WO2009073153A2 - Small molecule myristate inhibitors of bcr-abl and methods of use - Google Patents

Small molecule myristate inhibitors of bcr-abl and methods of use Download PDF

Info

Publication number
WO2009073153A2
WO2009073153A2 PCT/US2008/013219 US2008013219W WO2009073153A2 WO 2009073153 A2 WO2009073153 A2 WO 2009073153A2 US 2008013219 W US2008013219 W US 2008013219W WO 2009073153 A2 WO2009073153 A2 WO 2009073153A2
Authority
WO
WIPO (PCT)
Prior art keywords
optionally substituted
trifluoromethoxy
pyrimidin
phenyl
phenylamino
Prior art date
Application number
PCT/US2008/013219
Other languages
French (fr)
Other versions
WO2009073153A3 (en
Inventor
Nathanael S. Gray
Jianming Zhang
Barun Okram
Xianming Deng
Jaewon Chang
Amy Wojciechowski
Original Assignee
Dana Farber Cancer Institute
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 BRPI0819453-0A2A priority Critical patent/BRPI0819453A2/en
Priority to EA201070595A priority patent/EA019869B1/en
Application filed by Dana Farber Cancer Institute filed Critical Dana Farber Cancer Institute
Priority to US12/745,496 priority patent/US8921336B2/en
Priority to EP16197611.3A priority patent/EP3150593B8/en
Priority to CA2707046A priority patent/CA2707046A1/en
Priority to EP21184206.7A priority patent/EP3912973A3/en
Priority to ES08857261.5T priority patent/ES2610190T3/en
Priority to JP2010536015A priority patent/JP5779778B2/en
Priority to MX2010005671A priority patent/MX2010005671A/en
Priority to EP19170210.9A priority patent/EP3536687B8/en
Priority to AU2008331867A priority patent/AU2008331867B2/en
Priority to EP08857261.5A priority patent/EP2222162B1/en
Priority to CN200880123462.5A priority patent/CN101917849B/en
Publication of WO2009073153A2 publication Critical patent/WO2009073153A2/en
Publication of WO2009073153A3 publication Critical patent/WO2009073153A3/en
Priority to US14/540,919 priority patent/US9670214B2/en
Priority to US15/582,004 priority patent/US20170349595A1/en
Priority to US16/170,921 priority patent/US10787455B2/en
Priority to US16/933,095 priority patent/US11254682B2/en
Priority to US17/574,132 priority patent/US11932646B2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/517Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • A61K31/52Purines, e.g. adenine
    • 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
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7076Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines containing purines, e.g. adenosine, adenylic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • 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
    • 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
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/32One oxygen, sulfur or nitrogen atom
    • C07D239/42One nitrogen atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/70Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings condensed with carbocyclic rings or ring systems
    • C07D239/72Quinazolines; Hydrogenated quinazolines
    • C07D239/86Quinazolines; Hydrogenated quinazolines with hetero atoms directly attached in position 4
    • C07D239/94Nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • 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
    • C07D403/06Heterocyclic 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 carbon chain containing only aliphatic carbon atoms
    • 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
    • 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
    • C07D417/04Heterocyclic 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 directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D473/00Heterocyclic compounds containing purine ring systems
    • C07D473/26Heterocyclic compounds containing purine ring systems with an oxygen, sulphur, or nitrogen atom directly attached in position 2 or 6, but not in both
    • C07D473/32Nitrogen atom
    • C07D473/34Nitrogen atom attached in position 6, e.g. adenine
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D495/00Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
    • C07D495/02Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D495/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H19/00Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
    • C07H19/02Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
    • C07H19/04Heterocyclic radicals containing only nitrogen atoms as ring hetero atom
    • C07H19/16Purine radicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H19/00Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
    • C07H19/02Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
    • C07H19/04Heterocyclic radicals containing only nitrogen atoms as ring hetero atom
    • C07H19/16Purine radicals
    • C07H19/167Purine radicals with ribosyl as the saccharide radical
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the protein kinases represent a large family of proteins, which play a central role in the regulation of a wide variety of cellular processes and maintaining control over cellular function.
  • These kinases include receptor tyrosine kinases, such as platelet-derived growth factor receptor kinase (PDGF-R), the receptor kinase for stem cell factor, c- Kit, and nonreceptor tyrosine kinases, such as the fusion kinase Bcr-abl.
  • PDGF-R platelet-derived growth factor receptor kinase
  • Kit the receptor kinase for stem cell factor
  • nonreceptor tyrosine kinases such as the fusion kinase Bcr-abl.
  • Chronic myeloid leukemia is an extensively studied human cancer that is caused by a reciprocal translocation that fuses the AbI proto-oncogene on chromosome 9 with a gene on chromosome 22 called Bcr.
  • the resulting fusion protein Bcr-abl is capable of transforming B-cells by increasing mitogenic activity, reducing sensitivity to apoptosis and altering the adhesion and homing of CML progenitor cells.
  • STI-571 (Gleevec) is an inhibitor of the oncogenic Bcr-abl tyrosine kinase and is used for the treatment of chronic myeloid leukemia (CML).
  • CML chronic myeloid leukemia
  • some patients in the blast crisis stage of CML are resistant to STI-571 due to mutations in the Bcr-abl kinase.
  • novel compounds of this invention inhibit one or more kinases; in particular wild type and one or more of the mutant forms of Bcr-abl and are, therefore, useful in the treatment of kinase-associated diseases, particularly Bcr-abl kinase associated diseases.
  • the invention provides a compound of formula I, or pharmaceutically acceptable salt, solvate or hydrate thereof:
  • each of Xi, X 2 , or X 3 is independently NRi, CRi, C(O), O, or S; each Ri is independently selected from H, an optionally substituted Ci -6 alkyl, an optionally substituted C 3- i 2 cycloalkyl, an optionally substituted C 3 .i 2 heterocycloalkyl, an optionally substituted C 3- i 2 aryl, an optionally substituted C 3- i 2 heteroaryl, an optionally substituted Ci -6 aralkyl, an optionally substituted Ci -6 heteroaralkyl, an optionally substituted Ci -6 haloalkyl, an optionally substituted (CH 2 ) n C 3- i 2 heterocycloalkyl, an optionally substituted (CH 2 ) n C 3- i 2 aryl, an optionally substituted (CH 2 ) n C 3- i 2 heteroaryl, an optionally substituted (CH 2 ) n C 3- i 2 cycloalkyl, Ci -6 al
  • R 2 is H or Ci -6 alkyl
  • Y is H, OH, NH 2 , or CH 3 ; m is 1 or 2; and each n is independently 0, 1 or 2.
  • the invention provides a compound of formula II, or pharmaceutically acceptable salt, solvate or hydrate thereof: wherein
  • Ri is selected from H, an optionally substituted Ci -6 alkyl, an optionally substituted C 3 . i 2 cycloalkyl, an optionally substituted C 3- i 2 heterocycloalkyl, an optionally substituted C 3- i 2 aryl, an optionally substituted C 3 .i 2 heteroaryl, an optionally substituted C 1-6 aralkyl, an optionally substituted C ⁇ heteroaralkyl, an optionally substituted Ci ⁇ haloalkyl, an optionally substituted (CH 2 ) n C 3- i 2 heterocycloalkyl, an optionally substituted (CH 2 ) n C 3- i 2 aryl, an optionally substituted (CH 2 ) n C 3- i 2 heteroaryl, an optionally substituted (CH 2 ) n C 3- i 2 cycloalkyl, (CH 2 ) n C(O)R x , (CH 2 ) n C(O)OR x , (CH 2 ) n C
  • R x for each occurrence, is independently, H, an optionally substituted Ci -6 alkyl, an optionally substituted C 3- i 2 cycloalkyl, an optionally substituted C 3 .i 2 heterocycloalkyl, an optionally substituted and each n is independently 0, 1 or 2.
  • the invention provides a compound formula III, or pharmaceutically acceptable salt, solvate or hydrate thereof:
  • R] is selected from H, an optionally substituted Ci -6 alkyl, an optionally substituted C 3- i 2 cycloalkyl, an optionally substituted C 3- i 2 heterocycloalkyl, an optionally substituted C 3- i 2 aryl, an optionally substituted C 3- i 2 heteroaryl, an optionally substituted Ci -6 aralkyl, an optionally substituted Ci- ⁇ heteroaralkyl, an optionally substituted Ci- ⁇ haloalkyl, an optionally substituted (CH 2 ) n C 3- i 2 heterocycloalkyl, an optionally substituted (CH 2 ) n C 3 .i 2 aryl, an optionally substituted (CH 2 ) n C 3- i 2 heteroaryl, an optionally substituted (CH 2 ) n C 3-12 cycloalkyl, (CH 2 ) n C(O)R x , (CH 2 ) n C(O)OR x , (CH 2 ) n
  • R x for each occurrence, is independently, H, an optionally substituted Ci -6 alkyl, an optionally substituted C 3 -i 2 cycloalkyl, an optionally substituted Cs. ⁇ heterocycloalkyl, an optionally substituted C 3-12 aryl, an optionally substituted C 3- i 2 heteroaryl, or Ci -6 haloalkyl; and each n is independently 0, 1 or 2.
  • the invention provides a compound of formula IV, or pharmaceutically acceptable salt, solvate or hydrate thereof:
  • each R] is indepdently selected from H, an optionally substituted Ci -6 alkyl, an optionally substituted C 3-12 cycloalkyl, an optionally substituted C 3 -i 2 heterocycloalkyl, an optionally substituted C 3- i 2 aryl, an optionally substituted C 3- i 2 heteroaryl, an optionally substituted Ci -6 aralkyl, an optionally substituted an optionally substituted Ci -6 haloalkyl, an optionally substituted (CH 2 ) n C 3- i 2 heterocycloalkyl, an optionally substituted (CH 2 ) n C 3- i 2 aryl, an optionally substituted (CH 2 ) n C 3- i 2 heteroaryl, an optionally substituted (CH 2 ) n C 3-12 cycloalkyl, (CH 2 ) n C(O)R x , (CH 2 ) n C(O)OR ⁇ (CH 2 ) n C(O)NR x R
  • R x for each occurrence, is independently, H, an optionally substituted Ci -6 alkyl, an optionally substituted C 3 -i 2 cycloalkyl, an optionally substituted C 3- i 2 heterocycloalkyl, an optionally substituted C 3 .i 2 aryl, an optionally substituted C 3 -i 2 heteroaryl, or Cj- ⁇ haloalkyl; and each n is independently 0, 1 or 2.
  • the invention provides a compound of formula V, or pharmaceutically acceptable salt, solvate or hydrate thereof:
  • R] is independently selected from H, an optionally substituted Ci -6 alkyl, an optionally substituted C 3 -i 2 cycloalkyl, an optionally substituted Cs- ⁇ heterocycloalkyl, an optionally substituted C 3 -i 2 aryl, an optionally substituted C 3 -i 2 heteroaryl, an optionally substituted (CH 2 ) n C 3- i 2 heterocycloalkyl, -C(O)R", -OR", -SR X , -NR X R X , nitro, cyano, Ci -6 haloalkyl, C,.
  • R x for each occurrence, is independently H, an optionally substituted Ci -6 alkyl, an optionally substituted C 3- i 2 cycloalkyl, an optionally substituted C 3- i 2 heterocycloalkyl, an optionally substituted C 3-12 aryl, an optionally substituted C 3 -i 2 heteroaryl, NHNHR 2 , or C(NH)NH 2 ;
  • Ri' is selected from H, an optionally substituted Ci -6 alkyl, an optionally substituted C 3- i 2 cycloalkyl, an optionally substituted C 3 .i 2 heterocycloalkyl, an optionally substituted C 3 . i 2 aryl, an optionally substituted C 3 ,i 2 heteroaryl, an optionally substituted Ci -6 aralkyl, an optionally substituted Ci- ⁇ heteroaralkyl, an optionally substituted Ci ⁇ haloalkyl, an optionally substituted (CH 2 ) n C 3- i 2 heterocycloalkyl, an optionally substituted (CH 2 ) n C 3 .i 2 aryl, an optionally substituted (CH 2 ) n C 3- i 2 heteroaryl, an optionally substituted (CH 2 ) n C 3 -i 2 cycloalkyl, (CH 2 ) n C(0)R y , (CH 2 ) n C(O)OR y , (CH 2 )
  • R y for each occurrence, is independently, H, an optionally substituted Ci -6 alkyl, an optionally substituted C 3 -i 2 cycloalkyl, an optionally substituted C 3 -i 2 heterocycloalkyl, an optionally substituted C 3 .i 2 aryl, an optionally substituted C 3 .i 2 heteroaryl, or Ci -6 haloalkyl; each n is independently 0, 1 or 2; and
  • R 2 is H or CM alkyl.
  • Ri is independently selected from H, an optionally substituted Ci -6 alkyl, an optionally substituted C 3 .i 2 cycloalkyl, an optionally substituted C 3 . 12 heterocycloalkyl, an optionally substituted C 3-12 aryl, an optionally substituted C 3 .
  • nheteroaryl an optionally substituted (CH 2 ) n C 3- i 2 heterocycloalkyl, -C(O)R*, -OR X , -SR X , -NR X R X , nitro, cyano, Ci -6 haloalkyl, Ci -6 aminoalkyl, (CH 2 ) n C(O)R x , (CH 2 ) n C(O)OR x , (CH 2 ) n C(O)NR x R x , or -(CH 2 ) n C(O)NR x R x S(O) 2 R x ;
  • R x for each occurrence, is independently H, an optionally substituted Ci -6 alkyl, an optionally substituted C 3- i 2 cycloalkyl, an optionally substituted Cs- ⁇ heterocycloalkyl, an optionally substituted C 3- i 2 aryl, an optionally substituted C 3- i 2 heteroaryl, NHNHR 2 , or C(NH)NH 2 ; and
  • R 2 is H or C M alkyl.
  • R 1 ' is selected from H, an optionally substituted Ci- 6 alkyl, an optionally substituted C 3- i 2 cycloalkyl, an optionally substituted C 3 . i 2 heterocycloalkyl, an optionally substituted C 3- i 2 aryl, an optionally substituted C 3- i 2 heteroaryl, an optionally substituted Ci -6 aralkyl, an optionally substituted Ci -6 heteroaralkyl, an optionally substituted Ci -6 haloalkyl, an optionally substituted (CH 2 ) n C 3- i 2 heterocycloalkyl, an optionally substituted (CH 2 ) n C 3- i 2 aryl, an optionally substituted (CH 2 ) n C 3- i 2 heteroaryl, an optionally substituted (CH 2 ) n C 3 ., 2 cycloalkyl, (CH 2 ) n C(O)R y , (CH 2 ) n C(0)0R y ,
  • R y for each occurrence, is independently, H, an optionally substituted Ci -6 alkyl, an optionally substituted C 3- i 2 cycloalkyl, an optionally substituted C 3- i 2 heterocycloalkyl, an optionally substituted C 3- i 2 aryl, an optionally substituted C 3-12 heteroaryl, or Ci -6 haloalkyl;
  • the invention provides a compound of formula VI, or pharmaceutically acceptable salt, solvate or hydrate thereof:
  • Ri is independently selected from H, an optionally substituted Ci -6 alkyl, an optionally substituted C 3- i 2 cycloalkyl, an optionally substituted C 3- i 2 heterocycloalkyl, an optionally substituted C 3- i 2 aryl, an optionally substituted C 3- i 2 heteroaryl, an optionally substituted (CH 2 ) n C 3- i 2 heterocycloalkyl, -C(O)R", -OR ⁇ -SR", -NR X R X , nitro, cyano, Ci -6 haloalkyl, Ci- 6 aminoalkyl, (CH 2 ) n C(O)R x , (CH 2 ) n C(O)OR x , (CH 2 ) n C(O)NR x R x , or -(CH 2 ) n C(O)NR x R x S(O) 2 R x ;
  • R x for each occurrence, is independently H, an optionally substituted Ci -6 alkyl, an optionally substituted C 3- i 2 cycloalkyl, an optionally substituted C 3- i 2 heterocycloalkyl, an optionally substituted C 3- i 2 aryl, an optionally substituted C 3- i 2 heteroaryl, NHNHR 2 , C(NH)NH 2 ;
  • R 2 is H or Ci -4 alkyl
  • n is independently 0, 1, or 2.
  • the invention provides a compound of formula VII, or pharmaceutically acceptable salt, solvate or hydrate thereof:
  • Ri is selected from H, an optionally substituted Ci -6 alkyl, an optionally substituted C 3- i 2 cycloalkyl, an optionally substituted C 3 -i 2 heterocycloalkyl, an optionally substituted C 3- i 2 aryl, an optionally substituted C 3- i 2 heteroaryl, an optionally substituted (C ⁇ ) n C 3- i 2 heterocycloalkyl, -C(O)R X , -OR X , -SR", -NR X R X , nitro, cyano, Ci.
  • R x for each occurrence, is independently H, an optionally substituted Ci -6 alkyl, an optionally substituted C 3- i 2 cycloalkyl, an optionally substituted Cs- ⁇ heterocycloalkyl, an optionally substituted C 3- i 2 aryl, an optionally substituted C 3- i 2 heteroaryl, NHNHR 2 , or C(NH)NH 2 ;
  • R 2 is H or Ci -4 alkyl; m is 1 or 2; and each n is independently 0, 1 or 2.
  • the invention provides a compound of formula VIII, or pharmaceutically acceptable salt, solvate or hydrate thereof:
  • Ri is selected from H, an optionally substituted Ci -6 alkyl, an optionally substituted C 3- i 2 cycloalkyl, an optionally substituted C 3- i 2 heterocycloalkyl, an optionally substituted C 3- i 2 aryl, an optionally substituted C 3- i 2 heteroaryl, an optionally substituted Ci- ⁇ aralkyl, an optionally substituted Ci- ⁇ heteroaralkyl, an optionally substituted Ci -6 haloalkyl, an optionally substituted (CH 2 ) n C 3- i 2 heterocycloalkyl, an optionally substituted (CH 2 ) n C 3 -i 2 aryl, an optionally substituted (CH 2 ) n C 3-12 heteroaryl, an optionally substituted (CH 2 ) n C 3- i 2 cycloalkyl, (CH 2 ) n C(O)R x , (CH 2 ) n C(O)OR x , (CH 2 ) n C
  • R x for each occurrence, is independently, H, an optionally substituted Ci- ⁇ alkyl, an optionally substituted C 3- i 2 cycloalkyl, an optionally substituted C 3- i 2 heterocycloalkyl, an optionally substituted C 3 .i 2 aryl, an optionally substituted C 3- i 2 heteroaryl, or Ci -6 haloalkyl; and each n is independently 0, 1 or 2.
  • the invention provides a compound of formula IX, or pharmaceutically acceptable salt, solvate or hydrate thereof:
  • R 3 is H or C 1-4 alkyl
  • L is NR 3 , O, S, S(O), S(O) 2 , C(O), C(S), C(NR), C(NR X )NR X , C(O)NR", C(O)NR X NR X , C(0)0NR x , C(O)NR X O, C(O)O, OC(O), OC(O)O, or a bond
  • each R x for each occurrence, is independently H, an optionally substituted Ci ⁇ alkyl, an optionally substituted C 3- i 2 cycloalkyl, an optionally substituted C 3- i 2 heterocycloalkyl, an optionally substituted C 3-
  • A is an optionally substituted Ci -6 alkyl, an optionally substituted C 3- i 2 aryl, an optionally substituted C 3- i 2 heteroaryl, an optionally substituted C 3- i 2 Cycloalkyl, or an optionally substituted C 3- i 2 heterocycloalkyl.
  • A is an optionally substituted C]. 6 alkyl, an optionally substituted C 3- i 2 aryl, an optionally substituted C 3-12 heteroaryl, or an optionally substituted C 3- ⁇ 2 heterocycloalkyl.
  • A is an optionally substitued phenyl ring, an optionally substituted naphthyl ring, an optionally substituted piperidine ring, an optionally substituted imidazole ring, an optionally substituted pyrazole ring, an optionally substituted lH-pyrazole ring, an optionally substituted lH-Pyrrolo[2,3-b]pyridine ring, an optionally substituted pyrimidine ring, an optionally substituted pyrazine ring, an optionally substituted pyridine ring, an optionally substituted indole ring, an optionally substituted lH-indole ring, an optionally substituted furan ring, an optionally substituted benzo[b]thiophene ring, an optionally substituted dibenzofuran ring, an optionally substituted lH-Pyridin-2-one ring, an optionally substituted thiazole ring, or an ethyl group.
  • A is optionally substituted with Ci -6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3 -i 2 cycloalkyl, C 3 -i 2 heterocycloalkyl, C 3 -i 4 aralkyl, Q- ⁇ heteroaralkyl, C 3 - i 2 aryl, (CH 2 ) p C 3 -i 2 aryl, C 3 -i 2 heteroaryl, (CH 2 ) p C 3 -i 2 heteroaryl, halogen, haloalkyl, cyano, (CH 2 ) P CN, nitro, Ci -6 alkoxy, C 3 -i 2 aryloxy, C 1 -C 4 haloalkoxy, Ci-C 6 haloalkyl, hydroxyl, Ci- 6 hydroxylalkyl, Ci -6 carboxyl, CHO, amino, aminoalkyl, dialkylamino, OR 15 , SR 15 ,
  • the invention provides a compound of formula X, or pharmaceutically acceptable salt, solvate or hydrate thereof:
  • each Of X 2 or X 3 is independently NR b CR 1 , C(O), O, or S; each R] is independently selected from H, an optionally substituted Ci -6 alkyl, an optionally substituted C 3-12 cycloalkyl, an optionally substituted C 3- i 2 heterocycloalkyl, an optionally substituted C 3-12 aryl, an optionally substituted C 3 .i 2 heteroaryl, an optionally substituted C 2-6 aralkyl, an optionally substituted C 2- i 2 heteroaralkyl, an optionally substituted Ci -6 haloalkyl, an optionally substituted (CH 2 ) n C 3- i 2 aryl, an optionally substituted (CH 2 ), ⁇ .
  • R 2 is H or Ci -6 alkyl; and each n is independently 0, 1 or 2.
  • the invention provides a compound of formula XI, or pharmaceutically acceptable salt, solvate or hydrate thereof:
  • Ri is independently selected from H, an optionally substituted Ci -6 alkyl, an optionally substituted C 3- i 2 aryl, or an optionally substituted Cs- ⁇ heteroaryl.
  • the present invention is directed towards a method of treating a disease in a subject in which inhibition of kinase activity, particularly Bcr-abl activity, can prevent, inhibit or ameliorate the pathology and/or symptomology of certain disorders and/or diseases, which method comprises administering to the subject a therapeutically effective amount of a compound of the invention or pharmaceutically acceptable salt, solvate or hydrate thereof.
  • the invention provides a method of treating a kinase activity related disorder in a subject, comprising administering to said subject in need thereof, an effective amount of a compound, or pharmaceutically acceptable salt, solvate or hydrate thereof, of any one of the compounds disclosed herein.
  • the compound is a kinase inhibitor.
  • the compound interacts with Bcr-abl.
  • the compound targets the binding site of Bcr-abl.
  • the subject is identified as being in need of kinase inhibition, and the compound of the invention is administered to the identified subject.
  • the invention provides the method further comprising the step of identifying a subject in need of such treatment for a kinase activity related disorder and administering the compound to the identified subject.
  • the invention provides a method wherein the subject is suffering from a cell proliferative disorder or disease.
  • the disorder is cancer, tumor, neoplasm, neovascularization, vascularization, cardiovascular disease, metastasis, infection, blood clot, atherosclerosis, melanoma, skin disorder, rheumatoid arthritis, diabetic retinopathy, macular edema, or macular degeneration, inflammatory and arthritic disease, or osteosarcoma.
  • the subject is suffering from a solid tumor or disseminated cancer.
  • the cancer is leukemia, multiple myeloma or lymphoma. In certain embodiments, the leukemia is Chroic Myelogenous Leukemia.
  • the invention provides a method of treating cancer in a subject, the method comprising administering to said subject an effective amount of a compound, or pharmaceutically acceptable salt, solvate or hydrate thereof, of any one of the compounds disclosed herein.
  • the invention provides a method described above further comprising administering an additional therapeutic agent.
  • the additional therapeutic agent is an ATP-site inhibitor.
  • the ATP-site inhibitor is selected from the following: imatinib (STI571), Nilotinib (AMN 107), Dasatinib (BMS-354825), AP234464 (Ariad Pharmaceuticals), AZD0530 (Astrazeneca), and SKI-606 (Wyeth).
  • the subject is a mammal. In a further embodiment, the subject is a primate or human.
  • the invention provides a method wherein the step of administering the compound comprises administering the compound orally, topically, parentally, intravenously or intramuscularly. In a further embodiment, the method comprises the step of administering an effective amount of a composition comprising the compound and a pharmaceutically suitable excipient.
  • the step of administering the compound comprises administering the compound in a dosage of between about 0.01 ⁇ g/kg/day and 100 mg/kg/day.
  • the invention provides a method of inhibiting kinase activity in a subject, the method comprising the step of administering to the subject a compound, or pharmaceutically acceptable salt, solvate or hydrate thereof, of any one of the compounds disclosed herein.
  • the invention provides the use of a compound described herein in the manufacture of a medicament for treating a disease in a subject in which kinase activity, particularly Bcr-abl activity, contributes to the pathology and/or symptomology of the disease.
  • the invention provides a pharmaceutical composition comprising a compound, or pharmaceutically acceptable salt, solvate or hydrate thereof, of any of the compounds disclosed herein.
  • the invention provides a composition further comprising an additional therapeutic agent.
  • the additional therapeutic agent is an ATP-site inhibitor.
  • the ATP-site inhibitor is selected from the following: imatinib (STI571), Nilotinib (AMN107), Dasatinib (BMS-354825), AP234464 (Ariad Pharmaceuticals), AZD0530 (Astrazeneca), and SKI-606 (Wyeth).
  • the invention provides a kit comprising an effective amount of a compound disclosed herein, or pharmaceutically acceptable salt, solvate or hydrate thereof, in unit dosage form, together with instructions for administering the compound to a subject suffering from or susceptible to a cell proliferative disorder.
  • the present invention provides a method for inhibiting Bcr-abl activity, the method comprising contacting Bcr-abl with a compound of the invention that binds to a myristoyl binding pocket of Bcr-abl.
  • the invention provides for a method for identifying a compound which modulates the kinase activity of Bcr-abl, the method comprising: a) contacting Bcr-abl with a compound under conditions suitable for modulation of the kinase activity of Bcr-abl; and b) detecting modulation of the kinase activity of Bcr-abl by the compound; wherein the compound is capable of interacting with the myristate binding site of Bcr- abl.
  • the interaction of the compound with myristate binding site of Bcr-abl is a binding interaction.
  • the binding interaction is ionic, covalent, or a non-direct interaction.
  • the compound has a binding interaction with Ala452, Tyr454, Glu481, Pro484, Val487, Phe516, IleS21, or Val525 (Number based upon crystal structure with protein databank code: lOPL, reference: Nagar, B., Hantschel, O., Young, M.A., Scheffzek, K., Veach, D., Bornmann, W., Clarkson, B., Superti-Furga, G., Kuriyan, J. Structural basis for the autoinhibition of c- AbI tyrosine kinase Cell (Cambridge,Mass.) vl l2 pp. 859-871, 2003.
  • the modulation of the activity of the kinase activity is detected by direct binding of the compound to the myristate binding site of Bcr-abl.
  • the compound is designed de novo.
  • the compound is designed from a known ligand of the myristate binding site of Bcr-abl.
  • Exemplary compounds of the invention include the following, which are provided structurally in Tables 1-9.
  • Butane- 1 -sulfonic acid ⁇ 4-[6-(4-trifluoromethoxy-phenylamino)-pyrimidin-4-yl]-phenyl ⁇ - amide (153);
  • Pentane-2-sulfonic acid ⁇ 4-[6-(4-trifluoromethoxy-phenylamino)-pyrimidin-4-yl]-phenyl ⁇ - amide (155);
  • the compounds of formulae II and III were formed over a two-step reaction. First a chlorinated heteroaryl compound was coupled with a haloalkoxy substituted aniline derivative under basic conditions. The resulting compound was then treated with a hydrazine derivative to provide a mixture of compounds of formulae II and III, which were separated by chromatography (Scheme 1). Alternatively, hydrazine is added followed by addition of an electrophile under basic conditions to provide the compounds of formulae II and III.
  • Scheme 3 provides a synthetic route to arrive at the compounds of formulae V and VI.
  • a heteroaryl compound with adjacent chloro and aldehyde substitution was reacted with a substituted thiol derivative or secondary amine derivative to provide the fused bicyclic framework of the compounds of formulae V and VI. Additional functional group modification provided for the compounds of the invention.
  • the compounds of formula VII were prepared by coupling a halogenated fused bicyclic heteroaryl compound with a substituted aniline derivative. Additional functional group modification of the side chain provided the compounds of formula VII (Scheme 4).
  • the compounds of formula IX were synthesized by initially coupling a dihalogenated heteraryl compound with an aniline derivative, followed by another coupling under Mitsunobu conditions to provide the compounds of formula IX.
  • a second halo group on the heteroaryl ring is displaced by a nucleophile under basic conditions to provide a compound of formula IX (Scheme 5).
  • the aryl-substituted fused bicyclic compounds of formula X were synthesized according to Scheme 7.
  • a heteroaryl compound substituted with a halo group and an amino group was reacted with an alpha halo carbonyl compound, which produced a bicyclic heteroaryl compound.
  • Acids and bases useful in the methods herein are known in the art.
  • Acid catalysts are any acidic chemical, which can be inorganic (e.g., hydrochloric, sulfuric, nitric acids, aluminum trichloride) or organic (e.g., camphorsulfonic acid, p-toluenesulfonic acid, acetic acid, ytterbium triflate) in nature. Acids are useful in either catalytic or stoichiometric amounts to facilitate chemical reactions.
  • Bases are any basic chemical, which can be inorganic (e.g., sodium bicarbonate, potassium hydroxide) or organic (e.g., triethylamine, pyridine) in nature. Bases are useful in either catalytic or stoichiometric amounts to facilitate chemical reactions.
  • the compounds of the invention are defined herein by their chemical structures and/or chemical names. Where a compound is referred to by both a chemical structure and a chemical name, and the chemical structure and chemical name conflict, the chemical structure is determinative of the compound's identity.
  • compositions including solutions, capsules, cremes, or ointments for administration to a subject (e.g., human, animal).
  • compositions e.g., pharmaceuticals
  • Such compositions are useful for providing to the subject desirable health or other physiological benefits that are associated with such compounds.
  • Nucleophilic agents are known in the art and are described in the chemical texts and treatises referred to herein.
  • the chemicals used in the aforementioned methods may include, for example, solvents, reagents, catalysts, protecting group and deprotecting group reagents and the like.
  • the methods described above may also additionally include steps, either before or after the steps described specifically herein, to add or remove suitable protecting groups in order to ultimately allow synthesis of the compound of the formulae described herein.
  • the methods delineated herein contemplate converting compounds of one formula to compounds of another formula.
  • the process of converting refers to one or more chemical transformations, which can be performed in situ, or with isolation of intermediate compounds.
  • the transformations can include reacting the starting compounds or intermediates with additional reagents using techniques and protocols known in the art, including those in the references cited herein.
  • Intermediates can be used with or without purification (e.g., filtration, distillation, crystallization, chromatography).
  • Other embodiments relate to the intermediate compounds delineated herein, and their use in the methods (e.g., treatment, synthesis) delineated herein.
  • compounds of the invention in free form or in pharmaceutically acceptable salt form, exhibit valuable pharmacological properties, for example, as indicated by the in vitro tests described within "Assays", infra, and are therefore indicated for therapy of diseases and disorders associated with Bcr-abl activity.
  • compounds of the invention preferably show an IC 50 in the range of 1x10 " ' to 1x10 ⁇ 5 M, preferably less than 1 ⁇ M for wild-type Bcr-abl and at least two other Bcr-abl mutants (mutants selected from G250E, E255V, T315I, F317L and M351T).
  • the invention also provides a method for preventing or treating diseases or conditions comprising abnormal cell growth in a subject, including a human, comprising administering to the subject a compound of the invention in an amount effective to inhibit PDGF-R, c-Kit and/or Bcr-abl activity.
  • PDGF Platinum-derived Growth Factor
  • PDGF is a very commonly occurring growth factor, which plays an important role both in normal growth and also in pathological cell proliferation, such as is seen in carcinogenesis and in diseases of the smooth-muscle cells of blood vessels, for example in atherosclerosis and thrombosis.
  • Compounds of the invention can inhibit PDGF-R and are, therefore, also suitable for the treatment of tumor diseases, which are delineated infra.
  • the compounds of the present invention also inhibit cellular processes involving stem-cell factor (SCF, also known as the c-kit ligand or steel factor), such as SCF receptor (kit) autophosphorylation and the SCF-stimulated activationof MAPK kinase (mitogen- activated protein kinase).
  • SCF stem-cell factor
  • kit SCF receptor
  • MAPK kinase mitogen- activated protein kinase
  • M07e cells are a human promegakaryocytic leukemia cell line, which depends on SCF for proliferation.
  • a compound of the invention inhibits the autophosphorylationof SCF-R in the micromolar range.
  • abl kinase is inhibited by compounds of the present invention.
  • the compounds of the present invention also inhibit Bcr- abl kinase and are thus suitable for the treatment of Bcr-abl-positive cancer and tumor diseases, such as leukemias (especially chronic myeloid leukemia and acutelymphoblastic leukemia, where especially apoptotic mechanisms of action are found), and also shows effects on the subgroup of leukemic stem cells as well as potential for the purification of these cells in vitro after removal of said cells (for example, bone marrow removal) and reimplantation of the cells once they have been cleared of cancer cells (for example, reimplantation of purified bone marrow cells).
  • Bcr-abl abl kinase especially chronic myeloid leukemia and acutelymphoblastic leukemia, where especially apoptotic mechanisms of action are found
  • the present invention provides a method for inhibiting Bcr-abl activity, the method comprising contacting Bcr-abl with a compound of the invention that binds to a myristoyl binding pocket of Bcr-abl.
  • the compounds of the present invention can be used in combination with other anti-tumor agents.
  • Tumor is used to describe an abnormal growth in tissue which occurs when cellular proliferation is more rapid than normal tissue and continues to grow after the stimuli that initated the new growth cease. Tumors generally exhibit partial or complete lack of structural organization and functional coordination with the normal tissue, and usually form a distinct mass of tissue which may be benign (benign tumor) or malignant (carcinoma). Tumors tend to be highly vascularized.
  • carcinoma is used as a general term herein to describe malignant tumors or carcinoma. These malignant tumors may invade surrounding tissues, may metastasize to several sites and are likely to recur after attempted removal and to cause death of the patient unless adequately treated. As used herein, the terms carcinoma and cancer are subsumed under the term tumor.
  • sarcomas neoplasia
  • internal malignancies such as eye or ocular cancer
  • rectal cancer colon cancer
  • cervical cancer prostate cancer
  • breast cancer and bladder cancer benign and malignant tumors
  • various cancers such as, anal and oral cancers, stomach, rectal, liver, pancreatic, lung, cervix uteri, corpus uteri, ovary, prostate, testis, renal, mouth/pharynx, esophageal, larynx, kidney, brain/ens (e.g., gliomas), head and neck, throat, skin melanoma, acute lymphocytic leukemia, acute myelogenous leukemia, Ewing's Sarcoma, Kaposi's Sarcoma, basal cell carinoma and squamous cell carcinoma, small cell lung cancer, choriocarcinoma, rhabdomyosarcoma, angiosarcom
  • the inhibition of the kinase activity of Bcr-abl is an important aspect of the present invention. More particularly, the present invention relates to methods for treating cancers, including Chronic Myelogenous Leukemia, comprising exposing the cancer to an inhibitory or therapeutically effective amount or concentration of at least one of the disclosed compounds. This method may be used therapeutically, in comparison tests such as assays for determining the activities of related analogs as well as for determining the susceptibility of a patient's cancer to one or more of the compounds according to the present invention.
  • Kinase inhibiting compounds of the present invention are used to treat, ameliorate or prevent, among others, benign and malignant tumors, including various cancers such as, Chronic Myelogenous Leukemia, cervical, anal and oral cancers, stomach, colon, bladder, rectal, liver, pancreatic, lung, breast, cervix uteri, corpus uteri, ovary, prostate, testis, renal, brain/ens (e.g., gliomas), head and neck, eye or ocular, throat, skin melanoma, acute lymphocytic leukemia, acute myelogenous leukemia, Ewing's Sarcoma, Kaposi's Sarcoma, basal cell carinoma and squamous cell carcinoma, small cell lung cancer, choriocarcinoma, rhabdomyosarcoma, angiosarcoma, hemangioendothelioma, Wilms Tumor, neuroblastoma, mouth/pharynx, esoph
  • the compounds of the invention are useful in treating non-malignant proliferative disorders, such as atherosclerosis, thrombosis, psoriasis, scleroderma, and fibrosis, as well as for the protection of stem cells, for example to combat the hemotoxic effect of chemotherapeutic agents, such as 5-fluoruracil, and in asthma. It can especially be used for the treatment of diseases, which respond to an inhibition of the PDGF-R kinase.
  • Methods for treating, ameliorating, or preventing skin disorders such as psoriasis, acne, rosacea, warts, eczema, hemangiomas, lymphangiogenesis, neurofibromatosis, Sturge- Weber syndrome, venous ulcers of the skin, tuberous sclerosis, chronic inflammatory disease and arthritis, as well as inflammation such as chronic inflammatory disease, including arthritis, lupus and scleroderma are also contemplated by the present invention, such methods comprising administering a therapeutically effective amount of one or more of the disclosed compounds to a subject in need of such treatment.
  • skin disorders such as psoriasis, acne, rosacea, warts, eczema, hemangiomas, lymphangiogenesis, neurofibromatosis, Sturge- Weber syndrome, venous ulcers of the skin, tuberous sclerosis, chronic inflammatory disease and arthritis, as well as inflammation such as chronic inflammatory disease, including arthritis, lupus and
  • neovascularization inlcude optic disc neovascularization, iris neovascularization, retinal neovascularization, choroidal neovascularization, corneal neovascularization, and intravitreal neovascularization.
  • Diseases associated with corneal neovascularization and retinal/choroidal neovascularization that can be treated, ameliorated, or prevented, according to the present invention include but are not limited to, diabetic retinopathy, retinopathy of prematurity, corneal graft rejection, neovascular glaucoma and retrolental fibroplasias, epidemic keratoconjunctivitis, vitamin A deficiency, contact lens overwear, atopic keratitis, superior limbic keratitis, pterygium keratitis sicca, Sjogren's, acne rosacea, phylectenulosis, syphilis, Mycobacteria infections, lipid degeneration, chemical bums, bacterial ulcers, fungal ulcers, herpes simplex infections, herpes zoster infections, protozoan infections, Kaposi's sarcoma, Mooren's ulcer, Terrien's marginal degeneration, marginal keratolysis
  • diseases include, but are not limited to, diseases associated with rubeosis (neovascularization of the ankle), diseases caused by the abnormal proliferation of fibrovascular or fibrous tissue including all forms of proliferative vitreoretinopathy, whether or not associated with diabetes, and corneal graft rejection.
  • Diseases associated with chronic inflammation and arthritis can be treated, ameliorated or prevented by the compositions and methods of the present invention.
  • Diseases with symptoms of chronic inflammation include inflammatory bowel diseases such as Crohn's disease and ulcerative colitis, psoriasis, sarcoidosis, rheumatoid arthritis, osteoarthritis, lupus and scleroderma.
  • the compounds of the present invention show useful effects in the treatment of disorders arising as a result of transplantation, for example, allogenic transplantation, especially tissue rejection, such as especiallyobliterative bronchiolitis (OB), i.e., a chronic rejection of allogenic lung transplants.
  • allogenic transplantation especially tissue rejection, such as especiallyobliterative bronchiolitis (OB)
  • OB especiallyobliterative bronchiolitis
  • those with OB often show an elevated PDGF concentration in bronchoalveolar lavage fluids.
  • Synergistic effects with other immunomodulatory or anti- inflammatory substances are possible, for example when used in combination with cyclosporin, rapamycin, or ascomycin, or immunosuppressant analogues thereof, for example cyclosporin A (CsA), cyclosporin G, FK-506, rapamycin, or comparable compounds, corticosteroids, cyclophosphamide, azathioprine, methotrexate, brequinar, leflunomide, mizoribine, mycophenolic acid, mycophenolate mofetil, 15-deoxyspergualin, immunosuppressant antibodies, especially monoclonal antibodies for leukocyte receptors, for example MHC, CD2,CD3, CD4, CD7, CD25,CD28, B7, CD45, CD58 or their ligands, or other immunomodulatory compounds, such as CTLA41g.
  • CsA cyclosporin A
  • FK-506, rapamycin or comparable compounds
  • corticosteroids
  • the compounds of the present invention are also effective in diseases associated with vascular smooth-muscle cell migration and proliferation (where PDGF and PDGF-R often also play a role), such as restenosis and atherosclerosis.
  • compositions and dosage forms of the invention comprise one or more of the active ingredients disclosed herein.
  • Pharmaceutical compositions and dosage forms of the invention typically also comprise one or more pharmaceutically acceptable excipients or diluents.
  • pharmaceutically acceptable refers to a component that is, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and other mammals without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio.
  • pharmaceutically acceptable salt means any non-toxic salt that, upon administration to a recipient, is capable of providing, either directly or indirectly, a compound or a prodrug of a compound of this invention.
  • pharmaceutically acceptable counterion is an ionic portion of a salt that is not toxic when released from the salt upon administration to a recipient.
  • compositions comprising an effective amount of a composition containing a compound of the invention and an acceptable carrier.
  • a composition of this invention is formulated for pharmaceutical use ("a pharmaceutical composition"), wherein the carrier is a pharmaceutically acceptable carrier.
  • the carrier(s) must be "acceptable” in the sense of being compatible with the other ingredients of the formulation and, in the case of a pharmaceutically acceptable carrier, not deleterious to the recipient thereof in amounts typically used in medicaments.
  • Pharmaceutically acceptable carriers, adjuvants and vehicles that may be used in the pharmaceutical compositions of this invention 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 phosphat
  • Typical pharmaceutical compositions and dosage forms comprise one or more excipients.
  • Suitable excipients are well known to those skilled in the art of pharmacy, and non-limiting examples of suitable excipients are provided herein. Whether a particular excipient is suitable for incorporation into a pharmaceutical composition or dosage form depends on a variety of factors well known in the art including, but not limited to, the way in which the dosage form will be administered to a patient. Excipients can take a wide variety of forms depending on the form of preparation desired for administration.
  • excipients suitable for use in oral liquid or aerosol dosage forms include, but are not limited to, water, glycols, oils, alcohols, flavoring agents, preservatives, and coloring agents.
  • excipients suitable for use in solid oral dosage forms include, but are not limited to, starches, sugars, micro-crystalline cellulose, diluents, granulating agents, lubricants, binders, and disintegrating agents.
  • Such preparative methods include the step of bringing into association with the molecule to be administered ingredients such as the carrier that constitutes one or more accessory ingredients.
  • ingredients such as the carrier that constitutes one or more accessory ingredients.
  • the compositions are prepared by uniformly and intimately bringing into association the active ingredients with liquid carriers, liposomes or finely divided solid carriers or both, and then if necessary shaping the product.
  • compositions according to the invention are those suitable for enteral, such as oral or rectal, transdermal, topical, and parenteral administration to mammals, including man, to inhibit Bcr-abl activity, and for the treatment of Bcr-abl dependent disorders, and comprise an effective amount of a pharmacologically active compound of the present invention, alone or in combination, with one or more pharmaceutically acceptable carriers. More particularly, the pharmaceutical compositions comprise an effective Bcr-abl inhibiting amount of a compound of the present invention.
  • Single unit dosage forms of the invention are suitable for oral, mucosal (e.g., nasal, sublingual, vaginal, buccal, or rectal), parenteral (e.g., subcutaneous, intravenous, bolus injection, intramuscular, intraarterial, intracutaneous, intraarticular, intrasynovial, intrasternal, intrathecal, intralesional and intracranial injection or infusion techniques.), by inhalation spray, topically, rectally, nasally, buccally, vaginally, via an implanted reservoir, or transdermal administration to a patient.
  • parenteral e.g., subcutaneous, intravenous, bolus injection, intramuscular, intraarterial, intracutaneous, intraarticular, intrasynovial, intrasternal, intrathecal, intralesional and intracranial injection or infusion techniques.
  • inhalation spray topically, rectally, nasally, buccally, vaginally, via an implanted reservoir, or transdermal administration to a patient.
  • dosage forms include, but are not limited to: tablets; caplets; capsules, such as soft elastic gelatin capsules; cachets; troches; lozenges; dispersions; suppositories; ointments; cataplasms (poultices); pastes; powders; dressings; creams; plasters; solutions; patches; aerosols (e.g., nasal sprays or inhalers); gels; liquid dosage forms suitable for oral or mucosal administration to a patient, including suspensions (e.g., aqueous or non-aqueous liquid suspensions, oil-in-water emulsions, or a water-in-oil liquid emulsions), solutions, and elixirs; liquid dosage forms suitable for parenteral administration to a patient; and sterile solids (e.g., crystalline or amorphous solids) that can be reconstituted to provide liquid dosage forms suitable for parenteral administration to a patient.
  • suspensions e.g., aqueous
  • composition, shape, and type of dosage forms of the invention will typically vary depending on their use.
  • a dosage form used in the acute treatment of a disorder may contain larger amounts of one or more of the active ingredients it comprises than a dosage form used in the chronic treatment of the same disease.
  • a parenteral dosage form may contain smaller amounts of one or more of the active ingredients it comprises than an oral dosage form used to treat the same disease or disorder.
  • a composition for oral administration can be any orally acceptable dosage form including, but not limited to, capsules, tablets, emulsions and aqueous suspensions, dispersions and solutions.
  • carriers which are commonly used include lactose and corn starch.
  • Lubricating agents, such as magnesium stearate, are also typically added.
  • useful diluents include lactose and dried corn starch.
  • a nasal aerosol or inhalation composition can be prepared according to techniques well-known in the art of pharmaceutical formulation and can be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other solubilizing or dispersing agents known in the art.
  • a heterocyclic compound of this invention can also be administered in the form of suppositories for rectal administration.
  • tablets can be coated by standard aqueous or nonaqueous techniques.
  • dosage forms can be prepared by any of the methods of pharmacy.
  • pharmaceutical compositions and dosage forms are prepared by uniformly and intimately admixing the active ingredients with liquid carriers, finely divided solid carriers, or both, and then shaping the product into the desired presentation if necessary.
  • tablets and gelatin capsules comprising the active ingredient together with a) diluents, e. g., lactose, dextrose, sucrose, mannitol, sorbitol, cellulose and/or glycine; b) lubricants, e. g., silica, talcum, stearic acid, its magnesium or calcium salt and/or polyethyleneglycol; for tablets also c) binders, e. g., magnesium aluminum silicate, starch paste, gelatin, tragacanth, methylcellulose, sodium carboxymethylcellulose and or polyvinylpyrrolidone; if desired d) disintegrants, e.
  • diluents e. g., lactose, dextrose, sucrose, mannitol, sorbitol, cellulose and/or glycine
  • lubricants e. g., silica, talcum, stearic acid, its
  • Tablets may be either film coated or enteric coated according to methods known in the art.
  • a tablet can be prepared by compression or molding.
  • Compressed tablets can be prepared by compressing in a suitable machine the active ingredients in a free-flowing form such as powder or granules, optionally mixed with an excipient.
  • Molded tablets can be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent.
  • compositions are preferably aqueous isotonic solutions or suspensions, and suppositories are preferably prepared from fatty emulsions or suspensions.
  • the compositions may be sterilized and/or contain adjuvants, such as preserving, stabilizing, wetting or emulsifying agents, solution promoters, salts for regulating the osmotic pressureand/or buffers. In addition, they may also contain other therapeutically valuable substances.
  • Said compositions are prepared according to conventional mixing, granulating or coating methods, respectively, and contain about 0.1 to 75%, preferably about 1 to 50%, of the active ingredient.
  • a sterile injectable composition for example, a sterile injectable aqueous or oleaginous suspension, can be formulated according to techniques known in the art using suitable dispersing or wetting agents (such as, for example, Tween 80) and suspending agents.
  • the sterile injectable preparation can also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent.
  • suitable vehicles and solvents that can be employed are mannitol, water, Ringer's solution and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium (e.g., synthetic mono- or diglycerides).
  • 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.
  • oils such as olive oil or castor oil, especially in their polyoxyethylated versions.
  • These oil solutions or suspensions can also contain a long-chain alcohol diluent or dispersant, or carboxymethyl cellulose or similar dispersing agents.
  • Other commonly used surfactants such as Tweens or Spans or other similar emulsifying agents or bioavailability enhancers which are commonly used in the manufacture of pharmaceutically acceptable solid, liquid, or other dosage forms can also be used for the purposes of formulation.
  • Suitable formulations for transdermal application include an effective amount of a compound of the present invention with carrier.
  • Preferred carriers include absorbable pharmacologically acceptable solvents to assist passage through the skin of the host.
  • transdermal devices are in the form ofa bandage comprising a backing member, a reservoir containing the compound optionally with carriers, optionally a rate controlling barrier to deliver the compound to the skin of the host at a controlled and predetermined rate over a prolonged period of time, and means to secure the device to the skin.
  • Matrix transdermal formulations may also be used.
  • Suitable formulations for topical application are preferably aqueous solutions, ointments, creams or gels well-known in the art.
  • aqueous solutions e.g., ointments, creams or gels well-known in the art.
  • Such may containsolubilizers, stabilizers, tonicity enhancing agents, buffers and preservatives.
  • the pharmaceutical formulations contain an effective Bcr-abl inhibiting amount of a compound of the present invention as defined above, either alone or in combination with another therapeutic agent.
  • a compound of the present invention may be administered either simultaneously, before or after the other active ingredient, either separately by the same or different route of administration or together in the same pharmaceutical formulation.
  • the carrier in the pharmaceutical composition must be "acceptable” in the sense of being compatible with the active ingredient of the formulation (and preferably, capable of stabilizing it) and not deleterious to the subject to be treated.
  • solubilizing agents such as cyclodextrins, which form specific, more soluble complexes with the compounds of this invention, or one or more solubilizing agents, can be utilized as pharmaceutical excipients for delivery of the compounds of the invention.
  • examples of other carriers include colloidal silicon dioxide, magnesium stearate, cellulose, sodium lauryl sulfate, and D&C Yellow # 10.
  • animal As used herein, the terms "animal”, “subject” and “patient”, include, but are not limited to, a cow, monkey, horse, sheep, pig, chicken, turkey, quail, cat, dog, mouse, rat, rabbit, guinea pig and human (preferably, a human).
  • the pharmacologically active compounds of the present invention are useful in the manufacture of pharmaceutical compositions comprising an effective amount thereof in conjunction or mixture with excipients or carriers suitable for either enteral, parenteral, oral, mucosal, or topical application.
  • Body surface area may be approximately determined from height and weight of the patient. See, e.g., Scientific Tables, Geigy Pharmaceuticals, Ardley, N. Y., 1970, 537.
  • An effective amount of a compound of this invention can range from about 0.001 mg/kg to about 1000 mg/kg, more preferably 0.01 mg/kg to about 100 mg/kg, more preferably 0.1 mg/kg to about 10 mg/kg; or any range in which the low end of the range is any amount between 0.001 mg/kg and 900 mg/kg and the upper end of the range is any amount between 0.1 mg/kg and 1000 mg/kg (e.g., 0.005 mg/kg and 200 mg/kg, 0.5 mg/kg and 20 mg/kg).
  • Effective doses will also vary, as recognized by those skilled in the art, depending on the diseases treated, route of administration, excipient usage, and the possibility of co-usage with other therapeutic treatments such as use of other agents.
  • the dosage of active compound administered is dependent on the species of warmblooded animal (mammal), the body weight, age and individual condition, and on the form of administration.
  • a unit dosage for oral administration to a mammal of about 50 to 70 kg may contain between about 5 and 500 mg of the active ingredient.
  • a compound of the invention can, for example, be administered with a dosage ranging from about 0.5 to about 100 mg/kg of body weight, alternatively dosages between 1 mg and 1000 mg/dose, every 4 to 120 hours, or according to the requirements of the particular drug.
  • the methods herein contemplate administration of an effective amount of compound or compound composition to achieve the desired or stated effect.
  • the pharmaceutical compositions of this invention will be administered from about 1 to about 6 times per day or alternatively, as a continuous infusion. Such administration can be used as a chronic or acute therapy.
  • the amount of active ingredient that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration.
  • a typical preparation will contain from about 5% to about 95% active compound (w/w). Alternatively, such preparations contain from about 20% to about 80% active compound.
  • alkyl refers to a straight-chained or branched hydrocarbon group containing 1 to 12 carbon atoms.
  • the term “lower alkyl” refers to a C1-C6 alkyl chain. Examples of alkyl groups include methyl, ethyl, n-propyl, isopropyl, tert-butyl, and n-pentyl. Alkyl groups may be optionally substituted with one or more substituents.
  • alkenyl refers to an unsaturated hydrocarbon chain that may be a straight chain or branched chain, containing 2 to 12 carbon atoms and at least one carbon-carbon double bond. Alkenyl groups may be optionally substituted with one or more substituents.
  • alkynyl refers to an unsaturated hydrocarbon chain that may be a straight chain or branched chain, containing the 2 to 12 carbon atoms and at least one carbon-carbon triple bond. Alkynyl groups may be optionally substituted with one or more substituents.
  • the sp 2 or sp carbons of an alkenyl group and an alkynyl group, respectively, may optionally be the point of attachment of the alkenyl or alkynyl groups.
  • alkoxy refers to an -O-alkyl radical.
  • an “amido” is an -C(O)NH 2
  • an “N-alkyl-substituted amido” is of the formula C(O)NHR; where a divalent "amide” group is indicated, the group is -C(O)N- or -NC(O)-.
  • halogen means -F, -Cl, -Br or -I.
  • haloalkyl means an alkyl group in which one or more (including all) of the hydrogen radicals are replaced by a halo group, wherein each halo group is independently selected from -F, -Cl, -Br, and -I.
  • halomethyl means a methyl in which one to three hydrogen radical(s) have been replaced by a halo group.
  • Representative haloalkyl groups include trifluoromethyl, bromomethyl, 1,2-dichloroethyl, 4-iodobutyl, 2- fluoropentyl, and the like.
  • cycloalkyl refers to a hydrocarbon 3-8 membered monocyclic or 7-14 membered bicyclic ring system having at least one saturated ring, or hydrocarbon 3-8 membered monocyclic or 7-14 membered bicyclic ring system having at least one non- aromatic ring, wherein the non-aromatic ring has some degree of unsaturation.
  • Cycloalkyl groups may be optionally substituted with one or more substituents. In one embodiment, 0, 1, 2, 3, or 4 atoms of each ring of a cycloalkyl group may be substituted by a substituent.
  • cycloalkyl group include cyclopropyl, cyclopentyl, cyclohexyl, cyclobutyl, cycloheptyl, cyclooctyl, cyclononyl, cyclodecyl, cyclohexenyl, bicyclo[2.2.1]hept-2-enyl, dihydronaphthalenyl, benzocyclopentyl, cyclopentenyl, cyclopentadienyl, cyclohexenyl, cyclohexadienyl,cycloheptenyl, cycloheptadienyl, cycloheptatrienyl, cyclooctenyl, cyclooctadienyl, cyclooctatrienyl, cyclooctatetraenyl, cyclononenyl, cyclononadienyl, cyclodecenyl, cyclodecadie
  • aryl refers to a hydrocarbon monocyclic, bicyclic or tricyclic aromatic ring system.
  • Aryl groups may be optionally substituted with one or more substituents. In one embodiment, 0, 1, 2, 3, 4, 5 or 6 atoms of each ring of an aryl group may be substituted by a substituent. Examples of aryl groups include phenyl, naphthyl, anthracenyl, fluorenyl, indenyl, azulenyl, and the like.
  • aralkyl means an aryl group that is attached to another group by a (d-C 6 )alkylene group.
  • Aralkyl groups may be optionally substituted, either on the aryl portion of the aralkyl group or on the alkylene portion of the aralkyl group, with one or more substituents.
  • Representative aralkyl groups include benzyl, 2-phenyl-ethyl, naphth- 3-yl-methyl and the like.
  • alkylene refers to an alkyl group that has two points of attachment.
  • (Ci-C 6 )alkylene refers to an alkylene group that has from one to six carbon atoms.
  • alkylene groups include methylene (-CH 2 -), ethylene (-CH 2 CH 2 -), n-propylene (-CH 2 CH 2 CH 2 -), isopropylene (-CH 2 CH(CH 3 )-), and the like.
  • arylalkoxy refers to an alkoxy substituted with aryl.
  • heteroaryl refers to a 5-8 membered monocyclic, 8-12 membered bicyclic, or 11-14 membered tricyclic ring system having 1-4 ring heteroatoms if monocyclic, 1-6 heteroatoms if bicyclic, or 1-9 heteroatoms if tricyclic, said heteroatoms selected from O, N, or S, and the remainder ring atoms being carbon (with appropriate hydrogen atoms unless otherwise indicated), wherein at least one ring in the ring system is aromatic.
  • Heteroaryl groups may be optionally substituted with one or more substituents. In one embodiment, 0, 1, 2, 3, or 4 atoms of each ring of a heteroaryl group may be substituted by a substituent.
  • heteroaryl groups include pyridyl, 1-oxo-pyridyl, furanyl, benzo[l,3]dioxolyl, benzo[l,4]dioxinyl, thienyl, pyrrolyl, oxazolyl, oxadiazolyl, imidazolyl thiazolyl, isoxazolyl, quinolinyl, pyrazolyl, isothiazolyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl, triazolyl, thiadiazolyl, isoquinolinyl, indazolyl, benzoxazolyl, benzofuryl, indolizinyl, imidazopyridyl, tetrazolyl, benzimidazolyl, benzothiazolyl, benzothiadiazolyl, benzoxadiazolyl, indolyl, tetra
  • heteroarylkyl or “heteroarylalkyl” means a heteroaryl group that is attached to another group by a (Ci-C 6 )alkylene.
  • Heteroaralkyl groups may be optionally substituted, either on the heteroaryl portion of the heteroaralkyl group or on the alkylene portion of the heteroaralkyl group, with one or more substituent.
  • Representative heteroaralkyl groupss include 2-(pyridin-4-yl)-propyl, 2-(thien-3-yl)-ethyl, imidazol-4-yl- methyl and the like.
  • heterocycloalkyl refers to a nonaromatic 3-8 membered monocyclic, 7-12 membered bicyclic, or 10-14 membered tricyclic ring system comprising 1-3 heteroatoms if monocyclic, 1-6 heteroatoms if bicyclic, or 1-9 heteroatoms if tricyclic, said heteroatoms selected from O, N, S, B, P and Si, preferably O, N, and S, wherein the nonaromatic ring system is completely saturated.
  • heterocycloalkyl also refers to nonaromatic 5-8 membered monocyclic, 7-12 membered bicyclic, or 10-14 membered tricyclic ring system comprising 1-3 heteroatoms if monocyclic, 1-6 heteroatoms if bicyclic, or 1-9 heteroatoms if tricyclic, said heteroatoms selected from O, N, S, B, P or Si, wherein the nonaromatic ring system has some degree of unsaturation.
  • Bicyclic and tricyclic ring systems may be fused ring systems or spiro ring systems.
  • Heterocycloalkyl groups may be optionally substituted with one or more substituents.
  • heterocycloalkyl groups include piperidinyl, piperazinyl, 2-oxopiperazinyl, 2-oxopiperidinyl, 2- oxopyrrolidinyl, 4-piperidonyl, tetrahydropyranyl, tetrahydrothiopyranyl, tetrahydrothiopyranyl sulfone, morpholinyl, thiomorpholinyl, thiomorpholinyl sulfoxide, thiomorpholinyl sulfone, 1,3-dioxolane, l,4-dioxa-8-aza-spiro[4.5]dec-8-yl, tetrahydrofuranyl, tetrahydrothienyl, thiirene, thiirenyl, thiadiazirinyl,
  • alkylamino refers to an amino substituent which is further substituted with one or two alkyl groups.
  • aminoalkyl refers to an alkyl substituent which is further substituted with one or more amino groups.
  • an effective amount is used throughout the specification to describe concentrations or amounts of compounds according to the present invention which may be used to produce a favorable change in the disease or condition treated, whether that change is a remission, a decrease in growth or size of cancer, tumor or other growth, a favorable physiological result including the clearing up of skin or tissue, or the like, depending upon the disease or condition treated.
  • the terms "prevent,” “preventing,” “prevention,” and the like refer to reducing the probability of developing a disorder or condition in a subject, who does not have, but is at risk of or susceptible to developing a disorder or condition.
  • subject is used throughout the specification to describe an animal, preferably a human, to whom treatment, including prophylactic treatment, with the compounds according to the present invention is provided.
  • treatment including prophylactic treatment, with the compounds according to the present invention is provided.
  • patient refers to that specific animal. In most instances, the term patient refers to a human patient.
  • IC 50 is the concentration of a compound that results in 50% inhibition of activity of a peptide, protein, enzyme or biological process.
  • Myristoyl Binding Pocket is a region of Bcr-abl at which a myristoyl moiety can bind when the BCR-AbI protein is in an appropriate conformation for myristoyl binding.
  • Myristoyl binding pockets are described in, for example, Hantschel et al.,"A Myristoyl/Phosphotyrosine Switch Regulates c-Abl” Cell (2003), Vol. 112, 845-857 and Bhushan et al.,” Structural Basis for the Autoinlaibition of c-Abl Tyrosine Kinase” Cell (2003), Vol. 1 12, 859-871.
  • the fusion protein Bcr- AbI is a result of a reciprocal translocation that fuses the AbI proto-oncogene with the Bcr gene.
  • Bcr-abl is then capable of transforming B-cells through the increase of mitogenic activity. This increase results in a reduction of sensitivity to apoptosis, as well as altering the adhesion and homing of CML progenitor cells.
  • the present invention provides compounds, compositions and methods for the treatment of kinase related disease, particularly PDGF-R, c-Kit and Bcr-abl kinase related diseases.
  • substituted means that a hydrogen radical on a compound or group (such as, for example, alkyl, alkenyl, alkynyl, alkylene, aryl, aralkyl, heteroaryl, heteroaralkyl, cycloalkyl, or heterocycloalkyl group) is substituted or optionally substituted with any desired group that do not substantially adversely affect the stability of the compound.
  • desired substituents are those which do not adversely affect the activity of a compound.
  • substituted refers to one or more substituents (which may be the same or different), each replacing a hydrogen atom.
  • substituents include, but are not limited to, halogen (F, Cl, Br, or I), hydroxyl, amino, alkylamino, arylamino, dialkylamino, diarylamino, alkylarylamino, cyano, nitro, mercapto, thio, imino, formyl, carbamido, carbamyl, carboxyl, thioureido, thiocyanato, sulfoamido, sulfonylalkyl, sulfonylaryl, alkyl, alkenyl, alkoxy, mercaptoalkoxy, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, wherein alkyl, alkenyl, alkyloxy, alkoxyalkyl, aryl, heteroaryl, cycloalkyl, are heterocycloalkyl are optionally substituted with alkyl, aryl, heteroaryl, halogen,
  • substituents on any group can be at any atom of that group, wherein any group that can be substituted (such as, for example, alkyl, alkenyl, alkynyl, aryl, aralkyl, heteroaryl, heteroaralkyl, cycloalkyl, and heterocycloalkyl) can be optionally substituted with one or more substituents (which may be the same or different), each replacing a hydrogen atom.
  • substituents include, but not limited to alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aralkyl, heteroaralkyl, aryl, heteroaryl, halogen, haloalkyl, cyano, nitro, alkoxy, aryloxy, hydroxyl, hydroxylalkyl, oxo (i.e., carbonyl), carboxyl, formyl, alkylcarbonyl, alkylcarbonylalkyl, alkoxycarbonyl, alkylcarbonyloxy, aryloxycarbonyl, heteroaryloxy, heteroaryloxycarbonyl, thio, mercapto, mercaptoalkyl, arylsulfonyl, amino, aminoalkyl, dialkylamino, alkylcarbonylamino, alkylaminocarbonyl, or alkoxy carbonylamino; alkylamino, arylamino, diary
  • Each R 15 is independently hydrogen, Ci-C 6 alkyl optionally substituted with cycloalkyl, aryl, heterocycloalkyl, or heteroaryl.
  • Each R 16 is independently hydrogen, C 3 -C 6 cycloalkyl, aryl, heterocycloalkyl, heteroaryl, CpC 4 alkyl or Ci-C 4 alkyl substituted with C 3 - C 6 cycloalkyl, aryl, heterocycloalkyl or heteroaryl.
  • Each R 17 is independently C 3 -C 6 cycloalkyl, aryl, heterocycloalkyl, heteroaryl, Ci-C 4 alkyl or Ci-C 4 alkyl substituted with C 3 - C 6 cycloalkyl, aryl, heterocycloalkyl or heteroaryl.
  • Each C 3 -C 6 cycloalkyl, aryl, heterocycloalkyl, heteroaryl and Ci-C 4 alkyl in each R 15 , R 16 and R 17 can optionally be substituted with halogen, CN, Ci-C 4 alkyl, OH, Ci-C 4 alkoxy, COOH, C(O)OCi-C 4 alkyl, NH 2 , Ci-C 4 alkylamino, or C]-C 4 dialkylamino.
  • a compound of the invention may be optionally substituted with (i) alkyl, haloalkyl, aryl, halogen, hydroxyl, alkoxy, hydroxyalkyl, amino, mono-alkyl amine, di-alkyl amine, cyano, CONH 2 , C0 2 alkyl, SO 2 NH 2 , or (ii) heterocyclic or heteroaryl, selected from the following: morpholine,
  • an optionally substituted aryl or heteroaryl may be selected from the following: an optionally substituted phenyl, an optionally substituted naphthyl, an optionally substituted anthracenyl, an optionally substituted fluorenyl, an optionally substituted indenyl, an optionally substituted azulenyl, an optionally substituted pyridyl, an optionally substituted 1-oxo-pyridyl, an optionally substituted furanyl, an optionally substituted benzo[l,3]dioxolyl, an optionally substituted benzo[l,4]dioxinyl, an optionally substituted thienyl, an optionally substituted pyrrolyl, an optionally substituted oxazolyl, an optionally substituted imidazolyl, an optionally substituted thiazolyl, an optionally substituted isoxazolyl, an optionally substituted quinolinyl, an optionally substituted pyrazoly
  • an optionally substituted heterocycloalkyl may be selected from the following: an optionally substituted piperidinyl, an optionally substituted piperazinyl, an optionally substituted 2-oxopiperazinyl, an optionally substituted 2- oxopiperidinyl, an optionally substituted 2-oxopyrrolidinyl, an optionally substituted 4- piperidonyl, an optionally substituted tetrahydropyranyl, an optionally substituted oxazolidinyl, an optionally substituted 2-oxo-oxazolidinyl, an optionally substituted tetrahydrothiopyranyl, an optionally substituted tetrahydrothiopyranyl sulfone, an optionally substituted morpholinyl, an optionally substituted thiomorpholinyl, an optionally substituted thiomorpholinyl sulfoxide, an optionally substituted thiomorpholinyl sulfone, an optionally substituted 1,
  • the compounds of this invention include the compounds themselves, as well as their salts, solvate, clathrate, hydrate, polymorph, or prodrugs, if applicable.
  • the term "pharmaceutically acceptable salt,” is a salt formed from, for example, an acid and a basic group of a compound of any one of the formulae disclosed herein.
  • Illustrative salts include, but are not limited, to sulfate, citrate, acetate, oxalate, chloride, bromide, iodide, nitrate, bisulfate, phosphate, acid phosphate, isonicotinate, lactate, salicylate, acid citrate, tartrate, oleate, tannate, pantothenate, bitartrate, ascorbate, succinate, maleate, besylate, gentisinate, fumarate, gluconate, glucaronate, saccharate, formate, benzoate, glutamate, methanesulfonate, ethanesulfonate, benzenesulfonate, p- toluenesulfonate, and pamoate ⁇ i
  • Suitable bases include, but are not limited to, hydroxides of alkali metals such as sodium, potassium, and lithium; hydroxides of alkaline earth metal such as calcium and magnesium; hydroxides of other metals, such as aluminum and zinc; ammonia, and organic amines, such as unsubstituted or hydroxy-substituted mono-, di-, or trialkylamines; dicyclohexylamine; tributyl amine; pyridine; N-methyl,N-ethylamine; diethylamine; triethylamine; mono-, bis-, or tris-(2-hydroxy-lower alkyl amines), such as mono-, bis-, or tris-(2-hydroxyethyl)amine, 2- hydroxy
  • pharmaceutically acceptable salt also refers to a salt prepared from a compound of any one of the formulae disclosed herein having a basic functional group, such as an amino functional group, and a pharmaceutically acceptable inorganic or organic acid.
  • suitable acids include hydrogen sulfate, citric acid, acetic acid, oxalic acid, hydrochloric acid (HCl), hydrogen bromide (HBr), hydrogen iodide (HI), nitric acid, hydrogen bisulfide, phosphoric acid, lactic acid, salicylic acid, tartaric acid, bitartratic acid, ascorbic acid, succinic acid, maleic acid, besylic acid, fumaric acid, gluconic acid, glucaronic acid, formic acid, benzoic acid, glutamic acid, methanesulfonic acid, ethanesulfonic acid, benzenesulfonic acid, and/7-toluenesulfonic acid.
  • polymorph means solid crystalline forms of a compound of the present invention or complex thereof. Different polymorphs of the same compound can exhibit different physical, chemical and/or spectroscopic properties. Different physical properties include, but are not limited to stability (e.g., to heat or light), compressibility and density (important in formulation and product manufacturing), and dissolution rates (which can affect bioavailability).
  • Differences in stability can result from changes in chemical reactivity (e.g., differential oxidation, such that a dosage form discolors more rapidly when comprised of one polymorph than when comprised of another polymorph) or mechanical characteristics (e.g., tablets crumble on storage as a kinetically favored polymorph converts to thermodynamically more stable polymorph) or both (e.g., tablets of one polymorph are more susceptible to breakdown at high humidity).
  • Different physical properties of polymorphs can affect their processing. For example, one polymorph might be more likely to form solvates or might be more difficult to filter or wash free of impurities than another due to, for example, the shape or size distribution of particles of it.
  • the term "hydrate” means a compound of the present invention or a salt thereof, which further includes a stoichiometric or non-stoichiometric amount of water bound by non-covalent intermolecular forces.
  • clathrate means a compound of the present invention or a salt thereof in the form of a crystal lattice that contains spaces (e.g., channels) that have a guest molecule (e.g., a solvent or water) trapped within.
  • spaces e.g., channels
  • guest molecule e.g., a solvent or water
  • prodrug means a derivative of a compound that can hydrolyze, oxidize, or otherwise react under biological conditions (in vitro or in vivo) to provide a compound of this invention. Prodrugs may only become active upon such reaction under biological conditions, or they may have activity in their unreacted forms.
  • prodrugs contemplated in this invention include, but are not limited to, analogs or derivatives of compounds of any one of the formulae disclosed herein that comprise biohydrolyzable moieties such as biohydrolyzable amides, biohydrolyzable esters, biohydrolyzable carbamates, biohydrolyzable carbonates, biohydrolyzable ureides, and biohydrolyzable phosphate analogues.
  • biohydrolyzable moieties such as biohydrolyzable amides, biohydrolyzable esters, biohydrolyzable carbamates, biohydrolyzable carbonates, biohydrolyzable ureides, and biohydrolyzable phosphate analogues.
  • Other examples of prodrugs include derivatives of compounds of any one of the formulae disclosed herein that comprise -NO, -NO 2 , -ONO, or - ONO 2 moieties.
  • Prodrugs can typically be prepared using well-known methods, such as those described by 1 BURGER'S MEDICINAL CHEMISTRY AND DRUG DISCOVERY (1995) 172- 178, 949-982 (Manfred E. Wolff ed., 5 th ed).
  • some of the compounds of this invention have one or more double bonds, or one or more asymmetric centers. Such compounds can occur as racemates, racemic mixtures, single enantiomers, individual diastereomers, diastereomeric mixtures, and cis- or trans- or E- or Z- double isomeric forms. All such isomeric forms of these compounds are expressly included in the present invention.
  • the compounds of this invention may also be represented in multiple tautomeric forms, in such instances, the invention expressly includes all tautomeric forms of the compounds described herein (e.g., alkylation of a ring system may result in alkylation at multiple sites, the invention expressly includes all such reaction products). All such isomeric forms of such compounds are expressly included in the present invention. All crystal forms of the compounds described herein are expressly included in the present invention.
  • Example 1 Preparation of 2-(4-(4-(trifiuoromethoxy)phenylamino)-lH-pyrazolo[3,4- d]pyrimidin-l-yl)ethanol and 2-(4-(4-(trifluoromethoxy)phenylamino)-2H-pyrazolo[3,4- d]pyrimidin-2-yl)ethanol
  • Step 1 4-Chloro-6-(4-trifluoromethoxy-phenylamino)-pyrimidine-5-carbaldehyde:
  • Step 2 2-(4-(4-(trifiuoromethoxy)phenylamino)- 1 H-pyrazolo[3 ,4-d]pyrimidin- 1 -yl)ethanol and 2-(4-(4-(trifluoromethoxy)phenylamino)-2H-pyrazolo[3,4-d]pyrimidin-2-yl)ethanol:
  • 4-chloro-6-(4-trifluoromethoxy-phenylamino)-pyrimidine-5-carbaldehyde 50 g, 0.15 mmol
  • Na 2 CO 3 30 mg, 0.26 mmol
  • hydrazine 20 mg, 0.18 mmol
  • reaction was stirred at 60 0 C temperature for 8 hours.
  • the reaction mixture was partitioned in 100 ml water and 100 ml ethyl acetate, extracted with ethyl acetate three times.
  • the organic phase was combined and washed with brine, dried over Na 2 SO 4 .
  • the crude product was purified by silica gel flash chromatography by using hexane:ethyl acetate as eluent resulting 2-(4-(4-(trifluoromethoxy)phenylamino)-lH-pyrazolo[3,4-d]pyrimidin-l- yl)ethanol and 2-(4-(4-(trifluoromethoxy)phenylamino)-2H-pyrazolo[3,4-d]pyrimidin-2- yl)ethanol in the ratio of 4:1 as yellow solids.
  • Step 1 3-(4-Chloro-pyrrolo[2,3-d]pyrimidin-7-yl)-propionic acid ethyl ester:
  • Step 2 3-[4-(4-Trifluoromethoxy-phenylamino)-pyrrolo[2,3-d]pyrimidin-7-yl]-propionic acid ethyl ester:
  • Step 3 3-[4-(4-Trifluoromethoxy-phenylamino)-pyrrolo[2,3-d]pyrimidin-7-yl]-propionic acid:
  • Step 4 N-(2-Morpholin-4-yl-ethyl)-3-[4-(4-trifluoromethoxy-phenylamino)-pyrrolo[2,3- d]pyrimidin-7-yl] -propionamide :
  • Step 1 Methyl 4-(4-(trifluoromethoxy)phenylamino)thieno[2,3-d]pyrimidine-6-carboxylate:
  • Step 2 4-(4-(trifluoromethoxy)phenylamino)thieno[2,3-d]pyrimidine-6-carboxylic acid:
  • Step 3 N-(2-hydroxyethyl)-4-(4-(trifluoromethoxy)phenylamino)thieno[2,3-d]pyrimidine-6- carboxamide:
  • Step 1 Methyl 4-(4-(trifluoromethoxy)phenylamino)quinazoline-7-carboxylate:
  • Step 2 4-(4-(trifluoromethoxy)phenylamino)quinazoline-7-carboxylic acid:
  • Step 3 4-(4-(trifluoromethoxy)phenylamino)-N-(2-hydroxyethyl)quinazoline-7-carboxamide: 4-(4-(trifluoromethoxy)phenylamino)quinazoline-7-carboxylic acid (30 mg, 0.09 mmol) was mixed with DIEA (30 uL, 0.172 mmol) and HATU (39 mg, 0.1 mmol) in 2 ml DMF at room temperature. 2-Aminoethanol (6.1 mg, 0.1 mmol) was added into the reaction mixture 0.5 hour later. After stirring at room temperature for 2 hours, the reaction mixture was concentrated and purified by prep-HPLC to afford the title compound as TFA salt (29 mg, 82 % Yield). MS m/z 393.04 (M + 1).
  • Step 1 6-chloro-5-methyl-N-(4-(trifluoromethoxy)phenyl)pyrimidin-4-amine:
  • Step 2 4-(6-(4-(trifluoromethoxy)phenylamino)-5-methylpyrimidin-4-yl)benzoic acid:
  • Step 3 4-(6-(4-(trifluoromethoxy)phenylamino)-5-methylpyrimidin-4-yl)-N-(2- morpholinoethyl) benzamide:
  • Step 1 6-chloro-N-(4-(trifluoromethoxy)phenyl)pyrimidin-4-amine:
  • Step 2 6-( 1 H-pyrrolo [2,3 -bjpyridin- 1 -yl)-N-(4-(trifluoromethoxy)phenyl)pyrirnidin-4-amine : To a solution of compound 7-azaindole (18 mg, 0.15 mmol) in sec-BuOH (5 mL) was added DIEA (0.20 mL) and the reaction mixture was stirred for 15 min at RT. 6-Chloro-N-(4- (trifluoromethoxy)phenyl)pyrimidin-4-amine (29 mg, 0.1 mmol) was then added and the temperature was raised to 90 0 C and continued stirring for another 16 h. The solvent was removed and the title compound was purified by prep. HPLC (15 mg, 40 % Yield). MS m/z 372.10 (M + 1).
  • 6-Chloro-N-(4-(trifluoromethoxy)phenyl)pyrimidin-4-aniine intermediate (5.2 mg, 0.17 mmol), l,3,5-trimethyl-4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-lH-pyrazole (40 mg, 0.17 mmol), Pd(PPh 3 )., (10 mg, 0.085 mmol) and sodium carbonate (73 mg, 0.69 mmol) were dissolved in acetonitrile: water (v/v 1 :1, 10 ml). The flask was stirred at 95 °C for 5 hr.
  • Step 1 7-chloro-2-(3-nitrophenyl)imidazo[l,2-c]pyrimidine
  • 6-Chloropyrimidin-4-amine 50 mg, 0.39 mmol
  • 2-bromo-l-(3-nitrophenyl)ethanone 94 mg, 0.39 mmol
  • the reaction mixture was poured into 5 mL of water and it was then extracted with ethyl acetate.
  • the filtrate was dried (Na2SO4) and evaporated.
  • the compound was purified by silica gel column chromatography by using hexane/ethyl acetate (3:1) as eluent (60 mg, 56 % Yield). MS m/z 275.00 (M + 1).
  • Step 2 2-(3-Nitrophenyl)-N-(4-(trifluoromethoxy)phenyl)imidazo[l ,2-c]pyrimidin-7-amine
  • Interleukin-3 (IL-3) dependent murine pro-B cell line, Ba/F3, and murine myeloid precursor cell line, 32D were maintained in RPMI- 1640 medium supplemented with L-Glutamine, 10% FBS and 10 U/ml recombinant murine IL-3 (Roche).
  • Mo7e human megakarioblastic cell line was grown in RPMI- 1640 medium supplemented with L-Glutamine, 20% FBS and 5 ng/ml recombinant human granulocyte macrophage cell stimulating factor (GM-CSF) (R&D Systems) or 200 ng/ml stem cell factor (SCF) (Biosource).
  • GM-CSF granulocyte macrophage cell stimulating factor
  • SCF stem cell factor
  • Additional assays included Ba/F3 cells expressing the fusion protein kinases TEL- PDGFR ⁇ , TEL-JAKl, NPM-ALK, TEL-cKit, Tel-Bmx, Tel-FGR3, Tel-Lck and Ba/F3.TPR-met cells. All the above cell lines were maintained in RPMI- 1640 medium with L-Glutamine, 10% FBS and 1 mg/ml geneticin (Gibco).
  • Human leukemic cell lines K562 (p210 Bcr-abl expressing chronic myelogenous leukemia), HL-60 (acute promyelocyte leukemia), SUP/B15 (pi 90 Bcr-abl expressing acute lymphoblastic leukemia) and Jurkat (acute T cell leukemia) were purchased from the American Type Culture Collection (ATCC) and cultured following ATCC recommendations.
  • ATCC American Type Culture Collection
  • Cells grown for 24 or 48 hours in medium containing different concentrations of compound were collected by centrifugation (10 min, 4°C), washed twice with PBS and fixed in 70% ethanol at -20°C. After fixation, ethanol was removed by centrifugation (10 min, 4°C), and the cells were resuspended in PBS containing ImM EDTA and 100 ⁇ g/ml RNAse A (Sigma) and incubated 30 min at 37°C. Cells were stained with propidium iodide (10 ⁇ g/ml) and DNA content was determined by flow cytometry (FACScan, Becton Dickinson).
  • Bcr-abl expressing cells treated for 90 min with various concentrations of test compounds were homogenized in lysis buffer (50 mM Tris.HCl pH7.4, 150 mM NaCl, 5 mM EDTA, 1 mM EGTA, 1% NP-40, 2 mM Na 3 VO 4 and protease inhibitor cocktail (Roche)) and the lysates plated on 96 well plates containing adsorbed anti-Abl SH3 domain polyclonal antibody (Upstate Biotechnology). The plates were incubated for 4 h at 4 0 C and then washed with PBS/0.05% Tween 20 buffer.
  • lysis buffer 50 mM Tris.HCl pH7.4, 150 mM NaCl, 5 mM EDTA, 1 mM EGTA, 1% NP-40, 2 mM Na 3 VO 4 and protease inhibitor cocktail (Roche)
  • lysis buffer 50 mM Tris.HCl pH7.4, 150 mM NaCl,
  • alkaline phosphatase-conjugated monoclonal anti-phosphotyrosine antibody PY20, Zymed Laboratories
  • PY20 alkaline phosphatase-conjugated monoclonal anti-phosphotyrosine antibody
  • Ba/F3.p210 and Ba/F3.p210 E255V cells were incubated in the presence of various concentrations of GNF-2 and, after 1.5 h incubation, lysed in lysis buffer (20 mM Tris-HCl (pH 7.5), 150 mM NaCl, 1 mM Na 2 EDTA, 1 mM EGTA, 1% Triton, 2.5 mM sodium pyrophosphate, 1 mM beta- glycerophosphate, 1 mM Na 3 VO 4 , 1 ⁇ g/ml leupeptin, 1 mM PMSF).
  • lysis buffer 20 mM Tris-HCl (pH 7.5), 150 mM NaCl, 1 mM Na 2 EDTA, 1 mM EGTA, 1% Triton, 2.5 mM sodium pyrophosphate, 1 mM beta- glycerophosphate, 1 mM Na 3 VO 4 , 1 ⁇ g/ml leupeptin, 1 m
  • Equal amount of lysates (50 ⁇ g) were subjected to SDS-PAGE followed by immunoblotting with phospho-specific antibodies recognizing Bcr-abl phosphorylation sites: anti-phospho-c-Abl (Tyr 245), and anti- phospho-Stat5 (Tyr 694) (Cell signaling) antibodies or antibodies against Bcr-abl (Ab-3, Oncogene Science) and Stat5 (C- 17, Santa Cruz Biotechnology). Proteins were detected by enhanced chemioluminiscence (ECL-plus, Amersham), following manufacturer's guidelines.
  • Recombinant c-abl (residues 46-531) was expressed as a His 6 tag fusion protein in Sf9 insect cells using the Bac-to-Bac baculovirus expression system (Invitrogen) as previously described. Briefly, infected cells were grown for 48 h, harvested by centrifugation, suspended in lysis buffer (5OmM Tris pH 8.0, 30OmM NaCl, 5% glycerol, 1OmM 2- mercaptoethanol and protease inhibitor cocktail (Roche) and lysed by sonication. After centrifugation, the cell extract was loaded on a Ni-NTA affinity column and AbI was eluted with 25OmM imidazole.
  • lysis buffer 5OmM Tris pH 8.0, 30OmM NaCl, 5% glycerol, 1OmM 2- mercaptoethanol and protease inhibitor cocktail (Roche)
  • Fractions containing abl were pooled and the concentration of imidazole was reduced to 20 mM by dialysis.
  • the protein was treated with CiP alkaline phosphatase (Invitrogen) and purified by a second Ni affinity chromatography.
  • the His 6 tag was removed by treatment with TEV protease (Invitrogen) and the protein recovered from a Ni-NTA column with a purity of -90% by SDS-PAGE and coomassie blue staining.
  • In vitro kinase assays were carried out by using recombinant murine c-abl containing SH3, SH2 and kinase domains (residues 46-531) and full length immunoprecipitated Bcr-abl.
  • Recombinant abl was expressed in Sf9 insect cells and purified as described above.
  • Bcr-abl immune complexes were obtained with Ab-3 anti-abl monoclonal antibody (Oncogene Science) from Ba/F3.p210 lysates as previously described.
  • recombinant abl or immunoprecipitated Bcr-abl from 3x10 6 cells were incubated with various concentrations of test compound (0.1, 1 and 10 ⁇ M) in kinase buffer (50 mM Tris-HCl pH 7.5, 10 mM MgCl 2 , 100 mM EDTA, 1 mM DTT, 0.015% Brij 35), 100 ⁇ M ATP and 1 ⁇ Ci [ ⁇ - 32 P]-ATP for 30 min at 30°C (Calbiochem buffer and protocol). The reaction was stopped by addition of Laemmli buffer and the proteins were resolved by SDS- PAGE in a 4-20% gel. The phosphoproteins were visualized by autoradiography and the autophosphorylation quantitated using a phosphoimager (STORM, Molecular Devices). The results are provided in the following tables.

Abstract

The present invention provides novel heteroaryl compounds that are linked to an aryl group via an amine linker. Such compounds are useful for the treatment of cancers.

Description

SMALL MOLECULE MYRISTATE INHIBITORS OF BCR-ABL AND METHODS OF USE
RELATED APPLICATIONS
This application claims the benefit of U.S. Provisional Application No. 61/004,462, filed on November 28, 2007. The entire teachings of the above application are incorporated herein by reference.
BACKGROUND
The protein kinases represent a large family of proteins, which play a central role in the regulation of a wide variety of cellular processes and maintaining control over cellular function. These kinases include receptor tyrosine kinases, such as platelet-derived growth factor receptor kinase (PDGF-R), the receptor kinase for stem cell factor, c- Kit, and nonreceptor tyrosine kinases, such as the fusion kinase Bcr-abl.
Chronic myeloid leukemia (CML) is an extensively studied human cancer that is caused by a reciprocal translocation that fuses the AbI proto-oncogene on chromosome 9 with a gene on chromosome 22 called Bcr. The resulting fusion protein Bcr-abl is capable of transforming B-cells by increasing mitogenic activity, reducing sensitivity to apoptosis and altering the adhesion and homing of CML progenitor cells. STI-571 (Gleevec) is an inhibitor of the oncogenic Bcr-abl tyrosine kinase and is used for the treatment of chronic myeloid leukemia (CML). However, some patients in the blast crisis stage of CML are resistant to STI-571 due to mutations in the Bcr-abl kinase.
The novel compounds of this invention inhibit one or more kinases; in particular wild type and one or more of the mutant forms of Bcr-abl and are, therefore, useful in the treatment of kinase-associated diseases, particularly Bcr-abl kinase associated diseases.
BRIEF SUMMARY OF THE INVENTION
In one aspect, the invention provides a compound of formula I, or pharmaceutically acceptable salt, solvate or hydrate thereof:
Figure imgf000003_0001
wherein each of Xi, X2, or X3 is independently NRi, CRi, C(O), O, or S; each Ri is independently selected from H, an optionally substituted Ci-6alkyl, an optionally substituted C3-i2cycloalkyl, an optionally substituted C3.i2heterocycloalkyl, an optionally substituted C3-i2aryl, an optionally substituted C3-i2heteroaryl, an optionally substituted Ci-6aralkyl, an optionally substituted Ci-6heteroaralkyl, an optionally substituted Ci-6haloalkyl, an optionally substituted (CH2)nC3-i2heterocycloalkyl, an optionally substituted (CH2)nC3-i2aryl, an optionally substituted (CH2)nC3-i2heteroaryl, an optionally substituted (CH2)nC3-i2cycloalkyl, Ci-6haloalkyl, Ci-6aminoalkyl, hal, (CH2)nC(0)Rx, (CH2)nC(0)0Rx, (CH2)nC(O)NRxRx, -(CH2)nC(O)NRxRxS(O)2Rx; -C(O)RX, -C(S)RX, -C(NR)RX, -SRX, -S(O)RX, -S(O)2R", -ORX, -NRXRX, nitro, cyano, or absent; each Rx, for each occurrence, is independently H, an optionally substituted Ci-6alkyl, an optionally substituted C3.i2cycloalkyl, an optionally substituted C3-i2heterocycloalkyl, an optionally substituted C3-i2aryl, an optionally substituted C3-i2heteroaryl, Ci-6haloalkyl; NHNHR2, or C(NH)NH2;
R2 is H or Ci-6 alkyl;
Y is H, OH, NH2, or CH3; m is 1 or 2; and each n is independently 0, 1 or 2.
In one embodiment, the invention provides a compound of formula II, or pharmaceutically acceptable salt, solvate or hydrate thereof:
Figure imgf000004_0001
wherein
Ri is selected from H, an optionally substituted Ci-6alkyl, an optionally substituted C3. i2cycloalkyl, an optionally substituted C3-i2heterocycloalkyl, an optionally substituted C3- i2aryl, an optionally substituted C3.i2heteroaryl, an optionally substituted C1-6aralkyl, an optionally substituted C^heteroaralkyl, an optionally substituted Ci^haloalkyl, an optionally substituted (CH2)nC3-i2heterocycloalkyl, an optionally substituted (CH2)nC3-i2aryl, an optionally substituted (CH2)nC3-i2heteroaryl, an optionally substituted (CH2)nC3-i2cycloalkyl, (CH2)nC(O)Rx, (CH2)nC(O)ORx, (CH2)nC(0)NRxRx, -C(O)RX, -C(S)RX, -C(NR)RX, halo, C1- βhaloalkyl, -S(O)RX, or -S(O)2R";
Rx, for each occurrence, is independently, H, an optionally substituted Ci-6alkyl, an optionally substituted C3-i2cycloalkyl, an optionally substituted C3.i2heterocycloalkyl, an optionally substituted
Figure imgf000004_0002
and each n is independently 0, 1 or 2.
In another embodiment, the invention provides a compound formula III, or pharmaceutically acceptable salt, solvate or hydrate thereof:
Figure imgf000004_0003
wherein
R] is selected from H, an optionally substituted Ci-6alkyl, an optionally substituted C3- i2cycloalkyl, an optionally substituted C3-i2heterocycloalkyl, an optionally substituted C3- i2aryl, an optionally substituted C3-i2heteroaryl, an optionally substituted Ci-6 aralkyl, an optionally substituted Ci-βheteroaralkyl, an optionally substituted Ci-όhaloalkyl, an optionally substituted (CH2)nC3-i2heterocycloalkyl, an optionally substituted (CH2)nC3.i2aryl, an optionally substituted (CH2)nC3-i2heteroaryl, an optionally substituted (CH2)nC3-12cycloalkyl, (CH2)nC(O)Rx, (CH2)nC(O)ORx, (CH2)nC(O)NRxRx, -C(O)RX, -C(S)RX, -C(NR)RX, halo, Q- βhaloalkyl, -S(O)RX, or -S(O)2RX;
Rx, for each occurrence, is independently, H, an optionally substituted Ci-6alkyl, an optionally substituted C3-i2cycloalkyl, an optionally substituted Cs.^heterocycloalkyl, an optionally substituted C3-12aryl, an optionally substituted C3-i2heteroaryl, or Ci-6haloalkyl; and each n is independently 0, 1 or 2.
In certain embodiments, the invention provides a compound of formula IV, or pharmaceutically acceptable salt, solvate or hydrate thereof:
Figure imgf000005_0001
wherein each R] is indepdently selected from H, an optionally substituted Ci-6alkyl, an optionally substituted C3-12cycloalkyl, an optionally substituted C3-i2heterocycloalkyl, an optionally substituted C3-i2aryl, an optionally substituted C3-i2heteroaryl, an optionally substituted Ci-6aralkyl, an optionally substituted
Figure imgf000005_0002
an optionally substituted Ci-6haloalkyl, an optionally substituted (CH2)nC3-i2heterocycloalkyl, an optionally substituted (CH2)nC3-i2aryl, an optionally substituted (CH2)nC3-i2heteroaryl, an optionally substituted (CH2)nC3-12cycloalkyl, (CH2)nC(O)Rx, (CH2)nC(O)OR\ (CH2)nC(O)NRxRx, -C(O)RX, -C(S)RX, -C(NR)RX, halo, C]-6haloalkyl, -S(O)R", -S(O)2R", or absent;
Rx, for each occurrence, is independently, H, an optionally substituted Ci-6alkyl, an optionally substituted C3-i2cycloalkyl, an optionally substituted C3-i2heterocycloalkyl, an optionally substituted C3.i2aryl, an optionally substituted C3-i2heteroaryl, or Cj-βhaloalkyl; and each n is independently 0, 1 or 2. In another embodiment, the invention provides a compound of formula V, or pharmaceutically acceptable salt, solvate or hydrate thereof:
Figure imgf000006_0001
wherein
R] is independently selected from H, an optionally substituted Ci-6alkyl, an optionally substituted C3-i2cycloalkyl, an optionally substituted Cs-πheterocycloalkyl, an optionally substituted C3-i2aryl, an optionally substituted C3-i2heteroaryl, an optionally substituted (CH2)nC3-i2heterocycloalkyl, -C(O)R", -OR", -SRX, -NRXRX, nitro, cyano, Ci-6haloalkyl, C,. 6aminoalkyl, (CH2)nC(O)Rx, (CH2)nC(O)ORx, (CH2)nC(O)NRxRx, or -(CH2)nC(O)NRxRxS(O)2Rx;
Rx, for each occurrence, is independently H, an optionally substituted Ci-6alkyl, an optionally substituted C3-i2cycloalkyl, an optionally substituted C3-i2heterocycloalkyl, an optionally substituted C3-12aryl, an optionally substituted C3-i2heteroaryl, NHNHR2, or C(NH)NH2;
Ri' is selected from H, an optionally substituted Ci-6alkyl, an optionally substituted C3-i2cycloalkyl, an optionally substituted C3.i2heterocycloalkyl, an optionally substituted C3. i2aryl, an optionally substituted C3,i2heteroaryl, an optionally substituted Ci-6aralkyl, an optionally substituted Ci-βheteroaralkyl, an optionally substituted Ci^haloalkyl, an optionally substituted (CH2)nC3-i2heterocycloalkyl, an optionally substituted (CH2)nC3.i2aryl, an optionally substituted (CH2)nC3-i2heteroaryl, an optionally substituted (CH2)nC3-i2cycloalkyl, (CH2)nC(0)Ry, (CH2)nC(O)ORy, (CH2)nC(O)NRyRy, -C(O)Ry, -C(S)Ry, -C(NR)Ry, halo, C1- 6haloalkyl, -S(O)Ry, or -S(O)2Ry;
Ry, for each occurrence, is independently, H, an optionally substituted Ci-6alkyl, an optionally substituted C3-i2cycloalkyl, an optionally substituted C3-i2heterocycloalkyl, an optionally substituted C3.i2aryl, an optionally substituted C3.i2heteroaryl, or Ci-6haloalkyl; each n is independently 0, 1 or 2; and
R2 is H or CM alkyl. In a further embodiment, Ri is independently selected from H, an optionally substituted Ci-6alkyl, an optionally substituted C3.i2cycloalkyl, an optionally substituted C3. 12heterocycloalkyl, an optionally substituted C3-12aryl, an optionally substituted C3. nheteroaryl, an optionally substituted (CH2)nC3-i2heterocycloalkyl, -C(O)R*, -ORX, -SRX, -NRXRX, nitro, cyano, Ci-6haloalkyl, Ci-6aminoalkyl, (CH2)nC(O)Rx, (CH2)nC(O)ORx, (CH2)nC(O)NRxRx, or -(CH2)nC(O)NRxRxS(O)2Rx;
Rx, for each occurrence, is independently H, an optionally substituted Ci-6alkyl, an optionally substituted C3-i2cycloalkyl, an optionally substituted Cs-^heterocycloalkyl, an optionally substituted C3-i2aryl, an optionally substituted C3-i2heteroaryl, NHNHR2, or C(NH)NH2; and
R2 is H or CM alkyl.
In another further embodiment, R1' is selected from H, an optionally substituted Ci- 6alkyl, an optionally substituted C3-i2cycloalkyl, an optionally substituted C3. i2heterocycloalkyl, an optionally substituted C3-i2aryl, an optionally substituted C3- i2heteroaryl, an optionally substituted Ci-6aralkyl, an optionally substituted Ci-6heteroaralkyl, an optionally substituted Ci-6haloalkyl, an optionally substituted (CH2)nC3-i2heterocycloalkyl, an optionally substituted (CH2)nC3-i2aryl, an optionally substituted (CH2)nC3-i2heteroaryl, an optionally substituted (CH2)nC3.,2cycloalkyl, (CH2)nC(O)Ry, (CH2)nC(0)0Ry, (CH2)nC(0)NRyRy, -C(O)Ry, -C(S)Ry, -C(NR)Ry, halo, C,.6haloalkyl, -S(O)Ry, or -S(O)2Ry;
Ry, for each occurrence, is independently, H, an optionally substituted Ci-6alkyl, an optionally substituted C3-i2cycloalkyl, an optionally substituted C3-i2heterocycloalkyl, an optionally substituted C3-i2aryl, an optionally substituted C3-12heteroaryl, or Ci-6haloalkyl;
In other embodiments, the invention provides a compound of formula VI, or pharmaceutically acceptable salt, solvate or hydrate thereof:
Figure imgf000007_0001
wherein Ri is independently selected from H, an optionally substituted Ci-6alkyl, an optionally substituted C3-i2cycloalkyl, an optionally substituted C3-i2heterocycloalkyl, an optionally substituted C3-i2aryl, an optionally substituted C3-i2heteroaryl, an optionally substituted (CH2)nC3-i2heterocycloalkyl, -C(O)R", -OR\ -SR", -NRXRX, nitro, cyano, Ci-6haloalkyl, Ci- 6aminoalkyl, (CH2)nC(O)Rx, (CH2)nC(O)ORx, (CH2)nC(O)NRxRx, or -(CH2)nC(O)NRxRxS(O)2Rx;
Rx, for each occurrence, is independently H, an optionally substituted Ci-6alkyl, an optionally substituted C3-i2cycloalkyl, an optionally substituted C3-i2heterocycloalkyl, an optionally substituted C3-i2aryl, an optionally substituted C3-i2heteroaryl, NHNHR2, C(NH)NH2;
R2 is H or Ci-4 alkyl; and
Each n is independently 0, 1, or 2.
In still another embodiment, the invention provides a compound of formula VII, or pharmaceutically acceptable salt, solvate or hydrate thereof:
Figure imgf000008_0001
wherein
Ri is selected from H, an optionally substituted Ci-6alkyl, an optionally substituted C3- i2cycloalkyl, an optionally substituted C3-i2heterocycloalkyl, an optionally substituted C3- i2aryl, an optionally substituted C3-i2heteroaryl, an optionally substituted (C^)nC3- i2heterocycloalkyl, -C(O)RX, -ORX, -SR", -NRXRX, nitro, cyano, Ci.6haloalkyl, Ci-6aminoalkyl, (CH2)nC(O)Rx, (CH2)nC(O)ORx, (CH2)nC(O)NRxRx, or -(CH2)nC(O)NRxRxS(O)2Rx;
Rx, for each occurrence, is independently H, an optionally substituted Ci-6alkyl, an optionally substituted C3-i2cycloalkyl, an optionally substituted Cs-πheterocycloalkyl, an optionally substituted C3-i2aryl, an optionally substituted C3-i2heteroaryl, NHNHR2, or C(NH)NH2;
R2 is H or Ci-4 alkyl; m is 1 or 2; and each n is independently 0, 1 or 2. In another embodiment, the invention provides a compound of formula VIII, or pharmaceutically acceptable salt, solvate or hydrate thereof:
Figure imgf000009_0001
wherein
Ri is selected from H, an optionally substituted Ci-6alkyl, an optionally substituted C3- i2cycloalkyl, an optionally substituted C3-i2heterocycloalkyl, an optionally substituted C3- i2aryl, an optionally substituted C3-i2heteroaryl, an optionally substituted Ci-όaralkyl, an optionally substituted Ci-όheteroaralkyl, an optionally substituted Ci-6haloalkyl, an optionally substituted (CH2)nC3-i2heterocycloalkyl, an optionally substituted (CH2)nC3-i2aryl, an optionally substituted (CH2)nC3-12heteroaryl, an optionally substituted (CH2)nC3-i2cycloalkyl, (CH2)nC(O)Rx, (CH2)nC(O)ORx, (CH2)nC(O)NRxRx, -C(O)RX, -C(S)RX, -C(NR)RX, halo, Ci- 6haloalkyl, -S(O)RX, or -S(O)2R";
Rx, for each occurrence, is independently, H, an optionally substituted Ci-όalkyl, an optionally substituted C3-i2cycloalkyl, an optionally substituted C3-i2heterocycloalkyl, an optionally substituted C3.i2aryl, an optionally substituted C3-i2heteroaryl, or Ci-6haloalkyl; and each n is independently 0, 1 or 2.
In another aspect, the invention provides a compound of formula IX, or pharmaceutically acceptable salt, solvate or hydrate thereof:
Figure imgf000009_0002
wherein
R3 is H or C1-4 alkyl; L is NR3, O, S, S(O), S(O)2, C(O), C(S), C(NR), C(NRX)NRX, C(O)NR", C(O)NRXNRX, C(0)0NRx, C(O)NRXO, C(O)O, OC(O), OC(O)O, or a bond; each Rx, for each occurrence, is independently H, an optionally substituted Ci^alkyl, an optionally substituted C3-i2cycloalkyl, an optionally substituted C3-i2heterocycloalkyl, an optionally substituted C3-|2aryl, an optionally substituted C3-i2heteroaryl, Ci-6haloalkyl; NHNHR2, or C(NH)NH2;
A is an optionally substituted Ci-6alkyl, an optionally substituted C3-i2aryl, an optionally substituted C3-i2heteroaryl, an optionally substituted C3-i2Cycloalkyl, or an optionally substituted C3-i2heterocycloalkyl.
In one embodiment, A is an optionally substituted C].6alkyl, an optionally substituted C3-i2aryl, an optionally substituted C3-12heteroaryl, or an optionally substituted C3- ι2heterocycloalkyl.
In a further embodiment, A is an optionally substitued phenyl ring, an optionally substituted naphthyl ring, an optionally substituted piperidine ring, an optionally substituted imidazole ring, an optionally substituted pyrazole ring, an optionally substituted lH-pyrazole ring, an optionally substituted lH-Pyrrolo[2,3-b]pyridine ring, an optionally substituted pyrimidine ring, an optionally substituted pyrazine ring, an optionally substituted pyridine ring, an optionally substituted indole ring, an optionally substituted lH-indole ring, an optionally substituted furan ring, an optionally substituted benzo[b]thiophene ring, an optionally substituted dibenzofuran ring, an optionally substituted lH-Pyridin-2-one ring, an optionally substituted thiazole ring, or an ethyl group.
In another further embodiment, A is optionally substituted with Ci-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-i2cycloalkyl, C3-i2heterocycloalkyl, C3-i4aralkyl, Q-πheteroaralkyl, C3- i2aryl, (CH2)pC3-i2aryl, C3-i2heteroaryl, (CH2)pC3-i2heteroaryl, halogen, haloalkyl, cyano, (CH2)PCN, nitro, Ci-6alkoxy, C3-i2aryloxy, C1-C4 haloalkoxy, Ci-C6 haloalkyl, hydroxyl, Ci- 6hydroxylalkyl, Ci-6carboxyl, CHO, amino, aminoalkyl, dialkylamino, OR15, SR15, NR17R17C(O)OR15, C(O)NR15R16, OC(O)NR15R16, NR15C(O)NR15R16, NR17S(O)2R15, NR17(CH2)nR17, C(O)R17, C(O)NR17R17, NR15C(O)R17, S(O)R17, S(O)2R17, or S(O)2NRnR17; each R15 is independently hydrogen, C3-I4 aralkyl, Ci-6 alkyl, C3-I2 cycloalkyl, C3-I2 aryl, C3-I2 heterocycloalkyl, or C3-I2 heteroaryl; each R16 is independently hydrogen, C3-J4 aralkyl, or
Figure imgf000011_0001
alkyl; each R17 is independently H, Ci-6 alkyl, Ci-6 hydroxyalkyl, Cp6 alkoxy, C3-I2 cycloalkyl, C3-i2aryl, C3-i2heteroaryl, C3-12heterocycloalkyl, hydroxyl, or each R17 may form a heterocycloalkyl ring with another R17; wherein each A, R15, R16, and R17 may be further optionally substituted with C]-6 alkyl, Cp6 alkoxy, Cr6 haloalkoxy, C3-i2heterocycloalkyl, amino, halogen, d-6 haloalkyl, SO2alkyl, or NHC(O)aryl; and each p is independently 0-6.
In another aspect, the invention provides a compound of formula X, or pharmaceutically acceptable salt, solvate or hydrate thereof:
Figure imgf000011_0002
wherein each Of X2 or X3 is independently NRb CR1, C(O), O, or S; each R] is independently selected from H, an optionally substituted Ci-6alkyl, an optionally substituted C3-12cycloalkyl, an optionally substituted C3-i2heterocycloalkyl, an optionally substituted C3-12aryl, an optionally substituted C3.i2heteroaryl, an optionally substituted C2-6aralkyl, an optionally substituted C2-i2heteroaralkyl, an optionally substituted Ci-6haloalkyl, an optionally substituted (CH2)nC3-i2aryl, an optionally substituted (CH2),^. i2heteroaryl, an optionally substituted (CH2)nC3-i2cycloalkyl, an optionally substituted (CH2)nC3-i2heterocycloalkyl, d-6haloalkyl, Ci-6aminoalkyl, hal, (CH2)nC(O)Rx, (CH2)nC(O)ORx, (CH2)nC(O)NRxRx, -(CH2)nC(O)NRxRxS(O)2Rx; -C(O)RX, -C(S)RX, -C(NR)RX, -SRX, -S(O)RX, -S(O)2R", -ORX, -NRXRX, nitro, cyano, or absent; each Rx, for each occurrence, is independently H, an optionally substituted Ci^alkyl, an optionally substituted C3-i2cycloalkyl, an optionally substituted C3-i2heterocycloalkyl, an optionally substituted C3-i2aryl, an optionally substituted C3-i2heteroaryl, Ci-6haloalkyl; NHNHR2, or C(NH)NH2;
R2 is H or Ci-6 alkyl; and each n is independently 0, 1 or 2.
In one embodiment, the invention provides a compound of formula XI, or pharmaceutically acceptable salt, solvate or hydrate thereof:
Figure imgf000012_0001
wherein
Ri is independently selected from H, an optionally substituted Ci-6alkyl, an optionally substituted C3-i2aryl, or an optionally substituted Cs-πheteroaryl.
In certain aspects, the present invention is directed towards a method of treating a disease in a subject in which inhibition of kinase activity, particularly Bcr-abl activity, can prevent, inhibit or ameliorate the pathology and/or symptomology of certain disorders and/or diseases, which method comprises administering to the subject a therapeutically effective amount of a compound of the invention or pharmaceutically acceptable salt, solvate or hydrate thereof.
In one aspect, the invention provides a method of treating a kinase activity related disorder in a subject, comprising administering to said subject in need thereof, an effective amount of a compound, or pharmaceutically acceptable salt, solvate or hydrate thereof, of any one of the compounds disclosed herein.
In certain embodiments, the compound is a kinase inhibitor. In a further embodiment, the compound interacts with Bcr-abl. In another further embodiment, the compound targets the binding site of Bcr-abl.
In another embodiment, the subject is identified as being in need of kinase inhibition, and the compound of the invention is administered to the identified subject. In still another embodiment, the invention provides the method further comprising the step of identifying a subject in need of such treatment for a kinase activity related disorder and administering the compound to the identified subject.
In other embodiments, the invention provides a method wherein the subject is suffering from a cell proliferative disorder or disease. In a further embodiment, the disorder is cancer, tumor, neoplasm, neovascularization, vascularization, cardiovascular disease, metastasis, infection, blood clot, atherosclerosis, melanoma, skin disorder, rheumatoid arthritis, diabetic retinopathy, macular edema, or macular degeneration, inflammatory and arthritic disease, or osteosarcoma. In a further embodiment, the subject is suffering from a solid tumor or disseminated cancer. In another further embodiment, the cancer is leukemia, multiple myeloma or lymphoma. In certain embodiments, the leukemia is Chroic Myelogenous Leukemia.
In another aspect, the invention provides a method of treating cancer in a subject, the method comprising administering to said subject an effective amount of a compound, or pharmaceutically acceptable salt, solvate or hydrate thereof, of any one of the compounds disclosed herein.
In one embodiment, the invention provides a method described above further comprising administering an additional therapeutic agent. In a further embodiment, the additional therapeutic agent is an ATP-site inhibitor. In another further embodiment, the ATP-site inhibitor is selected from the following: imatinib (STI571), Nilotinib (AMN 107), Dasatinib (BMS-354825), AP234464 (Ariad Pharmaceuticals), AZD0530 (Astrazeneca), and SKI-606 (Wyeth).
In another embodiment, the subject is a mammal. In a further embodiment, the subject is a primate or human.
In one embodiment, the invention provides a method wherein the step of administering the compound comprises administering the compound orally, topically, parentally, intravenously or intramuscularly. In a further embodiment, the method comprises the step of administering an effective amount of a composition comprising the compound and a pharmaceutically suitable excipient.
In certain embodiments, the step of administering the compound comprises administering the compound in a dosage of between about 0.01 μg/kg/day and 100 mg/kg/day. In another aspect, the invention provides a method of inhibiting kinase activity in a subject, the method comprising the step of administering to the subject a compound, or pharmaceutically acceptable salt, solvate or hydrate thereof, of any one of the compounds disclosed herein.
In other aspects, the invention provides the use of a compound described herein in the manufacture of a medicament for treating a disease in a subject in which kinase activity, particularly Bcr-abl activity, contributes to the pathology and/or symptomology of the disease.
In other aspects, the invention provides a pharmaceutical composition comprising a compound, or pharmaceutically acceptable salt, solvate or hydrate thereof, of any of the compounds disclosed herein. In one embodiment, the invention provides a composition further comprising an additional therapeutic agent. In another embodiment, the additional therapeutic agent is an ATP-site inhibitor. In another further embodiment, the ATP-site inhibitor is selected from the following: imatinib (STI571), Nilotinib (AMN107), Dasatinib (BMS-354825), AP234464 (Ariad Pharmaceuticals), AZD0530 (Astrazeneca), and SKI-606 (Wyeth).
In certain aspects, the invention provides a kit comprising an effective amount of a compound disclosed herein, or pharmaceutically acceptable salt, solvate or hydrate thereof, in unit dosage form, together with instructions for administering the compound to a subject suffering from or susceptible to a cell proliferative disorder.
In certain aspects, the present invention provides a method for inhibiting Bcr-abl activity, the method comprising contacting Bcr-abl with a compound of the invention that binds to a myristoyl binding pocket of Bcr-abl.
In one aspect, the invention provides for a method for identifying a compound which modulates the kinase activity of Bcr-abl, the method comprising: a) contacting Bcr-abl with a compound under conditions suitable for modulation of the kinase activity of Bcr-abl; and b) detecting modulation of the kinase activity of Bcr-abl by the compound; wherein the compound is capable of interacting with the myristate binding site of Bcr- abl.
In one embodiment, the interaction of the compound with myristate binding site of Bcr-abl is a binding interaction. In another embodiment, the binding interaction is ionic, covalent, or a non-direct interaction. In still another embodiment, the compound has a binding interaction with Ala452, Tyr454, Glu481, Pro484, Val487, Phe516, IleS21, or Val525 (Number based upon crystal structure with protein databank code: lOPL, reference: Nagar, B., Hantschel, O., Young, M.A., Scheffzek, K., Veach, D., Bornmann, W., Clarkson, B., Superti-Furga, G., Kuriyan, J. Structural basis for the autoinhibition of c- AbI tyrosine kinase Cell (Cambridge,Mass.) vl l2 pp. 859-871, 2003.
In other embodiments, the modulation of the activity of the kinase activity is detected by direct binding of the compound to the myristate binding site of Bcr-abl.
In another embodiment, the compound is designed de novo.
In other embodiments, the compound is designed from a known ligand of the myristate binding site of Bcr-abl.
DETAILED DESCRIPTION
Compounds of the Invention
Exemplary compounds of the invention include the following, which are provided structurally in Tables 1-9.
2-(4-(4-(trifluoromethoxy)phenylamino)- 1 H-pyrazolo [3 ,4-d]pyrimidin- 1 -y l)ethanol ( 1 ); (lH-Pyrazolo[3,4-d]pyrimidin-4-yl)-(4-trifluoromethoxy-phenyl)-amine (2); ( 1 -Ethyl- 1 H-pyrazolo[3 ,4-d]pyrimidin-4-yl)-(4-trifluoromethoxy-phenyl)-amine (3); ( 1 -Ethanesulfonyl- 1 H-pyrazolo [3 ,4-d]pyrimidin-4-yl)-(4-trifluoromethoxy-phenyl)-amine
(4);
(1-Cyclohexyl-l H-pyrazolo [3 ,4-d]pyrimidin-4-yl)-(4-trifluoromethoxy-phenyl)-amine (5);
2-[4-(4-Trifluoromethoxy-phenylamino)-pyrazolo[3 ,4-d]pyrimidin- 1 -yl]-ethanol (6);
2-[4-(4-Trifluoromethoxy-phenylamino)-pyrazolo[3,4-d]pyrimidin-l-yl]-acetamide (7);
3-[4-(4-Trifluoromethoxy-phenylamino)-pyrazolo[3,4-d]pyrimidin-l-yl]-propionamide (8);
[l-(Tetrahydro-furan-3-ylmethyl)-lH-pyrazolo[3,4-d]pyrimidin-4-yl]-(4-trifluoromethoxy- phenyl)-amine (9);
3 -Dimethylamino-2- [4-(4-trifluoromethoxy-phenylamino)-pyrazolo [3 ,4-d]pyrimidin- 1 -yl] - propionitrile (10);
( 1 -Butyl- 1 H-pyrazolo [3 ,4-d]pyrimidin-4-y l)-(4-trifluoromethoxy-phenyl)-amine (1 1);
(l-tert-Butyl-lH-pyrazolo[3,4-d]pyrimidin-4-yl)-(4-trifluoromethoxy-phenyl)-amine (12); [l-(2,2,2-Trifluoro-ethyl)-lH-pyrazolo[3,4-d]pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)- amine (13);
( 1 -Benzyl- 1 H-pyrazolo[3 ,4-d]pyrimidin-4-yl)-(4-trifluoromethoxy-phenyl)-amine (14);
( 1 -Phenethyl- 1 H-pyrazolo [3 ,4-d]pyrimidin-4-yl)-(4-trifluoromethoxy-phenyl)-amine (15);
[l-(3-Fluoro-4-trifluoromethyl-benzyl)-lH-pyrazolo[3,4-d]pyrimidin-4-yl]-(4- trifluoromethoxy-phenyl)-amine (16);
[l-(3-Bromo-benzyl)-lH-pyrazolo[3,4-d]pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)-amine
(17);
3-[4-(4-Trifluoromethoxy-phenylamino)-pyrazolo[3,4-d]pyrimidin-l-ylmethyl]-phenol (18);
( 1 -Pyridin-3 -ylmethyl- 1 H-pyrazolo[3 ,4-d]pyrimidin-4-yl)-(4-trifluoromethoxy-phenyl)- amine (19);
( 1 -Pyridin-2 -ylmethyl- 1 H-pyrazolo[3 ,4-d]pyrimidin-4-yl)-(4-trifluoromethoxy-phenyl)- amine (20);
( 1 -Pyridin-4-ylmethyl- 1 H-pyrazolo [3 ,4-d]pyrimidin-4-yl)-(4-trifluoromethoxy-phenyl)- amine (21);
2-{2-[4-(4-Trifluoromethoxy-phenylamino)-pyrazolo[3,4-d]pyrimidin-l-yl]-ethyl}-isoindole- l,3-dione (22);
[l-(2-Amino-ethyl)-lH-pyrazolo[3,4-d]pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)-amine
(23);
(4-Trifluoromethoxy-phenyl)-[l-(4-trifluoro methyl-pyridin-2-yl)-lH-pyrazolo[3,4- d]pyrimidin-4-yl] -amine (24);
[ 1 -(2,3 -Dimethyl-pheny I)- 1 H-pyrazolo [3 ,4-d]pyrimidin-4-yl] -(4-trifluoromethoxy-phenyl)- amine (25);
[l-(2-Fluoro-phenyl)-lH-pyrazolo[3,4-d]pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)-amine
(26);
[ 1 -(2,4-Dichloro-phenyl)- 1 H-pyrazolo [3 ,4-d]pyrimidin-4-yl] -(4-trifluoromethoxy-phenyl)- amine (27);
[ 1 -(2-Ethyl-pheny I)- 1 H-pyrazolo [3 ,4-d]pyrimidin-4-y l]-(4-trifluoromethoxy-pheny l)-amine
(28);
(l-Pyridin-2-yl-lH-pyrazolo[3,4-d]pyrimidin-4-yl)-(4-trifluoromethoxy-phenyl)-amine (29);
3 - [4-(4-Trifluoromethoxy-pheny lamino)-pyrazolo [3 ,4-d]pyrimidin- 1 -yl] -benzenesulfonamide
(30); Table 1. Compounds of formula II.
Figure imgf000017_0001
Figure imgf000018_0001
Figure imgf000019_0001
2-(4-(4-(trifluoromethoxy)phenylamino)-2H-pyrazolo[3,4-d]pyrimidin-2-yl)ethanol (31); (2-Ethyl-2H-pyrazolo[3,4-d]pyrimidin-4-yl)-(4-trifluoromethoxy-phenyl)-amine (32); (2-Cyclohexyl-2H-pyrazolo[3,4-d]pyrimidin-4-yl)-(4-trifluoroniethoxy-phenyl)-amine (33); 2-[4-(4-Trifluoromethoxy-phenylamino)-pyrazolo[3,4-d]pyrimidin-2-yl]-ethanol (34); 2-[4-(4-Trifluoromethoxy-phenylamino)-pyrazolo[3,4-d]pyrimidin-2-yl]-acetamide (35); [2-(Tetrahydro-furan-3-ylmethyl)-2H-pyrazolo[3,4-d]pyrimidin-4-yl]-(4-trifluoromethoxy- phenyl)-amine (36);
(2-Butyl-2H-pyrazolo[3,4-d]pyrimidin-4-yl)-(4-trifluoromethoxy-phenyl)-amine (37); [2-(2,2,2-Trifluoro-ethyl)-2H-pyrazolo[3,4-d]pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)- amine (38);
(2-Benzyl-2H-pyrazolo[3,4-d]pyrimidin-4-yl)-(4-trifluoromethoxy-phenyl)-amine (39); (2-Phenethyl-2H-pyrazolo[3,4-d]pyrimidin-4-yl)-(4-trifluoromethoxy-phenyl)-amine (40); [2-(3-Fluoro-4-trifluoromethyl-benzyl)-2H-pyrazolo[3,4-d]pyrimidin-4-yl]-(4- trifluoromethoxy-phenyl)-amine (41);
[2-(3-Bromo-benzyl)-2H-pyrazolo[3,4-d]pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)-amine (42);
3-[4-(4-Trifluoromethoxy-phenylamino)-pyrazolo[3,4-d]pyrimidin-2-ylmethyl]-phenol (43); (2-Pyridin-3 -ylmethyl-2H-pyrazolo [3 ,4-d]pyrimidin-4-yl)-(4-trifluoromethoxy-phenyl)- amine (44); (4-Trifluoromethoxy-phenyl)-[2-(4-trifluoromethyl-pyridin-2-yl)-2H-pyrazolo[3,4- d]pyrimidin-4-yl]-amine (45);
[2-(2,3-Dimethyl-phenyl)-2H-pyrazolo[3,4-d]pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)- amine (46);
[2-(2-Fluoro-phenyl)-2H-pyrazolo[3,4-d]pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)-amine
(47);
[2-(2,4-Dichloro-phenyl)-2H-pyrazolo[3,4-d]pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)- amine (48);
[2-(2-Ethyl-phenyl)-2H-pyrazolo[3,4-d]pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)-amine
(49);
(2-Pyridin-2-yl-2H-pyrazolo[3,4-d]pyrimidin-4-yl)-(4-trifluoromethoxy-phenyl)-amine (50);
Table 2. Compounds of formula III.
Figure imgf000020_0001
Figure imgf000021_0001
(9-Methyl-9H-purin-6-yl)-(4-trifluoromethoxy-phenyl)-amine (51 ); (7-Methyl-7H-purin-6-yl)-(4-trifluoromethoxy-phenyl)-amine (52);
2-Hydroxymethyl-5-[6-(4-trifluoromethoxy-phenylamino)-purin-9-yl]-tetrahydro-furan-3,4- diol (53); [9-(2-Moφholin-4-yl-ethyl)-9H-purin-6-yl]-(4-trifluoromethoxy-phenyl)-amine (54);
Table 3. Compounds of formula IV.
Figure imgf000022_0001
N-(2-Morpholin-4-yl-ethyl)-3 - [4-(4-trifluoromethoxy-phenylamino)-pyrrolo [2,3 - d]pyrimidin-7-yl]-propionamide (55);
2-Hydroxymethyl-5-[4-(4-trifluoromethoxy-phenylaniino)-pyrrolo[2,3-d]pyrimidin-7-yl]- tetrahydro-furan-3,4-diol (56);
(7H-Pyrrolo [2,3 -d]pyrimidin-4-yl)-(4-trifluoromethoxy-phenyl)-amine (57) ;
(7-Methyl-7H-pyπOlo[2,3-d]pyrimidin-4-yl)-(4-trifluoromethoxy-phenyl)-aminev (58);
2-[4-(4-Trifluoromethoxy-phenylamino)-pyiτolo[2,3-d]pyrimidin-7-yl]-ethanol (59);
[7-(2-Moφholin-4-yl-ethyl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)- amine (60);
[7-(4-Methoxy-benzyl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)-amine
(61);
3-[4-(4-Trifluoromethoxy-phenylamino)-pyrrolo[2,3-d]pyrimidin-7-yl]-propionamide (62);
N-Ethyl-3-[4-(4-trifluoromethoxy-phenylamino)-pyrrolo[2,3-d]pyrimidin-7-yl]-propionamide
(63);
N-(3-Methoxy-propyl)-3-[4-(4-trifluoromethoxy-phenylamino)-pyrrolo[2,3-d]pyrimidin-7- yl]-propionamide (64);
N-(2-Moφholin-4-yl-ethyl)-3-[4-(4-trifluoromethoxy-phenylamino)-pyrrolo[2,3- d]pyrimidin-7-yl]-propionamide (65); 7-Methyl-4-(4-trifluoromethoxy-phenylamino)-7H-pyrrolo[2,3-d]pyrimidine-6-carboxylic acid ethyl ester (66);
(4-Bromo-piperidin- 1 -y I)- [7-methyl-4-(4-trifluoromethoxy-pheny lamino)-7H-pyrrolo [2,3 - d]pyrimidin-6-yl]-methanone (67); l-[7-Methyl-4-(4-trifluoromethoxy-phenylamino)-7H-pyrrolo[2,3-d]pyrimidine-6-carbonyl]- piperidine-4-carboxylic acid amide (68); l-[7-Methyl-4-(4-trifluoromethoxy-phenylamino)-7H-pyrrolo[2,3-d]pyrimidine-6-carbonyl]- piperidine-3-carboxylic acid amide (69); l-[7-Methyl-4-(4-trifluoromethoxy-phenylamino)-7H-pyrrolo[2,3-d]pyrimidine-6-carbonyl]- piperidine-3-carboxylic acid ethyl ester (70);
(3-Hydroxymethyl-piperidin-l-yl)-[7-methyl-4-(4-trifluoromethoxy-phenylamino)-7H- pyrrolo [2 , 3 -d]pyrimidin-6-y 1] -methanone (71);
8-[7-Methyl-4-(4-trifluoromethoxy-phenylamino)-7H-pyrrolo[2,3-d]pyrimidine-6-carbonyl]-
1 -phenyl- 1, 3, 8-triaza- spiro[4.5]decan-4-one (72);
[7-Methyl-4-(4-trifluoromethoxy-phenylamino)-7H-pyiτolo[2,3-d]pyrimidin-6-yl]-methanol
(73);
(2-Methoxymethy 1-pyrrolidin- 1 -yl)- [7-methyl-4-(4-trifluoromethoxy-phenylamino)-7H- pyrrolo [2 ,3 -d]pyrimidin-6-y 1] -methanone (74) ;
7-Methyl-4-(4-trifluoromethoxy-phenylamino)-7H-pyrrolo[2,3-d]pyrimidine-6-carboxylic acid amide (75);
7-Methyl-4-(4-trifluoromethoxy-phenylamino)-7H-pyrrolo[2,3-d]pyrimidine-6-carboxylic acid ethylamide (76);
7-Methyl-4-(4-trifluoromethoxy-phenylamino)-7H-pyiτolo[2,3-d]pyrimidine-6-carboxylic acid (1 -ethyl -pyrrolidin-2-yl)-amide (77);
N-[7-Methyl-4-(4-trifluoromethoxy-phenylamino)-7H-pyrrolo[2,3-d]pyrimidine-6-carbonyl]- guanidine (78);
7-Methyl-4-(4-trifluoromethoxy-phenylamino)-7H-pyπOlo[2,3-d]pyrimidine-6-carboxylic acid (4-diethylamino-l-methyl-butyl)-amide (79);
4-(2-Bromo-4-trifluoromethoxy-phenylamino)-l-(2-morpholin-4-yl-ethyl)-lH-pyrrolo[2,3- b]pyridine-5-carbaldehyde (80);
Table 4. Compounds of formula V.
Figure imgf000024_0001
Figure imgf000025_0001
N-(2-hydroxyethyl)-4-(4-(trifluoromethoxy)phenylamino)thieno[2,3-d]pyrimidine-6- carboxamide (81);
Thieno[2,3-d]pyrimidin-4-yl-(4-trifluoromethoxy-phenyl)-amine (82);
(6-Methyl-thieno[2,3-d]pyrimidin-4-yl)-(4-trifluoromethoxy-phenyl)-amine (83);
4-(4-Trifluoromethoxy-phenylamino)-thieno[2,3-d]pyrimidine-6-carboxylic acid methyl ester
(84);
4-(4-Trifluoromethoxy-phenylamino)-thieno[2,3-d]pyrimidine-6-carboxylic acid (85);
4-(4-Trifluoromethoxy-phenylaniino)-thieno[2,3-d]pyriniidine-6-carboxylic acid (2- morpholin-4-yl-ethyl)-amide (86);
[4-(4-Trifluoromethoxy-phenylamino)-thieno[2,3-d]pyrimidin-6-yl]-methanol (87);
4-(4-Trifluoromethoxy-phenylamino)-thieno[2,3-d]pyrimidine-6-carboxylic acid ethylamide
(88);
4-(4-Trifluoromethoxy-phenylamino)-thieno[2,3-d]pyrimidine-6-carboxylic acid (2-hydroxy- ethyl)-amide (89);
[6-(3-Dimethylamino-pyrrolidin-l-ylmethyl)-thieno[2,3-d]pyrimidin-4-yl]-(4- trifluoromethoxy-phenyl)-amine (90); (3 -Dimethy lamino-pyrrolidin- 1 -yl)- [4-(4-trifluoromethoxy-pheny lamino)-thieno [2,3 - d]pyrimidin-6-yl]-methanone (91);
4-(4-Trifluoromethoxy-phenylamino)-thieno[2,3-d]pyrimidine-6-carboxylic acid (1 -benzyl- pyrrolidin-3-yl)-amide (92);
(2-Hydroxymethyl-pyrrolidin-l-yl)-[4-(4-trifluoromethoxy-phenylamino)-thieno[2,3- d]pyrimidin-6-yl]-methanone (93);
Piperazin-l-yl-[4-(4-trifluoromethoxy-phenylamino)-thieno[2,3-d]pyrimidin-6-yl]- methanone (94);
(4-Pyrrolidin- 1 -yl-piperazin- 1 -yl)-[4-(4-trifluoromethoxy-phenylamino)-thieno[2,3- d]pyrimidin-6-yl]-methanone (95);
[4-(4-Trifluoromethoxy-phenylamino)-thieno[2,3-d]pyrimidin-6-yl]-[4-(3-trifluoromethyl- phenyl)-piperazin- 1 -yl]-methanone (96);
4-(4-Trifluoromethoxy-phenylamino)-thieno[2,3-d]pyrimidine-6-carboxylic acid (2- morpholin-4-yl-ethyl)-amide (97);
(4-Methyl-piperazin-l-yl)-[4-(4-trifluoromethoxy-phenylamino)-thieno[2,3-d]pyrimidin-6- yl]-methanone (98);
(4-Hydroxy-piperidin- 1 -yl)- [4-(4-trifluoromethoxy-phenylamino)-thieno [2,3 -d]pyrimidin-6- yl]-methanone (99);
(4-Methoxy-piperidin- 1 -yl)-[4-(4-trifluoromethoxy-phenylamino)-thieno[2,3 -d]pyrimidin-6- yl]-methanone (100);
4-(4-Trifluoromethoxy-phenylamino)-thieno[2,3-d]pyrimidine-6-carboxylic acid [2-methyl-
5-(3-trifluoromethyl-benzoylamino)-phenyl]-amide (101);
Table 5. Compounds of formula VI.
Figure imgf000026_0001
Figure imgf000027_0001
4-(4-(trifluoromethoxy)phenylamino)-N-(2-hydroxyethyl)quinazoline-7-carboxamide ( 102); 4-(4-Trifluoromethoxy-phenylamino)-quinazoline-7-carboxylic acid methyl ester (103); [4-(4-Trifluoromethoxy-phenylamino)-quinazolin-7-yl]-methanol (104); (7-Diethylaminomethyl-quinazolin-4-yl)-(4-trifluoromethoxy-phenyl)-amine (105); 4-(4-Trifluoromethoxy-phenylamino)-quinazoline-7-carboxylic acid diethylamide (106);
[4-(Furan-3 -carbonyl)-piperazin- 1 -yl] - [4-(4-trifluoromethoxy-phenylamino)-quinazolin-7- yl]-methanone (107);
4-(4-Trifluoromethoxy-phenylamino)-quinazoline-7-carboxylic acid (3 -trifluoromethyl- phenyl)-amide (108);
4-(4-Trifluoromethoxy-phenylamino)-quinazoline-7-carboxylic acid (3 -methoxy-propy I)- amide (109);
4-(4-Trifluoromethoxy-phenylamino)-quinazoline-7-carboxylic acid N'-ethyl-hydrazide
(110);
(2- { [4-(4-Trifluoromethoxy-phenylamino)-quinazoline-7-carbonyl]-amino } -thiazol-5-yl)- acetic acid ethyl ester (111);
(4-Methoxy-piperidin-l-yl)-[4-(4-trifluoromethoxy-phenylamino)-quinazolin-7-yl]- methanone (112);
(3-Hydroxy-pyrrolidin-l-yl)-[4-(4-trifluoromethoxy-phenylamino)-quinazolin-7-yl]- methanone (113);
(2-Hydroxymethyl-pyrrolidin- 1 -yl)- [4-(4-trifluoromethoxy-phenylamino)-quinazolin-7-yl] - methanone (114);
(3-Dimethylamino-pyπOlidin-l-yl)-[4-(4-trifluoromethoxy-phenylamino)-quinazolin-7-yl]- methanone (115);
4-(4-Trifluoromethoxy-phenylamino)-quinazoline-7-carboxylic acid (2-hydroxy-ethyl)- methyl-amide (1 16);
4-(4-Trifluoromethoxy-phenylamino)-quinazoline-7-carboxylic acid methyl-(l -methyl- piperidin-4-yl)-amide (117);
4-Methyl-N-[4-(4-trifluoromethoxy-phenylamino)-quinazoline-7-carbonyl]- benzenesulfonamide (118);
2-Methyl-propane-2-sulfonic acid [4-(4-trifluoromethoxy-phenylamino)-quinazoline-7- carbonyl]-amide (119);
(4-Methyl-[l,4]diazepan-l-yl)-[4-(4-trifluoromethoxy-phenylamino)-quinazolin-7-yl]- methanone (120);
(3,5-Dimethyl-piperazin-l-yl)-[4-(4-trifluoromethoxy-phenylamino)-quinazolin-7-yl]- methanone (121); (4-Hydroxy-piperidin-l-yl)-[4-(4-trifluoromethoxy-phenylamino)-quinazolin-7-yl]- methanone (122);
4-(4-Trifluoromethoxy-phenylamino)-quinazoline-7-carboxylic acid methyl-(2-methylamino- ethyl)-amide (123);
4-(4-Trifluoromethoxy-phenylamino)-quinazoline-7-carboxylic acid (2-hydroxy-ethyl)-amide
(124); l-Phenyl-8-[4-(4-trifluoromethoxy-phenylamino)-quinazoline-7-carbonyl]-l,3,8-triaza- spiro[4.5]decan-4-one (125);
(4-Bromo-piperidin-l-yl)-[4-(4-trifluoromethoxy-phenylamino)-quinazolin-7-yl]-methanone
(126);
1 - [4-(4-Trifluoromethoxy-pheny lamino)-quinazoline-7-carbonyl] -piperidine-4-carboxylic acid amide (127); l-[4-(4-Trifluoromethoxy-phenylamino)-quinazoline-7-carbonyl]-piperidine-3-carboxylic acid amide (128);
1 - [4-(4-Trifluoromethoxy-pheny lamino)-quinazoline-7-carbonyl] -piperidine-3 -carboxylic acid ethyl ester (129);
(3 -Hydroxymethyl-piperidin- 1 -yl)- [4-(4-trifluoromethoxy-phenylamino)-quinazolin-7-yl] - methanone (130);
Table 6. Compounds of formula VII.
Figure imgf000029_0001
Figure imgf000030_0001
4-(4-Trifluoromethoxy-phenylamino)-5,7-dihydro-pyrrolo[2,3-d]pyrimidin-6-one (131); 7-Methyl-4-(4-trifluoromethoxy-phenylamino)-5,7-dihydro-pyiτolo[2,3-d]pyrimidin-6-one (132);
Table 7. Compounds of formula VIII.
Figure imgf000031_0001
4-(6-(4-(trifluoromethoxy)phenylamino)-5-methylpyrimidin-4-yl)-N-(2-moφholinoethyl) benzamide (133);
6-( 1 H-pyrrolo[2,3-b]pyridin- 1 -yl)-N-(4-(trifluoromethoxy)phenyl)pyrimidin-4-amine (134);
N-(4-(trifluoromethoxy)phenyl)-6-(l,3,5-trimethyl-lH-pyrazol-4-yl)pyrimidin-4-amine
(135);
4-[6-(4-Trifluoromethoxy-phenylamino)-pyrimidin-4-yl]-benzenesulfonamide (136);
4- [5 -Methyl-6-(4-trifluoromethoxy-pheny lamino)-pyrimidin-4-y 1] -benzenesulfonamide
(137);
N-Methyl-4-[5-methyl-6-(4-trifluoromethoxy-phenylamino)-pyrimidin-4-yl]- benzenesulfonamide (138);
N-Ethyl-4-[6-(4-trifluoromethoxy-phenylamino)-pyrimidin-4-yl]-benzenesulfonamide (139);
N-(2-Hydroxy-ethyl)-4-[6-(4-trifluoromethoxy-phenylamino)-pyrimidin-4-yl]- benzenesulfonamide (140);
[6-(4-Methanesulfonyl-phenyl)-pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)-amine (141);
[6-(4-Methanesulfonyl-phenyl)-5-methyl-pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)-amine
(142);
{6-[4-(Propane-2-sulfonyl)-phenyl]-pyrimidin-4-yl}-(4-trifluoromethoxy-phenyl)-amine
(143);
{5-Methyl-6-[4-(propane-2-sulfonyl)-phenyl]-pyrimidin-4-yl}-(4-trifluoromethoxy-phenyl)- amine (144);
N,N-Diethyl-4-[6-(4-trifluoromethoxy-phenylamino)-pyrimidin-4-yl]-benzenesulfonamide
(145); N,N-Diethyl-4-[5-methyl-6-(4-trifluoromethoxy-phenylamino)-pyrimidin-4-yl]- benzenesulfonamide (146);
{ 6-[4-(Piperidine- 1 -sulfonyl)-phenyl] -pyrimidin-4-yl } -(4-trifluoromethoxy-pheny l)-amine
(147);
4-Methyl-N-{4-[6-(4-trifluoromethoxy-phenylamino)-pyrimidin-4-yl]-phenyl}- benzenesulfonamide (148);
4-Methyl-N- {4- [5 -methy l-6-(4-trifluoromethoxy-phenylamino)-pyrimidin-4-yl] -phenyl } - benzenesulfonamide (149);
{6-[4-(Morpholine-4-sulfonyl)-phenyl]-pyrimidin-4-yl}-(4-trifluoromethoxy-phenyl)-amine
(150);
N-{4-[6-(4-Trifluoromethoxy-phenylamino)-pyrimidin-4-yl]-phenyl}-methanesulfonamide
(151);
Ethanesulfonic acid {4-[6-(4-trifluoromethoxy-phenylamino)-pyrimidin-4-yl]-phenyl}-amide
(152);
Butane- 1 -sulfonic acid {4-[6-(4-trifluoromethoxy-phenylamino)-pyrimidin-4-yl]-phenyl} - amide (153);
Propane-2-sulfonic acid {4-[6-(4-trifluoromethoxy-phenylamino)-pyrimidin-4-yl]-phenyl } - amide (154);
Pentane-2-sulfonic acid {4-[6-(4-trifluoromethoxy-phenylamino)-pyrimidin-4-yl]-phenyl} - amide (155);
N-(2-Hydroxy-ethyl)-4-[5-methyl-6-(4-trifluoromethoxy-phenylamino)-pyrimidin-4-yl]- benzamide (156);
4-[5-Methyl-6-(4-trifluoromethoxy-phenylamino)-pyrimidin-4-yl]-N-(2-morpholin-4-yl- ethyl)-benzamide (157);
N-(3-Methoxy-propyl)-4-[5-methyl-6-(4-trifluoromethoxy-phenylamino)-pyrimidin-4-yl]- benzamide (158);
N- { 3 -[5 -Methyl-6-(4-trifluoromethoxy-phenylamino)-pyrimidin-4-yl]-phenyl } -acetamide
(159);
3-[5-Methyl-6-(4-trifluoromethoxy-phenylamino)-pyrimidin-4-yl]-benzamide (160);
3-[5-Methyl-6-(4-trifluoromethoxy-phenylamino)-pyrimidin-4-yl]-benzoic acid (161);
(6-Biphenyl-4-yl-pyrimidin-4-yl)-(4-trifluoromethoxy-phenyl)-amine ( 162);
(6-Biphenyl-4-yl-5-methyl-pyrimidin-4-yl)-(4-trifluoromethoxy-phenyl)-amine (163); [6-(5-Methoxy-naphthalen-2-yl)-pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)-amine (164);
[6-(5-Methoxy-naphthalen-2-yl)-5-methyl-pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)- amine (165);
{ 3 - [6-(4-Trifluoromethoxy-phenylamino)-pyrimidin-4-y 1] -phenyl } -acetonitrile ( 166);
[6-(3-Nitro-phenyl)-pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)-amine (167);
4-Methoxy-N-{4-[6-(4-trifluoromethoxy-phenylamino)-pyrimidin-4-yl]-phenyl}-benzamide
(168);
[6-(4-Aminomethyl-phenyl)-5-methyl-pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)-amine
(169);
5-Bromo-N-(2-moφholin-4-yl-ethyl)-N'-(4-trifluoromethoxy-phenyl)-pyrimidine-4,6- diamine (170);
1 - [5 -Bromo-6-(4-trifluoromethoxy-phenylamino)-pyrimidin-4-yl] -piperidine-3 -carboxylic acid amide (171);
{ 6-[2-(2-Moφholin-4-yl-ethylamino)-imidazol- 1 -yl] -pyrimidin-4-yl } -(4-trifluoromethoxy- phenyl)-amine (172);
(4-Trifluoromethoxy-phenyl)-{6-[2-(4-trifluoromethoxy-phenylamino)-imidazol-l-yl]- pyrimidin-4-yl} -amine (173);
N-(5 -Cyclopropyl- 1 H-pyrazol-3 -yl)-N'-(4-trifluoromethoxy-pheny l)-pyrimidine-4,6-diamine
(174);
[6-(3-Amino-5-cyclopropyl-pyrazol-l-yl)-pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)- amine (175);
N-(4-Trifluoromethoxy-phenyl)-N'-[ 1 -(3 -trifluoromethyl-phenyl)- 1 H-imidazol-2-yl]- pyrimidine-4,6-diamine (176);
(6-Pyrrolo[2,3-b]pyridin- 1 -yl-pyrimidin-4-yl)-(4-trifluoromethoxy-phenyl)-amine (177);
[6-(lH-Pyrazol-4-yl)-pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)-amine (178);
[6-( 1 -Methyl- 1 H-pyrazol-4-yl)-pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)-amine ( 179);
[6-(l-Isobutyl-lH-pyrazol-4-yl)-pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)-amine (180);
[6-(l-Isobutyl-lH-pyrazol-4-yl)-5-methyl-pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)- amine (181);
(4-Trifluoromethoxy-phenyl)-[6-(l,3,5-trimethyl-lH-pyrazol-4-yl)-pyrimidin-4-yl]-amine
(182); {6-[ 1 -(4-Methanesulfonyl-benzyl)- 1 H-pyrazol-4-yl]-pyrimidin-4-yl } -(4-trifluoromethoxy- phenyl)-amine (183);
[6-( 1 -Ethanesulfonyl- 1 H-pyrazol-4-yl)-pyrimidin-4-yl] -(4-trifluoromethoxy-pheny l)-amine
(184);
[6-( 1 -Pyridin-4-ylmethyl- 1 H-pyrazol-4-yl)-pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)- amine (185);
N6-(2-Morpholin-4-yl-ethyl)-N6'-(4-trifluoromethoxy-phenyl)-[4,4']bipyrimidinyl-6,6'- diamine (186);
[5-Methyl-6-(l-methyl-lH-pyrazol-4-yl)-pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)-amine
(187);
[6-(5 -Methyl- 1 -phenyl- 1 H-pyrazol-4-yl)-pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)-amine
(188);
[5-Methyl-6-(5-methyl- 1 -phenyl- 1 H-pyrazol-4-yl)-pyrimidin-4-yl]-(4-trifluoromethoxy- phenyl)-amine (189);
[6-( 1 H-Indol-4-yl)-pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)-amine ( 190);
[6-( 1 H-Indol-3-yl)-pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)-amine (191);
[6-(lH-Indol-5-yl)-pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)-amine (192);
4-[6-(4-Trifluoromethoxy-phenylamino)-pyrimidin-4-yl]-furan-2-carbaldehyde (193);
(6-Benzo[b]thiophen-4-yl-pyrimidin-4-yl)-(4-trifluoromethoxy-phenyl)-amine (194);
[6-( 1 H-Indol-4-yl)-5 -methyl-pyrimidin-4-yl] -(4-trifluoromethoxy-phenyl)-amine (195);
(6-Dibenzofuran-4-yl-pyrimidin-4-yl)-(4-trifluoromethoxy-phenyl)-amine ( 196);
[6-( 1 -Benzyl- 1 H-pyrazol-4-yl)-pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)-amine ( 197);
[6-( 1 -Benzyl- 1 H-pyrazol-4-yl)-5-methyl-pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)-amine
(198);
N6,N6l-Bis-(4-trifluoromethoxy-phenyl)-[4,4']bipyrimidinyl-6,6'-diamine ( 199);
N6-(2-Mθφholin-4-yl-ethyl)-N6l-(4-trifluoromethoxy-phenyl)-[4,4']bipyrimidinyl-6,6'- diamine (200);
N-(4-Methyl-3-{ l-[6-(4-trifluoromethoxy-phenylamino)-pyrimidin-4-yl]-lH-imidazol-2- ylamino}-phenyl)-benzamide (201);
N-(4-Methyl-3-{l-[6-(4-trifluoromethoxy-phenylamino)-pyrimidin-4-yl]-lH-imidazol-2- ylamino}-phenyl)-4-moφholin-4-ylmethyl-3-trifluoromethyl-benzamide (202); [6-(l-Pyridin-3-ylmethyl-lH-pyrazol-4-yl)-pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)- amine (203);
[6-(lH-Pyrrolo[2,3-b]pyridin-4-yl)-pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)-amine (204);
4-[6-(4-Trifluoroniethoxy-phenylamino)-pyrimidin-4-yl]-lH-pyridin-2-one (205); [6-(2-Methyl-thiazol-4-yl)-pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)-amine (206); 2-Fluoro-5-[6-(4-trifluoromethoxy-phenylamino)-pyrimidin-4-yl]-benzaldehyde (207); {6-[2-(4-Methyl-piperazin-l-yl)-pyridin-4-yl]-pyrimidin-4-yl}-(4-trifluoromethoxy-phenyl)- amine (208);
Table 8. Compounds of formula IX.
Figure imgf000035_0001
Figure imgf000036_0001
Figure imgf000037_0001
Figure imgf000038_0001
Figure imgf000039_0001
Figure imgf000040_0001
2-(3-nitrophenyl)-N-(4-(trifluoromethoxy)phenyl)imidazo[l,2-c]pyrimidin-7-amine (209); 2-Hydroxy-3-[7-(4-trifluoromethoxy-phenylamino)-imidazo[l,2-c]pyrimidin-2-yl]- benzamide (210);
Table 9. Compounds of formula X.
Figure imgf000041_0001
Synthesis of Compounds of the Invention
The compounds of formulae II and III were formed over a two-step reaction. First a chlorinated heteroaryl compound was coupled with a haloalkoxy substituted aniline derivative under basic conditions. The resulting compound was then treated with a hydrazine derivative to provide a mixture of compounds of formulae II and III, which were separated by chromatography (Scheme 1). Alternatively, hydrazine is added followed by addition of an electrophile under basic conditions to provide the compounds of formulae II and III.
Scheme 1
Figure imgf000041_0002
Compounds of formulae IV and V were prepared by reacting a halogenated fused bicyclic heteroaryl compound with an optionally substituted chlorinated alkyl group under basic conditions. The resulting substituted amine was coupled at the heteroaryl chloro position with an aniline derivative to provide the compounds of formulae IV or V (Scheme 2). Scheme 2 also provides for functional group modification of the side chain of the substituted amine to provide the variously substituted compounds of the invention.
Scheme 2
Figure imgf000042_0001
Scheme 3 provides a synthetic route to arrive at the compounds of formulae V and VI. A heteroaryl compound with adjacent chloro and aldehyde substitution was reacted with a substituted thiol derivative or secondary amine derivative to provide the fused bicyclic framework of the compounds of formulae V and VI. Additional functional group modification provided for the compounds of the invention.
Scheme 3
Figure imgf000043_0001
The compounds of formula VII were prepared by coupling a halogenated fused bicyclic heteroaryl compound with a substituted aniline derivative. Additional functional group modification of the side chain provided the compounds of formula VII (Scheme 4).
Scheme 4
Figure imgf000043_0002
The compounds of formula IX were synthesized by initially coupling a dihalogenated heteraryl compound with an aniline derivative, followed by another coupling under Mitsunobu conditions to provide the compounds of formula IX. Alternatively, in the second step, a second halo group on the heteroaryl ring is displaced by a nucleophile under basic conditions to provide a compound of formula IX (Scheme 5).
Scheme 5
Figure imgf000044_0001
In addition to Scheme 5, compounds of formula IX were also made by reactions described in Scheme 6, wherein the product of the initial coupling is further coupled under Mitsunobu conditions to a heteroaryl group, which is then subjected to functional group modification.
Scheme 6
Figure imgf000044_0002
The aryl-substituted fused bicyclic compounds of formula X were synthesized according to Scheme 7. A heteroaryl compound substituted with a halo group and an amino group was reacted with an alpha halo carbonyl compound, which produced a bicyclic heteroaryl compound. Subsequent coupling with an aniline derivative providing the compound of formula X.
Scheme 7
Figure imgf000045_0001
Acids and bases useful in the methods herein are known in the art. Acid catalysts are any acidic chemical, which can be inorganic (e.g., hydrochloric, sulfuric, nitric acids, aluminum trichloride) or organic (e.g., camphorsulfonic acid, p-toluenesulfonic acid, acetic acid, ytterbium triflate) in nature. Acids are useful in either catalytic or stoichiometric amounts to facilitate chemical reactions. Bases are any basic chemical, which can be inorganic (e.g., sodium bicarbonate, potassium hydroxide) or organic (e.g., triethylamine, pyridine) in nature. Bases are useful in either catalytic or stoichiometric amounts to facilitate chemical reactions.
The compounds of the invention are defined herein by their chemical structures and/or chemical names. Where a compound is referred to by both a chemical structure and a chemical name, and the chemical structure and chemical name conflict, the chemical structure is determinative of the compound's identity.
Combinations of substituents and variables envisioned by this invention are only those that result in the formation of stable compounds. The term "stable", as used herein, refers to compounds which possess stability sufficient to allow manufacture and which maintains the integrity of the compound for a sufficient period of time to be useful for the purposes detailed herein (e.g., formulation into therapeutic products, intermediates for use in production of therapeutic compounds, isolatable or storable intermediate compounds, treatment or prevention of disorders disclosed herein). The compounds produced by the methods herein can be incorporated into compositions, including solutions, capsules, cremes, or ointments for administration to a subject (e.g., human, animal). Such compositions (e.g., pharmaceuticals) are useful for providing to the subject desirable health or other physiological benefits that are associated with such compounds.
Nucleophilic agents are known in the art and are described in the chemical texts and treatises referred to herein. The chemicals used in the aforementioned methods may include, for example, solvents, reagents, catalysts, protecting group and deprotecting group reagents and the like. The methods described above may also additionally include steps, either before or after the steps described specifically herein, to add or remove suitable protecting groups in order to ultimately allow synthesis of the compound of the formulae described herein. The methods delineated herein contemplate converting compounds of one formula to compounds of another formula. The process of converting refers to one or more chemical transformations, which can be performed in situ, or with isolation of intermediate compounds. The transformations can include reacting the starting compounds or intermediates with additional reagents using techniques and protocols known in the art, including those in the references cited herein. Intermediates can be used with or without purification (e.g., filtration, distillation, crystallization, chromatography). Other embodiments relate to the intermediate compounds delineated herein, and their use in the methods (e.g., treatment, synthesis) delineated herein.
Methods of Treatment
The compounds of the invention in free form or in pharmaceutically acceptable salt form, exhibit valuable pharmacological properties, for example, as indicated by the in vitro tests described within "Assays", infra, and are therefore indicated for therapy of diseases and disorders associated with Bcr-abl activity. For Bcr-abl, compounds of the invention preferably show an IC50 in the range of 1x10"' to 1x10~5 M, preferably less than 1 μM for wild-type Bcr-abl and at least two other Bcr-abl mutants (mutants selected from G250E, E255V, T315I, F317L and M351T).
The invention also provides a method for preventing or treating diseases or conditions comprising abnormal cell growth in a subject, including a human, comprising administering to the subject a compound of the invention in an amount effective to inhibit PDGF-R, c-Kit and/or Bcr-abl activity.
PDGF (Platelet-derived Growth Factor) is a very commonly occurring growth factor, which plays an important role both in normal growth and also in pathological cell proliferation, such as is seen in carcinogenesis and in diseases of the smooth-muscle cells of blood vessels, for example in atherosclerosis and thrombosis.
Compounds of the invention can inhibit PDGF-R and are, therefore, also suitable for the treatment of tumor diseases, which are delineated infra.
The compounds of the present invention also inhibit cellular processes involving stem-cell factor (SCF, also known as the c-kit ligand or steel factor), such as SCF receptor (kit) autophosphorylation and the SCF-stimulated activationof MAPK kinase (mitogen- activated protein kinase). The compounds of the present invention thus inhibit also the autophosphorylation of SCF receptor (and c-kit, a proto-oncogene). M07e cells are a human promegakaryocytic leukemia cell line, which depends on SCF for proliferation. A compound of the invention inhibits the autophosphorylationof SCF-R in the micromolar range.
Also abl kinase, especially v-abl kinase, is inhibited by compounds of the present invention. By analogy, the compounds of the present invention also inhibit Bcr- abl kinase and are thus suitable for the treatment of Bcr-abl-positive cancer and tumor diseases, such as leukemias (especially chronic myeloid leukemia and acutelymphoblastic leukemia, where especially apoptotic mechanisms of action are found), and also shows effects on the subgroup of leukemic stem cells as well as potential for the purification of these cells in vitro after removal of said cells (for example, bone marrow removal) and reimplantation of the cells once they have been cleared of cancer cells (for example, reimplantation of purified bone marrow cells).
Furthermore, the present invention provides a method for inhibiting Bcr-abl activity, the method comprising contacting Bcr-abl with a compound of the invention that binds to a myristoyl binding pocket of Bcr-abl.
In addition, the compounds of the present invention can be used in combination with other anti-tumor agents.
Treatment of Diseases
The term "tumor" is used to describe an abnormal growth in tissue which occurs when cellular proliferation is more rapid than normal tissue and continues to grow after the stimuli that initated the new growth cease. Tumors generally exhibit partial or complete lack of structural organization and functional coordination with the normal tissue, and usually form a distinct mass of tissue which may be benign (benign tumor) or malignant (carcinoma). Tumors tend to be highly vascularized.
The term "cancer" is used as a general term herein to describe malignant tumors or carcinoma. These malignant tumors may invade surrounding tissues, may metastasize to several sites and are likely to recur after attempted removal and to cause death of the patient unless adequately treated. As used herein, the terms carcinoma and cancer are subsumed under the term tumor.
Diseases or disorders treated, ameliorated or prevented by the instant invention include the following: sarcomas, neoplasia, internal malignancies such as eye or ocular cancer, rectal cancer, colon cancer, cervical cancer, prostate cancer, breast cancer and bladder cancer, benign and malignant tumors, including various cancers such as, anal and oral cancers, stomach, rectal, liver, pancreatic, lung, cervix uteri, corpus uteri, ovary, prostate, testis, renal, mouth/pharynx, esophageal, larynx, kidney, brain/ens (e.g., gliomas), head and neck, throat, skin melanoma, acute lymphocytic leukemia, acute myelogenous leukemia, Ewing's Sarcoma, Kaposi's Sarcoma, basal cell carinoma and squamous cell carcinoma, small cell lung cancer, choriocarcinoma, rhabdomyosarcoma, angiosarcoma, hemangioendothelioma, Wilms Tumor, neuroblastoma, lymphoma, neurofibromatosis, tuberous sclerosis (each of which conditions produces benign tumors of the skin), hemangiomas, lymphangiogenesis, rhabdomyosarcomas, retinoblastoma, osteosarcoma, acoustic neuroma, neurofibroma, trachoma, pyogenic granulomas, blood-bom tumors such as leukemias, any of various acute or chronic neoplastic diseases of the bone marrow in which unrestrained proliferation of white blood cells occurs, usually accompanied by anemia, impaired blood clotting, and enlargement of the lymph nodes, liver, and spleen, psoriasis, acne, rosacea, warts, eczema, neurofibromatosis, Sturge- Weber syndrome, venous ulcers of the skin, tuberous sclerosis, chronic inflammatory disease, arthritis, lupus, scleroderma, diabetic retinopathy, retinopathy of prematurity, corneal graft rejection, neovascular glaucoma and retrolental fibroplasias, epidemic keratoconjunctivitis, vitamin A deficiency, contact lens overwear, atopic keratitis, superior limbic keratitis, pterygium, keratitis sicca, Sjogren's, phylectenulosis, syphilis, Mycobacteria infections, lipid degeneration, chemical burns, bacterial ulcers, fungal ulcers, herpes simplex infections, herpes zoster infections, protozoan infections, Mooren's ulcer, Terrien's marginal degeneration, marginal keratolysis, trauma, rheumatoid arthritis, systemic lupus, polyarteritis, Wegener's sarcoidosis, scleritis, Stevens- Johnson disease, pemphigoid, radial keratotomy, corneal graft rejection, diabetic retinopathy, macular edema, macular degeneration, sickle cell anemia, sarcoid, pseudoxanthoma elasticum, Paget's disease, vein occlusion, artery occlusion, carotid obstructive disease, chronic uveitis/vitritis, Lyme disease, systemic lupus erythematosus, Eales1 disease, Behcet's disease, infections causing a retinitis or choroiditis, presumed ocular histoplasmosis, Best's disease, myopia, optic pits, Stargardt's disease, pars planitis, chronic retinal detachment, hyperviscosity syndromes, toxoplasmosis, trauma, post-laser complications, rubeosis (neovascularization of the ankle), diseases caused by the abnormal proliferation of fibrovascular or fibrous tissue including all forms of proliferative vitreoretinopathy, whether or not associated with diabetes, neovascular disease, pannus, diabetic macular edema, vascular retinopathy, retinal degeneration, inflammatory diseases of the retina, proliferative vitreoretinopathy, diseases associated with rubeosis (neovascularization of the ankle), diseases caused by the abnormal proliferation of fibrovascular or fibrous tissue including all forms of proliferative vitreoretinopathy, Crohn's disease and ulcerative colitis, sarcoidosis, osteoarthritis, inflammatory bowel diseases, skin lesions, Osier- Weber-Rendu disease, or hereditary hemorrhagic telangiectasia, osteoarthritis, Sarcoidosis, skin lesions, acquired immune deficiency syndrome, and small bowel obstruction.
In a particular embodiment, the inhibition of the kinase activity of Bcr-abl is an important aspect of the present invention. More particularly, the present invention relates to methods for treating cancers, including Chronic Myelogenous Leukemia, comprising exposing the cancer to an inhibitory or therapeutically effective amount or concentration of at least one of the disclosed compounds. This method may be used therapeutically, in comparison tests such as assays for determining the activities of related analogs as well as for determining the susceptibility of a patient's cancer to one or more of the compounds according to the present invention.
Kinase inhibiting compounds of the present invention are used to treat, ameliorate or prevent, among others, benign and malignant tumors, including various cancers such as, Chronic Myelogenous Leukemia, cervical, anal and oral cancers, stomach, colon, bladder, rectal, liver, pancreatic, lung, breast, cervix uteri, corpus uteri, ovary, prostate, testis, renal, brain/ens (e.g., gliomas), head and neck, eye or ocular, throat, skin melanoma, acute lymphocytic leukemia, acute myelogenous leukemia, Ewing's Sarcoma, Kaposi's Sarcoma, basal cell carinoma and squamous cell carcinoma, small cell lung cancer, choriocarcinoma, rhabdomyosarcoma, angiosarcoma, hemangioendothelioma, Wilms Tumor, neuroblastoma, mouth/pharynx, esophageal, larynx, kidney and lymphoma, among others. In addition, conditions such as neurofibromatosis, tuberous sclerosis (each of which conditions produces benign tumors of the skin), hemangiomas and lymphangiogenesis, among others, may be treated effectively with compounds according to the present invention.
The compounds of the invention are useful in treating non-malignant proliferative disorders, such as atherosclerosis, thrombosis, psoriasis, scleroderma, and fibrosis, as well as for the protection of stem cells, for example to combat the hemotoxic effect of chemotherapeutic agents, such as 5-fluoruracil, and in asthma. It can especially be used for the treatment of diseases, which respond to an inhibition of the PDGF-R kinase.
Methods for treating, ameliorating, or preventing skin disorders such as psoriasis, acne, rosacea, warts, eczema, hemangiomas, lymphangiogenesis, neurofibromatosis, Sturge- Weber syndrome, venous ulcers of the skin, tuberous sclerosis, chronic inflammatory disease and arthritis, as well as inflammation such as chronic inflammatory disease, including arthritis, lupus and scleroderma are also contemplated by the present invention, such methods comprising administering a therapeutically effective amount of one or more of the disclosed compounds to a subject in need of such treatment.
Diseases associated with neovascularization inlcude optic disc neovascularization, iris neovascularization, retinal neovascularization, choroidal neovascularization, corneal neovascularization, and intravitreal neovascularization.
Diseases associated with corneal neovascularization and retinal/choroidal neovascularization that can be treated, ameliorated, or prevented, according to the present invention include but are not limited to, diabetic retinopathy, retinopathy of prematurity, corneal graft rejection, neovascular glaucoma and retrolental fibroplasias, epidemic keratoconjunctivitis, vitamin A deficiency, contact lens overwear, atopic keratitis, superior limbic keratitis, pterygium keratitis sicca, Sjogren's, acne rosacea, phylectenulosis, syphilis, Mycobacteria infections, lipid degeneration, chemical bums, bacterial ulcers, fungal ulcers, herpes simplex infections, herpes zoster infections, protozoan infections, Kaposi's sarcoma, Mooren's ulcer, Terrien's marginal degeneration, marginal keratolysis, trauma, rheumatoid arthritis, systemic lupus, polyarteritis, Wegener's sarcoidosis, scleritis, Stevens- Johnson disease, pemphigoid, radial keratotomy, macular edema, macular degeneration, sickle cell anemia, sarcoid, syphilis, pseudoxanthoma elasticum, Paget's disease, vein occlusion, artery occlusion, carotid obstructive disease, chronic uveitis/vitritis, mycobacterial infections, Lyme disease, systemic lupus erythematosus, retinopathy of prematurity, Eales' disease, Behcet's disease, infections causing a retinitis or choroiditis, presumed ocular histoplasmosis, Best's disease, myopia, optic pits, Stargardt's disease, pars planitis, chronic retinal detachment, hyperviscosity syndromes, toxoplasmosis, trauma and post-laser complications. Other diseases include, but are not limited to, diseases associated with rubeosis (neovascularization of the ankle), diseases caused by the abnormal proliferation of fibrovascular or fibrous tissue including all forms of proliferative vitreoretinopathy, whether or not associated with diabetes, and corneal graft rejection.
Diseases associated with chronic inflammation and arthritis can be treated, ameliorated or prevented by the compositions and methods of the present invention. Diseases with symptoms of chronic inflammation include inflammatory bowel diseases such as Crohn's disease and ulcerative colitis, psoriasis, sarcoidosis, rheumatoid arthritis, osteoarthritis, lupus and scleroderma.
In addition, the compounds of the present invention show useful effects in the treatment of disorders arising as a result of transplantation, for example, allogenic transplantation, especially tissue rejection, such as especiallyobliterative bronchiolitis (OB), i.e., a chronic rejection of allogenic lung transplants. In contrast to patients without OB, those with OB often show an elevated PDGF concentration in bronchoalveolar lavage fluids. Synergistic effects with other immunomodulatory or anti- inflammatory substances are possible, for example when used in combination with cyclosporin, rapamycin, or ascomycin, or immunosuppressant analogues thereof, for example cyclosporin A (CsA), cyclosporin G, FK-506, rapamycin, or comparable compounds, corticosteroids, cyclophosphamide, azathioprine, methotrexate, brequinar, leflunomide, mizoribine, mycophenolic acid, mycophenolate mofetil, 15-deoxyspergualin, immunosuppressant antibodies, especially monoclonal antibodies for leukocyte receptors, for example MHC, CD2,CD3, CD4, CD7, CD25,CD28, B7, CD45, CD58 or their ligands, or other immunomodulatory compounds, such as CTLA41g.
The compounds of the present invention are also effective in diseases associated with vascular smooth-muscle cell migration and proliferation (where PDGF and PDGF-R often also play a role), such as restenosis and atherosclerosis.
Pharmaceutical Compositions
Pharmaceutical compositions and dosage forms of the invention comprise one or more of the active ingredients disclosed herein. Pharmaceutical compositions and dosage forms of the invention typically also comprise one or more pharmaceutically acceptable excipients or diluents.
The term "pharmaceutically acceptable," as used herein, refers to a component that is, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and other mammals without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio. A "pharmaceutically acceptable salt" means any non-toxic salt that, upon administration to a recipient, is capable of providing, either directly or indirectly, a compound or a prodrug of a compound of this invention. A "pharmaceutically acceptable counterion" is an ionic portion of a salt that is not toxic when released from the salt upon administration to a recipient.
The invention also provides compositions comprising an effective amount of a composition containing a compound of the invention and an acceptable carrier. Preferably, a composition of this invention is formulated for pharmaceutical use ("a pharmaceutical composition"), wherein the carrier is a pharmaceutically acceptable carrier. The carrier(s) must be "acceptable" in the sense of being compatible with the other ingredients of the formulation and, in the case of a pharmaceutically acceptable carrier, not deleterious to the recipient thereof in amounts typically used in medicaments.
Pharmaceutically acceptable carriers, adjuvants and vehicles that may be used in the pharmaceutical compositions of this invention 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. Typical pharmaceutical compositions and dosage forms comprise one or more excipients. Suitable excipients are well known to those skilled in the art of pharmacy, and non-limiting examples of suitable excipients are provided herein. Whether a particular excipient is suitable for incorporation into a pharmaceutical composition or dosage form depends on a variety of factors well known in the art including, but not limited to, the way in which the dosage form will be administered to a patient. Excipients can take a wide variety of forms depending on the form of preparation desired for administration. For example, excipients suitable for use in oral liquid or aerosol dosage forms include, but are not limited to, water, glycols, oils, alcohols, flavoring agents, preservatives, and coloring agents. Examples of excipients suitable for use in solid oral dosage forms (e.g., powders, tablets, capsules, and caplets) include, but are not limited to, starches, sugars, micro-crystalline cellulose, diluents, granulating agents, lubricants, binders, and disintegrating agents.
Such preparative methods include the step of bringing into association with the molecule to be administered ingredients such as the carrier that constitutes one or more accessory ingredients. In general, the compositions are prepared by uniformly and intimately bringing into association the active ingredients with liquid carriers, liposomes or finely divided solid carriers or both, and then if necessary shaping the product.
The pharmaceutical compositions according to the invention are those suitable for enteral, such as oral or rectal, transdermal, topical, and parenteral administration to mammals, including man, to inhibit Bcr-abl activity, and for the treatment of Bcr-abl dependent disorders, and comprise an effective amount of a pharmacologically active compound of the present invention, alone or in combination, with one or more pharmaceutically acceptable carriers. More particularly, the pharmaceutical compositions comprise an effective Bcr-abl inhibiting amount of a compound of the present invention. Single unit dosage forms of the invention are suitable for oral, mucosal (e.g., nasal, sublingual, vaginal, buccal, or rectal), parenteral (e.g., subcutaneous, intravenous, bolus injection, intramuscular, intraarterial, intracutaneous, intraarticular, intrasynovial, intrasternal, intrathecal, intralesional and intracranial injection or infusion techniques.), by inhalation spray, topically, rectally, nasally, buccally, vaginally, via an implanted reservoir, or transdermal administration to a patient.
Examples of dosage forms include, but are not limited to: tablets; caplets; capsules, such as soft elastic gelatin capsules; cachets; troches; lozenges; dispersions; suppositories; ointments; cataplasms (poultices); pastes; powders; dressings; creams; plasters; solutions; patches; aerosols (e.g., nasal sprays or inhalers); gels; liquid dosage forms suitable for oral or mucosal administration to a patient, including suspensions (e.g., aqueous or non-aqueous liquid suspensions, oil-in-water emulsions, or a water-in-oil liquid emulsions), solutions, and elixirs; liquid dosage forms suitable for parenteral administration to a patient; and sterile solids (e.g., crystalline or amorphous solids) that can be reconstituted to provide liquid dosage forms suitable for parenteral administration to a patient.
The composition, shape, and type of dosage forms of the invention will typically vary depending on their use. For example, a dosage form used in the acute treatment of a disorder may contain larger amounts of one or more of the active ingredients it comprises than a dosage form used in the chronic treatment of the same disease. Similarly, a parenteral dosage form may contain smaller amounts of one or more of the active ingredients it comprises than an oral dosage form used to treat the same disease or disorder. These and other ways in which specific dosage forms encompassed by this invention will vary from one another will be readily apparent to those skilled in the art. See, e.g., Remington's Pharmaceutical Sciences, 18th ed., Mack Publishing, Easton Pa. (1990).
A composition for oral administration can be any orally acceptable dosage form including, but not limited to, capsules, tablets, emulsions and aqueous suspensions, dispersions and solutions. In the case of tablets for oral use, carriers which are commonly used include lactose and corn starch. Lubricating agents, such as magnesium stearate, are also typically added. For oral administration in a capsule form, useful diluents include lactose and dried corn starch. When aqueous suspensions or emulsions are administered orally, the active ingredient can be suspended or dissolved in an oily phase combined with emulsifying or suspending agents. If desired, certain sweetening, flavoring, or coloring agents can be added. A nasal aerosol or inhalation composition can be prepared according to techniques well-known in the art of pharmaceutical formulation and can be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other solubilizing or dispersing agents known in the art. A heterocyclic compound of this invention can also be administered in the form of suppositories for rectal administration.
If desired, tablets can be coated by standard aqueous or nonaqueous techniques. Such dosage forms can be prepared by any of the methods of pharmacy. In general, pharmaceutical compositions and dosage forms are prepared by uniformly and intimately admixing the active ingredients with liquid carriers, finely divided solid carriers, or both, and then shaping the product into the desired presentation if necessary.
Preferred are tablets and gelatin capsules comprising the active ingredient together with a) diluents, e. g., lactose, dextrose, sucrose, mannitol, sorbitol, cellulose and/or glycine; b) lubricants, e. g., silica, talcum, stearic acid, its magnesium or calcium salt and/or polyethyleneglycol; for tablets also c) binders, e. g., magnesium aluminum silicate, starch paste, gelatin, tragacanth, methylcellulose, sodium carboxymethylcellulose and or polyvinylpyrrolidone; if desired d) disintegrants, e. g., starches, agar, alginic acid or its sodium salt, or effervescent mixtures; and/or e) absorbents, colorants, flavors and sweeteners. Tablets may be either film coated or enteric coated according to methods known in the art.
For example, a tablet can be prepared by compression or molding. Compressed tablets can be prepared by compressing in a suitable machine the active ingredients in a free-flowing form such as powder or granules, optionally mixed with an excipient. Molded tablets can be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent.
Injectable compositions are preferably aqueous isotonic solutions or suspensions, and suppositories are preferably prepared from fatty emulsions or suspensions. The compositions may be sterilized and/or contain adjuvants, such as preserving, stabilizing, wetting or emulsifying agents, solution promoters, salts for regulating the osmotic pressureand/or buffers. In addition, they may also contain other therapeutically valuable substances. Said compositions are prepared according to conventional mixing, granulating or coating methods, respectively, and contain about 0.1 to 75%, preferably about 1 to 50%, of the active ingredient. A sterile injectable composition, for example, a sterile injectable aqueous or oleaginous suspension, can be formulated according to techniques known in the art using suitable dispersing or wetting agents (such as, for example, Tween 80) and suspending agents. The sterile injectable preparation can also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent. Among the acceptable vehicles and solvents that can be employed are mannitol, water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium (e.g., synthetic mono- or diglycerides). 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 can also contain a long-chain alcohol diluent or dispersant, or carboxymethyl cellulose or similar dispersing agents. Other commonly used surfactants such as Tweens or Spans or other similar emulsifying agents or bioavailability enhancers which are commonly used in the manufacture of pharmaceutically acceptable solid, liquid, or other dosage forms can also be used for the purposes of formulation.
Suitable formulations for transdermal application include an effective amount of a compound of the present invention with carrier. Preferred carriers include absorbable pharmacologically acceptable solvents to assist passage through the skin of the host. For example, transdermal devices are in the form ofa bandage comprising a backing member,a reservoir containing the compound optionally with carriers, optionally a rate controlling barrier to deliver the compound to the skin of the host at a controlled and predetermined rate over a prolonged period of time, and means to secure the device to the skin. Matrix transdermal formulations may also be used.
Suitable formulations for topical application, e.g., to the skin and eyes, are preferably aqueous solutions, ointments, creams or gels well-known in the art. Such may containsolubilizers, stabilizers, tonicity enhancing agents, buffers and preservatives.
The pharmaceutical formulations contain an effective Bcr-abl inhibiting amount of a compound of the present invention as defined above, either alone or in combination with another therapeutic agent. In conjunction with another active ingredient, a compound of the present invention may be administered either simultaneously, before or after the other active ingredient, either separately by the same or different route of administration or together in the same pharmaceutical formulation.
The carrier in the pharmaceutical composition must be "acceptable" in the sense of being compatible with the active ingredient of the formulation (and preferably, capable of stabilizing it) and not deleterious to the subject to be treated. For example, solubilizing agents such as cyclodextrins, which form specific, more soluble complexes with the compounds of this invention, or one or more solubilizing agents, can be utilized as pharmaceutical excipients for delivery of the compounds of the invention. Examples of other carriers include colloidal silicon dioxide, magnesium stearate, cellulose, sodium lauryl sulfate, and D&C Yellow # 10. As used herein, the terms "animal", "subject" and "patient", include, but are not limited to, a cow, monkey, horse, sheep, pig, chicken, turkey, quail, cat, dog, mouse, rat, rabbit, guinea pig and human (preferably, a human).
The pharmacologically active compounds of the present invention are useful in the manufacture of pharmaceutical compositions comprising an effective amount thereof in conjunction or mixture with excipients or carriers suitable for either enteral, parenteral, oral, mucosal, or topical application.
The interrelationship of dosages for animals and humans (based on milligrams per meter squared of body surface) is described in Freireich et al., (1966) Cancer Chemother Rep 50: 219. Body surface area may be approximately determined from height and weight of the patient. See, e.g., Scientific Tables, Geigy Pharmaceuticals, Ardley, N. Y., 1970, 537. An effective amount of a compound of this invention can range from about 0.001 mg/kg to about 1000 mg/kg, more preferably 0.01 mg/kg to about 100 mg/kg, more preferably 0.1 mg/kg to about 10 mg/kg; or any range in which the low end of the range is any amount between 0.001 mg/kg and 900 mg/kg and the upper end of the range is any amount between 0.1 mg/kg and 1000 mg/kg (e.g., 0.005 mg/kg and 200 mg/kg, 0.5 mg/kg and 20 mg/kg). Effective doses will also vary, as recognized by those skilled in the art, depending on the diseases treated, route of administration, excipient usage, and the possibility of co-usage with other therapeutic treatments such as use of other agents.
The dosage of active compound administered is dependent on the species of warmblooded animal (mammal), the body weight, age and individual condition, and on the form of administration. A unit dosage for oral administration to a mammal of about 50 to 70 kg may contain between about 5 and 500 mg of the active ingredient.
A compound of the invention can, for example, be administered with a dosage ranging from about 0.5 to about 100 mg/kg of body weight, alternatively dosages between 1 mg and 1000 mg/dose, every 4 to 120 hours, or according to the requirements of the particular drug. The methods herein contemplate administration of an effective amount of compound or compound composition to achieve the desired or stated effect. Typically, the pharmaceutical compositions of this invention will be administered from about 1 to about 6 times per day or alternatively, as a continuous infusion. Such administration can be used as a chronic or acute therapy. The amount of active ingredient that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration. A typical preparation will contain from about 5% to about 95% active compound (w/w). Alternatively, such preparations contain from about 20% to about 80% active compound.
Lower or higher doses than those recited above may be required. Specific dosage and treatment regimens 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 status, sex, diet, time of administration, rate of excretion, drug combination, the severity and course of the disease, condition or symptoms, the patient's disposition to the disease, condition or symptoms, and the judgment of the treating physician.
Definitions
As used herein, the term "alkyl" refers to a straight-chained or branched hydrocarbon group containing 1 to 12 carbon atoms. The term "lower alkyl" refers to a C1-C6 alkyl chain. Examples of alkyl groups include methyl, ethyl, n-propyl, isopropyl, tert-butyl, and n-pentyl. Alkyl groups may be optionally substituted with one or more substituents.
The term "alkenyl" refers to an unsaturated hydrocarbon chain that may be a straight chain or branched chain, containing 2 to 12 carbon atoms and at least one carbon-carbon double bond. Alkenyl groups may be optionally substituted with one or more substituents.
The term "alkynyl" refers to an unsaturated hydrocarbon chain that may be a straight chain or branched chain, containing the 2 to 12 carbon atoms and at least one carbon-carbon triple bond. Alkynyl groups may be optionally substituted with one or more substituents.
The sp2 or sp carbons of an alkenyl group and an alkynyl group, respectively, may optionally be the point of attachment of the alkenyl or alkynyl groups.
The term "alkoxy" refers to an -O-alkyl radical.
An "amido" is an -C(O)NH2, and an "N-alkyl-substituted amido" is of the formula C(O)NHR; where a divalent "amide" group is indicated, the group is -C(O)N- or -NC(O)-.
As used herein, the term "halogen," "halo," or "hal" means -F, -Cl, -Br or -I.
As used herein, the term "haloalkyl" means an alkyl group in which one or more (including all) of the hydrogen radicals are replaced by a halo group, wherein each halo group is independently selected from -F, -Cl, -Br, and -I. The term "halomethyl" means a methyl in which one to three hydrogen radical(s) have been replaced by a halo group. Representative haloalkyl groups include trifluoromethyl, bromomethyl, 1,2-dichloroethyl, 4-iodobutyl, 2- fluoropentyl, and the like. The term "cycloalkyl" refers to a hydrocarbon 3-8 membered monocyclic or 7-14 membered bicyclic ring system having at least one saturated ring, or hydrocarbon 3-8 membered monocyclic or 7-14 membered bicyclic ring system having at least one non- aromatic ring, wherein the non-aromatic ring has some degree of unsaturation. Cycloalkyl groups may be optionally substituted with one or more substituents. In one embodiment, 0, 1, 2, 3, or 4 atoms of each ring of a cycloalkyl group may be substituted by a substituent. Representative examples of cycloalkyl group include cyclopropyl, cyclopentyl, cyclohexyl, cyclobutyl, cycloheptyl, cyclooctyl, cyclononyl, cyclodecyl, cyclohexenyl, bicyclo[2.2.1]hept-2-enyl, dihydronaphthalenyl, benzocyclopentyl, cyclopentenyl, cyclopentadienyl, cyclohexenyl, cyclohexadienyl,cycloheptenyl, cycloheptadienyl, cycloheptatrienyl, cyclooctenyl, cyclooctadienyl, cyclooctatrienyl, cyclooctatetraenyl, cyclononenyl, cyclononadienyl, cyclodecenyl, cyclodecadienyl and the like.
The term "aryl" refers to a hydrocarbon monocyclic, bicyclic or tricyclic aromatic ring system. Aryl groups may be optionally substituted with one or more substituents. In one embodiment, 0, 1, 2, 3, 4, 5 or 6 atoms of each ring of an aryl group may be substituted by a substituent. Examples of aryl groups include phenyl, naphthyl, anthracenyl, fluorenyl, indenyl, azulenyl, and the like.
As used herein, the term "aralkyl" means an aryl group that is attached to another group by a (d-C6)alkylene group. Aralkyl groups may be optionally substituted, either on the aryl portion of the aralkyl group or on the alkylene portion of the aralkyl group, with one or more substituents. Representative aralkyl groups include benzyl, 2-phenyl-ethyl, naphth- 3-yl-methyl and the like.
As used herein, the term "alkylene" refers to an alkyl group that has two points of attachment. The term "(Ci-C6)alkylene" refers to an alkylene group that has from one to six carbon atoms. Non-limiting examples of alkylene groups include methylene (-CH2-), ethylene (-CH2CH2-), n-propylene (-CH2CH2CH2-), isopropylene (-CH2CH(CH3)-), and the like.
The term "arylalkoxy" refers to an alkoxy substituted with aryl.
The term "heteroaryl" refers to a 5-8 membered monocyclic, 8-12 membered bicyclic, or 11-14 membered tricyclic ring system having 1-4 ring heteroatoms if monocyclic, 1-6 heteroatoms if bicyclic, or 1-9 heteroatoms if tricyclic, said heteroatoms selected from O, N, or S, and the remainder ring atoms being carbon (with appropriate hydrogen atoms unless otherwise indicated), wherein at least one ring in the ring system is aromatic. Heteroaryl groups may be optionally substituted with one or more substituents. In one embodiment, 0, 1, 2, 3, or 4 atoms of each ring of a heteroaryl group may be substituted by a substituent. Examples of heteroaryl groups include pyridyl, 1-oxo-pyridyl, furanyl, benzo[l,3]dioxolyl, benzo[l,4]dioxinyl, thienyl, pyrrolyl, oxazolyl, oxadiazolyl, imidazolyl thiazolyl, isoxazolyl, quinolinyl, pyrazolyl, isothiazolyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl, triazolyl, thiadiazolyl, isoquinolinyl, indazolyl, benzoxazolyl, benzofuryl, indolizinyl, imidazopyridyl, tetrazolyl, benzimidazolyl, benzothiazolyl, benzothiadiazolyl, benzoxadiazolyl, indolyl, tetrahydroindolyl, azaindolyl, imidazopyridyl, quinazolinyl, purinyl, pyrrolo[2,3]pyrimidinyl, pyrazolo[3,4]pyrimidinyl, and benzo(b)thienyl, 3H-thiazolo[2,3-c][l,2,4]thiadiazolyl, imidazo[l,2-d]-l,2,4-thiadiazolyl, imidazo[2,l-b]-l,3,4-thiadiazolyl, lH,2H-furo[3,4-d]- 1,2,3-thiadiazolyl, lH-pyrazolo[5,l-c]-l,2,4-triazolyl, pyrrolo[3,4-d]-l,2,3-triazolyl, cyclopentatriazolyl, 3H-pyrrolo[3,4-c]isoxazolyl, lH,3H-pyrrolo[l,2-c]oxazolyl, pyrrolo[2,lb]oxazolyl, and the like.
As used herein, the term "heteroaralkyl" or "heteroarylalkyl" means a heteroaryl group that is attached to another group by a (Ci-C6)alkylene. Heteroaralkyl groups may be optionally substituted, either on the heteroaryl portion of the heteroaralkyl group or on the alkylene portion of the heteroaralkyl group, with one or more substituent. Representative heteroaralkyl groupss include 2-(pyridin-4-yl)-propyl, 2-(thien-3-yl)-ethyl, imidazol-4-yl- methyl and the like.
The term "heterocycloalkyl" refers to a nonaromatic 3-8 membered monocyclic, 7-12 membered bicyclic, or 10-14 membered tricyclic ring system comprising 1-3 heteroatoms if monocyclic, 1-6 heteroatoms if bicyclic, or 1-9 heteroatoms if tricyclic, said heteroatoms selected from O, N, S, B, P and Si, preferably O, N, and S, wherein the nonaromatic ring system is completely saturated. The term "heterocycloalkyl" also refers to nonaromatic 5-8 membered monocyclic, 7-12 membered bicyclic, or 10-14 membered tricyclic ring system comprising 1-3 heteroatoms if monocyclic, 1-6 heteroatoms if bicyclic, or 1-9 heteroatoms if tricyclic, said heteroatoms selected from O, N, S, B, P or Si, wherein the nonaromatic ring system has some degree of unsaturation. Bicyclic and tricyclic ring systems may be fused ring systems or spiro ring systems. Heterocycloalkyl groups may be optionally substituted with one or more substituents. In one embodiment, 0, 1, 2, 3, or 4 atoms of each ring of a heterocycloalkyl group may be substituted by a substituent. Representative heterocycloalkyl groups include piperidinyl, piperazinyl, 2-oxopiperazinyl, 2-oxopiperidinyl, 2- oxopyrrolidinyl, 4-piperidonyl, tetrahydropyranyl, tetrahydrothiopyranyl, tetrahydrothiopyranyl sulfone, morpholinyl, thiomorpholinyl, thiomorpholinyl sulfoxide, thiomorpholinyl sulfone, 1,3-dioxolane, l,4-dioxa-8-aza-spiro[4.5]dec-8-yl, tetrahydrofuranyl, tetrahydrothienyl, thiirene, thiirenyl, thiadiazirinyl, dioxazolyl, 1 ,3- oxathiolyl, 1 ,3-dioxolyl, 1,3-dithiolyl, oxathiazinyl, dioxazinyl, dithiazinyl, oxadiazinyl, thiadiazinyl, oxazinyl, thiazinyl, l,4-oxathiin,l,4-dioxin, 1,4-dithiin, lH-pyranyl, oxathiepinyl, 5H-l,4-dioxepinyl, 5H-l,4-dithiepinyl, 6H-isoxazolo[2,3-d]l,2,4-oxadiazolyl, 7aH-oxazolo[3,2-d]l,2,4-oxadiazolyl, and the like..
The term "alkylamino" refers to an amino substituent which is further substituted with one or two alkyl groups. The term "aminoalkyl" refers to an alkyl substituent which is further substituted with one or more amino groups.
The term "effective amount" is used throughout the specification to describe concentrations or amounts of compounds according to the present invention which may be used to produce a favorable change in the disease or condition treated, whether that change is a remission, a decrease in growth or size of cancer, tumor or other growth, a favorable physiological result including the clearing up of skin or tissue, or the like, depending upon the disease or condition treated.
As used herein, the terms "prevent," "preventing," "prevention," and the like refer to reducing the probability of developing a disorder or condition in a subject, who does not have, but is at risk of or susceptible to developing a disorder or condition.
The term "subject" is used throughout the specification to describe an animal, preferably a human, to whom treatment, including prophylactic treatment, with the compounds according to the present invention is provided. For treatment of those infections, conditions or disease states which are specific for a specific animal such as a human patient, the term patient refers to that specific animal. In most instances, the term patient refers to a human patient.
As used herein, the terms "treat," treating," "treatment," and the like refer to reducing or ameliorating a disorder and/or symptoms associated therewith. It will be appreciated that, although not precluded, treating a disorder or condition does not require that the disorder, condition or symptoms associated therewith be completely eliminated. The term "IC50" is the concentration of a compound that results in 50% inhibition of activity of a peptide, protein, enzyme or biological process.
"Myristoyl Binding Pocket" is a region of Bcr-abl at which a myristoyl moiety can bind when the BCR-AbI protein is in an appropriate conformation for myristoyl binding. Myristoyl binding pockets are described in, for example, Hantschel et al.,"A Myristoyl/Phosphotyrosine Switch Regulates c-Abl" Cell (2003), Vol. 112, 845-857 and Bhushan et al.," Structural Basis for the Autoinlaibition of c-Abl Tyrosine Kinase" Cell (2003), Vol. 1 12, 859-871.
The fusion protein Bcr- AbI is a result of a reciprocal translocation that fuses the AbI proto-oncogene with the Bcr gene. Bcr-abl is then capable of transforming B-cells through the increase of mitogenic activity. This increase results in a reduction of sensitivity to apoptosis, as well as altering the adhesion and homing of CML progenitor cells. The present invention provides compounds, compositions and methods for the treatment of kinase related disease, particularly PDGF-R, c-Kit and Bcr-abl kinase related diseases.
As used herein the term "substituent" or "substituted" means that a hydrogen radical on a compound or group (such as, for example, alkyl, alkenyl, alkynyl, alkylene, aryl, aralkyl, heteroaryl, heteroaralkyl, cycloalkyl, or heterocycloalkyl group) is substituted or optionally substituted with any desired group that do not substantially adversely affect the stability of the compound. In one embodiment, desired substituents are those which do not adversely affect the activity of a compound. The term "substituted" refers to one or more substituents (which may be the same or different), each replacing a hydrogen atom. Examples of substituents include, but are not limited to, halogen (F, Cl, Br, or I), hydroxyl, amino, alkylamino, arylamino, dialkylamino, diarylamino, alkylarylamino, cyano, nitro, mercapto, thio, imino, formyl, carbamido, carbamyl, carboxyl, thioureido, thiocyanato, sulfoamido, sulfonylalkyl, sulfonylaryl, alkyl, alkenyl, alkoxy, mercaptoalkoxy, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, wherein alkyl, alkenyl, alkyloxy, alkoxyalkyl, aryl, heteroaryl, cycloalkyl, are heterocycloalkyl are optionally substituted with alkyl, aryl, heteroaryl, halogen, hydroxyl, amino, mercapto, cyano, nitro, oxo (=0), thioxo (-S), imino (=NR), C(=N-NRk)Rk, or C(=N-ORk)Rk.
In other embodiments, substituents on any group (such as, for example, alkyl, alkenyl, alkynyl, aryl, aralkyl, heteroaryl, heteroaralkyl, cycloalkyl, and heterocycloalkyl) can be at any atom of that group, wherein any group that can be substituted (such as, for example, alkyl, alkenyl, alkynyl, aryl, aralkyl, heteroaryl, heteroaralkyl, cycloalkyl, heterocycloalkyl) can be optionally substituted with one or more substituents (which may be the same or different), each replacing a hydrogen atom. Examples of suitable substituents include, but not limited to alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aralkyl, heteroaralkyl, aryl, heteroaryl, halogen, haloalkyl, cyano, nitro, alkoxy, aryloxy, hydroxyl, hydroxylalkyl, oxo (i.e., carbonyl), carboxyl, formyl, alkylcarbonyl, alkylcarbonylalkyl, alkoxycarbonyl, alkylcarbonyloxy, aryloxycarbonyl, heteroaryloxy, heteroaryloxycarbonyl, thio, mercapto, mercaptoalkyl, arylsulfonyl, amino, aminoalkyl, dialkylamino, alkylcarbonylamino, alkylaminocarbonyl, or alkoxy carbonylamino; alkylamino, arylamino, diarylamino, alkylcarbonyl, or arylamino-substituted aryl; arylalkylamino, aralkylaminocarbonyl, amido, alkylaminosulfonyl, arylaminosulfonyl, dialkylaminosulfonyl, alkylsulfonylamino, arylsulfonylamino, imino, carbamido, carbamyl, thioureido, thiocyanato, sulfoamido, sulfonylalkyl, sulfonylaryl, or mercaptoalkoxy.
Additional suitable substituents for an alkyl, alkenyl, alkynyl, aryl, aralkyl, heteroaryl, heteroaralkyl, cycloalkyl, and heterocycloalkyl include, without limitation halogen, CN, NO2, OR15, SR15, S(O)2OR15, NR15R16, Ci-C2perfluoroalkyl, Ci-C2 perfluoroalkoxy, 1,2- methylenedioxy, (=0), (=S), (=NR15), C(O)OR15, C(O)NR15R16, OC(O)NR15R16, NR15C(O)NR15R16, C(NRI6)NR15R16, NR15C(NR16)NR15R16, S(O)2NR15R16, R17, C(O)H, C(O)R17, NR15C(O)R17, Si(R15)3, OSi(R15)3, Si(OH)2R15, B(OH)2, P(O)(OR15)2, S(O)R17, or S(O)2R17. Each R15 is independently hydrogen, Ci-C6 alkyl optionally substituted with cycloalkyl, aryl, heterocycloalkyl, or heteroaryl. Each R16 is independently hydrogen, C3-C6 cycloalkyl, aryl, heterocycloalkyl, heteroaryl, CpC4 alkyl or Ci-C4 alkyl substituted with C3- C6 cycloalkyl, aryl, heterocycloalkyl or heteroaryl. Each R17 is independently C3-C6 cycloalkyl, aryl, heterocycloalkyl, heteroaryl, Ci-C4 alkyl or Ci-C4 alkyl substituted with C3- C6 cycloalkyl, aryl, heterocycloalkyl or heteroaryl. Each C3-C6 cycloalkyl, aryl, heterocycloalkyl, heteroaryl and Ci-C4 alkyl in each R15, R16 and R17 can optionally be substituted with halogen, CN, Ci-C4 alkyl, OH, Ci-C4 alkoxy, COOH, C(O)OCi-C4 alkyl, NH2, Ci-C4 alkylamino, or C]-C4 dialkylamino.
In certain embodiments, a compound of the invention may be optionally substituted with (i) alkyl, haloalkyl, aryl, halogen, hydroxyl, alkoxy, hydroxyalkyl, amino, mono-alkyl amine, di-alkyl amine, cyano, CONH2, C02alkyl, SO2NH2, or (ii) heterocyclic or heteroaryl, selected from the following: morpholine,
Figure imgf000064_0001
In certain embodiments, an optionally substituted aryl or heteroaryl may be selected from the following: an optionally substituted phenyl, an optionally substituted naphthyl, an optionally substituted anthracenyl, an optionally substituted fluorenyl, an optionally substituted indenyl, an optionally substituted azulenyl, an optionally substituted pyridyl, an optionally substituted 1-oxo-pyridyl, an optionally substituted furanyl, an optionally substituted benzo[l,3]dioxolyl, an optionally substituted benzo[l,4]dioxinyl, an optionally substituted thienyl, an optionally substituted pyrrolyl, an optionally substituted oxazolyl, an optionally substituted imidazolyl, an optionally substituted thiazolyl, an optionally substituted isoxazolyl, an optionally substituted quinolinyl, an optionally substituted pyrazolyl, an optionally substituted isothiazolyl, an optionally substituted pyridazinyl, an optionally substituted pyrimidinyl, an optionally substituted pyrazinyl, an optionally substituted triazinyl, an optionally substituted triazolyl, an optionally substituted thiadiazolyl, an optionally substituted isoquinolinyl, an optionally substituted indazolyl, an optionally substituted benzoxazolyl, an optionally substituted benzofuryl, an optionally substituted indolizinyl, an optionally substituted imidazopyridyl, an optionally substituted tetrazolyl, an optionally substituted benzimidazolyl, an optionally substituted benzothiazolyl, an optionally substituted benzothiadiazolyl, an optionally substituted benzoxadiazolyl, an optionally substituted indolyl, an optionally substituted tetrahydroindolyl, an optionally substituted azaindolyl, an optionally substituted indazolyl, an optionally substituted imidazopyridyl, an optionally substituted quinazolinyl, an optionally substituted purinyl, an optionally substituted pyrrolo[2,3]pyrimidinyl, an optionally substituted pyrazolo[3,4] pyrimidinyl, or an optionally substituted benzo(b)thienyl.
In certain embodiments, an optionally substituted heterocycloalkyl may be selected from the following: an optionally substituted piperidinyl, an optionally substituted piperazinyl, an optionally substituted 2-oxopiperazinyl, an optionally substituted 2- oxopiperidinyl, an optionally substituted 2-oxopyrrolidinyl, an optionally substituted 4- piperidonyl, an optionally substituted tetrahydropyranyl, an optionally substituted oxazolidinyl, an optionally substituted 2-oxo-oxazolidinyl, an optionally substituted tetrahydrothiopyranyl, an optionally substituted tetrahydrothiopyranyl sulfone, an optionally substituted morpholinyl, an optionally substituted thiomorpholinyl, an optionally substituted thiomorpholinyl sulfoxide, an optionally substituted thiomorpholinyl sulfone, an optionally substituted 1,3-dioxolane, tetrahydrofuranyl, or an optionally substituted tetrahydrothienyl.
The recitation of a listing of chemical groups in any definition of a variable herein includes definitions of that variable as any single group or combination of listed groups.
The compounds of this invention include the compounds themselves, as well as their salts, solvate, clathrate, hydrate, polymorph, or prodrugs, if applicable.
As used herein, the term "pharmaceutically acceptable salt," is a salt formed from, for example, an acid and a basic group of a compound of any one of the formulae disclosed herein. Illustrative salts include, but are not limited, to sulfate, citrate, acetate, oxalate, chloride, bromide, iodide, nitrate, bisulfate, phosphate, acid phosphate, isonicotinate, lactate, salicylate, acid citrate, tartrate, oleate, tannate, pantothenate, bitartrate, ascorbate, succinate, maleate, besylate, gentisinate, fumarate, gluconate, glucaronate, saccharate, formate, benzoate, glutamate, methanesulfonate, ethanesulfonate, benzenesulfonate, p- toluenesulfonate, and pamoate {i.e., l,l'-methylene-bis-(2-hydroxy-3-naphthoate)) salts. The term "pharmaceutically acceptable salt" also refers to a salt prepared from a compound of any one of the formulae disclosed herein having an acidic functional group, such as a carboxylic acid functional group, and a pharmaceutically acceptable inorganic or organic base. Suitable bases include, but are not limited to, hydroxides of alkali metals such as sodium, potassium, and lithium; hydroxides of alkaline earth metal such as calcium and magnesium; hydroxides of other metals, such as aluminum and zinc; ammonia, and organic amines, such as unsubstituted or hydroxy-substituted mono-, di-, or trialkylamines; dicyclohexylamine; tributyl amine; pyridine; N-methyl,N-ethylamine; diethylamine; triethylamine; mono-, bis-, or tris-(2-hydroxy-lower alkyl amines), such as mono-, bis-, or tris-(2-hydroxyethyl)amine, 2- hydroxy-tert-butylamine, or tris-(hydroxymethyl)methylamine, N, N,-di-lower alkyl-N- (hydroxy lower alkyl)-amines, such as N,N-dimethyl-N-(2-hydroxyethyl)amine, or tri-(2- hydroxyethyl)amine; N-methyl-D-glucamine; and amino acids such as arginine, lysine, and the like. The term "pharmaceutically acceptable salt" also refers to a salt prepared from a compound of any one of the formulae disclosed herein having a basic functional group, such as an amino functional group, and a pharmaceutically acceptable inorganic or organic acid. Suitable acids include hydrogen sulfate, citric acid, acetic acid, oxalic acid, hydrochloric acid (HCl), hydrogen bromide (HBr), hydrogen iodide (HI), nitric acid, hydrogen bisulfide, phosphoric acid, lactic acid, salicylic acid, tartaric acid, bitartratic acid, ascorbic acid, succinic acid, maleic acid, besylic acid, fumaric acid, gluconic acid, glucaronic acid, formic acid, benzoic acid, glutamic acid, methanesulfonic acid, ethanesulfonic acid, benzenesulfonic acid, and/7-toluenesulfonic acid.
As used herein, the term "polymorph" means solid crystalline forms of a compound of the present invention or complex thereof. Different polymorphs of the same compound can exhibit different physical, chemical and/or spectroscopic properties. Different physical properties include, but are not limited to stability (e.g., to heat or light), compressibility and density (important in formulation and product manufacturing), and dissolution rates (which can affect bioavailability). Differences in stability can result from changes in chemical reactivity (e.g., differential oxidation, such that a dosage form discolors more rapidly when comprised of one polymorph than when comprised of another polymorph) or mechanical characteristics (e.g., tablets crumble on storage as a kinetically favored polymorph converts to thermodynamically more stable polymorph) or both (e.g., tablets of one polymorph are more susceptible to breakdown at high humidity). Different physical properties of polymorphs can affect their processing. For example, one polymorph might be more likely to form solvates or might be more difficult to filter or wash free of impurities than another due to, for example, the shape or size distribution of particles of it. As used herein, the term "hydrate" means a compound of the present invention or a salt thereof, which further includes a stoichiometric or non-stoichiometric amount of water bound by non-covalent intermolecular forces.
As used herein, the term "clathrate" means a compound of the present invention or a salt thereof in the form of a crystal lattice that contains spaces (e.g., channels) that have a guest molecule (e.g., a solvent or water) trapped within.
As used herein and unless otherwise indicated, the term "prodrug" means a derivative of a compound that can hydrolyze, oxidize, or otherwise react under biological conditions (in vitro or in vivo) to provide a compound of this invention. Prodrugs may only become active upon such reaction under biological conditions, or they may have activity in their unreacted forms. Examples of prodrugs contemplated in this invention include, but are not limited to, analogs or derivatives of compounds of any one of the formulae disclosed herein that comprise biohydrolyzable moieties such as biohydrolyzable amides, biohydrolyzable esters, biohydrolyzable carbamates, biohydrolyzable carbonates, biohydrolyzable ureides, and biohydrolyzable phosphate analogues. Other examples of prodrugs include derivatives of compounds of any one of the formulae disclosed herein that comprise -NO, -NO2, -ONO, or - ONO2 moieties. Prodrugs can typically be prepared using well-known methods, such as those described by 1 BURGER'S MEDICINAL CHEMISTRY AND DRUG DISCOVERY (1995) 172- 178, 949-982 (Manfred E. Wolff ed., 5th ed).
In addition, some of the compounds of this invention have one or more double bonds, or one or more asymmetric centers. Such compounds can occur as racemates, racemic mixtures, single enantiomers, individual diastereomers, diastereomeric mixtures, and cis- or trans- or E- or Z- double isomeric forms. All such isomeric forms of these compounds are expressly included in the present invention. The compounds of this invention may also be represented in multiple tautomeric forms, in such instances, the invention expressly includes all tautomeric forms of the compounds described herein (e.g., alkylation of a ring system may result in alkylation at multiple sites, the invention expressly includes all such reaction products). All such isomeric forms of such compounds are expressly included in the present invention. All crystal forms of the compounds described herein are expressly included in the present invention.
Examples The invention is further illustrated by the following examples which should in no way should be construed as being further limiting. The compounds of the invention were synthesized according to the examples provided herein and according to the reaction schemes provided supra.
Example 1: Preparation of 2-(4-(4-(trifiuoromethoxy)phenylamino)-lH-pyrazolo[3,4- d]pyrimidin-l-yl)ethanol and 2-(4-(4-(trifluoromethoxy)phenylamino)-2H-pyrazolo[3,4- d]pyrimidin-2-yl)ethanol
Figure imgf000068_0001
Step 1 : 4-Chloro-6-(4-trifluoromethoxy-phenylamino)-pyrimidine-5-carbaldehyde:
Figure imgf000068_0002
To a solution of 4,6-Dichloro-pyrimidine-5-carbaldehyde (3.06 g, 17.5 mmol) in THF (15 mL) was added 4-trifluoromethoxyaniline (3.09 g, 17.45 mmol) at 0 °C. The mixture was stirred for 2 h and ethyl acetate (100 mL) was added. The organic layer was separated from brine, dried (Na2SO4) and concentrated to yield crude title compound as yellow solids. The compound was then purified by silica gel column chromatography to give the title compound as yellow crystals. (2.9 g, 55%). MS m/z 318.2 (M + 1).
Step 2 : 2-(4-(4-(trifiuoromethoxy)phenylamino)- 1 H-pyrazolo[3 ,4-d]pyrimidin- 1 -yl)ethanol and 2-(4-(4-(trifluoromethoxy)phenylamino)-2H-pyrazolo[3,4-d]pyrimidin-2-yl)ethanol: To solution of 4-chloro-6-(4-trifluoromethoxy-phenylamino)-pyrimidine-5-carbaldehyde (50 g, 0.15 mmol) in sec-BuOH was added Na2CO3 (30 mg, 0.26 mmol) and hydrazine (20 mg, 0.18 mmol). The reaction was stirred at 60 0C temperature for 8 hours. The reaction mixture was partitioned in 100 ml water and 100 ml ethyl acetate, extracted with ethyl acetate three times. The organic phase was combined and washed with brine, dried over Na2SO4. The crude product was purified by silica gel flash chromatography by using hexane:ethyl acetate as eluent resulting 2-(4-(4-(trifluoromethoxy)phenylamino)-lH-pyrazolo[3,4-d]pyrimidin-l- yl)ethanol and 2-(4-(4-(trifluoromethoxy)phenylamino)-2H-pyrazolo[3,4-d]pyrimidin-2- yl)ethanol in the ratio of 4:1 as yellow solids.
2-(4-(4-(trifluoromethoxy)phenylainino)-lH-pyrazoIo[3,4-d]pyrimidin-l-yl)ethanol (20 mg): 1H NMR 400 MHz (DMSO-4$) δ 10.30 (s, IH), 8.45 {s, IH (6-Ar-H)}, 8.32 {s, IH (3- Ar-H), 7.98 (d, 2H, J= 9.6 Hz), 7.41 (d, 2H, J= 9.6 Hz), 4.40 (t, 2H, J= 3.0 Hz), 3.83 (/, 2H, J= 3.0 Hz); MS m/z 340.2 (M + 1). 2-(4-(4-(trifluoromethoxy)phenylamino)-2H-pyrazolo[3,4-d]pyrimidin-2-yl)ethanol (5 mg): 1H NMR 400 MHz (DMSO-J15) δ 10.75 (s, IH), 8.68 {s, IH (3-Ar-H)}, 8.57 {s, IH (6- Ar-H), 7.97 (d, 2H, J= 9.6 Hz), 7.45 {d, 2H, J= 9.6 Hz), 4.41 (t, 2H, J= 3.0 Hz), 3.85 (t, 2H, J= 3.0 Hz); MS m/z 340.2 (M + 1).
Example 2: N-(2-Morpholin-4-yl-ethyl)-3-[4-(4-trifluoromethoxy-phenylamino)- pyrrolo[2,3-d]pyrimidin-7-yl]-propionamide
Figure imgf000069_0001
Step 1 : 3-(4-Chloro-pyrrolo[2,3-d]pyrimidin-7-yl)-propionic acid ethyl ester:
Figure imgf000069_0002
To a solution of 4-Chloro-7H-pyrrolo[2,3-d]pyrimidine (1 g, 6.53 mmol) in DMF (30 mL), cooled at 0 0C, was added slowly NaH (1.8 g, 9.14 mmol). After stirring for 30 min at room temperature, the 4-(2-chloro-ethyl)-morpholine (1.28 g, 7.18 mmol) was added. The reaction mixture was stirring for 16 h at 40 0C. The mixture was brought to room temperature and then quenched with saturated NH4Cl solution and extracted with 150 ml of ethyl acetate. The organic extracts were washed with brine, dried over Na2SO4 and concentrated. The residue was purified by column chromatography (silica gel, eluting with hexanes) to afford the title compound (1.2 g, 72 %). 1H NMR 600 MHz (DMSO-^) δ 9.01 (s, IH), 6.26 (d, IH, J= 3.0 Hz), 5.91 (d, IH, J= 3.0 Hz), 4.24 (t, 2H, J= 5.4 Hz), 4.12 {t, 2H, J= 5.0 Hz), 2.73 (d, 2H, J = 5 Hz), 1.21 (d, 3H, J= 4 Hz); MS m/z 254.2 (M + 1).
Step 2: 3-[4-(4-Trifluoromethoxy-phenylamino)-pyrrolo[2,3-d]pyrimidin-7-yl]-propionic acid ethyl ester:
Figure imgf000070_0001
To a solution of 3-(4-Chloro-pyrrolo[2,3-d]pyrimidin-7-yl)-propionic acid ethyl ester (100 mg, 0.37 mmol) in sec-BuOH (3 mL) was added 4-trifluoromethoxyaniline (73 mg, 0.41 mmol). The mixture was heated at 120 0C and stirred for 2 h. The reaction mixture was concentrated and purified by prep-HPLC to afford the title compound as brown solid as TFA salt (1 14 mg, 60%). MS m/z 408.1 (M + 1).
Step 3: 3-[4-(4-Trifluoromethoxy-phenylamino)-pyrrolo[2,3-d]pyrimidin-7-yl]-propionic acid:
Figure imgf000070_0002
To a solution of 3-[4-(4-Trifluoromethoxy-phenylamino)-pyrrolo[2,3-d]pyrimidin-7-yl]- propionic acid ethyl ester (100 mg, 0.24 mmol) in 5 mL of THF:H2O (3:1)) was added CsCO3 (78 mg, 0.40 mmol). The mixture was heated at 80 °C and stirred for 2 h. The reaction mixture was concentrated and neutralized with acetic acid. The solid was filtered and dried to yield the tile compound as brown solid (67 mg, 77%). MS m/z 367.1 (M + 1).
Step 4: N-(2-Morpholin-4-yl-ethyl)-3-[4-(4-trifluoromethoxy-phenylamino)-pyrrolo[2,3- d]pyrimidin-7-yl] -propionamide :
3 - [4-(4-Trifluoromethoxy-pheny lamino)-pyrrolo [2,3 -d]pyrimidin-7-yl]-propionic acid (25 mg, 0.068 mmol) was mixed with DIEA (0.030 ml, 0.172 mmol) and HATU (28.5 mg, 0.071 mmol) in 2 ml DMF at room temperature. 2-Morpholin-4-yl-ethylamine (9.7 uL, 0.074 mmol) was added into the reaction mixture 0.5 hour later. After stirring at room temperature for 2 hours, the reaction mixture was concentrated and purified by Prep-HPLC to afford the title compound as TFA salt. MS m/z 479.2 (M + 1).
Example 3: Preparation of N-(2-hydroxyethyl)-4-(4-(trifluoromethoxy) phenylamino)thieno[2,3-d]pyrimidine-6-carboxamide
Figure imgf000071_0001
Step 1: Methyl 4-(4-(trifluoromethoxy)phenylamino)thieno[2,3-d]pyrimidine-6-carboxylate:
Figure imgf000071_0002
4-Chloro-6-(4-trifluoromethoxy-phenylamino)-pyrimidine-5-carbaldehyde (500 g, 1.5 mmol) in DMF (2 mL) was added to a suspension Of K2CO3 (620 mg, 4.5 mmol) in DMF (1 mL) at room temperature. After stirring for 30 min, methyl thioglucolate (210 mg , 1.9 mmol) was added slowly into the reaction mixture. The reaction mixture was then heated at 90 0C for 1 h. The reaction mixture was then cooled down at room temperature and poured onto ice cold water. The residue appeared was filtered, washed and dried to afford the title compound as white solid (40Og, 72 % Yield). MS m/z 370.2 (M + 1).
Step 2: 4-(4-(trifluoromethoxy)phenylamino)thieno[2,3-d]pyrimidine-6-carboxylic acid:
Figure imgf000072_0001
A mixture of methyl 4-(4-(trifluoromethoxy)phenylamino)thieno[2,3-d]pyrimidine-6- carboxylate (152 mg, 0.43 mmol), LiOH (25.5 mg, 1.07 mmol) was dissolved in THF (4 ml) and H2O (1 mL). The mixture was stirred at 60 °C for 3 h. Acidified with acetic acid resulted brown solid which was collected by filtration. It was then dried in vacuo and used for the next step without any further purification (115 mg, 75 % Yield). MS m/z 356.1 (M + 1)
Step 3 : N-(2-hydroxyethyl)-4-(4-(trifluoromethoxy)phenylamino)thieno[2,3-d]pyrimidine-6- carboxamide:
4-(4-(trifluoromethoxy)phenylamino)thieno[2,3-d]pyrimidine-6-carboxylic acid (30 mg, 0.08 mmol) was mixed with DIEA (30 uL, 0.172 mmol) and HATU (39 mg, 0.1 mmol) in 2 ml DMF at room temperature. 2-Aminoethanol (6.1 mg, 0.1 mmol) was added into the reaction mixture 0.5 hour later. After stirring at room temperature for 2 hours, the reaction mixture was concentrated and purified by prep-HPLC to afford the title compound as TFA salt (25 mg, 61% Yield). 1H NMR 400 MHz (DMSO-£/6) δ 10.10 (s, IH), 9.26 (s, IH), 8.82 (s, IH), 8.21 (d, 2H, J= 8.8 Hz), 7.85 (d, 2H, J= 8.8 Hz), 7.34 (s, IH), 3.02 (t, 2H, J= 6.4 Hz), 2.65 (t, 2H, J= 6.4 Hz). MS m/z 399.12 (M + 1). Example 4: 4-(4-(trifluoromethoxy)phenylamino)-N-(2-hydroxyethyl)quinazoline-7- carboxamide
Figure imgf000073_0001
Step 1 : Methyl 4-(4-(trifluoromethoxy)phenylamino)quinazoline-7-carboxylate:
Figure imgf000073_0002
To a solution of methyl 4-chloroquinazoline-7-carboxylate (610 mg, 2.75 mmol) in sec- BuOH (5 mL) was added 4-trifluoromethoxyaniline (535.5 mg, 3.02 mmol) at room temperature. The mixture was heated at 100 °C and stirred for 2 h. The solvent was evaporated and the compound was crystallized from CHCl3 and Et2O mixture (3:1). The white crystals were collected by filtration and washed with Et2O. (890 mg, 91 % Yield). MS m/z 364.10 (M + 1).
Step 2: 4-(4-(trifluoromethoxy)phenylamino)quinazoline-7-carboxylic acid:
Figure imgf000073_0003
A mixture of methyl 4-(4-(trifluoromethoxy)phenylamino)quinazoline-7-carboxylate (156 mg, 0.43 mmol), LiOH (25.5 mg, 1.07 mmol) was dissolved in THF (4 ml) and H2O (1 mL). The mixture was stirred at 60 0C for 3 h. Acidified with acetic acid resulted brown solid which was collected by filtration. It was then dried in vacuo and used for the next step without any further purification (120 mg, 80 % Yield). 1H NMR 400 MHz (DMSO-cfc) δ 12.25 (s, IH), 10.12 (5, IH), 8.68 (s, IH), 8.65 (d, IH, J= 1.6 Hz), 8.20-8.1 1 (m, 2H), 8.02 (d, 2 H, J= 8.8 Hz), 7.42 (d, 2H, J= 8.8 Hz. MS m/z 350.1 (M + 1).
Step 3 : 4-(4-(trifluoromethoxy)phenylamino)-N-(2-hydroxyethyl)quinazoline-7-carboxamide: 4-(4-(trifluoromethoxy)phenylamino)quinazoline-7-carboxylic acid (30 mg, 0.09 mmol) was mixed with DIEA (30 uL, 0.172 mmol) and HATU (39 mg, 0.1 mmol) in 2 ml DMF at room temperature. 2-Aminoethanol (6.1 mg, 0.1 mmol) was added into the reaction mixture 0.5 hour later. After stirring at room temperature for 2 hours, the reaction mixture was concentrated and purified by prep-HPLC to afford the title compound as TFA salt (29 mg, 82 % Yield). MS m/z 393.04 (M + 1).
Example 5: Preparation of 4-(6-(4-(trifluoromethoxy)phenylamino)- 5-methylpyrimidin-4-yl)-N-(2-morpholinoethyl)benzamide
Figure imgf000074_0001
Step 1 : 6-chloro-5-methyl-N-(4-(trifluoromethoxy)phenyl)pyrimidin-4-amine:
Figure imgf000074_0002
4,6-Dichloro-5-methylpyrimidine (534.6 mg, 3.3 mmol) and 4-trifluoromethoxy aniline (661 mg, 3.6 mmol) were dissolved in EtOH and DIEA (625 uL, 3.6 mmol) was added and the mixture was refluxed for 4 h. It was then cooled, concentrated and work-up with ethyl acetate. The organic layer was separated and dried (Na2SO4). Concentrated and purified by silica-gel column chromatography (Ethyl acetate/hexane). 800 mg (84% Yield), MS m/z 304.14 (M + 1).
Step 2: 4-(6-(4-(trifluoromethoxy)phenylamino)-5-methylpyrimidin-4-yl)benzoic acid:
Figure imgf000075_0001
Chloropyrimidine intermediate (521 mg, 1.72 mmol), 4-carboxyphenylboronic acid (287 mg, 1.72 mmol), Pd(PPh3 )4 (100 mg, 0.85 mmol) and sodium carbonate (730 mg, 6.9 mmol) were dissolved in acetonitrile: water (v/v 1 :1, 10 ml). The flask was stirred at 95 °C for 5 hr. The insoluble residues were removed by filtering the hot reaction solution. A solution of aq. HCl (6N) was added until ~pH <5. The solid was collected by filtration and rinsed with water and used without further purification (500 mg, 74 % Yield). MS m/z 390.20 (M + 1).
Step 3 : 4-(6-(4-(trifluoromethoxy)phenylamino)-5-methylpyrimidin-4-yl)-N-(2- morpholinoethyl) benzamide:
21 mg (0.55 mmol) of 2 ,2-aminoethanol (4.3 mg,0.066 mmol) and DIEA (37 uL, 0.21 mmol) were dissolved in DMF (1.5 ml) and stirred for 5 min. 26.5 mg (0.07 mmol) was added and the reaction mixture was continued to stir for another 6 at room temperature. The desired product was purified by mass triggered HPLC to give 22 mg (75 % yield) as TFA salt. MS m/z 502.23 (M + 1). Example 6: Preparation of 6-(lH-pyrrolo[2,3-b]pyridin-l-yl)-N-(4- (trifluoromethoxy)phenyl)pyrimidin-4-amine
Figure imgf000076_0001
Step 1 : 6-chloro-N-(4-(trifluoromethoxy)phenyl)pyrimidin-4-amine:
Figure imgf000076_0002
4,6-Dichloropyrimidine (250 mg, 3.3 mmol) and 4-trifluoromethoxy aniline (330.5 mg, 1.8 mmol) were dissolved in EtOH and DIEA (312.5 uL, 1.8 mmol) was added and the mixture was refluxed for 4 h. It was then cooled, concentrated and work-up with ethyl acetate. The organic layer was separated and dried (Na2SO4). Concentrated and purified by silica-gel column chromatography (Ethyl acetate/hexane). 400 mg (84% Yield). MS m/z 290.04 (M + 1).
Step 2 : 6-( 1 H-pyrrolo [2,3 -bjpyridin- 1 -yl)-N-(4-(trifluoromethoxy)phenyl)pyrirnidin-4-amine : To a solution of compound 7-azaindole (18 mg, 0.15 mmol) in sec-BuOH (5 mL) was added DIEA (0.20 mL) and the reaction mixture was stirred for 15 min at RT. 6-Chloro-N-(4- (trifluoromethoxy)phenyl)pyrimidin-4-amine (29 mg, 0.1 mmol) was then added and the temperature was raised to 90 0C and continued stirring for another 16 h. The solvent was removed and the title compound was purified by prep. HPLC (15 mg, 40 % Yield). MS m/z 372.10 (M + 1).
Example 7: Preparation of N-(4-(trifluoromethoxy)phenyl)-6-(l,3,5-trimethyl- lH-pyrazol-4-yl)pyrimidin-4-amine
Figure imgf000077_0001
6-Chloro-N-(4-(trifluoromethoxy)phenyl)pyrimidin-4-aniine intermediate (5.2 mg, 0.17 mmol), l,3,5-trimethyl-4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-lH-pyrazole (40 mg, 0.17 mmol), Pd(PPh3)., (10 mg, 0.085 mmol) and sodium carbonate (73 mg, 0.69 mmol) were dissolved in acetonitrile: water (v/v 1 :1, 10 ml). The flask was stirred at 95 °C for 5 hr. The solvent was removed and directly subjected to prep-HPLC to afford 65 mg (80% Yield) of the title compound as TFA salt. 1H NMR 400 MHz (OMSO-d6) δ 10.32 (s, IH), 8.75 (s, IH), 8.39 (s, 2H), 7.84 (d, 2H, J= 9.2 Hz), 7.45 (d, 2H, J= 9.2 Hz), 3.84 (s, 3H), 2.89 (s, 3H), 2.24 (s, 3H). MS m/z 364.11 (M + 1).
Example 8: Preparation of 2-(3-nitrophenyl)-N-(4-(trifluoromethoxy)phenyl) imidazo [ 1 ,2-c] py rimidin-7-amine
Figure imgf000077_0002
Step 1 : 7-chloro-2-(3-nitrophenyl)imidazo[l,2-c]pyrimidine
Figure imgf000077_0003
6-Chloropyrimidin-4-amine (50 mg, 0.39 mmol) and 2-bromo-l-(3-nitrophenyl)ethanone (94 mg, 0.39 mmol) were dissolved in 2 mL of DMSO and the mixture was stirred at room temperature for 3 days. The reaction mixture was poured into 5 mL of water and it was then extracted with ethyl acetate. The filtrate was dried (Na2SO4) and evaporated. The compound was purified by silica gel column chromatography by using hexane/ethyl acetate (3:1) as eluent (60 mg, 56 % Yield). MS m/z 275.00 (M + 1).
Step 2: 2-(3-Nitrophenyl)-N-(4-(trifluoromethoxy)phenyl)imidazo[l ,2-c]pyrimidin-7-amine
7-chloro-2-(3-nitrophenyl)imidazo[l,2-c]pyrimidine (30 mg, 0.1 1 mmol) and (58 mg, 0.32 mmol) were heated at 120 °C for 1 h. The residue was dissolved in DMSO and directly subjected to prep-HPLC to yield 8 mg (13 % Yield) of the title compound as TFA salt. 1H NMR 400 MHz (DMSO-^5) δ 10.42 (s, IH), 8.88 (s, IH), 8.67 (s, IH), 8.01-7.99 (m, 3H), 7.92 (d, 2H, J= 9.2 Hz), 7.53 (d, 2H, J= 9.2 Hz), 7.45 (s, IH), 7.22 (s, IH). MS m/z 416.12 (M + 1).
Example 9: Biological Data Cell culture
Interleukin-3 (IL-3) dependent murine pro-B cell line, Ba/F3, and murine myeloid precursor cell line, 32D, were maintained in RPMI- 1640 medium supplemented with L-Glutamine, 10% FBS and 10 U/ml recombinant murine IL-3 (Roche). Mo7e human megakarioblastic cell line was grown in RPMI- 1640 medium supplemented with L-Glutamine, 20% FBS and 5 ng/ml recombinant human granulocyte macrophage cell stimulating factor (GM-CSF) (R&D Systems) or 200 ng/ml stem cell factor (SCF) (Biosource).
Wild type Bcr-abl expressing 32D (32D.p210) and Ba/F3 (Ba/F3.p210) cells, and mutant Bcr-abl expressing cell lines Ba/F3.p210G250E, Ba/F3.p210E255V, Ba/F3.p210T3151, Ba/F3.p210F317L and Ba/F3.p210M351T, as well as Ba/F3 cells transformed with Flt-3-ITD kinase. Additional assays included Ba/F3 cells expressing the fusion protein kinases TEL- PDGFRβ, TEL-JAKl, NPM-ALK, TEL-cKit, Tel-Bmx, Tel-FGR3, Tel-Lck and Ba/F3.TPR-met cells. All the above cell lines were maintained in RPMI- 1640 medium with L-Glutamine, 10% FBS and 1 mg/ml geneticin (Gibco). Human leukemic cell lines K562 (p210 Bcr-abl expressing chronic myelogenous leukemia), HL-60 (acute promyelocyte leukemia), SUP/B15 (pi 90 Bcr-abl expressing acute lymphoblastic leukemia) and Jurkat (acute T cell leukemia) were purchased from the American Type Culture Collection (ATCC) and cultured following ATCC recommendations.
Proliferation assays
Cells (0.3-0.6xl06/ml) were plated in duplicate or triplicate in 96-well plates containing increasing drug concentrations (0.005-10 μM). After incubation at 37°C in 5% CO2 for 48 h, the effect of the compounds on cell viability was determined by MTT (Promega) colorimetric dye reduction method. Inhibition of cell proliferation was calculated as a percentage of growth of DMSO treated cells, and IC50 values were determined with Microsoft Excel XLfit3.
Simultaneous treatment of Ba/F3.p210 cells with imatinib and GNF-2 was carried out to evaluate the synergistic or additive effect of the drug combinations on cellular proliferation. Cells treated with increasing concentrations of each compound alone or in combination were incubated at 37°C in 5% CO2 for 48 h, and the cell viability determined by the MTT assay. The in vitro additive, synergistic or antagonistic effect was determined by calculating the combination index (CI) from two independent experiments using CalcuSyn Software.
Cell cycle analysis
Cells grown for 24 or 48 hours in medium containing different concentrations of compound were collected by centrifugation (10 min, 4°C), washed twice with PBS and fixed in 70% ethanol at -20°C. After fixation, ethanol was removed by centrifugation (10 min, 4°C), and the cells were resuspended in PBS containing ImM EDTA and 100 μg/ml RNAse A (Sigma) and incubated 30 min at 37°C. Cells were stained with propidium iodide (10 μg/ml) and DNA content was determined by flow cytometry (FACScan, Becton Dickinson).
Phosphotyrosine analysis
The total cellular tyrosine phosphorylation levels of Bcr-abl were first determined by using capture ELISA. Bcr-abl expressing cells treated for 90 min with various concentrations of test compounds were homogenized in lysis buffer (50 mM Tris.HCl pH7.4, 150 mM NaCl, 5 mM EDTA, 1 mM EGTA, 1% NP-40, 2 mM Na3VO4 and protease inhibitor cocktail (Roche)) and the lysates plated on 96 well plates containing adsorbed anti-Abl SH3 domain polyclonal antibody (Upstate Biotechnology). The plates were incubated for 4 h at 40C and then washed with PBS/0.05% Tween 20 buffer. To detect phosphotyrosine residues, alkaline phosphatase-conjugated monoclonal anti-phosphotyrosine antibody (PY20, Zymed Laboratories) was added to each well, and plates were incubated overnight at 40C. The wells were then washed with PBS/0.05% Tween 20 buffer, and 100 μl/well of CDP-Star® Substrate with Emerald-II™ enhancer substrate (Applied Biosystems) was added. After 45 min, light emission was quantified with a GeminiXS microplate reader (Molecular Devices). Bcr-abl phosphotyrosine content was calculated as a percentage of phosphotyrosine of non treated cells, and IC50 values were determined with Microsoft Excel XLfit3.
The state of tyrosine phosphorylation of cellular Bcr-abl and it substrate Stat5 after drug treatment was determined by western blot with phospho-specific antibodies. Ba/F3.p210 and Ba/F3.p210E255V cells were incubated in the presence of various concentrations of GNF-2 and, after 1.5 h incubation, lysed in lysis buffer (20 mM Tris-HCl (pH 7.5), 150 mM NaCl, 1 mM Na2EDTA, 1 mM EGTA, 1% Triton, 2.5 mM sodium pyrophosphate, 1 mM beta- glycerophosphate, 1 mM Na3VO4, 1 μg/ml leupeptin, 1 mM PMSF). Equal amount of lysates (50 μg) were subjected to SDS-PAGE followed by immunoblotting with phospho-specific antibodies recognizing Bcr-abl phosphorylation sites: anti-phospho-c-Abl (Tyr 245), and anti- phospho-Stat5 (Tyr 694) (Cell signaling) antibodies or antibodies against Bcr-abl (Ab-3, Oncogene Science) and Stat5 (C- 17, Santa Cruz Biotechnology). Proteins were detected by enhanced chemioluminiscence (ECL-plus, Amersham), following manufacturer's guidelines.
Protein expression and purification
Recombinant c-abl (residues 46-531) was expressed as a His6 tag fusion protein in Sf9 insect cells using the Bac-to-Bac baculovirus expression system (Invitrogen) as previously described. Briefly, infected cells were grown for 48 h, harvested by centrifugation, suspended in lysis buffer (5OmM Tris pH 8.0, 30OmM NaCl, 5% glycerol, 1OmM 2- mercaptoethanol and protease inhibitor cocktail (Roche) and lysed by sonication. After centrifugation, the cell extract was loaded on a Ni-NTA affinity column and AbI was eluted with 25OmM imidazole. Fractions containing abl were pooled and the concentration of imidazole was reduced to 20 mM by dialysis. The protein was treated with CiP alkaline phosphatase (Invitrogen) and purified by a second Ni affinity chromatography. The His6 tag was removed by treatment with TEV protease (Invitrogen) and the protein recovered from a Ni-NTA column with a purity of -90% by SDS-PAGE and coomassie blue staining.
Kinase assays
In vitro kinase assays were carried out by using recombinant murine c-abl containing SH3, SH2 and kinase domains (residues 46-531) and full length immunoprecipitated Bcr-abl. Recombinant abl was expressed in Sf9 insect cells and purified as described above. Bcr-abl immune complexes were obtained with Ab-3 anti-abl monoclonal antibody (Oncogene Science) from Ba/F3.p210 lysates as previously described.
1 μg of recombinant abl or immunoprecipitated Bcr-abl (from 3x106 cells) were incubated with various concentrations of test compound (0.1, 1 and 10 μM) in kinase buffer (50 mM Tris-HCl pH 7.5, 10 mM MgCl2, 100 mM EDTA, 1 mM DTT, 0.015% Brij 35), 100 μM ATP and 1 μCi [γ-32P]-ATP for 30 min at 30°C (Calbiochem buffer and protocol). The reaction was stopped by addition of Laemmli buffer and the proteins were resolved by SDS- PAGE in a 4-20% gel. The phosphoproteins were visualized by autoradiography and the autophosphorylation quantitated using a phosphoimager (STORM, Molecular Devices). The results are provided in the following tables.
Table 10
Figure imgf000081_0001
Figure imgf000082_0001
Figure imgf000083_0001
Figure imgf000084_0001
Figure imgf000085_0001
Figure imgf000086_0001
Figure imgf000087_0001
Figure imgf000088_0001
Figure imgf000089_0001
Figure imgf000090_0001
Figure imgf000091_0002
Table 11.
Figure imgf000091_0001
Figure imgf000092_0001
Table 12.
Figure imgf000092_0002
Figure imgf000093_0001
Figure imgf000094_0001
Figure imgf000095_0001
Figure imgf000096_0001
Figure imgf000097_0002
Table 13.
Figure imgf000097_0001
Figure imgf000098_0001
Figure imgf000099_0001
Figure imgf000100_0001
Figure imgf000101_0002
Table 14.
Figure imgf000101_0001
Figure imgf000102_0001
Figure imgf000103_0001
Figure imgf000104_0001
Figure imgf000105_0001
Figure imgf000106_0002
Table 15.
Figure imgf000106_0001
Table 16.
Figure imgf000106_0003
Figure imgf000107_0001
Figure imgf000108_0001
Figure imgf000109_0001
Figure imgf000110_0001
Figure imgf000111_0001
Figure imgf000112_0001
Figure imgf000113_0001
Figure imgf000114_0001
Figure imgf000115_0001
Figure imgf000116_0001
Figure imgf000117_0001
Figure imgf000118_0001
Figure imgf000119_0001
Figure imgf000120_0001
Figure imgf000121_0001
Table 17.
Figure imgf000121_0002
Figure imgf000122_0001
Incorporation by Reference
All references cited herein, whether in print, electronic, computer readable storage media or other form, are expressly incorporated by reference in their entirety, including but not limited to, abstracts, articles, journals, publications, texts, treatises, technical data sheets, internet web sites, databases, patents, patent applications, and patent publications.
Equivalents
Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents of the specific embodiments of the invention described herein. Such equivalents are intended to be encompassed by the following claims.

Claims

What is claimed is:
1. A compound of formula I, or pharmaceutically acceptable salt, solvate or hydrate thereof:
Figure imgf000123_0001
wherein each of Xi, X2, or X3 is independently NRi, CR1, C(O), O, or S; each Ri is independently selected from H, an optionally substituted Ci-6alkyl, an optionally substituted C3-i2cycloalkyl, an optionally substituted C3-i2heterocycloalkyl, an optionally substituted C3-i2aryl, an optionally substituted C3-i2heteroaryl, an optionally substituted Ci-6aralkyl, an optionally substituted Ci-6heteroaralkyl, an optionally substituted Ci-6haloalkyl, an optionally substituted (CH2)nC3-i2heterocycloalkyl, an optionally substituted (CH2)nC3-i2aryl, an optionally substituted (CH2)nC3-i2heteroaryl, an optionally substituted (CH2)nC3.12cycloalkyl, C,-6haloalkyl, Ci-6aminoalkyl, hal, (CH2)nC(O)Rx, (CH2)nC(O)ORx, (CH2)nC(O)NRxRx, -(CH2)nC(O)NRxRxS(O)2Rx; -C(O)RX, -C(S)RX, -C(NR)R", -SRX, -S(O)RX, -S(O)2RX, -ORX, -NRXRX, nitro, cyano, or absent; each Rx, for each occurrence, is independently H, an optionally substituted Ci-6alkyl, an optionally substituted C3-i2cycloalkyl, an optionally substituted C3-i2heterocycloalkyl, an optionally substituted C3-i2aryl, an optionally substituted C3-i2heteroaryl, Ci^haloalkyl; NHNHR2, or C(NH)NH2;
R2 is H or Ci-6 alkyl;
Y is H, OH, NH2 or CH3; m is 1 or 2; and each n is independently 0, 1 or 2.
2. The compound of claim 1 , of formula II: wherein
Ri is selected from H, an optionally substituted Ci-6alkyl, an optionally substituted C3- i2cycloalkyl, an optionally substituted C3.i2heterocycloalkyl, an optionally substituted C3- i2aryl, an optionally substituted C3-i2heteroaryl, an optionally substituted Ci-6aralkyl, an optionally substituted Ci-6heteroaralkyl, an optionally substituted Ci-6haloalkyl, an optionally substituted (CH2)nC3-i2heterocycloalkyl, an optionally substituted (CH2)nC3-i2aryl, an optionally substituted (CH2)nC3-i2heteroaryl, an optionally substituted (CH2)nC3-i2cycloalkyl, (CH2)nC(0)Rx, (CH2)nC(O)ORx, (CH2)nC(O)NRxRx, -C(O)RX, -C(S)R", -C(NR)R", halo, C1- βhaloalkyl, -S(O)R", or -S(O)2R";
Rx, for each occurrence, is independently, H, an optionally substituted Ci-6alkyl, an optionally substituted C3-i2cycloalkyl, an optionally substituted C3-i2heterocycloalkyl, an optionally substituted C3-i2aryl, an optionally substituted C3-i2heteroaryl, or Ci-6haloalkyl; and each n is independently 0, 1 or 2.
3. The compound of claim 2, selected from the following: 2-(4-(4-(trifluoromethoxy)phenylamino)-lH-pyrazolo[3,4-d]pyrimidin-l-yl)ethanol; (lH-Pyrazolo[3,4-d]pyrimidin-4-yl)-(4-trifluoromethoxy-phenyl)-amine; ( 1 -Ethyl- 1 H-pyrazolo[3 ,4-d]pyrimidin-4-yl)-(4-trifluoromethoxy-phenyl)-amine; (l-Ethanesulfonyl-lH-pyrazolo[3,4-d]pyrimidin-4-yl)-(4-trifluoromethoxy-phenyl)-amine; ( 1 -Cyclohexyl- 1 H-pyrazolo [3 ,4-d]pyrimidin-4-yl)-(4-trifluoromethoxy-phenyl)-amine; 2-[4-(4-Trifluoromethoxy-phenylamino)-pyrazolo[3,4-d]pyrimidin-l-yl]-ethanol; 2-[4-(4-Trifluoromethoxy-phenylamino)-pyrazolo[3,4-d]pyrimidin-l-yl]-acetamide; 3-[4-(4-Trifluoromethoxy-phenylamino)-pyrazolo[3,4-d]pyrimidin-l-yl]-propionamide; [ 1 -(Tetrahydro-furan-3 -y lmethyl)- 1 H-pyrazolo [3 ,4-d]pyrimidin-4-yl]-(4-trifluoromethoxy- phenyl)-amine; 3 -Dimethylamino-2- [4-(4-trifluoromethoxy-phenylamino)-pyrazolo [3 ,4-d]pyrimidin- 1 -yl] - propionitrile;
(l-Butyl-lH-pyrazolo[3,4-d]pyrimidin-4-yl)-(4-trifluoromethoxy-phenyl)-amine;
(l-tert-Butyl-lH-pyrazolo[3,4-d]pyrimidin-4-yl)-(4-trifluoromethoxy-phenyl)-amine;
(l-tert-Butyl-lH-pyrazolo[3,4-d]pyrimidin-4-yl)-(4-trifluoromethoxy-phenyl)-amine;
[l-(2,2,2-Trifluoro-ethyl)-lH-pyrazolo[3,4-d]pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)- amine;
( 1 -Benzyl- 1 H-pyrazolo [3 ,4-d]pyrimidin-4-y l)-(4-trifluoromethoxy-phenyl)-amine;
( 1 -Phenethyl- 1 H-pyrazolo[3 ,4-d]pyrimidin-4-yl)-(4-trifluoromethoxy-phenyl)-amine;
[ 1 -(3 -Fluoro-4-trifluoromethyl-benzyl)- 1 H-pyrazolo [3 ,4-d]pyrimidin-4-yl]-(4- trifluoromethoxy-phenyl)-amine;
[l-(3-Bromo-benzyl)-lH-pyrazolo[3,4-d]pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)-amine;
3-[4-(4-Trifluoromethoxy-phenylamino)-pyrazolo[3,4-d]pyrimidin-l-ylmethyl]-phenol
( 1 -Pyridin-3 -ylmethyl- 1 H-pyrazolo [3 ,4-d]pyrimidin-4-yl)-(4-trifluoromethoxy-phenyl)- amine;
(l-Pyridin-2-ylmethyl-lH-pyrazolo[3,4-d]pyrimidin-4-yl)-(4-trifluoromethoxy-phenyl)- amine;
( 1 -Pyridin-4-ylmethyl- 1 H-pyrazolo[3 ,4-d]pyrimidin-4-yl)-(4-trifluoromethoxy-phenyl)- amine;
2- { 2- [4-(4-Trifluoromethoxy-phenylamino)-pyrazolo [3 ,4-d]pyrimidin- 1 -yl]-ethyl } -isoindole-
1,3-dione;
[l-(2-Amino-ethyl)-lH-pyrazolo[3,4-d]pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)-amine;
(4-Trifluoromethoxy-phenyl)- [ 1 -(4-trifluoro methyl-pyridin-2-yl)- 1 H-pyrazolo [3 ,4- d]pyrimidin-4-yl]-amine;
[l-(2,3-Dimethyl-phenyl)-lH-pyrazolo[3,4-d]pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)- amine;
[l-(2-Fluoro-phenyl)-lH-pyrazolo[3,4-d]pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)-amine;
[l-(2,4-Dichloro-phenyl)-lH-pyrazolo[3,4-d]pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)- amine;
[l-(2-Ethyl-phenyl)-lH-pyrazolo[3,4-d]pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)-amine;
(l-Pyridin-2-yl-lH-pyrazolo[3,4-d]pyrimidin-4-yl)-(4-trifluoromethoxy-phenyl)-amine; 3 - [4-(4-Trifluoromethoxy-pheny lamino)-pyrazolo [3 ,4-d]pyrimidin- 1 -yl] - benzenesulfonamide;
4. The compound of claim 1, of formula III:
Figure imgf000126_0001
wherein
Ri is selected from H, an optionally substituted Ci-6alkyl, an optionally substituted C3- i2cycloalkyl, an optionally substituted C3-i2heterocycloalkyl, an optionally substituted C3- i2aryl, an optionally substituted C3-i2heteroaryl, an optionally substituted Ci-6 aralkyl, an optionally substituted Ci-6heteroaralkyl, an optionally substituted Ci-6haloalkyl, an optionally substituted (CH2)nC3-i2heterocycloalkyl, an optionally substituted (CH2)nC3-i2aryl, an optionally substituted (CH2)nC3-i2heteroaryl, an optionally substituted (CH2)nC3-i2cycloalkyl, (CH2)nC(O)Rx, (CH2)nC(O)ORx, (CH2)nC(O)NRxRx, -C(O)RX, -C(S)RX, -C(NR)RX, halo, Ci. ehaloalkyl, -S(O)R", or -S(O)2R";
R", for each occurrence, is independently, H, an optionally substituted Ci-6alkyl, an optionally substituted C3-i2cycloalkyl, an optionally substituted C3-i2heterocycloalkyl, an optionally substituted C3-i2aryl, an optionally substituted C3-i2heteroaryl, or Ci-6haloalkyl; and each n is independently 0, 1 or 2.
5. The compound of claim 4, selected from the following: 2-(4-(4-(trifluoromethoxy)phenylamino)-2H-pyrazolo[3,4-d]pyrimidin-2-yl)ethanol; (2-Ethyl-2H-pyrazolo[3,4-d]pyrimidin-4-yl)-(4-trifluoromethoxy-phenyl)-amine; (2-Cyclohexyl-2H-pyrazolo[3,4-d]pyrimidin-4-yl)-(4-trifluoromethoxy-phenyl)-amine; 2- [4-(4-Trifluoromethoxy-pheny lamino)-pyrazolo [3 ,4-d]pyrimidin-2-yl]-ethanol ; 2-[4-(4-Trifluoromethoxy-phenylamino)-pyrazolo[3,4-d]pyrimidin-2-yl]-acetamide; [2-(Tetrahydro-furan-3-ylmethyl)-2H-pyrazolo[3,4-d]pyrimidin-4-yl]-(4-trifluoromethoxy- phenyl)-amine; (2-Butyl-2H-pyrazolo[3,4-d]pyrimidin-4-yl)-(4-trifluoromethoxy-phenyl)-amine; [2-(2,2,2-Trifluoro-ethyl)-2H-pyrazolo[3,4-d]pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)- amine;
(2-Benzyl-2H-pyrazolo[3,4-d]pyrimidin-4-yl)-(4-trifluoromethoxy-phenyl)-amine;
(2-Phenethyl-2H-pyrazolo[3,4-d]pyrimidin-4-yl)-(4-trifluoromethoxy-phenyl)-amine;
[2-(3-Fluoro-4-trifluoromethyl-benzyl)-2H-pyrazolo[3,4-d]pyrimidin-4-yl]-(4- trifluoromethoxy-phenyl)-amine;
[2-(3-Bromo-benzyl)-2H-pyrazolo[3,4-d]pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)-amine;
3-[4-(4-Trifluoromethoxy-phenylamino)-pyrazolo[3,4-d]pyrimidin-2-ylmethyl]-phenol;
(2-Pyridin-3-ylmethyl-2H-pyrazolo[3,4-d]pyrimidin-4-yl)-(4-trifluoromethoxy-phenyl)- amine;
(4-Trifluoromethoxy-phenyl)-[2-(4-trifluoromethyl-pyridin-2-yl)-2H-pyrazolo[3,4- d]pyrimidin-4-yl]-amine;
[2-(2,3-Dimethyl-phenyl)-2H-pyrazolo[3,4-d]pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)- amine;
[2-(2-Fluoro-phenyl)-2H-pyrazolo[3,4-d]pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)-amine;
[2-(2,4-Dichloro-phenyl)-2H-pyrazolo[3,4-d]pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)- amine;
[2-(2-Ethyl-phenyl)-2H-pyrazolo[3,4-d]pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)-amine;
(2-Pyridin-2-yl-2H-pyrazolo[3,4-d]pyrimidin-4-yl)-(4-trifluoromethoxy-phenyl)-amine.
6. The compound of claim 1 , of formula IV:
Figure imgf000127_0001
wherein each Ri is indepdently selected from H, an optionally substituted Ci^alkyl, an optionally substituted C3.i2cycloalkyl, an optionally substituted C3-i2heterocycloalkyl, an optionally substituted C3-i2aryl, an optionally substituted C3-i2heteroaryl, an optionally substituted Ci-6aralkyl, an optionally substituted C|-6heteroaralkyl, an optionally substituted Ci-6haloalkyl, an optionally substituted (CH2)nC3-i2heterocycloalkyl, an optionally substituted (CH2)nC3.i2aryl, an optionally substituted (CH2)nC3.|2heteroaryl, an optionally substituted (CH2)nC3-12cycloalkyl, (CH2)nC(O)Rx, (CH2)nC(O)ORx, (CH2)nC(O)NRxRx, -C(O)RX, -C(S)R", -C(NR)R", halo, C,-6haloalkyl, -S(O)R", -S(O)2R", or absent;
R", for each occurrence, is independently, H, an optionally substituted C^aUcyl, an optionally substituted C3.i2cycloalkyl, an optionally substituted Cs-πheterocycloalkyl, an optionally substituted C3.i2aryl, an optionally substituted C3-i2heteroaryl, or Ci-6haloalkyl; and each n is independently 0, 1 or 2.
7. The compound of claim 6, selected from the following: (9-Methyl-9H-purin-6-yl)-(4-trifluoromethoxy-phenyl)-amine; (7-Methyl-7H-purin-6-yl)-(4-trifluoromethoxy-phenyl)-amine;
2-Hydroxymethyl-5-[6-(4-trifluoromethoxy-phenylamino)-purin-9-yl]-tetrahydro-furan-3,4- diol; [9-(2-Morpholin-4-yl-ethyl)-9H-purin-6-yl]-(4-trifluoromethoxy-phenyl)-amine.
8. The compound of claim 1 , of formula V:
Figure imgf000128_0001
wherein
Ri is independently selected from H, an optionally substituted Ci^alkyl, an optionally substituted C3-i2cycloalkyl, an optionally substituted C3-i2heterocycloalkyl, an optionally substituted C3- aTyI, an optionally substituted C3.i2heteroaryl, an optionally substituted (CH2)nC3.]2heterocycloalkyl, -C(O)R", -OR", -SR", -NR"R", nitro, cyano, C,-6haloalkyl, Ci- 6aminoalkyl, (CH2)nC(O)R", (CH2)nC(O)ORx, (CH2)nC(O)NR"R", or -(CH2)nC(O)NRxRxS(O)2Rx;
Rx, for each occurrence, is independently H, an optionally substituted Ci^alkyl, an optionally substituted C3.i2cycloalkyl, an optionally substituted C3.i2heterocycloalkyl, an optionally substituted C3-i2aryl, an optionally substituted C3.i2heteroaryl, NHNHR2, or C(NH)NH2;
Ri' is selected from H, an optionally substituted Ci-6alkyl, an optionally substituted C3-i2cycloalkyl, an optionally substituted C3.i2heterocycloalkyl, an optionally substituted C3- i2aryl, an optionally substituted C3-i2heteroaryl, an optionally substituted Ci-όaralkyl, an optionally substituted Ci-όheteroaralkyl, an optionally substituted Ci-6haloalkyl, an optionally substituted (CH2)nC3-i2heterocycloalkyl, an optionally substituted (CH2)nC3.i2aryl, an optionally substituted (CH2)nC3.i2heteroaryl, an optionally substituted (CH2)nC3-i2cycloalkyl, (CH2)nC(O)Ry, (CH2)nC(O)ORy, (CH2)nC(O)NRyRy, -C(O)Ry, -C(S)Ry, -C(NR)Ry, halo, C1- 6haloalkyl, -S(O)Ry, or -S(O)2Ry;
Ry, for each occurrence, is independently, H, an optionally substituted Ci-6alkyl, an optionally substituted C3.i2cycloalkyl, an optionally substituted C3-i2heterocycloalkyl, an optionally substituted C3-i2aryl, an optionally substituted C3.i2heteroaryl, or C1-6haloalkyl; each n is independently 0, 1 or 2; and
R2 is H or C-4 alkyl.
9. The compound of claim 8, wherein Ri is independently selected from H, an optionally substituted Ci^alkyl, an optionally substituted C3.i2cycloalkyl, an optionally substituted C3-i2heterocycloalkyl, an optionally substituted C3.i2aryl, an optionally substituted C3-i2heteroaryl, an optionally substituted (CH2)nC3-i2heterocycloalkyl, -C(O)R\ -ORX, -SRX, -NRXRX, nitro, cyano, C1-6haloalkyl, Ci-6aminoalkyl, (CH2)nC(O)Rx, (CH2)nC(O)ORx, (CH2)nC(O)NRxRx, or -(CH2)nC(O)NRxRxS(O)2Rx;
Rx, for each occurrence, is independently H, an optionally substituted Ci-6alkyl, an optionally substituted C3-i2cycloalkyl, an optionally substituted C3-i2heterocycloalkyl, an optionally substituted C3-i2aryl, an optionally substituted C3.i2heteroaryl, NHNHR2, or C(NH)NH2; and
R2 is H or Ci-4 alkyl.
10. The compound of claim 8, wherein Rj' is selected from H, an optionally substituted an optionally substituted C3-i2cycloalkyl, an optionally substituted C3. i2heterocycloalkyl, an optionally substituted C3-i2aryl, an optionally substituted C3. i2heteroaryl, an optionally substituted Ci-6aralkyl, an optionally substituted C|-6heteroaralkyl, an optionally substituted Ci-6haloalkyl, an optionally substituted (CH2)nC3-i2heterocycloalkyl, an optionally substituted (CH2)nC3.i2aryl, an optionally substituted (CH2)nC3.i2heteroaryl, an optionally substituted (CH2)nC3-i2cycloalkyl, (CH2)nC(O)Ry, (CH2)nC(O)ORy, (CH2)nC(O)NRyRy, -C(O)Ry, -C(S)Ry, -C(NR)Ry, halo, C1-6haloalkyl, -S(O)Ry, or -S(O)2Ry;
Ry, for each occurrence, is independently, H, an optionally substituted Ci-6alkyl, an optionally substituted C3-i2cycloalkyl, an optionally substituted C3-i2heterocycloalkyl, an optionally substituted C3-i2aryl, an optionally substituted C3-12heteroaryl, or
Figure imgf000130_0001
11. The compound of claim 8, selected from the following: N-(2-Morpholin-4-yl-ethyl)-3-[4-(4-trifluoromethoxy-phenylamino)-pyrrolo[2,3- d]pyrimidin-7-yl]-propionamide;
2-Hydroxymethyl-5-[4-(4-trifluoromethoxy-phenylamino)-pyrrolo[2,3-d]pyrimidin-7-yl]- tetrahydro-furan-3,4-diol;
(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)-(4-trifluoromethoxy-phenyl)-amine; (7-Methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)-(4-trifluoromethoxy-phenyl)-amine; 2-[4-(4-Trifluoromethoxy-phenylamino)-pyrrolo[2,3-d]pyrimidin-7-yl]-ethanol; [7-(2-Morpholin-4-yl-ethyl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)- amine;
[7-(4-Methoxy-benzyl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)- amine;
3-[4-(4-Trifluoromethoxy-phenylamino)-pyrrolo[2,3-d]pyrimidin-7-yl]-propionamide; N-Ethyl-3-[4-(4-trifluoromethoxy-phenylamino)-pyrrolo[2,3-d]pyrimidin-7-yl]- propionamide;
N-(3-Methoxy-propyl)-3-[4-(4-trifluoromethoxy-phenylamino)-pyrrolo[2,3-d]pyrimidin-7- y 1] -propionamide ;
N-(2-Morpholin-4-yl-ethyl)-3-[4-(4-trifluoromethoxy-phenylamino)-pyrrolo[2,3- d]pyrimidin-7-yl]-propionamide;
7-Methyl-4-(4-trifluoromethoxy-phenylamino)-7H-pyπOlo[2,3-d]pyrimidine-6-carboxylic acid ethyl ester;
(4-Bromo-piperidin-l-yl)-[7-methyl-4-(4-trifluoromethoxy-phenylamino)-7H-pyrrolo[2,3- d]pyrimidin-6-yl]-methanone; l-[7-Methyl-4-(4-trifluoromethoxy-phenylamino)-7H-pyrrolo[2,3-d]pyrimidine-6-carbonyl]- piperidine-4-carboxylic acid amide; l-[7-Methyl-4-(4-trifluoromethoxy-phenylamino)-7H-pyrrolo[2,3-d]pyrimidine-6-carbonyl]- piperidine-3-carboxylic acid amide; l-[7-Methyl-4-(4-trifluoromethoxy-phenylamino)-7H-pyrrolo[2,3-d]pyrimidine-6-carbonyl]- piperidine-3-carboxylic acid ethyl ester;
(3-Hydroxymethyl-piperidin-l-yl)-[7-methyl-4-(4-trifluoromethoxy-phenylamino)-7H- pyrrolo[2,3-d]pyrimidin-6-yl]-methanone;
8-[7-Methyl-4-(4-trifluoromethoxy-phenylamino)-7H-pyrrolo[2,3-d]pyrimidine-6-carbonyl]- l-phenyl-l,3,8-triaza- spiro[4.5]decan-4-one;
[7-Methyl-4-(4-trifluoromethoxy-phenylamino)-7H-pyrrolo[2,3-d]pyrimidin-6-yl]-methanol;
(2-Methoxymethyl-pyrrolidin-l-yl)-[7-methyl-4-(4-trifluoromethoxy-phenylamino)-7H- pyrrolo[2,3-d]pyrimidin-6-yl]-methanone;
7-Methyl-4-(4-trifluoromethoxy-phenylamino)-7H-pyrrolo[2,3-d]pyrimidine-6-carboxylic acid amide;
7-Methyl-4-(4-trifluoromethoxy-phenylamino)-7H-pyrrolo[2,3-d]pyrimidine-6-carboxylic acid ethylamide;
7-Methyl-4-(4-trifluoromethoxy-phenylamino)-7H-pyrrolo[2,3-d]pyrimidine-6-carboxylic acid ( 1 -ethyl-pyrrolidin-2-yl)-amide;
N-[7-Methyl-4-(4-trifluoromethoxy-phenylamino)-7H-pyrrolo[2,3-d]pyrimidine-6-carbonyl]- guanidine;
7-Methyl-4-(4-trifluoromethoxy-phenylamino)-7H-pyπOlo[2,3-d]pyrimidine-6-carboxylic acid (4-diethylamino- 1 -methyl-butyl)-amide;
4-(2-Bromo-4-trifluoromethoxy-phenylamino)-l-(2-morpholin-4-yl-ethyl)-lH-pyrrolo[2,3- b]pyridine- 5 -carbaldehyde .
12. The compound of claim 10, of formula VI:
Figure imgf000131_0001
wherein Ri is independently selected from H, an optionally substituted Ci^alkyl, an optionally substituted C3-i2cycloalkyl, an optionally substituted C3-i2heterocycloalkyl, an optionally substituted C3.i2aryl, an optionally substituted C3-i2heteroaryl, an optionally substituted (CH2)nC3-i2heterocycloalkyl, -C(O)RX, -ORX, -SRX, -NRXRX, nitro, cyano, C,-6haloalkyl, C,. 6aminoalkyl, (CH2)nC(O)Rx, (CH2)nC(O)ORx, (CH2)nC(O)NRxRx, or -(CH2)nC(O)NRxRxS(O)2Rx;
Rx, for each occurrence, is independently H, an optionally substituted Ci-6alkyl, an optionally substituted C3-i2cycloalkyl, an optionally substituted C3-i2heterocycloalkyl, an optionally substituted C3-i2aryl, an optionally substituted C3.i2heteroaryl, NHNHR2, C(NH)NH2;
R2 is H or Ci-4 alkyl; and
Each n is independently 0, 1, or 2.
13. The compound of claim 12, selected from the following: N-(2-hydroxyethyl)-4-(4-(trifluoromethoxy)phenylamino)thieno[2,3-d]pyrimidine-6- carboxamide;
Thieno[2,3-d]pyrimidin-4-yl-(4-trifluoromethoxy-phenyl)-amine; (6-Methyl-thieno[2,3-d]pyrimidin-4-yl)-(4-trifluoromethoxy-phenyl)-amine; 4-(4-Trifluoromethoxy-phenylamino)-thieno[2,3-d]pyrimidine-6-carboxylic acid methyl ester;
4-(4-Trifluoromethoxy-phenylamino)-thieno[2,3-d]pyrimidine-6-carboxylic acid; 4-(4-Trifluoromethoxy-phenylamino)-thieno[2,3-d]pyrimidine-6-carboxylic acid (2- morpholin-4-yl-ethyl)-amide;
[4-(4-Trifluoromethoxy-phenylamino)-thieno[2,3-d]pyrimidin-6-yl]-methanol; 4-(4-Trifluoromethoxy-phenylamino)-thieno[2,3-d]pyrimidine-6-carboxylic acid ethylamide; 4-(4-Trifluoromethoxy-phenylamino)-thieno[2,3-d]pyrimidine-6-carboxylic acid (2-hydroxy- ethyl)-amide;
[6-(3-Dimethylamino-pyrrolidin-l-ylmethyl)-thieno[2,3-d]pyrimidin-4-yl]-(4- trifluoromethoxy-phenyl)-amine;
(3-Dimethylamino-pyπOlidin-l-yl)-[4-(4-trifluoromethoxy-phenylamino)-thieno[2,3- d]pyrimidin-6-yl]-methanone; 4-(4-Trifluoromethoxy-phenylamino)-thieno[2,3-d]pyrimidine-6-carboxylic acid (1 -benzyl- pyrrolidin-3-yl)-amide;
(2-Hydroxymethyl-pyrrolidin-l-yl)-[4-(4-trifluoromethoxy-phenylamino)-thieno[2,3- d]pyrimidin-6-yl]-methanone;
Piperazin-l-yl-[4-(4-trifluoromethoxy-phenylamino)-thieno[2,3-d]pyrimidin-6-yl]- methanone;
(4-Pyrrolidin- 1 -yl-piperazin- 1 -yl)- [4-(4-trifluoromethoxy-pheny lamino)-thieno [2,3 - d]pyrimidin-6-yl]-methanone;
[4-(4-Trifluoromethoxy-phenylamino)-thieno[2,3-d]pyrimidin-6-yl]-[4-(3-trifluoromethyl- phenyl)-piperazin- 1 -yl]-methanone;
4-(4-Trifluoromethoxy-phenylamino)-thieno[2,3-d]pyrimidine-6-carboxylic acid (2- morpholin-4-yl-ethyl)-amide;
(4-Methyl-piperazin-l-yl)-[4-(4-trifluoromethoxy-phenylamino)-thieno[2,3-d]pyrimidin-6- yl]-methanone;
(4-Hydroxy-piperidin-l-yl)-[4-(4-trifluoromethoxy-phenylamino)-thieno[2,3-d]pyrimidin-6- yl]-methanone;
(4-Methoxy-piperidin-l-yl)-[4-(4-trifluoromethoxy-phenylatnino)-thieno[2,3-d]pyrimidin-6- yl]-methanone;
4-(4-Trifluoromethoxy-phenylamino)-thieno[2,3-d]pyrimidine-6-carboxylic acid [2-methyl-
5-(3-trifluoromethyl-benzoylamino)-phenyl]-amide.
14. The compound of claim 1 , of formula VII :
wherein
Ri is selected from H, an optionally substituted
Figure imgf000133_0002
an optionally substituted C3- i2cycloalkyl, an optionally substituted C^nheterocycloalkyl, an optionally substituted C3- i2aryl, an optionally substituted C3-i2heteroaryl, an optionally substituted (CH2)nC3- 12heterocycloalkyl, -C(O)RX, -ORX, -SRX, -NRXRX, nitro, cyano, Ci-6haloalkyl, Ci-6aminoalkyl, (CH2)nC(O)Rx, (CH2)nC(O)ORx, (CH2)nC(O)NRxRx, or -(CH2)nC(O)NRxRxS(O)2Rx;
Rx, for each occurrence, is independently H, an optionally substituted Ci-6alkyl, an optionally substituted C3-i2cycloalkyl, an optionally substituted C3-i2heterocycloalkyl, an optionally substituted C3-12aryl, an optionally substituted C3-i2heteroaryl, NHNHR2, or C(NH)NH2;
R2 is H or Ci-4 alkyl; and each n is independently 0, 1 or 2.
15. The compound of claim 14, selected from the following: 4-(4-(trifluoromethoxy)phenylamino)-N-(2-hydroxyethyl)quinazoline-7-carboxamide; 4-(4-Trifluoromethoxy-phenylamino)-quinazoline-7-carboxylic acid methyl ester; [4-(4-Trifluoromethoxy-phenylamino)-quinazolin-7-yl]-methanol; (7-Diethylaminomethyl-quinazolin-4-yl)-(4-trifluoromethoxy-phenyl)-amine; 4-(4-Trifluoromethoxy-phenylamino)-quinazoline-7-carboxylic acid diethylamide; [4-(Furan-3 -carbonyl)-piperazin- 1 -yl] - [4-(4-trifluoromethoxy-phenylamino)-quinazolin-7- yl]-methanone;
4-(4-Trifluoromethoxy-phenylamino)-quinazoline-7-carboxylic acid (3-trifluoromethyl- phenyl)-amide;
4-(4-Trifluoromethoxy-phenylamino)-quinazoline-7-carboxy lie acid (3 -methoxy-propy I)- amide;
4-(4-Trifluoromethoxy-phenylamino)-quinazoline-7-carboxylic acid N'-ethyl-hydrazide; (2- { [4-(4-Trifluoromethoxy-phenylamino)-quinazoline-7-carbonyl]-amino } -thiazol-5-yl)- acetic acid ethyl ester;
(4-Methoxy-piperidin-l-yl)-[4-(4-trifluoromethoxy-phenylamino)-quinazolin-7-yl]- methanone;
(3-Hydroxy-pyrrolidin-l-yl)-[4-(4-trifluoromethoxy-phenylamino)-quinazolin-7-yl]- methanone;
(2-Hydroxymethyl-pyrrolidin-l-yl)-[4-(4-trifluoromethoxy-phenylamino)-quinazolin-7-yl]- methanone;
(3-Dimethylamino-pyrrolidin-l-yl)-[4-(4-trifluoromethoxy-phenylamino)-quinazolin-7-yl]- methanone; 4-(4-Trifluoromethoxy-phenylamino)-quinazoline-7-carboxylic acid (2-hydroxy-ethyl)- methyl-amide;
4-(4-Trifluoromethoxy-phenylamino)-quinazoline-7-carboxylic acid methy l-( 1 -methyl- piperidin-4-yl)-amide;
4-Methyl-N-[4-(4-trifluoromethoxy-phenylamino)-quinazoline-7-carbonyl]- benzenesulfonamide;
2-Methyl-propane-2-sulfonic acid [4-(4-trifluoromethoxy-phenylamino)-quinazoline-7- carbony 1] -amide ;
(4-Methyl-[l,4]diazepan-l-yl)-[4-(4-trifluoromethoxy-phenylamino)-quinazolin-7-yl]- methanone;
(3,5-Dimethyl-piperazin-l-yl)-[4-(4-trifluoromethoxy-phenylamino)-quinazolin-7-yl]- methanone;
(4-Hydroxy-piperidin-l-yl)-[4-(4-trifluoromethoxy-phenylamino)-quinazolin-7-yl]- methanone;
4-(4-Trifluoromethoxy-phenylamino)-quinazoline-7-carboxylic acid methyl-(2-methylamino- ethyl)-amide;
4-(4-Trifluoromethoxy-phenylamino)-quinazoline-7-carboxylic acid (2-hydroxy-ethyl)- amide; l-Phenyl-8-[4-(4-trifluoromethoxy-phenylamino)-quinazoline-7-carbonyl]-l,3,8-triaza- spiro [4.5 ] decan-4-one ;
(4-Bromo-piperidin-l-yl)-[4-(4-trifluoromethoxy-phenylamino)-quinazolin-7-yl]-methanone; l-[4-(4-Trifluoromethoxy-phenylamino)-quinazoline-7-carbonyl]-piperidine-4-carboxylic acid amide;
1 - [4-(4-Trifluoromethoxy-phenylamino)-quinazoline-7-carbonyl] -piperidine-3 -carboxylic acid amide;
1 - [4-(4-Trifluoromethoxy-phenylamino)-quinazoline-7-carbonyl] -piperidine-3 -carboxylic acid ethyl ester;
(3-Hydroxymethyl-piperidin-l-yl)-[4-(4-trifluoromethoxy-phenylamino)-quinazolin-7-yl]- methanone.
16. The compound of claim 1 , of formula VIII:
Figure imgf000136_0001
wherein
Rι is selected from H, an optionally substituted Ci-6alkyl, an optionally substituted C3. i2cycloalkyl, an optionally substituted C3-i2heterocycloalkyl, an optionally substituted C3- i2aryl, an optionally substituted C3-12heteroaryl, an optionally substituted Ci-6aralkyl, an optionally substituted Ci-όheteroaralkyl, an optionally substituted C1-6haloalkyl, an optionally substituted (CH2)nC3-i2heterocycloalkyl, an optionally substituted (CH2)nC3-12aryl, an optionally substituted (CH2)nC3-12heteroaryl, an optionally substituted (CH2)nC3-i2cycloalkyl, (CH2)nC(0)Rx, (CH2)nC(O)ORx, (CH2)nC(O)NRxRx, -C(O)RX, -C(S)RX, -C(NR)R", halo, C1- βhaloalkyl, -S(O)R", or -S(O)2R";
R", for each occurrence, is independently, H, an optionally substituted Ci-6alkyl, an optionally substituted C3-i2cycloalkyl, an optionally substituted C3-i2heterocycloalkyl, an optionally substituted C3-i2aryl, an optionally substituted C3-12heteroaryl, or Ci-6haloalkyl; and each n is independently 0, 1 or 2.
17. The compound of claim 16, selected from the following: 4-(4-Trifluoromethoxy-phenylamino)-5,7-dihydro-pyrrolo[2,3-d]pyrimidin-6-one; 7-Methyl-4-(4-trifluoromethoxy-phenylamino)-5,7-dihydro-pyrrolo[2,3-d]pyrimidin-6-one.
18. A compound of formula IX, or pharmaceutically acceptable salt, solvate or hydrate thereof:
Figure imgf000136_0002
wherein
R3 is H or CM alkyl;
L is NR3, O, S, S(O), S(O)2, C(O), C(S), C(NR), C(NRX)NRX, C(O)NR", C(O)NRXNRX, C(O)ONR", C(0)NRx0, C(O)O, OC(O), OC(O)O, or a bond; each Rx, for each occurrence, is independently H, an optionally substituted Ci-6alkyl, an optionally substituted C3-i2cycloalkyl, an optionally substituted C3-i2heterocycloalkyl, an optionally substituted C3-i2aryl, an optionally substituted C3.i2heteroaryl, C1-6haloalkyl; NHNHR2, or C(NH)NH2;
A is an optionally substituted Ci^alkyl, an optionally substituted C3-i2aryl, an optionally substituted C3-12heteroaryl, an optionally substituted C3-12cycloalkyl, or an optionally substituted C3-i2heterocycloalkyl.
19. The compound of claim 18, wherein A is an optionally substituted Ci-6alkyl, an optionally substituted C3-i2aryl, an optionally substituted C3-i2heteroaryl, or an optionally substituted C3-i2heterocycloalkyl.
20. The compound of claim 19, wherein A is an optionally substitued phenyl ring, an optionally substituted naphthyl ring, an optionally substituted piperidine ring, an optionally substituted imidazole ring, an optionally substituted pyrazole ring, an optionally substituted lH-pyrazole ring, an optionally substituted lH-Pyrrolo[2,3-b]pyridine ring, an optionally substituted pyrimidine ring, an optionally substituted pyrazine ring, an optionally substituted pyridine ring, an optionally substituted indole ring, an optionally substituted IH- indole ring, an optionally substituted furan ring, an optionally substituted benzo[b]thiophene ring, an optionally substituted dibenzofuran ring, an optionally substituted lH-Pyridin-2-one ring, an optionally substituted thiazole ring, or an ethyl group.
21. The compound of claim 20, wherein A is optionally substituted with Ci-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-i2cycloalkyl, Crnheterocycloalkyl, C3-i4aralkyl, C3- i4heteroaralkyl, C3-i2aryl, (CH2)pC3-i2aryl, C3-i2heteroaryl, (CH2)pC3-i2heteroaryl, halogen, haloalkyl, cyano, (CH2)PCN, nitro, Ci^alkoxy, C3-i2aryloxy, Ci-C4 haloalkoxy, Ci-C6 haloalkyl, hydroxyl, Ci-6hydroxylalkyl, Ci-6carboxyl, CHO, amino, aminoalkyl, dialkylamino, OR15, SR15, NR17R17C(O)OR15, C(O)NR15R16, OC(O)NR15R16, NR15C(O)NR15R16, NR17S(O)2R15, NR17(CH2)nR17, C(O)R17, C(O)NR17R17, NR15C(O)R17, S(O)R17, S(O)2R17, or S(O)2NR17R17; each R15 is independently hydrogen, C3-H aralkyl, Cj-6 alkyl, C3-12 cycloalkyl, C3-I2 aryl, C3-I2 heterocycloalkyl, or C3-J2 heteroaryl; each R16 is independently hydrogen, C3-I4 aralkyl, or Cp6 alkyl; each R17 is independently H, Cr6 alkyl, Cj-6 hydroxyalkyl, C^6 alkoxy, C3-I2 cycloalkyl, C3-i2aryl, C3-i2heteroaryl, C3-i2heterocycloalkyl, hydroxyl, or each R17 may form a heterocycloalkyl ring with another R17; wherein each A, R15, R16, and R17 may be further optionally substituted with Cj-6 alkyl, Ci-6 alkoxy, Cp6 haloalkoxy, C3-i2heterocycloalkyl, amino, halogen, Ci-6 haloalkyl, S02alkyl, or NHC(O)aryl; and each p is independently 0-6.
22. The compound of claim 18, selected from the following: 4-(6-(4-(trifluoromethoxy)phenylamino)-5-methylpyrimidin-4-yl)-N-(2- morpholinoethyl)benzamide;
6-( 1 H-pyrrolo[2,3 -b]pyridin- 1 -yl)-N-(4-(trifluoromethoxy)phenyl)pyrimidin-4-amine; N-(4-(trifluoromethoxy)phenyl)-6-(l,3,5-trimethyl-lH-pyrazol-4-yl)pyrimidin-4-amine; 4-[6-(4-Trifluoromethoxy-phenylamino)-pyrimidin-4-yl]-benzenesulfonamide; 4-[5-Methyl-6-(4-trifluoromethoxy-phenylamino)-pyrimidin-4-yl]-benzenesulfonamide; N-Methyl-4-[5-methyl-6-(4-trifluoromethoxy-phenylarnino)-pyrimidin-4-yl]- benzenesulfonamide;
N-Ethyl-4-[6-(4-trifluoromethoxy-phenylamino)-pyrimidin-4-yl]-benzenesulfonamide; N-(2-Hydroxy-ethyl)-4-[6-(4-trifluoromethoxy-phenylamino)-pyrimidin-4-yl]- benzenesulfonamide;
[6-(4-Methanesulfonyl-phenyl)-pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)-amine; [6-(4-Methanesulfonyl-phenyl)-5-methyl-pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)- amine;
{6-[4-(Propane-2-sulfonyl)-phenyl]-pyrimidin-4-yl}-(4-trifluoromethoxy-phenyl)-amine; { 5-Methyl-6-[4-(propane-2-sulfonyl)-phenyl]-pyrimidin-4-yl } -(4-trifluoromethoxy-phenyl)- amine; N,N-Diethyl-4-[6-(4-trifluoromethoxy-phenylamino)-pyrimidin-4-yl]-benzenesulfonamide; N,N-Diethyl-4-[5-methyl-6-(4-trifluoromethoxy-phenylamino)-pyrimidin-4-yl]- benzenesulfonamide ;
{ 6- [4-(Piperidine- 1 -sulfonyl)-phenyl] -pyrimidin-4-yl } -(4-trifluoromethoxy-pheny l)-amine;
4-Methyl-N-{4-[6-(4-trifluoromethoxy-phenylamino)-pyrimidin-4-yl]-phenyl}- benzenesulfonamide;
4-Methyl-N-{4-[5-methyl-6-(4-trifluoromethoxy-phenylamino)-pyrimidin-4-yl]-phenyl}- benzenesulfonamide;
{ 6- [4-(Moφholine-4-sulfonyl)-phenyl] -pyrimidin-4-yl } -(4-trifluoromethoxy-pheny l)-amine;
N-{4-[6-(4-Trifluoromethoxy-phenylamino)-pyrimidin-4-yl]-phenyl}-methanesulfonamide;
Ethanesulfonic acid {4-[6-(4-trifluoromethoxy-phenylamino)-pyrimidin-4-yl]-phenyl}- amide;
Butane- 1 -sulfonic acid { 4- [6-(4-trifluoromethoxy-phenylamino)-pyrimidin-4-y l]-phenyl } - amide;
Propane-2-sulfonic acid {4-[6-(4-trifluoromethoxy-phenylarnino)-pyrimidin-4-yl]-phenyl} - amide;
Pentane-2-sulfonic acid { 4- [6-(4-trifluoromethoxy-phenylamino)-pyrimidin-4-yl]-phenyl } - amide;
N-(2-Hydroxy-ethyl)-4-[5-methyl-6-(4-trifluoromethoxy-phenylamino)-pyrimidin-4-yl]- benzamide;
4-[5-Methyl-6-(4-trifluoromethoxy-phenylamino)-pyrimidin-4-yl]-N-(2-moφholin-4-yl- ethyl)-benzamide;
N-(3-Methoxy-propyl)-4-[5-methyl-6-(4-trifluoromethoxy-phenylamino)-pyrimidin-4-yl]- benzamide;
N-{3-[5-Methyl-6-(4-trifluoromethoxy-phenylamino)-pyrimidin-4-yl]-phenyl}-acetamide;
3-[5-Methyl-6-(4-trifluoromethoxy-phenylamino)-pyrimidin-4-yl]-benzamide;
3-[5-Methyl-6-(4-trifluoromethoxy-phenylamino)-pyrimidin-4-yl]-benzoic acid;
(6-Biphenyl-4-yl-pyrimidin-4-yl)-(4-trifluoromethoxy-phenyl)-amine;
(6-Biphenyl-4-yl-5-methyl-pyrimidin-4-yl)-(4-trifluoromethoxy-phenyl)-amine;
[6-(5-Methoxy-naphthalen-2-yl)-pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)-amine;
[6-(5-Methoxy-naphthalen-2-yl)-5-methyl-pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)- amine;
{3-[6-(4-Trifluoromethoxy-phenylamino)-pyrimidin-4-yl]-phenyl}-acetonitrile; [6-(3-Nitro-phenyl)-pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)-amine;
4-Methoxy-N-{4-[6-(4-trifluoromethoxy-phenylamino)-pyrimidin-4-yl]-phenyl}-benzamide;
[6-(4-Aminomethyl-phenyl)-5-methyl-pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)-amine;
5-Bromo-N-(2-morpholin-4-yl-ethyl)-N'-(4-trifluoromethoxy-phenyl)-pyrimidine-4,6- diamine; l-[5-Bromo-6-(4-trifluoromethoxy-phenylamino)-pyrimidin-4-yl]-piperidine-3-carboxylic acid amide;
{ 6- [2-(2-Morpholin-4-yl-ethylamino)-imidazol- 1 -yl]-pyrimidin-4-yl } -(4-trifluoromethoxy- phenyl)-amine;
(4-Trifluoromethoxy-phenyl)- { 6- [2-(4-trifluoromethoxy-phenylamino)-imidazol- 1 -yl] - pyrimidin-4-yl} -amine;
N-(5-Cyclopropyl-lH-pyrazol-3-yl)-N'-(4-trifluoromethoxy-phenyl)-pyrimidine-4,6-diamine;
[6-(3-Amino-5-cyclopropyl-pyrazol-l-yl)-pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)- amine;
N-(4-Trifluoromethoxy-phenyl)-N'- [ 1 -(3 -trifluoromethyl -phenyl)- 1 H-imidazol-2-yl] - pyrimidine-4,6-diamine;
(6-PyπOlo[2,3-b]pyridin-l-yl-pyrimidin-4-yl)-(4-trifluoromethoxy-phenyl)-amine;
[6-(lH-Pyrazol-4-yl)-pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)-amine;
[6-( 1 -Methyl- 1 H-pyrazol-4-yl)-pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)-amine;
[6-( 1 -Isobutyl- 1 H-pyrazol-4-yl)-pyrimidin-4-yl] -(4-trifluoromethoxy-phenyl)-amine;
[6-(l-Isobutyl-lH-pyrazol-4-yl)-5-methyl-pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)- amine;
(4-Trifluoromethoxy-phenyl)-[6-(l,3,5-trimethyl-lH-pyrazol-4-yl)-pyrimidin-4-yl]-amine;
{ 6- [ 1 -(4-Methanesulfonyl-benzyl)- 1 H-pyrazol-4-y 1] -pyrimidin-4-yl } -(4-trifluoromethoxy- phenyl)-amine;
[6-( 1 -Ethanesulfonyl- 1 H-pyrazol-4-yl)-pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)-amine;
[6-( 1 -Pyridin-4-ylmethyl- 1 H-pyrazol-4-yl)-pyrimidin-4-yl]-(4-trifluorornethoxy-phenyl)- amine;
N6-(2-Mθφholin-4-yl-ethyl)-N6l-(4-trifluoromethoxy-phenyl)-[4,4']bipyrimidinyl-6,6'- diamine;
[5-Methyl-6-(l-methyl-lH-pyrazol-4-yl)-pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)- amine; [6-(5-Methyl- 1 -phenyl- 1 H-pyrazol-4-yl)-pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)-amine;
[5-Methyl-6-(5-methyl- 1 -phenyl- 1 H-pyrazol-4-yl)-pyrimidin-4-yl]-(4-trifluoromethoxy- phenyl)-amine;
[6-(lH-Indol-4-yl)-pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)-amine;
[6-( 1 H-Indol-3 -yl)-pyrimidin-4-yl] -(4-trifluoromethoxy-phenyl)-amine;
[6-(lH-Indol-5-yl)-pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)-amine;
4-[6-(4-Trifluoromethoxy-phenylamino)-pyrimidin-4-yl]-furan-2-carbaldehyde;
(6-Benzo[b]thiophen-4-yl-pyrimidin-4-yl)-(4-trifluoromethoxy-phenyl)-amine;
[6-(lH-Indol-4-yl)-5-methyl-pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)-amine;
(6-Dibenzofuran-4-yl-pyrimidin-4-yl)-(4-trifluoromethoxy-phenyl)-amine;
[6-(l -Benzyl- lH-pyrazol-4-yl)-pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)-amine;
[6-( 1 -Benzyl- 1 H-pyrazol-4-yl)-5-methyl-pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)-amine;
N6,N6'-Bis-(4-trifluoromethoxy-phenyl)-[4,4']bipyrimidinyl-6,6'-diamine;
N6-(2-Morpholin-4-yl-ethyl)-N6'-(4-trifluoromethoxy-phenyl)-[4,4']bipyrimidinyl-6,6'- diamine;
N-(4-Methyl-3-{l-[6-(4-trifluoromethoxy-phenylamino)-pyrimidin-4-yl]-lH-imidazol-2- ylamino} -phenyl)-benzamide;
N-(4-Methyl-3 - { 1 -[6-(4-trifluoromethoxy-phenylamino)-pyrimidin-4-yl] - 1 H-imidazol-2- ylamino}-phenyl)-4-morpholin-4-ylmethyl-3-trifluoromethyl-benzamide;
[6-(l-Pyridin-3-ylmethyl-lH-pyrazol-4-yl)-pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)- amine;
[6-(lH-Pyrrolo[2,3-b]pyridin-4-yl)-pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)-amine;
4-[6-(4-Trifluoromethoxy-phenylamino)-pyrimidin-4-yl]-lH-pyridin-2-one;
[6-(2-Methyl-thiazol-4-yl)-pyrimidin-4-yl]-(4-trifluoromethoxy-phenyl)-amine;
2-Fluoro-5-[6-(4-trifluoromethoxy-phenylamino)-pyrimidin-4-yl]-benzaldehyde;
{6-[2-(4-Methyl-piperazin-l-yl)-pyridin-4-yl]-pyrimidin-4-yl}-(4-trifluoromethoxy-phenyl)- amine.
23. A compound of formula X, or pharmaceutically acceptable salt, solvate or hydrate thereof:
Figure imgf000142_0001
wherein each ofX2 or X3 is independently NRi, CR1, C(O), O, or S; each R] is independently selected from H, an optionally substituted Ci-6alkyl, an optionally substituted C3-i2cycloalkyl, an optionally substituted Cs-πheterocycloalkyl, an optionally substituted C3-i2aryl, an optionally substituted C3-12heteroaryl, an optionally substituted C2-6aralkyl, an optionally substituted C2-i2heteroaralkyl, an optionally substituted Ci-6haloalkyl, an optionally substituted (CH2)nC3-12aryl, an optionally substituted (CH2)nC3- i2heteroaryl, an optionally substituted (CH2)nC3-i2cycloalkyl, an optionally substituted (CH2)nC3-i2heterocycloalkyl, Ci-6haloalkyl, Ci-6aminoalkyl, hal, (CH2)nC(0)Rx, (CH2)nC(O)ORx, (CH2)nC(O)NRxRx, -(CH2)nC(O)NRxRxS(O)2Rx; -C(O)RX, -C(S)RX, -C(NR)RX, -SRX, -S(O)RX, -S(O)2R", -ORX, -NRXRX, nitro, cyano, or absent; each Rx, for each occurrence, is independently H, an optionally substituted Ci-6alkyl, an optionally substituted C3-i2cycloalkyl, an optionally substituted C3-i2heterocycloalkyl, an optionally substituted C3-i2aryl, an optionally substituted C3-12heteroaryl, C1-6haloalkyl; NHNHR2, or C(NH)NH2;
R2 is H or Ci-6 alkyl; and each n is independently O, 1 or 2.
24. The compound of claim 23, of formula XI:
Figure imgf000142_0002
wherein
Ri is independently selected from H, an optionally substituted Ci-6alkyl, an optionally substituted C3-i2aryl, or an optionally substituted C3-i2heteroaryl.
25. The compound of claim 24, selected from the following: 2-(3-nitrophenyl)-N-(4-(trifluoromethoxy)phenyl)imidazo[l,2-c]pyrimidin-7-amine; 2-Hydroxy-3-[7-(4-trifluoromethoxy-phenylamino)-imidazo[l,2-c]pyrimidin-2-yl]- benzamide.
26. A method of treating a kinase activity related disorder in a subject, comprising administering to said subject in need thereof, an effective amount of a compound, or pharmaceutically acceptable salt, solvate or hydrate thereof, of any one of claims 1-25.
27. The method of claim 26, wherein the compound is a kinase inhibitor.
28. The method of claim 27, wherein the compound interacts with Bcr-abl.
29. The method of claim 28, wherein the compound targets the binding site of Bcr-abl.
30. The method of claim 27 wherein the subject is identified as being in need of kinase inhibition, and the compound is administered to the identified subject.
31. The method of claim 26 further comprising the step of identifying a subject in need of such treatment for a kinase activity related disorder and administering the compound to the identified subject.
32. The method of claim 26 wherein the subject is suffering from a cell proliferative disorder or disease.
33. The method of claim 32 wherein the disorder is cancer, tumor, neoplasm, neovascularization, vascularization, cardiovascular disease, metastasis, infection, blood clot, atherosclerosis, melanoma, skin disorder, rheumatoid arthritis, diabetic retinopathy, macular edema, or macular degeneration, inflammatory and arthritic disease, or osteosarcoma.
34. The method of claim 33 wherein the subject is suffering from a solid tumor or disseminated cancer.
35. The method of claim 34, wherein the cancer is leukemia, multiple myeloma or lymphoma.
36. The method of claim 35, wherein the leukemia is Chronic Myelogenous Leukemia.
37. A method of treating cancer in a subject, the method comprising administering to said subject an effective amount of a compound, or pharmaceutically acceptable salt, solvate or hydrate thereof, of any one of claims 1-25.
38. The method of claim 26 or claim 37, further comprising administering an additional therapeutic agent.
39. The method of claim 38, wherein the additional therapeutic agent is an ATP- site inhibitor.
40. The method of claim 39, wherein the ATP-site inhibitor is selected from the following: imatinib, Nilotinib, Dasatinib, AP234464, AZD0530, and SKI-606.
41. The method of claim 26 or claim 37, wherein the subject is a mammal.
42. The method of claim 41 wherein the subject is a primate or human.
43. The method of claim 26 or claim 37, wherein the step of administering the compound comprises administering the compound orally, topically, parentally, intravenously or intramuscularly.
44. The method of claim 43, comprising the step of administering an effective amount of a composition comprising the compound and a pharmaceutically suitable excipient.
45. The method of claim 26 or claim 37, wherein the step of administering the compound comprises administering the compound in a dosage of between about 0.01 μg/kg/day and 100 mg/kg/day.
46. A method of inhibiting kinase activity in a subject, the method comprising the step of administering to the subject a compound, or pharmaceutically acceptable salt, solvate or hydrate thereof, of any one of claims 1-25.
47. A pharmaceutical composition comprising a compound, or pharmaceutically acceptable salt, solvate or hydrate thereof, of any one of claims 1-25.
48. The composition of claim 47 further comprising an additional therapeutic agent.
49. The composition of claim 48 wherein the additional therapeutic agent is an ATP-site inhibitor.
50. A kit comprising an effective amount of a compound, or pharmaceutically acceptable salt, solvate or hydrate thereof, of any one of claims 1-25, in unit dosage form, together with instructions for administering the compound to a subject suffering from or susceptible to a cell proliferative disorder.
51. A method for identifying a compound which modulates the kinase activity of Bcr-abl, the method comprising: a) contacting Bcr-abl with a compound under conditions suitable for modulation of the kinase activity of Bcr-abl; and b) detecting modulation of the kinase activity of Bcr-abl by the compound; wherein the compound is capable of interacting with the myristate binding site of Bcr- abl.
52. The method of claim 51, wherein the interaction of the compound with myristate binding site of Bcr-abl is a binding interaction.
53. The method of claim 52, wherein the binding interaction is ionic, covalent, or a non-direct interaction.
54 The method of claim 52 wherein the compound has a binding interaction with Ala452, Tyr454, Glu481, Pro484, Val487, Phe516, Ile521, or Val525.
55. The method of claim 51, wherein said modulation of the activity of the kinase activity is detected by direct binding of the compound to the myristate binding site of Bcr-abl.
56. The method of claim 51, wherein the compound is designed de novo.
57. The method of claim 51 , wherein the compound is designed from a known ligand of the myristate binding site of Bcr-abl.
PCT/US2008/013219 2007-11-28 2008-11-28 Small molecule myristate inhibitors of bcr-abl and methods of use WO2009073153A2 (en)

Priority Applications (18)

Application Number Priority Date Filing Date Title
AU2008331867A AU2008331867B2 (en) 2007-11-28 2008-11-28 Small molecule myristate inhibitors of Bcr-abl and methods of use
EP19170210.9A EP3536687B8 (en) 2007-11-28 2008-11-28 Small molecule myristate inhibitors of bcr-abl and methods of use
EP08857261.5A EP2222162B1 (en) 2007-11-28 2008-11-28 Small molecule myristate inhibitors of bcr-abl and methods of use
EA201070595A EA019869B1 (en) 2007-11-28 2008-11-28 Small molecule myristate inhibitors of tyrosine kinase bcr-abl and methods of use thereof
CA2707046A CA2707046A1 (en) 2007-11-28 2008-11-28 Small molecule myristate inhibitors of bcr-abl and methods of use
EP21184206.7A EP3912973A3 (en) 2007-11-28 2008-11-28 Small molecule myristate inhibitors of bcr-abl and methods of use
ES08857261.5T ES2610190T3 (en) 2007-11-28 2008-11-28 Bcr-abl small molecule myristate inhibitors and methods of use
JP2010536015A JP5779778B2 (en) 2007-11-28 2008-11-28 BCR-ABL low molecular weight myristic acid ester inhibitor and method of using the same
MX2010005671A MX2010005671A (en) 2007-11-28 2008-11-28 Small molecule myristate inhibitors of bcr-abl and methods of use.
BRPI0819453-0A2A BRPI0819453A2 (en) 2007-11-28 2008-11-28 COMPOUND, METHODS TO TREAT A KINASE ACTIVITY DISORDER, TO TREAT CANCER IN A PATIENT, TO INHIBIT KINASE ACTIVITY AND TO IDENTIFY A COMPOUND THAT MODULATES BCR-ABL KINASE ACTIVITY, PHARMACEUTICAL COMPOSITION, KARMACEUTICAL COMPOSITION
EP16197611.3A EP3150593B8 (en) 2007-11-28 2008-11-28 Small molecule myristate inhibitors of bcr-abl and methods of use
US12/745,496 US8921336B2 (en) 2007-11-28 2008-11-28 Small molecule myristate inhibitors of BCR-ABL and methods of use
CN200880123462.5A CN101917849B (en) 2007-11-28 2008-11-28 Small molecule myristate inhibitors of BCR-ABL and methods of use
US14/540,919 US9670214B2 (en) 2007-11-28 2014-11-13 Small molecule myristate inhibitors of Bcr-abl and methods of use
US15/582,004 US20170349595A1 (en) 2007-11-28 2017-04-28 Small molecule myristate inhibitors of bcr-abl and abl and methods of use
US16/170,921 US10787455B2 (en) 2007-11-28 2018-10-25 Small molecule myristate inhibitors of BCR-ABL and methods of use
US16/933,095 US11254682B2 (en) 2007-11-28 2020-07-20 Small molecule myristate inhibitors of Bcr-abl and methods of use
US17/574,132 US11932646B2 (en) 2007-11-28 2022-01-12 Small molecule myristate inhibitors of Bcr-abl and methods of use

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US446207P 2007-11-28 2007-11-28
US61/004,462 2007-11-28

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US12/745,496 A-371-Of-International US8921336B2 (en) 2007-11-28 2008-11-28 Small molecule myristate inhibitors of BCR-ABL and methods of use
US14/540,919 Continuation US9670214B2 (en) 2007-11-28 2014-11-13 Small molecule myristate inhibitors of Bcr-abl and methods of use

Publications (2)

Publication Number Publication Date
WO2009073153A2 true WO2009073153A2 (en) 2009-06-11
WO2009073153A3 WO2009073153A3 (en) 2009-12-30

Family

ID=40718412

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2008/013219 WO2009073153A2 (en) 2007-11-28 2008-11-28 Small molecule myristate inhibitors of bcr-abl and methods of use

Country Status (12)

Country Link
US (6) US8921336B2 (en)
EP (4) EP3150593B8 (en)
JP (2) JP5779778B2 (en)
KR (1) KR20100130583A (en)
CN (3) CN104311563B (en)
AU (1) AU2008331867B2 (en)
BR (1) BRPI0819453A2 (en)
CA (1) CA2707046A1 (en)
EA (1) EA019869B1 (en)
ES (3) ES2893580T3 (en)
MX (3) MX2010005671A (en)
WO (1) WO2009073153A2 (en)

Cited By (37)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012066061A1 (en) * 2010-11-19 2012-05-24 F. Hoffmann-La Roche Ag Pyrazolopyridines and pyrazolopyridines and their use as tyk2 inhibitors
CN102574863A (en) * 2009-08-27 2012-07-11 拜奥克里斯特制药公司 Heterocyclic compounds as JANUS kinase inhibitors
EP2548878A1 (en) * 2011-07-21 2013-01-23 Laboratorios Del. Dr. Esteve, S.A. Pyrazolo[3,4-d]pyrimidine compounds, their preparation and use as sigma ligands
US8637526B2 (en) 2008-10-31 2014-01-28 Genentech, Inc. Pyrazolopyrimidine JAK inhibitor compounds and methods
US8697708B2 (en) 2010-09-15 2014-04-15 F. Hoffmann-La Roche Ag Azabenzothiazole compounds, compositions and methods of use
WO2014152716A1 (en) * 2013-03-15 2014-09-25 President And Fellows Of Harvard College Substituted heterocyclic compounds for treating or preventing viral infections
US8999998B2 (en) 2009-07-02 2015-04-07 Genentech, Inc. Pyrazolopyrimidine JAK inhibitor compounds and methods
US9266892B2 (en) 2012-12-19 2016-02-23 Incyte Holdings Corporation Fused pyrazoles as FGFR inhibitors
US9346815B2 (en) 2014-05-23 2016-05-24 Genentech, Inc. 5-chloro-2-difluoromethoxyphenyl pyrazolopyrimidine compounds, compositions and methods of use thereof
US9388185B2 (en) 2012-08-10 2016-07-12 Incyte Holdings Corporation Substituted pyrrolo[2,3-b]pyrazines as FGFR inhibitors
WO2016173557A1 (en) * 2015-04-30 2016-11-03 中国科学院上海药物研究所 Compound having kinase inhibition activity, and preparation method and uses
US9533954B2 (en) 2010-12-22 2017-01-03 Incyte Corporation Substituted imidazopyridazines and benzimidazoles as inhibitors of FGFR3
US9533984B2 (en) 2013-04-19 2017-01-03 Incyte Holdings Corporation Bicyclic heterocycles as FGFR inhibitors
US9580423B2 (en) 2015-02-20 2017-02-28 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US9611267B2 (en) 2012-06-13 2017-04-04 Incyte Holdings Corporation Substituted tricyclic compounds as FGFR inhibitors
US9708318B2 (en) 2015-02-20 2017-07-18 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US9890156B2 (en) 2015-02-20 2018-02-13 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
WO2018098280A1 (en) * 2016-11-22 2018-05-31 Dana-Farber Cancer Institute, Inc. Degradation of protein kinases by conjugation of protein kinase inhibitors with e3 ligase ligand and methods of use
US10307426B2 (en) 2017-05-22 2019-06-04 Genentech, Inc. Therapeutic compounds and compositions, and methods of use thereof
AU2015292818B2 (en) * 2014-07-21 2020-01-16 Dana-Farber Cancer Institute, Inc. Imidazolyl kinase inhibitors and uses thereof
US10611762B2 (en) 2017-05-26 2020-04-07 Incyte Corporation Crystalline forms of a FGFR inhibitor and processes for preparing the same
US10851105B2 (en) 2014-10-22 2020-12-01 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US10954242B2 (en) 2016-07-05 2021-03-23 The Broad Institute, Inc. Bicyclic urea kinase inhibitors and uses thereof
US11174257B2 (en) 2018-05-04 2021-11-16 Incyte Corporation Salts of an FGFR inhibitor
US11241435B2 (en) 2016-09-16 2022-02-08 The General Hospital Corporation Uses of salt-inducible kinase (SIK) inhibitors for treating osteoporosis
US11285158B2 (en) 2017-02-28 2022-03-29 The General Hospital Corporation Uses of pyrimidopyrimidinones as SIK inhibitors
WO2022087008A1 (en) * 2020-10-21 2022-04-28 Vivace Therapeutics, Inc. Tertiary carboxamide compounds
US11407750B2 (en) 2019-12-04 2022-08-09 Incyte Corporation Derivatives of an FGFR inhibitor
US11420935B2 (en) 2019-04-16 2022-08-23 Vivace Therapeutics, Inc. Bicyclic compounds
US11466004B2 (en) 2018-05-04 2022-10-11 Incyte Corporation Solid forms of an FGFR inhibitor and processes for preparing the same
US11566028B2 (en) 2019-10-16 2023-01-31 Incyte Corporation Bicyclic heterocycles as FGFR inhibitors
US11591329B2 (en) 2019-07-09 2023-02-28 Incyte Corporation Bicyclic heterocycles as FGFR inhibitors
US11607416B2 (en) 2019-10-14 2023-03-21 Incyte Corporation Bicyclic heterocycles as FGFR inhibitors
US11628162B2 (en) 2019-03-08 2023-04-18 Incyte Corporation Methods of treating cancer with an FGFR inhibitor
US11866431B2 (en) 2018-11-09 2024-01-09 Vivace Therapeutics, Inc. Bicyclic compounds
US11897891B2 (en) 2019-12-04 2024-02-13 Incyte Corporation Tricyclic heterocycles as FGFR inhibitors
US11939331B2 (en) 2021-06-09 2024-03-26 Incyte Corporation Tricyclic heterocycles as FGFR inhibitors

Families Citing this family (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104311563B (en) * 2007-11-28 2016-12-07 达那-法伯癌症研究所 The small molecule myristate inhibitors of BCR-ABL and using method thereof
US9273077B2 (en) 2008-05-21 2016-03-01 Ariad Pharmaceuticals, Inc. Phosphorus derivatives as kinase inhibitors
DK2300013T3 (en) 2008-05-21 2017-12-04 Ariad Pharma Inc PHOSPHORUS DERIVATIVES AS KINASE INHIBITORS
WO2011104340A1 (en) * 2010-02-26 2011-09-01 Boehringer Ingelheim International Gmbh Thienopyrimidines containing a substituted alkyl group for pharmaceutical compositions
BR112012021364A2 (en) * 2010-02-26 2016-10-25 Boehringer Ingelheim Int "thienopyrimidines-containing cycloalkyl compounds and pharmaceutical compositions".
UY33241A (en) * 2010-02-26 2011-09-30 Boehringer Ingelheim Int ? Tienopyrimidines containing heterocycloalkyl for pharmaceutical compositions ?.
CA2798578C (en) 2010-05-21 2015-12-29 Chemilia Ab Novel pyrimidine derivatives
CA2830129C (en) 2011-03-24 2016-07-19 Chemilia Ab Novel pyrimidine derivatives
CN103501612B (en) 2011-05-04 2017-03-29 阿里亚德医药股份有限公司 The compound that cell is bred in cancer caused by suppression EGF-R ELISA
EP2809671B1 (en) * 2012-01-30 2016-01-13 Cephalon, Inc. Imidazo [4, 5 - b]pyridine derivatives as alk and jak modulators for the treatment of proliferative disorders
US20150166591A1 (en) 2012-05-05 2015-06-18 Ariad Pharmaceuticals, Inc. Methods and compositions for raf kinase mediated diseases
CN102746308B (en) * 2012-07-09 2014-12-31 四川大学 Allopurinol derivative and preparation method and application thereof
CN102746306B (en) * 2012-07-09 2014-11-19 四川国康药业有限公司 Allopurinol derivative and preparation method and application thereof
US9611283B1 (en) 2013-04-10 2017-04-04 Ariad Pharmaceuticals, Inc. Methods for inhibiting cell proliferation in ALK-driven cancers
US10457691B2 (en) 2014-07-21 2019-10-29 Dana-Farber Cancer Institute, Inc. Macrocyclic kinase inhibitors and uses thereof
CN105561040A (en) * 2016-02-24 2016-05-11 梁功 Traditional Chinese medicine composition for treating hemiplegia
CN105777657A (en) * 2016-03-01 2016-07-20 张宏业 Medicine composition for treating hemiplegia
US10722484B2 (en) 2016-03-09 2020-07-28 K-Gen, Inc. Methods of cancer treatment
US20210130352A1 (en) * 2018-06-21 2021-05-06 Janssen Pharmaceutica Nv Oga inhibitor compounds
CA3222586A1 (en) * 2021-06-07 2022-12-15 The General Hospital Corporation Inhibitors of ttbk1

Family Cites Families (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IL112249A (en) * 1994-01-25 2001-11-25 Warner Lambert Co Pharmaceutical compositions containing di and tricyclic pyrimidine derivatives for inhibiting tyrosine kinases of the epidermal growth factor receptor family and some new such compounds
WO1998012184A1 (en) * 1996-09-19 1998-03-26 Nippon Soda Co., Ltd. Pyrimidine compounds, process for the preparation thereof, and pest controlling agents
US6413971B1 (en) * 1996-11-27 2002-07-02 Pfizer Inc Fused bicyclic pyrimidine derivatives
CZ20001709A3 (en) * 1997-11-11 2001-12-12 Pfizer Products Inc. Thienopyrimide and thienopyridine derivatives, pharmaceutical compositions and treatment methods based thereon
KR100415791B1 (en) * 1998-06-19 2004-01-24 화이자 프로덕츠 인코포레이티드 PYRROLO[2,3-d]PYRIMIDINE COMPOUNDS
US6258820B1 (en) * 1999-03-19 2001-07-10 Parker Hughes Institute Synthesis and anti-tumor activity of 6,7-dialkoxy-4-phenylamino-quinazolines
KR100790202B1 (en) 2000-07-06 2007-12-31 노벨리스 인코퍼레이티드 Method of making aluminum foil for fins
EP1351691A4 (en) * 2000-12-12 2005-06-22 Cytovia Inc Substituted 2-aryl-4-arylaminopyrimidines and analogs as activators of caspases and inducers of apoptosis and the use thereof
CZ294535B6 (en) * 2001-08-02 2005-01-12 Ústav Experimentální Botaniky Avčr Heterocyclic compounds based on N6-substituted adenine, processes of their preparation, their use in the preparation of medicaments, cosmetic compositions and growth regulators, as well as pharmaceutical preparations, cosmetic compositions and growth regulators in which these compounds are comprised
WO2003031406A2 (en) * 2001-10-12 2003-04-17 Irm Llc Kinase inhibitor scaffolds and methods for their preparation
JP2005526714A (en) * 2002-01-17 2005-09-08 ニューロジェン・コーポレーション Substituted quinazolin-4-ylamine analogs as modulators of capsaicin
TW200400034A (en) * 2002-05-20 2004-01-01 Bristol Myers Squibb Co Pyrazolo-pyrimidine aniline compounds useful as kinase inhibitors
WO2004058713A1 (en) * 2002-12-20 2004-07-15 Irm Llc Differential tumor cytotoxocity compounds and compositions
US20050014753A1 (en) 2003-04-04 2005-01-20 Irm Llc Novel compounds and compositions as protein kinase inhibitors
CN1798734A (en) * 2003-04-04 2006-07-05 Irm责任有限公司 Novel compounds and compositions as protein kinase inhibitors
AU2004260689B8 (en) * 2003-07-29 2008-05-15 Irm Llc Compounds and compositions as protein kinase inhibitors
US7338957B2 (en) * 2003-08-28 2008-03-04 Irm Llc Compounds and compositions as protein kinase inhibitors
GB0326168D0 (en) * 2003-11-10 2003-12-17 Arrow Therapeutics Ltd Chemical compounds
JP2008538544A (en) 2004-09-23 2008-10-30 レディ ユーエス セラピューティックス, インコーポレイテッド Novel pyrimidine compounds, processes for their preparation and compositions containing them
US20070009977A1 (en) * 2005-01-12 2007-01-11 Boyce James P Kinase-directed, activity-based probes
WO2006124874A2 (en) * 2005-05-12 2006-11-23 Kalypsys, Inc. Inhibitors of b-raf kinase
CA2655799A1 (en) * 2005-06-22 2006-12-28 Develogen Aktiengesellschaft Thienopyrimidines for pharmaceutical compositions
EP2044086A2 (en) * 2006-06-30 2009-04-08 Janssen Pharmaceutica N.V. Thiazolopyrimidine modulators of trpv1
CN104311563B (en) * 2007-11-28 2016-12-07 达那-法伯癌症研究所 The small molecule myristate inhibitors of BCR-ABL and using method thereof

Non-Patent Citations (1)

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

Cited By (69)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8637526B2 (en) 2008-10-31 2014-01-28 Genentech, Inc. Pyrazolopyrimidine JAK inhibitor compounds and methods
US8999998B2 (en) 2009-07-02 2015-04-07 Genentech, Inc. Pyrazolopyrimidine JAK inhibitor compounds and methods
CN102574863A (en) * 2009-08-27 2012-07-11 拜奥克里斯特制药公司 Heterocyclic compounds as JANUS kinase inhibitors
US8697708B2 (en) 2010-09-15 2014-04-15 F. Hoffmann-La Roche Ag Azabenzothiazole compounds, compositions and methods of use
CN103313987A (en) * 2010-11-19 2013-09-18 弗·哈夫曼-拉罗切有限公司 Pyrazolopyridines and pyrazolopyridines and their use as tyk2 inhibitors
JP2013542966A (en) * 2010-11-19 2013-11-28 エフ.ホフマン−ラ ロシュ アーゲー Pyrazolopyridines and their use as TYK2 inhibitors and their use
WO2012066061A1 (en) * 2010-11-19 2012-05-24 F. Hoffmann-La Roche Ag Pyrazolopyridines and pyrazolopyridines and their use as tyk2 inhibitors
US9309240B2 (en) 2010-11-19 2016-04-12 Genentech, Inc. Pyrazolopyridine compounds, compositions and methods of use
US10213427B2 (en) 2010-12-22 2019-02-26 Incyte Corporation Substituted imidazopyridazines and benzimidazoles as inhibitors of FGFR3
US10813930B2 (en) 2010-12-22 2020-10-27 Incyte Corporation Substituted imidazopyridazines and benzimidazoles as inhibitors of FGFR3
US9533954B2 (en) 2010-12-22 2017-01-03 Incyte Corporation Substituted imidazopyridazines and benzimidazoles as inhibitors of FGFR3
EP2548878A1 (en) * 2011-07-21 2013-01-23 Laboratorios Del. Dr. Esteve, S.A. Pyrazolo[3,4-d]pyrimidine compounds, their preparation and use as sigma ligands
WO2013010950A1 (en) * 2011-07-21 2013-01-24 Laboratorios Del Dr. Esteve, S.A. PYRAZOLO[3,4-d]PYRIMIDINE COMPOUNDS, THEIR PREPARATION AND USE AS SIGMA LIGANDS
US9567338B2 (en) 2011-07-21 2017-02-14 Laboratorios Del Dr. Esteve S.A. Pyrazolo[3,4-d]pyrimidine compounds, their preparation and use as sigma ligands
US11840534B2 (en) 2012-06-13 2023-12-12 Incyte Corporation Substituted tricyclic compounds as FGFR inhibitors
US9611267B2 (en) 2012-06-13 2017-04-04 Incyte Holdings Corporation Substituted tricyclic compounds as FGFR inhibitors
US10131667B2 (en) 2012-06-13 2018-11-20 Incyte Corporation Substituted tricyclic compounds as FGFR inhibitors
US11053246B2 (en) 2012-06-13 2021-07-06 Incyte Corporation Substituted tricyclic compounds as FGFR inhibitors
US9388185B2 (en) 2012-08-10 2016-07-12 Incyte Holdings Corporation Substituted pyrrolo[2,3-b]pyrazines as FGFR inhibitors
US9745311B2 (en) 2012-08-10 2017-08-29 Incyte Corporation Substituted pyrrolo[2,3-b]pyrazines as FGFR inhibitors
US9266892B2 (en) 2012-12-19 2016-02-23 Incyte Holdings Corporation Fused pyrazoles as FGFR inhibitors
WO2014152716A1 (en) * 2013-03-15 2014-09-25 President And Fellows Of Harvard College Substituted heterocyclic compounds for treating or preventing viral infections
US10150742B2 (en) 2013-03-15 2018-12-11 President And Fellows Of Harvard College Substituted heterocyclic compounds for treating or preventing viral infections
US11530214B2 (en) 2013-04-19 2022-12-20 Incyte Holdings Corporation Bicyclic heterocycles as FGFR inhibitors
US10947230B2 (en) 2013-04-19 2021-03-16 Incyte Corporation Bicyclic heterocycles as FGFR inhibitors
US10450313B2 (en) 2013-04-19 2019-10-22 Incyte Holdings Corporation Bicyclic heterocycles as FGFR inhibitors
US10040790B2 (en) 2013-04-19 2018-08-07 Incyte Holdings Corporation Bicyclic heterocycles as FGFR inhibitors
US9533984B2 (en) 2013-04-19 2017-01-03 Incyte Holdings Corporation Bicyclic heterocycles as FGFR inhibitors
US9604984B2 (en) 2014-05-23 2017-03-28 Genentech, Inc. 5-chloro-2-difluoromethoxyphenyl pyrazolopyrimidine compounds, compositions and methods of use thereof
US9346815B2 (en) 2014-05-23 2016-05-24 Genentech, Inc. 5-chloro-2-difluoromethoxyphenyl pyrazolopyrimidine compounds, compositions and methods of use thereof
AU2015292818B2 (en) * 2014-07-21 2020-01-16 Dana-Farber Cancer Institute, Inc. Imidazolyl kinase inhibitors and uses thereof
US10975058B2 (en) 2014-07-21 2021-04-13 Dana-Farber Cancer Institute, Inc. Imidazolyl kinase inhibitors and uses thereof
US10851105B2 (en) 2014-10-22 2020-12-01 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US10016438B2 (en) 2015-02-20 2018-07-10 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US9708318B2 (en) 2015-02-20 2017-07-18 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US10214528B2 (en) 2015-02-20 2019-02-26 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US11667635B2 (en) 2015-02-20 2023-06-06 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US10632126B2 (en) 2015-02-20 2020-04-28 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US10738048B2 (en) 2015-02-20 2020-08-11 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US9890156B2 (en) 2015-02-20 2018-02-13 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US9580423B2 (en) 2015-02-20 2017-02-28 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US9801889B2 (en) 2015-02-20 2017-10-31 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US11173162B2 (en) 2015-02-20 2021-11-16 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US10251892B2 (en) 2015-02-20 2019-04-09 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US11014923B2 (en) 2015-02-20 2021-05-25 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
WO2016173557A1 (en) * 2015-04-30 2016-11-03 中国科学院上海药物研究所 Compound having kinase inhibition activity, and preparation method and uses
US11725011B2 (en) 2016-07-05 2023-08-15 The General Hospital Corporation Bicyclic urea kinase inhibitors and uses thereof
US10954242B2 (en) 2016-07-05 2021-03-23 The Broad Institute, Inc. Bicyclic urea kinase inhibitors and uses thereof
US11241435B2 (en) 2016-09-16 2022-02-08 The General Hospital Corporation Uses of salt-inducible kinase (SIK) inhibitors for treating osteoporosis
US11578075B2 (en) 2016-11-22 2023-02-14 Dana-Farber Cancer Institute, Inc. Degradation of protein kinases by conjugation of protein kinase inhibitors with E3 ligase ligand and methods of use
WO2018098280A1 (en) * 2016-11-22 2018-05-31 Dana-Farber Cancer Institute, Inc. Degradation of protein kinases by conjugation of protein kinase inhibitors with e3 ligase ligand and methods of use
US10899768B2 (en) 2016-11-22 2021-01-26 Dana-Farber Cancer Institute, Inc. Degradation of protein kinases by conjugation of protein kinase inhibitors with E3 ligase ligand and methods of use
US11285158B2 (en) 2017-02-28 2022-03-29 The General Hospital Corporation Uses of pyrimidopyrimidinones as SIK inhibitors
US11878019B2 (en) 2017-02-28 2024-01-23 The General Hospital Corporation Uses of pyrimidopyrimidinones as SIK inhibitors
US10307426B2 (en) 2017-05-22 2019-06-04 Genentech, Inc. Therapeutic compounds and compositions, and methods of use thereof
US10611762B2 (en) 2017-05-26 2020-04-07 Incyte Corporation Crystalline forms of a FGFR inhibitor and processes for preparing the same
US11472801B2 (en) 2017-05-26 2022-10-18 Incyte Corporation Crystalline forms of a FGFR inhibitor and processes for preparing the same
US11466004B2 (en) 2018-05-04 2022-10-11 Incyte Corporation Solid forms of an FGFR inhibitor and processes for preparing the same
US11174257B2 (en) 2018-05-04 2021-11-16 Incyte Corporation Salts of an FGFR inhibitor
US11866431B2 (en) 2018-11-09 2024-01-09 Vivace Therapeutics, Inc. Bicyclic compounds
US11628162B2 (en) 2019-03-08 2023-04-18 Incyte Corporation Methods of treating cancer with an FGFR inhibitor
US11420935B2 (en) 2019-04-16 2022-08-23 Vivace Therapeutics, Inc. Bicyclic compounds
US11591329B2 (en) 2019-07-09 2023-02-28 Incyte Corporation Bicyclic heterocycles as FGFR inhibitors
US11607416B2 (en) 2019-10-14 2023-03-21 Incyte Corporation Bicyclic heterocycles as FGFR inhibitors
US11566028B2 (en) 2019-10-16 2023-01-31 Incyte Corporation Bicyclic heterocycles as FGFR inhibitors
US11407750B2 (en) 2019-12-04 2022-08-09 Incyte Corporation Derivatives of an FGFR inhibitor
US11897891B2 (en) 2019-12-04 2024-02-13 Incyte Corporation Tricyclic heterocycles as FGFR inhibitors
WO2022087008A1 (en) * 2020-10-21 2022-04-28 Vivace Therapeutics, Inc. Tertiary carboxamide compounds
US11939331B2 (en) 2021-06-09 2024-03-26 Incyte Corporation Tricyclic heterocycles as FGFR inhibitors

Also Published As

Publication number Publication date
WO2009073153A3 (en) 2009-12-30
EP2222162B1 (en) 2016-11-16
EP3150593B1 (en) 2019-04-24
JP2011504927A (en) 2011-02-17
US8921336B2 (en) 2014-12-30
AU2008331867B2 (en) 2014-05-22
US20110312908A1 (en) 2011-12-22
EP3912973A3 (en) 2022-02-16
EP3912973A2 (en) 2021-11-24
US20150094275A1 (en) 2015-04-02
US11932646B2 (en) 2024-03-19
BRPI0819453A2 (en) 2014-10-07
US20200347068A1 (en) 2020-11-05
MX2012003309A (en) 2012-11-20
JP5941513B2 (en) 2016-06-29
EP3536687A1 (en) 2019-09-11
ES2734288T3 (en) 2019-12-05
EA019869B1 (en) 2014-06-30
AU2008331867A1 (en) 2009-06-11
CN104311563A (en) 2015-01-28
US20170349595A1 (en) 2017-12-07
CN104311563B (en) 2016-12-07
CA2707046A1 (en) 2009-06-11
KR20100130583A (en) 2010-12-13
CN101917849B (en) 2014-11-05
EA201070595A1 (en) 2011-02-28
EP3150593A1 (en) 2017-04-05
ES2610190T3 (en) 2017-04-26
JP2015051977A (en) 2015-03-19
ES2893580T3 (en) 2022-02-09
CN104327084A (en) 2015-02-04
EP2222162A4 (en) 2012-01-18
US20190084990A1 (en) 2019-03-21
CN104327084B (en) 2017-06-06
MX2010005671A (en) 2010-06-25
EP3536687B8 (en) 2021-09-08
US11254682B2 (en) 2022-02-22
EP3150593B8 (en) 2019-08-21
CN101917849A (en) 2010-12-15
JP5779778B2 (en) 2015-09-16
EP2222162A2 (en) 2010-09-01
US10787455B2 (en) 2020-09-29
EP3536687B1 (en) 2021-07-14
US9670214B2 (en) 2017-06-06
MX2012003311A (en) 2013-05-08
US20220135573A1 (en) 2022-05-05

Similar Documents

Publication Publication Date Title
US11254682B2 (en) Small molecule myristate inhibitors of Bcr-abl and methods of use
KR101917972B1 (en) Thieno[2,3-c]pyrrol-4-one derivatives as erk inhibitors
US7390805B2 (en) 4-amino-6-phenyl-pyrrolo[2,3-d]pyrimidine derivatives
CA2585660A1 (en) 1,4 substituted pyrazolopyrimidines as kinase inhibitors
JP2012504157A (en) Heterocyclic JAK kinase inhibitors
US7323469B2 (en) 7H-pyrrolo[2,3-d]pyrimidine derivatives
US20240158403A1 (en) Small molecule myristate inhibitors of bcr-abl and methods of use
AU2014215991A1 (en) Small molecule myristate inhibitors of bcr-abl and methods of use
KR20180122493A (en) Novel TYRO 3 inhibitory compounds, preparation method thereof, pharmaceutical composition for use in preventing or treating TYRO 3 relating diseases containing the same as an active ingredient
KR20220090443A (en) Heteroaryl derivative compounds, and uses thereof

Legal Events

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

Ref document number: 200880123462.5

Country of ref document: CN

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

Ref document number: 08857261

Country of ref document: EP

Kind code of ref document: A2

WWE Wipo information: entry into national phase

Ref document number: 2008331867

Country of ref document: AU

Ref document number: MX/A/2010/005671

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 2707046

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2010536015

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2000/KOLNP/2010

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 201070595

Country of ref document: EA

ENP Entry into the national phase

Ref document number: 2008331867

Country of ref document: AU

Date of ref document: 20081128

Kind code of ref document: A

REEP Request for entry into the european phase

Ref document number: 2008857261

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2008857261

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 20107013953

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 12745496

Country of ref document: US

ENP Entry into the national phase

Ref document number: PI0819453

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20100528