WO2011038261A1 - Inhibiteurs hétérocycliques de la kinase - Google Patents

Inhibiteurs hétérocycliques de la kinase Download PDF

Info

Publication number
WO2011038261A1
WO2011038261A1 PCT/US2010/050248 US2010050248W WO2011038261A1 WO 2011038261 A1 WO2011038261 A1 WO 2011038261A1 US 2010050248 W US2010050248 W US 2010050248W WO 2011038261 A1 WO2011038261 A1 WO 2011038261A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
formula
raf
embodiment provides
acid
Prior art date
Application number
PCT/US2010/050248
Other languages
English (en)
Inventor
Jean-Michel Vernier
Patrick O'connor
William Ripke
David Matthews
Anthony Pinkerton
Original Assignee
Selexagen Therapeutics, 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 Selexagen Therapeutics, Inc. filed Critical Selexagen Therapeutics, Inc.
Publication of WO2011038261A1 publication Critical patent/WO2011038261A1/fr

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
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems

Definitions

  • Described herein are compounds, pharmaceutical compositions and methods for the inhibition of RAF, p38 and/or GSK3 kinase mediated signaling. Said compounds, pharmaceutical compositions and methods have utility in the treatment of human disease and disorders.
  • One embodiment provides a compound of Formula (I), or a tautomer, steroisomer, geometric isomer or a pharmaceutically acceptable salt, solvate, or hydrate thereof:
  • Z is N, NH or CH
  • Y is C or N
  • X is N, NH or CH
  • G is selected from: or
  • A is selected from Me, -CH 2 CH 2 OH, -CH 2 CH 2 N CH -CH 2 CH 2 OCH 3 , -OCH 2 CH 2 OH,
  • R 1 , R 2 , R 3 and R 4 are independently selected from hydrogen, F, CI, CN, CF 3 , CH 2 F, CHF 2 , C 2 F 5 , or N0 2 ;
  • R 5 is selected from hydrogen, substituted or unsubstituted C1-C4 alkyl, substituted or unsubstituted C 2 -C 4 alkenyl, substituted or unsubstituted C 2 -Cio acyl, CF 3 , CH 2 F, CHF 2 , C 2 F 5 , or a heterocyclic group.
  • Another embodiment provides a compound of Formula (I) wherein A is selected from - CH CH 2 OH, -CH 2 CH 2 N CH -CH 2 CH 2 OCH 3 , -OCH CH 2 OH, -OCH 2 CH 2 N(CH 3 ) 2 , -
  • Another embodiment provides a compound of Formula (I), wherein Z is NH, Y is C and X is N; or Z is N, Y is C and X is NH.
  • Another embodiment provides a compound of Formula (I), wherein Z is CH, Y is N and X is N.
  • Another embodiment provides a compound of Formula (I), wherein Z is N, Y is N and X is CH.
  • Another embodiment rovides a com oun of Formula I , wherein A is
  • Another embodiment provides a compound of Formula (I), wherein G is Another embodiment rovides a com ound of Formula I), wherein
  • Another embodiment provides a compound of Formula (I), wherein R 1 , R 2 , R 3 and R 4 are independently selected from hydrogen or fluorine.
  • Another embodiment provides a compound of Formula (I), wherein R is
  • Another embodiment provides a compound of Formula (I), wherein R is
  • Another embodiment provides a compound of Formula (I), wherein R is Another embodiment rovides a compound of Formula I), wherein A is
  • Another embodiment provides a compound of Formula (I), wherein G is 0017] Another embodiment provides a compound of Formula (I), wherein A is
  • Another embodiment provides a compound of Formula (I), wherein R 1 , R 2 , R 3 and R 4 independently selected from hydrogen or fluorine.
  • Another embodiment provides a compound of Formula (I), wherein R is
  • Another embodiment provides a compound of Formula (I), wherein R is
  • Another embodiment provides a compound of Formula (I), wherein R is
  • One embodiment provides a compound selected from:
  • example 80 example 81 example 82
  • One embodiment provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compond of Formula (I), or a stereoisomer, hydrate, solvate or pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable excipient.
  • One embodiment provides a method of inhibiting a protein kinase comprising contacting the protein kinase with an inhibitory concentration of a compound of Formula (I), or a stereoisomer, hydrate, solvate or pharmaceutically acceptable salt thereof.
  • Another embodiment provides the method wherein the protein kinase is selected from A-RAF, B-RAF, C-RAF, p38 and GSK3 kinases.
  • Another embodiment provides the method wherein the protein kinase is B- RAF.
  • Another embodiment provides the method wherein the protein kinase is the B-RAF V600E mutant.
  • Another embodiment provides the method wherein the protein kinase is the B- RAF G464V mutant.
  • Another embodiment provides the method wherein the protein kinase is p38 kinase.
  • Another embodiment provides the method wherein the protein kinase is GSK3P kinase.
  • One embodiment provides a method of inhibiting RAF kinase mediated signalling in a cell comprising contacting the cell with an inhibitory concentration of a compound of Formula (I), or a stereoisomer, hydrate, solvate or pharmaceutically acceptable salt thereof. Another embodiment provides the method wherein the cell is by increased activity of the RAS-RAF- MEK-ER pathway compared to a non-transformed cell. Another embodiment provides the method wherein the cell is characterized by a B-RAF gain-of-function mutation. Another embodiment provides the method wherein the cell is characterized by the presence of the B-RAF V600E mutant.
  • One embodiment provides a method of treating a human disease or disorder mediated by RAF kinase signalling comprising administering to a patient a therapeutically effective amount of a composition comprising a compound of Formula (I), or a stereoisomer, hydrate, solvate or pharmaceutically acceptable salt thereof.
  • Another embodiment provides the method wherein the RAF kinase is B-RAF kinase.
  • Another embodiment provides the method wherein the disease or disorder is a proliferative disease.
  • the proliferative disease is selected from melanoma, ovarian cancer, colorectal cancer, thyroid cancer, cholangiocarcinoma, or lung adenocarcinoma.
  • the protein kinase is selected from human A-RAF, B-RAF and C-RAF, or a homo log or an ortho log thereof.
  • RTKs cell membrane associated receptor tyrosine kinases
  • RAS-RAF-MEK-ER pathway that relays growth factor-mediated RTK signals to responder elements in the cytoplasm and/or nuclear compartments (Robinson, M.J. and Cobb, M.H., Curr. Opin. Cell Biol, 9: 180-186, 1997).
  • RAS and RAF members were initially discovered as viral oncogenes that transformed mammalian cells and such eventually lead to the identification of human homologs with similar oncogenic transforming activity (Rapp, U.R., et al., Proc. Natl. Acad. Sci., 80: 4218-4222, 1983: Malumbres, M. and Barbacid, M., Nat. Rev. Cancer, 3: 459- 465, 2003 and references therein).
  • RAF activation is normally regulated by an upstream RAS-GTP bound complex that orchestrates RAF binding to the cell membrane. Subsequent conformational changes induce RAF phosphorylation and kinase activity. The active RAF kinase then phosphorylates and activates MEK, that in-turn phosphorylates and activates ER 1/2 in a signaling cascade that is conserved across a wide variety of animal species (Kolch, W. Biochem. J. 351 : 289-305, 2000 and references therein).
  • A-RAF A-RAF
  • B-RAF B-RAF
  • C-RAF also known as c-RAF-1
  • signaling of RAF to MEK normally requires KSR, a RAF homo log lacking intrinsic kinase activity acting as a scaffold in protein-protein interactions.
  • B-RAF gain-of-function mutations identified to date involve substitution of a valine for a glutamic acid at position 600.
  • V600E this single amino acid substitution leads to constitutive kinase activity approximately 500-fold higher than basal wild-type B-RAF kinase activity (Wan, P.T.C., et al, Cell, 116: 855-867, 2004; Garnett, M.J. and Marais, R. Cancer Cell, 6: 313-319, 2004).
  • B-RAF (V600E) is by itself transforming, and increases tumor cell proliferation, survival and tumor growth in vivo (Davies, H., et al, Nature, 417: 949-954, 2002; Wellbrock, C, et al, Cancer Res., 64: 2338- 2342, 2004). Furthermore, B-RAF (V600E) mutations have been correlated with decreased response rates in cancer patients undergoing chemotherapy (Samowitz, W.S., et al, Cancer Research, 65: 6063-6069, 2005; Houben R., et al, J. Carcinogenesis, 3: 6-18, 2004).
  • siRNA directed to B-RAF (V600E) results in tumor cell growth arrest and/or apoptosis (Karasarides, M., et al, Oncogene, 23: 6292- 6298, 2004; Hingorani, S.R., et al, Cancer Res., 63: 5198-5202, 2003; Hoeflich, K.P., et al,
  • MAPK mitogen-activated protein kinase
  • cytokines pro-inflammatory cytokines
  • MAPKs mitogen-activated protein kinase family involved in the regulation of cell survival and cell stress responses, and are indeed also activated by pro-inflammatory cytokines. Inhibition of p38 MAPK activity may limit growth and/or survival of certain cancer cells expressing high basal activity of these kinases. However, in other cell types p38 MAPKs may be pro-survival signaling molecules whose inhibition then sensitizes cancer cells to apoptosis (Xia et al, Science, 270: 1326-1331, 1995; Potapova et al, J. Biol. Chem., 272: 14041-14044, 1997, and references therein). Taken together these results suggest that p38 MAPK inhibitors appropriately targeted to tumors dependent on signaling through the p38 MAPK pathway may have antitumor effects.
  • GSK3P glycogen synthase kinase 3 beta
  • AKT kinase 3 beta
  • PKC protein kinase 3 beta
  • S6K glycogen synthase kinase 3 beta
  • RSK glycogen synthase kinase 3 beta
  • the role of GSK3P in tumorigenesis depends on cellular context with promoting and inhibitory roles suggested (Mishra, Molecular Cancer, 9: 144, 2010).
  • GSK3P regulates a variety of cellular processes including stability of transcription factors that normally undergo proteosomal degradation including ⁇ -catenin, a target of the Wnt pathway (Ciani, et al, Nat. Rev.
  • GSK3P inhibitors appropriately targeted to tumors dependent on signaling through this kinase may have antitumor effects with examples including pancreatic cancer, colorectal cancer, multiple myeloma, and gastric cancer (Mishra, Molecular Cancer, 9: 144, 2010 and references therein).
  • One embodiment provides a compound having the structure of Formula (I), or a stereoisomer, hydrate, solvate or pharmaceutically acceptable salt thereof, wherein the compound of Formula (I) has the followin structure:
  • Y is C or N
  • R is selected from:
  • A is selected from Me -CH 2 CH 2 OH, -CH 2 CH 2 N(CH 3 ) 2 , -CH 2 CH 2 OCH 3 , -OCH 2 CH 2 OH,
  • R 1 , R 2 , R 3 and R 4 are independently selected from hydrogen, F, CI, CN, CF 3 , CH 2 F, CHF 2 , C 2 F 5 , or N0 2 ;
  • R 5 is selected from hydrogen, substituted or unsubstituted C1-C4 alkyl, substituted or unsubstituted C 2 -C 4 alkenyl, substituted or unsubstituted C 2 -Cio acyl, CF 3 , CH 2 F, CHF 2 ,
  • Another embodiment provides a compound of Formula (I) wherein A is selected from - CH 2 CH 2 OH, -CH 2 CH 2 N(CH 3 ) 2 , -CH 2 CH 2 OCH 3 , -OCH 2 CH 2 OH, -OCH 2 CH 2 N(CH 3 ) 2 , -
  • Another embodiment provides a compound of Formula (I), wherein Z is NH, Y is C and X is N; or Z is N, Y is C and X is NH.
  • Another embodiment provides a compound of Formula (I), wherein Z is CH, Y is N and X is N.
  • Another embodiment provides a compound of Formula (I), wherein Z is N, Y is N and X is CH.
  • Another embodiment provides a compoun of Formula (I), wherein A is
  • Another embodiment provides a compound of Formula (I), wherein G is
  • Another mbodiment pr ides a compound of Formula I), wherein
  • AAnnootthheer embodiment provides a compound of Formula (I), wherein R 1 , R 2 , R 3 and R 4 are independently selected from hydrogen or fluorine.
  • Another embodiment provides a compound of Formula (I), wherein R is [0046] Another embodiment provides a compound of Formula (I), wherein R
  • Another embodiment provides a compound of Formula (I), wherein R is bodiment provides a compound of Formula (I), wherein R is
  • Another embodiment provides a compound of Formula (I), wherein R is
  • Another embodiment provides a compound of Formula (I), wherein A is Another embodiment provides a compound of Formula (I), wherein A is
  • Another embodiment provides a compound of Formula (I), wherein R 1 , R 2 , R 3 and R 4 are independently selected from hydrogen or fluorine.
  • Another embodiment provides a compound of Formula (I), wherein R is
  • Another embodiment provides a compound of Formula (I), wherein R is
  • Another embodiment provides a compound of Formula (I), wherein R is
  • Another embodiment provides a compound of Formula (I), wherein R is Another embodiment provides a compound of Formula (I), wherein R is
  • One embodiment provides a compound selected from:
  • example 80 example 81 example 82
  • compounds of Formula (I) possess one or more stereocenters and each stereocenter exists independently in either the R or S configuration.
  • the compounds presented herein include all diastereomeric, enantiomeric, and epimeric forms as well as the appropriate mixtures thereof.
  • the compounds and methods provided herein include all cis, trans, syn, anti,
  • E
  • Z isomers as well as the appropriate mixtures thereof.
  • compounds of Formula (I) are prepared as their individual stereoisomers by reacting a racemic mixture of the compound with an optically active resolving agent to form a pair of diastereoisomeric compounds/salts, separating the diastereomers and recovering the optically pure enantiomers.
  • resolution of enantiomers is carried out using covalent diastereomeric derivatives of the compounds described herein.
  • diastereomers are seprated by separation/resolution techniques based upon differences in solubility.
  • separation of steroisomers is performed by chromatography or by the forming diastereomeric salts and separation by recrystallization, or chromatography, or any combination thereof.
  • stereoisomers are obtained by stereoselective synthesis.
  • compositions described herein include the use of amorphous forms as well as crystalline forms (also known as polymorphs).
  • compounds described herein are in the form of pharmaceutically acceptable salts.
  • active metabolites of these compounds having the same type of activity are included in the scope of the present disclosure.
  • the compounds described herein can exist in unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like.
  • the solvated forms of the compounds presented herein are also considered to be disclosed herein.
  • compounds described herein are prepared as prodrugs.
  • prodrug refers to an agent that is converted into the parent drug in vivo. Prodrugs are often useful because, in some situations, they may be easier to administer than the parent drug. They may, for instance, be bioavailable by oral administration whereas the parent is not. The prodrug may also have improved solubility in pharmaceutical compositions over the parent drug. In some embodiments, the design of a prodrug increases the effective water solubility.
  • An example, without limitation, of a prodrug is a compound described herein, which is administered as an ester (the "prodrug") to facilitate transmittal across a cell membrane where water solubility is detrimental to mobility but which then is metabolically hydrolyzed to the carboxylic acid, the active entity, once inside the cell where water-solubility is beneficial.
  • a further example of a prodrug might be a short peptide (polyaminoacid) bonded to an acid group where the peptide is metabolized to reveal the active moiety.
  • a prodrug upon in vivo administration, is chemically converted to the biologically, pharmaceutically or therapeutically active form of the compound.
  • a prodrug is enzymatically metabolized by one or more steps or processes to the biologically, pharmaceutically or therapeutically active form of the compound.
  • prodrugs are designed to alter the metabolic stability or the transport characteristics of a drug, to mask side effects or toxicity, to improve the flavor of a drug or to alter other characteristics or properties of a drug.
  • the design prodrugs of the compound is possible, (see, for example, Nogrady (1985) Medicinal Chemistry A Biochemical Approach, Oxford University Press, New York, pages 388-392; Silverman (1992), The Organic Chemistry of Drug Design and Drug Action, Academic Press, Inc., San Diego, pages 352-401, Rooseboom et al., Pharmacological Reviews, 56:53-102, 2004; Aesop Cho, "Recent Advances in Oral Prodrug Discovery", Annual Reports in Medicinal Chemistry, Vol. 41, 395-407, 2006; T. Higuchi and V. Stella, Pro-drugs as Novel Delivery Systems, Vol. 14 of the A.C
  • Prodrug forms of the herein described compounds, wherein the prodrug is metabolized in vivo to produce a compound of Formula (I) as set forth herein are included within the scope of the claims. In some cases, some of the herein-described compounds may be a prodrug for another derivative or active compound.
  • sites on the aromatic ring portion of compounds of Formula (I) are susceptible to various metabolic reactions Therefore incorporation of appropriate substituents on the aromatic ring structures will reduce, minimize or eliminate this metabolic pathway.
  • the appropriate substituent to decrease or eliminate the susceptibility of the aromatic ring to metabolic reactions is, by way of example only, a halogen, or an alkyl group.
  • the compounds described herein are labeled isotopically (e.g. with a radioisotope) or by another other means, including, but not limited to, the use of chromophores or fluorescent moieties, bioluminescent labels, or chemiluminescent labels.
  • Compounds described herein include isotopically-labeled compounds, which are identical to those recited in the various formulae and structures presented herein, but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature.
  • isotopes that can be incorporated into the present compounds include isotopes of hydrogen, carbon, nitrogen, oxygen, fluorine and chlorine, such as, for example, 2 H, 3 H, 13 C, 14 C, 15 N, 18 0, 17 0, 35 S, 18 F, 36 C1.
  • isotopically-labeled compounds described herein for example those into which radioactive isotopes such as 3 H and 14 C are incorporated, are useful in drug and/or substrate tissue distribution assays.
  • substitution with isotopes such as deuterium affords certain therapeutic advantages resulting from greater metabolic stability, such as, for example, increased in vivo half-life or reduced dosage requirements.
  • the compounds described herein are metabolized upon administration to an organism in need to produce a metabolite that is then used to produce a desired effect, including a desired therapeutic effect.
  • “Pharmaceutically acceptable,” as used herein, refers a material, such as a carrier or diluent, which does not abrogate the biological activity or properties of the compound, and is relatively nontoxic, i.e., the material may be administered to an individual without causing undesirable biological effects or interacting in a deleterious manner with any of the components of the composition in which it is contained.
  • pharmaceutically acceptable salt refers to a formulation of a compound that does not cause significant irritation to an organism to which it is administered and does not abrogate the biological activity and properties of the compound.
  • pharmaceutically acceptable salts are obtained by reacting a compound of Formula (I) with acids.
  • Pharmaceutically acceptable salts are also obtained by reacting a compound of Formula (I) with a base to form a salt.
  • Compounds described herein may be formed as, and/or used as, pharmaceutically acceptable salts.
  • the type of pharmaceutical acceptable salts include, but are not limited to: (1) acid addition salts, formed by reacting the free base form of the compound with a
  • inorganic acid such as, for example, hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, metaphosphoric acid, and the like
  • organic acid such as, for example, acetic acid, propionic acid, hexanoic acid
  • cyclopentanepropionic acid glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, trifluoroacetic acid, tartaric acid, citric acid, benzoic acid,
  • 3-(4-hydroxybenzoyl)benzoic acid cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, 1 ,2-ethanedisulfonic acid, 2-hydroxyethanesulfonic acid, benzenesulfonic acid, toluenesulfonic acid, 2-naphthalenesulfonic acid, 4-methylbicyclo-[2.2.2]oct-2-ene-l- carboxylic acid, glucoheptonic acid, 4,4'-methylenebis-(3-hydroxy-2-ene-l-carboxylic acid), 3- phenylpropionic acid, trimethylacetic acid, tertiary butylacetic acid, lauryl sulfuric acid, gluconic acid, glutamic acid, hydroxynaphthoic acid, salicylic acid, stearic acid, muconic acid, butyric acid, phenylacetic acid, phenylbutyric acid
  • compounds described herein may coordinate with an organic base, such as, but not limited to, ethanolamine, diethanolamine, triethanolamine, tromethamine, N- methylglucamine, dicyclohexylamine, tris(hydroxymethyl)methylamine.
  • compounds described herein may form salts with amino acids such as, but not limited to, arginine, lysine, and the like.
  • Acceptable inorganic bases used to form salts with compounds that include an acidic proton include, but are not limited to, aluminum hydroxide, calcium
  • hydroxide potassium hydroxide, sodium carbonate, sodium hydroxide, and the like.
  • a reference to a pharmaceutically acceptable salt includes the solvent addition forms or crystal forms thereof, particularly solvates or polymorphs.
  • Solvates contain either stoichiometric or non-stoichiometric amounts of a solvent, and may be formed during the process of crystallization with pharmaceutically acceptable solvents such as water, ethanol, and the like. Hydrates are formed when the solvent is water, or alcoholates are formed when the solvent is alcohol. Solvates of compounds described herein can be conveniently prepared or formed during the processes described herein.
  • the compounds provided herein can exist in unsolvated as well as solvated forms. In general, the solvated forms are considered equivalent to the unsolvated forms for the purposes of the compounds and methods provided herein.
  • Compounds described herein may be in various forms, including but not limited to, amorphous forms, milled forms and nano-particulate forms.
  • compounds described herein include crystalline forms, also known as polymorphs.
  • Polymorphs include the different crystal packing arrangements of the same elemental composition of a compound. Polymorphs usually have different X-ray diffraction patterns, melting points, density, hardness, crystal shape, optical properties, stability, and solubility. Various factors such as the recrystallization solvent, rate of crystallization, and storage temperature may cause a single crystal form to dominate.
  • alkyl group refers to an aliphatic hydrocarbon group.
  • the alkyl group may be a saturated alkyl group (which means that it does not contain any carbon-carbon double bonds or carbon-carbon triple bonds) or the the alkyl group may be an unsaturated alkyl group (which means that it contains at least one carbon-carbon double bonds or carbon-carbon triple bond).
  • the alkyl moiety, whether saturated or unsaturated, may be branched, or straight chain.
  • the "alkyl” group may have 1 to 10 carbon atoms (whenever it appears herein, a numerical range such as “1 to 10" refers to each integer in the given range; e.g. , "1 to 10 carbon atoms” means that the alkyl group may consist of 1 carbon atom, 2 carbon atoms, 3 carbon atoms, etc. , up to and including 10 carbon atoms, although the present definition also covers the occurrence of the term "alkyl” where no numerical range is designated).
  • the alkyl group of the compounds described herein may be designated as "Ci-C 6 alkyl" or similar designations.
  • Ci-C 6 alkyl indicates that there are one, two , three, four, five, or six carbon atoms in the alkyl chain.
  • the alkyl is selected from the group consisting of methyl, ethyl, propyl, iso-propyl, n-butyl, iso-butyl, sec-butyl, and t-butyl.
  • Typical alkyl groups include, but are in no way limited to, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, tertiary butyl, pentyl, neopentyl, hexyl, allyl, but-2-enyl, but-3-enyl, cyclopropylmethyl,
  • an alkyl is a Ci-C 6 alkyl.
  • alkylene refers to a divalent alkyl radical. Any of the above mentioned monovalent alkyl groups may be an alkylene by abstraction of a second hydrogen atom from the alkyl. In one aspect, an alkelene is a Ci-Cealkylene. In another apsect, an alkylene is a Ci- C 4 alkylene.
  • Typical alkylene groups include, but are not limited to, -CH 2 -, -CH(CH 3 )-, - C(CH 3 ) 2 -, -CH 2 CH 2 -, -CH 2 CH(CH 3 )-, -CH 2 C(CH 3 ) 2 -, -CH 2 CH 2 CH 2 -, -CH 2 CH 2 CH 2 CH 2 -, and the like.
  • alkoxy refers to a (alkyl)O- group, where alkyl is as defined herein.
  • aromatic refers to a planar ring having a delocalized ⁇ -electron system containing 4n+2 ⁇ electrons, where n is an integer. Aromatic rings can be formed from five, six, seven, eight, nine, ten, or more than ten atoms. Aromatics are optionally substituted.
  • aromatic includes both carbocyclic aryl ("aryl”, e.g., phenyl) and heterocyclic aryl (or “heteroaryl” or “heteroaromatic”) groups (e.g., pyridine).
  • aryl e.g., phenyl
  • heterocyclic aryl or “heteroaryl” or “heteroaromatic” groups
  • pyridine monocyclic or fused-ring polycyclic (i.e., rings which share adjacent pairs of carbon atoms) groups.
  • Carbocyclic or “carbocycle” refers to a ring or ring system where the atoms forming the backbone of the ring are all carbon atoms. The term thus distinguishes carbocyclic from heterocyclic rings in which the ring backbone contains at least one atom which is different from carbon.
  • aryl refers to an aromatic ring wherein each of the atoms forming the ring is a carbon atom.
  • Aryl rings are formed by five, six, seven, eight, nine, or more than nine carbon atoms.
  • Aryl groups are optionally substituted.
  • an aryl is a phenyl or a naphthalenyl.
  • an aryl is a phenyl.
  • an aryl is a C6-Cioaryl.
  • an aryl group can be a monoradical or a diradical (i.e., an arylene group).
  • an arylene is a C 6 -Ci 0 arylene.
  • Examplary arylenes include, but are not limited to, phenyl- 1,2-ene, phenyl-l,3-ene, and phenyl- 1,4-ene.
  • cycloalkyl refers to a monocyclic or polycyclic aliphatic, non-aromatic radical, wherein each of the atoms forming the ring (i.e. skeletal atoms) is a carbon atom. Cycloalkyls may be saturated, or partially unsaturated. Cycloalkyls may be fused with an aromatic ring, and the point of attachment is at a carbon that is not an aromatic ring carbon atom. Cycloalkyl groups include groups having from 3 to 10 ring atoms.
  • cycloalkyl groups are selected from among cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cycloheptyl, and cyclooctyl. Cycloalkyl groups may be substituted or unsubstituted.
  • a cycloalkyl group can be a monoradical or a diradical (i.e., an cycloalkylene group, such as, but not limited to, cyclopropan-l,l-diyl, cyclobutan-1,1- diyl, cyclopentan-l,l-diyl, cyclohexan-l,l-diyl, cyclohexan-l,4-diyl, cycloheptan-l,l-diyl, and the like).
  • a cycloalkyl is a C3-C 6 cycloalkyl.
  • halo or, alternatively, "halogen” or “halide” means fluoro, chloro, bromo or iodo.
  • haloalkyl refers to an alkyl group in which one or more hydrogen atoms are replaced by one or more halide atoms.
  • a haloalkyl is a Ci-C 4 haloalkyl.
  • haloalkylene refers to an alkylene group in which one or more hydrogen atoms are replaced by one or more halide atoms.
  • a haloalkylene is a Ci-
  • a haloalkylene is a Ci-C 4 haloalkylene.
  • fluoroalkyl refers to an alkyl in which one or more hydrogen atoms are replaced by a fluorine atom.
  • a fluoralkyl is a Ci-C 4 fluoroalkyl.
  • fluoroalkylene refers to an alkylene in which one or more hydrogen atoms are replaced by a fluorine atom.
  • a fluoralkylene is a Ci-Cefluoroalkylene.
  • a fluoralkylene is a Ci-C 4 fluoroalkylene.
  • heteroalkyl refers to an alkyl group in which one or more skeletal atoms of the alkyl are selected from an atom other than carbon, e.g., oxygen, nitrogen, sulfur, phosphorus or combinations thereof.
  • a heteroalkyl is a Ci-Ceheteroalkyl.
  • heteroalkylene refers to an alkylene group in which one or more skeletal atoms of the alkyl are selected from an atom other than carbon, e.g., oxygen, nitrogen, sulfur, phosphorus or combinations thereof.
  • a heteroalkylene is a Ci-Ceheteroalkylene.
  • a heteroalkylene is a Ci-C 4 heteroalkylene.
  • Examplary heteroalkylenes include, but are not limited to, -OCH 2 -, -OCH(CH 3 )-, -OC(CH 3 ) 2 -, -OCH 2 CH 2 -, -CH 2 0-, -CH(CH 3 )0-, - C(CH 3 ) 2 0-, -CH 2 CH 2 0-, -CH 2 OCH 2 -, -CH 2 OCH 2 CH 2 -, -CH 2 CH 2 OCH 2 -, -SCH 2 -, -SCH(CH 3 )-, -SC(CH 3 ) 2 -, -SCH 2 CH 2 -, -CH 2 S-, -CH(CH 3 )S-, -C(CH 3 ) 2 S-, -CH 2 CH 2 S-, -CH 2 SCH 2 -, - CH 2 SCH 2 CH 2 -, -CH 2 CH 2 SCH 2 -, -S0 2 CH 2 -, -S0
  • heterocycle refers to heteroaromatic rings (also known as heteroaryls) and heterocycloalkyl rings (also known as heteroalicyclic groups) containing one to four heteroatoms in the ring(s), where each heteroatom in the ring(s) is selected from O, S and N, wherein each heterocyclic group has from 4 to 10 atoms in its ring system, and with the proviso that the any ring does not contain two adjacent O or S atoms.
  • Non-aromatic heterocyclic groups also known as heterocycloalkyls
  • the heterocyclic groups include benzo-fused ring systems.
  • An example of a 3-membered heterocyclic group is aziridinyl.
  • An example of a 4-membered heterocyclic group is azetidinyl.
  • An example of a 5- membered heterocyclic group is thiazolyl.
  • An example of a 6-membered heterocyclic group is pyridyl, and an example of a 10-membered heterocyclic group is quinolinyl.
  • non- aromatic heterocyclic groups are pyrrolidinyl, tetrahydrofuranyl, dihydrofuranyl,
  • aromatic heterocyclic groups are pyridinyl, imidazolyl, pyrimidinyl, pyrazolyl, triazolyl, pyrazinyl, tetrazolyl, furyl, thienyl, isoxazolyl, thiazolyl, oxazolyl, isothiazolyl, pyrrolyl, quinolinyl, isoquinolinyl, indolyl, benzimidazolyl, benzofuranyl, cinnolinyl, indazolyl, indolizinyl, phthalazinyl, pyridazinyl, triazinyl, isoindolyl, pteridinyl, purinyl, oxadiazolyl, thiadiazolyl, furazanyl, benzofurazanyl, benzothiophenyl, benzothiazolyl, benzoxazolyl, quinazolinyl, quinox
  • the foregoing groups may be C-attached or N-attached where such is possible.
  • a group derived from pyrrole may be pyrrol- 1-yl (N- attached) or pyrrol-3-yl (C- attached).
  • a group derived from imidazole may be imidazol-l-yl or imidazol-3-yl (both N-attached) or imidazol-2-yl, imidazol-4-yl or imidazol-5-yl (all C-attached).
  • the heterocyclic groups include benzo-fused ring systems.
  • oxo 0
  • the terms “heteroaryl” or, alternatively, “heteroaromatic” refers to an aryl group that includes one or more ring heteroatoms selected from nitrogen, oxygen and sulfur.
  • heteroaryl groups include the following moieties:
  • a heteroaryl contains 0-3 N atoms. In another aspect, a heteroaryl contains 1-3 N atoms.
  • a heteroaryl contains 0-3 N atoms, 0-1 O atoms, and 0-1 S atoms.
  • a heteroaryl is a monocyclic or bicyclic heteroaryl.
  • heteroaryl is a Ci-Cgheteroaryl.
  • monocyclic heteroaryl is a Ci- Csheteroaryl.
  • monocyclic heteroaryl is a 5-membered or 6-membered heteroaryl.
  • bicyclic heteroaryl is a Ce-Cgheteroaryl.
  • a heteroaryl group can be a monoradical or a diradical (i.e., a heteroarylene group).
  • heteroarylene refers to a divalent heteroaryl radical. Any of the above mentioned monovalent heteroaryl groups may be a heteroarylene by abstraction of a second hydrogen atom from the heteroaryl group.
  • the divalent heteroaryl radical may be attached through two carbon atoms, or through one carbon atom and one heteroatom, or through two heteroatoms.
  • heterocycloalkyl or “heteroalicyclic” group refers to a cycloalkyl group that includes at least one heteroatom selected from nitrogen, oxygen and sulfur.
  • the radicals may be fused with an aryl or heteroaryl.
  • heterocycloalkyl groups also referred to as non-aromatic heterocycles, include:
  • the heterocycloalkyl is selected from oxazolidinonyl, pyrrolidinyl, tetrahydrofuranyl, tetrahydrothienyl, tetrahydropyranyl, tetrahydrothiopyranyl, piperidinyl, morpholinyl, thiomorpholinyl, piperazinyl, and indolinyl.
  • heteroalicyclic also includes all ring forms of the carbohydrates, including but not limited to the
  • a heterocycloalkyl is a C 2 -Cioheterocycloalkyl. In another aspect, a heterocycloalkyl is a C4-Cioheterocycloalkyl. In one aspect, a heterocycloalkyl contains 0-2 N atoms. In another aspect, a heterocycloalkyl contains 0-4 N atoms, 0-2 O atoms or 0-1 S atoms.
  • heterocycloalkylene refers to a divalent heterocycloalkyl radical. Any of the above mentioned monovalent heterocycloalkyl groups may be a heterocycloalkylene by abstraction of a second hydrogen atom from the heterocycloalkyl group.
  • the divalent heterocycloalkyl radical may be attached through two carbon atoms, or through one carbon atom and one heteroatom, or through two heteroatoms.
  • bond refers to a chemical bond between two atoms, or two moieties when the atoms joined by the bond are considered to be part of larger substructure.
  • bond when a group described herein is a bond, the referenced group is absent thereby allowing a bond to be formed between the remaining identified groups.
  • a "cyano" group refers to a -CN group.
  • membered ring includes any cyclic structure.
  • membered is meant to denote the number of skeletal atoms that constitute the ring.
  • cyclohexyl, pyridinyl, pyranyl and thiopyranyl are 6-membered rings and cyclopentyl, pyrrolyl, furanyl, and thienyl are 5-membered rings.
  • moiety refers to a specific segment or functional group of a molecule.
  • carboxylic acid bioisostere refers to a functional group or moiety that exhibits similar physical, biological and/or chemical properties as a carboxylic acid moiety.
  • Examples of carboxylic acid bioisosteres include, but are not limited to,
  • the term "optionally substituted” or “substituted” means that the referenced group may be substituted with one or more additional group(s) individually and independently selected from alkyl, cycloalkyl, aryl, heteroaryl, heteroalicyclic, hydroxy, alkoxy, aryloxy, alkylthio, arylthio, alkylsulfoxide, arylsulfoxide, alkylsulfone, arylsulfone, cyano, halo, nitro, haloalkyl, fluoroalkyl, fluoroalkoxy, and amino, including mono- and di-substituted amino groups, and the protected derivatives thereof.
  • substituents are selected from halogen, -CN, -NH 2 , -OH, -N(CH 3 ) 2 , alkyl, fluoroalkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, alkoxy, aryloxy, alkylthio, arylthio, alkylsulfoxide, arylsulfoxide, alkylsulfone, and arylsulfone.
  • an optional substituent is selected from halogen, -CN, -NH 2 , -OH, -NH(CH 3 ), -N(CH 3 ) 2 , -CH 3 , -CH 2 CH 3 , -CF 3 , -OCH 3 , and -OCF 3 .
  • the compounds presented herein possess one or more stereocenters and each center independently exists in either the R or S configuration.
  • the compounds presented herein include all diastereomeric, enantiomeric, and epimeric forms as well as the appropriate mixtures thereof.
  • Stereoisomers are obtained, if desired, by methods such as, stereoselective synthesis and/or the separation of stereoisomers by chiral chromatographic columns.
  • the methods and formulations described herein include the use of N-oxides (if appropriate), crystalline forms (also known as polymorphs), or pharmaceutically acceptable salts of compounds having the structure of Formula (I), as well as active metabolites of these compounds having the same type of activity.
  • compounds may exist as tautomers. All tautomers are included within the scope of the compounds presented herein.
  • the compounds described herein exist in solvated forms with
  • Schemes 1-6 illustrate the synthesis of imidazole-based kinase inhibitors.
  • Schemes 7-12 illustrate the synthesis of pyrazole-based kinase inhibitors.
  • module means to interact with a target either directly or indirectly so as to alter the activity of the target, including, by way of example only, to enhance the activity of the target, to inhibit the activity of the target, to limit the activity of the target, or to extend the activity of the target.
  • modulator refers to a molecule that interacts with a target either directly or indirectly.
  • the interactions include, but are not limited to, the interactions of an agonist, partial agonist, an inverse agonist and antagonist.
  • a modulator is an antagonist.
  • co-administration or the like, as used herein, are meant to encompass administration of the selected therapeutic agents to a single patient, and are intended to include treatment regimens in which the agents are administered by the same or different route of administration or at the same or different time.
  • an “effective amount” or “therapeutically effective amount,” as used herein, refer to a sufficient amount of an agent or a compound being administered which will relieve to some extent one or more of the symptoms of the disease or condition being treated. The result can be reduction and/or alleviation of the signs, symptoms, or causes of a disease, or any other desired alteration of a biological system.
  • an “effective amount” for therapeutic uses is the amount of the composition comprising a compound as disclosed herein required to provide a clinically significant decrease in disease symptoms.
  • An appropriate "effective" amount in any individual case may be determined using techniques, such as a dose escalation study.
  • the terms “enhance” or “enhancing,” as used herein, means to increase or prolong either in potency or duration a desired effect.
  • the term “enhancing” refers to the ability to increase or prolong, either in potency or duration, the effect of other therapeutic agents on a system.
  • An “enhancing-effective amount,” as used herein, refers to an amount adequate to enhance the effect of another therapeutic agent in a desired system.
  • subject or “patient” encompasses mammals and non-mammals.
  • mammals include, but are not limited to, any member of the Mammalian class: humans, non-human primates such as chimpanzees, and other apes and monkey species; farm animals such as cattle, horses, sheep, goats, swine; domestic animals such as rabbits, dogs, and cats; laboratory animals including rodents, such as rats, mice and guinea pigs, and the like.
  • the mammal is a human.
  • treat include alleviating, abating or ameliorating at least one symptom of a disease disease or condition, preventing additional symptoms, inhibiting the disease or condition, e.g., arresting the development of the disease or condition, relieving the disease or condition, causing regression of the disease or condition, relieving a condition caused by the disease or condition, or stopping the symptoms of the disease or condition either prophylactically and/or therapeutically.
  • Suitable routes of administration include, but are not limited to, oral, intravenous, rectal, aerosol, parenteral, ophthalmic, pulmonary, transmucosal, transdermal, vaginal, otic, nasal, and topical administration.
  • parenteral delivery includes intramuscular, subcutaneous, intravenous, intramedullary injections, as well as intrathecal, direct intraventricular, intraperitoneal, intralymphatic, and intranasal injections.
  • a compound as described herein is administered in a local rather than systemic manner, for example, via injection of the compound directly into an organ, often in a depot preparation or sustained release formulation.
  • long acting formulations are administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection.
  • the drug is delivered in a targeted drug delivery system, for example, in a liposome coated with
  • the liposomes are targeted to and taken up selectively by the organ.
  • the compound as described herein is provided in the form of a rapid release formulation, in the form of an extended release formulation, or in the form of an intermediate release formulation. In yet other embodiments, the compound described herein is administered topically.
  • the compounds described herein are formulated into pharmaceutical compositions.
  • Pharmaceutical compositions are formulated in a conventional manner using one or more pharmaceutically acceptable inactive ingredients that facilitate processing of the active compounds into preparations that can be used pharmaceutically. Proper formulation is dependent upon the route of administration chosen.
  • a summary of pharmaceutical compositions described herein can be found, for example, in Remington: The Science and Practice of Pharmacy, Nineteenth Ed (Easton, Pa.: Mack Publishing Company, 1995); Hoover, John E., Remington's Pharmaceutical Sciences, Mack Publishing Co., Easton, Pennsylvania 1975; Liberman, H.A.
  • compositions that include a compound of
  • Formula (I) and at least one pharmaceutically acceptable inactive ingredient are administered as pharmaceutical compositions in which compounds of Formula (I) are mixed with other active ingredients, as in combination therapy.
  • the pharmaceutical compositions include other medicinal or pharmaceutical agents, carriers, adjuvants, preserving, stabilizing, wetting or emulsifying agents, solution promoters, salts for regulating the osmotic pressure, and/or buffers.
  • the pharmaceutical compositions include other therapeutically valuable substances.
  • a pharmaceutical composition refers to a mixture of a compound of Formula (I) with other chemical components (i.e. pharmaceutically acceptable inactive ingredients), such as carriers, excipients, binders, filling agents, suspending agents, flavoring agents, sweetening agents, disintegrating agents, dispersing agents, surfactants, lubricants, colorants, diluents, solubilizers, moistening agents, plasticizers, stabilizers, penetration enhancers, wetting agents, anti-foaming agents, antioxidants, preservatives, or one or more combination thereof.
  • the pharmaceutical composition facilitates administration of the compound to an organism.
  • therapeutically effective amounts of compounds described herein are administered in a pharmaceutical composition to a mammal having a disease, disorder, or condition to be treated.
  • the mammal is a human.
  • a therapeutically effective amount can vary widely depending on the severity of the disease, the age and relative health of the subject, the potency of the compound used and other factors.
  • the compounds can be used singly or in combination with one or more therapeutic agents as components of mixtures.
  • the pharmaceutical formulations described herein are administered to a subject by appropriate administration routes, including but not limited to, oral, parenteral (e.g., intravenous, subcutaneous, intramuscular), intranasal, buccal, topical, rectal, or transdermal administration routes.
  • the pharmaceutical formulations described herein include, but are not limited to, aqueous liquid dispersions, self-emulsifying dispersions, solid solutions, liposomal dispersions, aerosols, solid dosage forms, powders, immediate release formulations, controlled release formulations, fast melt formulations, tablets, capsules, pills, delayed release formulations, extended release formulations, pulsatile release formulations, multiparticulate formulations, and mixed immediate and controlled release formulations.
  • compositions including a compound of Formula (I) are manufactured in a conventional manner, such as, by way of example only, by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or compression processes.
  • the pharmaceutical compositions will include at least one compound of Formula
  • compositions described herein include the use of N-oxides (if appropriate), crystalline forms, amorphous phases, as well as active metabolites of these compounds having the same type of activity.
  • compounds described herein exist in unsolvated form or in solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like. The solvated forms of the compounds presented herein are also considered to be disclosed herein.
  • compositions described herein which include a compound of Formula (I) are formulated into any suitable dosage form, including but not limited to, aqueous oral dispersions, liquids, gels, syrups, elixirs, slurries, suspensions, solid oral dosage forms, aerosols, controlled release formulations, fast melt formulations, effervescent formulations, lyophilized formulations, tablets, powders, pills, dragees, capsules, delayed release formulations, extended release formulations, pulsatile release formulations, multiparticulate formulations, and mixed immediate release and controlled release formulations.
  • aqueous oral dispersions liquids, gels, syrups, elixirs, slurries, suspensions, solid oral dosage forms, aerosols, controlled release formulations, fast melt formulations, effervescent formulations, lyophilized formulations, tablets, powders, pills, dragees, capsules, delayed release formulations, extended release formulations, pulsatile release formulations, multiparticulate
  • compositions for oral use are obtained by mixing one or more solid excipient with one or more of the compounds described herein, optionally grinding the resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores.
  • Suitable excipients include, for example, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methylcellulose, microcrystalline cellulose, hydroxypropylmethylcellulose, sodium carboxymethylcellulose; or others such as: polyvinylpyrrolidone (PVP or povidone) or calcium phosphate. If desired, disintegrating agents are added, such as the cross-linked croscarmellose sodium,
  • polyvinylpyrrolidone agar, or alginic acid or a salt thereof such as sodium alginate.
  • dyestuffs or pigments are added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.
  • compositions that are administered orally include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol.
  • the push-fit capsules contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers.
  • the active compounds are dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols. In some embodiments, stabilizers are added.
  • All formulations for oral administration are in dosages suitable for such administration.
  • solid oral soage forms are prepared by mixing a compound of
  • Formula (I) with one or more of the following: antioxidants, flavoring agents, and carrier materials such as binders, suspending agents, disintegration agents, filling agents, surfactants, solubilizers, stabilizers, lubricants, wetting agents, and diluents.
  • antioxidants such as binders, suspending agents, disintegration agents, filling agents, surfactants, solubilizers, stabilizers, lubricants, wetting agents, and diluents.
  • the solid dosage forms disclosed herein are in the form of a tablet, (including a suspension tablet, a fast-melt tablet, a bite-disintegration tablet, a rapid- disintegration tablet, an effervescent tablet, or a caplet), a pill, a powder, a capsule, solid dispersion, solid solution, bioerodible dosage form, controlled release formulations, pulsatile release dosage forms, multiparticulate dosage forms, beads, pellets, granules.
  • the pharmaceutical formulation is in the form of a powder.
  • the pharmaceutical formulation is in the form of a tablet.
  • pharmaceutical formulations of the compounds of Formula (I) are in the form of a capsule.
  • solid dosage forms e.g., tablets, effervescent tablets, and capsules
  • solid dosage forms are prepared by mixing particles of a compound of Formula (I) with one or more pharmaceutical excipients to form a bulk blend composition.
  • the bulk blend is readily subdivided into equally effective unit dosage forms, such as tablets, pills, and capsules.
  • the individual unit dosages include film coatings. These formulations are manufactured by conventional formulation techniques.
  • Conventional formulation techniques include, e.g., one or a combination of methods: (1) dry mixing, (2) direct compression, (3) milling, (4) dry or non-aqueous granulation, (5) wet granulation, or (6) fusion.
  • Other methods include, e.g., spray drying, pan coating, melt granulation, granulation, fluidized bed spray drying or coating (e.g., wurster coating), tangential coating, top spraying, tableting, extruding and the like.
  • Suitable carriers for use in the solid dosage forms described herein include, but are not limited to, acacia, gelatin, colloidal silicon dioxide, calcium glycerophosphate, calcium lactate, maltodextrin, glycerine, magnesium silicate, sodium caseinate, soy lecithin, sodium chloride, tricalcium phosphate, dipotassium phosphate, sodium stearoyl lactylate, carrageenan, monoglyceride, diglyceride, pregelatinized starch, hydroxypropylmethylcellulose,
  • hydroxypropylmethylcellulose acetate stearate sucrose, microcrystalline cellulose, lactose, mannitol and the like.
  • Suitable filling agents for use in the solid dosage forms described herein include, but are not limited to, lactose, calcium carbonate, calcium phosphate, dibasic calcium phosphate, calcium sulfate, microcrystalline cellulose, cellulose powder, dextrose, dextrates, dextran, starches, pregelatinized starch, hydroxypropylmethycellulose (HPMC),
  • HPLC HPLC
  • sucrose sucrose
  • xylitol lactitol
  • mannitol sorbitol
  • sodium chloride polyethylene glycol, and the like.
  • Suitable disintegrants for use in the solid dosage forms described herein include, but are not limited to, natural starch such as corn starch or potato starch, a pregelatinized starch, or sodium starch glycolate, a cellulose such as methylcrystalline cellulose, methylcellulose, microcrystalline cellulose, croscarmellose, or a cross-linked cellulose, such as cross-linked sodium carboxymethylcellulose, cross-linked carboxymethylcellulose, or cross-linked croscarmellose, a cross-linked starch such as sodium starch glycolate, a cross-linked polymer such as crospovidone, a cross-linked polyvinylpyrrolidone, alginate such as alginic acid or a salt of alginic acid such as sodium alginate, a gum such as agar, guar, locust bean, Karaya, pectin, or tragacanth, sodium starch glycolate, bentonite, sodium lauryl sulfate, sodium lauryl sulf
  • Binders impart cohesiveness to solid oral dosage form formulations: for powder filled capsule formulation, they aid in plug formation that can be filled into soft or hard shell capsules and for tablet formulation, they ensure the tablet remaining intact after compression and help assure blend uniformity prior to a compression or fill step.
  • Materials suitable for use as binders in the solid dosage forms described herein include, but are not limited to,
  • hydroxypropylmethylcellulose acetate stearate hydroxyethylcellulose, hydroxypropylcellulose, ethylcellulose, and microcrystalline cellulose, microcrystalline dextrose, amylose, magnesium aluminum silicate, polysaccharide acids, bentonites, gelatin, polyvinylpyrrolidone/vinyl acetate copolymer, crospovidone, povidone, starch, pregelatinized starch, tragacanth, dextrin, a sugar, such as sucrose, glucose, dextrose, molasses, mannitol, sorbitol, xylitol, lactose, a natural or synthetic gum such as acacia, tragacanth, ghatti gum, mucilage of isapol husks, starch, polyvinylpyrrolidone, larch arabogalactan, polyethylene glycol, waxes, sodium alginate, and the like.
  • binder levels of 20-70% are used in powder- filled gelatin capsule formulations. Binder usage level in tablet formulations varies whether direct compression, wet granulation, roller compaction, or usage of other excipients such as fillers which itself can act as moderate binder. Binder levels of up to 70% in tablet formulations is common.
  • Suitable lubricants or glidants for use in the solid dosage forms described herein include, but are not limited to, stearic acid, calcium hydroxide, talc, corn starch, sodium stearyl fumerate, alkali-metal and alkaline earth metal salts, such as aluminum, calcium, magnesium, zinc, stearic acid, sodium stearates, magnesium stearate, zinc stearate, waxes, Stearowet ® , boric acid, sodium benzoate, sodium acetate, sodium chloride, leucine, a polyethylene glycol or a methoxypolyethylene glycol such as CarbowaxTM, PEG 4000, PEG 5000, PEG 6000, propylene glycol, sodium oleate, glyceryl behenate, glyceryl palmitostearate, glyceryl benzoate, magnesium or sodium lauryl sulfate, and the like.
  • stearic acid calcium hydroxide, talc
  • Suitable diluents for use in the solid dosage forms described herein include, but are not limited to, sugars (including lactose, sucrose, and dextrose), polysaccharides (including dextrates and maltodextrin), polyols (including mannitol, xylitol, and sorbitol), cyclodextrins and the like.
  • Suitable wetting agents for use in the solid dosage forms described herein include, for example, oleic acid, glyceryl monostearate, sorbitan monooleate, sorbitan monolaurate, triethanolamine oleate, polyoxy ethylene sorbitan monooleate, polyoxy ethylene sorbitan monolaurate, quaternary ammonium compounds (e.g., Polyquat 10 ® ), sodium oleate, sodium lauryl sulfate, magnesium stearate, sodium docusate, triacetin, vitamin E TPGS and the like.
  • quaternary ammonium compounds e.g., Polyquat 10 ®
  • sodium oleate sodium lauryl sulfate
  • magnesium stearate sodium docusate
  • triacetin vitamin E TPGS and the like.
  • Suitable surfactants for use in the solid dosage forms described herein include, for example, sodium lauryl sulfate, sorbitan monooleate, polyoxyethylene sorbitan monooleate, polysorbates, polaxomers, bile salts, glyceryl monostearate, copolymers of ethylene oxide and propylene oxide, e.g., Pluronic ® (BASF), and the like.
  • Suitable suspending agents for use in the solid dosage forms described here include, but are not limited to, polyvinylpyrrolidone, e.g., polyvinylpyrrolidone K12,
  • polyvinylpyrrolidone K17 polyvinylpyrrolidone K25, or polyvinylpyrrolidone K30,
  • polyethylene glycol e.g., the polyethylene glycol can have a molecular weight of about 300 to about 6000, or about 3350 to about 4000, or about 7000 to about 5400, vinyl pyrrolidone/vinyl acetate copolymer (S630), sodium carboxymethylcellulose, methylcellulose, hydroxypropylmethylcellulose, polysorbate-80, hydroxyethylcellulose, sodium alginate, gums, such as, e.g., gum tragacanth and gum acacia, guar gum, xanthans, including xanthan gum, sugars, cellulosics, such as, e.g., sodium carboxymethylcellulose, methylcellulose, sodium
  • Suitable antioxidants for use in the solid dosage forms described herein include, for example, e.g., butylated hydroxytoluene (BHT), sodium ascorbate, and tocopherol.
  • BHT butylated hydroxytoluene
  • sodium ascorbate sodium ascorbate
  • tocopherol sodium ascorbate
  • additives used in the solid dosage forms described herein there is considerable overlap between additives used in the solid dosage forms described herein.
  • the above-listed additives should be taken as merely exemplary, and not limiting, of the types of additives that can be included in solid dosage forms of the pharmaceutical compositions described herein.
  • the amounts of such additives can be readily determined by one skilled in the art, according to the particular properties desired.
  • Compressed tablets are solid dosage forms prepared by compacting the bulk blend of the formulations described above.
  • tablets will include one or more flavoring agents.
  • the tablets will include a film surrounding the final compressed tablet.
  • the film coating can provide a delayed release of the compound of Formula (I) from the formulation.
  • the film coating aids in patient compliance (e.g., Opadry ® coatings or sugar coating). Film coatings including Opadry ® typically range from about 1% to about 3% of the tablet weight.
  • a capsule may be prepared, for example, by placing the bulk blend of the formulation of the compound described above, inside of a capsule.
  • the formulations non-aqueous suspensions and solutions
  • the formulations are placed in a soft gelatin capsule.
  • the formulations are placed in standard gelatin capsules or non-gelatin capsules such as capsules comprising HPMC.
  • the formulation is placed in a sprinkle capsule, wherein the capsule is swallowed whole or the capsule is opened and the contents sprinkled on food prior to eating.
  • the particles of the compound of Formula (I) and one or more excipients are dry blended and compressed into a mass, such as a tablet, having a hardness sufficient to provide a pharmaceutical composition that substantially disintegrates within less than about 30 minutes, less than about 35 minutes, less than about 40 minutes, less than about 45 minutes, less than about 50 minutes, less than about 55 minutes, or less than about 60 minutes, after oral administration, thereby releasing the formulation into the gastrointestinal fluid.
  • a powder including a compound of Formula (I) is formulated to include one or more pharmaceutical excipients and flavors.
  • a powder is prepared, for example, by mixing the the compound of Formula (I) and optional pharmaceutical excipients to form a bulk blend composition. Additional embodiments also include a suspending agent and/or a wetting agent. This bulk blend is uniformly subdivided into unit dosage packaging or multi-dosage packaging units.
  • effervescent powders are also prepared. Effervescent salts have been used to disperse medicines in water for oral administration.
  • the pharmaceutical solid oral dosage forms are formulated to provide a controlled release of the compound of Formula (I).
  • Controlled release refers to the release of the compound of Formula (I) from a dosage form in which it is incorporated according to a desired profile over an extended period of time.
  • Controlled release profiles include, for example, sustained release, prolonged release, pulsatile release, and delayed release profiles.
  • immediate release compositions controlled release compositions allow delivery of an agent to a subject over an extended period of time according to a predetermined profile.
  • Such release rates can provide therapeutically effective levels of agent for an extended period of time and thereby provide a longer period of pharmacologic response while minimizing side effects as compared to conventional rapid release dosage forms.
  • Such longer periods of response provide for many inherent benefits that are not achieved with the corresponding short acting, immediate release preparations.
  • the solid dosage forms described herein are formulated as enteric coated delayed release oral dosage forms, i.e., as an oral dosage form of a pharmaceutical composition as described herein which utilizes an enteric coating to affect release in the small intestine or large intestine.
  • the enteric coated dosage form is a compressed or molded or extruded tablet/mold (coated or uncoated) containing granules, powder, pellets, beads or particles of the active ingredient and/or other composition components, which are themselves coated or uncoated.
  • the enteric coated oral dosage form is in the form of a capsule containing pellets, beads or granules, which include a compound of Formula (I), that are coated or uncoated.
  • Coatings should be applied to a sufficient thickness such that the entire coating does not dissolve in the gastrointestinal fluids at pH below about 5, but does dissolve at pH about 5 and above. Coatings are typically selected from any of the following:
  • Shellac - this coating dissolves in media of pH >7;
  • Acrylic polymers - examples of suitable acrylic polymers include methacrylic acid copolymers and ammonium methacrylate copolymers.
  • the Eudragit series E, L, S, RL, RS and NE are available as solubilized in organic solvent, aqueous dispersion, or dry powders.
  • the Eudragit series RL, NE, and RS are insoluble in the gastrointestinal tract but are permeable and are used primarily for colonic targeting.
  • the Eudragit series E dissolve in the stomach.
  • the Eudragit series L, L-30D and S are insoluble in stomach and dissolve in the intestine; Poly Vinyl Acetate Phthalate (PVAP) - PVAP dissolves in pH >5, and it is much less permeable to water vapor and gastric fluids.
  • PVAP Poly Vinyl Acetate Phthalate
  • the formulations described herein are delivered using a pulsatile dosage form.
  • a pulsatile dosage form is capable of providing one or more immediate release pulses at predetermined time points after a controlled lag time or at specific sites.
  • the pulsatile dosage form includes at least two groups of particles, (i.e. multiparticulate) each containing the formulation described herein.
  • the first group of particles provides a substantially immediate dose of the compound of Formula (I) upon ingestion by a mammal.
  • the first group of particles can be either uncoated or include a coating and/or sealant.
  • the second group of particles comprises coated particles. The coating on the second group of particles provides a delay of from about 2 hours to about 7 hours following ingestion before release of the second dose. Suitable coatings for pharmaceutical compositions are described herein or known in the art.
  • compositions that include particles of a compound of Formula (I) and at least one dispersing agent or suspending agent for oral administration to a subject.
  • the formulations may be a powder and/or granules for suspension, and upon admixture with water, a substantially uniform suspension is obtained.
  • liquid formulation dosage forms for oral administration are in the form of aqueous suspensions selected from the group including, but not limited to,
  • the liquid dosage forms include additives, such as: (a) disintegrating agents; (b) dispersing agents; (c) wetting agents; (d) at least one preservative, (e) viscosity enhancing agents, (f) at least one sweetening agent, and (g) at least one flavoring agent.
  • the aqueous dispersions can further include a crystalline inhibitor.
  • compositions optionally include one or more pH adjusting agents or buffering agents, including acids such as acetic, boric, citric, lactic, phosphoric and hydrochloric acids; bases such as sodium hydroxide, sodium phosphate, sodium borate, sodium citrate, sodium acetate, sodium lactate and tris-hydroxymethylaminomethane; and buffers such as citrate/dextrose, sodium bicarbonate and ammonium chloride.
  • acids such as acetic, boric, citric, lactic, phosphoric and hydrochloric acids
  • bases such as sodium hydroxide, sodium phosphate, sodium borate, sodium citrate, sodium acetate, sodium lactate and tris-hydroxymethylaminomethane
  • buffers such as citrate/dextrose, sodium bicarbonate and ammonium chloride.
  • acids, bases and buffers are included in an amount required to maintain pH of the composition in an acceptable range.
  • compositions optionally include one or more salts in an amount required to bring osmolality of the composition into an acceptable range.
  • salts include those having sodium, potassium or ammonium cations and chloride, citrate, ascorbate, borate, phosphate, bicarbonate, sulfate, thiosulfate or bisulfite anions; suitable salts include sodium chloride, potassium chloride, sodium thiosulfate, sodium bisulfite and ammonium sulfate.
  • compositions optionally include one or more preservatives to inhibit microbial activity.
  • Suitable preservatives include mercury-containing substances such as merfen and thiomersal; stabilized chlorine dioxide; and quaternary ammonium compounds such as benzalkonium chloride, cetyltrimethylammonium bromide and cetylpyridinium chloride.
  • the aqueous suspensions and dispersions described herein remain in a homogenous state, as defined in The USP Pharmacists' Pharmacopeia (2005 edition, chapter 905), for at least 4 hours.
  • an aqueous suspension is re-suspended into a homogenous suspension by physical agitation lasting less than 1 minute.
  • no agitation is necessary to maintain a homogeneous aqueous dispersion.
  • a cross-linked starch such as sodium starch glycolate
  • a cross-linked polymer
  • the dispersing agents suitable for the aqueous suspensions and dispersions described herein include, for example, hydrophilic polymers, electrolytes, Tween ® 60 or 80, PEG, polyvinylpyrrolidone, and the carbohydrate-based dispersing agents such as, for example, hydroxypropylcellulose and hydroxypropyl cellulose ethers, hydroxypropyl methylcellulose and hydroxypropyl methylcellulose ethers, carboxymethylcellulose sodium, methylcellulose, hydroxyethylcellulose, hydroxypropylmethyl-cellulose phthalate,
  • hydroxypropylmethyl-cellulose acetate stearate noncrystalline cellulose
  • magnesium aluminum silicate triethanolamine
  • PVA polyvinyl alcohol
  • PVC polyvinylpyrrolidone/vinyl acetate copolymer
  • the dispersing agent is selected from a group not comprising one of the following agents: hydrophilic polymers; electrolytes; Tween ® 60 or 80; PEG; polyvinylpyrrolidone (PVP); hydroxypropylcellulose and hydroxypropyl cellulose ethers; hydroxypropyl methylcellulose and hydroxypropyl
  • methylcellulose ethers carboxymethylcellulose sodium; methylcellulose; hydroxyethylcellulose; hydroxypropylmethyl-cellulose phthalate; hydroxypropylmethyl-cellulose acetate stearate; noncrystalline cellulose; magnesium aluminum silicate; triethanolamine; polyvinyl alcohol (PVA); 4-(l,l,3,3-tetramethylbutyl)-phenol polymer with ethylene oxide and formaldehyde; poloxamers; or poloxamines.
  • PVA polyvinyl alcohol
  • Wetting agents suitable for the aqueous suspensions and dispersions described herein include, but are not limited to, cetyl alcohol, glycerol monostearate, polyoxyethylene sorbitan fatty acid esters (e.g., the commercially available Tweens ® such as e.g., Tween 20 ® and Tween 80 ® , and polyethylene glycols, oleic acid, glyceryl monostearate, sorbitan monooleate, sorbitan monolaurate, triethanolamine oleate, polyoxyethylene sorbitan monooleate,
  • Tweens ® such as e.g., Tween 20 ® and Tween 80 ®
  • polyethylene glycols e.g., oleic acid, glyceryl monostearate, sorbitan monooleate, sorbitan monolaurate, triethanolamine oleate, polyoxyethylene sorbitan monooleate
  • polyoxyethylene sorbitan monolaurate sodium oleate, sodium lauryl sulfate, sodium docusate, triacetin, vitamin E TPGS, sodium taurocholate, simethicone, phosphotidylcholine and the like
  • Suitable preservatives for the aqueous suspensions or dispersions described herein include, for example, potassium sorbate, parabens (e.g., methylparaben and propylparaben), benzoic acid and its salts, other esters of parahydroxybenzoic acid such as butylparaben, alcohols such as ethyl alcohol or benzyl alcohol, phenolic compounds such as phenol, or quaternary compounds such as benzalkonium chloride.
  • Preservatives, as used herein, are incorporated into the dosage form at a concentration sufficient to inhibit microbial growth.
  • Suitable viscosity enhancing agents for the aqueous suspensions or dispersions described herein include, but are not limited to, methyl cellulose, xanthan gum, carboxymethyl cellulose, hydroxypropyl cellulose, hydroxypropylmethyl cellulose, Plasdon ® S-630, carbomer, polyvinyl alcohol, alginates, acacia, chitosans and combinations thereof.
  • concentration of the viscosity enhancing agent will depend upon the agent selected and the viscosity desired.
  • sweetening agents suitable for the aqueous suspensions or dispersions described herein include, for example, acacia syrup, acesulfame K, alitame, aspartame, chocolate, cinnamon, citrus, cocoa, cyclamate, dextrose, fructose, ginger,
  • glycyrrhetinate glycyrrhiza (licorice) syrup, monoammonium glyrrhizinate (MagnaSweet ® ), maltol, mannitol, menthol, neohesperidine DC, neotame, Prosweet ® Powder, saccharin, sorbitol, stevia, sucralose, sucrose, sodium saccharin, saccharin, aspartame, acesulfame potassium, mannitol, sucralose, tagatose, thaumatin, vanilla, xylitol, or any combination thereof.
  • the liquid formulations also include inert diluents commonly used in the art, such as water or other solvents, solubilizing agents, and emulsifiers.
  • emulsifiers are ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propyleneglycol, 1,3-butyleneglycol, dimethylformamide, sodium lauryl sulfate, sodium doccusate, cholesterol, cholesterol esters, taurocholic acid,
  • oils such as cottonseed oil, groundnut oil, corn germ oil, olive oil, castor oil, and sesame oil, glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols, fatty acid esters of sorbitan, or mixtures of these substances, and the like.
  • Formulations that include a compound of Formula (I) are prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, fluorocarbons, and/or other solubilizing or dispersing agents known in the art. See, for example, Ansel, H. C. et al., Pharmaceutical Dosage Forms and Drug Delivery Systems, Sixth Ed. (1995). Preferably these compositions and formulations are prepared with suitable nontoxic
  • nasal dosage form generally contain large amounts of water in addition to the active ingredient. Minor amounts of other ingredients such as pH adjusters, emulsifiers or dispersing agents, preservatives, surfactants, gelling agents, or buffering and other stabilizing and solubilizing agents are optionally present.
  • the nasal dosage form should be isotonic with nasal secretions.
  • a compound of Formula (I) is formulated for use as an aerosol, a mist or a powder.
  • Pharmaceutical compositions described herein are
  • the dosage unit may be determined by providing a valve to deliver a metered amount.
  • Capsules and cartridges of, such as, by way of example only, gelatin for use in an inhaler or insufflator may be formulated containing a powder mix of the compound described herein and a suitable powder base such as lactose or starch.
  • buccal formulations that include a compound of Formula (I) are administered using a variety of formulations known in the art.
  • formulations include, but are not limited to, U.S. Pat. Nos. 4,229,447, 4,596,795, 4,755,386, and 5,739,136.
  • the buccal dosage forms described herein can further include a bioerodible (hydrolysable) polymeric carrier that also serves to adhere the dosage form to the buccal mucosa.
  • the compositions may take the form of tablets, lozenges, or gels formulated in a conventional manner.
  • transdermal formulations described herein include at least three components: (1) a formulation of a compound of Formula (I); (2) a penetration enhancer; and (3) an aqueous adjuvant.
  • the transdermal formulations include additional components such as, but not limited to, gelling agents, creams and ointment bases, and the like.
  • the transdermal formulation further include a woven or non-woven backing material to enhance absorption and prevent the removal of the transdermal formulation from the skin.
  • the transdermal formulations described herein can maintain a saturated or supersaturated state to promote diffusion into the skin.
  • formulations suitable for transdermal administration of compounds described herein employ transdermal delivery devices and transdermal delivery patches and can be lipophilic emulsions or buffered, aqueous solutions, dissolved and/or dispersed in a polymer or an adhesive.
  • patches are constructed for continuous, pulsatile, or on demand delivery of pharmaceutical agents.
  • transdermal delivery of the compounds described herein can be accomplished by means of iontophoretic patches and the like.
  • transdermal patches provide controlled delivery of the compound of Formula (I).
  • transdermal devices are in the form of a bandage comprising a backing member, a reservoir containing the compound optionally with carriers, optionally a rate controlling barrier to deliver the compound to the skin of the host at a controlled and predetermined rate over a prolonged period of time, and means to secure the device to the skin.
  • a compound of Formula (I) is formulated into a pharmaceutical composition suitable for intramuscular, subcutaneous, or intravenous injection.
  • formulations suitable for intramuscular, subcutaneous, or intravenous injection include physiologically acceptable sterile aqueous or non-aqueous solutions, dispersions, suspensions or emulsions, and sterile powders for reconstitution into sterile injectable solutions or dispersions.
  • aqueous and non-aqueous carriers examples include water, ethanol, polyols (propyleneglycol, polyethylene-glycol, glycerol, cremophor and the like), suitable mixtures thereof, vegetable oils (such as olive oil) and injectable organic esters such as ethyl oleate.
  • suitable aqueous and non-aqueous carriers, diluents, solvents, or vehicles include water, ethanol, polyols (propyleneglycol, polyethylene-glycol, glycerol, cremophor and the like), suitable mixtures thereof, vegetable oils (such as olive oil) and injectable organic esters such as ethyl oleate.
  • vegetable oils such as olive oil
  • injectable organic esters such as ethyl oleate.
  • Proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of
  • Prevention of the growth of microorganisms can be ensured by various antibacterial and antifungal agents, such as parabens, chlorobutanol, phenol, sorbic acid, and the like.
  • various antibacterial and antifungal agents such as parabens, chlorobutanol, phenol, sorbic acid, and the like.
  • isotonic agents such as sugars, sodium chloride, and the like.
  • Prolonged absorption of the injectable pharmaceutical form can be brought about by the use of agents delaying absorption, such as aluminum monostearate and gelatin.
  • compounds described herein are formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hank's solution, Ringer's solution, or physiological saline buffer.
  • physiologically compatible buffers such as Hank's solution, Ringer's solution, or physiological saline buffer.
  • penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art.
  • appropriate formulations include aqueous or nonaqueous solutions, preferably with physiologically compatible buffers or excipients. Such excipients are known.
  • Parenteral injections may involve bolus injection or continuous infusion.
  • Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative.
  • the pharmaceutical composition described herein may be in a form suitable for parenteral injection as a sterile suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • the active ingredient is in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
  • delivery systems for pharmaceutical compounds may be employed, such as, for example, liposomes and emulsions.
  • delivery systems for pharmaceutical compounds may be employed, such as, for example, liposomes and emulsions.
  • compositions provided herein can also include an mucoadhesive polymer, selected from among, for example, carboxymethylcellulose, carbomer (acrylic acid polymer),
  • the compounds described herein may be administered topically and can be formulated into a variety of topically administrable compositions, such as solutions, suspensions, lotions, gels, pastes, medicated sticks, balms, creams or ointments.
  • Such pharmaceutical compounds can contain solubilizers, stabilizers, tonicity enhancing agents, buffers and preservatives.
  • the compounds of Formula (I) are formulated in rectal compositions such as enemas, rectal gels, rectal foams, rectal aerosols, suppositories, jelly suppositories, or retention enemas, containing conventional suppository bases such as cocoa butter or other glycerides, as well as synthetic polymers such as polyvinylpyrrolidone, PEG, and the like.
  • rectal compositions such as enemas, rectal gels, rectal foams, rectal aerosols, suppositories, jelly suppositories, or retention enemas
  • conventional suppository bases such as cocoa butter or other glycerides
  • synthetic polymers such as polyvinylpyrrolidone, PEG, and the like.
  • a low-melting wax such as, but not limited to, a mixture of fatty acid glycerides, optionally in combination with cocoa butter is first melted.
  • One embodiment provides a method of inhibiting a protein kinase comprising contacting the protein kinase with an inhibitory concentration of a compound of Formula (I), or a stereoisomer, hydrate, solvate or pharmaceutically acceptable salt thereof, wherein the compound of Formula (I) has the followin structure:
  • Y is C or N
  • R is selected from:
  • A is selected from -CH 2 CH 2 OH, -CH 2 CH 2 N(CH 3 ) 2 , -CH 2 CH 2 OCH 3 , -OCH 2 CH 2 OH, -
  • R 1 , R 2 , R 3 and R 4 are independently selected from hydrogen, F, CI, CN, CF 3 , CH 2 F, CHF 2 , C 2 F 5 , or N0 2 ;
  • R 5 is selected from hydrogen, substituted or unsubstituted C1-C4 alkyl, substituted or unsubstituted C 2 -C 4 alkenyl, substituted or unsubstituted C 2 -Cio acyl, CF 3 , CH 2 F, CHF 2 ,
  • Another embodiment provides a method of inhibiting a protein kinase wherein the protein kinase is selected from A-RAF, B-RAF, C-RAF, p38 and GSK3 kinases.
  • Another embodiment provides a method of inhibiting a protein kinase wherein the protein kinase is B-RAF.
  • Another embodiment provides a method of inhibiting a protein kinase wherein the protein kinase is the B-RAF V600E mutant.
  • Another embodiment provides a method of inhibiting a protein kinase wherein the protein kinase is the B-RAF G464V mutant.
  • Another embodiment provides a method of inhibiting a protein kinase wherein the protein kinase is characterized by the presence of one or more mutations in addition to the B-RAF V600E mutation. Another embodiment provides the method wherein the protein kinase is p38 kinase. Another embodiment provides the method wherein the protein kinase is GSK3 kinase.
  • One embodiment provides a method of inhibiting RAF kinase mediated signalling in a cell comprising contacting the cell with an inhibitory concentration of a compound of Formula (I), or a stereoisomer, hydrate, solvate or pharmaceutically acceptable salt thereof, wherein the compound of Formula (I) has the followin structure:
  • Z is N, NH or CH
  • Y is C or N
  • X is N, NH or CH
  • R is selected from:
  • G is selected from: , or
  • A is selected from -CH 2 CH 2 OH, -CH 2 CH 2 N(CH 3 ) 2 , -CH 2 CH 2 OCH 3 , -OCH 2 CH 2 OH, -
  • R 1 , R 2 , R 3 and R 4 are independently selected from hydrogen, F, CI, CN, CF 3 , CH 2 F, CHF 2 , C 2 F 5 , or N0 2 ;
  • R 5 is selected from hydrogen, substituted or unsubstituted C1-C4 alkyl, substituted or unsubstituted C 2 -C 4 alkenyl, substituted or unsubstituted C 2 -Cio acyl, CF 3 , CH 2 F, CHF 2 , C 2 F 5 , or a heterocyclic group.
  • Another embodiment provides a method of inhibiting RAF kinase mediated signalling in a cell, wherein the cell is characterized by increased activity of the RAS-RAF- MEK-ER pathway compared to a non-transformed cell.
  • Another embodiment provides a method of inhibiting RAF kinase mediated signalling in a cell, wherein the inhibition RAF kinase mediated signalling occurs by inhibition of B-RAF kianse.
  • Another embodiment provides a method of inhibiting RAF kinase mediated signalling in a cell, wherein the cell is characterized by a B-RAF gain-of-function mutation.
  • Another embodiment provides a method of inhibiting B-RAF-mediated signalling in a cell, wherein the cell is characterized by the presence of the B-RAF V600E mutant.
  • One embodiment provides a method of inhibiting p38 kinase mediated signalling in a cell comprising contacting the cell with an inhibitory concentration of a compound of Formula (I), or a stereoisomer, hydrate, solvate or pharmaceutically acceptable salt thereof, wherein the compound of Formula (I) has the followin structure:
  • Z is N, NH or CH
  • Y is C or N
  • X is N, NH or CH
  • R is selected from:
  • A is selected from -CH 2 CH 2 OH, -CH 2 CH 2 N(CH 3 ) 2 , -CH 2 CH 2 OCH 3 , -OCH 2 CH 2 OH, -
  • R 1 , R 2 , R 3 and R 4 are independently selected from hydrogen, F, CI, CN, CF 3 , CH 2 F, CHF 2 , C 2 F 5 , or N0 2 ;
  • R 5 is selected from hydrogen, substituted or unsubstituted C1-C4 alkyl, substituted or unsubstituted C 2 -C 4 alkenyl, substituted or unsubstituted C 2 -Cio acyl, CF 3 , CH 2 F, CHF 2 , C 2 F 5 , or a heterocyclic group.
  • One embodiment provides a method of inhibiting GSK3 kinase mediated signalling in a cell comprising contacting the cell with an inhibitory concentration of a compound of Formula (I), or a stereoisomer, hydrate, solvate or pharmaceutically acceptable salt thereof, wherein the compound of Formula (I) has the following structure:
  • Z is N, NH or CH
  • Y is C or N
  • X is N, NH or CH
  • R is selected from:
  • A is selected from, -CH 2 CH 2 OH, -CH 2 CH N(CH 3 ) 2 , -CH 2 CH 2 OCH 3 , -OCH 2 CH 2 OH, -
  • R 1 , R 2 , R 3 and R 4 are independently selected from hydrogen, F, CI, CN, CF 3 , CH 2 F, CHF 2 , C 2 F 5 , or N0 2 ;
  • R 5 is selected from hydrogen, substituted or unsubstituted C1-C4 alkyl, substituted or unsubstituted C 2 -C 4 alkenyl, substituted or unsubstituted C 2 -Cio acyl, CF 3 , CH 2 F, CHF 2 , C 2 F 5 , or a heterocyclic group.
  • One embodiment provides a method of treating a human disease or disorder mediated by the RAF kinase signalling pathway comprising administering to a patient a therapeutically effective amount of a composition comprising a compound of Formula (I), or a stereoisomer, hydrate, solvate or pharmaceutically acceptable salt thereof, wherein the compound of Formula (I) has the followin structure:
  • Y is C or N
  • G is selected from: or
  • A is selected from, -CH 2 CH 2 OH, -CH 2 CH 2 N CH -CH 2 CH 2 OCH 3 , -OCH 2 CH 2 OH, -
  • R , R , R and R are independently selected from hydrogen, F, CI, CN, CF 3 , CH 2 F, CHF 2 , C 2 F 5 , or N0 2 ;
  • R 5 is selected from hydrogen, substituted or unsubstituted C1-C4 alkyl, substituted or unsubstituted C 2 -C 4 alkenyl, substituted or unsubstituted C 2 -Cio acyl, CF 3 , CH 2 F, CHF 2 , C 2 F 5 , or a heterocyclic group.
  • Another embodiment provides a method of treating a human disease or disorder mediated by RAF kinase signalling wherein the disease or disorder is a proliferative disease.
  • a further embodiment provides a method of treating proliferative disease wherein the proliferative disease is melanoma, ovarian cancer, colorectal cancer, thyroid cancer, cholangiocarcinoma, or lung adenocarcinoma.
  • One embodiment provides a method of treating a human proliferative disease or disorder selected from the group consisting of: oral cancer, prostate cancer, rectal cancer, non- small cell lung cancer, lip and oral cavity cancer, liver cancer, lung cancer, anal cancer, kidney cancer, vulvar cancer, breast cancer, oropharyngeal cancer, nasal cavity and paranasal sinus cancer, nasopharyngeal cancer, urethra cancer, small intestine cancer, bile duct cancer, bladder cancer, ovarian cancer, laryngeal cancer, hypopharyngeal cancer, gallbladder cancer, colon cancer, colorectal cancer, head and neck cancer, parathyroid cancer, penile cancer, vaginal cancer, thyroid cancer, pancreatic cancer, esophageal cancer, Hodgkin's lymphoma, leukemia- related disorders, mycosis fungoides, and myelodysplastic syndrome.
  • a human proliferative disease or disorder selected from the group consisting of: oral cancer, prostate cancer,
  • One embodiment provides a method of treating cancer wherein the cancer is a carcinoma, a tumor, a neoplasm, a lymphoma, a melanoma, a glioma, a sarcoma, and a blastoma.
  • the carcinoma is selected from the group consisting of: carcinoma, adenocarcinoma, adenoid cystic carcinoma, adenosquamous carcinoma,
  • adrenocortical carcinoma well differentiated carcinoma, squamous cell carcinoma, serous carcinoma, small cell carcinoma, invasive squamous cell carcinoma, large cell carcinoma, islet cell carcinoma, oat cell carcinoma, squamous carcinoma, undifferentiatied carcinoma, verrucous carcinoma, renal cell carcinoma, papillary serous adenocarcinoma, merkel cell carcinoma, hepatocellular carcinoma, soft tissue carcinomas, bronchial gland carcinomas, capillary carcinoma, bartholin gland carcinoma, basal cell carcinoma, carcinosarcoma,
  • papilloma/carcinoma clear cell carcinoma, endometrioid adenocarcinoma, mesothelial, metastatic carcinoma, mucoepidermoid carcinoma, cholangiocarcinoma, actinic keratoses, cystadenoma, and hepatic adenomatosis.
  • the tumor is selected from the group consisting of:
  • astrocytic tumors malignant mesothelial tumors, ovarian germ cell tumor, supratentorial primitive neuroectodermal tumors, Wilm's tumor, pituitary tumors, extragonadal germ cell tumor, gastrinoma, germ cell tumors, gestational trophoblastic tumor, brain tumors, pineal and supratentorial primitive neuroectodermal tumors, pituitary tumor, somatostatin-secreting tumor, endodermal sinus tumor, carcinoids, central cerebral astrocytoma, glucagonoma, hepatic adenoma, insulinoma, medulloepithelioma, plasmacytoma, vipoma, and pheochromocytoma.
  • the neoplasm is selected from the group consisting of: intaepithelial neoplasia, multiple myeloma/plasma cell neoplasm, plasma cell neoplasm, interepithelial squamous cell neoplasia, endometrial hyperplasia, focal nodular hyperplasia, hemangioendothelioma, and malignant thymoma.
  • the lymphoma is selected from the group consisting of: nervous system lymphoma, AIDS-related lymphoma, cutaneous T-cell lymphoma, non-
  • lymphoma Hodgkin's lymphoma, lymphoma, and Waldenstrom's macroglobulinemia.
  • the melanoma is selected from the group consisting of: acral lentiginous melanoma, superficial spreading melanoma, uveal melanoma, lentigo maligna melanomas, melanoma, intraocular melanoma, adenocarcinoma nodular melanoma, and hemangioma.
  • the sarcoma is selected from the group consisting of: adenomas, adenosarcoma, chondosarcoma, endometrial stromal sarcoma, Ewing's sarcoma, Kaposi's sarcoma, leiomyosarcoma, , rhabdomyosarcoma, sarcoma, uterine sarcoma, osteosarcoma, and pseudosarcoma.
  • the glioma is selected from the group consisting of: glioma, brain stem glioma, and hypothalamic and visual pathway glioma.
  • the blastoma is selected from the group consisting of: pulmonary blastoma, pleuropulmonary blastoma, retinoblastoma, neuroblastoma,
  • medulloblastoma medulloblastoma, glioblastoma, and hemangiblastomas.
  • One embodiment provides a method of treating a veterinary disease or disorder mediated by the RAF kinase signalling pathway comprising administering to a patient a therapeutically effective amount of a composition comprising a compound of Formula (I), or a stereoisomer, hydrate, solvate or pharmaceutically acceptable salt thereof, wherein the compound of Formula (I) has the following structure:
  • Z is N, NH or CH
  • Y is C or N
  • X is N, NH or CH;
  • G is selected from: or
  • A is selected from, -CH 2 CH 2 OH, -CH 2 CH 2 N CH -CH 2 CH 2 OCH 3 , -OCH 2 CH 2 OH, -
  • R , R , R and R are independently selected from hydrogen, F, CI, CN, CF 3 , CH 2 F, CHF 2 , C 2 F 5 , or N0 2 ;
  • R 5 is selected from hydrogen, substituted or unsubstituted C1-C4 alkyl, substituted or unsubstituted C 2 -C 4 alkenyl, substituted or unsubstituted C 2 -Cio acyl, CF 3 , CH 2 F, CHF 2 , C 2 F 5 , or a heterocyclic group.
  • One embodiment provides a method of treating a parasitic disease or fungal infection in humans or animals comprising administering to a subject a therapeutically effective amount of a composition comprising a compound of Formula (I), or a stereoisomer, hydrate, solvate or pharmaceutically acceptable salt thereof, wherein the compound of Formula (I) has the following structure:
  • Z is N, NH or CH
  • Y is C or N
  • X is N, NH or CH
  • A is selected from, -CH 2 CH 2 OH, -CH 2 CH 2 N CH -CH 2 CH 2 OCH 3 , -OCH 2 CH 2 OH, -
  • R 1 , R 2 , R 3 and R 4 are independently selected from hydrogen, F, CI, CN, CF 3 , CH 2 F, CHF 2 , C 2 F 5 , or N0 2 ;
  • R 5 is selected from hydrogen, substituted or unsubstituted C 1 -C4 alkyl, substituted or unsubstituted C 2 -C 4 alkenyl, substituted or unsubstituted C2-C10 acyl, CF 3 , CH 2 F, CHF 2 , C 2 F 5 , or a heterocyclic group.
  • Step 1 4-(2-phenyl-l-((2-(trimethylsilyl)ethoxy)methyl)-lH-imidazol-4-
  • Step 2 4-(5-bromo-2-phenyl-l-((2-(trimethylsilyl)ethoxy)methyl)-lH-imidazol-
  • Step 3 methyl 4-(2-phenyl-4-(pyridin-4-yl)- 1 -((2-(trimethylsilyl)ethoxy)methyl)- lH-imidazol-5-yl)benzoate
  • Step 4 (4-(2-phenyl-4-(pyridin-4-yl)- 1 -((2-(trimethylsilyl)ethoxy)methyl)- 1 ⁇ - imidazol-5 -yl)phenyl)methanol
  • Step 5 4-(2-phenyl-4-(pyridin-4-yl)- 1 -((2-(trimethylsilyl)ethoxy)methyl)- 1 H- imidazol-5-yl)benzyl methanesulfonate
  • Step 6 4-(5-(4-(azidomethyl)phenyl)-2-phenyl-l-((2-azidomethyl)phenyl)-2-phenyl-l-((2-azidomethyl)phenyl)-2-phenyl-l-((2-azidomethyl)phenyl)-2-phenyl-l-((2-azidomethyl)phenyl)-2-phenyl-l-((2-azidomethyl)phenyl)-2-phenyl-l-((2-azidomethyl)phenyl)-2-phenyl-l-(2-azidomethyl)phenyl)-2-phenyl-l-(2-azidomethyl)phenyl)-2-phenyl-l-(2-azidomethyl)phenyl)-2-phenyl-l-((2-azidomethyl)phenyl)-2-phenyl-l-(2-azidomethyl)phenyl)-2-phenyl-l
  • Step 7 (4-(2-phenyl-4-(pyridin-4-yl)- 1 -((2-(trimethylsilyl)ethoxy)methyl)- 1 H- imidazol-5-yl)phenyl)methanami
  • Step 8 N-(4-(2-phenyl-4-(pyridin-4-yl)-l-((2-(trimethylsilyl)ethoxy)methyl)-lH- imidazol-5-yl)benzyl)methanesulfonamide
  • Step 9 N-(4-(2-phenyl-4-(pyridin-4-yl)-lH-imidazol-5- yl)benzyl)methanesulfonamide
  • Example 8 Example 9
  • Example 10 Example 11
  • Step 1 4-(5-(4-nitrophenyl)-2-phenyl-l-((2-(trimethylsilyl)ethoxy)methyl)-lH-
  • Step 2 4-(2-phenyl-4-(pyridin-4-yl)- 1 -((2-(trimethylsilyl)ethoxy)methyl)- 1 H- imidazol-5 -yl)aniline
  • Step 3 N-(4-(2-phenyl-4-(pyridin-4-y 1)- 1 -((2-(trimethylsilyl)ethoxy)methyl)- 1 H- imidazol-5-yl)phenyl)methanesulfonamide
  • Step 4 N-(4-(2-phenyl-4-(pyridin-4-yl)-lH-imidazol-5- yl)phenyl)methanesulfonamide
  • Step 1 4-(2-phenyl-4-(pyridin-4-yl)-lH-imidazol-5-yl)aniline:
  • Step 1 methyl 4-(2-phenyl-4-(pyridin-4-yl)- 1 -((2-(trimethylsilyl)ethoxy)methyl)- lH-imidazol-5-yl)benzoate
  • step 2 4-(5-Bromo-2-phenyl-l-((2-(trimethylsilyl)ethoxy)methyl)-lH-imidazol-4-yl)pyridine (step 2, example 1) (1100 mg, 2.56 mmol) was dissolved in DME (15 mL) and 2M aqueous sodium carbonate (5 mL) and degassed with nitrogen for 5 minutes. Then, 4- methoxycarbonylphenylboronic acid (691 mg, 3.84 mmol) and palladium
  • Step 2 4-(2-phenyl-4-(pyridin-4-yl)- 1 -((2-(trimethylsilyl)ethoxy)methyl)- 1 ⁇ - imidazol-5-yl)benzoic acid
  • Step 3 4-(2-phenyl-4-(pyridin-4-yl)-l H-imidazol-5 -yl)benzoic acid
  • Example 1 using 4-(4-bromo-l -phenyl- lH-pyrazol-3-yl)pyridine as starting material: example 34 example 35 example 37 example 38
  • Example 1 using 4-(4-bromo-l-methy-lH-pyrazol-3-yl)pyridine as starting material:
  • example 80 example 81 example 82
  • example 95 example 96 example 97 e following compounds are made according to the following scheme.
  • example 110 example 111 example 112 following compounds are made according to the following scheme.
  • the ability of compounds described herein to inhibit kinase activity of RAF, p38 or GSK3 can be determined from biochemical kinase assays using recombinant RAF, p38 or GSK3 proteins as known in the art.
  • the ability of compounds described herein to selectively inhibit cell growth of cultured cells for example those containing either V600E activated B-RAF or wild-type B-RAF is performed as described below.
  • MTT 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide
  • Cell lines are obtained from the American Type Tissue Culture Collection (Maryland, USA) and cultured in media containing heat-inactivated 10% fetal bovine serum. Cell cultures are also maintained in 10 U/mL penicillin, 100 ⁇ g/mL streptomycin and 2 mM glutamine. At least triplicate
  • MTT 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide
  • Cell lines are obtained from the American Type Tissue Culture Collection (Maryland, USA) and cultured in media containing heat-inactivated 10% fetal bovine serum. Cell cultures are also maintained in 10 U/mL penicillin, 100 ⁇ g/mL streptomycin and 2 mM glutamine. At least triplicate

Landscapes

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

Abstract

L'invention concerne des composés, des compositions pharmaceutiques et des méthodes d'inhibition de signalisation de la kinase induites par les kinases A-RAF, B-RAF,C-RAF, p38, ou GSK3β. Les composés, les compositions pharmaceutiques et les méthodes sont utilisés pour des maladies et des troubles humains.
PCT/US2010/050248 2009-09-24 2010-09-24 Inhibiteurs hétérocycliques de la kinase WO2011038261A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US24560609P 2009-09-24 2009-09-24
US61/245,606 2009-09-24

Publications (1)

Publication Number Publication Date
WO2011038261A1 true WO2011038261A1 (fr) 2011-03-31

Family

ID=43796235

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2010/050248 WO2011038261A1 (fr) 2009-09-24 2010-09-24 Inhibiteurs hétérocycliques de la kinase

Country Status (1)

Country Link
WO (1) WO2011038261A1 (fr)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20130040983A1 (en) * 2010-01-08 2013-02-14 Jean-Michel Vernier Raf kinase inhibitors
GB2517988A (en) * 2013-09-09 2015-03-11 Redx Pharma Ltd Compounds
US11034669B2 (en) 2018-11-30 2021-06-15 Nuvation Bio Inc. Pyrrole and pyrazole compounds and methods of use thereof
US11497738B2 (en) 2016-04-29 2022-11-15 Fgh Biotech, Inc. Di-substituted pyrazole compounds for the treatment of diseases

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5434178A (en) * 1993-11-30 1995-07-18 G.D. Searle & Co. 1,3,5 trisubstituted pyrazole compounds for treatment of inflammation
US5486534A (en) * 1994-07-21 1996-01-23 G. D. Searle & Co. 3,4-substituted pyrazoles for the treatment of inflammation
US5620999A (en) * 1994-07-28 1997-04-15 Weier; Richard M. Benzenesulfonamide subtituted imidazolyl compounds for the treatment of inflammation
US20050049288A1 (en) * 2003-06-26 2005-03-03 Fryszman Olga M. 5-MEMBERED HETEROCYCLE-BASED p38 KINASE INHIBITORS
US20080004270A1 (en) * 2004-07-05 2008-01-03 Astex Therapeutics Limited 3,4-Disubstituted Pyrazoles as Cyclin Dependent Kinases (Cdk) or Aurora Kinase or Glycogen Synthase 3 (Gsk-3) Inhibitors

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5434178A (en) * 1993-11-30 1995-07-18 G.D. Searle & Co. 1,3,5 trisubstituted pyrazole compounds for treatment of inflammation
US5486534A (en) * 1994-07-21 1996-01-23 G. D. Searle & Co. 3,4-substituted pyrazoles for the treatment of inflammation
US5620999A (en) * 1994-07-28 1997-04-15 Weier; Richard M. Benzenesulfonamide subtituted imidazolyl compounds for the treatment of inflammation
US20050049288A1 (en) * 2003-06-26 2005-03-03 Fryszman Olga M. 5-MEMBERED HETEROCYCLE-BASED p38 KINASE INHIBITORS
US20080004270A1 (en) * 2004-07-05 2008-01-03 Astex Therapeutics Limited 3,4-Disubstituted Pyrazoles as Cyclin Dependent Kinases (Cdk) or Aurora Kinase or Glycogen Synthase 3 (Gsk-3) Inhibitors

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
PERRY ET AL.: "p38-alpha MAP Kinase C-Terminal Domain Binding Pocket Characterized by Crystallographic and Computational Analyses.", J MOL BIOL., vol. 391, no. 1, 7 August 2009 (2009-08-07), pages 1 - 11, XP026321946, DOI: doi:10.1016/j.jmb.2009.06.005 *
YOUNG ET AL.: "Pyridinyl Imidazole Inhibitors of p38 Mitogen-activated Protein Kinase Bind in the ATP Site.", J. BIOLOGICAL CHEMISTRY, vol. 272, no. 18, 1997, pages 12116 - 12121, XP055024337, DOI: doi:10.1074/jbc.272.18.12116 *

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20130040983A1 (en) * 2010-01-08 2013-02-14 Jean-Michel Vernier Raf kinase inhibitors
GB2517988A (en) * 2013-09-09 2015-03-11 Redx Pharma Ltd Compounds
US11497738B2 (en) 2016-04-29 2022-11-15 Fgh Biotech, Inc. Di-substituted pyrazole compounds for the treatment of diseases
US11034669B2 (en) 2018-11-30 2021-06-15 Nuvation Bio Inc. Pyrrole and pyrazole compounds and methods of use thereof

Similar Documents

Publication Publication Date Title
WO2011085269A1 (fr) Inhibiteurs de la kinase raf
WO2012125981A2 (fr) Inhibiteurs des kinases raf
RU2528408C2 (ru) Сп0соб получения соединений дигидроинденамида, фармацевтические композии, содержащие данные соединение и их применение в качестве ингибитора протеинкиназы
US20210087167A1 (en) Ask1 inhibitor compounds and uses thereof
TW201825472A (zh) 新穎化合物類
TWI672296B (zh) 單環吡啶衍生物
US20110263541A1 (en) Kinase inhibitors and their use as pharmaceutical agents
CN111315747A (zh) 二氢吡唑酮并嘧啶类化合物及其制备方法和用途
KR20020093848A (ko) GSK3β억제제로서의 2-아미노-3-(알킬)-피리미돈유도체
CN111132984A (zh) 凋亡信号调节激酶1抑制剂的盐及其晶型
CN111484481A (zh) 哒嗪酮类衍生物、其制备方法及其在医药上的用途
WO2021213317A1 (fr) Inhibiteur de hpk1, son procédé de préparation et son utilisation
CN112300227B (zh) 杂环酰胺类化合物及其制备方法和应用
WO2015058661A1 (fr) Inhibiteur de bcr-abl kinase et application correspondante
KR20230012597A (ko) Ampk 활성화제
WO2011038261A1 (fr) Inhibiteurs hétérocycliques de la kinase
BR112016005606B1 (pt) Composto, composição farmacêutica, e usos de um composto
WO2016023330A1 (fr) Dérivé de quinazoline
US10807983B2 (en) Imidazo-fused heterocycles and uses thereof
WO2014172639A1 (fr) Inhibiteurs des kinases raf
US11434249B1 (en) ASK1 inhibitor compounds and uses thereof
TW202128686A (zh) 稠合雜芳基類衍生物、其製備方法及其在醫藥上的應用
WO2023097189A1 (fr) Activateurs de l'ampk
WO2022089400A1 (fr) Composé pour la dégradation ciblée de btk et son utilisation antitumorale
CN115197221A (zh) 二氢吡唑并嘧啶酮类大环衍生物及其用途

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: 10819556

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: 10819556

Country of ref document: EP

Kind code of ref document: A1