -C 3 )-alkyl; and
  • R 4 is (C5-C 6 )-heteroaryl; wherein the (Cs-C 6 )-heteroaryl substituent may be optionally substituted with one or more substituents independently selected from the group consisting of alkyl and hydroxyalkyl.
  • V is CH and W is selected from the group consisting of CH and N;
  • R 1 is phenyl optionally substituted with one or more halo substituents;
  • R 3 is selected from the group consisting of methyl and fluoro;
  • R 4 is selected from the group consisting of pyridine and pyrimidine; wherein the pyridine and pyrimidine substituents may be optionally substituted with one or more substituents independently selected from the group consisting of alkyl and hydroxyalkyl; and
  • R 5 is selected from the group consisting of hydrogen and fluoro.
  • V is CH and W is selected from the group consisting of CH and N;
  • R 1 is phenyl optionally substituted with one or more fluoro substituents;
  • R 3 is selected from the group consisting of methyl and fluoro;
  • R 4 is selected from the group consisting of pyridine and pyrimidine; wherein the pyridine and pyrimidine substituents may be optionally substituted with one or more substituents independently selected from the group consisting of alkyl and hydroxyalkyl; and
  • R 5 is selected from the group consisting of hydrogen and fluoro.
  • the present invention is also directed to a subclass of compounds, including pharmaceutically acceptable salts of the compounds, wherein the compounds have the structure of Formula IV:
  • W is selected from the group consisting of CH and N;
  • R is selected from the group consisting of methyl and fluoro
  • R 5 is selected from the group consisting of hydrogen and fluoro
  • R 10 and R 11 are independently selected from the group consisting of hydrogen and fluoro;
  • R 40 is selected from the group consisting of methyl and hydroxy.
  • the compounds of Formula IV are selected from the group consisting of:
  • the present invention further comprises methods for treating a condition in a subject having or susceptible to having such a condition, by administering to the subject a therapeutically-effective amount of one or more compounds as described above.
  • the treatment is preventative treatment.
  • the treatment is palliative treatment.
  • the treatment is restorative treatment.
  • the conditions that can be treated in accordance with the present invention include, but are not limited to, autoimmune disorders, chronic inflammatory disorders, acute inflammatory disorders, auto-inflammatory disorders, pain, atherosclerosis, diabetes, fibrotic diseases, metabolic disorders, cancer, neoplasia, leukemia, lymphoma and the like.
  • the methods described herein are used to treat patients with disorders arising from dysregulated cytokine, enzymes and/or inflammatory mediator production, stability, secretion, posttranslational processing.
  • cytokines that may be dysregulated include interleukins 1, 2, 6, 8, 10, 12, 17, 22 and 23 along with tumor necrosis factor alpha and interferons alpha, beta and gamma.
  • inflammatory mediators that may be dysregulated include nitric oxide, prostaglandins and leukotrienes.
  • enzymes include cyclo-oxygenase, nitric oxide synthase and matrixmetallopro tease.
  • the methods described herein are used to treat patients with dysregulated p38 activity, activation, biosynthesis or pathway function.
  • the methods described herein are used to treat a patient in need thereof suffering from an autoimmune disorder, chronic and/or acute inflammatory disorder and/or auto-inflammatory disorder.
  • disorders include, but are not limited to colitis, multiple sclerosis, arthritis, rheumatoid arthritis, osteoarthritis, juvenile arthritis, psoriatic arthritis, cryopyrin associated periodic syndromes, Muckle- Wells Syndrome, Familial Cold Auto-inflammatory Syndrome, neonatal-onset multisystem inflammatory disease, TNF receptor associated periodic syndrome, acute pancreatitis, chronic pancreatitis, atherosclerosis, inflammatory bowel disease, Crohn's disease, ulcerative colitis, Diabetes mellitus type 1, Diabetes mellitus type 2, diabetic retinopathy, Still's disease, multiple sclerosis, vasculitis, sarcoidosis, pulmonary inflammation, acute respiratory distress syndrome, wet and dry age- related macular degeneration, autoimmune hemolytic syndromes, autoimmune hepatitis, autoimmune neuropathy,
  • host reaction i.e., graft vs. host disease
  • allograft rejections e.g., acute allograft rejection, and chronic allograft rejection
  • early transplantation rejection e.g., acute allograft rejection
  • reperfusion injury acute pain, chronic pain, neuropathic pain, Fibromyalgia ,pancreatitis, chronic infections, meningitis, encephalitis, myocarditis, gingivitis, post surgical trauma, tissue injury, traumatic brain injury, hepatitis, enterocolitis, sinusitis, uveitis, ocular inflammation, optic neuritis, gastric ulcers, esophagitis, peritonitis, periodontitis, dermatomyositis, gastritis, myositis, polymyalgia, pneumonia and bronchitis.
  • Fibrotic diseases including but not limited Obesity, steroid-resistance, glucose intolerance, metabolic syndrome.
  • the methods described herein can be used to treat a patient in need thereof and suffering from neoplasia.
  • Examples of these conditions include but not limited to angiogenesis, multiple myeloma, leukemia, B cell lymphoma, T cell lymphoma, mast cell tumors, lymphoma, Hodgkin's disease, cancer of the bone, mouth/pharynx, oesophagus, larynx, stomach, intestine, colon, rectum, lung, liver, pancreas, nerve, brain, head and neck, throat, ovary, uterus, prostate, testis, bladder, kidney, breast non- small cell lung carcinoma, melanoma, skin cancer, teratoma, rhabdomyosarcoma, glioma, metastatic and bone disorders.
  • the disease associated with dysregulated p38 include Cardiovascular and Cerebrovascular diseases, including but not limited to Atherosclerosis, restenosis of an atherosclerotic coronary artery, Acute coronary syndrome, myocardial infarction, cardiac-allograft vasculopathy and stroke; central nervous system disorders with an inflammatory or apoptotic component, Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, spinal cord injury, neuronal ischemia and peripheral neuropathy.
  • the term patient refers to both humans and nonhuman animals with the abovementioned conditions. Nonhuman animals could be companion animals such as, but not limited to canine and feline species.
  • Suitable subjects to be treated according to the present invention include mammalian subjects.
  • Mammals according to the present invention include, but are not limited to, human, canine, feline, bovine, caprine, equine, ovine, porcine, rodents, lagomorphs, primates, and the like, and encompass mammals in utero. Subjects may be of either gender and at any stage of development.
  • the compounds of the present invention are generally administered in a therapeutically effective amount.
  • the compounds of the present invention can be administered by any suitable route in the form of a pharmaceutical composition adapted to such a route, and in a dose effective for the treatment intended.
  • An effective dosage is typically in the range of about 0.001 to about 100 mg per kg body weight per day, preferably about 0.01 to about 30 mg/kg/day, in single or divided doses. Depending on age, species and condition being treated, dosage levels below the lower limit of this range may be suitable. In other cases, still larger doses may be used without harmful side effects. Larger doses may also be divided into several smaller doses, for administration throughout the day.
  • the compounds of the present invention may be administered orally, including by swallowing, so that the compound enters the gastrointestinal tract, or absorbed into the blood stream directly from the mouth (e.g., buccal or sublingual administration).
  • compositions for oral administration include solid formulations such as tablets, lozenges and capsules, which can contain liquids, gels, or powders.
  • compositions for oral administration may be formulated as immediate or modified release, including delayed or sustained release, optionally with enteric coating.
  • Liquid formulations can include solutions, syrups and suspensions, which can be used in soft or hard capsules.
  • Such formulations may include a pharmaceutically acceptable carrier, for example, water, ethanol, polyethylene glycol, cellulose, or an oil.
  • the formulation may also include one or more emulsifying agents and/or suspending agents.
  • a tablet dosage form the amount of drug present may be from about 0.05% to about 95% by weight, more typically from about 2% to about 50% by weight of the dosage form.
  • tablets may contain a disintegrant, comprising from about 0.5% to about 35% by weight, more typically from about 2% to about 25% of the dosage form.
  • disintegrants include methyl cellulose, sodium or calcium carboxymethyl cellulose, croscarmellose sodium, polyvinylpyrrolidone, hydroxypropyl cellulose, starch and the like.
  • Suitable lubricants for use in a tablet, may be present in amounts from about 0.1% to about 5% by weight, and include calcium, zinc or magnesium stearate, sodium stearyl fumarate and the like.
  • Suitable binders for use in a tablet, include gelatin, polyethylene glycol, sugars, gums, starch, hydroxypropyl cellulose and the like.
  • Suitable diluents for use in a tablet, include mannitol, xylitol, lactose, dextrose, sucrose, sorbitol and starch.
  • Suitable surface active agents and glidants for use in a tablet, may be present in amounts from about 0.1% to about 3% by weight, and include polysorbate 80, sodium dodecyl sulfate, talc and silicon dioxide.
  • Compounds of the present invention may be administered directly into the blood stream, muscle, or internal organs.
  • Suitable means for parenteral administration include intravenous, intra- muscular, subcutaneous intraarterial, intraperitoneal, intrathecal, intracranial, and the like.
  • Suitable devices for parenteral administration include injectors (including needle and needle-free injectors) and infusion methods.
  • compositions for parenteral administration may be formulated as immediate or modified release, including delayed or sustained release.
  • parenteral formulations are aqueous solutions containing excipients, including salts, buffering agents and carbohydrates.
  • Parenteral formulations may also be prepared in a dehydrated form (e.g., by lyophilization) or as sterile non-aqueous solutions. These formulations can be used with a suitable vehicle, such as sterile water. Solubility-enhancing agents may also be used in preparation of parenteral solutions.
  • Compounds of the present invention may be administered topically to the skin or transdermally.
  • Formulations for this topical administration can include lotions, solutions, creams, gels, hydrogels, ointments, foams, implants, patches and the like.
  • Pharmaceutically acceptable carriers for topical administration formulations can include water, alcohol, mineral oil, glycerin, polyethylene glycol and the like. Topical administration can also be performed by electroporation, iontophoresis, phonophoresis and the like.
  • compositions for topical administration may be formulated as immediate or modified release, including delayed or sustained release.
  • the compounds of the present invention can be used, alone or in combination with other pharmaceutically active compounds, to treat conditions such as those previously described above.
  • the compound(s) of the present invention and other pharmaceutically active compound(s) can be administered simultaneously (either in the same dosage form or in separate dosage forms) or sequentially.
  • the present invention comprises methods for treating a condition by administering to the subject a therapeutically-effective amount of one or more compounds of the present invention and one or more additional pharmaceutically active compounds.
  • the present invention comprises a pharmaceutical composition comprising one or more compounds of the present invention, one or more additional pharmaceutically active compounds, and a pharmaceutically acceptable carrier.
  • the one or more additional pharmaceutically active compounds is selected from the group consisting of anti-inflammatory drugs, anti-atherosclerotic drugs, immunosuppressive drugs, immunomodulatory drugs, cytostatic drugs, anti-proliferative agents, angiogenesis inhibitors, kinase inhibitors, cytokine blockers and inhibitors of cell adhesion molecules.
  • p38 inhibitor compositions described herein are also optionally used in combination with other therapeutic reagents that are selected for their therapeutic value for the condition to be treated.
  • the compositions described herein and, in embodiments where combinational therapy is employed, other agents do not have to be administered in the same pharmaceutical composition, and, because of different physical and chemical characteristics, are optionally administered by different routes.
  • the initial administration is generally made according to established protocols, and then, based upon the observed effects, the dosage, modes of administration and times of administration subsequently modified. In certain instances, it is appropriate to administer a p38 inhibitor composition as described herein in combination with another therapeutic agent.
  • a p38 inhibitor composition as described herein is rash, then it is appropriate to administer an anti-histamine agent in combination with the initial therapeutic agent.
  • the therapeutic effectiveness of a p38 inhibitor is enhanced by administration of another therapeutic agent (which also includes a therapeutic regimen) that also has therapeutic benefit.
  • another therapeutic agent which also includes a therapeutic regimen
  • the overall benefit experienced by the patient is either simply additive of the two therapeutic agents or the patient experiences a synergistic benefit.
  • Therapeutically effective dosages vary when the drugs are used in treatment combinations. Methods for experimentally determining therapeutically effective dosages of drugs and other agents for use in combination treatment regimens are documented methodologies. Combination treatment further includes periodic treatments that start and stop at various times to assist with the clinical management of the patient.
  • the multiple therapeutic agents one of which is a p38 inhibitor as described herein
  • the multiple therapeutic agents are administered in any order, or even simultaneously. If simultaneously, the multiple therapeutic agents are optionally provided in a single, unified form, or in multiple forms (by way of example only, either as a single pill or as two separate pills).
  • one of the therapeutic agents is given in multiple doses, or both are given as multiple doses. If not simultaneous, the timing between the multiple doses optionally varies from more than zero weeks to less than twelve weeks.
  • the combination methods, compositions and formulations are not to be limited to the use of only two agents, the use of multiple therapeutic combinations are also envisioned. It is understood that the dosage regimen to treat, prevent, or ameliorate the condition(s) for which relief is sought, is optionally modified in accordance with a variety of factors. These factors include the disorder from which the subject suffers, as well as the age, weight, sex, diet, and medical condition of the subject. Thus, the dosage regimen actually employed varies widely, in some embodiments, and therefore deviates from the dosage regimens set forth herein.
  • the pharmaceutical agents which make up the combination therapy disclosed herein are optionally a combined dosage form or in separate dosage forms intended for substantially simultaneous administration.
  • the pharmaceutical agents that make up the combination therapy are optionally also administered sequentially, with either agent being administered by a regimen calling for two-step administration.
  • the two-step administration regimen optionally calls for sequential administration of the active agents or spaced-apart administration of the separate active agents.
  • the time period between the multiple administration steps ranges from, a few minutes to several hours, depending upon the properties of each pharmaceutical agent, such as potency, solubility, bioavailability, plasma half-life and kinetic profile of the pharmaceutical agent. Circadian variation of the target molecule concentration is optionally used to determine the optimal dose interval.
  • a p38 inhibitor is optionally used in combination with procedures that provide additional or synergistic benefit to the patient.
  • a p38 inhibitor and the additional therapy(ies) are optionally administered before, during or after the occurrence of a disease or condition, and the timing of administering the composition containing a p38 inhibitor varies in some embodiments.
  • a p38 inhibitor is used as a prophylactic and is administered continuously to subjects with a propensity to develop conditions or diseases in order to prevent the occurrence of the disease or condition.
  • a p38 inhibitor and compositions are optionally administered to a subject during or as soon as possible after the onset of the symptoms.
  • a p38 inhibitor can be used in combination with drugs from the following classes: NSAIDs, immunosuppressive drugs, immunomodulatory drugs, cytostatic drugs, angiogenesis inhibitors, biological agents, steroids, vitamin D3 analogs, retinoids, other kinase inhbitors, cytokine blockers, corticosteroids and inhibitors of cell adhesion molecules.
  • drugs from the following classes NSAIDs, immunosuppressive drugs, immunomodulatory drugs, cytostatic drugs, angiogenesis inhibitors, biological agents, steroids, vitamin D3 analogs, retinoids, other kinase inhbitors, cytokine blockers, corticosteroids and inhibitors of cell adhesion molecules.
  • NSAIDs drugs from the following classes: NSAIDs, immunosuppressive drugs, immunomodulatory drugs, cytostatic drugs, angiogenesis inhibitors, biological agents, steroids, vitamin D3 analogs, retinoids, other kinase inhbitors, cytokine blockers, cortic
  • therapeutic agents/treatments for treating atherosclerosis or a condition that is associated with atherosclerosis include, but are not limited to any of the following: torcetrapib, aspirin, niacin, HMG CoA reductase inhibitors (e.g., atorvastatin, fluvastatin, lovastatin, pravastatin, rosuvastatin and simvastatin), colesevelam, cholestyramine, colestipol, gemfibrozil, probucol and clofibrate.
  • HMG CoA reductase inhibitors e.g., atorvastatin, fluvastatin, lovastatin, pravastatin, rosuvastatin and simvastatin
  • colesevelam cholestyramine
  • colestipol gemfibrozil
  • probucol and clofibrate examples include, but are not limited to any of the following: torcetrapib, aspirin, niaci
  • a p38 inhibitor composition described herein is optionally used together with one or more agents or methods for treating an inflammatory condition in any combination.
  • agents/treatments for treating an autoimmune and/or inflammatory condition include, but are not limited to any of the following: corticosteroids, nonsteroidal antiinflammatory drugs (NSAID) (e.g.
  • ibuprofen ibuprofen, naproxen, acetominophen, aspirin, Fenoprofen (Nalfon), Flurbiprofen (Ansaid), Ketoprofen, Oxaprozin (Daypro), Diclofenac sodium (Voltaren), Diclofenac potassium (Cataflam), Etodolac (Lodine), Indomethacin (Indocin), Ketorolac (Toradol), Sulindac (Clinoril), Tolmetin (Tolectin), Meclofenamate (Meclomen), Mefenamic acid (Ponstel), Nabumetone (Relafen), Piroxicam (Feldene), cox-2 inhibitors (e.g.
  • celecoxib (Celebrex))
  • immunosuppressants e.g. methotrexate (Rheumatrex), leflunomide (Arava), azathioprine (Imuran), cyclosporine (Neoral, Sandimmune), tacrolimus and cyclophosphamide (Cytoxan
  • CD20 blockers Rituximab
  • Tumor Necrosis Factor (TNF) blockers e.g. etanercept (Enbrel), infliximab (Remicade) and adalimumab (Humira)
  • Abatacept CLA4-Ig
  • interleukin- 1 receptor antagonists e.g.
  • Anakinra (Kineret), interleukin 6 inhibitors (e.g. Actemra), interleukin 17 inhibitors (e.g. AIN457), Janus kinase inhibitors (e.g. Tasocitinib), syk inhibitors (e.g. R788), chloroquine and its derivatives.
  • interleukin 6 inhibitors e.g. Actemra
  • interleukin 17 inhibitors e.g. AIN457
  • Janus kinase inhibitors e.g. Tasocitinib
  • syk inhibitors e.g. R788
  • a p38 inhibitor is optimally used together with one or more of the following classes of drugs: wherein the anti-cancer agent is an EGFR kinase inhibitor, MEK inhibitor, VEGFR inhibitor, anti-VEGFR2 antibody, KDR antibody, AKT inhibitor, PDK-1 inhibitor, PI3K inhibitor, c-kit/Kdr tyrosine kinase inhibitor, Bcr-Abl tyrosine kinase inhibitor, VEGFR2 inhibitor, PDGFR-beta inhibitor, KIT inhibitor, Flt3 tyrosine kinase inhibitor, PDGF receptor family inhibitor, Flt3 tyrosine kinase inhibitor, RET tyrosine kinase receptor family inhibitor, VEGF-3 receptor antagonist, Raf protein kinase family inhibitor, angiogenesis inhibitor, Erb2 inhibitor, mTOR inhibitor, IGF-1R antibody, NFkB inhibitor, proteosome inhibitor, chemotherapy
  • kits that are suitable for use in performing the methods of treatment or prevention described above.
  • the kit contains a first dosage form comprising one or more of the compounds of the present invention and a container for the dosage, in quantities sufficient to carry out the methods of the present invention.
  • the invention relates to the novel intermediates useful for preparing the compounds of the present invention.
  • the compounds of the present invention can be prepared using the methods illustrated in the general synthetic schemes and experimental procedures detailed below. These general synthetic schemes and experimental procedures are presented for purposes of illustration and are not intended to be limiting.
  • the starting materials used to prepare the compounds of the present invention are commercially available or can be prepared using routine methods known in the art.
  • Step A Compound 1 may be prepared by coupling a dihalogenated pyrimidine with a substituted aniline, compound 4 as exemplified in schemes 2 and 3, in the presence of a catalyst such as palladium acetate and a ligand such as BINAP and a solvent such as toluene, DMA or THF.
  • a catalyst such as palladium acetate and a ligand such as BINAP
  • a solvent such as toluene, DMA or THF.
  • the halopyridine may be displaced using the aniline and a base such as cesium carbonate in a solvent such as DMF, DMA or NMP with heating to provide compound 1.
  • Step B Compound 2 may be prepared via Suzuki coupling between a 2- bromopyrimidine (Compound 1) and a boronic acid in the presence of a catalyst such as tetrakis(triphenylphosphine)palladium and a base such as sodium carbonate, sodium t-butoxide, potassium t-butoxide or cesium carbonate and a solvent such as ethanol/water.
  • a catalyst such as tetrakis(triphenylphosphine)palladium
  • a base such as sodium carbonate, sodium t-butoxide, potassium t-butoxide or cesium carbonate
  • a solvent such as ethanol/water.
  • a boronic acid derivative of pyrimidine may be coupled with a substituted aryl halide under conditions listed above.
  • Step C Compound 3 may be prepared via reaction with an activated carbonyl such as phosgene or triphosgene and subsequenently treating the carbonyl with ammonium hydroxide in a solvent such as toluene or dimethoxyethane.
  • an activated carbonyl such as phosgene or triphosgene
  • ammonium hydroxide such as toluene or dimethoxyethane.
  • a suitable boronic acid or ester may be used to synthesize the desired aryl or alkyl R 4 in the presence of a suitable catalyst such as palladium chloride or tetrakis(triphenylphosphine)palladium(0), a base such as sodium carbonate and a solvent such as ethanol.
  • a suitable catalyst such as palladium chloride or tetrakis(triphenylphosphine)palladium(0)
  • a base such as sodium carbonate
  • a solvent such as ethanol.
  • X may be a boronic acid or ester and a suitable haloalkyl or haloaryl may be used with conditions listed above to prepare compound 4.
  • Step A When X is a carboxylate group, the ⁇ , ⁇ -dimethyamide may be formed via activation with thionyl chloride or 2-chloro-4,6-dimethoxytriazine or a carbodiimide activating agent such as CD I, EDC or DCC in the presence of HOBt or N-hydroxysuccinimide in a solvent such as dichloromethane, DMF or THF.
  • Step B The amide may be treated with a Grignard reagent in the presence of a solvent such as diethyl ether or THF to provide the appropriate ketone derivative.
  • Step C The ketone may be reacted with dimethylformamide acetal in DMF to provide the enamine intermediate.
  • Step D The enamine may be reacted with a guanidine, amidine or hydrazine with the appropriate substitution in DMF to provide the desired heteroaryl R 4 .
  • P38 inhibitory potency and P38/MK2 substrate selectivity The novel, MK2 substrate-selective inhibitory mechanism of compounds is evaluated in enzyme assays comparing inhibitor potency in blocking p38/MK2 versus p38/PRAK induced phosphorylation of an HSP-27 derived peptide substrate.
  • the ability of compounds to inhibit activated phospho- p38a is evaluated using a p38a/MK2 and a p38a/PRAK cascade assay format.
  • the kinase activity of p38a is determined by its ability to phosphorylate GST-MK2 or GST-PRAK.
  • Activation of MK2 or PRAK by p38a is quantitated by measuring the phosphorylation of a fluorescently-labeled, MK2 specific peptide substrate, Hsp27 peptide (FITC- KKKALSRQLSVAA).
  • Hsp27 peptide FITC- KKKALSRQLSVAA
  • the phosphorylation of the Hsp27 peptide is quantified using the Caliper LabChip 3000.
  • Kinase reactions are carried out in a 384-well plate (Matrical, MPlOl-1- PP) in 20 mM HEPES pH 7.5, 10 mM MgC12, 0.0005% Tween-20, 0.01% BSA, 1 mM DTT, and 2% DMSO.
  • the inhibitor concentration is varied between 0.02-30,000 nM, while the Hsp27 peptide substrate and MgATP are held constant at 1 ⁇ and 10 ⁇ , respectively.
  • Activated p38a is added to a final concentration of 20 pM for reactions with nonphosphorylated 1 nM His6-MK2 in the cascade reaction.
  • unactivated GST-PRAK is held constant at 1 nM while p38 is added in to a final concentration of 20 pM.
  • Kinase reactions are incubated at room temperature and quenched after 30 minutes by the addition of stop buffer (180 mM HEPES, 30 mM EDTA, and 0.2% Coating Reagent-3).
  • Hsp27 peptide is phosphorylated. Reactions are initiated by the addition of activated p38a except for preincubation experiments, where reactions are initiated by the addition of Hsp27 peptide and MgATP. Preincubation of p38a with inhibitor or p38a with unactivated His6-MK2 or unactivated GST-PRAK and inhibitor are performed at 2X final assay concentrations at room temperature 240 minutes prior to adding ATP and Hsp27 peptide to initiate catalysis.
  • p38a compound inhibitory potency is quantitated from dose-response IC50 values or Ki values from p38a/MK2 cascade assays while the substrate selectivity is calculated as a ratio of p38a/PRAK:p38a/MK2 IC50 values.
  • Species compounds of Formula I, described hereinabove, evaluated in this assay, are expected to provide a therapeutic benefit in the treatment of p38 kinase mediated diseases, such as autoimmune diseases and lymphoma.
  • Cytokine regulation in human monocytes The p38 pathway has been shown to be critical for the biosynthesis of a number of proinflammatory cytokines including TNFa, IL- ⁇ and IL-6. Evaluation of the potency and efficacy of p38 inhibitors to block cytokine production is carried out using the human U937 cell line.
  • the U937 human pre-monocytic cell line will be obtained from the American Type Culture Collection (Rockville, MD). These cells are differentiated to a monocytic/macrophage phenotype as described by Burnette (Burnette et al, (2009).
  • SD0006 a potent, selective and orally available inhibitor of p38 kinase, Pharmacology 84(l):42-60).
  • Differentiated U937 cells are seeded into 96-well tissue culture plates (200,000 cells/well) in complete media. After 24 hours, the cells are pretreated for 60 minutes in the presence or absence of compound and then stimulated with LPS (0.1 ⁇ g/mL) for 4 hours. Culture media are then collected for determination of TNFa, IL-6 or IL- ⁇ levels by ELISA. Cytokine concentrations are extrapolated from recombinant protein standard curves using a four- parameter logistic model and solving for IC 50 after iterating to the best least-squares fit. Species compounds of Formula I, described hereinabove, evaluated in this assay, are expected to provide a therapeutic benefit in the treatment of p38 kinase mediated diseases, such as lymphoma or inflammation.
  • Rheumatoid arthritis synovial fibroblasts are derived from the inflamed synovium of a female RA patient who was undergoing total knee replacement. Synovial tissue is teased away from adjacent cartilage and dispersed into single cells with collagenase. Cells are expanded and banked. RASF cells are further cultured as described by Burnette supra. RASF cells are seeded into 96-well tissue culture plates (5xl0 4 cells/well) in complete growth medium. After 24 hours, the medium is replaced with fresh growth medium containing 1% FBS.
  • Substrate selectivity in HUVEC cells When a compound is identified from the biochemical characterization step with selective inhibition of p38/MK2, it is next placed into a cell-based assay to verify enzyme to cell translatability. These assays utilize human umbilical vein endothelial cells (HUVEC) to demonstrate inhibition of Hsp27 phosphorylation (a biomarker of p38/MK2 activation) while sparing production of tissue factor (TF), which is linked to another downstream substrate of p38, MSK.
  • Hsp27 phosphorylation a biomarker of p38/MK2 activation
  • TF tissue factor
  • adherent HUVEC (at 5 passages or less) are treated for 1 hour with serially-diluted compounds, including a nonselective p38 inhibitor as a reference, or vehicle for controls.
  • Hsp27 phosphorylation cells are then stimulated with 500 pg/mL IL- ⁇ for 0.5 hours, media is removed, cells are lysed, and phospho-Hsp27 in the lysate is quantitated by enzyme-linked immunosorbent assay (ELISA)(Life Technologies, Carlsbad, CA).
  • ELISA enzyme-linked immunosorbent assay
  • the procedure for TF release is a similar ELISA- based assay (American Diagnostica, Stanford, CT), except that IL- ⁇ stimulation proceeds for 5 hours.
  • the ratio of TF inhibition IC50:HSP27 phosphorylation inhibition IC50 is defined as the substrate selectivity index in these cells.
  • Species compounds of Formula I, described hereinabove, evaluated in this assay, are expected to provide a therapeutic benefit in the treatment of p38 kinase mediated diseases, such as lymphoma and autoinflammatory disease.
  • Canine B cell growth regulation p38 inhibitors have been shown to uniquely inhibit canine B cell proliferation and survival. This selective effect on canine B cells may be exploited in therapeutic treatment for canine B cell lymphoma, a fatal disease that impacts >40,000 companion animals in the United States. Quantitation of impact of p38 inhibitors on B cell growth is a cellular indicator of efficacy in B cell lymphoma. Species compounds of Formula I, described hereinabove, evaluated in this assay, are expected to provide a therapeutic benefit in the treatment of p38 kinase mediated diseases, such as lymphoma.
  • Leukocytes are isolated from splenocytes by centrifugation through Histopaque 1077. To evaluate effect on proliferation, leukocytes are then cultured for 48 hours in 96-well plates in the presence of vehicle or test compounds. Cells are stimulated with LPS for TLR4 stimulation, Staphylococcus aureus B cell mitogen, or concanavalin-A T cell mitogen, then proliferation is quantitated with a BRDU incorporation ELISA (Roche, Mannheim, Germany).
  • leukocytes are plated on 96-well polypropylene U bottom plates and treated with p38 MAPK inhibitors or staurosporine (as a positive control) for up to 24 hours in the absence or presence of actinomycin D or cycloheximide (if needed to increase apoptosis rate).
  • Apoptosis is determined using Caspase-Glo 3/7 luminescent assay (Promega, Madison, WI). In both assays, values generated after incubation with increasing concentrations of the inhibitors are compared to a negative control without inhibitors.
  • LPS Induced TNFa Production in rats Rats are fasted eighteen hours prior to oral dosing, and allowed free access to water throughout the experiment. Each treatment group consists of five animals. Compounds are prepared as a suspension in a vehicle consisting of 0.5% methylcellulose, (Sigma Aldrich, St. Louis, MO), 0.025% Tween 20 (Sigma Aldrich). The compound or vehicle is administered by oral gavage in a volume of 1 mL. Two vehicle groups are used per experiment to control for intra-experiment variability. LPS (E.
  • coli serotype 0111 :B4, Sigma Aldrich is administered four hours after compound intravenous injection at a dose of 1 mg/kg in 0.5 mL sterile saline (Baxter Healthcare, Deerfield, IL). Blood is collected in serum separator tubes via cardiac puncture ninety minutes after LPS injection, a time point corresponding to maximal TNFa and IL- ⁇ production. After clotting, serum is withdrawn and stored at -20 °C and IL- ⁇ and TNFa levels quantitated by ELISA (Burnette supra). Species compounds of Formula I, described hereinabove, evaluated in this assay, are expected to provide a therapeutic benefit in the treatment of p38 kinase mediated diseases, such as lymphoma or inflammation.
  • p38 kinase mediated diseases such as lymphoma or inflammation.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present disclosure provides pyrimidine urea compounds useful in the treatment of p38 kinase mediated diseases, such as lymphoma and auto-inflammatory disease, having the structure of Formula (I): wherein R1, R3, R4, R5, V and W are as defined in the detailed description; pharmaceutical compositions comprising at least one of the compounds; and methods for treating p38 kinase mediated diseases using the compound.

Description

SUBSTITUTED PYRIMIDINE UREA COMPOUNDS
CROSS-REFERENCE TO RELATED APPLICATIONS
[0001] This application claims the benefit of U.S. Provisional Application No. 61/420,084, filed on December 6, 2010. The entire disclosure of the above application is incorporated herein by reference.
FIELD
[0002] The present disclosure generally relates to a compound having enzyme inhibitory activity, pharmaceutical compositions comprising the compound, and methods useful for treating diseases. More specifically, the present disclosure relates to a class of pyrimidine urea compounds, pharmaceutical compositions comprising the compound, and methods useful for treating p38 kinase mediated diseases.
BACKGROUND
[0003] Mitogen-activated protein kinases (MAPK) are a conserved family of enzymes that relay and propagate external stimuli, using phosphorylation cascades to generate a coordinated cellular response to the environment. The MAPK are proline-directed serine/threonine-specific protein kinases that regulate cellular activities, such as gene expression, mitosis, differentiation, and cell survival/apoptosis. To date, 4 distinct classes of mammalian MAPK have been identified: the extracellular signaling kinases (ERK1 and 2), the c-jun N-terminal kinase- 1 (JNK1-3), the p38 MAPK (ρ38α, β, γ, and δ), and ERK5. The MAPK are activated by the dual phosphorylation of Thr and Tyr residues within a TXY activation motif by coordinated dual- specificity MAPKK, where X is Glu, Pro, and Gly in ERK, JNK, and p38 MAPK, respectively. MAPK are 60-70% identical to each other, yet differ in their activation loop sequences and sizes. The activation loop is adjacent to the enzyme-active site, and its phosphorylation allows the enzyme to reposition active-site residues into the optimal orientation for substrate binding and catalysis. Downstream substrates of MAPK include mitogen-activated protein-kinase-activated protein (MAPKAP) kinases and transcription factors, the phosphorylation of which, either directly or indirectly, regulates gene expression at several points, including transcription, nuclear export, and mRNA stability and translation. The cellular consequences of MAPK activation include inflammation, apoptosis, differentiation, and proliferation.
[0004] Distinct genes encode 4 p38 MAPK in humans: ρ38α, β, γ, and δ. Significant amino acid sequence homology is observed among the 4 isoforms, with 60 -75 overall sequence identity and > 90% identity within the kinase domains. Tissue- selective expression is observed, with ρ38γ found predominantly in skeletal muscle, ρ38δ in the testes, pancreas, and small intestine. In contrast, p38a and β are more ubiquitously expressed.
[0005] An understanding of the broad biologic and pathophysiological roles of p38 MAPK family members has grown significantly over the past decade, as has the complexity of the signaling network leading to their activation. Scientific exploration of this pathway from biological, cellular, and in vivo perspectives was largely enabled by the availability of well- behaved, selective, small-molecule inhibitors of p38 MAPK that target the a and, to a lesser extent, β isoforms. p38 MAPK is the major isoform involved in the immune and inflammatory response. As such its function is critical for the production and activity of multiple proinflammatory cytokines, including TNFa, IL-1, IL-6, and IL-8, in cells such as macrophages, monocytes, synovial cells, and endothelial cells. p38 MAPK is also responsible for the induction of key inflammatory enzymes such as COX2 and iNOS, the major sources of eicosanoids and nitric oxide at sites of inflammation, respectively. Additionally, the p38 MAPK pathway regulates the expression of matrix metalloproteinases (MMP), including MMP2, MMP9, and MMP13.
[0006] The use of selective and potent inhibitors has facilitated the discovery of several families of p38 MAPK substrates, including transcription factors, MAPKAP kinases, and other enzymes. p38 MAPK can directly phosphorylate several transcription factors, such as myocyte - specific enhancer binding factor 2C (MEF2C), CHOP, peroxisome proliferator- activated receptor (PPAR) a, PPAR γ co-activator 1 and p53. These transcription factors are involved in cellular functions such as apoptosis, gluconeogenesis, and synthesis of enzymes involved in fatty acid oxidation. p38 MAPK is also involved in the direct or indirect phosphorylation of enzyme substrates, such as cytosolic phospholipase A2, and the Cdc25 phosphatases, which are involved in the activation of cyclin-dependent protein kinase activity and cell-cycle regulation. Therefore in addition to its role in the inflammatory response, p38 MAPK has other functions associated with normal and abnormal cell growth and survival as well as cellular function and homeostasis. [0007] The MAPKAP kinases— MK2, MK-3, and PRAK— are selectively phosphorylated by p38 MAPK, while the phosphorylation of MSK1/2, MNK1/2, and RSKb is catalyzed by both p38 MAPK and ERK. Activation of RSKb is thought to play a role in cell survival, although the identification of substrates has been difficult, due to the lack of specific inhibitors. MNK is involved in the phosphorylation of eukaryotic initiation factor-4E, which binds to the 'cap' structure of mRNA and enhances protein translation. MNK phosphorylates the mRNA binding protein hnRNP-AO, a protein that regulates mRNA stability of transcripts encoding inflammatory proteins. MSK1/2 is involved in the phosphorylation of the transcription factors CREB and ATF- 1, which regulate AP-1 binding proteins. In addition, MSK1/2 can phosphorylate Histone H3, which is involved in chromatin remodeling. While evidence suggests that MSK and MNK play a role in the mediation of pro-inflammatory cytokines, in vivo data with selective inhibitors and/or knockout mice are lacking.
[0008] MK-2, MK-3, and PRAK, once phosphorylated and activated by p38 MAPK, share similar substrate specificities. All of these kinases can phosphorylate the small heat-shock protein Hsp27. Studies have shown that the PRAK- and MK3-deficient mice do not display any resistance to endotoxic shock or a decrease in lipopolysaccharide-(LPS)-induced cytokine production. In contrast, MK-2-deficient mice show a resistance to endotoxic shock and an impaired inflammatory response, as well as a significantly decreased production of cytokines such as TNFa, IFNyD and IL-6. Thus, the p38/MK2 axis specifically is necessary and sufficient for mediating pro-inflammatory responses.
[0009] Recently, Davidson et al (2004) Discovery and characterization of a substrate selective p38alpha inhibitor, Biochemistry 43: 11658-71, described a novel approach for increasing selectivity of a p38 MAPK inhibitors. In these studies, a high throughput screen was carried out using an assay that measured the p38-dependent phosphorylation and activation of MK2. The p38:MK2 complex is very stable with a Kd of 6 nM. The binding affinity of p38 for MK2 is driven by the C-terminal domain of MK2 containing several positively charged amino acid residues. Crystallographic studies of the p38:MK2 complex demonstrated that the C- terminal region of MK2 wraps around p38a and binds to the negatively charged ED binding site. The tight binding of p38 to MK2 may give rise to conformational changes providing additional binding pockets for inhibitors that would specifically be dependent upon the p38:MK2 interaction. [0010] Taking advantage of the p38:MK2 interaction and using MK2 as the p38 substrate, a novel inhibitor of p38a was discovered exhibiting interesting properties (Davidson et al). This inhibitor demonstrated substrate selectivity by preventing the p38a dependent phosphorylation of MK2 (Ki app 300 nM) while sparing the p38a dependent phosphorylation of ATF2 (Ki app > 20 uM). This novel inhibitor is functionally unique compared with traditional p38 ATP competitive inhibitors that block the p38-dependent phosphorylation of all p38 substrates. A second independent study also describes p38 inhibitors with unique mechanistic properties. This work demonstrates a novel mechanism for the selective inhibition of the p38 dependent phosphorylation of MK2. Unlike the previous study of Davidson et al., these mechanistically unique compounds are competitive with ATP and stabilize the p38/MK2 complex. Taken together, these two studies clearly prove the concept that selective p38/MK2 axis blockade is achievable with small molecule inhibitors. In comparison to traditional p38 MAPK inhibitors these p38/MK2 inhibitors should retain or enhance potency and exhibit improved safety features in animal models of disease or in human clinical settings.
[0011] The p38/MK2 role in the regulation of inflammatory cytokines (TNFa, IL-Ιβ, IL-6) and enzymes responsible for inflammation (COX-2, iNOS, and MMPs) makes it an attractive drug target. Several classical p38 MAPK inhibitors have progressed to testing in clinical trials. Some of these candidates have failed, for safety or other reasons, but several have reported clinical data in diseases such as rheumatoid arthritis, pain, Crohn's disease, acute coronary syndrome, multiple myeloma and chronic obstructive pulmonary disease. In addition to these diseases several IL-Ιβ mediated diseases could be impacted by a p38 inhibitor based upon the key role for the p38 MAPK pathway in the biosynthesis and activity of this cytokine. These diseases include the family of cryopyrin associated periodic disorders (CAPS), chronic gout, diabetes, Still's disease, Familial Mediterranean Fever among others.
[0012] In addition to human inflammatory pathways, p38 MAPK has been linked to canine B cell growth and survival. The role of p38 MAPK in B cell growth suggests that inhibition of this enzyme may be therapeutically beneficial for the treatment of canine B cell lymphoma. Canine lymphoma is one of the most common malignancies diagnosed in companion animals representing 10-25% of canine neoplasms and >80% of the hematopoietic tumors. An orally available, selective B cell growth inhibitor would meet a significant unmet medical need. [0013] Compounds useful for treating diseases and conditions caused or exacerbated by unregulated p38 MAP Kinase and/or TNF activity are described in WO 2000/017175 published 30 March 2000. The compounds described therein include a class of substituted urea compounds.
[0014] Compounds useful for treating diseases and conditions caused or exacerbated by unregulated p38 MAP Kinase and/or TNF activity are described in WO 2000/071535 published 30 November 2000. The compounds described therein include a class of indole-type compounds.
[0015] Compounds useful for treating diseases and conditions caused or exacerbated by unregulated p38 MAP Kinase and/or TNF activity are described in WO 2002/042292 published 30 May 2002. The compounds described therein include a class of coupled indole-type derivatives.
[0016] Compounds useful for prophylaxis or treatment of circulatory diseases, metabolic diseases and/or central nervous system diseases are described in WO 2008/062905 published 29 May 2008. The compounds described therein include an alkyl-pyrimidinone-phenyl compounds wherein the phenyl fragment is substituted with a cyclopropyl radical, e.g., 6-butyl-3-(3- cyclopropylphenyl)-2methyl-5-{ [2'-(5-oxo-4,5-dihydro-l,2,4-oxadizol-3-yl)biphenyl-4- yl]methyl}pyrimidin-4(3H)-one.
[0017] Various potential inhibitors or modulators of p38 kinase and the p38 kinase pathway are described in WO 2005/018557 published 3 March 2005. The compounds described therein include di-fluorophenyl-methoxy-pyridinone-pyridyl compounds wherein the pyridyl fragment is substituted with various radicals including: alkyl, alkenyl, hydroxyalkyl, halo, cyano, amino, carboxy, carbamoyl, methoxycarbonyl and hydroxyalkenylimino radicals.
[0018] Compounds useful for treating diseases and conditions caused or exacerbated by unregulated p38 MAP Kinase and/or TNF activity are described in US 2007/0167621 published 19 July 2007. The compounds described therein include di-fluorophenyl-methoxy-pyrimidinone- phenyl compounds wherein the phenyl fragment is substituted with methyl amido radical.
[0019] Compounds useful for treating diseases and conditions caused or exacerbated by unregulated p38 MAP Kinase and/or TNF activity are described in WO 2004/087677 published 14 October 2004. The compounds described therein include di-fluorophenyl-methoxy- pyrimidinone-phenyl compounds wherein the phenyl fragment is substituted with piperazinyl or a morpholinyl radical through a carbonyl bridge. [0020] Pyrimidinone derivatives (as inhibitors of protein kinases and useful in treating disorders related to abnormal protein kinase activities such as inflammatory diseases and certain types of cancer), are described in WO 2007/081901 published 19 July 2008. The compounds described therein include di-fluorophenyl-methoxy-pyrimidinone-phenyl compounds wherein the phenyl fragment is substituted with a cyclopropanyl or a morpholinyl radical through an amidoalkylamido bridge.
[0021] Pyrimidinone derivatives (as inhibitors of protein kinases and useful in treating disorders related to abnormal protein kinase activities such as inflammatory diseases and certain types of cancer) are described in WO 2008/153942 published 18 December 2008. The compounds described therein include di-fluorophenyl-methoxy-pyrimidinone-phenyl compounds where the phenyl radical is substituted with cyclopentyl or a cyclohexyl radical through an amido bridge.
[0022] Compounds useful for treating diseases and conditions caused or exacerbated by unregulated p38 MAP Kinase and/or TNF activity are described in 7,067,540 published 27 June 2007. The compounds described therein include di-fluorophenyl-methoxy-pyridinone-phenyl compounds wherein the phenyl radical is substituted with a Cs-heteroaryl radical (e.g., pyrazolyl or imidazolyl).
SUMMARY
[0023] In one embodiment, there is rovided a compound of Formula (I):
Figure imgf000007_0001
and the pharmaceutically acceptable salts thereof; wherein R1, R3, R4, R5, W and V are as defined in the Detailed Description. [0024] In another embodiment, there is provided a pharmaceutical composition comprising a compound of Formula (I) or a pharmaceutically acceptable salt thereof, and a pharmaceutically- acceptable carrier.
[0025] In various embodiments, the pharmaceutical composition further comprises one or more additional pharmaceutically active compounds.
[0026] In yet another embodiment, there is provided a method for treating a condition comprising administering to a subject a therapeutically effective amount of a compound of Formula (I), wherein the condition to be treated includes, but is not limited to, autoimmune disorders, chronic inflammatory disorders, acute inflammatory disorders, auto-inflammatory disorders, pain, atherosclerosis, diabetes, fibrotic diseases, metabolic disorders, cancer, neoplasia, leukemia, and lymphoma.
[0027] In various embodiments, the method comprises administering a combination of a compound of Formula (I) and at least one additional pharmaceutically active compound.
[0028] In another embodiment, the invention comprises use of a compound having the structure of Formula I, or pharmaceutically acceptable salts thereof, for the manufacture of a medicament for the treatment of a condition in a subject. The conditions that can be treated in accordance with the present invention include autoimmune disorders, chronic inflammatory disorders, acute inflammatory disorders, auto-inflammatory disorders, pain, atherosclerosis, diabetes, fibrotic diseases, metabolic disorders, cancer, neoplasia, leukemia, lymphoma and the like.
[0029] In another embodiment, the invention comprises methods for making a compound having the structure of Formula I, or pharmaceutically acceptable salts thereof.
[0030] In another embodiment, the invention comprises intermediates useful in making a compound having the structure of Formula I, or pharmaceutically acceptable salts thereof.
DETAILED DESCRIPTION
[0031] The following description is merely exemplary in nature and is not intended to limit the present disclosure, application, or uses.
A. Definitions
[0032] The use of generic terms in the description of the compounds are herein defined for clarity. [0033] This specification uses the terms "substituent", "radical", "group", "moiety", and "fragment" interchangeably.
[0034] The term "hydrido" denotes a single -H atom (H) and may be used interchangeably with the symbol "H" or the term "hydrogen".
[0035] If a substituent is described as being "optionally substituted," the substituent may be either (1) not substituted or (2) substituted. If a substitutable position is not substituted, the default substituent is a hydrido radical.
[0036] As used herein, the singular forms "a" and "an" may include plural reference unless the context clearly dictates otherwise.
[0037] The term "alkyl", either alone or within other terms such as "haloalkyl" and "alkylaryl", refers to an acyclic alkyl radical containing 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 carbon atoms. In some embodiments, alkyl is a C Qo alkyl group or a Ci-C6 alkyl group. Examples of alkyl groups include, but are not limited to, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, t- butyl, pentyl, hexyl, heptyl, octyl, nonyl and decyl.
[0038] The term "alkoxy" is RO- where R is alkyl as defined herein. Non-limiting examples of alkoxy groups include methoxy, ethoxy and propoxy. The terms alkyloxy and alkoxy may be used interchangeably.
[0039] The term "alkoxyalkyl" refers to an alkyl moiety substituted with an alkoxy group. Examples of alkoxyalkyl groups include methoxymethyl, methoxyethyl, methoxypropyl and ethoxyethyl.
[0040] The term "aralkoxy" embraces an arylalkyl radical attached through an oxygen atom to the parent molecular scaffold. The terms "arylalkoxy" and "aralkoxy" may be used interchangeable.
[0041] The term "aryl" refers to any monocyclic, bicyclic or tricyclic carbon ring of up to 6 atoms in each ring, wherein at least one ring is aromatic, or an aromatic ring system of 5 to 14 carbons atoms which includes a carbocyclic aromatic group fused with a 5-or 6-membered cycloalkyl group. Examples of aryl groups include, but are not limited to, phenyl, naphthyl, tetrahydronaphthyl and indanyl.
[0042] The term "arylalkyl" embraces an aryl-substituted alkyl radical and may be used interchangeably with the term "aralkyl". Examples include benzyl, diphenylmethyl, triphenylmethyl, phenylethyl and diphenylethyl. The terms benzyl and phenylmethyl are interchangeable.
[0043] The term "aryloxy" is RO-, where R is aryl. "Arylthio" is RS-, where R is aryl.
[0044] The term "aryloxyalkyl" embraces an aryloxy radical attached to an alkyl group.
[0045] The term "cyano" denotes a carbon radical having 3 of 4 covalent bonds shared by a nitrogen atom.
[0046] The term "cycloalkyl" is a hydrocarbyl group containing at least one saturated or partially unsaturated ring structure, and attached via a ring carbon. In various embodiments, it refers to a saturated or a partially unsaturated C3-C12 cyclic moiety. Examples of cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cycloheptyl and cyclooctyl.
[0047] The term "halo" refers to fluoro (-F), chloro (-C1), bromo (-Br), or iodo (-1).
[0048] The term "haloalkyl" refers to an alkyl moiety substituted with one or more halo groups. Examples of haloalkyl groups include -CF3 and -CHF2.
[0049] The term "haloaralkoxy" refers to aralkoxy group substituted with one or more halo radicals. Examples of haloaralkoxy groups include fluorobenzyloxy, difluorobenzyloxy. In various embodiments of the invention, haloaralkoxy is 4-fluorobenzyloxy or 2,4- difluorobenzyloxy.
[0050] The term "heterocyclyl" includes the heteroaryls defined below and refers to a saturated or partially unsaturated monocyclic, bicyclic or tricyclic group of 2 to 14 ring-carbon atoms and, in addition to ring-carbon atoms, 1 to 4 heteroatoms selected from P, N, O and S. In various embodiments the heterocyclic group is attached to another moiety through carbon or through a heteroatom, and is optionally substituted on carbon or a heteroatom. Examples of heterocyclyl include azetidinyl, benzoimidazolyl, benzofuranyl, benzofurazanyl, benzopyrazolyl, benzotriazolyl, benzothiophenyl, benzoxazolyl, carbazolyl, carbolinyl, cinnolinyl, furanyl, imidazolyl, indolinyl, indolyl, indolazinyl, indazolyl, isobenzofuranyl, isoindolyl, isoquinolyl, isothiazolyl, isoxazolyl, naphthpyridinyl, oxadiazolyl, oxazolyl, oxazoline, isoxazoline, oxetanyl, pyranyl, pyrazinyl, pyrazolyl, pyridazinyl, pyridopyridinyl, pyridazinyl, pyridyl, pyrimidyl, pyrrolyl, quinazolinyl, quinolyl, quinoxalinyl, tetrahydropyranyl, tetrahydrothiopyranyl, tetrahydroisoquinolinyl, tetrazolyl, tetrazolopyridyl, thiadiazolyl, thiazolyl, thienyl, triazolyl, azetidinyl, 1,4-dioxanyl, hexahydroazepinyl, piperazinyl, piperidinyl, pyridin-2-onyl, pyrrolidinyl, morpholinyl, thiomorpholinyl, dihydrobenzoimidazolyl, dihydrobenzofuranyl, dihydrobenzothiophenyl, dihydrobenzoxazolyl, dihydrofuranyl, dihydroimidazolyl, dihydroindolyl, dihydroisooxazolyl, dihydroisothiazolyl, dihydrooxadiazolyl, dihydrooxazolyl, dihydropyrazinyl, dihydropyrazolyl, dihydropyridinyl, dihydropyrimidinyl, dihydropyrrolyl, dihydroquinolinyl, dihydrotetrazolyl, dihydrothiadiazolyl, dihydrothiazolyl, dihydrothienyl, dihydrotriazolyl, dihydroazetidinyl, methylenedioxybenzoyl, tetrahydrofuranyl, and tetrahydrothienyl, and N-oxides thereof.
[0051] The term "heteroaryl" refers to a monocyclic, bicyclic or tricyclic ring having up to 6 atoms in each ring, wherein at least one ring is aromatic and contains from 1 to 4 heteroatoms in the ring selected from the group consisting of N, O and S. Non-limiting examples of heteroaryl include pyridyl, thienyl, furanyl, pyrimidyl, imidazolyl, pyranyl, pyrazolyl, thiazolyl, thiadiazolyl, isothiazolyl, oxazolyl, isoxazoyl, pyrrolyl, pyridazinyl, pyrazinyl, quinolinyl, isoquinolinyl, benzofuranyl, dibenzofuranyl, dibenzothiophenyl, benzothienyl, indolyl, benzothiazolyl, benzooxazolyl, benzimidazolyl, isoindolyl, benzotriazolyl, purinyl, thianaphthenyl and pyrazinyl. Attachment of heteroaryl can occur via an aromatic ring, or, if heteroaryl is bicyclic or tricyclic and one of the rings is not aromatic or contains no heteroatoms, through a non-aromatic ring or a ring containing no heteroatoms. "Heteroaryl" is also understood to include the N-oxide derivative of any nitrogen containing heteroaryl.
[0052] The term "heteroaralkoxy" embraces a heteroarylalkyl radical attached through an oxygen atom to the molecular scaffold. A class of preferred heteroaralkoxy radicals is "lower heteroaralkoxy" radicals having an alkyl range of 1-3 carbon atoms. A preferred class of C6- heteroarylalkoxy radicals is (pyridin-2-yl)methoxy.
[0053] The term "heteroaryloxy" is RO-, where R is heteroaryl as defined herein. Examples include thiophen-2-yl-oxy, pyridin-2-yl-oxy, pyridin-3-yl-oxy, and pyridin-4-yl-oxy.
[0054] The term "heteroaryloxyalkyl" is a heteroaryloxy radical further attached to an alkyl radical.
[0055] The term "hydroxyl" refers to -OH radical and may be used interchangeably with "hydroxyl".
[0056] The term "hydroxyalkyl" refers to a linear or branched monovalent C Qo hydrocarbon group substituted with at least one hydroxy group and examples of hydroxyalkyl groups include, but are not limited to, hydroxymethyl, hydroxyethyl, hydroxypropyl and hydroxybutyl. [0057] The number of carbon atoms in a hydrocarbyl substituent can be indicated by the prefix "Cx-Cy" where X is the minimum and Y is the maximum number of carbon atoms in the substituent.
[0058] The term "pharmaceutically-acceptable" means suitable for use in pharmaceutical preparations, generally considered as safe for such use, officially approved by a regulatory agency of a national or state government for such use, or being listed in the U. S. Pharmacopoeia or other generally recognized pharmacopoeia for use in animals, and more particularly in humans.
[0059] The term "pharmaceutically-acceptable salt" refers to a salt which may enhance desired pharmacological activity. Examples of pharmaceutically-acceptable salts include acid addition salts formed with inorganic or organic acids, metal salts and amine salts. Examples of acid addition salts formed with inorganic acids include salts with hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid and phosphoric acid. Examples of acid addition salts formed with organic acids such as acetic acid, propionic acid, hexanoic acid, heptanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, o-(4-hydroxy- benzoyl)-benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, 1,2-ethanedisulfonic acid, 2-hydroxyethane-sulfonic acid, benzenesulfonic acid, p- chlorobenzenesulfonic acid, 2-naphthalenesulfonic acid, p-toluenesulfonic acid, camphorsulfonic acid, 4-methyl-bicyclo[2.2.2]oct-2-enel-carboxylic acid, gluco-heptonic acid, 4,4'- methylenebis(3-hydroxy-2-naphthoic) acid, 3-phenylpropionic acid, trimethyl- acetic acid, tertiary butylacetic acid, lauryl sulfuric acid, gluconic acid, glutamic acid, hydroxy-naphthoic acids, salicylic acid, stearic acid and muconic acid. Examples of metal salts include salts with sodium, potassium, calcium, magnesium, aluminum, iron, and zinc ions. Examples of amine salts include salts with ammonia and organic nitrogenous bases strong enough to form salts with carboxylic acids.
[0060] The term "therapeutically-effective amount" refers to an amount of a compound that, when administered to a subject for treating a disease, is sufficient to effect treatment for the disease. "Therapeutically effective amount" can vary depending on the compound, the disease and its severity, the age, the weight, etc. of the subject to be treated.
[0061] Compounds of the present invention can exist in tautomeric, geometric or stereoisomeric forms. The compounds' corresponding esters, metabolites, oximes, prodrugs, oniums and N-oxides are also embraced by the invention. The present invention contemplates all such compounds, including cis- and trans-geometric isomers, E- and Z-geometric isomers, R- and S-enantiomers, diastereomers, d-isomers, 1-isomers, mixtures of isomers and racemates thereof, as falling within the scope of the invention.
[0062] The terms "cis" and "trans" denote a form of geometric isomerism in which two carbon atoms connected by a double bond will each have a radical atom on the same side of the double bond ("cis") or on opposite sides of the double bond ("trans").
[0063] Some of the compounds described contain one or more stereocenters and are meant to include R, S and mixtures of R and S forms for each stereocenter present.
[0064] The compounds of the invention may also exist as atropisomers, i.e., chiral rotational isomers. The invention encompasses the racemic, resolved atropisomers, and mixtures thereof.
B. Compounds
[0065] The present disclosure provides a class of compounds, including pharmaceutically acceptable salts of the compounds wherein the compounds have the structure of Formula I:
Figure imgf000013_0001
wherein:
V and W are independently selected from the group consisting of CH and N;
R1 is selected from the group consisting of cycloalkyl, aryl, heterocyclyl and heteroaryl; wherein the cycloalkyl and aryl substituents may be optionally substituted with one or more substituents independently selected from the group consisting of halo, cyano, alkyl and alkoxy; and wherein the heterocyclyl and heteroaryl substituents may be optionally substituted with one or more substituents independently selected from the group consisting of cyano and alkyl;
R 3J and R 5J are independently selected from the group consisting of alkyl, halo and hydrogen; and R4 is selected from the group consisting of cycloalkyl, aryl, heterocyclyl and heteroaryl; wherein the cycloalkyl and aryl substituents may be optionally substituted with one or more substituents independently selected from the group consisting of halo, hydroxy, cyano, alkyl, alkoxy, hydroxyalkyl, alkoxyalkyl and aminoalkyl; and wherein the heterocyclyl and heteroaryl substituents may be optionally substituted with one or more substituents independently selected from the group consisting of cyano, alkyl, hydroxyalkyl, alkoxyalkyl and aminoalkyl.
[0066] In one embodiment of the compounds of Formula I, V is CH and W is selected from the group consisting of CH and N; R1 is selected from the group consisting of heterocyclyl and heteroaryl; wherein the heterocyclyl and heteroaryl substituents may be optionally substituted with one or more substituents independently selected from the group consisting of (Ci-C3)-alkyl and cyano; R 3J and R 5J are independently selected from the group consisting of hydrogen, halo and (C1-C3)-alkyl; and R4 is (Cs-C6)-heteroaryl; wherein the (Cs-C6)-heteroaryl substituent may be optionally substituted with one or more substituents independently selected from the group consisting of alkyl and hydroxyalkyl.
[0067] In another embodiment of the compounds of Formula I, V is CH and W is selected from the group consisting of CH and N; R1 is (Cs-C6)-heteroaryl; wherein the (Cs-C6)-heteroaryl substituent may be optionally substituted with one or more substituents independently selected from (Ci-C3)-alkyl; R3 is selected from the group consisting of methyl and fluoro; R4 is selected from the group consisting of pyridine and pyrimidine; wherein the pyridine and pyrimidine substituents may be optionally substituted with one or more substituents independently selected from the group consisting of alkyl and hydroxyalkyl; and R5 is selected from the group consisting of hydrogen and fluoro.
[0068] In still another embodiment of the compounds of Formula I, V is CH and W is selected from the group consisting of CH and N; R1 is Cs-heteroaryl, comprising one or more heteroatoms selected from the group consisting of O, N and S; and wherein the Cs-heteroaryl may be optionally substituted with one or more methyl substituents; R is selected from the group consisting of methyl and fluoro; R4 is selected from the group consisting of pyridine and pyrimidine; wherein the pyridine and pyrimidine substituents may be optionally substituted with one or more substituents independently selected from the group consisting of alkyl and hydroxyalkyl; and R5 is selected from the group consisting of hydrogen and fluoro. [0069] The present invention is also directed to a subclass of compounds, including pharmaceutically acceptable salts of the compounds, wherein the compounds have the structure of Formula II:
Figure imgf000015_0001
wherein:
W is selected from the group consisting of CH and N;
X is selected from the group consisting of O, S and CH;
Y is selected from the group consisting of N and C;
R is selected from the group consisting of methyl and fluoro;
R5 is selected from the group consisting of hydrogen and fluoro;
R10 is selected from (C1-C3)-alkyl; and
R40 is selected from the group consisting of methyl and hydroxy.
[0070] Non-limiting examples of Formula (II) compounds include the following compounds and pharmaceutically acceptable salts thereof:
Figure imgf000015_0002
[0071] In another embodiment of the compounds of Formula I, V is CH and W is selected from the group consisting of CH and N; R 1 is pyridine; R 3 is selected from the group consisting of methyl and fluoro; R is selected from the group consisting of pyridine and pyrimidine; wherein the pyridine and pyrimidine substituents may be optionally substituted with one or more substituents independently selected from the group consisting of alkyl and hydroxyalkyl; and R5 is selected from the group consisting of hydrogen and fluoro.
[0072] The present invention is also directed to a subclass of compounds, including pharmaceutically acceptable salts of the compounds, wherein the compounds have the structure of Formula III:
Figure imgf000016_0001
wherein:
W is selected from the group consisting of CH and N;
R is selected from the group consisting of methyl and fluoro;
R5 is selected from the group consisting of hydrogen and fluoro; and
R40 is selected from the group consisting of methyl and hydroxy.
[0073] In another embodiment, the compound of Formula III is l-(5-(2-(2-hydroxypropan-2- yl)pyrimidin-4-yl)-2-methylphenyl)-l-(2-(pyridin-2-yl)pyrimidin-4-yl)urea.
[0074] In another embodiment of the compounds of Formula I, V is CH and W is selected from the group consisting of CH and N; R1 is selected from the group consisting of cycloalkyl and aryl; wherein the cycloalkyl and aryl substituents may be optionally substituted with one or more substituents independently selected from the group consisting of (Ci-C3)-alkyl and halo; R and R5 are independently selected from the group consisting of hydrogen, halo and (C1-C3)-alkyl; and R4 is (C5-C6)-heteroaryl; wherein the (Cs-C6)-heteroaryl substituent may be optionally substituted with one or more substituents independently selected from the group consisting of alkyl and hydroxyalkyl.
[0075] In another embodiment of the compounds of Formula I, V is CH and W is selected from the group consisting of CH and N; R1 is phenyl optionally substituted with one or more halo substituents; R3 is selected from the group consisting of methyl and fluoro; R4 is selected from the group consisting of pyridine and pyrimidine; wherein the pyridine and pyrimidine substituents may be optionally substituted with one or more substituents independently selected from the group consisting of alkyl and hydroxyalkyl; and R5 is selected from the group consisting of hydrogen and fluoro.
[0076] In still another embodiment of the compounds of Formula I, V is CH and W is selected from the group consisting of CH and N; R1 is phenyl optionally substituted with one or more fluoro substituents; R3 is selected from the group consisting of methyl and fluoro; R4 is selected from the group consisting of pyridine and pyrimidine; wherein the pyridine and pyrimidine substituents may be optionally substituted with one or more substituents independently selected from the group consisting of alkyl and hydroxyalkyl; and R5 is selected from the group consisting of hydrogen and fluoro.
[0077] The present invention is also directed to a subclass of compounds, including pharmaceutically acceptable salts of the compounds, wherein the compounds have the structure of Formula IV:
Figure imgf000017_0001
wherein:
W is selected from the group consisting of CH and N;
R is selected from the group consisting of methyl and fluoro;
R5 is selected from the group consisting of hydrogen and fluoro;
R10 and R11 are independently selected from the group consisting of hydrogen and fluoro; and
R40 is selected from the group consisting of methyl and hydroxy.
[0078] In another embodiment, the compounds of Formula IV are selected from the group consisting of:
Figure imgf000018_0001
C. Methods of Treatment
[0079] The present invention further comprises methods for treating a condition in a subject having or susceptible to having such a condition, by administering to the subject a therapeutically-effective amount of one or more compounds as described above. In one embodiment, the treatment is preventative treatment. In another embodiment, the treatment is palliative treatment. In another embodiment, the treatment is restorative treatment.
1. Conditions
[0080] The conditions that can be treated in accordance with the present invention include, but are not limited to, autoimmune disorders, chronic inflammatory disorders, acute inflammatory disorders, auto-inflammatory disorders, pain, atherosclerosis, diabetes, fibrotic diseases, metabolic disorders, cancer, neoplasia, leukemia, lymphoma and the like.
[0081] In some embodiments the methods described herein are used to treat patients with disorders arising from dysregulated cytokine, enzymes and/or inflammatory mediator production, stability, secretion, posttranslational processing. Examples of cytokines that may be dysregulated include interleukins 1, 2, 6, 8, 10, 12, 17, 22 and 23 along with tumor necrosis factor alpha and interferons alpha, beta and gamma. Examples of inflammatory mediators that may be dysregulated include nitric oxide, prostaglandins and leukotrienes. Examples of enzymes include cyclo-oxygenase, nitric oxide synthase and matrixmetallopro tease.
[0082] In some embodiments the methods described herein are used to treat patients with dysregulated p38 activity, activation, biosynthesis or pathway function.
[0083] In some embodiments, the methods described herein are used to treat a patient in need thereof suffering from an autoimmune disorder, chronic and/or acute inflammatory disorder and/or auto-inflammatory disorder. Examples of disorders include, but are not limited to colitis, multiple sclerosis, arthritis, rheumatoid arthritis, osteoarthritis, juvenile arthritis, psoriatic arthritis, cryopyrin associated periodic syndromes, Muckle- Wells Syndrome, Familial Cold Auto-inflammatory Syndrome, neonatal-onset multisystem inflammatory disease, TNF receptor associated periodic syndrome, acute pancreatitis, chronic pancreatitis, atherosclerosis, inflammatory bowel disease, Crohn's disease, ulcerative colitis, Diabetes mellitus type 1, Diabetes mellitus type 2, diabetic retinopathy, Still's disease, multiple sclerosis, vasculitis, sarcoidosis, pulmonary inflammation, acute respiratory distress syndrome, wet and dry age- related macular degeneration, autoimmune hemolytic syndromes, autoimmune hepatitis, autoimmune neuropathy, autoimmune ovarian failure, autoimmune orchitis, autoimmune thrombocytopenia, reactive arthritis, ankylosing spondylitis, silicone implant associated autoimmune disease, Sjogren's syndrome, Familial Mediterranean Fever, systemic lupus erythematosus, vasculitis syndromes (such as, for example, giant cell arteritis, Behcet's disease & Wegener's granulomatosis), Vitiligo, secondary hematologic manifestation of autoimmune diseases (such as, for example, anemias), drug-induced autoimmunity, Hashimoto's thyroiditis, hypophysitis, idiopathic thrombocytic pupura, metal-induced autoimmunity, myasthenia gravis, pemphigus, autoimmune deafness (including, for example, Meniere's disease), Goodpasture's syndrome, Graves' disease, HW-related autoimmune syndromes and Gullain-Barre disease; Examples of inflammatory conditions include, but are not limited to sepsis, septic shock, endotoxic shock, exotoxin-induced toxic shock, gram negative sepsis, toxic shock syndrome, glomerulonephritis, peritonitis, interstitial cystitis, psoriasis, atopic dermatitis, hyperoxia-induced inflammations, asthma, chronic obstructive pulmonary disease (COPD), vasculitis, graft vs. host reaction (i.e., graft vs. host disease), allograft rejections (e.g., acute allograft rejection, and chronic allograft rejection), early transplantation rejection (e.g., acute allograft rejection), reperfusion injury, acute pain, chronic pain, neuropathic pain, Fibromyalgia ,pancreatitis, chronic infections, meningitis, encephalitis, myocarditis, gingivitis, post surgical trauma, tissue injury, traumatic brain injury, hepatitis, enterocolitis, sinusitis, uveitis, ocular inflammation, optic neuritis, gastric ulcers, esophagitis, peritonitis, periodontitis, dermatomyositis, gastritis, myositis, polymyalgia, pneumonia and bronchitis. Fibrotic diseases; Metabolic disorders, including but not limited Obesity, steroid-resistance, glucose intolerance, metabolic syndrome. In some embodiments, the methods described herein can be used to treat a patient in need thereof and suffering from neoplasia. Examples of these conditions include but not limited to angiogenesis, multiple myeloma, leukemia, B cell lymphoma, T cell lymphoma, mast cell tumors, lymphoma, Hodgkin's disease, cancer of the bone, mouth/pharynx, oesophagus, larynx, stomach, intestine, colon, rectum, lung, liver, pancreas, nerve, brain, head and neck, throat, ovary, uterus, prostate, testis, bladder, kidney, breast non- small cell lung carcinoma, melanoma, skin cancer, teratoma, rhabdomyosarcoma, glioma, metastatic and bone disorders. In some embodiments, the disease associated with dysregulated p38 include Cardiovascular and Cerebrovascular diseases, including but not limited to Atherosclerosis, restenosis of an atherosclerotic coronary artery, Acute coronary syndrome, myocardial infarction, cardiac-allograft vasculopathy and stroke; central nervous system disorders with an inflammatory or apoptotic component, Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, spinal cord injury, neuronal ischemia and peripheral neuropathy. The term patient refers to both humans and nonhuman animals with the abovementioned conditions. Nonhuman animals could be companion animals such as, but not limited to canine and feline species.
2. Subjects
[0084] Suitable subjects to be treated according to the present invention include mammalian subjects. Mammals according to the present invention include, but are not limited to, human, canine, feline, bovine, caprine, equine, ovine, porcine, rodents, lagomorphs, primates, and the like, and encompass mammals in utero. Subjects may be of either gender and at any stage of development.
3. Administration and Dosing
[0085] The compounds of the present invention are generally administered in a therapeutically effective amount.
[0086] The compounds of the present invention can be administered by any suitable route in the form of a pharmaceutical composition adapted to such a route, and in a dose effective for the treatment intended. An effective dosage is typically in the range of about 0.001 to about 100 mg per kg body weight per day, preferably about 0.01 to about 30 mg/kg/day, in single or divided doses. Depending on age, species and condition being treated, dosage levels below the lower limit of this range may be suitable. In other cases, still larger doses may be used without harmful side effects. Larger doses may also be divided into several smaller doses, for administration throughout the day.
D. Pharmaceutical Compositions
[0087] For the treatment of the conditions referred to above, the compounds of described herein can be administered as follows:
[0088] Oral Administration
[0089] The compounds of the present invention may be administered orally, including by swallowing, so that the compound enters the gastrointestinal tract, or absorbed into the blood stream directly from the mouth (e.g., buccal or sublingual administration).
[0090] Suitable compositions for oral administration include solid formulations such as tablets, lozenges and capsules, which can contain liquids, gels, or powders.
[0091] Compositions for oral administration may be formulated as immediate or modified release, including delayed or sustained release, optionally with enteric coating.
[0092] Liquid formulations can include solutions, syrups and suspensions, which can be used in soft or hard capsules. Such formulations may include a pharmaceutically acceptable carrier, for example, water, ethanol, polyethylene glycol, cellulose, or an oil. The formulation may also include one or more emulsifying agents and/or suspending agents.
[0093] In a tablet dosage form the amount of drug present may be from about 0.05% to about 95% by weight, more typically from about 2% to about 50% by weight of the dosage form. In addition, tablets may contain a disintegrant, comprising from about 0.5% to about 35% by weight, more typically from about 2% to about 25% of the dosage form. Examples of disintegrants include methyl cellulose, sodium or calcium carboxymethyl cellulose, croscarmellose sodium, polyvinylpyrrolidone, hydroxypropyl cellulose, starch and the like.
[0094] Suitable lubricants, for use in a tablet, may be present in amounts from about 0.1% to about 5% by weight, and include calcium, zinc or magnesium stearate, sodium stearyl fumarate and the like. [0095] Suitable binders, for use in a tablet, include gelatin, polyethylene glycol, sugars, gums, starch, hydroxypropyl cellulose and the like. Suitable diluents, for use in a tablet, include mannitol, xylitol, lactose, dextrose, sucrose, sorbitol and starch.
[0096] Suitable surface active agents and glidants, for use in a tablet, may be present in amounts from about 0.1% to about 3% by weight, and include polysorbate 80, sodium dodecyl sulfate, talc and silicon dioxide.
[0097] Parenteral Administration
[0098] Compounds of the present invention may be administered directly into the blood stream, muscle, or internal organs. Suitable means for parenteral administration include intravenous, intra- muscular, subcutaneous intraarterial, intraperitoneal, intrathecal, intracranial, and the like. Suitable devices for parenteral administration include injectors (including needle and needle-free injectors) and infusion methods.
[0099] Compositions for parenteral administration may be formulated as immediate or modified release, including delayed or sustained release.
[00100] Most parenteral formulations are aqueous solutions containing excipients, including salts, buffering agents and carbohydrates.
[00101] Parenteral formulations may also be prepared in a dehydrated form (e.g., by lyophilization) or as sterile non-aqueous solutions. These formulations can be used with a suitable vehicle, such as sterile water. Solubility-enhancing agents may also be used in preparation of parenteral solutions.
[00102] Topical Administration
[00103] Compounds of the present invention may be administered topically to the skin or transdermally. Formulations for this topical administration can include lotions, solutions, creams, gels, hydrogels, ointments, foams, implants, patches and the like. Pharmaceutically acceptable carriers for topical administration formulations can include water, alcohol, mineral oil, glycerin, polyethylene glycol and the like. Topical administration can also be performed by electroporation, iontophoresis, phonophoresis and the like.
[00104] Compositions for topical administration may be formulated as immediate or modified release, including delayed or sustained release.
E. Combinations and Combination Therapy
[00105] The compounds of the present invention can be used, alone or in combination with other pharmaceutically active compounds, to treat conditions such as those previously described above. The compound(s) of the present invention and other pharmaceutically active compound(s) can be administered simultaneously (either in the same dosage form or in separate dosage forms) or sequentially. Accordingly, in one embodiment, the present invention comprises methods for treating a condition by administering to the subject a therapeutically-effective amount of one or more compounds of the present invention and one or more additional pharmaceutically active compounds.
[00106] In another embodiment, the present invention comprises a pharmaceutical composition comprising one or more compounds of the present invention, one or more additional pharmaceutically active compounds, and a pharmaceutically acceptable carrier.
[00107] In another embodiment, the one or more additional pharmaceutically active compounds is selected from the group consisting of anti-inflammatory drugs, anti-atherosclerotic drugs, immunosuppressive drugs, immunomodulatory drugs, cytostatic drugs, anti-proliferative agents, angiogenesis inhibitors, kinase inhibitors, cytokine blockers and inhibitors of cell adhesion molecules.
[00108] p38 inhibitor compositions described herein are also optionally used in combination with other therapeutic reagents that are selected for their therapeutic value for the condition to be treated. In general, the compositions described herein and, in embodiments where combinational therapy is employed, other agents do not have to be administered in the same pharmaceutical composition, and, because of different physical and chemical characteristics, are optionally administered by different routes. The initial administration is generally made according to established protocols, and then, based upon the observed effects, the dosage, modes of administration and times of administration subsequently modified. In certain instances, it is appropriate to administer a p38 inhibitor composition as described herein in combination with another therapeutic agent. By way of example only, if one of the side effects experienced by a patient upon receiving a p38 inhibitor composition as described herein is rash, then it is appropriate to administer an anti-histamine agent in combination with the initial therapeutic agent. Or, by way of example only, the therapeutic effectiveness of a p38 inhibitor is enhanced by administration of another therapeutic agent (which also includes a therapeutic regimen) that also has therapeutic benefit In any case, regardless of the disease, disorder or condition being treated, the overall benefit experienced by the patient is either simply additive of the two therapeutic agents or the patient experiences a synergistic benefit.
[00109] Therapeutically effective dosages vary when the drugs are used in treatment combinations. Methods for experimentally determining therapeutically effective dosages of drugs and other agents for use in combination treatment regimens are documented methodologies. Combination treatment further includes periodic treatments that start and stop at various times to assist with the clinical management of the patient. In any case, the multiple therapeutic agents (one of which is a p38 inhibitor as described herein) are administered in any order, or even simultaneously. If simultaneously, the multiple therapeutic agents are optionally provided in a single, unified form, or in multiple forms (by way of example only, either as a single pill or as two separate pills).
[00110] In some embodiments, one of the therapeutic agents is given in multiple doses, or both are given as multiple doses. If not simultaneous, the timing between the multiple doses optionally varies from more than zero weeks to less than twelve weeks.
[00111] In addition, the combination methods, compositions and formulations are not to be limited to the use of only two agents, the use of multiple therapeutic combinations are also envisioned. It is understood that the dosage regimen to treat, prevent, or ameliorate the condition(s) for which relief is sought, is optionally modified in accordance with a variety of factors. These factors include the disorder from which the subject suffers, as well as the age, weight, sex, diet, and medical condition of the subject. Thus, the dosage regimen actually employed varies widely, in some embodiments, and therefore deviates from the dosage regimens set forth herein.
[00112] The pharmaceutical agents which make up the combination therapy disclosed herein are optionally a combined dosage form or in separate dosage forms intended for substantially simultaneous administration. The pharmaceutical agents that make up the combination therapy are optionally also administered sequentially, with either agent being administered by a regimen calling for two-step administration. The two-step administration regimen optionally calls for sequential administration of the active agents or spaced-apart administration of the separate active agents. The time period between the multiple administration steps ranges from, a few minutes to several hours, depending upon the properties of each pharmaceutical agent, such as potency, solubility, bioavailability, plasma half-life and kinetic profile of the pharmaceutical agent. Circadian variation of the target molecule concentration is optionally used to determine the optimal dose interval.
[00113] In another embodiment, a p38 inhibitor is optionally used in combination with procedures that provide additional or synergistic benefit to the patient. A p38 inhibitor and the additional therapy(ies) are optionally administered before, during or after the occurrence of a disease or condition, and the timing of administering the composition containing a p38 inhibitor varies in some embodiments. Thus, for example, a p38 inhibitor is used as a prophylactic and is administered continuously to subjects with a propensity to develop conditions or diseases in order to prevent the occurrence of the disease or condition. A p38 inhibitor and compositions are optionally administered to a subject during or as soon as possible after the onset of the symptoms. While embodiments of the present invention have been shown and described herein, it will be obvious to those skilled in the art that such embodiments are provided by way of example only. Numerous variations, changes, and substitutions will now occur to those skilled in the art without departing from the invention. It should be understood that in some embodiments of the invention various alternatives to the embodiments described herein are employed in practicing the invention.
[00114] A p38 inhibitor can be used in combination with drugs from the following classes: NSAIDs, immunosuppressive drugs, immunomodulatory drugs, cytostatic drugs, angiogenesis inhibitors, biological agents, steroids, vitamin D3 analogs, retinoids, other kinase inhbitors, cytokine blockers, corticosteroids and inhibitors of cell adhesion molecules. Where a subject is suffering from or at risk of suffering from atherosclerosis or a condition that is associated with atherosclerosis, a p38 inhibitor composition described herein is optionally used together with one or more agents or methods for treating atherosclerosis or a condition that is associated with atherosclerosis in any combination. Examples of therapeutic agents/treatments for treating atherosclerosis or a condition that is associated with atherosclerosis include, but are not limited to any of the following: torcetrapib, aspirin, niacin, HMG CoA reductase inhibitors (e.g., atorvastatin, fluvastatin, lovastatin, pravastatin, rosuvastatin and simvastatin), colesevelam, cholestyramine, colestipol, gemfibrozil, probucol and clofibrate.
[00115] Where a subject is suffering from or at risk of suffering from an inflammatory condition, a p38 inhibitor composition described herein is optionally used together with one or more agents or methods for treating an inflammatory condition in any combination. Examples of therapeutic agents/treatments for treating an autoimmune and/or inflammatory condition include, but are not limited to any of the following: corticosteroids, nonsteroidal antiinflammatory drugs (NSAID) (e.g. ibuprofen, naproxen, acetominophen, aspirin, Fenoprofen (Nalfon), Flurbiprofen (Ansaid), Ketoprofen, Oxaprozin (Daypro), Diclofenac sodium (Voltaren), Diclofenac potassium (Cataflam), Etodolac (Lodine), Indomethacin (Indocin), Ketorolac (Toradol), Sulindac (Clinoril), Tolmetin (Tolectin), Meclofenamate (Meclomen), Mefenamic acid (Ponstel), Nabumetone (Relafen), Piroxicam (Feldene), cox-2 inhibitors (e.g. celecoxib (Celebrex))), immunosuppressants (e.g. methotrexate (Rheumatrex), leflunomide (Arava), azathioprine (Imuran), cyclosporine (Neoral, Sandimmune), tacrolimus and cyclophosphamide (Cytoxan), CD20 blockers (Rituximab), Tumor Necrosis Factor (TNF) blockers (e.g. etanercept (Enbrel), infliximab (Remicade) and adalimumab (Humira)), Abatacept (CTLA4-Ig) and interleukin- 1 receptor antagonists (e.g. Anakinra (Kineret), interleukin 6 inhibitors (e.g. Actemra), interleukin 17 inhibitors (e.g. AIN457), Janus kinase inhibitors (e.g. Tasocitinib), syk inhibitors (e.g. R788), chloroquine and its derivatives.
[00116] For use in cancer and neoplastic diseases a p38 inhibitor is optimally used together with one or more of the following classes of drugs: wherein the anti-cancer agent is an EGFR kinase inhibitor, MEK inhibitor, VEGFR inhibitor, anti-VEGFR2 antibody, KDR antibody, AKT inhibitor, PDK-1 inhibitor, PI3K inhibitor, c-kit/Kdr tyrosine kinase inhibitor, Bcr-Abl tyrosine kinase inhibitor, VEGFR2 inhibitor, PDGFR-beta inhibitor, KIT inhibitor, Flt3 tyrosine kinase inhibitor, PDGF receptor family inhibitor, Flt3 tyrosine kinase inhibitor, RET tyrosine kinase receptor family inhibitor, VEGF-3 receptor antagonist, Raf protein kinase family inhibitor, angiogenesis inhibitor, Erb2 inhibitor, mTOR inhibitor, IGF-1R antibody, NFkB inhibitor, proteosome inhibitor, chemotherapy agent, or glucose reduction agent.
F. Kits
[00117] The present invention further comprises kits that are suitable for use in performing the methods of treatment or prevention described above. In one embodiment, the kit contains a first dosage form comprising one or more of the compounds of the present invention and a container for the dosage, in quantities sufficient to carry out the methods of the present invention.
G. Intermediates
[00118] In another embodiment, the invention relates to the novel intermediates useful for preparing the compounds of the present invention. H. General Synthetic Schemes
[00119] The compounds of the present invention can be prepared using the methods illustrated in the general synthetic schemes and experimental procedures detailed below. These general synthetic schemes and experimental procedures are presented for purposes of illustration and are not intended to be limiting. The starting materials used to prepare the compounds of the present invention are commercially available or can be prepared using routine methods known in the art.
[00120] Schemes 1 and 2 illustrate the application of the process to the synthesis of claims of this invention.
Urea Scheme 1
Figure imgf000027_0001
4 1
Figure imgf000027_0002
[00121] The various steps illustrated in Scheme A may be briefly described as follows:
[00122] Step A: Compound 1 may be prepared by coupling a dihalogenated pyrimidine with a substituted aniline, compound 4 as exemplified in schemes 2 and 3, in the presence of a catalyst such as palladium acetate and a ligand such as BINAP and a solvent such as toluene, DMA or THF. Alternatively, the halopyridine may be displaced using the aniline and a base such as cesium carbonate in a solvent such as DMF, DMA or NMP with heating to provide compound 1.
[00123] Step B: Compound 2 may be prepared via Suzuki coupling between a 2- bromopyrimidine (Compound 1) and a boronic acid in the presence of a catalyst such as tetrakis(triphenylphosphine)palladium and a base such as sodium carbonate, sodium t-butoxide, potassium t-butoxide or cesium carbonate and a solvent such as ethanol/water. Alternatively, a boronic acid derivative of pyrimidine may be coupled with a substituted aryl halide under conditions listed above. [00124] Step C: Compound 3 may be prepared via reaction with an activated carbonyl such as phosgene or triphosgene and subsequenently treating the carbonyl with ammonium hydroxide in a solvent such as toluene or dimethoxyethane.
Urea Scheme 2
Figure imgf000028_0001
[00125] When X is an activated group, such as a halogen or triflate, a suitable boronic acid or ester may be used to synthesize the desired aryl or alkyl R4 in the presence of a suitable catalyst such as palladium chloride or tetrakis(triphenylphosphine)palladium(0), a base such as sodium carbonate and a solvent such as ethanol. Conversely, X may be a boronic acid or ester and a suitable haloalkyl or haloaryl may be used with conditions listed above to prepare compound 4.
Urea Scheme 3
Figure imgf000028_0002
4
[00126] Step A: When X is a carboxylate group, the Ν,Ο-dimethyamide may be formed via activation with thionyl chloride or 2-chloro-4,6-dimethoxytriazine or a carbodiimide activating agent such as CD I, EDC or DCC in the presence of HOBt or N-hydroxysuccinimide in a solvent such as dichloromethane, DMF or THF.
[00127] Step B: The amide may be treated with a Grignard reagent in the presence of a solvent such as diethyl ether or THF to provide the appropriate ketone derivative.
[00128] Step C: The ketone may be reacted with dimethylformamide acetal in DMF to provide the enamine intermediate.
[00129] Step D: The enamine may be reacted with a guanidine, amidine or hydrazine with the appropriate substitution in DMF to provide the desired heteroaryl R4.
EXAMPLES
[00130] The following examples are merely illustrative, and do not limit this disclosure in any way.
Figure imgf000029_0001
Figure imgf000030_0001
Example 7. Biological Assays
[00131] P38 inhibitory potency and P38/MK2 substrate selectivity: The novel, MK2 substrate-selective inhibitory mechanism of compounds is evaluated in enzyme assays comparing inhibitor potency in blocking p38/MK2 versus p38/PRAK induced phosphorylation of an HSP-27 derived peptide substrate. The ability of compounds to inhibit activated phospho- p38a is evaluated using a p38a/MK2 and a p38a/PRAK cascade assay format. The kinase activity of p38a is determined by its ability to phosphorylate GST-MK2 or GST-PRAK. Activation of MK2 or PRAK by p38a is quantitated by measuring the phosphorylation of a fluorescently-labeled, MK2 specific peptide substrate, Hsp27 peptide (FITC- KKKALSRQLSVAA). The phosphorylation of the Hsp27 peptide is quantified using the Caliper LabChip 3000. Kinase reactions are carried out in a 384-well plate (Matrical, MPlOl-1- PP) in 20 mM HEPES pH 7.5, 10 mM MgC12, 0.0005% Tween-20, 0.01% BSA, 1 mM DTT, and 2% DMSO. The inhibitor concentration is varied between 0.02-30,000 nM, while the Hsp27 peptide substrate and MgATP are held constant at 1 μΜ and 10 μΜ, respectively. Activated p38a is added to a final concentration of 20 pM for reactions with nonphosphorylated 1 nM His6-MK2 in the cascade reaction. For the p38a/PRAK cascade, unactivated GST-PRAK is held constant at 1 nM while p38 is added in to a final concentration of 20 pM. Kinase reactions are incubated at room temperature and quenched after 30 minutes by the addition of stop buffer (180 mM HEPES, 30 mM EDTA, and 0.2% Coating Reagent-3). Under these conditions, approximately 20% of the substrate Hsp27 peptide is phosphorylated. Reactions are initiated by the addition of activated p38a except for preincubation experiments, where reactions are initiated by the addition of Hsp27 peptide and MgATP. Preincubation of p38a with inhibitor or p38a with unactivated His6-MK2 or unactivated GST-PRAK and inhibitor are performed at 2X final assay concentrations at room temperature 240 minutes prior to adding ATP and Hsp27 peptide to initiate catalysis. The p38a compound inhibitory potency is quantitated from dose-response IC50 values or Ki values from p38a/MK2 cascade assays while the substrate selectivity is calculated as a ratio of p38a/PRAK:p38a/MK2 IC50 values. Species compounds of Formula I, described hereinabove, evaluated in this assay, are expected to provide a therapeutic benefit in the treatment of p38 kinase mediated diseases, such as autoimmune diseases and lymphoma.
[00132] Cytokine regulation in human monocytes: The p38 pathway has been shown to be critical for the biosynthesis of a number of proinflammatory cytokines including TNFa, IL-Ιβ and IL-6. Evaluation of the potency and efficacy of p38 inhibitors to block cytokine production is carried out using the human U937 cell line. The U937 human pre-monocytic cell line will be obtained from the American Type Culture Collection (Rockville, MD). These cells are differentiated to a monocytic/macrophage phenotype as described by Burnette (Burnette et al, (2009). SD0006: a potent, selective and orally available inhibitor of p38 kinase, Pharmacology 84(l):42-60). Differentiated U937 cells are seeded into 96-well tissue culture plates (200,000 cells/well) in complete media. After 24 hours, the cells are pretreated for 60 minutes in the presence or absence of compound and then stimulated with LPS (0.1 μg/mL) for 4 hours. Culture media are then collected for determination of TNFa, IL-6 or IL-Ιβ levels by ELISA. Cytokine concentrations are extrapolated from recombinant protein standard curves using a four- parameter logistic model and solving for IC50 after iterating to the best least-squares fit. Species compounds of Formula I, described hereinabove, evaluated in this assay, are expected to provide a therapeutic benefit in the treatment of p38 kinase mediated diseases, such as lymphoma or inflammation.
[00133] ILip induced prostaglandin production in Rheumatoid arthritis synovial fibroblasts (RASF): Rheumatoid arthritis synovial fibroblasts (RASF) are derived from the inflamed synovium of a female RA patient who was undergoing total knee replacement. Synovial tissue is teased away from adjacent cartilage and dispersed into single cells with collagenase. Cells are expanded and banked. RASF cells are further cultured as described by Burnette supra. RASF cells are seeded into 96-well tissue culture plates (5xl04 cells/well) in complete growth medium. After 24 hours, the medium is replaced with fresh growth medium containing 1% FBS. Cells are treated with serial concentrations (30,000-0.01 nM) of compound or dimethyl- sulfoxide (DMSO) vehicle control for 1 hour then stimulated with Ing/mL IL-Ιβ (R&D Systems, Minneapolis, MN) for 18-20 hours at 37 °C and conditioned media collected. PGE2 levels the in cultured media are quantitated by ELISA (Cayman Chemical, Ann Arbor, MI). Species compounds of Formula I, described hereinabove, evaluated in this assay, are expected to provide a therapeutic benefit in the treatment of p38 kinase mediated diseases, such as lymphoma or rheumatoid arthritis.
[00134] Substrate selectivity in HUVEC cells: When a compound is identified from the biochemical characterization step with selective inhibition of p38/MK2, it is next placed into a cell-based assay to verify enzyme to cell translatability. These assays utilize human umbilical vein endothelial cells (HUVEC) to demonstrate inhibition of Hsp27 phosphorylation (a biomarker of p38/MK2 activation) while sparing production of tissue factor (TF), which is linked to another downstream substrate of p38, MSK. In a 96-well format, adherent HUVEC (at 5 passages or less) are treated for 1 hour with serially-diluted compounds, including a nonselective p38 inhibitor as a reference, or vehicle for controls. For Hsp27 phosphorylation, cells are then stimulated with 500 pg/mL IL-Ιβ for 0.5 hours, media is removed, cells are lysed, and phospho-Hsp27 in the lysate is quantitated by enzyme-linked immunosorbent assay (ELISA)(Life Technologies, Carlsbad, CA). The procedure for TF release is a similar ELISA- based assay (American Diagnostica, Stanford, CT), except that IL-Ιβ stimulation proceeds for 5 hours. The ratio of TF inhibition IC50:HSP27 phosphorylation inhibition IC50 is defined as the substrate selectivity index in these cells. Species compounds of Formula I, described hereinabove, evaluated in this assay, are expected to provide a therapeutic benefit in the treatment of p38 kinase mediated diseases, such as lymphoma and autoinflammatory disease.
[00135] Canine B cell growth regulation: p38 inhibitors have been shown to uniquely inhibit canine B cell proliferation and survival. This selective effect on canine B cells may be exploited in therapeutic treatment for canine B cell lymphoma, a fatal disease that impacts >40,000 companion animals in the United States. Quantitation of impact of p38 inhibitors on B cell growth is a cellular indicator of efficacy in B cell lymphoma. Species compounds of Formula I, described hereinabove, evaluated in this assay, are expected to provide a therapeutic benefit in the treatment of p38 kinase mediated diseases, such as lymphoma. These assays utilize beagle dog spleens obtained with protocols approved by the Saint Louis University Animal Care and Use Committee in collaboration with Seventh Wave Laboratories. Leukocytes are isolated from splenocytes by centrifugation through Histopaque 1077. To evaluate effect on proliferation, leukocytes are then cultured for 48 hours in 96-well plates in the presence of vehicle or test compounds. Cells are stimulated with LPS for TLR4 stimulation, Staphylococcus aureus B cell mitogen, or concanavalin-A T cell mitogen, then proliferation is quantitated with a BRDU incorporation ELISA (Roche, Mannheim, Germany). For apoptosis experiments, leukocytes are plated on 96-well polypropylene U bottom plates and treated with p38 MAPK inhibitors or staurosporine (as a positive control) for up to 24 hours in the absence or presence of actinomycin D or cycloheximide (if needed to increase apoptosis rate). Apoptosis is determined using Caspase-Glo 3/7 luminescent assay (Promega, Madison, WI). In both assays, values generated after incubation with increasing concentrations of the inhibitors are compared to a negative control without inhibitors.
[00136] LPS Induced TNFa Production in rats: Rats are fasted eighteen hours prior to oral dosing, and allowed free access to water throughout the experiment. Each treatment group consists of five animals. Compounds are prepared as a suspension in a vehicle consisting of 0.5% methylcellulose, (Sigma Aldrich, St. Louis, MO), 0.025% Tween 20 (Sigma Aldrich). The compound or vehicle is administered by oral gavage in a volume of 1 mL. Two vehicle groups are used per experiment to control for intra-experiment variability. LPS (E. coli serotype 0111 :B4, Sigma Aldrich) is administered four hours after compound intravenous injection at a dose of 1 mg/kg in 0.5 mL sterile saline (Baxter Healthcare, Deerfield, IL). Blood is collected in serum separator tubes via cardiac puncture ninety minutes after LPS injection, a time point corresponding to maximal TNFa and IL- Ιβ production. After clotting, serum is withdrawn and stored at -20 °C and IL- Ιβ and TNFa levels quantitated by ELISA (Burnette supra). Species compounds of Formula I, described hereinabove, evaluated in this assay, are expected to provide a therapeutic benefit in the treatment of p38 kinase mediated diseases, such as lymphoma or inflammation.
[00137] All mentioned documents are incorporated by reference as if herein written. When introducing elements of the present invention or the exemplary embodiment(s) thereof, the articles "a," "an," "the" and "said" are intended to mean that there are one or more of the elements. The terms "comprising," "including" and "having" are intended to be inclusive and mean that there may be additional elements other than the listed elements. Although this invention has been described with respect to specific embodiments, the details of these embodiments are not to be construed as limitations.

Claims

WHAT IS CLAIMED IS:
1. A compound, or a pharmaceutically acceptable salt of the compound, wherein the compound has the structure of Formula I:
Figure imgf000035_0001
wherein:
V and W are independently selected from the group consisting of CH and N;
R1 is selected from the group consisting of cycloalkyl, aryl, heterocyclyl and heteroaryl; wherein the cycloalkyl and aryl substituents may be optionally substituted with one or more substituents independently selected from the group consisting of halo, cyano, alkyl and alkoxy; and wherein the heterocyclyl and heteroaryl substituents may be optionally substituted with one or more substituents independently selected from the group consisting of cyano and alkyl;
R 3J and R 5J are independently selected from the group consisting of alkyl, halo and hydrogen; and
R4 is selected from the group consisting of cycloalkyl, aryl, heterocyclyl and heteroaryl; wherein the cycloalkyl and aryl substituents may be optionally substituted with one or more substituents independently selected from the group consisting of halo, hydroxy, cyano, alkyl, alkoxy, hydroxyalkyl, alkoxyalkyl and aminoalkyl; and wherein the heterocyclyl and heteroaryl substituents may be optionally substituted with one or more substituents
independently selected from the group consisting of cyano, alkyl, hydroxyalkyl, alkoxyalkyl and aminoalkyl.
2. A compound according to Claim 1, wherein V is CH.
3. A compound according to Claim 2, wherein:
R1 is selected from the group consisting of heterocyclyl and heteroaryl; wherein the heterocyclyl and heteroaryl substituents may be optionally substituted with one or more substituents independently selected from the group consisting of (Ci-C3)-alkyl and cyano;
R 3J and R 5J are independently selected from the group consisting of hydrogen, halo and (Ci-C3)-alkyl; and
R4 is (C5-C6)-heteroaryl; wherein the (Cs-C6)-heteroaryl substituent may be optionally substituted with one or more substituents independently selected from the group consisting of alkyl and hydroxyalkyl.
4. A compound according to Claim 3, wherein:
R1 is (C5-C6)-heteroaryl; wherein the (Cs-C6)-heteroaryl substituent may be optionally substituted with one or more substituents independently selected from (Ci-C3)-alkyl;
R is selected from the group consisting of methyl and fluoro;
R4 is selected from the group consisting of pyridine and pyrimidine; wherein the pyridine and pyrimidine substituents may be optionally substituted with one or more substituents independently selected from the group consisting of alkyl and hydroxyalkyl; and
R5 is selected from the group consisting of hydrogen and fluoro.
5. A compound according to Claim 4, wherein R1 is Cs-heteroaryl, comprising one or more heteroatoms selected from the group consisting of O, N and S; and wherein the Cs-heteroaryl may be optionally substituted with one or more methyl substituents.
6. A compound according to Claim 5, wherein the compound has the structure of Formula II:
Figure imgf000037_0001
wherein:
W is selected from the group consisting of CH and N;
X is selected from the group consisting of O, S and CH;
Y is selected from the group consisting of N and C;
R is selected from the group consisting of methyl and fluoro;
R5 is selected from the group consisting of hydrogen and fluoro;
R10 is selected from (Ci-C3)-alkyl; and
R40 is selected from the group consisting of methyl and hydroxy.
7. A compound according to Claim 6, selected from the group consisting of:
l-(5-(2-(2-hydroxypropan-2-yl)pyrimidin-4-yl)-2-methylphenyl)-l-(2-(2-methyloxazol-4- yl)pyrimidin-4-yl)urea;
l-(5-(2-(2-hydroxypropan-2-yl)pyrimidin-4-yl)-2-methylphenyl)-l-(2-(2-methylthiazol-4- yl)pyrimidin-4-yl)urea; and
l-(5-(2-(2-hydroxypropan-2-yl)pyrimidin-4-yl)-2-methylphenyl)-l-(2-(l-methyl-lH-pyrazol-3- yl)pyrimidin-4-yl)urea.
8. A compound according to Claim 4 wherein R1 is pyridine.
9. A compound according to Claim 8, wherein the compound has the structure of Formula VII:
Figure imgf000038_0001
wherein:
W is selected from the group consisting of CH and N;
R is selected from the group consisting of methyl and fluoro;
R5 is selected from the group consisting of hydrogen and fluoro; and
R40 is selected from the group consisting of methyl and hydroxy.
10. A compound according to Claim 9, where the compound is l-(5-(2-(2-hydroxypropan-2- yl)pyrimidin-4-yl)-2-methylphenyl)-l-(2-(pyridin-2-yl)pyrimidin-4-yl)urea.
11. A compound according to Claim 2, wherein:
R1 is selected from the group consisting of cycloalkyl and aryl; wherein the cycloalkyl and aryl substituents may be optionally substituted with one or more substituents independently selected from the group consisting of (C1-C3)-alkyl and halo;
R 3J and R 5J are independently selected from the group consisting of hydrogen, halo and (Ci-C3)-alk l; and
R4 is (C5-C6)-heteroaryl; wherein the (Cs-C6)-heteroaryl substituent may be optionally substituted with one or more substituents independently selected from the group consisting of alkyl and hydroxyalkyl.
12. A compound according to Claim 11, wherein: R1 is (C5-C6)-aryl; wherein the (C5-C6)-aryl substituents may be optionally substituted with one or more halo substituents;
R is selected from the group consisting of methyl and fluoro;
R4 is selected from the group consisting of pyridine and pyrimidine; wherein the pyridine and pyrimidine substituents may be optionally substituted with one or more substituents independently selected from the group consisting of alkyl and hydroxyalkyl; and
R5 is selected from the group consisting of hydrogen and fluoro.
13. A compound according to Claim 12, wherein R is phenyl optionally substituted with one or more fluoro substituents.
14. A compound accordin to Claim 13, wherein the compound has the structure of Formula IV:
Figure imgf000039_0001
wherein:
W is selected from the group consisting of CH and N;
R is selected from the group consisting of methyl and fluoro;
R5 is selected from the group consisting of hydrogen and fluoro;
R and R , 1111 are independently selected from the group consisting of hydrogen and fluoro; and
selected from the group consisting of methyl and hydroxy.
15. A compound according to Claim 14, selected from the group consisting of:
l-(2-(4-fluorophenyl)pyrimidin-4-yl)-l-(5-(2-(2-hydroxypropan-2-yl)pyrimidin-4-yl)-2- methylphenyl)urea; and
l-(2-(2,4-difluorophenyl)pyrimidin-4-yl)-l-(5-(2-(2-hydroxypropan-2-yl)pyrimidin-4-yl)-2- methylphenyl)urea.
16. A pharmaceutical composition comprising a therapeutically effective amount of a compound of Claim 1 or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
17. The pharmaceutical composition of Claim 16, further comprising a therapeutically effective amount of one or more compounds selected from the group consisting of anti-inflammatory drugs, anti-atherosclerotic drugs, immunosuppressive drugs, immunomodulatory drugs, cytostatic drugs, angiogenesis inhibitors, kinase inhbitors, cytokine blockers and inhibitors of cell adhesion molecules.
18. A method of treating a condition selected from the group consisting of autoimmune disorders, chronic inflammatory disorders, acute inflammatory disorders, auto-inflammatory disorders, atherosclerosis, diabetes, fibrotic diseases, metabolic disorders, cancer, neoplasia, leukemia and lymphoma comprising administering a therapeutically effective amount of a compound of Claim 1 or a pharmaceutically acceptable salt thereof, to a subject in need thereof.
19. The method of Claim 18, wherein the subject is a mammal.
20. The method of Claim 19, wherein the mammalian subject is a canine.
21. The method of Claim 20, wherein the condition is lymphoma.
PCT/US2011/063611 2010-12-06 2011-12-06 Substituted pyrimidine urea compounds WO2012078687A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US42008410P 2010-12-06 2010-12-06
US61/420,084 2010-12-06

Publications (1)

Publication Number Publication Date
WO2012078687A1 true WO2012078687A1 (en) 2012-06-14

Family

ID=46207489

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2011/063611 WO2012078687A1 (en) 2010-12-06 2011-12-06 Substituted pyrimidine urea compounds

Country Status (1)

Country Link
WO (1) WO2012078687A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TWI654189B (en) 2016-06-28 2019-03-21 韓美藥品股份有限公司 Novel heterocyclic derivative compound and use thereof

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050143398A1 (en) * 2003-10-22 2005-06-30 Jagabandhu Das Phenyl-aniline substituted bicyclic compounds useful as kinase inhibitors
US20060178388A1 (en) * 2005-02-04 2006-08-10 Wrobleski Stephen T Phenyl-substituted pyrimidine compounds useful as kinase inhibitors

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050143398A1 (en) * 2003-10-22 2005-06-30 Jagabandhu Das Phenyl-aniline substituted bicyclic compounds useful as kinase inhibitors
US20060178388A1 (en) * 2005-02-04 2006-08-10 Wrobleski Stephen T Phenyl-substituted pyrimidine compounds useful as kinase inhibitors

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
WILSON ET AL.: "Crystal Structure of p38 Mitogen-activated Protein Kinase", JOURNAL BIOLOGICAL CHEMISTRY, vol. 271, 1996, pages 27696 - 27700, XP002339160, DOI: doi:10.1074/jbc.271.44.27696 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TWI654189B (en) 2016-06-28 2019-03-21 韓美藥品股份有限公司 Novel heterocyclic derivative compound and use thereof

Similar Documents

Publication Publication Date Title
US9365546B2 (en) Substituted pyridinone-pyridinyl compounds
JP7146968B2 (en) Methyl/fluoro-pyridinyl-methoxy-substituted pyridinone-pyridinyl compounds and fluoro-pyrimidinyl-methoxy-substituted pyridinone-pyridinyl compounds
US9056110B2 (en) Substituted pyrimidinone-phenyl-pyrimidinyl compounds
US9359300B2 (en) Methyl/difluorophenyl-methoxy substituted pyridinone-pyridinyl compounds, methyl-pyridinyl-methoxy substituted pyridinone-pyridinyl compounds, and methyl-pyrimidinyl-methoxy substituted pyridinone-pyridinyl compounds
US8563558B2 (en) Substituted pyridine urea compounds
WO2012078687A1 (en) Substituted pyrimidine urea compounds
US8507499B2 (en) Substituted indole/indazole-pyrimidinyl compounds

Legal Events

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

Ref document number: 11847824

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 11847824

Country of ref document: EP

Kind code of ref document: A1