WO2007056163A2 - Aminopyrimidines useful as kinase inhibitors - Google Patents

Aminopyrimidines useful as kinase inhibitors Download PDF

Info

Publication number
WO2007056163A2
WO2007056163A2 PCT/US2006/042993 US2006042993W WO2007056163A2 WO 2007056163 A2 WO2007056163 A2 WO 2007056163A2 US 2006042993 W US2006042993 W US 2006042993W WO 2007056163 A2 WO2007056163 A2 WO 2007056163A2
Authority
WO
WIPO (PCT)
Prior art keywords
compound
compounds
pharmaceutically acceptable
alkyl
patient
Prior art date
Application number
PCT/US2006/042993
Other languages
French (fr)
Other versions
WO2007056163A3 (en
Inventor
Hayley Binch
Michael Mortimore
Damien Fraysse
Alistair Rutherford
Original Assignee
Vertex Pharmaceuticals Incorporated
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to CN2006800444148A priority Critical patent/CN101316843B/en
Priority to AU2006311830A priority patent/AU2006311830B2/en
Priority to BRPI0619708-6A priority patent/BRPI0619708A2/en
Priority to MX2008005717A priority patent/MX2008005717A/en
Priority to JP2008539070A priority patent/JP5249770B2/en
Priority to CA002627808A priority patent/CA2627808A1/en
Application filed by Vertex Pharmaceuticals Incorporated filed Critical Vertex Pharmaceuticals Incorporated
Priority to EP06827465.3A priority patent/EP1951715B1/en
Priority to ES06827465T priority patent/ES2435081T3/en
Publication of WO2007056163A2 publication Critical patent/WO2007056163A2/en
Publication of WO2007056163A3 publication Critical patent/WO2007056163A3/en
Priority to IL191044A priority patent/IL191044A0/en
Priority to NO20082517A priority patent/NO20082517L/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4151,2-Diazoles
    • A61K31/41551,2-Diazoles non condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/14Decongestants or antiallergics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/32One oxygen, sulfur or nitrogen atom
    • C07D239/38One sulfur atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings

Definitions

  • the present invention relates to compounds useful as inhibitors of Aurora protein kinases .
  • the invention also relates to pharmaceutically acceptable compositions comprising the compounds of the invention, methods of using the compounds and compositions in the treatment of various disorders, and processes for preparing the compounds.
  • Aurora proteins are a family of three related serine/threonine kinases (termed Aurora-A, -B and -C) that are essential for progression through the mitotic phase of cell cycle. Specifically Aurora-A plays a crucial role in centrosome maturation and segregation, formation of the mitotic spindle and faithful segregation of chromosomes .
  • Aurora-B is a chromosomal passenger protein that plays a central role in regulating the alignment of chromosomes on the meta-phase plate, the spindle assembly checkpoint and for the correct completion of cytokinesis.
  • Aurora kinases are attractive targets due to their association with numerous human cancers and the roles they play in the proliferation of these cancer cells. It would be desirable to have an Aurora kinase inhibitor with favorable drug-like properties, such as stability in human liver microsomes. Accordingly, there is a need for compounds that inhibit Aurora kinases and also exhibit favorable drug-like properties .
  • This invention provides compounds and pharmaceutically acceptable compositions thereof that are useful as inhibitors of Aurora protein kinases. These compounds are represented by formula I :
  • R 2 , R 9 , and p are as defined herein.
  • These compounds and pharmaceutically acceptable compositions thereof are useful for inhibiting kinases in vitro, in vivo, and ex vivo.
  • Such uses include treating or preventing myeloproliferative disorders and proliferative disorders such as melanoma, myeloma, leukemia, lymphoma, neuroblastoma, and cancer.
  • Other uses include the study of kinases in biological and pathological phenomena; the study of intracellular signal transduction pathways mediated by such kinases; and the comparative evaluation of new kinase inhibitors .
  • R 2 is C h alky1 or cyclopropyl
  • R 9 is halo, C ⁇ alkyl, -O- (Ci- 3 alkyl) , -S- (Ci_ 3 alkyl) , -OCF 3 , or
  • R 2 is methyl
  • p is 1.
  • R 9 is substituted in the ortho position.
  • R 9 is CF 3 , halo, Ci_ 3 alkyl, or -S- (Ci_ 3 alkyl) . In some embodiments, R 9 is F, Cl, or CF 3 .
  • a specified number range of atoms includes any integer therein.
  • a group having from 1-4 atoms could have 1, 2, 3, or 4 atoms.
  • compounds of the invention may optionally be substituted with one or more substituents, such as are illustrated generally above, or as exemplified by particular classes, subclasses, and species of the invention. It will be appreciated that the phrase “optionally substituted” is used interchangeably with the phrase “substituted or unsubstituted. " In general, the term “substituted”, whether preceded by the term “optionally” or not, refers to the replacement of hydrogen radicals in a given structure with the radical of a specified substituent.
  • an optionally substituted group may have a substituent at each substitutable position of the group, and when more than one position in any given structure may be substituted with more than one substituent selected from a specified group, the substituent may be either the same or different at every position.
  • Combinations of substituents envisioned by this invention are preferably those that result in the formation of stable or chemically feasible compounds .
  • stable refers to compounds that are not substantially altered when subjected to conditions to allow for their production, detection, and preferably their recovery, purification, and use for one or more of the purposes disclosed herein.
  • a stable compound or chemically feasible compound is one that is not substantially altered when kept at a temperature of 40 0 C or less, in the absence of moisture or other chemically reactive conditions, for at least a week.
  • alkyl as used herein, means an unbranched or branched, straight-chain hydrocarbon that is completely saturated and has a single point of attachment to the rest of the molecule.
  • alkyl groups include, but are not limited to, methyl, ethyl, isopropyl, n-propyl, and sec-butyl .
  • cycloalkyl refers to a monocyclic hydrocarbon that is completely saturated and has a single point of attachment to the rest of the molecule.
  • Suitable cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, and cyclopentyl.
  • haloalkyl means an alkyl substituted with one or more halogen atoms . This includes perfluorinated alkyl groups , such as CF 3 .
  • halogen means F, Cl, Br, or I .
  • protecting group refers to an agent used to temporarily block one or more desired reactive sites in a multifunctional compound.
  • a protecting group has one or more, or preferably all, of the following characteristics: a) reacts selectively in good yield to give a protected substrate that is stable to the reactions occurring at one or more of the other reactive sites; and b) is selectively removable in good yield by reagents that do not attack the regenerated functional group.
  • Exemplary protecting groups are detailed in Greene, T. W., Wuts, P.
  • nitrogen protecting group refers to an agents used to temporarily block one or more desired nitrogen reactive sites in a multifunctional compound.
  • Preferred nitrogen protecting groups also possess the characteristics exemplified above, and certain exemplary nitrogen protecting groups are also detailed in Chapter 7 in Greene, T.W., Wuts, P. G in "Protective Groups in Organic Synthesis", Third Edition, John Wiley & Sons, New York: 1999, the entire contents of which are hereby incorporated by reference.
  • structures depicted herein are also meant to include all isomeric (e.g., enantiomeric, diastereomeric, and geometric (or conformational) ) forms of the structure; for example, the R and S configurations for each asymmetric center, (Z) and (E) double bond isomers, and (Z) and (E) conformational isomers. Therefore, single stereochemical isomers as well as enantiomeric, diastereomeric, and geometric (or conformational) mixtures of the present compounds are within the scope of the invention. [0026] Unless otherwise indicated, all tautomeric forms of the compounds of the invention are within the scope of the invention. As would be understood by a skilled practitioner, a pyrazole group can be represented in a variety of ways . For
  • a structure drawn a also represents other
  • a substituent can freely rotate around any rotatable bonds.
  • a substituent can freely rotate around any rotatable bonds.
  • structures depicted herein are also meant to include compounds that differ only in the presence of one or more isotopically enriched atoms.
  • compounds having the present structures except for the replacement of hydrogen by deuterium or tritium, or the replacement of a carbon by a 13 C- or 14 C- enriched carbon are within the scope of this invention.
  • Such compounds are useful, for example, as analytical tools or probes in biological assays.
  • the compounds of this invention may be prepared in light of the specification using steps generally known to those of ordinary skill in the art. Those compounds may be analyzed by known methods, including but not limited to LCMS (liquid chromatography mass spectrometry) and NMR (nuclear magnetic resonance) . It should be understood that the specific conditions shown below are only examples, and are not meant to limit the scope of the conditions that can be used for making compounds of this invention. Instead, this invention also includes conditions that would be apparent to those skilled in that art in light of this specification for making the compounds of this invention. Unless otherwise indicated, all variables in the following schemes are as defined herein.
  • DIPEA is diisopropylethylamine
  • DMF is dimethylformamide
  • ⁇ -BUOH is ⁇ -butanol
  • t-BuOH is tert-butanol
  • MeOH is methanol
  • EtOAc is ethyl acetate
  • TFA is trifluoroacetic acid
  • DMSO is dimethyl sulfoxide
  • Rt is retention time
  • DCM is dichloromethane
  • MeCN is acetonitrile
  • THF is tetrahydrofuran
  • TBTU 2- (lH-Benzotriazole-1-yl) -1, 1, 3, 3-tetramethyluronium tetrafluoroborate
  • HPLC high performance liquid chromatography LCMS liquid chromatography mass spectrometry 1 H NMR is nuclear magnetic resonance
  • Scheme I above shows a general route for the preparation of compounds of formula 4 (in Scheme I) , wherein the variables are as defined herein.
  • the dichlorinated pyrimidine of formula 1 is combined with HQ-R 1 to form a compound of formula 2.
  • the two compounds are heated in the presence of a suitable solvent (e.g. t-BuOH) for 16 hours.
  • the two compounds are mixed at 0 °C in the presence of acetonitrile and triethylamine for 1 hour.
  • the compound of formula 2 is then heated in the presence of a suitable solvent (e.g. DMF) and a suitable base (e.g.
  • DIPEA/Nal DIPEA/Nal
  • an optionally substituted aminopyrazole to form a compound of formula 3
  • azetidine in the presence of a suitable solvent (e.g. n-BuOH) to form a compound of formula 4.
  • a suitable solvent e.g. n-BuOH
  • Scheme II above shows a general route for the preparation of compounds of formula 6 (in Scheme II) , wherein R 2 and R 5 are as defined herein.
  • the compound of formula 5 is combined with a suitable acid chloride (wherein X" is Cl) in the presence of pyridine to form an intermediate compound that, upon mixing in the presence of sodium methoxide and methanol, forms the compound of formula 6.
  • X" can be OH, in which case a suitable acid coupling reagent is used to couple the acid to the amine.
  • suitable acid coupling reagents include, but are not limited to, EDC, DCI, and HOBT.
  • Suitable solvents for these coupling reactions include, but are not limited to, THF, CH 2 Cl 2 , and dioxane .
  • the activity of the compounds as protein kinase inhibitors may be assayed in vitro, in vivo or in a cell line.
  • In vitro assays include assays that determine inhibition of either the kinase activity or ATPase activity of the activated kinase. Alternate in vitro assays guantitate the ability of the inhibitor to bind to the protein kinase and may be measured either by radiolabelling the inhibitor prior to binding, isolating the inhibitor/kinase complex and determining the amount of radiolabel bound, or by running a competition experiment where new inhibitors are incubated with the kinase bound to known radioligands .
  • Another aspect of the invention relates to inhibiting kinase activity in a biological sample, which method comprises contacting said biological sample with a compound of formula I or a composition comprising said compound.
  • biological sample means an in vitro or an ex vivo sample, including, without limitation, cell cultures or extracts thereof; biopsied material obtained from a mammal or extracts thereof; and blood, saliva, urine, feces, semen, tears, or other body fluids or extracts thereof.
  • Inhibition of kinase activity in a biological sample is useful for a variety of purposes that are known to one of skill in the art. Examples of such purposes include, but are not limited to, blood transfusion, organ-transplantation, biological specimen storage, and biological assays.
  • Inhibition of kinase activity in a biological sample is also useful for the study of kinases in biological and pathological phenomena; the study of intracellular signal transduction pathways mediated by such kinases,- and the comparative evaluation of new kinase inhibitors .
  • the Aurora protein kinase inhibitors or pharmaceutical salts thereof may be formulated into pharmaceutical compositions for administration to animals or humans . These pharmaceutical compositions, which comprise an amount of the Aurora protein inhibitor effective to treat or prevent an Aurora-mediated condition and a pharmaceutically acceptable carrier, are another embodiment of the present invention.
  • Aurora-mediated condition or “Aurora- mediated disease” as used herein means any disease or other deleterious condition in which Aurora (Aurora A, Aurora B, and Aurora C) is known to play a role.
  • Such conditions include, without limitation, cancer, proliferative disorders, and myeloproliferative disorders.
  • myeloproliferative disorders include, but are not limited, to, polycythemia vera, thrombocythemia, myeloid metaplasia with myelofibrosis, chronic myelogenous leukaemia (CML) , chronic myelomonocytic leukemia, hypereosinophilic syndrome, juvenile myelomonocytic leukemia, and systemic mast cell disease.
  • CML chronic myelogenous leukaemia
  • chronic myelomonocytic leukemia chronic myelomonocytic leukemia
  • hypereosinophilic syndrome juvenile myelomonocytic leukemia
  • systemic mast cell disease systemic mast cell disease
  • cancer also includes, but is not limited to, the following cancers: epidermoid Oral : buccal cavity, lip, tongue, mouth, pharynx; Cardiac : sarcoma (angiosarcoma, fibrosarcoma, rhabdomyosarcoma, liposarcoma) , myxoma, rhabdomyoma, fibroma, lipoma and teratoma; Lung: bronchogenic carcinoma (squamous cell or epidermoid, undifferentiated small cell, undifferentiated large cell, adenocarcinoma) , alveolar (bronchiolar) carcinoma, bronchial adenoma, sarcoma, lymphoma, chondromatous hamartoma, mesothelioma; Gastrointestinal : esophagus (squamous cell carcinoma, larynx, adenocarcinoma, lei
  • the term "cancerous cell” as provided herein includes a cell afflicted by any one of the above-identified conditions.
  • the cancer is selected from colorectal, thyroid, or breast cancer.
  • the compounds of this invention are useful for treating cancer, such as colorectal, thyroid, breast, and lung cancer; and myeloproliferative disorders, such as polycythemia vera, thrombocythemia, myeloid metaplasia with myelofibrosis, chronic myelogenous leukemia, chronic myelomonocytic leukemia, hypereosinophilic syndrome, juvenile myelomonocytic leukemia, and systemic mast cell disease.
  • the compounds of this invention are useful for treating hematopoietic disorders, in particular, acute-myelogenous leukemia (AML) , chronic- myelogenous leukemia (CML) , acute-promyelocytic leukemia (APL) , and acute lymphocytic leukemia (ALL) .
  • AML acute-myelogenous leukemia
  • CML chronic- myelogenous leukemia
  • APL acute-promyelocytic leukemia
  • ALL acute lymphocytic leukemia
  • pharmaceutically acceptable derivatives or prodrugs of the compounds of this invention may also be employed in compositions to treat or prevent the above-identified disorders .
  • a "pharmaceutically acceptable derivative or prodrug” means any pharmaceutically acceptable ester, salt of an ester or other derivative of a compound of this invention which, upon administration to a recipient, is capable of providing, either directly or indirectly, a compound of this invention or an inhibitorily active metabolite or residue thereof .
  • Such derivatives or prodrugs include those that increase the bioavailability of the compounds of this invention when such compounds are administered to a patient (e.g., by allowing an orally administered compound to be more readily absorbed into the blood) or which enhance delivery of the parent compound to a biological compartment (e.g., the brain or lymphatic system) relative to the parent species .
  • Examples of pharmaceutically acceptable prodrugs of the compounds of this invention include, without limitation, esters, amino acid esters, phosphate esters, metal salts and sulfonate esters .
  • the compounds of this invention can exist in free form for treatment, or where appropriate, as a pharmaceutically acceptable salt .
  • the term "pharmaceutically acceptable salt” refers to salts of a compound which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio.
  • Pharmaceutically acceptable salts of the compounds of this invention include those derived from suitable inorganic and organic acids and bases. These salts can be prepared in situ during the final isolation and purification of the compounds. Acid addition salts can be prepared by 1) reacting the purified compound in its free-based form with a suitable organic or inorganic acid and 2) isolating the salt thus formed.
  • Suitable acid salts include acetate, adipate, alginate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, citrate, camphorate, camphorsulfonate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptanoate, glycerophosphate, glycolate, hemisulfate, heptanoate, hexanoate, hydrochloride, hydrobromide , hydroiodide, 2- hydroxyethanesulfonate, lactate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oxalate, palmoate, pectinate, persulfate, 3-phenylpropionate, phosphate, picrate
  • Base addition salts can be prepared by 1) reacting the purified compound in its acid form with a suitable organic or inorganic base and 2) isolating the salt thus formed.
  • Salts derived from appropriate bases include alkali metal (e.g., sodium and potassium), alkaline earth metal (e.g., magnesium), ammonium and N + (Ci. 4 alkyl) 4 salts. This invention also envisions the quaternization of any basic nitrogen-containing groups of the compounds disclosed herein. Water or oil-soluble or dispersible products may be obtained by such quaternization.
  • Base addition salts also include alkali or alkaline earth metal salts.
  • Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like.
  • Further pharmaceutically acceptable salts include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, loweralkyl sulfonate and aryl sulfonate.
  • Other acids and bases while not in themselves pharmaceutically acceptable, may be employed in the preparation of salts useful as intermediates in obtaining the compounds of the invention and their pharmaceutically acceptable acid or base addition salts.
  • Pharmaceutically acceptable carriers that may be used in these pharmaceutical compositions include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates , waxes, polyethylene- polyoxypropylene-block polymers, polyethylene glycol and wool fat.
  • ion exchangers alumina, aluminum stearate, lecithin
  • serum proteins such as human serum albumin
  • buffer substances such as phosphates, glycine, sorb
  • compositions of the present invention may be administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir.
  • parenteral as used herein includes subcutaneous, intravenous, intramuscular, intraarticular, intra-synovial, intrasternal, intrathecal, intraperitoneal, intrahepatic, intralesional and intracranial injection or infusion techniques.
  • Sterile injectable forms of the compositions of this invention may be aqueous or oleaginous suspension. These suspensions may be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a nontoxic parenterally-acceptable diluent or solvent, for example as a solution in 1, 3-butanediol .
  • the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • a bland fixed oil may be employed including synthetic mono- or di-glycerides .
  • Fatty acids such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions.
  • These oil solutions or suspensions may also contain a long-chain alcohol diluent or dispersant, such as carboxymethyl cellulose or similar dispersing agents which are commonly used in the formulation of pharmaceutically acceptable dosage forms including emulsions and suspensions.
  • a long-chain alcohol diluent or dispersant such as carboxymethyl cellulose or similar dispersing agents which are commonly used in the formulation of pharmaceutically acceptable dosage forms including emulsions and suspensions.
  • Other commonly used surfactants such as Tweens, Spans and other emulsifying agents or bioavailability enhancers which are commonly used in the manufacture of pharmaceutically acceptable solid, liquid, or other dosage forms may also be used for the purposes of formulation.
  • compositions of this invention may be orally administered in any orally acceptable dosage form including, but not limited to, capsules, tablets, aqueous suspensions or solutions .
  • carriers commonly used may include lactose and corn starch.
  • Lubricating agents such as magnesium stearate, may also be added.
  • useful diluents may include lactose and dried cornstarch.
  • the active ingredient may be combined with emulsifying and suspending agents. If desired, certain sweetening, flavoring or coloring agents may also be added.
  • compositions of this invention may be administered in the form of suppositories for rectal administration.
  • suppositories for rectal administration.
  • a suitable non-irritating excipient which is solid at room temperature but liquid at rectal temperature and therefore will melt in the rectum to release the drug.
  • suitable non-irritating excipient may include cocoa butter, beeswax and polyethylene glycols.
  • compositions of this invention may also be administered topically, especially when the target of treatment includes areas or organs readily accessible by topical application, including diseases of the eye, the skin, or the lower intestinal tract. Suitable topical formulations may be prepared for each of these areas or organs .
  • Topical application for the lower intestinal tract can be effected in a rectal suppository formulation (see above) or in a suitable enema formulation. Topically-transdermal patches may also be used.
  • the pharmaceutical compositions may be formulated in a suitable ointment containing the active component suspended or dissolved in one or more carriers .
  • Carriers for topical administration of the compounds of this invention may include, but are not limited to, mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene, polyoxypropylene compound, emulsifying wax and water.
  • the pharmaceutical compositions may be formulated in a suitable lotion or cream containing the active components suspended or dissolved in one or more pharmaceutically acceptable carriers.
  • Suitable carriers may include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water.
  • the pharmaceutical compositions may be formulated as micronized suspensions in isotonic, pH adjusted sterile saline, or as solutions in isotonic, pH adjusted sterile saline, either with or without a preservative such as benzylalkonium • chloride.
  • the pharmaceutical compositions may be formulated in an ointment such as petrolatum.
  • compositions of this invention may also be administered by nasal aerosol or inhalation.
  • Such compositions may be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons , and/or other conventional solubilizing or dispersing agents.
  • compositions should be formulated so that a dosage of between 0.01 - 100 mg/kg body weight/day of the inhibitor can be administered to a patient receiving these compositions. In another embodiment, the compositions should be formulated so that a dosage of between 0.1 - 100 mg/kg body weight/day of the inhibitor can be administered to a patient receiving these compositions .
  • a specific dosage and treatment regimen for any particular patient will depend upon a variety of factors, including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, rate of excretion, drug combination, and the judgment of the treating physician and the severity of the particular disease being treated.
  • the amount of inhibitor will also depend upon the particular compound in the composition.
  • the invention provides methods for treating or preventing cancer, a proliferative disorder, or a myeloproliferative disorder comprising the step of administering to a patient one of the herein-described compounds or pharmaceutical compositions.
  • patient means an animal, including a human.
  • said method is used to treat or prevent a hematopoietic disorder, such as acute-myelogenous leukemia (AML) , acute-promyelocytic leukemia (APL) , chronic- myelogenous leukemia (CML) , or acute lymphocytic leukemia
  • AML acute-myelogenous leukemia
  • APL acute-promyelocytic leukemia
  • CML chronic- myelogenous leukemia
  • said method is used to treat or prevent myeloproliferative disorders, such as polycythemia vera, thrombocythemia, myeloid metaplasia with myelofibrosis, chronic myelogenous leukaemia (CML) , chronic myelomonocytic leukemia, hypereosinophilic syndrome, juvenile myelomonocytic leukemia, and systemic mast cell disease.
  • myeloproliferative disorders such as polycythemia vera, thrombocythemia, myeloid metaplasia with myelofibrosis, chronic myelogenous leukaemia (CML) , chronic myelomonocytic leukemia, hypereosinophilic syndrome, juvenile myelomonocytic leukemia, and systemic mast cell disease.
  • said method is used to treat or prevent cancer, such as cancers of the breast, colon, prostate, skin, pancreas, brain, genitourinary tract, lymphatic system, stomach, larynx and lung, including lung adenocarcinoma, small cell lung cancer, and non-small cell lung cancer.
  • cancer such as cancers of the breast, colon, prostate, skin, pancreas, brain, genitourinary tract, lymphatic system, stomach, larynx and lung, including lung adenocarcinoma, small cell lung cancer, and non-small cell lung cancer.
  • Another embodiment provides a method of treating or preventing cancer comprising the step of administering to a patient a compound of formula I or a composition comprising said compound .
  • Another aspect of the invention relates to inhibiting kinase activity in a patient, which method comprises administering to the patient a compound of formula I or a composition comprising said compound.
  • said kinase is an Aurora kinase (Aurora A, Aurora B, Aurora C), AbI, Arg, FGFRl, MELK, MLKl, MuSK, Ret, or TrkA.
  • additional drugs may be administered together with the compounds of this invention.
  • these additional drugs are normally administered to treat or prevent the same condition.
  • chemotherapeutic agents or other anti-proliferative agents may be combined with the compounds of this invention to treat proliferative diseases .
  • Another aspect of this invention is directed towards a method of treating cancer in a subject in need thereof, comprising the sequential or co-administration of a compound of this invention or a pharmaceutically acceptable salt thereof, and another therapeutic agent.
  • said additional therapeutic agent is selected from an anticancer agent, an anti-proliferative agent, or a chemotherapeutic agent.
  • said additional therapeutic agent is selected from camptothecin, the MEK inhibitor: U0126, a KSP (kinesin spindle protein) inhibitor, adriamycin, interferons, and platinum derivatives, such as Cisplatin.
  • said additional therapeutic agent is selected from taxanes; inhibitors of bcr-abl (such as Gleevec, dasatinib, and nilotinib) ; inhibitors of EGFR (such as Tarceva and Iressa) ; DNA damaging agents (such as cisplatin, oxaliplatin, carboplatin, topoisomerase inhibitors, and anthracyclines) ; and antimetabolites (such as AraC and 5-FU) .
  • inhibitors of bcr-abl such as Gleevec, dasatinib, and nilotinib
  • inhibitors of EGFR such as Tarceva and Iressa
  • DNA damaging agents such as cisplatin, oxaliplatin, carboplatin, topoisomerase inhibitors, and anthracyclines
  • antimetabolites such as AraC and 5-FU
  • said additional therapeutic agent is selected from camptothecin, doxorubicin, idarubicin, Cisplatin, taxol, taxotere, vincristine, tarceva, the MEK inhibitor, U0126, a KSP inhibitor, vorinostat, Gleevec, dasatinib, and nilotinib.
  • said additional therapeutic agent is selected from Her-2 inhibitors (such as Herceptin) ; HDAC inhibitors (such as vorinostat) , VEGFR inhibitors (such as Avastin) , c-KIT and FLT-3 inhibitors (such as sunitinib) , BRAF inhibitors (such as Bayer's BAY 43-9006) MEK inhibitors (such as Pfizer' s PD0325901) ; and spindle poisons (such as Epothilones and paclitaxel protein-bound particles (such as Abraxane ®) .
  • Her-2 inhibitors such as Herceptin
  • HDAC inhibitors such as vorinostat
  • VEGFR inhibitors such as Avastin
  • c-KIT and FLT-3 inhibitors such as sunitinib
  • BRAF inhibitors such as Bayer's BAY 43-9006
  • MEK inhibitors such as Pfizer' s PD0325901
  • spindle poisons such as Epothilone
  • a compound of the instant invention may also be useful for treating cancer in combination with the following therapeutic agents: abarelix (Plenaxis depot ® ); aldesleukin (Prokine ® ) ; Aldesleukin (Proleukin ® ) ; Alemtuzumabb (Campath ® ) ; alitretinoin (Panretin ® ) ; allopurinol (Zyloprim ® ) ; altretamine (Hexalen ® ) ; amifostine (Ethyol ® ) ; anastrozole (Arimidex ® ) ; arsenic trioxide (Trisenox ® ) ; asparaginase (Elspar ® ) ; azacitidine (Vidaza ® ) ; bevacuzimab (Avastin ® ) ; bexarotene capsules (Targretin ® ) ; bexa
  • paclitaxel protein-bound particles (Neumega ® ) ; oxaliplatin (Eloxatin ® ) ; paclitaxel (Paxene ® ) ; paclitaxel (Taxol ® ) ; paclitaxel protein-bound particles
  • Another embodiment provides a simultaneous, separate or sequential use of a combined preparation.
  • Those additional agents may be administered separately, as part of a multiple dosage regimen, from the kinase inhibitor-containing compound or composition. Alternatively, those agents may be part of a single dosage form, mixed together with the kinase inhibitor in a single composition.
  • Rt(tnin) refers to the HPLC retention time, in minutes, associated with the compound. Unless otherwise indicated, the HPLC method utilized to obtain the reported retention time is as follows: Column: ACE C8 column, 4.6 x 150 mm
  • Mass spec samples were analyzed on a MicroMass Quattro Micro mass spectrometer operated in single MS mode with electrospray ionization. Samples were introduced into the mass spectrometer using chromatography. Mobile phase for all mass spec, analyses consisted of 1OmM pH 7 ammonium acetate and a 1:1 acetonitrile-methanol mixture, column gradient conditions was 5%-100% acetonitrile-methanol over 3.5 mins gradient time and 5 mins run time on an ACE C8 3.0 x 75mm column. Flow rate was 1.2 ml/min.
  • Table 2 below depicts data for compounds made according to the method described in Scheme I and in Examples 1-3. Compound numbers correspond to those compounds depicted in Table 1.
  • Table 3 below depicts data for certain exemplary compounds made according to the method described in Scheme I and in Examples 4-6. Compound numbers correspond to those compounds depicted in Table 1.
  • tert-butyl 4- (4- (5-methyl-lH-pyrazol-3-ylamino) -6- (azetidin-1-yl) pyrimidin-2-ylthio) phenylcarbamate (prepared using method similar to that described for example 6) (2.53g, 5.6 mmol) was dissolved in 1:1 TFA-DCM (20 mL) and the resulting solution allowed to stand overnight at room temperature. The solution was concentrated in vacuo. The residue was taken up in EtOAc and washed with saturated aqueous sodium bicarbonate solution (x2) then brine and dried over sodium sulfate. The resulting tan solid (1.8g, 91%) [MS (ES+) 354] was used without further purification or characterization in the next step.
  • reaction mixture was concentrated in vacuo and the residue taken up in methanol (3mL) .
  • Sodium methoxide (25% w/w solution in MeOH, ImL) was added and the resulting cloudy solution stirred at room temperature for 15 minutes .
  • the reaction mixture was purified directly by chromatography (silica, 5- 100% EtOAc-petrol gradient elution) to give the title compound (89mg, 33%) as a white solid.
  • Table 4 depicts data for certain exemplary compounds made according to the method described in Scheme II and in Examples 7-8. Compound numbers correspond to those compounds depicted in Table 1. Table 4
  • the solid was then collected by vacuum filtration and washed with water (390 mL) .
  • the solid was taken up in CH 2 Cl 2 (520 mL) and washed with saturated aqueous NaHCO 3 .
  • the organic layer was dried over Na 2 SO 4 , filtered, and concentrated to give the desired material (174 g, 87%) .
  • Example 10 Aurora-2 (Aurora A) Inhibition Assay
  • Compounds were screened for their ability to inhibit Aurora-2 using a standard coupled enzyme assay (Fox et al . , Protein Sci., (1998) 7, 2249). Assays were carried out in a mixture of 10OmM Hepes (pH7.5), 1OmM MgCl 2 , ImM DTT, 25mM NaCl, 2.5mM phosphoenolpyruvate , 300 ⁇ M NADH, 30 ⁇ g/ml pyruvate kinase and 10 ⁇ g/ml lactate dehydrogenase.
  • An assay stock buffer solution was prepared containing all of the reagents listed above, with the exception of Aurora-2 and the test compound of interest. 55 ⁇ l of the stock solution was placed in a 96 well plate followed by addition of 2 ⁇ l of DMSO stock containing serial dilutions of the test compound (typically starting from a final concentration of 7.5 ⁇ M) . The plate was preincubated for 10 minutes at 30 "C and the reaction initiated by addition of 10 ⁇ l of Aurora-2. Initial reaction rates were determined with a Molecular Devices SpectraMax Plus plate reader over a 10 minute time course. IC50 and Ki data were calculated from non- linear regression analysis using the Prism software package (GraphPad Prism version 3.0cx for Macintosh, GraphPad Software, San Diego California, USA) .
  • Example 11 Aurora-1 (Aurora B) Inhibition Assay (radiometric) [00108]
  • An assay buffer solution was prepared which consisted of 25 mM HEPES (pH 7.5), 10 mM MgCl 2 , 0.1% BSA and 10% glycerol.
  • a 22 nM Aurora-B solution also containing 1.7 mM DTT and 1.5 mM Kemptide (LRRASLG), was prepared in assay buffer.
  • the enzyme reaction was initiated by the addition of 16 ⁇ l stock [ ⁇ - 33 P] -ATP solution (-20 nCi/ ⁇ L) prepared in assay buffer, to a final assay concentration of 800 ⁇ M. The reaction was stopped after 3 hours by the addition of 16 ⁇ L 500 mM phosphoric acid and the levels of 33 P incorporation into the peptide substrate were determined by the following method.
  • a phosphocellulose 96-well plate (Millipore, Cat no. MAPHNOB50) was pre-treated with 100 ⁇ L of a 100 mM phosphoric acid prior to the addition of the enzyme reaction mixture (40 ⁇ L) .
  • Ki values were calculated from initial rate data by non-linear regression using the Prism software package (Prism 3.0, Graphpad Software, San Diego, CA) .
  • Example 12 Itk Inhibition Assay: Radioactivity-based Assay [00110] The compounds of the present invention were evaluated as inhibitors of human Itk kinase using a radioactivity-based assay.
  • Assays were carried out in a mixture of 20 mM MOPS (pH 7.0), 1OmM MgCl 2 , 0.1% BSA and ImM DTT. Final substrate concentrations in the assay were 7.5 ⁇ M [ ⁇ - 33 P]ATP (400 ⁇ Ci 33 P ATP/ ⁇ mol ATP, Amersham Pharmacia Biotech / Sigma Chemicals) and 3 ⁇ M peptide (SAM68 protein ⁇ 332-443) . Assays were carried out at 25 0 C. in the presence of 50 nM Itk. An assay stock buffer solution was prepared containing all of the reagents listed above, with the exception of ATP and the test compound of interest.
  • 50 ⁇ L of the stock solution was placed in a 96 well plate followed by addition of 2 ⁇ L of DMSO stock containing serial dilutions of the test compound (typically starting from a final concentration of 50 ⁇ M with 2-fold serial dilutions) in duplicate (final DMSO concentration 2%) .
  • the plate was pre-incubated for 10 minutes at 25°C and the reaction initiated by addition of 50 ⁇ L [ ⁇ - 33 P]ATP (final concentration 7.5 ⁇ M) .
  • Ki(app) data were calculated from non-linear regression analysis of the initial rate data using the Prism software package (GraphPad Prism version 3. Ocx for Macintosh, GraphPad Software, San Diego California, USA) .
  • the assays are as described above in Example 33 except that JAK-2 enzyme was used, the final poly (GIu) 4 Tyr concentration was 15 ⁇ M, and final ATP concentration was 12 ⁇ M.
  • the final concentration of a compound of the present invention was generally between 0.01 and 5 ⁇ M.
  • a 12-point titration was conducted by preparing serial dilutions from 10 mM DMSO stock of test compound. Reactions were carried out at room temperature .
  • Solution 1 contains 100 mM HEPES (pH 7.5), 10 mM MgCl 2 , 25 mM NaCl, 1 mg/ml pE4Y and 180 mM ATP (containing 0.3mCi of [ ⁇ -33 p ]ATP for each reaction).
  • Solution 2 contains 100 mM HEPES (pH 7.5), 10 mM MgCl 2 , 25 mM NaCl, 2 mM DTT, 0.02% BSA and 3 nM FLT-3.
  • the assay was run on a 96 well plate by mixing 50 ⁇ l each of Solution 1 and 2.5 ml of the compounds of the present invention. The reaction was initiated with Solution 2.
  • Microsomal stability was monitored by generation of depletion-time profiles in microsomes from a range of species (male CD-I mouse, male Sprague-Dawley rat, male Beagle dog, male Cynomolgus monkey and pooled mixed gender human) .
  • Compound spiking solutions were made up by diluting down the compound stock solution in DMSO (typically 10 mM) to give a solution in acetonitrile (0.5 mM) .
  • the reaction was initiated by the addition (250 ⁇ L) of the pre-incubated RGS to the pre-incubated microsome/VRT/PB mixture (pre-incubation in both instances was for 10 minutes at 37 0 C) .
  • Samples were incubated within Eppendorf vials (1.5 ml) on a heater shaker (DPC Micromix 5 (settings; form 20, amplitude 4) modified to be heated, to 37 0 C, by two plate heaters fixed to the deck and controlled by a Packard Manual Heater) attached to a Multiprobe II HT Ex automated liquid handler.
  • the liquid handler was programmed (WinPREP software) to sample the microsomal incubation mixture after 0, 2, 10, 30 and 60 minutes of incubation and transfer an aliquot (100 ⁇ L) to a stop block (96-well block) containing 100 ⁇ L of chilled methanol.
  • the % organic in the stop mixture was optimized for analysis by addition of appropriate volumes of aqueous/organic (typically 100 ⁇ L of 50:50 methanol: water) .
  • aqueous/organic typically 100 ⁇ L of 50:50 methanol: water
  • Prior to analysis the stop block was placed on a shaker (DPC Micromix 5; 10 min, form 20, amplitude 5) to precipitate out proteins . The block was then centrifuged (Jouan GR412; 2000 rpm, 15 min, 4 0 C).
  • a sample aliquot (200 ⁇ L) was then transferred to an analysis block and the block was centrifuged again (Jouan GR412; 2000 rpm, 5 min, 4 0 C) prior to being sent for analysis.
  • Depletion profiles were determined by monitoring the disappearance of VRT by liquid chromatography-tandem mass spectrometry (LC-MS/MS) .
  • Samples were injected (20 ⁇ L; Agilent 1100 liquid chromatographic system equipped with autosampler) onto an analytical column.
  • Mobile phase consisted of Water + 0.05% (v/v) formic acid (A) and methanol + 0.05% (v/v) formic acid (B) .
  • Running a gradient method optimized for the compound of interest carried out the compound elution from analytical column The total run time was 6 minutes with a flow rate of 0.35 mL/min. The entire column effluent entered the electrospray ionization source (positive mode) of a Micromass Quattro LC tandem mass spectrometer between 0.5 and 5.9 min of the run. The mass spectrometry was optimized for the compound of interest. All incubations were conducted in duplicate and results were expressed as % parent remaining at either 30 minutes or 60 minutes relative to 0 minutes sample.
  • Example 17 Analysis of cell proliferation and viability [00123] Compounds were screened for their ability to inhibit cell proliferation and their effects on cell viability using
  • Colo205 cells obtained from ECACC and using the assay shown below. [00124] Colo205 cells were seeded in 96 well plates and serially diluted compound was added to the wells in duplicate.
  • Control groups included untreated cells, the compound diluent (0.1% DMSO alone) and culture medium without cells. The cells were then incubated for 72 or 96 hrs at 37C in an atmosphere of 5% CO2/95% humidity.
  • Example 18 AbI Kinase Activity Inhibition Assay and Determination of the Inhibition Constant Ki
  • Reactions were carried out at 30 0 C and 21 nM AbI kinase. Final concentrations of the components of the coupled enzyme system were 2.5 mM phosphoenolpyruvate, 200 ⁇ M NADH, 60 ⁇ g/ml pyruvate kinase and 20 ⁇ g/ml lactate dehydrogenase.
  • An assay stock buffer solution was prepared containing all of the reagents listed above with the exception of ATP and the test compound of interest. The assay stock buffer solution (60 ⁇ l) was incubated in a 96 well plate with 2 ⁇ l of the test compound of interest at final concentrations typically spanning 0.002 ⁇ M to 30 ⁇ M at 30 0 C for 10 min.
  • a 12 point titration was prepared by serial dilutions (from 1 mM compound stocks) with DMSO of the test compounds in daughter plates .
  • the reaction was initiated by the addition of 5 ⁇ l of ATP (final concentration 110 ⁇ M) .
  • Rates of reaction were obtained using a Molecular Devices Spectramax plate reader (Sunnyvale, CA) over 10 rain at 30 0 C.
  • the Ki values were determined from the residual rate data as a function of inhibitor concentration using nonlinear regression
  • Example 19 Mutant AbI Kinase (T315I) Activity Inhibition Assay and Determination of the Inhibition Constant IC50
  • Example 20 Arg (AbI-2) , FGFRl, MELK, MLKl, MuSK, Ret, and TrkA Inhibition Assay
  • Compound 1-1 was screened for its ability to inhibit Arg (Abl-2) , FGFRl, MELK, MLKl, MuSK, Ret, and TrkA using screening methods known to one of ordinary skill in the art. All of the above enzymes were screened with ATP concentrations at or close to the K m for ATP.
  • Table 6 below depicts data from Examples 10-11 and 16 described above.

Abstract

The present invention relates to compounds useful as inhibitors of Aurora protein kinases . The invention also provides pharmaceutically acceptable compositions comprising those compounds and methods of using the compounds and compositions in the treatment of various disease, conditions, and disorders . The invention also provides processes for preparing compounds of the invention.

Description

AMINOPYRIMIDINES USEFUL AS KINASE INHIBITORS
TECHNICAL FIELD OF THE INVENTION
[0001] The present invention relates to compounds useful as inhibitors of Aurora protein kinases . The invention also relates to pharmaceutically acceptable compositions comprising the compounds of the invention, methods of using the compounds and compositions in the treatment of various disorders, and processes for preparing the compounds.
BACKGROUND OF THE INVENTION
[0002] The Aurora proteins are a family of three related serine/threonine kinases (termed Aurora-A, -B and -C) that are essential for progression through the mitotic phase of cell cycle. Specifically Aurora-A plays a crucial role in centrosome maturation and segregation, formation of the mitotic spindle and faithful segregation of chromosomes . Aurora-B is a chromosomal passenger protein that plays a central role in regulating the alignment of chromosomes on the meta-phase plate, the spindle assembly checkpoint and for the correct completion of cytokinesis.
[0003] Overexpression of Aurora-A, -B or -C has been observed in a range of human cancers including colorectal, ovarian, gastric and invasive duct adenocarcinomas. [0004] A number of studies have now demonstrated that depletion or inhibition of Aurora-A or -B in human cancer cell lines by siRNA, dominant negative antibodies or neutralizing antibodies disrupts progression through mitosis with accumulation of cells with 4N DNA, and in some cases this is followed by endoreduplication and cell death.
[0005] The Aurora kinases are attractive targets due to their association with numerous human cancers and the roles they play in the proliferation of these cancer cells. It would be desirable to have an Aurora kinase inhibitor with favorable drug-like properties, such as stability in human liver microsomes. Accordingly, there is a need for compounds that inhibit Aurora kinases and also exhibit favorable drug-like properties .
SUMMARY OF THE INVENTION
[0006] This invention provides compounds and pharmaceutically acceptable compositions thereof that are useful as inhibitors of Aurora protein kinases. These compounds are represented by formula I :
Figure imgf000003_0001
or a pharmaceutically acceptable salt thereof, wherein R2, R9, and p are as defined herein.
[0007] These compounds and pharmaceutically acceptable compositions thereof are useful for inhibiting kinases in vitro, in vivo, and ex vivo. Such uses include treating or preventing myeloproliferative disorders and proliferative disorders such as melanoma, myeloma, leukemia, lymphoma, neuroblastoma, and cancer. Other uses include the study of kinases in biological and pathological phenomena; the study of intracellular signal transduction pathways mediated by such kinases; and the comparative evaluation of new kinase inhibitors .
DETAILED DESCRIPTION OF THE INVENTION [0008] The present invention provides a compound of formula I:
Figure imgf000004_0001
or a pharmaceutically acceptable salt thereof, wherein:
R2 is Chalky1 or cyclopropyl;
R9 is halo, C^alkyl, -O- (Ci-3alkyl) , -S- (Ci_3alkyl) , -OCF3, or
CF3; and p is 1-2.
[0009] In some embodiments, R2 is methyl.
[0010] In other embodiments, p is 1.
[0011] In some embodiments, R9 is substituted in the ortho position.
[0012] In some aspects of the invention, R9 is CF3, halo, Ci_3alkyl, or -S- (Ci_3alkyl) . In some embodiments, R9 is F, Cl, or CF3.
[0013] One aspect provides a compound selected from Table 1
(or a pharmaceutically acceptable salt thereof) :
Table 1:
Figure imgf000005_0001
Figure imgf000006_0001
Figure imgf000007_0001
Figure imgf000008_0001
1- 23 1 - 24
[0014] For purposes of this invention, the chemical elements are identified in accordance with the Periodic Table of the Elements, CAS version, Handbook of Chemistry and Physics, 75th Ed. Additionally, general principles of organic chemistry are described in texts known to those of ordinary skill in the art, including, for example, "Organic Chemistry", Thomas Sorrell, University Science Books, Sausalito: 1999, and "March's Advanced Organic Chemistry", 5th Ed., Ed.: Smith, M. B. and March, J., John Wiley & Sons, New York: 2001, the entire contents of which are hereby incorporated by reference.
[0015] As described herein, a specified number range of atoms includes any integer therein. For example, a group having from 1-4 atoms could have 1, 2, 3, or 4 atoms.
[0016] As described herein, compounds of the invention may optionally be substituted with one or more substituents, such as are illustrated generally above, or as exemplified by particular classes, subclasses, and species of the invention. It will be appreciated that the phrase "optionally substituted" is used interchangeably with the phrase "substituted or unsubstituted. " In general, the term "substituted", whether preceded by the term "optionally" or not, refers to the replacement of hydrogen radicals in a given structure with the radical of a specified substituent. Unless otherwise indicated, an optionally substituted group may have a substituent at each substitutable position of the group, and when more than one position in any given structure may be substituted with more than one substituent selected from a specified group, the substituent may be either the same or different at every position. Combinations of substituents envisioned by this invention are preferably those that result in the formation of stable or chemically feasible compounds .
[0017] The term "stable", as used herein, refers to compounds that are not substantially altered when subjected to conditions to allow for their production, detection, and preferably their recovery, purification, and use for one or more of the purposes disclosed herein. In some embodiments, a stable compound or chemically feasible compound is one that is not substantially altered when kept at a temperature of 40 0C or less, in the absence of moisture or other chemically reactive conditions, for at least a week.
[0018] The term "alkyl" as used herein, means an unbranched or branched, straight-chain hydrocarbon that is completely saturated and has a single point of attachment to the rest of the molecule. Specific examples of alkyl groups include, but are not limited to, methyl, ethyl, isopropyl, n-propyl, and sec-butyl .
[0019] The term "cycloalkyl" refers to a monocyclic hydrocarbon that is completely saturated and has a single point of attachment to the rest of the molecule. Suitable cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, and cyclopentyl.
[0021] The term "unsaturated", as used herein, means that a moiety has one or more units of unsaturation.
[0022] The terms "haloalkyl" means an alkyl substituted with one or more halogen atoms . This includes perfluorinated alkyl groups , such as CF3.
[0023] The term "halogen" means F, Cl, Br, or I . [0024] The term "protecting group", as used herein, refers to an agent used to temporarily block one or more desired reactive sites in a multifunctional compound. In certain embodiments, a protecting group has one or more, or preferably all, of the following characteristics: a) reacts selectively in good yield to give a protected substrate that is stable to the reactions occurring at one or more of the other reactive sites; and b) is selectively removable in good yield by reagents that do not attack the regenerated functional group. Exemplary protecting groups are detailed in Greene, T. W., Wuts, P. G in "Protective Groups in Organic Synthesis", Third Edition, John Wiley & Sons, New York: 1999, and other editions of this book, the entire contents of which are hereby incorporated by reference. The term "nitrogen protecting group" , as used herein, refers to an agents used to temporarily block one or more desired nitrogen reactive sites in a multifunctional compound. Preferred nitrogen protecting groups also possess the characteristics exemplified above, and certain exemplary nitrogen protecting groups are also detailed in Chapter 7 in Greene, T.W., Wuts, P. G in "Protective Groups in Organic Synthesis", Third Edition, John Wiley & Sons, New York: 1999, the entire contents of which are hereby incorporated by reference.
[0025] Unless otherwise indicated, structures depicted herein are also meant to include all isomeric (e.g., enantiomeric, diastereomeric, and geometric (or conformational) ) forms of the structure; for example, the R and S configurations for each asymmetric center, (Z) and (E) double bond isomers, and (Z) and (E) conformational isomers. Therefore, single stereochemical isomers as well as enantiomeric, diastereomeric, and geometric (or conformational) mixtures of the present compounds are within the scope of the invention. [0026] Unless otherwise indicated, all tautomeric forms of the compounds of the invention are within the scope of the invention. As would be understood by a skilled practitioner, a pyrazole group can be represented in a variety of ways . For
example, a structure drawn a also represents other
possible tautoraers, such as
Figure imgf000011_0001
, Likewise, a structure
drawn as
Figure imgf000011_0002
also represents other possible tautomers, such
Figure imgf000011_0003
[0027] Unless otherwise indicated, a substituent can freely rotate around any rotatable bonds. For example, a substituent
drawn as
Figure imgf000011_0004
also represents . Likewise, a
substituent drawn as
Figure imgf000011_0005
also represents [0028] Additionally, unless otherwise indicated, structures depicted herein are also meant to include compounds that differ only in the presence of one or more isotopically enriched atoms. For example, compounds having the present structures except for the replacement of hydrogen by deuterium or tritium, or the replacement of a carbon by a 13C- or 14C- enriched carbon are within the scope of this invention. Such compounds are useful, for example, as analytical tools or probes in biological assays.
[0029] The compounds of this invention may be prepared in light of the specification using steps generally known to those of ordinary skill in the art. Those compounds may be analyzed by known methods, including but not limited to LCMS (liquid chromatography mass spectrometry) and NMR (nuclear magnetic resonance) . It should be understood that the specific conditions shown below are only examples, and are not meant to limit the scope of the conditions that can be used for making compounds of this invention. Instead, this invention also includes conditions that would be apparent to those skilled in that art in light of this specification for making the compounds of this invention. Unless otherwise indicated, all variables in the following schemes are as defined herein.
[0030] The following abbreviations are used: DIPEA is diisopropylethylamine DMF is dimethylformamide Ώ-BUOH is π-butanol t-BuOH is tert-butanol MeOH is methanol EtOAc is ethyl acetate TFA is trifluoroacetic acid DMSO is dimethyl sulfoxide Rt is retention time DCM is dichloromethane MeCN is acetonitrile THF is tetrahydrofuran
TBTU is 2- (lH-Benzotriazole-1-yl) -1, 1, 3, 3-tetramethyluronium tetrafluoroborate
HPLC is high performance liquid chromatography LCMS liquid chromatography mass spectrometry 1H NMR is nuclear magnetic resonance
General Scheme
Figure imgf000013_0001
NH HQR1
Lu base, solvent
J0-6
Method A Method B
Figure imgf000013_0002
[0031] The general scheme above shows some methods of making compounds of this invention.
Figure imgf000013_0003
4
[0032] Scheme I above shows a general route for the preparation of compounds of formula 4 (in Scheme I) , wherein the variables are as defined herein. The dichlorinated pyrimidine of formula 1 is combined with HQ-R1 to form a compound of formula 2. In some embodiments, the two compounds are heated in the presence of a suitable solvent (e.g. t-BuOH) for 16 hours. In other embodiments, the two compounds are mixed at 0 °C in the presence of acetonitrile and triethylamine for 1 hour. The compound of formula 2 is then heated in the presence of a suitable solvent (e.g. DMF) and a suitable base (e.g. DIPEA/Nal) with an optionally substituted aminopyrazole to form a compound of formula 3, which is heated in the presence of azetidine in the presence of a suitable solvent (e.g. n-BuOH) to form a compound of formula 4.
Scheme II
Figure imgf000014_0001
5 6
[0033] Scheme II above shows a general route for the preparation of compounds of formula 6 (in Scheme II) , wherein R2 and R5 are as defined herein. The compound of formula 5 is combined with a suitable acid chloride (wherein X" is Cl) in the presence of pyridine to form an intermediate compound that, upon mixing in the presence of sodium methoxide and methanol, forms the compound of formula 6. In some embodiments, X" can be OH, in which case a suitable acid coupling reagent is used to couple the acid to the amine. Examples of suitable acid coupling reagents include, but are not limited to, EDC, DCI, and HOBT. Suitable solvents for these coupling reactions include, but are not limited to, THF, CH2Cl2, and dioxane . [0034] Accordingly, this invention relates to processes for making the compounds of this invention.
[0035] Methods for evaluating the activity of the compounds of this invention (e.g., kinase assays) are known in the art and are also described in the examples set forth.
[0036] The activity of the compounds as protein kinase inhibitors may be assayed in vitro, in vivo or in a cell line. In vitro assays include assays that determine inhibition of either the kinase activity or ATPase activity of the activated kinase. Alternate in vitro assays guantitate the ability of the inhibitor to bind to the protein kinase and may be measured either by radiolabelling the inhibitor prior to binding, isolating the inhibitor/kinase complex and determining the amount of radiolabel bound, or by running a competition experiment where new inhibitors are incubated with the kinase bound to known radioligands .
[0037] Another aspect of the invention relates to inhibiting kinase activity in a biological sample, which method comprises contacting said biological sample with a compound of formula I or a composition comprising said compound. The term "biological sample", as used herein, means an in vitro or an ex vivo sample, including, without limitation, cell cultures or extracts thereof; biopsied material obtained from a mammal or extracts thereof; and blood, saliva, urine, feces, semen, tears, or other body fluids or extracts thereof.
[0038] Inhibition of kinase activity in a biological sample is useful for a variety of purposes that are known to one of skill in the art. Examples of such purposes include, but are not limited to, blood transfusion, organ-transplantation, biological specimen storage, and biological assays.
[0039] Inhibition of kinase activity in a biological sample is also useful for the study of kinases in biological and pathological phenomena; the study of intracellular signal transduction pathways mediated by such kinases,- and the comparative evaluation of new kinase inhibitors . [0040] The Aurora protein kinase inhibitors or pharmaceutical salts thereof may be formulated into pharmaceutical compositions for administration to animals or humans . These pharmaceutical compositions, which comprise an amount of the Aurora protein inhibitor effective to treat or prevent an Aurora-mediated condition and a pharmaceutically acceptable carrier, are another embodiment of the present invention. [0041] The term "Aurora-mediated condition" or "Aurora- mediated disease" as used herein means any disease or other deleterious condition in which Aurora (Aurora A, Aurora B, and Aurora C) is known to play a role. Such conditions include, without limitation, cancer, proliferative disorders, and myeloproliferative disorders.
[0042] Examples of myeloproliferative disorders include, but are not limited, to, polycythemia vera, thrombocythemia, myeloid metaplasia with myelofibrosis, chronic myelogenous leukaemia (CML) , chronic myelomonocytic leukemia, hypereosinophilic syndrome, juvenile myelomonocytic leukemia, and systemic mast cell disease.
[0043] The term "cancer" also includes, but is not limited to, the following cancers: epidermoid Oral : buccal cavity, lip, tongue, mouth, pharynx; Cardiac : sarcoma (angiosarcoma, fibrosarcoma, rhabdomyosarcoma, liposarcoma) , myxoma, rhabdomyoma, fibroma, lipoma and teratoma; Lung: bronchogenic carcinoma (squamous cell or epidermoid, undifferentiated small cell, undifferentiated large cell, adenocarcinoma) , alveolar (bronchiolar) carcinoma, bronchial adenoma, sarcoma, lymphoma, chondromatous hamartoma, mesothelioma; Gastrointestinal : esophagus (squamous cell carcinoma, larynx, adenocarcinoma, leiomyosarcoma, lymphoma) , stomach (carcinoma, lymphoma, leiomyosarcoma) , pancreas (ductal adenocarcinoma, insulinoma, glucagonoma, gastrinoma, carcinoid tumors, vipoma), small bowel or small intestines (adenocarcinoma, lymphoma, carcinoid tumors, Karposi's sarcoma, leiomyoma, hemangioma, lipoma, neurofibroma, fibroma) , large bowel or large intestines (adenocarcinoma, tubular adenoma, villous adenoma, hamartoma, leiomyoma) , colon, colon-rectum, colorectal; rectum, Genitourinary tract: kidney (adenocarcinoma, WiIm1 s tumor [nephroblastoma] , lymphoma, leukemia) , bladder and urethra (squamous cell carcinoma, transitional cell carcinoma, adenocarcinoma) , prostate (adenocarcinoma, sarcoma) , testis (seminoma, teratoma, embryonal carcinoma, - teratocarcinoma, choriocarcinoma, sarcoma, interstitial cell carcinoma, fibroma, fibroadenoma, adenomatoid tumors, lipoma); Liver: hepatoma (hepatocellular carcinoma) , cholangiocarcinoma, hepatoblastoma, angiosarcoma, hepatocellular adenoma, hemangioma, biliary passages; Bone : osteogenic sarcoma (osteosarcoma) , fibrosarcoma, malignant fibrous histiocytoma, chondrosarcoma, Ewing ' s sarcoma, malignant lymphoma (reticulum cell sarcoma) , multiple myeloma, malignant giant cell tumor chordoma, osteochronfroma (osteocartilaginous exostoses) , benign chondroma, chondroblastoma, chondromyxofibroma, osteoid osteoma and giant cell tumors; Nervous system: skull (osteoma, hemangioma, granuloma, xanthoma, osteitis deformans) , meninges (meningioma, meningiosarcoma, gliomatosis) , brain (astrocytoma, medulloblastoma, glioma, ependymoma, germinoma [pinealoma] , glioblastoma multiform, oligodendroglioma, schwannoma, retinoblastoma, congenital tumors) , spinal cord neurofibroma, meningioma, glioma, sarcoma) ; Gynecological : uterus (endometrial carcinoma) , cervix (cervical carcinoma, pre-tumor cervical dysplasia) , ovaries (ovarian carcinoma [serous cystadenocarcinoma, mucinous cystadenocarcinoma, unclassified carcinoma], granulosa-thecal cell tumors, Sertoli-Leydig cell tumors, dysgerminoma, malignant teratoma), vulva (squamous cell carcinoma, intraepithelial carcinoma, adenocarcinoma, fibrosarcoma, melanoma) , vagina (clear cell carcinoma, squamous cell carcinoma, botryoid sarcoma (embryonal rhabdomyosarcoma) , fallopian tubes (carcinoma) , breast; Hematologic : blood (myeloid leukemia [acute and chronic] , acute lymphoblastic leukemia, chronic lymphocytic leukemia, myeloproliferative diseases, multiple myeloma, myelodysplastic syndrome), Hodgkin's disease, non-Hodgkin1 s lymphoma [malignant lymphoma] hairy cell; lymphoid disorders; Skin: malignant melanoma, basal cell carcinoma, squamous cell carcinoma, Karposi's sarcoma, keratoacanthoma, moles dysplastic nevi, lipoma, angioma, dermatofibroma, keloids, psoriasis, Thyroid gland: papillary thyroid carcinoma, follicular thyroid carcinoma; medullary thyroid carcinoma, undifferentiated thyroid cancer, multiple endocrine neoplasia type 2A, multiple endocrine neoplasia type 2B, familial medullary thyroid cancer, pheochromocytoma, paraganglioma; and Adrenal glands : neuroblastoma. Thus, the term "cancerous cell" as provided herein, includes a cell afflicted by any one of the above-identified conditions. In some embodiments, the cancer is selected from colorectal, thyroid, or breast cancer. [0044] In some embodiments, the compounds of this invention are useful for treating cancer, such as colorectal, thyroid, breast, and lung cancer; and myeloproliferative disorders, such as polycythemia vera, thrombocythemia, myeloid metaplasia with myelofibrosis, chronic myelogenous leukemia, chronic myelomonocytic leukemia, hypereosinophilic syndrome, juvenile myelomonocytic leukemia, and systemic mast cell disease. [0045] In some embodiments, the compounds of this invention are useful for treating hematopoietic disorders, in particular, acute-myelogenous leukemia (AML) , chronic- myelogenous leukemia (CML) , acute-promyelocytic leukemia (APL) , and acute lymphocytic leukemia (ALL) . [0046] In addition to the compounds of this invention, pharmaceutically acceptable derivatives or prodrugs of the compounds of this invention may also be employed in compositions to treat or prevent the above-identified disorders .
[0047] A "pharmaceutically acceptable derivative or prodrug" means any pharmaceutically acceptable ester, salt of an ester or other derivative of a compound of this invention which, upon administration to a recipient, is capable of providing, either directly or indirectly, a compound of this invention or an inhibitorily active metabolite or residue thereof . Such derivatives or prodrugs include those that increase the bioavailability of the compounds of this invention when such compounds are administered to a patient (e.g., by allowing an orally administered compound to be more readily absorbed into the blood) or which enhance delivery of the parent compound to a biological compartment (e.g., the brain or lymphatic system) relative to the parent species .
[0048] Examples of pharmaceutically acceptable prodrugs of the compounds of this invention include, without limitation, esters, amino acid esters, phosphate esters, metal salts and sulfonate esters .
[0049] The compounds of this invention can exist in free form for treatment, or where appropriate, as a pharmaceutically acceptable salt .
[0050] As used herein, the term "pharmaceutically acceptable salt" refers to salts of a compound which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio. [0051] Pharmaceutically acceptable salts of the compounds of this invention include those derived from suitable inorganic and organic acids and bases. These salts can be prepared in situ during the final isolation and purification of the compounds. Acid addition salts can be prepared by 1) reacting the purified compound in its free-based form with a suitable organic or inorganic acid and 2) isolating the salt thus formed.
[0052] Examples of suitable acid salts include acetate, adipate, alginate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, citrate, camphorate, camphorsulfonate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptanoate, glycerophosphate, glycolate, hemisulfate, heptanoate, hexanoate, hydrochloride, hydrobromide , hydroiodide, 2- hydroxyethanesulfonate, lactate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oxalate, palmoate, pectinate, persulfate, 3-phenylpropionate, phosphate, picrate, pivalate, propionate, salicylate, succinate, sulfate, tartrate, thiocyanate, tosylate and undecanoate . Other acids, such as oxalic, while not in themselves pharmaceutically acceptable, may be employed in the preparation of salts useful as intermediates in obtaining the compounds of the invention and their pharmaceutically acceptable acid addition salts .
[0053] Base addition salts can be prepared by 1) reacting the purified compound in its acid form with a suitable organic or inorganic base and 2) isolating the salt thus formed. [0054] Salts derived from appropriate bases include alkali metal (e.g., sodium and potassium), alkaline earth metal (e.g., magnesium), ammonium and N+(Ci.4 alkyl)4 salts. This invention also envisions the quaternization of any basic nitrogen-containing groups of the compounds disclosed herein. Water or oil-soluble or dispersible products may be obtained by such quaternization.
[0055] Base addition salts also include alkali or alkaline earth metal salts. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like. Further pharmaceutically acceptable salts include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, loweralkyl sulfonate and aryl sulfonate. Other acids and bases, while not in themselves pharmaceutically acceptable, may be employed in the preparation of salts useful as intermediates in obtaining the compounds of the invention and their pharmaceutically acceptable acid or base addition salts. [0056] Pharmaceutically acceptable carriers that may be used in these pharmaceutical compositions include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates , waxes, polyethylene- polyoxypropylene-block polymers, polyethylene glycol and wool fat.
[0057] The compositions of the present invention may be administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir. The term "parenteral" as used herein includes subcutaneous, intravenous, intramuscular, intraarticular, intra-synovial, intrasternal, intrathecal, intraperitoneal, intrahepatic, intralesional and intracranial injection or infusion techniques.
[0058] Sterile injectable forms of the compositions of this invention may be aqueous or oleaginous suspension. These suspensions may be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution or suspension in a nontoxic parenterally-acceptable diluent or solvent, for example as a solution in 1, 3-butanediol . Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose, a bland fixed oil may be employed including synthetic mono- or di-glycerides . Fatty acids, such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions. These oil solutions or suspensions may also contain a long-chain alcohol diluent or dispersant, such as carboxymethyl cellulose or similar dispersing agents which are commonly used in the formulation of pharmaceutically acceptable dosage forms including emulsions and suspensions. Other commonly used surfactants, such as Tweens, Spans and other emulsifying agents or bioavailability enhancers which are commonly used in the manufacture of pharmaceutically acceptable solid, liquid, or other dosage forms may also be used for the purposes of formulation.
[0059] The pharmaceutical compositions of this invention may be orally administered in any orally acceptable dosage form including, but not limited to, capsules, tablets, aqueous suspensions or solutions . In the case of tablets for oral use, carriers commonly used may include lactose and corn starch. Lubricating agents, such as magnesium stearate, may also be added. For oral administration in a capsule form, useful diluents may include lactose and dried cornstarch. When aqueous suspensions are required for oral use, the active ingredient may be combined with emulsifying and suspending agents. If desired, certain sweetening, flavoring or coloring agents may also be added. [0060] Alternatively, the pharmaceutical compositions of this invention may be administered in the form of suppositories for rectal administration. These can be prepared by mixing the agent with a suitable non-irritating excipient which is solid at room temperature but liquid at rectal temperature and therefore will melt in the rectum to release the drug. Such materials may include cocoa butter, beeswax and polyethylene glycols.
[0061] The pharmaceutical compositions of this invention may also be administered topically, especially when the target of treatment includes areas or organs readily accessible by topical application, including diseases of the eye, the skin, or the lower intestinal tract. Suitable topical formulations may be prepared for each of these areas or organs .
[0062] Topical application for the lower intestinal tract can be effected in a rectal suppository formulation (see above) or in a suitable enema formulation. Topically-transdermal patches may also be used.
[0063] For topical applications, the pharmaceutical compositions may be formulated in a suitable ointment containing the active component suspended or dissolved in one or more carriers . Carriers for topical administration of the compounds of this invention may include, but are not limited to, mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene, polyoxypropylene compound, emulsifying wax and water. Alternatively, the pharmaceutical compositions may be formulated in a suitable lotion or cream containing the active components suspended or dissolved in one or more pharmaceutically acceptable carriers. Suitable carriers may include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water.
[0064] For ophthalmic use, the pharmaceutical compositions may be formulated as micronized suspensions in isotonic, pH adjusted sterile saline, or as solutions in isotonic, pH adjusted sterile saline, either with or without a preservative such as benzylalkonium • chloride. Alternatively, for ophthalmic uses, the pharmaceutical compositions may be formulated in an ointment such as petrolatum.
[0065] The pharmaceutical compositions of this invention may also be administered by nasal aerosol or inhalation. Such compositions may be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons , and/or other conventional solubilizing or dispersing agents.
[0066] The amount of kinase inhibitor that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated, the particular mode of administration, and the indication. In an embodiment, the compositions should be formulated so that a dosage of between 0.01 - 100 mg/kg body weight/day of the inhibitor can be administered to a patient receiving these compositions. In another embodiment, the compositions should be formulated so that a dosage of between 0.1 - 100 mg/kg body weight/day of the inhibitor can be administered to a patient receiving these compositions .
[0067] It should also be understood that a specific dosage and treatment regimen for any particular patient will depend upon a variety of factors, including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, rate of excretion, drug combination, and the judgment of the treating physician and the severity of the particular disease being treated. The amount of inhibitor will also depend upon the particular compound in the composition.
[0068] According to another embodiment, the invention provides methods for treating or preventing cancer, a proliferative disorder, or a myeloproliferative disorder comprising the step of administering to a patient one of the herein-described compounds or pharmaceutical compositions.
[0069] The term "patient", as used herein, means an animal, including a human.
[0070] In some embodiments, said method is used to treat or prevent a hematopoietic disorder, such as acute-myelogenous leukemia (AML) , acute-promyelocytic leukemia (APL) , chronic- myelogenous leukemia (CML) , or acute lymphocytic leukemia
(ALL) .
[0071] In other embodiments, said method is used to treat or prevent myeloproliferative disorders, such as polycythemia vera, thrombocythemia, myeloid metaplasia with myelofibrosis, chronic myelogenous leukaemia (CML) , chronic myelomonocytic leukemia, hypereosinophilic syndrome, juvenile myelomonocytic leukemia, and systemic mast cell disease.
[0072] In yet other embodiments, said method is used to treat or prevent cancer, such as cancers of the breast, colon, prostate, skin, pancreas, brain, genitourinary tract, lymphatic system, stomach, larynx and lung, including lung adenocarcinoma, small cell lung cancer, and non-small cell lung cancer.
[0073] Another embodiment provides a method of treating or preventing cancer comprising the step of administering to a patient a compound of formula I or a composition comprising said compound .
[0074] Another aspect of the invention relates to inhibiting kinase activity in a patient, which method comprises administering to the patient a compound of formula I or a composition comprising said compound. In some embodiments, said kinase is an Aurora kinase (Aurora A, Aurora B, Aurora C), AbI, Arg, FGFRl, MELK, MLKl, MuSK, Ret, or TrkA.
[0075] Depending upon the particular conditions to be treated or prevented, additional drugs may be administered together with the compounds of this invention. In some cases, these additional drugs are normally administered to treat or prevent the same condition. For example, chemotherapeutic agents or other anti-proliferative agents may be combined with the compounds of this invention to treat proliferative diseases . [0076] Another aspect of this invention is directed towards a method of treating cancer in a subject in need thereof, comprising the sequential or co-administration of a compound of this invention or a pharmaceutically acceptable salt thereof, and another therapeutic agent. In some embodiments, said additional therapeutic agent is selected from an anticancer agent, an anti-proliferative agent, or a chemotherapeutic agent.
[0077] In some embodiments, said additional therapeutic agent is selected from camptothecin, the MEK inhibitor: U0126, a KSP (kinesin spindle protein) inhibitor, adriamycin, interferons, and platinum derivatives, such as Cisplatin.
[0078] In other embodiments, said additional therapeutic agent is selected from taxanes; inhibitors of bcr-abl (such as Gleevec, dasatinib, and nilotinib) ; inhibitors of EGFR (such as Tarceva and Iressa) ; DNA damaging agents (such as cisplatin, oxaliplatin, carboplatin, topoisomerase inhibitors, and anthracyclines) ; and antimetabolites (such as AraC and 5-FU) .
[0079] In yet other embodiments, said additional therapeutic agent is selected from camptothecin, doxorubicin, idarubicin, Cisplatin, taxol, taxotere, vincristine, tarceva, the MEK inhibitor, U0126, a KSP inhibitor, vorinostat, Gleevec, dasatinib, and nilotinib.
[0080] In another embodiment, said additional therapeutic agent is selected from Her-2 inhibitors (such as Herceptin) ; HDAC inhibitors (such as vorinostat) , VEGFR inhibitors (such as Avastin) , c-KIT and FLT-3 inhibitors (such as sunitinib) , BRAF inhibitors (such as Bayer's BAY 43-9006) MEK inhibitors (such as Pfizer' s PD0325901) ; and spindle poisons (such as Epothilones and paclitaxel protein-bound particles (such as Abraxane®) .
[0081] Other therapies or anticancer agents that may be used in combination with the inventive anticancer agents of the present invention include surgery, radiotherapy (in but a few examples, gamma-radiation, neutron beam radiotherapy, electron beam radiotherapy, proton therapy, brachytherapy, and systemic radioactive isotopes, to name a few), endocrine therapy, biologic response modifiers (interferons, interleukins, and tumor necrosis factor (TNF) to name a few) , hyperthermia and cryotherapy, agents to attenuate any adverse effects (e.g., antiemetics), and other approved chemotherapeutic drugs, including, but not limited to, alkylating drugs (mechlorethamine, chlorambucil, Cyclophosphamide, Melphalan, Ifosfamide) , antimetabolites (Methotrexate) , purine antagonists and pyrimidine antagonists (6-Mercaptopurine, 5- Fluorouracil, Cytarabile, Gemcitabine) , spindle poisons (Vinblastine, Vincristine, Vinorelbine, Paclitaxel) , podophylIotoxins (Etoposide, Irinotecan, Topotecan) , antibiotics (Doxorubicin, Bleomycin, Mitomycin) , nitrosoureas (Carmustine, Lomustine) , inorganic ions (Cisplatin, Carboplatin) , enzymes (Asparaginase) , and hormones (Tamoxifen, Leuprolide, Flutamide, and Megestrol) , Gleevec™, dexamethasone, and cyclophosphamide.
[0082] A compound of the instant invention may also be useful for treating cancer in combination with the following therapeutic agents: abarelix (Plenaxis depot®); aldesleukin (Prokine®) ; Aldesleukin (Proleukin®) ; Alemtuzumabb (Campath®) ; alitretinoin (Panretin®) ; allopurinol (Zyloprim®) ; altretamine (Hexalen®) ; amifostine (Ethyol®) ; anastrozole (Arimidex®) ; arsenic trioxide (Trisenox®) ; asparaginase (Elspar®) ; azacitidine (Vidaza®) ; bevacuzimab (Avastin®) ; bexarotene capsules (Targretin®) ; bexarotene gel (Targretin®) ; bleomycin (Blenoxane®) ; bortezomib (Velcade®) ; busulfan intravenous (Busulfex®) ; busulfan oral (Myleran®) ; calusterone (Methosarb®) ; capecitabine (Xeloda®) ; carboplatin (Paraplatin®) ; carmustine (BCNU®, BiCNU®) ; carmustine (Gliadel®) ; carmustine with Polifeprosan 20 Implant (Gliadel Wafer®) ; celecoxib (Celebrex®) ; cetuximab (Erbitux®) ; chlorambucil (Leukeran®) ; cisplatin (Platinol®) ; cladribine (Leustatin®, 2-CdA®) ; clofarabine (Clolar®) ; cyclophosphamide (Cytoxan®, Neosar®) ; cyclophosphamide (Cytoxan Injection®) ; cyclophosphamide (Cytoxan Tablet®) ; cytarabine (Cytosar-ϋ®) ; cytarabine liposomal (DepoCyt®) ; dacarbazine (DTIC-Dome®) ; dactinomycin, actinomycin D (Cosmegen®) ; Darbepoetin alfa (Aranesp®) ; daunorubicin liposomal (DanuoXome®) ; daunorubicin, daunomycin (Daunorubicin®) ; daunorubicin, daunomycin (Cerubidine®) ; Denileukin diftitox (Ontak®) ; dexrazoxane (Zinecard®) ; docetaxel (Taxotere®) ; doxorubicin (Adriamycin PFS®) ; doxorubicin (Adriamycin®, Rubex®) ; doxorubicin (Adriamycin PFS Injection®); doxorubicin liposomal (Doxil®) ; dromostanolone propionate (dromostanolone®) ; dromostanolone propionate (masterone injection®); Elliott's B Solution (Elliott's B Solution®); epirubicin (Ellence®) ; Epoetin alfa (epogen®) ; erlotinib (Tarceva®) ; estramustine (Emcyt®) ; etoposide phosphate (Etopophos®) ; etoposide, VP-16 (Vepesid®) ; exemestane (Aromasin®) ; Filgrastim (Neupogen®) ; floxuridine (intraarterial) (FUDR®) ; fludarabine (Fludara®) ; fluorouracil, 5-FU (Adrucil®) ; fulvestrant (Faslodex®) ; gefitinib (Iressa®) ; gemcitabine (Gemzar®) ; gemtuzumab ozogamicin (Mylotarg®) ; goserelin acetate (Zoladex Implant®) ; goserelin acetate (Zoladex®) ; histrelin acetate (Histrelin implant®) ; hydroxyurea (Hydrea®) ; Ibritumomab Tiuxetan (Zevalin®) ; idarubicin (Idamycin®) ; ifosfamide (IFEX®) ; imatinib mesylate (Gleevec®) ; interferon alfa 2a (Roferon A®) ; Interferon alfa- 2b (Intron A®) ; irinotecan (Camptosar®) ; lenalidomide (Revlimid®) ; letrozole (Femara®) ; leucovorin (Wellcovorin®, Leucovorin®) ; Leuprolide Acetate (Eligard®) ; levamisole
(Ergamisol®) ; lomustine, CCNU (CeeBU®) ; meclorethamine, nitrogen mustard (Mustargen®) ; megestrol acetate (Megace®) ; melphalan, L-PAM (Alkeran®) ; mercaptopurine , 6-MP
(Purinethol®) ; mesna (Mesnex®) ; mesna (Mesnex tabs®) ; methotrexate (Methotrexate®) ; methoxsalen (Uvadex®) ; mitomycin C (Mutamycin®) ; mitotane (Lysodren®) ; mitoxantrone
(Novantrone®) ; nandrolone phenpropionate (Durabolin-50®) ; nelarabine (Arranon®) ; Nofetumomab (Verluma®) ; Oprelvekin
(Neumega®) ; oxaliplatin (Eloxatin®) ; paclitaxel (Paxene®) ; paclitaxel (Taxol®) ; paclitaxel protein-bound particles
(Abraxane®) ; palifermin (Kepivance®) ; pamidronate (Aredia®) ; pegademase (Adagen (Pegademase Bovine)®); pegaspargase
(Oncaspar®) ; Pegfilgrastim (Neulasta®) ; pemetrexed disodium
(Alimta®) ; pentostatin (Nipent®) ; pipobroman (Vercyte®) ; plicamycin, mithramycin (Mithracin®) ; porfimer sodium
(Photofrin®) ; procarbazine (Matulane®) ; quinacrine
(Atabrine®) ; Rasburicase (Elitek®) ; Rituximab (Rituxan®) ; sargramostim (Leukine®) ; Sargramostim (Prokine®) ; sorafenib
(Nexavar®) ; streptozocin (Zanosar®) ; sunitinib maleate
(Sutent®) ; talc (Sclerosol®) ; tamoxifen (Nolvadex®) ; temozolomide (Temodar®) ; teniposide, VM-26 (Vumon®) ; testolactone (Teslac®) ; thioguanine, 6-TG (Thioguanine®) ; thiotepa (Thioplex®) ; topotecan (Hycamtin®) ; toremifene
(Fareston®) ; Tositumomab (Bexxar®) ; Tositumomab/I-131 tositumomab (Bexxar®) ; Trastuzumab (Herceptin®) ; tretinoin, ATRA (Vesanoid®) ; Uracil Mustard (Uracil Mustard Capsules®) ; valrubicin (VaIstar®) ; vinblastine (Velban®) ; vincristine
(Oncovin®) ; vinorelbine (Navelbine®) ; zoledronate (Zometa®) and vorinostat (Zolinza®) . [0083] For a comprehensive discussion of updated cancer therapies see, http://www.nci.nih.gov/, a list of the FDA approved oncology drugs at http://www.fda.gov/cder/cancer/druglistframe.htm, and The Merck Manual, Seventeenth Ed. 1999, the entire contents of which are hereby incorporated by reference.
[0084] Another embodiment provides a simultaneous, separate or sequential use of a combined preparation.
[0085] Those additional agents may be administered separately, as part of a multiple dosage regimen, from the kinase inhibitor-containing compound or composition. Alternatively, those agents may be part of a single dosage form, mixed together with the kinase inhibitor in a single composition.
[0086] In order that this invention be more fully understood, the following preparative and testing examples are set forth. These examples are for the purpose of illustration only and are not to be construed as limiting the scope of the invention in any way. All documents cited herein are hereby incorporated by reference.
EXAMPLES
[0087] As used herein, the term "Rt(tnin)" refers to the HPLC retention time, in minutes, associated with the compound. Unless otherwise indicated, the HPLC method utilized to obtain the reported retention time is as follows: Column: ACE C8 column, 4.6 x 150 mm
Gradient: 0-100% acetonitrile+methanol 60:40 (2OmM Tris phosphate)
Flow rate : 1.5 mL/minute Detection: 225 nm.
[0088] Mass spec, samples were analyzed on a MicroMass Quattro Micro mass spectrometer operated in single MS mode with electrospray ionization. Samples were introduced into the mass spectrometer using chromatography. Mobile phase for all mass spec, analyses consisted of 1OmM pH 7 ammonium acetate and a 1:1 acetonitrile-methanol mixture, column gradient conditions was 5%-100% acetonitrile-methanol over 3.5 mins gradient time and 5 mins run time on an ACE C8 3.0 x 75mm column. Flow rate was 1.2 ml/min.
[0089] 1H-NMR spectra were recorded at 400 MHz using a Bruker DPX 400 instrument. The following compounds of formula I were prepared and analyzed as follows.
Example 1
Figure imgf000031_0001
2 , 6-Difluoro -N- [4- (4 , 6-dichloro-pyrimidin-2 -ylsulfanyl) - phenyl] -benzamide :
[0090] A 250ml round bottom flask equipped with a condenser was charged with 4 , 6-dichloro-2-methanesulfonyl pyrimidine
(4.2g, 18. δmmol) , 2 , 6-difluoro-ltf- (4-mercapto-phenyl) -benzamide
(4.98g, 18.8mmol) and tert-butanol (75ml) under nitrogen. The reaction mixture was degassed thoroughly and then heated at 900C for 2h. The reaction mixture was allowed to cool to room temperature and concentrated in vacuo. The solid residue was taken up in ethyl acetate (50ml) and washed with saturated sodium bicarbonate solution and brine. The organic was dried over magnesium sulfate, filtered and concentrated until the product began to precipitate. The mixture was then cooled and aged for 12 hrs . The product was collected by filtration, washed with cold ethyl acetate and dried. This gave the title compound as an off-white solid (2.7g, 35%) . IH NMR (DMSO) 7.32 (2H, m) , 7.61 (3H, m) , 7.79 (IH, s), 7.82 (2H, d) , 10.9
(IH, s) . MS (ES+) : 412.19
Example 2
Figure imgf000032_0001
2, 6-Difluoro-JV-{4- [4-chloro-6- (5-methyl-2iϊ-pyrazol-3~ylamino) - pyrimidin-2-ylsulfanyl] -phenyl}-benzamide:
[0091] A 50ml round bottom flask was charged with 2,6- difluoro-JV- [4- (4, 6-dichloro-pyrimidin-2-ylsulfanyl) -phenyl] - benzamide (l.Og, 2.3mmol), 5-methyl-2H-pyrazol-3-ylamine
(250mg, 2.58mmol), sodium iodide (351mg, 2.34mmol), diisopropylethyl amine (333mg, 2.58mmol) and dimethylformamide
(5ml) under nitrogen. The reaction mixture was stirred at 900C for 18h, then allowed to cool to room temperature. The reaction mixture was diluted with ethyl acetate (25ml) , washed with saturated sodium bicarbonate solution and brine. The organic was dried over magnesium sulfate, filtered and concentrated in vacuo. The compound was purified by flash chromatography (75 to 80% ethyl acetate/petrol) to give the title compound (1.08g, 98%). IH NMR (DMSO) 2.00 (3H, s) , 5.25
(IH, brs) , 6.48 (IH, brs) , 7.30-7.97 (7H, m) , 10.28 (IH, s), 10.89 (IH, S) , 11.90 (IH, s) ; MS(ES+): 473.4.
Example 3
Figure imgf000032_0002
iV-{4- [4-Azetidin-l-yl-6- (5-methyl-2iJ-pyrazol-3-ylamino) - pyrimidin-2-ylsulfanyl] -phenyl}-2, 6-Difluoro -benzamide:
(Compound 1-2)
[0092] A 10ml round bottom flask was charged with 2,6- difluoro-iV- {4- [4-chloro-6- (5-methyl-2H-pyrazol-3-ylamino) - pyrimidin-2-ylsulfanyl] -phenyl} -benzamide (150mg, 0.31mmol), azetidine (35rag, 0.62mmol), diisopropyl ethylamine (80rag, 0.62mmol) and n-butanol (1.5ml). The reaction mixture was stirred at 800C for 4h, then cooled and concentrated in vacuo. The compound was purified by preparative HPLC (MeCN/water + 0.05% TFA 10/90 to 100/0 over 10 min) to give the title compound as the trifluoroacetic acid salt (54mg, 29%) . IH NMR
(DMSO) 2.05 (3H, s) , 2.25-2.36 (2H, m) , 3.70-3.98 (4H, m, masked), 5.31 (IH, s) , 5.52 (IH, brs) , 7.39 (2H, m) , 7.46-7.67
(3H, m) , 7.79 (2H, d) , 9.35 (IH, brs), 11.04 (IH, s) , 11.80
(IH, brs); MS (ES+):494.5.
[0093] Table 2 below depicts data for compounds made according to the method described in Scheme I and in Examples 1-3. Compound numbers correspond to those compounds depicted in Table 1.
Table 2
Figure imgf000033_0002
Example 4
Figure imgf000033_0001
2 -ChIoro-iV- [4- (4, 6-dichloro-pyrimidin-2-ylsulfanyl) -phenyl] - benzamide :
[0094] A 250 mL round bottom flask was charged with 4,6- dichloro-2-methanesulfonylpyrimidine (7.00 g, 26.6 mmol) , 2- chloro-2\r- (4-mercapto-phenyl) -benzamide (6.33 g, 27.9 mmol) and acetonitrile (100 mL) under nitrogen. Once the solid had dissolved, the reaction mixture was cooled to 00C and triethylamine (3.7 mL, 26.6 mmol) was added dropwise. The solution was stirred at 00C for 10 min and then allowed to warm to room temperature and stirred for 1 h. After this time, water (50 mL) was added and a white solid precipitated and the reaction mixture stirred for an additional 4 h. After this time, the reaction mixture was filtered and the solid washed with acetonitrile (2 x 10 mL) to give the title compound as a white solid (8.03 g, 74%). IH NMR (DMSO) 7.4- 7.6 (5H, m) , 7.7 (IH, s) , 7.80-7.85 (2H, d) , 10.9 (IH, s). MS (ES+) : 412
Example 5
Figure imgf000034_0001
2-Chloro-JV-{4- [4-chloro-6- (5-methyl-2H-pyrazol-3-ylamino) - pyrimidin-2-ylsulfanyl] -phenyl} -benzamide:
[0095] A 250 mL round bottom flask was charged with 2-chloro- N- [4- (4, 6-dichloro-pyrimidin-2-ylsulfanyl) -phenyl] -benzamide
(12.5 g, 30.4 mmol) , 5-methyl-2H-pyrazol~3-ylamine (3.55 g, 36.5 mmol), sodium iodide (4.56 g, 30.4 mmol), N, N- diisopropylethylamine (6.9 mL, 40.0 mmol) and N, N- dimethylformamide (125 mL) under nitrogen. The reaction mixture was stirred at 90°C for 5 h, then allowed to cool to room temperature. Water (600 mL) was added and the resulting suspension stirred at room temperature for 2 h and the solid collected by filtration and dried. The resulting white solid was triturated with hot ethyl acetate (50 mL) , filtered and washed with ethyl acetate (1 x 20 mL) to give the title compound as a white solid (11.76 g, 82 %) . IH NMR (DMSO) : 2.16 (3H, S), 5.30 (IH, s) , 6.48 (IH, s) , 7.49-7.62 (6H, m) , 7.89 (2H, m) , 10,28 (IH, s) , 10.84 (IH, s) , 11.93 (IH, s); MS
(ES+) : 471. Example 6
Figure imgf000035_0001
JW-{4- [4-Azetidin-l-yl-6- (5-methyl-2H~pyrazol-3-ylamino) - pyrimidin-2-ylsulfanyl] -phenyl} -2 -chloro-benzamide: (Compound 1-1)
[0096] A 500 mL round bottom flask was charged with 2-chloro- N- {4- [4-chloro-6- (5-methyl-2J:f-pyrazol-3-ylamino) -pyrimidin-2- ylsulfanyl] -phenyl} -benzamide (16.0 g, 34.0 mmol) , azetidine
(3.87 g, 68.0 mmol), JV, N-diisopropylethylamine (13.0 mL, 74.7 mmol) and n-butanol (250 mL) . The reaction mixture was stirred at 900C for 5 h. The reaction mixture was cooled and concentrated in vacuo. Diethyl "ether (200 mL) was added and a light brown solid precipitated. The solution was filtered and the solid recrystallized from ethanol to give the pure product as a white solid (9.42 g, 52%) IH NMR (DMSO): 2.04 (3H, s) , 2.32 (2H, m) , 3.87 (4H, m) , 5.39 (IH, s) , 5.66 (IH, br s) , 7.48-7.59 (6H, m) , 7.82 (2H, m) , 9.87 (IH, s) , 10.74 (IH, s) , 11.68 (IH, s) ; MS (ES+): 492.
[0097] Another method used to prepare example 6 is described below:
[0098] To a suspension of
Figure imgf000035_0002
{4- [4-chloro-6- (5-methyl- 2ff-pyrazol-3-ylamino) -pyrimidin-2-ylsulfanyl] -phenyl} - benzamide (169 g, 0.36 mol) in 2-propanol (1.3 L) azetidine
(100 g, 1.76 mol) was added portion wise. The reaction mixture was heated to 80-82 °C. After 24 hours, di-isopropylethylamine
(73.4 g, 0.57 mol) was added. The progress of the reaction was monitored by HPLC. The reaction mixture was concentrated under reduce pressure to dryness, azeotroped with methanol three times (3 x 650 mL) , stirred for 2 hours in methanol (1 L) at 400C, and cooled to 100C. The resulting off-white solid was filtered. The isolated material was slurried in refluxing acetonitrile for 3 hours, cooled to 20-250C, filtered and dried in a vacuum oven overnight. The material was slurried again in refluxing acetonitrile for 3 hours, cooled to 20- 250C, and filtered. The material was allowed to dry until it was a constant weight. The desired product was isolated as an off-white solid (154 g, 86%) .
[0099] Table 3 below depicts data for certain exemplary compounds made according to the method described in Scheme I and in Examples 4-6. Compound numbers correspond to those compounds depicted in Table 1.
Table 3
Figure imgf000036_0001
Figure imgf000037_0002
Example 7
Figure imgf000037_0001
2- (4-aminophenylthio) -6- (azetidin-1-yl) -JV- (3 -methyl -IH- pyrazol-5-yl) pyrimidin-4-amine.
[00100] tert-butyl 4- (4- (5-methyl-lH-pyrazol-3-ylamino) -6- (azetidin-1-yl) pyrimidin-2-ylthio) phenylcarbamate (prepared using method similar to that described for example 6) (2.53g, 5.6 mmol) was dissolved in 1:1 TFA-DCM (20 mL) and the resulting solution allowed to stand overnight at room temperature. The solution was concentrated in vacuo. The residue was taken up in EtOAc and washed with saturated aqueous sodium bicarbonate solution (x2) then brine and dried over sodium sulfate. The resulting tan solid (1.8g, 91%) [MS (ES+) 354] was used without further purification or characterization in the next step.
Example 8
Figure imgf000038_0001
N- (4- (4- (3-methyl-lH-pyrazol-5-ylamino) -6- (azetidin-1- yl) pyrirαidin-2 -ylthio) phenyl) -3-methylbenzamide (1-4) [00101] 2- (4-Aminophenylthio) -6- (azetidin-1-yl) -N- (3 -methyl- lH-pyrazol-5-yl) pyrimidin-4-amine (200mg, 0.57 mmol) was taken up in pyridine (2mL) and m-toluoyl chloride (0.187 mL, 1.42 mmol) was added dropwise at room temperature. After 15 minutes the reaction mixture was concentrated in vacuo and the residue taken up in methanol (3mL) . Sodium methoxide (25% w/w solution in MeOH, ImL) was added and the resulting cloudy solution stirred at room temperature for 15 minutes . The reaction mixture was purified directly by chromatography (silica, 5- 100% EtOAc-petrol gradient elution) to give the title compound (89mg, 33%) as a white solid. IH NMR: (400 MHz, DMSO) 1.99 (3H, brs) , 2.31 (2H, qn) , 2.42 (3H, s) , 3.88 (4H, t) , 5.39 (IH, brs), 5.67 (IH, vbrs) , 7.43-7.46 (2H, m) , 7.54 (2H, d) , 7.73-7.76 (2H, m) , 7.91 (2H, d) , 9.23 (IH, brs), 10.42 (IH, s) , 11.67 (IH, brs). ES+ 472.
[00102] Table 4 below depicts data for certain exemplary compounds made according to the method described in Scheme II and in Examples 7-8. Compound numbers correspond to those compounds depicted in Table 1. Table 4
Figure imgf000039_0001
[00103] The experimentals shown below describe the preparation of some of the compounds used in the examples described herein. Compound a
Figure imgf000040_0001
2-chloro-jW- (4-mercaptophenyl) benzamide
Figure imgf000040_0002
S-4- (2 -chlorobenzamido) phenyl 2-chlorobenzothioate [00104] Degassed EtOAc (3.2 L) was charged in a flask. The solvent was cooled to 00C under nitrogen. 4-aminobenzenethiol (435 g, 3.48 mol) was melted and added directly to the flask. Triethylamine (773 g, 7.65 mol) was added over 30 minutes forming a precipitate. Then, 2-chlorobenzoyl chloride (1340 g, 7.65 mol) was added neat keeping the temperature below 5°C. After complete addition, the mixture was heated to 200C for one hour. The slurry was filtered and the cake washed with EtOAc (780 mL) . The material was dried at 500C under vacuum with a nitrogen sweep until a constant weight was obtained. The compound was carried on to the next reaction without further purification.
Figure imgf000040_0003
2 -chloro-N- (4 -mercaptophenyl) benzamide [00105] S-4- (2 -chlorobenzamido) phenyl 2-chlorobenzothioate (305 g, 0.76 mol) , EtOAc (325 mL) , and water (S5 mL) were charged to a flask fitted with a reflux condenser. A solution of NaOH (3 eq. , 50% aq.) was added and the mixture heated to 7O0C for 30-40 minutes. EtOAc was removed by distillation at 100mm Hg and the mixture was cooled to 50C. The mixture was acidified with 6N HCl to pH 2. The solid was then collected by vacuum filtration and washed with water (390 mL) . The solid was taken up in CH2Cl2 (520 mL) and washed with saturated aqueous NaHCO3. The organic layer was dried over Na2SO4, filtered, and concentrated to give the desired material (174 g, 87%) .
Example 10: Aurora-2 (Aurora A) Inhibition Assay [00106] Compounds were screened for their ability to inhibit Aurora-2 using a standard coupled enzyme assay (Fox et al . , Protein Sci., (1998) 7, 2249). Assays were carried out in a mixture of 10OmM Hepes (pH7.5), 1OmM MgCl2, ImM DTT, 25mM NaCl, 2.5mM phosphoenolpyruvate , 300 μM NADH, 30 μg/ml pyruvate kinase and 10 μg/ml lactate dehydrogenase. Final substrate concentrations in the assay were 400μM ATP (Sigma Chemicals) and 570μM peptide (Kemptide, American Peptide, Sunnyvale, CA) . Assays were carried out at 30 0C and in the presence of 4OnM Aurora-2.
[00107] An assay stock buffer solution was prepared containing all of the reagents listed above, with the exception of Aurora-2 and the test compound of interest. 55 μl of the stock solution was placed in a 96 well plate followed by addition of 2 μl of DMSO stock containing serial dilutions of the test compound (typically starting from a final concentration of 7.5μM) . The plate was preincubated for 10 minutes at 30 "C and the reaction initiated by addition of 10 μl of Aurora-2. Initial reaction rates were determined with a Molecular Devices SpectraMax Plus plate reader over a 10 minute time course. IC50 and Ki data were calculated from non- linear regression analysis using the Prism software package (GraphPad Prism version 3.0cx for Macintosh, GraphPad Software, San Diego California, USA) .
Example 11: Aurora-1 (Aurora B) Inhibition Assay (radiometric) [00108] An assay buffer solution was prepared which consisted of 25 mM HEPES (pH 7.5), 10 mM MgCl2, 0.1% BSA and 10% glycerol. A 22 nM Aurora-B solution, also containing 1.7 mM DTT and 1.5 mM Kemptide (LRRASLG), was prepared in assay buffer. To 22 μL of the Aurora-B solution, in a 96-well plate, was added 2 μl of a compound stock solution in DMSO and the mixture allowed to equilibrate for 10 minutes at 250C. The enzyme reaction was initiated by the addition of 16 μl stock [γ-33P] -ATP solution (-20 nCi/μL) prepared in assay buffer, to a final assay concentration of 800 μM. The reaction was stopped after 3 hours by the addition of 16 μL 500 mM phosphoric acid and the levels of 33P incorporation into the peptide substrate were determined by the following method. [00109] A phosphocellulose 96-well plate (Millipore, Cat no. MAPHNOB50) was pre-treated with 100 μL of a 100 mM phosphoric acid prior to the addition of the enzyme reaction mixture (40 μL) . The solution was left to soak on to the phosphocellulose membrane for 30 minutes and the plate subsequently washed four times with 200 μL of a 100 mM phosphoric acid. To each well of the dry plate was added 30 μL of Optiphase "SuperMix' liquid scintillation cocktail (Perkin Elmer) prior to scintillation counting (1450 Microbeta Liquid Scintillation Counter, Wallac) . Levels of non-enzyme catalyzed background radioactivity were determined by adding 16 μL of the 500 mM phosphoric acid to control wells, containing all assay components (which acts to denature the enzyme) , prior to the addition of the [γ-33P] -ATP solution. Levels of enzyme catalyzed 33P incorporation were calculated by subtracting mean background counts from those measured at each inhibitor concentration. For each Ki determination 8 data points, typically covering the concentration range 0 - 10 μM compound, were obtained in duplicate (DMSO stocks were prepared from an initial compound stock of 10 mM with subsequent 1:2.5 serial dilutions) . Ki values were calculated from initial rate data by non-linear regression using the Prism software package (Prism 3.0, Graphpad Software, San Diego, CA) .
Example 12: Itk Inhibition Assay: Radioactivity-based Assay [00110] The compounds of the present invention were evaluated as inhibitors of human Itk kinase using a radioactivity-based assay.
[00111] Assays were carried out in a mixture of 20 mM MOPS (pH 7.0), 1OmM MgCl2, 0.1% BSA and ImM DTT. Final substrate concentrations in the assay were 7.5 μM [γ-33P]ATP (400μCi 33P ATP/ μmol ATP, Amersham Pharmacia Biotech / Sigma Chemicals) and 3μM peptide (SAM68 protein Δ332-443) . Assays were carried out at 25 0C. in the presence of 50 nM Itk. An assay stock buffer solution was prepared containing all of the reagents listed above, with the exception of ATP and the test compound of interest. 50 μL of the stock solution was placed in a 96 well plate followed by addition of 2μL of DMSO stock containing serial dilutions of the test compound (typically starting from a final concentration of 50μM with 2-fold serial dilutions) in duplicate (final DMSO concentration 2%) . The plate was pre-incubated for 10 minutes at 25°C and the reaction initiated by addition of 50μL [γ-33P]ATP (final concentration 7.5μM) .
[00112] The reaction was stopped after 10 minutes by the addition of lOOμL 0.2M phosphoric acid + 0.01% TWEEN 20. A multiscreen phosphocellulose filter 96-well plate (Millipore, Cat no. MAPHN0B50) was pretreated with lOOμL 0.2M phosphoric acid + 0.01% TWEEN 20 prior to the addition of 170μL of the stopped assay mixture. The plate was washed with 4 x 200μL 0.2M phosphoric acid + 0.01% TWEEN 20. After drying, 30μL Optiphase "SuperMix' liquid scintillation cocktail (Perkin ■ Elmer) was added to the well prior to scintillation counting
(1450 Microbeta Liquid Scintillation Counter, Wallac) .
[00113] Ki(app) data were calculated from non-linear regression analysis of the initial rate data using the Prism software package (GraphPad Prism version 3. Ocx for Macintosh, GraphPad Software, San Diego California, USA) .
Example 13 : JAK3 Inhibition Assay
[00114] Compounds were screened for their ability to inhibit JAK using the assay shown below. Reactions were carried out in a kinase buffer containing 100 mM HEPES (pH 7.4), 1 mM DTT, 10 mM MgCl2, 25 mM NaCl, and 0.01% BSA. Substrate concentrations in the assay were 5 μM ATP (200 uCi/μmole ATP) and 1 μM poly (GIu) 4Tyr. Reactions were carried out at 25 °C and 1 nM JAK3.
[00115] To each well of a 96 well polycarbonate plate was added 1.5 μl of a candidate JAK3 inhibitor along with 50 μl of kinase buffer containing 2 μM poly (GIu) 4Tyr and 10 μM ATP. This was then mixed and 50μl of kinase buffer containing 2 nM JAK3 enzyme was added to start the reaction. After 20 minutes at room temperature (25C) , the reaction was stopped with 50μl of 20% trichloroacetic acid (TCA) that also contained 0.4 mM ATP. The entire contents of each well were then transferred to a 96 well glass fiber filter plate using a TomTek Cell Harvester. After washing, 60 μl of scintillation fluid was added and 33P incorporation detected on a Perkin Elmer TopCount .
Example 14 : JAK2 Inhibition Assay
[00116] The assays are as described above in Example 33 except that JAK-2 enzyme was used, the final poly (GIu) 4Tyr concentration was 15 μM, and final ATP concentration was 12 μM.
Example 15: FLT-3 Inhibition Assay
[00117] Compounds were screened for their ability to inhibit FLT-3 activity using a radiometric filter-binding assay. This assay monitors the 33P incorporation into a substrate poly (GIu, Tyr) 4:1 (pE4Y) . Reactions were carried out in a solution containing 100 mM HEPES (pH 7.5), 10 mM MgCl2, 25 mM NaCl, 1 mM DTT, 0.01% BSA and 2.5% DMSO. Final substrate concentrations in the assay were 90 μM ATP and 0.5mg/ml pE4Y
(both from Sigma Chemicals, St Louis, MO) . The final concentration of a compound of the present invention was generally between 0.01 and 5 μM. Typically, a 12-point titration was conducted by preparing serial dilutions from 10 mM DMSO stock of test compound. Reactions were carried out at room temperature .
[00118] Two assay solutions were prepared. Solution 1 contains 100 mM HEPES (pH 7.5), 10 mM MgCl2, 25 mM NaCl, 1 mg/ml pE4Y and 180 mM ATP (containing 0.3mCi of [γ-33p]ATP for each reaction). Solution 2 contains 100 mM HEPES (pH 7.5), 10 mM MgCl2, 25 mM NaCl, 2 mM DTT, 0.02% BSA and 3 nM FLT-3. The assay was run on a 96 well plate by mixing 50μl each of Solution 1 and 2.5 ml of the compounds of the present invention. The reaction was initiated with Solution 2. After incubation for 20 minutes at room temperature, the reaction was stopped with 50μl of 20% TCA containing 0.4mM of ATP. All of the reaction volume was then transferred to a filter plate and washed with 5% TCA by a Harvester 9600 from TOMTEC
(Hamden, CT) . The amount of 33P incorporation into pE4y was analyzed by a Packard Top Count Microplate Scintillation Counter (Meriden, CT) . The data was fitted using Prism software to get an IC50 or Ki. Example 16: Microsomal Stability Assay
[00119] Microsomal stability was monitored by generation of depletion-time profiles in microsomes from a range of species (male CD-I mouse, male Sprague-Dawley rat, male Beagle dog, male Cynomolgus monkey and pooled mixed gender human) . Compound spiking solutions were made up by diluting down the compound stock solution in DMSO (typically 10 mM) to give a solution in acetonitrile (0.5 mM) . Compound (to give final concentration of 5 μM) was incubated with a final reaction mixture (1000 μL) consisting of liver microsome protein (1 mg/mL) and a β-nicotinamide adenine dinucleotide phosphate, reduced form (NADPH) -regenerating system (RGS) [consisting of 2 mM β-nicotinamide adenine dinucleotide phosphate (NADP) , 20.5 mM isocitric acid, 0.5 U of isocitrate dehydrogenase/mL, 30 mM magnesium chloride, and 0.1 M phosphate buffer (PB) pH 7.4] in the presence of 0.1 M PB (pH 7.4) .
[00120] The reaction was initiated by the addition (250 μL) of the pre-incubated RGS to the pre-incubated microsome/VRT/PB mixture (pre-incubation in both instances was for 10 minutes at 37 0C) . Samples were incubated within Eppendorf vials (1.5 ml) on a heater shaker (DPC Micromix 5 (settings; form 20, amplitude 4) modified to be heated, to 37 0C, by two plate heaters fixed to the deck and controlled by a Packard Manual Heater) attached to a Multiprobe II HT Ex automated liquid handler. The liquid handler was programmed (WinPREP software) to sample the microsomal incubation mixture after 0, 2, 10, 30 and 60 minutes of incubation and transfer an aliquot (100 μL) to a stop block (96-well block) containing 100 μL of chilled methanol. The % organic in the stop mixture was optimized for analysis by addition of appropriate volumes of aqueous/organic (typically 100 μL of 50:50 methanol: water) . [00121] Prior to analysis the stop block was placed on a shaker (DPC Micromix 5; 10 min, form 20, amplitude 5) to precipitate out proteins . The block was then centrifuged (Jouan GR412; 2000 rpm, 15 min, 4 0C). A sample aliquot (200 μL) was then transferred to an analysis block and the block was centrifuged again (Jouan GR412; 2000 rpm, 5 min, 4 0C) prior to being sent for analysis. Depletion profiles were determined by monitoring the disappearance of VRT by liquid chromatography-tandem mass spectrometry (LC-MS/MS) . Samples were injected (20 μL; Agilent 1100 liquid chromatographic system equipped with autosampler) onto an analytical column. Mobile phase consisted of Water + 0.05% (v/v) formic acid (A) and methanol + 0.05% (v/v) formic acid (B) .
[00122] Running a gradient method optimized for the compound of interest carried out the compound elution from analytical column: The total run time was 6 minutes with a flow rate of 0.35 mL/min. The entire column effluent entered the electrospray ionization source (positive mode) of a Micromass Quattro LC tandem mass spectrometer between 0.5 and 5.9 min of the run. The mass spectrometry was optimized for the compound of interest. All incubations were conducted in duplicate and results were expressed as % parent remaining at either 30 minutes or 60 minutes relative to 0 minutes sample.
Example 17: Analysis of cell proliferation and viability [00123] Compounds were screened for their ability to inhibit cell proliferation and their effects on cell viability using
Colo205 cells obtained from ECACC and using the assay shown below. [00124] Colo205 cells were seeded in 96 well plates and serially diluted compound was added to the wells in duplicate.
Control groups included untreated cells, the compound diluent (0.1% DMSO alone) and culture medium without cells. The cells were then incubated for 72 or 96 hrs at 37C in an atmosphere of 5% CO2/95% humidity.
[00125] To measure proliferation, 3 h prior to the end of the experiment 0.5 μCi of 3H thymidine was added to each well . Cells were then harvested and the incorporated radioactivity counted on a Wallac microplate beta-counter. Cell viability was assessed using Promega CellTiter 96AQ to measure MTS conversion. Dose response curves were calculated using either Prism 3.0 (GraphPad) or SoftMax Pro 4.3.1 LS (Molecular Devices) software.
Example 18 : AbI Kinase Activity Inhibition Assay and Determination of the Inhibition Constant Ki
[00126] Compounds were screened for their ability to inhibit N-terminally truncated (Δ 27) AbI kinase activity using a standard coupled enzyme system (Fox et al . , Protein Sci . , 7, pp. 2249 (1998)) . Reactions were carried out in a solution containing 100 mM HEPES (pH 7.5), 10 mM MgCl2, 25 mM NaCl, 300 μM NADH, 1 mM DTT and 3% DMSO. Final substrate concentrations in the assay were 110 μM ATP (Sigma Chemicals, St Louis, MO) and 70 μM peptide (EAIYAAPFAKKK, American Peptide, Sunnyvale, CA) . Reactions were carried out at 30 0C and 21 nM AbI kinase. Final concentrations of the components of the coupled enzyme system were 2.5 mM phosphoenolpyruvate, 200 μM NADH, 60 μg/ml pyruvate kinase and 20 μg/ml lactate dehydrogenase. [00127] An assay stock buffer solution was prepared containing all of the reagents listed above with the exception of ATP and the test compound of interest. The assay stock buffer solution (60 μl) was incubated in a 96 well plate with 2 μl of the test compound of interest at final concentrations typically spanning 0.002 μM to 30 μM at 30 0C for 10 min. Typically, a 12 point titration was prepared by serial dilutions (from 1 mM compound stocks) with DMSO of the test compounds in daughter plates . The reaction was initiated by the addition of 5 μl of ATP (final concentration 110 μM) . Rates of reaction were obtained using a Molecular Devices Spectramax plate reader (Sunnyvale, CA) over 10 rain at 30 0C. The Ki values were determined from the residual rate data as a function of inhibitor concentration using nonlinear regression
(Prism 3.0, Graphpad Software, San Diego, CA) .
[00128] Compound 14 was found to inhibit AbI kinase
Example 19: Mutant AbI Kinase (T315I) Activity Inhibition Assay and Determination of the Inhibition Constant IC50
[00129] Compounds were screened for their ability to inhibit the T315I mutant form of human AbI at Upstate Cell Signaling Solutions (Dundee, UK) . In a final reaction volume of 25 μl, the T315I mutant of human AbI (5-10 mU) was incubated with 8 mM MOPS pH 7.0, 0.2 mM EDTA, 50 μM EAIYAAPFAKKK, 10 mM Mg Acetate, [γ-33P-ATP] (specific activity approx. 500 cpm/pmol, 1OmM final assay concentration) and the test compound of interest at final concentrations over the range 0-4μnM. The reaction was initiated by the addition of the MgATP mix. After incubation for 40 minutes at room temperature, the reaction was stopped by the addition of 5 μl of , a 3% phosphoric acid solution. 10 μl of the reaction was then spotted onto a P30 filtermat and washed three times for 5 minutes in 75 mM phosphoric acid and once in methanol prior to drying and scintillation counting. Inhibition IC50 values were determined from non-linear regression analysis of the residual enzyme activities as a function of inhibitor concentration
(Prism 3.0, Graphpad Software, San Diego, CA) .
Example 20: Arg (AbI-2) , FGFRl, MELK, MLKl, MuSK, Ret, and TrkA Inhibition Assay
[00130] Compound 1-1 was screened for its ability to inhibit Arg (Abl-2) , FGFRl, MELK, MLKl, MuSK, Ret, and TrkA using screening methods known to one of ordinary skill in the art. All of the above enzymes were screened with ATP concentrations at or close to the Km for ATP.
[00131] Table 5 below depicts the Ki values obtained from Example 20:
Table 5
Figure imgf000050_0001
[00132] Table 6 below depicts data from Examples 10-11 and 16 described above.
Table 6
Figure imgf000050_0002
[00133] Table 7 below depicts data from Examples 13-15 and
18-19.
Table 7
Figure imgf000051_0001
[00134] While we have described a number of embodiments of this invention, it is apparent that our basic examples may be altered to provide other embodiments that utilize or encompass the compounds, methods, and processes of this invention. Therefore, it will be appreciated that the scope -of this invention is to be defined by the appended claims .

Claims

We claim :
1 . A compound of formula I :
Figure imgf000052_0001
or a pharmaceutically acceptable salt thereof, wherein: R2 is Cα_3alkyl or cyclopropyl;
R9 is halo, Ci_3alkyl, -0- (d-3alkyl) , -S- (Ci_3alkyl) , or CF3; and p is 1-2.
2. The compound of claim 1, wherein R2 is methyl.
3. The compound of claim 2, wherein p is 1.
4. The compound of claim 2, wherein R9 is substituted in the ortho position.
5. The compound claim 3, wherein R9 is substituted in the ortho position.
6. The compound of claim 5, wherein R9 is F, Cl, or CF3.
The compound of claim 1 selected from the following:
Figure imgf000053_0001
1-2
1-1
Figure imgf000053_0002
1-3 1-4
Figure imgf000053_0003
1-5
1-6
Figure imgf000053_0004
1-7
I-i
Figure imgf000054_0001
Figure imgf000055_0001
Figure imgf000056_0001
1-23 1- 24
8. A composition comprising a compound of any one of claims 1-7 and a pharmaceutically acceptable carrier, adjuvant, or vehicle .
9. A method of inhibiting Aurora protein kinase activity in a biological sample comprising contacting said biological sample with a compound of any one of claims 1-7.
10. A method of treating a proliferative disorder in a patient comprising the step of administering to said patient a compound of any one of claims 1-7.
11. The method according to claim 10, wherein said proliferative disorder is selected from melanoma, myeloma, leukemia, lymphoma, neuroblastoma, or a cancer selected from colon, breast, gastric, ovarian, cervical, lung, central nervous system (CNS), renal, prostate, bladder, pancreatic, brain (gliomas) , head and neck, kidney, liver, melanoma, sarcoma, or thyroid cancer in a patient in need thereof wherein said method comprises administering to said patient a compound of any one of claims 1-7.
12. A method of treating cancer in a subject in need thereof, comprising the sequential or co-administration of a compound of any one of claims 1-7 or a pharmaceutically acceptable salt thereof, and another therapeutic agent.
13. The method according to claim 12, wherein said therapeutic agent is selected from taxanes, inhibitors of bcr- abl, inhibitors of EGFR, DNA damaging agents, and antimetabolites .
14. The method according to claim 12, wherein said therapeutic agent is selected from Paclitaxel, Gleevec, dasatinib, nilotinib, Tarceva, Iressa, cisplatin, oxaliplatin, carboplatin, anthracyclines, AraC and 5 -FU.
15. The method according to claim 12, wherein said therapeutic agent is selected from camptothecin, doxorubicin, idarubicin, Cisplatin, taxol, taxotere, vincristine, tarceva, the MEK inhibitor, U0126, a KSP inhibitor, vorinostat, Gleevec, dasatinib, and nilotinib.
PCT/US2006/042993 2005-11-03 2006-11-03 Aminopyrimidines useful as kinase inhibitors WO2007056163A2 (en)

Priority Applications (10)

Application Number Priority Date Filing Date Title
AU2006311830A AU2006311830B2 (en) 2005-11-03 2006-11-03 Aminopyrimidines useful as kinase inhibitors
BRPI0619708-6A BRPI0619708A2 (en) 2005-11-03 2006-11-03 compound, composition, method for inhibiting aurora kinase activity in a biological sample, method for treating proliferative disorder and method for treating cancer
MX2008005717A MX2008005717A (en) 2005-11-03 2006-11-03 Aminopyrimidines useful as kinase inhibitors.
JP2008539070A JP5249770B2 (en) 2005-11-03 2006-11-03 Aminopyridines useful as kinase inhibitors
CA002627808A CA2627808A1 (en) 2005-11-03 2006-11-03 Aminopyrimidines useful as kinase inhibitors
CN2006800444148A CN101316843B (en) 2005-11-03 2006-11-03 Aminopyrimidines useful as kinase inhibitors
EP06827465.3A EP1951715B1 (en) 2005-11-03 2006-11-03 Aminopyrimidines useful as kinase inhibitors
ES06827465T ES2435081T3 (en) 2005-11-03 2006-11-03 Aminopyrimidines useful as kinase inhibitors
IL191044A IL191044A0 (en) 2005-11-03 2008-04-27 Aminopyrimidine derivatives and pharmaceutical compositions containing the same
NO20082517A NO20082517L (en) 2005-11-03 2008-05-30 Aminopyrimidines useful as kinase inhibitors

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US73295105P 2005-11-03 2005-11-03
US60/732,951 2005-11-03
US73355705P 2005-11-04 2005-11-04
US60/733,557 2005-11-04

Publications (2)

Publication Number Publication Date
WO2007056163A2 true WO2007056163A2 (en) 2007-05-18
WO2007056163A3 WO2007056163A3 (en) 2008-03-27

Family

ID=38023840

Family Applications (3)

Application Number Title Priority Date Filing Date
PCT/US2006/042994 WO2007056164A2 (en) 2005-11-03 2006-11-03 Aminopyrimidines useful as kinase inhibitors
PCT/US2006/042993 WO2007056163A2 (en) 2005-11-03 2006-11-03 Aminopyrimidines useful as kinase inhibitors
PCT/US2006/043096 WO2007056221A2 (en) 2005-11-03 2006-11-03 Aminopyrimidines useful as kinase inhibitors

Family Applications Before (1)

Application Number Title Priority Date Filing Date
PCT/US2006/042994 WO2007056164A2 (en) 2005-11-03 2006-11-03 Aminopyrimidines useful as kinase inhibitors

Family Applications After (1)

Application Number Title Priority Date Filing Date
PCT/US2006/043096 WO2007056221A2 (en) 2005-11-03 2006-11-03 Aminopyrimidines useful as kinase inhibitors

Country Status (18)

Country Link
US (7) US7767672B2 (en)
EP (3) EP1948193B1 (en)
JP (5) JP5328361B2 (en)
KR (3) KR20080067693A (en)
CN (3) CN103145702A (en)
AR (3) AR057874A1 (en)
AU (3) AU2006311831C1 (en)
BR (3) BRPI0619706A2 (en)
CA (3) CA2627857A1 (en)
ES (2) ES2435081T3 (en)
IL (3) IL191043A0 (en)
MX (3) MX2008005814A (en)
NO (3) NO20082514L (en)
NZ (4) NZ594385A (en)
RU (5) RU2008122070A (en)
SG (2) SG166827A1 (en)
TW (3) TW200736249A (en)
WO (3) WO2007056164A2 (en)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008137621A1 (en) * 2007-05-02 2008-11-13 Vertex Pharmaceuticals Incorporated Aminopyrimidines useful as kinase inhibitors
WO2008147626A2 (en) * 2007-05-24 2008-12-04 Vertex Pharmaceuticals Incorporated Thiazoles and pyrazoles useful as kinase inhibitors
US8410133B2 (en) 2007-03-09 2013-04-02 Vertex Pharmaceuticals Incorporated Aminopyridines useful as inhibitors of protein kinases
US8524720B2 (en) 2000-09-15 2013-09-03 Vertex Pharmaceuticals Incorporated Substituted N-(pyrazol-5-yl)-pyrrolo[3,2-D]pyrimidin-4-amine useful as protein kinase inhibitors
US8557833B2 (en) 2005-11-03 2013-10-15 Vertex Pharmaceuticals Incorporated Aminopyrimidines useful as kinase inhibitors
US8653088B2 (en) 2003-02-06 2014-02-18 Vertex Pharmaceuticals Incorporated Compositions useful as inhibitors of protein kinases
US8664219B2 (en) 2007-03-09 2014-03-04 Vertex Pharmaceuticals Incorporated Aminopyrimidines useful as inhibitors of protein kinases
US8785444B2 (en) 2007-05-02 2014-07-22 Vertex Pharmaceuticals Incorporated Thiazoles and pyrazoles useful as kinase inhibitors
JP2018104473A (en) * 2008-03-17 2018-07-05 ザ スクリプス リサーチ インスティテュート Combined chemical and genetic approaches for generation of induced pluripotent stem cells

Families Citing this family (63)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2001291013A1 (en) 2000-09-15 2002-03-26 Vertex Pharmaceuticals Incorporated Pyrazole compounds useful as protein kinase inhibitors
SE0104140D0 (en) * 2001-12-07 2001-12-07 Astrazeneca Ab Novel Compounds
SI1853588T1 (en) * 2005-02-16 2008-10-31 Astrazeneca Ab Chemical compounds
MX2007014328A (en) * 2005-05-16 2008-02-12 Astrazeneca Ab Chemical compounds.
WO2007048088A2 (en) * 2005-10-18 2007-04-26 Janssen Pharmaceutica N.V. Method of inhibiting flt3 kinase
EP2388259A1 (en) * 2005-10-28 2011-11-23 AstraZeneca AB 4- (3-aminopyrazole) pyrimidine derivatives for use as tyrosine kinase inhibitors in the treatment of cancer
CN101360740A (en) * 2005-11-16 2009-02-04 沃泰克斯药物股份有限公司 Aminopyrimidines useful as kinase inhibitors
WO2007124321A1 (en) * 2006-04-20 2007-11-01 Janssen Pharmaceutica N.V. Inhibitors of c-fms kinase
US8697716B2 (en) * 2006-04-20 2014-04-15 Janssen Pharmaceutica Nv Method of inhibiting C-KIT kinase
DE602007004750D1 (en) 2006-11-02 2010-03-25 Vertex Pharma AS INHIBITORS OF PROTEIN KINASES SUITABLE AMINOPYRIDINES AND AMINOPYRIMIDINES
ES2347721T3 (en) 2006-12-19 2010-11-03 Vertex Pharmaceuticals, Inc. USEFUL AMINOPIRIMIDINS AS PROTEIN QUINASE INHIBITORS.
NZ579485A (en) * 2007-03-09 2012-02-24 Vertex Pharma Aminopyrimidines useful as inhibitors of protein kinases
WO2008128009A2 (en) 2007-04-13 2008-10-23 Vertex Pharmaceuticals Incorporated Aminopyrimidines useful as kinase inhibitors
US20100204246A1 (en) * 2007-04-18 2010-08-12 Astrazeneca Ab 5-aminopyrazol-3-yl-3h-imidazo (4,5-b) pyridine derivatives and their use for the treatment of cancer
MX2009011811A (en) * 2007-05-02 2010-01-14 Vertex Pharma Aminopyrimidines useful as kinase inhibitors.
US20100137398A1 (en) * 2007-05-04 2010-06-03 Novartis Ag Use of hdac inhibitors for the treatment of gastrointestinal cancers
UA99459C2 (en) * 2007-05-04 2012-08-27 Астразенека Аб 9-(pyrazol-3-yl)- 9h-purine-2-amine and 3-(pyraz0l-3-yl)-3h-imidazo[4,5-b]pyridin-5-amine derivatives and their use for the treatment of cancer
JP2010529193A (en) * 2007-06-11 2010-08-26 ミイカナ セラピューティクス インコーポレイテッド Substituted pyrazole compounds
JP5553751B2 (en) 2007-07-31 2014-07-16 バーテックス ファーマシューティカルズ インコーポレイテッド Process for preparing 5-fluoro-1H-pyrazolo [3,4-b] pyridin-3-amine and its derivatives
AU2008287339A1 (en) * 2007-08-15 2009-02-19 Vertex Pharmaceuticals Incorporated 4-(9-(3, 3-difluorocyclopentyl) -5, 7, 7-trimethyl-6-oxo-6, 7, 8, 9-tetrahydro-5H-pyrimido [4, 5-b][1, 4] diazepin-2-ylamino)-3-methoxybenzamide Derivatives as Inhibitors of the Human Protein Kinases PLK1 to PLK4 for the Treatment of Proliferative Diseases
BRPI0816881A2 (en) 2007-09-21 2015-03-17 Array Biopharma Inc Compound, pharmaceutical composition, use of a compound, and methods for treating diseases or disorders and for preparing a compound.
WO2009086012A1 (en) * 2007-12-20 2009-07-09 Curis, Inc. Aurora inhibitors containing a zinc binding moiety
WO2009114703A2 (en) * 2008-03-12 2009-09-17 Fox Chase Cancer Center Combination therapy for the treatment of cancer
EP2265601B1 (en) * 2008-03-13 2012-02-01 Bristol-Myers Squibb Company Pyridazine derivatives as factor xia inhibitors
CN102119157A (en) * 2008-06-11 2011-07-06 阿斯利康(瑞典)有限公司 Tricyclic 2,4-diamino-L,3,5-triazine derivatives useful for the treatment of cancer and myeloproliferative disorders
US20100185419A1 (en) * 2008-09-05 2010-07-22 Avila Therapeutics, Inc. Algorithm for designing irreversible inhibitors
MX2011003447A (en) * 2008-09-30 2011-07-29 Astrazeneca Ab Heterocyclic jak kinase inhibitors.
KR101741168B1 (en) 2008-12-22 2017-05-29 밀레니엄 파머슈티컬스 인코퍼레이티드 Combination of aurora kinase inhibitors and anti-cd20 antibodies
KR20120016676A (en) * 2009-06-09 2012-02-24 아브락시스 바이오사이언스, 엘엘씨 Ureidophenyl substituted triazine derivatives and their therapeutical applications
US9556426B2 (en) 2009-09-16 2017-01-31 Celgene Avilomics Research, Inc. Protein kinase conjugates and inhibitors
WO2011082285A1 (en) 2009-12-30 2011-07-07 Avila Therapeutics, Inc. Ligand-directed covalent modification of protein
CA2803446C (en) 2010-07-02 2019-06-04 University Health Network Use of plk4 antagonists for treating pten mutant diseases
DK2597955T3 (en) * 2010-07-30 2016-03-14 Onco Therapy Science Inc QUINOLINE DERIVATIVES AND MILK INHIBITORS CONTAINING THESE
WO2012112674A2 (en) 2011-02-15 2012-08-23 The Johns Hopkins University Compounds and methods of use thereof for treating neurodegenerative disorders
BR122014026094B1 (en) 2011-07-27 2020-10-13 Astrazeneca Ab compounds of formula ii, iii, iv and vi
CN104024248A (en) 2011-08-25 2014-09-03 霍夫曼-拉罗奇有限公司 Serine/threonine pak1 inhibitors
US9221798B2 (en) * 2011-09-05 2015-12-29 Zhejian Hisun Pharmaceutical Co., Ltd. 4-substituted-(3-substituted-1H-pyrazole-5-amino)-pyrimidine derivatives having activity of inhibiting protein kinase and use thereof
CA3131037A1 (en) 2011-11-30 2013-06-06 Emory University Antiviral jak inhibitors useful in treating or preventing retroviral and other viral infections
US8815877B2 (en) 2011-12-22 2014-08-26 Genentech, Inc. Serine/threonine kinase inhibitors
CA2867469A1 (en) 2012-03-16 2013-09-19 Axikin Pharmaceuticals, Inc. 3,5-diaminopyrazole kinase inhibitors
NZ731797A (en) 2012-04-24 2018-08-31 Vertex Pharma Dna-pk inhibitors
US20130310340A1 (en) 2012-05-16 2013-11-21 Rigel Pharmaceuticals, Inc. Method of treating muscular degradation
US9539259B2 (en) 2012-05-23 2017-01-10 The Johns Hopkins University Compounds and methods of use thereof for treating neurodegenerative disorders
AU2013280644B2 (en) 2012-06-26 2018-08-02 Jeffrey A. BACHA Methods for treating tyrosine-kinase-inhibitor-resistant malignancies in patients with genetic polymorphisms or AHI1 dysregulations or mutations employing dianhydrogalactitol, diacetyldianhydrogalactitol, dibromodulcitol, or analogs or derivatives thereof
CA2890018A1 (en) 2012-11-05 2014-05-08 Nant Holdings Ip, Llc Substituted indol-5-ol derivatives and their therapeutical applications
ES2900061T3 (en) 2013-03-12 2022-03-15 Vertex Pharma DNA-PK inhibitors
CN105377261B (en) 2013-03-15 2017-12-15 南特生物科学公司 The substituted amphyl of indoles 5 and their treatment use
AU2014251038A1 (en) 2013-04-08 2015-11-26 Dennis M. Brown Therapeutic benefit of suboptimally administered chemical compounds
NZ631142A (en) 2013-09-18 2016-03-31 Axikin Pharmaceuticals Inc Pharmaceutically acceptable salts of 3,5-diaminopyrazole kinase inhibitors
DK3424920T3 (en) 2013-10-17 2020-06-08 Vertex Pharma CO CRYSTALS OF (S) -N-METHYL-8- (1 - ((2'-METHYL- [4,5'-BIPYRIMIDIN] -6-YL) AMINO) PROPAN-2-YL) QUINOLIN-4-CARBOXAMIDE AND DEUTERATED DERIVATIVES THEREOF AS DNA-PK INHIBITORS
WO2015085289A1 (en) 2013-12-06 2015-06-11 Millennium Pharmaceuticals, Inc. Combination of aurora kinase inhibitors and anti-cd30 antibodies
HUE049801T2 (en) 2014-12-23 2020-10-28 Sma Therapeutics Inc 3,5-diaminopyrazole kinase inhibitors
US10118904B2 (en) 2015-06-05 2018-11-06 Vertex Pharmaceuticals Incorporated Triazoles for the treatment of Demyelinating Diseases
US10722484B2 (en) 2016-03-09 2020-07-28 K-Gen, Inc. Methods of cancer treatment
AU2017335648B2 (en) 2016-09-27 2022-02-17 Vertex Pharmaceuticals Incorporated Method for treating cancer using a combination of DNA-damaging agents and DNA-PK inhibitors
CA3045951A1 (en) * 2016-12-14 2018-06-21 Intervet International B.V. Aminopyrazoles as selective janus kinase inhibitors
PL3562821T3 (en) 2016-12-28 2021-06-14 Minoryx Therapeutics S.L. Isoquinoline compounds, methods for their preparation, and therapeutic uses thereof in conditions associated with the alteration of the activity of beta galactosidase
CN106841420A (en) * 2016-12-30 2017-06-13 广州中大南沙科技创新产业园有限公司 A kind of method that LC MS/MS determine Ly 7u concentration in rat plasma
US11874276B2 (en) 2018-04-05 2024-01-16 Dana-Farber Cancer Institute, Inc. STING levels as a biomarker for cancer immunotherapy
CN110132795A (en) * 2019-05-20 2019-08-16 华核(天津)新技术开发有限公司 Radioactive decontamination Efficiency test method
WO2021041532A1 (en) 2019-08-26 2021-03-04 Dana-Farber Cancer Institute, Inc. Use of heparin to promote type 1 interferon signaling
WO2021178485A1 (en) * 2020-03-06 2021-09-10 National Health Research Institutes Pyrimidine compounds and their pharmaceutical uses
WO2021222637A1 (en) 2020-04-30 2021-11-04 United States Government As Represented By The Department Of Veterans Affairs Krüppel-like factor 15 (klf15) small molecule agonists in kidney disease

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004000833A1 (en) 2002-06-20 2003-12-31 Vertex Pharmaceuticals Incorporated Processes for preparing substituted pyrimidines and pyrimidine derivatives as inhibitors of protein kinase

Family Cites Families (104)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3133081A (en) * 1964-05-12 J-aminoindazole derivatives
US3935183A (en) * 1970-01-26 1976-01-27 Imperial Chemical Industries Limited Indazole-azo phenyl compounds
BE754242A (en) * 1970-07-15 1971-02-01 Geigy Ag J R DIAMINO-S-TRIAZINES AND DINITRO-S-TRIAZINES
US3998951A (en) * 1974-03-13 1976-12-21 Fmc Corporation Substituted 2-arylquinazolines as fungicides
DE2458965C3 (en) 1974-12-13 1979-10-11 Bayer Ag, 5090 Leverkusen 3-Amino-indazole-N-carboxylic acid derivatives, process for their preparation and pharmaceuticals containing them
MA18829A1 (en) 1979-05-18 1980-12-31 Ciba Geigy Ag PYRIMIDINE DERIVATIVES, METHODS FOR THEIR PREPARATION, PHARMACEUTICAL COMPOSITIONS CONTAINING THESE COMPOUNDS AND THEIR THERAPEUTIC USE
DOP1981004033A (en) 1980-12-23 1990-12-29 Ciba Geigy Ag PROCEDURE TO PROTECT CROP PLANTS FROM PHYTOTOXIC ACTION OF HERBICIDES.
SE8102194L (en) 1981-04-06 1982-10-07 Pharmacia Ab THERAPEUTIC ACTIVE ORGANIC ASSOCIATION AND PHARMACEUTICAL PREPARATION INCLUDING THIS
SE8102193L (en) 1981-04-06 1982-10-07 Pharmacia Ab THERAPEUTIC ACTIVE ORGANIC ASSOCIATION AND ITS USE
JPS58124773A (en) * 1982-01-20 1983-07-25 Mitsui Toatsu Chem Inc 5-methylthiopyrimidine derivative, its preparation and fungicide for agricultural and horticultural purposes
EP0136976A3 (en) 1983-08-23 1985-05-15 Ciba-Geigy Ag Use of phenyl pyrimidines as plant regulators
DE3725638A1 (en) 1987-08-03 1989-02-16 Bayer Ag NEW ARYLOXY (OR THIO) AMINOPYRIMIDINE
US5710158A (en) 1991-05-10 1998-01-20 Rhone-Poulenc Rorer Pharmaceuticals Inc. Aryl and heteroaryl quinazoline compounds which inhibit EGF and/or PDGF receptor tyrosine kinase
US5597920A (en) 1992-04-30 1997-01-28 Neurogen Corporation Gabaa receptor subtypes and methods for screening drug compounds using imidazoquinoxalines and pyrrolopyrimidines to bind to gabaa receptor subtypes
JPH0665237A (en) 1992-05-07 1994-03-08 Nissan Chem Ind Ltd Substituted pyrazole derivative and germicide for agriculture and horticulture
JP3588116B2 (en) 1993-10-01 2004-11-10 ノバルティス アクチェンゲゼルシャフト Pharmacologically active pyrimidineamine derivatives and process for their production
ATE262902T1 (en) 1994-11-10 2004-04-15 Millennium Pharm Inc USE OF PYRAZOLE COMPOUNDS TO TREAT GLOMERULONEPHRITIS, CANCER, ATHEROSCLERosis OR RESTENOSIS
IL117659A (en) * 1995-04-13 2000-12-06 Dainippon Pharmaceutical Co Substituted 2-phenyl pyrimidino amino acetamide derivative process for preparing the same and a pharmaceutical composition containing same
AU726058B2 (en) 1995-09-01 2000-10-26 Signal Pharmaceuticals, Inc. Pyrimidine carboxylates and related compounds and methods for treating inflammatory conditions
US5935966A (en) 1995-09-01 1999-08-10 Signal Pharmaceuticals, Inc. Pyrimidine carboxylates and related compounds and methods for treating inflammatory conditions
GB9523675D0 (en) 1995-11-20 1996-01-24 Celltech Therapeutics Ltd Chemical compounds
US6716575B2 (en) * 1995-12-18 2004-04-06 Sugen, Inc. Diagnosis and treatment of AUR1 and/or AUR2 related disorders
PT912559E (en) 1996-07-13 2003-03-31 Glaxo Group Ltd HETEROCYCLIC COMPOUNDS FUSED AS PROTEIN INHIBITORS TYROSINE KINASE
JPH10130150A (en) 1996-09-05 1998-05-19 Dainippon Pharmaceut Co Ltd Medicine comprising acetic acid amide derivative
GB9619284D0 (en) 1996-09-16 1996-10-30 Celltech Therapeutics Ltd Chemical compounds
DE69732780T2 (en) 1996-10-02 2006-04-06 Novartis Ag PYRIMIDERIVATES AND METHOD FOR THE PRODUCTION THEREOF
WO1998016502A1 (en) 1996-10-11 1998-04-23 Warner-Lambert Company ASPARTATE ESTER INHIBITORS OF INTERLEUKIN-1β CONVERTING ENZYME
BR9814817A (en) 1997-10-10 2002-01-08 Cytovia Inc Dipeptide apoptosis inhibitors and their use
US6267952B1 (en) 1998-01-09 2001-07-31 Geltex Pharmaceuticals, Inc. Lipase inhibiting polymers
JP2000026421A (en) 1998-01-29 2000-01-25 Kumiai Chem Ind Co Ltd Diaryl sulfide derivative and pest controlling agent
DE69909756D1 (en) * 1998-02-17 2003-08-28 Tularik Inc ANTIVIRAL PYRIMIDINE DERIVATIVES
EP1076563B1 (en) 1998-03-16 2005-05-11 Cytovia, Inc. Dipeptide caspase inhibitors and the use thereof
TR200003513T2 (en) 1998-06-02 2001-06-21 Osi Pharmaceuticals, Inc. Pirolo [2,3d] Pyrimidine compositions and uses
WO1999065897A1 (en) 1998-06-19 1999-12-23 Chiron Corporation Inhibitors of glycogen synthase kinase 3
EP1105394A1 (en) 1998-08-21 2001-06-13 Du Pont Pharmaceuticals Company ISOXAZOLO 4,5-d]PYRIMIDINES AS CRF ANTAGONISTS
US6184226B1 (en) 1998-08-28 2001-02-06 Scios Inc. Quinazoline derivatives as inhibitors of P-38 α
ES2241324T3 (en) 1998-10-08 2005-10-16 Astrazeneca Ab DERIVATIVES OF QUINAZOLINA.
GB9828640D0 (en) 1998-12-23 1999-02-17 Smithkline Beecham Plc Novel method and compounds
GB9828511D0 (en) 1998-12-24 1999-02-17 Zeneca Ltd Chemical compounds
US6921748B1 (en) 1999-03-29 2005-07-26 Uutech Limited Analogs of gastric inhibitory polypeptide and their use for treatment of diabetes
GB9914258D0 (en) * 1999-06-18 1999-08-18 Celltech Therapeutics Ltd Chemical compounds
US20020065270A1 (en) 1999-12-28 2002-05-30 Moriarty Kevin Joseph N-heterocyclic inhibitors of TNF-alpha expression
AU782775B2 (en) * 2000-02-05 2005-08-25 Vertex Pharmaceuticals Incorporated Pyrazole compositions useful as inhibitors of ERK
CN1429222A (en) * 2000-02-17 2003-07-09 安姆根有限公司 Kinase inhibitors
GB0004887D0 (en) * 2000-03-01 2000-04-19 Astrazeneca Uk Ltd Chemical compounds
PT1268519E (en) 2000-04-03 2005-08-31 Vertex Pharma PROTEASE SERINE INHIBITORS, PARTICULARLY PROTEASE NS3 HEPATITIS C VIRUS
AU2001266505A1 (en) * 2000-06-28 2002-01-08 Astrazeneca Ab Substituted quinazoline derivatives and their use as inhibitors
JP4298289B2 (en) 2000-07-21 2009-07-15 シェーリング コーポレイション Novel peptides as NS3-serine protease inhibitors of hepatitis C virus
IL154016A0 (en) 2000-08-31 2003-07-31 Pfizer Prod Inc Pyrazole derivatives and their use as protein kinase inhibitors
US6660731B2 (en) 2000-09-15 2003-12-09 Vertex Pharmaceuticals Incorporated Pyrazole compounds useful as protein kinase inhibitors
US6610677B2 (en) 2000-09-15 2003-08-26 Vertex Pharmaceuticals Incorporated Pyrazole compounds useful as protein kinase inhibitors
US7473691B2 (en) * 2000-09-15 2009-01-06 Vertex Pharmaceuticals Incorporated Pyrazole compounds useful as protein kinase inhibitors
US6495582B1 (en) 2000-09-15 2002-12-17 Vertex Pharmaceuticals Incorporated Isoxazole compositions useful as inhibitors of ERK
US6613776B2 (en) * 2000-09-15 2003-09-02 Vertex Pharmaceuticals Incorporated Pyrazole compounds useful as protein kinase inhibitors
AU2001291013A1 (en) * 2000-09-15 2002-03-26 Vertex Pharmaceuticals Incorporated Pyrazole compounds useful as protein kinase inhibitors
US6641579B1 (en) * 2000-09-29 2003-11-04 Spectrasonics Imaging, Inc. Apparatus and method for ablating cardiac tissue
DE10061863A1 (en) 2000-12-12 2002-06-13 Basf Ag Preparation of triethylenediamine, useful for making pharmaceuticals and polymers, by reacting ethylenediamine over specific zeolite catalyst
US6716851B2 (en) 2000-12-12 2004-04-06 Cytovia, Inc. Substituted 2-aryl-4-arylaminopyrimidines and analogs as activators or caspases and inducers of apoptosis and the use thereof
AU2001297619B2 (en) 2000-12-21 2006-06-08 Vertex Pharmaceuticals Incorporated Pyrazole compounds useful as protein kinase inhibitors
MY130778A (en) * 2001-02-09 2007-07-31 Vertex Pharma Heterocyclic inhibitiors of erk2 and uses thereof
US6759983B2 (en) * 2001-03-28 2004-07-06 Strategic Analysis, Inc. Method and device for precise geolocation of low-power, broadband, amplitude-modulated signals
EP1373257B9 (en) 2001-03-29 2008-10-15 Vertex Pharmaceuticals Incorporated Inhibitors of c-jun n-terminal kinases (jnk) and other protein kinases
US6642227B2 (en) 2001-04-13 2003-11-04 Vertex Pharmaceuticals Incorporated Inhibitors of c-Jun N-terminal kinases (JNK) and other protein kinases
WO2002085909A1 (en) * 2001-04-20 2002-10-31 Vertex Pharmaceuticals Incorporated 9-deazaguanine derivatives as inhibitors of gsk-3
AU2002308748A1 (en) * 2001-05-16 2002-11-25 Vertex Pharmaceuticals Incorporated Heterocyclic substituted pyrazoles as inhibitors of src and other protein kinases
CA2450769A1 (en) * 2001-06-15 2002-12-27 Vertex Pharmaceuticals Incorporated 5-(2-aminopyrimidin-4-yl) benzisoxazoles as protein kinase inhibitors
US6689778B2 (en) 2001-07-03 2004-02-10 Vertex Pharmaceuticals Incorporated Inhibitors of Src and Lck protein kinases
US6698980B2 (en) 2001-07-30 2004-03-02 Stewart Mining Products Inc. Rock stabilizing apparatus and method
WO2003026665A1 (en) 2001-09-26 2003-04-03 Bayer Pharmaceuticals Corporation 2-phenylamino-4-(5-pyrazolylamino)-pyrimidine derivatives as kinase inhibitors, in particular, src kinase inhibitors
US6569499B2 (en) 2001-10-02 2003-05-27 Xerox Corporation Apparatus and method for coating photoreceptor substrates
EP2198867A1 (en) * 2001-12-07 2010-06-23 Vertex Pharmaceuticals, Inc. Pyrimidine-based compounds useful as GSK-3 inhibitors
US7863282B2 (en) 2003-03-14 2011-01-04 Vertex Pharmaceuticals Incorporated Compositions useful as inhibitors of protein kinases
EP1485100B1 (en) * 2002-03-15 2010-05-05 Vertex Pharmaceuticals Incorporated Azinylaminoazoles as inhibitors of protein kinases
US6846928B2 (en) * 2002-03-15 2005-01-25 Vertex Pharmaceuticals Incorporated Compositions useful as inhibitors of protein kinases
EP1485381B8 (en) 2002-03-15 2010-05-12 Vertex Pharmaceuticals Incorporated Azolylaminoazine as inhibitors of protein kinases
DE60332604D1 (en) * 2002-03-15 2010-07-01 Vertex Pharma AZOLYLAMINOAZINE AS PROTEIN KINASE INHIBITOR
US20030207873A1 (en) 2002-04-10 2003-11-06 Edmund Harrington Inhibitors of Src and other protein kinases
EP1506189A1 (en) 2002-04-26 2005-02-16 Vertex Pharmaceuticals Incorporated Pyrrole derivatives as inhibitors of erk2 and uses thereof
WO2004005283A1 (en) * 2002-07-09 2004-01-15 Vertex Pharmaceuticals Incorporated Imidazoles, oxazoles and thiazoles with protein kinase inhibiting activities
CA2494100C (en) 2002-08-02 2011-10-11 Vertex Pharmaceuticals Incorporated Pyrazole compositions useful as inhibitors of gsk-3
AU2003286711A1 (en) 2002-10-25 2004-05-13 Vertex Pharmaceuticals Incorporated Indazolinone compositions useful as kinase inhibitors
JP2004224944A (en) * 2003-01-24 2004-08-12 Kuraray Plast Co Ltd Thermoplastic resin composition excellent in high-frequency adhesiveness
JP2007510626A (en) 2003-10-17 2007-04-26 アストラゼネカ アクチボラグ 4- (Pyrazol-3-ylamino) pyrimidine derivatives for use in the treatment of cancer
MY141220A (en) * 2003-11-17 2010-03-31 Astrazeneca Ab Pyrazole derivatives as inhibitors of receptor tyrosine kinases
GB0402653D0 (en) * 2004-02-06 2004-03-10 Cyclacel Ltd Compounds
EP1910379B1 (en) 2005-07-28 2011-10-19 Vertex Pharmaceuticals Incorporated Caspase inhibitor prodrugs
EP1917259B1 (en) 2005-08-18 2012-01-25 Vertex Pharmaceuticals Incorporated Pyrazine kinase inhibitors
WO2007023382A2 (en) 2005-08-25 2007-03-01 Pfizer Inc. Pyrimidine amino pyrazole compounds, potent kinase inhibitors
KR20140025610A (en) * 2005-09-30 2014-03-04 미카나 테라퓨틱스, 인크. Substituted pyrazole compounds
CN103145702A (en) * 2005-11-03 2013-06-12 顶点医药品公司 Aminopyrimidines useful as kinase inhibitors
CN101360740A (en) 2005-11-16 2009-02-04 沃泰克斯药物股份有限公司 Aminopyrimidines useful as kinase inhibitors
DE602007004750D1 (en) 2006-11-02 2010-03-25 Vertex Pharma AS INHIBITORS OF PROTEIN KINASES SUITABLE AMINOPYRIDINES AND AMINOPYRIMIDINES
ES2347721T3 (en) 2006-12-19 2010-11-03 Vertex Pharmaceuticals, Inc. USEFUL AMINOPIRIMIDINS AS PROTEIN QUINASE INHIBITORS.
ES2618057T3 (en) * 2006-12-29 2017-06-20 Janssen Sciences Ireland Uc HIV-inhibited 5,6-substituted pyrimidines
NZ579483A (en) 2007-03-09 2012-07-27 Vertex Pharma Aminopyridines useful as inhibitors of protein kinases
EP2134707B1 (en) 2007-03-09 2013-09-11 Vertex Pharmaceuticals, Inc. Aminopyrimidines useful as inhibitors of protein kinases
NZ579485A (en) 2007-03-09 2012-02-24 Vertex Pharma Aminopyrimidines useful as inhibitors of protein kinases
JP2010522194A (en) 2007-03-20 2010-07-01 バーテックス ファーマシューティカルズ インコーポレイテッド Aminopyrimidines useful as kinase inhibitors
WO2008131103A2 (en) 2007-04-17 2008-10-30 Vertex Pharmaceuticals Incorporated Drug discovery methods for aurora kinase inhibitors
MX2009011811A (en) 2007-05-02 2010-01-14 Vertex Pharma Aminopyrimidines useful as kinase inhibitors.
CA2685876A1 (en) 2007-05-02 2008-11-13 Vertex Pharmaceuticals Incorporated Thiazoles and pyrazoles useful as kinase inhibitors
MX2009011812A (en) 2007-05-02 2010-01-14 Vertex Pharma Aminopyrimidines useful as kinase inhibitors.
AU2008257044A1 (en) 2007-05-24 2008-12-04 Vertex Pharmaceuticals Incorporated Thiazoles and pyrazoles useful as kinase inhibitors
NZ624460A (en) 2009-03-20 2015-12-24 Vertex Pharma Process for making modulators of cystic fibrosis transmembrane conductance regulator

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004000833A1 (en) 2002-06-20 2003-12-31 Vertex Pharmaceuticals Incorporated Processes for preparing substituted pyrimidines and pyrimidine derivatives as inhibitors of protein kinase

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8524720B2 (en) 2000-09-15 2013-09-03 Vertex Pharmaceuticals Incorporated Substituted N-(pyrazol-5-yl)-pyrrolo[3,2-D]pyrimidin-4-amine useful as protein kinase inhibitors
US8653088B2 (en) 2003-02-06 2014-02-18 Vertex Pharmaceuticals Incorporated Compositions useful as inhibitors of protein kinases
US8637511B2 (en) 2005-11-03 2014-01-28 Vertex Pharmaceuticals Incorporated Aminopyrimidines useful as kinase inhibitors
US8557833B2 (en) 2005-11-03 2013-10-15 Vertex Pharmaceuticals Incorporated Aminopyrimidines useful as kinase inhibitors
US8664219B2 (en) 2007-03-09 2014-03-04 Vertex Pharmaceuticals Incorporated Aminopyrimidines useful as inhibitors of protein kinases
US8410133B2 (en) 2007-03-09 2013-04-02 Vertex Pharmaceuticals Incorporated Aminopyridines useful as inhibitors of protein kinases
US8383633B2 (en) 2007-05-02 2013-02-26 Vertex Pharmaceuticals Incorporated Aminopyrimidines useful as kinase inhibitors
WO2008137621A1 (en) * 2007-05-02 2008-11-13 Vertex Pharmaceuticals Incorporated Aminopyrimidines useful as kinase inhibitors
US8785444B2 (en) 2007-05-02 2014-07-22 Vertex Pharmaceuticals Incorporated Thiazoles and pyrazoles useful as kinase inhibitors
WO2008147626A3 (en) * 2007-05-24 2009-03-19 Vertex Pharma Thiazoles and pyrazoles useful as kinase inhibitors
WO2008147626A2 (en) * 2007-05-24 2008-12-04 Vertex Pharmaceuticals Incorporated Thiazoles and pyrazoles useful as kinase inhibitors
JP2018104473A (en) * 2008-03-17 2018-07-05 ザ スクリプス リサーチ インスティテュート Combined chemical and genetic approaches for generation of induced pluripotent stem cells
JP2021143194A (en) * 2008-03-17 2021-09-24 ザ スクリプス リサーチ インスティテュート Combined chemical and genetic approaches for generation of induced pluripotent stem cells

Also Published As

Publication number Publication date
NZ594383A (en) 2013-05-31
MX2008005818A (en) 2008-10-15
CN103145702A (en) 2013-06-12
JP2013049731A (en) 2013-03-14
US20080032963A1 (en) 2008-02-07
AU2006311797A1 (en) 2007-05-18
AU2006311831B2 (en) 2013-04-18
CA2627830A1 (en) 2007-05-18
EP1954277A4 (en) 2011-11-09
WO2007056221A2 (en) 2007-05-18
US20070249031A1 (en) 2007-10-25
NZ594385A (en) 2013-02-22
US20090181938A1 (en) 2009-07-16
EP1954277A2 (en) 2008-08-13
RU2423361C2 (en) 2011-07-10
AU2006311830B2 (en) 2013-03-14
US20120142660A1 (en) 2012-06-07
MX2008005717A (en) 2008-10-15
JP2013075904A (en) 2013-04-25
RU2427578C2 (en) 2011-08-27
ES2533241T3 (en) 2015-04-08
US20110020469A1 (en) 2011-01-27
SG166827A1 (en) 2010-12-29
EP1951715A4 (en) 2011-11-09
RU2008122046A (en) 2009-12-10
IL191043A0 (en) 2008-12-29
ES2435081T3 (en) 2013-12-18
CN101316597B (en) 2013-04-17
US8557833B2 (en) 2013-10-15
US20070259869A1 (en) 2007-11-08
AU2006311831C1 (en) 2013-11-07
AR057874A1 (en) 2007-12-26
CN101316843B (en) 2013-01-02
TW200736250A (en) 2007-10-01
EP1948193A4 (en) 2011-11-09
WO2007056163A3 (en) 2008-03-27
NO20082517L (en) 2008-07-18
WO2007056164A3 (en) 2007-12-13
JP2009514880A (en) 2009-04-09
RU2008122048A (en) 2009-12-10
CA2627808A1 (en) 2007-05-18
MX2008005814A (en) 2008-10-15
NZ606263A (en) 2014-03-28
JP5328361B2 (en) 2013-10-30
RU2008122070A (en) 2009-12-10
US8637511B2 (en) 2014-01-28
EP1948193A2 (en) 2008-07-30
US20100310675A1 (en) 2010-12-09
CN101316843A (en) 2008-12-03
EP1951715B1 (en) 2013-09-04
RU2011118227A (en) 2012-11-10
IL191044A0 (en) 2008-12-29
AR056764A1 (en) 2007-10-24
EP1948193B1 (en) 2015-01-07
BRPI0619708A2 (en) 2011-10-11
JP5249770B2 (en) 2013-07-31
EP1954277B1 (en) 2017-01-18
BRPI0619704A2 (en) 2011-10-11
US8129399B2 (en) 2012-03-06
EP1951715A2 (en) 2008-08-06
RU2011112948A (en) 2012-10-10
KR20080067693A (en) 2008-07-21
WO2007056221A3 (en) 2007-12-06
KR20080067694A (en) 2008-07-21
SG166828A1 (en) 2010-12-29
NO20082508L (en) 2008-07-31
CA2627857A1 (en) 2007-05-18
NO20082514L (en) 2008-07-18
BRPI0619706A2 (en) 2011-10-11
JP2009514887A (en) 2009-04-09
JP2009514881A (en) 2009-04-09
AU2006311831A1 (en) 2007-05-18
TW200736249A (en) 2007-10-01
KR20080066069A (en) 2008-07-15
US7820685B2 (en) 2010-10-26
CN101316597A (en) 2008-12-03
AU2006311830A1 (en) 2007-05-18
NZ606259A (en) 2014-06-27
TW200734327A (en) 2007-09-16
WO2007056164A2 (en) 2007-05-18
US7528142B2 (en) 2009-05-05
AR056763A1 (en) 2007-10-24
JP5249771B2 (en) 2013-07-31
IL191140A0 (en) 2008-12-29
US7767672B2 (en) 2010-08-03

Similar Documents

Publication Publication Date Title
US7820685B2 (en) Aminopyrimidines useful as kinase inhibitors
US8383633B2 (en) Aminopyrimidines useful as kinase inhibitors
US20110046104A1 (en) Aminopyrimidines useful as kinase inhibitors
EP2148931A2 (en) Drug discovery methods for aurora kinase inhibitors
AU2013201058A1 (en) Aminopyrimidines useful as kinase inhibitors
AU2013201630B2 (en) Aminopyrimidines useful as kinase inhibitors
AU2016203378A1 (en) Aminopyrimidines useful as kinase inhibitors
US20140037754A1 (en) Aminopyrimidines useful as kinase inhibitors
US20140303137A1 (en) Aminopyrimidines useful as kinase inhibitors

Legal Events

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

Ref document number: 200680044414.8

Country of ref document: CN

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 191044

Country of ref document: IL

ENP Entry into the national phase

Ref document number: 2627808

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2008539070

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: MX/a/2008/005717

Country of ref document: MX

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2068/KOLNP/2008

Country of ref document: IN

Ref document number: 2006311830

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 568582

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 2006827465

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2008122048

Country of ref document: RU

Ref document number: 1020087013424

Country of ref document: KR

ENP Entry into the national phase

Ref document number: 2006311830

Country of ref document: AU

Date of ref document: 20061103

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: PI0619708

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20080502